Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Alzheimers Dement ; 12(1): 34-48, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26327236

RESUMO

INTRODUCTION: Blast traumatic brain injury (B-TBI) affects military and civilian personnel. Presently, there are no approved drugs for blast brain injury. METHODS: Exendin-4 (Ex-4), administered subcutaneously, was evaluated as a pretreatment (48 hours) and postinjury treatment (2 hours) on neurodegeneration, behaviors, and gene expressions in a murine open field model of blast injury. RESULTS: B-TBI induced neurodegeneration, changes in cognition, and genes expressions linked to dementia disorders. Ex-4, administered preinjury or postinjury, ameliorated B-TBI-induced neurodegeneration at 72 hours, memory deficits from days 7-14, and attenuated genes regulated by blast at day 14 postinjury. DISCUSSION: The present data suggest shared pathologic processes between concussive and B-TBI, with end points amenable to beneficial therapeutic manipulation by Ex-4. B-TBI-induced dementia-related gene pathways and cognitive deficits in mice somewhat parallel epidemiologic studies of Barnes et al. who identified a greater risk in US military veterans who experienced diverse TBIs, for dementia in later life.


Assuntos
Traumatismos por Explosões/tratamento farmacológico , Concussão Encefálica/tratamento farmacológico , Transtornos Cognitivos/prevenção & controle , Peptídeo 1 Semelhante ao Glucagon/agonistas , Peptídeos/uso terapêutico , Peçonhas/uso terapêutico , Animais , Traumatismos por Explosões/patologia , Concussão Encefálica/metabolismo , Concussão Encefálica/patologia , Cognição/efeitos dos fármacos , Exenatida , Expressão Gênica/efeitos dos fármacos , Injeções Subcutâneas , Masculino , Camundongos , Camundongos Endogâmicos ICR , Fármacos Neuroprotetores/administração & dosagem , Peptídeos/farmacologia , Peçonhas/farmacologia
2.
J Neuroinflammation ; 9: 106, 2012 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-22642825

RESUMO

BACKGROUND: Neuroinflammation is associated with virtually all major neurodegenerative disorders, including Alzheimer's disease (AD). Although it remains unclear whether neuroinflammation is the driving force behind these disorders, compelling evidence implicates its role in exacerbating disease progression, with a key player being the potent proinflammatory cytokine TNF-α. Elevated TNF-α levels are commonly detected in the clinic and animal models of AD. METHODS: The potential benefits of a novel TNF-α-lowering agent, 3,6'-dithiothalidomide, were investigated in cellular and rodent models of neuroinflammation with a specific focus on AD. These included central and systemic inflammation induced by lipopolysaccharide (LPS) and Aß(1-42) challenge, and biochemical and behavioral assessment of 3xTg-AD mice following chronic 3,6'-dithiothaliodmide. RESULTS: 3,6'-Dithiothaliodmide lowered TNF-α, nitrite (an indicator of oxidative damage) and secreted amyloid precursor protein (sAPP) levels in LPS-activated macrophage-like cells (RAW 264.7 cells). This translated into reduced central and systemic TNF-α production in acute LPS-challenged rats, and to a reduction of neuroinflammatory markers and restoration of neuronal plasticity following chronic central challenge of LPS. In mice centrally challenged with A(ß1-42) peptide, prior systemic 3,6'-dithiothalidomide suppressed Aß-induced memory dysfunction, microglial activation and neuronal degeneration. Chronic 3,6'-dithiothalidomide administration to an elderly symptomatic cohort of 3xTg-AD mice reduced multiple hallmark features of AD, including phosphorylated tau protein, APP, Aß peptide and Aß-plaque number along with deficits in memory function to levels present in younger adult cognitively unimpaired 3xTg-AD mice. Levels of the synaptic proteins, SNAP25 and synaptophysin, were found to be elevated in older symptomatic drug-treated 3xTg-AD mice compared to vehicle-treated ones, indicative of a preservation of synaptic function during drug treatment. CONCLUSIONS: Our data suggest a strong beneficial effect of 3,6'-dithiothalidomide in the setting of neuroinflammation and AD, supporting a role for neuroinflammation and TNF-α in disease progression and their targeting as a means of clinical management.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/patologia , Modelos Animais de Doenças , Talidomida/análogos & derivados , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/fisiologia , Doença de Alzheimer/fisiopatologia , Animais , Biomarcadores/metabolismo , Inflamação/tratamento farmacológico , Inflamação/patologia , Inflamação/fisiopatologia , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Ratos , Ratos Endogâmicos F344 , Talidomida/farmacologia , Talidomida/uso terapêutico
3.
Proc Natl Acad Sci U S A ; 106(4): 1285-90, 2009 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-19164583

RESUMO

Glucagon-like peptide-1 (GLP-1) is an endogenous insulinotropic peptide secreted from the gastrointestinal tract in response to food intake. It enhances pancreatic islet beta-cell proliferation and glucose-dependent insulin secretion, and lowers blood glucose and food intake in patients with type 2 diabetes mellitus (T2DM). A long-acting GLP-1 receptor (GLP-1R) agonist, exendin-4 (Ex-4), is the first of this new class of antihyperglycemia drugs approved to treat T2DM. GLP-1Rs are coupled to the cAMP second messenger pathway and, along with pancreatic cells, are expressed within the nervous system of rodents and humans, where receptor activation elicits neurotrophic actions. We detected GLP-1R mRNA expression in both cultured embryonic primary cerebral cortical and ventral mesencephalic (dopaminergic) neurons. These cells are vulnerable to hypoxia- and 6-hydroxydopamine-induced cell death, respectively. We found that GLP-1 and Ex-4 conferred protection in these cells, but not in cells from Glp1r knockout (-/-) mice. Administration of Ex-4 reduced brain damage and improved functional outcome in a transient middle cerebral artery occlusion stroke model. Ex-4 treatment also protected dopaminergic neurons against degeneration, preserved dopamine levels, and improved motor function in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of Parkinson's disease (PD). Our findings demonstrate that Ex-4 can protect neurons against metabolic and oxidative insults, and they provide preclinical support for the therapeutic potential for Ex-4 in the treatment of stroke and PD.


Assuntos
Citoproteção , Dopamina/metabolismo , Neurônios/patologia , Doença de Parkinson/patologia , Receptores de Glucagon/metabolismo , Acidente Vascular Cerebral/patologia , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina , Animais , Infarto Encefálico/tratamento farmacológico , Infarto Encefálico/patologia , Morte Celular/efeitos dos fármacos , Hipóxia Celular/efeitos dos fármacos , Células Cultivadas , Córtex Cerebral/citologia , Citoproteção/efeitos dos fármacos , Modelos Animais de Doenças , Embrião de Mamíferos/citologia , Exenatida , Regulação da Expressão Gênica/efeitos dos fármacos , Receptor do Peptídeo Semelhante ao Glucagon 1 , Humanos , Mesencéfalo/citologia , Camundongos , Neurônios/efeitos dos fármacos , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/metabolismo , Peptídeos/farmacologia , Peptídeos/uso terapêutico , Ratos , Receptores de Glucagon/genética , Acidente Vascular Cerebral/tratamento farmacológico , Acidente Vascular Cerebral/metabolismo , Resultado do Tratamento , Peçonhas/farmacologia , Peçonhas/uso terapêutico
4.
Bioorg Med Chem ; 19(13): 3965-72, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21658960

RESUMO

Eight novel 2-(2,6-dioxopiperidin-3-yl)phthalimidine EM-12 dithiocarbamates 9 and 10, N-substituted 3-(phthalimidin-2-yl)-2,6-dioxopiperidines 11-14 and 3-substituted 2,6-dioxopiperidines 16 and 18 were synthesized as tumor necrosis factor-α (TNF-α) synthesis inhibitors. Synthesis involved utilization of a novel condensation approach, a one-pot reaction involving addition, iminium rearrangement and elimination, to generate the phthalimidine ring required for the creation of compounds 9-14. Agents were, thereafter, quantitatively assessed for their ability to suppress the synthesis on TNF-α in a lipopolysaccharide (LPS)-challenged mouse macrophage-like cellular screen, utilizing cultured RAW 264.7 cells. Whereas compounds 9, 14 and 16 exhibited potent TNF-α lowering activity, reducing TNF-α by up to 48% at 30 µM, compounds 12, 17 and 18 presented moderate TNF-α inhibitory action. The TNF-α lowering properties of these analogs proved more potent than that of revlimid (3) and thalidomide (1). In particular, N-dithiophthalimidomethyl-3-(phthalimidin-2-yl)-2,6-dioxopiperidine 14 not only possessed the greatest potency of the analogs to reduce TNF-α synthesis, but achieved this with minor cellular toxicity at 30 µM. The pharmacological focus of the presented compounds is towards the development of well-tolerated agents to ameliorate the neuroinflammation, that is, commonly associated with neurodegenerative disorders, epitomized by Alzheimer's disease and Parkinson's disease.


Assuntos
Ftalimidas/química , Piperidinas/química , Talidomida/análogos & derivados , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Animais , Linhagem Celular Tumoral , Desenho de Fármacos , Lenalidomida , Camundongos , Piperidinas/síntese química , Piperidinas/toxicidade , Talidomida/síntese química , Talidomida/química , Talidomida/toxicidade , Fator de Necrose Tumoral alfa/metabolismo
5.
J Neurochem ; 113(6): 1621-31, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20374430

RESUMO

Increasing evidence suggests that glucagon-like peptide-1 (GLP-1), an incretin hormone of current interest in type 2 diabetes, is neuroprotective in both cell culture and animal models. To characterize the neuroprotective properties of GLP-1 and associated underlying mechanisms, we over-expressed the GLP-1 receptor (GLP-1R) on human neuroblastoma SH-SY5Y cells to generate a neuronal culture system featuring enhanced GLP-1R signaling. In GLP-1R over-expressing SH-SY5Y (SH-hGLP-1R#9) cells, GLP-1 and the long-acting agonist exendin-4 stimulated cell proliferation and increased cell viability by 2-fold at 24 h at physiologically relevant concentrations. This GLP-1R-dependent action was mediated via the protein kinase A and phosphoinositide 3-kinase signaling pathways, with the MAPK pathway playing a minor role. GLP-1 and exendin-4 pretreatment dose-dependently protected SH-hGLP-1R#9 cells from hydrogen peroxide (H(2)O(2))- and 6-hydroxydopamine-induced cell death. This involved amelioration of elevated caspase 3 activity, down-regulation of pro-apoptotic Bax and up-regulation of anti-apoptotic Bcl-2 protein. In the presence of 6-hydroxydopamine, GLP-1's ability to lower caspase-3 activity was abolished with the phosphoinositide 3-kinase inhibitor, LY2940002, and partly reduced with the protein kinase A inhibitor, H89. Hence, GLP-1R mediated neurotrophic and anti-apoptotic actions co-contribute to the neuroprotective property of GLP-1 in neuronal cell cultures, and reinforce the potential therapeutic value of GLP-1R agonists in neurodegenerative disorders involving oxidative stress.


Assuntos
Proliferação de Células/efeitos dos fármacos , Peptídeo 1 Semelhante ao Glucagon/farmacologia , Fármacos Neuroprotetores/farmacologia , Receptores de Glucagon/metabolismo , Transdução de Sinais/fisiologia , Fator 4 Ativador da Transcrição/metabolismo , Adrenérgicos/toxicidade , Apoptose/efeitos dos fármacos , Bromodesoxiuridina/metabolismo , Butadienos/farmacologia , Caspase 3/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Colforsina/farmacologia , AMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Exenatida , Receptor do Peptídeo Semelhante ao Glucagon 1 , Humanos , Peróxido de Hidrogênio/toxicidade , Hipoglicemiantes/farmacologia , Neuroblastoma , Nitrilas/farmacologia , Oxidantes/toxicidade , Oxidopamina/toxicidade , Peptídeos/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Receptores de Glucagon/genética , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Transfecção/métodos , Peçonhas/farmacologia , Proteína X Associada a bcl-2/metabolismo
6.
Bioorg Med Chem ; 18(13): 4687-93, 2010 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-20627738

RESUMO

The N-monophenylcarbamate analogues of neostigmine methyl sulfate (6) and pyridostigmine bromide (8) together with their precursors (5), (7), and the N(1)-methylammonium analogues of (-)-phenserine (12), (-)-tolserine (14), (-)-cymserine (16) and (-)-phenethylcymserine (18) were synthesized to produce long-acting peripheral inhibitors of acetylcholinesterase or butyrylcholinesterase. Evaluation of their cholinesterase inhibition against human enzyme ex vivo demonstrated that, whereas compounds 5-8 possessed only marginal activity, 12, 14, 16 and 18 proved to be potent anticholinesterases. An extended duration of cholinesterase inhibition was determined in rodent, making them of potential interest as long-acting agents for myasthenia gravis.


Assuntos
Acetilcolinesterase/química , Butirilcolinesterase/química , Inibidores da Colinesterase/síntese química , Miastenia Gravis/tratamento farmacológico , Neostigmina/química , Fenilcarbamatos/química , Fisostigmina/química , Brometo de Piridostigmina/química , Acetilcolinesterase/metabolismo , Animais , Butirilcolinesterase/metabolismo , Inibidores da Colinesterase/química , Inibidores da Colinesterase/uso terapêutico , Humanos , Camundongos , Fenilcarbamatos/síntese química , Fenilcarbamatos/uso terapêutico
7.
Neuromolecular Med ; 9(4): 315-23, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17999205

RESUMO

Uric acid is a major antioxidant in the blood of humans that can protect cultured neurons against oxidative and metabolic insults. However, uric acid has a very low solubility which compromises its potential clinical use for neurodegenerative disorders. Here we describe the synthesis, characterization and preclinical development of neuroprotective methyl- and sulfur-containing analogs of uric acid with increased solubility. In vitro and cell culture screening identified 1,7-dimethyluric acid (mUA2) and 6,8-dithiouric acid (sUA2) as two analogs with high antioxidant and neuroprotective activities. When administered intravenously in mice, uric acid analogs mUA2 and sUA2 lessened damage to the brain and improved functional outcome in an ischemia-reperfusion mouse model of stroke. Analogs sUA2 and mUA2 were also effective in reducing damage to the cerebral cortex when administered up to 4 h after stroke onset in a permanent middle cerebral artery occlusion mouse model. These findings suggest a therapeutic potential for soluble analogs of uric acid in the treatment of stroke and related neurodegenerative conditions.


Assuntos
Antioxidantes/uso terapêutico , Lesões Encefálicas/tratamento farmacológico , Isquemia Encefálica/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Ácido Úrico/análogos & derivados , Ácido Úrico/uso terapêutico , Animais , Velocidade do Fluxo Sanguíneo/efeitos dos fármacos , Velocidade do Fluxo Sanguíneo/fisiologia , Técnicas de Cultura de Células , Circulação Cerebrovascular/efeitos dos fármacos , Modelos Animais de Doenças , Camundongos
8.
Sci Rep ; 7(1): 3735, 2017 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-28623327

RESUMO

Mild blast traumatic brain injury (B-TBI) induced lasting cognitive impairments in novel object recognition and less severe deficits in Y-maze behaviors. B-TBI significantly reduced the levels of synaptophysin (SYP) protein staining in cortical (CTX) and hippocampal (HIPP) tissues. Treatment with exendin-4 (Ex-4) delivered by subcutaneous micro-osmotic pumps 48 hours prior to or 2 hours immediately after B-TBI prevented the induction of both cognitive deficits and B-TBI induced changes in SYP staining. The effects of a series of biaxial stretch injuries (BSI) on a neuronal derived cell line, HT22 cells, were assessed in an in vitro model of TBI. Biaxial stretch damage induced shrunken neurites and cell death. Treatment of HT22 cultures with Ex-4 (25 to 100 nM), prior to injury, attenuated the cytotoxic effects of BSI and preserved neurite length similar to sham treated cells. These data imply that treatment with Ex-4 may represent a viable option for the management of secondary events triggered by blast-induced, mild traumatic brain injury that is commonly observed in militarized zones.


Assuntos
Traumatismos por Explosões/metabolismo , Lesões Encefálicas Traumáticas/prevenção & controle , Disfunção Cognitiva/prevenção & controle , Exenatida/farmacologia , Hipocampo/metabolismo , Sinaptofisina/metabolismo , Animais , Traumatismos por Explosões/patologia , Traumatismos por Explosões/prevenção & controle , Lesões Encefálicas Traumáticas/metabolismo , Lesões Encefálicas Traumáticas/patologia , Linhagem Celular , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/patologia , Modelos Animais de Doenças , Hipocampo/patologia , Masculino , Camundongos
9.
J Med Chem ; 49(7): 2174-85, 2006 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-16570913

RESUMO

A new enantiomeric synthesis utilizing classical resolution provided two novel series of optically active inhibitors of cholinesterase: (-)- and (+)-O-carbamoyl phenols of tetrahydrofurobenzofuran and methanobenzodioxepine. An additional two series of (-)- and (+)-O-carbamoyl phenols of pyrroloindole and furoindole were obtained by known procedures, and their anticholinesterase actions were similarly quantified against freshly prepared human acetyl- (AChE) and butyrylcholinesterase (BChE). Both enantiomeric forms of each series demonstrated potent cholinesterase inhibitory activity (with IC(50) values as low as 10 nM for AChE and 3 nM for BChE), with the exception of the (+)-O-carbamoyl phenols of pyrroloindole, which lacked activity (IC(50) values >1 microM). Based on the biological data of these four series, a structure-activity relationship (SAR) analysis was provided by molecular volume calculations. In addition, a probable transition-state model was established according to the known X-ray structure of a transition-state complex of Torpedo californica AChE-m-(N,N,N-trimethylammonio)-2,2,2-trifluoroacetophenone (TcAChE-TMTFA). This model proved valuable in explaining the enantioselectivity and enzyme subtype selectivity of each series. These carbamates are more potent than, or similarly potent to, anticholinesterases in current clinical use, providing not only inhibitors of potential clinical relevance but also pharmacological tools to define drug-enzyme binding interactions within an enzyme crucial in the maintenance of cognition and numerous systemic physiological functions in health, aging, and disease.


Assuntos
Acetilcolinesterase/química , Benzofuranos/síntese química , Butirilcolinesterase/química , Carbamatos/síntese química , Inibidores da Colinesterase/química , Furanos/síntese química , Compostos Heterocíclicos com 3 Anéis/síntese química , Oxepinas/síntese química , Acetofenonas/química , Animais , Benzofuranos/química , Carbamatos/química , Cristalografia por Raios X , Furanos/química , Compostos Heterocíclicos com 3 Anéis/química , Humanos , Modelos Moleculares , Oxepinas/química , Estereoisomerismo , Relação Estrutura-Atividade , Torpedo
10.
J Alzheimers Dis ; 10(1): 43-51, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16988481

RESUMO

An explosion in the incidence of neurodegenerative diseases, particularly Alzheimer's disease (AD), is predicted in coming decades. Hence, the need to devise and assess new treatment strategies has never been more acute. AD, although an irreversible and progressive disorder, is currently treated with palliative, symptomatic therapy: primarily with acetylcholinesterase (AChE) inhibitors to amplify remaining cholinergic activity. New agents that, additionally, affect disease progression are sorely needed. Inhibition of brain butyrylcholinesterase (BuChE) represents a new drug target for AD treatment. Therefore, hand-in-hand with the development of selective ligands to inhibit BuChE in brain, it is fundamental to optimize assay conditions for kinetic studies of human BuChE. Kinetic analysis of serum BuChE, which is structurally similar to brain enzyme, was performed at dual substrate (butyrylthiocholine iodide) concentration ranges: 3-80 microM (low) and 25-800 microM (optimal) by use of the Ellman technique. Interaction of BuChE with a novel experimental AD therapeutic, bisnorcymserine (BNC; 0.06-2.0 nM) was also studied ex vivo. The IC_{50} and other key kinetic constants were determined for human serum BuChE inhibition by BNC, which proved to be a highly potent inhibitor in comparison to its structural analogue, cymserine. BNC may, additionally, lower the amyloid plaque-associated protein, amyloid-beta peptide. In synopsis, the characterization of the kinetic parameters of BuChE and BNC, described herein, is both aiding in the design of novel agents and optimizing their translation toward clinical use.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Butirilcolinesterase/metabolismo , Inibidores da Colinesterase/farmacocinética , Inibidores da Colinesterase/uso terapêutico , Fisostigmina/análogos & derivados , Fisostigmina/uso terapêutico , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/enzimologia , Encéfalo/patologia , Inibidores da Colinesterase/farmacologia , Humanos , Fisostigmina/farmacocinética , Fisostigmina/farmacologia , Fatores de Tempo
11.
PLoS One ; 11(6): e0156493, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27254111

RESUMO

Traumatic brain injury (TBI), often caused by a concussive impact to the head, affects an estimated 1.7 million Americans annually. With no approved drugs, its pharmacological treatment represents a significant and currently unmet medical need. In our prior development of the anti-cholinesterase compound phenserine for the treatment of neurodegenerative disorders, we recognized that it also possesses non-cholinergic actions with clinical potential. Here, we demonstrate neuroprotective actions of phenserine in neuronal cultures challenged with oxidative stress and glutamate excitotoxicity, two insults of relevance to TBI. These actions translated into amelioration of spatial and visual memory impairments in a mouse model of closed head mild TBI (mTBI) two days following cessation of clinically translatable dosing with phenserine (2.5 and 5.0 mg/kg BID x 5 days initiated post mTBI) in the absence of anti-cholinesterase activity. mTBI elevated levels of thiobarbituric acid reactive substances (TBARS), a marker of oxidative stress. Phenserine counteracted this by augmenting homeostatic mechanisms to mitigate oxidative stress, including superoxide dismutase [SOD] 1 and 2, and glutathione peroxidase [GPx], the activity and protein levels of which were measured by specific assays. Microarray analysis of hippocampal gene expression established that large numbers of genes were exclusively regulated by each individual treatment with a substantial number of them co-regulated between groups. Molecular pathways associated with lipid peroxidation were found to be regulated by mTBI, and treatment of mTBI animals with phenserine effectively reversed injury-induced regulations in the 'Blalock Alzheimer's Disease Up' pathway. Together these data suggest that multiple phenserine-associated actions underpin this compound's ability to ameliorate cognitive deficits caused by mTBI, and support the further evaluation of the compound as a therapeutic for TBI.


Assuntos
Concussão Encefálica/tratamento farmacológico , Disfunção Cognitiva/tratamento farmacológico , Estresse Oxidativo/efeitos dos fármacos , Fisostigmina/análogos & derivados , Animais , Concussão Encefálica/complicações , Concussão Encefálica/patologia , Colinérgicos/administração & dosagem , Inibidores da Colinesterase/administração & dosagem , Disfunção Cognitiva/etiologia , Disfunção Cognitiva/patologia , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Ácido Glutâmico/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Humanos , Peroxidação de Lipídeos/efeitos dos fármacos , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/patologia , Fisostigmina/administração & dosagem
12.
J Med Chem ; 48(4): 986-94, 2005 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-15715468

RESUMO

Reductive cyclization of 5-hydroxy-3-methyl-3-methoxycarbonylmethylenebenzofuran-2(3H)-one (4) gave 5-hydroxy-3a-methyl-2,3,3a,8a-tatrahydrofuro[2,3-b]benzofuran (5) and the rearrangement product 7-hydroxy-4,5-dihydro-2,5-methano-1,3-benzodioxepine (6). Reaction of compounds 5 and 6 with different isocyanates provided two series novel carbamates (7-12) whose structures were confirmed by X-ray crystallography. These were assessed for anticholinesterase action against freshly prepared human enzyme and proved to be potent inhibitors of either acetyl- (AChE) or butyrylcholinesterase (BChE) with specific compounds exhibiting remarkable selectivity. Because the two series of carbamates (7-12) differ in their phenolic moieties, their respective potency and selectivity for AChE versus BChE was governed by their N-substituted groups. This same characteristic was also present in a series of physovenine analogues (1, 13, 15, 17) and physostigmine analogues (2, 14, 16, 18). These structure-activity relations proved valuable in elucidating the mechanisms underpinning the interaction between carbamate-based cholinesterase inhibitors and their enzyme target. In addition, because physostigmine analogues have demonstrated activity in lowering the Alzheimer's disease protein, amyloid precursor protein (APP), examples of the two new series of carbamates were characterized in culture assays of quantifying cell viability and synthesis of APP.


Assuntos
Acetilcolinesterase/metabolismo , Benzofuranos/síntese química , Carbamatos/síntese química , Inibidores da Colinesterase/síntese química , Oxepinas/síntese química , Acetilcolinesterase/química , Precursor de Proteína beta-Amiloide/biossíntese , Precursor de Proteína beta-Amiloide/metabolismo , Benzofuranos/química , Benzofuranos/farmacologia , Butirilcolinesterase/química , Carbamatos/química , Carbamatos/farmacologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Inibidores da Colinesterase/química , Inibidores da Colinesterase/farmacologia , Cristalografia por Raios X , Humanos , Modelos Moleculares , Estrutura Molecular , Neurônios/citologia , Oxepinas/química , Oxepinas/farmacologia , Relação Estrutura-Atividade
13.
J Med Chem ; 48(6): 1919-29, 2005 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-15771436

RESUMO

Tacrine heterobivalent ligands were designed as novel and reversible inhibitors of cholinesterases. On the basis of the investigation of the active site gorge topology of butyrylcholinesterase (BuChE) and acetylcholinesterase (AChE) and by using flexible docking procedures, molecular modeling studies formulated the hypothesis of extra interaction sites in the active gorge of hBuChE, namely, a mid-gorge interaction site and a peripheral interaction site. The design strategy led to novel BuChE inhibitors, balancing potency and selectivity. Among the compounds identified, the heterobivalent ligand 4m, containing an amide nitrogen and a sulfur atom at the 8-membered tether level, is one of the most potent and selective BuChE inhibitors described to date. The novel inhibitors, bearing postulated key features, validated the hypothesis of the presence of extra interaction sites within the hBuChE active site gorge.


Assuntos
Acridinas/síntese química , Butirilcolinesterase/química , Inibidores da Colinesterase/síntese química , Acetilcolinesterase/metabolismo , Acridinas/química , Acridinas/farmacologia , Sítios de Ligação , Butirilcolinesterase/metabolismo , Inibidores da Colinesterase/química , Inibidores da Colinesterase/farmacologia , Desenho de Fármacos , Humanos , Técnicas In Vitro , Ligantes , Modelos Moleculares , Sondas Moleculares , Relação Estrutura-Atividade
14.
Curr Alzheimer Res ; 2(4): 483-92, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16248851

RESUMO

OBJECTIVE: To evaluate the safety, maximum tolerated dose (MTD), pharmacokinetics (PK), and pharmacodynamics (PD) of the acetyl-selective anticholinesterase, phenserine tartrate, in healthy elderly subjects. METHODS: 32 healthy elderly volunteers received single oral doses of phenserine tartrate (5-20 mg). Physical and vital signs were monitored over the ensuing 24 hours. Analyses were performed on plasma samples to determine PK, and PD were assessed using an erythrocyte acetylcholinesterase (AChE) inhibition assay. RESULTS: No serious adverse events (AEs) occurred; the most common were headache and vomiting. The MTD of phenserine tartrate was 10 mg. The Cmax and AUC(0-24) of phenserine increased with dose, but neither were dose-proportional. Subjects receiving 10 mg of phenserine tartrate had a Cmax of 1.95 ng/mL at 1.5 hours, and the mean peak inhibition (Imax) of AChE was 26% (range: 18-34%) at 1.75 hours (tImax) following dosing. The half-life of AChE inhibition (tI1/2) was 11 hours. Evaluation of PK/PD relationships suggested a linear correlation between plasma phenserine concentration and AChE inhibition in the blood. CONCLUSIONS: Phenserine tartrate was safe and well tolerated when administered as a single oral dose of either 5 mg or 10 mg. An increase in the severity and frequency of AEs occurred at the 20 mg dose level.


Assuntos
Inibidores da Colinesterase/efeitos adversos , Inibidores da Colinesterase/farmacocinética , Fisostigmina/análogos & derivados , Acetilcolinesterase/efeitos dos fármacos , Idoso , Área Sob a Curva , Eritrócitos/efeitos dos fármacos , Eritrócitos/enzimologia , Feminino , Humanos , Masculino , Dose Máxima Tolerável , Pessoa de Meia-Idade , Fisostigmina/efeitos adversos , Fisostigmina/farmacocinética
15.
Eur J Pharmacol ; 517(3): 186-90, 2005 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-15967428

RESUMO

Butyrylcholinesterase is a major cocaine-metabolizing enzyme in humans and other primates, catalyzing hydrolysis to ecgonine methylester. Increasing butyrylcholinesterase activity may be a treatment for cocaine addiction. We evaluated the effect of 30-min pretreatment with horse-derived butyrylcholinesterase (5-15,000 U i.v.) or with the selective butyrylcholinesterase inhibitor cymserine (10 mg/kg i.v.) on the metabolism of cocaine (17 mg/kg i.p.) in anesthetized rats. Venous blood samples were collected for two hours after cocaine administration and later assayed for cocaine and metabolites by gas chromatography/mass spectroscopy. Whole brains were collected after the last blood sample and similarly assayed. Butyrylcholinesterase significantly increased plasma and brain ecgonine methylester levels and decreased cocaine plasma half-life from 26.2 min (saline) to 16.4 min (15,000 U). Butyrylcholinesterase had no significant effect on plasma or brain cocaine or benzoylecgonine levels. Cymserine had no effect on any variable. These findings suggest that butyrylcholinesterase treatment may have benefits in enhancing cocaine metabolism and in increasing levels of ecgonine methylester, which may have a protective action against cocaine.


Assuntos
Encéfalo/metabolismo , Butirilcolinesterase/farmacologia , Cocaína/farmacocinética , Animais , Butirilcolinesterase/administração & dosagem , Butirilcolinesterase/metabolismo , Inibidores da Colinesterase/administração & dosagem , Inibidores da Colinesterase/farmacologia , Cocaína/análogos & derivados , Cocaína/sangue , Cocaína/metabolismo , Relação Dose-Resposta a Droga , Cromatografia Gasosa-Espectrometria de Massas , Meia-Vida , Injeções Intraperitoneais , Injeções Intravenosas , Masculino , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Vasoconstritores/sangue , Vasoconstritores/metabolismo , Vasoconstritores/farmacocinética
16.
Life Sci ; 76(10): 1073-81, 2005 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-15620572

RESUMO

Male Fischer-344 rats (n = 38) at 5 months old were tested in a Morris water maze to determine if treatment with the cholinesterase inhibitor, phenserine (PHEN), would overcome a learning impairment induced by scopolamine (SCOP), a muscarinic cholinergic receptor antagonist. Each rat was randomly assigned to one of five groups to receive two intraperitoneal injections 60 and 30 min, prior to testing, respectively, as follows: (1) saline-saline (SAL); (2) saline-1.0 mg/kg (SCOP); (3) 2 mg/kg PHEN- SCOP (PHEN2); (4) 4 mg/kg PHEN-SCOP (PHEN4); and (5) 1 mg/kg PHEN-SAL (PHEN1). Maze testing occurred across 5 days with 4 days of acquisition trials (4 trials per day) and a fifth day consisting of a single 120 sec probe trial. PHEN1 and SAL were combined into one control (CON) group for purposes of statistical analysis for both acquisition and probe trials as comparison of the two groups revealed that they did not significantly differ on any measure. SCOP-treated rats were significantly impaired compared to CON in learning the location of the submerged platform as measured by latency to locate the platform and the distance traversed to find the platform across days of testing. The PHEN4 group had significantly lower latencies and traveled a shorter distance to reach the submerged platform when compared to SCOP on the fourth day of trials while the PHEN2 group traveled more directly to the submerged platform but did not have shorter latencies than the SCOP group. For probe trials, CON rats swam closer to the target area (a measure of proximity to the removed platform) than did all other groups, and the PHEN4 group swam in an area more proximate to the target area than did the SCOP-treated group. These findings demonstrate the ability of this drug to improve learning when cholinergic function has been impaired in a spatial memory task.


Assuntos
Inibidores da Colinesterase/farmacologia , Aprendizagem em Labirinto/efeitos dos fármacos , Fisostigmina/análogos & derivados , Fisostigmina/farmacologia , Escopolamina/farmacologia , Animais , Masculino , Ratos , Ratos Endogâmicos F344
17.
Eur J Neurosci ; 4(7): 653-662, 1992.
Artigo em Inglês | MEDLINE | ID: mdl-12106329

RESUMO

After unilateral destruction of the nucleus basalis magnocellularis (NBM) in 3-month-old rats, which reduces cholinergic inputs to the ipsilateral frontoparietal neocortex, regional cerebral metabolic rates for glucose (rCMRglc) of denervated cortex are initially reduced, but nearly normalize by 2 weeks. To examine functional reorganization of the brain after unilateral destruction of the NBM, a correlation analysis of rCMRglc was performed on two groups of 16 young rats 2 weeks after stereotaxic ablation of the right NBM with ibotenate or sham surgery. rCMRglc was measured in 117 brain regions of awake rats with the [14C]deoxyglucose method. For each region pair, a partial correlation coefficient was calculated for rCMRglc across animals. Most correlations between cholinergic nuclei of both left and right forebrain (medial septum and diagonal band) and right (66/72, mean increase 0.44) but not left (39/72) frontoparietal cortical regions were larger (P < 0.001) in lesioned rats, as were those between most frontoparietal region pairs (516/630, P < 0.001). These results suggest that, after unilateral NBM ablation, (1) functional interactions are established between the remaining cholinergic forebrain and the deafferented cortex, (2) the neocortex becomes more integrated, and (3) functional reorganization involves both cortical hemispheres. These changes do not correspond to those reported to occur in Alzheimer's disease.

18.
J Med Chem ; 45(17): 3684-91, 2002 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-12166941

RESUMO

A series of phenylcarbamate analogues of geneserine (8, 10, 12, 14) were synthesized from their counterparts, the phenylcarbamate analogues of physostigmine (2-5), by oxidation. The geneserine analogues can undergo tautomerism between N-oxide and 1,2-oxazine structures in a pH- and time-dependent manner. Assessment by (1)H NMR indicated that the N-oxide structure is adopted at neutral pH and that the compound exists in an equilibrium between several epimers. Evaluation of their biological action to inhibit human acetylcholinesterase (AChE) and butyrylcholinesterase (BChE), ex vivo, demonstrated that the N-oxide (7, 9, 11, 13, 15) and 1,2-oxazine (6, 8, 10, 12, 14) structures possessed similar potencies against AChE, but the latter structures were more potent against BChE. With the exception of the BChE selective inhibitor, 12, none of the geneserine analogues were as potent or enzyme subtype selective as their physostigmine analogue counterparts.


Assuntos
Alcaloides/química , Inibidores da Colinesterase/síntese química , Óxidos N-Cíclicos/síntese química , Oxazinas/síntese química , Acetilcolinesterase/química , Butirilcolinesterase/química , Inibidores da Colinesterase/química , Óxidos N-Cíclicos/química , Humanos , Isomerismo , Oxazinas/química , Relação Estrutura-Atividade
19.
J Med Chem ; 45(23): 5090-7, 2002 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-12408720

RESUMO

Tumor suppressor protein, p53, is an intracellular protein that is critical within the biochemical cascade that leads to cell death via apoptosis. Recent studies identified the tetrahydrobenzothiazole analogue, pifithrin-alpha (2), as a p53 inhibitor that was effective in protecting neuronal cells against a variety of lethal insults and reducing the side effects of anticancer drugs. As up-regulation of p53 has been described as a common feature of several neurodegenerative disorders, including Alzheimer's disease, 2 and novel analogues (3-16) were synthesized to (i) assess the value of tetrahydrobenzothiazole analogues as neuroprotective agents and (ii) define the structural requirements for p53 inactivation. Not only did 2 exhibit neuroprotective activity in both tissue culture and in vivo stroke models but also compounds 6, 7, 10, 13, 15, and 16 proved to be highly potent in protecting PC12 cells and compounds 3, 4, and 6 were highly potent in protecting primary hippocampal cells against death induced by the DNA-damaging agent, camptothecin.


Assuntos
Benzoxazóis/síntese química , Fármacos Neuroprotetores/síntese química , Tiazóis/síntese química , Fatores de Transcrição/antagonistas & inibidores , Proteína Supressora de Tumor p53/antagonistas & inibidores , Animais , Apoptose/efeitos dos fármacos , Benzoxazóis/química , Benzoxazóis/farmacologia , Camptotecina/toxicidade , Técnicas de Cultura , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Iminas/síntese química , Iminas/química , Iminas/farmacologia , Ataque Isquêmico Transitório/tratamento farmacológico , Ataque Isquêmico Transitório/prevenção & controle , Fármacos Neuroprotetores/química , Fármacos Neuroprotetores/farmacologia , Células PC12 , Ratos , Ratos Sprague-Dawley , Relação Estrutura-Atividade , Tiazóis/química , Tiazóis/farmacologia
20.
J Med Chem ; 46(24): 5222-9, 2003 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-14613324

RESUMO

Thalidomide is being increasingly used in the clinical management of a wide spectrum of immunologically-mediated and infectious diseases, and cancers. However, the mechanisms underlying its pharmacological action are still under investigation. In this regard, oral thalidomide is clinically valuable in the treatment of erythema nodosum leprosum (ENL) and multiple myeloma and effectively reduces tumor necrosis factor-alpha (TNF-alpha) levels and angiogenesis in vivo. This contrasts with its relatively weak effects on TNF-alpha and angiogenesis in in vitro studies and implies that active metabolites contribute to its in vivo pharmacologic action and that specific analogues would be endowed with potent activity. Our focus in the structural modification of thalidomide is toward the discovery of novel isosteric active analogues. In this regard, a series of thiothalidomides and analogues were synthesized and evaluated for their TNF-alpha inhibitory activity against lipopolysacharide (LPS)-stimulated peripheral blood mononuclear cells (PBMC), This was combined with a PBMC viability assay to differentiate reductions in TNF-alpha secretion from cellular toxicity. Two isosteric analogues of thalidomide, compounds 15 and 16, that mostly reflect the parent compound, together with the simple structure, dithioglutarimide 19, potently inhibited TNF-alpha secretion, compared to thalidomide, 1. The mechanism underpinning this most likely is posttranscriptional, as each of these compounds decreased TNF-alpha mRNA stability via its 3'-UTR. The potency of 19 warrants further study and suggests that replacement of the amide carbonyl with a thiocarbonyl may be beneficial for increased TNF-alpha inhibitory action. In addition, an intact phthalimido moiety appeared to be requisite for TNF-alpha inhibitory activity.


Assuntos
Piperidinas/síntese química , Talidomida/análogos & derivados , Talidomida/síntese química , Tionas/síntese química , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Regiões 3' não Traduzidas , Animais , Linhagem Celular , Genes Reporter , Humanos , Técnicas In Vitro , Lipopolissacarídeos/farmacologia , Luciferases/genética , Luciferases/metabolismo , Camundongos , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Piperidinas/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Relação Estrutura-Atividade , Talidomida/farmacologia , Tionas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA