Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
J Pharmacol Exp Ther ; 378(2): 184-196, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34011530

RESUMEN

Ferroptosis is a form of oxidative cell death that is increasingly recognized as a key mechanism not only in neurodegeneration but also in regulated cell death, causing disease in other tissues. In neurons, major hallmarks of ferroptosis involve the accumulation of lipid reactive oxygen species (ROS) and impairment of mitochondrial morphology and function. Compounds that interfere with ferroptosis could provide novel treatment options for neurodegenerative disorders and other diseases involving ferroptosis. In the present study, we developed new compounds by refining structural elements of the BH3 interacting-domain death agonist inhibitor BI-6c9, which was previously demonstrated to block ferroptosis signaling at the level of mitochondria. Here, we inserted an antioxidative diphenylamine (DPA) structure to the BI-6c9 structure. These DPA compounds were then tested in models of erastin, and Ras-selective lethal small molecule 3 induced ferroptosis in neuronal HT22 cells. The DPA compounds showed an increased protective potency against ferroptotic cell death compared with the scaffold molecule BI-6c9. Moreover, hallmarks of ferroptosis such as lipid, cytosolic, and mitochondrial ROS formation were abrogated in a concentration- and time-dependent manner. Additionally, mitochondrial parameters such as mitochondrial morphology, mitochondrial membrane potential, and mitochondrial respiration were preserved by the DPA compounds, supporting the conclusion that lipid ROS toxicity and mitochondrial impairment are closely related in ferroptosis. Our findings confirm that the DPA compounds are very effective agents in preventing ferroptotic cell death by blocking ROS production and, in particular, via mitochondrial protection. SIGNIFICANCE STATEMENT: Preventing neuronal cells from different forms of oxidative cell death was previously described as a promising strategy for treatment against several neurodegenerative diseases. This study reports novel compounds based on a diphenylamine structure that strongly protects neuronal HT22 cells from ferroptotic cell death upon erastin and Ras-selective lethal small molecule 3 induction by preventing the development of different reactive oxygen species and by protecting mitochondria from ferroptotic impairments.


Asunto(s)
Ferroptosis , Muerte Celular , Difenilamina , Mitocondrias
2.
Nervenarzt ; 88(8): 888-894, 2017 Aug.
Artículo en Alemán | MEDLINE | ID: mdl-28497256

RESUMEN

BACKGROUND: This overview focuses on the aspects of the pharmacotherapy of Parkinson's disease, which is one of the most common disorders of the nervous system. This article presents the complexity of the pharmacotherapy of geriatric patients with neurological diseases. OBJECTIVES: Information about the potential risk factors and aspects of drug safety in the pharmacotherapy of Parkinson's disease. MATERIALS AND METHODS: Selective literature search using PubMed and the scientific-clinical experience of the authors. RESULTS: Patients with Parkinson's disease are usually geriatric patients with concomitant diseases. As a result they are often treated with comedication which leads to a complex medication regime with more than five drugs. Such polypharmacy increases the risk of adverse drug events due to the rising number of possible interactions and contraindications. To control this risk and maintain a safe therapy, certain measures should be considered. This implies additional need for educational work in order to create awareness regarding potential adverse drug events. In certain cases of diagnosed comorbidities or relevant drug prescriptions in the medication regime, follow-up examinations should be conducted. CONCLUSION: Specific parameters of Parkinson's disease, the health-related quality of life of affected patients and the quality of pharmacotherapeutic drug safety can be improved by targeted monitoring of the medication regime. As a result, the overall drug safety can be increased.


Asunto(s)
Antiparkinsonianos/efectos adversos , Antiparkinsonianos/uso terapéutico , Enfermedad de Parkinson/tratamiento farmacológico , Anciano , Biomarcadores Farmacológicos , Comorbilidad , Interacciones Farmacológicas , Adhesión a Directriz , Humanos , Cumplimiento de la Medicación , Errores de Medicación , Enfermedad de Parkinson/diagnóstico , Factores de Riesgo
3.
Apoptosis ; 19(11): 1545-58, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25146045

RESUMEN

It is well-established that activation of proteases, such as caspases, calpains and cathepsins are essential components in signaling pathways of programmed cell death (PCD). Although these proteases have also been linked to mechanisms of neuronal cell death, they are dispensable in paradigms of intrinsic death pathways, e.g. induced by oxidative stress. However, emerging evidence implicated a particular role for serine proteases in mechanisms of PCD in neurons. Here, we investigated the role of trypsin-like serine proteases in a model of glutamate toxicity in HT-22 cells. In these cells glutamate induces oxytosis, a form of caspase-independent cell death that involves activation of the pro-apoptotic protein BH3 interacting-domain death agonist (Bid), leading to mitochondrial demise and ensuing cell death. In this model system, the trypsin-like serine protease inhibitor Nα-tosyl-l-lysine chloromethyl ketone hydrochloride (TLCK) inhibited mitochondrial damage and cell death. Mitochondrial morphology alterations, the impairment of the mitochondrial membrane potential and ATP depletion were prevented and, moreover, lipid peroxidation induced by glutamate was completely abolished. Strikingly, truncated Bid-induced cell death was not affected by TLCK, suggesting a detrimental activity of serine proteases upstream of Bid activation and mitochondrial demise. In summary, this study demonstrates the protective effect of serine protease inhibition by TLCK against oxytosis-induced mitochondrial damage and cell death. These findings indicate that TLCK-sensitive serine proteases play a crucial role in cell death mechanisms upstream of mitochondrial demise and thus, may serve as therapeutic targets in diseases, where oxidative stress and intrinsic pathways of PCD mediate neuronal cell death.


Asunto(s)
Mitocondrias/efectos de los fármacos , Neuronas/efectos de los fármacos , Inhibidores de Serina Proteinasa/farmacología , Clorometilcetona Tosilisina/farmacología , Animales , Apoptosis/efectos de los fármacos , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Ácido Glutámico/farmacología , Peroxidación de Lípido/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Neuronas/metabolismo , Estrés Oxidativo , Transducción de Señal , Clorometilcetona de Tosilfenilalanila/farmacología
4.
Anaesthesiologie ; 73(2): 85-92, 2024 02.
Artículo en Alemán | MEDLINE | ID: mdl-38289347

RESUMEN

BACKGROUND: The administration of intravenous fluids includes various indications, e.g., fluid replacement, nutritional therapy or as a solvent for drugs and is a common routine in the intensive care unit (ICU); however, overuse of intravenous fluids can lead to fluid overload, which can be associated with a poorer outcome in critically ill patients. OBJECTIVE: The aim of this survey was to find out the current status of the use and management of intravenous fluids as well as the interprofessional cooperation involving clinical pharmacists on German ICUs. METHODS: An online survey with 33 questions was developed. The answers of 62 participants from the Scientific Working Group on Intensive Care Medicine of the German Society for Anesthesiology and Intensive Care Medicine were evaluated. RESULTS: Fluid overload occurs "frequently" in 62.9% (39/62) and "very frequently" in 9.7% (6/62) of the ICUs of respondents. An established standard for an infusion management system is unknown to 71.0% (44/62) of participants and 45.2% of the respondents stated that they did not have a patient data management system. In addition, the participants indicated how they define fluid overload. This was defined by the presence of edema by 50.9% (28/55) and by positive fluid balance by 30.9% (17/55). According to the participants septic patients (38/60; 63.3%) and cardiological/cardiac surgical patients (26/60; 43.3%) are most susceptible to the occurrence of fluid overload. Interprofessional collaboration among intensive care physicians, critical care nurses, and clinical pharmacists to optimize fluid therapy was described as "relevant" by 38.7% (24/62) and "very relevant" by 45.2% (28/62). Participants with clinical pharmacists on the wards (24/62; 38.7%) answered this question more often as "very relevant" with 62.5% (15/24). CONCLUSION: Fluid overload is a frequent and relevant problem in German intensive care units. Yet there are few established standards in this area. There is also a lack of validated diagnostic parameters and a clear definition of fluid overload. These are required to ensure appropriate and effective treatment that is tailored to the patient and adapted to the respective situation. Intravenous fluids should be considered as drugs that may exert side effects or can be overdosed with severe adverse consequences for the patients. One approach to optimize fluid therapy could be achieved by a fluid stewardship corresponding to comparable established procedures of the antibiotic stewardship. In particular, fluid stewardship will contribute to drug safety of intravenous fluids profiting from joined expertise in a setting of interprofessional collaboration. An important principle of fluid stewardship is to consider intravenous fluids in the same way as medication in terms of their importance. Furthermore, more in-depth studies are needed to investigate the effects of interprofessional fluid stewardship in a prospective and controlled manner.


Asunto(s)
Médicos , Desequilibrio Hidroelectrolítico , Humanos , Estudios Prospectivos , Unidades de Cuidados Intensivos , Cuidados Críticos/métodos , Fluidoterapia/efectos adversos , Desequilibrio Hidroelectrolítico/etiología
5.
Apoptosis ; 17(10): 1027-38, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22865232

RESUMEN

Previous studies established a major role for apoptosis inducing factor (AIF) in neuronal cell death after acute brain injury. For example, AIF translocation from mitochondria to the nucleus determined delayed neuronal death, whereas reduced AIF expression provided neuroprotective effects in models of cerebral ischemia or brain trauma. The question remains, however, why reduced AIF levels are sufficient to mediate neuroprotection, since only very little AIF translocation to the nucleus is required for induction of cell death. Thus, the present study addresses the question, whether AIF gene silencing affects intrinsic death pathways upstream of nuclear translocation at the level of the mitochondria. Using MTT assays and real-time cell impedance measurements we confirmed the protective effect of AIF siRNA against glutamate toxicity in immortalized mouse hippocampal HT-22 neurons. Further, AIF siRNA prevented glutamate-induced mitochondrial fragmentation and loss of mitochondrial membrane potential. The protection of mitochondrial integrity was associated with preserved ATP levels, attenuated increases in lipid peroxidation and reduced complex I expression levels. Notably, low concentrations of the complex I inhibitor rotenone (20 nM), provided similar protective effects against glutamate toxicity at the mitochondrial level. These results expose a preconditioning effect as a mechanism for neuroprotection mediated by AIF depletion. In particular, they point out an association between mitochondrial complex I and AIF, which regulate each other's stability in mitochondria. Overall, these findings postulate that AIF depletion mediates a preconditioning effect protecting neuronal cells from subsequent glutamate toxicity through reduced levels of complex I protein.


Asunto(s)
Factor Inductor de la Apoptosis/genética , Mitocondrias/metabolismo , Neuronas/efectos de los fármacos , Neuronas/fisiología , Animales , Apoptosis/efectos de los fármacos , Factor Inductor de la Apoptosis/fisiología , Muerte Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Células Cultivadas , Regulación hacia Abajo , Complejo I de Transporte de Electrón/genética , Complejo I de Transporte de Electrón/metabolismo , Silenciador del Gen , Ácido Glutámico/toxicidad , Hipocampo , Ratones , Mitocondrias/efectos de los fármacos , ARN Interferente Pequeño/farmacología , Rotenona/farmacología
6.
Cell Death Differ ; 15(10): 1553-63, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18535584

RESUMEN

Mitochondrial dysfunction and release of pro-apoptotic factors such as cytochrome c or apoptosis-inducing factor (AIF) from mitochondria are key features of neuronal cell death. The precise mechanisms of how these proteins are released from mitochondria and their particular role in neuronal cell death signaling are however largely unknown. Here, we demonstrate by fluorescence video microscopy that 8-10 h after induction of glutamate toxicity, AIF rapidly translocates from mitochondria to the nucleus and induces nuclear fragmentation and cell death within only a few minutes. This markedly fast translocation of AIF to the nucleus is preceded by increasing translocation of the pro-apoptotic bcl-2 family member Bid (BH3-interacting domain death agonist) to mitochondria, perinuclear accumulation of Bid-loaded mitochondria, and loss of mitochondrial membrane integrity. A small molecule Bid inhibitor preserved mitochondrial membrane potential, prevented nuclear translocation of AIF, and abrogated glutamate-induced neuronal cell death, as shown by experiments using Bid small interfering RNA (siRNA). Cell death induced by truncated Bid was inhibited by AIF siRNA, indicating that caspase-independent AIF signaling is the main pathway through which Bid mediates cell death. This was further supported by experiments showing that although caspase-3 was activated, specific caspase-3 inhibition did not protect neuronal cells against glutamate toxicity. In conclusion, Bid-mediated mitochondrial release of AIF followed by rapid nuclear translocation is a major mechanism of glutamate-induced neuronal death.


Asunto(s)
Factor Inductor de la Apoptosis/metabolismo , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/metabolismo , Muerte Celular/fisiología , Mitocondrias/metabolismo , Neuronas/fisiología , Animales , Factor Inductor de la Apoptosis/genética , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/antagonistas & inhibidores , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/genética , Caspasas/metabolismo , Activación Enzimática , Silenciador del Gen , Ácido Glutámico/toxicidad , Humanos , Ratones , Microscopía Fluorescente , Microscopía por Video , Neuronas/citología , Neuronas/efectos de los fármacos , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
7.
Oncogene ; 26(2): 284-9, 2007 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-16847457

RESUMEN

Solid tumors often have an inadequate blood supply, which results in large regions that are subjected to hypoxic or anoxic stress. Hypoxia-inducible factor-1 (HIF-1) is a transcription factor that regulates much of the transcriptional response of cells to hypoxia. Activating transcription factor 3 (ATF3) is another transcription factor that responds to a variety of stresses and is often upregulated in cancer. We investigated the regulation of ATF3 by oxygen deprivation. ATF3 induction occurred most robustly under anoxia, is common, and it is not dependent on presence of HIF-1 or p53, but is sensitive to the inhibition of c-Jun NH2-terminal kinase activation and the antioxidant N-acetylcystein. ATF3 could also be induced by desferrioxamine but not by the mitochondrial poison cyanide or the nonspecific 2-oxoglutarate dioxygenase inhibitor dimethyloxalylglycine. We also show that anoxic ATF3 mRNA is more stable than normoxic mRNA providing a mechanism for this induction. Thus, this study demonstrates that the regulation of ATF3 under anoxia is independent of 2-oxoglutarate dioxygenase, HIF-1 and p53, presumably involving multiple regulatory pathways.


Asunto(s)
Factor de Transcripción Activador 3/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Hipoxia/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Acetilcisteína/farmacología , Factor de Transcripción Activador 3/genética , Aminoácidos Dicarboxílicos/farmacología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Células Cultivadas/metabolismo , Células Cultivadas/patología , Cianuros/farmacología , Deferoxamina/farmacología , Activación Enzimática , Depuradores de Radicales Libres/farmacología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Melanoma/metabolismo , Melanoma/patología , Oxigenasas de Función Mixta/antagonistas & inhibidores , Oxigenasas de Función Mixta/metabolismo , Neuronas/metabolismo , Neuronas/patología , Oxígeno/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Sideróforos/farmacología , Factores de Transcripción/genética , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/genética , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo
8.
Cell Death Differ ; 14(8): 1529-41, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17464322

RESUMEN

Acute and chronic neurodegeneration, for example, following brain injury or Alzheimer's disease, is characterized by programmed death of neuronal cells. The present study addresses the role and interaction of p53- and NF-kappaB-dependent mechanisms in delayed neurodegeneration following traumatic brain injury (TBI). After experimental TBI in mice p53 rapidly accumulated in the injured brain tissue and translocated to the nucleus of damaged neurons, whereas NF-kappaB transcriptional activity simultaneously declined. Post-traumatic neurodegeneration correlated with the increase in p53 levels and was significantly reduced by the selective p53 inhibitor pifithrin-alpha (PFT). Strikingly, this protective effect was observed even when PFT treatment was delayed up to 6 h after trauma. Inhibition of p53 activity resulted in the concomitant increase in NF-kappaB transcriptional activity and upregulation of NF-kappaB-target proteins, for example X-chromosomal-linked inhibitor of apoptosis (XIAP). It is interesting to note that inhibition of XIAP abolished the neuroprotective effects of PFT in cultured neurons exposed to camptothecin, glutamate, or oxygen glucose deprivation. In conclusion, delayed neuronal cell death after brain trauma is mediated by p53-dependent mechanisms that involve inhibition of NF-kappaB transcriptional activity. Hence, p53 inhibition provides a promising approach for the treatment of acute brain injury, since it blocks apoptotic pathways and concomitantly triggers survival signaling with a therapeutic window relevant for clinical applications.


Asunto(s)
Apoptosis/fisiología , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , FN-kappa B/genética , Proteína p53 Supresora de Tumor/metabolismo , Animales , Apoptosis/genética , Benzotiazoles/farmacología , Lesiones Encefálicas/genética , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Degeneración Nerviosa/genética , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Embarazo , Ratas , Ratas Sprague-Dawley , Tolueno/análogos & derivados , Tolueno/farmacología , Transcripción Genética , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo
9.
Cell Death Differ ; 14(4): 775-84, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17039248

RESUMEN

Nine-day-old harlequin (Hq) mice carrying the hypomorphic apoptosis-inducing factor (AIF)(Hq) mutation expressed 60% less AIF, 18% less respiratory chain complex I and 30% less catalase than their wild-type (Wt) littermates. Compared with Wt, the infarct volume after hypoxia-ischemia (HI) was reduced by 53 and 43% in male (YX(Hq)) and female (X(Hq)X(Hq)) mice, respectively (P<0.001). The Hq mutation did not inhibit HI-induced mitochondrial release of cytochrome c or activation of calpain and caspase-3. The broad-spectrum caspase inhibitor quinoline-Val-Asp(OMe)-CH(2)-PH (Q-VD-OPh) decreased the activation of all detectable caspases after HI, both in Wt and Hq mice. Q-VD-OPh reduced the infarct volume equally in Hq and in Wt mice, and the combination of Hq mutation and Q-VD-OPh treatment showed an additive neuroprotective effect. Oxidative stress leading to nitrosylation and lipid peroxidation was more pronounced in ischemic brain areas from Hq than Wt mice. The antioxidant edaravone decreased oxidative stress in damaged brains, more pronounced in the Hq mice, and further reduced brain injury in Hq but not in Wt mice. Thus, two distinct strategies can enhance the neuroprotection conferred by the Hq mutation, antioxidants, presumably compensating for a defect in AIF-dependent redox detoxification, and caspase inhibitors, presumably interrupting a parallel pathway leading to cellular demise.


Asunto(s)
Factor Inductor de la Apoptosis/metabolismo , Apoptosis , Hipoxia-Isquemia Encefálica/metabolismo , Hipoxia-Isquemia Encefálica/patología , Neuronas/patología , Clorometilcetonas de Aminoácidos/farmacología , Animales , Animales Recién Nacidos , Antipirina/análogos & derivados , Antipirina/farmacología , Apoptosis/efectos de los fármacos , Factor Inductor de la Apoptosis/deficiencia , Inhibidores de Caspasas , Caspasas/metabolismo , Citocromos c/metabolismo , Edaravona , Femenino , Depuradores de Radicales Libres/farmacología , Hipoxia-Isquemia Encefálica/genética , Masculino , Ratones , Ratones Mutantes , Mitocondrias/metabolismo , Necrosis/genética , Necrosis/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Estrés Oxidativo , Quinolinas/farmacología
10.
Cell Death Dis ; 7: e2214, 2016 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-27148687

RESUMEN

Mitochondrial impairment induced by oxidative stress is a main characteristic of intrinsic cell death pathways in neurons underlying the pathology of neurodegenerative diseases. Therefore, protection of mitochondrial integrity and function is emerging as a promising strategy to prevent neuronal damage. Here, we show that pharmacological inhibition of hypoxia-inducible factor prolyl-4-hydroxylases (HIF-PHDs) by adaptaquin inhibits lipid peroxidation and fully maintains mitochondrial function as indicated by restored mitochondrial membrane potential and ATP production, reduced formation of mitochondrial reactive oxygen species (ROS) and preserved mitochondrial respiration, thereby protecting neuronal HT-22 cells in a model of glutamate-induced oxytosis. Selective reduction of PHD1 protein using CRISPR/Cas9 technology also reduced both lipid peroxidation and mitochondrial impairment, and attenuated glutamate toxicity in the HT-22 cells. Regulation of activating transcription factor 4 (ATF4) expression levels and related target genes may mediate these beneficial effects. Overall, these results expose HIF-PHDs as promising targets to protect mitochondria and, thereby, neurons from oxidative cell death.


Asunto(s)
Hidroxiquinolinas/farmacología , Prolina Dioxigenasas del Factor Inducible por Hipoxia/antagonistas & inhibidores , Neuronas/efectos de los fármacos , Procolágeno-Prolina Dioxigenasa/genética , Inhibidores de Prolil-Hidroxilasa/farmacología , Factor de Transcripción Activador 4/genética , Factor de Transcripción Activador 4/metabolismo , Adenosina Trifosfato/agonistas , Adenosina Trifosfato/biosíntesis , Animales , Apoptosis/efectos de los fármacos , Sistemas CRISPR-Cas , Línea Celular , Regulación de la Expresión Génica , Ácido Glutámico/toxicidad , Hipocampo/citología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Prolina Dioxigenasas del Factor Inducible por Hipoxia/genética , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Potencial de la Membrana Mitocondrial/genética , Ratones , Neuronas/citología , Neuronas/metabolismo , Fosforilación Oxidativa/efectos de los fármacos , Estrés Oxidativo , Procolágeno-Prolina Dioxigenasa/deficiencia , Procolágeno-Prolina Dioxigenasa/metabolismo , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal
11.
Cell Death Differ ; 23(5): 814-27, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26586570

RESUMEN

Alteration of endoplasmic reticulum (ER) Ca(2+) homeostasis leads to excessive cytosolic Ca(2+) accumulation and delayed neuronal cell death in acute and chronic neurodegenerative disorders. While our recent studies established a protective role for SK channels against excessive intracellular Ca(2+) accumulation, their functional role in the ER has not been elucidated yet. We show here that SK2 channels are present in ER membranes of neuronal HT-22 cells, and that positive pharmacological modulation of SK2 channels with CyPPA protects against cell death induced by the ER stressors brefeldin A and tunicamycin. Calcium imaging of HT-22 neurons revealed that elevated cytosolic Ca(2+) levels and decreased ER Ca(2+) load during sustained ER stress could be largely prevented by SK2 channel activation. Interestingly, SK2 channel activation reduced the amount of the unfolded protein response transcription factor ATF4, but further enhanced the induction of CHOP. Using siRNA approaches we confirmed a detrimental role for ATF4 in ER stress, whereas CHOP regulation was dispensable for both, brefeldin A toxicity and CyPPA-mediated protection. Cell death induced by blocking Ca(2+) influx into the ER with the SERCA inhibitor thapsigargin was not prevented by CyPPA. Blocking the K(+) efflux via K(+)/H(+) exchangers with quinine inhibited CyPPA-mediated neuroprotection, suggesting an essential role of proton uptake and K(+) release in the SK channel-mediated neuroprotection. Our data demonstrate that ER SK2 channel activation preserves ER Ca(2+) uptake and retention which determines cell survival in conditions where sustained ER stress contributes to progressive neuronal death.


Asunto(s)
Calcio/metabolismo , Muerte Celular , Estrés del Retículo Endoplásmico , Retículo Endoplásmico/metabolismo , Homeostasis , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo , Línea Celular , Supervivencia Celular , Humanos
12.
J Neurosci ; 21(3): 798-811, 2001 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-11157066

RESUMEN

To test the hypothesis of an involvement of tachykinins in destabilization and hyperexcitation of neuronal circuits, gliosis, and neuroinflammation during cerebral ischemia, we investigated cell-specific expressional changes of the genes encoding substance P (SP), neurokinin B (NKB), and the tachykinin/neurokinin receptors (NK1, NK2, and NK3) after middle cerebral artery occlusion (MCAO) in the rat. Our analysis by quantitative in situ hybridization, immunohistochemistry, and confocal microscopy was concentrated on cerebrocortical areas that survive primary infarction but undergo secondary damage. Here, SP-encoding preprotachykinin-A and NK1 mRNA levels and SP-like immunoreactivity were transiently increased in GABAergic interneurons at 2 d after MCAO. Coincidently, MCAO caused a marked expression of SP and NK1 in a subpopulation of glutamatergic pyramidal cells, and in some neurons SP and NK1 mRNAs were coinduced. Elevated levels of the NKB-encoding preprotachykinin-B mRNA and of NKB-like immunoreactivity at 2 and 7 d after MCAO were confined to GABAergic interneurons. In parallel, the expression of NK3 was markedly downregulated in pyramidal neurons. MCAO caused transient NK1 expression in activated cerebrovenular endothelium within and adjacent to the infarct. NK1 expression was absent from activated astroglia or microglia. The differential ischemia-induced plasticity of the tachykinin system in distinct inhibitory and excitatory cerebrocortical circuits suggests that it may be involved in the balance of endogenous neuroprotection and neurotoxicity by enhancing GABAergic inhibitory circuits or by facilitating glutamate-mediated hyperexcitability. The transient induction of NK1 in cerebrovenular endothelium may contribute to ischemia-induced edema and leukocyte diapedesis. Brain tachykinin receptors are proposed as potential drug targets in stroke.


Asunto(s)
Isquemia Encefálica/metabolismo , Corteza Cerebral/metabolismo , Endotelio Vascular/metabolismo , Receptores de Taquicininas/biosíntesis , Taquicininas/biosíntesis , Animales , Isquemia Encefálica/genética , Isquemia Encefálica/patología , Corteza Cerebral/irrigación sanguínea , Corteza Cerebral/patología , Circulación Cerebrovascular , Regulación de la Expresión Génica , Ácido Glutámico/metabolismo , Infarto de la Arteria Cerebral Media/genética , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/patología , Masculino , Neuroquinina B/genética , Neuroquinina B/metabolismo , Plasticidad Neuronal , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Long-Evans , Receptores de Neuroquinina-1/genética , Receptores de Neuroquinina-1/metabolismo , Receptores de Neuroquinina-3/genética , Receptores de Neuroquinina-3/metabolismo , Receptores de Taquicininas/genética , Sustancia P/genética , Sustancia P/metabolismo , Taquicininas/genética , Taquicininas/metabolismo , Vénulas/metabolismo , Vénulas/patología , Ácido gamma-Aminobutírico/metabolismo
13.
J Cereb Blood Flow Metab ; 18(9): 1032-9, 1998 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-9740107

RESUMEN

We have previously demonstrated that the neuroprotective effect of the beta2-adrenoceptor agonist clenbuterol in vitro and in vivo was most likely mediated by an increased nerve growth factor (NGF) expression. In the present study, we examined whether clenbuterol was capable of inhibiting apoptosis caused by ischemia. Transient forebrain ischemia was performed in male Wistar rats (300 to 350 g) by clamping both common carotid arteries and reducing the blood pressure to 40 mm Hg for 10 minutes. Clenbuterol (0.1, 0.5, and 1.0 mg/kg intraperitoneally) was administered 3 hours before ischemia or immediately after ischemia. The brains were removed for histologic evaluation 7 days after ischemia. The time course of DNA fragmentation was determined 1, 2, 3 and 4 days after ischemia. Staining with terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick end-labeling (TUNEL) was used for further analysis of DNA fragments in situ 3 days after ischemia. The NGF protein was assayed by enzyme-linked immunosorbent assay. Ten-minute forebrain ischemia damaged 80% to 90% of the neurons in the hippocampal CA1 region evaluated 7 days after ischemia. Pretreatment with clenbuterol (0.5 and 1.0 mg/kg) reduced the neuronal damage by 18.1% (P < 0.01) and 13.1% (P < 0.05), respectively. The neuroprotective effect also was found when clenbuterol (0.5 mg/kg) was administered immediately after ischemia (P < 0.05). The DNA laddering appeared in striatum 1 day and in hippocampus 2 days after ischemia and peaked on the third day in both regions. The DNA laddering was nearly abolished in the hippocampus and partially blocked in striatum and cortex by 0.5 mg/kg clenbuterol. These results were confirmed by TUNEL staining. Clenbuterol (0.5 mg/kg intraperitoneally) elevated the NGF protein level by 33% (P < 0.05) in the hippocampus and 41% (P < 0.05) in the cortex 6 hours after ischemia. Three days after ischemia, the NGF levels in these regions were no longer different between the clenbuterol-treated and control groups. This study clearly demonstrates that clenbuterol possesses a neuroprotective activity and a marked capacity to inhibit DNA degradation after global ischemia. The results suggest that clenbuterol increases NGF expression during the first hours after global ischemia and thereby protects neurons against apoptotic damage.


Asunto(s)
Agonistas Adrenérgicos beta/uso terapéutico , Apoptosis/efectos de los fármacos , Encéfalo/efectos de los fármacos , Clenbuterol/uso terapéutico , Ataque Isquémico Transitorio/tratamiento farmacológico , Prosencéfalo/irrigación sanguínea , Animales , Encéfalo/patología , Fragmentación del ADN , ADN Nucleotidilexotransferasa/metabolismo , Nucleótidos de Desoxiuracil/metabolismo , Ensayo de Inmunoadsorción Enzimática , Masculino , Factores de Crecimiento Nervioso/biosíntesis , Neuronas/efectos de los fármacos , Neuronas/patología , Ratas , Ratas Wistar , Estimulación Química
14.
J Cereb Blood Flow Metab ; 21(4): 334-43, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11323519

RESUMEN

After a stroke many neurons in the ischemic brain tissue die by a process called apoptosis, a form of cell death that may be preventable. The specific molecular cascades that mediate ischemic neuronal death are not well understood. The authors recently identified prostate apoptosis response-4 (Par-4) as a protein that participates in the death of cultured hippocampal neurons induced by trophic factor withdrawal and exposure to glutamate. Here, the authors show that Par-4 levels increase in vulnerable populations of hippocampal and striatal neurons in rats after transient forebrain ischemia; Par-4 levels increased within 6 hours of reperfusion and remained elevated in neurons undergoing apoptosis 3 days later. After transient focal ischemia in mice, Par-4 levels were increased 6 to 12 hours after reperfusion in the infarcted cortex and the striatum, and activation of caspase-8 occurred with a similar time course. Par-4 immunoreactivity was localized predominantly in cortical neurons at the border of the infarct area. A Par-4 antisense oligonucleotide protected cultured hippocampal neurons against apoptosis induced by chemical hypoxia and significantly reduced focal ischemic damage in mice. The current data suggest that early up-regulation of Par-4 plays a pivotal role in ischemic neuronal death in animal models of stroke and cardiac arrest.


Asunto(s)
Apoptosis/fisiología , Proteínas Portadoras/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Ataque Isquémico Transitorio/metabolismo , Neuronas/citología , Animales , Proteínas Reguladoras de la Apoptosis , Proteínas Portadoras/genética , Caspasas/metabolismo , Células Cultivadas , Cuerpo Estriado/citología , Hipocampo/citología , Infarto de la Arteria Cerebral Media/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/enzimología , Oligodesoxirribonucleótidos Antisentido/farmacología , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Daño por Reperfusión/metabolismo , Accidente Cerebrovascular/metabolismo
15.
J Cereb Blood Flow Metab ; 19(11): 1263-9, 1999 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-10566973

RESUMEN

Estrogens have been suggested for the treatment of neurodegenerative disorders, including stroke, because of their neuroprotective activities against various neurotoxic stimuli such as glutamate, glucose deprivation, iron, or beta-amyloid. Here, the authors report that 17beta-estradiol (0.3 to 30 mg/kg) and 2-OH-estradiol (0.003 to 30 mg/kg) reduced brain tissue damage after permanent occlusion of the middle cerebral artery in male NMRI mice. In vitro, 17beta-estradiol (1 to 10 micromol/L) and 2-OH-estradiol (0.01 to 1 micromol/L) reduced the percentage of damaged chick embryonic neurons treated with FeSO4. In these primary neurons exposed to FeSO4, the authors also found reactive oxygen species to be diminished after treatment with 17beta-estradiol (1 to 10 micromol/L) or 2-OH-estradiol (0.01 to 10 micromol/L), suggesting a strong antioxidant activity of the estrogens that were used. Neither the neuroprotective effect nor the free radical scavenging properties of the estrogens were influenced by the estrogen receptor antagonist tamoxifen. The authors conclude that estrogens protect neurons against damage by radical scavenging rather than through estrogen receptor activation.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Estradiol/farmacología , Animales , Antioxidantes/metabolismo , Antioxidantes/farmacología , Isquemia Encefálica/metabolismo , Células Cultivadas , Estradiol/uso terapéutico , Antagonistas de Estrógenos/farmacología , Masculino , Ratones , Receptores de Estradiol/metabolismo , Tamoxifeno/farmacología
16.
Neuroscience ; 123(4): 897-906, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-14751283

RESUMEN

Prevention of neuronal apoptosis has been introduced as a new therapeutic strategy for neurodegenerative disorders. We have previously reported anti-apoptotic effects of transforming growth factor-beta1 (TGF-beta1), a multifunctional cytokine, in models of cerebral ischemia and in cultured neurons and recently focused on the mechanisms underlying the anti-apoptotic effect of TGF-beta1. The anti-apoptotic transcriptional factor nuclear factor kappa B (NF-kappaB) shows high impact in the cell survival function of multiple cytokines and growth factors. The present study explored whether NF-kappabeta is a target of TGF-beta1 and which signaling pathways involved in the activation of NF-kappabeta are triggered by TGF-beta1. We demonstrated that TGF-beta1 increased the transcriptional activity of NF-kappabeta in cultured hippocampal neurons in a time- and concentration-dependent manner. Furthermore, TGF-beta1 induced translocation of p65/NF-kappabeta to the nucleus and enhanced NF-kappabeta transcriptional activity in the presence of apoptotic stimuli. TGF-beta1-mediated NF-kappabeta activation was blocked by wortmannin and U0126, indicating the involvement of both phosphatidylinositol-3-OH kinase (PI3k)/Akt and mitogen-activated protein kinase (MAPK)/extracellular-signal regulated kinase (Erk)1,2 pathways in the action of TGF-beta1. TGF-beta1 produced a concomitant increase in the phosphorylations of Ikappabeta kinase (IKKalpha/beta) and Ikappabetaalpha with a subsequent degradation of Ikappabetaalpha. Interestingly, the increased phosphorylation of IKKalpha/beta and Ikappabetaalpha was abrogated by wortmannin, but not by U0126, suggesting that PI3k/Akt and MAPK/Erk1,2 pathways triggered by TGF-beta1 regulated the activation of NF-kappabeta through different mechanisms. Of note, wortmannin and U0126, as well as kappabeta-decoy DNA, abolished the anti-apoptotic effect of TGF-beta1, corroborating the notion that both PI3k/Akt and MAPK/Erk1,2 pathways, and NF-kappabeta activity are necessary for the anti-apoptotic activity of TGF-beta1.


Asunto(s)
Proteínas de Unión al Calcio , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/fisiología , FN-kappa B/metabolismo , Fármacos Neuroprotectores/farmacología , Fosfatidilinositol 3-Quinasas/fisiología , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas/fisiología , Factor de Crecimiento Transformador beta/farmacología , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Western Blotting/métodos , Recuento de Células , Células Cultivadas , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Embrión de Mamíferos , Inhibidores Enzimáticos/farmacología , Hipocampo/citología , Inmunohistoquímica/métodos , Sistema de Señalización de MAP Quinasas/fisiología , Glicoproteínas de Membrana/metabolismo , Ratones , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas del Tejido Nervioso/metabolismo , Neuronas/efectos de los fármacos , Neuronas/fisiología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt , Ratas , Ratas Endogámicas F344 , Estaurosporina/toxicidad , Sinaptotagmina I , Sinaptotagminas , Factores de Tiempo , Factor de Crecimiento Transformador beta1 , Factor de Necrosis Tumoral alfa/farmacología
17.
Neuroscience ; 107(4): 593-602, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11720783

RESUMEN

A protective capacity of transforming growth factor-beta1 (TGF-beta1) against various insults inducing neurone cell death in vitro and in vivo has been well established. We have recently shown the rapid up-regulation and persistent expression of TGF-beta1 in surviving CA1 pyramidal cells after cerebral ischemia suggesting an endogenous mechanism of neuroprotection by this multifunctional cytokine. In the present study, we demonstrated that intraperitoneal administration of clenbuterol, a lipophilic beta(2)-adrenoceptor agonist, caused an increase in TGF-beta1 expression in non-ischemic rats and further enhanced TGF-beta1 protein levels in rat CA1 pyramidal neurones after transient forebrain ischemia. In the hippocampus neuroprotection by clenbuterol (0.5 mg/kg) was accompanied by increased TGF-beta1 immunoreactivity as early as 3 h, and remained elevated up to 2 days after ischemia. The corresponding increased TGF-beta1 mRNA levels after ischemia were not further enhanced by clenbuterol, suggesting post-transcriptional regulation of TGF-beta1 protein after beta(2)-adrenoceptor stimulation. In saline-treated rats latent TGF-beta-binding protein-1 (LTBP-1) immunoreactivity was moderately elevated 3 and 6 h after ischemia, and returned to control levels after 1 day of reperfusion. In parallel with the up-regulation of TGF-beta1 immunoreactivity, LTBP-1 levels in the hippocampus were considerably increased by clenbuterol from 3 h to 2 days after ischemia. Our data demonstrate a concomitant increase in LTBP-1 and TGF-beta1 expression in the ischemic hippocampus after stimulation of beta(2)-adrenoceptors.


Asunto(s)
Proteínas Portadoras/genética , Hipocampo/fisiología , Péptidos y Proteínas de Señalización Intracelular , Ataque Isquémico Transitorio/fisiopatología , Receptores Adrenérgicos beta 2/metabolismo , Factor de Crecimiento Transformador beta/genética , Agonistas Adrenérgicos beta/farmacología , Animales , Proteínas Portadoras/análisis , Clenbuterol/farmacología , Expresión Génica/fisiología , Hipocampo/irrigación sanguínea , Hipocampo/química , Ataque Isquémico Transitorio/tratamiento farmacológico , Proteínas de Unión a TGF-beta Latente , Masculino , ARN Mensajero/análisis , Ratas , Ratas Wistar , Factor de Crecimiento Transformador beta/análisis , Factor de Crecimiento Transformador beta1
18.
Neuroscience ; 90(4): 1255-63, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-10338295

RESUMEN

It is well known that proteins encoded by the Bcl-2 gene family play a major role in the regulation of apoptosis. We have demonstrated previously that neuronal apoptosis can be induced in the hippocampus and striatum after global ischemia. Clenbuterol, a beta2-adrenoceptor agonist, showed considerable activity against neuronal apoptosis. In the present study, we attempted to find out whether the members of the Bcl-2 family are induced after ischemia, and whether expression of these genes could be altered by clenbuterol. Transient forebrain ischemia was performed in male Wistar rats by clamping both common carotid arteries and reducing the blood pressure to 40 mmHg for 10 min. Clenbuterol (0.5 mg/kg, i.p.) or vehicle were injected 3 h before onset of ischemia or in non-ischemic rats. The hippocampus and striatum were taken from non-ischemic rats 3, 6 and 24 h after injection of clenbuterol, as well as from drug-treated and untreated rats 6 and 24 h after ischemia. Eighty micrograms/lane total protein were loaded on a 15% sodium dodecyl sulfate-polyacrylamide gel for western blotting. Bcl-2, Bax and Bcl-xl proteins were detectable in the non-ischemic hippocampus and the striatum. Clenbuterol up-regulated the expression of Bcl-2 protein at 3, 6 and 24 h after administration. Enhanced Bcl-xl signals were found in the non-ischemic striatum 3, 6 and 24 h after clenbuterol treatment, but no change of Bcl-xl expression by clenbuterol was seen in the non-ischemic hippocampus. Bax expression was not altered by clenbuterol in the non-ischemic hippocampus and striatum. Bcl-2 was up-regulated in both detected regions at 24 h after ischemia, while the increase in Bax and Bcl-xl protein expression had appeared already at 6 h and also 24 h after ischemia. Clenbuterol further increased the expression of Bcl-2 at 6 and 24 h after ischemia. In contrast, Bax protein level was down-regulated by clenbuterol at 6 and 24 h after ischemia. Clenbuterol also increased Bcl-xl level in the ischemic striatum. The results suggest that global ischemia induces proto-oncogenes which are associated with apoptosis. Clenbuterol not only increased Bcl-2 expression in the non-ischemic hippocampus and striatum, but also up-regulated Bcl-2 and down-regulated Bax expression in the ischemic hippocampus and striatum. The increase in the ratio of Bcl-2 and Bax may contribute to the anti-apoptotic effect of clenbuterol. The present study indicates that pharmacological modulation of Bcl-2 family member expression could become a new strategy to interfere with neuronal damage.


Asunto(s)
Agonistas Adrenérgicos beta/farmacología , Clenbuterol/farmacología , Ataque Isquémico Transitorio/metabolismo , Prosencéfalo/irrigación sanguínea , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Animales , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Ratas , Ratas Wistar , Valores de Referencia , Proteína X Asociada a bcl-2 , Proteína bcl-X
19.
Neuroscience ; 115(4): 1089-108, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12453482

RESUMEN

The role of the common neurotrophin receptor p75 (p75NTR) in neuronal survival and cell death remains controversial. On the one hand, p75NTR provides a positive modulatory influence on nerve growth factor (NGF) signaling through the high affinity neurotrophin receptor TrkA, and hence increases NGF survival signaling. However, p75NTR may also signal independently of TrkA, causing cell death or cell survival, depending on the cell type and stage of development. Here we demonstrate that TrkA is expressed in primary cultures of hippocampal neurons and is activated by NGF within 10 min of exposure. In primary hippocampal cultures neuroprotection by NGF against glutamate toxicity was mediated by NF-kappaB and accompanied by an increased expression of neuroprotective NF-kappaB target genes Bcl-2 and Bcl-xl. In mouse hippocampal cells lacking p75NTR (p75NTR-/-) activation of TrkA by NGF was not detectable. Moreover, neuroprotection by NGF against glutamate toxicity was abolished in p75NTR-/- neurons, and the expression of bcl-2 and bcl-xl was markedly reduced as compared to wildtype cells. NGF increased TrkA phosphorylation in hippocampal neurons and provided protection that required phosphoinositol-3-phosphate (PI3)-kinase activity and Akt phosphorylation, whereas the mitogen-activated protein kinases (MAPK), extracellular-regulated kinases (Erk) 1/2, were not involved. P75NTR signaling independent of TrkA, such as increased neutral sphingomyelinase (NSMase) activity causing enhanced levels of ceramide, were not detected after exposure of hippocampal neurons to NGF. Interestingly, inhibition of sphingosine-kinase blocked the neuroprotective effect of NGF, suggesting that sphingosine-1-phosphate was also involved in NGF-mediated survival in our cultured hippocampal neurons. Overall, our results indicate an essential role for p75NTR in supporting NGF-triggered TrkA signaling pathways mediating neuronal survival in hippocampal neurons.


Asunto(s)
Supervivencia Celular/genética , Hipocampo/metabolismo , Factor de Crecimiento Nervioso/metabolismo , Neuronas/metabolismo , Proteínas Serina-Treonina Quinasas , Receptor trkA/metabolismo , Receptores de Factor de Crecimiento Nervioso/deficiencia , Transducción de Señal/genética , Animales , Factor Neurotrófico Derivado del Encéfalo/farmacología , Supervivencia Celular/efectos de los fármacos , Agonistas de Aminoácidos Excitadores/farmacología , Femenino , Hipocampo/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , FN-kappa B/efectos de los fármacos , FN-kappa B/metabolismo , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/metabolismo , Fármacos Neuroprotectores/farmacología , Células PC12 , Fosfatidilinositol 3-Quinasas/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Proteínas Proto-Oncogénicas/efectos de los fármacos , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor de Factor de Crecimiento Nervioso , Receptor trkA/efectos de los fármacos , Receptor trkA/genética , Receptores de Factor de Crecimiento Nervioso/genética , Transducción de Señal/efectos de los fármacos
20.
J Mol Neurosci ; 17(1): 45-58, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11665862

RESUMEN

Adenosine monophosphate-activated protein kinase (AMPK) is a member of metabolite-sensing kinase family that plays important roles in responses of muscle cells to metabolic stress. AMPK is a heterotrimer of a catalytic alpha subunit (alpha1 or alpha2), and beta (beta1 or beta2) and gamma (gamma1 or gamma2) subunits. Because the brain has a high metabolic rate and is sensitive to changes in the supply of glucose and oxygen, we investigated the expression of AMPK in rat embryonic and adult brain and its role in modifying neuronal survival under conditions of cellular stress. We report that catalytic (alpha1 and alpha2) and noncatalytic (beta2 and gamma1) subunits of AMPK are present at high levels in embryonic hippocampal neurons in vivo and in cell culture. In the adult rat brain, the catalytic subunits alpha1 and alpha2 are present in neurons throughout the brain. The AMPK-activating agent AICAR protected hippocampal neurons against death induced by glucose deprivation, chemical hypoxia, and exposure to glutamate and amyloid beta-peptide. Suppression of levels of the AMPK alpha1 and alpha2 subunits using antisense technology resulted in enhanced neuronal death following glucose deprivation, and abolished the neuroprotective effect of AICAR. These findings suggest that AMPK can protect neurons against metabolic and excitotoxic insults relevant to the pathogenesis of several different neurodegenerative conditions.


Asunto(s)
Aminoimidazol Carboxamida/análogos & derivados , Encéfalo/crecimiento & desarrollo , Supervivencia Celular/fisiología , Glucosa/metabolismo , Complejos Multienzimáticos/metabolismo , Neuronas/enzimología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Quinasas Activadas por AMP , Aminoimidazol Carboxamida/farmacología , Animales , Apoptosis/fisiología , Encéfalo/citología , Encéfalo/embriología , Encéfalo/enzimología , Células Cultivadas , Hipoglucemiantes/farmacología , Inmunohistoquímica , Masculino , Complejos Multienzimáticos/química , Complejos Multienzimáticos/genética , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/fisiología , Fármacos Neuroprotectores/farmacología , Oligonucleótidos Antisentido/metabolismo , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/genética , Subunidades de Proteína , Ratas , Ratas Sprague-Dawley , Ribonucleótidos/farmacología , Triazinas/farmacología , Triazoles/farmacología , Xantinas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA