Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
J Biol Chem ; 292(43): 17963-17974, 2017 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-28860188

RESUMEN

Aberrant activation of matrix metalloproteinases (MMPs) is a common feature of pathological cascades observed in diverse disorders, such as cancer, fibrosis, immune dysregulation, and neurodegenerative diseases. MMP-9, in particular, is highly dynamically regulated in several pathological processes. Development of MMP inhibitors has therefore been an attractive strategy for therapeutic intervention. However, a long history of failed clinical trials has demonstrated that broad-spectrum MMP inhibitors have limited clinical utility, which has spurred the development of inhibitors selective for individual MMPs. Attaining selectivity has been technically challenging because of sequence and structural conservation across the various MMPs. Here, through a biochemical and structural screening paradigm, we have identified JNJ0966, a highly selective compound that inhibited activation of MMP-9 zymogen and subsequent generation of catalytically active enzyme. JNJ0966 had no effect on MMP-1, MMP-2, MMP-3, MMP-9, or MMP-14 catalytic activity and did not inhibit activation of the highly related MMP-2 zymogen. The molecular basis for this activity was characterized as an interaction of JNJ0966 with a structural pocket in proximity to the MMP-9 zymogen cleavage site near Arg-106, which is distinct from the catalytic domain. JNJ0966 was efficacious in reducing disease severity in a mouse experimental autoimmune encephalomyelitis model, demonstrating the viability of this therapeutic approach. This discovery reveals an unprecedented pharmacological approach to MMP inhibition, providing an opportunity to improve selectivity of future clinical drug candidates. Targeting zymogen activation in this manner may also allow for pharmaceutical exploration of other enzymes previously viewed as intractable drug targets.


Asunto(s)
Precursores Enzimáticos/antagonistas & inhibidores , Precursores Enzimáticos/química , Metaloproteinasa 9 de la Matriz/química , Inhibidores de la Metaloproteinasa de la Matriz/química , Regulación Alostérica , Animales , Células COS , Dominio Catalítico , Chlorocebus aethiops , Precursores Enzimáticos/genética , Precursores Enzimáticos/metabolismo , Humanos , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Dominios Proteicos
2.
Eur J Neurosci ; 39(7): 1225-33, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24713001

RESUMEN

Amyloid beta (Aß), a key component in the pathophysiology of Alzheimer's disease, is thought to target excitatory synapses early in the disease. However, the mechanism by which Aß weakens synapses is not well understood. Here we showed that the PDZ domain protein, protein interacting with C kinase 1 (PICK1), was required for Aß to weaken synapses. In mice lacking PICK1, elevations of Aß failed to depress synaptic transmission in cultured brain slices. In dissociated cultured neurons, Aß failed to reduce surface α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor subunit 2, a subunit of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors that binds with PICK1 through a PDZ ligand-domain interaction. Lastly, a novel small molecule (BIO922) discovered through structure-based drug design that targets the specific interactions between GluA2 and PICK1 blocked the effects of Aß on synapses and surface receptors. We concluded that GluA2-PICK1 interactions are a key component of the effects of Aß on synapses.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Proteínas Portadoras/metabolismo , Potenciales Postsinápticos Excitadores , Proteínas Nucleares/metabolismo , Fragmentos de Péptidos/toxicidad , Sinapsis/metabolismo , Animales , Proteínas Portadoras/genética , Proteínas de Ciclo Celular , Células Cultivadas , Hipocampo/citología , Hipocampo/metabolismo , Hipocampo/fisiología , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/fisiología , Proteínas Nucleares/genética , Unión Proteica , Ratas , Receptores AMPA/metabolismo , Sinapsis/efectos de los fármacos , Sinapsis/fisiología
3.
J Pharmacol Exp Ther ; 341(1): 274-84, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22267202

RESUMEN

Oxidative stress is central to the pathology of several neurodegenerative diseases, including multiple sclerosis, and therapeutics designed to enhance antioxidant potential could have clinical value. The objective of this study was to characterize the potential direct neuroprotective effects of dimethyl fumarate (DMF) and its primary metabolite monomethyl fumarate (MMF) on cellular resistance to oxidative damage in primary cultures of central nervous system (CNS) cells and further explore the dependence and function of the nuclear factor (erythroid-derived 2)-like 2 (Nrf2) pathway in this process. Treatment of animals or primary cultures of CNS cells with DMF or MMF resulted in increased nuclear levels of active Nrf2, with subsequent up-regulation of canonical antioxidant target genes. DMF-dependent up-regulation of antioxidant genes in vivo was lost in mice lacking Nrf2 [Nrf2(-/-)]. DMF or MMF treatment increased cellular redox potential, glutathione, ATP levels, and mitochondrial membrane potential in a concentration-dependent manner. Treating astrocytes or neurons with DMF or MMF also significantly improved cell viability after toxic oxidative challenge in a concentration-dependent manner. This effect on viability was lost in cells that had eliminated or reduced Nrf2. These data suggest that DMF and MMF are cytoprotective for neurons and astrocytes against oxidative stress-induced cellular injury and loss, potentially via up-regulation of an Nrf2-dependent antioxidant response. These data also suggest DMF and MMF may function through improving mitochondrial function. The clinical utility of DMF in multiple sclerosis is being explored through phase III trials with BG-12, which is an oral therapeutic containing DMF as the active ingredient.


Asunto(s)
Sistema Nervioso Central/citología , Sistema Nervioso Central/metabolismo , Citoprotección/genética , Fumaratos/farmacología , Factor 2 Relacionado con NF-E2/fisiología , Neuronas/metabolismo , Estrés Oxidativo/genética , Transducción de Señal/genética , Animales , Células Cultivadas , Sistema Nervioso Central/efectos de los fármacos , Citoprotección/efectos de los fármacos , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 2 Relacionado con NF-E2/deficiencia , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
4.
Mol Neurobiol ; 59(4): 2171-2189, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35060064

RESUMEN

Stearoyl-CoA desaturase (SCD) is a potential therapeutic target for Parkinson's and related neurodegenerative diseases. SCD inhibition ameliorates neuronal toxicity caused by aberrant α-synuclein, a lipid-binding protein implicated in Parkinson's disease. Its inhibition depletes monounsaturated fatty acids, which may modulate α-synuclein conformations and membrane interactions. Herein, we characterize the pharmacokinetic and pharmacodynamic properties of YTX-7739, a clinical-stage SCD inhibitor. Administration of YTX-7739 to rats and monkeys for 15 days caused a dose-dependent increase in YTX-7739 concentrations that were well-tolerated and associated with concentration-dependent reductions in the fatty acid desaturation index (FADI), the ratio of monounsaturated to saturated fatty acids. An approximate 50% maximal reduction in the carbon-16 desaturation index was observed in the brain, with comparable responses in the plasma and skin. A study with a diet supplemented in SCD products indicates that changes in brain C16 desaturation were due to local SCD inhibition, rather than to changes in systemic fatty acids that reach the brain. Assessment of pharmacodynamic response onset and reversibility kinetics indicated that approximately 7 days of dosing were required to achieve maximal responses, which persisted for at least 2 days after cessation of dosing. YTX-7739 thus achieved sufficient concentrations in the brain to inhibit SCD and produce pharmacodynamic responses that were well-tolerated in rats and monkeys. These results provide a framework for evaluating YTX-7739 pharmacology clinically as a disease-modifying therapy to treat synucleinopathies.


Asunto(s)
Enfermedad de Parkinson , Estearoil-CoA Desaturasa , Animales , Ácidos Grasos/metabolismo , Ácidos Grasos/farmacología , Metabolismo de los Lípidos/fisiología , Ratas , Estearoil-CoA Desaturasa/metabolismo , alfa-Sinucleína/metabolismo
5.
PLoS One ; 17(12): e0277532, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36454869

RESUMEN

There are currently no preventive or disease-modifying therapies for Parkinson's Disease (PD). Failures in clinical trials necessitate a re-evaluation of existing pre-clinical models in order to adopt systems that better recapitulate underlying disease mechanisms and better predict clinical outcomes. In recent years, models utilizing patient-derived induced pluripotent stem cells (iPSC) have emerged as attractive models to recapitulate disease-relevant neuropathology in vitro without exogenous overexpression of disease-related pathologic proteins. Here, we utilized iPSC derived from patients with early-onset PD and dementia phenotypes that harbored either a point mutation (A53T) or multiplication at the α-synuclein/SNCA gene locus. We generated a three-dimensional (3D) cortical neurosphere culture model to better mimic the tissue microenvironment of the brain. We extensively characterized the differentiation process using quantitative PCR, Western immunoblotting and immunofluorescence staining. Differentiated and aged neurospheres revealed alterations in fatty acid profiles and elevated total and pathogenic phospho-α-synuclein levels in both A53T and the triplication lines compared to their isogenic control lines. Furthermore, treatment of the neurospheres with a small molecule inhibitor of stearoyl CoA desaturase (SCD) attenuated the protein accumulation and aberrant fatty acid profile phenotypes. Our findings suggest that the 3D cortical neurosphere model is a useful tool to interrogate targets for PD and amenable to test small molecule therapeutics.


Asunto(s)
Células Madre Pluripotentes Inducidas , Enfermedad de Parkinson , Humanos , alfa-Sinucleína/genética , Enfermedad de Parkinson/genética , Organoides , Ácidos Grasos
6.
Neurotherapeutics ; 19(3): 1018-1036, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35445353

RESUMEN

Increasing evidence has shown that Parkinson's disease (PD) impairs midbrain dopaminergic, cortical and other neuronal subtypes in large part due to the build-up of lipid- and vesicle-rich α-synuclein (αSyn) cytotoxic inclusions. We previously identified stearoyl-CoA desaturase (SCD) as a potential therapeutic target for synucleinopathies. A brain-penetrant SCD inhibitor, YTX-7739, was developed and has entered Phase 1 clinical trials. Here, we report the efficacy of YTX-7739 in reversing pathological αSyn phenotypes in various in vitro and in vivo PD models. In cell-based assays, YTX-7739 decreased αSyn-mediated neuronal death, reversed the abnormal membrane interaction of amplified E46K ("3K") αSyn, and prevented pathological phenotypes in A53T and αSyn triplication patient-derived neurospheres, including dysregulated fatty acid profiles and pS129 αSyn accumulation. In 3K PD-like mice, YTX-7739 crossed the blood-brain barrier, decreased unsaturated fatty acids, and prevented progressive motor deficits. Both YTX-7739 treatment and decreasing SCD activity through deletion of one copy of the SCD1 gene (SKO) restored the physiological αSyn tetramer-to-monomer ratio, dopaminergic integrity, and neuronal survival in 3K αSyn mice. YTX-7739 efficiently reduced pS129 + and PK-resistant αSyn in both human wild-type αSyn and 3K mutant mice similar to the level of 3K-SKO. Together, these data provide further validation of SCD as a PD therapeutic target and YTX-7739 as a clinical candidate for treating human α-synucleinopathies.


Asunto(s)
Enfermedad de Parkinson , alfa-Sinucleína , Animales , Encéfalo/metabolismo , Humanos , Ratones , Neuronas/metabolismo , Enfermedad de Parkinson/genética , Estearoil-CoA Desaturasa/genética , Estearoil-CoA Desaturasa/metabolismo , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
7.
Bioorg Med Chem Lett ; 21(21): 6485-90, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-21925881

RESUMEN

The discovery of potent N-hydroxyl caprolactam matrix metalloproteinase (MMP) inhibitors (6) based on the natural product Cobactin-T (2) is described. The synthetic method, which utilizes the ring closing metathesis reaction, is compatible to provide complementary (R) and (S) enantiomers. These compounds tested against MMP-2 and 9, show that the R stereochemistry (i.e., 16), which is opposite for that found in the natural product Cobactin-T is >1000-fold more active with IC(50) values of 0.2-0.6nM against both enzymes. The variation in the incorporation of the sulfonamide enzyme recognition element (Ar(2)XAr(1)SO(2)N(R(1)), 6), along with alterations in the RCM/double bond chemistry (R(2)) provided a series of sub nanomolar MMP inhibitors. For example, compounds 16 and 34 were found to be the most potent with IC(50) values against MMP-2 and MMP-9 found to be between 0.2 and 0.6nM with 34 being the most potent compound discovered (MMP-2 IC(50)=0.39nM and MMP-9 IC(50)=0.22nM). Compounds 16 and 34 showed acceptable drug-like properties in vivo with compound 34 showing oral bioavailability.


Asunto(s)
Azepinas/farmacología , Inhibidores de la Metaloproteinasa de la Matriz , Inhibidores de Proteasas/farmacología , Azepinas/farmacocinética , Disponibilidad Biológica , Ciclización , Descubrimiento de Drogas , Concentración 50 Inhibidora , Inhibidores de Proteasas/farmacocinética , Estereoisomerismo
8.
Bioorg Med Chem Lett ; 21(24): 7277-80, 2011 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-22061640

RESUMEN

Starting from literature examples of nonsteroidal anti-inflammatory drugs (NSAIDs)-type carboxylic acid γ-secretase modulators (GSMs) and using a scaffold design approach, we identified 4-aminomethylphenylacetic acid 4 with a desirable γ-secretase modulation profile. Scaffold optimization led to the discovery of a novel chemical series, represented by 6b, having improved brain penetration. Further SAR studies provided analog 6q that exhibited a good pharmacological profile. Oral administration of 6q significantly reduced brain Aß42 levels in mice and rats.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Antiinflamatorios no Esteroideos/química , Inhibidores Enzimáticos/química , Fenilacetatos/química , Piperidinas/química , Administración Oral , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Antiinflamatorios no Esteroideos/síntesis química , Antiinflamatorios no Esteroideos/farmacocinética , Encéfalo/metabolismo , Diseño de Fármacos , Evaluación Preclínica de Medicamentos , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacocinética , Ratones , Fragmentos de Péptidos/metabolismo , Fenilacetatos/síntesis química , Fenilacetatos/farmacocinética , Piperidinas/síntesis química , Piperidinas/farmacocinética , Ratas
9.
Neuron ; 41(4): 587-98, 2004 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-14980207

RESUMEN

The family of calcium binding proteins called KChIPs associates with Kv4 family K(+) channels and modulates their biophysical properties. Here, using mutagenesis and X-ray crystallography, we explore the interaction between Kv4 subunits and KChIP1. Two regions in the Kv4.2 N terminus, residues 7-11 and 71-90, are necessary for KChIP1 modulation and interaction with Kv4.2. When inserted into the Kv1.2 N terminus, residues 71-90 of Kv4.2 are also sufficient to confer association with KChIP1. To provide a structural framework for these data, we solved the crystal structures of Kv4.3N and KChIP1 individually. Taken together with the mutagenesis data, the individual structures suggest that that the Kv4 N terminus is required for stable association with KChIP1, perhaps through a hydrophobic surface interaction, and that residues 71-90 in Kv4 subunits form a contact loop that mediates the specific association of KChIPs with Kv4 subunits.


Asunto(s)
Proteínas de Unión al Calcio/química , Membrana Celular/química , Canales de Potasio con Entrada de Voltaje , Canales de Potasio/química , Secuencia de Aminoácidos/fisiología , Animales , Sitios de Unión/genética , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Línea Celular , Membrana Celular/genética , Membrana Celular/metabolismo , Cristalografía por Rayos X , Humanos , Proteínas de Interacción con los Canales Kv , Potenciales de la Membrana/genética , Modelos Moleculares , Mutagénesis Sitio-Dirigida/genética , Oocitos/metabolismo , Técnicas de Placa-Clamp , Canales de Potasio/genética , Canales de Potasio/metabolismo , Unión Proteica/genética , Estructura Terciaria de Proteína/genética , Subunidades de Proteína , Canales de Potasio Shal
10.
Stroke ; 39(12): 3372-7, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18927459

RESUMEN

BACKGROUND AND PURPOSE: Thrombolysis with tPA is the only FDA-approved therapy for acute ischemic stroke. But its widespread application remains limited by narrow treatment time windows and the related risks of cerebral hemorrhage. In this study, we ask whether minocycline can prevent tPA-associated cerebral hemorrhage and extend the reperfusion window in an experimental stroke model in rats. METHODS: Spontaneously hypertensive rats were subjected to embolic focal ischemia using homologous clots and treated with: saline at 1 hour; early tPA at 1 hour, delayed tPA at 6 hours; minocycline at 4 hours; combined minocycline at 4 hours plus tPA at 6 hours. Infarct volumes and hemorrhagic transformation were quantified at 24 hours. Gelatin zymography was used to measure blood levels of circulating matrix metalloproteinase-9 (MMP-9). RESULTS: Early 1-hour thrombolysis restored perfusion and reduced infarction. Late 6-hour tPA did not decrease infarction but instead worsened hemorrhagic conversion. Combining minocycline with delayed 6-hour tPA decreased plasma MMP-9 levels, reduced infarction, and ameliorated brain hemorrhage. Blood levels of MMP-9 were also significantly correlated with volumes of infarction and hemorrhage. CONCLUSIONS: Combination therapy with minocycline may extend tPA treatment time windows in ischemic stroke.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Fibrinolíticos/uso terapéutico , Embolia Intracraneal/tratamiento farmacológico , Inhibidores de la Metaloproteinasa de la Matriz , Minociclina/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Terapia Trombolítica/métodos , Activador de Tejido Plasminógeno/uso terapéutico , Animales , Biomarcadores , Barrera Hematoencefálica/efectos de los fármacos , Isquemia Encefálica/sangre , Isquemia Encefálica/enzimología , Isquemia Encefálica/etiología , Hemorragia Cerebral/sangre , Hemorragia Cerebral/inducido químicamente , Hemorragia Cerebral/enzimología , Hemorragia Cerebral/prevención & control , Infarto Cerebral/sangre , Infarto Cerebral/enzimología , Infarto Cerebral/etiología , Infarto Cerebral/prevención & control , Esquema de Medicación , Evaluación Preclínica de Medicamentos , Fibrinolíticos/administración & dosificación , Fibrinolíticos/efectos adversos , Embolia Intracraneal/sangre , Embolia Intracraneal/complicaciones , Embolia Intracraneal/enzimología , Masculino , Metaloproteinasa 9 de la Matriz/sangre , Minociclina/administración & dosificación , Fármacos Neuroprotectores/administración & dosificación , Ratas , Ratas Endogámicas SHR , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/efectos adversos , Proteínas Recombinantes/uso terapéutico , Reperfusión , Terapia Trombolítica/efectos adversos , Factores de Tiempo , Activador de Tejido Plasminógeno/administración & dosificación , Activador de Tejido Plasminógeno/efectos adversos
11.
Bioorg Med Chem Lett ; 18(3): 1135-9, 2008 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-18086526

RESUMEN

A new series of beta-N-biaryl ether sulfonamide hydroxamates as novel gelatinase inhibitors is described. These compounds exhibit good potency for MMP-2 and MMP-9 without inhibiting MMP-1. The structure-activity relationships (SAR) reveal the biaryl ether type P1' moiety together with methanesulfonamide is the optimal combination that provides inhibitory activity of MMP-9 in the single-digit nanomolar range.


Asunto(s)
Gelatinasas/antagonistas & inhibidores , Ácidos Hidroxámicos/síntesis química , Ácidos Hidroxámicos/farmacología , Inhibidores de la Metaloproteinasa de la Matriz , Pirazinas/síntesis química , Pirazinas/farmacología , Animales , Diseño de Fármacos , Humanos , Ácidos Hidroxámicos/química , Microsomas Hepáticos/efectos de los fármacos , Estructura Molecular , Pirazinas/química , Ratas , Relación Estructura-Actividad , Sulfonamidas/síntesis química , Sulfonamidas/química , Sulfonamidas/farmacología
12.
Bioorg Med Chem Lett ; 18(3): 1140-5, 2008 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-18083558

RESUMEN

The introduction and the optimization of an alpha-amino substituent based on a series of alpha-hydroxy-beta-N-biaryl ether sulfonamide hydroxamates is described. The modification leads to a new series of MMP-2/MMP-9 inhibitors with enhanced inhibitory activities and improved ADME properties. An efficacy study on reducing the ischemia-induced brain edema in the rat transient middle cerebral artery occlusion (tMCAo) model is also demonstrated.


Asunto(s)
Aminoácidos/química , Gelatinasas/antagonistas & inhibidores , Ácidos Hidroxámicos/síntesis química , Ácidos Hidroxámicos/farmacología , Inhibidores de la Metaloproteinasa de la Matriz , Pirazinas/síntesis química , Pirazinas/farmacología , Animales , Edema Encefálico/inducido químicamente , Modelos Animales de Enfermedad , Diseño de Fármacos , Humanos , Ácidos Hidroxámicos/química , Microsomas Hepáticos/efectos de los fármacos , Arteria Cerebral Media/efectos de los fármacos , Estructura Molecular , Pirazinas/química , Ratas , Estereoisomerismo , Relación Estructura-Actividad , Sulfonamidas/síntesis química , Sulfonamidas/química , Sulfonamidas/farmacología
13.
Bioorg Med Chem Lett ; 18(1): 409-13, 2008 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-17981034

RESUMEN

Matrix metalloproteinase-9 (MMP-9) has been implicated in the breakdown of the blood-brain barrier during cerebral ischemia. As a result, inhibition of MMP-9 may have utility as a therapeutic intervention in stroke. Towards this end, we have synthesized a series of 1-hydroxy-2-pyridinones that have excellent in vitro potency in inhibiting MMP-9 in addition to MMP-2. Representative compounds also demonstrate good efficacy in the mouse transient mid-cerebral artery occlusion (tMCAO) model of cerebral ischemia.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Inhibidores de la Metaloproteinasa de la Matriz , Inhibidores de Proteasas/síntesis química , Piridonas/síntesis química , Piridonas/farmacología , Administración Oral , Animales , Disponibilidad Biológica , Modelos Animales de Enfermedad , Concentración 50 Inhibidora , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Inhibidores de Proteasas/química , Inhibidores de Proteasas/farmacocinética , Inhibidores de Proteasas/farmacología , Piridonas/química , Piridonas/farmacocinética , Relación Estructura-Actividad , Sulfonamidas/síntesis química , Sulfonamidas/química , Sulfonamidas/farmacocinética , Sulfonamidas/farmacología , Zinc/química , Zinc/metabolismo
14.
Bioorg Med Chem Lett ; 18(1): 405-8, 2008 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-17980583

RESUMEN

Several classes of arylsulfone-based MMP-2/-9 inhibitors utilizing 6- to 8-membered heterocyclic rings as zinc-binding groups (ZBGs) have been synthesized and their enzyme inhibitory activities were evaluated. Although a number of 6- and 7-membered heterocycles were effective, the most potent arylsulfone inhibitors are based on the rigid 1- or 3-hydroxypyridone ZBG.


Asunto(s)
Compuestos Heterocíclicos/síntesis química , Compuestos Heterocíclicos/farmacología , Inhibidores de la Metaloproteinasa de la Matriz , Inhibidores de Proteasas/síntesis química , Sulfonas/síntesis química , Sulfonas/farmacología , Zinc/química , Compuestos Heterocíclicos/química , Lactamas/síntesis química , Lactamas/química , Lactamas/farmacología , Inhibidores de Proteasas/química , Inhibidores de Proteasas/farmacología , Piridonas/química , Piridonas/farmacología , Relación Estructura-Actividad , Sulfonas/química
15.
Cell Rep ; 25(10): 2742-2754.e31, 2018 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-30517862

RESUMEN

The lack of disease-modifying treatments for neurodegenerative disease stems in part from our rudimentary understanding of disease mechanisms and the paucity of targets for therapeutic intervention. Here we used an integrated discovery paradigm to identify a new therapeutic target for diseases caused by α-synuclein (α-syn), a small lipid-binding protein that misfolds and aggregates in Parkinson's disease and other disorders. Using unbiased phenotypic screening, we identified a series of compounds that were cytoprotective against α-syn-mediated toxicity by inhibiting the highly conserved enzyme stearoyl-CoA desaturase (SCD). Critically, reducing the levels of unsaturated membrane lipids by inhibiting SCD reduced α-syn toxicity in human induced pluripotent stem cell (iPSC) neuronal models. Taken together, these findings suggest that inhibition of fatty acid desaturation has potential as a therapeutic approach for the treatment of Parkinson's disease and other synucleinopathies.


Asunto(s)
Estearoil-CoA Desaturasa/antagonistas & inhibidores , alfa-Sinucleína/toxicidad , Animales , Citoprotección/efectos de los fármacos , Ácidos Grasos/metabolismo , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Oxadiazoles/química , Oxadiazoles/farmacología , Agregado de Proteínas , Ratas , Saccharomyces cerevisiae/efectos de los fármacos , Estearoil-CoA Desaturasa/metabolismo , Triglicéridos/metabolismo
16.
Sci Rep ; 8(1): 13438, 2018 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-30194389

RESUMEN

Protein interacting with C kinase (PICK1) is a scaffolding protein that is present in dendritic spines and interacts with a wide array of proteins through its PDZ domain. The best understood function of PICK1 is regulation of trafficking of AMPA receptors at neuronal synapses via its specific interaction with the AMPA GluA2 subunit. Disrupting the PICK1-GluA2 interaction has been shown to alter synaptic plasticity, a molecular mechanism of learning and memory. Lack of potent, selective inhibitors of the PICK1 PDZ domain has hindered efforts at exploring the PICK1-GluA2 interaction as a therapeutic target for neurological diseases. Here, we report the discovery of PICK1 small molecule inhibitors using a structure-based drug design strategy. The inhibitors stabilized surface GluA2, reduced Aß-induced rise in intracellular calcium concentrations in cultured neurons, and blocked long term depression in brain slices. These findings demonstrate that it is possible to identify potent, selective PICK1-GluA2 inhibitors which may prove useful for treatment of neurodegenerative disorders.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Proteínas Portadoras/antagonistas & inhibidores , Espinas Dendríticas/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Proteínas Nucleares/antagonistas & inhibidores , Sinapsis/metabolismo , Animales , Encéfalo/patología , Calcio/metabolismo , Señalización del Calcio , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular , Espinas Dendríticas/patología , Diseño de Fármacos , Ratones , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/patología , Proteínas Nucleares/metabolismo , Dominios PDZ , Receptores AMPA/metabolismo , Sinapsis/patología
17.
Circ Res ; 96(4): 451-8, 2005 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-15662035

RESUMEN

Voltage-gated K+ (Kv) channel accessory (beta) subunits associate with pore-forming Kv alpha subunits and modify the properties and/or cell surface expression of Kv channels in heterologous expression systems. There is very little presently known, however, about the functional role(s) of Kv beta subunits in the generation of native cardiac Kv channels. Exploiting mice with a targeted disruption of the Kvbeta1 gene (Kvbeta1-/-), the studies here were undertaken to explore directly the role of Kvbeta1 in the generation of ventricular Kv currents. Action potential waveforms and peak Kv current densities are indistinguishable in myocytes isolated from the left ventricular apex (LVA) of Kvbeta1-/- and wild-type (WT) animals. Analysis of Kv current waveforms, however, revealed that mean+/-SEM I(to,f) density is significantly (P< or =0.01) lower in Kvbeta1-/- (21.0+/-0.9 pA/pF; n=68), than in WT (25.3+/-1.4 pA/pF; n=42), LVA myocytes, and that mean+/-SEM I(K,slow) density is significantly (P< or =0.01) higher in Kvbeta1-/- (19.1+/-0.9 pA/pF; n=68), compared with WT (15.9+/-0.7 pA/pF; n=42), LVA cells. Pharmacological studies demonstrated that the TEA-sensitive component of I(K,slow), I(K,slow2,) is selectively increased in Kvbeta1-/- LVA myocytes. In parallel with the alterations in I(to,f) and I(K,slow2) densities, Kv4.3 expression is decreased and Kv2.1 expression is increased in Kvbeta1-/- ventricles. Taken together, these results demonstrate that Kvbeta1 differentially regulates the functional cell surface expression of myocardial I(to,f) and I(K,slow2) channels.


Asunto(s)
Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Canales de Potasio Calcio-Activados/fisiología , Canales de Potasio con Entrada de Voltaje/fisiología , Potasio/metabolismo , Potenciales de Acción , Empalme Alternativo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Proteínas de Unión al Calcio/metabolismo , Membrana Celular/metabolismo , Exones , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/metabolismo , Intrones , Activación del Canal Iónico/efectos de los fármacos , Proteínas de Interacción con los Canales Kv , Canal de Potasio Kv.1.1 , Canal de Potasio Kv.1.2 , Canal de Potasio Kv1.4 , Canal de Potasio Kv1.5 , Canales de Potasio de Gran Conductancia Activados por el Calcio , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Técnicas de Placa-Clamp , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio Calcio-Activados/biosíntesis , Canales de Potasio Calcio-Activados/química , Canales de Potasio Calcio-Activados/deficiencia , Canales de Potasio Calcio-Activados/genética , Canales de Potasio con Entrada de Voltaje/biosíntesis , Canales de Potasio con Entrada de Voltaje/química , Canales de Potasio con Entrada de Voltaje/efectos de los fármacos , Canales de Potasio con Entrada de Voltaje/genética , Canales de Potasio con Entrada de Voltaje/metabolismo , Mapeo de Interacción de Proteínas , Subunidades de Proteína , Alineación de Secuencia , Canales de Potasio Shab , Canales de Potasio Shal
18.
Neurosci Lett ; 417(1): 1-5, 2007 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-17386975

RESUMEN

Plasminogen activators are used in thrombolytic stroke therapy. However, it is increasingly recognized that they have other actions besides fibrinolysis. In this study, we assess potential pro-inflammatory effects of tissue-type plasminogen activator (tPA) and urokinase-type plasminogen activator (uPA) in rat cortical astrocytes. Both uPA and tPA induced rapid dose-dependent upregulation in MMP-2 and MMP-9, as demonstrated by zymography of conditioned media. In addition, a multiplex ELISA array demonstrated that patterned responses in chemokines and cytokines were also evoked. Exposure to tPA induced elevations in secreted MIP-2, MCP-1 and GRO/KC. Exposure to uPA induced elevations in secreted IFN-gamma, TNF-alpha, GMCSF, MIP-1alpha, MIP-2, MIP-3alpha, MCP-1, RANTES and fractalkine. These data suggest that plasminogen activators may trigger selected pro-inflammatory responses at the neurovascular interface. Whether these effects influence thrombolytic stroke therapy warrants further investigation.


Asunto(s)
Astrocitos/efectos de los fármacos , Corteza Cerebral/efectos de los fármacos , Encefalitis/inducido químicamente , Mediadores de Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Activadores Plasminogénicos/efectos adversos , Animales , Animales Recién Nacidos , Astrocitos/inmunología , Astrocitos/metabolismo , Biomarcadores/análisis , Biomarcadores/metabolismo , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/inmunología , Células Cultivadas , Corteza Cerebral/inmunología , Corteza Cerebral/metabolismo , Quimiocinas/inmunología , Quimiocinas/metabolismo , Citocinas/inmunología , Citocinas/metabolismo , Relación Dosis-Respuesta a Droga , Encefalitis/inmunología , Encefalitis/metabolismo , Gliosis/inducido químicamente , Gliosis/inmunología , Gliosis/metabolismo , Trombosis Intracraneal/tratamiento farmacológico , Metaloproteinasa 2 de la Matriz/efectos de los fármacos , Metaloproteinasa 2 de la Matriz/inmunología , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/efectos de los fármacos , Metaloproteinasa 9 de la Matriz/inmunología , Metaloproteinasa 9 de la Matriz/metabolismo , Metaloproteinasas de la Matriz/efectos de los fármacos , Metaloproteinasas de la Matriz/inmunología , Metaloproteinasas de la Matriz/metabolismo , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/inmunología
20.
J Comp Neurol ; 496(3): 289-302, 2006 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-16566008

RESUMEN

Ca(2+)-activated voltage-dependent K(+) channels (Slo1, KCa1.1, Maxi-K, or BK channel) play a crucial role in controlling neuronal signaling by coupling channel activity to both membrane depolarization and intracellular Ca(2+) signaling. In mammalian brain, immunolabeling experiments have shown staining for Slo1 channels predominantly localized to axons and presynaptic terminals of neurons. We have developed anti-Slo1 mouse monoclonal antibodies that have been extensively characterized for specificity of staining against recombinant Slo1 in heterologous cells, and native Slo1 in mammalian brain, and definitively by the lack of detectable immunoreactivity against brain samples from Slo1 knockout mice. Here we provide precise immunolocalization of Slo1 in rat brain with one of these monoclonal antibodies and show that Slo1 is accumulated in axons and synaptic terminal zones associated with glutamatergic synapses in hippocampus and GABAergic synapses in cerebellum. By using cultured hippocampal pyramidal neurons as a model system, we show that heterologously expressed Slo1 is initially targeted to the axonal surface membrane, and with further development in culture, become localized in presynaptic terminals. These studies provide new insights into the polarized localization of Slo1 channels in mammalian central neurons and provide further evidence for a key role in regulating neurotransmitter release in glutamatergic and GABAergic terminals.


Asunto(s)
Axones/metabolismo , Encéfalo/citología , Encéfalo/metabolismo , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Terminaciones Nerviosas/metabolismo , Neuronas/citología , Animales , Animales Recién Nacidos , Western Blotting/métodos , Calbindinas , Células Cultivadas , Homólogo 4 de la Proteína Discs Large , Embrión de Mamíferos , Guanilato-Quinasas , Inmunohistoquímica/métodos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Canal de Potasio Kv1.4/metabolismo , Canales de Potasio de Gran Conductancia Activados por el Calcio/deficiencia , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas de Neurofilamentos/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Ratas , Proteína G de Unión al Calcio S100/metabolismo , Canales de Potasio Shab/metabolismo , Sinapsinas/metabolismo , Proteínas tau/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA