Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
CNS Neurol Disord Drug Targets ; 16(7): 820-827, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28176640

RESUMEN

BACKGROUND: Selective butyrylcholinesterase (BuChE)-inhibition, increases acetylcholine (ACh) levels. In rodents, this inhibition is known to boost cognition. Also, this occurs without the typical unwanted adverse effects of acetylcholinesterase-inhibitors or AChE-Is. The novel compound, fluorobenzylcymserine (FBC), is derived from our effort to design a selective BuChE-inhibitor. Also, we wanted to check whether butyrylcholinesterase-inhibitors (BuChE-Is) possessed an edge over AChE-Is in Alzheimer's disease (AD) in terms of efficacy and/or tolerance. METHOD: FBC was synthesized as reported earlier while enzymatic activity of BuChE was calculated by Ellman-technique. Molecular docking was performed using Autodock4.2. We applied classical as well as innovative analyses of enzyme-kinetics for exploring "FBC:human BuChE-interaction". The mode of inhibition and kinetic parameters were also determined. RESULTS: Docking results displayed two strong interacting sites for FBC. One of these binding sites was previously identified as a deep narrow groove having polar aromatic residues while a second site was identified during this study which displayed better interaction and was lined with aliphatic and sulphur containing residues. At low concentrations of BuChE, the IC50 was found to be very low i.e. 4.79 and 6.10 nM for 12 and 36 µg, respectively, whereas it increased exponentially by increasing the units of BuChE. CONCLUSION: These analyses indicate that FBC is an interesting AD drug candidate that could provide a potent and partial mixed type of inhibition of human BuChE.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Butirilcolinesterasa/efectos de los fármacos , Inhibidores de la Colinesterasa/farmacología , Fisostigmina/análogos & derivados , Sitios de Unión , Butirilcolinesterasa/metabolismo , Humanos , Concentración 50 Inhibidora , Cinética , Simulación del Acoplamiento Molecular , Fisostigmina/farmacología
2.
Neurobiol Dis ; 96: 216-226, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27553877

RESUMEN

Traumatic brain injury (TBI) is a major cause of death and disability worldwide. Neuronal apoptosis in the hippocampus has been detected after TBI. The hippocampal dysfunction may result in cognitive deficits in learning, memory, and spatial information processing. Our previous studies demonstrated that a p53 inhibitor, pifithrin-α oxygen analogue (PFT-α (O)), significantly reduced cortical cell death, which is substantial following controlled cortical impact (CCI) TBI, and improved neurological functional outcomes via anti-apoptotic mechanisms. In the present study, we examined the effect of PFT-α (O) on CCI TBI-induced hippocampal cellular pathophysiology in light of this brain region's role in memory. To investigate whether p53-dependent apoptosis plays a role in hippocampal neuronal loss and associated cognitive deficits and to define underlying mechanisms, SD rats were subjected to experimental CCI TBI followed by the administration of PFT-α or PFT-α (O) (2mg/kg, i.v.) or vehicle at 5h after TBI. Magnetic resonance imaging (MRI) scans were acquired at 24h and 7days post-injury to assess evolving structural hippocampal damage. Fluoro-Jade C was used to stain hippocampal sub-regions, including CA1 and dentate gyrus (DG), for cellular degeneration. Neurological functions, including motor and recognition memory, were assessed by behavioral tests at 7days post injury. p53, p53 upregulated modulator of apoptosis (PUMA), 4-hydroxynonenal (4-HNE), cyclooxygenase-IV (COX IV), annexin V and NeuN were visualized by double immunofluorescence staining with cell-specific markers. Levels of mRNA encoding for caspase-3, p53, PUMA, Bcl-2, Bcl-2-associated X protein (BAX) and superoxide dismutase (SOD) were measured by RT-qPCR. Our results showed that post-injury administration of PFT-α and, particularly, PFT-α (O) at 5h dramatically reduced injury volumes in the ipsilateral hippocampus, improved motor outcomes, and ameliorated cognitive deficits at 7days after TBI, as evaluated by novel object recognition and open-field test. PFT-α and especially PFT-α (O) significantly reduced the number of FJC-positive cells in hippocampus CA1 and DG subregions, versus vehicle treatment, and significantly decreased caspase-3 and PUMA mRNA expression. PFT-α (O), but not PFT-α, treatment significantly lowered p53 and elevated SOD2 mRNA expression. Double immunofluorescence staining demonstrated that PFT-α (O) treatment decreased p53, annexin V and 4-HNE positive neurons in the hippocampal CA1 region. Furthermore, PUMA co-localization with the mitochondrial maker COX IV, and the upregulation of PUMA were inhibited by PFT-α (O) after TBI. Our data suggest that PFT-α and especially PFT-α (O) significantly reduce hippocampal neuronal degeneration, and ameliorate neurological and cognitive deficits in vivo via antiapoptotic and antioxidative properties.


Asunto(s)
Benzotiazoles/uso terapéutico , Lesiones Traumáticas del Encéfalo/complicaciones , Trastornos del Conocimiento , Tolueno/análogos & derivados , Proteína p53 Supresora de Tumor/metabolismo , Aldehídos/metabolismo , Animales , Anexina A5/genética , Anexina A5/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Benzotiazoles/química , Benzotiazoles/farmacología , Encéfalo/diagnóstico por imagen , Lesiones Traumáticas del Encéfalo/diagnóstico por imagen , Trastornos del Conocimiento/diagnóstico por imagen , Trastornos del Conocimiento/tratamiento farmacológico , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/patología , Modelos Animales de Enfermedad , Complejo IV de Transporte de Electrones/metabolismo , Conducta Exploratoria/efectos de los fármacos , Fluoresceínas/metabolismo , Imagen por Resonancia Magnética , Masculino , Oxígeno , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Reconocimiento en Psicología/efectos de los fármacos , Factores de Tiempo , Tolueno/química , Tolueno/farmacología , Tolueno/uso terapéutico , Proteína p53 Supresora de Tumor/genética
3.
PLoS One ; 11(6): e0156493, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27254111

RESUMEN

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.


Asunto(s)
Conmoción Encefálica/tratamiento farmacológico , Disfunción Cognitiva/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos , Fisostigmina/análogos & derivados , Animales , Conmoción Encefálica/complicaciones , Conmoción Encefálica/patología , Colinérgicos/administración & dosificación , Inhibidores de la Colinesterasa/administración & dosificación , Disfunción Cognitiva/etiología , Disfunción Cognitiva/patología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Ácido Glutámico/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Humanos , Peroxidación de Lípido/efectos de los fármacos , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Neuronas/efectos de los fármacos , Neuronas/patología , Fisostigmina/administración & dosificación
4.
PLoS One ; 9(6): e99818, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24955574

RESUMEN

Organophosphorus (OP) nerve agents are deadly chemical weapons that pose an alarming threat to military and civilian populations. The irreversible inhibition of the critical cholinergic degradative enzyme acetylcholinesterase (AChE) by OP nerve agents leads to cholinergic crisis. Resulting excessive synaptic acetylcholine levels leads to status epilepticus that, in turn, results in brain damage. Current countermeasures are only modestly effective in protecting against OP-induced brain damage, supporting interest for evaluation of new ones. (-)-Phenserine is a reversible AChE inhibitor possessing neuroprotective and amyloid precursor protein lowering actions that reached Phase III clinical trials for Alzheimer's Disease where it exhibited a wide safety margin. This compound preferentially enters the CNS and has potential to impede soman binding to the active site of AChE to, thereby, serve in a protective capacity. Herein, we demonstrate that (-)-phenserine protects neurons against soman-induced neuronal cell death in rats when administered either as a pretreatment or post-treatment paradigm, improves motoric movement in soman-exposed animals and reduces mortality when given as a pretreatment. Gene expression analysis, undertaken to elucidate mechanism, showed that (-)-phenserine pretreatment increased select neuroprotective genes and reversed a Homer1 expression elevation induced by soman exposure. These studies suggest that (-)-phenserine warrants further evaluation as an OP nerve agent protective strategy.


Asunto(s)
Sustancias para la Guerra Química/toxicidad , Inhibidores de la Colinesterasa/farmacología , Fisostigmina/análogos & derivados , Soman/toxicidad , Estado Epiléptico , Animales , Proteínas Portadoras/biosíntesis , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas de Andamiaje Homer , Masculino , Fisostigmina/farmacología , Ratas , Ratas Sprague-Dawley , Estado Epiléptico/inducido químicamente , Estado Epiléptico/tratamiento farmacológico , Estado Epiléptico/metabolismo
5.
Curr Alzheimer Res ; 11(6): 608-22, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24359497

RESUMEN

Increasing evidence suggests that elevated production and/or reduced clearance of amyloid-ß peptide (Aß) drives the early pathogenesis of Alzheimer's disease (AD). Aß soluble oligomers trigger a neurotoxic cascade that leads to neuronal dysfunction, neurodegeneration and, ultimately, clinical dementia. Inflammation, both within brain and systemically, together with a deficiency in the neurotransmitter acetylcholine (ACh) that underpinned the development of anticholinesterases for AD symptomatic treatment, are invariable hallmarks of the disease. The inter-relation between Aß, inflammation and cholinergic signaling is complex, with each feeding back onto the others to drive disease progression. To elucidate these interactions plasma samples and peripheral blood mononuclear cells (PBMCs) were evaluated from healthy controls (HC) and AD patients. Plasma levels of acetylcholinesterase (AChE), butyrylcholinesterase (BuChE) and Aß were significantly elevated in AD vs. HC subjects, and ACh showed a trend towards reduced levels. Aß challenge of PBMCs induced a greater release of inflammatory cytokines interleukin-1ß (IL-1ß), monocyte chemotactic protein-1 (MCP-1) and tumor necrosis factor-alpha (TNF-α) from AD vs. HC subjects, with IL-10 being similarly affected. THP-1 monocytic cells, a cell culture counterpart of PBMCs and brain microglial cells, responded similarly to Aß as well as to phytohaemagglutinin (PHA) challenge, to allow preliminary analysis of the cellular and molecular pathways underpinning Aß-induced changes in cytokine expression. As amyloid-ß precursor protein expression, and hence Aß, has been reported regulated by particular cytokines and anticholinesterases, the latter were evaluated on Aß- and PHA-induced chemocytokine expression. Co-incubation with selective AChE/BuChE inhibitors, (-)-phenserine (AChE) and (-)-cymserine analogues (BuChE), mitigated the rise in cytokine levels and suggest that augmentation of the cholinergic anti-inflammatory pathway may prove valuable in AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/inmunología , Péptidos beta-Amiloides/metabolismo , Quimiocinas/metabolismo , Inhibidores de la Colinesterasa/farmacología , Citocinas/metabolismo , Acetilcolinesterasa/metabolismo , Anciano , Butirilcolinesterasa/metabolismo , Células Cultivadas , Quimiocina CCL2/metabolismo , Femenino , Humanos , Interleucina-10/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/fisiología , Masculino , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo
6.
PLoS One ; 8(11): e79837, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24312187

RESUMEN

With parallels to concussive mild traumatic brain injury (mTBI) occurring in humans, anesthetized mice subjected to a single 30 g weight drop mTBI event to the right parietal cortex exhibited significant diffuse neuronal degeneration that was accompanied by delayed impairments in recognition and spatial memory. To elucidate the involvement of reversible p53-dependent apoptosis in this neuronal loss and associated cognitive deficits, mice were subjected to experimental mTBI followed by the systemic administration of the tetrahydrobenzothiazole p53 inactivator, PFT-α, or vehicle. Neuronal loss was quantified immunohistochemically at 72 hr. post-injury by the use of fluoro-Jade B and NeuN within the dentate gyrus on both sides of the brain, and recognition and spatial memory were assessed by novel object recognition and Y-maze paradigms at 7 and 30 days post injury. Systemic administration of a single dose of PFT-α 1 hr. post-injury significantly ameliorated both neuronal cell death and cognitive impairments, which were no different from sham control animals. Cellular studies on human SH-SY5Y cells and rat primary neurons challenged with glutamate excitotoxicity and H2O2 induced oxidative stress, confirmed the ability of PFT-α and a close analog to protect against these TBI associated mechanisms mediating neuronal loss. These studies suggest that p53-dependent apoptotic mechanisms underpin the neuronal and cognitive losses accompanying mTBI, and that these are potentially reversible by p53 inactivation.


Asunto(s)
Benzotiazoles/farmacología , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/fisiopatología , Disfunción Cognitiva/etiología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Tolueno/análogos & derivados , Proteína p53 Supresora de Tumor/metabolismo , Animales , Lesiones Encefálicas/complicaciones , Muerte Celular/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Giro Dentado/efectos de los fármacos , Giro Dentado/metabolismo , Giro Dentado/patología , Ácido Glutámico/metabolismo , Ácido Glutámico/toxicidad , Humanos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Neuronas/patología , Estrés Oxidativo , Cultivo Primario de Células , Ratas , Tolueno/farmacología
7.
Biochem Biophys Res Commun ; 433(4): 602-6, 2013 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-23537652

RESUMEN

Nuclear factor E2-related factor 2 (Nrf2)/antioxidant response element (ARE) pathway is an important cellular stress response pathway involved in neuroprotection. We previously screened several natural phytochemicals and identified plumbagin as a novel activator of the Nrf2/ARE pathway that can protect neurons against ischemic injury. Here we extended our studies to natural and synthetic derivatives of plumbagin. We found that 5,8-dimethoxy-1,4-naphthoquinone (naphthazarin) is a potent activator of the Nrf2/ARE pathway, up-regulates the expression of Nrf2-driven genes in primary neuronal and glial cultures, and protects neurons against glutamate-induced excitotoxicity.


Asunto(s)
Muerte Celular , Ácido Glutámico/efectos adversos , Factor 2 Relacionado con NF-E2/metabolismo , Naftoquinonas/farmacología , Neuronas/efectos de los fármacos , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Astrocitos/patología , Proteínas Portadoras/metabolismo , Supervivencia Celular , Relación Dosis-Respuesta a Droga , Genes Reporteros , Células Hep G2 , Humanos , Proteínas de Microfilamentos/metabolismo , Factor 2 Relacionado con NF-E2/genética , Neuronas/metabolismo , Neuronas/patología , Fármacos Neuroprotectores/farmacología , Cultivo Primario de Células , Proteolisis , Ratas , Ratas Sprague-Dawley
8.
PLoS One ; 8(1): e54887, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23382994

RESUMEN

Neuronal dysfunction and demise together with a reduction in neurogenesis are cardinal features of Alzheimer's disease (AD) induced by a combination of oxidative stress, toxic amyloid-ß peptide (Aß) and a loss of trophic factor support. Amelioration of these was assessed with the Aß lowering AD experimental drugs (+)-phenserine and (-)-phenserine in neuronal cultures, and actions in mice were evaluated with (+)-phenserine. Both experimental drugs together with the metabolite N1-norphenserine induced neurotrophic actions in human SH-SY5Y cells that were mediated by the protein kinase C (PKC) and extracellular signal-regulated kinases (ERK) pathways, were evident in cells expressing amyloid precursor protein Swedish mutation (APP(SWE)), and retained in the presence of Aß and oxidative stress challenge. (+)-Phenserine, together with its (-) enantiomer as well as its N1- and N8-norphenserine and N1,N8-bisnorphenserine metabolites, likewise provided neuroprotective activity against oxidative stress and glutamate toxicity via the PKC and ERK pathways. These neurotrophic and neuroprotective actions were evident in primary cultures of subventricular zone (SVZ) neural progenitor cells, whose neurosphere size and survival were augmented by (+)-phenserine. Translation of these effects in vivo was assessed in wild type and AD APPswe transgenic (Tg2576) mice by doublecortin (DCX) immunohistochemical analysis of neurogenesis in the SVZ, which was significantly elevated by 16 day systemic (+)-phenserine treatment, in the presence of a (+)-phenserine-induced elevation in brain- derived neurotrophic factor (BDNF).


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Fármacos Neuroprotectores/química , Fármacos Neuroprotectores/farmacología , Fisostigmina/análogos & derivados , Péptidos beta-Amiloides/farmacología , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Proteína Doblecortina , Descubrimiento de Drogas , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Ratones Transgénicos , Mutación , Células-Madre Neurales/citología , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/metabolismo , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fármacos Neuroprotectores/uso terapéutico , Estrés Oxidativo/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Fisostigmina/química , Fisostigmina/farmacología , Fisostigmina/uso terapéutico , Estereoisomerismo
9.
Artículo en Inglés | MEDLINE | ID: mdl-23360256

RESUMEN

A major pathological hallmark of Alzheimer disease (AD) is the appearance in the brain of senile plaques that are primarily composed of aggregated forms of ß-amyloid peptide (Aß) that derive from amyloid precursor protein (APP). Posiphen (1) tartrate is an experimental AD drug in current clinical trials that reduces Aß levels by lowering the rate of APP synthesis without toxicity. To support the clinical development of Posiphen (1) and elucidate its efficacy, its three major metabolic products, (+)-N1-norPosiphen (15), (+)-N8-norPosiphen (17) and (+)-N1, N8-bisnorPosiphen (11), were required in high chemical and optical purity. The efficient transformation of Posiphen (1) into these metabolic products, 15, 17 and 11, is described. The biological activity of these metabolites together with Posiphen (1) and its enantiomer, the AD drug candidate (-)-phenserine (2), was assessed against APP,α-synuclein and classical cholinergic targets. All the compounds potently inhibited the generation of APP and α-synuclein in neuronal cultures. In contrast, metabolites 11 and 15, and (-)-phenserine (2) but not Posiphen (1) or 17, possessed acetyl cholinesterase inhibitory action and no compounds bound either nicotinic or muscarinic receptors. As Posiphen (1) lowered CSF markers of inflammation in a recent clinical trial, the actions of 1 and 2 on proinflammatory cytokine interleukin (IL)-1ß release human peripheral blood mononuclear cells was evaluated, and found to be potently inhibited by both agents.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/fisiopatología , Fisostigmina/análogos & derivados , Precursor de Proteína beta-Amiloide/biosíntesis , Humanos , Interleucina-1beta/biosíntesis , Fisostigmina/química , Fisostigmina/farmacocinética , Fisostigmina/farmacología , Placa Amiloide/metabolismo , Receptores Muscarínicos/metabolismo , Receptores Nicotínicos/metabolismo , Estereoisomerismo , alfa-Sinucleína/biosíntesis
10.
Biochem J ; 444(2): 269-77, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22390827

RESUMEN

Natural and synthetic carbamates act as pseudo-irreversible inhibitors of AChE (acetylcholinesterase) as well as BChE (butyrylcholinesterase), two enzymes involved in neuronal function as well as in the development and progression of AD (Alzheimer's disease). The AChE mode of action is characterized by a rapid carbamoylation of the active-site Ser(200) with release of a leaving group followed by a slow regeneration of enzyme action due to subsequent decarbamoylation. The experimental AD therapeutic bisnorcymserine, a synthetic carbamate, shows an interesting activity and selectivity for BChE, and its clinical development is currently being pursued. We undertook detailed kinetic studies on the activity of the carbamate bisnorcymserine with Tc (Torpedo californica) AChE and, on the basis of the results, crystallized the complex between TcAChE and bisnorcymserine. The X-ray crystal structure showed only the leaving group, bisnoreseroline, trapped at the bottom of the aromatic enzyme gorge. Specifically, bisnoreseroline interacts in a non-covalent way with Ser(200) and His(440), disrupting the existing interactions within the catalytic triad, and it stacks with Trp(84) at the bottom of the gorge, giving rise to an unprecedented hydrogen-bonding contact. These interactions point to a dominant reversible inhibition mechanism attributable to the leaving group, bisnoreseroline, as revealed by kinetic analysis.


Asunto(s)
Acetilcolinesterasa/metabolismo , Inhibidores de la Colinesterasa/farmacocinética , Cristalografía por Rayos X , Fisostigmina/análogos & derivados , Torpedo , Acetilcolinesterasa/química , Animales , Dominio Catalítico , Inhibidores de la Colinesterasa/química , Cristalografía por Rayos X/métodos , Humanos , Enlace de Hidrógeno , Fisostigmina/química , Fisostigmina/farmacocinética
11.
Behav Brain Res ; 225(1): 222-9, 2011 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-21820013

RESUMEN

The cholinesterase inhibitor, rivastigmine, ameliorates cognitive dysfunction and is approved for the treatment of Alzheimer's disease (AD). Rivastigmine is a dual inhibitor of acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE); however, the impact of BuChE inhibition on cognitive dysfunction remains to be determined. We compared the effects of a selective BuChE inhibitor, N1-phenethyl-norcymserine (PEC), rivastigmine and donepezil (an AChE-selective inhibitor) on cognitive dysfunction induced by amyloid-ß peptide (Aß(1-40)) in mice. Five-week-old imprinting control region (ICR) mice were injected intracerebroventricularly (i.c.v.) with either Aß(1-40) or the control peptide Aß(40-1) on Day 0, and their recognition memory was analyzed by a novel object recognition test. Treatment with donepezil (1.0mg/kg), rivastigmine (0.03, 0.1, 0.3mg/kg) or PEC (1.0, 3.0mg/kg) 20min prior to, or immediately after the acquisition session (Day 4) ameliorated the Aß(1-40) induced memory impairment, indicating a beneficial effect on memory acquisition and consolidation. In contrast, none of the investigated drugs proved effective when administrated before the retention session (Day 5). Repeated daily administration of donepezil, rivastigmine or PEC, on Days 0-3 inclusively, ameliorated the cognitive dysfunction in Aß(1-40) challenged mice. Consistent with the reversal of memory impairments, donepezil, rivastigmine or PEC treatment significantly reduced Aß(1-40) induced tyrosine nitration of hippocampal proteins, a marker of oxidative damage. These results indicate that BuChE inhibition, as well as AChE inhibition, is a viable therapeutic strategy for cognitive dysfunction in AD.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Butirilcolinesterasa/metabolismo , Inhibidores de la Colinesterasa/uso terapéutico , Trastornos del Conocimiento/inducido químicamente , Trastornos del Conocimiento/tratamiento farmacológico , Fragmentos de Péptidos/toxicidad , Fenilcarbamatos/uso terapéutico , Análisis de Varianza , Animales , Modelos Animales de Enfermedad , Donepezilo , Relación Dosis-Respuesta a Droga , Indanos/uso terapéutico , Masculino , Ratones , Ratones Endogámicos ICR , Actividad Motora/efectos de los fármacos , Piperidinas/uso terapéutico , Reconocimiento en Psicología/efectos de los fármacos , Rivastigmina
12.
Bioorg Med Chem ; 19(13): 3965-72, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21658960

RESUMEN

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.


Asunto(s)
Ftalimidas/química , Piperidinas/química , Talidomida/análogos & derivados , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Diseño de Fármacos , Lenalidomida , Ratones , Piperidinas/síntesis química , Piperidinas/toxicidad , Talidomida/síntesis química , Talidomida/química , Talidomida/toxicidad , Factor de Necrosis Tumoral alfa/metabolismo
13.
ACS Med Chem Lett ; 1(3): 105-109, 2010 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-20694189

RESUMEN

As a clinical medication for the treatment of hyperkinetic movement disorders, in conditions such as Huntington's disease, tetrabenazine (TBZ) has been always used in its racemic form. To establish whether or not its beneficial therapeutic actions are enantiospecific, a practical total synthetic route was developed to yield each enantiomeric form to allow their chemical and pharmacological characterization. We briefly summarize the total synthesis of TBZ and report a detailed procedure for resolution of TBZ into its enantiomers, (+)-TBZ and (-)-TBZ. This allowed determination of the optical rotation and absolute configurations of each TBZ enantiomer, based on X-ray crystallographic analysis, together with characterization of their inhibitory action at the vesicular monoamine transporter 2, where (+)-TBZ proved three-fold more active than (-)-TBZ.

14.
Bioorg Med Chem ; 18(13): 4687-93, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-20627738

RESUMEN

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.


Asunto(s)
Acetilcolinesterasa/química , Butirilcolinesterasa/química , Inhibidores de la Colinesterasa/síntesis química , Miastenia Gravis/tratamiento farmacológico , Neostigmina/química , Fenilcarbamatos/química , Fisostigmina/química , Bromuro de Piridostigmina/química , Acetilcolinesterasa/metabolismo , Animales , Butirilcolinesterasa/metabolismo , Inhibidores de la Colinesterasa/química , Inhibidores de la Colinesterasa/uso terapéutico , Humanos , Ratones , Fenilcarbamatos/síntesis química , Fenilcarbamatos/uso terapéutico
15.
J Neurochem ; 115(6): 1337-49, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20456019

RESUMEN

Plumbagin (5-hydroxy-2-methyl-1,4 naphthoquinone) is a naturally occurring low molecular weight lipophilic phytochemical derived from roots of plants of the Plumbago genus. Plumbagin has been reported to have several clinically relevant biological activities in non-neural cells, including anti-atherosclerotic, anticoagulant, anticarcinogenic, antitumor, and bactericidal effects. In a recent screen of a panel of botanical pesticides, we identified plumbagin as having neuroprotective activity. In this study, we determined if plumbagin could modify the developmental fate of rat E14.5 embryonic neural progenitor cells (NPC). Plumbagin exhibited no cytotoxicity when applied to cultured NPC at concentrations below 1 µM. At a concentration of 0.1 µM, plumbagin significantly enhanced the proliferation of NPC as indicated by a 17% increase in the percentage of cells incorporating bromo-deoxyuridine. Plumbagin at a concentration of 0.1 pM (but not 0.1 µM), stimulated the production of astrocytes as indicated by increased GFAP expression. Plumbagin selectively induced the proliferation and differentiation of glial progenitor cells without affecting the proliferation or differentiation of neuron-restricted progenitors. Plumbagin (0.1 pM) rapidly activated the transcription factor signal transducer and activator of transcription 3 (Stat3) in NPC, and a Stat3 inhibitor peptide prevented both plumbagin-induced astrocyte formation and proliferation. These findings demonstrate the ability of a low molecular weight naturally occurring phytochemical to control the fate of glial progenitor cells by a mechanism involving the Stat3 signaling pathway.


Asunto(s)
Astrocitos/metabolismo , Células Madre Embrionarias/metabolismo , Naftoquinonas/farmacología , Neuronas/metabolismo , Fármacos Neuroprotectores/farmacología , Factor de Transcripción STAT3/metabolismo , Médula Espinal/metabolismo , Células Madre/metabolismo , Animales , Supervivencia Celular/fisiología , Femenino , Embarazo , Ratas , Ratas Sprague-Dawley , Médula Espinal/citología
16.
PLoS One ; 5(4): e10044, 2010 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-20386608

RESUMEN

Wound healing is a complex process involving intrinsic dermal and epidermal cells, and infiltrating macrophages and leukocytes. Excessive oxidative stress and associated inflammatory processes can impair wound healing, and antioxidants have been reported to improve wound healing in animal models and human subjects. Uric acid (UA) is an efficient free radical scavenger, but has a very low solubility and poor tissue penetrability. We recently developed novel UA analogs with increased solubility and excellent free radical-scavenging properties and demonstrated their ability to protect neural cells against oxidative damage. Here we show that the uric acid analog (6, 8 dithio-UA, but not equimolar concentrations of UA or 1, 7 dimethyl-UA) modified the behaviors of cultured vascular endothelial cells, keratinocytes and fibroblasts in ways consistent with enhancement of the wound healing functions of all three cell types. We further show that 6, 8 dithio-UA significantly accelerates the wound healing process when applied topically (once daily) to full-thickness wounds in mice. Levels of Cu/Zn superoxide dismutase were increased in wound tissue from mice treated with 6, 8 dithio-UA compared to vehicle-treated mice, suggesting that the UA analog enhances endogenous cellular antioxidant defenses. These results support an adverse role for oxidative stress in wound healing and tissue repair, and provide a rationale for the development of UA analogs in the treatment of wounds and for modulation of angiogenesis in other pathological conditions.


Asunto(s)
Piel/lesiones , Ácido Úrico/análogos & derivados , Cicatrización de Heridas/efectos de los fármacos , Animales , Antioxidantes , Células Cultivadas , Depuradores de Radicales Libres , Ratones , Neovascularización Fisiológica , Estrés Oxidativo , Piel/patología , Solubilidad , Compuestos de Sulfhidrilo , Superóxido Dismutasa/efectos de los fármacos , Ácido Úrico/administración & dosificación , Ácido Úrico/farmacología , Ácido Úrico/uso terapéutico
17.
J Neurochem ; 112(5): 1316-26, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20028456

RESUMEN

Many phytochemicals function as noxious agents that protect plants against insects and other damaging organisms. However, at subtoxic doses, the same phytochemicals may activate adaptive cellular stress response pathways that can protect cells against a variety of adverse conditions. We screened a panel of botanical pesticides using cultured human and rodent neuronal cell models, and identified plumbagin as a novel potent activator of the nuclear factor E2-related factor 2 (Nrf2)/antioxidant response element (ARE) pathway. In vitro, plumbagin increases nuclear localization and transcriptional activity of Nrf2, and induces the expression of the Nrf2/ARE-dependent genes, such as heme oxygenase 1 in human neuroblastoma cells. Plumbagin specifically activates the Nrf2/ARE pathway in primary mixed cultures from ARE-human placental alkaline phosphatase reporter mice. Exposure of neuroblastoma cells and primary cortical neurons to plumbagin provides protection against subsequent oxidative and metabolic insults. The neuroprotective effects of plumbagin are abolished by RNA interference-mediated knockdown of Nrf2 expression. In vivo, administration of plumbagin significantly reduces the amount of brain damage and ameliorates-associated neurological deficits in a mouse model of focal ischemic stroke. Our findings establish precedence for the identification and characterization of neuroprotective phytochemicals based upon their ability to activate adaptive cellular stress response pathways.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Hipoxia/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Naftoquinonas/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Células Cultivadas , Corteza Cerebral/citología , Infarto Cerebral/etiología , Infarto Cerebral/prevención & control , Modelos Animales de Enfermedad , Embrión de Mamíferos , Glucosa/deficiencia , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Factor 2 Relacionado con NF-E2/genética , Naftoquinonas/metabolismo , Naftoquinonas/farmacología , Neuroblastoma , Examen Neurológico , Neuronas , Fármacos Neuroprotectores/metabolismo , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo , Transfección/métodos
18.
J Neurosci Methods ; 183(2): 182-7, 2009 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-19583982

RESUMEN

Neuroinflammation is a common facet of both acute and chronic neurodegenerative conditions, exemplified by stroke and by Alzheimer's and Parkinson's disease, and the presence of elevated levels of the proinflammatory cytokine, tumor necrosis factor-alpha (TNF-alpha), has been documented in each. Although initial TNF-alpha generation is associated with a protective compensatory response, its unregulated chronic elevation is generally detrimental and can drive the disease process. In such circumstances, therapeutic strategies that can both gain access to the brain and target the production of TNF-alpha are predicted to be of clinical benefit. An in vitro mouse macrophage-like cellular screen, utilizing RAW 264.7 cells, was hence developed to identify novel TNF-alpha lowering agents incorporating lipophilic physicochemical characteristics predicted to allow penetration of the blood-brain barrier. Cultured RAW 264.7 cells exposed to lipopolysaccharide (LPS) induced a rapid, marked and concentration-dependent cellular release of TNF-alpha into the cell culture media, which was readily detected by enzyme linked immunosorbent assay (ELISA). The effects of four characterized thalidomide-based TNF-alpha lowering agents were assessed alongside 10 novel uncharacterized compounds synthesized on the same backbone. One of these new analogs possessed activity of sufficient magnitude to warrant further investigation. Activity determined in the cellular model translated to an in vivo rodent model of acute LPS-induced TNF-alpha elevation. The utility of the TNF-alpha cellular assay lies in its simplicity and robust nature, providing a tool for initial pharmacological screening to allow for the rapid identification novel TNF-alpha lowering agents.


Asunto(s)
Inflamación/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Línea Celular Transformada , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Inmunosupresores/química , Inmunosupresores/farmacología , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratas , Ratas Endogámicas F344 , Estadísticas no Paramétricas , Talidomida/análogos & derivados , Talidomida/química , Talidomida/farmacología , Factores de Tiempo
19.
Neurobiol Aging ; 30(7): 1061-8, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18068871

RESUMEN

Presenilin-1 (PS1) mutations cause many cases of early-onset inherited Alzheimer's disease, in part, by increasing the production of neurotoxic forms of amyloid beta-peptide (Abeta). However, Abeta-independent effects of mutant PS1 on neuronal Ca(2+) homeostasis and sensitivity to excitatory neurotransmitters have been reported. Here we show that cholinergic modulation of hippocampal synaptic plasticity is impaired in PS1 mutant knockin (PS1KI) mice. Whereas activation of muscarinic receptors enhances LTP at CA1 synapses of normal mice, it impairs LTP in PS1KI mice. Similarly, mutant PS1 impairs the ability of the cholinesterase inhibitor phenserine to enhance LTP. The NMDA current is decreased in CA1 neurons of PS1KI mice and is restored by intracellular Ca(2+)chelation. Similar alterations in acetylcholine and NMDA receptor-mediated components of synaptic plasticity are evident in 3xTgAD mice with PS1, amyloid precursor protein and tau mutations, suggesting that the adverse effects of mutant PS1 on synaptic plasticity can occur in the absence or presence of amyloid and tau pathologies.


Asunto(s)
Acetilcolina/metabolismo , Enfermedad de Alzheimer/metabolismo , Hipocampo/metabolismo , Plasticidad Neuronal/genética , Presenilina-1/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/metabolismo , Animales , Núcleo Basal de Meynert/metabolismo , Núcleo Basal de Meynert/fisiopatología , Fibras Colinérgicas/metabolismo , Fibras Colinérgicas/patología , Inhibidores de la Colinesterasa/farmacología , Técnicas de Sustitución del Gen , Potenciación a Largo Plazo/genética , Masculino , Trastornos de la Memoria/genética , Trastornos de la Memoria/metabolismo , Trastornos de la Memoria/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación/genética , Vías Nerviosas/metabolismo , Vías Nerviosas/patología , Técnicas de Cultivo de Órganos , Receptores Muscarínicos/efectos de los fármacos , Receptores Muscarínicos/metabolismo , Transmisión Sináptica/genética , Proteínas tau/metabolismo
20.
J Chromatogr A ; 1216(13): 2730-8, 2009 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-18950780

RESUMEN

Focus of this work was the development and characterization of a new immobilized enzyme reactor (IMER) containing human recombinant butyrylcholinesterase (rBChE) for the on-line kinetic characterization of specific, pseudo-irreversible and brain-targeted BChE inhibitors as potential drug candidates for Alzheimer's disease (AD). Specifically, a rBChE-IMER containing 0.99 U of covalently bound target enzyme was purposely developed and inserted into a HPLC system connected to a UV-vis detector. Selected reversible cholinesterase inhibitors, (-)-phenserine and (-)-cymserine analogues, were then kinetically characterized by rBChE-IMER, and by classical in solution assays and their carbamoylation and decarbamoylation constants were determined. The results support the elucidation of the potency, inhibition duration, mode of action and specific structure/activity relations of these agents and allow cross-validation of the two assay techniques.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Butirilcolinesterasa/química , Inhibidores de la Colinesterasa/aislamiento & purificación , Enzimas Inmovilizadas/química , Butirilcolinesterasa/efectos de los fármacos , Inhibidores de la Colinesterasa/farmacología , Cromatografía Líquida de Alta Presión , Evaluación Preclínica de Medicamentos , Enzimas Inmovilizadas/efectos de los fármacos , Humanos , Proteínas Recombinantes/química , Proteínas Recombinantes/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...