Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
J Pharmacol Sci ; 153(4): 197-207, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37973217

RESUMO

Osteoclasts are multinucleated, specializes bone-resorbing cells that are derived from the monocyte/macrophage lineage. Excessive resorbing activities of osteoclasts are involved in destructive bone diseases. The detailed mechanism of acidification at the bone adhesion surface during the bone resorption process of osteoclasts remains to be defined. During glycolysis, pyruvate proceeds to the tricarboxylic cycle under aerobic conditions and pyruvate is converted to lactate via lactate dehydrogenase A (LDHA) under anaerobic conditions. However, tumor cells produce ATP during aerobic glycolysis and large amounts of pyruvate are converted to lactate and H+ by LDHA. Lactate and H+ are excreted outside the cell, whereby they are involved in invasion of tumor cells due to the pH drop around the cell. In this study, we focused on aerobic glycolysis and investigated the production of lactate by LDHA in osteoclasts. Expression of LDHA and monocarboxylate transporter 4 (MCT4) was upregulated during osteoclast differentiation. Intracellular and extracellular lactate levels increased with upregulation of LDHA and MCT4, respectively. FX11 (an LDHA inhibitor) inhibited osteoclast differentiation and suppressed the bone-resorbing activity of osteoclasts. We propose that inhibition of LDHA may represent a novel therapeutic strategy for controlling excessive bone resorption in osteoporosis and rheumatoid arthritis.


Assuntos
Reabsorção Óssea , Osteogênese , Humanos , Lactato Desidrogenase 5/metabolismo , Osteoclastos/fisiologia , Reabsorção Óssea/prevenção & controle , Reabsorção Óssea/metabolismo , Lactatos/metabolismo , Glicólise , Piruvatos/metabolismo , L-Lactato Desidrogenase/metabolismo
2.
Biochem Biophys Res Commun ; 530(2): 455-461, 2020 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-32553625

RESUMO

Osteoclasts are multinucleated bone-resorbing cells derived from monocyte/macrophage progenitor cells. Excessive formation and resorbing activities of osteoclasts are involved in the bone-destructive pathologies of rheumatoid arthritis and osteoporosis. Recently, it has been found that nuclear factor erythroid 2-related factor 2 (Nrf2), a transcription factor for anti-oxidative stress genes, functions in osteoclastogenesis. Dimethyl fumarate (DMF) is a potent activator of Nrf2 and has been shown to inhibit osteoclastogenesis. Here, we investigated the mechanisms of this inhibition by examining the activation of several signalling pathways during the differentiation of bone marrow-derived macrophages into osteoclasts. DMF inhibited the differentiation of osteoclasts in a dose-dependent manner and suppressed the bone-resorbing activity of osteoclasts. DMF treatment decreased the expression of nuclear factor of activated T-cells cytoplasmic-1, and significantly decreased phosphorylation of extracellular signal-regulated kinase and p38 mitogen-activated protein kinase in osteoclasts. We also found that DMF inhibited the extracellular release of high mobility group box 1, associated with an up-regulation of heme oxygenase-1, likely mediated through Nrf2 activation. Our results indicate that DMF inhibits osteoclast differentiation through multiple pathways.


Assuntos
Fumarato de Dimetilo/farmacologia , Proteína HMGB1/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Fatores de Transcrição NFATC/metabolismo , Osteogênese/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Animais , Células Cultivadas , Regulação para Baixo/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteína HMGB1/análise , Masculino , Camundongos Endogâmicos C57BL , Fatores de Transcrição NFATC/análise , Proteínas Quinases p38 Ativadas por Mitógeno/análise , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
3.
Biochem Biophys Res Commun ; 473(2): 620-4, 2016 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-27038546

RESUMO

CD147, a membrane glycoprotein of the immunoglobulin superfamily, is highly upregulated during dynamic cellular events including tissue remodelling. Elevated CD147 expression is present in the joint of rheumatoid arthritis patients. However, the role of CD147 in bone destruction remains unclear. To determine whether CD147 is involved in osteoclastogenesis, we studied its expression in mouse osteoclasts and its role in osteoclast differentiation and function. CD147 expression was markedly upregulated during osteoclast differentiation. To investigate the role of CD147 in receptor activator of nuclear factor-kappa B ligand (RANKL)-induced osteoclastogenesis and bone resorption activity, osteoclast precursor cells were transfected with CD147 siRNA. Decreased CD147 expression inhibited osteoclast formation and bone resorption, inhibited RANKL-induced nuclear translocation of the nuclear factor of activated T cells (NFAT) c1 and decreased the expression of the d2 isoform of vacuolar ATPase Vo domain and cathepsin K. Therefore, CD147 plays a critical role in the differentiation and function of osteoclasts by upregulating NFATc1 through the autoamplification of its expression in osteoclastogenesis.


Assuntos
Basigina/metabolismo , Fatores de Transcrição NFATC/metabolismo , Osteoclastos/metabolismo , Transdução de Sinais , Animais , Basigina/genética , Reabsorção Óssea/genética , Reabsorção Óssea/metabolismo , Células Cultivadas , Masculino , Camundongos Endogâmicos C57BL , Osteoclastos/citologia , Transporte Proteico , Ligante RANK/metabolismo , Regulação para Cima
4.
Drug Alcohol Depend ; 257: 111139, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38430788

RESUMO

BACKGROUND: The recreational drug ±3,4-methylenedioxymethamphetamine (MDMA; also known as "ecstasy") has unusual subjective prosocial and empathogenic effects, and has exhibited potential as an adjunct to psychotherapy in recent years. However, there has been some concern regarding possible neuropsychiatric symptoms, such as cognitive impairment and dependence, emerging after abstinence. Therefore, this study aimed to evaluate the mechanism underlying cognitive impairment during MDMA withdrawal. To achieve this, we focused on the arachidonic acid cascade, which is related to addiction to some abusive drugs. METHODS: A novel object recognition task was used to investigate cognitive function in mice. Furthermore, we quantified prostaglandin E2 during MDMA withdrawal. RESULTS: The recognition index significantly decreased during withdrawal after repeated administration of MDMA (10mg/kg, i.p., once daily for 7 days), but not following co-administration of diclofenac (10mg/kg, i.p.), a cyclooxygenase inhibitor. On day 1, following repeated MDMA treatment, prostaglandin E2 content significantly increased in the hippocampus but not in the prefrontal cortex and striatum. CONCLUSIONS: Our findings indicate that activation of the arachidonic acid cascade at least in the hippocampus is likely involved in the development of recognition memory impairment during MDMA withdrawal. Therefore, co-use of cyclooxygenase inhibitors with MDMA may reduce concerns regarding MDMA-induced impairment of recognition memory.


Assuntos
N-Metil-3,4-Metilenodioxianfetamina , Camundongos , Animais , N-Metil-3,4-Metilenodioxianfetamina/efeitos adversos , Ácido Araquidônico/farmacologia , Cognição , Hipocampo , Prostaglandinas/farmacologia
5.
Biol Pharm Bull ; 36(3): 492-5, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23449334

RESUMO

The blood-brain barrier (BBB) is formed by brain endothelial cells. Many immortalized brain endothelial cell lines have been established; these have been used as in vitro BBB models. The aim of the present study was to assess the paracellular barrier properties of the immortalized mouse brain endothelial cell lines bEND.3, bEND.5 cells, and mouse brain endothelial cell 4 (MBEC4), and those of the primary mouse brain endothelial cells pMBECs. bEND.3 cells showed low permeability to sodium fluorescein and obvious staining of tight junction proteins (claudin-5, occludin and ZO-1) similar to pMBECs; these barrier properties of MBEC4 and bEND.5 cells were low. In addition, bEND.3 cells expressed the highest level of claudin-5 among all cells. These results suggest that bEND.3 cells are a convenient and useful model for evaluating BBB function, especially the paracellular barrier.


Assuntos
Barreira Hematoencefálica , Encéfalo/irrigação sanguínea , Células Endoteliais/metabolismo , Proteínas de Junções Íntimas/análise , Animais , Linhagem Celular , Sobrevivência Celular , Claudina-5/análise , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos ICR , Ocludina/análise , Proteína da Zônula de Oclusão-1/análise
6.
J Neuroinflammation ; 8: 106, 2011 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-21867555

RESUMO

BACKGROUND: Increased matrix metalloproteinase (MMP)-9 in the plasma and brain is associated with blood-brain barrier (BBB) disruption through proteolytic activity in neuroinflammatory diseases. MMP-9 is present in the brain microvasculature and its vicinity, where brain microvascular endothelial cells (BMECs), pericytes and astrocytes constitute the BBB. Little is known about the cellular source and role of MMP-9 at the BBB. Here, we examined the ability of pericytes to release MMP-9 and migrate in response to inflammatory mediators in comparison with BMECs and astrocytes, using primary cultures isolated from rat brains. METHODS: The culture supernatants were collected from primary cultures of rat brain endothelial cells, pericytes, or astrocytes. MMP-9 activities and levels in the supernatants were measured by gelatin zymography and western blot, respectively. The involvement of signaling molecules including mitogen-activated protein kinases (MAPKs) and phosphoinositide-3-kinase (PI3K)/Akt in the mediation of tumor necrosis factor (TNF)-α-induced MMP-9 release was examined using specific inhibitors. The functional activity of MMP-9 was evaluated by a cell migration assay. RESULTS: Zymographic and western blot analyses demonstrated that TNF-α stimulated pericytes to release MMP-9, and this release was much higher than from BMECs or astrocytes. Other inflammatory mediators [interleukin (IL)-1ß, interferon-γ, IL-6 and lipopolysaccharide] failed to induce MMP-9 release from pericytes. TNF-α-induced MMP-9 release from pericytes was found to be mediated by MAPKs and PI3K. Scratch wound healing assay showed that in contrast to BMECs and astrocytes the extent of pericyte migration was significantly increased by TNF-α. This pericyte migration was inhibited by anti-MMP-9 antibody. CONCLUSION: These findings suggest that pericytes are most sensitive to TNF-α in terms of MMP-9 release, and are the major source of MMP-9 at the BBB. This pericyte-derived MMP-9 initiated cellular migration of pericytes, which might be involved in pericyte loss in the damaged BBB.


Assuntos
Barreira Hematoencefálica/citologia , Barreira Hematoencefálica/metabolismo , Movimento Celular/fisiologia , Metaloproteinase 9 da Matriz/metabolismo , Pericitos/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Células Cultivadas , Meios de Cultura/química , Inibidores Enzimáticos/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Pericitos/citologia , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Ratos , Ratos Wistar , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo
7.
Microvasc Res ; 81(1): 103-7, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21036181

RESUMO

The blood-brain barrier (BBB) is the interface that separates the central nervous system (CNS) from the peripheral circulation. An increase in blood-borne substances including cytokines in plasma and brain affects BBB function, and this is associated with the development of pathogenesis of a number of diseases. Plasminogen activator inhibitor (PAI)-1 regulates the plasminogen activator/plasmin system as a serpin in the periphery and the CNS. We investigated whether PAI-1 alters BBB function using in vitro models of the BBB consisting of rat primary brain endothelial cells (RBECs) alone and co-cultured with pericytes. We found that PAI-1 increased the tightness of the brain endothelial barrier in a time- and dose-dependent manner, as shown by an increase in the transendothelial electrical resistance (TEER) and a decrease in the permeability to sodium fluorescein (Na-F). RBECs responded equally to PAI-1 in the blood-facing and brain-facing sides of the brain, leading to a decrease in Na-F permeability. In addition, RBECs constitutively released PAI-1 into the blood-facing (luminal) and brain-facing (abluminal) sides. This release was polarized in favor of the luminal side and facilitated by serum. The neutralization of PAI-1 by an antibody to PAI-1 in RBEC/pericyte co-culture more robustly reduced TEER of RBECs than in RBEC monolayers. These findings suggest that PAI-1 derived from the neurovascular unit and peripheral vascular system participates as a positive regulator of the BBB in facilitating the barrier function of the endothelial tight junctions.


Assuntos
Comunicação Autócrina/fisiologia , Barreira Hematoencefálica/fisiologia , Comunicação Parácrina/fisiologia , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Animais , Anticorpos/imunologia , Anticorpos/farmacologia , Barreira Hematoencefálica/efeitos dos fármacos , Encéfalo/citologia , Permeabilidade Capilar/efeitos dos fármacos , Permeabilidade Capilar/fisiologia , Células Cultivadas , Técnicas de Cocultura , Meios de Cultura Livres de Soro/metabolismo , Meios de Cultura Livres de Soro/farmacologia , Impedância Elétrica , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Fluoresceína/metabolismo , Pericitos/citologia , Pericitos/efeitos dos fármacos , Pericitos/metabolismo , Inibidor 1 de Ativador de Plasminogênio/genética , Inibidor 1 de Ativador de Plasminogênio/imunologia , Inibidor 1 de Ativador de Plasminogênio/farmacologia , Ratos , Ratos Wistar , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacologia , Soro/fisiologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia
8.
Cell Mol Neurobiol ; 31(7): 985-9, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21681559

RESUMO

Olanzapine is known to be advantageous with respect to outcome and drug compliance in patients with schizophrenia. However, olanzapine has adverse effects, including a higher incidence of weight gain and metabolic disturbances, when compared with those of other antipsychotic agents. The mechanisms underlying these adverse events remain obscure. Female rats were orally administered olanzapine (2 mg/kg) or vehicle once a day for 2 weeks to ascertain if hypothalamic AMP-activated protein kinase (AMPK) mediates olanzapine-induced weight gain and hyperphagia. Body weight and food intake in each rat were evaluated every day and every two days, respectively. After the termination of drug treatment, we measured the protein levels of AMPK and phosphorylated AMPK in the hypothalamus using western blot analyses. Olanzapine significantly increased body weight and food intake. The phosphorylation levels of AMPK were significantly elevated by olanzapine. These results suggest that activation of hypothalamic AMPK may mediate hyperphagia and weight gain induced by chronic treatment with olanzapine.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Antipsicóticos/farmacologia , Benzodiazepinas/farmacologia , Hiperfagia/induzido quimicamente , Hipotálamo/efeitos dos fármacos , Hipotálamo/enzimologia , Aumento de Peso/efeitos dos fármacos , Animais , Peso Corporal/efeitos dos fármacos , Ingestão de Alimentos/efeitos dos fármacos , Feminino , Hiperfagia/metabolismo , Olanzapina , Ratos , Ratos Sprague-Dawley
9.
Cell Mol Neurobiol ; 30(2): 247-53, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19728078

RESUMO

The blood-brain barrier (BBB) is formed by brain capillary endothelial cells, astrocytes, pericytes, microglia, and neurons. BBB disruption under pathological conditions such as neurodegenerative disease and inflammation is observed in parallel with microglial activation. To test whether activation of microglia is linked to BBB dysfunction, we evaluated the effect of lipopolysaccharide (LPS) on BBB functions in an in vitro co-culture system with rat brain microvascular endothelial cells (RBEC) and microglia. When LPS was added for 6 h to the abluminal side of RBEC/microglia co-culture at a concentration showing no effects on the RBEC monolayer, transendothelial electrical resistance was decreased and permeability to sodium-fluorescein was increased in RBEC. Immunofluorescence staining for tight junction proteins demonstrated that zonula occludens-1-, claudin-5-, and occludin-like immunoreactivities at the intercellular borders of RBEC were fragmented in the presence of LPS-activated microglia. These functional changes induced by LPS-activated microglia were blocked by the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase inhibitor, diphenyleneiodonium chloride. The present findings suggest that LPS activates microglia to induce dysfunction of the BBB by producing reactive oxygen species through NADPH oxidase.


Assuntos
Barreira Hematoencefálica , Técnicas de Cocultura , Células Endoteliais/fisiologia , Lipopolissacarídeos/farmacologia , Microglia/efeitos dos fármacos , Microglia/fisiologia , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/fisiopatologia , Permeabilidade da Membrana Celular , Células Endoteliais/citologia , Microglia/citologia , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo
10.
J Pharmacol Sci ; 112(2): 251-4, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20118615

RESUMO

The present study was designed to elucidate the involvement of tumor necrosis factor-alpha (TNF-alpha) release from activated microglia in the induction of blood-brain barrier (BBB) dysfunction in an in vitro co-culture system with mouse brain capillary endothelial cells (MBEC4) and microglia. Lipopolysaccharide (LPS)-activated microglia increased the permeability of MBEC4 cells to sodium-fluorescein, and this hyper-permeability was blocked by a neutralizing antibody against TNF-alpha. LPS stimulated microglia to facilitate TNF-alpha release. These findings suggested that TNF-alpha released from activated microglia is attributable to BBB dysfunction.


Assuntos
Barreira Hematoencefálica/fisiopatologia , Microglia/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Barreira Hematoencefálica/metabolismo , Encéfalo/irrigação sanguínea , Encéfalo/metabolismo , Capilares/metabolismo , Permeabilidade da Membrana Celular , Técnicas de Cocultura , Endotélio Vascular/metabolismo , Fluoresceína/farmacocinética , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos BALB C
11.
Biomed Res ; 41(6): 269-277, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33268671

RESUMO

Osteoporosis disturbs the balance of bone metabolism, and excessive bone resorption causes a decrease in bone density, thus increasing the risk of fracture. (-)-Epigallocatechin-3-gallate (EGCG) is the most abundant catechin contained in green tea. EGCG has a variety of pharmacological activities. Recently, it was reported that EGCG inhibits osteoclast differentiation, but the details of the mechanism underlying the EGCG-mediated suppression of osteoclastogenesis are unknown. In this study, we investigated the effects of EGCG on several signaling pathways in osteoclastogenesis. EGCG suppressed the expression of the nuclear factor of activated T cells cytoplasmic-1 (NFATc1), the master regulator of osteoclastogenesis. EGCG decreased the expression of cathepsin K, c-Src, and ATP6V0d2 and suppressed bone resorption. We also found that EGCG upregulated heme oxygenase-1 (HO-1) and suppressed the extracellular release of high-mobility group box 1 (HMGB1). In addition, EGCG decreased the expression of the receptor for advanced glycation end products (RAGE), which is the receptor of HMGB1, in osteoclastogenesis. In summary, our study showed that EGCG could inhibit osteoclast differentiation through the downregulation of NFATc1 and the suppression of the HO-1-HMGB1-RAGE pathway. EGCG might have the potential to be a lead compound that suppresses bone resorption in the treatment of osteoporosis.


Assuntos
Conservadores da Densidade Óssea/farmacologia , Reabsorção Óssea/prevenção & controle , Catequina/análogos & derivados , Regulação da Expressão Gênica/efeitos dos fármacos , Osteoclastos/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Osteoporose/tratamento farmacológico , Animais , Densidade Óssea/efeitos dos fármacos , Reabsorção Óssea/genética , Reabsorção Óssea/metabolismo , Reabsorção Óssea/patologia , Proteína Tirosina Quinase CSK/genética , Proteína Tirosina Quinase CSK/metabolismo , Catequina/farmacologia , Catepsina K/genética , Catepsina K/metabolismo , Diferenciação Celular/efeitos dos fármacos , Fêmur/efeitos dos fármacos , Fêmur/metabolismo , Fêmur/patologia , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Heme Oxigenase-1/genética , Heme Oxigenase-1/metabolismo , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Transcrição NFATC/genética , Fatores de Transcrição NFATC/metabolismo , Osteoclastos/metabolismo , Osteoclastos/patologia , Osteogênese/genética , Osteoporose/genética , Osteoporose/metabolismo , Osteoporose/patologia , Cultura Primária de Células , Ligante RANK/antagonistas & inibidores , Ligante RANK/farmacologia , Receptor para Produtos Finais de Glicação Avançada/genética , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Transdução de Sinais , Tíbia/efeitos dos fármacos , Tíbia/metabolismo , Tíbia/patologia , Resultado do Tratamento , ATPases Vacuolares Próton-Translocadoras/genética , ATPases Vacuolares Próton-Translocadoras/metabolismo
12.
Cell Mol Neurobiol ; 29(3): 309-16, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-18987969

RESUMO

The blood-brain barrier (BBB) is highly restrictive of the transport of substances between blood and the central nervous system. Brain pericytes are one of the important cellular constituents of the BBB and are multifunctional, polymorphic cells that lie within the microvessel basal lamina. The present study aimed to evaluate the role of pericytes in the mediation of BBB disruption using a lipopolysaccharide (LPS)-induced model of septic encephalopathy in mice. ICR mice were injected intraperitoneally with LPS or saline and were sacrificed at 1, 3, 6, and 24 h after injection. Sodium fluorescein accumulated with time in the hippocampus after LPS injection; this hyperpermeability was supported by detecting the extravasation of fibrinogen. Microglia were activated and the number of microglia increased with time after LPS injection. LPS-treated mice exhibited a broken basal lamina and pericyte detachment from the basal lamina at 6-24 h after LPS injection. The disorganization in the pericyte and basal lamina unit was well correlated with increased microglial activation and increased cerebrovascular permeability in LPS-treated mice. These findings suggest that pericyte detachment and microglial activation may be involved in the mediation of BBB disruption due to inflammatory responses in the damaged brain.


Assuntos
Membrana Basal/patologia , Barreira Hematoencefálica/patologia , Pericitos/patologia , Sepse/patologia , Animais , Hipocampo/patologia , Lipopolissacarídeos , Camundongos , Camundongos Endogâmicos ICR , Microglia/patologia , Sepse/induzido quimicamente
13.
Neurosci Lett ; 449(1): 71-5, 2009 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-18983892

RESUMO

Brain pericytes are known to embrace the abluminal endothelial surfaces of cerebral microvessels. The rich expression of contractile proteins in these cells suggests pericytal regulation of cerebral blood flow. Here, we investigated the molecular mechanisms by which an endothelium-derived relaxing factor, adrenomedullin, was able to induce the relaxation of rat primary cultured brain pericytes. Adrenomedullin increased the relative proportion of pericytes that were relaxed, as shown by an increased cell surface area. A smaller fragment of adrenomedullin (adrenomedullin(22-52)) blocked the adrenomedullin-induced relaxation. Adrenomedullin increased intracellular cAMP concentrations and decreased the phosphorylation of myosin light chain (MLC). H89 (a PKA inhibitor) inhibited the adrenomedullin-induced increase in the number of relaxed pericytes, and returned the level of phosphorylation of MLC to the control level. The results of the present study suggest that adrenomedullin-induced relaxation of brain pericytes is related to the reduced phosphorylation of MLC through cAMP/PKA.


Assuntos
Adrenomedulina/farmacologia , Encéfalo/citologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Cadeias Leves de Miosina/metabolismo , Pericitos/efeitos dos fármacos , Vasodilatadores/farmacologia , 4-(3-Butoxi-4-metoxibenzil)-2-imidazolidinona/farmacologia , Animais , Células Cultivadas , AMP Cíclico/farmacologia , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Isoquinolinas/farmacologia , Pericitos/metabolismo , Fosforilação/efeitos dos fármacos , Ratos , Transdução de Sinais/efeitos dos fármacos , Sulfonamidas/farmacologia , Fatores de Tempo
14.
Endocrinology ; 149(10): 5177-88, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18556352

RESUMO

Peripheral administration of bacterial endotoxin [lipopolysaccharide (LPS)] to rodents produces an innate immune response and hypothalamic-pituitary-adrenal axis stimulation. Renin-angiotensin-aldosterone system inhibition by angiotensin II AT1 receptor blockade has antiinflammatory effects in the vasculature. We studied whether angiotensin II receptor blockers (ARBs) prevent the LPS response. We focused on the adrenal gland, one organ responsive to LPS and expressing a local renin-angiotensin-aldosterone system. LPS (50 microg/kg, ip) produced a generalized inflammatory response with increased release of TNF-alpha and IL-6 to the circulation, enhanced adrenal aldosterone synthesis and release, and enhanced adrenal cyclooxygenase-2, IL-6, and TNF-alpha gene expression. ACTH and corticosterone release were also increased by LPS. Pretreatment with the ARB candesartan (1 mg/kg.d, sc for 3 d before the LPS administration) decreased LPS-induced cytokine release to the circulation, adrenal aldosterone synthesis and release, and cyclooxygenase-2 and IL-6 gene expression. Candesartan did not prevent the LPS-induced ACTH and corticosterone release. Our results suggest that AT1 receptors are essential for the development of the full innate immune and stress responses to bacterial endotoxin. The ARB decreased the general peripheral inflammatory response to LPS, partially decreased the inflammatory response in the adrenal gland, prevented the release of the pro-inflammatory hormone aldosterone, and protected the antiinflammatory effects of glucocorticoid release. An unrestricted innate immune response to the bacterial endotoxin may have deleterious effects for the organism and may lead to development of chronic inflammatory disease. We postulate that the ARBs may have therapeutic effects on inflammatory conditions.


Assuntos
Glândulas Suprarrenais/imunologia , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Benzimidazóis/farmacologia , Inflamação/tratamento farmacológico , Receptor Tipo 1 de Angiotensina/metabolismo , Tetrazóis/farmacologia , Glândulas Suprarrenais/efeitos dos fármacos , Hormônio Adrenocorticotrópico/sangue , Aldosterona/sangue , Animais , Biomarcadores/sangue , Compostos de Bifenilo , Pressão Sanguínea/efeitos dos fármacos , Corticosterona/sangue , Inflamação/induzido quimicamente , Inflamação/imunologia , Interleucina-6/sangue , Receptores de Lipopolissacarídeos/metabolismo , Lipopolissacarídeos/farmacologia , Masculino , Ratos , Ratos Wistar , Sistema Renina-Angiotensina/efeitos dos fármacos , Sistema Renina-Angiotensina/imunologia , Fator de Necrose Tumoral alfa/sangue
15.
J Neurochem ; 105(4): 1394-404, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18221376

RESUMO

Cathepsin E is an endolysosomal aspartic proteinase predominantly expressed in cells of the immune system, but physiological functions of this protein in the brain remains unclear. In this study, we investigate the behavioral effect of disrupting the gene encoding cathepsin E in mice. We found that the cathepsin E-deficient (CatE-/-) mice were behaviorally normal when housed communally, but they became more aggressive compared with the wild-type littermates when housed individually in a single cage. The increased aggressive response of CatE-/- mice was reduced to the level comparable to that seen for CatE+/+ mice by pretreatment with an NK-1-specific antagonist. Consistent with this, the neurotransmitter substance P (SP) level in affective brain areas including amygdala, hypothalamus, and periaqueductal gray was significantly increased in CatE-/- mice compared with CatE+/+ mice, indicating that the increased aggressive behavior of CatE-/- mice by isolation housing followed by territorial challenge is mainly because of the enhanced SP/NK-1 receptor signaling system. Double immunofluorescence microscopy also revealed the co-localization of SP with synaptophysin but not with microtubule-associated protein-2. Our data thus indicate that cathepsin E is associated with the SP/NK-1 receptor signaling system and thereby regulates the aggressive response of the animals to stressors such as territorial challenge.


Assuntos
Agressão/fisiologia , Catepsina E/deficiência , Territorialidade , Agressão/psicologia , Animais , Catepsina E/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Atividade Motora/fisiologia , Isolamento Social/psicologia
16.
Neurosci Lett ; 441(2): 163-6, 2008 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-18603369

RESUMO

Oncostatin M (OSM), a member of the interleukin-6 family, is produced by monocytes and macrophages in the peripheral blood and microglia in the brain. The present study aimed to elucidate a regulatory role of OSM in the functions of blood-brain barrier (BBB) comprised of rat brain capillary endothelial cells (RBECs). OSM decreased the transendothelial electrical resistance of RBEC monolayers in a concentration- and time-dependent manner. Immunocytochemical observations of ZO-1 and claudin-5 in OSM-treated RBECs showed a zipper-like and/or zigzag shape along the junctions between cells, in contrast with the smooth and linear shape in vehicle-treated cultures. When RBECs were pre-treated with anti-OSM antibody, OSM failed to evoke these changes. The cellular constituents producing OSM in the brain and peripheral blood could be implicated in the functional and structural impairment of the BBB.


Assuntos
Barreira Hematoencefálica/citologia , Células Endoteliais/efeitos dos fármacos , Inibidores do Crescimento/toxicidade , Oncostatina M/toxicidade , Animais , Anticorpos/farmacologia , Células Cultivadas , Relação Dose-Resposta a Droga , Impedância Elétrica , Células Endoteliais/patologia , Inibidores do Crescimento/imunologia , Proteínas de Membrana/metabolismo , Camundongos , Oncostatina M/imunologia , Ratos , Ratos Wistar , Fatores de Tempo
17.
Am J Chin Med ; 45(7): 1497-1511, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29025293

RESUMO

Berberine (BBR), an isoquinoline alkaloid, is a well-known bioactive compound contained in medicinal plants used in traditional and folk medicines. In this study, we investigated the subcellular localization and the apoptotic mechanisms of BBR were elucidated. First, we confirmed the incorporation of BBR into the cell visually. BBR showed antiproliferative activity and promptly localized to the nucleus from 5[Formula: see text]min to 15[Formula: see text]min after BBR treatment in HL-60 human promyelocytic leukemia cells. Next, we examined the antiproliferative activity of BBR (1) and its biosynthetically related compounds (2-7) in HL-60 cells. BBR exerted strongest antiproliferative activity among 1-7 and the results of structures and activity relation suggested that a methylenedioxyl group in ring A, an [Formula: see text]-alkyl group at C-9 position, and the frame of isoquinoline may be necessary for antiproliferative activity. Moreover, BBR showed the most potent antiproliferative activity in HL-60 cells among human cancer and normal cell lines tested. Next, we examined the effect of BBR on molecular events known as apoptosis induction. In HL-60 cells, BBR induced chromatin condensation and DNA fragmentation, and triggered the activation of PARP, caspase-3 and caspase-8 without the activation of caspase-9. BBR-induced DNA fragmentation was abolished by pretreatment with inhibitors against caspase-3 and caspase-8, but not against caspase-9. ERK and p38 were promptly phosphorylated after 15 min of BBR treatment, and this was correlated with time of localization to the nucleus of BBR. These results demonstrated that BBR translocated into nucleus immediately after treatments and induced apoptotic cell death by activation of caspase-3 and caspase-8.


Assuntos
Apoptose/efeitos dos fármacos , Berberina/metabolismo , Berberina/farmacologia , Caspase 3/metabolismo , Caspase 8/metabolismo , Núcleo Celular/metabolismo , Leucemia Promielocítica Aguda/patologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Cromatina/metabolismo , Fragmentação do DNA/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Células HL-60 , Humanos , Leucemia Promielocítica Aguda/metabolismo , Fosforilação/efeitos dos fármacos , Relação Estrutura-Atividade , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
18.
Stroke ; 37(5): 1271-6, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16601219

RESUMO

BACKGROUND AND PURPOSE: Blockade of angiotensin II AT1 receptors in cerebral microvessels protects against brain ischemia and inflammation. In this study, we tried to clarify the presence and regulation of the local renin-angiotensin system (RAS) in brain microvessels in hypertension. METHODS: Spontaneously hypertensive rats (SHR) and Wistar Kyoto (WKY) controls were treated with an AT1 receptor antagonist (candesartan, 0.3 mg/kg per day) via subcutaneous osmotic minipumps for 4 weeks. The expression and localization of RAS components and the effect of AT1 receptor blockade were assessed by Affymetrix microarray, qRT-PCR, Western blots, immunohistochemistry and immunofluorescence. RESULTS: We found transcripts of most of RAS components in our microarray database, and confirmed their expression by qRT-PCR. Angiotensinogen (Aogen), angiotensin-converting enzyme (ACE) and AT1 receptors were localized to the endothelium. There was no evidence of AT2 receptor localization in the microvascular endothelium. In SHR, (pro)renin receptor mRNA and AT1 receptor mRNA and protein expression were higher, whereas Aogen, ACE mRNA and AT2 receptor mRNA and protein expression were lower than in WKY rats. Candesartan treatment increased Aogen, ACE and AT2 receptor in SHR, and increased ACE and decreased Aogen in WKY rats, without affecting the (pro)renin and AT1 receptors. CONCLUSIONS: Increased (pro)renin and AT1 receptor expression in SHR substantiates the importance of the local RAS overdrive in the cerebrovascular pathophysiology in hypertension. AT1 receptor blockade and increased AT2 receptor stimulation after administration of candesartan may contribute to the protection against brain ischemia and inflammation.


Assuntos
Angiotensina II/metabolismo , Encéfalo/irrigação sanguínea , Hipertensão/metabolismo , Microcirculação/metabolismo , Receptor Tipo 1 de Angiotensina/metabolismo , Transdução de Sinais , Angiotensina II/antagonistas & inibidores , Angiotensina II/genética , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Animais , Benzimidazóis/farmacologia , Compostos de Bifenilo , Regulação da Expressão Gênica , Hipertensão/fisiopatologia , Masculino , Microcirculação/fisiopatologia , Ratos , Ratos Endogâmicos SHR , Receptor Tipo 1 de Angiotensina/genética , Sistema Renina-Angiotensina/genética , Transdução de Sinais/efeitos dos fármacos , Tetrazóis/farmacologia
19.
J Inflamm (Lond) ; 10(1): 32, 2013 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-24107300

RESUMO

BACKGROUND: BMP4, a member of the transforming growth factor-beta superfamily, is upregulated in the aortas of diabetic db/db mice. However, little is known about its role in diabetic atherosclerosis. Therefore, we examined the roles of BMP4 in the formation of diabetic atherosclerosis in apolipoprotein E knockout (ApoE KO) mice and in the uptake of oxidized low density lipoprotein (oxLDL) in peritoneal macrophages of wild-type mice. METHODS: To induce diabetes, ApoE KO mice were intraperitoneally injected with streptozotocin. Diabetic and non-diabetic ApoE KO mice were then fed a high-fat diet for 4 weeks. Next, to investigate a role of BMP4 in the peritoneal macrophages, we examined the uptake of oxLDL in BMP4-treated macrophages. RESULTS: Diabetic ApoE KO mice showed accelerated progression of aortic plaques accompanied by increased luminal plaque area. Western blot analysis showed that BMP4 expression in the whole aorta was greatly increased in diabetic ApoE KO mice, than non-diabetic mice. Western blot analysis showed that the BMP4/SMAD1/5/8 signaling pathway was strongly activated in the aorta from diabetic ApoE KO mice, compared with control ApoE KO mice. Double immunofluorescence staining showed that BMP4 was expressed in MOMA2-labeled macrophage in the aortic lesions of ApoE KO mice. BMP4 significantly increased the uptake of oxLDL into peritoneal macrophages in vitro. CONCLUSION: We show that in the aorta of diabetic ApoE KO mice, BMP4 is increased and activates SMAD1/5/8. Our in vitro findings indicate that BMP4 enhances oxLDL uptake in mouse peritoneal macrophages, suggesting BMP4 may be involved in aortic plaque formation in diabetic ApoE KO mice. Targeting BMP4 may offer a new strategy for inhibition of plaque progression and stabilization of atherosclerotic lesions.

20.
Neurosci Lett ; 531(2): 114-9, 2012 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-23127852

RESUMO

Recently, 3-hydroxy-3-methyl glutaryl coenzyme A (HMG-CoA) reductase inhibitors were reported to induce neurite outgrowth in vitro. However, the mechanism underlying this effect remains unclear. Cellular prion protein (PrP(C)) is a ubiquitous glycoprotein present on the surfaces of various cells, including neurons, and is suggested to be involved in neurite outgrowth. Therefore, the present study aimed to determine whether PrP(C) mediates neurite outgrowth induced by HMG-CoA reductase inhibitors. Atorvastatin, a strong HMG-CoA reductase inhibitor, induced neurite outgrowth and increased PrP(C) levels in Neuro2a cells in a time- and dose-dependent manner. PrP(C) mRNA expression was also increased by atorvastatin. Farnesol, a non-sterol mevalonate derivative, attenuated the atorvastatin-induced neurite outgrowth and increase in PrP(C). Neuro2a cells overexpressing PrP(C) showed a remarkable enhancement of atorvastatin-induced neurite outgrowth compared with mock cells transfected with empty pCI-neo vector. These findings suggest that PrP(C) contributes, at least in part, to atorvastatin-induced neurite outgrowth. This phenomenon may be included among the mechanisms underlying decreased risk of Alzheimer's disease in patients treated with HMG-CoA reductase inhibitors.


Assuntos
Ácidos Heptanoicos/farmacologia , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Neuritos/metabolismo , Príons/metabolismo , Pirróis/farmacologia , Animais , Atorvastatina , Linhagem Celular Tumoral , Camundongos , Neuritos/efeitos dos fármacos , Neuroblastoma , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA