Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 569
Filtrar
1.
Sci Rep ; 11(1): 21962, 2021 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-34753987

RESUMO

Neuromyelitis optica spectrum disorder (NMOSD) is an autoimmune inflammatory disease of the central nervous system. Most NMOSD patients are seropositive for immunoglobulin G (IgG) autoantibodies against astrocyte water channel aquaporin-4 (AQP4), called AQP4-IgG. AQP4-IgG binding to aquaporin-4 causes complement-dependent cytotoxicity (CDC), leading to inflammation and demyelination. Here, CDC was measured in AQP4-expressing cells exposed to human complement and heat-inactivated sera from 108 AQP4-IgG seropositive NMOSD subjects and 25 non-NMOSD controls. AQP4-IgG positive sera produced a wide range of CDC, with 50% maximum cytotoxicity produced by as low as 0.2% serum concentration. Unexpectedly, 58 samples produced no cytotoxicity, and of those, four sera were cytoprotective against cytotoxic AQP4-IgG. Cytoprotection was found against different cytotoxic monoclonal AQP4-IgGs and NMOSD patient sera, and in primary astrocyte cultures. Mechanistic studies revealed that the protective factor is an IgG antibody that did not inhibit complement directly, but interfered with binding of cytotoxic AQP4-IgG to AQP4 and consequent C1q binding and complement activation. Further studies suggested that non-pathogenic AQP4-IgG, perhaps with altered glycosylation, may contribute to reduced or ineffectual binding of cytotoxic AQP4-IgG, as well as reduced cell-surface AQP4. The presence of natural cytoprotective antibodies in AQP4-IgG seropositive sera reveals an added level of complexity in NMOSD disease pathogenesis, and suggests the potential therapeutic utility of 'convalescent' serum or engineered protective antibody to interfere with pathogenic antibody in AQP4-IgG seropositive NMOSD.


Assuntos
Aquaporina 4/imunologia , Neuromielite Óptica/imunologia , Animais , Aquaporina 4/sangue , Autoanticorpos/sangue , Autoanticorpos/imunologia , Biomarcadores/sangue , Células CHO , Cricetulus , Progressão da Doença , Humanos , Soros Imunes , Imunoglobulina G/sangue , Neuromielite Óptica/sangue , Neuromielite Óptica/patologia
2.
Neuropharmacology ; 133: 345-353, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29428821

RESUMO

Intravenous human immunoglobulin G (IVIG) may have therapeutic benefit in neuromyelitis optica spectrum disorders (herein called NMO), in part because of the anti-inflammatory properties of the IgG Fc region. Here, we evaluated recombinant Fc hexamers consisting of the IgM µ-tailpiece fused with the Fc region of human IgG1. In vitro, the Fc hexamers prevented cytotoxicity in aquaporin-4 (AQP4) expressing cells and in rat spinal cord slice cultures exposed to NMO anti-AQP4 autoantibody (AQP4-IgG) and complement, with >500-fold greater potency than IVIG or monomeric Fc fragments. Fc hexamers at low concentration also prevented antibody-dependent cellular cytotoxicity produced by AQP4-IgG and natural killer cells. Serum from rats administered a single intravenous dose of Fc hexamers at 50 mg/kg taken at 8 h did not produce complement-dependent cytotoxicity when added to AQP4-IgG-treated AQP4-expressing cell cultures. In an experimental rat model of NMO produced by intracerebral injection of AQP4-IgG, Fc hexamers at 50 mg/kg administered before and at 12 h after AQP4-IgG fully prevented astrocyte injury, complement activation, inflammation and demyelination. These results support the potential therapeutic utility of recombinant IgG1 Fc hexamers in AQP4-IgG seropositive NMO.


Assuntos
Fragmentos Fc das Imunoglobulinas/uso terapêutico , Imunoglobulina G/uso terapêutico , Neuromielite Óptica/terapia , Administração Intravenosa , Animais , Aquaporina 4/genética , Aquaporina 4/imunologia , Aquaporina 4/metabolismo , Aquaporina 4/toxicidade , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Astrócitos/patologia , Autoanticorpos/uso terapêutico , Células CHO , Complemento C1q/metabolismo , Cricetulus , Nucleotídeos de Desoxiuracil/imunologia , Nucleotídeos de Desoxiuracil/metabolismo , Nucleotídeos de Desoxiuracil/uso terapêutico , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Fragmentos Fc das Imunoglobulinas/genética , Imunoglobulina G/química , Técnicas In Vitro , Mutação/genética , Neuromielite Óptica/imunologia , Neuromielite Óptica/patologia , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Medula Espinal/patologia , Estatísticas não Paramétricas , Fatores de Tempo , Transfecção
3.
Lab Chip ; 17(5): 887-895, 2017 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-28184395

RESUMO

Water transport across epithelial monolayers is of central importance in mammalian fluid homeostasis, and epithelial aquaporin (AQP) water channels are potential drug targets. Current methods to measure transepithelial water permeability based on indicator dilution have limited accuracy and can require hours for a single measurement. We report here a microfluidics platform for rapid and accurate measurement of water transport across a conventionally cultured epithelial monolayer on a porous filter requiring only a single image obtained using a standard laboratory fluorescence microscope. The undersurface of a porous polyester filter containing cultured epithelial cells on top is contacted with a perfused microfluidic channel of 100 µm width, 20 µm height and 10 cm length with folded geometry, with in-plane size of 3.2 × 3.2 mm2 for visualization with a 2× objective lens. Osmotic water permeability is measured from the steady-state concentration profile along the length of the channel of a membrane-impermeant fluorescent dye in the perfusate, in which an osmotic gradient is imposed by an anisosmolar solution overlying the epithelial monolayer; diffusional water permeability is measured using a D2O/H2O-sensing fluorescent dye in the perfusate with a D2O-containing isosmolar solution overlying the cell layer. Permeability values are deduced from single fluorescence images. The method, named fluid transport on a chip (FT-on-Chip), was applied to measure transepithelial osmotic and diffusional water permeability in control and AQP4-expressing epithelial cell monolayers. FT-on-Chip allows for rapid, accurate and repeated measurements of transepithelial water permeability, and is generalizable to transport measurements of ions and solutes using suitable indicator dyes.


Assuntos
Permeabilidade da Membrana Celular/fisiologia , Células Epiteliais/metabolismo , Técnicas Analíticas Microfluídicas/instrumentação , Água/análise , Água/metabolismo , Animais , Aquaporina 4/metabolismo , Linhagem Celular , Células Epiteliais/citologia , Desenho de Equipamento , Corantes Fluorescentes/análise , Corantes Fluorescentes/metabolismo , Dispositivos Lab-On-A-Chip , Técnicas Analíticas Microfluídicas/métodos , Osmose , Ratos
4.
Acta Neuropathol Commun ; 4(1): 42, 2016 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-27117475

RESUMO

Neuromyelitis optica (NMO) is an inflammatory demyelinating disease of the central nervous system that can produce marked neurological deficit. Current NMO therapies include immunosuppressants, plasma exchange and B-cell depletion. Here, we evaluated 14 potential remyelinating drugs emerging from prior small molecule screens done to identify drugs for repurposing in multiple sclerosis and other demyelinating neurological diseases. Compounds were initially evaluated in oligodendrocyte precursor cell (OPC) and cerebellar slice cultures, and then in a mouse model of NMO produced by intracerebral injection of anti-AQP4 autoantibody (AQP4-IgG) and human complement characterized by demyelination with minimal axonal damage. The FDA-approved drug clobetasol promoted differentiation in OPC cultures and remyelination in cerebellar slice cultures and in mice. Intraperitoneal administration of 2 mg/kg/day clobetasol reduced myelin loss by ~60 %, even when clobetasol was administered after demyelination occurred. Clobetasol increased the number of mature oligodendrocytes within lesions without significantly altering initial astrocyte damage or inflammation. These results provide proof-of-concept for the potential utility of a remyelinating approach in the treatment of NMO.


Assuntos
Clobetasol/farmacologia , Bainha de Mielina/efeitos dos fármacos , Neuromielite Óptica/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/patologia , Astrócitos/fisiologia , Células Cultivadas , Cerebelo/efeitos dos fármacos , Cerebelo/patologia , Cerebelo/fisiopatologia , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Masculino , Camundongos , Bainha de Mielina/patologia , Bainha de Mielina/fisiologia , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/patologia , Células-Tronco Neurais/fisiologia , Neurogênese/efeitos dos fármacos , Neurogênese/fisiologia , Neuromielite Óptica/patologia , Neuromielite Óptica/fisiopatologia , Técnicas de Cultura de Tecidos
5.
Cell Mol Gastroenterol Hepatol ; 2(3): 317-327, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27127798

RESUMO

BACKGROUND & AIMS: Constipation is a common clinical problem that negatively impacts quality of life and is associated with significant health care costs. Activation of the cystic fibrosis transmembrane regulator (CFTR) chloride channel is the primary pathway that drives fluid secretion in the intestine, which maintains lubrication of luminal contents. We hypothesized that direct activation of CFTR would cause fluid secretion and reverse the excessive dehydration of stool found in constipation. METHODS: A cell-based high-throughput screen was done for 120,000 drug-like, synthetic small molecules. Active compounds were characterized for mechanism of action and one lead compound was tested in a loperamide-induced constipation model in mice. RESULTS: Several classes of novel CFTR activators were identified, one of which, the phenylquinoxalinone CFTRact-J027, fully activated CFTR chloride conductance with EC50 ~ 200 nM, without causing elevation of cytoplasmic cAMP. Orally administered CFTRact-J027 normalized stool output and water content in a loperamide-induced mouse model of constipation with ED50 ~0.5 mg/kg; CFTRact-J027 was without effect in cystic fibrosis mice lacking functional CFTR. Short-circuit current, fluid secretion and motility measurements in mouse intestine indicated a pro-secretory action of CFTRact-J027 without direct stimulation of intestinal motility. Oral administration of 10 mg/kg CFTRact-J027 showed minimal bioavailability, rapid hepatic metabolism and blood levels <200 nM, and without apparent toxicity after chronic administration. CONCLUSIONS: CFTRact-J027 or alternative small-molecule CFTR-targeted activators may be efficacious for the treatment of constipation.

6.
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
7.
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
8.
FASEB J ; 30(5): 1789-97, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26842854

RESUMO

Dry eye disorders, including Sjögren's syndrome, constitute a common problem in the aging population, with limited effective therapeutic options available. The cAMP-activated Cl(-) channel cystic fibrosis transmembrane conductance regulator (CFTR) is a major prosecretory channel at the ocular surface. We investigated whether compounds that target CFTR can correct the abnormal tear film in dry eye. Small-molecule activators of human wild-type CFTR identified by high-throughput screening were evaluated in cell culture and in vivo assays, to select compounds that stimulate Cl(-)-driven fluid secretion across the ocular surface in mice. An aminophenyl-1,3,5-triazine, CFTRact-K089, fully activated CFTR in cell cultures with EC50 ∼250 nM and produced an ∼8.5 mV hyperpolarization in ocular surface potential difference. When delivered topically, CFTRact-K089 doubled basal tear volume for 4 h and had no effect in CF mice. CFTRact-K089 showed sustained tear film bioavailability without detectable systemic absorption. In a mouse model of aqueous-deficient dry eye produced by lacrimal ablation, topical administration of 0.1 nmol CFTRact-K089 3 times daily restored tear volume to basal levels, preventing corneal epithelial disruption when initiated at the time of surgery and reversing it when started after development of dry eye. Our results support the potential utility of CFTR-targeted activators as a novel prosecretory treatment for dry eye.-Flores, A. M., Casey, S. D., Felix, C. M., Phuan, P. W., Verkman, A. S., Levin, M. H. Small-molecule CFTR activators increase tear secretion and prevent experimental dry eye disease.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/agonistas , Síndromes do Olho Seco/prevenção & controle , Regulação da Expressão Gênica/efeitos dos fármacos , Lágrimas/metabolismo , Animais , Células Cultivadas , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Estrutura Molecular , Mutação , Ratos , Ratos Endogâmicos F344
9.
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
10.
Lab Chip ; 15(16): 3380-90, 2015 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-26159099

RESUMO

Cell membrane water permeability is an important determinant of epithelial fluid secretion, tissue swelling, angiogenesis, tumor spread and other biological processes. Cellular water channels, aquaporins, are important drug targets. Water permeability is generally measured from the kinetics of cell volume change in response to an osmotic gradient. Here, we developed a microfluidic platform in which cells expressing a cytoplasmic, volume-sensing fluorescent dye are rapidly subjected to an osmotic gradient by solution mixing inside a ~0.1 nL droplet surrounded by oil. The solution mixing time was <10 ms. Osmotic water permeability was deduced from a single, time-integrated fluorescence image of an observation area in which the time after mixing was determined through spatial position. Water permeability was accurately measured in aquaporin-expressing erythrocytes with half-times for osmotic equilibration down to <50 ms. Compared with conventional water permeability measurements using costly stopped-flow instrumentation, the microfluidic platform here utilizes sub-microliter blood sample volume, does not suffer from mixing artifacts, and replaces challenging kinetic measurements by single image capture using a standard laboratory fluorescence microscope.


Assuntos
Eritrócitos/metabolismo , Técnicas Analíticas Microfluídicas/métodos , Água/metabolismo , Animais , Aquaporinas/deficiência , Aquaporinas/genética , Permeabilidade da Membrana Celular , Eritrócitos/química , Eritrócitos/citologia , Fluoresceínas/química , Proteínas de Membrana Transportadoras/deficiência , Proteínas de Membrana Transportadoras/genética , Camundongos , Camundongos Knockout , Técnicas Analíticas Microfluídicas/instrumentação , Microscopia de Fluorescência , Óleos/química , Água/química
11.
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.

12.
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
13.
Nat Commun ; 6: 7454, 2015 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-26100668

RESUMO

Aquaporin 3 (AQP3), a water/glycerol channel protein, has been found to transport hydrogen peroxide (H2O2). Here, we show that H2O2, imported via AQP3, is involved in nuclear factor-κB (NF-κB) signalling in keratinocytes and in the pathogenesis of psoriasis. IL-23-mediated induction of psoriasis is reduced in AQP3 knockout mice (AQP3(-/-)), and is accompanied by impaired NF-κB activation and intracellular H2O2 accumulation. In primary keratinocyte cultures, cellular import of H2O2 produced by membrane NADPH oxidase 2 (Nox2) in response to TNF-α is facilitated by AQP3 and required for NF-κB activation by regulation of protein phosphatase 2A. As AQP3 associates with Nox2, we propose that this interplay constitutes H2O2-mediated signalling in response to TNF-α stimulation. Collectively, these data indicate that AQP3-facilitated H2O2 transport is required for NF-κB activation in keratinocytes in the development of psoriasis.


Assuntos
Aquaporina 3/genética , Peróxido de Hidrogênio/metabolismo , Queratinócitos/metabolismo , NF-kappa B/metabolismo , Psoríase/genética , Animais , Aquaporina 3/metabolismo , Estudos de Casos e Controles , Células Cultivadas , Humanos , Immunoblotting , Imuno-Histoquímica , Imunoprecipitação , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , NADPH Oxidase 2 , NADPH Oxidases/metabolismo , Psoríase/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo
14.
Glia ; 63(10): 1860-9, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25944186

RESUMO

The astrocyte water channel aquaporin-4 (AQP4) regulates extracellular space (ECS) K(+) concentration ([K(+)]e) and volume dynamics following neuronal activation. Here, we investigated how AQP4-mediated changes in [K(+)]e and ECS volume affect the velocity, frequency, and amplitude of cortical spreading depression (CSD) depolarizations produced by surface KCl application in wild-type (AQP4(+/+)) and AQP4-deficient (AQP4(-/-)) mice. In contrast to initial expectations, both the velocity and the frequency of CSD were significantly reduced in AQP4(-/-) mice when compared with AQP4(+/+) mice, by 22% and 32%, respectively. Measurement of [K(+)]e with K(+)-selective microelectrodes demonstrated an increase to ∼35 mM during spreading depolarizations in both AQP4(+/+) and AQP4(-/-) mice, but the rates of [K(+)]e increase (3.5 vs. 1.5 mM/s) and reuptake (t1/2 33 vs. 61 s) were significantly reduced in AQP4(-/-) mice. ECS volume fraction measured by tetramethylammonium iontophoresis was greatly reduced during depolarizations from 0.18 to 0.053 in AQP4(+/+) mice, and 0.23 to 0.063 in AQP4(-/-) mice. Analysis of the experimental data using a mathematical model of CSD propagation suggested that the reduced velocity of CSD depolarizations in AQP4(-/-) mice was primarily a consequence of the slowed increase in [K(+)]e during neuronal depolarization. These results demonstrate that AQP4 effects on [K(+)]e and ECS volume dynamics accelerate CSD propagation.


Assuntos
Aquaporina 4/metabolismo , Encéfalo/fisiologia , Depressão Alastrante da Atividade Elétrica Cortical/genética , Camundongos Transgênicos/metabolismo , Análise de Variância , Animais , Aquaporina 4/genética , Fracionamento Celular , Depressão Alastrante da Atividade Elétrica Cortical/efeitos dos fármacos , Estimulação Elétrica , Espaço Extracelular/metabolismo , Eletrodos Seletivos de Íons , Camundongos , Camundongos Transgênicos/genética , Modelos Teóricos , Potássio/farmacologia
15.
FASEB J ; 29(4): 1551-63, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25573755

RESUMO

Water channel aquaporin-1 (AQP1) is expressed at epithelial cell plasma membranes in renal proximal tubules and thin descending limb of Henle. Recently, AQP1 was reported to interact with ß-catenin. Here we investigated the relationship between AQP1 and Wnt signaling in in vitro and in vivo models of autosomal dominant polycystic kidney disease (PKD). AQP1 overexpression decreased ß-catenin and cyclinD1 expression, suggesting down-regulation of Wnt signaling, and coimmunoprecipitation showed AQP1 interaction with ß-catenin, glycogen synthase kinase 3ß, LRP6, and Axin1. AQP1 inhibited cyst development and promoted branching in matrix-grown MDCK cells. In embryonic kidney cultures, AQP1 deletion increased cyst development by up to ∼ 40%. Kidney size and cyst number were significantly greater in AQP1-null PKD mice than in AQP1-expressing PKD mice, with the difference mainly attributed to a greater number of proximal tubule cysts. Biochemical analysis revealed decreased ß-catenin phosphorylation and increased ß-catenin expression in AQP1-null PKD mice, suggesting enhanced Wnt signaling. These results implicate AQP1 as a novel determinant in renal cyst development that may involve inhibition of Wnt signaling by an AQP1-macromolecular signaling complex.


Assuntos
Aquaporina 1/metabolismo , Rim Policístico Autossômico Dominante/metabolismo , Rim Policístico Autossômico Dominante/patologia , Via de Sinalização Wnt , Animais , Aquaporina 1/deficiência , Aquaporina 1/genética , Adesão Celular , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Cães , Rim/embriologia , Rim/metabolismo , Rim/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Biológicos , Complexos Multiproteicos/metabolismo , Rim Policístico Autossômico Dominante/genética , beta Catenina/metabolismo
16.
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
17.
Exp Neurol ; 265: 152-9, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25585012

RESUMO

Dorsal root ganglion (DRG) neurons transduce peripheral pain signals through small-diameter, non-myelinated C-fibers, which, when injured, can regenerate to restore pain sensation. Water channel aquaporin-1 (AQP1) is expressed at the plasma membrane of cell bodies and axons of DRG neurons, where it modulates the sensing of certain types of pain. Here, we found that AQP1 is also involved in DRG axonal growth and regeneration by a mechanism that may involve water transport-facilitated extension of axonal outgrowths. Spontaneous and nerve growth factor-stimulated axonal extension was reduced in cultures of AQP1-deficient DRG neurons and DRG explants compared to the wildtype. Axonal growth in AQP1-deficient DRG cultures was rescued by transfection with AQP1 or a different water-transporting AQP (AQP4), but not by a non-water-transporting AQP1 mutant. Following sciatic nerve compression injury AQP1 expression was increased in DRG neurons in wildtype mice, and DRG axonal growth was impaired in AQP1-deficient mice. Our results indicate AQP1 as a novel determinant of DRG axonal regeneration and hence a potential therapeutic target to accelerate neuronal regeneration.


Assuntos
Aquaporina 1/metabolismo , Axônios/metabolismo , Gânglios Espinais/metabolismo , Regeneração Nervosa/fisiologia , Neurônios/metabolismo , Água/metabolismo , Animais , Células Cultivadas , Gânglios Espinais/citologia , Camundongos , Camundongos Knockout , Permeabilidade
18.
Neurosci Lett ; 584: 368-72, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25449874

RESUMO

Aquaporin-4 (AQP4) is a water channel expressed in astrocyte end-feet lining the blood-brain barrier. AQP4 deletion in mice is associated with improved outcomes in global cerebral ischemia produced by transient carotid artery occlusion, and focal cerebral ischemia produced by permanent middle cerebral artery occlusion (MCAO). Here, we investigated the consequences of 1-h transient MCAO produced by intraluminal suture blockade followed by 23 h of reperfusion. In nine AQP4(+/+) and nine AQP4(-/-) mice, infarct volume was significantly reduced by an average of 39 ± 4% at 24h in AQP4(-/-) mice, cerebral hemispheric edema was reduced by 23 ± 3%, and Evans Blue extravasation was reduced by 31 ± 2% (mean ± SEM). Diffusion-weighted magnetic resonance imaging showed greatest reduction in apparent diffusion coefficient around the occlusion site after reperfusion, with remarkably lesser reduction in AQP4(-/-) mice. The reduced infarct volume in AQP4(-/-) mice following transient MCAO supports the potential utility of therapeutic AQP4 inhibition in stroke.


Assuntos
Aquaporina 4/genética , Edema Encefálico/patologia , Infarto Encefálico/patologia , Ataque Isquêmico Transitório/patologia , Animais , Edema Encefálico/metabolismo , Infarto Encefálico/metabolismo , Imagem de Difusão por Ressonância Magnética , Ataque Isquêmico Transitório/metabolismo , Masculino , Camundongos Knockout
19.
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
20.
PLoS One ; 9(9): e106824, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25191939

RESUMO

Neuromyelitis optica (NMO) is an autoimmune demyelinating disease of the central nervous system in which binding of anti-aquaporin-4 (AQP4) autoantibodies (NMO-IgG) to astrocytes causes complement-dependent cytotoxicity (CDC) and inflammation resulting in oligodendrocyte and neuronal injury. There is compelling evidence for a central role of complement in NMO pathogenesis. Here, we evaluated the potential of C1-esterase inhibitor (C1-inh) for complement-targeted therapy of NMO. C1-inh is an anti-inflammatory plasma protein with serine protease inhibition activity that has a broad range of biological activities on the contact (kallikrein), coagulation, fibrinolytic and complement systems. C1-inh is approved for therapy of hereditary angioedema (HAE) and has been studied in a small safety trial in acute NMO relapses (NCT 01759602). In vitro assays of NMO-IgG-dependent CDC showed C1-inh inhibition of human and rat complement, but with predicted minimal complement inhibition activity at a dose of 2000 units in humans. Inhibition of complement by C1-inh was potentiated by ∼10-fold by polysulfated macromolecules including heparin and dextran sulfate. In rats, intravenous C1-inh at a dose 30-fold greater than that approved to treat HAE inhibited serum complement activity by <5%, even when supplemented with heparin. Also, high-dose C1-inh did not reduce pathology in a rat model of NMO produced by intracerebral injection of NMO-IgG. Therefore, although C1r and C1s are targets of C1-inh, our in vitro data with human serum and in vivo data in rats suggest that the complement inhibition activity of C1-inh in serum is too low to confer clinical benefit in NMO.


Assuntos
Aquaporina 4/imunologia , Astrócitos/imunologia , Proteína Inibidora do Complemento C1/administração & dosagem , Complemento C1/antagonistas & inibidores , Neuromielite Óptica/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Autoanticorpos/sangue , Células CHO , Proteína Inibidora do Complemento C1/farmacologia , Cricetulus , Sulfato de Dextrana/farmacologia , Modelos Animais de Doenças , Sinergismo Farmacológico , Heparina/farmacologia , Humanos , Imunoglobulina G/efeitos adversos , Imunoglobulina G/imunologia , Técnicas In Vitro , Terapia de Alvo Molecular , Neuromielite Óptica/sangue , Neuromielite Óptica/patologia , Ratos , Ratos Endogâmicos Lew
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...