Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
J Pharmacol Exp Ther ; 386(1): 4-14, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-36958846

RESUMO

Divalent metal transporter 1 (DMT1) cotransports ferrous iron and protons and is the primary mechanism for uptake of nonheme iron by enterocytes. Inhibitors are potentially useful as therapeutic agents to treat iron overload disorders such as hereditary hemochromatosis or ß-thalassemia intermedia, provided that inhibition can be restricted to the duodenum. We used a calcein quench assay to identify human DMT1 inhibitors. Dimeric compounds were made to generate more potent compounds with low systemic exposure. Direct block of DMT1 was confirmed by voltage clamp measurements. The lead compound, XEN602, strongly inhibits dietary nonheme iron uptake in both rats and pigs yet has negligible systemic exposure. Efficacy is maintained for >2 weeks in a rat subchronic dosing assay. Doses that lowered iron content in the spleen and liver by >50% had no effect on the tissue content of other divalent cations except for cobalt. XEN602 represents a powerful pharmacological tool for understanding the physiologic function of DMT1 in the gut. SIGNIFICANCE STATEMENT: This report introduces methodology to develop potent, gut-restricted inhibitors of divalent metal transporter 1 (DMT1) and identifies XEN602 as a suitable compound for in vivo studies. We also report novel animal models to quantify the inhibition of dietary uptake of iron in both rodents and pigs. This research shows that inhibition of DMT1 is a promising means to treat iron overload disorders.


Assuntos
Sobrecarga de Ferro , Humanos , Ratos , Animais , Suínos , Sobrecarga de Ferro/tratamento farmacológico , Ferro/metabolismo , Transporte Biológico , Proteínas de Ligação ao Ferro/metabolismo , Modelos Animais
2.
Elife ; 112022 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-35234610

RESUMO

NBI-921352 (formerly XEN901) is a novel sodium channel inhibitor designed to specifically target NaV1.6 channels. Such a molecule provides a precision-medicine approach to target SCN8A-related epilepsy syndromes (SCN8A-RES), where gain-of-function (GoF) mutations lead to excess NaV1.6 sodium current, or other indications where NaV1.6 mediated hyper-excitability contributes to disease (Gardella and Møller, 2019; Johannesen et al., 2019; Veeramah et al., 2012). NBI-921352 is a potent inhibitor of NaV1.6 (IC500.051 µM), with exquisite selectivity over other sodium channel isoforms (selectivity ratios of 756 X for NaV1.1, 134 X for NaV1.2, 276 X for NaV1.7, and >583 Xfor NaV1.3, NaV1.4, and NaV1.5). NBI-921352is a state-dependent inhibitor, preferentially inhibiting inactivatedchannels. The state dependence leads to potent stabilization of inactivation, inhibiting NaV1.6 currents, including resurgent and persistent NaV1.6 currents, while sparing the closed/rested channels. The isoform-selective profile of NBI-921352 led to a robust inhibition of action-potential firing in glutamatergic excitatory pyramidal neurons, while sparing fast-spiking inhibitory interneurons, where NaV1.1 predominates. Oral administration of NBI-921352 prevented electrically induced seizures in a Scn8a GoF mouse,as well as in wild-type mouse and ratseizure models. NBI-921352 was effective in preventing seizures at lower brain and plasma concentrations than commonly prescribed sodium channel inhibitor anti-seizure medicines (ASMs) carbamazepine, phenytoin, and lacosamide. NBI-921352 waswell tolerated at higher multiples of the effective plasma and brain concentrations than those ASMs. NBI-921352 is entering phase II proof-of-concept trials for the treatment of SCN8A-developmental epileptic encephalopathy (SCN8A-DEE) and adult focal-onset seizures.


Assuntos
Epilepsia , Canal de Sódio Disparado por Voltagem NAV1.6 , Animais , Mutação com Ganho de Função , Camundongos , Mutação , Canal de Sódio Disparado por Voltagem NAV1.6/genética , Neurônios/fisiologia , Ratos , Sódio , Bloqueadores dos Canais de Sódio/farmacologia
3.
J Med Chem ; 64(6): 2953-2966, 2021 03 25.
Artigo em Inglês | MEDLINE | ID: mdl-33682420

RESUMO

Nav1.7 is an extensively investigated target for pain with a strong genetic link in humans, yet in spite of this effort, it remains challenging to identify efficacious, selective, and safe inhibitors. Here, we disclose the discovery and preclinical profile of GDC-0276 (1) and GDC-0310 (2), selective Nav1.7 inhibitors that have completed Phase 1 trials. Our initial search focused on close-in analogues to early compound 3. This resulted in the discovery of GDC-0276 (1), which possessed improved metabolic stability and an acceptable overall pharmacokinetics profile. To further derisk the predicted human pharmacokinetics and enable QD dosing, additional optimization of the scaffold was conducted, resulting in the discovery of a novel series of N-benzyl piperidine Nav1.7 inhibitors. Improvement of the metabolic stability by blocking the labile benzylic position led to the discovery of GDC-0310 (2), which possesses improved Nav selectivity and pharmacokinetic profile over 1.


Assuntos
Azetidinas/farmacologia , Benzamidas/farmacologia , Descoberta de Drogas , Canal de Sódio Disparado por Voltagem NAV1.7/metabolismo , Sulfonamidas/farmacologia , Bloqueadores do Canal de Sódio Disparado por Voltagem/farmacologia , Animais , Azetidinas/química , Azetidinas/farmacocinética , Benzamidas/química , Benzamidas/farmacocinética , Células Cultivadas , Células HEK293 , Humanos , Piperidinas/química , Piperidinas/farmacocinética , Piperidinas/farmacologia , Ratos Sprague-Dawley , Sulfonamidas/química , Sulfonamidas/farmacocinética , Bloqueadores do Canal de Sódio Disparado por Voltagem/química , Bloqueadores do Canal de Sódio Disparado por Voltagem/farmacocinética
4.
J Med Chem ; 62(21): 9618-9641, 2019 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-31525968

RESUMO

Nonselective antagonists of voltage-gated sodium (NaV) channels have been long used for the treatment of epilepsies. The efficacy of these drugs is thought to be due to the block of sodium channels on excitatory neurons, primarily NaV1.6 and NaV1.2. However, these currently marketed drugs require high drug exposure and suffer from narrow therapeutic indices. Selective inhibition of NaV1.6, while sparing NaV1.1, is anticipated to provide a more effective and better tolerated treatment for epilepsies. In addition, block of NaV1.2 may complement the anticonvulsant activity of NaV1.6 inhibition. We discovered a novel series of aryl sulfonamides as CNS-penetrant, isoform-selective NaV1.6 inhibitors, which also displayed potent block of NaV1.2. Optimization focused on increasing selectivity over NaV1.1, improving metabolic stability, reducing active efflux, and addressing a pregnane X-receptor liability. We obtained compounds 30-32, which produced potent anticonvulsant activity in mouse seizure models, including a direct current maximal electroshock seizure assay.


Assuntos
Amidas/química , Sistema Nervoso Central/metabolismo , Epilepsia/tratamento farmacológico , Canal de Sódio Disparado por Voltagem NAV1.6/metabolismo , Bloqueadores dos Canais de Sódio/metabolismo , Bloqueadores dos Canais de Sódio/farmacologia , Animais , Cães , Células Hep G2 , Humanos , Células Madin Darby de Rim Canino , Camundongos , Modelos Moleculares , Canal de Sódio Disparado por Voltagem NAV1.6/química , Domínios Proteicos , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Bloqueadores dos Canais de Sódio/química , Bloqueadores dos Canais de Sódio/uso terapêutico , Relação Estrutura-Atividade
5.
J Med Chem ; 62(2): 908-927, 2019 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-30499663

RESUMO

Herein, we report the discovery and optimization of a series of orally bioavailable acyl sulfonamide NaV1.7 inhibitors that are selective for NaV1.7 over NaV1.5 and highly efficacious in in vivo models of pain and hNaV1.7 target engagement. An analysis of the physicochemical properties of literature NaV1.7 inhibitors suggested that acyl sulfonamides with high fsp3 could overcome some of the pharmacokinetic (PK) and efficacy challenges seen with existing series. Parallel library syntheses lead to the identification of analogue 7, which exhibited moderate potency against NaV1.7 and an acceptable PK profile in rodents, but relatively poor stability in human liver microsomes. Further, design strategy then focused on the optimization of potency against hNaV1.7 and improvement of human metabolic stability, utilizing induced fit docking in our previously disclosed X-ray cocrystal of the NaV1.7 voltage sensing domain. These investigations culminated in the discovery of tool compound 33, one of the most potent and efficacious NaV1.7 inhibitors reported to date.


Assuntos
Analgésicos/química , Canal de Sódio Disparado por Voltagem NAV1.7/química , Sulfonamidas/química , Bloqueadores do Canal de Sódio Disparado por Voltagem/química , Analgésicos/metabolismo , Analgésicos/uso terapêutico , Animais , Sítios de Ligação , Desenho de Fármacos , Meia-Vida , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Microssomos Hepáticos/metabolismo , Simulação de Acoplamento Molecular , Canal de Sódio Disparado por Voltagem NAV1.7/metabolismo , Dor/induzido quimicamente , Dor/tratamento farmacológico , Dor/patologia , Estrutura Terciária de Proteína , Ratos , Ratos Sprague-Dawley , Relação Estrutura-Atividade , Sulfonamidas/metabolismo , Sulfonamidas/uso terapêutico , Bloqueadores do Canal de Sódio Disparado por Voltagem/metabolismo , Bloqueadores do Canal de Sódio Disparado por Voltagem/uso terapêutico
6.
Cell Rep ; 24(12): 3133-3145, 2018 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-30231997

RESUMO

Selective block of NaV1.7 promises to produce non-narcotic analgesic activity without motor or cognitive impairment. Several NaV1.7-selective blockers have been reported, but efficacy in animal pain models required high multiples of the IC50 for channel block. Here, we report a target engagement assay using transgenic mice that has enabled the development of a second generation of selective Nav1.7 inhibitors that show robust analgesic activity in inflammatory and neuropathic pain models at low multiples of the IC50. Like earlier arylsulfonamides, these newer acylsulfonamides target a binding site on the surface of voltage sensor domain 4 to achieve high selectivity among sodium channel isoforms and steeply state-dependent block. The improved efficacy correlates with very slow dissociation from the target channel. Chronic dosing increases compound potency about 10-fold, possibly due to reversal of sensitization arising during chronic injury, and provides efficacy that persists long after the compound has cleared from plasma.


Assuntos
Analgésicos/uso terapêutico , Canal de Sódio Disparado por Voltagem NAV1.7/metabolismo , Neuralgia/tratamento farmacológico , Bloqueadores dos Canais de Sódio/uso terapêutico , Sulfonamidas/uso terapêutico , Analgésicos/farmacocinética , Animais , Sítios de Ligação , Células Cultivadas , Células HEK293 , Humanos , Concentração Inibidora 50 , Camundongos , Canal de Sódio Disparado por Voltagem NAV1.7/química , Ligação Proteica , Bloqueadores dos Canais de Sódio/farmacocinética , Sulfonamidas/farmacocinética
7.
J Med Chem ; 61(11): 4810-4831, 2018 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-29737846

RESUMO

The sodium channel NaV1.7 has emerged as a promising target for the treatment of pain based on strong genetic validation of its role in nociception. In recent years, a number of aryl and acyl sulfonamides have been reported as potent inhibitors of NaV1.7, with high selectivity over the cardiac isoform NaV1.5. Herein, we report on the discovery of a novel series of N-([1,2,4]triazolo[4,3- a]pyridin-3-yl)methanesulfonamides as selective NaV1.7 inhibitors. Starting with the crystal structure of an acyl sulfonamide, we rationalized that cyclization to form a fused heterocycle would improve physicochemical properties, in particular lipophilicity. Our design strategy focused on optimization of potency for block of NaV1.7 and human metabolic stability. Lead compounds 10, 13 (GNE-131), and 25 showed excellent potency, good in vitro metabolic stability, and low in vivo clearance in mouse, rat, and dog. Compound 13 also displayed excellent efficacy in a transgenic mouse model of induced pain.


Assuntos
Desenho de Fármacos , Canal de Sódio Disparado por Voltagem NAV1.7/metabolismo , Dor/tratamento farmacológico , Sulfonamidas/química , Sulfonamidas/farmacologia , Bloqueadores do Canal de Sódio Disparado por Voltagem/química , Bloqueadores do Canal de Sódio Disparado por Voltagem/farmacologia , Sequência de Aminoácidos , Animais , Cães , Estabilidade de Medicamentos , Humanos , Cinética , Camundongos , Conformação Molecular , Dor/metabolismo , Ratos , Sulfonamidas/farmacocinética , Sulfonamidas/uso terapêutico , Bloqueadores do Canal de Sódio Disparado por Voltagem/farmacocinética , Bloqueadores do Canal de Sódio Disparado por Voltagem/uso terapêutico
8.
ACS Med Chem Lett ; 7(3): 277-82, 2016 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-26985315

RESUMO

We report on a novel series of aryl sulfonamides that act as nanomolar potent, isoform-selective inhibitors of the human sodium channel hNaV1.7. The optimization of these inhibitors is described. We aimed to improve potency against hNaV1.7 while minimizing off-target safety concerns and generated compound 3. This agent displayed significant analgesic effects in rodent models of acute and inflammatory pain and demonstrated that binding to the voltage sensor domain 4 site of NaV1.7 leads to an analgesic effect in vivo. Our findings corroborate the importance of hNaV1.7 as a drug target for the treatment of pain.

9.
Science ; 350(6267): aac5464, 2015 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-26680203

RESUMO

Voltage-gated sodium (Nav) channels propagate action potentials in excitable cells. Accordingly, Nav channels are therapeutic targets for many cardiovascular and neurological disorders. Selective inhibitors have been challenging to design because the nine mammalian Nav channel isoforms share high sequence identity and remain recalcitrant to high-resolution structural studies. Targeting the human Nav1.7 channel involved in pain perception, we present a protein-engineering strategy that has allowed us to determine crystal structures of a novel receptor site in complex with isoform-selective antagonists. GX-936 and related inhibitors bind to the activated state of voltage-sensor domain IV (VSD4), where their anionic aryl sulfonamide warhead engages the fourth arginine gating charge on the S4 helix. By opposing VSD4 deactivation, these compounds inhibit Nav1.7 through a voltage-sensor trapping mechanism, likely by stabilizing inactivated states of the channel. Residues from the S2 and S3 helices are key determinants of isoform selectivity, and bound phospholipids implicate the membrane as a modulator of channel function and pharmacology. Our results help to elucidate the molecular basis of voltage sensing and establish structural blueprints to design selective Nav channel antagonists.


Assuntos
Canal de Sódio Disparado por Voltagem NAV1.7/química , Bloqueadores dos Canais de Sódio/química , Bloqueadores dos Canais de Sódio/farmacologia , Sulfonamidas/química , Sulfonamidas/farmacologia , Tiadiazóis/química , Tiadiazóis/farmacologia , Sequência de Aminoácidos , Membrana Celular/química , Cristalização/métodos , Cristalografia por Raios X , Análise Mutacional de DNA , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Canal de Sódio Disparado por Voltagem NAV1.7/genética , Percepção da Dor/efeitos dos fármacos , Engenharia de Proteínas , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/química , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína
10.
Bioorg Med Chem Lett ; 24(18): 4397-4401, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-25176194

RESUMO

The voltage gated sodium channel Nav1.7 represents an interesting target for the treatment of pain. Human genetic studies have identified the crucial role of Nav1.7 in pain signaling. Herein, we report the design and synthesis of a novel series of benzenesulfonamide-based Nav1.7 inhibitors. Structural-activity relationship (SAR) studies were undertaken towards improving Nav1.7 activity and minimizing CYP inhibition. These efforts resulted in the identification of compound 12k, a highly potent Nav1.7 inhibitor with a thousand-fold selectivity over Nav1.5 and negligible CYP inhibition.


Assuntos
Citocromo P-450 CYP3A/metabolismo , Descoberta de Drogas , Inibidores Enzimáticos/farmacologia , Canal de Sódio Disparado por Voltagem NAV1.7/metabolismo , Sulfonamidas/farmacologia , Bloqueadores do Canal de Sódio Disparado por Voltagem/farmacologia , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Humanos , Estrutura Molecular , Relação Estrutura-Atividade , Sulfonamidas/síntese química , Sulfonamidas/química , Bloqueadores do Canal de Sódio Disparado por Voltagem/síntese química , Bloqueadores do Canal de Sódio Disparado por Voltagem/química , Benzenossulfonamidas
12.
Life Sci ; 91(11-12): 402-408, 2012 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-22906633

RESUMO

AIMS: The present work investigated the underlying mechanism for the positive inotropic effect of liguzinediol (LZDO) in isolated rat hearts. MAIN METHODS: Isolated rat heart perfusion, intracellular action potential recording, patch clamp and Ca2+ imaging were used to measure the isolated rat heart contractility, action potential duration, L-type Ca2+ current and sarcoplasmic reticulum (SR) Ca2+ transient in rat cardiomyocyte, respectively. KEY FINDINGS: LZDO (1, 10, and 100µM) significantly enhanced the inotropy of isolated rat hearts, but not heart rates. Nimodipine (1µM, an L-type Ca2+ channel antagonist), ruthenium red (5µM, a ryanodine receptor inhibitor) and thapsigargin (2µM, an irreversible SR Ca2+ ATPase inhibitor) completely blocked the positive inotropic effect of LZDO. LZDO significantly enhanced the intracellular Ca2+ transient in rat cardiomyocyte. However, LZDO (100µM) did not increase L-type Ca2+ channel current. Moreover, LZDO (100µM) restored the depletion effect of caffeine on Ca2+ transient. The following compounds also failed to block the positive inotropic effect of LZDO (100µM): ß-AR antagonist (propranolol 1µM), phosphodiesterase (PDE) inhibitor (IBMX 5µM), Na+-K+ ATPase inhibitor (ouabain 1µM), α(1)-AR antagonist (prazosin 1µM), dopamine D1 receptor antagonist (SCH23390 1µM) and Na+-Ca2+ exchange inhibitor (KB-R7943 1µM). SIGNIFICANCE: The positive inotropic effect of LZDO in isolated rat hearts was mediated through an elevation of SR Ca2+ transient, which may act on SR Ca2+ ATPase. LZDO has a unique biological mechanism that may prove effective in treating heart failure in clinic.


Assuntos
Cardiotônicos/farmacologia , Coração/efeitos dos fármacos , Pirazinas/farmacologia , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/efeitos dos fármacos , 1-Metil-3-Isobutilxantina/farmacologia , Animais , Cafeína/farmacologia , Cardiotônicos/antagonistas & inibidores , Relação Dose-Resposta a Droga , Coração/fisiologia , Masculino , Contração Miocárdica/efeitos dos fármacos , Contração Miocárdica/fisiologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/fisiologia , Nimodipina/farmacologia , Ouabaína/farmacologia , Prazosina/farmacologia , Propranolol/farmacologia , Ratos , Ratos Sprague-Dawley , Rutênio Vermelho/farmacologia , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/antagonistas & inibidores , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/fisiologia , Tapsigargina/farmacologia
13.
Pain ; 153(1): 80-85, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22035805

RESUMO

Mutations in the SCN9A gene leading to deficiency of its protein product, Na(v)1.7, cause congenital indifference to pain (CIP). CIP is characterized by the absence of the ability to sense pain associated with noxious stimuli. In contrast, the opposite phenotype to CIP, inherited erythromelalgia (IEM), is a disorder of spontaneous pain caused by missense mutations resulting in gain-of-function in Na(v)1.7 that promote neuronal hyperexcitability. The primary aim of this study was to demonstrate that Na(v)1.7 antagonism could alleviate the pain of IEM, thereby demonstrating the utility of this opposite phenotype model as a tool for rapid proof-of-concept for novel analgesics. An exploratory, randomized, double-blind, 2-period crossover study was conducted in 4 SCN9A mutation-proven IEM patients. In each treatment period (2days), separated by a 2-day washout period, patients were orally administered XEN402 (400mg twice daily) or matching placebo. In 3 patients, pain was induced by heat or exercise during each treatment arm. A fourth patient, in constant severe pain, required no induction. Patient-reported outcomes of pain intensity and/or relief were recorded, and the time taken to induce pain was measured. The ability to induce pain in IEM patients was significantly attenuated by XEN402 compared with placebo. XEN402 increased the time to maximal pain induction and significantly reduced the amount of pain (42% less) after induction (P=.014). This pilot study showed that XEN402 blocks Na(v)1.7-mediated pain associated with IEM, thereby demonstrating target engagement in humans and underscoring the use of rare genetic disorders with mutant target channels as a novel approach to rapid proof-of-concept.


Assuntos
Eritromelalgia/tratamento farmacológico , Bloqueadores dos Canais de Sódio/uso terapêutico , Canais de Sódio/genética , Adulto , Método Duplo-Cego , Eritromelalgia/genética , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Canal de Sódio Disparado por Voltagem NAV1.7 , Medição da Dor , Projetos Piloto , Resultado do Tratamento
14.
PLoS Genet ; 5(9): e1000650, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19763162

RESUMO

Cornelia de Lange Syndrome (CdLS) is a multi-organ system birth defects disorder linked, in at least half of cases, to heterozygous mutations in the NIPBL gene. In animals and fungi, orthologs of NIPBL regulate cohesin, a complex of proteins that is essential for chromosome cohesion and is also implicated in DNA repair and transcriptional regulation. Mice heterozygous for a gene-trap mutation in Nipbl were produced and exhibited defects characteristic of CdLS, including small size, craniofacial anomalies, microbrachycephaly, heart defects, hearing abnormalities, delayed bone maturation, reduced body fat, behavioral disturbances, and high mortality (75-80%) during the first weeks of life. These phenotypes arose despite a decrease in Nipbl transcript levels of only approximately 30%, implying extreme sensitivity of development to small changes in Nipbl activity. Gene expression profiling demonstrated that Nipbl deficiency leads to modest but significant transcriptional dysregulation of many genes. Expression changes at the protocadherin beta (Pcdhb) locus, as well as at other loci, support the view that NIPBL influences long-range chromosomal regulatory interactions. In addition, evidence is presented that reduced expression of genes involved in adipogenic differentiation may underlie the low amounts of body fat observed both in Nipbl+/- mice and in individuals with CdLS.


Assuntos
Síndrome de Cornélia de Lange/genética , Síndrome de Cornélia de Lange/patologia , Regulação da Expressão Gênica , Heterozigoto , Especificidade de Órgãos/genética , Fatores de Transcrição/genética , Transcrição Gênica , Animais , Animais Recém-Nascidos , Desenvolvimento Ósseo , Osso e Ossos/anormalidades , Osso e Ossos/patologia , Caderinas/genética , Caderinas/metabolismo , Proteínas de Ciclo Celular , Anormalidades Craniofaciais/complicações , Anormalidades Craniofaciais/genética , Anormalidades Craniofaciais/patologia , Anormalidades Craniofaciais/fisiopatologia , Síndrome de Cornélia de Lange/complicações , Síndrome de Cornélia de Lange/fisiopatologia , Modelos Animais de Doenças , Embrião de Mamíferos/anormalidades , Embrião de Mamíferos/patologia , Cardiopatias Congênitas/complicações , Cardiopatias Congênitas/genética , Cardiopatias Congênitas/patologia , Cardiopatias Congênitas/fisiopatologia , Camundongos , Mutação/genética , Malformações do Sistema Nervoso/complicações , Malformações do Sistema Nervoso/genética , Malformações do Sistema Nervoso/fisiopatologia , Fenótipo , Troca de Cromátide Irmã , Análise de Sobrevida
15.
Int J Neuropsychopharmacol ; 9(4): 457-63, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16316483

RESUMO

Recent studies indicate that levels of presynaptic proteins are altered in the post-mortem brain in schizophrenia. In particular, the hippocampus exhibits reduced levels of synaptophysin and the SNARE protein SNAP-25. The effects of treatment with antipsychotic drugs on levels of SNAP-25 in the hippocampus remains unknown. To determine the effects of typical antipsychotic drugs on levels of synaptophysin and SNAP-25 in the hippocampus, rats were treated with chlorpromazine, haloperidol or trifluoperazine for 21 d. Quantitative immunohistochemistry was used to measure immunoreactivity within the trisynaptic circuit of the hippocampus. Trifluoperazine decreased synaptophysin within the Schaffer collateral region of the radiatum lacunosum in CA1, while haloperidol and chlorpromazine increased SNAP-25 throughout the trisynaptic pathway of the hippocampus, with strongest effects in the mossy fibre region of CA3. These results indicate that presynaptic proteins represent a potential molecular substrate for the effects of antipsychotic drugs on hippocampal synaptic connectivity.


Assuntos
Antipsicóticos/farmacologia , Hipocampo/efeitos dos fármacos , Sinapses/efeitos dos fármacos , Sinaptofisina/biossíntese , Proteína 25 Associada a Sinaptossoma/biossíntese , Animais , Hipocampo/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Sinapses/metabolismo
16.
Eur J Neurosci ; 21(12): 3310-20, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16026469

RESUMO

The mesocortical dopamine (DA) input to the prefrontal cortex (PFC) is crucial for processing short-term working memory (STWM) to guide forthcoming behavior. Short-term plasticity-like post-tetanic potentiation (PTP, < 3 min) and short-term potentiation (STP, < 10 min) may underlie STWM. Using whole-cell voltage-clamp recordings, mixed glutamatergic excitatory postsynaptic currents (EPSCs) evoked by layer III or layer V stimulation (0.5 or 0.067 Hz) were recorded from layer V pyramidal neurons. With 0.5 Hz basal stimulation of layer III, brief tetani (2 x 50 Hz) induced a homosynaptic PTP (decayed: approximately 1 min). The D1-like antagonist SCH23390 (1 microm) increased the PTP amplitude and decay time without inducing changes to the tetanic response. The tetani may evoke endogenous DA release, which activates a presynaptic D1-like receptor to inhibit glutamate release to modulate PTP. With a slower (0.067 Hz) basal stimulation, the same tetani induced STP (lasting approximately 4 min, but only at 2x intensity only) that was insignificantly suppressed by SCH23390. With stimulation of layer-V-->V inputs at 0.5 Hz, layer V tetani yielded inconsisitent responses. However, at 0.067 Hz, tetani at double the intensity resulted in an STP (lasting approximately 6 min), but a long-term depression after SCH23390 application. Endogenous DA released by tetanic stimulation can interact with a D1-like receptor to induce STP in layer V-->V synapses that receive slower (0.067 Hz) frequency inputs, but suppresses PTP at layer III-->V synapses that receive higher (0.5 Hz) frequency inputs. This D1-like modulation of layer- and frequency-specific synaptic responses in the PFC may contribute to STWM processing.


Assuntos
Plasticidade Neuronal/fisiologia , Neurônios/fisiologia , Córtex Pré-Frontal/citologia , Receptores de Dopamina D1/fisiologia , Animais , Animais Recém-Nascidos , Área Sob a Curva , Benzazepinas/farmacologia , Antagonistas de Dopamina/farmacologia , Relação Dose-Resposta à Radiação , Estimulação Elétrica/métodos , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Potenciais Pós-Sinápticos Excitadores/efeitos da radiação , Masculino , Plasticidade Neuronal/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Técnicas de Patch-Clamp/métodos , Ratos , Ritmo Teta/efeitos dos fármacos , Ritmo Teta/efeitos da radiação , Fatores de Tempo
17.
Arch Gen Psychiatry ; 62(3): 263-72, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15753239

RESUMO

BACKGROUND: Converging neuroimaging and postmortem evidence indicates synaptic terminals are abnormal in schizophrenia. A putative molecular mechanism implicates abnormalities of proteins involved in the presynaptic secretory machinery, including the modulator proteins complexin I and complexin II. OBJECTIVES: To determine the amount and distribution of complexin proteins in the hippocampus of subjects with schizophrenia, in parallel with markers for excitatory and inhibitory nerve terminals. The functional implications were also investigated. DESIGN: We used immunocytochemistry to study complexin I and complexin II proteins in hippocampus, as well as the vesicular transporters for gamma-aminobutyric acid (GABA) and for glutamate. Immunocytochemical findings were correlated with cognitive function assessed through medical record review. To further explore the implications of the human findings, we studied rats exposed to haloperidol, amphetamine, and ketamine as well as rats trained in memory tasks. SUBJECTS: We studied hippocampal sections from 12 subjects with schizophrenia and 12 subjects with no known neuropsychiatric disorder. RESULTS: The absolute values and ratio of the hippocampal presynaptic proteins complexin II-complexin I were lower in subjects with schizophrenia. Disturbances in the complexin proteins in subjects with schizophrenia were greater than those observed for vesicular gamma-aminobutyric acid or vesicular glutamate transporters. The lower complexin II-complexin I ratio in several hippocampal subfields in subjects with schizophrenia was inversely correlated with the severity of antemortem cognitive impairment. In contrast, the hippocampal complexin II-complexin I ratio was higher in rats trained in a memory task compared with untrained rats. Treatment of rats with antipsychotic drugs or with the psychotomimetic drugs amphetamine or ketamine did not alter the complexin II-complexin I ratio. CONCLUSIONS: The pathology of hippocampal complexin proteins might play an important role in schizophrenia, especially concerning cognitive disturbances.


Assuntos
Transtornos Cognitivos/diagnóstico , Hipocampo/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Esquizofrenia/diagnóstico , Psicologia do Esquizofrênico , Proteínas Adaptadoras de Transporte Vesicular , Idoso , Animais , Antipsicóticos/farmacologia , Comportamento Animal/efeitos dos fármacos , Transtornos Cognitivos/induzido quimicamente , Transtornos Cognitivos/metabolismo , Modelos Animais de Doenças , Feminino , Haloperidol/farmacologia , Hipocampo/fisiopatologia , Humanos , Imuno-Histoquímica , Ketamina/farmacologia , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Proteínas de Membrana Transportadoras/metabolismo , Pessoa de Meia-Idade , Proteínas do Tecido Nervoso/fisiologia , Terminações Pré-Sinápticas/metabolismo , Terminações Pré-Sinápticas/fisiologia , Ratos , Ratos Sprague-Dawley , Esquizofrenia/metabolismo , Esquizofrenia/fisiopatologia , Proteína Vesicular 1 de Transporte de Glutamato , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores
18.
Eur J Neurosci ; 20(1): 303-7, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15245502

RESUMO

Post-weaning social isolation-rearing of rats leads to behavioural and neurochemical sequelae that model aspects of schizophrenia, and it may be useful to test hypotheses related to putative molecular mechanisms of the illness. In humans, the presynaptic protein CDCrel-1 represents an interesting candidate molecule for the mechanism and aetiology of schizophrenia. CDCrel-1 modulates dopamine neurotransmission, binds to the SNARE protein syntaxin and maps onto a region of chromosome 22q11 deleted in velo-cardio-facial and DiGeorge syndromes, which are associated with increased prevalence of schizophrenia. Using the isolation-rearing model, we measured immunoreactivity of the synaptic proteins CDCrel-1, synaptophysin and syntaxin. Male, Sprague-Dawley rats were raised in groups or in isolation for 12 weeks from weaning. Synaptic protein immunoreactivities were measured in striatal and hippocampal homogenates, using a sensitive enzyme-linked immunoadsorbent assay with monoclonal antibodies. Isolation-rearing produced region- and protein-specific effects. CDCrel-1 immunoreactivity was significantly lower in the striatum and marginally higher in the hippocampus of isolation-reared compared with socially reared animals. There were no statistically significant differences in synaptophysin immunoreactivity in either region. Confocal microscopy demonstrated a high degree of colocalization between the two presynaptic proteins. In striatum, a robust relationship between CDCrel-1 and syntaxin immunoreactivities was observed in socially reared rats, this was lost in the isolation-reared animals. Altered levels of the septin CDCrel-1 in isolation-reared rats may contribute to changes in neuronal connectivity and neurotransmission, and suggest a potential role for CDCrel-1 in schizophrenia related to chromosome 22q11 deletion syndrome.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Neostriado/metabolismo , Isolamento Social , Animais , Comportamento Animal , Western Blotting/métodos , Proteínas de Ciclo Celular/imunologia , Ensaio de Imunoadsorção Enzimática , Hipocampo/metabolismo , Imuno-Histoquímica/métodos , Masculino , Proteínas de Membrana/metabolismo , Proteínas Qa-SNARE , Ratos , Ratos Sprague-Dawley , Septinas , Sinaptofisina/metabolismo
20.
J Neurosci ; 24(1): 8-23, 2004 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-14715933

RESUMO

To determine the nature of dopamine modulation of dendritic Ca2+ signaling in layers V-VI prefrontal cortex (PFC) neurons, whole-cell Ca2+ potentials were evoked after blockade of Na+ and K+ channels. Soma-dendritic Ca2+ spikes evoked by suprathreshold depolarizing pulses, which could be terminated by superimposed brief intrasomatic hyperpolarizing pulses, are blocked by the L-type Ca2+ channel antagonist nimodipine (1 microM). The D1/D5 receptor agonist dihydrexidine (DHX) (0.01-10 microM; 5 min) or R-(+)SKF81291 (10 microM) induced a prolonged (>30 min) dose-dependent peak suppression of these Ca2+ spikes. This effect was dependent on [Ca2+]i- and protein kinase C (PKC)-dependent mechanisms because [Ca2+]i chelation by BAPTA or inhibition of PKC by bisindolymaleimide (BiM1), but not inhibition of [Ca2+]i release with heparin or Xestospongin C, prevented the D1-mediated suppression of Ca2+ spikes. Depolarizing pulses subthreshold to activating a Ca2+ spike evoked a nimodipine-sensitive Ca2+ "hump" potential. D1/D5 stimulation induced an N-[2-((o-bromocinamyl)amino)ethyl]-5-isoquinolinesulfonamide (H-89)- or internal PKA inhibitory peptide[5-24]-sensitive (PKA-dependent) transient (approximately 7 min) potentiation of the hump potential to full Ca2+ spike firing. Furthermore, application of DHX in the presence of the PKC inhibitor BiM1 or internal PKC inhibitory peptide[19-36] resulted in persistent firing of full Ca2+ spike bursts, suggesting that a D1/D5-PKA mechanism switches subthreshold Ca2+ hump potential to fire full Ca2+ spikes, which are eventually turned off by a D1/D5-Ca2+-dependent PKC mechanism. This depolarizing state-dependent, D1/D5-activated, bi-directional switching of soma-dendritic L-type Ca2+ channels via PKA-dependent potentiation and PKC-dependent suppression may provide spatiotemporal regulation of synaptic integration and plasticity in PFC.


Assuntos
Sinalização do Cálcio , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Córtex Pré-Frontal/fisiologia , Proteína Quinase C/metabolismo , Células Piramidais/fisiologia , Receptores de Dopamina D1/metabolismo , Animais , Cálcio/metabolismo , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio/metabolismo , Canais de Cálcio Tipo L/metabolismo , Células Cultivadas , Dendritos/enzimologia , Dendritos/fisiologia , Ativação Enzimática , Potenciais Evocados , Receptores de Inositol 1,4,5-Trifosfato , Transporte de Íons , Cinética , Masculino , Plasticidade Neuronal , Nimodipina/farmacologia , Técnicas de Patch-Clamp , Canais de Potássio/metabolismo , Córtex Pré-Frontal/citologia , Células Piramidais/citologia , Células Piramidais/enzimologia , Ratos , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores de Dopamina D5
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA