Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros

Métodos Terapéuticos y Terapias MTCI
Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Pharmacol Res ; 152: 104597, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31838078

RESUMEN

Neuroinflammation is increasingly associated to the onset and progression of neurodegenerative diseases. Furthermore, several lines of evidence have demonstrated the capacity of aberrant protein aggregates to activate the immune response, accelerating the advance of the disease. Compound ITH12674 is a melatonin-sulforaphane hybrid designed to exert a dual drug-prodrug mechanism of action that combines potent NRF2 induction and free radical scavenger activity. ITH12674 also showed neuroprotective properties in oxidative stress related models, that were dependant on its NRF2 inducing properties. Given the high impact of neuroinflammation in the pathogenesis of neurodegeneration, we foresaw to study the anti-inflammatory properties of ITH12674. ITH12674 reduced inflammatory markers in glial cell cultures and hippocampal tissue after LPS administration. The anti-inflammatory effect was related to inhibition of TLR4 receptors due to a direct interaction with the TLR4/MD2 complex at the hydrophobic cavity of MD2. ITH12674 is endowed with anti-inflammatory properties, that are complementary to the NRF2 inducing activity and neuroprotective properties. Thus, ITH12674 could be of potential interest for the treatment of diseases with chronic neuroinflammation.


Asunto(s)
Antiinflamatorios/farmacología , Isotiocianatos/farmacología , Antígeno 96 de los Linfocitos/metabolismo , Melatonina/análogos & derivados , Neuroglía/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Receptor Toll-Like 4/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Células Cultivadas , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Interleucina-1beta/genética , Lipopolisacáridos/farmacología , Locomoción/efectos de los fármacos , Masculino , Melatonina/farmacología , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Moleculares , Factor 2 Relacionado con NF-E2/genética , Neuroglía/metabolismo , Ratas Sprague-Dawley , Interacción Social/efectos de los fármacos , Factor de Necrosis Tumoral alfa/genética
2.
J Pineal Res ; 67(1): e12578, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30943316

RESUMEN

Alterations in autophagy are increasingly being recognized in the pathogenesis of proteinopathies like Alzheimer's disease (AD). This study was conducted to evaluate whether melatonin treatment could provide beneficial effects in an Alzheimer model related to tauopathy by improving the autophagic flux and, thereby, prevent cognitive decline. The injection of AAV-hTauP301L viral vectors and treatment/injection with okadaic acid were used to achieve mouse and human ex vivo, and in vivo tau-related models. Melatonin (10 µmol/L) impeded oxidative stress, tau hyperphosphorylation, and cell death by restoring autophagy flux in the ex vivo models. In the in vivo studies, intracerebroventricular injection of AAV-hTauP301L increased oxidative stress, neuroinflammation, and tau hyperphosphorylation in the hippocampus 7 days after the injection, without inducing cognitive impairment; however, when animals were maintained for 28 days, cognitive decline was apparent. Interestingly, late melatonin treatment (10 mg/kg), starting once the alterations mentioned above were established (from day 7 to day 28), reduced oxidative stress, neuroinflammation, tau hyperphosphorylation, and caspase-3 activation; these observations correlated with restoration of the autophagy flux and memory improvement. This study highlights the importance of autophagic dysregulation in tauopathy and how administration of pharmacological doses of melatonin, once tauopathy is initiated, can restore the autophagy flux, reduce proteinopathy, and prevent cognitive decline. We therefore propose exogenous melatonin supplementation or the development of melatonin derivatives to improve autophagy flux for the treatment of proteinopathies like AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Muerte Celular Autofágica/efectos de los fármacos , Disfunción Cognitiva/tratamiento farmacológico , Melatonina/farmacología , Estrés Oxidativo/efectos de los fármacos , Enfermedad de Alzheimer/inducido químicamente , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Animales , Disfunción Cognitiva/inducido químicamente , Disfunción Cognitiva/genética , Disfunción Cognitiva/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratas , Ratas Sprague-Dawley
3.
ACS Chem Neurosci ; 8(1): 67-81, 2017 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-27731633

RESUMEN

During brain ischemia, oxygen and glucose deprivation induces calcium overload, extensive oxidative stress, neuroinflammation, and, finally, massive neuronal loss. In the search of a neuroprotective compound to mitigate this neuronal loss, we have designed and synthesized a new multitarget hybrid (ITH14001) directed at the reduction of calcium overload by acting on two regulators of calcium homeostasis; the mitochondrial Na+/Ca2+ exchanger (mNCX) and L-type voltage dependent calcium channels (VDCCs). This compound is a hybrid of CGP37157 (mNCX inhibitor) and nimodipine (L-type VDCCs blocker), and its pharmacological evaluation revealed a moderate ability to selectively inhibit both targets. These activities conferred concentration-dependent neuroprotection in two models of Ca2+ overload, such as toxicity induced by high K+ in the SH-SY5Y cell line (60% protection at 30 µM) and veratridine in hippocampal slices (26% protection at 10 µM). It also showed neuroprotective effect against oxidative stress, an activity related to its nitrogen radical scavenger effect and moderate induction of the Nrf2-ARE pathway. Its Nrf2 induction capability was confirmed by the increase of the expression of the antioxidant and anti-inflammatory enzyme heme-oxygenase I (3-fold increase). In addition, the multitarget profile of ITH14001 led to anti-inflammatory properties, shown by the reduction of nitrites production induced by lipopolysaccharide in glial cultures. Finally, it showed protective effect in two acute models of cerebral ischemia in hippocampal slices, excitotoxicity induced by glutamate (31% protection at 10 µM) and oxygen and glucose deprivation (76% protection at 10 µM), reducing oxidative stress and iNOS deleterious induction. In conclusion, our hybrid derivative showed improved neuroprotective properties when compared to its parent compounds CGP37157 and nimodipine.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Calcio/metabolismo , Nimodipina/farmacología , Nimodipina/uso terapéutico , Estrés Oxidativo/efectos de los fármacos , Tiazepinas/uso terapéutico , Animales , Animales Recién Nacidos , Benzodiazepinonas/química , Benzodiazepinonas/farmacología , Benzodiazepinonas/uso terapéutico , Isquemia Encefálica/patología , Bovinos , Hipoxia de la Célula/efectos de los fármacos , Línea Celular Tumoral , Células Cultivadas , Células Cromafines , Modelos Animales de Enfermedad , Embrión de Mamíferos , Hipocampo/efectos de los fármacos , Hipocampo/patología , Masculino , Neuroblastoma/patología , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Nimodipina/análogos & derivados , Nimodipina/química , Ratas , Ratas Sprague-Dawley , Tiazepinas/química , Tiazepinas/farmacología
4.
Future Med Chem ; 7(15): 1961-9, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26496465

RESUMEN

BACKGROUND: Neurodegenerative diseases share many pathological pathways, such as abnormal protein aggregation, mitochondrial dysfunction, extensive oxidative stress and neuroinflammation. Cells have an intrinsic mechanism of protection, the Nrf2 transcriptional factor, known as the master regulator of redox homeostasis. RESULTS: Based on the common features of these diseases we have designed a multi-target hybrid structure derived from melatonin and ethyl cinnamate. The obtained derivatives were Nrf2 inducers and potent-free radical scavengers. These new compounds showed a very interesting neuroprotective profile in several in vitro models of oxidative stress, Alzheimer's disease and brain ischemia. CONCLUSION: We have designed a new hybrid structure with complementary activities. We have identified compound 5h as an interesting Nrf2 inducer, very potent antioxidant and neuroprotectant.


Asunto(s)
Antioxidantes/farmacología , Cinamatos/farmacología , Melatonina/farmacología , Factor 2 Relacionado con NF-E2/metabolismo , Enfermedades Neurodegenerativas/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Animales , Antioxidantes/síntesis química , Antioxidantes/química , Cinamatos/química , Humanos , Melatonina/química , Estructura Molecular , Fármacos Neuroprotectores/síntesis química , Fármacos Neuroprotectores/química , Estrés Oxidativo/efectos de los fármacos , Ratas , Especies Reactivas de Oxígeno/metabolismo , Relación Estructura-Actividad
5.
Neuropharmacology ; 99: 187-95, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26188145

RESUMEN

Stopping the ischemic cascade by targeting its components is a potential strategy for acute ischemic stroke treatment. During ischemia and especially over reperfusion, oxidative stress plays a major role in causing neuronal cell death. Melatonin has been previously reported to provide neuroprotective effects in in vivo models of stroke by a mechanism that implicates melatonin receptors. In this context, this study was planned to test the potential neuroprotective effects of the novel melatonin MT1/MT2 receptor agonist, Neu-P11, against brain ischemia in in vitro and in vivo models, and to elucidate its underlying mechanism of action. Neu-P11 proved to be a good antioxidant, to protect against glutamate-induced excitotoxicity and oxygen and glucose deprivation in hippocampal slices, and to reduce infarct volume in an in vivo stroke model. Regarding its mechanism of action, the protective effect of Neu-P11 was reverted by luzindole (melatonin receptor antagonist), AG490 (JAK2 inhibitor), LY294002 (PI3/AKT inhibitor) and PD98059 (MEK/ERK1/2 inhibitor). In conclusion, Neu-P11 affords neuroprotection against brain ischemia in in vitro and in vivo models by activating a pro-survival signaling pathway that involves melatonin receptors, JAK/STAT, PI3K/Akt and MEK/ERK1/2.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Indoles/farmacología , Fármacos Neuroprotectores/farmacología , Piranos/farmacología , Animales , Antioxidantes/farmacología , Encéfalo/efectos de los fármacos , Encéfalo/patología , Encéfalo/fisiopatología , Isquemia Encefálica/patología , Isquemia Encefálica/fisiopatología , Hipoxia de la Célula/efectos de los fármacos , Hipoxia de la Célula/fisiología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Glucosa/deficiencia , Humanos , Masculino , Melatonina/análogos & derivados , Ratones Endogámicos C57BL , Distribución Aleatoria , Ratas Sprague-Dawley , Receptores de Melatonina/agonistas , Receptores de Melatonina/antagonistas & inhibidores , Receptores de Melatonina/metabolismo , Técnicas de Cultivo de Tejidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA