Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
1.
Neuropharmacology ; 202: 108835, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34648772

RESUMO

Peripheral nerve injuries (PNIs) often result in persistent neuropathic pain, seriously affecting quality of life. Existing therapeutic interventions for PNI-induced neuropathic pain are far from satisfactory. Extracellular signal-regulated kinases (ERKs) and p38 have been found to participate in triggering and maintaining PNI-induced neuropathic pain. However, ERK and p38 also contribute to axonal regeneration and motor function recovery after PNI, making it difficult to inhibit ERK and p38 for therapeutic purposes. In this study, we simultaneously characterized neuropathic pain and motor function recovery in a mouse sciatic nerve crush injury model to identify the time window for therapeutic interventions. We further demonstrated that delayed delivery of a combination of ERK and p38 inhibitors at three weeks after PNI could significantly alleviate PNI-induced neuropathic pain without affecting motor function recovery. Additionally, the combined use of these two inhibitors could suppress pain markedly better than either inhibitor alone, possibly reducing the required dose of each inhibitor and alleviating the side effects and risks of the inhibitors when used individually.


Assuntos
Butadienos/farmacologia , Butadienos/uso terapêutico , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Imidazóis/farmacologia , Imidazóis/uso terapêutico , Neuralgia/tratamento farmacológico , Neuralgia/etiologia , Nitrilas/farmacologia , Nitrilas/uso terapêutico , Traumatismos dos Nervos Periféricos/complicações , Traumatismos dos Nervos Periféricos/fisiopatologia , Piridinas/farmacologia , Piridinas/uso terapêutico , Nervo Isquiático/lesões , Nervo Isquiático/fisiopatologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia , Animais , Axônios/fisiologia , Modelos Animais de Doenças , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Regeneração Nervosa/genética , Neuralgia/genética , Recuperação de Função Fisiológica , Resultado do Tratamento , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
2.
Sci Rep ; 11(1): 11993, 2021 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-34099834

RESUMO

In acute ischemic stroke, the only FDA-approved drug; recombinant tissue plasminogen activator (rt-PA) is limited by restricted time-window due to an enhanced risk of hemorrhagic transformation which is thought to be caused by metalloproteinase (MMP). In experimental stroke inhibitors of the mitogen-activated protein kinase kinase extracellular signal-regulated kinase kinase (MEK) 1/2 pathways reduce the MMPs. This study evaluated whether a MEK1/2 inhibitor in combination with rt-PA can prevent the detrimental effects of delayed rt-PA therapy in stroke. Thromboembolic stroke was induced in C57 black/6J mice and the MEK1/2 inhibitor U0126 was administrated 3.5 h and rt-PA 4 h post stroke-onset. Treatment with rt-PA demonstrated enhanced MMP-9 protein levels and hemorrhagic transformation which was prevented when U0126 was given in conjunction with rt-PA. By blocking the MMP-9 with U0126 the safety of rt-PA administration was improved and demonstrates a promising adjuvant strategy to reduce the harmful effects of delayed rt-PA treatment in acute ischemic stroke.


Assuntos
Butadienos/farmacocinética , Hemorragia/prevenção & controle , AVC Isquêmico/tratamento farmacológico , Nitrilas/farmacocinética , Ativador de Plasminogênio Tecidual/farmacologia , Animais , Butadienos/uso terapêutico , Modelos Animais de Doenças , Descoberta de Drogas , Quimioterapia Combinada , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/prevenção & controle , Humanos , Masculino , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Nitrilas/uso terapêutico , Transdução de Sinais , Ativador de Plasminogênio Tecidual/uso terapêutico , Resultado do Tratamento
3.
Biochem Biophys Res Commun ; 522(2): 512-517, 2020 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-31784089

RESUMO

Combined LXR ligand (T0901317) and MEK1/2 inhibitor (U0126) not only reduces atherosclerosis in apoE deficient mice, but also blocks LXR ligand-induced fatty liver and hypertriglyceridemia. However, the atheroprotective function of combined T0901317 and U0126 should be further investigated in LDLR deficient (LDLR-/-) mice since deficiency of LDLR not apoE can occur to humans with a high frequency. Herein, we validated the effectiveness of this combinational therapy on the development of atherosclerosis in LDLR-/- mice to demonstrate its potential application in clinic. We found although T0901317 or U0126 alone reduced atherosclerotic plaques in en face and aortic root areas in HFD-fed LDLR-/- mice, their combination inhibited lesions in a synergistic manner. Combined U0126 and T0901317 had no effect on serum total cholesterol levels. T0901317 deceased HDL-cholesterol levels, which was restored by combined U0126. Meanwhile, U0126 alleviated T0901317-induced triglyceride accumulation, the major adverse effect of T0901317 which limits its clinical utility. Mechanistically, U0126 reduced fatty acid de novo synthesis by inhibiting hepatic fatty acid synthase (FASN) expression, thereby correcting T0901317-induced triglyceride overproduction. In conclusion, our study demonstrates that combination of MEK1/2 inhibitor and LXR ligand can synergistically reduce atherosclerosis in LDLR deficient mice without lipogenic side effects.


Assuntos
Aterosclerose/tratamento farmacológico , Receptores X do Fígado/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Inibidores de Proteínas Quinases/uso terapêutico , Receptores de LDL/deficiência , Animais , Aterosclerose/sangue , Aterosclerose/complicações , Butadienos/farmacologia , Butadienos/uso terapêutico , Sinergismo Farmacológico , Fígado Gorduroso/sangue , Fígado Gorduroso/complicações , Fígado Gorduroso/tratamento farmacológico , Feminino , Hipertrigliceridemia/sangue , Hipertrigliceridemia/complicações , Ligantes , Lipídeos/sangue , Masculino , Camundongos Knockout , Nitrilas/farmacologia , Nitrilas/uso terapêutico , Placa Aterosclerótica/sangue , Placa Aterosclerótica/complicações , Placa Aterosclerótica/tratamento farmacológico , Placa Aterosclerótica/patologia , Receptores de LDL/metabolismo
4.
Cell Signal ; 63: 109381, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31374291

RESUMO

Multidrug resistance is a major treatment obstacle for recurrent and metastatic bladder cancer, which often leads to disease progression and poor clinical outcome. Although overexpression of interleukin-6 (IL-6) appears to play a critical role in the development of chemotherapy resistance, inhibitors for IL-6 alone have not improved clinical outcomes. Since the IL-6/IL-6R/GP130 complex is involved in multidrug resistance, another strategy would be to focus on glycoprotein-130 (GP130) since it dimerizes with IL-6R/CD26 as a membrane-bound signaling transducer receptor and initiates subsequent signaling activation and may be a potential therapeutic target. Currently, the role of GP130 in chemoresistant bladder cancer is unknown. In the present study, we demonstrate that GP130 is over-expressed in cisplatin and gemcitabine-resistant bladder cancer cells, and that the inhibition of GP130 expression significantly reduces cell viability, survival and migration. Downstream of GP130 is PI3K/AKT/mTOR signaling, which is inactivated by SC144, a GP130 inhibitor. However, Raf/MEK/ERK signaling, which also is downstream of GP130 is activated by SC144. This activation is likely based on a mTOR/S6K1/PI3K/ERK negative feedback loop, which is presumed to counteract the inhibitory effect of SC144 on tumor aggressiveness. Blocking both GP130 and pERK resulted in synergistic inhibition of cytotoxicity, clonal survival rates and cell migration in our chemotherapy resistant bladder cancer cells. This vertical inhibition offers a novel therapeutic strategy for targeting human chemoresistant bladder cancer.


Assuntos
Carcinoma de Células de Transição/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Glicoproteínas/antagonistas & inibidores , Hidrazinas/farmacologia , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Quinoxalinas/farmacologia , Neoplasias da Bexiga Urinária/tratamento farmacológico , Butadienos/farmacologia , Butadienos/uso terapêutico , Carcinoma de Células de Transição/patologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sinergismo Farmacológico , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glicoproteínas/fisiologia , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteína Quinase 1 Ativada por Mitógeno/fisiologia , Nitrilas/farmacologia , Nitrilas/uso terapêutico , Neoplasias da Bexiga Urinária/patologia
5.
J Neurochem ; 151(2): 166-184, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31314915

RESUMO

The specific mechanisms underlying cyclin-dependent kinase 5 (Cdk5)-mediated neuropathic pain at the spinal cord level remain elusive. The aim of the present study was to explore the role of crosstalk between Cdk5/p35 and extracellular signal-regulated kinase 1/2 (ERK1/2) signalling in mediating spinal astrocyte activity via the PPARγ pathway in a rat model of chronic constriction injury (CCI). Here, we quantified pain behaviour after CCI; detected the localization of p35, Cdk5, phosphorylated ERK1/2 (pERK1/2), phosphorylated peroxisome proliferator-activated receptor γ (pPPARγ), neuronal nuclei (a neuronal marker), glial fibrillary acidic protein (GFAP, an activated astrocyte marker) and ionized calcium binding adaptor molecule 1 (a microglial marker) in the dorsal horn using immunofluorescence; measured the protein levels of Cdk5, p35, pERK1/2, pPPARγ and GFAP using western blot analysis; and gauged the enzyme activity of Cdk5/p35 kinase using a Cdk5/p35 kinase activity assay kit. Tumour necrosis factor-α, interleukin (IL)-1ß and IL-6 levels were measured using enzyme-linked immunosorbent assay (ELISA). Ligation of the right sciatic nerve induced mechanical allodynia; thermal hyperalgesia; and the time-dependent upregulation of p35, pERK1/2 and GFAP and downregulation of pPPARγ. p35 colocalized with Cdk5, pERK1/2, pPPARγ, neurons and astrocytes but not microglia. Meanwhile, intrathecal injection of the Cdk5 inhibitor roscovitine, the mitogen-activated ERK kinase (MEK) inhibitor U0126 and the PPARγ agonist pioglitazone prevented or reversed behavioural allodynia, increased pPPARγ expression, inhibited astrocyte activation and alleviated proinflammatory cytokine (tumour necrosis factor-α, IL-1ß, and IL-6) release from activated astrocytes. Furthermore, crosstalk between the Cdk5/p35 and ERK1/2 pathways was observed with CCI. Blockade of either Cdk5/p35 or ERK1/2 inhibited Cdk5 activity. These findings indicate that spinal crosstalk between the Cdk5/p35 and ERK1/2 pathways mediates astrocyte activity via the PPARγ pathway in CCI rats and that targeting this crosstalk could be an effective strategy to attenuate CCI and astrocyte-derived neuroinflammation.


Assuntos
Astrócitos/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , PPAR gama/biossíntese , Fosfotransferases/biossíntese , Neuropatia Ciática/metabolismo , Medula Espinal/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Butadienos/farmacologia , Butadienos/uso terapêutico , Constrição Patológica/tratamento farmacológico , Constrição Patológica/metabolismo , Quinase 5 Dependente de Ciclina/antagonistas & inibidores , Quinase 5 Dependente de Ciclina/biossíntese , Quinase 5 Dependente de Ciclina/genética , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Nitrilas/farmacologia , Nitrilas/uso terapêutico , PPAR gama/genética , Fosfotransferases/antagonistas & inibidores , Fosfotransferases/genética , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Ratos , Ratos Sprague-Dawley , Roscovitina/farmacologia , Roscovitina/uso terapêutico , Neuropatia Ciática/tratamento farmacológico , Medula Espinal/efeitos dos fármacos
6.
Oncogene ; 38(1): 88-102, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30082909

RESUMO

B7-H3 is a tumor-promoting glycoprotein that is expressed at low levels in most normal tissues, but is overexpressed in various human cancers which is associated with disease progression and poor patient outcome. Although numerous publications have reported the correlation between B7-H3 and cancer progression in many types of cancers, mechanistic studies on how B7-H3 regulates cancer malignancy are rare, and the mechanisms underlying the role of B7-H3 in drug resistance are almost unknown. Here we report a novel finding that upregulation of B7-H3 increases the breast cancer stem cell population and promotes cancer development. Depletion of B7-H3 in breast cancer significantly inhibits the cancer stem cells. By immunoprecipitation and mass spectrometry, we found that B7-H3 is associated with the major vault protein (MVP) and activates MEK through MVP-enhancing B-RAF and MEK interaction. B7-H3 expression increases stem cell population by binding to MVP which regulates the activation of the MAPK kinase pathway. Depletion of MVP blocks the activation of MEK induced by B7-H3 and dramatically inhibits B7-H3 induced stem cells. This study reports novel functions of B7-H3 in regulating breast cancer stem cell enrichment. The novel mechanism for B7-H3-induced stem cell propagation by regulating MVP/MEK signaling axis independent of the classic Ras pathway may have important implications in the development of strategies for overcoming cancer cell resistance to chemotherapy.


Assuntos
Antígenos B7/fisiologia , Neoplasias da Mama/metabolismo , Resistencia a Medicamentos Antineoplásicos/fisiologia , MAP Quinase Quinase Quinases/fisiologia , Proteínas de Neoplasias/fisiologia , Células-Tronco Neoplásicas/metabolismo , Partículas de Ribonucleoproteínas em Forma de Abóbada/fisiologia , Animais , Antígenos B7/antagonistas & inibidores , Antígenos B7/química , Antígenos B7/genética , Neoplasias da Mama/patologia , Butadienos/farmacologia , Butadienos/uso terapêutico , Sistemas CRISPR-Cas , Linhagem Celular Tumoral , Polaridade Celular , Ativação Enzimática , Feminino , Humanos , MAP Quinase Quinase Quinases/antagonistas & inibidores , Camundongos , Camundongos Nus , Proteína Homeobox Nanog/biossíntese , Proteína Homeobox Nanog/genética , Invasividade Neoplásica , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Nitrilas/farmacologia , Nitrilas/uso terapêutico , Domínios Proteicos , Mapeamento de Interação de Proteínas , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Proto-Oncogênicas B-raf/metabolismo , Interferência de RNA , RNA Guia de Cinetoplastídeos/genética , RNA Interferente Pequeno/farmacologia , Proteínas Recombinantes/metabolismo , Fatores de Transcrição SOXB1/biossíntese , Fatores de Transcrição SOXB1/genética , Deleção de Sequência , Esferoides Celulares , Transfecção , Regulação para Cima
7.
Int J Mol Sci ; 19(9)2018 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-30200486

RESUMO

Infantile myofibromatosis represents one of the most common proliferative fibrous tumors of infancy and childhood. More effective treatment is needed for drug-resistant patients, and targeted therapy using specific protein kinase inhibitors could be a promising strategy. To date, several studies have confirmed a connection between the p.R561C mutation in gene encoding platelet-derived growth factor receptor beta (PDGFR-beta) and the development of infantile myofibromatosis. This study aimed to analyze the phosphorylation of important kinases in the NSTS-47 cell line derived from a tumor of a boy with infantile myofibromatosis who harbored the p.R561C mutation in PDGFR-beta. The second aim of this study was to investigate the effects of selected protein kinase inhibitors on cell signaling and the proliferative activity of NSTS-47 cells. We confirmed that this tumor cell line showed very high phosphorylation levels of PDGFR-beta, extracellular signal-regulated kinases (ERK) 1/2 and several other protein kinases. We also observed that PDGFR-beta phosphorylation in tumor cells is reduced by the receptor tyrosine kinase inhibitor sunitinib. In contrast, MAPK/ERK kinases (MEK) 1/2 and ERK1/2 kinases remained constitutively phosphorylated after treatment with sunitinib and other relevant protein kinase inhibitors. Our study showed that sunitinib is a very promising agent that affects the proliferation of tumor cells with a p.R561C mutation in PDGFR-beta.


Assuntos
Mutação , Miofibromatose/congênito , Inibidores de Proteínas Quinases/administração & dosagem , Receptor beta de Fator de Crescimento Derivado de Plaquetas , Sunitinibe/administração & dosagem , Butadienos/administração & dosagem , Butadienos/uso terapêutico , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Criança , Cloridrato de Erlotinib/administração & dosagem , Cloridrato de Erlotinib/uso terapêutico , Feminino , Humanos , Lactente , Masculino , Miofibromatose/tratamento farmacológico , Miofibromatose/genética , Nitrilas/administração & dosagem , Nitrilas/uso terapêutico , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/uso terapêutico , Pirazóis/administração & dosagem , Pirazóis/uso terapêutico , Piridazinas/administração & dosagem , Piridazinas/uso terapêutico , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Sunitinibe/uso terapêutico
8.
Brain Res ; 1678: 174-179, 2018 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-29074343

RESUMO

BACKGROUND: Sevoflurane has been shown to stimulate or depress memory in adult rats; however, the cellular mechanism of this bidirectional effect has not been fully investigated. METHODS: We used an intra-hippocampal microinfusion of U0126 to suppress ERK activation. Male SD rats were randomly assigned to four groups: Sham, 0.11%SEV, 0.3%SEV and 0.3%+U0126. They received bilateral injections of U0126 or saline. Rats were anesthetized, and Inhibitory Avoidance (IA) training was performed immediately after anesthesia. The memory retention latency was observed 24 h later. In another experiment, the hippocampus was removed 45 min after IA training to assess ARC expression, the synapsin 1 protein levels and the phosphorylation level of ERK. RESULTS: Treatment with 0.11%SEV led to rapid phosphorylation of ERK, while 0.3%SEV inhibited phosphorylation; the latter change was reversed by the microinfusion of U0126 in the hippocampus. The memory latency result had similar tendencies. The local infusion of U0126 abolished the 0.3%SEV-induced memory impairment and ERK inhibition. Selective upregulations of ARC and synapsin 1 proteins were observed in the 0.3%SEV group compared with the 0.11%SEV group. CONCLUSIONS: The results indicate that different doses of sevoflurane trigger synaptic plasticity-related cytoskeleton proteins through the ERK signaling pathway. This novel modulation by inhalational agents may help to reduce their side-effects on memory function.


Assuntos
Complexo Relacionado com a AIDS/metabolismo , Anestésicos Inalatórios/toxicidade , Hipocampo/metabolismo , Deficiências da Aprendizagem/induzido quimicamente , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Transtornos da Memória/induzido quimicamente , Éteres Metílicos/toxicidade , Animais , Butadienos/uso terapêutico , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/uso terapêutico , Reação de Fuga/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Deficiências da Aprendizagem/tratamento farmacológico , Deficiências da Aprendizagem/metabolismo , Deficiências da Aprendizagem/patologia , Masculino , Transtornos da Memória/tratamento farmacológico , Transtornos da Memória/metabolismo , Transtornos da Memória/patologia , Nitrilas/uso terapêutico , Ratos , Ratos Sprague-Dawley , Sevoflurano , Sinapsinas/metabolismo
9.
J Mol Neurosci ; 62(3-4): 380-394, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28741142

RESUMO

Subarachnoid hemorrhage (SAH) is a serious clinical condition where leakage of blood into the subarachnoid space causes an acute rise in intracranial pressure and reduces cerebral blood flow, which may lead to delayed cerebral ischemia and poor outcome. In experimental SAH, we have previously shown that the outcome can be significantly improved by early inhibition of the MAPK/ERK kinase/extracellular signal-regulated kinase (MEK/ERK1/2) pathway. The aim of this study was to apply mass spectrometry to investigate the overall late effects of experimental SAH on cerebrovascular protein expression. SAH was induced in rats that were treated with the MEK1/2 inhibitor U0126 or vehicle. Neurological outcome was assessed using a battery of behavioral tests. Specific protein expression of large cerebral arteries was analyzed quantitatively with high-throughput tandem mass spectrometry. SAH resulted in a marked reduction of neurological scores, which was counteracted by U0126 treatment. Mass spectrometry analysis demonstrated regulation of 184 proteins after SAH, regulations that were in part prevented by U0126 treatment. Network analysis identified several protein networks including a strong structural network centered around 14-3-3. Additionally, protein networks with functions in mRNA metabolism and protein folding were identified. Treatment with U0126 inhibited cerebral vessel wall pERK1/2 expression and significantly improved outcome of the rats. In conclusion, we show that SAH induces a broad array of specific changes in the overall protein networks in cerebral artery smooth muscle cells and suggest that this is essential for understanding the vascular pathophysiology after SAH.


Assuntos
Artérias Cerebrais/metabolismo , Sistema de Sinalização das MAP Quinases , Proteoma/metabolismo , Hemorragia Subaracnóidea/metabolismo , Animais , Butadienos/uso terapêutico , Inibidores Enzimáticos/uso terapêutico , Masculino , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Nitrilas/uso terapêutico , Proteoma/genética , Ratos , Ratos Sprague-Dawley , Hemorragia Subaracnóidea/tratamento farmacológico , Hemorragia Subaracnóidea/patologia
10.
Brain Res ; 1648(Pt A): 90-100, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27402431

RESUMO

This study was undertaken to examine the function of extracellular signal-regulated kinase (ERK) signaling pathway on the proliferation and activation of microglia/macrophage and astrocytes after brain injury in mice. The result of Western blot showed that p-ERK was immediately activated after injury (<4h), but the duration was short (<4 days). According to immunofluorescence double staining, it was found that at 4 and 8h after injury, p-ERK was expressed in microglia/macrophages, and that more cells were co-expressed by p-ERK and IBA-1 (microglia/macrophage marker) at 8h; at days 1 and 4, p-ERK was expressed in astrocytes, and more cells were co-expressed by p-ERK and GFAP (astrocyte marker) at day 4. After injury, the mice were injected with U0126 (MAPK/ERK signaling pathway inhibitor) via the femoral vein. Compared with those injected with DMSO, the cell number co-expressed by p-ERK and IBA-1 or GFAP significantly decreased (P<0.05). The increase of microglia/macrophage and astrocyte caused by injury was remitted, and the positive cell number significantly decreased (P<0.05). Western blot showed that the expression quantity of IBA-1 and GFAP significantly decreased (P<0.05). Furthermore, the ERK signaling pathway was involved in the proliferation and activation of the two glial cells types and improved long-term neurobehavioral function after brain injury. Therefore, the exploration of the formation mechanism of glial scar after injury and further research on the therapeutic method of neural regeneration are essential.


Assuntos
Sistema de Sinalização das MAP Quinases/fisiologia , Neuroglia/metabolismo , Animais , Astrócitos/metabolismo , Western Blotting , Lesões Encefálicas/terapia , Butadienos/farmacologia , Butadienos/uso terapêutico , Proliferação de Células/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Macrófagos/metabolismo , Masculino , Camundongos , Microglia/metabolismo , Neuralgia/tratamento farmacológico , Neuroglia/fisiologia , Nitrilas/farmacologia , Nitrilas/uso terapêutico , Fosforilação/fisiologia , Transdução de Sinais
11.
Eur J Pharmacol ; 788: 280-285, 2016 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-27343376

RESUMO

Myocardial ischemia is one of the main causes of sudden cardiac death worldwide. Depending on the cell type and stimulus, ERK activity mediates different anti-proliferative events, such as apoptosis, autophagy, and senescence. The aim of this study was to determine the protective effect of 1,4-diamino-2,3-dicyano-1,4-bis[2-aminophenylthio] butadiene (U0126), an ERK kinase inhibitor, on myocardial ischemia/reperfusion (I/R) injury and the mechanisms involved. An I/R model was established in vivo in C57BL/6 mice and in vitro using mouse cardiomyocytes, respectively. To evaluate the protective effects of U0126 on I/R injury, we measured the myocardial infarct area, apoptosis, and autophagy. Our data indicated that pretreatment with U0126 significantly reduced the infarct area caused by I/R. Moreover, U0126 reduced the caspase-3 activity and the number of TUNEL-positive cardiomyocytes, which together indicate decreased apoptosis. Additionally, U0126 remarkable reduced the level of Beclin-1 and LC3 and increased p62 expression, which indicates that U0126 suppressed H/R-induced autophagy. Furthermore, the relationship between U0126 and MEK/ERK pathway activation in H/R-induced cardiomyocytes was also investigated. U0126 ameliorated H/R injury through inhibition of the MEK/ERK pathway and by suppressing in the downstream EGR-1 expression. Together, our research suggests that U0126 may protect against H/R injury by preventing H/R-induced myocardium apoptosis and autophagy via the MEK/ERK/EGR-1 pathway, and may be a potential therapeutic approach for attenuating myocardial I/R injury.


Assuntos
Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Butadienos/farmacologia , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/patologia , Nitrilas/farmacologia , Animais , Butadienos/uso terapêutico , Camundongos , Miocárdio/patologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Nitrilas/uso terapêutico
12.
Australas Phys Eng Sci Med ; 38(3): 399-412, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26108891

RESUMO

This feasibility study aims to determine if a low-cost 3D printer (BitsFromBytes 3D Touch) with ABS plastic can print custom mould structures and catheter channels defined in a brachytherapy treatment planning system (Nucletron Oncentra) for patient-specific treatment. Printer accuracy was evaluated through physical measurement, and print quality was investigated by adjusting print parameters (print speed, layer thickness, percentage infill). Catheter positioning and reproducibility were measured over repeated insertions. ABS plastic water equivalency was investigated by comparing Ir-192 HDR source dose distributions, measured with radiochromic film, in ABS plastic and in water. Structures and catheter channels were printed accurately to within 0.5 mm laterally and 1 mm in the vertical print direction. Adjusting print parameters could reduce print time, albeit with reduced print quality. 3.5 mm channel diameters allowed for easy catheter insertion. Catheter positioning was reproducible to within 0.5 mm but, because of catheter flex within the channel, was on average 1 mm offset from defined TPS positions. This offset could be accounted for by repeating the treatment planning CT scan with the printed mould positioned on the patient. Dose attenuation in ABS plastic and in water was equivalent to within the measurement limitations. While clinical uses for this particular low-cost printer and ABS plastic are limited by print size restrictions and non-certification for biocompatibility, it has been demonstrated that a low-cost 3D printer set-up can accurately create custom moulds and catheter channels potentially acceptable for clinical use.


Assuntos
Resinas Acrílicas/uso terapêutico , Braquiterapia/instrumentação , Butadienos/uso terapêutico , Plásticos/uso terapêutico , Poliestirenos/uso terapêutico , Medicina de Precisão/instrumentação , Impressão Tridimensional/instrumentação , Estudos de Viabilidade , Cabeça/anatomia & histologia , Humanos , Modelos Biológicos , Imagens de Fantasmas , Tomografia Computadorizada por Raios X
13.
Oncotarget ; 5(5): 1162-73, 2014 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-24811485

RESUMO

The pro-apoptotic protein Bnip3 is induced by hypoxia and is present in the core regions of most solid tumors. Bnip3 induces programmed necrosis by an intrinsic caspase independent mitochondrial pathway. Many tumor cells have evolved pathways to evade Bnip3-mediated death attesting to the physiological relevance of the survival threat imposed by Bnip3. We have reported that acidosis can trigger the Bnip3 death pathway in hypoxic cells therefore we hypothesized that manipulation of intracellular pH by pharmacological inhibition of the vacuolar (v)ATPase proton pump, a significant pH control pathway, may activate Bnip3 and promote death of hypoxic cells within the tumor. Here we confirm that bafilomycin A1 (BafA1), a selective vATPase inhibitor, significantly increased death of breast cancer cells in a hypoxia and Bnip3-dependent manner and significantly reduced tumor growth in MCF7 and MDA-MB-231 mouse xenografts. Combined treatment of cells with BafA1 and the ERK1/2 inhibitor U0126 further augmented cell death. Combined treatment of mice containing MDA-MB-231 xenografts with BafA1 and the ERK1/2 inhibitor sorafenib was superior to either treatment alone and supported tumor regression. BafA1 and sorafenib treatments alone reduced MDA-MB-231 cell metastasis and again the combination was significantly more effective than either treatment alone and was without apparent side effects. These results present a novel mechanism to destroy hypoxic tumor cells that may help reverse the resistance of hypoxic tumors to radiation and chemotherapy and perhaps target tumor stem cells.


Assuntos
Apoptose , Neoplasias da Mama/tratamento farmacológico , Hipóxia Celular , Inibidores Enzimáticos/farmacologia , Macrolídeos/farmacologia , Proteínas de Membrana/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , ATPases Vacuolares Próton-Translocadoras/antagonistas & inibidores , Animais , Antineoplásicos/uso terapêutico , Apoptose/genética , Neoplasias da Mama/patologia , Butadienos/farmacologia , Butadienos/uso terapêutico , Inibidores Enzimáticos/uso terapêutico , Técnicas de Silenciamento de Genes , Xenoenxertos , Humanos , Concentração de Íons de Hidrogênio , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Células MCF-7 , Macrolídeos/uso terapêutico , Proteínas de Membrana/genética , Camundongos , Metástase Neoplásica , Niacinamida/análogos & derivados , Niacinamida/uso terapêutico , Nitrilas/farmacologia , Nitrilas/uso terapêutico , Compostos de Fenilureia/uso terapêutico , Proteínas Proto-Oncogênicas/genética , Sorafenibe , Carga Tumoral/efeitos dos fármacos , ATPases Vacuolares Próton-Translocadoras/genética
14.
J Cell Physiol ; 229(11): 1660-72, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24615622

RESUMO

Cachexia is a relevant comorbid condition of chronic diseases including cancer. Inflammation, oxidative stress, autophagy, ubiquitin-proteasome system, nuclear factor (NF)-κB, and mitogen-activated protein kinases (MAPK) are involved in the pathophysiology of cancer cachexia. Currently available treatment is limited and data demonstrating effectiveness in in vivo models are lacking. Our objectives were to explore in respiratory and limb muscles of lung cancer (LC) cachectic mice whether proteasome, NF-κB, and MAPK inhibitors improve muscle mass and function loss through several molecular mechanisms. Body and muscle weights, limb muscle force, protein degradation and the ubiquitin-proteasome system, signaling pathways, oxidative stress and inflammation, autophagy, contractile and functional proteins, myostatin and myogenin, and muscle structure were evaluated in the diaphragm and gastrocnemius of LC (LP07 adenocarcinoma) bearing cachectic mice (BALB/c), with and without concomitant treatment with NF-κB (sulfasalazine), MAPK (U0126), and proteasome (bortezomib) inhibitors. Compared to control animals, in both respiratory and limb muscles of LC cachectic mice: muscle proteolysis, ubiquitinated proteins, autophagy, myostatin, protein oxidation, FoxO-1, NF-κB and MAPK signaling pathways, and muscle abnormalities were increased, while myosin, creatine kinase, myogenin, and slow- and fast-twitch muscle fiber size were decreased. Pharmacological inhibition of NF-κB and MAPK, but not the proteasome system, induced in cancer cachectic animals, a substantial restoration of muscle mass and force through a decrease in muscle protein oxidation and catabolism, myostatin, and autophagy, together with a greater content of myogenin, and contractile and functional proteins. Attenuation of MAPK and NF-κB signaling pathway effects on muscles is beneficial in cancer-induced cachexia.


Assuntos
Autofagia , Caquexia/tratamento farmacológico , Caquexia/etiologia , Neoplasias Pulmonares/complicações , Neoplasias Pulmonares/tratamento farmacológico , Debilidade Muscular/tratamento farmacológico , Músculo Esquelético/patologia , Proteólise , Animais , Autofagia/efeitos dos fármacos , Ácidos Borônicos/farmacologia , Ácidos Borônicos/uso terapêutico , Bortezomib , Butadienos/farmacologia , Butadienos/uso terapêutico , Linhagem Celular Tumoral , Inflamação/complicações , Inflamação/patologia , Inflamação/fisiopatologia , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/fisiopatologia , Camundongos , Camundongos Endogâmicos BALB C , Contração Muscular/efeitos dos fármacos , Proteínas Musculares/metabolismo , Debilidade Muscular/etiologia , Debilidade Muscular/patologia , Debilidade Muscular/fisiopatologia , Músculo Esquelético/anormalidades , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/fisiopatologia , NF-kappa B/metabolismo , Nitrilas/farmacologia , Nitrilas/uso terapêutico , Oxirredução , Proteólise/efeitos dos fármacos , Pirazinas/farmacologia , Pirazinas/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Sulfassalazina/farmacologia , Sulfassalazina/uso terapêutico , Tirosina/metabolismo , Aumento de Peso/efeitos dos fármacos
15.
PLoS One ; 9(3): e92417, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24642693

RESUMO

BACKGROUND: Global cerebral ischemia following cardiac arrest is associated with increased cerebral vasoconstriction and decreased cerebral blood flow, contributing to delayed neuronal cell death and neurological detriments in affected patients. We hypothesize that upregulation of contractile ETB and 5-HT1B receptors, previously demonstrated in cerebral arteries after experimental global ischemia, are a key mechanism behind insufficient perfusion of the post-ischemic brain, proposing blockade of this receptor upregulation as a novel target for prevention of cerebral hypoperfusion and delayed neuronal cell death after global cerebral ischemia. The aim was to characterize the time-course of receptor upregulation and associated neuronal damage after global ischemia and investigate whether treatment with the MEK1/2 inhibitor U0126 can prevent cerebrovascular receptor upregulation and thereby improve functional outcome after global cerebral ischemia. Incomplete global cerebral ischemia was induced in Wistar rats and the time-course of enhanced contractile responses and the effect of U0126 in cerebral arteries were studied by wire myography and the neuronal cell death by TUNEL. The expression of ETB and 5-HT1B receptors was determined by immunofluorescence. RESULTS: Enhanced vasoconstriction peaked in fore- and midbrain arteries 3 days after ischemia. Neuronal cell death appeared initially in the hippocampus 3 days after ischemia and gradually increased until 7 days post-ischemia. Treatment with U0126 normalised cerebrovascular ETB and 5-HT1B receptor expression and contractile function, reduced hippocampal cell death and improved survival rate compared to vehicle treated animals. CONCLUSIONS: Excessive cerebrovascular expression of contractile ETB and 5-HT1B receptors is a delayed response to global cerebral ischemia peaking 3 days after the insult, which likely contributes to the development of delayed neuronal damage. The enhanced cerebrovascular contractility can be prevented by treatment with the MEK1/2 inhibitor U0126, diminishes neuronal damage and improves survival rate, suggesting MEK1/2 inhibition as a novel strategy for early treatment of neurological consequences following global cerebral ischemia.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Butadienos/farmacologia , Hipóxia Encefálica/prevenção & controle , Nitrilas/farmacologia , Receptor de Endotelina B/metabolismo , Receptor 5-HT1B de Serotonina/metabolismo , Animais , Isquemia Encefálica/patologia , Butadienos/uso terapêutico , Circulação Cerebrovascular/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , MAP Quinase Quinase 1/antagonistas & inibidores , MAP Quinase Quinase 1/metabolismo , MAP Quinase Quinase 2/antagonistas & inibidores , MAP Quinase Quinase 2/metabolismo , Nitrilas/uso terapêutico , Ratos , Ratos Wistar , Receptor de Endotelina B/genética , Receptor 5-HT1B de Serotonina/genética , Resultado do Tratamento , Regulação para Cima/efeitos dos fármacos , Vasoconstrição/efeitos dos fármacos
16.
Cell Physiol Biochem ; 32(5): 1528-40, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24335178

RESUMO

BACKGROUND: The p53:miR-34a:E2F positive feed-forward loop and the p53:miR-605:Mdm2 positive feed-back loop have been identified to be crucial oncogenesis/tumor suppressor-regulating signaling pathways. In this study, we sought to examine the hypothesis that neoadjuvant chemotherapy (NAC) is a better approach with improved prognosis and outcomes after laparoscopical radical hysterectomy (LRH) on patients with cervical cancer and to elucidate the potential roles of the p53:miR-34a:E2F1 and the p53:miR-605:Mdm2 signaling pathways in this therapy. METHODS: Twenty-one patients with stage IIB cervical cancer were recruited to this study and they were randomly divided into two groups: LRH (n=10) and NAC+LRH (n=11) groups. The NAC+LRH group consisted of 4 cycles of cisplatin, paclitaxel and carboplatin. Complication rates and NAC outcomes (tumor size changes, 2-year disease-free survival rate, and 2-year overall survival rate) were compared between the two groups. Expression of p53, Mdm2, E2F1, miR-34a, and miR-605 at mRNA and protein levels from the tumor tissues was analyzed. RESULTS: We observed that the diameter of tumors following chemotherapy was substantially smaller in the NAC+LRH patients than in LRH patients. No recurrence or metastasis after surgery was observed in the NAC+LRH patients, whereas 2 out of 10 LRH patients had recurrences and 1 had metastasis. The 2-year disease-free and overall survival rates were apparently higher in the NAC+LRH group than in the LRH group. Furthermore, molecular biology analyses revealed that the protein and mRNA levels of p53 were both markedly increased in patients who received NAC than those who did not, and oppositely, the levels of E2F1 and Mdm2 were significantly lower in the NAC+LRH patients than in the LRH patients. The levels of miR-34a and miR-605 were considerably higher with NAC relative to without NAC. Among the three anti-cancer drugs included in NAC, cisplatin was found to be the main component that caused increases in p53 protein levels, miR-34a and miR-605 miRNA levels, and decreases in Mdm2 and E2F1 protein levels. Furthermore, ERK1/2 inhibitor U0126 or TAB1 siRNA mitigated these changes induced by cisplatin. CONCLUSION: These findings not only indicate NAC as a rational approach for better treatment of cervical cancer with improved therapeutic outcomes, due partly to the ability of cisplatin to promote the p53:miR-34a:E2F1 positive feed-forward loop and the p53:miR-605:Mdm2 positive feedback loop.


Assuntos
Neoplasias do Colo do Útero/tratamento farmacológico , Neoplasias do Colo do Útero/cirurgia , Proteínas Adaptadoras de Transdução de Sinal/genética , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Butadienos/uso terapêutico , Cisplatino/uso terapêutico , Intervalo Livre de Doença , Fator de Transcrição E2F1/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Genes p53 , Humanos , Histerectomia , MicroRNAs/genética , Pessoa de Meia-Idade , Terapia Neoadjuvante , Nitrilas/uso terapêutico , Prognóstico , Proteínas Proto-Oncogênicas c-mdm2/genética , Neoplasias do Colo do Útero/genética , Neoplasias do Colo do Útero/mortalidade , Neoplasias do Colo do Útero/patologia
17.
J Int Med Res ; 41(2): 300-6, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23569008

RESUMO

OBJECTIVE: To test the hypothesis that chemoresistance in pancreatic cancer is mediated via extracellular signal-regulated protein kinase (ERK) 1/2 overactivity. METHODS: The human pancreatic cancer cell lines BxPC3, PANC-1 and a stably gemcitabine-resistant subline, PANC1(GemRes), were treated with combinations of gemcitabine and the ERK1/2 inhibitor, U0126. Phosphorylated (p)ERK1/2 was examined by Western blotting; cell proliferation and apoptosis were quantified. A nude mouse xenograft model was established with each cell line, and the therapeutic efficacy of gemcitabine and U0126 alone or in combination was examined. RESULTS: Gemcitabine treatment visibly increased pERK1/2 levels in BxPC-3 and PANC-1 cells. PANC-1(GemRes) constitutively produced high levels of pERK1/2. U0126 treatment reversed the gemcitabine-associated increase in cell proliferation and reduction in apoptosis, in all three cell lines. Combination treatment with U0126 and gemcitabine inhibited tumour growth and promoted apoptosis in xenograft tumours derived from all three cell lines. CONCLUSIONS: ERK1/2 activity may protect pancreatic cancer cells from chemotherapy-induced apoptosis. The combined use of an ERK1/2 inhibitor (such as U0126) together with gemcitabine may result in synergistic therapeutic effects at tolerable gemcitabine doses.


Assuntos
Desoxicitidina/análogos & derivados , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/enzimologia , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/enzimologia , Adenocarcinoma/patologia , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Butadienos/farmacologia , Butadienos/uso terapêutico , Linhagem Celular Tumoral , Desoxicitidina/farmacologia , Desoxicitidina/uso terapêutico , Humanos , Concentração Inibidora 50 , Camundongos , Nitrilas/farmacologia , Nitrilas/uso terapêutico , Neoplasias Pancreáticas/patologia , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Carga Tumoral/efeitos dos fármacos , Gencitabina
18.
PLoS One ; 8(3): e59879, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23533654

RESUMO

Phosphoinositide 3-kinase (PI3K)/AKT/mammalian target of rapamycin inhibitor (mTOR) pathway is often constitutively activated in human tumor cells and thus has been considered as a promising drug target. To ascertain a therapeutical approach of nasopharyngeal carcinoma (NPC), we hypothesized NVP-BEZ235, a novel and potent imidazo[4,5-c] quinolone derivative, that dually inhibits both PI3K and mTOR kinases activities, had antitumor activity in NPC. Expectedly, we found that NVP-BEZ235 selectively inhibited proliferation of NPC cells rather than normal nasopharyngeal cells using MTT assay. In NPC cell lines, with the extended exposure, NVP-BEZ235 selectively inhibited proliferation of NPC cells harboring PIK3CA mutation, compared to cells with wild-type PIK3CA. Furthermore, exposure of NPC cells to NVP-BEZ235 resulted in G1 growth arrest by Propidium iodide uptake assay, reduction of cyclin D1and CDK4, and increased levels of P27 and P21 by Western blotting, but negligible apoptosis. Moreover, we found that cisplatin (CDDP) activated PI3K/AKT and mTORC1 pathways and NVP-BEZ235 alleviated the activation by CDDP through dually targeting PI3K and mTOR kinases. Also, NVP-BEZ235 combining with CDDP synergistically inhibited proliferation and induced apoptosis in NPC cells. In CNE2 and HONE1 nude mice xenograft models, orally NVP-BEZ235 efficiently attenuated tumor growth with no obvious toxicity. In combination with NVP-BEZ235 and CDDP, there was dramatic synergy in shrinking tumor volumes and inducing apoptosis through increasing Noxa, Bax and decreasing Mcl-1, Bcl-2. Based on the above results, NVP-BEZ235, which has entered phase I/II clinical trials in patients with advanced solid tumors, has a potential as a monotherapy or in combination with CDDP for NPC treatment.


Assuntos
Cisplatino/uso terapêutico , Imidazóis/uso terapêutico , Neoplasias Nasofaríngeas/tratamento farmacológico , Fosfatidilinositol 3-Quinases/metabolismo , Quinolinas/uso terapêutico , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Butadienos/farmacologia , Butadienos/uso terapêutico , Carcinoma , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Cisplatino/farmacologia , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Feminino , Humanos , Imidazóis/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/enzimologia , Neoplasias Nasofaríngeas/metabolismo , Nitrilas/farmacologia , Nitrilas/uso terapêutico , Inibidores de Fosfoinositídeo-3 Quinase , Quinolinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/antagonistas & inibidores , Ensaios Antitumorais Modelo de Xenoenxerto
19.
J Neuroinflammation ; 9: 274, 2012 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-23259581

RESUMO

BACKGROUND: Subarachnoid hemorrhage (SAH) is associated with high morbidity and mortality. It is suggested that the associated inflammation is mediated through activation of the mitogen-activated protein kinase (MAPK) pathway which plays a crucial role in the pathogenesis of delayed cerebral ischemia after SAH. The aim of this study was first to investigate the timecourse of altered expression of proinflammatory cytokines and matrix metalloproteinase in the cerebral arteries walls following SAH. Secondly, we investigated whether administration of a specific mitogen-activated protein kinase kinase (MEK)1/2 inhibitor, U0126, given at 6 h after SAH prevents activation of the MEK/extracellular signal-regulated kinase 1/2 pathway and the upregulation of cerebrovascular inflammatory mediators and improves neurological function. METHODS: SAH was induced in rats by injection of 250 µl of autologous blood into basal cisterns. U0126 was given intracisternally using two treatment regimens: (A) treatments at 6, 12, 24 and 36 h after SAH and experiments terminated at 48 h after SAH, or (B) treatments at 6, 12, and 24 h after SAH and terminated at 72 h after SAH. Cerebral arteries were harvested and interleukin (IL)-6, IL-1ß, tumor necrosis factor α (TNF)α, matrix metalloproteinase (MMP)-9 and phosphorylated ERK1/2 (pERK1/2) levels investigated by immunohistochemistry. Early activation of pERK1/2 was measured by western blot. Functional neurological outcome after SAH was also analyzed. RESULTS: Expression levels of IL-1ß, IL-6, MMP-9 and pERK1/2 proteins were elevated over time with an early increase at around 6 h and a late peak at 48 to 72 h post-SAH in cerebral arteries. Enhanced expression of TNFα in cerebral arteries started at 24 h and increased until 96 h. In addition, SAH induced sensorimotor and spontaneous behavior deficits in the animals. Treatment with U0126 starting at 6 h after SAH prevented activation of MEK-ERK1/2 signaling. Further, U0126 significantly decreased the upregulation of inflammation proteins at 48 and 72 h following SAH and improved neurological function. We found no differences between treatment regimens A and B. CONCLUSIONS: These results show that SAH induces early activation of the MEK-ERK1/2 pathway in cerebral artery walls, which is associated with upregulation of proinflammatory cytokines and MMP-9. Inhibition of the MEK-ERK1/2 pathway by U0126 starting at 6 h post-SAH prevented upregulation of cytokines and MMP-9 in cerebral vessels, and improved neurological outcome.


Assuntos
Artérias Cerebrais/enzimologia , Citocinas/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Transdução de Sinais/fisiologia , Hemorragia Subaracnóidea/patologia , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/enzimologia , Transfusão de Sangue Autóloga/efeitos adversos , Butadienos/uso terapêutico , Artérias Cerebrais/patologia , Citocinas/genética , Modelos Animais de Doenças , Inibidores Enzimáticos/uso terapêutico , Comportamento Exploratório/efeitos dos fármacos , Proteína Glial Fibrilar Ácida/metabolismo , Masculino , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Atividade Motora/efeitos dos fármacos , Doenças do Sistema Nervoso/tratamento farmacológico , Doenças do Sistema Nervoso/etiologia , Nitrilas/uso terapêutico , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Estatísticas não Paramétricas , Hemorragia Subaracnóidea/complicações , Hemorragia Subaracnóidea/tratamento farmacológico , Hemorragia Subaracnóidea/metabolismo , Fatores de Tempo , Fator de Necrose Tumoral alfa/metabolismo
20.
FEBS Lett ; 586(24): 4296-302, 2012 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-23137704

RESUMO

In this study, we investigated the role of PTEN (phosphatase and tensin homolog deleted on chromosome 10) in a platelet-activating factor (PAF)-induced experimental pulmonary tumor metastasis model. An adenovirus carrying PTEN cDNA (Ad-PTEN) reversed PAF-induced increase in phosphorylation of AKT as well as pulmonary metastasis of B16F10. PAF-induced pulmonary metastasis was inhibited by MAPK inhibitors, but not by PI3K inhibitor. Ad-PTEN abrogated PAF-induced phosphorylation of MAPKs. These data indicate PTEN/MAPK pathways play a key role in PAF-induced tumor metastasis.


Assuntos
Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/secundário , Sistema de Sinalização das MAP Quinases , Melanoma Experimental/secundário , PTEN Fosfo-Hidrolase/metabolismo , Fator de Ativação de Plaquetas/metabolismo , Androstadienos/uso terapêutico , Animais , Antracenos/uso terapêutico , Butadienos/uso terapêutico , Linhagem Celular , Humanos , Imidazóis/uso terapêutico , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Nitrilas/uso terapêutico , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Fator de Ativação de Plaquetas/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Piridinas/uso terapêutico , Wortmanina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA