Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 96
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Environ Res ; 169: 280-296, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30497003

RESUMEN

Although wetland environmental protection plans are synonymous with wetland management and erosion control plans, the public perceptions of such plans often focus on their impact on the human enjoyment of wetland areas. These plans are affected by many interrelated influence factors, such as human welfare, property, safety, management, operations, maintenance, ecology, environment, artificial structures, climate control, and sustainable development. The purpose of this paper is to probe how to use qualitative and quantitative measurements of wetland environments to create plan indexes using criteria/attributes as well as how to help these indexes for achieving the aspiration levels in each criterion/attribute. Previous studies that attempted to measure environmental evaluations and plans have assumed that these criteria are independent, but this assumption does not hold in real-world applications of real problems. Therefore, in this proof-of-concept study, using an empirical exam among various attributes and to measure and evaluate the real conditions for improving the wetland environmental problems. A DEMATEL technique can be used to construct the INRM, the basic concept of an ANP was modified to determine the influential weights of criteria/dimensions in our research alternative, called DANP (DEMATEL-based ANP). Then we can construct a decision-making model via a hybrid modified VIKOR method to improve wetlands environmental management manager strategy formulation in performance evaluation toward for achieving the aspiration level. Using these techniques, a proposed model appeared, which can be used to explain interdependence and feedback problems. Based on the final results, we can also propose a gap improvement in the development of a sustainable development plan for the environment while taking comfort and safety into account to improve standards and achieve human welfare expectations.


Asunto(s)
Desarrollo Sostenible , Humedales , Conservación de los Recursos Naturales , Toma de Decisiones , Humanos , Modelos Teóricos
2.
J Cell Mol Med ; 22(9): 4387-4398, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29992759

RESUMEN

Kallistatin, a plasma protein, protects against vascular and organ injury. This study is aimed to investigate the role and mechanism of kallistatin in endothelial senescence. Kallistatin inhibited H2 O2 -induced senescence in human endothelial cells, as indicated by reduced senescence-associated-ß-galactosidase activity, p16INK4a and plasminogen activator inhibitor-1 expression, and elevated telomerase activity. Kallistatin blocked H2 O2 -induced superoxide formation, NADPH oxidase levels and VCAM-1, ICAM-1, IL-6 and miR-34a synthesis. Kallistatin reversed H2 O2 -mediated inhibition of endothelial nitric oxide synthase (eNOS), SIRT1, catalase and superoxide dismutase (SOD)-2 expression, and kallistatin alone stimulated the synthesis of these antioxidant enzymes. Moreover, kallistatin's anti-senescence and anti-oxidant effects were attributed to SIRT1-mediated eNOS pathway. Kallistatin, via interaction with tyrosine kinase, up-regulated Let-7g, whereas Let-7g inhibitor abolished kallistatin's effects on miR-34a and SIRT1/eNOS synthesis, leading to inhibition of senescence, oxidative stress and inflammation. Furthermore, lung endothelial cells isolated from endothelium-specific kallistatin knockout mice displayed marked reduction in mouse kallistatin levels. Kallistatin deficiency in mouse endothelial cells exacerbated senescence, oxidative stress and inflammation compared to wild-type mouse endothelial cells, and H2 O2 treatment further magnified these effects. Kallistatin deficiency caused marked reduction in Let-7g, SIRT1, eNOS, catalase and SOD-1 mRNA levels, and elevated miR-34a synthesis in mouse endothelial cells. These findings indicate that endogenous kallistatin through novel mechanisms protects against endothelial senescence by modulating Let-7g-mediated miR-34a-SIRT1-eNOS pathway.


Asunto(s)
Células Endoteliales/metabolismo , MicroARNs/genética , Óxido Nítrico Sintasa de Tipo III/genética , Serpinas/genética , Sirtuina 1/genética , Animales , Catalasa/genética , Catalasa/metabolismo , Senescencia Celular/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Regulación de la Expresión Génica , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Peróxido de Hidrógeno/farmacología , Pulmón/citología , Pulmón/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Cultivo Primario de Células , Serpina E2/genética , Serpina E2/metabolismo , Serpinas/deficiencia , Serpinas/farmacología , Transducción de Señal , Sirtuina 1/metabolismo , Superóxido Dismutasa-1/genética , Superóxido Dismutasa-1/metabolismo , Telomerasa/genética , Telomerasa/metabolismo , beta-Galactosidasa/genética , beta-Galactosidasa/metabolismo
3.
Biol Chem ; 398(12): 1309-1317, 2017 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-28742513

RESUMEN

Kallistatin, via its two structural elements - an active site and a heparin-binding domain - displays a double-edged function in angiogenesis, apoptosis and oxidative stress. First, kallistatin has both anti-angiogenic and pro-angiogenic effects. Kallistatin treatment attenuates angiogenesis and tumor growth in cancer-bearing mice. Kallistatin via its heparin-binding site inhibits angiogenesis by blocking vascular endothelial growth factor (VEGF)-induced growth, migration and adhesion of endothelial cells. Conversely, kallistatin via the active site promotes neovascularization by stimulating VEGF levels in endothelial progenitor cells. Second, kallistatin inhibits or induces apoptosis depending on cell types. Kallistatin attenuates organ injury and apoptosis in animal models, and its heparin-binding site is essential for blocking tumor necrosis factor (TNF)-α-induced apoptosis in endothelial cells. However, kallistatin via its active site induces apoptosis in breast cancer cells by up-regulating miR-34a and down-regulating miR-21 and miR-203 synthesis. Third, kallistatin can act as an antioxidant or pro-oxidant. Kallistatin treatment inhibits oxidative stress and tissue damage in animal models and cultured cells. Kallistatin via the heparin-binding domain antagonizes TNF-α-induced oxidative stress, whereas its active site is crucial for stimulating antioxidant enzyme expression. In contrast, kallistatin provokes oxidant formation, leading to blood pressure reduction and bacterial killing. Kallistatin-mediated vasodilation is partly mediated by H2O2, as the effect is abolished by the antioxidant enzyme catalase. Moreover, kallistatin exerts a bactericidal effect by stimulating superoxide production in neutrophils of mice with microbial infection as well as in cultured immune cells. Thus, kallistatin's dual roles in angiogenesis, apoptosis and oxidative stress contribute to its beneficial effects in various diseases.


Asunto(s)
Apoptosis , Neovascularización Patológica/metabolismo , Estrés Oxidativo , Serpinas/metabolismo , Animales , Humanos
4.
Exp Cell Res ; 340(2): 305-14, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26790955

RESUMEN

Kallistatin is an endogenous protein that regulates differential signaling pathways and biological functions. Our previous studies showed that kallistatin gene therapy inhibited angiogenesis, tumor growth and metastasis in mice, and kallistatin protein suppressed Wnt-mediated growth, migration and invasion by blocking Wnt/ß-catenin signaling pathway in breast cancer cells. In this study, we show that kallistatin reduced cell viability, and increased apoptotic cell death and caspase-3 activity in MDA-MB-231 breast cancer cells. Kallistatin also induced cancer cell autophagy, as evidenced by increased LC3B levels and elevated Atg5 and Beclin-1 expression; however, co-administration of Wnt or PPARγ antagonist GW9662 abolished these effects. Moreover, kallistatin via its heparin-binding site antagonized Wnt3a-induced cancer cell proliferation and increased PPARγ expression. Kallistatin inhibited oncogenic miR-21 synthesis associated with reduced Akt phosphorylation and Bcl-2 synthesis, but increased BAX expression. Kallistatin via PKC-ERK activation reduced miR-203 levels, leading to increased expression of suppressor of cytokine signaling 3 (SOCS3), a tumor suppressor. Conversely, kallistatin stimulated expression of the tumorigenic suppressors miR-34a and p53. Kallistatin's active site is essential for suppressing miR-21 and miR-203, and stimulating miR-34a and SOCS3 expression. This is the first study to demonstrate that kallistatin's heparin-binding site is essential for inhibiting Wnt-mediated effects, and its active site plays a key role in regulating miR-21, miR-203, miR-34a and SOCS3 synthesis in breast cancer cells. These findings reveal novel mechanisms of kallistatin in inducing apoptosis and autophagy in breast cancer cells, thus inhibiting tumor progression by regulation of Wnt/PPARγ signaling, as well as miR-21, miR-203 and miR-34a synthesis.


Asunto(s)
Apoptosis/genética , Autofagia/genética , Neoplasias de la Mama/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , MicroARNs/genética , Serpinas/metabolismo , Vía de Señalización Wnt/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular/fisiología , Humanos , MicroARNs/biosíntesis
5.
Exp Cell Res ; 337(1): 103-10, 2015 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-26156753

RESUMEN

Kallistatin, an endogenous protein, consists of two structural elements: active site and heparin-binding domain. Kallistatin exerts beneficial effects on fibrosis by suppressing transforming growth factor (TGF)-ß synthesis in animal models. TGF-ß is the most potent inducer of endothelial-mesenchymal transition (EndMT), which contributes to fibrosis and cancer. MicroRNA (miR)-21 is an important player in organ fibrosis and tumor invasion. Here we investigated the potential role of kallistatin in EndMT via modulation of miR-21 in endothelial cells. Human kallistatin treatment blocked TGF-ß-induced EndMT, as evidenced by morphological changes as well as increased endothelial and reduced mesenchymal marker expression. Kallistatin also inhibited TGF-ß-mediated reactive oxygen species (ROS) formation and NADPH oxidase expression and activity. Moreover, kallistatin antagonized TGF-ß-induced miR-21 and Snail1 synthesis, Akt phosphorylation, NF-κB activation, and matrix metalloproteinase 2 (MMP2) synthesis and activation. Kallistatin via its heparin-binding site blocked TGF-ß-induced miR-21, Snail1 expression, and ROS formation, as wild-type kallistatin, but not heparin-binding site mutant kallistatin, exerted the effect. Conversely, kallistatin through its active site stimulated the synthesis of endothelial nitric oxide synthase (eNOS), sirtuin 1 (Sirt1) and forkhead box O1 (FoxO1); however, these effects were blocked by genistein, a tyrosine kinase inhibitor. This is the first study to demonstrate that kallistatin's heparin-binding site is crucial for preventing TGF-ß-induced miR-21 and oxidative stress, while its active site is key for stimulating the expression of antioxidant genes via interaction with an endothelial surface tyrosine kinase. These findings reveal novel mechanisms of kallistatin in protection against fibrosis and cancer by suppressing EndMT.


Asunto(s)
Células Endoteliales/fisiología , Transición Epitelial-Mesenquimal , MicroARNs/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Serpinas/fisiología , Factor de Crecimiento Transformador beta/fisiología , Dominio Catalítico , Expresión Génica , Regulación Enzimológica de la Expresión Génica , Células HEK293 , Humanos , MicroARNs/genética , NADPH Oxidasas/metabolismo , Óxido Nítrico Sintasa de Tipo III/genética , Especies Reactivas de Oxígeno/metabolismo , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
6.
Antimicrob Agents Chemother ; 59(9): 5619-30, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26149981

RESUMEN

Proteolytic cleavage of the hemagglutinin (HA) of influenza virus by host trypsin-like proteases is required for viral infectivity. Some serine proteases are capable of cleaving influenza virus HA, whereas some serine protease inhibitors (serpins) inhibit the HA cleavage in various cell types. Kallikrein-related peptidase 1 (KLK1, also known as tissue kallikrein) is a widely distributed serine protease. Kallistatin, a serpin synthesized mainly in the liver and rapidly secreted into the circulation, forms complexes with KLK1 and inhibits its activity. Here, we investigated the roles of KLK1 and kallistatin in influenza virus infection. We show that the levels of KLK1 increased, whereas those of kallistatin decreased, in the lungs of mice during influenza virus infection. KLK1 cleaved H1, H2, and H3 HA molecules and consequently enhanced viral production. In contrast, kallistatin inhibited KLK1-mediated HA cleavage and reduced viral production. Cells transduced with the kallistatin gene secreted kallistatin extracellularly, which rendered them more resistant to influenza virus infection. Furthermore, lentivirus-mediated kallistatin gene delivery protected mice against lethal influenza virus challenge by reducing the viral load, inflammation, and injury in the lung. Taking the data together, we determined that KLK1 and kallistatin contribute to the pathogenesis of influenza virus by affecting the cleavage of the HA peptide and inflammatory responses. This study provides a proof of principle for the potential therapeutic application of kallistatin or other KLK1 inhibitors for influenza. Since proteolytic activation also enhances the infectivity of some other viruses, kallistatin and other kallikrein inhibitors may be explored as antiviral agents against these viruses.


Asunto(s)
Antivirales/uso terapéutico , Hemaglutininas Virales/metabolismo , Gripe Humana/tratamiento farmacológico , Serpinas/uso terapéutico , Calicreínas de Tejido/metabolismo , Animales , Western Blotting , Línea Celular , Perros , Ensayo de Inmunoadsorción Enzimática , Humanos , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
Crit Care ; 19: 200, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25930108

RESUMEN

INTRODUCTION: Kallistatin levels in the circulation are reduced in patients with sepsis and liver disease. Transgenic mice expressing kallistatin are resistant to lipopolysaccharide (LPS)-induced mortality. Here, we investigated the effect of kallistatin on survival and organ damage in mouse models of established sepsis. METHODS: Mice were rendered septic by cecal ligation and puncture (CLP), or endotoxemic by LPS injection. Recombinant human kallistatin was administered intravenously six hours after CLP, or intraperitoneally four hours after LPS challenge. The effect of kallistatin treatment on organ damage was examined one day after sepsis initiation, and mouse survival was monitored for four to six days. RESULTS: Human kallistatin was detected in mouse serum of kallistatin-treated mice. Kallistatin significantly reduced CLP-induced renal injury as well as blood urea nitrogen, serum creatinine, interleukin-6 (IL-6), and high mobility group box-1 (HMGB1) levels. In the lung, kallistatin decreased malondialdehyde levels and HMGB1 and toll-like receptor-4 (TLR4) synthesis, but increased suppressor of cytokine signaling-3 (SOCS3) expression. Moreover, kallistatin attenuated liver injury, serum alanine transaminase (ALT) levels and hepatic tumor necrosis factor-α (TNF-α) synthesis. Furthermore, delayed kallistatin administration improved survival in CLP mice by 38%, and LPS-treated mice by 42%. In LPS-induced endotoxemic mice, kallistatin attenuated kidney damage in association with reduced serum creatinine, IL-6 and HMGB1 levels, and increased renal SOCS3 expression. Kallistatin also decreased liver injury in conjunction with diminished serum ALT levels and hepatic TNF-α and TLR4 expression. In cultured macrophages, kallistatin through its active site increased SOCS3 expression, but this effect was blocked by inhibitors of tyrosine kinase, protein kinase C and extracellular signal-regulated kinase (ERK), indicating that kallistatin stimulates a tyrosine-kinase-protein kinase C-ERK signaling pathway. CONCLUSIONS: This is the first study to demonstrate that delayed human kallistatin administration is effective in attenuating multi-organ injury, inflammation and mortality in mouse models of polymicrobial infection and endotoxemia. Thus, kallistatin therapy may provide a promising approach for the treatment of sepsis in humans.


Asunto(s)
Lesión Renal Aguda/prevención & control , Hígado/efectos de los fármacos , Proteínas Recombinantes/farmacología , Sepsis/tratamiento farmacológico , Serpinas/farmacología , Alanina Transaminasa/sangre , Animales , Nitrógeno de la Urea Sanguínea , Células Cultivadas , Creatinina/sangre , Modelos Animales de Enfermedad , Endotoxemia/metabolismo , Proteína HMGB1/metabolismo , Humanos , Interleucina-6/sangre , Hígado/metabolismo , Pulmón/metabolismo , Macrófagos/metabolismo , Malondialdehído/metabolismo , Ratones , Sepsis/metabolismo , Serpinas/sangre , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Receptor Toll-Like 4/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
8.
Immunology ; 142(2): 216-26, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24467264

RESUMEN

Kallistatin, a plasma protein, has been shown to exert multi-factorial functions including inhibition of inflammation, oxidative stress and apoptosis in animal models and cultured cells. Kallistatin levels are reduced in patients with sepsis and in lipopolysaccharide (LPS)-induced septic mice. Moreover, transgenic mice expressing kallistatin are more resistant to LPS-induced mortality. Here, we investigated the effects of human kallistatin on organ injury and survival in a mouse model of polymicrobial sepsis. In this study, mice were injected intravenously with recombinant kallistatin (KS3, 3 mg/kg; or KS10, 10 mg/kg body weight) and then rendered septic by caecal ligation and puncture 30 min later. Kallistatin administration resulted in a > 10-fold reduction of peritoneal bacterial counts, and significantly decreased serum tumour necrosis factor-α, interleukin-6 and high mobility group box-1 (HMGB1) levels. Kallistatin also inhibited HMGB1 and toll-like receptor-4 gene expression in the lung and kidney. Administration of kallistatin attenuated renal damage and decreased blood urea nitrogen and serum creatinine levels, but increased endothelial nitric oxide synthase and nitric oxide levels in the kidney. In cultured endothelial cells, human kallistatin via its heparin-binding site inhibited HMGB1-induced nuclear factor-κB activation and inflammatory gene expression. Moreover, kallistatin significantly reduced apoptosis and caspase-3 activity in the spleen. Furthermore, kallistatin treatment markedly improved the survival of septic mice by 23% (KS3) and 41% (KS10). These results indicate that kallistatin is a unique protecting agent in sepsis-induced organ damage and mortality by inhibiting inflammation and apoptosis, as well as enhancing bacterial clearance in a mouse model of polymicrobial sepsis.


Asunto(s)
Modelos Animales de Enfermedad , Riñón/efectos de los fármacos , Riñón/patología , Sepsis/tratamiento farmacológico , Sepsis/patología , Serpinas/farmacología , Serpinas/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Nitrógeno de la Urea Sanguínea , Caspasa 3/metabolismo , Creatina/sangre , Proteína HMGB1/metabolismo , Humanos , Inflamación/tratamiento farmacológico , Inflamación/patología , Inyecciones Intravenosas , Interleucina-6/metabolismo , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Masculino , Ratones , FN-kappa B/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Serpinas/administración & dosificación , Bazo/efectos de los fármacos , Bazo/metabolismo , Tasa de Supervivencia , Receptor Toll-Like 4/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
9.
Prog Drug Res ; 69: 37-57, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25130039

RESUMEN

Tissue kallikrein is a serine proteinase that cleaves low molecular weight kininogen to produce kinin peptides, which in turn activate kinin receptors to trigger multiple biological functions. In addition to its kinin-releasing activity, tissue kallikrein directly interacts with the kinin B2 receptor, protease-activated receptor-1, and gamma-epithelial Na channel. The tissue kallikrein-kinin system (KKS) elicits a wide spectrum of biological activities, including reducing hypertension, cardiac and renal damage, restenosis, ischemic stroke, and skin wound injury. Both loss-of-function and gain-of-function studies have shown that the KKS plays an important endogenous role in the protection against health pathologies. Tissue kallikrein/kinin treatment attenuates cardiovascular, renal, and brain injury by inhibiting oxidative stress, apoptosis, inflammation, hypertrophy, and fibrosis and promoting angiogenesis and neurogenesis. Approaches that augment tissue kallikrein-kinin activity might provide an effective strategy for the treatment of hypertension and associated organ damage.


Asunto(s)
Antihipertensivos/uso terapéutico , Hipertensión/tratamiento farmacológico , Cininas/uso terapéutico , Calicreínas de Tejido/uso terapéutico , Animales , Antihipertensivos/metabolismo , Diabetes Mellitus/tratamiento farmacológico , Diabetes Mellitus/enzimología , Modelos Animales de Enfermedad , Cardiopatías/enzimología , Cardiopatías/etiología , Cardiopatías/prevención & control , Humanos , Hipertensión/complicaciones , Hipertensión/enzimología , Enfermedades Renales/enzimología , Enfermedades Renales/etiología , Enfermedades Renales/prevención & control , Cininas/metabolismo , Transducción de Señal/efectos de los fármacos , Calicreínas de Tejido/metabolismo , Lesiones del Sistema Vascular/enzimología , Lesiones del Sistema Vascular/prevención & control
10.
Lab Invest ; 93(5): 577-91, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23508045

RESUMEN

Endothelial progenitor cells (EPCs) have been shown to enhance angiogenesis not only by incorporating into the vasculature but also by secreting cytokines, thereby serving as an ideal vehicle for gene transfer. As tissue kallikrein (TK) has pleiotropic effects in inhibiting apoptosis and oxidative stress, and promoting angiogenesis, we evaluated the salutary potential of kallikrein-modified human EPCs (hEPCs; Ad.hTK-hEPCs) after acute myocardial infarction (MI). We genetically modified hEPCs with a TK gene and evaluated cell survival, engraftment, revascularization, and functional improvement in a nude mouse left anterior descending ligation model. hEPCs were manipulated to overexpress the TK gene. In vitro, the antiapoptotic and paracrine effects were assessed under oxidative stress. TK protects hEPCs from oxidative stress-induced apoptosis via inhibition of activation of caspase-3 and -9, induction of Akt phosphorylation, and secretion of vascular endothelial growth factor. In vivo, the Ad.hTK-hEPCs were transplanted after MI via intracardiac injection. The surviving cells were tracked after transplantation using near-infrared optical imaging. Left ventricular (LV) function was evaluated by transthoracic echocardiography. Capillary density was quantified using immunohistochemical staining. Engrafted Ad.hTK-hEPCs exhibited advanced protection against ischemia by increasing LV ejection fraction. Compared with Ad.Null-hEPCs, transplantation with Ad.hTK-hEPCs significantly decreased cardiomyocyte apoptosis in association with increased retention of transplanted EPCs in the myocardium. Capillary density and arteriolar density in the infarct border zone was significantly higher in Ad.hTK-hEPC-transplanted mice than in Ad.Null-hEPC-treated mice. Transplanted hEPCs were clearly incorporated into CD31(+) capillaries. These results indicate that implantation of kallikrein-modified EPCs in the heart provides advanced benefits in protection against ischemia-induced MI by enhanced angiogenesis and reducing apoptosis.


Asunto(s)
Células Endoteliales/enzimología , Células Endoteliales/trasplante , Infarto del Miocardio/cirugía , Proteínas Proto-Oncogénicas c-akt/metabolismo , Trasplante de Células Madre/métodos , Células Madre/enzimología , Calicreínas de Tejido/metabolismo , Adenoviridae , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Presión Sanguínea/fisiología , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Sangre Fetal/citología , Técnica del Anticuerpo Fluorescente , Ingeniería Genética , Terapia Genética , Humanos , Peróxido de Hidrógeno/farmacología , Masculino , Ratones , Ratones Desnudos , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Neovascularización Fisiológica/fisiología , Imagen Óptica , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Distribución Aleatoria , Células Madre/citología , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Calicreínas de Tejido/genética , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/biosíntesis
11.
Antimicrob Agents Chemother ; 57(11): 5366-72, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23959316

RESUMEN

Group A streptococcus (GAS) infection may cause severe life-threatening diseases, including necrotizing fasciitis and streptococcal toxic shock syndrome. Despite the availability of effective antimicrobial agents, there has been a worldwide increase in the incidence of invasive GAS infection. Kallistatin (KS), originally found to be a tissue kallikrein-binding protein, has recently been shown to possess anti-inflammatory properties. However, its efficacy in microbial infection has not been explored. In this study, we transiently expressed the human KS gene by hydrodynamic injection and investigated its anti-inflammatory and protective effects in mice via air pouch inoculation of GAS. The results showed that KS significantly increased the survival rate of GAS-infected mice. KS treatment reduced local skin damage and bacterial counts compared with those in mice infected with GAS and treated with a control plasmid or saline. While there was a decrease in immune cell infiltration of the local infection site, cell viability and antimicrobial factors such as reactive oxygen species actually increased after KS treatment. The efficiency of intracellular bacterial killing in neutrophils was directly enhanced by KS administration. Several inflammatory cytokines, including tumor necrosis factor alpha, interleukin 1ß, and interleukin 6, in local infection sites were reduced by KS. In addition, KS treatment reduced vessel leakage, bacteremia, and liver damage after local infection. Therefore, our study demonstrates that KS provides protection in GAS-infected mice by enhancing bacterial clearance, as well as reducing inflammatory responses and organ damage.


Asunto(s)
Inmunomodulación , Neutrófilos/inmunología , Serpinas/inmunología , Infecciones Estreptocócicas/inmunología , Streptococcus pyogenes/inmunología , Animales , Expresión Génica , Humanos , Interleucina-1beta/antagonistas & inhibidores , Interleucina-1beta/biosíntesis , Interleucina-6/antagonistas & inhibidores , Interleucina-6/biosíntesis , Ratones , Neutrófilos/microbiología , Serpinas/genética , Serpinas/metabolismo , Infecciones Estreptocócicas/microbiología , Infecciones Estreptocócicas/mortalidad , Streptococcus pyogenes/patogenicidad , Análisis de Supervivencia , Transgenes , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/biosíntesis
12.
Mol Cell Biochem ; 379(1-2): 295-301, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23666756

RESUMEN

Kallistatin, a plasma protein, exerts pleiotropic effects in inhibiting angiogenesis, inflammation and tumor growth. Canonical Wnt signaling is the primary pathway for oncogenesis in the mammary gland. In this study, we demonstrate that kallistatin bound to the Wnt coreceptor low-density lipoprotein receptor-related protein 6 (LRP6), thus, blocking Wnt/ß-catenin signaling and Wnt-mediated growth and migration in MDA-MB-231 breast cancer cells. Kallistatin inhibited Wnt3a-induced proliferation, migration, and invasion of cultured breast cancer cells. Moreover, kallistatin was bound to LRP6 in breast cancer cells, as identified by immunoprecipitation followed by western blot. Kallistatin suppressed Wnt3a-mediated phosphorylation of LRP6 and glycogen synthase kinase-3ß, and the elevation of cytosolic ß-catenin levels. Furthermore, kallistatin antagonized Wnt3a-induced expression of c-Myc, cyclin D1, and vascular endothelial growth factor. These findings indicate a novel role of kallistatin in preventing breast tumor growth and mobility by direct interaction with LRP6, leading to blockade of the canonical Wnt signaling pathway.


Asunto(s)
Movimiento Celular , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/metabolismo , Serpinas/fisiología , Vía de Señalización Wnt , Neoplasias de la Mama , Línea Celular Tumoral , Proliferación Celular , Ciclina D1/genética , Ciclina D1/metabolismo , Femenino , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Unión Proteica , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteína Wnt3A/metabolismo
13.
Circ J ; 77(8): 2134-44, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23697984

RESUMEN

BACKGROUND: Genetically modified mesenchymal stem cells (MSCs) are a promising approach to the treatment of cardiac injury after myocardial infarction (MI). METHODS AND RESULTS: Rat MSCs were transduced with adenovirus containing human tissue kallikrein (TK) gene (TK-MSCs), and they secreted human TK into culture medium. Cultured TK-MSCs were more resistant to hypoxia-induced apoptosis and exhibited reduced caspase-3 activity compared to control GFP-MSCs. The effect of TK-MSC injection on cardiac injury was evaluated in rats at 1 and 14 days after MI. At 1 day after MI, human TK expression in the myocardium was associated with improved cardiac function and decreased inflammatory cell accumulation, proinflammatory gene expression and apoptosis. The beneficial effect of TK-MSCs against apoptosis was verified in cultured cardiomyocytes, as TK-MSC-conditioned medium suppressed hypoxia-induced apoptosis and caspase-3 activity, and increased Akt phosphorylation. At 2 weeks after MI, TK-MSCs improved cardiac function, decreased infarct size, attenuated cardiac remodeling, and promoted neovascularization, as compared to GFP-MSCs. Furthermore, the TK-MSC-conditioned medium, containing elevated vascular endothelial growth factor levels, stimulated the proliferation, migration and tube formation of cultured human endothelial cells. CONCLUSIONS: Our results indicate that TK-modified MSCs provide enhanced protection against cardiac injury, apoptosis and inflammation, and promote neovascularization after MI, leading to cardiac function improvement.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Infarto del Miocardio/terapia , Isquemia Miocárdica/prevención & control , Neovascularización Fisiológica , Calicreínas de Tejido/biosíntesis , Adenoviridae , Animales , Apoptosis/genética , Caspasa 3/genética , Caspasa 3/metabolismo , Humanos , Células Madre Mesenquimatosas/patología , Infarto del Miocardio/complicaciones , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/fisiopatología , Isquemia Miocárdica/etiología , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/patología , Isquemia Miocárdica/fisiopatología , Ratas , Ratas Sprague-Dawley , Calicreínas de Tejido/genética , Transducción Genética
14.
Crit Care ; 17(1): R27, 2013 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-23394256

RESUMEN

INTRODUCTION: Community-acquired pneumonia (CAP) requiring intensive care unit (ICU) treatment commonly causes acute respiratory failure with high mortality. Kallistatin, an endogenous tissue kallikrein inhibitor, has been reported to be protective in various human diseases. The aim of this study was to assess the correlations of kallistatin with other biomarkers and to determine whether kallistatin levels have a prognostic value in severe CAP. METHODS: Plasma samples and clinical data were prospectively collected from 54 patients with severe CAP requiring ICU admission. Seventeen healthy control subjects were included for comparison. Plasma kallistatin, kallikrein, and other biomarkers of inflammation (tumor necrosis factor-α (TNF-α), interleukin (IL)-1ß, IL-6, IL-8, C-reactive protein (CRP)), and anti-coagulation (protein C, anti-thrombin III) were measured on days 1 and 4 of ICU admission. Comparison between survivors (n = 41) and nonsurvivors (n = 13) was performed. RESULTS: Plasma kallistatin was significantly consumed in severe CAP patients compared with healthy individuals. Lower day 1 kallistatin levels showed a strong trend toward increased mortality (P = 0.018) and higher day 1 CURB-65 scores (P = 0.004). Plasma kallistatin levels on day 1 of ICU admission were significantly decreased in patients who developed septic shock (P = 0.017) and who had acute respiratory distress syndrome (P = 0.044). In addition, kallistatin levels were positively correlated with anti-thrombin III and protein C and inversely correlated with IL-1ß, IL-6, and CRP levels. In a multivariate logistic regression analysis, higher day 1 CURB-65 scores were independent predictors of mortality (odds ratio = 29.9; P = 0.009). Also, higher day 1 kallistatin levels were independently associated with a decreased risk of death (odds ratio, 0.1) with a nearly significant statistical difference (P = 0.056). Furthermore, we found that a cutoff level of 6.5 µg/ml of day 1 kallistatin determined by receiver operating characteristic curves could be used to distinguish between patients who survived in 60 days and those who did not. CONCLUSIONS: These results suggest that kallistatin may serve as a novel marker for severe CAP prognosis and may be involved in the pathogenesis of CAP through antiinflammatory and anticoagulation effects.


Asunto(s)
Neumonía/sangre , Neumonía/diagnóstico , Serpinas/sangre , Índice de Severidad de la Enfermedad , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores/sangre , Infecciones Comunitarias Adquiridas/sangre , Infecciones Comunitarias Adquiridas/diagnóstico , Infecciones Comunitarias Adquiridas/mortalidad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mortalidad/tendencias , Neumonía/mortalidad , Estudios Prospectivos , Adulto Joven
15.
PLoS One ; 18(3): e0282891, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36913383

RESUMEN

Abrupt thrombosis is a form of thrombosis that occurs unexpectedly and without being preceded by hemodialysis fistula (AVF) dysfunction during dialysis. We found that AVFs with a history of abrupt thrombosis (abtAVF) appeared to have more episodes of thrombosis and required more frequent interventions than those without such history. Therefore, we sought to characterize the abtAVFs and examined our follow-up protocols to determine which one is optimal. We performed a retrospective cohort study using routinely collected data. The thrombosis rate, AVF loss rate, thrombosis-free primary patency, and secondary patency were calculated. Additionally, the restenosis rates of the AVFs under the follow-up protocol/sub-protocols and the abtAVFs were determined. The thrombosis rate, procedure rate, AVF loss rate, thrombosis-free primary patency, and secondary patency of the abtAVFs were 0.237/pt-yr, 2.702/pt-yr, 0.027/pt-yr, 78.3%, and 96.0%, respectively. The restenosis rate for AVFs in the abtAVF group and the angiographic follow-up sub-protocol were similar. However, the abtAVF group had a significantly higher thrombosis rate and AVF loss rate than AVFs without a history of abrupt thrombosis (n-abtAVF). The lowest thrombosis rate was observed for n-abtAVFs, followed up periodically under the outpatient or angiographic sub-protocols. AVFs with a history of abrupt thrombosis had a high restenosis rate, and periodic angiographic follow-up with a mean interval of 3 months was presumed appropriate. For selected populations, such as salvage-challenging AVFs, periodic outpatient or angiographic follow-up was mandatory to extend their usable lives for hemodialysis.


Asunto(s)
Derivación Arteriovenosa Quirúrgica , Fístula , Trombosis , Humanos , Diálisis Renal/efectos adversos , Diálisis Renal/métodos , Estudios Retrospectivos , Estudios de Seguimiento , Grado de Desobstrucción Vascular , Derivación Arteriovenosa Quirúrgica/efectos adversos , Derivación Arteriovenosa Quirúrgica/métodos , Trombosis/etiología , Resultado del Tratamiento
16.
J Vasc Access ; 24(4): 639-645, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34524025

RESUMEN

BACKGROUND: Quantitative physical examination (PE) indicators, including palpable pulsatility length and outflow scores, can be used to quantify stenosis severity at hemodialysis vascular access sites. It is known that the risk of high-shear-related thrombosis is increased when the minimal luminal diameter (MLD) of stenosis decreases. At present, MLD is measured using sonography or angiography. This study sought to determine the relationship between quantitative PE indicators and MLD and report their diagnostic performance in detecting patients with stenosis at a high risk of thrombosis. METHODS: We performed a retrospective case-control study using routinely collected data. We used the post-stenosis palpable pulsatility length (sPPL) and pulse-and-thrill based outflow score to assess the severity of AVF inflow and outflow stenosis, respectively. We recorded paired quantitative PE indicators and MLD before and after angioplasty in patients enrolled over a 4-month period. RESULTS: A total of 249 paired PE indicators and MLD measurements were obtained from 163 patients. A receiver operating characteristic curve analysis showed that an MLD cutoff value of <1.55 mm and an MLD of <1.95 mm discriminated sPPL = 0 and PESOS (physical examination significant outflow stenosis)/1- of the outflow score, respectively, from all other measurements, with the area under the curve values of 0.8922 and 0.9618, respectively. With sPPL = 0 and PESOS/1- of the outflow score as diagnostic tools to detect inflow stenosis with an MLD of ⩽1.5 mm and outflow stenosis with an MLD of ⩽1.9 mm at vascular access sites, sensitivity = 86.00% and 88.46%; specificity = 97.67% and 92.11%; positive predictive values of 97.73% and 92.00% and negative predictive values of 85.71% and 88.61%, respectively, were observed. CONCLUSIONS: Our preliminary results showed that physical examination can potentially be a diagnostic tool in detecting patients with stenosis who are at a high risk of thrombosis at hemodialysis vascular access sites with high diagnostic accuracy.


Asunto(s)
Derivación Arteriovenosa Quirúrgica , Trombosis , Humanos , Constricción Patológica/etiología , Estudios Retrospectivos , Estudios de Casos y Controles , Examen Físico , Diálisis Renal/efectos adversos , Trombosis/diagnóstico por imagen , Trombosis/etiología , Derivación Arteriovenosa Quirúrgica/efectos adversos
17.
Am J Physiol Renal Physiol ; 303(8): F1230-8, 2012 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-22811485

RESUMEN

Kallistatin (KS) levels are reduced in the kidney and blood vessels under oxidative stress conditions. To determine the function of endogenous KS in the renal and cardiovascular systems, KS levels were depleted by daily injection of anti-rat KS antibody into DOCA-salt hypertensive rats for 10 days. Administration of anti-KS antibody resulted in reduced KS levels in the circulation but increased levels of serum thiobarbituric acid reactive substances (an indicator of lipid peroxidation) as well as superoxide formation in the aorta. Moreover, anti-KS antibody injection resulted in increased NADH oxidase activity and superoxide production but decreased nitric oxide levels in the kidney and heart. Endogenous KS blockade aggravated renal dysfunction, damage, hypertrophy, inflammation, and fibrosis as evidenced by decreased creatinine clearance and increased serum creatinine, blood urea nitrogen and urinary protein levels, tubular dilation, protein cast formation, glomerulosclerosis, glomerular enlargement, inflammatory cell accumulation, and collagen deposition. In addition, rats receiving anti-KS antibody had enhanced cardiac injury as indicated by cardiomyocyte hypertrophy, inflammation, myofibroblast accumulation, and fibrosis. Renal and cardiac injury caused by endogenous KS depletion was accompanied by increases in the expression of the proinflammatory genes tumor necrosis factor-α and intercellular adhesion molecule-1 and the profibrotic genes collagen I and III, transforming growth factor-ß, and tissue inhibitor of metalloproteinase-1. Taken together, these results implicate an important role for endogenous KS in protection against salt-induced renal and cardiovascular injury in rats by suppressing oxidative stress, inflammation, hypertrophy, and fibrosis.


Asunto(s)
Hipertensión/metabolismo , Inflamación/metabolismo , Enfermedades Renales/metabolismo , Estrés Oxidativo/fisiología , Serpinas/metabolismo , Animales , Desoxicorticosterona/farmacología , Hipertensión/patología , Inflamación/patología , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/patología , Enfermedades Renales/patología , Peroxidación de Lípido/efectos de los fármacos , Peroxidación de Lípido/fisiología , Masculino , Complejos Multienzimáticos/metabolismo , NADH NADPH Oxidorreductasas/metabolismo , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Wistar , Cloruro de Sodio Dietético/farmacología , Superóxidos/metabolismo
18.
Exp Cell Res ; 316(3): 376-89, 2010 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-19879874

RESUMEN

Biological functions of tissue kallikrein (TK, KLK1) are mainly mediated by kinin generation and subsequent kinin B2 receptor activation. In this study, we investigated the potential role of TK and its signaling pathways in cultured human keratinocyte migration and in a rat skin wound healing model. Herein, we show that TK promoted cell migration and proliferation in a concentration- and time-dependent manner. Inactive TK or kinin had no significant effect on cell migration. Interestingly, cell migration induced by active TK was not blocked by icatibant or L-NAME, indicating an event independent of kinin B2 receptor and nitric oxide formation. TK's stimulatory effect on cell migration was inhibited by small interfering RNA for proteinase-activated receptor 1 (PAR(1)), and by PAR(1) inhibitor. TK-induced migration was associated with increased phosphorylation of epidermal growth factor receptor (EGFR) and extracellular signal-regulated kinase (ERK), which was blocked by inhibition of protein kinase C (PKC), Src, EGFR and ERK. TK-induced cell migration and EGFR phosphorylation were blocked by metalloproteinase (MMP) inhibitor, heparin, and antibodies against EGFR external domain, heparin-binding EGF-like growth factor (HB-EGF) and amphiregulin (AR). Local application of TK promoted skin wound healing in rats, whereas icatibant and EGFR inhibitor blocked TK's effect. Skin wound healing was further delayed by aprotinin and neutralizing TK antibody. This study demonstrates a novel role of TK in skin wound healing and uncovers new signaling pathways mediated by TK in promoting keratinocyte migration through activation of the PAR(1)-PKC-Src-MMP pathway and HB-EGF/AR shedding-dependent EGFR transactivation.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Receptores ErbB/metabolismo , Queratinocitos/citología , Queratinocitos/enzimología , Receptor PAR-1/metabolismo , Transducción de Señal/efectos de los fármacos , Calicreínas de Tejido/farmacología , Animales , Proliferación Celular/efectos de los fármacos , Receptores ErbB/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Queratinocitos/efectos de los fármacos , Cininas/metabolismo , Ligandos , Metaloproteinasas de la Matriz/metabolismo , Óxido Nítrico/metabolismo , Fosforilación/efectos de los fármacos , Proteína Quinasa C/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Ratas , Receptor de Bradiquinina B2/metabolismo , Piel/efectos de los fármacos , Piel/patología , Activación Transcripcional/efectos de los fármacos , Activación Transcripcional/genética , Cicatrización de Heridas/efectos de los fármacos
19.
J Biol Chem ; 284(51): 35471-8, 2009 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-19858207

RESUMEN

Kallistatin is a plasma protein that exhibits pleiotropic effects in vasodilation, anti-angiogenesis, and anti-inflammation. To isolate a kallistatin-binding protein that mediates the vascular actions of kallistatin, we screened and identified a positive clone from a human heart cDNA expression library by using an alkaline phosphatase-kallistatin fusion protein binding assay. Sequence analysis revealed that kallistatin-binding protein is human Kruppel-like factor 4 (KLF4). KLF4 was localized on the plasma membrane of HEK-293 cells and endothelial cells overexpressing KLF4. KLF4 and kallistatin complex formation was identified in endothelial cells by immunoprecipitation followed by immunoblotting. We showed that kallistatin inhibits tumor necrosis factor-alpha-induced NF-kappaB activation, as well as vascular cell adhesion molecule-1 and monocyte chemoattractant protein-1 expression in endothelial cells, whereas knockdown of KLF4 by small interfering RNA oligonucleotide abolished the effect of kallistatin. Kallistatin increased endothelial nitric-oxide synthase (eNOS) expression and nitric oxide levels, and these effects were also blocked by KLF4 small interfering RNA oligonucleotide. Moreover, inhibition of eNOS by RNA interference or by NOS inhibitor abolished the blocking effect of kallistatin on vascular cell adhesion molecule-1 and monocyte chemoattractant protein-1 expression. In summary, we identified KLF4 as a kallistatin-binding protein, which has a novel role in mediating the anti-inflammatory actions of kallistatin via increasing eNOS expression in endothelial cells. This study provides a new target for modulating endothelial function in vascular disease.


Asunto(s)
Endotelio Vascular/metabolismo , Regulación Enzimológica de la Expresión Génica , Factores de Transcripción de Tipo Kruppel/metabolismo , Óxido Nítrico Sintasa de Tipo III/biosíntesis , Serpinas/metabolismo , Línea Celular , Membrana Celular/genética , Membrana Celular/metabolismo , Membrana Celular/patología , Quimiocina CCL2/metabolismo , Quimiocina CCL2/farmacología , Endotelio Vascular/patología , Inhibidores Enzimáticos/farmacología , Técnicas de Silenciamiento del Gen , Biblioteca de Genes , Humanos , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Miocardio/metabolismo , Miocardio/patología , FN-kappa B/genética , FN-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo III/genética , Unión Proteica , Serpinas/genética , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Molécula 1 de Adhesión Celular Vascular/biosíntesis , Molécula 1 de Adhesión Celular Vascular/genética , Enfermedades Vasculares/genética , Enfermedades Vasculares/metabolismo , Enfermedades Vasculares/patología
20.
Am J Physiol Renal Physiol ; 298(4): F1033-40, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20089675

RESUMEN

Levels of tissue kallikrein (TK) are significantly lower in the urine of patients with kidney failure, and TK expression is specifically diminished in rat kidney after recovery from ischemia-reperfusion injury. In this study, we investigated the functional consequence of blocking endogenous TK activity in a rat model of chronic kidney disease. Inhibition of endogenous TK levels for 10 days by neutralizing TK antibody injection in DOCA-salt rats caused a significant increase in blood urea nitrogen and urinary protein levels, and a decrease in creatinine clearance. Kidney sections from anti-TK antibody-treated rats displayed a marked rise in tubular dilation and protein cast accumulation as well as glomerular sclerosis and size. TK blockade also increased inflammatory cell infiltration, myofibroblast and collagen accumulation, and collagen fraction volume. Elevated renal inflammation and fibrosis by anti-TK antibody were associated with increased expression of tumor necrosis factor-alpha, intercellular adhesion molecule-1, tissue inhibitor of metalloproteinase-2 (TIMP-2), and plasminogen activator inhibitor-1 (PAI-1). Moreover, the detrimental effect of TK blockade resulted in reduced nitric oxide (NO) levels as well as increased serum lipid peroxidation, renal NADH oxidase activity, and superoxide formation. In cultured proximal tubular cells, TK inhibited angiotensin II-induced superoxide production and NADH oxidase activity via NO formation. In addition, TK markedly increased matrix metalloproteinase-2 activity with a parallel reduction of TIMP-2 and PAI-1 synthesis. These findings indicate that endogenous TK has the propensity to preserve kidney structure and function in rats with chronic renal disease by inhibiting oxidative stress and activating matrix degradation pathways.


Asunto(s)
Enfermedades Renales/metabolismo , Calicreínas de Tejido/antagonistas & inhibidores , Angiotensina II , Animales , Anticuerpos , Línea Celular , Humanos , Túbulos Renales Proximales/citología , Masculino , Metaloproteasas/metabolismo , Estrés Oxidativo , Inhibidor 1 de Activador Plasminogénico/genética , Inhibidor 1 de Activador Plasminogénico/metabolismo , Ratas , Ratas Endogámicas WF , Especies Reactivas de Oxígeno , Inhibidor Tisular de Metaloproteinasa-2/genética , Inhibidor Tisular de Metaloproteinasa-2/metabolismo , Calicreínas de Tejido/metabolismo , Factor de Crecimiento Transformador beta/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA