Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Host Microbe ; 32(6): 925-944.e10, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38754417

RESUMEN

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.


Asunto(s)
Microbioma Gastrointestinal , Glucuronidasa , Homeostasis , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Ratones , Glucuronidasa/metabolismo , Ratones Endogámicos C57BL , Serotonina/metabolismo , Glucurónidos/metabolismo , Humanos , Intestinos/microbiología , Masculino , Vida Libre de Gérmenes
2.
Plasmid ; 128: 102708, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37967733

RESUMEN

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.


Asunto(s)
Replicación del ADN , Staphylococcus aureus , Plásmidos/genética , Staphylococcus aureus/genética , Secuencia de Bases , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , ADN , ARN Mensajero
3.
Cell Chem Biol ; 30(11): 1402-1413.e7, 2023 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-37633277

RESUMEN

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.


Asunto(s)
Microbioma Gastrointestinal , Insuficiencia Renal Crónica , Humanos , Ratones , Animales , Indicán/farmacología , Indicán/metabolismo , Triptofanasa , Microbioma Gastrointestinal/fisiología , Indoles/farmacología , Indoles/metabolismo , Insuficiencia Renal Crónica/tratamiento farmacológico , Mamíferos/metabolismo
4.
Sci Adv ; 9(18): eadg3390, 2023 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-37146137

RESUMEN

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.


Asunto(s)
Microbioma Gastrointestinal , Microbiota , Enfermedades Periodontales , Periodontitis , Humanos , Glucuronidasa/metabolismo , Microbioma Gastrointestinal/fisiología , Enfermedades Periodontales/etiología , Periodontitis/microbiología , Inhibidores Enzimáticos/farmacología
5.
Nat Microbiol ; 7(11): 1817-1833, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36266335

RESUMEN

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.


Asunto(s)
Proteínas de Arabidopsis , Arabidopsis , Microbiota , Reguladores del Crecimiento de las Plantas/metabolismo , Ácidos Indolacéticos/metabolismo , Arabidopsis/genética , Proteínas de Arabidopsis/genética , Plantas/metabolismo
6.
Science ; 373(6553): 420-425, 2021 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-34140391

RESUMEN

Plant nucleotide-binding leucine-rich repeat receptors (NLRs) regulate immunity and cell death. In Arabidopsis, a subfamily of "helper" NLRs is required by many "sensor" NLRs. Active NRG1.1 oligomerized, was enriched in plasma membrane puncta, and conferred cytoplasmic calcium ion (Ca2+) influx in plant and human cells. NRG1.1-dependent Ca2+ influx and cell death were sensitive to Ca2+ channel blockers and were suppressed by mutations affecting oligomerization or plasma membrane enrichment. Ca2+ influx and cell death mediated by NRG1.1 and ACTIVATED DISEASE RESISTANCE 1 (ADR1), another helper NLR, required conserved negatively charged N-terminal residues. Whole-cell voltage-clamp recordings demonstrated that Arabidopsis helper NLRs form Ca2+-permeable cation channels to directly regulate cytoplasmic Ca2+ levels and consequent cell death. Thus, helper NLRs transduce cell death signals directly.


Asunto(s)
Proteínas de Arabidopsis/química , Canales de Calcio/química , Calcio/metabolismo , Péptidos y Proteínas de Señalización Intracelular/química , Proteínas NLR/química , Arabidopsis , Proteínas de Arabidopsis/metabolismo , Canales de Calcio/metabolismo , Señalización del Calcio , Muerte Celular , Membrana Celular/metabolismo , Células HEK293 , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas NLR/metabolismo , Técnicas de Placa-Clamp , Dominios Proteicos , Estructura Secundaria de Proteína
7.
Biochemistry ; 59(40): 3939-3950, 2020 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-32993284

RESUMEN

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.


Asunto(s)
Bacterias/enzimología , Microbioma Gastrointestinal , Microbiota , Sulfatasas/metabolismo , Bacterias/química , Bacterias/genética , Bacterias/metabolismo , Dominio Catalítico , Heces/microbiología , Genes Bacterianos , Humanos , Modelos Moleculares , Conformación Proteica , Sulfatasas/química , Sulfatasas/genética
8.
Proc Natl Acad Sci U S A ; 117(13): 7374-7381, 2020 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-32170007

RESUMEN

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.


Asunto(s)
Microbioma Gastrointestinal/efectos de los fármacos , Glucuronidasa/antagonistas & inhibidores , Glucuronidasa/efectos de los fármacos , Animales , Antineoplásicos Fitogénicos/farmacología , Bacterias/efectos de los fármacos , Modelos Animales de Enfermedad , Disbiosis/tratamiento farmacológico , Inhibidores Enzimáticos/farmacología , Femenino , Glucuronidasa/metabolismo , Humanos , Irinotecán/farmacología , Ratones , Ratones Desnudos , Neoplasias/tratamiento farmacológico
9.
EMBO Mol Med ; 12(4): e11621, 2020 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-32153125

RESUMEN

The human PXR (pregnane X receptor), a master regulator of drug metabolism, has essential roles in intestinal homeostasis and abrogating inflammation. Existing PXR ligands have substantial off-target toxicity. Based on prior work that established microbial (indole) metabolites as PXR ligands, we proposed microbial metabolite mimicry as a novel strategy for drug discovery that allows exploiting previously unexplored parts of chemical space. Here, we report functionalized indole derivatives as first-in-class non-cytotoxic PXR agonists as a proof of concept for microbial metabolite mimicry. The lead compound, FKK6 (Felix Kopp Kortagere 6), binds directly to PXR protein in solution, induces PXR-specific target gene expression in cells, human organoids, and mice. FKK6 significantly represses pro-inflammatory cytokine production cells and abrogates inflammation in mice expressing the human PXR gene. The development of FKK6 demonstrates for the first time that microbial metabolite mimicry is a viable strategy for drug discovery and opens the door to underexploited regions of chemical space.


Asunto(s)
Imitación Molecular , Receptor X de Pregnano/química , Animales , Células Cultivadas , Citocinas , Humanos , Inflamación , Intestinos , Ligandos , Ratones , Organoides
10.
J Med Chem ; 63(7): 3701-3712, 2020 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-32160459

RESUMEN

Pregnane X receptor (PXR) is a master xenobiotic-sensing transcription factor and a validated target for immune and inflammatory diseases. The identification of chemical probes to investigate the therapeutic relevance of the receptor is still highly desired. In fact, currently available PXR ligands are not highly selective and can exhibit toxicity and/or potential off-target effects. In this study, we have identified garcinoic acid as a selective and efficient PXR agonist. The properties of this natural molecule as a specific PXR agonist were demonstrated by the screening on a panel of nuclear receptors, the assessment of the physical and thermodynamic binding affinity, and the determination of the PXR-garcinoic acid complex crystal structure. Cytotoxicity, transcriptional, and functional properties were investigated in human liver cells, and compound activity and target engagement were confirmed in vivo in mouse liver and gut tissue. In conclusion, garcinoic acid is a selective natural agonist of PXR and a promising lead compound toward the development of new PXR-regulating modulators.


Asunto(s)
Benzopiranos/farmacología , Receptor X de Pregnano/agonistas , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Animales , Benzopiranos/metabolismo , Benzopiranos/toxicidad , Línea Celular Tumoral , Cristalografía por Rayos X , Citocromo P-450 CYP3A/metabolismo , Expresión Génica/efectos de los fármacos , Humanos , Hígado/metabolismo , Masculino , Ratones Endogámicos C57BL , Receptor X de Pregnano/metabolismo
11.
ACS Chem Biol ; 15(1): 217-225, 2020 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-31774274

RESUMEN

It is increasingly clear that interindividual variability in human gut microbial composition contributes to differential drug responses. For example, gastrointestinal (GI) toxicity is not observed in all patients treated with the anticancer drug irinotecan, and it has been suggested that this variability is a result of differences in the types and levels of gut bacterial ß-glucuronidases (GUSs). GUS enzymes promote drug toxicity by hydrolyzing the inactive drug-glucuronide conjugate back to the active drug, which damages the GI epithelium. Proteomics-based identification of the exact GUS enzymes responsible for drug reactivation from the complexity of the human microbiota has not been accomplished, however. Here, we discover the specific bacterial GUS enzymes that generate SN-38, the active and toxic metabolite of irinotecan, from human fecal samples using a unique activity-based protein profiling (ABPP) platform. We identify and quantify gut bacterial GUS enzymes from human feces with an ABPP-enabled proteomics pipeline and then integrate this information with ex vivo kinetics to pinpoint the specific GUS enzymes responsible for SN-38 reactivation. Furthermore, the same approach also reveals the molecular basis for differential gut bacterial GUS inhibition observed between human fecal samples. Taken together, this work provides an unprecedented technical and bioinformatics pipeline to discover the microbial enzymes responsible for specific reactions from the complexity of human feces. Identifying such microbial enzymes may lead to precision biomarkers and novel drug targets to advance the promise of personalized medicine.


Asunto(s)
Proteínas Bacterianas/metabolismo , Ciclohexanoles/química , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/metabolismo , Inhibidores Enzimáticos/química , Microbioma Gastrointestinal/fisiología , Glucuronidasa/metabolismo , Irinotecán/química , Animales , Biomarcadores/metabolismo , Biología Computacional , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/microbiología , Inhibidores Enzimáticos/metabolismo , Heces/química , Femenino , Glucurónidos/metabolismo , Humanos , Hidrólisis , Irinotecán/metabolismo , Cinética , Masculino , Metaboloma , Ratones , Modelos Moleculares , Medicina de Precisión , Unión Proteica , Conformación Proteica
12.
ACS Chem Biol ; 14(12): 2737-2744, 2019 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-31663730

RESUMEN

Regorafenib (Stivarga) is an oral small molecule kinase inhibitor used to treat metastatic colorectal cancer, hepatocellular carcinomas, and gastrointestinal stromal tumors. Diarrhea is one of the most frequently observed adverse reactions associated with regorafenib. This toxicity may arise from the reactivation of the inactive regorafenib-glucuronide to regorafenib by gut microbial ß-glucuronidase (GUS) enzymes in the gastrointestinal tract. We sought to unravel the molecular basis of regorafenib-glucuronide processing by human intestinal GUS enzymes and to examine the potential inhibition of these enzymes. Using a panel of 31 unique gut microbial GUS enzymes derived from the 279 mapped from the human gut microbiome, we found that only four were capable of regorafenib-glucuronide processing. Using crystal structures as a guide, we pinpointed the molecular features unique to these enzymes that confer regorafenib-glucuronide processing activity. Furthermore, a pilot screen identified the FDA-approved drug raloxifene as an inhibitor of regorafenib reactivation by the GUS proteins discovered. Novel synthetic raloxifene analogs exhibited improved potency in both in vitro and ex vivo studies. Taken together, these data establish that regorafenib reactivation is exclusively catalyzed by gut microbial enzymes and that these enzymes are amenable to targeted inhibition. Our results unravel key molecular details of regorafenib reactivation in the GI tract and provide a potential pathway to improve clinical outcomes with regorafenib.


Asunto(s)
Inhibidores Enzimáticos/toxicidad , Microbioma Gastrointestinal , Glucuronidasa/antagonistas & inhibidores , Intestinos/enzimología , Compuestos de Fenilurea/toxicidad , Piridinas/toxicidad , Animales , Glucurónidos/química , Ratones , Compuestos de Fenilurea/química , Piridinas/química
13.
mSystems ; 4(4)2019 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-31455640

RESUMEN

Gut microbial ß-glucuronidase (GUS) enzymes play important roles in drug efficacy and toxicity, intestinal carcinogenesis, and mammalian-microbial symbiosis. Recently, the first catalog of human gut GUS proteins was provided for the Human Microbiome Project stool sample database and revealed 279 unique GUS enzymes organized into six categories based on active-site structural features. Because mice represent a model biomedical research organism, here we provide an analogous catalog of mouse intestinal microbial GUS proteins-a mouse gut GUSome. Using metagenome analysis guided by protein structure, we examined 2.5 million unique proteins from a comprehensive mouse gut metagenome created from several mouse strains, providers, housing conditions, and diets. We identified 444 unique GUS proteins and organized them into six categories based on active-site features, similarly to the human GUSome analysis. GUS enzymes were encoded by the major gut microbial phyla, including Firmicutes (60%) and Bacteroidetes (21%), and there were nearly 20% for which taxonomy could not be assigned. No differences in gut microbial gus gene composition were observed for mice based on sex. However, mice exhibited gus differences based on active-site features associated with provider, location, strain, and diet. Furthermore, diet yielded the largest differences in gus composition. Biochemical analysis of two low-fat-associated GUS enzymes revealed that they are variable with respect to their efficacy of processing both sulfated and nonsulfated heparan nonasaccharides containing terminal glucuronides.IMPORTANCE Mice are commonly employed as model organisms of mammalian disease; as such, our understanding of the compositions of their gut microbiomes is critical to appreciating how the mouse and human gastrointestinal tracts mirror one another. GUS enzymes, with importance in normal physiology and disease, are an attractive set of proteins to use for such analyses. Here we show that while the specific GUS enzymes differ at the sequence level, a core GUSome functionality appears conserved between mouse and human gastrointestinal bacteria. Mouse strain, provider, housing location, and diet exhibit distinct GUSomes and gus gene compositions, but sex seems not to affect the GUSome. These data provide a basis for understanding the gut microbial GUS enzymes present in commonly used laboratory mice. Further, they demonstrate the utility of metagenome analysis guided by protein structure to provide specific sets of functionally related proteins from whole-genome metagenome sequencing data.

14.
Biochemistry ; 58(9): 1311-1317, 2019 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-30729778

RESUMEN

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.


Asunto(s)
Microbioma Gastrointestinal/fisiología , Glucuronidasa/química , Glicósido Hidrolasas/química , Dominio Catalítico , Cristalografía por Rayos X , Heces/microbiología , Glucuronidasa/genética , Glucuronidasa/metabolismo , Glicósido Hidrolasas/genética , Glicósido Hidrolasas/metabolismo , Bacilos Gramnegativos Anaerobios Facultativos/genética , Humanos , Mutagénesis Sitio-Dirigida , Conformación Proteica
15.
Sci Rep ; 9(1): 825, 2019 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-30696850

RESUMEN

Bacterial ß-glucuronidase (GUS) enzymes cause drug toxicity by reversing Phase II glucuronidation in the gastrointestinal tract. While many human gut microbial GUS enzymes have been examined with model glucuronide substrates like p-nitrophenol-ß-D-glucuronide (pNPG), the GUS orthologs that are most efficient at processing drug-glucuronides remain unclear. Here we present the crystal structures of GUS enzymes from human gut commensals Lactobacillus rhamnosus, Ruminococcus gnavus, and Faecalibacterium prausnitzii that possess an active site loop (Loop 1; L1) analogous to that found in E. coli GUS, which processes drug substrates. We also resolve the structure of the No Loop GUS from Bacteroides dorei. We then compare the pNPG and diclofenac glucuronide processing abilities of a panel of twelve structurally diverse GUS proteins, and find that the new L1 GUS enzymes presented here process small glucuronide substrates inefficiently compared to previously characterized L1 GUS enzymes like E. coli GUS. We further demonstrate that our GUS inhibitors, which are effective against some L1 enzymes, are not potent towards all. Our findings pinpoint active site structural features necessary for the processing of drug-glucuronide substrates and the inhibition of such processing.


Asunto(s)
Microbioma Gastrointestinal/fisiología , Tracto Gastrointestinal/microbiología , Glucuronidasa/antagonistas & inhibidores , Glucuronidasa/metabolismo , Glucurónidos/metabolismo , Bacteroides/enzimología , Dominio Catalítico , Clostridiales/enzimología , Cristalografía por Rayos X , Inhibidores Enzimáticos/farmacología , Faecalibacterium prausnitzii/enzimología , Tracto Gastrointestinal/metabolismo , Humanos , Lacticaseibacillus rhamnosus/enzimología , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Relación Estructura-Actividad
16.
J Mol Biol ; 431(5): 970-980, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30658055

RESUMEN

The human gut microbiota encodes ß-glucuronidases (GUSs) that play key roles in health and disease via the metabolism of glucuronate-containing carbohydrates and drugs. Hundreds of putative bacterial GUS enzymes have been identified by metagenomic analysis of the human gut microbiome, but less than 10% have characterized structures and functions. Here we describe a set of unique gut microbial GUS enzymes that bind flavin mononucleotide (FMN). First, we show using mass spectrometry, isothermal titration calorimetry, and x-ray crystallography that a purified GUS from the gut commensal microbe Faecalibacterium prausnitzii binds to FMN on a surface groove located 30 Šaway from the active site. Second, utilizing structural and functional data from this FMN-binding GUS, we analyzed the 279 unique GUS sequences from the Human Microbiome Project database and identified 14 putative FMN-binding GUSs. We characterized four of these hits and solved the structure of two, the GUSs from Ruminococcus gnavus and Roseburia hominis, which confirmed that these are FMN binders. Third, binding and kinetic analysis of the FMN-binding site mutants of these five GUSs show that they utilize a conserved site to bind FMN that is not essential for GUS activity, but can affect KM. Lastly, a comprehensive structural review of the PDB reveals that the FMN-binding site employed by these enzymes is unlike any structurally characterized FMN binders to date. These findings reveal the first instance of an FMN-binding glycoside hydrolase and suggest a potential link between FMN and carbohydrate metabolism in the human gut microbiota.


Asunto(s)
Mononucleótido de Flavina/metabolismo , Microbioma Gastrointestinal/fisiología , Glucuronidasa/metabolismo , Dominio Catalítico/fisiología , Clostridiales/metabolismo , Humanos , Cinética , Metagenoma/fisiología , Microbiota/fisiología , Ruminococcus/metabolismo
17.
J Biol Chem ; 293(48): 18559-18573, 2018 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-30301767

RESUMEN

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.


Asunto(s)
Bacteroides/enzimología , Microbioma Gastrointestinal , Glucuronidasa/química , Glucuronidasa/metabolismo , Isoenzimas/química , Isoenzimas/metabolismo , Secuencia de Aminoácidos , Dominio Catalítico , Inhibidores Enzimáticos/farmacología , Ácido Glucárico/análogos & derivados , Glucuronidasa/antagonistas & inhibidores , Humanos , Conformación Proteica
18.
Protein Sci ; 27(12): 2010-2022, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30230652

RESUMEN

ß-Glucuronidase (GUS) enzymes in the gastrointestinal tract are involved in maintaining mammalian-microbial symbiosis and can play key roles in drug efficacy and toxicity. Parabacteroides merdae GUS was identified as an abundant mini-Loop 2 (mL2) type GUS enzyme in the Human Microbiome Project gut metagenomic database. Here, we report the crystal structure of P. merdae GUS and highlight the differences between this enzyme and extant structures of gut microbial GUS proteins. We find that P. merdae GUS exhibits a distinct tetrameric quaternary structure and that the mL2 motif traces a unique path within the active site, which also includes two arginines distinctive to this GUS. We observe two states of the P. merdae GUS active site; a loop repositions itself by more than 50 Å to place a functionally-relevant residue into the enzyme's catalytic site. Finally, we find that P. merdae GUS is able to bind to homo and heteropolymers of the polysaccharide alginic acid. Together, these data broaden our understanding of the structural and functional diversity in the GUS family of enzymes present in the human gut microbiome and point to specialization as an important feature of microbial GUS orthologs.


Asunto(s)
Bacteroidaceae/enzimología , Microbioma Gastrointestinal , Glucuronidasa/metabolismo , Dominio Catalítico , Cristalografía por Rayos X , Humanos , Modelos Moleculares , Conformación Proteica
19.
PLoS One ; 13(9): e0203621, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30216370

RESUMEN

SPLUNC1 is a multifunctional protein of the airway with antimicrobial properties. We previously reported that it displayed antibiofilm activities against P. aeruginosa. The goal of this study was to determine whether (1) the antibiofilm property is broad (including S. aureus, another prevalent organism in cystic fibrosis); (2) the α4 region is responsible for such activity; and (3), if so, this motif could be structurally optimized as an antimicrobial peptide with enhanced activities. We used S. aureus biofilm-prevention assays to determine bacterial biomass in the presence of SPLUNC1 and SPLUNC1Δα4 recombinant proteins, or SPLUNC1-derived peptides (α4 and α4M1), using the well-established crystal-violet biofilm detection assay. The SPLUNC1Δα4 showed markedly reduced biofilm prevention compared to the parent protein. Surprisingly, the 30-residue long α4 motif alone demonstrated minimal biofilm prevention activities. However, structural optimization of the α4 motif resulted in a modified peptide (α4M1) with significantly enhanced antibiofilm properties against methicillin-sensitive (MSSA) and-resistant (MRSA) S. aureus, including six different clinical strains of MRSA and the well-known USA300. Hemolytic activity was undetectable at up to 100µM for the peptides. The data warrant further investigation of α4-derived AMPs to explore the potential application of antimicrobial peptides to combat bacterial biofilm-related infections.


Asunto(s)
Antiinfecciosos/química , Antiinfecciosos/farmacología , Biopelículas/efectos de los fármacos , Glicoproteínas/química , Péptidos/química , Péptidos/farmacología , Fosfoproteínas/química , Staphylococcus aureus/efectos de los fármacos , Humanos , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Pruebas de Sensibilidad Microbiana
20.
ACS Cent Sci ; 4(7): 868-879, 2018 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-30062115

RESUMEN

Microbial ß-glucuronidases (GUSs) cause severe gut toxicities that limit the efficacy of cancer drugs and other therapeutics. Selective inhibitors of bacterial GUS have been shown to alleviate these side effects. Using structural and chemical biology, mass spectrometry, and cell-based assays, we establish that piperazine-containing GUS inhibitors intercept the glycosyl-enzyme catalytic intermediate of these retaining glycosyl hydrolases. We demonstrate that piperazine-based compounds are substrate-dependent GUS inhibitors that bind to the GUS-GlcA catalytic intermediate as a piperazine-linked glucuronide (GlcA, glucuronic acid). We confirm the GUS-dependent formation of inhibitor-glucuronide conjugates by LC-MS and show that methylated piperazine analogs display significantly reduced potencies. We further demonstrate that a range of approved piperazine- and piperidine-containing drugs from many classes, including those for the treatment of depression, infection, and cancer, function by the same mechanism, and we confirm through gene editing that these compounds selectively inhibit GUS in living bacterial cells. Together, these data reveal a unique mechanism of GUS inhibition and show that a range of therapeutics may impact GUS activities in the human gut.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...