Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros











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.
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
3.
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
4.
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
5.
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
6.
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.

7.
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
8.
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
9.
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.

10.
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
11.
Nat Commun ; 8: 14118, 2017 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-28165446

RESUMEN

Asthma is a chronic airway disease characterized by inflammation, mucus hypersecretion and abnormal airway smooth muscle (ASM) contraction. Bacterial permeability family member A1, BPIFA1, is a secreted innate defence protein. Here we show that BPIFA1 levels are reduced in sputum samples from asthmatic patients and that BPIFA1 is secreted basolaterally from healthy, but not asthmatic human bronchial epithelial cultures (HBECs), where it suppresses ASM contractility by binding to and inhibiting the Ca2+ influx channel Orai1. We have localized this effect to a specific, C-terminal α-helical region of BPIFA1. Furthermore, tracheas from Bpifa1-/- mice are hypercontractile, and this phenotype is reversed by the addition of recombinant BPIFA1. Our data suggest that BPIFA1 deficiency in asthmatic airways promotes Orai1 hyperactivity, increased ASM contraction and airway hyperresponsiveness. Strategies that target Orai1 or the BPIFA1 deficiency in asthma may lead to novel therapies to treat this disease.


Asunto(s)
Asma/fisiopatología , Glicoproteínas/fisiología , Contracción Muscular/fisiología , Músculo Liso/fisiopatología , Proteína ORAI1/metabolismo , Fosfoproteínas/fisiología , Adulto , Anciano , Animales , Bronquios/citología , Células Epiteliales/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Glicoproteínas/química , Células HEK293 , Humanos , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Simulación del Acoplamiento Molecular , Proteína ORAI1/química , Proteína ORAI1/genética , Fosfoproteínas/química , ARN Interferente Pequeño/metabolismo , Proteínas Recombinantes/metabolismo , Mucosa Respiratoria/citología , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/fisiopatología , Esputo/fisiología , Adulto Joven
12.
Antimicrob Agents Chemother ; 60(10): 6003-12, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27458217

RESUMEN

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.


Asunto(s)
Antibacterianos/farmacología , Biopelículas/efectos de los fármacos , Complejo Burkholderia cepacia/efectos de los fármacos , Glicoproteínas/farmacología , Elastasa de Leucocito/química , Fosfoproteínas/farmacología , Antibacterianos/química , Antibacterianos/metabolismo , Sitios de Unión , Biopelículas/crecimiento & desarrollo , Complejo Burkholderia cepacia/crecimiento & desarrollo , Clonación Molecular , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Glicoproteínas/química , Glicoproteínas/genética , Glicoproteínas/metabolismo , Humanos , Pruebas de Sensibilidad Microbiana , Fosfoproteínas/química , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Unión Proteica , Conformación Proteica en Hélice alfa , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Relación Estructura-Actividad
13.
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
14.
Am J Physiol Lung Cell Mol Physiol ; 305(12): L990-L1001, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24124190

RESUMEN

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.


Asunto(s)
Absorción/fisiología , Fibrosis Quística/metabolismo , Células Epiteliales/metabolismo , Glicoproteínas/metabolismo , Fosfoproteínas/metabolismo , Sodio/metabolismo , Células Cultivadas , Canales Epiteliales de Sodio/metabolismo , Glicoproteínas/genética , Humanos , Transporte Iónico/fisiología , Elastasa de Leucocito/metabolismo , Pulmón/metabolismo , Fosfoproteínas/genética , Mucosa Respiratoria/metabolismo
15.
Proc Natl Acad Sci U S A ; 110(40): 15973-8, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-24043776

RESUMEN

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.


Asunto(s)
Fibrosis Quística/patología , Deshidratación/metabolismo , Canales Epiteliales de Sodio/metabolismo , Glicoproteínas/química , Modelos Moleculares , Moco/química , Fosfoproteínas/química , Mucosa Respiratoria/química , Adulto , Análisis de Varianza , Células Cultivadas , Cristalización , Fibrosis Quística/complicaciones , Deshidratación/etiología , Deshidratación/patología , Técnicas de Silenciamiento del Gen , Glicoproteínas/genética , Glicoproteínas/metabolismo , Humanos , Concentración de Iones de Hidrógeno , North Carolina , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología
16.
FASEB J ; 26(10): 4348-59, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22798424

RESUMEN

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.


Asunto(s)
Canales Iónicos Sensibles al Ácido/metabolismo , Fibrosis Quística/metabolismo , Canales Epiteliales de Sodio/metabolismo , Sodio/metabolismo , Absorción/efectos de los fármacos , Animales , Western Blotting , Línea Celular , Dicroismo Circular , Electrofisiología , Glicoproteínas/metabolismo , Humanos , Oocitos , Péptidos/farmacología , Fosfoproteínas/metabolismo , Estructura Terciaria de Proteína , Xenopus
17.
Stem Cells ; 25(2): 279-88, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17008427

RESUMEN

Hematopoietic stem cells (HSCs) have enormous potential for use in transplantation and gene therapy. However, the frequency of repopulating HSCs is often very low; thus, highly effective techniques for cell enrichment and maintenance are required to obtain sufficient cell numbers for therapeutic use and for studies of HSC physiology. Common methods of HSC enrichment use antibodies recognizing HSC surface marker antigens. Because antibodies are known to alter the physiology of other cell types, we investigated the effect of such enrichment strategies on the physiology and lineage commitment of HSCs. We sorted HSCs using a method that does not require antibodies: exclusion of Hoechst 33342 to isolate side population (SP) cells. To elucidate the effect of antibody binding on this HSC population, we compared untreated SP cells with SP cells treated with the Sca-1(+)c-Kit(+)Lin(-) (SKL) antibody cocktail prior to SP sorting. Our findings revealed that HSCs incubated with the antibody cocktail had decreased expression of the stem cell-associated genes c-Kit, Cd34, Tal-1, and Slamf1 relative to untreated SP cells or to cells treated with polyclonal isotype control antibodies. Moreover, SKL antibodies induced cycling in SP cells and diminished their ability to confer long-term hematopoietic engraftment in lethally irradiated mice. Taken together, these data suggest that antibody-based stem cell isolation procedures can have negative effects on HSC physiology.


Asunto(s)
Anticuerpos/inmunología , Anticuerpos/farmacología , Antígenos de Diferenciación/inmunología , Trasplante de Células Madre Hematopoyéticas , Células Madre/efectos de los fármacos , Células Madre/inmunología , Animales , Antígenos de Diferenciación/genética , Ataxina-1 , Ataxinas , Bencimidazoles , Ciclo Celular/efectos de los fármacos , Ciclo Celular/efectos de la radiación , Separación Celular , Femenino , Citometría de Flujo , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/efectos de la radiación , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/inmunología , Proteínas Nucleares/inmunología , Proteínas Proto-Oncogénicas c-kit/inmunología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Células Madre/efectos de la radiación , Irradiación Corporal Total
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA