Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 143
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
CA Cancer J Clin ; 67(4): 326-344, 2017 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-28481406

RESUMEN

Answer questions and earn CME/CNE The human body harbors enormous numbers of microbiota that influence cancer susceptibility, in part through their prodigious metabolic capacity and their profound influence on immune cell function. Microbial pathogens drive tumorigenesis in 15% to 20% of cancer cases. Even larger numbers of malignancies are associated with an altered composition of commensal microbiota (dysbiosis) based on microbiome studies using metagenomic sequencing. Although association studies cannot distinguish whether changes in microbiota are causes or effects of cancer, a causative role is supported by rigorously controlled preclinical studies using gnotobiotic mouse models colonized with one or more specific bacteria. These studies demonstrate that microbiota can alter cancer susceptibility and progression by diverse mechanisms, such as modulating inflammation, inducing DNA damage, and producing metabolites involved in oncogenesis or tumor suppression. Evidence is emerging that microbiota can be manipulated for improving cancer treatment. By incorporating probiotics as adjuvants for checkpoint immunotherapy or by designing small molecules that target microbial enzymes, microbiota can be harnessed to improve cancer care. CA Cancer J Clin 2017;67:326-344. © 2017 American Cancer Society.


Asunto(s)
Microbiota , Neoplasias/microbiología , Neoplasias/terapia , Animales , Carcinogénesis , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Susceptibilidad a Enfermedades , Disbiosis , Humanos , Metagenómica , Medicina de Precisión
2.
Xenobiotica ; : 1-12, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38794972

RESUMEN

In vitro studies using rat, mouse, and human microsomes and hepatocytes on the bacterial ß-glucuronidase inhibitor 1-((6,8-dimethyl-2-oxo-1,2-dihydroquinolin-3-yl)methyl)-3-(4-ethoxyphenyl)-1-(2-hydroxyethyl)thiourea) (Inh 1) revealed extensive metabolism in all species.The intrinsic clearances of Inh 1 in human, mouse, and rat hepatic microsomes were 30.9, 67.8, and 201 µL/min/mg, respectively. For intact hepatocytes intrinsic clearances of 21.6, 96.0, and 129 µL/min/106 cells were seen for human, mouse and rat, respectively.The metabolism of Inh 1 involved an uncommon desulphurisation reaction in addition to oxidation, deethylation, and conjugation reactions at multiple sites. Six metabolites were detected in microsomal incubations in human and rat, and seven for the mouse. With hepatocytes, 18 metabolites were characterised, 9 for human, and 11 for mouse and rat.Following IV administration to mice (3 mg/kg), plasma concentrations of Inh 1 exhibited a monophasic decline with a terminal elimination half-life of 0.91 h and low systemic clearance (11.8% of liver blood flow). After PO dosing to mice (3 mg/kg), peak observed Inh 1 concentrations of 495 ng/mL were measured 0.5 h post dose, declining to under 10 ng/mL at 8 h post dose. The absolute oral bioavailability of Inh 1 in the mouse was ca. 26%.

3.
Drug Metab Dispos ; 51(4): 427-435, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36623880

RESUMEN

Testosterone exhibits high variability in pharmacokinetics and glucuronidation after oral administration. Although testosterone metabolism has been studied for decades, the impact of UGT2B17 gene deletion and the role of gut bacterial ß-glucuronidases on its disposition are not well characterized. We first performed an exploratory study to investigate the effect of UGT2B17 gene deletion on the global liver proteome, which revealed significant increases in proteins from multiple biological pathways. The most upregulated liver proteins were aldoketoreductases [AKR1D1, AKR1C4, AKR7A3, AKR1A1, and 7-dehydrocholesterol reductase (DHCR7)] and alcohol or aldehyde dehydrogenases (ADH6, ADH1C, ALDH1A1, ALDH9A1, and ALDH5A). In vitro assays revealed that AKR1D1 and AKR1C4 inactivate testosterone to 5ß-dihydrotestosterone (5ß-DHT) and 3α,5ß-tetrahydrotestosterone (3α,5ß-THT), respectively. These metabolites also appeared in human hepatocytes treated with testosterone and in human serum collected after oral testosterone dosing in men. Our data also suggest that 5ß-DHT and 3α, 5ß-THT are then eliminated through glucuronidation by UGT2B7 in UGT2B17 deletion individuals. Second, we evaluated the potential reactivation of testosterone glucuronide (TG) after its secretion into the intestinal lumen. Incubation of TG with purified gut microbial ß-glucuronidase enzymes and with human fecal extracts confirmed testosterone reactivation into testosterone by gut bacterial enzymes. Both testosterone metabolic switching and variable testosterone activation by gut microbial enzymes are important mechanisms for explaining the disposition of orally administered testosterone and appear essential to unraveling the molecular mechanisms underlying UGT2B17-associated pathophysiological conditions. SIGNIFICANCE STATEMENT: This study investigated the association of UGT2B17 gene deletion and gut bacterial ß-glucuronidases with testosterone disposition in vitro. The experiments revealed upregulation of AKR1D1 and AKR1C4 in UGT2B17 deletion individuals, and the role of these enzymes to inactivate testosterone to 5ß-dihydrotestosterone and 3α, 5ß-tetrahydrotestosterone, respectively. Key gut bacterial species responsible for testosterone glucuronide activation were identified. These data are important for explaining the disposition of exogenously administered testosterone and appear essential to unraveling the molecular mechanisms underlying UGT2B17-associated pathophysiological conditions.


Asunto(s)
Dihidrotestosterona , Glucuronidasa , Masculino , Humanos , Dihidrotestosterona/metabolismo , Testosterona/metabolismo , Hígado/metabolismo , Glucuronosiltransferasa/genética , Glucuronosiltransferasa/metabolismo
4.
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
5.
Immunity ; 41(2): 296-310, 2014 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-25065623

RESUMEN

Intestinal microbial metabolites are conjectured to affect mucosal integrity through an incompletely characterized mechanism. Here we showed that microbial-specific indoles regulated intestinal barrier function through the xenobiotic sensor, pregnane X receptor (PXR). Indole 3-propionic acid (IPA), in the context of indole, is a ligand for PXR in vivo, and IPA downregulated enterocyte TNF-α while it upregulated junctional protein-coding mRNAs. PXR-deficient (Nr1i2(-/-)) mice showed a distinctly "leaky" gut physiology coupled with upregulation of the Toll-like receptor (TLR) signaling pathway. These defects in the epithelial barrier were corrected in Nr1i2(-/-)Tlr4(-/-) mice. Our results demonstrate that a direct chemical communication between the intestinal symbionts and PXR regulates mucosal integrity through a pathway that involves luminal sensing and signaling by TLR4.


Asunto(s)
Intestinos/inmunología , Receptores de Esteroides/inmunología , Uniones Estrechas/inmunología , Receptor Toll-Like 4/inmunología , Uniones Adherentes/genética , Uniones Adherentes/inmunología , Animales , Antiinflamatorios no Esteroideos/farmacología , Anticuerpos/inmunología , Complejo CD3/inmunología , Células CACO-2 , Línea Celular , Femenino , Células HEK293 , Humanos , Indoles , Indometacina/farmacología , Inflamación/inmunología , Intestinos/microbiología , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C57BL , Microbiota/inmunología , Receptor X de Pregnano , Interferencia de ARN , ARN Mensajero , ARN Interferente Pequeño , Receptores de Esteroides/genética , Daño por Reperfusión/inmunología , Transducción de Señal/inmunología , Uniones Estrechas/genética , Receptor Toll-Like 4/genética , Factor de Necrosis Tumoral alfa/biosíntesis
6.
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
7.
Xenobiotica ; 52(8): 904-915, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-36149349

RESUMEN

In vitro incubation of the bacterial ß-glucuronidase inhibitor UNC10201652 (4-(8-(piperazin-1-yl)-1,2,3,4-tetrahydro-[1,2,3]triazino[4',5':4,5]thieno[2,3-c]isoquinolin-5-yl)morpholine) with mouse, rat, and human liver microsomes and hepatocytes generated metabolites at multiple sites via deethylations, oxidations and glucuronidation.Two UNC10201652 metabolites were detected in human, and four in mouse and rat liver microsomal incubations. Intrinsic clearances of UNC10201652 in human, mouse, and rat liver microsomes were 48.1, 115, and 194 µL/min/mg respectively.Intrinsic clearances for human, mouse, and rat hepatocytes were 20.9, 116, and 140 µL/min/106 cells respectively and 24 metabolites were characterised: 9 for human and 11 for both rodent species.Plasma clearance was 324.8 mL/min/kg with an elimination half-life of 0.66 h following IV administration of UNC10201652 to Swiss Albino mice (3 mg/kg). Pre-treatment with 1-aminobenzotriazole (ABT) decreased clearance to 127.43 mL/min/kg, increasing the t1/2 to 3.66 h.Comparison of profiles after oral administration of UNC10201652 to control and pre-treated mice demonstrated a large increase in Cmax (from 15.2 ng/mL to 184.0 ng/mL), a delay in Tmax from 0.25 to 1 h and increased AUC from 20.1 to 253 h ng/ml. ABT pre-treatment increased oral bioavailability from 15% to >100% suggesting that CYP450's contributed significantly to UNC10201652 clearance in mice.


Asunto(s)
Inhibidores Enzimáticos , Animales , Humanos , Ratones , Ratas , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacocinética , Hepatocitos/metabolismo , Microsomas Hepáticos/metabolismo , Morfolinas/metabolismo , Morfolinas/farmacología , Piperazinas/metabolismo , Piperazinas/farmacocinética
8.
Proc Natl Acad Sci U S A ; 116(34): 16961-16970, 2019 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-31391308

RESUMEN

Intestinal epithelial cell (IEC) shedding is a fundamental response to intestinal damage, yet underlying mechanisms and functions have been difficult to define. Here we model chronic intestinal damage in zebrafish larvae using the nonsteroidal antiinflammatory drug (NSAID) Glafenine. Glafenine induced the unfolded protein response (UPR) and inflammatory pathways in IECs, leading to delamination. Glafenine-induced inflammation was augmented by microbial colonization and associated with changes in intestinal and environmental microbiotas. IEC shedding was a UPR-dependent protective response to Glafenine that restricts inflammation and promotes animal survival. Other NSAIDs did not induce IEC delamination; however, Glafenine also displays off-target inhibition of multidrug resistance (MDR) efflux pumps. We found a subset of MDR inhibitors also induced IEC delamination, implicating MDR efflux pumps as cellular targets underlying Glafenine-induced enteropathy. These results implicate IEC delamination as a protective UPR-mediated response to chemical injury, and uncover an essential role for MDR efflux pumps in intestinal homeostasis.


Asunto(s)
Antiinflamatorios no Esteroideos , Enterocitos/metabolismo , Microbioma Gastrointestinal , Glafenina/efectos adversos , Enfermedades Intestinales , Pez Cebra , Transportadoras de Casetes de Unión a ATP/antagonistas & inhibidores , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Antiinflamatorios no Esteroideos/efectos adversos , Antiinflamatorios no Esteroideos/farmacología , Enterocitos/microbiología , Enterocitos/patología , Glafenina/farmacología , Inflamación/inducido químicamente , Inflamación/metabolismo , Inflamación/microbiología , Inflamación/patología , Enfermedades Intestinales/inducido químicamente , Enfermedades Intestinales/metabolismo , Enfermedades Intestinales/microbiología , Enfermedades Intestinales/patología , Pez Cebra/metabolismo , Pez Cebra/microbiología
9.
Drug Metab Dispos ; 49(8): 683-693, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34074730

RESUMEN

The anticancer drug irinotecan shows serious dose-limiting gastrointestinal toxicity regardless of intravenous dosing. Although enzymes and transporters involved in irinotecan disposition are known, quantitative contributions of these mechanisms in complex in vivo disposition of irinotecan are poorly understood. We explained intestinal disposition and toxicity of irinotecan by integrating 1) in vitro metabolism and transport data of irinotecan and its metabolites, 2) ex vivo gut microbial activation of the toxic metabolite SN-38, and 3) the tissue protein abundance data of enzymes and transporters relevant to irinotecan and its metabolites. Integration of in vitro kinetics data with the tissue enzyme and transporter abundance predicted that carboxylesterase (CES)-mediated hydrolysis of irinotecan is the rate-limiting process in the liver, where the toxic metabolite formed is rapidly deactivated by glucuronidation. In contrast, the poor SN-38 glucuronidation rate as compared with its efficient formation by CES2 in the enterocytes is the key mechanism of the intestinal accumulation of the toxic metabolite. The biliary efflux and organic anion transporting polypeptide-2B1-mediated enterocyte uptake can also synergize buildup of SN-38 in the enterocytes, whereas intestinal P-glycoprotein likely facilitates SN-38 detoxification in the enterocytes. The higher SN-38 concentration in the intestine can be further nourished by ß-d-glucuronidases. Understanding the quantitative significance of the key metabolism and transport processes of irinotecan and its metabolites can be leveraged to alleviate its intestinal side effects. Further, the proteomics-informed quantitative approach to determine intracellular disposition can be extended to determine susceptibility of cancer cells over normal cells for precision irinotecan therapy. SIGNIFICANCE STATEMENT: This work provides a deeper insight into the quantitative relevance of irinotecan hydrolysis (activation), conjugation (deactivation), and deconjugation (reactivation) by human or gut microbial enzymes or transporters. The results of this study explain the characteristic intestinal exposure and toxicity of irinotecan. The quantitative tissue-specific in vitro to in vivo extrapolation approach presented in this study can be extended to cancer cells.


Asunto(s)
Microbioma Gastrointestinal/efectos de los fármacos , Eliminación Hepatobiliar , Inactivación Metabólica/efectos de los fármacos , Irinotecán , Transportadores de Anión Orgánico/metabolismo , Antineoplásicos/farmacocinética , Antineoplásicos/toxicidad , Carboxilesterasa/metabolismo , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Glucuronidasa/metabolismo , Eliminación Hepatobiliar/efectos de los fármacos , Eliminación Hepatobiliar/fisiología , Humanos , Irinotecán/análogos & derivados , Irinotecán/farmacocinética , Irinotecán/toxicidad , Hígado/enzimología , Inhibidores de Topoisomerasa I/farmacocinética , Inhibidores de Topoisomerasa I/toxicidad
10.
Proc Natl Acad Sci U S A ; 115(2): E152-E161, 2018 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-29269393

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/genética , Enterobacteriaceae/genética , Microbioma Gastrointestinal/genética , Regulación Bacteriana de la Expresión Génica , Glucuronidasa/genética , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Sitios de Unión , Cristalografía por Rayos X , ADN/química , ADN/genética , ADN/metabolismo , Enterobacteriaceae/enzimología , Escherichia coli/genética , Escherichia coli/metabolismo , Genes Reguladores/genética , Ácido Glucurónico/química , Ácido Glucurónico/metabolismo , Glucuronidasa/química , Glucuronidasa/metabolismo , Humanos , Mutación , Operón/genética , Homología de Secuencia de Aminoácido
11.
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
12.
J Biol Chem ; 294(49): 18586-18599, 2019 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-31636122

RESUMEN

Gut microbial ß-glucuronidase (GUS) enzymes have been suggested to be involved in the estrobolome, the collection of microbial reactions involving estrogens. Furthermore, bacterial GUS enzymes within the gastrointestinal tract have been postulated to be a contributing factor in hormone-driven cancers. However, to date, there has been no experimental evidence to support these hypotheses. Here we provide the first in vitro analysis of the ability of 35 human gut microbial GUS enzymes to reactivate two distinct estrogen glucuronides, estrone-3-glucuronide and estradiol-17-glucuronide, to estrone and estradiol, respectively. We show that certain members within the Loop 1, mini-Loop 1, and FMN-binding classes of gut microbial GUS enzymes can reactivate estrogens from their inactive glucuronides. We provide molecular details of key interactions that facilitate these catalytic processes and present the structures of two novel human gut microbial GUS enzymes related to the estrobolome. Further, we demonstrate that estrogen reactivation by Loop 1 bacterial GUS enzymes can be inhibited both in purified enzymes and in fecal preparations of mixed murine fecal microbiota. Finally, however, despite these in vitro and ex vivo data, we show that a Loop 1 GUS-specific inhibitor is not capable of reducing the development of tumors in the PyMT mouse model of breast cancer. These findings validate that gut microbial GUS enzymes participate in the estrobolome but also suggest that the estrobolome is a multidimensional set of processes on-going within the mammalian gastrointestinal tract that likely involves many enzymes, including several distinct types of GUS proteins.


Asunto(s)
Estrógenos/metabolismo , Glucuronidasa/metabolismo , Animales , Cromatografía Líquida de Alta Presión , Estrona/metabolismo , Femenino , Microbioma Gastrointestinal/fisiología , Glucuronidasa/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Mutagénesis Sitio-Dirigida
13.
Clin Infect Dis ; 70(10): 2045-2053, 2020 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-31504285

RESUMEN

BACKGROUND: Neisseria meningitidis (Nm) is a nasopharyngeal commensal carried by healthy individuals. However, invasive infections occurs in a minority of individuals, with devastating consequences. There is evidence that common polymorphisms are associated with invasive meningococcal disease (IMD), but the contributions of rare variants other than those in the complement system have not been determined. METHODS: We identified familial cases of IMD in the UK meningococcal disease study and the European Union Life-Threatening Infectious Disease Study. Candidate genetic variants were identified by whole-exome sequencing of 2 patients with familial IMD. Candidate variants were further validated by in vitro assays. RESULTS: Exomes of 2 siblings with IMD identified a novel heterozygous missense mutation in BPIFA1/SPLUNC1. Sequencing of 186 other nonfamilial cases identified another unrelated IMD patient with the same mutation. SPLUNC1 is an innate immune defense protein expressed in the nasopharyngeal epithelia; however, its role in invasive infections is unknown. In vitro assays demonstrated that recombinant SPLUNC1 protein inhibits biofilm formation by Nm, and impedes Nm adhesion and invasion of human airway cells. The dominant negative mutant recombinant SPLUNC1 (p.G22E) showed reduced antibiofilm activity, increased meningococcal adhesion, and increased invasion of cells, compared with wild-type SPLUNC1. CONCLUSIONS: A mutation in SPLUNC1 affecting mucosal attachment, biofilm formation, and invasion of mucosal epithelial cells is a new genetic cause of meningococcal disease.


Asunto(s)
Glicoproteínas/genética , Infecciones Meningocócicas/genética , Infecciones Meningocócicas/microbiología , Neisseria meningitidis , Fosfoproteínas/genética , Proteínas del Sistema Complemento , Células Epiteliales , Humanos , Mutación , Neisseria meningitidis/genética
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.
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
16.
Infect Immun ; 87(7)2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31036600

RESUMEN

Enterococcus faecalis strains are resident intestinal bacteria associated with invasive infections, inflammatory bowel diseases, and colon cancer. Although factors promoting E. faecalis colonization of intestines are not fully known, one implicated pathway is a phosphotransferase system (PTS) in E. faecalis strain OG1RF that phosphorylates gluconate and contains the genes OG1RF_12399 to OG1RF_12402 (OG1RF_12399-12402). We hypothesize that this PTS permits growth in gluconate, facilitates E. faecalis intestinal colonization, and exacerbates colitis. We generated E. faecalis strains containing deletions/point mutations in this PTS and measured bacterial growth and PTS gene expression in minimal medium supplemented with selected carbohydrates. We show that E. faecalis upregulates OG1RF_12399 transcription specifically in the presence of gluconate and that E. faecalis strains lacking, or harboring a single point mutation in, OG1RF_12399-12402 are unable to grow in minimal medium containing gluconate. We colonized germfree wild-type and colitis-prone interleukin-10-deficient mice with defined bacterial consortia containing the E. faecalis strains and measured inflammation and bacterial abundance in the colon. We infected macrophage and intestinal epithelial cell lines with the E. faecalis strains and measured intracellular bacterial survival and proinflammatory cytokine secretion. The presence of OG1RF_12399-12402 is not required for E. faecalis colonization of the mouse intestine but is associated with an accelerated onset of experimental colitis in interleukin-10-deficient mice, altered bacterial composition in the colon, enhanced E. faecalis survival within macrophages, and increased proinflammatory cytokine secretion by colon tissue and macrophages. Further studies of bacterial carbohydrate metabolism in general, and E. faecalis PTS-gluconate in particular, during inflammation may identify new mechanisms of disease pathogenesis.


Asunto(s)
Proteínas Bacterianas/metabolismo , Colitis/microbiología , Enterococcus faecalis/enzimología , Macrófagos/inmunología , Fosfotransferasas/metabolismo , Animales , Proteínas Bacterianas/genética , Colitis/genética , Colitis/inmunología , Enterococcus faecalis/genética , Enterococcus faecalis/crecimiento & desarrollo , Femenino , Gluconatos/metabolismo , Humanos , Interleucina-10/genética , Interleucina-10/inmunología , Intestinos/inmunología , Intestinos/microbiología , Macrófagos/microbiología , Masculino , Ratones , Operón , Fosfotransferasas/genética
17.
FASEB J ; 32(5): 2478-2491, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29295861

RESUMEN

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.


Asunto(s)
Membrana Celular/metabolismo , Células Epiteliales/metabolismo , Canales Epiteliales de Sodio/metabolismo , Glicoproteínas/metabolismo , Complejos Multiproteicos/química , Mucosa Nasal/metabolismo , Fosfoproteínas/metabolismo , Regulación Alostérica/fisiología , Membrana Celular/química , Membrana Celular/genética , Células Epiteliales/química , Canales Epiteliales de Sodio/química , Canales Epiteliales de Sodio/genética , Transferencia Resonante de Energía de Fluorescencia , Glicoproteínas/química , Glicoproteínas/genética , Células HEK293 , Humanos , Proteínas Luminiscentes , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Mucosa Nasal/química , Fosfoproteínas/química , Fosfoproteínas/genética , Proteína Fluorescente Roja
18.
Nucleic Acids Res ; 45(9): 5333-5348, 2017 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-28369583

RESUMEN

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.


Asunto(s)
Daño del ADN , Reparación del ADN , ADN Cruciforme/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/enzimología , Endonucleasas/metabolismo , Resolvasas de Unión Holliday/metabolismo , Animales , Secuencia de Bases , Citoplasma/metabolismo , Replicación del ADN , Proteínas de Unión al ADN/metabolismo , Embrión no Mamífero/metabolismo , Humanos , Modelos Biológicos , Mutación/genética , Multimerización de Proteína , Transporte de Proteínas , Proteínas de Schizosaccharomyces pombe/metabolismo , Especificidad por Sustrato
19.
J Biol Chem ; 292(21): 8569-8576, 2017 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-28389557

RESUMEN

The intestinal milieu is astonishingly complex and home to a constantly changing mixture of small and large molecules, along with an abundance of bacteria, viral particles, and eukaryotic cells. Such complexity makes it difficult to develop testable molecular hypotheses regarding host-microbe interactions. Fortunately, mammals and their associated gastrointestinal (GI) microbes contain complementary systems that are ideally suited for mechanistic studies. Mammalian systems inactivate endobiotic and xenobiotic compounds by linking them to a glucuronic acid sugar for GI excretion. In the GI tract, the microbiota express ß-glucuronidase enzymes that remove the glucuronic acid as a carbon source, effectively reversing the actions of mammalian inactivation. Thus, by probing the actions of microbial ß-glucuronidases, and by understanding which substrate glucuronides they process, molecular insights into mammalian-microbial symbioses may be revealed amid the complexity of the intestinal tract. Here, we focus on glucuronides in the gut and the microbial proteins that process them.


Asunto(s)
Microbioma Gastrointestinal/fisiología , Glucurónidos/metabolismo , Intestinos/microbiología , Simbiosis/fisiología , Animales , Proteínas Bacterianas/metabolismo , Glucuronidasa/metabolismo , Humanos , Xenobióticos/metabolismo
20.
Biochim Biophys Acta ; 1859(9): 1155-1169, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-26855179

RESUMEN

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.


Asunto(s)
ADN/metabolismo , Lisina/metabolismo , Procesamiento Proteico-Postraduccional , Receptores de Esteroides/química , Sirtuina 1/metabolismo , Activación Transcripcional , Factores de Transcripción p300-CBP/metabolismo , Acetilación , Clonación Molecular , ADN/química , Escherichia coli/genética , Escherichia coli/metabolismo , Genes Reporteros , Células HEK293 , Células HeLa , Células Hep G2 , Humanos , Luciferasas/genética , Luciferasas/metabolismo , Lisina/química , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Receptor X de Pregnano , Multimerización de Proteína , Estructura Secundaria de Proteína , Receptores de Esteroides/genética , Receptores de Esteroides/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Elementos de Respuesta , Sirtuina 1/genética , Homología Estructural de Proteína , Relación Estructura-Actividad , Factores de Transcripción p300-CBP/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA