Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
2.
Nature ; 537(7618): 50-6, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27582220

RESUMO

Alzheimer's disease (AD) is characterized by deposition of amyloid-ß (Aß) plaques and neurofibrillary tangles in the brain, accompanied by synaptic dysfunction and neurodegeneration. Antibody-based immunotherapy against Aß to trigger its clearance or mitigate its neurotoxicity has so far been unsuccessful. Here we report the generation of aducanumab, a human monoclonal antibody that selectively targets aggregated Aß. In a transgenic mouse model of AD, aducanumab is shown to enter the brain, bind parenchymal Aß, and reduce soluble and insoluble Aß in a dose-dependent manner. In patients with prodromal or mild AD, one year of monthly intravenous infusions of aducanumab reduces brain Aß in a dose- and time-dependent manner. This is accompanied by a slowing of clinical decline measured by Clinical Dementia Rating-Sum of Boxes and Mini Mental State Examination scores. The main safety and tolerability findings are amyloid-related imaging abnormalities. These results justify further development of aducanumab for the treatment of AD. Should the slowing of clinical decline be confirmed in ongoing phase 3 clinical trials, it would provide compelling support for the amyloid hypothesis.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/psicologia , Peptídeos beta-Amiloides/antagonistas & inibidores , Peptídeos beta-Amiloides/metabolismo , Anticorpos Monoclonais Humanizados/uso terapêutico , Placa Amiloide/tratamento farmacológico , Placa Amiloide/metabolismo , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Amiloide/efeitos dos fármacos , Amiloide/metabolismo , Peptídeos beta-Amiloides/química , Animais , Anticorpos Monoclonais Humanizados/administração & dosagem , Anticorpos Monoclonais Humanizados/efeitos adversos , Anticorpos Monoclonais Humanizados/farmacocinética , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Ensaios Clínicos Fase III como Assunto , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Modelos Biológicos , Placa Amiloide/patologia , Agregação Patológica de Proteínas/tratamento farmacológico , Solubilidade
3.
J Biol Chem ; 292(43): 17963-17974, 2017 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-28860188

RESUMO

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.


Assuntos
Precursores Enzimáticos/antagonistas & inibidores , Precursores Enzimáticos/química , Metaloproteinase 9 da Matriz/química , Inibidores de Metaloproteinases de Matriz/química , Regulação Alostérica , Animais , Células COS , Domínio Catalítico , Chlorocebus aethiops , Precursores Enzimáticos/genética , Precursores Enzimáticos/metabolismo , Humanos , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Domínios Proteicos
4.
Eur J Neurosci ; 39(7): 1225-33, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24713001

RESUMO

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.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Proteínas de Transporte/metabolismo , Potenciais Pós-Sinápticos Excitadores , Proteínas Nucleares/metabolismo , Fragmentos de Peptídeos/toxicidade , Sinapses/metabolismo , Animais , Proteínas de Transporte/genética , Proteínas de Ciclo Celular , Células Cultivadas , Hipocampo/citologia , Hipocampo/metabolismo , Hipocampo/fisiologia , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/fisiologia , Proteínas Nucleares/genética , Ligação Proteica , Ratos , Receptores de AMPA/metabolismo , Sinapses/efeitos dos fármacos , Sinapses/fisiologia
6.
SLAS Technol ; 28(2): 63-69, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36455858

RESUMO

The development of phenotypic assays with appropriate analyses is an important step in the drug discovery process. Assays using induced pluripotent stem cell (iPSC)-derived human neurons are emerging as powerful tools for drug discovery in neurological disease. We have previously shown that longitudinal single cell tracking enabled the quantification of survival and death of neurons after overexpression of α-synuclein with a familial Parkinson's disease mutation (A53T). The reliance of this method on manual counting, however, rendered the process labor intensive, time consuming and error prone. To overcome these hurdles, we have developed automated detection algorithms for neurons using the BioStation CT live imaging system and CL-Quant software. In the current study, we use these algorithms to successfully measure the risk of neuronal death caused by overexpression of α-synuclein (A53T) with similar accuracy and improved consistency as compared to manual counting. This novel method also provides additional key readouts of neuronal fitness including total neurite length and the number of neurite nodes projecting from the cell body. Finally, the algorithm reveals the neuroprotective effects of brain-derived neurotrophic factor (BDNF) treatment in neurons overexpressing α-synuclein (A53T). These data show that an automated algorithm improves the consistency and considerably shortens the analysis time of assessing neuronal health, making this method advantageous for small molecule screening for inhibitors of synucleinopathy and other neurodegenerative diseases.


Assuntos
Sinucleinopatias , alfa-Sinucleína , Humanos , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , Sinucleinopatias/metabolismo , Rastreamento de Células , Neurônios/metabolismo , Algoritmos
7.
J Pharmacol Exp Ther ; 341(1): 274-84, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22267202

RESUMO

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.


Assuntos
Sistema Nervoso Central/citologia , Sistema Nervoso Central/metabolismo , Citoproteção/genética , Fumaratos/farmacologia , Fator 2 Relacionado a NF-E2/fisiologia , Neurônios/metabolismo , Estresse Oxidativo/genética , Transdução de Sinais/genética , Animais , Células Cultivadas , Sistema Nervoso Central/efeitos dos fármacos , Citoproteção/efeitos dos fármacos , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 2 Relacionado a NF-E2/deficiência , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
8.
Mol Neurobiol ; 59(4): 2171-2189, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35060064

RESUMO

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.


Assuntos
Doença de Parkinson , Estearoil-CoA Dessaturase , Animais , Ácidos Graxos/metabolismo , Ácidos Graxos/farmacologia , Metabolismo dos Lipídeos/fisiologia , Ratos , Estearoil-CoA Dessaturase/metabolismo , alfa-Sinucleína/metabolismo
9.
PLoS One ; 17(12): e0277532, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36454869

RESUMO

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.


Assuntos
Células-Tronco Pluripotentes Induzidas , Doença de Parkinson , Humanos , alfa-Sinucleína/genética , Doença de Parkinson/genética , Organoides , Ácidos Graxos
10.
Neurotherapeutics ; 19(3): 1018-1036, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35445353

RESUMO

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.


Assuntos
Doença de Parkinson , alfa-Sinucleína , Animais , Encéfalo/metabolismo , Humanos , Camundongos , Neurônios/metabolismo , Doença de Parkinson/genética , Estearoil-CoA Dessaturase/genética , Estearoil-CoA Dessaturase/metabolismo , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
11.
Chem Res Toxicol ; 24(7): 1012-30, 2011 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-21667953

RESUMO

2-Amino-4-phenyl-8-pyrrolidin-1-ylmethyl-indeno[1,2-d]pyrimidin-5-one (1) is a novel and potent selective dual A(2A)/A(1) adenosine receptor antagonist from the arylindenopyrimidine series that was determined to be genotoxic in both the Ames and Mouse Lymphoma L5178Y assays only following metabolic activation. Compound 1 was identified as a frame-shift mutagen in Salmonella typhimurium tester strain TA1537 as indicated by a significant dose-dependent increase in revertant colonies as compared to the vehicle control. The metabolic activation-dependent irreversible covalent binding of radioactivity to DNA, recovery of 1 and its enamine metabolite from acid hydrolysis of covalently modified DNA, and protection of covalent binding to DNA by both cyanide ion and methoxylamine suggest that the frame-shift mutation in TA1537 strain involved covalent binding instead of simple intercalation to DNA. Compound 1 was bioactivated to endocyclic iminium ion, aldehyde, epoxide, and α,ß-unsaturated keto reactive intermediates from the detection of cyano, oxime, and glutathione conjugates by data-dependent high resolution accurate mass measurements. Collision-induced dissociation of these conjugates provided evidence for bioactivation of the pyrrolidine ring of 1. The epoxide and α,ß-unsaturated keto reactive intermediates were unlikely to cause the genotoxicity of 1 because the formation of their glutathione adducts did not ameliorate the binding of compound related material to DNA. Instead, the endocyclic iminium ions and amino aldehydes were likely candidates responsible for genotoxicity based on, first, the protection afforded by both cyanide ion and methoxylamine, which reduced the potential to form covalent adducts with DNA, and, second, analogues of 1 designed with low probability to form these reactive intermediates were not genotoxic. It was concluded that 1 also had the potential to be mutagenic in humans based on observing the endocyclic iminium ion following incubation with a human liver S9 preparation and the commensurate detection of DNA adducts. An understanding of this genotoxicity mechanism supported an evidence-based approach to selectively modify the structure of 1 which resulted in analogues being synthesized that were devoid of a genotoxic liability. In addition, potency and selectivity against both adenosine A(2A) and A(1) receptors were maintained.


Assuntos
Antagonistas do Receptor A1 de Adenosina/toxicidade , Antagonistas do Receptor A2 de Adenosina/toxicidade , Iminas/química , Indenos/toxicidade , Pirimidinas/química , Pirimidinas/toxicidade , Pirrolidinas/toxicidade , Receptor A1 de Adenosina/química , Receptor A2A de Adenosina/química , Antagonistas do Receptor A1 de Adenosina/química , Antagonistas do Receptor A1 de Adenosina/metabolismo , Antagonistas do Receptor A2 de Adenosina/química , Antagonistas do Receptor A2 de Adenosina/metabolismo , Animais , Biotransformação , Cromatografia Líquida de Alta Pressão , DNA/química , DNA/metabolismo , Adutos de DNA/química , Adutos de DNA/metabolismo , Humanos , Indenos/química , Íons/química , Espectrometria de Massas , Camundongos , Testes de Mutagenicidade , Pirrolidinas/química , Pirrolidinas/metabolismo , Ratos , Receptor A1 de Adenosina/metabolismo , Receptor A2A de Adenosina/metabolismo , Salmonella typhimurium/efeitos dos fármacos , Salmonella typhimurium/genética
12.
Bioorg Med Chem Lett ; 21(21): 6485-90, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-21925881

RESUMO

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.


Assuntos
Azepinas/farmacologia , Inibidores de Metaloproteinases de Matriz , Inibidores de Proteases/farmacologia , Azepinas/farmacocinética , Disponibilidade Biológica , Ciclização , Descoberta de Drogas , Concentração Inibidora 50 , Inibidores de Proteases/farmacocinética , Estereoisomerismo
13.
Bioorg Med Chem Lett ; 21(24): 7277-80, 2011 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-22061640

RESUMO

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.


Assuntos
Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Anti-Inflamatórios não Esteroides/química , Inibidores Enzimáticos/química , Fenilacetatos/química , Piperidinas/química , Administração Oral , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Anti-Inflamatórios não Esteroides/síntese química , Anti-Inflamatórios não Esteroides/farmacocinética , Encéfalo/metabolismo , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/farmacocinética , Camundongos , Fragmentos de Peptídeos/metabolismo , Fenilacetatos/síntese química , Fenilacetatos/farmacocinética , Piperidinas/síntese química , Piperidinas/farmacocinética , Ratos
15.
Bioorg Med Chem Lett ; 20(9): 2868-71, 2010 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-20338760

RESUMO

Two reactive metabolites were identified in vivo for the dual A(2A)/A(1) receptor antagonist 1. Two strategies were implemented to successfully mitigate the metabolic liabilities associated with 1. Optimization of the arylindenopyrimidines led to a number of amide, ether, and amino analogs having comparable in vitro and in vivo activity.


Assuntos
Antagonistas do Receptor A1 de Adenosina , Antagonistas do Receptor A2 de Adenosina , Neurotransmissores/química , Pirimidinas/química , Animais , Catalepsia/tratamento farmacológico , Modelos Animais de Doenças , Camundongos , Neurotransmissores/síntese química , Neurotransmissores/uso terapêutico , Pirimidinas/síntese química , Pirimidinas/uso terapêutico , Receptor A1 de Adenosina/metabolismo , Receptor A2A de Adenosina/metabolismo , Relação Estrutura-Atividade
16.
Neuron ; 41(4): 587-98, 2004 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-14980207

RESUMO

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.


Assuntos
Proteínas de Ligação ao Cálcio/química , Membrana Celular/química , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/química , Sequência de Aminoácidos/fisiologia , Animais , Sítios de Ligação/genética , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Linhagem Celular , Membrana Celular/genética , Membrana Celular/metabolismo , Cristalografia por Raios X , Humanos , Proteínas Interatuantes com Canais de Kv , Potenciais da Membrana/genética , Modelos Moleculares , Mutagênese Sítio-Dirigida/genética , Oócitos/metabolismo , Técnicas de Patch-Clamp , Canais de Potássio/genética , Canais de Potássio/metabolismo , Ligação Proteica/genética , Estrutura Terciária de Proteína/genética , Subunidades Proteicas , Canais de Potássio Shal
17.
Stroke ; 39(12): 3372-7, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18927459

RESUMO

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.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Fibrinolíticos/uso terapêutico , Embolia Intracraniana/tratamento farmacológico , Inibidores de Metaloproteinases de Matriz , Minociclina/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Terapia Trombolítica/métodos , Ativador de Plasminogênio Tecidual/uso terapêutico , Animais , Biomarcadores , Barreira Hematoencefálica/efeitos dos fármacos , Isquemia Encefálica/sangue , Isquemia Encefálica/enzimologia , Isquemia Encefálica/etiologia , Hemorragia Cerebral/sangue , Hemorragia Cerebral/induzido quimicamente , Hemorragia Cerebral/enzimologia , Hemorragia Cerebral/prevenção & controle , Infarto Cerebral/sangue , Infarto Cerebral/enzimologia , Infarto Cerebral/etiologia , Infarto Cerebral/prevenção & controle , Esquema de Medicação , Avaliação Pré-Clínica de Medicamentos , Fibrinolíticos/administração & dosagem , Fibrinolíticos/efeitos adversos , Embolia Intracraniana/sangue , Embolia Intracraniana/complicações , Embolia Intracraniana/enzimologia , Masculino , Metaloproteinase 9 da Matriz/sangue , Minociclina/administração & dosagem , Fármacos Neuroprotetores/administração & dosagem , Ratos , Ratos Endogâmicos SHR , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/efeitos adversos , Proteínas Recombinantes/uso terapêutico , Reperfusão , Terapia Trombolítica/efeitos adversos , Fatores de Tempo , Ativador de Plasminogênio Tecidual/administração & dosagem , Ativador de Plasminogênio Tecidual/efeitos adversos
18.
Bioorg Med Chem Lett ; 18(3): 1135-9, 2008 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-18086526

RESUMO

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.


Assuntos
Gelatinases/antagonistas & inibidores , Ácidos Hidroxâmicos/síntese química , Ácidos Hidroxâmicos/farmacologia , Inibidores de Metaloproteinases de Matriz , Pirazinas/síntese química , Pirazinas/farmacologia , Animais , Desenho de Fármacos , Humanos , Ácidos Hidroxâmicos/química , Microssomos Hepáticos/efeitos dos fármacos , Estrutura Molecular , Pirazinas/química , Ratos , Relação Estrutura-Atividade , Sulfonamidas/síntese química , Sulfonamidas/química , Sulfonamidas/farmacologia
19.
Bioorg Med Chem Lett ; 18(3): 1140-5, 2008 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-18083558

RESUMO

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.


Assuntos
Aminoácidos/química , Gelatinases/antagonistas & inibidores , Ácidos Hidroxâmicos/síntese química , Ácidos Hidroxâmicos/farmacologia , Inibidores de Metaloproteinases de Matriz , Pirazinas/síntese química , Pirazinas/farmacologia , Animais , Edema Encefálico/induzido quimicamente , Modelos Animais de Doenças , Desenho de Fármacos , Humanos , Ácidos Hidroxâmicos/química , Microssomos Hepáticos/efeitos dos fármacos , Artéria Cerebral Média/efeitos dos fármacos , Estrutura Molecular , Pirazinas/química , Ratos , Estereoisomerismo , Relação Estrutura-Atividade , Sulfonamidas/síntese química , Sulfonamidas/química , Sulfonamidas/farmacologia
20.
Bioorg Med Chem Lett ; 18(1): 409-13, 2008 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-17981034

RESUMO

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.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Inibidores de Metaloproteinases de Matriz , Inibidores de Proteases/síntese química , Piridonas/síntese química , Piridonas/farmacologia , Administração Oral , Animais , Disponibilidade Biológica , Modelos Animais de Doenças , Concentração Inibidora 50 , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Moleculares , Inibidores de Proteases/química , Inibidores de Proteases/farmacocinética , Inibidores de Proteases/farmacologia , Piridonas/química , Piridonas/farmacocinética , Relação Estrutura-Atividade , Sulfonamidas/síntese química , Sulfonamidas/química , Sulfonamidas/farmacocinética , Sulfonamidas/farmacologia , Zinco/química , Zinco/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa