Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Plasmid ; 128: 102708, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37967733

RESUMO

The majority of large multiresistance plasmids of Staphylococcus aureus utilise a RepA_N-type replication initiation protein, the expression of which is regulated by a small antisense RNA (RNAI) that overlaps the rep mRNA leader. The pSK41/pGO1-family of conjugative plasmids additionally possess a small (86 codon) divergently transcribed ORF (orf86) located upstream of the rep locus. The product of pSK41 orf86 was predicted to have a helix-turn-helix motif suggestive of a likely function in transcriptional repression. In this study, we investigated the effect of Orf86 on transcription of thirteen pSK41 backbone promoters. We found that Orf86 only repressed transcription from the rep promoter, and hence now redesignate the product as Cop. Over-expression of Cop in trans reduced the copy number of pSK41 mini-replicons, both in the presence and absence of rnaI. in vitro protein-DNA binding experiments with purified 6 × His-Cop demonstrated specific DNA binding, adjacent to, and partially overlapping the -35 hexamer of the rep promoter. The crystal structure of Cop revealed a dimeric structure similar to other known transcriptional regulators. Cop mRNA was found to result from "read-through" transcription from the strong RNAI promoter that escapes the rnaI terminator. Thus, PrnaI is responsible for transcription of two distinct negative regulators of plasmid copy number; the antisense RNAI that primarily represses Rep translation, and Cop protein that can repress rep transcription. Deletion of cop in a native plasmid did not appear to impact copy number, indicating a cryptic auxiliary role.


Assuntos
Replicação do DNA , Staphylococcus aureus , Plasmídeos/genética , Staphylococcus aureus/genética , Sequência de Bases , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , DNA , RNA Mensageiro
2.
Proc Natl Acad Sci U S A ; 117(13): 7374-7381, 2020 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-32170007

RESUMO

Irinotecan treats a range of solid tumors, but its effectiveness is severely limited by gastrointestinal (GI) tract toxicity caused by gut bacterial ß-glucuronidase (GUS) enzymes. Targeted bacterial GUS inhibitors have been shown to partially alleviate irinotecan-induced GI tract damage and resultant diarrhea in mice. Here, we unravel the mechanistic basis for GI protection by gut microbial GUS inhibitors using in vivo models. We use in vitro, in fimo, and in vivo models to determine whether GUS inhibition alters the anticancer efficacy of irinotecan. We demonstrate that a single dose of irinotecan increases GI bacterial GUS activity in 1 d and reduces intestinal epithelial cell proliferation in 5 d, both blocked by a single dose of a GUS inhibitor. In a tumor xenograft model, GUS inhibition prevents intestinal toxicity and maintains the antitumor efficacy of irinotecan. Remarkably, GUS inhibitor also effectively blocks the striking irinotecan-induced bloom of Enterobacteriaceae in immune-deficient mice. In a genetically engineered mouse model of cancer, GUS inhibition alleviates gut damage, improves survival, and does not alter gut microbial composition; however, by allowing dose intensification, it dramatically improves irinotecan's effectiveness, reducing tumors to a fraction of that achieved by irinotecan alone, while simultaneously promoting epithelial regeneration. These results indicate that targeted gut microbial enzyme inhibitors can improve cancer chemotherapeutic outcomes by protecting the gut epithelium from microbial dysbiosis and proliferative crypt damage.


Assuntos
Microbioma Gastrointestinal/efeitos dos fármacos , Glucuronidase/antagonistas & inibidores , Glucuronidase/efeitos dos fármacos , Animais , Antineoplásicos Fitogênicos/farmacologia , Bactérias/efeitos dos fármacos , Modelos Animais de Doenças , Disbiose/tratamento farmacológico , Inibidores Enzimáticos/farmacologia , Feminino , Glucuronidase/metabolismo , Humanos , Irinotecano/farmacologia , Camundongos , Camundongos Nus , Neoplasias/tratamento farmacológico
3.
Proc Natl Acad Sci U S A ; 115(2): E152-E161, 2018 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-29269393

RESUMO

The gut microbiota harbor diverse ß-glucuronidase (GUS) enzymes that liberate glucuronic acid (GlcA) sugars from small-molecule conjugates and complex carbohydrates. However, only the Enterobacteriaceae family of human gut-associated Proteobacteria maintain a GUS operon under the transcriptional control of a glucuronide repressor, GusR. Despite its potential importance in Escherichia, Salmonella, Klebsiella, Shigella, and Yersinia opportunistic pathogens, the structure of GusR has not been examined. Here, we explore the molecular basis for GusR-mediated regulation of GUS expression in response to small-molecule glucuronides. Presented are 2.1-Å-resolution crystal structures of GusRs from Escherichia coli and Salmonella enterica in complexes with a glucuronide ligand. The GusR-specific DNA operator site in the regulatory region of the E. coli GUS operon is identified, and structure-guided GusR mutants pinpoint the residues essential for DNA binding and glucuronide recognition. Interestingly, the endobiotic estradiol-17-glucuronide and the xenobiotic indomethacin-acyl-glucuronide are found to exhibit markedly differential binding to these GusR orthologs. Using structure-guided mutations, we are able to transfer E. coli GusR's preferential DNA and glucuronide binding affinity to S. enterica GusR. Structures of putative GusR orthologs from GUS-encoding Firmicutes species also reveal functionally unique features of the Enterobacteriaceae GusRs. Finally, dominant-negative GusR variants are validated in cell-based studies. These data provide a molecular framework toward understanding the control of glucuronide utilization by opportunistic pathogens in the human gut.


Assuntos
Proteínas de Bactérias/genética , Enterobacteriaceae/genética , Microbioma Gastrointestinal/genética , Regulação Bacteriana da Expressão Gênica , Glucuronidase/genética , Sequência de Aminoácidos , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Cristalografia por Raios X , DNA/química , DNA/genética , DNA/metabolismo , Enterobacteriaceae/enzimologia , Escherichia coli/genética , Escherichia coli/metabolismo , Genes Reguladores/genética , Ácido Glucurônico/química , Ácido Glucurônico/metabolismo , Glucuronidase/química , Glucuronidase/metabolismo , Humanos , Mutação , Óperon/genética , Homologia de Sequência de Aminoácidos
4.
Biochemistry ; 59(40): 3939-3950, 2020 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-32993284

RESUMO

Phase II drug metabolism inactivates xenobiotics and endobiotics through the addition of either a glucuronic acid or sulfate moiety prior to excretion, often via the gastrointestinal tract. While the human gut microbial ß-glucuronidase enzymes that reactivate glucuronide conjugates in the intestines are becoming well characterized and even controlled by targeted inhibitors, the sulfatases encoded by the human gut microbiome have not been comprehensively examined. Gut microbial sulfatases are poised to reactivate xenobiotics and endobiotics, which are then capable of undergoing enterohepatic recirculation or exerting local effects on the gut epithelium. Here, using protein structure-guided methods, we identify 728 distinct microbiome-encoded sulfatase proteins from the 4.8 million unique proteins present in the Human Microbiome Project Stool Sample database and 1766 gut microbial sulfatases from the 9.9 million sequences in the Integrated Gene Catalogue. We purify a representative set of these sulfatases, elucidate crystal structures, and pinpoint unique structural motifs essential to endobiotic sulfate processing. Gut microbial sulfatases differentially process sulfated forms of the neurotransmitters serotonin and dopamine, and the hormones melatonin, estrone, dehydroepiandrosterone, and thyroxine in a manner dependent both on variabilities in active site architecture and on markedly distinct oligomeric states. Taken together, these data provide initial insights into the structural and functional diversity of gut microbial sulfatases, providing a path toward defining the roles these enzymes play in health and disease.


Assuntos
Bactérias/enzimologia , Microbioma Gastrointestinal , Microbiota , Sulfatases/metabolismo , Bactérias/química , Bactérias/genética , Bactérias/metabolismo , Domínio Catalítico , Fezes/microbiologia , Genes Bacterianos , Humanos , Modelos Moleculares , Conformação Proteica , Sulfatases/química , Sulfatases/genética
5.
Biochemistry ; 58(9): 1311-1317, 2019 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-30729778

RESUMO

The human gut microbiome is a ripe space for the discovery of new proteins and novel functions. Many genes in the gut microbiome encode glycoside hydrolases that help bacteria scavenge sugars present in the human gut. Glycoside hydrolase family 2 (GH2) is one group of sugar-scavenging proteins, which includes ß-glucuronidases (GUS) and ß-galacturonidases (GalAses), enzymes that cleave the sugar conjugates of the epimers glucuronate and galacturonate. Here we structurally and functionally characterize a GH2 GalAse and a hybrid GUS/GalAse, which reveal the molecular details that enable these GHs to differentiate a single stereocenter. First, we characterized a previously annotated GUS from Eisenbergiella tayi and demonstrated that it is, in fact, a GalAse. We determined the crystal structure of this GalAse, identified the key residue that confers GalAse activity, and convert this GalAse into a GUS by mutating a single residue. We performed bioinformatic analysis of 279 putative GUS enzymes from the human gut microbiome and identified 12 additional putative GH2 GalAses, one of which we characterized and confirmed is a GalAse. Lastly, we report the structure of a hybrid GUS/GalAse from Fusicatenibacter saccharivorans, which revealed a unique hexamer that positions the N-terminus of adjacent protomers in the aglycone binding site. Taken together, these data reveal a new class of bacterial GalAses in the human gut microbiome and unravel the structural details that differentiate GH2 GUSs and GalAses.


Assuntos
Microbioma Gastrointestinal/fisiologia , Glucuronidase/química , Glicosídeo Hidrolases/química , Domínio Catalítico , Cristalografia por Raios X , Fezes/microbiologia , Glucuronidase/genética , Glucuronidase/metabolismo , Glicosídeo Hidrolases/genética , Glicosídeo Hidrolases/metabolismo , Bacilos Gram-Negativos Anaeróbios Facultativos/genética , Humanos , Mutagênese Sítio-Dirigida , Conformação Proteica
6.
J Biol Chem ; 293(48): 18559-18573, 2018 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-30301767

RESUMO

The glycoside hydrolases encoded by the human gut microbiome play an integral role in processing a variety of exogenous and endogenous glycoconjugates. Here we present three structurally and functionally distinct ß-glucuronidase (GUS) glycoside hydrolases from a single human gut commensal microbe, Bacteroides uniformis We show using nine crystal structures, biochemical, and biophysical data that whereas these three proteins share similar overall folds, they exhibit different structural features that create three structurally and functionally unique enzyme active sites. Notably, quaternary structure plays an important role in creating distinct active site features that are hard to predict via structural modeling methods. The enzymes display differential processing capabilities toward glucuronic acid-containing polysaccharides and SN-38-glucuronide, a metabolite of the cancer drug irinotecan. We also demonstrate that GUS-specific and nonselective inhibitors exhibit varying potencies toward each enzyme. Together, these data highlight the diversity of GUS enzymes within a single Bacteroides gut commensal and advance our understanding of how structural details impact the specific roles microbial enzymes play in processing drug-glucuronide and glycan substrates.


Assuntos
Bacteroides/enzimologia , Microbioma Gastrointestinal , Glucuronidase/química , Glucuronidase/metabolismo , Isoenzimas/química , Isoenzimas/metabolismo , Sequência de Aminoácidos , Domínio Catalítico , Inibidores Enzimáticos/farmacologia , Ácido Glucárico/análogos & derivados , Glucuronidase/antagonistas & inibidores , Humanos , Conformação Proteica
7.
FASEB J ; 32(5): 2478-2491, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29295861

RESUMO

Cystic fibrosis (CF) is a common genetic disease with significantly increased mortality. CF airways exhibit ion transport abnormalities, including hyperactivity of the epithelial Na+ channel (ENaC). Short-palate lung and nasal epithelial clone 1 (SPLUNC1) is a multifunctional innate defense protein that is secreted into the airway lumen. We have previously demonstrated that SPLUNC1 binds to and inhibits ENaC to maintain fluid homeostasis in airway epithelia and that this process fails in CF airways. Despite this, how SPLUNC1 actually regulates ENaC is unknown. Here, we found that SPLUNC1 caused αγ-ENaC to internalize, whereas SPLUNC1 and ß-ENaC remained at the plasma membrane. Additional studies revealed that SPLUNC1 increased neural precursor cell-expressed developmentally down-regulated protein 4-2-dependent ubiquitination of α- but not ß- or γ-ENaC. We also labeled intracellular ENaC termini with green fluorescent protein and mCherry, and found that extracellular SPLUNC1 altered intracellular ENaC Forster resonance energy transfer. Taken together, our data indicate that SPLUNC1 is an allosteric regulator of ENaC that dissociates αßγ-ENaC to generate a new SPLUNC1-ß-ENaC complex. These data indicate a novel mode for regulating ENaC at the plasma membrane.-Kim, C. S., Ahmad, S., Wu, T., Walton, W. G., Redinbo, M. R., Tarran, R. SPLUNC1 is an allosteric modulator of the epithelial sodium channel.


Assuntos
Membrana Celular/metabolismo , Células Epiteliais/metabolismo , Canais Epiteliais de Sódio/metabolismo , Glicoproteínas/metabolismo , Complexos Multiproteicos/química , Mucosa Nasal/metabolismo , Fosfoproteínas/metabolismo , Regulação Alostérica/fisiologia , Membrana Celular/química , Membrana Celular/genética , Células Epiteliais/química , Canais Epiteliais de Sódio/química , Canais Epiteliais de Sódio/genética , Transferência Ressonante de Energia de Fluorescência , Glicoproteínas/química , Glicoproteínas/genética , Células HEK293 , Humanos , Proteínas Luminescentes , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Mucosa Nasal/química , Fosfoproteínas/química , Fosfoproteínas/genética , Proteína Vermelha Fluorescente
8.
Nucleic Acids Res ; 45(9): 5333-5348, 2017 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-28369583

RESUMO

Human GEN1 and yeast Yen1 are endonucleases with the ability to cleave Holliday junctions (HJs), which are proposed intermediates in recombination. In vivo, GEN1 and Yen1 function secondarily to Mus81, which has weak activity on intact HJs. We show that the genetic relationship is reversed in Drosophila, with Gen mutants having more severe defects than mus81 mutants. In vitro, DmGen, like HsGEN1, efficiently cleaves HJs, 5΄ flaps, splayed arms, and replication fork structures. We find that the cleavage rates for 5΄ flaps are significantly higher than those for HJs for both DmGen and HsGEN1, even in vast excess of enzyme over substrate. Kinetic studies suggest that the difference in cleavage rates results from a slow, rate-limiting conformational change prior to HJ cleavage: formation of a productive dimer on the HJ. Despite the stark difference in vivo that Drosophila uses Gen over Mus81 and humans use MUS81 over GEN1, we find the in vitro activities of DmGen and HsGEN1 to be strikingly similar. These findings suggest that simpler branched structures may be more important substrates for Gen orthologs in vivo, and highlight the utility of using the Drosophila model system to further understand these enzymes.


Assuntos
Dano ao DNA , Reparo do DNA , DNA Cruciforme/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/enzimologia , Endonucleases/metabolismo , Resolvases de Junção Holliday/metabolismo , Animais , Sequência de Bases , Citoplasma/metabolismo , Replicação do DNA , Proteínas de Ligação a DNA/metabolismo , Embrião não Mamífero/metabolismo , Humanos , Modelos Biológicos , Mutação/genética , Multimerização Proteica , Transporte Proteico , Proteínas de Schizosaccharomyces pombe/metabolismo , Especificidade por Substrato
9.
Biochim Biophys Acta ; 1859(9): 1155-1169, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-26855179

RESUMO

Pregnane X receptor (PXR) is a major transcriptional regulator of xenobiotic metabolism and transport pathways in the liver and intestines, which are critical for protecting organisms against potentially harmful xenobiotic and endobiotic compounds. Inadvertent activation of drug metabolism pathways through PXR is known to contribute to drug resistance, adverse drug-drug interactions, and drug toxicity in humans. In both humans and rodents, PXR has been implicated in non-alcoholic fatty liver disease, diabetes, obesity, inflammatory bowel disease, and cancer. Because of PXR's important functions, it has been a therapeutic target of interest for a long time. More recent mechanistic studies have shown that PXR is modulated by multiple PTMs. Herein we provide the first investigation of the role of acetylation in modulating PXR activity. Through LC-MS/MS analysis, we identified lysine 109 (K109) in the hinge as PXR's major acetylation site. Using various biochemical and cell-based assays, we show that PXR's acetylation status and transcriptional activity are modulated by E1A binding protein (p300) and sirtuin 1 (SIRT1). Based on analysis of acetylation site mutants, we found that acetylation at K109 represses PXR transcriptional activity. The mechanism involves loss of RXRα dimerization and reduced binding to cognate DNA response elements. This mechanism may represent a promising therapeutic target using modulators of PXR acetylation levels. This article is part of a Special Issue entitled: Xenobiotic nuclear receptors: New Tricks for An Old Dog, edited by Dr. Wen Xie.


Assuntos
DNA/metabolismo , Lisina/metabolismo , Processamento de Proteína Pós-Traducional , Receptores de Esteroides/química , Sirtuína 1/metabolismo , Ativação Transcricional , Fatores de Transcrição de p300-CBP/metabolismo , Acetilação , Clonagem Molecular , DNA/química , Escherichia coli/genética , Escherichia coli/metabolismo , Genes Reporter , Células HEK293 , Células HeLa , Células Hep G2 , Humanos , Luciferases/genética , Luciferases/metabolismo , Lisina/química , Modelos Moleculares , Mutagênese Sítio-Dirigida , Receptor de Pregnano X , Multimerização Proteica , Estrutura Secundária de Proteína , Receptores de Esteroides/genética , Receptores de Esteroides/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Elementos de Resposta , Sirtuína 1/genética , Homologia Estrutural de Proteína , Relação Estrutura-Atividade , Fatores de Transcrição de p300-CBP/genética
10.
Biochemistry ; 55(21): 2979-91, 2016 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-27145151

RESUMO

SPLUNC1 is an abundantly secreted innate immune protein in the mammalian respiratory tract that exerts bacteriostatic and antibiofilm effects, binds to lipopolysaccharide (LPS), and acts as a fluid-spreading surfactant. Here, we unravel the structural elements essential for the surfactant and antimicrobial functions of human SPLUNC1 (short palate lung nasal epithelial clone 1). A unique α-helix (α4) that extends from the body of SPLUNC1 is required for the bacteriostatic, surfactant, and LPS binding activities of this protein. Indeed, we find that mutation of just four leucine residues within this helical motif to alanine is sufficient to significantly inhibit the fluid spreading abilities of SPLUNC1, as well as its bacteriostatic actions against Gram-negative pathogens Burkholderia cenocepacia and Pseudomonas aeruginosa. Conformational flexibility in the body of SPLUNC1 is also involved in the bacteriostatic, surfactant, and LPS binding functions of the protein as revealed by disulfide mutants introduced into SPLUNC1. In addition, SPLUNC1 exerts antibiofilm effects against Gram-negative bacteria, although α4 is not involved in this activity. Interestingly, though, the introduction of surface electrostatic mutations away from α4 based on the unique dolphin SPLUNC1 sequence, and confirmed by crystal structure, is shown to impart antibiofilm activity against Staphylococcus aureus, the first SPLUNC1-dependent effect against a Gram-positive bacterium reported to date. Together, these data pinpoint SPLUNC1 structural motifs required for the antimicrobial and surfactant actions of this protective human protein.


Assuntos
Anti-Infecciosos/farmacologia , Brônquios/efeitos dos fármacos , Burkholderia cenocepacia/efeitos dos fármacos , Glicoproteínas/química , Glicoproteínas/metabolismo , Lipopolissacarídeos/metabolismo , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Pseudomonas aeruginosa/efeitos dos fármacos , Sequência de Aminoácidos , Biofilmes/efeitos dos fármacos , Brônquios/citologia , Burkholderia cenocepacia/imunologia , Células Cultivadas , Cristalização , Cristalografia por Raios X , Glicoproteínas/genética , Humanos , Imunidade Inata/efeitos dos fármacos , Fosfoproteínas/genética , Conformação Proteica , Pseudomonas aeruginosa/imunologia , Surfactantes Pulmonares/química , Surfactantes Pulmonares/metabolismo
11.
Antimicrob Agents Chemother ; 60(10): 6003-12, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27458217

RESUMO

The opportunistic bacteria of the Burkholderia cepacia complex (Bcc) are extremely pathogenic to cystic fibrosis (CF) patients, and acquisition of Bcc bacteria is associated with a significant increase in mortality. Treatment of Bcc infections is difficult because the bacteria are multidrug resistant and able to survive in biofilms. Short palate, lung, and nasal epithelial clone 1 (SPLUNC1) is an innate defense protein that is secreted by the upper airways and pharynx. While SPLUNC1 is known to have antimicrobial functions, its effects on Bcc strains are unclear. We therefore tested the hypothesis that SPLUNC1 is able to impair Bcc growth and biofilm formation. We found that SPLUNC1 exerted bacteriostatic effects against several Bcc clinical isolates, including B. cenocepacia strain J2315 (50% inhibitory concentration [IC50] = 0.28 µM), and reduced biofilm formation and attachment (IC50 = 0.11 µM). We then determined which domains of SPLUNC1 are responsible for its antimicrobial activity. Deletions of SPLUNC1's N terminus and α6 helix did not affect its function. However, deletion of the α4 helix attenuated antimicrobial activity, while the corresponding α4 peptide displayed antimicrobial activity. Chronic neutrophilia is a hallmark of CF lung disease, and neutrophil elastase (NE) cleaves SPLUNC1. However, we found that the ability of SPLUNC1 to disrupt biofilm formation was significantly potentiated by NE pretreatment. While the impact of CF on SPLUNC1-Bcc interactions is not currently known, our data suggest that understanding this interaction may have important implications for CF lung disease.


Assuntos
Antibacterianos/farmacologia , Biofilmes/efeitos dos fármacos , Complexo Burkholderia cepacia/efeitos dos fármacos , Glicoproteínas/farmacologia , Elastase de Leucócito/química , Fosfoproteínas/farmacologia , Antibacterianos/química , Antibacterianos/metabolismo , Sítios de Ligação , Biofilmes/crescimento & desenvolvimento , Complexo Burkholderia cepacia/crescimento & desenvolvimento , Clonagem Molecular , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Glicoproteínas/química , Glicoproteínas/genética , Glicoproteínas/metabolismo , Humanos , Testes de Sensibilidade Microbiana , Fosfoproteínas/química , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Ligação Proteica , Conformação Proteica em alfa-Hélice , Domínios e Motivos de Interação entre Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Relação Estrutura-Atividade
12.
Proc Natl Acad Sci U S A ; 110(37): 15055-60, 2013 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-23980177

RESUMO

Members of the CsrA family of prokaryotic mRNA-binding proteins alter the translation and/or stability of transcripts needed for numerous global physiological processes. The previously described CsrA family member in Pseudomonas aeruginosa (RsmA) plays a central role in determining infection modality by reciprocally regulating processes associated with acute (type III secretion and motility) and chronic (type VI secretion and biofilm formation) infection. Here we describe a second, structurally distinct RsmA homolog in P. aeruginosa (RsmF) that has an overlapping yet unique regulatory role. RsmF deviates from the canonical 5 ß-strand and carboxyl-terminal α-helix topology of all other CsrA proteins by having the α-helix internally positioned. Despite striking changes in topology, RsmF adopts a tertiary structure similar to other CsrA family members and binds a subset of RsmA mRNA targets, suggesting that RsmF activity is mediated through a conserved mechanism of RNA recognition. Whereas deletion of rsmF alone had little effect on RsmA-regulated processes, strains lacking both rsmA and rsmF exhibited enhanced RsmA phenotypes for markers of both type III and type VI secretion systems. In addition, simultaneous deletion of rsmA and rsmF resulted in superior biofilm formation relative to the wild-type or rsmA strains. We show that RsmF translation is derepressed in an rsmA mutant and demonstrate that RsmA specifically binds to rsmF mRNA in vitro, creating a global hierarchical regulatory cascade that operates at the posttranscriptional level.


Assuntos
Proteínas de Bactérias/metabolismo , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/metabolismo , Proteínas de Ligação a RNA/metabolismo , Sequência de Aminoácidos , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Sequência de Bases , Biofilmes/crescimento & desenvolvimento , Genes Bacterianos , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Estrutura Terciária de Proteína , Processamento Pós-Transcricional do RNA , RNA Bacteriano/genética , RNA Bacteriano/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/genética , Regulon , Homologia de Sequência de Aminoácidos
13.
Proc Natl Acad Sci U S A ; 110(8): 2804-9, 2013 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-23359708

RESUMO

Multidrug-resistant Staphylococcus aureus infections pose a significant threat to human health. Antibiotic resistance is most commonly propagated by conjugative plasmids like pLW1043, the first vancomycin-resistant S. aureus vector identified in humans. We present the molecular basis for resistance transmission by the nicking enzyme in S. aureus (NES), which is essential for conjugative transfer. NES initiates and terminates the transfer of plasmids that variously confer resistance to a range of drugs, including vancomycin, gentamicin, and mupirocin. The NES N-terminal relaxase-DNA complex crystal structure reveals unique protein-DNA contacts essential in vitro and for conjugation in S. aureus. Using this structural information, we designed a DNA minor groove-targeted polyamide that inhibits NES with low micromolar efficacy. The crystal structure of the 341-residue C-terminal region outlines a unique architecture; in vitro and cell-based studies further establish that it is essential for conjugation and regulates the activity of the N-terminal relaxase. This conclusion is supported by a small-angle X-ray scattering structure of a full-length, 665-residue NES-DNA complex. Together, these data reveal the structural basis for antibiotic multiresistance acquisition by S. aureus and suggest novel strategies for therapeutic intervention.


Assuntos
Resistência Microbiana a Medicamentos , Resistência a Múltiplos Medicamentos , Staphylococcus aureus/efeitos dos fármacos , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/química , Biocatálise , DNA Bacteriano/genética , Modelos Moleculares , Nylons/farmacologia , Plasmídeos , Espalhamento a Baixo Ângulo , Staphylococcus aureus/enzimologia , Staphylococcus aureus/genética , Difração de Raios X
14.
Proc Natl Acad Sci U S A ; 110(40): 15973-8, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-24043776

RESUMO

The ability to maintain proper airway surface liquid (ASL) volume homeostasis is vital for mucus hydration and clearance, which are essential aspects of the mammalian lung's innate defense system. In cystic fibrosis (CF), one of the most common life-threatening genetic disorders, ASL dehydration leads to mucus accumulation and chronic infection. In normal airways, the secreted protein short palate lung and nasal epithelial clone 1 (SPLUNC1) effectively inhibits epithelial Na(+) channel (ENaC)-dependent Na(+) absorption and preserves ASL volume. In CF airways, it has been hypothesized that increased ENaC-dependent Na(+) absorption contributes to ASL depletion, and hence increased disease. However, this theory is controversial, and the mechanism for abnormal ENaC regulation in CF airways has remained elusive. Here, we show that SPLUNC1 is a pH-sensitive regulator of ENaC and is unable to inhibit ENaC in the acidic CF airway environment. Alkalinization of CF airway cultures prevented CF ASL hyperabsorption, and this effect was abolished when SPLUNC1 was stably knocked down. Accordingly, we resolved the crystal structure of SPLUNC1 to 2.8 Å. Notably, this structure revealed two pH-sensitive salt bridges that, when removed, rendered SPLUNC1 pH-insensitive and able to regulate ASL volume in acidic ASL. Thus, we conclude that ENaC hyperactivity is secondary to reduced CF ASL pH. Together, these data provide molecular insights into the mucosal dehydration associated with a range of pulmonary diseases, including CF, and suggest that future therapy be directed toward alkalinizing the pH of CF airways.


Assuntos
Fibrose Cística/patologia , Desidratação/metabolismo , Canais Epiteliais de Sódio/metabolismo , Glicoproteínas/química , Modelos Moleculares , Muco/química , Fosfoproteínas/química , Mucosa Respiratória/química , Adulto , Análise de Variância , Células Cultivadas , Cristalização , Fibrose Cística/complicações , Desidratação/etiologia , Desidratação/patologia , Técnicas de Silenciamento de Genes , Glicoproteínas/genética , Glicoproteínas/metabolismo , Humanos , Concentração de Íons de Hidrogênio , North Carolina , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Mucosa Respiratória/metabolismo , Mucosa Respiratória/patologia
15.
Cell Host Microbe ; 32(6): 925-944.e10, 2024 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-38754417

RESUMO

Hormones and neurotransmitters are essential to homeostasis, and their disruptions are connected to diseases ranging from cancer to anxiety. The differential reactivation of endobiotic glucuronides by gut microbial ß-glucuronidase (GUS) enzymes may influence interindividual differences in the onset and treatment of disease. Using multi-omic, in vitro, and in vivo approaches, we show that germ-free mice have reduced levels of active endobiotics and that distinct gut microbial Loop 1 and FMN GUS enzymes drive hormone and neurotransmitter reactivation. We demonstrate that a range of FDA-approved drugs prevent this reactivation by intercepting the catalytic cycle of the enzymes in a conserved fashion. Finally, we find that inhibiting GUS in conventional mice reduces free serotonin and increases its inactive glucuronide in the serum and intestines. Our results illuminate the indispensability of gut microbial enzymes in sustaining endobiotic homeostasis and indicate that therapeutic disruptions of this metabolism promote interindividual response variabilities.


Assuntos
Microbioma Gastrointestinal , Glucuronidase , Homeostase , Animais , Microbioma Gastrointestinal/efeitos dos fármacos , Camundongos , Glucuronidase/metabolismo , Camundongos Endogâmicos C57BL , Serotonina/metabolismo , Glucuronídeos/metabolismo , Humanos , Intestinos/microbiologia , Masculino , Vida Livre de Germes
16.
Am J Physiol Lung Cell Mol Physiol ; 305(12): L990-L1001, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24124190

RESUMO

The epithelial sodium channel (ENaC) is responsible for Na(+) and fluid absorption across colon, kidney, and airway epithelia. Short palate lung and nasal epithelial clone 1 (SPLUNC1) is a secreted, innate defense protein and an autocrine inhibitor of ENaC that is highly expressed in airway epithelia. While SPLUNC1 has a bactericidal permeability-increasing protein (BPI)-type structure, its NH2-terminal region lacks structure. Here we found that an 18 amino acid peptide, S18, which corresponded to residues G22-A39 of the SPLUNC1 NH2 terminus inhibited ENaC activity to a similar degree as full-length SPLUNC1 (∼2.5 fold), while SPLUNC1 protein lacking this region was without effect. S18 did not inhibit the structurally related acid-sensing ion channels, indicating specificity for ENaC. However, S18 preferentially bound to the ßENaC subunit in a glycosylation-dependent manner. ENaC hyperactivity is contributory to cystic fibrosis (CF) lung disease. Unlike control, CF human bronchial epithelial cultures (HBECs) where airway surface liquid (ASL) height was abnormally low (4.2 ± 0.6 µm), addition of S18 prevented ENaC-led ASL hyperabsorption and maintained CF ASL height at 7.9 ± 0.6 µm, even in the presence of neutrophil elastase, which is comparable to heights seen in normal HBECs. Our data also indicate that the ENaC inhibitory domain of SPLUNC1 may be cleaved away from the main molecule by neutrophil elastase, suggesting that it may still be active during inflammation or neutrophilia. Furthermore, the robust inhibition of ENaC by the S18 peptide suggests that this peptide may be suitable for treating CF lung disease.


Assuntos
Absorção/fisiologia , Fibrose Cística/metabolismo , Células Epiteliais/metabolismo , Glicoproteínas/metabolismo , Fosfoproteínas/metabolismo , Sódio/metabolismo , Células Cultivadas , Canais Epiteliais de Sódio/metabolismo , Glicoproteínas/genética , Humanos , Transporte de Íons/fisiologia , Elastase de Leucócito/metabolismo , Pulmão/metabolismo , Fosfoproteínas/genética , Mucosa Respiratória/metabolismo
17.
FASEB J ; 26(10): 4348-59, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22798424

RESUMO

The epithelial sodium channel (ENaC) is responsible for Na+ and fluid absorption across colon, kidney, and airway epithelia. We have previously identified SPLUNC1 as an autocrine inhibitor of ENaC. We have now located the ENaC inhibitory domain of SPLUNC1 to SPLUNC1's N terminus, and a peptide corresponding to this domain, G22-A39, inhibited ENaC activity to a similar degree as full-length SPLUNC1 (∼2.5 fold). However, G22-A39 had no effect on the structurally related acid-sensing ion channels, indicating specificity for ENaC. G22-A39 preferentially bound to the ß-ENaC subunit in a glycosylation-dependent manner. ENaC hyperactivity is contributory to cystic fibrosis (CF) lung disease. Addition of G22-A39 to CF human bronchial epithelial cultures (HBECs) resulted in an increase in airway surface liquid height from 4.2±0.6 to 7.9±0.6 µm, comparable to heights seen in normal HBECs, even in the presence of neutrophil elastase. Our data also indicate that the ENaC inhibitory domain of SPLUNC1 may be cleaved away from the main molecule by neutrophil elastase, which suggests that it may still be active during inflammation or neutrophilia. Furthermore, the robust inhibition of ENaC by the G22-A39 peptide suggests that this peptide may be suitable for treating CF lung disease.


Assuntos
Canais Iônicos Sensíveis a Ácido/metabolismo , Fibrose Cística/metabolismo , Canais Epiteliais de Sódio/metabolismo , Sódio/metabolismo , Absorção/efeitos dos fármacos , Animais , Western Blotting , Linhagem Celular , Dicroísmo Circular , Eletrofisiologia , Glicoproteínas/metabolismo , Humanos , Oócitos , Peptídeos/farmacologia , Fosfoproteínas/metabolismo , Estrutura Terciária de Proteína , Xenopus
18.
Cell Chem Biol ; 30(11): 1402-1413.e7, 2023 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-37633277

RESUMO

Indoxyl sulfate is a microbially derived uremic toxin that accumulates in late-stage chronic kidney disease and contributes to both renal and cardiovascular toxicity. Indoxyl sulfate is generated by the metabolism of indole, a compound created solely by gut microbial tryptophanases. Here, we characterize the landscape of tryptophanase enzymes in the human gut microbiome and find remarkable structural and functional similarities across diverse taxa. We leverage this homology through a medicinal chemistry campaign to create a potent pan-inhibitor, (3S) ALG-05, and validate its action as a transition-state analog. (3S) ALG-05 successfully reduces indole production in microbial culture and displays minimal toxicity against microbial and mammalian cells. Mice treated with (3S) ALG-05 show reduced cecal indole and serum indoxyl sulfate levels with minimal changes in other tryptophan-metabolizing pathways. These studies present a non-bactericidal pan-inhibitor of gut microbial tryptophanases with potential promise for reducing indoxyl sulfate in chronic kidney disease.


Assuntos
Microbioma Gastrointestinal , Insuficiência Renal Crônica , Humanos , Camundongos , Animais , Indicã/farmacologia , Indicã/metabolismo , Triptofanase , Microbioma Gastrointestinal/fisiologia , Indóis/farmacologia , Indóis/metabolismo , Insuficiência Renal Crônica/tratamento farmacológico , Mamíferos/metabolismo
19.
Sci Adv ; 9(18): eadg3390, 2023 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-37146137

RESUMO

Periodontitis is a chronic inflammatory disease associated with persistent oral microbial dysbiosis. The human ß-glucuronidase (GUS) degrades constituents of the periodontium and is used as a biomarker for periodontitis severity. However, the human microbiome also encodes GUS enzymes, and the role of these factors in periodontal disease is poorly understood. Here, we define the 53 unique GUSs in the human oral microbiome and examine diverse GUS orthologs from periodontitis-associated pathogens. Oral bacterial GUS enzymes are more efficient polysaccharide degraders and processers of biomarker substrates than the human enzyme, particularly at pHs associated with disease progression. Using a microbial GUS-selective inhibitor, we show that GUS activity is reduced in clinical samples obtained from individuals with untreated periodontitis and that the degree of inhibition correlates with disease severity. Together, these results establish oral GUS activity as a biomarker that captures both host and microbial contributions to periodontitis, facilitating more efficient clinical monitoring and treatment paradigms for this common inflammatory disease.


Assuntos
Microbioma Gastrointestinal , Microbiota , Doenças Periodontais , Periodontite , Humanos , Glucuronidase/metabolismo , Microbioma Gastrointestinal/fisiologia , Doenças Periodontais/etiologia , Periodontite/microbiologia , Inibidores Enzimáticos/farmacologia
20.
Nat Microbiol ; 7(11): 1817-1833, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36266335

RESUMO

Chemical signalling in the plant microbiome can have drastic effects on microbial community structure, and on host growth and development. Previously, we demonstrated that the auxin metabolic signal interference performed by the bacterial genus Variovorax via an auxin degradation locus was essential for maintaining stereotypic root development in an ecologically relevant bacterial synthetic community. Here, we dissect the Variovorax auxin degradation locus to define the genes iadDE as necessary and sufficient for indole-3-acetic acid (IAA) degradation and signal interference. We determine the crystal structures and binding properties of the operon's MarR-family repressor with IAA and other auxins. Auxin degradation operons were identified across the bacterial tree of life and we define two distinct types on the basis of gene content and metabolic products: iac-like and iad-like. The structures of MarRs from representatives of each auxin degradation operon type establish that each has distinct IAA-binding pockets. Comparison of representative IAA-degrading strains from diverse bacterial genera colonizing Arabidopsis plants show that while all degrade IAA, only strains containing iad-like auxin-degrading operons interfere with auxin signalling in a complex synthetic community context. This suggests that iad-like operon-containing bacterial strains, including Variovorax species, play a key ecological role in modulating auxins in the plant microbiome.


Assuntos
Proteínas de Arabidopsis , Arabidopsis , Microbiota , Reguladores de Crescimento de Plantas/metabolismo , Ácidos Indolacéticos/metabolismo , Arabidopsis/genética , Proteínas de Arabidopsis/genética , Plantas/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa