Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Biochemistry ; 62(8): 1342-1346, 2023 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-37021938

RESUMO

Some bacteria survive in nutrient-poor environments and resist killing by antimicrobials by forming spores. The cortex layer of the peptidoglycan cell wall that surrounds mature spores contains a unique modification, muramic-δ-lactam, that is essential for spore germination and outgrowth. Two proteins, the amidase CwlD and the deacetylase PdaA, are required for muramic-δ-lactam synthesis in cells, but their combined ability to generate muramic-δ-lactam has not been directly demonstrated. Here we report an in vitro reconstitution of cortex peptidoglycan biosynthesis, and we show that CwlD and PdaA together are sufficient for muramic-δ-lactam formation. Our method enables characterization of the individual reaction steps, and we show for the first time that PdaA has transamidase activity, catalyzing both the deacetylation of N-acetylmuramic acid and cyclization of the product to form muramic-δ-lactam. This activity is unique among peptidoglycan deacetylases and is notable because it may involve the direct ligation of a carboxylic acid with a primary amine. Our reconstitution products are nearly identical to the cortex peptidoglycan found in spores, and we expect that they will be useful substrates for future studies of enzymes that act on the spore cortex.


Assuntos
Peptidoglicano , Esporos Bacterianos , Esporos Bacterianos/química , Esporos Bacterianos/metabolismo , Peptidoglicano/química , Bactérias/metabolismo , Parede Celular/química , Lactamas/metabolismo , Proteínas de Bactérias/metabolismo
2.
J Am Chem Soc ; 141(33): 12994-12997, 2019 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-31386359

RESUMO

The bacterial cell wall is composed of peptidoglycan, and its biosynthesis is an established target for antibiotics. Peptidoglycan is assembled from a glycopeptide precursor, Lipid II, that is polymerized by peptidoglycan glycosyltransferases into glycan strands that are subsequently cross-linked to form the mature cell wall. For decades bacteria were thought to contain only one family of enzymes that polymerize Lipid II, but recently, the ubiquitous Shape, Elongation, Division, and Sporulation (SEDS)-family proteins RodA and FtsW were shown to be peptidoglycan polymerases. Because RodA and FtsW are essential in nearly all bacteria, these enzymes are promising targets for new antibiotics. However, almost nothing is known about the mechanisms of these polymerases. Here, we report that SEDS proteins synthesize peptidoglycan by adding new Lipid II monomers to the reducing end of the growing glycan chain. Using substrates that can only react at the reducing end, we also show that the glycosyl donor and acceptor in the polymerization reaction have distinct lipid requirements. These findings provide the first fundamental insights into the mechanism of SEDS-family polymerases and lay the groundwork for future biochemical and structural studies.


Assuntos
Proteínas de Bactérias/metabolismo , Peptidoglicano Glicosiltransferase/metabolismo , Peptidoglicano/metabolismo , Staphylococcus aureus/metabolismo , Vias Biossintéticas , Humanos , Peptidoglicano/química , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/química , Especificidade por Substrato
3.
Artigo em Inglês | MEDLINE | ID: mdl-30323039

RESUMO

New antibiotics are needed to combat the growing problem of resistant bacterial infections. An attractive avenue toward the discovery of such next-generation therapies is to identify novel inhibitors of clinically validated targets, like cell wall biogenesis. We have therefore developed a pathway-directed whole-cell screen for small molecules that block the activity of the Rod system of Escherichia coli This conserved multiprotein complex is required for cell elongation and the morphogenesis of rod-shaped bacteria. It is composed of cell wall synthases and membrane proteins of unknown function that are organized by filaments of the actin-like MreB protein. Our screen takes advantage of the conditional essentiality of the Rod system and the ability of the beta-lactam mecillinam (also known as amdinocillin) to cause a toxic malfunctioning of the machinery. Rod system inhibitors can therefore be identified as molecules that promote growth in the presence of mecillinam under conditions permissive for the growth of Rod- cells. A screen of ∼690,000 compounds identified 1,300 compounds that were active against E. coli Pathway-directed screening of a majority of this subset of compounds for Rod inhibitors successfully identified eight analogs of the MreB antagonist A22. Further characterization of the A22 analogs identified showed that their antibiotic activity under conditions where the Rod system is essential was strongly correlated with their ability to suppress mecillinam toxicity. This result combined with those from additional biological studies reinforce the notion that A22-like molecules are relatively specific for MreB and suggest that the lipoprotein transport factor LolA is unlikely to be a physiologically relevant target as previously proposed.


Assuntos
Antibacterianos/farmacologia , Parede Celular/metabolismo , Escherichia coli/efeitos dos fármacos , Peptidoglicano/metabolismo , Andinocilina/farmacologia , Andinocilina/toxicidade , Proteínas de Bactérias/antagonistas & inibidores , Proteínas do Citoesqueleto/antagonistas & inibidores , Farmacorresistência Bacteriana Múltipla/genética , Escherichia coli/genética , Proteínas de Escherichia coli/antagonistas & inibidores , Testes de Sensibilidade Microbiana , Proteínas de Ligação às Penicilinas/antagonistas & inibidores , Proteínas de Ligação às Penicilinas/metabolismo
4.
J Am Chem Soc ; 139(49): 17727-17730, 2017 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-29182854

RESUMO

Penicillin-binding proteins (PBPs) are enzymes involved in the assembly of the bacterial cell wall, a major target for antibiotics. These proteins are classified by mass into high-molecular-weight PBPs, which are transpeptidases that form peptidoglycan cross-links, and low-molecular-weight PBPs, which are typically hydrolases. We report a functionally unique family of low-molecular-weight PBPs that act as transpeptidases rather than hydrolases, but they do not cross-link peptidoglycan. We show that these PBPs can exchange d-amino acids bearing chemical tags or affinity handles into peptidoglycan precursors, including Lipid II, enabling biochemical studies of proteins involved in cell wall assembly. We report that, in two organisms, the PBPs incorporate lysine into cellular peptidoglycan and that, further, the PBPs have the unprecedented ability to transfer the primary ε-amine of lysine to peptidoglycan.


Assuntos
Proteínas de Bactérias/classificação , Proteínas de Bactérias/metabolismo , Proteínas de Ligação às Penicilinas/classificação , Proteínas de Ligação às Penicilinas/metabolismo , Peptidil Transferases/química , Peptidil Transferases/metabolismo , Aminas/metabolismo , Proteínas de Bactérias/química , Domínio Catalítico , Parede Celular/química , Parede Celular/metabolismo , Enterococcus faecalis/enzimologia , Lisina/química , Lisina/metabolismo , Peso Molecular , Proteínas de Ligação às Penicilinas/química , Peptidoglicano/química , Peptidoglicano/metabolismo , Streptococcus gordonii/enzimologia , Uridina Difosfato Ácido N-Acetilmurâmico/análogos & derivados , Uridina Difosfato Ácido N-Acetilmurâmico/metabolismo
5.
Adv Funct Mater ; 26(21): 3599-3611, 2016 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-28713229

RESUMO

Many types of slippery liquid-infused porous surfaces (or 'SLIPS') can resist adhesion and colonization by microorganisms. These 'slippery' materials thus offer new approaches to prevent fouling on a range of commercial and industrial surfaces, including biomedical devices. However, while SLIPS can prevent fouling on surfaces to which they are applied, they can currently do little to prevent the proliferation of non-adherent (planktonic) organisms, stop them from colonizing other surfaces, or prevent them from engaging in other behaviors that could lead to infection and associated burdens. Here, we report an approach to the design of multi-functional SLIPS that addresses these issues and expands the potential utility of slippery surfaces in antimicrobial contexts. Our approach is based on the incorporation and controlled release of small-molecule antimicrobial agents from the porous matrices used to host infused slippery oil phases. We demonstrate that SLIPS fabricated using nanoporous polymer multilayers can prevent short- and longer-term colonization and biofilm formation by four common fungal and bacterial pathogens (Candida albicans, Pseudomonas aeruginosa, Escherichia coli, and Staphylococcus aureus), and that the polymer and oil phases comprising these materials can be exploited to load and sustain the release of triclosan, a model hydrophobic and broad-spectrum antimicrobial agent, into surrounding media. This approach both improves the inherent anti-fouling properties of these materials and endows them with the ability to efficiently kill planktonic pathogens. Finally, we show that this approach can be used to fabricate dual-action SLIPS on complex surfaces, including the luminal surfaces of flexible catheter tubes. This strategy has the potential to be general; we anticipate that the materials, strategies, and concepts reported here will enable new approaches to the design of slippery surfaces with improved anti-fouling properties and open the door to new applications of slippery liquid-infused materials that host or promote the release of a variety of other active agents.

6.
J Am Chem Soc ; 137(4): 1510-9, 2015 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-25574853

RESUMO

The opportunistic pathogen Pseudomonas aeruginosa uses three interwoven quorum-sensing (QS) circuits-Las, Rhl, and Pqs-to regulate the global expression of myriad virulence-associated genes. Interception of these signaling networks with small molecules represents an emerging strategy for the development of anti-infective agents against this bacterium. In the current study, we applied a chemical approach to investigate how the Las-Rhl-Pqs QS hierarchy coordinates key virulence phenotypes in wild-type P. aeruginosa. We screened a focused library of synthetic, non-native N-acyl l-homoserine lactones and identified compounds that can drastically alter production of two important virulence factors: pyocyanin and rhamnolipid. We demonstrate that these molecules act by targeting RhlR in P. aeruginosa, a QS receptor that has seen far less scrutiny to date relative to other circuitry. Unexpectedly, modulation of RhlR activity by a single compound induces inverse regulation of pyocyanin and rhamnolipid, a result that was not predicted using genetic approaches to interrogate QS in P. aeruginosa. Further, we show that certain RhlR agonists strongly repress Pqs signaling, revealing disruption of Rhl-Pqs cross-regulation as a novel mechanism for QS inhibition. These compounds significantly expand the known repertoire of chemical probes available to study RhlR in P. aeruginosa. Moreover, our results suggest that designing chemical agents to disrupt Rhl-Pqs crosstalk could be an effective antivirulence strategy to fight this common pathogen.


Assuntos
Acil-Butirolactonas/química , Acil-Butirolactonas/farmacologia , Antibacterianos/química , Antibacterianos/farmacologia , Pseudomonas aeruginosa/efeitos dos fármacos , Percepção de Quorum/efeitos dos fármacos , Glicolipídeos/metabolismo , Humanos , Infecções por Pseudomonas/tratamento farmacológico , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/fisiologia , Piocianina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fatores de Virulência/metabolismo
7.
J Am Chem Soc ; 137(46): 14626-39, 2015 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-26491787

RESUMO

Quorum sensing (QS) is a chemical signaling mechanism that allows bacterial populations to coordinate gene expression in response to social and environmental cues. Many bacterial pathogens use QS to initiate infection at high cell densities. Over the past two decades, chemical antagonists of QS in pathogenic bacteria have attracted substantial interest for use both as tools to further elucidate QS mechanisms and, with further development, potential anti-infective agents. Considerable recent research has been devoted to the design of small molecules capable of modulating the LasR QS receptor in the opportunistic pathogen Pseudomonas aeruginosa. These molecules hold significant promise in a range of contexts; however, as most compounds have been developed independently, comparative activity data for these compounds are scarce. Moreover, the mechanisms by which the bulk of these compounds act are largely unknown. This paucity of data has stalled the choice of an optimal chemical scaffold for further advancement. Herein, we submit the best-characterized LasR modulators to standardized cell-based reporter and QS phenotypic assays in P. aeruginosa, and we report the first comprehensive set of comparative LasR activity data for these compounds. Our experiments uncovered multiple interesting mechanistic phenomena (including a potential alternative QS-modulatory ligand binding site/partner) that provide new, and unexpected, insights into the modes by which many of these LasR ligands act. The lead compounds, data trends, and mechanistic insights reported here will significantly aid the design of new small molecule QS inhibitors and activators in P. aeruginosa, and in other bacteria, with enhanced potencies and defined modes of action.


Assuntos
Pseudomonas aeruginosa/fisiologia , Percepção de Quorum , Transporte Biológico , Ligantes , Pseudomonas aeruginosa/metabolismo
8.
Breast Cancer Res ; 16(3): R57, 2014 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-24890385

RESUMO

INTRODUCTION: Transforming growth factor-ßs (TGF-ßs) play a dual role in breast cancer, with context-dependent tumor-suppressive or pro-oncogenic effects. TGF-ß antagonists are showing promise in early-phase clinical oncology trials to neutralize the pro-oncogenic effects. However, there is currently no way to determine whether the tumor-suppressive effects of TGF-ß are still active in human breast tumors at the time of surgery and treatment, a situation that could lead to adverse therapeutic responses. METHODS: Using a breast cancer progression model that exemplifies the dual role of TGF-ß, promoter-wide chromatin immunoprecipitation and transcriptomic approaches were applied to identify a core set of TGF-ß-regulated genes that specifically reflect only the tumor-suppressor arm of the pathway. The clinical significance of this signature and the underlying biology were investigated using bioinformatic analyses in clinical breast cancer datasets, and knockdown validation approaches in tumor xenografts. RESULTS: TGF-ß-driven tumor suppression was highly dependent on Smad3, and Smad3 target genes that were specifically enriched for involvement in tumor suppression were identified. Patterns of Smad3 binding reflected the preexisting active chromatin landscape, and target genes were frequently regulated in opposite directions in vitro and in vivo, highlighting the strong contextuality of TGF-ß action. An in vivo-weighted TGF-ß/Smad3 tumor-suppressor signature was associated with good outcome in estrogen receptor-positive breast cancer cohorts. TGF-ß/Smad3 effects on cell proliferation, differentiation and ephrin signaling contributed to the observed tumor suppression. CONCLUSIONS: Tumor-suppressive effects of TGF-ß persist in some breast cancer patients at the time of surgery and affect clinical outcome. Carefully tailored in vitro/in vivo genomic approaches can identify such patients for exclusion from treatment with TGF-ß antagonists.


Assuntos
Neoplasias da Mama/genética , Proteína Smad3/genética , Fator de Crescimento Transformador beta/genética , Proteínas Supressoras de Tumor/genética , Neoplasias da Mama/patologia , Diferenciação Celular , Linhagem Celular Tumoral , Proliferação de Células , Efrinas/metabolismo , Feminino , Humanos , Regiões Promotoras Genéticas/genética , Interferência de RNA , RNA Interferente Pequeno , Receptor EphA2/metabolismo , Proteína Smad2/genética , Proteína Smad3/biossíntese , Fator de Crescimento Transformador beta/antagonistas & inibidores , Fator de Crescimento Transformador beta/biossíntese , Proteínas Supressoras de Tumor/antagonistas & inibidores
9.
Breast Cancer Res ; 12(5): R83, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20942910

RESUMO

INTRODUCTION: Molecular dissection of the signaling pathways that underlie complex biological responses in the mammary epithelium is limited by the difficulty of propagating large numbers of mouse mammary epithelial cells, and by the inability of ribonucleic acid interference (RNAi)-based knockdown approaches to fully ablate gene function. Here we describe a method for the generation of conditionally immortalized mammary epithelial cells with defined genetic defects, and we show how such cells can be used to investigate complex signal transduction processes using the transforming growth factor beta (TGFß/Smad pathway as an example. METHODS: We intercrossed the previously described H-2Kb-tsA58 transgenic mouse (Immortomouse) which expresses a temperature-sensitive mutant of the simian virus-40 large T-antigen (tsTAg), with mice of differing Smad genotypes. A panel of conditionally immortalized mammary epithelial cell (IMEC) cultures were derived from the virgin mammary glands of offspring of these crosses and used to assess the Smad dependency of different biological responses to TGFß. RESULTS: IMECs could be propagated indefinitely at permissive temperatures and had a stable epithelial phenotype, resembling primary mammary epithelial cells with respect to several criteria, including responsiveness to TGFß. Using this panel of cells, we demonstrated that Smad3, but not Smad2, is necessary for TGFß-induced apoptotic, growth inhibitory and EMT responses, whereas either Smad can support TGFß-induced invasion as long as a threshold level of total Smad is exceeded. CONCLUSIONS: This work demonstrates the practicality and utility of generating conditionally immortalized mammary epithelial cell lines from genetically modified Immortomice for detailed investigation of complex signaling pathways in the mammary epithelium.


Assuntos
Células Epiteliais/metabolismo , Glândulas Mamárias Animais/metabolismo , Transdução de Sinais , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Apoptose/genética , Diferenciação Celular , Movimento Celular , Células Cultivadas , Transição Epitelial-Mesenquimal/genética , Feminino , Técnicas de Inativação de Genes , Glândulas Mamárias Animais/citologia , Camundongos , Camundongos Transgênicos , Proteína Smad2/genética , Proteína Smad3/genética , Fator de Crescimento Transformador beta/genética
10.
Elife ; 92020 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-32011233

RESUMO

Increasing Neisseria gonorrhoeae resistance to ceftriaxone, the last antibiotic recommended for empiric gonorrhea treatment, poses an urgent public health threat. However, the genetic basis of reduced susceptibility to ceftriaxone is not completely understood: while most ceftriaxone resistance in clinical isolates is caused by target site mutations in penA, some isolates lack these mutations. We show that penA-independent ceftriaxone resistance has evolved multiple times through distinct mutations in rpoB and rpoD. We identify five mutations in these genes that each increase resistance to ceftriaxone, including one mutation that arose independently in two lineages, and show that clinical isolates from multiple lineages are a single nucleotide change from ceftriaxone resistance. These RNA polymerase mutations cause large-scale transcriptional changes without altering susceptibility to other antibiotics, reducing growth rate, or deranging cell morphology. These results underscore the unexpected diversity of pathways to resistance and the importance of continued surveillance for novel resistance mutations.


Assuntos
Resistência às Cefalosporinas/genética , RNA Polimerases Dirigidas por DNA/genética , Mutação de Sentido Incorreto , Neisseria gonorrhoeae/efeitos dos fármacos , Antibacterianos/farmacologia , Proteínas de Bactérias/genética , Cefalosporinas/farmacologia , Genes Bacterianos , Testes de Sensibilidade Microbiana , Neisseria gonorrhoeae/genética
11.
Clin Cancer Res ; 26(3): 643-656, 2020 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-31582516

RESUMO

PURPOSE: TGFßs are overexpressed in many advanced cancers and promote cancer progression through mechanisms that include suppression of immunosurveillance. Multiple strategies to antagonize the TGFß pathway are in early-phase oncology trials. However, TGFßs also have tumor-suppressive activities early in tumorigenesis, and the extent to which these might be retained in advanced disease has not been fully explored. EXPERIMENTAL DESIGN: A panel of 12 immunocompetent mouse allograft models of metastatic breast cancer was tested for the effect of neutralizing anti-TGFß antibodies on lung metastatic burden. Extensive correlative biology analyses were performed to assess potential predictive biomarkers and probe underlying mechanisms. RESULTS: Heterogeneous responses to anti-TGFß treatment were observed, with 5 of 12 models (42%) showing suppression of metastasis, 4 of 12 (33%) showing no response, and 3 of 12 (25%) showing an undesirable stimulation (up to 9-fold) of metastasis. Inhibition of metastasis was immune-dependent, whereas stimulation of metastasis was immune-independent and targeted the tumor cell compartment, potentially affecting the cancer stem cell. Thus, the integrated outcome of TGFß antagonism depends on a complex balance between enhancing effective antitumor immunity and disrupting persistent tumor-suppressive effects of TGFß on the tumor cell. Applying transcriptomic signatures derived from treatment-naïve mouse primary tumors to human breast cancer datasets suggested that patients with breast cancer with high-grade, estrogen receptor-negative disease are most likely to benefit from anti-TGFß therapy. CONCLUSIONS: Contrary to dogma, tumor-suppressive responses to TGFß are retained in some advanced metastatic tumors. Safe deployment of TGFß antagonists in the clinic will require good predictive biomarkers.


Assuntos
Antineoplásicos Imunológicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Regulação Neoplásica da Expressão Gênica , Genes Supressores de Tumor , Neoplasias Pulmonares/tratamento farmacológico , Células-Tronco Neoplásicas/efeitos dos fármacos , Fator de Crescimento Transformador beta/antagonistas & inibidores , Animais , Neoplasias da Mama/imunologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Humanos , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Células-Tronco Neoplásicas/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/imunologia , Fator de Crescimento Transformador beta/metabolismo , Resultado do Tratamento
12.
Nat Microbiol ; 4(4): 587-594, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30692671

RESUMO

The peptidoglycan cell wall is essential for the survival and morphogenesis of bacteria1. For decades, it was thought that only class A penicillin-binding proteins (PBPs) and related enzymes effected peptidoglycan synthesis. Recently, it was shown that RodA-a member of the unrelated SEDS protein family-also acts as a peptidoglycan polymerase2-4. Not all bacteria require RodA for growth; however, its homologue, FtsW, is a core member of the divisome complex that appears to be universally essential for septal cell wall assembly5,6. FtsW was previously proposed to translocate the peptidoglycan precursor lipid II across the cytoplasmic membrane7,8. Here, we report that purified FtsW polymerizes lipid II into peptidoglycan, but show that its polymerase activity requires complex formation with its partner class B PBP. We further demonstrate that the polymerase activity of FtsW is required for its function in vivo. Thus, our findings establish FtsW as a peptidoglycan polymerase that works with its cognate class B PBP to produce septal peptidoglycan during cell division.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Ligação às Penicilinas/metabolismo , Peptidoglicano/metabolismo , Staphylococcus aureus/enzimologia , Streptococcus thermophilus/enzimologia , Proteínas de Bactérias/genética , Divisão Celular , Parede Celular/genética , Parede Celular/metabolismo , Proteínas de Membrana/genética , Proteínas de Ligação às Penicilinas/genética , Ligação Proteica , Staphylococcus aureus/citologia , Staphylococcus aureus/genética , Streptococcus thermophilus/citologia , Streptococcus thermophilus/genética
13.
Elife ; 72018 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-30324906

RESUMO

In most well-studied rod-shaped bacteria, peptidoglycan is primarily crosslinked by penicillin-binding proteins (PBPs). However, in mycobacteria, crosslinks formed by L,D-transpeptidases (LDTs) are highly abundant. To elucidate the role of these unusual crosslinks, we characterized Mycobacterium smegmatis cells lacking all LDTs. We find that crosslinks generate by LDTs are required for rod shape maintenance specifically at sites of aging cell wall, a byproduct of polar elongation. Asymmetric polar growth leads to a non-uniform distribution of these two types of crosslinks in a single cell. Consequently, in the absence of LDT-mediated crosslinks, PBP-catalyzed crosslinks become more important. Because of this, Mycobacterium tuberculosis (Mtb) is more rapidly killed using a combination of drugs capable of PBP- and LDT- inhibition. Thus, knowledge about the spatial and genetic relationship between drug targets can be exploited to more effectively treat this pathogen.


Assuntos
Reagentes de Ligações Cruzadas/metabolismo , Mycobacterium smegmatis/metabolismo , Peptidoglicano/metabolismo , Aminoácidos/metabolismo , Aminoaciltransferases/metabolismo , Amoxicilina/farmacologia , Bacillus/metabolismo , Parede Celular/metabolismo , Escherichia coli/metabolismo , Fluorescência , Cinética , Meropeném/farmacologia , Viabilidade Microbiana , Modelos Biológicos , Mycobacterium smegmatis/efeitos dos fármacos , Proteínas de Ligação às Penicilinas/metabolismo , Peptidoglicano/química
14.
FEMS Microbiol Rev ; 40(5): 774-94, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27268906

RESUMO

Bacteria can utilize chemical signals to coordinate the expression of group-beneficial behaviors in a method of cell-cell communication called quorum sensing (QS). The discovery that QS controls the production of virulence factors and biofilm formation in many common pathogens has driven an explosion of research aimed at both deepening our fundamental understanding of these regulatory networks and developing chemical agents that can attenuate QS signaling. The inherently chemical nature of QS makes studying these pathways with small molecule tools a complementary approach to traditional microbiology techniques. Indeed, chemical tools are beginning to yield new insights into QS regulation and provide novel strategies to inhibit QS. Here, we review the most recent advances in the development of chemical probes of QS systems in Gram-negative bacteria, with an emphasis on the opportunistic pathogen Pseudomonas aeruginosa We first describe reports of novel small molecule modulators of QS receptors and QS signal synthases. Next, in several case studies, we showcase how chemical tools have been deployed to reveal new knowledge of QS biology and outline lessons for how researchers might best target QS to combat bacterial virulence. To close, we detail the outstanding challenges in the field and suggest strategies to overcome these issues.


Assuntos
Proteínas de Bactérias/antagonistas & inibidores , Pseudomonas aeruginosa/patogenicidade , Percepção de Quorum/efeitos dos fármacos , Percepção de Quorum/fisiologia , Proteínas Repressoras/antagonistas & inibidores , Transativadores/antagonistas & inibidores , Fatores de Transcrição/antagonistas & inibidores , Acil-Butirolactonas/metabolismo , Biofilmes/crescimento & desenvolvimento , Pseudomonas aeruginosa/metabolismo , Transdução de Sinais , Fatores de Virulência/biossíntese
15.
Cell Chem Biol ; 23(3): 361-9, 2016 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-26905657

RESUMO

Nutritional cues differentially influence the activities of the three quorum sensing (QS) circuits-Las, Rhl, and Pqs-in the pathogen Pseudomonas aeruginosa. A full understanding of how these systems work together to tune virulence factor production to the environment is lacking. Here, we used chemical probes to evaluate the contribution of each QS circuit to virulence in wild-type P. aeruginosa under defined environmental conditions. Our results indicate that Rhl and Pqs drive virulence factor production in phosphate- and iron-limiting environments, while Las has a minor influence. Consequently, simultaneous inhibition of Rhl and Pqs can attenuate virulence in environments where Las inhibition fails. The activity trends generated in this study can be extrapolated to predict QS inhibitor activity in infection-relevant environments, such as cystic fibrosis sputum. These results indicate that environmental signals can drastically alter the efficacy of small-molecule QS inhibitors in P. aeruginosa and possibly other pathogens.


Assuntos
Pseudomonas aeruginosa/efeitos dos fármacos , Percepção de Quorum/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Estrutura Molecular , Pseudomonas aeruginosa/crescimento & desenvolvimento , Pseudomonas aeruginosa/patogenicidade , Bibliotecas de Moléculas Pequenas/química , Fatores de Virulência/antagonistas & inibidores , Fatores de Virulência/biossíntese
16.
ACS Infect Dis ; 2(7): 509-17, 2016 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-27626103

RESUMO

Surfaces that can both prevent bacterial biofouling and inhibit the expression of virulence phenotypes in surrounding planktonic bacteria are of interest in a broad range of contexts. Here, we report new slippery-liquid infused porous surfaces (SLIPS) that resist bacterial colonization (owing to inherent "slippery" surface character) and also attenuate virulence phenotypes in non-adherent cells by gradually releasing small-molecule quorum sensing inhibitors (QSIs). QSIs active against Pseudomonas aeruginosa can be loaded into SLIPS without loss of their slippery and antifouling properties, and imbedded agents can be released into surrounding media over hours to days depending on the structures of the loaded agent. This controlled-release approach is useful for inhibiting virulence factor production and can also inhibit bacterial biofilm formation on nearby, non-SLIPS-coated surfaces. Finally, we demonstrate that this approach is compatible with the simultaneous release of more than one type of QSI, enabling greater control over virulence and suggesting new opportunities to tune the antifouling properties of these slippery surfaces.


Assuntos
Antibacterianos/farmacologia , Incrustação Biológica/prevenção & controle , Bombas de Infusão/microbiologia , Plâncton/efeitos dos fármacos , Pseudomonas aeruginosa/efeitos dos fármacos , Antibacterianos/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Contaminação de Equipamentos/prevenção & controle , Plâncton/genética , Plâncton/patogenicidade , Plâncton/fisiologia , Porosidade , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/patogenicidade , Pseudomonas aeruginosa/fisiologia , Percepção de Quorum/efeitos dos fármacos , Propriedades de Superfície , Virulência/efeitos dos fármacos
17.
ACS Chem Biol ; 7(10): 1719-28, 2012 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-22853441

RESUMO

Many bacterial pathogens use quorum sensing (QS) to control virulence. As a result, the development of methods to intercept QS has attracted significant interest as a potential anti-infective therapy. Acinetobacter baumannii has emerged as a pan-drug-resistant pathogen and displays a remarkable ability to persist in hospital settings despite desiccation and antimicrobial treatment. Recent studies have shown that A. baumannii QS mutants have limited motility and fail to form mature biofilms; these phenotypes are linked to its ability to persist on biotic and abiotic surfaces and increase its pathogenicity. A. baumannii uses N-(3-hydroxydodecanoyl)-l-homoserine lactone (OH-dDHL) and its putative cognate receptor, AbaR, for QS. We sought to identify non-native ligands capable of blocking or promoting AbaR activity in A. baumannii for use as chemical probes to modulate QS phenotypes in this pathogen. We screened a focused library of synthetic, non-native N-acyl homoserine lactones (AHLs) to identify such compounds, and several highly potent antagonists and agonists were uncovered, with IC(50) and EC(50) values in the low micromolar range, respectively. The strongest AbaR antagonists largely contained aromatic acyl groups, whereas the AbaR agonists closely resembled OH-dDHL. Notably, the 10 most potent AbaR antagonists also strongly inhibited A. baumannii motility, and five antagonists reduced biofilm formation in A. baumannii by up to 40%. The discovery of these compounds is significant, as they represent, to our knowledge, the first non-native modulators of QS in A. baumannii to be reported and could find utility as new tools to study the role and timing of QS phenotypes in A. baumannii infections.


Assuntos
4-Butirolactona/análogos & derivados , Acinetobacter baumannii/efeitos dos fármacos , Percepção de Quorum/efeitos dos fármacos , 4-Butirolactona/química , 4-Butirolactona/farmacologia , Acilação , Biofilmes , Farmacorresistência Bacteriana Múltipla/efeitos dos fármacos , Genes Reporter/genética , Proteínas Repressoras/antagonistas & inibidores , Relação Estrutura-Atividade , beta-Galactosidase/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA