Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Cell Mol Neurobiol ; 38(6): 1271-1281, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29948553

RESUMO

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.


Assuntos
Sistema Hipotálamo-Hipofisário/metabolismo , Sistema Hipófise-Suprarrenal/metabolismo , Serotonina/metabolismo , Estresse Fisiológico/fisiologia , Animais , Ansiedade , Encéfalo/metabolismo , Corticosterona/metabolismo , Hormônio Liberador da Corticotropina/metabolismo , Masculino , Camundongos Transgênicos , Núcleo Hipotalâmico Paraventricular , Receptores da Neurocinina-1/genética , Receptores da Neurocinina-1/metabolismo
2.
Clin Sci (Lond) ; 128(9): 567-78, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25487516

RESUMO

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.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Leiomiossarcoma/metabolismo , Receptor Tipo 2 de Angiotensina/agonistas , Neoplasias Uterinas/metabolismo , Antineoplásicos/metabolismo , Antineoplásicos/toxicidade , Proteínas Reguladoras de Apoptose/metabolismo , Linhagem Celular Tumoral , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Membrana Celular/patologia , Proliferação de Células , Feminino , Humanos , Leiomiossarcoma/patologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Permeabilidade , Receptor Tipo 2 de Angiotensina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Neoplasias Uterinas/patologia
3.
Mol Cell Neurosci ; 49(3): 300-10, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22206897

RESUMO

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.


Assuntos
Encéfalo/metabolismo , Citocromos c/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Mitocôndrias/metabolismo , Peptídeos/farmacologia , Poli(ADP-Ribose) Polimerases/metabolismo , Animais , Apoptose/fisiologia , Citocromos c/efeitos dos fármacos , Modelos Animais de Doenças , Hipocampo/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Mitocôndrias/efeitos dos fármacos , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Fosforilação/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Proteína X Associada a bcl-2/metabolismo
4.
Naunyn Schmiedebergs Arch Pharmacol ; 396(11): 3315-3326, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37284895

RESUMO

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.


Assuntos
Neoplasias de Próstata Resistentes à Castração , Neoplasias da Próstata , Humanos , Masculino , Dipeptídeos/farmacologia , Compostos Heterocíclicos com 1 Anel/farmacologia , Compostos Heterocíclicos com 1 Anel/química , Antígeno Prostático Específico , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/radioterapia , Neoplasias da Próstata/metabolismo , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Neoplasias de Próstata Resistentes à Castração/radioterapia
5.
J Mol Med (Berl) ; 99(8): 1073-1083, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33864097

RESUMO

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.


Assuntos
Antioxidantes/farmacologia , Isquemia Encefálica/metabolismo , Fármacos Neuroprotetores/farmacologia , Pioglitazona/farmacologia , Transdução de Sinais/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Biomarcadores , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/etiologia , Isquemia Encefálica/patologia , Circulação Cerebrovascular/efeitos dos fármacos , Modelos Animais de Doenças , Suscetibilidade a Doenças , Expressão Gênica , Masculino , Fator 2 Relacionado a NF-E2/metabolismo , Neuroproteção/efeitos dos fármacos , Células PC12 , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos
6.
Trends Pharmacol Sci ; 28(5): 244-9, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17416424

RESUMO

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.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Sistemas de Liberação de Medicamentos , Fármacos Neuroprotetores/farmacologia , PPAR gama/agonistas , Acidente Vascular Cerebral/tratamento farmacológico , Animais , Isquemia Encefálica/fisiopatologia , Diabetes Mellitus Tipo 2/tratamento farmacológico , Vias de Administração de Medicamentos , Humanos , Hipoglicemiantes/farmacologia , Hipoglicemiantes/uso terapêutico , Microglia/metabolismo , Neurônios/metabolismo , Fármacos Neuroprotetores/efeitos adversos , Fármacos Neuroprotetores/uso terapêutico , PPAR gama/metabolismo , Pioglitazona , Acidente Vascular Cerebral/fisiopatologia , Tiazolidinedionas/farmacologia , Tiazolidinedionas/uso terapêutico
7.
Eur J Neurosci ; 28(9): 1786-94, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18973594

RESUMO

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.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Infarto Cerebral/tratamento farmacológico , Encefalite/tratamento farmacológico , Interleucina-6/antagonistas & inibidores , PPAR gama/agonistas , Tiazolidinedionas/farmacologia , Animais , Anti-Inflamatórios/farmacologia , Isquemia Encefálica/imunologia , Isquemia Encefálica/fisiopatologia , Infarto Cerebral/imunologia , Infarto Cerebral/fisiopatologia , Modelos Animais de Doenças , Encefalite/imunologia , Encefalite/fisiopatologia , Hipoglicemiantes/farmacologia , Infarto da Artéria Cerebral Média/imunologia , Infarto da Artéria Cerebral Média/metabolismo , Infarto da Artéria Cerebral Média/fisiopatologia , Interleucina-6/imunologia , Masculino , Degeneração Neural/imunologia , Degeneração Neural/metabolismo , Degeneração Neural/fisiopatologia , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , PPAR gama/imunologia , PPAR gama/metabolismo , Pioglitazona , Ratos , Ratos Wistar , Traumatismo por Reperfusão/imunologia , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/fisiopatologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia
8.
J Hypertens ; 25(1): 187-96, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17143191

RESUMO

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.


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Benzimidazóis/farmacologia , Isquemia Encefálica/tratamento farmacológico , Encéfalo/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Traumatismo por Reperfusão/prevenção & controle , Traumatismo por Reperfusão/fisiopatologia , Acidente Vascular Cerebral/tratamento farmacológico , Tetrazóis/farmacologia , Bloqueadores do Receptor Tipo 1 de Angiotensina II/administração & dosagem , Animais , Benzimidazóis/uso terapêutico , Compostos de Bifenilo , Pressão Sanguínea/efeitos dos fármacos , Encéfalo/patologia , Edema Encefálico/prevenção & controle , Isquemia Encefálica/etiologia , Isquemia Encefálica/patologia , Isquemia Encefálica/fisiopatologia , Circulação Cerebrovascular/efeitos dos fármacos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Esquema de Medicação , Frequência Cardíaca/efeitos dos fármacos , Infarto da Artéria Cerebral Média/complicações , Masculino , Atividade Motora/efeitos dos fármacos , Fármacos Neuroprotetores/administração & dosagem , Desempenho Psicomotor/efeitos dos fármacos , Ratos , Ratos Wistar , Reflexo/efeitos dos fármacos , Acidente Vascular Cerebral/complicações , Acidente Vascular Cerebral/etiologia , Acidente Vascular Cerebral/fisiopatologia , Tetrazóis/uso terapêutico , Fatores de Tempo
9.
FASEB J ; 20(8): 1162-75, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16770015

RESUMO

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.


Assuntos
Isquemia Encefálica/metabolismo , Ciclo-Oxigenase 2/metabolismo , Neurônios/metabolismo , PPAR gama/metabolismo , Animais , Isquemia Encefálica/enzimologia , Sobrevivência Celular , Córtex Cerebral/química , Córtex Cerebral/citologia , Córtex Cerebral/enzimologia , Circulação Cerebrovascular/efeitos dos fármacos , Ciclo-Oxigenase 1/metabolismo , Ciclo-Oxigenase 2/análise , Masculino , Neurônios/enzimologia , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo , PPAR gama/agonistas , PPAR gama/análise , Pioglitazona , Ratos , Ratos Wistar , Tiazolidinedionas/farmacologia , Fator de Necrose Tumoral alfa/metabolismo
10.
Naunyn Schmiedebergs Arch Pharmacol ; 390(9): 949-959, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28669009

RESUMO

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.


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Isquemia Encefálica/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Acidente Vascular Cerebral/tratamento farmacológico , Bloqueadores do Receptor Tipo 1 de Angiotensina II/administração & dosagem , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacocinética , Animais , Anti-Hipertensivos/administração & dosagem , Anti-Hipertensivos/farmacocinética , Anti-Hipertensivos/farmacologia , Benzimidazóis/administração & dosagem , Benzimidazóis/farmacocinética , Benzimidazóis/farmacologia , Compostos de Bifenilo/administração & dosagem , Compostos de Bifenilo/farmacocinética , Compostos de Bifenilo/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Irbesartana , Losartan/administração & dosagem , Losartan/farmacocinética , Losartan/farmacologia , Masculino , Fármacos Neuroprotetores/administração & dosagem , Fármacos Neuroprotetores/farmacocinética , Ratos , Ratos Wistar , Receptor Tipo 1 de Angiotensina/efeitos dos fármacos , Receptor Tipo 1 de Angiotensina/metabolismo , Tetrazóis/administração & dosagem , Tetrazóis/farmacocinética , Tetrazóis/farmacologia , Distribuição Tecidual
11.
Naunyn Schmiedebergs Arch Pharmacol ; 390(1): 37-48, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27664035

RESUMO

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.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Leiomiossarcoma/tratamento farmacológico , Mitocôndrias/efeitos dos fármacos , PPAR gama/agonistas , Tiazolidinedionas/farmacologia , Neoplasias Uterinas/tratamento farmacológico , Proteínas Reguladoras de Apoptose/metabolismo , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ativação Enzimática , Feminino , Humanos , Leiomiossarcoma/metabolismo , Leiomiossarcoma/patologia , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , PPAR gama/metabolismo , Fosforilação , Pioglitazona , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Neoplasias Uterinas/metabolismo , Neoplasias Uterinas/patologia
12.
FASEB J ; 19(6): 617-9, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15665034

RESUMO

Several lines of clinical and experimental evidence suggest an important role of the renin-angiotensin system in ischemic brain injury although the cellular regulation of the angiotensin AT1 and AT2 receptors and their potential relevance in this condition have not yet been clearly defined. We first assessed the regulation of brain AT1 and AT2 receptors in response to transient unilateral medial cerebral artery occlusion in rats by real-time RT-PCR, Western blot, and immunofluorescence labeling. AT2 receptors in the peri-infarct zone were significantly upregulated 2 days after transient focal cerebral ischemia. Increased AT2 receptors, which were abundantly distributed in a large number of brain regions adjacent to the infarct area including cerebral frontal cortex, piriform cortex, striatum, and hippocampus, were exclusively expressed in neurons. By contrast, AT1 receptors, which remained unaltered, were mainly expressed in astrocytes. In neurons of ischemic striatum, increased AT2 receptors were associated with intense neurite outgrowth. Blockade of central AT2 receptors with PD123177 abolished the neuroprotective effects of central AT1 receptor blockade with irbesartan on infarct size and neurological outcome. In primary cortical neurons, stimulation of AT2 receptors supported neuronal survival and neurite outgrowth. Our data indicate that cerebral AT2 receptors exert neuroprotective actions in response to ischemia-induced neuronal injury, possibly by supporting neuronal survival and neurite outgrowth in peri-ischemic brain areas.


Assuntos
Encefalopatias/etiologia , Encefalopatias/prevenção & controle , Isquemia Encefálica/complicações , Neurônios/fisiologia , Receptor Tipo 2 de Angiotensina/fisiologia , Angiotensina II/farmacologia , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Bloqueadores do Receptor Tipo 2 de Angiotensina II , Animais , Astrócitos/química , Western Blotting , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Córtex Cerebral/citologia , Infarto Cerebral/patologia , Imunofluorescência , Expressão Gênica , Masculino , Neuritos/fisiologia , Neurônios/química , Neurônios/citologia , Ratos , Ratos Wistar , Receptor Tipo 1 de Angiotensina/análise , Receptor Tipo 1 de Angiotensina/genética , Receptor Tipo 1 de Angiotensina/fisiologia , Receptor Tipo 2 de Angiotensina/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Distribuição Tecidual
13.
Drug Metab Pharmacokinet ; 31(2): 146-55, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26948953

RESUMO

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.


Assuntos
Elafina/farmacocinética , Inibidores Enzimáticos/farmacocinética , Tecnécio/química , Animais , Elafina/química , Elafina/metabolismo , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Humanos , Elastase Pancreática/antagonistas & inibidores , Elastase Pancreática/metabolismo , Ratos , Distribuição Tecidual
14.
J Hypertens ; 23(12): 2277-85, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16269970

RESUMO

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.


Assuntos
Inibidores da Enzima Conversora de Angiotensina/administração & dosagem , Captopril/administração & dosagem , Hipertensão/tratamento farmacológico , Cloreto de Lítio/administração & dosagem , Acidente Vascular Cerebral/prevenção & controle , Albuminúria/etiologia , Albuminúria/prevenção & controle , Animais , Anti-Hipertensivos/administração & dosagem , Pressão Sanguínea/efeitos dos fármacos , Hipertensão/complicações , Hipertensão/fisiopatologia , Lítio/sangue , Masculino , Potássio/urina , Ratos , Ratos Endogâmicos SHR , Renina/sangue , Sódio/urina , Sódio na Dieta/administração & dosagem , Acidente Vascular Cerebral/etiologia
15.
Naunyn Schmiedebergs Arch Pharmacol ; 388(9): 939-51, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25953200

RESUMO

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.


Assuntos
Inibidores da Enzima Conversora de Angiotensina/administração & dosagem , Isquemia Encefálica/metabolismo , Piridinas/administração & dosagem , Acidente Vascular Cerebral/metabolismo , Tiazepinas/administração & dosagem , Administração Intravenosa , Administração Oral , Inibidores da Enzima Conversora de Angiotensina/farmacocinética , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Infusões Intraventriculares , Masculino , Neprilisina/antagonistas & inibidores , Peptidil Dipeptidase A/metabolismo , Piridinas/farmacocinética , Piridinas/farmacologia , Ratos , Ratos Wistar , Tiazepinas/farmacocinética , Tiazepinas/farmacologia
16.
Stroke ; 34(5): 1287-92, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12677021

RESUMO

BACKGROUND AND PURPOSE: In vitro and in vivo studies have demonstrated neuroprotective actions of lithium. The present study investigated the effect of a low dose of lithium on infarct volume and neurological outcome as well as on apoptotic and inflammatory processes in rats exposed to focal ischemia. METHODS: Cerebral ischemia was induced by middle cerebral artery occlusion (MCAO) for 90 minutes followed by reperfusion. Lithium (1 mmol/kg) was given subcutaneously daily for 14 days before the onset of MCAO and 2 days thereafter. Blood parameters and cerebral blood flow were assessed before, during, and after MCAO. Rats were examined for neurological deficits 24 and 48 hours after MCAO. Two days after MCAO, the brains were removed for immunohistochemical evaluation of caspase-3, terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL), activated microglia, and the expression of AP-1 proteins (c-Fos and c-Jun). Infarct volume was assessed by cresyl violet staining. RESULTS: Pretreatment with lithium did not alter cerebral blood flow or blood parameters. Neurological deficits were significantly decreased in rats treated with lithium at 24 and 48 hours after ischemia. Infarct volume was reduced in rats treated with lithium at 48 hours after ischemia. Lithium significantly decreased the ischemia-induced caspase-3 immunoreactivity and TUNEL staining as well as the AP-1 protein expression in the penumbra of the ischemic cortex. No changes in activated microglia were observed. CONCLUSIONS: The present study demonstrates that chronic treatment with lithium at a low dose exhibits neuroprotection in transient focal cerebral ischemia. Antiapoptotic mechanisms are involved in the lithium-induced neuroprotective effects.


Assuntos
Apoptose/efeitos dos fármacos , Isquemia Encefálica/tratamento farmacológico , Infarto da Artéria Cerebral Média/tratamento farmacológico , Carbonato de Lítio/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Animais , Biomarcadores , Edema Encefálico/etiologia , Edema Encefálico/patologia , Edema Encefálico/prevenção & controle , Isquemia Encefálica/etiologia , Isquemia Encefálica/patologia , Caspase 3 , Caspases/análise , Circulação Cerebrovascular/efeitos dos fármacos , Depressão Química , Esquema de Medicação , Avaliação Pré-Clínica de Medicamentos , Marcação In Situ das Extremidades Cortadas , Infarto da Artéria Cerebral Média/complicações , Infarto da Artéria Cerebral Média/patologia , Carbonato de Lítio/administração & dosagem , Masculino , Microglia/patologia , Proteínas do Tecido Nervoso/análise , Fármacos Neuroprotetores/administração & dosagem , Ratos , Ratos Wistar , Traumatismo por Reperfusão/prevenção & controle , Método Simples-Cego , Fator de Transcrição AP-1/análise
17.
J Cereb Blood Flow Metab ; 24(5): 536-47, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15129186

RESUMO

In the present study, we investigate whether a long-term blockade of brain AT1 receptors in male Wistar rats before and after ischemic injury exerts neuroprotective effects and modulates apoptosis and inflammatory responses, which are associated with the post-ischemic progression of brain damage. The AT1 receptor antagonist irbesartan was continuously infused intracerebroventricularly using osmotic minipumps over a 5-day period before and for 3 or 7 days after middle cerebral artery occlusion (MCAO) for 90 minutes. Neurologic status was evaluated daily, starting 24 hours after MCAO. After MCAO (3 and 7 days), brains were removed for the measurement of infarct size and immunohistochemical evaluation of apoptosis and accumulation of reactive microglia and macrophages. Treatment with irbesartan before ischemia improved motor functions, whereas post-ischemic treatment improved sensory functions. Blockade of brain AT1 receptors reduced the infarct size on days 3 and 7 after MCAO. In the peri-infarct cortex, irbesartan treatment decreased the number of apoptotic cells on day 3 and attenuated the invasion of activated microg-lia and macrophages on days 3 and 7 after ischemia. Long-term blockade of brain AT1 receptors improves the recovery from cerebral ischemia. Antiapoptotic mechanisms and inhibition of post-ischemic inflammation are involved in the AT1 receptor blockade-induced neuroprotective effects in ischemic brain tissue.


Assuntos
Apoptose/fisiologia , Isquemia Encefálica/metabolismo , Inflamação , Receptor Tipo 1 de Angiotensina/metabolismo , Bloqueadores do Receptor Tipo 1 de Angiotensina II , Animais , Compostos de Bifenilo/farmacologia , Encéfalo/citologia , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Encéfalo/fisiologia , Isquemia Encefálica/patologia , Hemodinâmica , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Infarto da Artéria Cerebral Média , Irbesartana , Masculino , Fármacos Neuroprotetores/farmacologia , Fragmentos de Peptídeos/metabolismo , Poli(ADP-Ribose) Polimerase-1 , Poli(ADP-Ribose) Polimerases , Proteínas/metabolismo , Ratos , Ratos Wistar , Tetrazóis/farmacologia
18.
J Hypertens ; 21(11): 2175-82, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14597862

RESUMO

OBJECTIVE: In the present study, we investigated whether systemic pretreatment with the AT1 receptor antagonist, candesartan, reduces neuronal injury after cerebral ischaemia in rats. DESIGN AND METHODS: Focal cerebral ischaemia in male, normotensive Wistar rats was induced by 90 min middle cerebral artery occlusion (MCAO) followed by reperfusion. Experiment 1: Candesartan was injected intravenously (i.v.) at doses of 0.1 or 0.3 mg/kg, 4 h prior to ischaemic injury. Experiment 2: Rats were treated with candesartan [0.1 mg/kg, subcutaneously (s.c.) twice daily], on 5 consecutive days prior to ischaemia. The last injection was administered 12 h before MCAO. Mean arterial pressure (MAP) was measured before, during and after ischaemic injury. Twenty-four hours after ischaemia, neurological outcome, infarct volume and brain oedema were evaluated. RESULTS: Acute i.v. pretreatment with candesartan, 0.1 and 0.3 mg/kg, dose-dependently decreased MAP before, during and after ischaemic injury but did not improve recovery from brain ischaemia. Systemic long-term s.c. pretreatment with 0.1 mg/kg candesartan, reduced MAP during and after ischaemia to the same extent as did the i.v. dose of 0.1 mg administered 4 h before MCAO, but significantly improved neurological outcome and reduced infarction size and oedema of the ipsilateral hemisphere when compared with the vehicle-treated group. CONCLUSION: Long-term blockade of AT1 receptors improves neurological outcome of focal cerebral ischaemia and protects brain tissue against ischaemic injury.


Assuntos
Benzimidazóis/administração & dosagem , Isquemia Encefálica/fisiopatologia , Tetrazóis/administração & dosagem , Bloqueadores do Receptor Tipo 1 de Angiotensina II , Animais , Compostos de Bifenilo , Pressão Sanguínea/efeitos dos fármacos , Infarto Cerebral/patologia , Relação Dose-Resposta a Droga , Esquema de Medicação , Injeções Intravenosas , Injeções Subcutâneas , Masculino , Sistema Nervoso/efeitos dos fármacos , Sistema Nervoso/fisiopatologia , Doenças do Sistema Nervoso/prevenção & controle , Ratos , Ratos Wistar , Recuperação de Função Fisiológica
19.
J Hypertens ; 20(5): 909-18, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-12011652

RESUMO

OBJECTIVE: In the present study, we investigated the ability of the peripherally administered angiotensin II type 1 (AT1) receptor antagonist, candesartan cilexetil, to block central effects of angiotensin II (Ang II) in conscious rats. DESIGN AND METHODS: Candesartan cilexetil was administered orally by gavage at doses of 0.1, 1, 10 and 30 mg/kg. Drinking response, pressor response and release of vasopressin into the circulation following intracerebroventricular (i.c.v.) Ang II (10 or 100 ng) were measured at 0.5, 2, 4 and 24 h following the drug application. The same parameters were measured after chronic treatment with candesartan cilexetil for 1 week. In a separate experiment, the release of vasopressin induced by microinjection of Ang II (100 ng) into the paraventricular nucleus (PVN) was determined 4 h after oral administration of candesartan cilexetil (1 mg/kg) or vehicle. RESULTS: Oral treatment with candesartan cilexetil inhibited all central responses to i.c.v. Ang II in a dose- and time-dependent manner. The Ang II-induced responses were inhibited 4 h after acute or chronic treatment with 0.1 mg/kg candesartan cilexetil, but had returned to control levels 24 h after drug application. In contrast, the highest dose of candesartan cilexetil (30 mg/kg) nearly abolished the central responses to Ang II for 24 h. Candesartan cilexetil completely blocked vasopressin release into the circulation induced by Ang II microinjection into the PVN. CONCLUSIONS: Our results demonstrate that the AT1 receptor antagonist, candesartan cilexetil, very effectively inhibits the centrally mediated effects of Ang II upon peripheral application.


Assuntos
Angiotensina II/farmacologia , Antagonistas de Receptores de Angiotensina , Benzimidazóis/administração & dosagem , Compostos de Bifenilo/administração & dosagem , Encéfalo/efeitos dos fármacos , Tetrazóis , Administração Oral , Angiotensina II/administração & dosagem , Angiotensina II/antagonistas & inibidores , Animais , Arginina Vasopressina/metabolismo , Benzimidazóis/farmacologia , Compostos de Bifenilo/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Relação Dose-Resposta a Droga , Injeções Intraventriculares , Masculino , Microinjeções , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/fisiologia , Ratos , Ratos Wistar , Receptor Tipo 1 de Angiotensina
20.
Int J Pharm ; 477(1-2): 167-75, 2014 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-25455769

RESUMO

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.


Assuntos
Barreira Hematoencefálica/metabolismo , Pentamidina/análogos & derivados , Succinatos/farmacocinética , Tripanossomicidas/farmacocinética , Administração Oral , Animais , Disponibilidade Biológica , Encéfalo/metabolismo , Humanos , Masculino , Pentamidina/farmacocinética , Pró-Fármacos , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Distribuição Tecidual , Tomografia Computadorizada de Emissão de Fóton Único/métodos , Tripanossomíase Africana/tratamento farmacológico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA