Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
RSC Med Chem ; 14(11): 2342-2347, 2023 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-37974969

RESUMO

A loss of prosecretory Cl- channel CFTR activity in the intestine is considered as the key cause of gastrointestinal problems in cystic fibrosis (CF): meconium ileus, distal intestinal obstruction syndrome (DIOS) and constipation. Since CFTR modulators have minimal effects on gastrointestinal symptoms, there is an unmet need for novel treatments for CF-associated gastrointestinal disorders. Meconium ileus and DIOS mainly affect the ileum (distal small intestine). SLC26A6 (putative anion transporter 1, PAT1) is a Cl-/HCO3- exchanger at the luminal membrane of small intestinal epithelial cells which facilitates Cl- and fluid absorption. We recently identified first-in-class PAT1 inhibitors by high-throughput screening. Isoxazolopyrimidine PAT1inh-A01 was a hit compound, which had low potency (IC50 5.2 µM) for SLC26A6 inhibition precluding further preclinical development. Here we performed structure-activity relationship studies to optimize isoxazolopyrimidine SLC26A6 inhibitors and tested a potent inhibitor in mouse models of intestinal fluid absorption. Structure-activity studies of 377 isoxazolopyrimidine analogs identified PAT1inh-A0030 (ethyl 4-(benzyl(methyl)amino)-3-methylisoxazolo[5,4-d]pyrimidine-6-carboxylate) as the most potent SLC26A6 inhibitor with a 1.0 µM IC50. Selectivity studies showed that PAT1inh-A030 has no activity on relevant ion transporters/channels (SLC26A3, SLC26A4, SLC26A9, CFTR, TMEM16A). In a closed-loop model of intestinal fluid absorption, intraluminal PAT1inh-A0030 treatment inhibited fluid absorption in the ileum of wild-type and CF mice (CftrdelF508/delF508) with >90% prevention of a decrease in loop fluid volume and loop weight/length ratio at 30 minutes. These results suggest that SLC26A6 is the key transporter mediating Cl- and fluid absorption in the ileum and SLC26A6 inhibitors are novel drug candidates for treatment of CF-associated small intestinal disorders.

2.
Eur J Med Chem ; 249: 115149, 2023 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-36724632

RESUMO

The anion exchanger protein SLC26A3 (down-regulated in adenoma, DRA) is expressed in the luminal membrane of intestinal epithelial cells in colon, where it facilitates the absorption of Cl- and oxalate. We previously identified a 4,8-dimethylcoumarin class of SLC26A3 inhibitors that act from the SLC26A3 cytoplasmic surface, and demonstrated their efficacy in mouse models of constipation and hyperoxaluria. Here, screening of 50,000 new compounds and 1740 chemical analogs of active compounds from the primary screen produced five novel classes of SLC26A3-selective inhibitors (1,3-dioxoisoindoline-amides; N-(5-sulfamoyl-1,3,4-thiadiazol-2-yl)acetamides; thiazolo-pyrimidin-5-ones; 3-carboxy-2-phenylbenzofurans and benzoxazin-4-ones) with IC50 down to 100 nM. Kinetic washout and onset of action studies revealed an extracellular site of action for the thiazolo-pyrimidin-5-one and 3-carboxy-2-phenylbenzofuran inhibitors. Molecular docking computations revealed putative binding sites for these inhibitors. In a loperamide model of constipation in mice, orally administered 7-(2-chloro-phenoxymethyl)-3-phenyl-thiazolo [3,2-a]pyrimidin-5-one (3a) significantly increased stool weight, pellet number and water content. SLC26A3 inhibitors with an extracellular site of action offer the possibility of creating non-absorbable, luminally acting inhibitors with minimal systemic exposure following oral administration. Our findings also suggest that inhibitors of related SLC26 anion transporters with an extracellular site of action might be identified for pharmacological modulation of selected epithelial ion transport processes.


Assuntos
Antiporters , Constipação Intestinal , Camundongos , Animais , Antiporters/química , Antiporters/metabolismo , Antiporters/farmacologia , Simulação de Acoplamento Molecular , Transporte Biológico , Ânions , Cloretos/metabolismo , Transportadores de Sulfato/metabolismo
3.
Open Biol ; 11(6): 200400, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34186010

RESUMO

Wnt gradients elicit distinct cellular responses, such as proliferation, specification, differentiation and survival in a dose-dependent manner. Porcupine (PORCN), a membrane-bound O-acyl transferase (MBOAT) that resides in the endoplasmic reticulum, catalyses the addition of monounsaturated palmitate to Wnt proteins and is required for Wnt gradient formation and signalling. In humans, PORCN mutations are causal for focal dermal hypoplasia (FDH), an X-linked dominant syndrome characterized by defects in mesodermal and endodermal tissues. PORCN is also an emerging target for cancer therapeutics. Despite the importance of this enzyme, its structure remains poorly understood. Recently, the crystal structure of DltB, an MBOAT family member from bacteria, was solved. In this report, we use experimental data along with homology modelling to DltB to determine the membrane topology of PORCN. Our studies reveal that PORCN has 11 membrane domains, comprising nine transmembrane spanning domains and two reentrant domains. The N-terminus is oriented towards the lumen while the C-terminus is oriented towards the cytosol. Like DltB, PORCN has a funnel-like structure that is encapsulated by multiple membrane-spanning helices. This new model for PORCN topology allows us to map residues that are important for biological activity (and implicated in FDH) onto its three-dimensional structure.


Assuntos
Aciltransferases/metabolismo , Membrana Celular/química , Membrana Celular/metabolismo , Proteínas de Membrana/metabolismo , Via de Sinalização Wnt , Aciltransferases/química , Algoritmos , Animais , Linhagem Celular , Biologia Computacional/métodos , Sequência Consenso , Imunofluorescência , Glicosilação , Humanos , Proteínas de Membrana/química , Modelos Moleculares , Simulação de Dinâmica Molecular , Ligação Proteica , Conformação Proteica , Relação Estrutura-Atividade
4.
Kidney Int ; 100(2): 311-320, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33836171

RESUMO

Hypertension is a major cause of cardiovascular morbidity and mortality, despite the availability of antihypertensive drugs with different targets and mechanisms of action. Here, we provide evidence that pharmacological inhibition of TMEM16A (ANO1), a calcium-activated chloride channel expressed in vascular smooth muscle cells, blocks calcium-activated chloride currents and contraction in vascular smooth muscle in vitro and decreases blood pressure in spontaneously hypertensive rats. The acylaminocycloalkylthiophene TMinh-23 fully inhibited calcium-activated TMEM16A chloride current with nanomolar potency in Fischer rat thyroid cells expressing TMEM16A, and in primary cultures of rat vascular smooth muscle cells. TMinh-23 reduced vasoconstriction caused by the thromboxane mimetic U46619 in mesenteric resistance arteries of wild-type and spontaneously hypertensive rats, with a greater inhibition in spontaneously hypertensive rats. Blood pressure measurements by tail-cuff and telemetry showed up to a 45-mmHg reduction in systolic blood pressure lasting for four-six hours in spontaneously hypertensive rats after a single dose of TMinh-23. A minimal effect on blood pressure was seen in wild-type rats or mice treated with TMinh-23. Five-day twice daily treatment of spontaneously hypertensive rats with TMinh-23 produced sustained reductions of 20-25 mmHg in daily mean systolic and diastolic blood pressure. TMinh-23 action was reversible, with blood pressure returning to baseline in spontaneously hypertensive rats by three days after treatment discontinuation. Thus, our studies provide validation for TMEM16A as a target for antihypertensive therapy and demonstrate the efficacy of TMinh-23 as an antihypertensive with a novel mechanism of action.


Assuntos
Anoctamina-1/antagonistas & inibidores , Hipertensão , Músculo Liso Vascular , Vasoconstrição , Animais , Pressão Sanguínea/efeitos dos fármacos , Canais de Cloreto , Hipertensão/tratamento farmacológico , Contração Muscular/efeitos dos fármacos , Ratos , Ratos Endogâmicos SHR
7.
Bioorg Med Chem Lett ; 32: 127683, 2021 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-33227414

RESUMO

The protozoan parasite Plasmodium falciparum causes the most severe form of human malaria and is estimated to kill 400,000 people a year. The parasite infects and replicates in host red blood cells (RBCs), where it expresses an array of proteases to carry out multiple essential processes. We are investigating the function of falcilysin (FLN), a protease known to be required for parasite development in the RBC. We previously developed a piperazine-based hydroxamic acid scaffold to generate the first inhibitors of FLN, and the current study reports the optimization of the lead compound from that series. A range of substituents were tested at the N1 and N4 positions of the piperazine core, and inhibitors with significantly improved potency against purified FLN and cultured P. falciparum were identified. Computational studies were also performed to understand the mode of binding for these compounds, and predicted a binding model consistent with the biochemical data and the distinctive SAR observed at both the N1 and N4 positions.


Assuntos
Antimaláricos/química , Ácidos Hidroxâmicos/química , Metaloendopeptidases/antagonistas & inibidores , Piperazina/química , Proteínas de Protozoários/antagonistas & inibidores , Antimaláricos/metabolismo , Antimaláricos/farmacologia , Sítios de Ligação , Ácidos Hidroxâmicos/metabolismo , Ácidos Hidroxâmicos/farmacologia , Metaloendopeptidases/metabolismo , Simulação de Acoplamento Molecular , Plasmodium falciparum/efeitos dos fármacos , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/metabolismo , Relação Estrutura-Atividade
8.
J Med Chem ; 63(20): 11902-11919, 2020 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-32945666

RESUMO

Malaria remains one of the most deadly infectious diseases, causing hundreds of thousands of deaths each year, primarily in young children and pregnant mothers. Here, we report the discovery and derivatization of a series of pyrazolo[3,4-b]pyridines targeting Plasmodium falciparum, the deadliest species of the malaria parasite. Hit compounds in this series display sub-micromolar in vitro activity against the intraerythrocytic stage of the parasite as well as little to no toxicity against the human fibroblast BJ and liver HepG2 cell lines. In addition, our hit compounds show good activity against the liver stage of the parasite but little activity against the gametocyte stage. Parasitological profiles, including rate of killing, docking, and molecular dynamics studies, suggest that our compounds may target the Qo binding site of cytochrome bc1.


Assuntos
Antimaláricos/farmacologia , Plasmodium falciparum/efeitos dos fármacos , Pirazóis/farmacologia , Piridinas/farmacologia , Antimaláricos/síntese química , Antimaláricos/química , Linhagem Celular , Relação Dose-Resposta a Droga , Células Hep G2 , Humanos , Modelos Moleculares , Estrutura Molecular , Testes de Sensibilidade Parasitária , Pirazóis/síntese química , Pirazóis/química , Piridinas/síntese química , Piridinas/química , Relação Estrutura-Atividade
9.
FASEB J ; 33(10): 11247-11257, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31299174

RESUMO

Interstitial cells of Cajal, which express the calcium-activated chloride channel transmembrane member 16A (TMEM16A), are an important determinant of gastrointestinal (GI) motility. We previously identified the acylaminocycloalkylthiophene class of TMEM16A inhibitors, which, following medicinal chemistry, gave analog 2-bromodifluoroacetylamino-5,6,7,8-tetrahydro-4H-cyclohepta[b]thiophene-3-carboxylic acid o-tolylamide (TMinh-23) with 30 nM half-maximal inhibitory concentration. Here, we tested the efficacy of TMinh-23 for inhibition of GI motility in mice. In isolated murine gastric antrum, TMinh-23 strongly inhibited spontaneous and carbachol-stimulated rhythmic contractions. Pharmacokinetic analysis showed predicted therapeutic concentrations of TMinh-23 for at least 4 h following a single oral or intraperitoneal dose at 10 mg/kg. Gastric emptying, as assessed following an oral bolus of phenol red or independently by [99mTc]-diethylenetriamine pentaacetic acid scintigraphy, was reduced by TMinh-23 by ∼60% at 20 min. Interestingly, there was little effect of TMinh-23 on baseline whole-gut transit time or time to diarrhea induced by castor oil. Consequent to the delay in gastric emptying, TMinh-23 administration significantly reduced the elevation in blood sugar in mice following an oral but not intraperitoneal glucose load. These results provide pharmacological evidence for involvement of TMEM16A in gastric emptying and suggest the utility of TMEM16A inhibition in disorders of accelerated gastric emptying, such as dumping syndrome, and potentially for improving glucose tolerance in diabetes mellitus/metabolic syndrome and enhancing satiety in obesity.-Cil, O., Anderson, M. O., Yen, R., Kelleher, B., Huynh, T. L., Seo, Y., Nilsen, S. P., Turner, J. R., Verkman, A. S. Slowed gastric emptying and improved oral glucose tolerance produced by a nanomolar-potency inhibitor of calcium-activated chloride channel TMEM16A.


Assuntos
Anoctamina-1/metabolismo , Cálcio/metabolismo , Agonistas dos Canais de Cloreto/farmacologia , Canais de Cloreto/metabolismo , Esvaziamento Gástrico/efeitos dos fármacos , Glucose/metabolismo , Proteínas de Neoplasias/metabolismo , Animais , Glicemia/efeitos dos fármacos , Cloretos/metabolismo , Feminino , Motilidade Gastrointestinal/efeitos dos fármacos , Teste de Tolerância a Glucose/métodos , Humanos , Camundongos
10.
J Mol Model ; 25(7): 194, 2019 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-31209577

RESUMO

The Zika virus has recently become a subject of acute interest after the discovery of the link between viral infection and microcephaly in infants. Though a number of treatments are under active investigation, there are currently no approved treatments for the disease. To address this critical need, we screened more than 7 million compounds targeting the NS2B-NS3 protease in an attempt to identify promising inhibitor candidates. Starting with commercially and freely available compounds, we identified six hits utilizing an exhaustive consensus screening protocol, followed by molecular dynamics simulation and binding energy estimation using MM/GBSA and MM/PBSA methods. These compounds feature a variety of cores and functionalities, and all are predicted to have good pharmacokinetic profiles, making them promising candidates for screening assays. Graphical abstract Virtual screen of potential Zika virus NS2B-NS3 protease inhibitors.


Assuntos
Antivirais/química , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Peptídeo Hidrolases/química , Inibidores de Proteases/química , Proteínas não Estruturais Virais/química , Zika virus/metabolismo , Antivirais/farmacologia , Avaliação Pré-Clínica de Medicamentos , Humanos , Conformação Molecular , Estrutura Molecular , Inibidores de Proteases/farmacologia , Ligação Proteica , Proteínas não Estruturais Virais/antagonistas & inibidores , Zika virus/efeitos dos fármacos
11.
J Med Chem ; 61(7): 3209-3217, 2018 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-29589443

RESUMO

Urea transporter A (UT-A) isoforms encoded by the Slc14a2 gene are expressed in kidney tubule epithelial cells, where they facilitate urinary concentration. UT-A1 inhibition is predicted to produce a unique salt-sparing diuretic action in edema and hyponatremia. Here we report the discovery of 1,2,4-triazoloquinoxalines and the analysis of 37 synthesized analogues. The most potent compound, 8ay, containing 1,2,4-triazolo[4,3- a]quinoxaline-substituted benzenesulfonamide linked by an aryl ether, rapidly and reversibly inhibited UT-A1 urea transport by a noncompetitive mechanism with IC50 ≈ 150 nM; the IC50 was ∼2 µM for the related urea transporter UT-B encoded by the Slc14a1 gene. Molecular modeling suggested a putative binding site on the UT-A1 cytoplasmic domain. In vitro metabolism showing quinoxaline ring oxidation prompted the synthesis of metabolically stable 7,8-difluoroquinoxaline analogue 8bl, which when administered to rats produced marked diuresis and reduced urinary osmolality. 8bl has substantially improved UT-A1 inhibition potency and metabolic stability compared with prior compounds.


Assuntos
Rim/metabolismo , Proteínas de Membrana Transportadoras/efeitos dos fármacos , Quinoxalinas/síntese química , Quinoxalinas/farmacologia , Animais , Diurese/efeitos dos fármacos , Diuréticos/síntese química , Diuréticos/farmacologia , Cães , Humanos , Rim/efeitos dos fármacos , Células Madin Darby de Rim Canino , Masculino , Modelos Moleculares , Conformação Molecular , Simulação de Acoplamento Molecular , Concentração Osmolar , Quinoxalinas/farmacocinética , Ratos , Ratos Wistar , Relação Estrutura-Atividade , Ureia/metabolismo , Transportadores de Ureia
12.
Expert Opin Ther Targets ; 21(12): 1161-1170, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29072508

RESUMO

INTRODUCTION: Aquaporin-4 (AQP4) is a water transporting protein expressed at the plasma membrane of astrocytes throughout the central nervous system (CNS). Analysis of AQP4 knockout mice has suggested its broad involvement in brain water balance, neuroexcitation, glial scarring, neuroinflammation, and even neurodegenerative and neuropsychiatric disorders. Broad clinical utility of AQP4 modulators has been speculated. Area covered: This review covers the biology of AQP4, evidence for its roles in normal CNS function and neurological disorders, and progress in AQP4 drug discovery. Expert opinion: Critical examination of available data reduces the lengthy potential applications list to AQP4 inhibitors for early therapy of ischemic stroke and perhaps for reduction of glial scarring following CNS injury. Major challenges in identification and clinical development of AQP4 inhibitors include the apparent poor druggability of AQPs, the many homologous AQP isoforms with broad tissue distribution and functions, technical issues with water transport assays, predicted undesired CNS and non-CNS actions, and the need for high blood-brain barrier permeation. To date, despite considerable effort, validated small-molecule AQP4 inhibitors have not been advanced. However, a biologic ('aquaporumab') is in development for neuromyelitis optica, an autoimmune inflammatory demyelinating disease where CNS pathology is initiated by binding of anti-AQP4 autoantibodies to astrocyte AQP4.


Assuntos
Aquaporina 4/antagonistas & inibidores , Doenças do Sistema Nervoso Central/tratamento farmacológico , Desenho de Fármacos , Animais , Aquaporina 4/metabolismo , Astrócitos/metabolismo , Barreira Hematoencefálica/metabolismo , Sistema Nervoso Central/fisiologia , Sistema Nervoso Central/fisiopatologia , Fármacos do Sistema Nervoso Central/farmacocinética , Fármacos do Sistema Nervoso Central/farmacologia , Doenças do Sistema Nervoso Central/genética , Doenças do Sistema Nervoso Central/fisiopatologia , Descoberta de Drogas/métodos , Humanos , Camundongos , Camundongos Knockout , Terapia de Alvo Molecular
13.
J Med Chem ; 60(11): 4626-4635, 2017 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-28493701

RESUMO

Transmembrane protein 16A (TMEM16A), also called anoctamin 1 (ANO1), is a calcium-activated chloride channel expressed widely mammalian cells, including epithelia, vascular smooth muscle tissue, electrically excitable cells, and some tumors. TMEM16A inhibitors have been proposed for treatment of disorders of epithelial fluid and mucus secretion, hypertension, asthma, and possibly cancer. Herein we report, by screening, the discovery of 2-acylaminocycloalkylthiophene-3-carboxylic acid arylamides (AACTs) as inhibitors of TMEM16A and analysis of 48 synthesized analogs (10ab-10bw) of the original AACT compound (10aa). Structure-activity studies indicated the importance of benzene substituted as 2- or 4-methyl, or 4-fluoro, and defined the significance of thiophene substituents and size of the cycloalkylthiophene core. The most potent compound (10bm), which contains an unusual bromodifluoroacetamide at the thiophene 2-position, had IC50 of ∼30 nM, ∼3.6-fold more potent than the most potent previously reported TMEM16A inhibitor 4 (Ani9), and >10-fold improved metabolic stability. Direct and reversible inhibition of TMEM16A by 10bm was demonstrated by patch-clamp analysis. AACTs may be useful as pharmacological tools to study TMEM16A function and as potential drug development candidates.


Assuntos
Canais de Cloreto/antagonistas & inibidores , Tiofenos/química , Tiofenos/farmacologia , Animais , Anoctamina-1 , Ácidos Carboxílicos/química , Células Cultivadas , Humanos , Ratos , Ratos Endogâmicos F344 , Relação Estrutura-Atividade
14.
Adv Exp Med Biol ; 969: 239-250, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28258578

RESUMO

Drugs targeting aquaporins have broad potential clinical applications, including cancer, obesity, edema, glaucoma, skin diseases and others. The astrocyte water channel aquaporin-4 is a particularly compelling target because of its role of brain water movement, neuroexcitation and glia scarring, and because it is the target of pathogenic autoantibodies in the neuroinflammatory demyelinating disease neuromyelitis optica . There has been considerable interest in the identification of small molecule inhibitors of aquaporins, with various candidates emerging from testing of known ion transport inhibitors, as well as compound screening and computational chemistry. However, in general, the activity of reported aquaporin inhibitors has not been confirmed on retesting, which may be due to technical problems in water transport assays used in the original identification studies, and the challenges in modulating the activity of small, compact, pore-containing membrane proteins. We review here the state of the field of aquaporin-modulating small molecules and biologics, and the challenges and opportunities in moving forward.


Assuntos
Aquaporina 4/antagonistas & inibidores , Edema Encefálico/tratamento farmacológico , Neuromielite Óptica/tratamento farmacológico , Tiadiazóis/farmacologia , Água/metabolismo , Animais , Aquaporina 4/genética , Aquaporina 4/metabolismo , Transporte Biológico , Edema Encefálico/genética , Edema Encefálico/metabolismo , Edema Encefálico/patologia , Regulação da Expressão Gênica , Humanos , Terapia de Alvo Molecular , Neuromielite Óptica/genética , Neuromielite Óptica/metabolismo , Neuromielite Óptica/patologia , Concentração Osmolar , Pressão Osmótica , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Sumatriptana/farmacologia , Triazóis/farmacologia , Triptaminas/farmacologia
15.
PLoS Pathog ; 12(5): e1005628, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27191388

RESUMO

[This corrects the article DOI: 10.1371/journal.ppat.1003576.].

16.
Mol Pharmacol ; 89(6): 686-93, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26993802

RESUMO

The aquaporin-1 (AQP1) water channel is a potentially important drug target, as AQP1 inhibition is predicted to have therapeutic action in edema, tumor growth, glaucoma, and other conditions. Here, we measured the AQP1 inhibition efficacy of 12 putative small-molecule AQP1 inhibitors reported in six recent studies, and one AQP1 activator. Osmotic water permeability was measured by stopped-flow light scattering in human and rat erythrocytes that natively express AQP1, in hemoglobin-free membrane vesicles from rat and human erythrocytes, and in plasma membrane vesicles isolated from AQP1-transfected Chinese hamster ovary cell cultures. As a positive control, 0.3 mM HgCl2 inhibited AQP1 water permeability by >95%. We found that none of the tested compounds at 50 µM significantly inhibited or increased AQP1 water permeability in these assays. Identification of AQP1 inhibitors remains an important priority.


Assuntos
Aquaporina 1/antagonistas & inibidores , Bibliotecas de Moléculas Pequenas/farmacologia , Animais , Aquaporina 1/metabolismo , Células CHO , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Permeabilidade da Membrana Celular/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Cricetinae , Cricetulus , Eritrócitos/efeitos dos fármacos , Eritrócitos/metabolismo , Fluoresceínas/metabolismo , Hemoglobinas/metabolismo , Humanos , Osmose/efeitos dos fármacos , Ratos Wistar , Bibliotecas de Moléculas Pequenas/química , Água/metabolismo
17.
J Enzyme Inhib Med Chem ; 31(6): 1362-8, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26796863

RESUMO

Transmembrane protein 16A (TMEM16A), also called Ano1, is a Ca(2+) activated Cl(-) channel expressed widely in mammalian epithelia, as well as in vascular smooth muscle and some tumors and electrically excitable cells. TMEM16A inhibitors have potential utility for treatment of disorders of epithelial fluid and mucus secretion, hypertension, some cancers and other diseases. 4-Aryl-2-amino thiazole T16Ainh-01 was previously identified by high-throughput screening. Here, a library of 47 compounds were prepared that explored the 5,6-disubstituted pyrimidine scaffold found in T16Ainh-01. TMEM16A inhibition activity was measured using fluorescence plate reader and short-circuit current assays. We found that very little structural variation of T16Ainh-01 was tolerated, with most compounds showing no activity at 10 µM. The most potent compound in the series, 9bo, which substitutes 4-methoxyphenyl in T16Ainh-01 with 2-thiophene, had IC50 ∼1 µM for inhibition of TMEM16A chloride conductance.


Assuntos
Canais de Cloreto/antagonistas & inibidores , Tiazóis/síntese química , Tiazóis/farmacologia , Animais , Anoctamina-1 , Espectroscopia de Ressonância Magnética Nuclear de Carbono-13 , Linhagem Celular , Espectroscopia de Prótons por Ressonância Magnética , Ratos , Ratos Endogâmicos F344 , Espectrometria de Massas por Ionização por Electrospray
18.
Medchemcomm ; 6: 1278-1284, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26191399

RESUMO

Kidney urea transporters are targets for development of small-molecule inhibitors with action as salt-sparing diuretics. A cell-based, functional high-throughput screen identified 2,7-bisacetamido fluorenone 3 as a novel inhibitor of urea transporters UT-A1 and UT-B. Here, we synthesized twenty-two 2,7-disubstituted fluorenone analogs by acylation. Structure-activity relationship analysis revealed: (a) the carbonyl moiety at C9 is required for UT inhibition; (b) steric limitation on C2, 7-substituents; and (c) the importance of a crescent-shape structure. The most potent fluorenones inhibited UT-A1 and UT-B urea transport with IC50 ~ 1 µM. Analysis of in vitro metabolic stability in hepatic microsomes indicated metabolism of 2,7-disubstituted fluorenones by reductase and subsequent elimination. Computational docking to a homology model of UT-A1 suggested UT inhibitor binding to the UT cytoplasmic domain at a site that does not overlap with the putative urea binding site.

19.
Mol Pharmacol ; 88(4): 689-96, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26174774

RESUMO

We previously reported that benzopyrimido-pyrrolo-oxazinedione BPO-27 [6-(5-bromofuran-2-yl)-7,9-dimethyl-8,10-dioxo-11-phenyl-7,8,9,10-tetrahydro-6H-benzo[b]pyrimido [4',5':3,4]pyrrolo [1,2-d][1,4]oxazine-2-carboxylic acid] inhibits the cystic fibrosis transmembrane conductance regulator (CFTR) chloride channel with low nanomolar potency and reduces cystogenesis in a model of polycystic kidney disease. We used computational chemistry and patch-clamp to show that enantiomerically pure (R)-BPO-27 inhibits CFTR by competition with ATP, whereas (S)-BPO-27 is inactive. Docking computations using a homology model of CFTR structure suggested that (R)-BPO-27 binds near the canonical ATP binding site, and these findings were supported by molecular dynamics simulations showing a lower binding energy for the (R) versus (S) stereoisomers. Three additional lower-potency BPO-27 analogs were modeled in a similar fashion, with the binding energies predicted in the correct order. Whole-cell patch-clamp studies showed linear CFTR currents with a voltage-independent (R)-BPO-27 block mechanism. Single-channel recordings in inside-out patches showed reduced CFTR channel open probability and increased channel closed time by (R)-BPO-27 without altered unitary channel conductance. At a concentration of (R)-BPO-27 that inhibited CFTR chloride current by ∼50%, the EC50 for ATP activation of CFTR increased from 0.27 to 1.77 mM but was not changed by CFTRinh-172 [4-[[4-oxo-2-thioxo-3-[3-trifluoromethyl)phenyl]-5-thiazolidinylidene]methyl]benzoic acid], a thiazolidinone CFTR inhibitor that acts at a site distinct from the ATP binding site. Our results suggest that (R)-BPO-27 inhibition of CFTR involves competition with ATP.


Assuntos
Trifosfato de Adenosina/metabolismo , Ligação Competitiva/fisiologia , Regulador de Condutância Transmembrana em Fibrose Cística/antagonistas & inibidores , Regulador de Condutância Transmembrana em Fibrose Cística/fisiologia , Pirimidinas/metabolismo , Sítios de Ligação/fisiologia , Ligação Competitiva/efeitos dos fármacos , Relação Dose-Resposta a Droga , Células HEK293 , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/fisiologia , Oxazinas/química , Oxazinas/metabolismo , Oxazinas/farmacologia , Estrutura Secundária de Proteína , Pirimidinas/química , Pirimidinas/farmacologia
20.
Kidney Int ; 88(2): 311-20, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25993324

RESUMO

Inhibitors of kidney urea transporter (UT) proteins have potential use as salt-sparing diuretics ('urearetics') with a different mechanism of action than diuretics that target salt transporters. To study UT inhibition in rats, we screened about 10,000 drugs, natural products and urea analogs for inhibition of rat UT-A1. Drug and natural product screening found nicotine, sanguinarine and an indolcarbonylchromenone with IC50 of 10-20 µM. Urea analog screening found methylacetamide and dimethylthiourea (DMTU). DMTU fully and reversibly inhibited rat UT-A1 and UT-B by a noncompetitive mechanism with IC50 of 2-3 mM. Homology modeling and docking computations suggested DMTU binding sites on rat UT-A1. Following a single intraperitoneal injection of 500 mg/kg DMTU, peak plasma concentration was 9 mM with t1/2 of about 10 h, and a urine concentration of 20-40 mM. Rats chronically treated with DMTU had a sustained, reversible reduction in urine osmolality from 1800 to 600 mOsm, a 3-fold increase in urine output, and mild hypokalemia. DMTU did not impair urinary concentrating function in rats on a low protein diet. Compared to furosemide-treated rats, the DMTU-treated rats had greater diuresis and reduced urinary salt loss. In a model of syndrome of inappropriate antidiuretic hormone secretion, DMTU treatment prevented hyponatremia and water retention produced by water-loading in dDAVP-treated rats. Thus, our results establish a rat model of UT inhibition and demonstrate the diuretic efficacy of UT inhibition.


Assuntos
Diurese/efeitos dos fármacos , Proteínas de Membrana Transportadoras/metabolismo , Cloreto de Sódio/urina , Tioureia/análogos & derivados , Animais , Sítios de Ligação , Modelos Animais de Doenças , Diuréticos/farmacologia , Cães , Avaliação Pré-Clínica de Medicamentos , Feminino , Furosemida/farmacologia , Hipopotassemia/induzido quimicamente , Hiponatremia/etiologia , Hiponatremia/prevenção & controle , Síndrome de Secreção Inadequada de HAD/complicações , Síndrome de Secreção Inadequada de HAD/tratamento farmacológico , Concentração Inibidora 50 , Células Madin Darby de Rim Canino , Proteínas de Membrana Transportadoras/química , Estrutura Molecular , Concentração Osmolar , Ratos , Ratos Wistar , Tioureia/sangue , Tioureia/química , Tioureia/farmacologia , Tioureia/uso terapêutico , Fatores de Tempo , Urina/química , Transportadores de Ureia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...