Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Acta Neuropathol ; 136(4): 621-639, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30046897

RESUMEN

Parkinson's disease (PD), the most common neurodegenerative movement disorder, is characterized by the progressive loss of nigral dopamine neurons. The deposition of fibrillary aggregated α-synuclein in Lewy bodies (LB), that is considered to play a causative role in the disease, constitutes another key neuropathological hallmark of PD. We have recently described that synapsin III (Syn III), a synaptic phosphoprotein that regulates dopamine release in cooperation with α-synuclein, is present in the α-synuclein insoluble fibrils composing the LB of patients affected by PD. Moreover, we observed that silencing of Syn III gene could prevent α-synuclein fibrillary aggregation in vitro. This evidence suggests that Syn III might be crucially involved in α-synuclein pathological deposition. To test this hypothesis, we studied whether mice knock-out (ko) for Syn III might be protected from α-synuclein aggregation and nigrostriatal neuron degeneration resulting from the unilateral injection of adeno-associated viral vectors (AAV)-mediating human wild-type (wt) α-synuclein overexpression (AAV-hαsyn). We found that Syn III ko mice injected with AAV-hαsyn did not develop fibrillary insoluble α-synuclein aggregates, showed reduced amount of α-synuclein oligomers detected by in situ proximity ligation assay (PLA) and lower levels of Ser129-phosphorylated α-synuclein. Moreover, the nigrostriatal neurons of Syn III ko mice were protected from both synaptic damage and degeneration triggered by the AAV-hαsyn injection. Our observations indicate that Syn III constitutes a crucial mediator of α-synuclein aggregation and toxicity and identify Syn III as a novel therapeutic target for PD.


Asunto(s)
Neostriado/patología , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Sustancia Negra/patología , Sinapsis/patología , Sinapsinas/deficiencia , alfa-Sinucleína/metabolismo , Anfetamina/farmacología , Animales , Estimulantes del Sistema Nervioso Central/farmacología , Dependovirus/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Degeneración Nerviosa/genética , Degeneración Nerviosa/patología , Neuronas/patología , Conducta Estereotipada/efectos de los fármacos , Proteína 2 de Membrana Asociada a Vesículas/metabolismo
2.
Handb Exp Pharmacol ; 245: 85-110, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-28965171

RESUMEN

The formation of protein aggregates and inclusions in the brain and spinal cord is a common neuropathological feature of a number of neurodegenerative diseases including Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), and many others. These are commonly referred as neurodegenerative proteinopathies or protein-misfolding diseases. The main characteristic of protein aggregates in these disorders is the fact that they are enriched in amyloid fibrils. Since protein aggregation is considered to play a central role for the onset of neurodegenerative proteinopathies, research is ongoing to develop strategies aimed at preventing or removing protein aggregation in the brain of affected patients. Numerous studies have shown that small molecule-based approaches may be potentially the most promising for halting protein aggregation in neurodegenerative diseases. Indeed, several of these compounds have been found to interact with intrinsically disordered proteins and promote their clearing in experimental models. This notwithstanding, at present small molecule inhibitors still awaits achievements for clinical translation. Hopefully, if we determine whether the formation of insoluble inclusions is effectively neurotoxic and find a valid biomarker to assess their protein aggregation-inhibitory activity in the human central nervous system, the use of small molecule inhibitors will be considered as a cure for neurodegenerative protein-misfolding diseases.


Asunto(s)
Enfermedades Neurodegenerativas/tratamiento farmacológico , Agregación Patológica de Proteínas/tratamiento farmacológico , Péptidos beta-Amiloides/antagonistas & inhibidores , Péptidos beta-Amiloides/química , Humanos , Enfermedades Neurodegenerativas/etiología , Proteínas Priónicas/antagonistas & inhibidores , Proteínas Priónicas/química , Superóxido Dismutasa-1/antagonistas & inhibidores , Superóxido Dismutasa-1/química , alfa-Sinucleína/antagonistas & inhibidores , alfa-Sinucleína/química
3.
Neural Plast ; 2018: 4196961, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29531524

RESUMEN

The antiparkinsonian ropinirole and pramipexole are D3 receptor- (D3R-) preferring dopaminergic (DA) agonists used as adjunctive therapeutics for the treatment resistant depression (TRD). While the exact antidepressant mechanism of action remains uncertain, a role for D3R in the restoration of impaired neuroplasticity occurring in TRD has been proposed. Since D3R agonists are highly expressed on DA neurons in humans, we studied the effect of ropinirole and pramipexole on structural plasticity using a translational model of human-inducible pluripotent stem cells (hiPSCs). Two hiPSC clones from healthy donors were differentiated into midbrain DA neurons. Ropinirole and pramipexole produced dose-dependent increases of dendritic arborization and soma size after 3 days of culture, effects antagonized by the selective D3R antagonists SB277011-A and S33084 and by the mTOR pathway kinase inhibitors LY294002 and rapamycin. All treatments were also effective in attenuating the D3R-dependent increase of p70S6-kinase phosphorylation. Immunoneutralisation of BDNF, inhibition of TrkB receptors, and blockade of MEK-ERK signaling likewise prevented ropinirole-induced structural plasticity, suggesting a critical interaction between BDNF and D3R signaling pathways. The highly similar profiles of data acquired with DA neurons derived from two hiPSC clones underpin their reliability for characterization of pharmacological agents acting via dopaminergic mechanisms.


Asunto(s)
Antiparkinsonianos/administración & dosificación , Benzotiazoles/administración & dosificación , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Neuronas Dopaminérgicas , Indoles/administración & dosificación , Plasticidad Neuronal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Animales , Neuronas Dopaminérgicas/citología , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/fisiología , Ratones , Pramipexol , Transducción de Señal
4.
Int J Mol Sci ; 19(1)2018 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-29316653

RESUMEN

Histone deacetylation, together with altered acetylation of NF-κB/RelA, encompassing the K310 residue acetylation, occur during brain ischemia. By restoring the normal acetylation condition, we previously reported that sub-threshold doses of resveratrol and entinostat (MS-275), respectively, an activator of the AMP-activated kinase (AMPK)-sirtuin 1 pathway and an inhibitor of class I histone deacetylases (HDACs), synergistically elicited neuroprotection in a mouse model of ischemic stroke. To improve the translational power of this approach, we investigated the efficacy of MS-275 replacement with valproate, the antiepileptic drug also reported to be a class I HDAC blocker. In cortical neurons previously exposed to oxygen glucose deprivation (OGD), valproate elicited neuroprotection at 100 nmol/mL concentration when used alone and at 1 nmol/mL concentration when associated with resveratrol (3 nmol/mL). Resveratrol and valproate restored the acetylation of histone H3 (K9/18), and they reduced the RelA(K310) acetylation and the Bim level in neurons exposed to OGD. Chromatin immunoprecipitation analysis showed that the synergistic drug association impaired the RelA binding to the Bim promoter, as well as the promoter-specific H3 (K9/18) acetylation. In mice subjected to 60 min of middle cerebral artery occlusion (MCAO), the association of resveratrol 680 µg/kg and valproate 200 µg/kg significantly reduced the infarct volume as well as the neurological deficits. The present study suggests that valproate and resveratrol may represent a promising ready-to-use strategy to treat post-ischemic brain damage.


Asunto(s)
Inhibidores de Histona Desacetilasas/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Estilbenos/uso terapéutico , Accidente Cerebrovascular/tratamiento farmacológico , Ácido Valproico/uso terapéutico , Acetilación/efectos de los fármacos , Animales , Proteína 11 Similar a Bcl2/genética , Proteína 11 Similar a Bcl2/metabolismo , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Inhibidores de Histona Desacetilasas/farmacología , Histonas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fármacos Neuroprotectores/farmacología , Regiones Promotoras Genéticas , Unión Proteica , Resveratrol , Estilbenos/farmacología , Accidente Cerebrovascular/patología , Factor de Transcripción ReIA/metabolismo , Ácido Valproico/farmacología
5.
J Cell Sci ; 128(13): 2231-43, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25967550

RESUMEN

The main neuropathological features of Parkinson's disease are dopaminergic nigrostriatal neuron degeneration, and intraneuronal and intraneuritic proteinaceous inclusions named Lewy bodies and Lewy neurites, respectively, which mainly contain α-synuclein (α-syn, also known as SNCA). The neuronal phosphoprotein synapsin III (also known as SYN3), is a pivotal regulator of dopamine neuron synaptic function. Here, we show that α-syn interacts with and modulates synapsin III. The absence of α-syn causes a selective increase and redistribution of synapsin III, and changes the organization of synaptic vesicle pools in dopamine neurons. In α-syn-null mice, the alterations of synapsin III induce an increased locomotor response to the stimulation of synapsin-dependent dopamine overflow, despite this, these mice show decreased basal and depolarization-dependent striatal dopamine release. Of note, synapsin III seems to be involved in α-syn aggregation, which also coaxes its increase and redistribution. Furthermore, synapsin III accumulates in the caudate and putamen of individuals with Parkinson's disease. These findings support a reciprocal modulatory interaction of α-syn and synapsin III in the regulation of dopamine neuron synaptic function.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Sinapsis/metabolismo , Sinapsinas/metabolismo , alfa-Sinucleína/metabolismo , Animales , Cocaína/administración & dosificación , Cuerpo Estriado , Dopamina/metabolismo , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Neuronas Dopaminérgicas/ultraestructura , Silenciador del Gen , Humanos , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Biológicos , Actividad Motora , Proteínas Mutantes/metabolismo , Enfermedad de Parkinson , Terminales Presinápticos , Agregado de Proteínas , Unión Proteica , Putamen , Fracciones Subcelulares/metabolismo , Sinapsis/ultraestructura , Vesículas Sinápticas/metabolismo , alfa-Sinucleína/deficiencia
6.
J Pharmacol Exp Ther ; 363(2): 164-175, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28899992

RESUMEN

Loss-of-function mutations in the progranulin (PGRN) gene are a common cause of familial frontotemporal lobar degeneration (FTLD). This age-related neurodegenerative disorder, characterized by brain atrophy in the frontal and temporal lobes and such typical symptoms as cognitive and memory impairment, profound behavioral abnormalities, and personality changes is thought to be related to connectome dysfunctions. Recently, PGRN reduction has been found to induce a behavioral phenotype reminiscent of FTLD symptoms in mice by affecting neuron spine density and morphology, suggesting that the protein can influence neuronal structural plasticity. Here, we evaluated whether a partial haploinsufficiency-like PGRN depletion, achieved by using RNA interference in primary mouse cortical neurons, could modulate GluN2B-containing N-methyl-d-aspartate (NMDA) receptors and tau phosphorylation, which are crucially involved in the regulation of the structural plasticity of these cells. In addition, we studied the effect of PGRN decrease on neuronal cell arborization both in the presence and absence of GluN2B-containing NMDA receptor stimulation. We found that PGRN decline diminished GluN2B-containing NMDA receptor levels and density as well as NMDA-dependent tau phosphorylation. These alterations were accompanied by a marked drop in neuronal arborization that was prevented by an acute GluN2B-containing NMDA receptor stimulation. Our findings support that PGRN decrease, resulting from pathogenic mutations, might compromise the trophism of cortical neurons by affecting GluN2B-contaning NMDA receptors. These mechanisms might be implicated in the pathogenesis of FTLD.


Asunto(s)
Corteza Cerebral/citología , Péptidos y Proteínas de Señalización Intercelular/deficiencia , Plasticidad Neuronal , Neuronas/metabolismo , Interferencia de ARN , Receptores de N-Metil-D-Aspartato/metabolismo , Proteínas tau/metabolismo , Envejecimiento , Animales , Secuencia de Bases , Supervivencia Celular , Regulación de la Expresión Génica , Glicosilación , Granulinas , Hipocampo/citología , Péptidos y Proteínas de Señalización Intercelular/química , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Ratones , Ratones Endogámicos C57BL , Peso Molecular , Neuronas/citología , Fosforilación , Progranulinas
7.
Neural Plast ; 2017: 5012129, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28133550

RESUMEN

Synaptopathies are diseases with synapse defects as shared pathogenic features, encompassing neurodegenerative disorders such as Parkinson's disease (PD). In sporadic PD, the most common age-related neurodegenerative movement disorder, nigrostriatal dopaminergic deficits are responsible for the onset of motor symptoms that have been related to α-synuclein deposition at synaptic sites. Indeed, α-synuclein accumulation can impair synaptic dopamine release and induces the death of nigrostriatal neurons. While in physiological conditions the protein can interact with and modulate synaptic vesicle proteins and membranes, numerous experimental evidences have confirmed that its pathological aggregation can compromise correct neuronal functioning. In addition, recent findings indicate that α-synuclein pathology spreads into the brain and can affect the peripheral autonomic and somatic nervous system. Indeed, monomeric, oligomeric, and fibrillary α-synuclein can move from cell to cell and can trigger the aggregation of the endogenous protein in recipient neurons. This novel "prion-like" behavior could further contribute to synaptic failure in PD and other synucleinopathies. This review describes the major findings supporting the occurrence of α-synuclein pathology propagation in PD and discusses how this phenomenon could induce or contribute to synaptic injury and degeneration.


Asunto(s)
Encéfalo/metabolismo , Encéfalo/patología , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Sinapsis/metabolismo , Sinapsis/patología , alfa-Sinucleína/metabolismo , Animales , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/patología , Humanos , Neuronas/metabolismo , Neuronas/patología , Enfermedad de Parkinson/fisiopatología , Sinapsis/fisiología
8.
Int J Mol Sci ; 18(1)2017 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-28106772

RESUMEN

CSP-1103 (formerly CHF5074) has been shown to reverse memory impairment and reduce amyloid plaque as well as inflammatory microglia activation in preclinical models of Alzheimer's disease. Moreover, it was found to improve cognition and reduce brain inflammation in patients with mild cognitive impairment. Recent evidence suggests that CSP-1103 acts through a single molecular target, the amyloid precursor protein intracellular domain (AICD), a transcriptional regulator implicated in inflammation and apoptosis. We here tested the possible anti-apoptotic and neuroprotective activity of CSP-1103 in a cell-based model of post-ischemic injury, wherein the primary mouse cortical neurons were exposed to oxygen-glucose deprivation (OGD). When added after OGD, CSP-1103 prevented the apoptosis cascade by reducing cytochrome c release and caspase-3 activation and the secondary necrosis. Additionally, CSP-1103 limited earlier activation of p38 and nuclear factor κB (NF-κB) pathways. These results demonstrate that CSP-1103 is neuroprotective in a model of post-ischemic brain injury and provide further mechanistic insights as regards its ability to reduce apoptosis and potential production of pro-inflammatory cytokines. In conclusion, these findings suggest a potential use of CSP-1103 for the treatment of brain ischemia.


Asunto(s)
Apoptosis/efectos de los fármacos , Ciclopropanos/farmacología , Flurbiprofeno/análogos & derivados , Glucosa/deficiencia , Neuronas/patología , Fármacos Neuroprotectores/farmacología , Oxígeno/farmacología , Animales , Caspasa 3/metabolismo , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Corteza Cerebral/patología , Citocromos c/metabolismo , Activación Enzimática/efectos de los fármacos , Flurbiprofeno/farmacología , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Ibuprofeno/farmacología , Ratones Endogámicos C57BL , Necrosis , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Factor de Transcripción ReIA/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
9.
Neuropathol Appl Neurobiol ; 42(1): 77-94, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26613567

RESUMEN

Parkinson's disease (PD) is a common neurodegenerative disorder with prominent loss of nigro-striatal dopaminergic neurons. The resultant dopamine (DA) deficiency underlies the onset of typical motor symptoms (MS). Nonetheless, individuals affected by PD usually show a plethora of nonmotor symptoms (NMS), part of which may precede the onset of motor signs. Besides DA neuron degeneration, a key neuropathological alteration in the PD brain is Lewy pathology. This is characterized by abnormal intraneuronal (Lewy bodies) and intraneuritic (Lewy neurites) deposits of fibrillary aggregates mainly composed of α-synuclein. Lewy pathology has been hypothesized to progress in a stereotypical pattern over the course of PD and α-synuclein mutations and multiplications have been found to cause monogenic forms of the disease, thus raising the question as to whether this protein is pathogenic in this disorder. Findings showing that the majority of α-synuclein aggregates in PD are located at presynapses and this underlies the onset of synaptic and axonal degeneration, coupled to the fact that functional connectivity changes correlate with disease progression, strengthen this idea. Indeed, by altering the proper action of key molecules involved in the control of neurotransmitter release and re-cycling as well as synaptic and structural plasticity, α-synuclein deposition may crucially impair axonal trafficking, resulting in a series of noxious events, whose pressure may inevitably degenerate into neuronal damage and death. Here, we provide a timely overview of the molecular features of synaptic loss in PD and disclose their possible translation into clinical symptoms through functional disconnection.


Asunto(s)
Degeneración Nerviosa/patología , Enfermedad de Parkinson/patología , Sinapsis/patología , Conectoma , Progresión de la Enfermedad , Humanos
10.
Pharmacol Res ; 102: 12-21, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26361727

RESUMEN

The zebrafish (Danio rerio) is a very popular vertebrate model system, especially embryos represent a valuable tool for in vivo pharmacological assays. This is mainly due to the zebrafish advantages when compared to other animal models. Erythropoietin is a glycoprotein hormone that acts principally on erythroid progenitors, stimulating their survival, proliferation and differentiation. Recombinant human erythropoietin (rhEPO) has been widely used in medicine to treat anemia and it is one of the best-selling biotherapeutics worldwide. The recombinant molecule, industrially produced in CHO cells, has the same amino acid sequence of endogenous human erythropoietin, but differs in the glycosylation pattern. This may influence efficacy and safety, particularly immunogenicity, of the final product. We employed the zebrafish embryo as a vertebrate animal model to perform in vivo pharmacological assays. We conducted a functional analysis of rhEPO alpha Eprex(®) and two biosimilars, the erythropoietin alpha Binocrit(®) and zeta Retacrit(®). By in silico analysis and 3D modeling we proved the interaction between recombinant human erythropoietin and zebrafish endogenous erythropoietin receptor. Then we treated zebrafish embryos with the 3 rhEPOs and we investigated their effect on erythrocytes production with different assays. By real time-PCR we observed the relative upregulation of gata1 (2.4 ± 0.3 fold), embryonic α-Hb (1.9 ± 0.2 fold) and ß-Hb (1.6 ± 0.1 fold) transcripts. A significant increase in Stat5 phosphorylation was also assessed in embryos treated with rhEPOs when compared with the negative controls. Live imaging in tg (kdrl:EGFP; gata1:ds-red) embryos, o-dianisidine positive area quantification and cyanomethemoglobin content quantification revealed a 1.8 ± 0.3 fold increase of erythrocytes amount in embryos treated with rhEPOs when compared with the negative controls. Finally, we verified that recombinant human erythropoietins did not cause any inflammatory response in the treated embryos. Our data showed that zebrafish embryo can be a valuable tool to study in vivo effects of complex pharmacological compounds, such as recombinant human glycoproteins, allowing to perform fast and reproducible pharmacological assays with excellent results.


Asunto(s)
Biosimilares Farmacéuticos/farmacología , Eritropoyetina/metabolismo , Pez Cebra/metabolismo , Secuencia de Aminoácidos , Animales , Biología Computacional/métodos , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/metabolismo , Epoetina alfa/farmacología , Factor de Transcripción GATA1/metabolismo , Humanos , Modelos Animales , Datos de Secuencia Molecular , Receptores de Eritropoyetina/metabolismo , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Regulación hacia Arriba/efectos de los fármacos
11.
Exp Cell Res ; 323(1): 100-111, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24589892

RESUMEN

Non-metastatic glycoprotein melanoma protein B (GPNMB), also known as osteoactivin (OA) is expressed in a wide array of tumors and represents an emerging target for drug development. In this study, we investigated the role of GPNMB/OA in the progression of human metastatic DU145 and PC3 prostate cancer cells. GPNMB/OA contribution in PCa malignant phenotype has been analyzed by small interfering RNA-induced GPNMB/OA silencing. We found that following GPNMB/OA silencing the migration capability of both DU145 and PC3 cells, evaluated by using in vitro invasivity assay, as well as the metalloproteinases MMP-2 and MMP-9 activity were equally strongly inhibited. By contrast knocking down GPNMB/OA weakly attenuated cell proliferation rate of DU145, an effect that paralleled with an increase number of apoptotic cells. However, PC3 cell growth seems to be not affected by GPNMB/OA. Together, these data reveal that GPNMB/OA acts as a critical molecular mediator promoting the acquisition of the more aggressive, pro-metastatic phenotype distinctive of human DU145 and PC3 cell lines.


Asunto(s)
Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Glicoproteínas de Membrana/metabolismo , Neoplasias de la Próstata/patología , Apoptosis/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Expresión Génica , Humanos , Masculino , Glicoproteínas de Membrana/genética , Invasividad Neoplásica , Metástasis de la Neoplasia , Fosforilación , Interferencia de ARN , ARN Mensajero/biosíntesis , ARN Interferente Pequeño , Inhibidor Tisular de Metaloproteinasa-1/biosíntesis , Inhibidor Tisular de Metaloproteinasa-2/biosíntesis
12.
Mol Pharmacol ; 83(6): 1176-89, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23543412

RESUMEN

Although long-term exposure to nicotine is highly addictive, one beneficial consequence of chronic tobacco use is a reduced risk for Parkinson's disease. Of interest, these effects both reflect structural and functional plasticity of brain circuits controlling reward and motor behavior and, specifically, recruitment of nicotinic acetylcholine receptors (nAChR) in mesencephalic dopaminergic neurons. Because the underlying cellular mechanisms are poorly understood, we addressed this issue with use of primary cultures of mouse mesencephalic dopaminergic neurons. Exposure to nicotine (1-10 µM) for 72 hours in vitro increased dendritic arborization and soma size in primary cultures. These effects were blocked by mecamylamine and dihydro-ß-erythroidine, but not methyllycaconitine. The involvement of α4ß2 nAChR was supported by the lack of nicotine-induced structural remodeling in neurons from α4 null mutant mice (KO). Challenge with nicotine triggered phosphorylation of the extracellular signal-regulated kinase (ERK) and the thymoma viral proto-oncogene (Akt), followed by activation of the mammalian target of rapamycin complex 1 (mTORC1)-dependent p70 ribosomal S6 protein kinase. Upstream pathway blockade using the phosphatidylinositol 3-kinase inhibitor LY294002 [2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one hydrochloride] resulted in suppression of nicotine-induced phosphorylations and structural plasticity. These effects were dependent on functional DA D3 receptor (D3R), because nicotine was inactive both in cultures from D3R KO mice and after pharmacologic blockade with D3R antagonist trans-N-4-2-(6-cyano-1,2,3, 4-tetrahydroisoquinolin-2-yl)ethylcyclohexyl-4-quinolinecarboxamide (SB-277011-A) (50 nM). Finally, exposure to nicotine in utero (5 mg/kg/day for 5 days) resulted in increased soma area of DAergic neurons of newborn mice, effects not observed in D3 receptor null mutant mice mice. These findings indicate that nicotine-induced structural plasticity at mesencephalic dopaminergic neurons involves α4ß2 nAChRs together with dopamine D3R-mediated recruitment of ERK/Akt-mTORC1 signaling.


Asunto(s)
Neuronas Dopaminérgicas/efectos de los fármacos , Mesencéfalo/efectos de los fármacos , Nicotina/farmacología , Proteínas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Dopamina D3/metabolismo , Animales , Animales Recién Nacidos , Células Cultivadas , Neuronas Dopaminérgicas/citología , Neuronas Dopaminérgicas/metabolismo , Activación Enzimática , Femenino , Sistema de Señalización de MAP Quinasas , Exposición Materna , Intercambio Materno-Fetal , Diana Mecanicista del Complejo 1 de la Rapamicina , Mesencéfalo/citología , Mesencéfalo/metabolismo , Ratones , Ratones Noqueados , Complejos Multiproteicos , Embarazo , Receptores de Dopamina D3/genética , Receptores Nicotínicos/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR
13.
Neurobiol Dis ; 49: 177-89, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22971966

RESUMEN

UNLABELLED: Nuclear factor-kappaB (NF-κB) p50/RelA is a key molecule with a dual effect in the progression of ischemic stroke. In harmful ischemia, but not in preconditioning insult, neurotoxic activation of p50/RelA is characterized by RelA-specific acetylation at Lys310 (K310) and deacetylation at other Lys residues. The derangement of RelA acetylation is associated with activation of Bim promoter. OBJECTIVE: With the aim of producing neuroprotection by correcting altered acetylation of RelA in brain ischemia, we combined the pharmacological inhibition of histone deacetylase (HDAC) 1-3, the enzymes known to reduce global RelA acetylation, and the activation of sirtuin 1, endowed with a specific deacetylase activity on the K310 residue of RelA. To afford this aim, we tested the clinically used HDAC 1-3 inhibitor entinostat (MS-275) and the sirtuin 1 activator resveratrol. METHODS: We used the mouse model of transient middle cerebral artery occlusion (MCAO) and primary cortical neurons exposed to oxygen glucose deprivation (OGD). RESULTS: The combined use of MS-275 and resveratrol, by restoring normal RelA acetylation, elicited a synergistic neuroprotection in neurons exposed to OGD. This effect correlated with MS-275 capability to increase total RelA acetylation and resveratrol capability to reduce RelA K310 acetylation through the activation of an AMP-activated protein kinase-sirtuin 1 pathway. The synergistic treatment reproduced the acetylation state of RelA peculiar of preconditioning ischemia. Neurons exposed to the combined drugs totally recovered the optimal histone H3 acetylation. Neuroprotection was reproduced in mice subjected to MCAO and treated with MS-275 (20µg/kg and 200µg/kg) or resveratrol (6800µg/kg) individually. However, the administration of lowest doses of MS-275 (2µg/kg) and resveratrol (68µg/kg) synergistically reduced infarct volume and neurological deficits. Importantly, the treatment was effective even when administered 7h after the stroke onset. Chromatin immunoprecipitation analysis of cortices harvested from treated mice showed that the RelA binding and histone acetylation increased at the Bcl-xL promoter and decreased at the Bim promoter. CONCLUSION: Our study reveals that epigenetic therapy shaping acetylation of both RelA and histones may be a promising strategy to limit post-ischemic injury with an extended therapeutic window.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Epigénesis Genética/efectos de los fármacos , Histonas/metabolismo , FN-kappa B/metabolismo , Fármacos Neuroprotectores/farmacología , Factor de Transcripción ReIA/metabolismo , Acetilación/efectos de los fármacos , Animales , Benzamidas/farmacología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Hipoxia de la Célula/efectos de los fármacos , Hipoxia de la Célula/fisiología , Modelos Animales de Enfermedad , Epigénesis Genética/fisiología , Glucosa/deficiencia , Inhibidores de Histona Desacetilasas/farmacología , Infarto de la Arteria Cerebral Media , Masculino , Ratones Endogámicos C57BL , Neuronas/metabolismo , Neuronas/patología , Piridinas/farmacología , Resveratrol , Sirtuina 1/metabolismo , Estilbenos/farmacología
14.
Pharmacol Res ; 74: 1-6, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23628881

RESUMEN

While acetylcholine (ACh) and muscarinic receptors in the bladder are mainly known for their role in the regulation of smooth muscle contractility, in other tissues they are involved in tissue remodelling and promote cell growth and proliferation. In the present study we have used primary cultures of human detrusor smooth muscle cells (HDSMCs), in order to investigate the role of muscarinic receptors in HDSMC proliferation. Samples were obtained as discarded tissue from men >65 years undergoing radical cystectomy for bladder cancer and cut in pieces that were either immediately frozen or placed in culture medium for the cell culture establishment. HDSMCs were isolated from samples, propagated and maintained in culture. [(3)H]-QNB radioligand binding on biopsies revealed the presence of muscarinic receptors, with a Kd of 0.10±0.02nM and a Bmax of 72.8±0.1fmol/mg protein. The relative expression of muscarinic receptor subtypes, based on Q-RT-PCR, was similar in biopsies and HDSMC with a rank order of M2≥M3>M1>M4>M5. The cholinergic agonist carbachol (CCh, 1-100µM) concentration-dependently increased [(3)H]-thymidine incorporation (up to 46±4%). This was concentration-dependently inhibited by the general muscarinic receptor antagonist atropine and by subtype-preferring antagonists with an order of potency of darifenacin >4-DAMP>AF-DX 116. The CCh-induced cell proliferation was blocked by selective PI-3 kinase and ERK activation inhibitors, strongly suggesting that these intracellular pathways mediate, at least in part, the muscarinic receptor-mediated cell proliferation. This work shows that M2 and M3 receptors can mediate not only HDSM contraction but also proliferation; they may also contribute bladder remodelling including detrusor hypertrophy.


Asunto(s)
Proliferación Celular , Miocitos del Músculo Liso/metabolismo , Receptores Muscarínicos/fisiología , Vejiga Urinaria/citología , Anciano , Atropina/farmacología , Benzofuranos/farmacología , Carbacol/farmacología , Células Cultivadas , Agonistas Colinérgicos/farmacología , Expresión Génica , Humanos , Masculino , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Antagonistas Muscarínicos/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Piperidinas/farmacología , Pirenzepina/análogos & derivados , Pirenzepina/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirrolidinas/farmacología , ARN Mensajero/metabolismo
15.
Ann Vasc Surg ; 26(3): 387-95, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22285343

RESUMEN

BACKGROUND: To present our experience of extended endovascular management for thromboangiitis obliterans (Buerger's disease) patients with critical limb ischemia (CLI). METHODS: Between January 2005 and July 2010, a consecutive series of 17 Buerger's disease patients with CLI in 20 limbs were admitted and the diagnosis confirmed. The mean age of the patients was 41.5 years (standard error: ±1.7). All patients presented with history of smoking, one patient presented with hypertension, and eight patients presented with dyslipidemia. According to Rutherford classification, all patients were found to be between grades 3 and 5. Ultrasonography first, and angiography examination later, confirmed a severe arterial disease involving almost exclusively below-the-knee and foot arteries in all cases. A new approach for revascularization, defined as extended angioplasty of each tibial and foot artery obstruction, was performed to achieve direct perfusion of at least one foot artery. RESULTS: An extensive endovascular treatment was intended in all patients with success in 19 of 20 limbs, achieving a technical success in 95%. No mortality or complication related to the procedure was observed. During a mean follow-up of 23 months (standard error: ±4.05), amputation-free survival with no need of major amputation in any case and sustained clinical improvement was achieved in 16 of the 19 limbs (84.2%) successfully treated, resulting in a 100% limb salvage rate (19/19). CONCLUSION: In this first experience, in patients with thromboangiitis obliterans, extended endovascular intervention was a feasible and effective revascularization procedure in case of CLI. High technical success, amputation-free survival, and sustained clinical improvement rates were achieved at midterm follow-up was achieved.


Asunto(s)
Angioplastia de Balón , Pie/irrigación sanguínea , Isquemia/terapia , Tromboangitis Obliterante/terapia , Arterias Tibiales/fisiopatología , Adulto , Amputación Quirúrgica , Angioplastia de Balón/efectos adversos , Angioplastia de Balón/instrumentación , Enfermedad Crítica , Estudios de Factibilidad , Femenino , Humanos , Isquemia/diagnóstico , Isquemia/etiología , Isquemia/fisiopatología , Italia , Recuperación del Miembro , Masculino , Flujo Sanguíneo Regional , Estudios Retrospectivos , Stents , Tromboangitis Obliterante/complicaciones , Tromboangitis Obliterante/diagnóstico , Tromboangitis Obliterante/fisiopatología , Terapia Trombolítica , Factores de Tiempo , Resultado del Tratamiento , Grado de Desobstrucción Vascular
16.
Pharmacology ; 89(1-2): 99-102, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22343422

RESUMEN

Personalized medicine is becoming an important tool in oncology, both in preventing disease and in optimizing the treatment of existing cancers. Here we describe the cases of 2 patients with relevant systemic toxicity following 5-fluorouracil (5-FU) therapy and we study the more frequent polymorphisms in the target genes, in particular: (1) the variability in the number of 28-base repetitions present in the 5'-untranslated sequence of the thymidine synthase gene; (2) the presence of single-nucleotide polymorphisms in the methylene tetrahydrofolate reductase gene, and (3) the presence of mRNA splicing in intron 14 of the hepatic enzyme dihydropyrimidine dehydrogenase. The 5-FU gene profile of our patients strongly suggested that the polymorphisms expressed may contribute to the adverse effects seen during the therapy. To what extent these polymorphisms induced adverse effects cannot be established at present; however, our results strengthen the relevance of the 5-FU-related pharmacogenomic profile to predict the response outcome and the chemotherapy toxicity.


Asunto(s)
Dihidrouracilo Deshidrogenasa (NADP)/genética , Fluorouracilo/efectos adversos , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Polimorfismo de Nucleótido Simple , Timidilato Sintasa/genética , Adulto , Femenino , Humanos , Masculino , Empalme del ARN , Secuencias Repetidas Terminales
17.
J Neurochem ; 117(2): 253-63, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21272002

RESUMEN

We report a novel mechanism for dopamine D(1) receptor (D(1) R)-mediated extracellular signal-regulated kinases (Erk) activation in rat striatum. Erk signaling depends on phosphorylation and dephosphorylation events mediated by specific kinases and phosphatases. The tyrosine phosphatase Shp-2, that is required for Erk activation by tyrosine kinase receptors, has been recently shown to regulate signaling downstream of few G protein-coupled receptors. We show that the D(1) R interacts with Shp-2, that D(1) R stimulation results in Shp-2 tyrosine phosphorylation and activation in primary striatal neuronal cultures and that D(1) R/Shp-2 interaction is required for transmitting D(1) R-dependent signaling to Erk1/2 activation. D(1) R-mediated Erk1/2 phosphorylation in cultured striatal neurons is in fact abolished by over-expression of the inactive Shp-2(C/S) mutant and by small interfering RNA-induced Shp-2 silencing. Moreover, by using selective inhibitors we show that both D(1) R-induced Shp-2 activation and Erk1/2 phosphorylation are dependent on the cyclic AMP/protein kinase A pathway and require Src. These results, which were substantiated also in transfected human embryonic kidney 293 cells, provide a novel mechanism by which to converge D(1) R signaling to the Erk pathway and suggest that Shp-2 or the D(1) R/Shp-2 interface could represent a potential drug target for disorders of dopamine transmission involving malfunctioning of D(1) R signaling.


Asunto(s)
Cuerpo Estriado/citología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Neuronas/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Receptores de Dopamina D1/metabolismo , Transducción de Señal/fisiología , Adenilil Ciclasas/metabolismo , Análisis de Varianza , Animales , Anticuerpos/farmacología , Benzazepinas/farmacología , Células Cultivadas , Agonistas de Dopamina/farmacología , Antagonistas de Dopamina/farmacología , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Embrión de Mamíferos , Humanos , Inmunoprecipitación/métodos , Neuronas/efectos de los fármacos , Fosforilación/efectos de los fármacos , Fosforilación/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 11/inmunología , ARN Interferente Pequeño/farmacología , Ratas , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Transfección/métodos , Tirosina/metabolismo
18.
J Neurochem ; 116(4): 588-605, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21166675

RESUMEN

Accumulation of misfolded proteins in the endoplasmic reticulum (ER) is the main event leading to the induction of the ER stress-related unfolded protein response (UPR). Recent postmortem evaluation, showing that the UPR pathway is activated in nigral dopaminergic neurons bearing α-synuclein inclusions in the brain of Parkinson's disease (PD) patients, suggests that the activation of the UPR may be induced by the accumulation of α-synuclein. In this study, we show that the misfolded protein-sensor/UPR activator glucose-regulated protein 78/immunoglobulin heavy chain-binding protein was bound to α-synuclein and was increased in 'in vitro' and 'in vivo' models showing aggregated α-synuclein accumulation. Moreover, α-synuclein accumulation induced the expression of the UPR-related activating transcription factor 4/cAMP-responsive element-2. These findings indicate that activation of the UPR pathway in the PD brain is associated with α-synuclein accumulation occurring in part within the ER.


Asunto(s)
Modelos Animales de Enfermedad , Retículo Endoplásmico/metabolismo , Enfermedad de Parkinson/metabolismo , Respuesta de Proteína Desplegada/fisiología , alfa-Sinucleína/metabolismo , Animales , Línea Celular Tumoral , Retículo Endoplásmico/patología , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Enfermedad de Parkinson/patología , Transducción de Señal/fisiología
19.
J Neurochem ; 116(6): 1148-59, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21210815

RESUMEN

This study was designed to test the hypothesis that improved mitochondrial biogenesis could help reducing ischemic cerebral injury. We found that levels of proliferator-activated receptor γ coactivator 1α and nuclear respiratory factor-1, mitochondrial DNA content and other markers of mitochondrial biogenesis and function were reduced in primary mouse cortical neurons under oxygen-glucose deprivation (OGD). The glycogen synthase kinase-3 (GSK-3) inhibitor SB216763 activated an efficient mitochondrial biogenesis program in control cortical neurons and counteracted the OGD-mediated mitochondrial biogenesis impairment. This was accompanied by the activation of an antioxidant response that reduced mitochondrial reactive oxygen species generation and ischemic neuronal damage. The in vitro effects of SB216763 were mimicked by two other structurally unrelated GSK-3 inhibitors. The protective effects of SB216763 on OGD-mediated neuronal damage were abolished in the presence of diverse mitochondrial inhibitors. Finally, when systemically administered in vivo, SB216763 reduced the infarct size and recovered the loss of mitochondrial DNA in mice subjected to permanent middle cerebral artery occlusion. We conclude that GSK-3 inhibition by SB216763 might pave the way of novel promising therapies aimed at stimulating the renewal of functional mitochondria and reducing reactive oxygen species-mediated damage in ischemic stroke.


Asunto(s)
Glucógeno Sintasa Quinasa 3/metabolismo , Infarto de la Arteria Cerebral Media/enzimología , Mitocondrias/efectos de los fármacos , Neuronas/ultraestructura , Biogénesis de Organelos , Especies Reactivas de Oxígeno/metabolismo , Animales , Células Cultivadas , Corteza Cerebral/citología , ADN Mitocondrial/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Embrión de Mamíferos , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Glucosa/deficiencia , Proteínas del Grupo de Alta Movilidad/genética , Proteínas del Grupo de Alta Movilidad/metabolismo , Hipoxia , Indoles/farmacología , Indoles/uso terapéutico , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/patología , L-Lactato Deshidrogenasa/metabolismo , Maleimidas/farmacología , Maleimidas/uso terapéutico , Ratones , Mitocondrias/metabolismo , Mutación/genética , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Factor Nuclear 1 de Respiración/genética , Factor Nuclear 1 de Respiración/metabolismo , ARN Mensajero/metabolismo , Transfección/métodos
20.
Pharmacol Res ; 64(4): 420-5, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21718784

RESUMEN

The widespread non-neuronal synthesis of acetylcholine (ACh) has changed the paradigm of ACh acting solely as a neurotransmitter. Indeed, the presence of ACh in many types of proliferating cells suggests a role for this neurotransmitter in the control of cell division. The parasympathetic system is a major pathway regulating micturition, but ACh-mediated control plays a more complex role than previously described, acting not only in the detrusor muscle, but also influencing detrusor function through the activity of urothelial muscarinic receptors. Here we investigated the role of muscarinic receptors in mediating cell proliferation in the human UROtsa cell line, which is a widely used experimental model to study urothelium physiology and pathophysiology. Our results demonstrate that UROtsa cells express the machinery for ACh synthesis and that muscarinic receptors, with the rank order of M3>M2>M5>M1=M4, are present and functionally linked to their known second messengers. Indeed, the cholinergic receptor agonist carbachol (CCh) (1-100 µM) concentration-dependently raised IP(3) levels, reaching 66±5% over basal. The forskolin-mediated adenylyl cyclase activation was reduced by CCh exposure (forskolin: 1.4±0.14 pmol/ml; forskolin+100 µM CCh: 0.84±0.12 pmol/ml). CCh (1-100 µM) concentration-dependently increased UROtsa cell proliferation and this effect was inhibited by the non-selective antagonist atropine and the M(3)-selective antagonists darifenacin and J104129. Finally, CCh-induced cell proliferation was blocked by selective PI-3 kinase and ERK activation inhibitors, strongly suggesting that these intracellular pathways mediate, at least in part, the muscarinic receptor-mediated cell proliferation.


Asunto(s)
Proliferación Celular , Receptores Muscarínicos/metabolismo , Urotelio/citología , Acetilcolina/metabolismo , Línea Celular , Humanos , Fosfatidilinositol 3-Quinasas/metabolismo , Sistemas de Mensajero Secundario
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA