Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
Bioorg Med Chem Lett ; 29(16): 2119-2123, 2019 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-31281021

RESUMO

Pendrin is a transmembrane chloride/anion antiporter that is strongly upregulated in the airways in rhinoviral infection, asthma, cystic fibrosis and chronic rhinosinusitis. Based on its role in the regulation of airway surface liquid depth, pendrin inhibitors have potential indications for treatment of inflammatory airways diseases. Here, a completely regioselective route to tetrahydro-pyrazolopyridine pendrin inhibitors based on 1,3-diketone and substituted hydrazine condensation was been developed. Structure-activity relationships at the tetrahydropyridyl nitrogen were investigated using a focused library, establishing the privileged nature of N-phenyl ureas and improving inhibitor potency by greater than 2-fold.


Assuntos
Pirazóis/farmacologia , Piridinas/farmacologia , Transportadores de Sulfato/antagonistas & inibidores , Animais , Camundongos , Estrutura Molecular , Pirazóis/síntese química , Piridinas/síntese química , Ratos Endogâmicos F344 , Bibliotecas de Moléculas Pequenas/síntese química , Bibliotecas de Moléculas Pequenas/farmacologia , Relação Estrutura-Atividade
2.
J Biol Chem ; 292(3): 771-785, 2017 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-27895116

RESUMO

W1282X is the fifth most common cystic fibrosis transmembrane regulator (CFTR) mutation that causes cystic fibrosis. Here, we investigated the utility of a small molecule corrector/potentiator strategy, as used for ΔF508-CFTR, to produce functional rescue of the truncated translation product of the W1282X mutation, CFTR1281, without the need for read-through. In transfected cell systems, certain potentiators and correctors, including VX-809 and VX-770, increased CFTR1281 activity. To identify novel correctors and potentiators with potentially greater efficacy on CFTR1281, functional screens were done of ∼30,000 synthetic small molecules and drugs/nutraceuticals in CFTR1281-transfected cells. Corrector scaffolds of 1-arylpyrazole-4-arylsulfonyl-piperazine and spiro-piperidine-quinazolinone classes were identified with up to ∼5-fold greater efficacy than VX-809, some of which were selective for CFTR1281, whereas others also corrected ΔF508-CFTR. Several novel potentiator scaffolds were identified with efficacy comparable with VX-770; remarkably, a phenylsulfonamide-pyrrolopyridine acted synergistically with VX-770 to increase CFTR1281 function ∼8-fold over that of VX-770 alone, normalizing CFTR1281 channel activity to that of wild type CFTR. Corrector and potentiator combinations were tested in primary cultures and conditionally reprogrammed cells generated from nasal brushings from one W1282X homozygous subject. Although robust chloride conductance was seen with correctors and potentiators in homozygous ΔF508 cells, increased chloride conductance was not found in W1282X cells despite the presence of adequate transcript levels. Notwithstanding the negative data in W1282X cells from one human subject, we speculate that corrector and potentiator combinations may have therapeutic efficacy in cystic fibrosis caused by the W1282X mutation, although additional studies are needed on human cells from W1282X subjects.


Assuntos
Aminofenóis/farmacologia , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Mutação de Sentido Incorreto , Piperazinas/farmacologia , Quinolonas/farmacologia , Substituição de Aminoácidos , Animais , Células Cultivadas , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Humanos , Ratos , Ratos Endogâmicos F344
3.
FASEB J ; 31(2): 751-760, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27871064

RESUMO

Secretory diarrheas caused by bacterial enterotoxins, including cholera and traveler's diarrhea, remain a major global health problem. Inappropriate activation of the cystic fibrosis transmembrane conductance regulator (CFTR) chloride channel occurs in these diarrheas. We previously reported that the benzopyrimido-pyrrolo-oxazinedione (R)-BPO-27 inhibits CFTR chloride conductance with low-nanomolar potency. Here, we demonstrate using experimental mouse models and human enterocyte cultures the potential utility of (R)-BPO-27 for treatment of secretory diarrheas caused by cholera and Escherichia coli enterotoxins. (R)-BPO-27 fully blocked CFTR chloride conductance in epithelial cell cultures and intestine after cAMP agonists, cholera toxin, or heat-stable enterotoxin of E. coli (STa toxin), with IC50 down to ∼5 nM. (R)-BPO-27 prevented cholera toxin and STa toxin-induced fluid accumulation in small intestinal loops, with IC50 down to 0.1 mg/kg. (R)-BPO-27 did not impair intestinal fluid absorption or inhibit other major intestinal transporters. Pharmacokinetics in mice showed >90% oral bioavailability with sustained therapeutic serum levels for >4 h without the significant toxicity seen with 7-d administration at 5 mg/kg/d. As evidence to support efficacy in human diarrheas, (R)-BPO-27 blocked fluid secretion in primary cultures of enteroids from human small intestine and anion current in enteroid monolayers. These studies support the potential utility of (R)-BPO-27 for therapy of CFTR-mediated secretory diarrheas.-Cil, O., Phuan, P.-W., Gillespie, A. M., Lee, S., Tradtrantip, L., Yin, J., Tse, M., Zachos, N. C., Lin, R., Donowitz, M., Verkman, A. S. Benzopyrimido-pyrrolo-oxazine-dione CFTR inhibitor (R)-BPO-27 for antisecretory therapy of diarrheas caused by bacterial enterotoxins.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/antagonistas & inibidores , Diarreia/induzido quimicamente , Diarreia/tratamento farmacológico , Oxazinas/farmacologia , Pirimidinonas/farmacologia , Pirróis/farmacologia , Animais , Relação Dose-Resposta a Droga , Feminino , Humanos , Intestinos/efeitos dos fármacos , Camundongos , Estrutura Molecular , Oxazinas/síntese química , Pirimidinonas/síntese química , Pirróis/síntese química
4.
FASEB J ; 30(6): 2187-97, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26932931

RESUMO

Pendrin (SLC26A4) is a Cl(-)/anion exchanger expressed in the epithelium of inflamed airways where it is thought to facilitate Cl(-) absorption and HCO3 (-) secretion. Studies using pendrin knockout mice and airway epithelial cells from hearing-impaired subjects with pendrin loss of function suggest involvement of pendrin in inflammatory lung diseases, including cystic fibrosis (CF), perhaps by regulation of airway surface liquid (ASL) volume. Here we identified small-molecule pendrin inhibitors and demonstrated their efficacy in increasing ASL volume. A cell-based, functional high-throughput screen of ∼36,000 synthetic small molecules produced 3 chemical classes of inhibitors of human pendrin. After structure-activity studies, tetrahydropyrazolopyridine and pyrazolothiophenesulfonamide compounds reversibly inhibited pendrin-facilitated Cl(-) exchange with SCN(-), I(-), NO3 (-), and HCO3 (-) with drug concentration causing 50% inhibition down to ∼2.5 µM. In well-differentiated primary cultures of human airway epithelial cells from non-CF and CF subjects, treatment with IL-13, which causes inflammation with strong pendrin up-regulation, strongly increased Cl(-)/HCO3 (-) exchange and the increase was blocked by pendrin inhibition. Pendrin inhibition significantly increased ASL depth (by ∼8 µm) in IL-13-treated non-CF and CF cells but not in untreated cells. These studies implicate the involvement of pendrin-facilitated Cl(-)/HCO3 (-) in the regulation of ASL volume and suggest the utility of pendrin inhibitors in inflammatory lung diseases, including CF.-Haggie, P. M., Phuan, P.-W., Tan, J.-A., Zlock, L., Finkbeiner, W. E., Verkman, A. S. Inhibitors of pendrin anion exchange identified in a small molecule screen increase airway surface liquid volume in cystic fibrosis.


Assuntos
Antiportadores de Cloreto-Bicarbonato/metabolismo , Fibrose Cística/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas de Membrana Transportadoras/metabolismo , Piridinas/farmacologia , Sulfonamidas/farmacologia , Animais , Células Cultivadas , Antiportadores de Cloreto-Bicarbonato/antagonistas & inibidores , Antiportadores de Cloreto-Bicarbonato/genética , Chlorocebus aethiops , Células Epiteliais/efeitos dos fármacos , Humanos , Interleucina-13/farmacologia , Proteínas de Membrana Transportadoras/genética , Piridinas/química , Ratos , Sistema Respiratório/efeitos dos fármacos , Sistema Respiratório/metabolismo , Relação Estrutura-Atividade , Transportadores de Sulfato , Sulfonamidas/química
5.
J Am Soc Nephrol ; 27(12): 3706-3714, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27153921

RESUMO

Pendrin is a Cl-/HCO3- exchanger expressed in type B and non-A, non-B intercalated cells in the distal nephron, where it facilitates Cl- absorption and is involved in Na+ absorption and acid-base balance. Pendrin-knockout mice show no fluid-electrolyte abnormalities under baseline conditions, although mice with double knockout of pendrin and the Na+/Cl- cotransporter (NCC) manifest profound salt wasting. Thus, pendrin may attenuate diuretic-induced salt loss, but this function remains unconfirmed. To clarify the physiologic role of pendrin under conditions not confounded by gene knockout, and to test the potential utility of pendrin inhibitors for diuretic therapy, we tested in mice a small-molecule pendrin inhibitor identified from a high-throughput screen. In vitro, a pyrazole-thiophenesulfonamide, PDSinh-C01, inhibited Cl-/anion exchange mediated by mouse pendrin with a 50% inhibitory concentration of 1-3 µM, without affecting other major kidney tubule transporters. Administration of PDSinh-C01 to mice at predicted therapeutic doses, determined from serum and urine pharmacokinetics, did not affect urine output, osmolality, salt excretion, or acid-base balance. However, in mice treated acutely with furosemide, administration of PDSinh-C01 produced a 30% increase in urine output, with increased Na+ and Cl- excretion. In mice treated long term with furosemide, in which renal pendrin is upregulated, PDSinh-C01 produced a 60% increase in urine output. Our findings clarify the role of pendrin in kidney function and suggest pendrin inhibition as a novel approach to potentiate the action of loop diuretics. Such combination therapy might enhance diuresis and salt excretion for treatment of hypertension and edema, perhaps including diuretic-resistant edema.


Assuntos
Proteínas de Transporte de Ânions/antagonistas & inibidores , Diuréticos/farmacologia , Furosemida/farmacologia , Sulfonamidas/farmacologia , Tiofenos/farmacologia , Animais , Sinergismo Farmacológico , Feminino , Camundongos , Transportadores de Sulfato
6.
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
7.
Biochim Biophys Acta ; 1848(5): 1075-80, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25613743

RESUMO

Small-molecule inhibitors of urea transporter (UT) proteins in kidney have potential application as novel salt-sparing diuretics. The urea analog dimethylthiourea (DMTU) was recently found to inhibit the UT isoforms UT-A1 (expressed in kidney tubule epithelium) and UT-B (expressed in kidney vasa recta endothelium) with IC50 of 2-3 mM, and was shown to have diuretic action when administered to rats. Here, we measured UT-A1 and UT-B inhibition activity of 36 thiourea analogs, with the goal of identifying more potent and isoform-selective inhibitors, and establishing structure-activity relationships. The analog set systematically explored modifications of substituents on the thiourea including alkyl, heterocycles and phenyl rings, with different steric and electronic features. The analogs had a wide range of inhibition activities and selectivities. The most potent inhibitor, 3-nitrophenyl-thiourea, had an IC50 of ~0.2 mM for inhibition of both UT-A1 and UT-B. Some analogs such as 4-nitrophenyl-thiourea were relatively UT-A1 selective (IC50 1.3 vs. 10 mM), and others such as thioisonicotinamide were UT-B selective (IC50>15 vs. 2.8 mM).


Assuntos
Moduladores de Transporte de Membrana/farmacologia , Proteínas de Membrana Transportadoras/efeitos dos fármacos , Tioureia/farmacologia , Ureia/metabolismo , Animais , Cães , Relação Dose-Resposta a Droga , Células Madin Darby de Rim Canino , Moduladores de Transporte de Membrana/química , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Estrutura Molecular , Ratos , Relação Estrutura-Atividade , Tioureia/análogos & derivados , Tioureia/química , Transfecção , Transportadores de Ureia
8.
Mol Pharmacol ; 88(4): 791-9, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26245207

RESUMO

Combination drug therapies under development for cystic fibrosis caused by the ∆F508 mutation in cystic fibrosis transmembrane conductance regulator (CFTR) include a "corrector" to improve its cellular processing and a "potentiator" to improve its chloride channel function. Recently, it was reported that the approved potentiator N-(2,4-di-tert-butyl-5-hydroxyphenyl)-4-oxo-1,4-dihydroquinoline-3-carboxamide (Ivacaftor) reduces ∆F508-CFTR cellular stability and the efficacy of investigational correctors, including 3-(6-[([1-(2,2-difluoro-1,3-benzodioxol-5-yl)cyclopropyl]carbonyl) amino]-3-methyl-2-pyridinyl)-benzoic acid and 1-(2,2-difluoro-1,3-benzodioxol-5-yl)-N-(1-[(2R)-2,3-dihydroxypropyl]-6-fluoro-2-(2-hydroxy-1,1-dimethylethyl)-1H-indol-5-yl), which might contribute to the modest reported efficacy of combination therapy in clinical trials. Here, we report the identification and characterization of potentiators that do not interfere with ∆F508-CFTR stability or corrector action. High-throughput screening and structure-activity analysis identified several classes of potentiators that do not impair corrector action, including tetrahydrobenzothiophenes, thiooxoaminothiazoles, and pyrazole-pyrrole-isoxazoles. The most potent compounds have an EC(50) for ∆F508-CFTR potentiation down to 18 nM and do not reduce corrector efficacy in heterologous ∆F508-CFTR-expressing cells or primary cultures of ∆F508/∆F508 human bronchial epithelia. The ΔF508-CFTR potentiators also activated wild-type and G551D CFTR, albeit weakly. The efficacy of combination therapy for cystic fibrosis caused by the ∆F508 mutation may be improved by replacement of Ivacaftor with a potentiator that does not interfere with corrector action.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Ativação do Canal Iônico/fisiologia , Aminopiridinas/química , Aminopiridinas/metabolismo , Aminopiridinas/farmacologia , Animais , Benzodioxóis/química , Benzodioxóis/metabolismo , Benzodioxóis/farmacologia , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Células Cultivadas , Fibrose Cística/metabolismo , Fibrose Cística/patologia , Regulador de Condutância Transmembrana em Fibrose Cística/agonistas , Regulador de Condutância Transmembrana em Fibrose Cística/química , Regulador de Condutância Transmembrana em Fibrose Cística/farmacologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Ratos , Ratos Endogâmicos F344 , Relação Estrutura-Atividade
9.
FASEB J ; 28(9): 3878-90, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24843071

RESUMO

Urea transport (UT) proteins of the UT-A class are expressed in epithelial cells in kidney tubules, where they are required for the formation of a concentrated urine by countercurrent multiplication. Here, using a recently developed high-throughput assay to identify UT-A inhibitors, a screen of 50,000 synthetic small molecules identified UT-A inhibitors of aryl-thiazole, γ-sultambenzosulfonamide, aminocarbonitrile butene, and 4-isoxazolamide chemical classes. Structure-activity analysis identified compounds that inhibited UT-A selectively by a noncompetitive mechanism with IC50 down to ∼1 µM. Molecular modeling identified putative inhibitor binding sites on rat UT-A. To test compound efficacy in rats, formulations and administration procedures were established to give therapeutic inhibitor concentrations in blood and urine. We found that intravenous administration of an indole thiazole or a γ-sultambenzosulfonamide at 20 mg/kg increased urine output by 3-5-fold and reduced urine osmolality by ∼2-fold compared to vehicle control rats, even under conditions of maximum antidiuresis produced by 1-deamino-8-D-arginine vasopressin (DDAVP). The diuresis was reversible and showed urea > salt excretion. The results provide proof of concept for the diuretic action of UT-A-selective inhibitors. UT-A inhibitors are first in their class salt-sparing diuretics with potential clinical indications in volume-overload edemas and high-vasopressin-associated hyponatremias.


Assuntos
Transporte Biológico/efeitos dos fármacos , Diurese/efeitos dos fármacos , Capacidade de Concentração Renal/efeitos dos fármacos , Proteínas de Membrana Transportadoras/química , Bibliotecas de Moléculas Pequenas/farmacologia , Urina/química , Animais , Cromatografia Líquida , Diurese/fisiologia , Cães , Ensaios de Triagem em Larga Escala , Células Madin Darby de Rim Canino , Masculino , Proteínas de Membrana Transportadoras/metabolismo , Modelos Moleculares , Concentração Osmolar , Ratos , Ratos Wistar , Bibliotecas de Moléculas Pequenas/síntese química , Bibliotecas de Moléculas Pequenas/farmacocinética , Cloreto de Sódio , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Relação Estrutura-Atividade , Distribuição Tecidual , Sistema Urinário/efeitos dos fármacos , Sistema Urinário/metabolismo , Transportadores de Ureia
10.
Cell Microbiol ; 16(1): 1-14, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23910799

RESUMO

Pseudomonas aeruginosa infections are commonly associated with cystic fibrosis, pneumonias, neutropenia and burns. The P. aeruginosa quorum sensing molecule N-(3-oxo-dodecanoyl) homoserine lactone (C12) cause multiple deleterious host responses, including repression of NF-κB transcriptional activity and apoptosis. Inhibition of C12-mediated host responses is predicted to reduce P. aeruginosa virulence. We report here a novel, host-targeted approach for potential adjunctive anti-Pseudomonal therapy based on inhibition of C12-mediated host responses. A high-throughput screen was developed to identify C12 inhibitors that restore NF-κB activity in C12-treated, lipopolysaccharide (LPS)-stimulated cells. Triazolo[4,3-a]quinolines with nanomolar potency were identified as C12-inhibitors that restore NF-κB-dependent luciferase expression in LPS- and TNF-stimulated cell lines. In primary macrophages and fibroblasts, triazolo[4,3-a]quinolines inhibited C12 action to restore cytokine secretion in LPS-stimulated cells. Serendipitously, in the absence of an inflammatory stimulus, triazolo[4,3-a]quinolines prevented C12-mediated responses, including cytotoxicity, elevation of cytoplasmic calcium, and p38 MAPK phosphorylation. In vivo efficacy was demonstrated in a murine model of dermal inflammation involving intradermalC12 administration. The discovery of triazolo[4,3-a]quinolines provides a pharmacological tool to investigate C12-mediated host responses, and a potential host-targeted anti-Pseudomonal therapy.


Assuntos
4-Butirolactona/análogos & derivados , Antibacterianos/isolamento & purificação , Pseudomonas aeruginosa/fisiologia , Quinolinas/isolamento & purificação , Fatores de Virulência/antagonistas & inibidores , 4-Butirolactona/antagonistas & inibidores , 4-Butirolactona/metabolismo , 4-Butirolactona/toxicidade , Animais , Antibacterianos/farmacologia , Células Cultivadas , Avaliação Pré-Clínica de Medicamentos , Ensaios de Triagem em Larga Escala , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Camundongos , NF-kappa B/metabolismo , Pseudomonas aeruginosa/imunologia , Quinolinas/farmacologia , Transdução de Sinais , Fatores de Virulência/metabolismo , Fatores de Virulência/toxicidade
11.
Mol Pharmacol ; 86(1): 42-51, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24737137

RESUMO

The most prevalent cystic fibrosis transmembrane conductance regulator (CFTR) mutation causing cystic fibrosis, ΔF508, impairs folding of nucleotide binding domain (NBD) 1 and stability of the interface between NBD1 and the membrane-spanning domains. The interfacial stability defect can be partially corrected by the investigational drug VX-809 (3-[6-[[[1-(2,2-difluoro-1,3-benzodioxol-5-yl)cyclopropyl]carbonyl]amino]-3-methyl-2-pyridinyl]-benzoic acid) or the R1070W mutation. Second-generation ΔF508-CFTR correctors are needed to improve on the modest efficacy of existing cystic fibrosis correctors. We postulated that a second corrector targeting a distinct folding/interfacial defect might act in synergy with VX-809 or the R1070W suppressor mutation. A biochemical screen for ΔF508-CFTR cell surface expression was developed in a human lung epithelium-derived cell line (CFBE41o(-)) by expressing chimeric CFTRs with a horseradish peroxidase (HRP) in the fourth exofacial loop in either the presence or absence of R1070W. Using a luminescence readout of HRP activity, screening of approximately 110,000 small molecules produced nine novel corrector scaffolds that increased cell surface ∆F508-CFTR expression by up to 200% in the presence versus absence of maximal VX-809. Further screening of 1006 analogs of compounds identified from the primary screen produced 15 correctors with an EC50 < 5 µM. Eight chemical scaffolds showed synergy with VX-809 in restoring chloride permeability in ∆F508-expressing A549 cells. An aminothiazole increased chloride conductance in human bronchial epithelial cells from a ΔF508 homozygous subject beyond that of maximal VX-809. Mechanistic studies suggested that NBD2 is required for the aminothiazole rescue. Our results provide proof of concept for synergy screening to identify second-generation correctors, which, when used in combination, may overcome the "therapeutic ceiling" of first-generation correctors.


Assuntos
Aminopiridinas/farmacologia , Benzodioxóis/farmacologia , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Mucosa Respiratória/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Animais , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/genética , Brônquios/efeitos dos fármacos , Brônquios/metabolismo , Linhagem Celular , Cloretos/metabolismo , Fibrose Cística/tratamento farmacológico , Fibrose Cística/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Cães , Sinergismo Farmacológico , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Peroxidase do Rábano Silvestre/metabolismo , Humanos , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Células Madin Darby de Rim Canino , Mutação/efeitos dos fármacos , Mutação/genética , Permeabilidade/efeitos dos fármacos , Dobramento de Proteína/efeitos dos fármacos , Estrutura Terciária de Proteína/efeitos dos fármacos , Estrutura Terciária de Proteína/genética , Mucosa Respiratória/metabolismo , Relação Estrutura-Atividade
12.
Bioorg Med Chem Lett ; 24(24): 5840-5844, 2014 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-25452003

RESUMO

The most common mutation causing cystic fibrosis (CF) is deletion of phenylalanine residue 508 in the cystic fibrosis transmembrane regulator conductance (CFTR) protein. Small molecules that are able to correct the misfolding of defective ΔF508-CFTR have considerable promise for therapy. Reported here are the design, preparation, and evaluation of five more hydrophilic bisazole analogs of previously identified bithiazole CF corrector 1. Interestingly, bisazole ΔF508-CFTR corrector activity was not increased by incorporation of more H-bond acceptors (O or N), but correlated best with the overall bisazole molecular geometry. The structure activity data, together with molecular modeling, suggested that active bisazole correctors adopt a U-shaped conformation, and that corrector activity depends on the molecule's ability to access this molecular geometry.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/química , Imidazóis/química , Oxidiazóis/química , Oxazóis/química , Tiadiazóis/química , Fibrose Cística/metabolismo , Fibrose Cística/patologia , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Humanos , Imidazóis/metabolismo , Cinética , Conformação Molecular , Oxidiazóis/metabolismo , Oxazóis/metabolismo , Ligação Proteica , Relação Estrutura-Atividade , Termodinâmica , Tiadiazóis/metabolismo , Tiazóis/química , Água/química
13.
J Biol Chem ; 287(17): 13829-39, 2012 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-22393049

RESUMO

Neuromyelitis optica (NMO) is an inflammatory demyelinating disease of the central nervous system in which binding of pathogenic autoantibodies (NMO-IgG) to astrocyte aquaporin-4 (AQP4) causes complement-dependent cytotoxicity (CDC) and inflammation. We previously reported a wide range of binding affinities of NMO-IgGs to AQP4 in separate tetramers versus intramembrane aggregates (orthogonal arrays of particles, OAPs). We report here a second, independent mechanism by which CDC is affected by AQP4 assembly. Utilizing lactate dehydrogenase release and live/dead cell cytotoxicity assays, we found in different cell lines, and with different monoclonal and patient-derived NMO-IgGs, that CDC was greatly (>100-fold) reduced in cells expressing M1- versus M23-AQP4. Studies using a M23-AQP4 mutant containing an OAP-disrupting mutation, and in cells expressing AQP4 in different M1/M23 ratios, indicated that NMO-IgG-dependent CDC requires AQP4 OAP assembly. In contrast, antibody-dependent cell-mediated cytotoxicity produced by natural killer cells did not depend on AQP4 OAP assembly. Measurements of C1q binding and complement attack complex (C9neo) supported the conclusion that the greatly enhanced CDC by OAPs is due to efficient, multivalent binding of C1q to clustered NMO-IgG on OAPs. We conclude that AQP4 assembly in OAPs is required for CDC in NMO, establishing a new mechanism of OAP-dependent NMO pathogenesis. Disruption of AQP4 OAPs may greatly reduce NMO-IgG dependent CDC and NMO pathology.


Assuntos
Aquaporina 4/metabolismo , Neuromielite Óptica/metabolismo , Animais , Astrócitos/citologia , Autoanticorpos/química , Células CHO , Linhagem Celular Tumoral , Ativação do Complemento , Complemento C1q/química , Proteínas do Sistema Complemento , Cricetinae , Cricetulus , Humanos , Imunoglobulina G/química , Ligação Proteica
14.
J Biol Chem ; 287(44): 36837-44, 2012 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-22989877

RESUMO

Neuromyelitis optica (NMO) is an inflammatory demyelinating disease of the central nervous system caused by binding of anti-aquaporin-4 (AQP4) autoantibodies (NMO-IgG) to AQP4 on astrocytes. A screen was developed to identify inhibitors of NMO-IgG-dependent, complement-dependent cytotoxicity. Screening of 50,000 synthetic small molecules was done using CHO cells expressing human AQP4 and a human NMO recombinant monoclonal antibody (rAb-53). The screen yielded pyrano[2,3-c]pyrazoles that blocked rAb-53 binding to AQP4 and prevented cytotoxicity in cell culture and spinal cord slice models of NMO. Structure-activity analysis of 82 analogs yielded a blocker with IC(50) ∼ 6 µm. Analysis of the blocker mechanism indicated idiotype specificity, as (i) pyrano[2,3-c]pyrazoles did not prevent AQP4 binding or cytotoxicity of other NMO-IgGs, and (ii) surface plasmon resonance showed specific rAb-53 binding. Antibody structure modeling and docking suggested a putative binding site near the complementarity-determining regions. Small molecules with idiotype-specific antibody targeting may be useful as research tools and therapeutics.


Assuntos
Aquaporina 4/imunologia , Autoanticorpos/metabolismo , Imunoglobulina G/metabolismo , Neuromielite Óptica/imunologia , Animais , Citotoxicidade Celular Dependente de Anticorpos/efeitos dos fármacos , Aquaporina 4/metabolismo , Autoanticorpos/imunologia , Sítios de Ligação , Células CHO , Cricetinae , Ensaios de Triagem em Larga Escala , Humanos , Imunoglobulina G/imunologia , Idiótipos de Imunoglobulinas/imunologia , Idiótipos de Imunoglobulinas/metabolismo , Camundongos , Camundongos Knockout , Simulação de Acoplamento Molecular , Neuromielite Óptica/metabolismo , Ligação Proteica/efeitos dos fármacos , Piranos/farmacologia , Pirazóis/farmacologia , Medula Espinal/efeitos dos fármacos , Medula Espinal/imunologia , Medula Espinal/patologia , Relação Estrutura-Atividade , Ressonância de Plasmônio de Superfície , Técnicas de Cultura de Tecidos
15.
Ann Neurol ; 71(3): 314-22, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22271321

RESUMO

OBJECTIVE: Neuromyelitis optica (NMO) is an inflammatory demyelinating disease of the central nervous system. Circulating autoantibodies (NMO-immunoglobulin [Ig]G) against astrocyte water channel aquaporin-4 (AQP4) cause complement- and cell-mediated astrocyte damage with consequent neuroinflammation and demyelination. Current NMO therapies, which have limited efficacy, include immunosuppression and plasma exchange. The objective of this study was to develop a potential new NMO therapy based on blocking of pathogenic NMO-IgG binding to its target, AQP4. METHODS: We generated nonpathogenic recombinant monoclonal anti-AQP4 antibodies that selectively block NMO-IgG binding to AQP4. These antibodies comprise a tight-binding anti-AQP4 Fab and a mutated Fc that lacks functionality for complement- and cell-mediated cytotoxicity. The efficacy of the blocking antibodies was studied using cell culture, spinal cord slice, and in vivo mouse models of NMO. RESULTS: In AQP4-expressing cell cultures, the nonpathogenic competing antibodies blocked binding of NMO-IgG in human sera, reducing to near zero complement- and cell-mediated cytotoxicity. The antibodies prevented the development of NMO lesions in an ex vivo spinal cord slice model of NMO and in an in vivo mouse model, without causing cytotoxicity. INTERPRETATION: Our results provide proof of concept for a therapy of NMO with blocking antibodies. The broad efficacy of antibody inhibition is likely due to steric competition because of its large physical size compared to AQP4. Blocker therapy to prevent binding of pathogenic autoantibodies to their targets may be useful for treatment of other autoimmune diseases as well.


Assuntos
Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais/uso terapêutico , Aquaporina 4/antagonistas & inibidores , Aquaporina 4/metabolismo , Neuromielite Óptica/tratamento farmacológico , Neuromielite Óptica/metabolismo , Animais , Animais Recém-Nascidos , Aquaporina 4/imunologia , Sítios de Ligação de Anticorpos , Ligação Competitiva/imunologia , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Humanos , Imunoglobulina G/metabolismo , Camundongos , Camundongos Knockout , Neuromielite Óptica/imunologia , Técnicas de Cultura de Órgãos , Ligação Proteica/imunologia , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/uso terapêutico
16.
Acta Neuropathol ; 125(6): 829-40, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23677375

RESUMO

Neuromyelitis optica (NMO) is an autoimmune disorder with inflammatory demyelinating lesions in the central nervous system, particularly in the spinal cord and optic nerve. NMO pathogenesis is thought to involve binding of anti-aquaporin-4 (AQP4) autoantibodies to astrocytes, which causes complement-dependent cytotoxicity (CDC) and downstream inflammation leading to oligodendrocyte and neuronal injury. Vasculocentric deposition of activated complement is a prominent feature of NMO pathology. Here, we show that a neutralizing monoclonal antibody against the C1q protein in the classical complement pathway prevents AQP4 autoantibody-dependent CDC in cell cultures and NMO lesions in ex vivo spinal cord slice cultures and in mice. A monoclonal antibody against human C1q with 11 nM binding affinity prevented CDC caused by NMO patient serum in AQP4-transfected cells and primary astrocyte cultures, and prevented complement-dependent cell-mediated cytotoxicity (CDCC) produced by natural killer cells. The anti-C1q antibody prevented astrocyte damage and demyelination in mouse spinal cord slice cultures exposed to AQP4 autoantibody and human complement. In a mouse model of NMO produced by intracerebral injection of AQP4 autoantibody and human complement, the inflammatory demyelinating lesions were greatly reduced by intracerebral administration of the anti-C1q antibody. These results provide proof-of-concept for C1q-targeted monoclonal antibody therapy in NMO. Targeting of C1q inhibits the classical complement pathway directly and causes secondary inhibition of CDCC and the alternative complement pathway. As C1q-targeted therapy leaves the lectin complement activation pathway largely intact, its side-effect profile is predicted to differ from that of therapies targeting downstream complement proteins.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Complemento C1q/antagonistas & inibidores , Fatores Imunológicos/uso terapêutico , Neuromielite Óptica/patologia , Neuromielite Óptica/prevenção & controle , Animais , Aquaporina 4/fisiologia , Técnicas de Cultura de Células , Ativação do Complemento , Cricetulus , Modelos Animais de Doenças , Humanos , Camundongos , Neuromielite Óptica/etiologia
17.
FASEB J ; 26(5): 2197-208, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22319008

RESUMO

Neuromyelitis optica (NMO) is an inflammatory demyelinating disease of spinal cord and optic nerve caused by pathogenic autoantibodies (NMO-IgG) against astrocyte aquaporin-4 (AQP4). We developed a high-throughput screen to identify blockers of NMO-IgG binding to human AQP4 using a human recombinant monoclonal NMO-IgG and transfected Fisher rat thyroid cells stably expressing human M23-AQP4. Screening of ∼60,000 compounds yielded the antiviral arbidol, the flavonoid tamarixetin, and several plant-derived berbamine alkaloids, each of which blocked NMO-IgG binding to AQP4 without affecting AQP4 expression, array assembly, or water permeability. The compounds inhibited NMO-IgG binding to AQP4 in NMO patient sera and blocked NMO-IgG-dependent complement- and cell-mediated cytotoxicity with IC(50) down to ∼5 µM. Docking computations identified putative sites of blocker binding at the extracellular surface of AQP4. The blockers did not affect complement-dependent cytotoxicity caused by anti-GD3 antibody binding to ganglioside GD3. The blockers reduced by >80% the severity of NMO lesions in an ex vivo spinal cord slice culture model of NMO and in mice in vivo. Our results provide proof of concept for a small-molecule blocker strategy to reduce NMO pathology. Small-molecule blockers may also be useful for other autoimmune diseases caused by binding of pathogenic autoantibodies to defined targets.


Assuntos
Aquaporina 4/metabolismo , Astrócitos/metabolismo , Imunoglobulina G/metabolismo , Neuromielite Óptica/patologia , Bibliotecas de Moléculas Pequenas , Animais , Camundongos , Ratos , Ratos Endogâmicos F344
18.
J Am Soc Nephrol ; 23(7): 1210-20, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22491419

RESUMO

Urea transport (UT) proteins facilitate the concentration of urine by the kidney, suggesting that inhibition of these proteins could have therapeutic use as a diuretic strategy. We screened 100,000 compounds for UT-B inhibition using an optical assay based on the hypotonic lysis of acetamide-loaded mouse erythrocytes. We identified a class of triazolothienopyrimidine UT-B inhibitors; the most potent compound, UTB(inh)-14, fully and reversibly inhibited urea transport with IC(50) values of 10 nM and 25 nM for human and mouse UT-B, respectively. UTB(inh)-14 competed with urea binding at an intracellular site on the UT-B protein. UTB(inh)-14 exhibited low toxicity and high selectivity for UT-B over UT-A isoforms. After intraperitoneal administration of UTB(inh)-14 in mice to achieve predicted therapeutic concentrations in the kidney, urine osmolality after administration of 1-deamino-8-D-arginine-vasopressin was approximately 700 mosm/kg H(2)O lower in UTB(inh)-14-treated mice than vehicle-treated mice. UTB(inh)-14 also increased urine output and reduced urine osmolality in mice given free access to water. UTB(inh)-14 did not reduce urine osmolality in UT-B knockout mice. In summary, these data provide proof of concept for the potential utility of UT inhibitors to reduce urinary concentration in high-vasopressin, fluid-retaining conditions. The diuretic mechanism of UT inhibitors may complement the action of conventional diuretics, which target sodium transport.


Assuntos
Proteínas de Transporte/antagonistas & inibidores , Capacidade de Concentração Renal/fisiologia , Rim/fisiologia , Proteínas de Membrana Transportadoras/fisiologia , Fenômenos Fisiológicos do Sistema Urinário , Animais , Aquaporina 1/deficiência , Aquaporina 1/genética , Aquaporina 1/fisiologia , Simulação por Computador , Ensaios de Triagem em Larga Escala , Humanos , Masculino , Proteínas de Membrana Transportadoras/deficiência , Proteínas de Membrana Transportadoras/genética , Camundongos , Camundongos Knockout , Modelos Animais , Transportadores de Ureia
19.
J Biol Chem ; 286(3): 2365-74, 2011 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-21084298

RESUMO

TMEM16A (ANO1) functions as a calcium-activated chloride channel (CaCC). We developed pharmacological tools to investigate the contribution of TMEM16A to CaCC conductance in human airway and intestinal epithelial cells. A screen of ∼110,000 compounds revealed four novel chemical classes of small molecule TMEM16A inhibitors that fully blocked TMEM16A chloride current with an IC(50) < 10 µM, without interfering with calcium signaling. Following structure-activity analysis, the most potent inhibitor, an aminophenylthiazole (T16A(inh)-A01), had an IC(50) of ∼1 µM. Two distinct types of inhibitors were identified. Some compounds, such as tannic acid and the arylaminothiophene CaCC(inh)-A01, fully inhibited CaCC current in human bronchial and intestinal cells. Other compounds, including T16A(inh)-A01 and digallic acid, inhibited total CaCC current in these cells poorly, but blocked mainly an initial, agonist-stimulated transient chloride current. TMEM16A RNAi knockdown also inhibited mainly the transient chloride current. In contrast to the airway and intestinal cells, all TMEM16A inhibitors fully blocked CaCC current in salivary gland cells. We conclude that TMEM16A carries nearly all CaCC current in salivary gland epithelium, but is a minor contributor to total CaCC current in airway and intestinal epithelia. The small molecule inhibitors identified here permit pharmacological dissection of TMEM16A/CaCC function and are potential development candidates for drug therapy of hypertension, pain, diarrhea, and excessive mucus production.


Assuntos
Canais de Cloreto/metabolismo , Células Epiteliais/metabolismo , Mucosa Intestinal/metabolismo , Proteínas de Membrana/antagonistas & inibidores , Moduladores de Transporte de Membrana/farmacologia , Proteínas de Neoplasias/antagonistas & inibidores , Mucosa Respiratória/metabolismo , Anoctamina-1 , Linhagem Celular , Cloretos/metabolismo , Diarreia/tratamento farmacológico , Diarreia/metabolismo , Avaliação Pré-Clínica de Medicamentos/métodos , Humanos , Hipertensão/tratamento farmacológico , Hipertensão/metabolismo , Transporte de Íons/efeitos dos fármacos , Proteínas de Membrana/metabolismo , Moduladores de Transporte de Membrana/química , Proteínas de Neoplasias/metabolismo , Dor/tratamento farmacológico , Dor/metabolismo , Glândulas Salivares/metabolismo
20.
Bioorg Med Chem Lett ; 22(4): 1602-5, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-22281185

RESUMO

(19)F-modified bithiazole correctors and phenylglycine potentiators of the ΔF508-CFTR chloride channel were synthesized and their function assayed in cells expressing human ΔF508-CFTR and a halide-sensitive fluorescent protein. Fluorine was incorporated into each scaffold using prosthetic groups for future biodistribution imaging studies using positron emission tomography (PET). The ΔF508-CFTR corrector and potentiator potencies of the fluorinated analogs were comparable to or better than those of the original compounds.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/química , Radioisótopos de Flúor , Tomografia por Emissão de Pósitrons/métodos , Humanos , Estrutura Molecular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA