Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
1.
Life Sci ; 330: 122008, 2023 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-37549828

RESUMO

AIM: Sepsis-induced cardiac dysfunction is the leading cause of higher morbidity and mortality with poor prognosis in septic patients. Our recent previous investigation provides evidence of the hallmarks of signal transducer and activator of transcription3 (STAT3) activation in sepsis and targeting of STAT3 with Stattic, a small-molecule inhibitor of STAT3, has beneficial effects in various septic tissues. We investigated the possible cardioprotective effects of Stattic on cardiac inflammation and dysfunction in mice with cecal ligation and puncture (CLP)-induced sepsis. MAIN METHODS: A polymicrobial sepsis model was induced by CLP in mice and Stattic (25 mg/kg) was intraperitoneally given at one and twelve hours after CLP operation. The cecum was exposed in sham-control mice without CLP. After 18 h of surgery, electrocardiogram (ECG) for anaesthized mice was registered followed by collecting of samples of blood and tissues for bimolecular and histopathological assessments. Myeloperoxidase, a marker of neutrophil infiltration, was assessed immunohistochemically. KEY FINDINGS: CLP profoundly impaired cardiac functions as evidenced by ECG changes in septic mice as well as elevation of cardiac enzymes, and inflammatory markers with myocardial histopathological and immunohistochemical alterations. While, Stattic markedly reversed the CLP-induced cardiac abnormalities and restored the cardiac function by its anti-inflammatory activities. SIGNIFICANCE: Stattic treatment had potential beneficial effects against sepsis-induced cardiac inflammation, dysfunction and damage. Its cardioprotective effects were possibly attributed to its anti-inflammatory activities by targeting STAT3 and downregulation of IL-6 and gp130. Our investigations suggest that Stattic could be a promising target for management of cardiac sepsis and inflammation-related cardiac damage.


Assuntos
Traumatismos Cardíacos , Sepse , Animais , Camundongos , Anti-Inflamatórios/farmacologia , Ceco/cirurgia , Ceco/lesões , Receptor gp130 de Citocina/metabolismo , Inflamação/etiologia , Interleucina-6/metabolismo , Ligadura/efeitos adversos , Punções/efeitos adversos , Sepse/tratamento farmacológico , Transdução de Sinais
2.
Nihon Yakurigaku Zasshi ; 157(4): 226-231, 2022.
Artigo em Japonês | MEDLINE | ID: mdl-35781449

RESUMO

The acute respiratory distress syndrome (ARDS) is an important cause of respiratory failure in critically ill patients and may become a life-threatening condition where inflammation of the lungs may begin in one lung but eventually affects both, leading to damage to the alveoli and surrounding small blood vessels. ARDS is particularly characterized by noncardiogenic pulmonary edema caused by an increase in pulmonary capillary permeability. Several clinical disorders can precipitate in ARDS, including pneumonia, sepsis, aspiration of gastric contents, and major trauma. The most common cause of ARDS is sepsis, which is a serious and widespread infection of the bloodstream and is now defined as life-threatening organ dysfunction due to a dysregulated reponse of the host to infection. In sepsis, a number of vascular hyperpermeable factors, such as histamine, nitric oxide, thromboxane A2, and vascular endothelial growth factor, can be overproducted and contribute to the development of pulmonary edema. Given that sepsis can be regarded as a gene-related disorder, the nucleic-acid based gene therapeutic strategy to regulate some transcription factors involved in expression of vascular hyperpermeable genes may be considered to be a promising novel approach for treatment of ARDS in sepsis.


Assuntos
Edema Pulmonar , Síndrome do Desconforto Respiratório , Sepse , Humanos , Pulmão , Edema Pulmonar/etiologia , Síndrome do Desconforto Respiratório/etiologia , Sepse/complicações , Fator A de Crescimento do Endotélio Vascular
3.
Naunyn Schmiedebergs Arch Pharmacol ; 394(10): 2129-2139, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34402957

RESUMO

Cumulative evidence has established that macrophages orchestrate inflammatory responses that crucially contribute to the pathogenesis of insulin-resistant obesity and type 2 diabetes. In the present study, we examined the impact of hyperglycemia on macrophage pro-inflammatory responses under an inflammatory stimulus. To conduct this study, RAW264.7 macrophages were cultured under normal- (5.5 mM) or high-glucose (22 or 40 mM) conditions for 7 days and stimulated with lipopolysaccharide (LPS). Long-term exposure to high glucose significantly enhanced the increase in the production of pro-inflammatory cytokines, including tumor necrosis-α, interleukin (IL)-1ß, and IL-6, when macrophages were stimulated with LPS. The LPS-induced increases in inducible nitric oxide (NO) synthase (iNOS) expression and NO production were also significantly enhanced by long-term exposure of macrophages to high glucose. Treatment with N-acetyl-L-cysteine, a widely used thiol-containing antioxidant, blunted the enhancement of the LPS-induced upregulation of pro-inflammatory cytokine production, iNOS expression, and NO production in macrophages. When intracellular reactive oxygen species (ROS) were visualized using the fluorescence dye 5-(and-6)-chloromethyl-2',7'-dichlorofluorescein diacetate, acetyl ester, a significant increase in ROS generation was found after stimulation of macrophages with LPS, and this increased ROS generation was exacerbated under long-term high-glucose conditions. LPS-induced translocation of phosphorylated nuclear factor-κB (NF-κB), a transcription factor regulating many pro-inflammatory genes, into the nucleus was promoted under long-term high-glucose conditions. Altogether, the present results indicate that a long-term high-glucose environment can enhance activation of NF-κB in LPS-stimulated macrophages possibly due to excessive ROS production, thereby leading to increased macrophage pro-inflammatory responses.


Assuntos
Glucose/farmacologia , Lipopolissacarídeos/farmacologia , NF-kappa B/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Animais , Citocinas/metabolismo , Heme Oxigenase-1/metabolismo , Hiperglicemia/induzido quimicamente , Hiperglicemia/metabolismo , Inflamação/induzido quimicamente , Inflamação/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Fator 2 Relacionado a NF-E2/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Células RAW 264.7
4.
Intern Med ; 60(5): 755-759, 2021 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-33028772

RESUMO

Keishibukuryogan is a Kampo medicine that induces vasodilation and improves the blood flow velocity in subcutaneous blood vessels. We herein report two cases in which keishibukuryogan completely diminished subcutaneous hematoma after cardiac resynchronization therapy pacemaker implantation and defibrillator battery replacement within a month. Keishibukuryogan can be a good option for treating or preventing subcutaneous hematoma after surgical procedures for devices.


Assuntos
Terapia de Ressincronização Cardíaca , Desfibriladores Implantáveis , Medicamentos de Ervas Chinesas , Marca-Passo Artificial , Desfibriladores Implantáveis/efeitos adversos , Hematoma/diagnóstico por imagem , Hematoma/etiologia , Humanos , Marca-Passo Artificial/efeitos adversos
5.
Naunyn Schmiedebergs Arch Pharmacol ; 393(12): 2365-2374, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32696151

RESUMO

Vascular endothelial growth factor (VEGF) is a prime regulator of vascular permeability. Acute lung injury (ALI) is characterized by high-permeability pulmonary edema in addition to refractory hypoxemia and diffuse pulmonary infiltrates. In this study, we examined whether VEGF can be implicated as a pulmonary vascular permeability factor in sepsis-associated ALI. We found that a great increase in lung vascular leak occurred in mice instilled intranasally with lipopolysaccharide (LPS), as assessed by IgM levels in bronchoalveolar lavage fluid. Treatment with the VEGF-neutralizing monoclonal antibody bevacizumab significantly reduced this hyperpermeability response, suggesting active participation of VEGF in non-cardiogenic lung edema associated with LPS-induced ALI. However, this was not solely attributable to excessive levels of intrapulmonary VEGF. Expression levels of VEGF were significantly reduced in lung tissues from mice with both intranasal LPS administration and cecal ligation and puncture (CLP)-induced sepsis, which may stem from decreases in non-endothelial cells-dependent VEGF production in the lungs. In support of this assumption, stimulation with LPS and interferon-γ (IFN-γ) significantly increased VEGF in human pulmonary microvascular endothelial cells (HPMECs) at mRNA and protein levels. Furthermore, a significant rise in plasma VEGF levels was observed in CLP-induced septic mice. The increase in VEGF released from HPMECs after LPS/IFN-γ challenge was completely blocked by either specific inhibitor of mitogen-activated protein kinase (MAPK) subgroups. Taken together, our results indicate that VEGF can contribute to the development of non-cardiogenic lung edema in sepsis-associated ALI due to increased VEGF secretion from pulmonary vascular endothelial cells through multiple MAPK-dependent pathways.


Assuntos
Lesão Pulmonar Aguda/sangue , Permeabilidade Capilar/fisiologia , Pulmão/irrigação sanguínea , Pulmão/metabolismo , Sepse/sangue , Fator A de Crescimento do Endotélio Vascular/sangue , Lesão Pulmonar Aguda/etiologia , Lesão Pulmonar Aguda/patologia , Animais , Linhagem Celular Transformada , Humanos , Pulmão/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Sepse/complicações , Sepse/patologia
7.
Int Immunopharmacol ; 62: 120-131, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30005227

RESUMO

Cilostazol, a selective inhibitor of phosphodiesterase type III with anti-platelet, anti-mitogenic, and vasodilating properties, is widely used to treat ischemic symptoms of peripheral vascular disease. Ample evidence has suggested that cilostazol also exhibits an anti-inflammatory effect, but its anti-inflammatory mechanism is not fully understood. Here, we showed that cilostazol specifically inhibited expression of cytokines, which are induced by nuclear factor-κB (NF-κB) activation, in RAW264.7 macrophage cells stimulated with different Toll-like receptor (TLR) ligands. Cilostazol was found to significantly reduce TLR-4 and TLR-3 ligands-stimulated NF-κB transcriptional activity, which was quantified by luciferase reporter assays. However, cilostazol was without effect on IκBα degradation and NF-κB p65 phosphorylation and nuclear translocation after challenge with the TLR-4 ligand lipopolysaccharide (LPS). Cilostazol did not also prevent the LPS-induced increase in phosphorylated levels of the mitogen-activated protein kinase (MAPK) family. On the other hand, using chromatin immunoprecipitation assays, we demonstrated that cilostazol reduced the LPS-induced transcriptional activities of interleukin-6 and tumor necrosis factor-α by preventing the recruitment of NF-κB p65 to these gene promoters. When cilostazol was given to mice by oral gavage daily for 7 days, LPS-induced aberrant pro-inflammatory cytokine production and end-organ tissue injury were significantly reduced. The results of this study suggest that cilostazol is capable of directly interrupting DNA binding activity of NF-κB proteins from the TLR signaling pathways. The therapy to specifically intervene in this pathway may be potentially beneficial for the prevention of different inflammatory disorders.


Assuntos
Anti-Inflamatórios/farmacologia , Cilostazol/farmacologia , DNA/metabolismo , NF-kappa B/metabolismo , Receptores Toll-Like/metabolismo , Animais , Citocinas/genética , Camundongos , Inibidor de NF-kappaB alfa/metabolismo , NF-kappa B/genética , Células RAW 264.7 , Transdução de Sinais , Transcrição Gênica/efeitos dos fármacos
8.
J Neurochem ; 145(6): 474-488, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29500815

RESUMO

Sepsis-associated encephalopathy (SAE), characterized as diffuse brain dysfunction and neurological manifestations secondary to sepsis, is a common complication in critically ill patients and can give rise to poor outcome, but understanding the molecular basis of this disorder remains a major challenge. Given the emerging role of G protein-coupled receptor 2 (GRK2), first identified as a G protein-coupled receptor (GPCR) regulator, in the regulation of non-G protein-coupled receptor-related molecules contributing to diverse cellular functions and pathology, including inflammation, we tested the hypothesis that GRK2 may be linked to the neuropathogenesis of SAE. When mouse MG6 microglial cells were challenged with lipopolysaccharide (LPS), GRK2 cytosolic expression was highly up-regulated. The ablation of GRK2 by small interfering RNAs (siRNAs) prevented an increase in intracellular reactive oxygen species generation in LPS-stimulated MG6 cells. Furthermore, the LPS-induced up-regulation of inducible nitric-oxide synthase expression and increase in nitric oxide production were negated by GRK2 inhibitor or siRNAs. However, GRK2 inhibition was without effect on overproduction of tumor necrosis factor-α, interleukin (IL)-6, and IL-1ß in LPS-stimulated MG cells. In mice with cecal ligation and puncture-induced sepsis, treatment with GRK2 inhibitor reduced high levels of oxidative and nitrosative stress in the mice brains, where GRK2 expression was up-regulated, alleviated neurohistological damage observed in cerebral cortex sections, and conferred a significant survival advantage to CLP mice. Altogether, these results uncover the novel role for GRK2 in regulating cellular oxidative and nitrosative stress during inflammation and suggest that GRK2 may have a potential as an intriguing therapeutic target to prevent or treat SAE.


Assuntos
Quinase 2 de Receptor Acoplado a Proteína G/fisiologia , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Encefalopatia Associada a Sepse/patologia , Animais , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/patologia , Citocinas/biossíntese , Inibidores Enzimáticos/uso terapêutico , Quinase 2 de Receptor Acoplado a Proteína G/antagonistas & inibidores , Lipopolissacarídeos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Microglia/enzimologia , Óxido Nítrico Sintase Tipo II/metabolismo , RNA Interferente Pequeno/farmacologia , Encefalopatia Associada a Sepse/complicações , Encefalopatia Associada a Sepse/tratamento farmacológico , Regulação para Cima/efeitos dos fármacos
9.
Crit Care Med ; 46(5): e435-e442, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29406423

RESUMO

OBJECTIVES: Inflammation and apoptosis are decisive mechanisms for the development of end-organ injury in sepsis. Activator protein-1 may play a key role in regulating expression of harmful genes responsible for the pathophysiology of septic end-organ injury along with the major transcription factor nuclear factor-κB. We investigated whether in vivo introduction of circular dumbbell activator protein-1 decoy oligodeoxynucleotides can provide benefits for reducing septic end-organ injury. DESIGN: Laboratory and animal/cell research. SETTINGS: University research laboratory. SUBJECTS: Male BALB/c mice (8-10 wk old). INTERVENTIONS: Activator protein-1 decoy oligodeoxynucleotides were effectively delivered into tissues of septic mice in vivo by preparing into a complex with atelocollagen given 1 hour after surgery. MATERIALS AND MAIN RESULTS: Polymicrobial sepsis was induced by cecal ligation and puncture in mice. Activator protein-1 decoy oligodeoxynucleotide transfection inhibited abnormal production of proinflammatory and chemotactic cytokines after cecal ligation and puncture. Histopathologic changes in lung, liver, and kidney tissues after cecal ligation and puncture were improved by activator protein-1 decoy oligodeoxynucleotide administration. When activator protein-1 decoy oligodeoxynucleotides were given, apoptosis induction was strikingly suppressed in lungs, livers, kidneys, and spleens of cecal ligation and puncture mice. These beneficial effects of activator protein-1 decoy oligodeoxynucleotides led to a significant survival advantage in mice after cecal ligation and puncture. Apoptotic gene profiling indicated that activator protein-1 activation was involved in the up-regulation of many of proapoptotic and antiapoptotic genes in cecal ligation and puncture-induced sepsis. CONCLUSIONS: Our results indicate a detrimental role of activator protein-1 in the sepsis pathophysiology and the potential usefulness of activator protein-1 decoy oligodeoxynucleotides for the prevention and treatment of septic end-organ failure.


Assuntos
Oligodesoxirribonucleotídeos/uso terapêutico , Sepse/terapia , Fator de Transcrição AP-1/uso terapêutico , Transfecção/métodos , Animais , Apoptose , Citocinas/sangue , Modelos Animais de Doenças , Ensaio de Desvio de Mobilidade Eletroforética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Oligodesoxirribonucleotídeos/genética , Reação em Cadeia da Polimerase em Tempo Real , Sepse/genética , Sepse/mortalidade , Sepse/patologia , Fator de Transcrição AP-1/genética
10.
Biochem Biophys Res Commun ; 493(3): 1342-1348, 2017 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-28970068

RESUMO

Aberrant activation of Wnt signaling plays a pivotal role in the development of human cancers including colon cancer. Small compounds that regulate Wnt signaling are attractive candidate for the colon cancer therapy. Here, we showed that SKL2001, which has been identified as an activator for Wnt signaling by disrupting the Axin/ß-Catenin complex, negatively regulates growth of colon cancer spheroids cultured in the 3D condition that simulates tumor microenvironment in vivo. SKL2001 inhibited proliferation of colon cancer cells cultured in 3D spheroid and induced them accumulation in the G0/G1 phase of the cell cycle with a reduced c-myc level. To examine the potential of arrested cells to recover, colon cancer spheroids that were treated with SKL2001 were then cultured in the SKL2001-free medium. We found that SKL2001-treated cells were resumed cell cycle progression and proliferated in the SKL2001-free medium. Notably, SKL2001 facilitated round-shape spheroid formation. This was associated with upregulated expressions of E-cadherin and ß-Catenin. These findings suggest that SKL2001 can suppress colon cancer spheroid growth through regulating cell cycle progression and cadherin/catenin mediated cell-cell contact.


Assuntos
Caderinas/metabolismo , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/patologia , Imidazóis/farmacologia , Isoxazóis/farmacologia , beta Catenina/metabolismo , Antígenos CD , Antineoplásicos/farmacologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Neoplasias do Colo/metabolismo , Células HCT116 , Humanos , Esferoides Celulares/efeitos dos fármacos , Células Tumorais Cultivadas , Microambiente Tumoral
11.
Toxicol Appl Pharmacol ; 334: 55-65, 2017 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-28887131

RESUMO

Prostacyclin (PGI2) serves as a protective, anti-inflammatory mediator and PGI2 mimetics may be useful as a hepatoprotective agent. We examined whether two PGI2 mimetics, ONO-1301 and beraprost, are beneficial in acute liver injury and attempted to delineate the possible mechanism underlying the hepatoprotective effect. Acute liver injury was induced by lipopolysaccharide/d-galactosamine (LPS/GalN) in mice. Mice were given an intraperitoneal injection of PGI2 mimetics 1h before LPS/GalN challenge. Both ONO-1301 and beraprost significantly declined the LPS/GalN-induced increase in serum aminotransferase activity. ONO-1301 and, to a lesser extent, beraprost inhibited hepatic gene expression levels of pro-inflammatory cytokines, which were sharply elevated by LPS/GalN. The hepatoprotective effects of ONO-1301, to a lesser extent, of beraprost were also supported by liver histopathological examinations. The PGI2 receptor antagonist CAY10441 abrogated their hepatoprotective effects. The mechanisms behind the benefit of PGI2 mimetics in reducing LPS/GalN-induced liver injury involved, in part, their suppressive effects on increased generation of reactive oxygen species (ROS), since their ability to prevent LPS/GalN-induced hepatic apoptosis was mimicked by the antioxidant N-acetyl-l-cysteine. They significantly diminished LPS/GalN-induced activation of signal transducers and activators of transcription 3 (STAT3) in liver tissues, an effect which was highly associated with their hepatoprotective effects. We indicate that IP receptor activation with PGI2 mimetics can rescue the damage in the liver induced by LPS/GalN by undermining activation of STAT3 and leading to a lower production of ROS. Our findings point to PGI2 mimetics, especially ONO-1301, as a potential novel therapeutic modality for the treatment of acute liver injury.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Epoprostenol/análogos & derivados , Galactosamina/toxicidade , Lipopolissacarídeos/toxicidade , Piridinas/farmacologia , Animais , Compostos de Benzil/farmacologia , Proteína de Ligação a CREB/genética , Proteína de Ligação a CREB/metabolismo , Epoprostenol/farmacologia , Galactosamina/administração & dosagem , Regulação da Expressão Gênica , Imidazóis/farmacologia , Lipopolissacarídeos/administração & dosagem , Camundongos , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Prostaglandinas I/química , Prostaglandinas I/farmacologia , Espécies Reativas de Oxigênio , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais
12.
Am J Physiol Heart Circ Physiol ; 312(6): H1224-H1237, 2017 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-28455289

RESUMO

Dobutamine has been used in septic shock for many years as an only inotrope, but its benefit has been questioned. We weighed the effects of dobutamine and milrinone as inotropes in mice with cecal ligation and puncture (CLP)-induced polymicrobial sepsis. CLP-induced septic mice exhibited significant cardiac inflammation, as indicated by greatly increased mRNAs of proinflammatory cytokines and robust infiltration of inflammatory cells in the ventricular myocardium. Elevations of plasma cardiac troponin-I showed cardiac injury in CLP mice. Noninvasive echocardiographic assessment of cardiac function revealed that despite preserved left ventricular function in the presence of fluid replacement, the dobutamine inotropic response was significantly impaired in CLP mice compared with sham-operated controls. By contrast, milrinone exerted inotropic effects in sham-operated and CLP mice in an equally effective manner. Surface expression levels of ß1-adrenoceptors and α-subunits of three main G protein families in the myocardium were unaffected by CLP-induced sepsis. Plasma cAMP levels were significantly elevated in both sham-operated and CLP mice in response to milrinone but only in sham-operated controls in response to dobutamine. Of phosphodiesterase (PDE) isoforms, PDE4D, but not PDE3A, both of which are responsible for cardiac cAMP hydrolysis, was significantly upregulated in CLP mouse myocardium. We define a novel mechanism for the impaired responsiveness to dobutamine as an inotrope in sepsis, and understanding the role of PDE4D in modulating cardiac functional responsiveness in sepsis may open the potential of a PDE4D-targeted therapeutic option in septic patients with low cardiac output who have a need for inotropic support.NEW & NOTEWORTHY Advisability of the usefulness of dobutamine in septic shock management is limited. Here, we reveal that the effect of dobutamine as a positive inotrope is impaired in mice with cecal ligation and puncture-induced sepsis without changes in cardiac ß1-adrenoceptor signaling as a result of cAMP breakdown achieved by upregulated phosphodiesterase 4D.


Assuntos
Cardiotônicos/farmacologia , Ceco/cirurgia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/metabolismo , Dobutamina/farmacologia , Milrinona/farmacologia , Contração Miocárdica/efeitos dos fármacos , Miocárdio/enzimologia , Inibidores da Fosfodiesterase 3/farmacologia , Sepse/tratamento farmacológico , Sepse/enzimologia , Adenilil Ciclases/metabolismo , Animais , Ceco/microbiologia , AMP Cíclico/sangue , Nucleotídeo Cíclico Fosfodiesterase do Tipo 3/genética , Nucleotídeo Cíclico Fosfodiesterase do Tipo 3/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/genética , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Hidrólise , Mediadores da Inflamação/metabolismo , Ligadura , Masculino , Camundongos Endogâmicos BALB C , Punções , Receptores Adrenérgicos beta 1/genética , Receptores Adrenérgicos beta 1/metabolismo , Sepse/microbiologia , Sepse/fisiopatologia , Transdução de Sinais , Regulação para Cima
13.
Int J Oncol ; 50(3): 787-797, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28197625

RESUMO

The effect of resveratrol on various human cancer cells was investigated with special focus on apoptotic cell death, in an attempt to further characterize its mechanism of action. There were great differences in the anti-viability effect of resveratrol between different types of human cancer cells. While the inhibition of cell viability by resveratrol was marked in U937 and MOLT-4 leukemia cells, resveratrol moderately inhibited cell viability in MCF-7 breast, HepG2 liver, and A549 lung cancer cells, and the effect was slight on cell viability in Caco-2, HCT116, and SW480 colon cancer cells. Following resveratrol treatment, U937 and MOLT-4 markedly increased the population of late apoptotic cells but MCF-7 and HepG2 underwent apoptosis with an increased population of early apoptosis, and resveratrol-induced DNA fragmentation was observed only in leukemic cells. Activation of sirtuin 1 and adenosine-monophosphate-activated protein kinase was not responsible for resveratrol-induced cancer cell death. Instead, resveratrol significantly reduced Akt activation with the downregulation of H-Ras, resulting in facilitation of Bax translocation to mitochondria in leukemic cells. This study suggests that resveratrol can induce apoptotic cell death in human leukemic cells to a greater extent than in human solid tumor cells via reducing Akt activation due to Ras downregulation.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Apoptose/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Fragmentação do DNA/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Estilbenos/farmacologia , Células A549 , Células CACO-2 , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Células HCT116 , Células Hep G2 , Humanos , Células MCF-7 , Mitocôndrias/metabolismo , Neoplasias/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Resveratrol , Sirtuína 1/metabolismo , Proteína X Associada a bcl-2/metabolismo
14.
PLoS Genet ; 12(10): e1006349, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27736879

RESUMO

Zinc transporters play a critical role in spatiotemporal regulation of zinc homeostasis. Although disruption of zinc homeostasis has been implicated in disorders such as intestinal inflammation and aberrant epithelial morphology, it is largely unknown which zinc transporters are responsible for the intestinal epithelial homeostasis. Here, we show that Zrt-Irt-like protein (ZIP) transporter ZIP7, which is highly expressed in the intestinal crypt, is essential for intestinal epithelial proliferation. Mice lacking Zip7 in intestinal epithelium triggered endoplasmic reticulum (ER) stress in proliferative progenitor cells, leading to significant cell death of progenitor cells. Zip7 deficiency led to the loss of Olfm4+ intestinal stem cells and the degeneration of post-mitotic Paneth cells, indicating a fundamental requirement for Zip7 in homeostatic intestinal regeneration. Taken together, these findings provide evidence for the importance of ZIP7 in maintenance of intestinal epithelial homeostasis through the regulation of ER function in proliferative progenitor cells and maintenance of intestinal stem cells. Therapeutic targeting of ZIP7 could lead to effective treatment of gastrointestinal disorders.


Assuntos
Proteínas de Transporte de Cátions/genética , Proliferação de Células/genética , Estresse do Retículo Endoplasmático/genética , Zinco/metabolismo , Animais , Apoptose/genética , Proteínas de Transporte de Cátions/biossíntese , Retículo Endoplasmático/genética , Células Epiteliais/metabolismo , Trato Gastrointestinal/crescimento & desenvolvimento , Trato Gastrointestinal/metabolismo , Técnicas de Inativação de Genes , Homeostase , Mucosa Intestinal/crescimento & desenvolvimento , Mucosa Intestinal/metabolismo , Camundongos , Organoides/crescimento & desenvolvimento , Celulas de Paneth/metabolismo , Células-Tronco/metabolismo
15.
Genom Data ; 8: 126-30, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27275413

RESUMO

Radiation exposure such as A-bomb or radiation therapy is considered a major health-risk factor for cardiovascular disease. In order to understand the molecular mechanisms underlying the inflammatory reaction frequently encountered in the vascular system after exposure to ionizing radiation, we carried out a global scale microarray and computational gene expression analyses on human umbilical endothelial cells (HUVECs) exposed to X-ray (2.5 Gy). The gene ontology analysis revealed that the down-regulated genes were associated with cell cycle regulation, whereas the up-regulated genes were associated with inflammatory responses, in particular, the type 1 interferon response. The computational analysis using ingenuity pathway analysis also identified a gene network containing the interferon response factor 7 (IRF7) and its transcriptional targets such as interferon-induced transcripts (IFITs) and Mx1, which have been known to be associated with inflammation in endothelial cells. The up-regulated genes and the gene network identified here may explain the inflammatory response induced by X-irradiation. These findings uncover part of the molecular basis of the mechanism(s) of the inflammatory disorder in response to X-irradiation in HUVECs. The dataset is publicly available at the Gene Expression Omnibus (GEO) repository (http://www.ncbi.nlm.nih.gov/geo/) with accession number GSE76484.

16.
Life Sci ; 150: 24-31, 2016 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-26924495

RESUMO

Glycemic fluctuations may play a critical role in the pathogenesis of diabetic complications, such as cardiovascular disease. We investigated whether the oxycarotenoid astaxanthin can reduce the detrimental effects of fluctuating glucose on vascular endothelial cells. Human umbilical venous endothelial cells were incubated for 3 days in media containing 5.5mM glucose, 22 mM glucose, or 5.5mM glucose alternating with 22 mM glucose in the absence or presence of astaxanthin or N-acetyl-L-cysteine (NAC). Constant high glucose increased reactive oxygen species (ROS) generation, but such an effect was more pronounced in fluctuating glucose. This was associated with up-regulated p22(phox) expression and down-regulated peroxisome proliferator activated receptor-γ coactivator (PGC-1α) expression. Astaxanthin inhibited ROS generation, p22(phox) up-regulation, and PGC-1α down-regulation by the stimuli of glucose fluctuation. Fluctuating glucose, but not constant high glucose, significantly decreased the endothelial nitric oxide synthase (eNOS) phosphorylation level at Ser-1177 without affecting total eNOS expression, which was prevented by astaxanthin as well as by the anti-oxidant NAC. Transferase-mediated dUTP nick end labeling (TUNEL) showed increased cell apoptosis in fluctuating glucose. Glucose fluctuation also resulted in up-regulating gene expression of pro-inflammatory mediators, interleukin-6 and intercellular adhesion molecule-1. These adverse changes were subdued by astaxanthin. The phosphorylation levels of c-Jun N-terminal kinase (JNK) and p38 were significantly increased by glucose fluctuations, and astaxanthin significantly inhibited the increase in JNK and p38 phosphorylation. Taken together, our results suggest that astaxanthin can protect vascular endothelial cells against glucose fluctuation by reducing ROS generation.


Assuntos
Antioxidantes/farmacologia , Glucose/farmacologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Acetilcisteína/farmacologia , Apoptose/efeitos dos fármacos , Glucose/metabolismo , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Inflamação/genética , Proteínas Quinases Ativadas por Mitógeno/biossíntese , Proteínas Quinases Ativadas por Mitógeno/genética , NADPH Oxidases/biossíntese , NADPH Oxidases/genética , Óxido Nítrico Sintase Tipo III/biossíntese , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Espécies Reativas de Oxigênio/metabolismo , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , Xantofilas/farmacologia
17.
Nihon Rinsho ; 74(10): 1761-1768, 2016 10.
Artigo em Japonês | MEDLINE | ID: mdl-30551294

RESUMO

G protein-coupled receptor kinase 2(GRK2) is a ubiquitous member of the family of GRKs that are serine/threonine kinases originally discovered for their role in the process of desensitization of agonist-activated G protein-coupled receptors (GPCRs). However, emerging evidence suggests that GRK2 can phosphorylate a large number of non-GPCR substrates and interact with a plethora of proteins involved in signaling and trafficking, suggesting that GRK2 would participate in the regulation of diverse cellular responses in a phosphorylation-dependent and -independent manner. Alternations in GRK2 levels and/or activity are demonstrated in an array of relevant cardiovascular, metabolic, inflammatory, or cancer pathologies. These changes are assumed to contribute to the onset and/or development of such pathologies. Thus, GRK2 may serve as a potentially interesting therapeutic target and those drugs targeted for GRK2 may constitute a novel therapeutic strategy for several intractable diseases, including sepsis.


Assuntos
Quinase 2 de Receptor Acoplado a Proteína G/metabolismo , Quinase 2 de Receptor Acoplado a Proteína G/genética , Humanos
18.
Crit Care Med ; 43(11): e508-20, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26468714

RESUMO

OBJECTIVES: The calcium sensitizer levosimendan is used in treatment of decompensated heart failure and may also exhibit anti-inflammatory properties. We examined whether treatment with levosimendan is substantially beneficial in mice with cecal ligation and puncture-induced polymicrobial sepsis, and its arbitration mechanism was explored in the mouse macrophage cell line RAW264.7. DESIGN: Laboratory and animal/cell research. SETTING: University research laboratory. SUBJECTS: BALB/c mice (8-10 wk old) and mouse macrophage cell line RAW264.7 cells. INTERVENTIONS: Levosimendan (0.5 µg/kg/min) was administered to mice through an osmotic pump that was implanted into the peritoneal cavity immediately following surgery. In RAW264.7 cells, levosimendan was added to the culture medium 30 minutes before lipopolysaccharide. MEASUREMENTS AND MAIN RESULTS: When levosimendan was continuously administered to cecal ligation and puncture-induced septic mice, a significant improvement of left ventricular function was found without any change in heart rate, and hypotension was significantly mitigated. Furthermore, levosimendan conferred substantial protection against sepsis-associated inflammation in mice, as indicated by reduced lung injury and decreased blood proinflammatory and chemotactic cytokine levels. These beneficial effects of levosimendan led to a significant improvement of survival in mice after cecal ligation and puncture. In endotoxin-stimulated RAW264.7 macrophages, treatment with levosimendan and pimobendan suppressed overproduction of proinflammatory and chemotactic cytokines. Levosimendan and pimobendan were without effect on activation of the nuclear factor-κB, mitogen-activated protein kinase, and Akt pathways. Instead, levosimendan and pimobendan prevented high mobility group box 1 release from the nucleus to the extracellular space in macrophages. This was associated with inhibition of the Rho kinase signaling pathway. The elevated serum high mobility group box 1 levels in cecal ligation and puncture-induced septic mice were also inhibited by continued administration of levosimendan and pimobendan. CONCLUSIONS: We define a novel mechanism for the anti-inflammatory action of levosimendan and suggest that the pharmacological profiles of levosimendan as both an inotrope and an anti-inflammatory agent could contribute to its clinical benefit in patients with sepsis with heart problems.


Assuntos
Anti-Inflamatórios/farmacologia , Citocinas/metabolismo , Hidrazonas/farmacologia , Macrófagos/efeitos dos fármacos , Piridazinas/farmacologia , Sepse/tratamento farmacológico , Sepse/patologia , Animais , Biópsia por Agulha , Western Blotting , Ceco/cirurgia , Células Cultivadas , Citocinas/efeitos dos fármacos , Modelos Animais de Doenças , Ecocardiografia/métodos , Ensaio de Imunoadsorção Enzimática , Imuno-Histoquímica , Injeções Intravenosas , Ligadura/métodos , Lipopolissacarídeos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Distribuição Aleatória , Reação em Cadeia da Polimerase em Tempo Real , Valores de Referência , Sepse/mortalidade , Simendana , Estatísticas não Paramétricas , Taxa de Sobrevida
19.
J Pharmacol Exp Ther ; 354(3): 240-50, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26136420

RESUMO

Macrophages are a well recognized player of both innate and adaptive immunity and have emerged as a key regulator of systemicmetabolism, hematopoiesis, vasculogenesis, apoptosis, malignancy, and reproduction. Such pleiotropic roles of macrophages are mirrored by their protean features. Upon environmental. challenges, macrophages redistribute and differentiate in situ and contribute to the multiple disease states by exerting protective and pathogenic effects. The environmental challenges include cytokines, chemokines, lipid mediators, and extrinsic insults, such as food and pathogenic bacteria. In addition, homeostasis and the activation state of macrophages are influenced by various metabolites from a commensal microbe that colonizes epithelial and mucosal surfaces, such as the lungs, intestines, and skin. In this review, we describe macrophage differentiation, polarization, and various functions in chronic disease states, including chronic inflammatory bowel disease, tumorigenesis, metabolism and obesity, and central nervous system demyelinating disorders. Controlling the macrophage dynamics to affect the pathologic states is considered to be an important therapeutic approach for many clinical disorders involving chronic inflammation.


Assuntos
Doença Crônica/terapia , Inflamação/patologia , Macrófagos/patologia , Animais , Diferenciação Celular/imunologia , Diferenciação Celular/fisiologia , Humanos , Inflamação/imunologia , Inflamação/metabolismo , Ativação de Macrófagos/imunologia , Ativação de Macrófagos/fisiologia , Macrófagos/imunologia , Macrófagos/metabolismo
20.
Eur J Pharmacol ; 761: 44-54, 2015 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-25934567

RESUMO

Direct antitumor effects of bisphosphonates (BPs) have been demonstrated in various cancer cells in vitro. However, the effective concentrations of BPs are typically much higher than their clinically relevant concentrations. Oral cancers frequently invade jawbone and may lead to the release of Ca(2+) in primary lesions. We investigated the effects of the combined application of zoledronic acid (ZA) and Ca(2+) on proliferation and apoptosis of oral cancer cells. Human oral cancer cells, breast cancer cells, and colon cancer cells were treated with ZA at a wide range of concentrations in different Ca(2+) concentration environments. Under a standard Ca(2+) concentration (0.6mM), micromolar concentrations of ZA were required to inhibit oral cancer cell proliferation. Increasing extracellular Ca(2+) concentrations greatly enhanced the potency of the ZA cytocidal effect. The ability of Ca(2+) to enhance the cytocidal effects of ZA was negated by the Ca(2+)-selective chelator EGTA. In contrast, the cytocidal effect of ZA was less pronounced in breast and colon cancer cells regardless of whether extracellular Ca(2+) was elevated. In oral cancer cells incubated with 1.6mM Ca(2+), ZA up-regulated mitochondrial Bax expression and increased mitochondrial Ca(2+) uptake. This was associated with decreased mitochondrial membrane potential and increased release of cytochrome c. We suggest that ZA can specifically produce potent cytocidal activity in oral cancer cells in an extracellular Ca(2+)-dependent manner, implying that BPs may be useful for treatment of oral squamous cell carcinoma with jawbone invasion leading to the hypercalcemic state.


Assuntos
Antineoplásicos/farmacologia , Cálcio/farmacologia , Carcinoma de Células Escamosas/tratamento farmacológico , Difosfonatos/farmacologia , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Imidazóis/farmacologia , Neoplasias Bucais/tratamento farmacológico , Apoptose/efeitos dos fármacos , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Cálcio/metabolismo , Quelantes de Cálcio/farmacologia , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Citocromos c/metabolismo , Relação Dose-Resposta a Droga , Neoplasias de Cabeça e Pescoço/metabolismo , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Células MCF-7 , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Neoplasias Bucais/metabolismo , Neoplasias Bucais/patologia , Carcinoma de Células Escamosas de Cabeça e Pescoço , Ácido Zoledrônico , Proteína X Associada a bcl-2/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA