Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Chin Med Sci J ; 32(4): 253-259, 2017 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-29301601

RESUMO

Hypoxic pulmonary hypertension (HPH) is a common complication in patients with chronic obstructive pulmonary disease (COPD), sleep-disordered breathing, or dwellers in high altitude. The exact mechanisms underlying the development of HPH still remain unclear. Reactive oxygen species (ROS), hypoxia inducible factors (HIF), and potassium channels (KV) are believed as the main factors during the development of HPH. We propose that the "ROS/Kv/HIF axis" may play an important initiating role in the development of HPH. Being formed under a hypoxic condition, ROS affects the expression and function of HIFs or KV, and consequently triggers multiple downstream signaling pathways and genes expression that participate in promoting pulmonary vasoconstriction and arterial remodeling. Thus, further study determining the initiating role of "ROS/Kv/HIF axis" in the development of HPH could provide theoretic evidences to better understand the underlying mechanisms of HPH, and help identify new potential targets in the treatment of HPH.


Assuntos
Hipertensão Pulmonar , Fator 1 Induzível por Hipóxia/metabolismo , Hipóxia , Canais de Potássio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Animais , Humanos , Hipertensão Pulmonar/etiologia , Hipertensão Pulmonar/metabolismo , Hipertensão Pulmonar/patologia , Hipertensão Pulmonar/fisiopatologia , Hipóxia/complicações , Hipóxia/metabolismo , Hipóxia/patologia , Hipóxia/fisiopatologia
2.
Int J Med Sci ; 13(12): 942-954, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27994500

RESUMO

Resveratrol, a plant-derived polyphenolic compound and a phytoestrogen, was shown to possess multiple protective effects including anti-inflammatory response and anti-oxidative stress. Hypoxic pulmonary hypertension (HPH) is a progressive disease characterized by sustained vascular resistance and marked pulmonary vascular remodeling. The exact mechanisms of HPH are still unclear, but inflammatory response and oxidative stress was demonstrated to participate in the progression of HPH. The present study was designed to investigate the effects of resveratrol on HPH development. Sprague-Dawley rats were challenged by hypoxia exposure for 28 days to mimic hypoxic pulmonary hypertension along with treating resveratrol (40 mg/kg/day). Hemodynamic and pulmonary pathomorphology data were then obtained, and the anti-proliferation effect of resveratrol was determined by in vitro assays. The anti-inflammation and anti-oxidative effects of resveratrol were investigated in vivo and in vitro. The present study showed that resveratrol treatment alleviated right ventricular systolic pressure and pulmonary arterial remodeling induced by hypoxia. In vitro experiments showed that resveratrol notably inhibited proliferation of pulmonary arterial smooth muscle cells in an ER-independent manner. Data showed that resveratrol administration inhibited HIF-1 α expression in vivo and in vitro, suppressed inflammatory cells infiltration around the pulmonary arteries, and decreased ROS production induced by hypoxia in PAMSCs. The inflammatory cytokines' mRNA levels of tumor necrosis factor α, interleukin 6, and interleukin 1ß were all suppressed by resveratrol treatment. The in vitro assays showed that resveratrol inhibited the expression of HIF-1 α via suppressing the MAPK/ERK1 and PI3K/AKT pathways. The antioxidant axis of Nuclear factor erythroid-2 related factor 2/ Thioredoxin 1 (Nrf-2/Trx-1) was up-regulated both in lung tissues and in cultured PASMCs. In general, the current study demonstrated that resveratrol may prevent pulmonary hypertension through its anti-proliferation, anti-inflammation and antioxidant effects. Hence, the present data may offer novel targets and promising pharmacological perspective for treating hypoxic pulmonary hypertension.


Assuntos
Antioxidantes/uso terapêutico , Hipertensão Pulmonar/tratamento farmacológico , Hipertensão Pulmonar/metabolismo , Estilbenos/uso terapêutico , Animais , Hipóxia/tratamento farmacológico , Hipóxia/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Distribuição Aleatória , Ratos , Espécies Reativas de Oxigênio/metabolismo , Resveratrol , Tiorredoxinas/metabolismo
3.
Mol Med Rep ; 14(6): 5033-5040, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27779669

RESUMO

Nuclear factor of activated T cells 5 (NFAT5) is a transcription factor that can be activated by extracellular tonicity. It has been reported that NFAT5 may increase the transcription of certain osmoprotective genes in the renal system, and the aim of the current study was to explore the role of NFAT5 in seawater inhalation­induced acute lung injury. Though establishing the model of seawater inhalation­induced acute lung injury, it was demonstrated that seawater inhalation enhanced the transcription and protein expression of NFAT5 (evaluated by reverse transcription­polymerase chain reaction, immunohistochemistry stain and western blotting) and activation of nuclear factor (NF)­κB (evaluated by western blotting and mRNA expression levels of three NF­κB­dependent genes) both in lung tissue and rat alveolar macrophage cells (NR8383 cells). When expression of NFAT5 was reduced in NR8383 cells using an siRNA targeted to NFAT5, the phosphorylation of NF­κB and transcription of NF­κB­dependent genes were significantly reduced. In addition, the elevated content of certain inflammatory cytokines [tumor necrosis factor α, interleukin (IL)­1 and IL­8] were markedly reduced. In conclusion, NFAT5 serves an important pathophysiological role in seawater inhalation­induced acute lung injury by modulating NF­κB activity, and these data suggest that NFAT5 may be a promising therapeutic target.


Assuntos
Lesão Pulmonar Aguda/etiologia , Lesão Pulmonar Aguda/metabolismo , NF-kappa B/metabolismo , Fatores de Transcrição NFATC/metabolismo , Água do Mar/efeitos adversos , Lesão Pulmonar Aguda/patologia , Animais , Biomarcadores , Biópsia , Linhagem Celular , Citocinas/metabolismo , Modelos Animais de Doenças , Ativação Enzimática , Expressão Gênica , Regulação da Expressão Gênica , Imuno-Histoquímica , Mediadores da Inflamação/metabolismo , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/metabolismo , Masculino , Fatores de Transcrição NFATC/genética , RNA Interferente Pequeno/genética , Ratos
4.
Mol Med Rep ; 13(3): 1989-98, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26796050

RESUMO

Calcium is an important second messenger and it is widely recognized that acute lung injury (ALI) is often caused by oscillations of cytosolic free Ca2+. Previous studies have indicated that the activation of transient receptor potential­vanilloid (TRPV) channels and subsequent Ca2+ entry initiates an acute calcium­dependent permeability increase during ALI. However, whether seawater exposure induces such an effect through the activation of TRPV channels remains unknown. In the current study, the effect of calcium, a component of seawater, on the inflammatory reactions that occur during seawater drowning­induced ALI, was examined. The results demonstrated that a high concentration of calcium ions in seawater increased lung tissue myeloperoxidase activity and the secretion of inflammatory mediators, such as tumor necrosis factor­α (TNF­α) and interleukin (IL)­1ß and IL­6. Further study demonstrated that the seawater challenge elevated cytosolic Ca2+ concentration, indicated by [Ca2+]c, by inducing calcium influx from the extracellular medium via TRPV1 channels. The elevated [Ca2+c] may have resulted in the increased release of TNF­α and IL­1ß via increased phosphorylation of nuclear factor­κB (NF­κB). It was concluded that a high concentration of calcium in seawater exacerbated lung injury, and TRPV1 channels were notable mediators of the calcium increase initiated by the seawater challenge. Calcium influx through TRPV1 may have led to greater phosphorylation of NF­κB and increased release of TNF­α and IL­1ß.


Assuntos
Canais de Cátion TRPV/metabolismo , Citoesqueleto de Actina/efeitos dos fármacos , Citoesqueleto de Actina/metabolismo , Lesão Pulmonar Aguda , Administração por Inalação , Animais , Cálcio/metabolismo , Quelantes de Cálcio/farmacologia , Sinalização do Cálcio , Capsaicina/análogos & derivados , Capsaicina/farmacologia , Linhagem Celular Tumoral , Citosol/metabolismo , Progressão da Doença , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Espaço Extracelular/metabolismo , Fluorescência , Humanos , Mediadores da Inflamação/metabolismo , Interleucina-1beta/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/patologia , Masculino , NF-kappa B/metabolismo , Fosforilação , Prolina/análogos & derivados , Prolina/farmacologia , Ratos Sprague-Dawley , Água do Mar , Tiocarbamatos/farmacologia , Fator de Necrose Tumoral alfa/metabolismo
5.
Int J Med Sci ; 11(6): 578-86, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24782646

RESUMO

Pulmonary fibrosis (PF) is a common complication in those interstitial lung diseases patients, which will result in poor prognosis and short survival. Traditional therapeutic methods such as glucocorticoid and cytotoxic drugs are insufficient for treating PF and may cause severe side effects. Recent studies showed that traditional Chinese herbal abstraction such as Tanshinone IIA (TIIA) was displayed significant anti-PF effects in animal models. However, the exact mechanisms underlying the protective effects of TIIA were not fully understood. Here we further investigated the protective effects of TIIA and its mechanisms underlying. PF models of rat were induced by bleomycin (BLM); TIIA was administered subsequently. The PF changes were identified by histopathological analyses. The results showed that BLM resulted in severe PF and alveolar inflammation; together with significant elevation of transforming growth factor-ß 1 (TGF-ß1). Angiotensin-converting enzyme 2 (ACE-2) together with angiotensin-(1-7) [ANG-(1-7)] were both greatly reduced after BLM administration. TIIA treatment notably attenuated BLM induced PF and inflammation, decreased expression of TGF-ß1 and reversed ACE-2 and ANG-(1-7) production in rat lungs. Thus we may draw the conclusion that TIIA may exert protective effects on BLM induced PF in rats, and the ACE-2/ANG-(1-7) axis may ascribe to those protective effects.


Assuntos
Abietanos/administração & dosagem , Angiotensina I/metabolismo , Fragmentos de Peptídeos/metabolismo , Peptidil Dipeptidase A/metabolismo , Fibrose Pulmonar/tratamento farmacológico , Enzima de Conversão de Angiotensina 2 , Animais , Bleomicina/toxicidade , Humanos , Inflamação/induzido quimicamente , Inflamação/tratamento farmacológico , Inflamação/patologia , Fibrose Pulmonar/induzido quimicamente , Ratos , Fator de Crescimento Transformador beta/metabolismo
6.
Free Radic Biol Med ; 69: 198-207, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24440469

RESUMO

Pulmonary hypertension is a progressive disease characterized by marked pulmonary arterial remodeling and increased vascular resistance. Inflammation and oxidative stress promote the development of pulmonary hypertension. Oxymatrine, one of the main active components of the Chinese herb Sophora flavescens Ait. (Kushen), plays anti-inflammatory and antioxidant protective roles, which effects on pulmonary arteries remain unclear. This study aimed to investigate the effects of oxymatrine on pulmonary hypertension development. Sprague-Dawley rats were exposed to hypoxia for 28 days or injected with monocrotaline, to develop pulmonary hypertension, along with administration of oxymatrine (50mg/kg/day). Hemodynamics and pulmonary arterial remodeling data from the rats were then obtained. The antiproliferative effect of oxymatrine was verified by in vitro assays. The inflammatory cytokine mRNA levels and leukocyte and T cell accumulation in lung tissue were detected. The antioxidative effects of oxymatrine were explored in vitro. Our study shows that oxymatrine treatment attenuated right-ventricular systolic pressure and pulmonary arterial remodeling induced by hypoxia or monocrotaline and inhibited proliferation of pulmonary arterial smooth muscle cells (PASMCs). Increased expression of inflammatory cytokine mRNA and accumulation of leukocytes and T cells around the pulmonary arteries were suppressed with oxymatrine administration. Under hypoxic conditions, oxymatrine significantly upregulated Nrf2 and antioxidant protein SOD1 and HO-1 expression, but downregulated hydroperoxide levels in PASMCs. In summary, this study indicates that oxymatrine may prevent pulmonary hypertension through its antiproliferative, anti-inflammatory, and antioxidant effects, thus providing a promising pharmacological avenue for treating pulmonary hypertension.


Assuntos
Alcaloides , Hipertensão Pulmonar , Hipóxia , Miócitos de Músculo Liso , Quinolizinas , Animais , Humanos , Ratos , Alcaloides/administração & dosagem , Proliferação de Células/efeitos dos fármacos , Heme Oxigenase-1/biossíntese , Hipertensão Pulmonar/induzido quimicamente , Hipertensão Pulmonar/tratamento farmacológico , Hipertensão Pulmonar/patologia , Hipóxia/tratamento farmacológico , Hipóxia/patologia , Monocrotalina/toxicidade , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Artéria Pulmonar/metabolismo , Quinolizinas/administração & dosagem , Superóxido Dismutase/biossíntese , Superóxido Dismutase-1 , Fator 2 Relacionado a NF-E2/biossíntese
7.
Acta Pharmacol Sin ; 34(12): 1515-25, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24122010

RESUMO

AIM: To investigate the protective effects of hydrogen sulfide (H2S) against inflammation, oxidative stress and apoptosis in a rat model of resuscitated hemorrhagic shock. METHODS: Hemorrhagic shock was induced in adult male SD rats by drawing blood from the femoral artery for 10 min. The mean arterial pressure was maintained at 35-40 mmHg for 1.5 h. After resuscitation the animals were observed for 200 min, and then killed. The lungs were harvested and bronchoalveolar lavage fluid was prepared. The levels of relevant proteins were examined using Western blotting and immunohistochemical analyses. NaHS (28 µmol/kg, ip) was injected before the resuscitation. RESULTS: Resuscitated hemorrhagic shock induced lung inflammatory responses and significantly increased the levels of inflammatory cytokines IL-6, TNF-α, and HMGB1 in bronchoalveolar lavage fluid. Furthermore, resuscitated hemorrhagic shock caused marked oxidative stress in lung tissue as shown by significant increases in the production of reactive oxygen species H2O2 and ·OH, the translocation of Nrf2, an important regulator of antioxidant expression, into nucleus, and the decrease of thioredoxin 1 expression. Moreover, resuscitated hemorrhagic shock markedly increased the expression of death receptor Fas and Fas-ligand and the number apoptotic cells in lung tissue, as well as the expression of pro-apoptotic proteins FADD, active-caspase 3, active-caspase 8, Bax, and decreased the expression of Bcl-2. Injection with NaHS significantly attenuated these pathophysiological abnormalities induced by the resuscitated hemorrhagic shock. CONCLUSION: NaHS administration protects rat lungs against inflammatory responses induced by resuscitated hemorrhagic shock via suppressing oxidative stress and the Fas/FasL apoptotic signaling pathway.


Assuntos
Apoptose/efeitos dos fármacos , Pneumonia/prevenção & controle , Ressuscitação , Choque Hemorrágico/complicações , Sulfetos/farmacologia , Animais , Líquido da Lavagem Broncoalveolar , Masculino , Pneumonia/complicações , Ratos , Ratos Sprague-Dawley
8.
Am J Respir Cell Mol Biol ; 49(6): 1057-63, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23855826

RESUMO

Leptin is reported to be involved in acute lung injury (ALI). However, the role and underlying mechanisms of leptin in ALI remain unclear. The aim of this study was to determine whether leptin deficiency promoted the development of ALI. LPS or oleic acid (OA) were administered to wild-type and leptin deficient (ob/ob) mice to induce ALI. Leptin level, survival rate, and lung injury were examined. Results showed that leptin levels were predominantly increased in the lung, but also in the heart, liver, kidney, and adipose tissue after LPS adminiatration. Compared with wild-type mice, LPS- or OA-induced lung injury was worse and the survival rate was lower in ob/ob mice. Moreover, leptin deficiency promoted the release of proinflammatory cytokines. Exogenous administration of leptin reduced lethality in ob/ob mice and ameliorated lung injury partly through inhibiting the activation of NF-κB, p38, and ERK pathways. These results indicated that leptin deficiency contributed to the development of lung injury by enhancing inflammatory response, and a high level of leptin improved survival and protected against ALI.


Assuntos
Lesão Pulmonar Aguda/etiologia , Lesão Pulmonar Aguda/prevenção & controle , Leptina/fisiologia , Lipopolissacarídeos/toxicidade , Ácido Oleico/toxicidade , Lesão Pulmonar Aguda/fisiopatologia , Animais , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Mediadores da Inflamação/metabolismo , Leptina/deficiência , Leptina/genética , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , NF-kappa B/metabolismo , Regulação para Cima/efeitos dos fármacos
9.
Int J Med Sci ; 10(6): 771-81, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23630443

RESUMO

Exogenous estrogen was shown to exert various beneficial effects on multiple diseases including hypoxia-induced pulmonary hypertension (HPH). However, the effect of endogenous estrogen on HPH was seldom investigated. In the present study, we explored the protective effects and mechanisms of endogenous estrogen on hypoxia-induced pulmonary hypertension. Male, female, pregnant and ovariectomized rats were housed in a hypoxic condition for 21 days, and then hemodynamic together with morphologic indexes of pulmonary circulation were measured. The right ventricular systolic pressure, mean pulmonary artery pressure, right ventricular hypertrophy index, and arterial remodeling index were significantly elevated after chronic hypoxia exposure. Experimental data showed less severity in female, especially in pregnant rats. In vitro, artery rings of different sex or estrus cycle rats were obtained, and then artery rings experiments were performed to investigate pulmonary vasoconstriction by recording the maximum phase II vasoconstriction. Data showed that the vasoconstriction was milder in proestrus female than diestrus female or male groups, which could be leveled by treating U0126 (a MAPK pathway inhibitor). Pulmonary arterial smooth muscle cells isolated from different sex or estrus cycle rats were cultured in the condition of 2% oxygen for 24 hours, and cell proliferation was evaluated by the [3H]-thymidine incorporation assay. Cells from proestrus rats exhibited lower proliferation than the other groups, which could be countered by both U0126 and raloxifene (a selective estrogen receptor modulator). Serum estradiol levels were detected, and rats with higher levels showed less severity of pulmonary hypertension. Conclusively, endogenous estrogen may alleviate hypoxia-induced pulmonary hypertension by attenuating vasoconstriction through non-genomic mechanisms and inhibiting smooth muscle cells proliferation through both genomic and non-genomic mechanisms.


Assuntos
Estrogênios/administração & dosagem , Hipertensão Pulmonar/patologia , Miócitos de Músculo Liso/efeitos dos fármacos , Artéria Pulmonar/crescimento & desenvolvimento , Animais , Proliferação de Células/efeitos dos fármacos , Estradiol/sangue , Feminino , Humanos , Hipertensão Pulmonar/tratamento farmacológico , Hipóxia/patologia , Masculino , Ovariectomia , Gravidez , Artéria Pulmonar/citologia , Artéria Pulmonar/efeitos dos fármacos , Ratos , Vasoconstrição/efeitos dos fármacos
10.
Respir Physiol Neurobiol ; 188(2): 214-22, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23623946

RESUMO

Inhibiting reactive oxygen species (ROS) has been viewed as a therapeutic target for the treatment of acute lung injury (ALI). Osthole, an active component in Chinese herbal medicine, has drawn increasing attention because of its various pharmacological functions, including anti-inflammatory and anti-oxidative activities. The aim of the present study was to examine the effects of osthole on ALI induced by lipopolysaccharide (LPS) through intratracheal instillation. The mRNA and protein expression levels of thioredoxin 1 (Trx1) and the nuclear factor erythroid-2 related factor 2 (Nrf2) were detected by real-time PCR, reverse transcription PCR (RT-PCR) and Western blot, respectively. ROS production was measured by flow cytometry. Our results showed that osthole treatment improved the mice survival rates in the middle and high dosage groups, compared with the untreated LPS group. Moreover, osthole treatment significantly improved LPS-induced lung pathological damage, and it decreased the lung injury scores, lung wet/dry ratios and the total protein level in Bronchoalveolar lavage fluid (BALF). Osthole treatment dramatically reduced the H2O2, MDA and OH levels in the lung homogenates. LDH and ROS were markedly reduced in the osthole+LPS group in vitro. Furthermore, osthole increased Nrf2 and Trx1 expression in terms of mRNA and protein in vivo and in vitro. Nrf2 siRNA (siNrf2) could suppress the beneficial effects of osthole on ALI. In conclusion, the current study demonstrates that osthole exerted protective effects on LPS-induced ALI by up-regulating the Nrf-2/Trx-1 pathway.


Assuntos
Lesão Pulmonar Aguda/tratamento farmacológico , Bloqueadores dos Canais de Cálcio/uso terapêutico , Cumarínicos/uso terapêutico , Fator 2 Relacionado a NF-E2/metabolismo , Tiorredoxinas/metabolismo , Regulação para Cima/efeitos dos fármacos , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/metabolismo , Lesão Pulmonar Aguda/mortalidade , Animais , Líquido da Lavagem Broncoalveolar , Bloqueadores dos Canais de Cálcio/farmacologia , Linhagem Celular Transformada , Sobrevivência Celular/efeitos dos fármacos , Cumarínicos/farmacologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Lipopolissacarídeos/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Fator 2 Relacionado a NF-E2/genética , Tamanho do Órgão/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Estatísticas não Paramétricas , Análise de Sobrevida , Tiorredoxinas/genética , Fatores de Tempo , Transfecção
11.
PLoS One ; 8(2): e56774, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23437233

RESUMO

We previously showed that tanshinone IIA ameliorated the hypoxia-induced pulmonary hypertension (HPH) partially by attenuating pulmonary artery remodeling. The hypoxia-induced proliferation of pulmonary artery smooth muscle cells (PASMCs) is one of the major causes for pulmonary arterial remodeling, therefore the present study was performed to explore the effects and underlying mechanism of tanshinone IIA on the hypoxia-induced PASMCs proliferation. PASMCs were isolated from male Sprague-Dawley rats and cultured in normoxic (21%) or hypoxic (3%) condition. Cell proliferation was measured with 3 - (4, 5 - dimethylthiazal - 2 - yl) - 2, 5 - diphenyltetrazoliumbromide assay and cell counting. Cell cycle was measured with flow cytometry. The expression of of p27, Skp-2 and the phosphorylation of Akt were measured using western blot and/or RT-PCR respectively. The results showed that tanshinone IIA significantly inhibited the hypoxia-induced PASMCs proliferation in a concentration-dependent manner and arrested the cells in G1/G0-phase. Tanshinone IIA reversed the hypoxia-induced reduction of p27 protein, a cyclin-dependent kinase inhibitor, in PASMCs by slowing down its degradation. Knockdown of p27 with specific siRNA abolished the anti-proliferation of tanshinone IIA. Moreover, tanshinone IIA inhibited the hypoxia-induced increase of S-phase kinase-associated protein 2 (Skp2) and the phosphorylation of Akt, both of which are involved in the degradation of p27 protein. In vivo tanshinone IIA significantly upregulated the hypoxia-induced p27 protein reduction and downregulated the hypoxia-induced Skp2 increase in pulmonary arteries in HPH rats. Therefore, we propose that the inhibition of tanshinone IIA on hypoxia-induce PASMCs proliferation may be due to arresting the cells in G1/G0-phase by slowing down the hypoxia-induced degradation of p27 via Akt/Skp2-associated pathway. The novel information partially explained the anti-remodeling property of tanshinone IIA on pulmonary artery in HPH.


Assuntos
Abietanos/farmacologia , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Artéria Pulmonar/efeitos dos fármacos , Artéria Pulmonar/metabolismo , Proteínas Quinases Associadas a Fase S/metabolismo , Animais , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Hipóxia Celular , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p27/genética , Hipertensão Pulmonar/genética , Hipertensão Pulmonar/metabolismo , Masculino , Modelos Biológicos , Fosforilação/efeitos dos fármacos , Proteólise , Interferência de RNA , Ratos , Transdução de Sinais/efeitos dos fármacos
12.
Eur J Pharm Sci ; 48(4-5): 819-24, 2013 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-23321685

RESUMO

The renin-angiotensin-aldosterone system (RAAS) plays an important role in the pathogenesis of acute lung injury (ALI)/acute respiratory distress syndrome (ARDS). Angiotensin converting enzyme 2 (ACE2) plays a protective role in acute lung injury. Osthole, a natural coumarin derivative extracted from traditional Chinese medicines, is known to have anti-inflammatory effect, but the effect of osthole on the ALI is largely unknown. The aim of this study is to explore whether and by what mechanisms osthole protects lipopolysaccharide(LPS)-induced acute lung injury. Herein, we found that osthole had a beneficial effect on LPS-induced ALI in mice. As revealed by survival study, pretreatment with high doses of osthole reduced the mortality of mice from ALI. Osthole pretreatment significantly improved LPS-induced lung pathological changes, reduced lung wet/dry weight ratios and total protein in BALF. Osthole also inhibited the release of inflammatory mediators TNF-α and IL-6. Meanwhile, osthole markedly prevented the loss of ACE2 and Ang1-7 in lung tissue of ALI mice. ACE2 inhibitor blocked the protective effect of osthole in NR 8383 cell lines. Taken together, our study showed that osthole improved survival rate and attenuated LPS-induced ALI and ACE2 may play a role in it.


Assuntos
Lesão Pulmonar Aguda/tratamento farmacológico , Anti-Inflamatórios/uso terapêutico , Cumarínicos/uso terapêutico , Peptidil Dipeptidase A/metabolismo , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/metabolismo , Lesão Pulmonar Aguda/patologia , Angiotensina I/metabolismo , Enzima de Conversão de Angiotensina 2 , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Animais , Anti-Inflamatórios/farmacologia , Líquido da Lavagem Broncoalveolar/química , Linhagem Celular , Cumarínicos/farmacologia , Regulação para Baixo/efeitos dos fármacos , Interleucina-6/metabolismo , Lipopolissacarídeos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Fragmentos de Peptídeos/metabolismo , Peptidil Dipeptidase A/genética , RNA Mensageiro/metabolismo , Ratos , Fator de Necrose Tumoral alfa/metabolismo
13.
Pulm Pharmacol Ther ; 25(6): 472-7, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22982561

RESUMO

Insulin is a main glucose homeostatic hormone in the body. Previous reports showed that insulin also exerted anti-inflammatory actions and attenuated systemic inflammatory response. Here, we observed the effects and the underlying mechanisms of insulin on lipopolysaccharide (LPS)-induced acute lung injury (ALI). As revealed by survival study, insulin reduced mortality of rats and prolonged their survival time. Meanwhile, insulin significantly reduced the levels of inflammatory cytokines including tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß), interleukin-6 (IL-6), and high mobility group box 1 (HMGB1) in bronchoalveolar lavage fluid (BALF). Besides, insulin markedly inhibited the expression of toll-like receptor 2 (TLR2), toll-like receptor 4 (TLR4) and nuclear factor-κB (NF-κB). Taken together, these data provided information that insulin attenuated LPS-induced ALI may attribute partly to the inhibition of the production of cytokines, and the expression of TLR2, TLR4 and NF-κB.


Assuntos
Lesão Pulmonar Aguda/tratamento farmacológico , Anti-Inflamatórios/farmacologia , Inflamação/tratamento farmacológico , Insulina/farmacologia , Lesão Pulmonar Aguda/patologia , Animais , Líquido da Lavagem Broncoalveolar , Citocinas/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Inflamação/patologia , Lipopolissacarídeos/toxicidade , Masculino , NF-kappa B/genética , Ratos , Ratos Sprague-Dawley , Taxa de Sobrevida , Receptor 2 Toll-Like/genética , Receptor 4 Toll-Like/genética
14.
Biol Pharm Bull ; 35(7): 1029-34, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22791148

RESUMO

Hemorrhagic shock (HS) is a common condition and leading cause of death in trauma patients universally. Severe inflammatory responses during HS finally lead to multiple-organ failure. Hydrogen sulphide (H2S) is increasingly recognized as an important signaling molecule with various protective effects. In the present study, we investigated the antiinflammatory and cardioprotective effects of an exogenous H2S donor, sodium hydrosulfide (NaHS), in an HS rat model. Male Sprague-Dawley rats were randomly divided into the sham-operated, sham-operated treated with NaHS (28 µmol/kg, intraperitoneally (i.p.)), HS, and HS treated with NaHS (28 µmol/kg, i.p.) groups. The HS groups were subjected to mimicked HS for 1 h and then treated with NaHS or left untreated. The rats were then resuscitated with Ringer lactate solution for 1 h. Myocardial enzymes and inflammatory cytokines were evaluated. Morphologic changes in cardiac tissue and ultrastructural injury were also analyzed. HS resulted in significant hemodynamic deterioration and increased myocardial enzyme and inflammatory cytokine levels. Intraperitoneal administration of NaHS significantly prevented hemodynamic deterioration and decreased the elevation of myocardial enzymes. NaHS also inhibited the nuclear factor κB inhibitor kinase (IKK)/nuclear factor κB inhibitor (IκB)/nuclear factor κB (NF-κB) signaling pathway. The results suggest that NaHS exerts cardioprotective effects against HS. The protective effects of NaHS may occur via down-regulation of the IKK/IκB/NF-κB signaling pathway.


Assuntos
Cardiotônicos/farmacologia , Quinase I-kappa B/antagonistas & inibidores , Proteínas I-kappa B/antagonistas & inibidores , NF-kappa B/antagonistas & inibidores , Choque Hemorrágico/metabolismo , Sulfetos/farmacologia , Animais , Creatina Quinase/sangue , Modelos Animais de Doenças , Sulfeto de Hidrogênio/metabolismo , Quinase I-kappa B/metabolismo , Proteínas I-kappa B/metabolismo , Interleucina-6/sangue , L-Lactato Desidrogenase/sangue , Masculino , NF-kappa B/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/sangue
15.
Respir Physiol Neurobiol ; 182(2-3): 71-80, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22609371

RESUMO

Our previous study showed that seawater can cause lung tissue cell apoptosis; in the present study, the immunohistochemistry and Western blot analysis results demonstrated that Fas, FasL, and cleaved caspase-8 and caspase-3 were up-regulated in the rat lungs exposed to seawater. We found that seawater-induced human lung alveolar epithelial A549 cell apoptosis was concentration and time dependent. Moreover, seawater increased the expression of Fas, FasL, and cleaved caspase-8 and caspase-3 in A549 cells. The incubation of A549 cells in the presence of FasL-neutralising antibody (NOK-2) or caspase-8 inhibitor (Z-IETD-FMK) resulted in a decrease of seawater-induced cell apoptosis. NOK-2 inhibited Fas/FasL interaction and reduced the cleavage of caspase-8 and caspase-3, and Z-IETD-FMK blocked caspase-8 and caspase-3 activation. Seawater similarly produced a significant increase in rat alveolar type II cell apoptosis and expression of Fas and cleaved caspase-8. In summary, the Fas/FasL pathway involved in alveolar epithelial cell (AEC) apoptosis could be important in the pathogenesis of seawater-induced acute lung injury (SW-ALI).


Assuntos
Apoptose/fisiologia , Células Epiteliais/metabolismo , Proteína Ligante Fas/metabolismo , Alvéolos Pulmonares/metabolismo , Água do Mar/efeitos adversos , Receptor fas/metabolismo , Animais , Western Blotting , Caspase 3/metabolismo , Caspase 8/metabolismo , Afogamento/metabolismo , Afogamento/fisiopatologia , Humanos , Imuno-Histoquímica , Ratos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
16.
Mediators Inflamm ; 2012: 840737, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22363104

RESUMO

Pulmonary hypertension (PH) contributes to the mortality of patients with lung and heart diseases. However, the underlying mechanism has not been completely elucidated. Accumulating evidence suggests that inflammatory response may be involved in the pathogenesis of PH. Macrophage migration inhibitory factor (MIF) is a critical upstream inflammatory mediator which promotes a broad range of pathophysiological processes. The aim of the study was to investigate the role of MIF in the pulmonary vascular remodeling of hypoxia-induced PH. We found that MIF mRNA and protein expression was increased in the lung tissues from hypoxic pulmonary hypertensive rats. Intensive immunoreactivity for MIF was observed in smooth muscle cells of large pulmonary arteries (PAs), endothelial cells of small PAs, and inflammatory cells of hypoxic lungs. MIF participated in the hypoxia-induced PASMCs proliferation, and it could directly stimulate proliferation of these cells. MIF-induced enhanced growth of PASMCs was attenuated by MEK and JNK inhibitor. Besides, MIF antagonist ISO-1 suppressed the ERK1/2 and JNK phosphorylation induced by MIF. In conclusion, the current finding suggested that MIF may act on the proliferation of PASMCs through the activation of the ERK1/2 and JNK pathways, which contributes to hypoxic pulmonary hypertension.


Assuntos
Hipertensão Pulmonar/metabolismo , Fatores Inibidores da Migração de Macrófagos/metabolismo , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/metabolismo , Animais , Apoptose/genética , Apoptose/fisiologia , Proliferação de Células , Humanos , Fatores Inibidores da Migração de Macrófagos/genética
17.
Acta Histochem ; 114(4): 311-7, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21782216

RESUMO

PerClot(®) is a hemostatic material made of polysaccharide from modified starch and has been shown to assist in topical hemostasis. The principal goal in treating surgical and non-surgical wounds is the need for rapid closure of the lesion. This study investigated whether topical application of PerClot(®) could improve impaired wound healing in Sprague-Dawley (SD) rats. Full-thickness skin wounds were created on the back of the rats. Immediately, PerClot(®) was introduced into the wound bed, while wounds receiving starch or nothing served as controls. Wound closure was monitored using well-recognized wound-healing parameters: histological examination for inflammatory cells and fibroblast infiltration, newly formed capillaries, and collagen deposition. Meanwhile, transforming growth factor (TGF-ß1) was measured by immunochemistry. Wound closure was significantly accelerated by local application of PerClot(®). Furthermore, PerClot(®)-treated wounds showed significantly increased fibroblast numbers at 5 days post-wounding, and newly formed capillaries at 7 days post-wounding, and collagen regeneration at 7 and 14 days post-wounding. The number of infiltrating fibroblasts expressing TGF-ß1 was significantly higher than that in the controls at 7 and 14 days post-wounding. PerClot(®) can improve the wound healing and this effect might involve an increase in the activity of fibroblasts and increased release of TGF-ß1.


Assuntos
Hemostáticos/uso terapêutico , Pele , Cicatrização/efeitos dos fármacos , Ferimentos Penetrantes/tratamento farmacológico , Administração Tópica , Animais , Capilares/fisiologia , Contagem de Células , Movimento Celular/efeitos dos fármacos , Colágeno/biossíntese , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Hemostasia/efeitos dos fármacos , Hemostasia/fisiologia , Hemostáticos/administração & dosagem , Masculino , Neovascularização Fisiológica , Ratos , Ratos Sprague-Dawley , Pele/irrigação sanguínea , Pele/efeitos dos fármacos , Pele/lesões , Fator de Crescimento Transformador beta1/biossíntese , Cicatrização/fisiologia , Ferimentos Penetrantes/patologia
18.
Microvasc Res ; 83(2): 205-12, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22005047

RESUMO

BACKGROUND: Hypoxic pulmonary vasoconstriction may lead to pulmonary hypertension, but the underlying mechanisms of persistent vasoconstriction are still unclear. There is evidence that pulmonary inflammation contributes to the abnormalities of function in the pulmonary artery (PA) following chronic hypoxia exposure. Macrophage migration inhibitory factor (MIF) is an important pro-inflammatory cytokine, and we found that expression of MIF was increased in the smooth muscle of PA from hypoxic pulmonary hypertensive rats. Therefore, the aim of the study was to investigate the role of MIF in modulating vasoreactivity of isolated PA rings. METHODS: Sprague-Dawley rats were challenged by intermittent chronic hypoxia exposure for 4 weeks to establish hypoxic pulmonary hypertension models. Subsequently, immunohistochemistry and western blot assay were used to examine the MIF expression in pulmonary artery. Moreover, isometric force displacement was measured in isolated intrapulmonary artery. RESULTS: In the isolated PA, our results showed that MIF mediated the enhanced pulmonary arterial vasoconstriction in response to chronic hypoxia, and the delayed hypoxic constriction in a biphasic pattern of constriction occurs in response to acute hypoxia. We also present the finding that MIF had no effect on force on its own, but concentration-dependently potentiated constrictions pre-evoked by phenylephrine under normoxic condition. The potentiation was independent of the endothelium. MIF-induced potentiation of phenylephrine-evoked constriction was partially inhibited by PKC inhibitor chelerythrine, p38 inhibitor SB 203580, ERK1/2 inhibitor U0126, respectively. CONCLUSIONS: Our results suggested that MIF enhanced vasoconstriction of pulmonary artery elicited by agonist through PKC, p38 and ERK1/2 signal pathways, which may contributes to hypoxic pulmonary vasoconstriction.


Assuntos
Hipertensão Pulmonar/etiologia , Hipóxia/complicações , Oxirredutases Intramoleculares/metabolismo , Fatores Inibidores da Migração de Macrófagos/metabolismo , Artéria Pulmonar/metabolismo , Artéria Pulmonar/fisiopatologia , Vasoconstrição , Animais , Western Blotting , Células Cultivadas , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Hipertensão Pulmonar Primária Familiar , Hipertensão Pulmonar/metabolismo , Hipertensão Pulmonar/fisiopatologia , Hipóxia/metabolismo , Hipóxia/fisiopatologia , Imuno-Histoquímica , Masculino , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Cadeias Leves de Miosina/metabolismo , Fosforilação , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Artéria Pulmonar/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Vasoconstrição/efeitos dos fármacos , Vasoconstritores/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
19.
Biol Pharm Bull ; 34(7): 1052-7, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21720012

RESUMO

Inflammation takes responsibility for the seawater aspiration-induced lung injury. Tanshinone IIA (TIIA) can protect lipopolysaccharide-induced lung injury in mice through the inhibition of inflammation, but it is not reported whether TIIA have a protective effect on lung injury induced by seawater aspiration. Macrophage migration inhibitory factor (MIF) plays an important role in acute lung injury. In this study, we observed the effect of TIIA on the seawater aspiration-induced lung injury and the role of MIF in it. Seawater was aspirated into trachea of rats to make the lung injury model. TIIA was administered to investigate its beneficial effect on seawater-induced acute lung injury. The results showed that seawater aspiration led to hyoxemia, pulmonary edema, neutrophil infiltration, and lung histopathologic changes, with the elevated MIF expression in the lung tissues and plasma. However, these changes were attenuated by TIIA. In macrophage cells we also demonstrated that TIIA could inhibit MIF expression, nuclear factor κB (NF-κB) activity and release of interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) induced by seawater. Besides, pretreatment with (S,R)-3-(4-hydroxyphenyl)-4,5-dihydro-5-isoxazole acetic acid (ISO-1), the MIF antagonist, elevated NF-κB and cytokines induced by seawater were also reduced markedly. Furthermore, rMIF treatment alone increased the phosphorylation level of NF-κB and release of cytokines, which was almost abolished by TIIA. Taken together, our results suggested that TIIA exert a protective effect on the seawater aspiration-induced lung injury partly through downregulation of MIF and the subsequent NF-κB activity, as well as expression of IL-6 and TNF-α.


Assuntos
Abietanos/farmacologia , Lesão Pulmonar/prevenção & controle , Fatores Inibidores da Migração de Macrófagos/antagonistas & inibidores , Água do Mar , Animais , Ensaio de Imunoadsorção Enzimática , Lesão Pulmonar/patologia , Masculino , Neutrófilos/patologia , Ratos , Ratos Sprague-Dawley
20.
Am J Respir Cell Mol Biol ; 45(5): 1028-35, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21622293

RESUMO

Inhibiting hypoxia-inducible factor (HIF)-1α activity has been proposed as a novel therapeutic target in LPS-induced sepsis syndrome. We have reported that tanshinone IIA (TIIA) can reduce LPS-induced lethality and lung injury in mice, but the precise mechanisms have not been fully described. Therefore, the present study investigated whether the protective effect of TIIA was related to the inhibition of LPS-induced HIF-1α expression and what mechanisms accounted for it. This study showed that TIIA pretreatment improved LPS-induced biochemical and cellular changes and reduced the production of inflammatory cytokines. Pretreatment with TIIA decreased LPS-induced HIF-1α expression in vivo and in vitro. TIIA did not affect the LPS-induced HIF-1α mRNA level but inhibited HIF-1α protein translation by the inhibition of the PI3K/AKT and MAPK pathways and related protein translational regulators, such as p70S6K1, S6 ribosomal protein, 4E-BP1, and eIF4E, and promoted HIF-1α protein degradation via the proteasomal pathway in LPS-stimulated macrophages. These observations partially explain the antiinflammatory effects of TIIA, which provides scientific basis for its application for the treatment of acute lung injury/acute respiratory distress syndrome or sepsis.


Assuntos
Abietanos/farmacologia , Lesão Pulmonar Aguda/tratamento farmacológico , Anti-Inflamatórios não Esteroides/farmacologia , Endotoxemia/tratamento farmacológico , Subunidade alfa do Fator 1 Induzível por Hipóxia/biossíntese , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Ciclo Celular , Proteínas de Ligação a DNA/antagonistas & inibidores , Endotoxemia/induzido quimicamente , Fatores de Iniciação em Eucariotos , Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Lipopolissacarídeos/toxicidade , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Fosfoproteínas/antagonistas & inibidores , Complexo de Endopeptidases do Proteassoma/efeitos dos fármacos , Biossíntese de Proteínas/efeitos dos fármacos , Proteína S6 Ribossômica/antagonistas & inibidores , Proteínas Quinases S6 Ribossômicas 70-kDa/antagonistas & inibidores , Fatores de Transcrição/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...