Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
bioRxiv ; 2024 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-38645173

RESUMEN

Alcohol use disorders (AUDs) impose an enormous societal and financial burden, and world-wide, alcohol misuse is the 7th leading cause of premature death1. Despite this, there are currently only 3 FDA approved pharmacological treatments for the treatment of AUDs in the United States. The neurotensin (Nts) system has long been implicated in modulating behaviors associated with alcohol misuse. Recently, a novel compound, SBI-553, that biases the action of Nts receptor 1 (NTSR1) activation, has shown promise in preclinical models of psychostimulant misuse. Here we investigate the efficacy of this compound to alter ethanol-mediated behaviors in a comprehensive battery of experiments assessing ethanol consumption, behavioral responses to ethanol, sensitivity to ethanol, and ethanol metabolism. Additionally, we investigated behavior in avoidance and cognitive assays to monitor potential side effects of SBI-553. We find that SBI-553 reduces binge-like ethanol consumption in mice without altering avoidance behavior or novel object recognition. We also observe sex-dependent differences in physiological responses to sequential ethanol injections in mice. In rats, we show that SBI-553 attenuates sensitivity to the interoceptive effects of ethanol (using a Pavlovian drug discrimination task). Our data suggest that targeting NTSR1 signaling may be promising to attenuate alcohol misuse, and adds to a body of literature that suggests NTSR1 may be a common downstream target involved in the psychoactive effects of multiple reinforcing substances.

2.
Glia ; 71(2): 245-258, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36106533

RESUMEN

Fractalkine (FKN) is a membrane-bound chemokine that can be cleaved by proteases such as ADAM 10, ADAM 17, and cathepsin S to generate soluble fragments. Studies using different forms of the soluble FKN yield conflicting results in vivo. These observations prompted us to investigate the function and pharmacology of two commonly used isoforms of FKN, a human full-length soluble FKN (sFKN), and a human chemokine domain only FKN (cdFKN). Both are prevalent in the literature and are often assumed to be functionally equivalent. We observed that recombinant sFKN and cdFKN exhibit similar potencies in a cell-based cAMP assay, but binding affinity for CX3CR1 was modestly different. There was a 10-fold difference in potency between sFKN and cdFKN when assessing their ability to stimulate ß-arrestin recruitment. Interestingly, high concentrations of FKN, regardless of cleavage variant, were ineffective at reducing pro-inflammatory microglial activation and may induce a pro-inflammatory response. This effect was observed in mouse and rat primary microglial cells as well as microglial cell lines. The inflammatory response was exacerbated in aged microglia, which is known to exhibit age-related inflammatory phenotypes. We observed the same effects in Cx3cr1-/- primary microglia and therefore speculate that an alternative FKN receptor may exist. Collectively, these data provide greater insights into the function and pharmacology of these common FKN reagents, which may clarify conflicting reports and urge greater caution in the selection of FKN peptides for use in in vitro and in vivo studies and the interpretation of results obtained using these differing peptides.


Asunto(s)
Quimiocina CX3CL1 , Microglía , Ratones , Ratas , Humanos , Animales , Anciano , Quimiocina CX3CL1/metabolismo , Microglía/metabolismo , Proteolisis , Receptor 1 de Quimiocinas CX3C/genética , Receptor 1 de Quimiocinas CX3C/metabolismo , Línea Celular
3.
Antimicrob Agents Chemother ; 66(4): e0210921, 2022 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-35266827

RESUMEN

In Plasmodium, the first two and rate-limiting enzymes of the pentose phosphate pathway, glucose 6-phosphate dehydrogenase (G6PD) and the 6-phosphogluconolactonase, are bifunctionally fused to a unique enzyme named GluPho, differing structurally and mechanistically from the respective human orthologs. Consistent with the enzyme's essentiality for malaria parasite proliferation and propagation, human G6PD deficiency has immense impact on protection against severe malaria, making PfGluPho an attractive antimalarial drug target. Herein we report on the optimized lead compound N-(((2R,4S)-1-cyclobutyl-4-hydroxypyrrolidin-2-yl)methyl)-6-fluoro-4-methyl-11-oxo-10,11-dihydrodibenzo[b,f][1,4]thiazepine-8-carboxamide (SBI-0797750), a potent and fully selective PfGluPho inhibitor with robust nanomolar activity against recombinant PfGluPho, PvG6PD, and P. falciparum blood-stage parasites. Mode-of-action studies have confirmed that SBI-0797750 disturbs the cytosolic glutathione-dependent redox potential, as well as the cytosolic and mitochondrial H2O2 homeostasis of P. falciparum blood stages, at low nanomolar concentrations. Moreover, SBI-0797750 does not harm red blood cell (RBC) integrity and phagocytosis and thus does not promote anemia. SBI-0797750 is therefore a very promising antimalarial lead compound.


Asunto(s)
Antimaláricos , Deficiencia de Glucosafosfato Deshidrogenasa , Malaria Falciparum , Malaria Vivax , Malaria , Antimaláricos/farmacología , Antimaláricos/uso terapéutico , Hidrolasas de Éster Carboxílico , Glucosa/metabolismo , Glucosafosfato Deshidrogenasa/metabolismo , Humanos , Peróxido de Hidrógeno/metabolismo , Malaria Falciparum/tratamiento farmacológico , Malaria Vivax/tratamiento farmacológico , Fosfatos , Plasmodium falciparum/metabolismo , Plasmodium vivax
4.
Cardiovasc Res ; 118(7): 1771-1784, 2022 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-34132787

RESUMEN

AIMS: Hydrogen sulfide (H2S) is a potent signalling molecule that activates diverse cardioprotective pathways by post-translational modification (persulfidation) of cysteine residues in upstream protein targets. Heart failure patients with reduced ejection fraction (HFrEF) exhibit low levels of H2S. Sulfide:quinone oxidoreductase (SQOR) catalyses the first irreversible step in the metabolism of H2S and plays a key role in regulating H2S-mediated signalling. Here, the aim of this study was to discover a first-in-class inhibitor of human SQOR and evaluate its cardioprotective effect in an animal model of HFrEF. METHODS AND RESULTS: We identified a potent inhibitor of human SQOR (STI1, IC50 = 29 nM) by high-throughput screening of a small-molecule library, followed by focused medicinal chemistry optimization and structure-based design. STI1 is a competitive inhibitor that binds with high selectivity to the coenzyme Q-binding pocket in SQOR. STI1 exhibited very low cytotoxicity and attenuated the hypertrophic response of neonatal rat ventricular cardiomyocytes and H9c2 cells induced by neurohormonal stressors. A mouse HFrEF model was produced by transverse aortic constriction (TAC). Treatment of TAC mice with STI1 mitigated the development of cardiomegaly, pulmonary congestion, dilatation of the left ventricle, and cardiac fibrosis and decreased the pressure gradient across the aortic constriction. Moreover, STI1 dramatically improved survival, preserved cardiac function, and prevented the progression to HFrEF by impeding the transition from compensated to decompensated left ventricle hypertrophy. CONCLUSION: We demonstrate that the coenzyme Q-binding pocket in human SQOR is a druggable target and establish proof of concept for the potential of SQOR inhibitors to provide a novel therapeutic approach for the treatment of HFrEF.


Asunto(s)
Insuficiencia Cardíaca , Animales , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/prevención & control , Humanos , Ratones , Ratas , Volumen Sistólico , Sulfuros/farmacología , Ubiquinona/uso terapéutico , Remodelación Ventricular
5.
Bioorg Med Chem Lett ; 54: 128443, 2021 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-34763081

RESUMEN

Here we report the first small-molecule inhibitors of human sulfide:quinone oxidoreductase (SQOR) that decrease the rate of breakdown of hydrogen sulfide (H2S), a potent cardioprotective signaling molecule. SQOR is a mitochondrial membrane-bound protein that catalyzes a two-electron oxidation of H2S to sulfane sulfur (S0), using glutathione (or sulfite) and coenzyme Q (CoQ) as S0 and electron acceptor, respectively. Inhibition of SQOR may constitute a new approach for the treatment of heart failure with reduced ejection fraction. Starting from top hits identified in a high-throughput screen, we conducted SAR development guided by docking of lead candidates into our crystal structure of SQOR. We identified potent SQOR inhibitors such as 19 which has an IC50 of 29 nM for SQOR inhibition and favorable pharmacokinetic and ADME properties required for in vivo efficacy testing.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Hidrocarburos Aromáticos/farmacología , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Humanos , Hidrocarburos Aromáticos/síntesis química , Hidrocarburos Aromáticos/química , Estructura Molecular , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/metabolismo , Bibliotecas de Moléculas Pequeñas/síntesis química , Bibliotecas de Moléculas Pequeñas/química , Relación Estructura-Actividad
6.
J Med Chem ; 64(9): 5645-5653, 2021 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-33914534

RESUMEN

Obesity-associated insulin resistance plays a central role in the pathogenesis of type 2 diabetes. A promising approach to decrease insulin resistance in obesity is to inhibit the protein tyrosine phosphatases that negatively regulate insulin receptor signaling. The low-molecular-weight protein tyrosine phosphatase (LMPTP) acts as a critical promoter of insulin resistance in obesity by inhibiting phosphorylation of the liver insulin receptor activation motif. Here, we report development of a novel purine-based chemical series of LMPTP inhibitors. These compounds inhibit LMPTP with an uncompetitive mechanism and are highly selective for LMPTP over other protein tyrosine phosphatases. We also report the generation of a highly orally bioavailable purine-based analogue that reverses obesity-induced diabetes in mice.


Asunto(s)
Inhibidores Enzimáticos/química , Proteínas Tirosina Fosfatasas/antagonistas & inhibidores , Purinas/química , Administración Oral , Animales , Sitios de Unión , Cristalografía por Rayos X , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/etiología , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Semivida , Humanos , Resistencia a la Insulina , Cinética , Simulación de Dinámica Molecular , Obesidad/complicaciones , Obesidad/patología , Fosforilación/efectos de los fármacos , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Purinas/metabolismo , Purinas/farmacología , Purinas/uso terapéutico , Transducción de Señal/efectos de los fármacos , Relación Estructura-Actividad
7.
Bioorg Med Chem Lett ; 41: 128007, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33798699

RESUMEN

NAD+ is a crucial cellular factor that plays multifaceted roles in wide ranging biological processes. Low levels of NAD+ have been linked to numerous diseases including metabolic disorders, cardiovascular disease, neurodegeneration, and muscle wasting disorders. A novel strategy to boost NAD+ is to activate nicotinamide phosphoribosyltransferase (NAMPT), the putative rate-limiting step in the NAD+ salvage pathway. We previously showed that NAMPT activators increase NAD+ levels in vitro and in vivo. Herein we describe the optimization of our NAMPT activator prototype (SBI-0797812) leading to the identification of 1-(4-((4-chlorophenyl)sulfonyl)phenyl)-3-(oxazol-5-ylmethyl)urea (34) that showed far more potent NAMPT activation and improved oral bioavailability.


Asunto(s)
Citocinas/metabolismo , Nicotinamida Fosforribosiltransferasa/metabolismo , Urea/farmacología , Relación Dosis-Respuesta a Droga , Humanos , Estructura Molecular , Relación Estructura-Actividad , Urea/análogos & derivados , Urea/química
8.
Am Psychol ; 76(7): 1196-1197, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34990176

RESUMEN

A survey by Thornewill et al. (2020) produced potentially useful information about how national security roles of psychologists are viewed both within and beyond the discipline. However, a closer examination of the data, and of the authors' interpretations of that data, raises some important questions. (PsycInfo Database Record (c) 2022 APA, all rights reserved).


Asunto(s)
Medidas de Seguridad
9.
Br J Cancer ; 124(6): 1098-1109, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33318657

RESUMEN

BACKGROUND: The BCL2 inhibitor venetoclax has shown efficacy in several hematologic malignancies, with the greatest response rates in indolent blood cancers such as chronic lymphocytic leukaemia. There is a lower response rate to venetoclax monotherapy in diffuse large B-cell lymphoma (DLBCL). METHODS: We tested inhibitors of cap-dependent mRNA translation for the ability to sensitise DLBCL and mantle cell lymphoma (MCL) cells to apoptosis by venetoclax. We compared the mTOR kinase inhibitor (TOR-KI) MLN0128 with SBI-756, a compound targeting eukaryotic translation initiation factor 4G1 (eIF4G1), a scaffolding protein in the eIF4F complex. RESULTS: Treatment of DLBCL and MCL cells with SBI-756 synergised with venetoclax to induce apoptosis in vitro, and enhanced venetoclax efficacy in vivo. SBI-756 prevented eIF4E-eIF4G1 association and cap-dependent translation without affecting mTOR substrate phosphorylation. In TOR-KI-resistant DLBCL cells lacking eIF4E binding protein-1, SBI-756 still sensitised to venetoclax. SBI-756 selectively reduced translation of mRNAs encoding ribosomal proteins and translation factors, leading to a reduction in protein synthesis rates in sensitive cells. When normal lymphocytes were treated with SBI-756, only B cells had reduced viability, and this correlated with reduced protein synthesis. CONCLUSIONS: Our data highlight a novel combination for treatment of aggressive lymphomas, and establishes its efficacy and selectivity using preclinical models.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Resistencia a Antineoplásicos/efectos de los fármacos , Factor 4E Eucariótico de Iniciación/antagonistas & inhibidores , Linfoma de Células B/tratamiento farmacológico , Terapia Molecular Dirigida , Animales , Apoptosis , Compuestos Bicíclicos Heterocíclicos con Puentes/administración & dosificación , Proliferación Celular , Femenino , Humanos , Lactamas/administración & dosificación , Linfoma de Células B/metabolismo , Linfoma de Células B/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Quinolonas/administración & dosificación , Sulfonamidas/administración & dosificación , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
10.
J Med Chem ; 62(17): 8357-8363, 2019 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-31390201
11.
Structure ; 27(5): 794-805.e4, 2019 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-30905673

RESUMEN

Hydrogen sulfide (H2S) is a gasotransmitter exhibiting pivotal functions in diverse biological processes, including activation of multiple cardioprotective pathways. Sulfide:quinone oxidoreductase (SQOR) is an integral membrane flavoprotein that catalyzes the first step in the mitochondrial metabolism of H2S. As such, it plays a critical role in controlling physiological levels of the gasotransmitter and has attracted keen interest as a potential drug target. We report the crystal structure of human SQOR, unraveling the molecular basis for the enzyme's ability to catalyze sulfane sulfur transfer reactions with structurally diverse acceptors. We demonstrate that human SQOR contains unique features: an electropositive surface depression implicated as a binding site for sulfane sulfur acceptors and postulated to funnel negatively charged substrates to a hydrophilic H2S-oxidizing active site, which is connected to a hydrophobic internal tunnel that binds coenzyme Q. These findings support a proposed model for catalysis and open the door for structure-based drug design.


Asunto(s)
Sulfuro de Hidrógeno/química , Mitocondrias/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/química , Oxígeno/química , Sitios de Unión , Dominio Catalítico , Cristalografía por Rayos X , Cisteína/análogos & derivados , Cisteína/química , Humanos , Enlace de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Oxidación-Reducción , Unión Proteica , Dominios Proteicos , Estructura Cuaternaria de Proteína
12.
Cancer Lett ; 449: 145-162, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30771432

RESUMEN

Inhibition of ubiquitin ligases with small molecule remains a very challenging task, given the lack of catalytic activity of the target and the requirement of disruption of its interactions with other proteins. Siah1/2, which are E3 ubiquitin ligases, are implicated in melanoma and prostate cancer and represent high-value drug targets. We utilized three independent screening approaches in our efforts to identify small-molecule Siah1/2 inhibitors: Affinity Selection-Mass Spectrometry, a protein thermal shift-based assay and an in silico based screen. Inhibitors were assessed for their effect on viability of melanoma and prostate cancer cultures, colony formation, prolyl-hydroxylase-HIF1α signaling, expression of selected Siah2-related transcripts, and Siah2 ubiquitin ligase activity. Several analogs were further characterized, demonstrating improved efficacy. Combination of the top hits identified in the different assays demonstrated an additive effect, pointing to complementing mechanisms that underlie each of these Siah1/2 inhibitors.


Asunto(s)
Melanoma/tratamiento farmacológico , Proteínas Nucleares/antagonistas & inhibidores , Neoplasias de la Próstata/tratamiento farmacológico , Bibliotecas de Moléculas Pequeñas/administración & dosificación , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Simulación por Computador , Regulación hacia Abajo , Ensayos de Selección de Medicamentos Antitumorales , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Espectrometría de Masas , Melanoma/genética , Ratones , Proteínas Nucleares/genética , Neoplasias de la Próstata/genética , Bibliotecas de Moléculas Pequeñas/aislamiento & purificación , Bibliotecas de Moléculas Pequeñas/farmacología , Ubiquitina-Proteína Ligasas/genética , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Bioorg Med Chem Lett ; 28(1): 31-34, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29174347

RESUMEN

Tissue-nonspecific alkaline phosphatase (TNAP) is an ectoenzyme crucial for bone matrix mineralization via its ability to hydrolyze extracellular inorganic pyrophosphate (ePPi), a potent mineralization inhibitor, to phosphate (Pi). By the controlled hydrolysis of ePPi, TNAP maintains the correct ratio of Pi to ePPi and therefore enables normal skeletal and dental calcification. In other areas of the body low ePPi levels lead to the development of pathological soft-tissue calcification, which can progress to a number of disorders. TNAP inhibitors have been shown to prevent these processes via an increase of ePPi. Herein we describe the use of a whole blood assay to optimize a previously described series of TNAP inhibitors resulting in 5-((5-chloro-2-methoxyphenyl)sulfonamido)nicotinamide (SBI-425), a potent, selective and oral bioavailable compound that robustly inhibits TNAP in vivo.


Asunto(s)
Fosfatasa Alcalina/antagonistas & inhibidores , Inhibidores Enzimáticos/química , Niacinamida/análogos & derivados , Niacinamida/química , Sulfonamidas/química , Administración Oral , Fosfatasa Alcalina/metabolismo , Animales , Evaluación Preclínica de Medicamentos , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacocinética , Semivida , Concentración 50 Inhibidora , Ratones , Niacinamida/metabolismo , Niacinamida/farmacocinética , Relación Estructura-Actividad , Sulfonamidas/metabolismo , Sulfonamidas/farmacocinética
14.
Am Psychol ; 72(4): 395-396, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28481588

RESUMEN

Drawing on Kuhn's (1970) analysis, Melchert (2016) argued that current professional psychology exists in a preparadigmatic state and that a transition to a unified clinical science based on the paradigm of the behavioral and neurosciences is now possible. But Melchert's analysis makes questionable assumptions about reducibility and neglects several crucial aspects of Kuhn's analysis. A close examination of psychological work on problems such as violence against women indicates that different research paradigms and their associated exemplars identify strengths and weaknesses of specific treatment resources that cannot be entirely encompassed within a single paradigmatic perspective (Jackson, 2015b) and additionally suggests that psychological knowledge is governed by at least 3 overarching research paradigms, as well as a variety of subparadigms encompassing applied and mixed methods research and many current orientations to professional psychology (Jackson, 2015a). (PsycINFO Database Record


Asunto(s)
Neurociencias , Psicología , Investigación , Humanos , Conocimiento , Trastornos Mentales , Ciencia
15.
Biochemistry ; 56(7): 986-996, 2017 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-28107627

RESUMEN

Hydrogen sulfide (H2S) is an endogenously synthesized signaling molecule that is enzymatically metabolized in mitochondria. The metabolism of H2S maintains optimal concentrations of the gasotransmitter and produces sulfane sulfur (S0)-containing metabolites that may be functionally important in signaling. Sulfide:quinone oxidoreductase (SQOR) catalyzes the initial two-electron oxidation of H2S to S0 using coenzyme Q as the electron acceptor in a reaction that requires a third substrate to act as the acceptor of S0. We discovered that sulfite is a highly efficient acceptor and proposed that sulfite is the physiological acceptor in a reaction that produces thiosulfate, a known metabolic intermediate. This model has been challenged by others who assume that the intracellular concentration of sulfite is very low, a scenario postulated to favor reaction of SQOR with a considerably poorer acceptor, glutathione. In this study, we measured the intracellular concentration of sulfite and other metabolites in mammalian tissues. The values observed for sulfite in rat liver (9.2 µM) and heart (38 µM) are orders of magnitude higher than previously assumed. We discovered that the apparent kinetics of oxidation of H2S by SQOR with glutathione as the S0 acceptor reflect contributions from other SQOR-catalyzed reactions, including a novel glutathione:CoQ reductase reaction. We used observed metabolite levels and steady-state kinetic parameters to simulate rates of oxidation of H2S by SQOR at physiological concentrations of different S0 acceptors. The results show that the reaction with sulfite as the S0 acceptor is a major pathway in liver and heart and provide insight into the potential dynamics of H2S metabolism.


Asunto(s)
Sulfuro de Hidrógeno/metabolismo , Hígado/metabolismo , Miocardio/metabolismo , Quinona Reductasas/metabolismo , Anaerobiosis , Animales , Cisteína/metabolismo , Glutatión/metabolismo , Cinética , Masculino , Redes y Vías Metabólicas , Quinona Reductasas/química , Ratas Wistar , Sulfitos/metabolismo , Ubiquinona/metabolismo
16.
J Biomol Screen ; 20(10): 1232-45, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26265713

RESUMEN

Endoplasmic reticulum (ER) stress activates three distinct signal transducers on the ER membrane. Inositol-requiring protein 1 (IRE1), the most conserved signal transducer, plays a key role in ER stress-mediated signaling. During ER stress, IRE1 initiates two discrete signaling cascades: the "adaptive" signaling cascade mediated by the XBP1 pathway and the "alarm" signaling cascade mediated by stress-activated protein kinase pathways. Fine-tuning of the balance between the adaptive and alarm signals contributes significantly to cellular fate under ER stress. Thus, we propose that the design of high-throughput screening (HTS) assays to selectively monitor IRE1 mediated-signaling would be desirable for drug discovery. To this end, we report the generation of stable human neural cell lines and development of cell-based HTS luciferase (Luc) reporter gene assays for the identification of pathway-specific chemical modulators of IRE1. We implemented a cell-based Luc assay using a chimeric CHOP-Gal4 transcription factor in 384-well format for monitoring IRE1 kinase-mediated p38MAPK activation and an unfolded response pathway element (URPE)-Luc cell-based assay in 1536-well format for monitoring IRE1's RNase-mediated activation of XBP1. Chemical library screening was successfully conducted with both the CHOP/Gal4-Luc cells and UPRE-Luc engineered cells. The studies demonstrate the feasibility of using these HTS assays for discovery of pathway-selective modulators of IRE1.


Asunto(s)
Endorribonucleasas/antagonistas & inhibidores , Ensayos Analíticos de Alto Rendimiento , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas , Línea Celular Tumoral , Proteínas de Unión al ADN/metabolismo , Estrés del Retículo Endoplásmico , Endorribonucleasas/fisiología , Activación Enzimática , Genes Reporteros , Células HeLa , Humanos , Luciferasas/análisis , Luciferasas/genética , Sistema de Señalización de MAP Quinasas , Neuronas , Proteínas Serina-Treonina Quinasas/fisiología , Factores de Transcripción del Factor Regulador X , Tapsigargina/metabolismo , Factores de Transcripción/metabolismo , Proteína 1 de Unión a la X-Box
17.
Cell ; 161(6): 1252-65, 2015 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-26046436

RESUMEN

Small-molecule probes can illuminate biological processes and aid in the assessment of emerging therapeutic targets by perturbing biological systems in a manner distinct from other experimental approaches. Despite the tremendous promise of chemical tools for investigating biology and disease, small-molecule probes were unavailable for most targets and pathways as recently as a decade ago. In 2005, the NIH launched the decade-long Molecular Libraries Program with the intent of innovating in and broadening access to small-molecule science. This Perspective describes how novel small-molecule probes identified through the program are enabling the exploration of biological pathways and therapeutic hypotheses not otherwise testable. These experiences illustrate how small-molecule probes can help bridge the chasm between biological research and the development of medicines but also highlight the need to innovate the science of therapeutic discovery.


Asunto(s)
Descubrimiento de Drogas , Bibliotecas de Moléculas Pequeñas , Animales , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Ensayos Analíticos de Alto Rendimiento , Humanos , National Institutes of Health (U.S.) , Estados Unidos
18.
Methods Enzymol ; 554: 255-70, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25725526

RESUMEN

The first step in the mammalian metabolism of H2S is catalyzed by sulfide:quinone oxidoreductase (SQOR). Human SQOR is an integral membrane protein, which presumably interacts with the inner mitochondrial membrane in a monotopic fashion. The enzyme is a member of a family of flavoprotein disulfide oxidoreductases (e.g., glutathione reductase) that utilize a Cys-S-S-Cys disulfide bridge as an additional redox center. SQOR catalyzes a two-electron oxidation of H2S to sulfane sulfur using coenzyme Q as electron acceptor. The enzyme also requires a third substrate to act as the acceptor of the sulfane sulfur from a cysteine persulfide intermediate. Here, we describe a method for the bacterial expression of human SQOR as a catalytically active membrane-bound protein, procedures for solubilization and purification of the recombinant protein to >95% homogeneity, and spectrophotometric assays to monitor SQOR-mediated H2S oxidation in reactions with different sulfane sulfur acceptors.


Asunto(s)
Sulfuro de Hidrógeno/metabolismo , Quinona Reductasas/fisiología , Biocatálisis , Pruebas de Enzimas , Escherichia coli , Humanos , Sulfuro de Hidrógeno/química , Cinética , Oxidación-Reducción , Quinona Reductasas/química , Quinona Reductasas/aislamiento & purificación
19.
J Bone Miner Res ; 30(5): 824-36, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25428889

RESUMEN

Medial vascular calcification (MVC) is a pathological phenomenon that causes vascular stiffening and can lead to heart failure; it is common to a variety of conditions, including aging, chronic kidney disease, diabetes, obesity, and a variety of rare genetic diseases. These conditions share the common feature of tissue-nonspecific alkaline phosphatase (TNAP) upregulation in the vasculature. To evaluate the role of TNAP in MVC, we developed a mouse model that overexpresses human TNAP in vascular smooth muscle cells in an X-linked manner. Hemizygous overexpressor male mice (Tagln-Cre(+/-) ; Hprt(ALPL) (/Y) or TNAP-OE) show extensive vascular calcification, high blood pressure, and cardiac hypertrophy, and have a median age of death of 44 days, whereas the cardiovascular phenotype is much less pronounced and life expectancy is longer in heterozygous (Tagln-Cre(+/-) ; Hprt(ALPL) (/-) ) female TNAP-OE mice. Gene expression analysis showed upregulation of osteoblast and chondrocyte markers and decreased expression of vascular smooth muscle markers in the aortas of TNAP-OE mice. Through medicinal chemistry efforts, we developed inhibitors of TNAP with drug-like pharmacokinetic characteristics. TNAP-OE mice were treated with the prototypical TNAP inhibitor SBI-425 or vehicle to evaluate the feasibility of TNAP inhibition in vivo. Treatment with this inhibitor significantly reduced aortic calcification and cardiac hypertrophy, and extended lifespan over vehicle-treated controls, in the absence of secondary effects on the skeleton. This study shows that TNAP in the vasculature contributes to the pathology of MVC and that it is a druggable target.


Asunto(s)
Fosfatasa Alcalina/metabolismo , Músculo Liso Vascular/enzimología , Músculo Liso Vascular/fisiopatología , Calcificación Vascular/enzimología , Calcificación Vascular/fisiopatología , Fosfatasa Alcalina/antagonistas & inhibidores , Animales , Animales Recién Nacidos , Aorta/enzimología , Aorta/patología , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacocinética , Inhibidores Enzimáticos/farmacología , Masculino , Ratones Transgénicos , Músculo Liso Vascular/efectos de los fármacos , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Resultado del Tratamiento , Ultrasonografía , Calcificación Vascular/sangre , Calcificación Vascular/diagnóstico por imagen
20.
Biochemistry ; 53(28): 4739-53, 2014 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-24981631

RESUMEN

Human sulfide:quinone oxidoreductase (SQOR) catalyzes the conversion of H2S to thiosulfate, the first step in mammalian H2S metabolism. SQOR's inability to produce the glutathione persulfide (GSS(-)) substrate for sulfur dioxygenase (SDO) suggested that a thiosulfate:glutathione sulfurtransferase (TST) was required to provide the missing link between the SQOR and SDO reactions. Although TST could be purified from yeast, attempts to isolate the mammalian enzyme were not successful. We used bioinformatic approaches to identify genes likely to encode human TST (TSTD1) and its yeast ortholog (RDL1). Recombinant TSTD1 and RDL1 catalyze a predicted thiosulfate-dependent conversion of glutathione to GSS(-). Both enzymes contain a rhodanese homology domain and a single catalytically essential cysteine, which is converted to cysteine persulfide upon reaction with thiosulfate. GSS(-) is a potent inhibitor of TSTD1 and RDL1, as judged by initial rate accelerations and ≥25-fold lower Km values for glutathione observed in the presence of SDO. The combined action of GSS(-) and SDO is likely to regulate the biosynthesis of the reactive metabolite. SDO drives to completion p-toluenethiosulfonate:glutathione sulfurtransferase reactions catalyzed by TSTD1 and RDL1. The thermodynamic coupling of the irreversible SDO and reversible TST reactions provides a model for the physiologically relevant reaction with thiosulfate as the sulfane donor. The discovery of bacterial Rosetta Stone proteins that comprise fusions of SDO and TSTD1 provides phylogenetic evidence of the association of these enzymes. The presence of adjacent bacterial genes encoding SDO-TSTD1 fusion proteins and human-like SQORs suggests these prokaryotes and mammals exhibit strikingly similar pathways for H2S metabolism.


Asunto(s)
Sulfuro de Hidrógeno/química , Proteínas de Neoplasias/química , Proteínas de Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/enzimología , Humanos , Sulfuro de Hidrógeno/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Estructura Terciaria de Proteína , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Homología Estructural de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA