Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Naunyn Schmiedebergs Arch Pharmacol ; 396(11): 3315-3326, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37284895

RESUMEN

The human prostate-specific membrane antigen (PSMA) is substantially up-regulated in metastatic prostate cancer (PCa) cells. PSMA can be targeted by 177Lu conjugated to PSMA-617, a high-affinity ligand for the PSMA. The binding of the radioligand, 177Lu-PSMA-617, results in its internalisation and delivery of ß-radiation into the cancer cells. However, PSMA-617, a component of the final product in the synthesis of the radioligand, may also play a role in the pathophysiology of PCa cells. The present study aimed to clarify the effects of PSMA-617 (10, 50 and 100 nM) on the expression of PSMA in PSMA-positive LNCaP cells, their proliferation, 177Lu-PSMA-617-induced cell death by WST-1 and lactate dehydrogenase assays, immunohistochemistry, western blotting, immunofluorescence staining and uptake of 177Lu-PSMA-617. PSMA-617 at 100 nM concentration induced cell-growth arrest, down-regulated cyclin D1 and cyclin E1 (by 43 and 36%, respectively) and up-regulated the cyclin-dependent kinase inhibitor p21Waf1/Cip1 (by 48%). Immunofluorescence staining demonstrated reduced content of DNA, pointing to a lower rate of cell division. PSMA-617 (up to 100 nM) did not alter the uptake of 177Lu-PSMA-617 into the LNCaP cells. Interestingly, simultaneous treatment with 177Lu-PSMA-617 and PSMA-617 for 24 and 48 h substantially potentiated the cell-death promoting effects of the radioligand. In conclusion, the combination of impeding tumour cell proliferation by PSMA-617 and its potentiation of the radiation-induced cell death brought about by 177Lu-PSMA-617 in PCa cells may considerably improve the outcome of the radiation therapy with 177Lu-PSMA-617, especially in patients with decreased radiosensitivity of PCa cells to the radioligand.


Asunto(s)
Neoplasias de la Próstata Resistentes a la Castración , Neoplasias de la Próstata , Humanos , Masculino , Dipéptidos/farmacología , Compuestos Heterocíclicos con 1 Anillo/farmacología , Compuestos Heterocíclicos con 1 Anillo/química , Antígeno Prostático Específico , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/radioterapia , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/radioterapia
2.
J Mol Med (Berl) ; 99(8): 1073-1083, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33864097

RESUMEN

The present study elucidates the neuroprotective mechanisms of the PPARγ (peroxisome proliferator-activated receptor γ) agonist pioglitazone in survival of ischemic neurons following middle cerebral artery occlusion with reperfusion (MCAO). Intracerebroventricular infusion of pioglitazone over 5 days before and 24 or 48 h after MCAO alleviated neurological impairments, inhibited apoptosis 24 h, and activated the PI3K/Akt pathway along with increased phosphorylation of Akt (ser473) and GSK-3ß (ser9) in the peri-infarct cortical areas 48 h after MCAO. In primary cortical neurons, pioglitazone suppressed the glutamate-induced release of lactate dehydrogenase by a PPARγ-dependent mechanism. This protective effect was reversed after co-treatment with PI3K and Akt inhibitors, LY294002 and SH-6, respectively. Pioglitazone enhanced the expression of the antioxidative transcription factor Nrf2 and its target gene protein, heme oxidase-1, in the peri-infarct area. Pioglitazone also increased activation of the antioxidant response element (ARE) in neuronal PC12 cells transfected with the pNQO1-rARE plasmid. We demonstrate in primary cortical neurons from Nrf2 knockout mice that the lack of Nrf2 completely abolished the neuroprotective effects of pioglitazone against oxidative and excitotoxic damage. Our results strongly suggest that the neuroprotective effects of PPARγ in peri-infarct brain tissues comprise the concomitant activation of the PI3K/Akt and Nrf2/ARE pathways. KEY MESSAGES: Pioglitazone inhibits apoptosis in ischemic brain tissue.  Pioglitazone acting on PPARγ activates PI3K/Akt pathway in ischemic brain tissue. Pioglitazone activates via Nrf2 the antioxidant defense pathway in injured neurons. Pioglitazone activates the antioxidant response element in neuronal PC12 cells. Pioglitazone fails to protect primary neurons lacking Nrf2 against oxidative damage. Activation of PPARγ supports the survival of viable neurons in peri-infarct regions.


Asunto(s)
Antioxidantes/farmacología , Isquemia Encefálica/metabolismo , Fármacos Neuroprotectores/farmacología , Pioglitazona/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Biomarcadores , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/etiología , Isquemia Encefálica/patología , Circulación Cerebrovascular/efectos de los fármacos , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Expresión Génica , Masculino , Factor 2 Relacionado con NF-E2/metabolismo , Neuroprotección/efectos de los fármacos , Células PC12 , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas
3.
Cell Mol Neurobiol ; 38(6): 1271-1281, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29948553

RESUMEN

Mice lacking the substance P (SP) neurokinin-1 (NK1) receptor (NK1R-/-mice) were used to investigate whether SP affects serotonin (5-HT) function in the brain and to assess the effects of acute immobilisation stress on the hypothalamic-pituitary-adrenocortical (HPA) axis and 5-HT turnover in individual brain nuclei. Basal HPA activity and the expression of hypothalamic corticotropin-releasing hormone (CRH) in wild-type (WT)- and NK1R-/- mice were identical. Stress-induced increases in plasma ACTH concentration were considerably higher in NK1R-/- mice than in WT mice while corticosterone concentrations were equally elevated in both mouse lines. Acute stress did not alter the expression of CRH. In the dorsal raphe nucleus (DRN), basal 5-HT turnover was increased in NK1R-/- mice and a 15 min stress further magnified 5-HT utilisation in this region. In the frontoparietal cortex, medial prefrontal cortex, central nucleus of amygdala, and the hippocampal CA1 region, stress increased 5-HT and/or 5-hydroxyindoleacetic acid (5-HIAA) concentrations to a similar extent in WT and NK1R-/- mice. 5-HT turnover in the hypothalamic paraventricular nucleus was not affected by stress, but stress induced similar increases in 5-HT and 5-HIAA in the ventromedial and dorsomedial hypothalamic nuclei in WT and NK1R-/- mice. Our findings indicate that NK1 receptor activation suppresses ACTH release during acute stress but does not exert sustained inhibition of the HPA axis. Genetic deletion of the NK1 receptor accelerates 5-HT turnover in DRN under basal and stress conditions. No differences between the responses of serotonergic system to acute stress in WT and NK1R-/- mice occur in forebrain nuclei linked to the regulation of anxiety and neuroendocrine stress responses.


Asunto(s)
Sistema Hipotálamo-Hipofisario/metabolismo , Sistema Hipófiso-Suprarrenal/metabolismo , Serotonina/metabolismo , Estrés Fisiológico/fisiología , Animales , Ansiedad , Encéfalo/metabolismo , Corticosterona/metabolismo , Hormona Liberadora de Corticotropina/metabolismo , Masculino , Ratones Transgénicos , Núcleo Hipotalámico Paraventricular , Receptores de Neuroquinina-1/genética , Receptores de Neuroquinina-1/metabolismo
4.
Naunyn Schmiedebergs Arch Pharmacol ; 390(9): 949-959, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28669009

RESUMEN

The present study conducted in rats defines the requirements for neuroprotective effects of systemically administered AT1 receptor blockers (ARBs) in acute ischaemic stroke. The inhibition of central effects to angiotensin II (ANG II) after intravenous (i.v.) treatment with candesartan (0.3 and 3 mg/kg) or irbesartan and losartan (3 and 30 mg/kg) was employed to study the penetration of these ARBs across the blood-brain barrier. Verapamil and probenecid were used to assess the role of the transporters, P-glycoprotein and the multidrug resistance-related protein 2, in the entry of losartan and irbesartan into the brain. Neuroprotective effects of i.v. treatment with the ARBs were investigated after transient middle cerebral artery occlusion (MCAO) for 90 min. The treatment with the ARBs was initiated 3 h after the onset of MCAO and continued for two consecutive days. Blood pressure was continuously recorded before and during MCAO until 5.5 h after the onset of reperfusion. The higher dose of candesartan completely abolished, and the lower dose of candesartan and higher doses of irbesartan and losartan partially inhibited the drinking response to intracerebroventricular ANG II. Only 0.3 mg/kg candesartan improved the recovery from ischaemic stroke, and 3 mg/kg candesartan did not exert neuroprotective effects due to marked blood pressure reduction during reperfusion. Both doses of irbesartan and losartan had not any effect on the stroke outcome. An effective, long-lasting blockade of brain AT1 receptors after systemic treatment with ARBs without extensive blood pressure reductions is the prerequisite for neuroprotective effects in ischaemic stroke.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Isquemia Encefálica/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Accidente Cerebrovascular/tratamiento farmacológico , Bloqueadores del Receptor Tipo 1 de Angiotensina II/administración & dosificación , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacocinética , Animales , Antihipertensivos/administración & dosificación , Antihipertensivos/farmacocinética , Antihipertensivos/farmacología , Bencimidazoles/administración & dosificación , Bencimidazoles/farmacocinética , Bencimidazoles/farmacología , Compuestos de Bifenilo/administración & dosificación , Compuestos de Bifenilo/farmacocinética , Compuestos de Bifenilo/farmacología , Presión Sanguínea/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Irbesartán , Losartán/administración & dosificación , Losartán/farmacocinética , Losartán/farmacología , Masculino , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/farmacocinética , Ratas , Ratas Wistar , Receptor de Angiotensina Tipo 1/efectos de los fármacos , Receptor de Angiotensina Tipo 1/metabolismo , Tetrazoles/administración & dosificación , Tetrazoles/farmacocinética , Tetrazoles/farmacología , Distribución Tisular
5.
Naunyn Schmiedebergs Arch Pharmacol ; 390(1): 37-48, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27664035

RESUMEN

The peroxisome proliferator-activated receptor γ (PPARγ) agonists, thiazolidinediones, including pioglitazone (PIO) exhibit anti-tumour activities in cancer cells. The present study investigates the effects of PIO on cell proliferation and apoptosis in SK-UT-1 cells, a human uterine leiomyosarcoma cell line, and human uterine smooth muscle cells (HUtSMC). The proliferation and viability of SK-UT-1 cells treated with vehicle or PIO were assessed by cell counting and WST-1 assay. The activity of MEK/ERK and p38 MAPK signalling pathways and the expression of p53, the cyclin-dependent kinase inhibitor, p21, Bax, Bad and Bim proteins and cleaved caspase-3 were analysed by Western blotting. Quiescent SK-UT-1 cells intensively proliferate and display high levels of phosphorylated, activated MEK1/2, ERK1/2 and p38 MAPK. PIO (10 or 25 µM) induced time- and dose-dependently cell-growth arrest, reduced the cell numbers and effectively suppressed the over-activated MEK/ERK and p38 MAPK signalling pathways as evidenced by the abolished levels of phosphorylated MEK1/2, ERK1/2 and p38 MAPK. PIO activated the intrinsic apoptotic pathway, i.e. up-regulated the p53, p21, Bax and Bad proteins and cleaved caspase-3. PIO also reduced cell numbers of highly proliferative SK-UT-1 cells cultured in growth medium. The anti-proliferative and pro-apoptotic actions of PIO were not PPARγ dependent and exclusive for SK-UT-1 cells as PIO did not interfere with the proliferation of HUtSMC. The pronounced anti-tumorigenic effects of PIO in SK-UT-1 cells address an important issue about the relevance of the PPARγ agonist in the treatment of the human uterine leiomyosarcoma.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Leiomiosarcoma/tratamiento farmacológico , Mitocondrias/efectos de los fármacos , PPAR gamma/agonistas , Tiazolidinedionas/farmacología , Neoplasias Uterinas/tratamiento farmacológico , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Activación Enzimática , Femenino , Humanos , Leiomiosarcoma/metabolismo , Leiomiosarcoma/patología , Mitocondrias/metabolismo , Mitocondrias/patología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , PPAR gamma/metabolismo , Fosforilación , Pioglitazona , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Neoplasias Uterinas/metabolismo , Neoplasias Uterinas/patología
6.
Drug Metab Pharmacokinet ; 31(2): 146-55, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26948953

RESUMEN

Elafin is a potent reversible inhibitor of the pro-inflammatory proteases leukocyte elastase and protease 3. It is currently in clinical development for the use in postoperative inflammatory diseases. We investigated the pharmacokinetics of (99m)Tc-labeled elafin ((99m)Tc-Elafin) in blood and individual organs in rat after bolus intravenous injection using the single photon emission tomography (SPECT). (99m)Tc-Elafin predominantly accumulated in the kidney reaching a maximum of 8.5% ± 0.1% of the injected dose per gram (ID/g) at 5 min post injection (p.i) and decreased only slowly during 24 h. In contrast, the initially high radio activity recorded in the other organs rapidly decreased parallel to the radioactivity detected in blood. The blood kinetics fits to a two compartment kinetics model. The radio activity in the dissected kidney was 4.98 ± 1.24%ID/g 24 h p.i, while in other organs, including the brain, no accumulation of (99m)Tc-Elafin was detected. At this time point 30% of the detected radioactivity in the kidney was identified to be not metabolized (99m)Tc-Elafin. In conclusion, the blood and organ-specific kinetic data provide a basis for planning of adequate dosing regimens and the high accumulation of intact elafin in the kidney favors clinical developments targeting inflammatory kidney diseases, such as chronic allograft nephropathy after kidney transplantation.


Asunto(s)
Elafina/farmacocinética , Inhibidores Enzimáticos/farmacocinética , Tecnecio/química , Animales , Elafina/química , Elafina/metabolismo , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Humanos , Elastasa Pancreática/antagonistas & inhibidores , Elastasa Pancreática/metabolismo , Ratas , Distribución Tisular
7.
Naunyn Schmiedebergs Arch Pharmacol ; 388(9): 939-51, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25953200

RESUMEN

Omapatrilat (OMA), which simultaneously inhibits the angiotensin-converting enzyme (ACE) and the neutral endopeptidase (neprilysin (NEP)), is widely used in experimental protocols related to hypertension and heart failure. The penetration of OMA across the blood-brain barrier (BBB) and the effects of ACE/NEP inhibition on the recovery from ischaemic stroke have not yet been investigated. Angiotensin (Ang) I injected intracerebroventricularly (ICV) or intravenously (IV) is converted to Ang II by ACE and induces an immediate increase in blood pressure. The pressor responses to OMA administered ICV, orally or IV were studied in male Wistar rats instrumented with an ICV and arterial and venous catheters. OMA infused ICV rapidly appeared in the systemic circulation and more effectively attenuated the systemic than the central pressor responses to Ang I. OMA administered orally (5, 25, 100 µmol/kg body weight) or IV (0.5, 1, 5, 25 µmol/kg body weight) completely abolished increases in blood pressure to IV Ang I up to 2 h after treatment. The pressor responses to ICV Ang I were not altered, indicating that systemically administered OMA does not cross the BBB. To study the effects of ACE and NEP inhibition in the brain on the recovery from ischaemic stroke, OMA was infused ICV over a 5-day period before and 24 h after the occlusion of the middle cerebral artery (MCAO) for 90 min. ICV application of OMA had no effect on infarction volume and marginally improved neurological outcome. We demonstrate for the first time that simultaneous inhibition of ACE and NEP in the brain tissue does not alter the recovery from ischaemic stroke.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/administración & dosificación , Isquemia Encefálica/metabolismo , Piridinas/administración & dosificación , Accidente Cerebrovascular/metabolismo , Tiazepinas/administración & dosificación , Administración Intravenosa , Administración Oral , Inhibidores de la Enzima Convertidora de Angiotensina/farmacocinética , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Animales , Presión Sanguínea/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Infusiones Intraventriculares , Masculino , Neprilisina/antagonistas & inhibidores , Peptidil-Dipeptidasa A/metabolismo , Piridinas/farmacocinética , Piridinas/farmacología , Ratas , Ratas Wistar , Tiazepinas/farmacocinética , Tiazepinas/farmacología
8.
Clin Sci (Lond) ; 128(9): 567-78, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25487516

RESUMEN

The presence of angiotensin type 2 (AT2) receptors in mitochondria and their role in NO generation and cell aging were recently demonstrated in various human and mouse non-tumour cells. We investigated the intracellular distribution of AT2 receptors including their presence in mitochondria and their role in the induction of apoptosis and cell death in cultured human uterine leiomyosarcoma (SK-UT-1) cells and control human uterine smooth muscle cells (HutSMC). The intracellular levels of the AT2 receptor are low in proliferating SK-UT-1 cells but the receptor is substantially up-regulated in quiescent SK-UT-1 cells with high densities in mitochondria. Activation of the cell membrane AT2 receptors by a concomitant treatment with angiotensin II and the AT1 receptor antagonist, losartan, induces apoptosis but does not affect the rate of cell death. We demonstrate for the first time that the high-affinity, non-peptide AT2 receptor agonist, Compound 21 (C21), penetrates the cell membrane of quiescent SK-UT-1 cells, activates intracellular AT2 receptors and induces rapid cell death; approximately 70% of cells died within 24 h. The cells, which escaped cell death, displayed activation of the mitochondrial apoptotic pathway, i.e. down-regulation of the Bcl-2 protein, induction of the Bax protein and activation of caspase-3. All quiescent SK-UT-1 cells died within 5 days after treatment with a single dose of C21. C21 was devoid of cytotoxic effects in proliferating SK-UT-1 cells and in quiescent HutSMC. Our results point to a new, unique approach enabling the elimination non-cycling uterine leiomyosarcoma cells providing that they over-express the AT2 receptor.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Leiomiosarcoma/metabolismo , Receptor de Angiotensina Tipo 2/agonistas , Neoplasias Uterinas/metabolismo , Antineoplásicos/metabolismo , Antineoplásicos/toxicidad , Proteínas Reguladoras de la Apoptosis/metabolismo , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Membrana Celular/patología , Proliferación Celular , Femenino , Humanos , Leiomiosarcoma/patología , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/patología , Permeabilidad , Receptor de Angiotensina Tipo 2/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Neoplasias Uterinas/patología
9.
Int J Pharm ; 477(1-2): 167-75, 2014 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-25455769

RESUMEN

UNLABELLED: Pentamidine is an effective antiparasitic agent and approved drug for the treatment of African trypanosomiasis (sleeping sickness). However, pentamidine suffers from poor orally bioavailability and lacks central nervous system (CNS) delivery. Therefore its applicability is limited to intravenous or intramuscular treatment of the first stage of the African trypanosomiasis. For this reason, several new pentamidine pro-drugs have been developed with the aim of providing improved orally availability and CNS penetration. AIM: this work aims to measure and to compare the distribution, bioavailability, and ability to cross the blood-brain barrier of [(123)I]-labeled pentamidine and its pro-drugs, N,N'-dihydroxypentamidine and N,N'­bis(succinyloxy) pentamidine, using SPECT (single photon emission computed tomography) after intravenously and per orally administration in rats. METHODS: a total of 60 male Sprague Dawley rats were examined. Each [(123)I]-labeled substance (n=3) was applied to 12 rats (n=6 i.v. and n=6 orally). In two additional test series both [(123)I]iodopentamidine (n=6) and N,N'-bis(succinyloxy)-[(123)I]iodopentamidine (n=6) were administered orally together with the non-radioactive homologues. To evaluate the in vivo stability of the labeled compounds, [(123)I]NaI solution was administered intravenously (n=6) and orally (n=6). In vivo SPECT images were acquired after 30 min, 4h, and 24h and blood samples were taken over 24h. The SPECT images were fusioned with previously acquired magnetic resonance images. After the last SPECT the rats were perfused, sacrificed and the organ γ-radiation levels were determined with a γ-counter. Analysis and quantification of the reconstructed SPECT images was performed using the region of interest technique. RESULTS AND CONCLUSION: the data showed a highly improved oral bioavailability of the [(123)I]-labeled pro-drugs compared to [(123)I]-labeled pentamidine. While [(123)I]iodopentamidine was mainly renally eliminated the pro-drugs were primarily metabolized in the liver and underwent biliary elimination. Considering pentamidine's nephrotoxicity this feature has to be seen as an advantage of the pro-drug principle. Moreover, a significantly higher concentration in the brain was detected after intravenous injection of N,N'-dihydroxy[(123)I]iodopentamidine compared to [(123)I]iodopentamidine. The feasibility of an effective treatment of second stage African trypanosomiasis, in which the parasites already infected the brain, with the herein investigated pro-drugs remains to be clarified with infected animals in additional in vivo studies.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Pentamidina/análogos & derivados , Succinatos/farmacocinética , Tripanocidas/farmacocinética , Administración Oral , Animales , Disponibilidad Biológica , Encéfalo/metabolismo , Humanos , Masculino , Pentamidina/farmacocinética , Profármacos , Ratas , Ratas Sprague-Dawley , Factores de Tiempo , Distribución Tisular , Tomografía Computarizada de Emisión de Fotón Único/métodos , Tripanosomiasis Africana/tratamiento farmacológico
10.
Exp Neurol ; 238(2): 243-53, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22995601

RESUMEN

Thiazolidinediones (TZDs), pioglitazone, rosiglitazone and troglitazone, the synthetic agonists for the PPARγ, administered prior or during ischemic insult improve stroke outcome in rodents, post-occlusion treatments yielded inconsistent results. In the present experiments carried out according to the Stroke Therapy Academic Industry Roundtable (STAIR) guidelines, we studied the effects of post-ischemic pioglitazone treatment on the outcome of focal cerebral ischemia, inflammatory and apoptotic processes, neuronal degeneration and regeneration, blood pressure, heart rate and physiological variables in blood. Male Wistar rats were subjected to a 90 min middle cerebral artery occlusion (MCAO). Subcutaneous (SC) treatment with vehicle or pioglitazone was initiated 90 min after MCAO, i.e. in the post-ischemic, reperfusion phase and continued on 2 (2 day-experiment, protocol 1) or 5 (5-day experiment, protocol 2) consecutive days. In the 2-day experiment, pioglitazone at a dose of 2.5 mg/kg body weight (bw) reduced infarct volume by 31% and oedema by 43% on day 2 after MCAO and attenuated the infiltration of ischemic cortical tissue with activated microglia and macrophages. The slight reduction in infarct volume by approximately 18%, detected in rats treated with 10 mg/kg bw pioglitazone did not reach statistical significance. The neurological scores of sham-operated rats treated with vehicle or 10 mg/kg bw pioglitazone were not significantly different. In rats subjected to cerebral ischemia, post-ischemic treatment with either dose of pioglitazone alleviated particular motor deficits and sensory impairments on day 2 after MCAO. A single injection of 10 mg/kg bw pioglitazone in the reperfusion phase (90 min after the onset of reperfusion) did not modify systolic and diastolic blood pressure, heart rate and physiological variables compared to vehicle-treated rats at any time point after MCAO. In the 5-day experiment, continuous post-occlusion treatment with 2.5 mg/kg body weight pioglitazone significantly reduced cerebral infarction by 29% and improved the partial paralysis of the forelimb and alleviated sensory deficits. In the peri-infarct cortex, pioglitazone effectively suppressed the accumulation of activated microglia/macrophages, inhibited neuronal degeneration and promoted neuroregeneration and formation of neuronal networks. The current results provide evidence that pioglitazone treatment in the post-ischemic, reperfusion phase improves the recovery from ischemic stroke. Neuroprotective effects of pioglitazone are mediated by inhibition of post-ischemic inflammation and neuronal degeneration, protection of neurones against ischemic injury and by promoting of neuronal regeneration. Our data together with previous findings favour the view that pioglitazone is a promising candidate for clinical stroke trials.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Hipoglucemiantes/uso terapéutico , Daño por Reperfusión , Tiazolidinedionas/uso terapéutico , Análisis de Varianza , Animales , Infarto Encefálico/etiología , Infarto Encefálico/prevención & control , Isquemia Encefálica/complicaciones , Isquemia Encefálica/mortalidad , Isquemia Encefálica/patología , Caspasa 9/metabolismo , Corteza Cerebral/irrigación sanguínea , Corteza Cerebral/efectos de los fármacos , Ciclooxigenasa 2/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Ectodisplasinas/metabolismo , Frecuencia Cardíaca/efectos de los fármacos , Masculino , Enfermedades del Sistema Nervioso/etiología , Enfermedades del Sistema Nervioso/prevención & control , Pioglitazona , Poli(ADP-Ribosa) Polimerasas/metabolismo , Ratas , Ratas Wistar , Flujo Sanguíneo Regional/efectos de los fármacos , Factores de Tiempo , Factor de Necrosis Tumoral alfa/metabolismo
11.
Neuropharmacology ; 63(6): 958-65, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22771977

RESUMEN

Mice lacking the NK(1) receptor (NK(1)R-/- mice) and selective, high-affinity, non-peptide, NK(1), NK(2) and NK(3) receptor antagonists were used to identify the tachykinin receptor subtype(s) mediating the central responses induced by neurokinin A (NKA). The peptides, substance P (SP), NKA and senktide and the antagonists were injected intracerebroventricularly (ICV) through an implanted cannula. NKA (50 pmol) was as potent as SP (50 pmol) in inducing grooming behaviour (face washing and hind limb grooming) in wild-type mice, but both peptides failed to induce behavioural responses in NK(1)R-/- mice. In wild-type mice, the NK(1) receptor antagonist, RP 67580 (2 nmol), effectively inhibited grooming behaviour elicited by SP, but was inactive against grooming induced by NKA, which in turn was abolished after pre-treatment with the selective NK(2) receptor agonist, SR 48968 (2 nmol). Unlike NKA, the selective NK(2) receptor agonists, (ß Ala(8)) NKA 4-10 and (NLeu(10)) NKA 4-10, injected ICV at doses of 50 or 100 pmol did not elicit any behavioural response in wild-type mice. The NK(3) receptor antagonist, SR 142801, inhibited behaviours induced by the NK(3) receptor agonist, senktide, but did not alter behavioural responses to either SP or NKA in wild-type mice. The present findings demonstrate that central biological actions of SP and senktide are mediated by activation of NK(1) and NK(3) receptors, respectively. Our results also indicate that NK(1) receptors are essential for generating central actions induced by NKA, which are most probably mediated by a cross-talk between the NK(1) and NK(2) receptors.


Asunto(s)
Encéfalo/efectos de los fármacos , Neuroquinina A/farmacología , Receptor Cross-Talk/efectos de los fármacos , Receptores de Neuroquinina-1/genética , Receptores de Neuroquinina-2/genética , Animales , Conducta Animal/efectos de los fármacos , Benzamidas/farmacología , Relación Dosis-Respuesta a Droga , Aseo Animal/efectos de los fármacos , Inyecciones Intraventriculares , Isoindoles/farmacología , Masculino , Ratones , Ratones Noqueados , Antagonistas del Receptor de Neuroquinina-1 , Fragmentos de Péptidos/farmacología , Piperidinas/farmacología , Receptores de Neuroquinina-1/agonistas , Receptores de Neuroquinina-2/agonistas , Receptores de Neuroquinina-2/antagonistas & inhibidores , Receptores de Neuroquinina-3/agonistas , Receptores de Neuroquinina-3/antagonistas & inhibidores , Estereoisomerismo , Sustancia P/análogos & derivados , Sustancia P/farmacología
12.
Mol Cell Neurosci ; 49(3): 300-10, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22206897

RESUMEN

Kainic acid (KA) induced seizures provokes an extensive neuronal degeneration initiated by c-Jun N-terminal kinases (JNK) as central mediators of excitotoxicity. However, the actions of their individual isoforms in cellular organelles including mitochondria remain to be elucidated. Here, we have studied the activation of JNK1, JNK2 and JNK3 and their activators, mitogen-activated protein kinase kinase (MKK) 4/7, in brain mitochondria, cytosolic and nuclear fractions after KA seizures. In the mitochondrial fraction, KA significantly increased the presence of JNK1, JNK3 and MKK4 and stimulated their phosphorylation i.e. activation. The pro-apoptotic proteins, Bim and Bax were induced and, consequently, the ratio Bcl-2-Bax decreased. These changes were paralleled by the release of cytochrome c and cleavage of poly(ADP-ribose)-polymerase (PARP). The JNK peptide inhibitor, D-JNKI-1 (XG-102) reversed these pathological events in the mitochondria and almost completely abolished cytochrome c release and PARP cleavage. Importantly, JNK3, but not JNK1 or JNK2, was associated with Bim in mitochondria and D-JNKI-1 prevented the formation of this apoptotic complex. Apart from of the attenuation of c-Jun phosphorylation in the nucleus, D-JNKI-1 did not affect the level of JNK3 isoform in the nuclear and cytosolic fractions. These findings provide novel insights into the mode of action of individual JNK isoforms in cell organelles and points to the JNK3 pool in mitochondria as a target of the JNK inhibitor D-JNKI-1 to confer neuroprotection.


Asunto(s)
Encéfalo/metabolismo , Citocromos c/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Mitocondrias/metabolismo , Péptidos/farmacología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Animales , Apoptosis/fisiología , Citocromos c/efectos de los fármacos , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Mitocondrias/efectos de los fármacos , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Fosforilación/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Proteína X Asociada a bcl-2/metabolismo
13.
PLoS One ; 5(11): e15052, 2010 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-21124781

RESUMEN

OBJECTIVE: Pre-treatment with angiotensin receptor blockers is known to improve neurological outcome after stroke. This study investigated for the first time, whether the renin inhibitor aliskiren has similar neuroprotective effects. METHODS: Since aliskiren specifically blocks human renin, double transgenic rats expressing human renin and angiotensinogen genes were used. To achieve a systolic blood pressure of 150 or 130 mmHg animals were treated with aliskiren (7.5 or 12.5 mg/kg*d) or candesartan (1.5 or 10 mg/kg*d) via osmotic minipump starting five days before middle cerebral artery occlusion with reperfusion. Infarct size was determined by magnetic resonance imaging. mRNA of inflammatory marker genes was studied in different brain regions. RESULTS: The mortality of 33.3% (7 of 21 animals) in the vehicle group was reduced to below 10% by treatment with candesartan or aliskiren (p<0.05). Aliskiren-treated animals had a better neurological outcome 7 days post-ischemia, compared to candesartan (Garcia scale: 9.9±0.7 vs. 7.3±0.7; p<0.05). The reduction of infarct size in the aliskiren group did not reach statistical significance compared to candesartan and vehicle (24 h post-ischemia: 314±81 vs. 377±70 and 403±70 mm(3) respectively). Only aliskiren was able to significantly reduce stroke-induced gene expression of CXC chemokine ligand 1, interleukin-6 and tumor necrosis factor-alpha in the ischemic core. CONCLUSIONS: Head-to-head comparison suggests that treatment with aliskiren before and during cerebral ischemia is at least as effective as candesartan in double transgenic rats. The improved neurological outcome in the aliskiren group was blood pressure independent. Whether this effect is due to primary anti-inflammatory mechanisms has to be investigated further.


Asunto(s)
Amidas/farmacología , Angiotensinógeno/fisiología , Fumaratos/farmacología , Renina/fisiología , Accidente Cerebrovascular/tratamiento farmacológico , Angiotensinógeno/antagonistas & inhibidores , Angiotensinógeno/genética , Animales , Animales Modificados Genéticamente , Antihipertensivos/farmacología , Bencimidazoles/farmacología , Compuestos de Bifenilo , Presión Sanguínea/efectos de los fármacos , Encéfalo/irrigación sanguínea , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Isquemia Encefálica/genética , Isquemia Encefálica/fisiopatología , Enfermedades Arteriales Cerebrales/fisiopatología , Trastornos Cerebrovasculares/fisiopatología , Quimiocina CXCL1/genética , Expresión Génica/efectos de los fármacos , Humanos , Interleucina-6/genética , Ratas , Renina/antagonistas & inhibidores , Renina/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Accidente Cerebrovascular/genética , Accidente Cerebrovascular/fisiopatología , Tetrazoles/farmacología , Factor de Necrosis Tumoral alfa/genética
14.
J Hypertens ; 28(7): 1488-97, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20410836

RESUMEN

OBJECTIVE: The imbalance between the production and release of interleukin-1 (IL-1) ligands, IL-1alpha, IL-1beta and IL-1 receptor antagonist (IL-1ra) in ischaemic brain exaggerates inflammatory responses and contributes to neuronal death. Cerebral ischaemia also upregulates the peroxisome-proliferator-activated receptor (PPAR) gamma. We studied in rats the effects of the PPARgamma agonist, pioglitazone, on the regulation of IL-1beta, IL-1ra and IL-1 receptor I (IL-1RI) expression in ischaemic brain after occlusion of the middle cerebral artery for 90 min. METHODS: Pioglitazone or vehicle was infused intracerebroventricularly over a 5-day period before, during and 24 or 48 h after middle cerebral artery occlusion. The expression of IL-1beta, IL-1ra and IL-1RI in the peri-infarct cortex was investigated by immunohistochemistry, Western blotting and immunofluorescence staining. The mechanisms of the IL-1ra regulation by pioglitazone and the neuroprotection under excitotoxic neuronal injury were studied in primary cortical neurones expressing PPARgamma and PPAR beta/delta. RESULTS: Cerebral ischaemia increased the expression of IL-1beta, IL-1RI and IL-1ra in the ischaemic cortex. Pioglitazone reduced IL-1beta, but upregulated IL-1ra and increased the number of IL-1ra immunoreactive cells. In primary cortical neurones, pioglitazone stimulated the IL-1ra production via activation of the PPARbeta/delta, but prevented excitotoxic neuronal injury and death by a PPARgamma-dependent mechanism. CONCLUSION: Our data demonstrate that activation of PPARgamma and PPAR beta/delta by proglitazone in neurones triggers diverse neuroprotective mechanisms. The restoration of the equilibrium between I1-1beta and IL-1ra in ischaemic brain tissue limits IL-1beta signalling, reduces inflammatory responses and is an important mechanism by which thiazolidinediones improve the recovery from ischaemic stroke.


Asunto(s)
Isquemia Encefálica/metabolismo , Proteína Antagonista del Receptor de Interleucina 1/metabolismo , Fármacos Neuroprotectores/uso terapéutico , PPAR gamma/metabolismo , Receptores Activados del Proliferador del Peroxisoma/metabolismo , Tiazolidinedionas/farmacología , Animales , Encéfalo/metabolismo , Infarto Cerebral/metabolismo , Infarto Cerebral/prevención & control , Infarto de la Arteria Cerebral Media/metabolismo , Proteína Antagonista del Receptor de Interleucina 1/farmacología , Interleucina-1/metabolismo , Interleucina-1/farmacología , Masculino , Fármacos Neuroprotectores/farmacología , PPAR gamma/farmacología , PPAR-beta/metabolismo , Receptores Activados del Proliferador del Peroxisoma/farmacología , Pioglitazona , Ratas , Ratas Wistar , Receptores de Interleucina-1/metabolismo , Tiazolidinedionas/metabolismo , Regulación hacia Arriba/efectos de los fármacos
15.
Eur J Neurosci ; 28(9): 1786-94, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18973594

RESUMEN

Interleukin-6 (IL-6) exerts neuroprotective effects after cerebral ischaemia but can also exacerbate inflammation and induce neuronal death. The current study investigates the role of cerebral peroxisome proliferator-activated receptor(s) gamma (PPARgamma) in the regulation of IL-6 expression in the peri-infarct cortical tissue in rats exposed to focal cerebral ischaemia. Pioglitazone, a high-affinity PPARgamma ligand, was infused intracerebroventricularly (i.c.v.) via osmotic minipumps over a 5-day period before, during and 24 h or 48 h after middle cerebral artery occlusion (MCAO) for 90 min followed by reperfusion. The expression of PPARgamma and IL-6 in cortical tissue adjacent to the ischaemic core was studied 24 h and 48 h after MCAO. Pioglitazone augmented the ischaemia-induced upregulation of PPARgamma at both time points. Cerebral ischaemia substantially increased IL-6 expression in the peri-infarct cortical tissue. Twenty-four hours after MCAO, the majority of microglial cells/macrophages showed an intense IL-6 immunoreactivity. IL-6 was also localized in neurons, but the distribution of neurons positively stained for IL-6 at the border of the infarct was very heterogeneous. Pioglitazone effectively decreased the number of IL-6-immunoreactive cells and IL-6 protein levels at 24 h but not at 48 h after MCAO. Pioglitazone treatment reduced the infarct size and improved neurological functions. The present study demonstrates that cerebral PPARgamma suppresses the expression of IL-6 in ischaemic brain tissue during the initial phase of ischaemic stroke, in which the overproduction of IL-6 may aggravate neuronal damage, but not at later time points, when IL-6 promotes neuroprotection and inhibits neuronal death.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Infarto Cerebral/tratamiento farmacológico , Encefalitis/tratamiento farmacológico , Interleucina-6/antagonistas & inhibidores , PPAR gamma/agonistas , Tiazolidinedionas/farmacología , Animales , Antiinflamatorios/farmacología , Isquemia Encefálica/inmunología , Isquemia Encefálica/fisiopatología , Infarto Cerebral/inmunología , Infarto Cerebral/fisiopatología , Modelos Animales de Enfermedad , Encefalitis/inmunología , Encefalitis/fisiopatología , Hipoglucemiantes/farmacología , Infarto de la Arteria Cerebral Media/inmunología , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/fisiopatología , Interleucina-6/inmunología , Masculino , Degeneración Nerviosa/inmunología , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/fisiopatología , Neuroglía/efectos de los fármacos , Neuroglía/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , PPAR gamma/inmunología , PPAR gamma/metabolismo , Pioglitazona , Ratas , Ratas Wistar , Daño por Reperfusión/inmunología , Daño por Reperfusión/metabolismo , Daño por Reperfusión/fisiopatología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología
16.
Trends Pharmacol Sci ; 28(5): 244-9, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17416424

RESUMEN

The peroxisome proliferator activated receptors (PPARs), which belong to the nuclear receptor superfamily, are key regulators of glucose and fat metabolism. The PPAR-gamma isoform is involved in the regulation of cellular glucose uptake, protection against atherosclerosis and control of immune reactions. In addition, the activation of PPAR-gamma effectively attenuates neurodegenerative and inflammatory processes in the brain. Here, we review a novel aspect of beneficial and clinically relevant PPAR-gamma actions: neuroprotection against ischemic injury mediated by intracerebral PPAR-gamma, which is expressed in neurons and microglia. Together with the recent observation that the PPAR-gamma ligand pioglitazone reduces the incidence of stroke in patients with type 2 diabetes, this review supports the concept that activators of PPAR-gamma are effective drugs against ischemic injury.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Sistemas de Liberación de Medicamentos , Fármacos Neuroprotectores/farmacología , PPAR gamma/agonistas , Accidente Cerebrovascular/tratamiento farmacológico , Animales , Isquemia Encefálica/fisiopatología , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Vías de Administración de Medicamentos , Humanos , Hipoglucemiantes/farmacología , Hipoglucemiantes/uso terapéutico , Microglía/metabolismo , Neuronas/metabolismo , Fármacos Neuroprotectores/efectos adversos , Fármacos Neuroprotectores/uso terapéutico , PPAR gamma/metabolismo , Pioglitazona , Accidente Cerebrovascular/fisiopatología , Tiazolidinedionas/farmacología , Tiazolidinedionas/uso terapéutico
17.
J Hypertens ; 25(1): 187-96, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17143191

RESUMEN

OBJECTIVE: The effects of candesartan treatment starting early (3 h) and delayed (24 h) after middle cerebral artery occlusion (MCAO) with reperfusion was investigated in normotensive rats. METHODS: Subcutaneous treatment with candesartan (0.3 and 3 mg/kg) or vehicle was initiated 3 or 24 h after the onset of MCAO and continued for seven consecutive days (n=20 per group and timepoint). Neurological outcome was evaluated daily using two different scoring systems. Infarct and oedema volumes were determined in rats 2 or 7 days after MCAO. Mean arterial, systolic and diastolic blood pressures were recorded before and after the application of candesartan. RESULTS: Mean arterial, systolic and diastolic blood pressures were markedly decreased with the high dose, but only moderately decreased with the low dose of candesartan. Vehicle-treated rats showed marked neurological deficits 24 h after MCAO, which gradually improved with time. Candesartan improved neurological outcomes at all timepoints only when treatment was started 3, but not 24 h after MCAO. The infarct volume was reduced on days 2 and 7 after MCAO in rats treated with the low but not the high dose of candesartan. CONCLUSION: The present study demonstrates that only an early but not a delayed onset of treatment with candesartan exerts neuroprotection after focal ischaemia. The degree of neurological impairments did not correlate with the infarct volume, which was reduced only after the low dose of candesartan. The high dose of candesartan failed to reduce the infarct volume, probably because of an excessive blood pressure decrease.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Bencimidazoles/farmacología , Isquemia Encefálica/tratamiento farmacológico , Encéfalo/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Daño por Reperfusión/prevención & control , Daño por Reperfusión/fisiopatología , Accidente Cerebrovascular/tratamiento farmacológico , Tetrazoles/farmacología , Bloqueadores del Receptor Tipo 1 de Angiotensina II/administración & dosificación , Animales , Bencimidazoles/uso terapéutico , Compuestos de Bifenilo , Presión Sanguínea/efectos de los fármacos , Encéfalo/patología , Edema Encefálico/prevención & control , Isquemia Encefálica/etiología , Isquemia Encefálica/patología , Isquemia Encefálica/fisiopatología , Circulación Cerebrovascular/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Frecuencia Cardíaca/efectos de los fármacos , Infarto de la Arteria Cerebral Media/complicaciones , Masculino , Actividad Motora/efectos de los fármacos , Fármacos Neuroprotectores/administración & dosificación , Desempeño Psicomotor/efectos de los fármacos , Ratas , Ratas Wistar , Reflejo/efectos de los fármacos , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/etiología , Accidente Cerebrovascular/fisiopatología , Tetrazoles/uso terapéutico , Factores de Tiempo
18.
FASEB J ; 20(8): 1162-75, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16770015

RESUMEN

Up-regulation of cyclooxygenase (COX)-2 exacerbates neuronal injury after cerebral ischemia and contributes to neuronal cell death. The present study clarifies the function of cerebral peroxisome-proliferator-activated receptor(s) gamma (PPARgamma) in the expression of COX-2 in neurons of the rat brain after middle cerebral artery occlusion (MCAO) with reperfusion by immunohistochemistry, Western blot, and immunofluorescence staining. In peri-infarct cortical areas the PPARgamma was located in both microglia and neurons, whereas COX-2 was almost exclusively expressed in neurons. PPARgamma immunolabeling reached the peak 12 h after MCAO, whereas the number of COX-2 immunostained cells gradually rose and reached its peak at 48 h. Intracerebroventricular infusion of pioglitazone, an agonist of the PPARgamma, over a 5-day period before and 2 days after MCAO, reduced the infarct size, the expression of tumor necrosis factor alpha (TNF-alpha), COX-2, and the number of cells positively stained for COX-1 and COX-2 in the peri-infarct cortical regions. COX-2 induction was also attenuated in the ipsilateral but not in the contralateral hippocampus. In primary cortical neurons expressing the PPARgamma, pioglitazone suppressed COX-2 expression in response to oxidative stress. This protective effect was reversed after cotreatment with GW 9662, a selective antagonist of the PPARgamma, clearly demonstrating a PPARgamma-dependent mechanism. Our data provide evidence that activation of neuronal PPARgamma considerably contributes to neuroprotection by prevention of COX-2 up-regulation in vitro and in peri-infarct brain areas.


Asunto(s)
Isquemia Encefálica/metabolismo , Ciclooxigenasa 2/metabolismo , Neuronas/metabolismo , PPAR gamma/metabolismo , Animales , Isquemia Encefálica/enzimología , Supervivencia Celular , Corteza Cerebral/química , Corteza Cerebral/citología , Corteza Cerebral/enzimología , Circulación Cerebrovascular/efectos de los fármacos , Ciclooxigenasa 1/metabolismo , Ciclooxigenasa 2/análisis , Masculino , Neuronas/enzimología , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo , PPAR gamma/agonistas , PPAR gamma/análisis , Pioglitazona , Ratas , Ratas Wistar , Tiazolidinedionas/farmacología , Factor de Necrosis Tumoral alfa/metabolismo
19.
J Hypertens ; 23(12): 2277-85, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16269970

RESUMEN

OBJECTIVE: A number of potential interactions between angiotensin-converting enzyme inhibitors and lithium have been described in the literature. In the present study, we investigated the effects of a low-dose combination treatment with lithium and captopril on survival and stroke prevention in salt-loaded, stroke-prone spontaneously hypertensive rats (SHRSP). METHODS: Eight-week-old saline-drinking SHRSP (n = 21 per group) were treated with vehicle, LiCl (1 mmol/kg per day), captopril (25 mg/kg per day) and captopril plus LiCl for up to 37 weeks. Body weight, salt water intake blood pressure and mortality were recorded throughout the experimental period. Plasma renin activity, plasma lithium concentration and urinary excretion of albumin, sodium and potassium were measured at different time points. RESULTS: Captopril treatment doubled the life expectancy when compared with vehicle-treated rats. Lithium alone had minor effects on survival but led to a dramatic increase in survival when added to captopril (mean survival time > 237 versus 147 days, P < 0.001). Systolic blood pressure increased with age in all treatment groups but was comparable in the captopril-treated and the captopril-plus-lithium-treated groups. Plasma renin activity as well as urinary sodium and potassium excretion did not differ between both groups. In the captopril group a striking fivefold increase of albuminuria occurred between 14 and 26 weeks of age, while this progression was completely abolished by the addition of lithium. CONCLUSIONS: Our results demonstrate that the addition of lithium to captopril dramatically prolong the effects of the angiotensin-converting enzyme inhibitor on survival in salt-loaded SHRSP. This effect was independent of a reduction in blood pressure.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/administración & dosificación , Captopril/administración & dosificación , Hipertensión/tratamiento farmacológico , Cloruro de Litio/administración & dosificación , Accidente Cerebrovascular/prevención & control , Albuminuria/etiología , Albuminuria/prevención & control , Animales , Antihipertensivos/administración & dosificación , Presión Sanguínea/efectos de los fármacos , Hipertensión/complicaciones , Hipertensión/fisiopatología , Litio/sangre , Masculino , Potasio/orina , Ratas , Ratas Endogámicas SHR , Renina/sangre , Sodio/orina , Sodio en la Dieta/administración & dosificación , Accidente Cerebrovascular/etiología
20.
J Neurochem ; 94(5): 1395-401, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15992368

RESUMEN

The angiotensin type 2 (AT2) receptor has been previously demonstrated to exert neuroprotective actions possibly by inducing neuronal cell differentiation involving neurite outgrowth. The nuclear hormone receptor peroxisome proliferator-activated receptor gamma (PPARgamma) is an important transcriptional regulator of cell differentiation. The aim of the present study was to clarify whether PPARgamma is involved in AT2-receptor-mediated morphological neuronal cell differentiation. To investigate AT2-receptor-mediated morphological neuronal cell differentiation, rat pheochromocytoma cells (PC12W cells) expressing AT2 but not AT1 receptors, were stimulated with angiotensin II (Ang II, 100 nmol/L) +/- the PPARgamma antagonists GW9662 (3 micromol/L) and bisphenol A diglycidyl ether (BADGE, 1 micromol/L), and neurite outgrowth of these cells was assessed. Ang II induced neurite outgrowth by 19 +/- 1.6-fold (p < 0.01). Antagonizing PPARgamma activity by GW9662 or BADGE potently blocked Ang II-induced neurite outgrowth (Ang II + GW9662: 6.6 +/- 1.5-fold, p < 0.05; Ang II + BADGE: 1.3 +/- 0.7-fold, p < 0.01). AT2 receptor activation by Ang II markedly induced mRNA and protein expression of the PPARgamma2 isoform and enhanced ligand-induced PPARgamma activity in transactivation assays. In conclusion, the present study demonstrates that Ang II induces PPARgamma expression and ligand-mediated PPARgamma activity via AT2 receptor activation, which appears to be a crucial process in AT2 receptor mediated neurite outgrowth. AT2 receptor/PPARgamma-dependent neurite outgrowth may play an important role during neuroprotective processes.


Asunto(s)
Angiotensina II/farmacología , PPAR gamma/biosíntesis , Receptor de Angiotensina Tipo 2/fisiología , Animales , Anexina A2/metabolismo , Humanos , Ligandos , Neuritas/efectos de los fármacos , Neuritas/fisiología , Células PC12 , PPAR gamma/fisiología , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA