Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
FASEB J ; 34(12): 16163-16178, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33063394

RESUMEN

Hepatitis is an important health problem worldwide. Novel molecular targets are in demand for detection and management of hepatitis. Hepatoma-derived growth factor (HDGF) has been delineated to participate in hepatic fibrosis and liver carcinogenesis. However, the relationship between hepatitis and HDGF remains unclear. This study aimed to elucidate the role of HDGF during hepatitis using concanavalin A (ConA)-induced hepatitis model. In cultured hepatocytes, ConA treatment-elicited HDGF upregulation at transcriptional level and promoted HDGF secretion while reducing intracellular HDGF protein level and cellular viability. Similarly, mice receiving ConA administration exhibited reduced hepatic HDGF expression and elevated circulating HDGF level, which was positively correlated with serum aspartate aminotransferase (AST) and alanine aminotransferase (ALT) levels. By using HDGF knockout (KO) mice, it was found the ConA-evoked cell death was prominently alleviated in KO compared with control. Besides, it was delineated HDGF ablation conferred protection by suppressing the ConA-induced neutrophils recruitment in livers. Above all, the ConA-mediated activation of tumor necrosis factor-α (TNF-α)/interleukin-1ß (IL-1ß)/interleukin-6 (IL-6)/cyclooxygenase-2 (COX-2) inflammatory signaling was significantly abrogated in KO mice. Treatment with recombinant HDGF (rHDGF) dose-dependently stimulated the expression of TNF-α/IL-1ß/IL-6/COX-2 in hepatocytes, further supporting the pro-inflammatory function of HDGF. Finally, application of HDGF antibody not only attenuated the ConA-mediated inflammatory cascade in hepatocytes, but also ameliorated the ConA-induced hepatic necrosis and AST elevation in mice. In summary, HDGF participates in ConA-induced hepatitis via neutrophils recruitment and may constitute a therapeutic target for acute hepatitis.


Asunto(s)
Concanavalina A/farmacología , Hepatitis Animal/inducido químicamente , Hepatitis Animal/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Alanina Transaminasa/metabolismo , Animales , Aspartato Aminotransferasas/metabolismo , Células Cultivadas , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Cirrosis Hepática/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infiltración Neutrófila/efectos de los fármacos , Ratas , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba/efectos de los fármacos
2.
Biochem Pharmacol ; 178: 114064, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32492449

RESUMEN

Osteosarcoma (OS) is the most common solid tumor of the bone that most often affects adolescents. The introduction of chemotherapy for the treatment of OS has largely improved the survival rates of patients with localized tumors. However, the 5-year survival rate of OS patients with relapsed or metastatic disease is only 10 to 20%. In this study, the antimicrobial peptide tilapia piscidin 3 (TP3), isolated from Nile tilapia (Oreochromis niloticus), was treated to OS MG63 cells. Our findings showed that TP3 concentration as low as 1 µM induced significant inhibition of cell viability and increased DNA fragmentation, as determined by the MTT and TUNEL assays, respectively. The protein expression levels of cleaved caspases 3/9 were increased. An in situ live-cell time-lapse video and cell tomographic microscopy images showed cellular blebbing, shrinkage, nuclear fragmentation, and chromatin condensation, with the formation of beaded apoptopodia. Moreover, there were significant increase in the production of TP3-induced mitochondrial and cellular reactive oxygen species (ROS), as well as down-regulated mitochondrial oxygen consumption and extracellular acidification rates. Additionally, TP3 enhanced mitochondrial fission, whereas fusion was attenuated. Furthermore, after administration of the mitochondria targeted antioxidant mitoTempo, TP3-induced ROS oxidant levels and alterations in cleaved caspases 3/9 expression were rescued. TP3 promoted mitochondria-modulated intrinsic apoptosis through the induction of ROS production, activation of caspases 3/9, and the down-regulation of mitochondrial oxygen consumption and extracellular acidification rates, suggesting that TP3 has potential as an innovative alternative for OS treatment.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/farmacología , Apoptosis/efectos de los fármacos , Proteínas de Peces/farmacología , Mitocondrias/efectos de los fármacos , Osteosarcoma , Microambiente Tumoral/efectos de los fármacos , Animales , Péptidos Catiónicos Antimicrobianos/aislamiento & purificación , Péptidos Catiónicos Antimicrobianos/uso terapéutico , Apoptosis/fisiología , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Proteínas de Peces/aislamiento & purificación , Proteínas de Peces/uso terapéutico , Humanos , Mitocondrias/patología , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/patología , Tilapia , Microambiente Tumoral/fisiología
3.
Int J Mol Sci ; 19(12)2018 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-30513637

RESUMEN

α-melanocyte-stimulating hormone (α-MSH) has been characterized as a novel angiogenesis inhibitor. The homeostasis of nitric oxide (NO) plays an important role in neovascularization. However, it remains unclear whether α-MSH mitigates angiogenesis through modulation of NO and its signaling pathway. The present study elucidated the function and mechanism of NO signaling in α-MSH-induced angiogenesis inhibition using cultured human umbilical vein endothelial cells (HUVECs), rat aorta rings, and transgenic zebrafish. By Griess reagent assay, it was found α-MSH dose-dependently reduced the NO release in HUVECs. Immunoblotting and immunofluorescence analysis revealed α-MSH potently suppressed endothelial and inducible nitric oxide synthase (eNOS/iNOS) expression, which was accompanied with inhibition of nuclear factor kappa B (NF-κB) activities. Excessive supply of NO donor l-arginine reversed the α-MSH-induced angiogenesis inhibition in vitro and in vivo. By using antibody neutralization and RNA interference, it was delineated that melanocortin-1 receptor (MC1-R) and melanocortin-2 receptor (MC2-R) participated in α-MSH-induced inhibition of NO production and NF-κB/eNOS/iNOS signaling. This was supported by pharmaceutical inhibition of protein kinase A (PKA), the downstream effector of MC-Rs signaling, using H89 abolished the α-MSH-mediated suppression of NO release and eNOS/iNOS protein level. Therefore, α-MSH exerts anti-angiogenic function by perturbing NO bioavailability and eNOS/iNOS expression in endothelial cells.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , FN-kappa B/metabolismo , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Receptores de Melanocortina/metabolismo , alfa-MSH/uso terapéutico , Células Endoteliales de la Vena Umbilical Humana , Humanos , Óxido Nítrico , Interferencia de ARN , Transducción de Señal/efectos de los fármacos
4.
Int J Mol Sci ; 19(10)2018 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-30274378

RESUMEN

Choroidal neovascularization (CNV) is a key pathological feature of several leading causes of vision loss including neovascular age-related macular degeneration. Here, we show that a calreticulin anti-angiogenic domain (CAD)-like peptide 27, CAD27, inhibited in vitro angiogenic activities, including tube formation, migration of endothelial cells, and vascular sprouting from rat aortic ring explants. In a rat model of laser-induced CNV, we demonstrate that intravitreal injection of CAD27 significantly attenuated the formation of CNV lesions as measured via fundus fluorescein angiography and choroid flat-mounts (19.5% and 22.4% reductions at 10 µg and 20 µg of CAD27 injected, respectively). Similarly, the reduction of CNV lesions was observed in rats that had received topical applications of CAD27 (choroid flat-mounts: 17.9% and 32.5% reductions at 10 µg/mL and 20 µg/mL of CAD27 instilled, respectively). Retinal function was unaffected, as measured using electroretinography in both groups receiving interareal injection or topical applications of CAD27 for at least fourteen days. These findings show that CAD27 can be used as a potential therapeutic alternative for targeting CNV in diseases such as neovascular age-related macular degeneration.


Asunto(s)
Calreticulina/química , Neovascularización Coroidal/tratamiento farmacológico , Péptidos/uso terapéutico , Administración Tópica , Secuencia de Aminoácidos , Inhibidores de la Angiogénesis/farmacología , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Aorta/patología , Neovascularización Coroidal/patología , Neovascularización Coroidal/fisiopatología , Angiografía con Fluoresceína , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Inyecciones Intravítreas , Rayos Láser , Masculino , Neovascularización Fisiológica/efectos de los fármacos , Péptidos/administración & dosificación , Péptidos/química , Péptidos/farmacología , Dominios Proteicos , Ratas Sprague-Dawley , Retina/efectos de los fármacos , Retina/patología , Retina/fisiopatología
5.
Cancer Med ; 7(6): 2567-2580, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29683262

RESUMEN

Epirubicin is a chemotherapy agent for hepatocellular carcinoma (HCC). However, the outcome of HCC patients receiving epirubicin remains unsatisfactory. Moreover, our previous study indicated that celecoxib suppresses HCC progression and liver cancer stemness. This study evaluated the potential of celecoxib to serve as a complementary therapy during epirubicin treatment. Cell proliferation, apoptosis, invasiveness, and anchorage-independent growth were analyzed in hepatoma cells. Therapeutic efficacy was validated in rat orthotopic Novikoff hepatoma. After animal sacrifice, the antitumor mechanism of celecoxib and epirubicin combined therapy was investigated by histological analysis. Celecoxib enhanced the cytotoxic activity of epirubicin in HCC cells by promoting apoptosis. Besides, celecoxib potentiated the antineoplastic function of epirubicin in inhibiting the invasiveness and anchorage-independent growth of HCC cells. Ultrasound monitoring showed that combined therapy was more potent than either therapy alone in perturbing HCC progression. Consistently, the size and weight of dissected HCC tissues from rats receiving combined therapy were smallest among all groups. HCC treated with combined therapy exhibited the highest prevalence of apoptotic cells, which was accompanied by reduced proliferating and angiogenic activities in tumor tissues. Moreover, the expression levels of cancer stemness markers (CD44 and CD133) and drug transporter MDR-1 were significantly diminished in rats receiving combined therapy. Besides, celecoxib treatment increased the infiltration of cytotoxic T lymphocytes (CTLs) and reduced the number of regulatory T cells (Tregs), tumor-associated macrophages (TAMs), and the expression of immune checkpoint PD-L1 in HCC tissues during epirubicin therapy. Celecoxib augmented the therapeutic efficacy while modulated cancer stemness and antitumor immunity. Thus, celecoxib may serve as complementary therapy to improve the outcome of patients with advanced HCC during epirubicin treatment.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Celecoxib/farmacología , Inhibidores de la Ciclooxigenasa 2/farmacología , Epirrubicina/farmacología , Inhibidores de Topoisomerasa II/farmacología , Animales , Apoptosis/efectos de los fármacos , Biomarcadores , Carcinoma Hepatocelular/diagnóstico por imagen , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/patología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Humanos , Inmunomodulación/efectos de los fármacos , Neoplasias Hepáticas Experimentales , Ratas , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Mar Drugs ; 13(2): 861-78, 2015 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-25668036

RESUMEN

BACKGROUND: WA-25 (dihydroaustrasulfone alcohol, a synthetic derivative of marine compound WE-2) suppresses atherosclerosis in rats by reducing neointima formation. Because angiogenesis plays a critical role in the pathogenesis of atherosclerosis, the present study investigated the angiogenic function and mechanism of WA-25. METHODS: The angiogenic effect of WA-25 was evaluated using a rat aortic ring assay and transgenic zebrafish models were established using transgenic Tg(fli-1:EGFP)y1 and Tg(kdrl:mCherryci5-fli1a:negfpy7) zebrafish embryos. In addition, the effect of WA-25 on distinct angiogenic processes, including matrix metalloproteinase (MMP) expression, endothelial cell proliferation and migration, as well as tube formation, was studied using human umbilical vein endothelial cells (HUVECs). The effect of WA-25 on the endothelial vascular endothelial growth factor (VEGF) signaling pathway was elucidated using qRT-PCR, immunoblot analysis, immunofluorescence and flow cytometric analyses. RESULTS: The application of WA-25 perturbed the development of intersegmental vessels in transgenic zebrafish. Moreover, WA-25 potently suppressed microvessel sprouting in organotypic rat aortic rings. Among cultured endothelial cells, WA-25 significantly and dose-dependently inhibited MMP-2/MMP-9 expression, proliferation, migration and tube formation in HUVECs. Mechanistic studies revealed that WA-25 significantly reduced the VEGF release by reducing VEGF expression at the mRNA and protein levels. In addition, WA-25 reduced surface VEGF receptor 2 (VEGFR2/Flk-1) expression by repressing the VEGFR2 mRNA level. Finally, an exogenous VEGF supply partially rescued the WA-25-induced angiogenesis blockage in vitro and in vivo. CONCLUSIONS: WA-25 is a potent angiogenesis inhibitor that acts through the down-regulation of VEGF and VEGFR2 in endothelial cells. GENERAL SIGNIFICANCE: WA-25 may constitute a novel anti-angiogenic drug that acts by targeting endothelial VEGF/VEGFR2 signaling.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Antozoos/química , Butanonas/farmacología , Sulfonas/farmacología , Factor A de Crecimiento Endotelial Vascular/efectos de los fármacos , Receptor 2 de Factores de Crecimiento Endotelial Vascular/efectos de los fármacos , Animales , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Masculino , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Neovascularización Patológica/prevención & control , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Pez Cebra
7.
Int J Mol Sci ; 15(9): 16611-27, 2014 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-25244013

RESUMEN

Angiogenesis, the process of neovascularization, plays an important role in physiological and pathological conditions. ST104P is a soluble polysulfated-cyclo-tetrachromotropylene compound with anti-viral and anti-thrombotic activities. However, the functions of ST104P in angiogenesis have never been explored. In this study, we investigated the effects of ST104P in angiogenesis in vitro and in vivo. Application of ST104P potently suppressed the microvessels sprouting in aortic rings ex vivo. Furthermore, ST104P treatment significantly disrupted the vessels' development in transgenic zebrafish in vivo. Above all, repeated administration of ST104P resulted in delayed tumor growth and prolonged the life span of mice bearing Lewis lung carcinoma. Mechanistic studies revealed that ST104P potently inhibited the migration, tube formation and wound closure of human umbilical endothelial cells (HUVECs). Moreover, ST104P treatment inhibited the secretion and expression of matrix metalloproteinase-2 (MMP-2) in a dose-dependent manner. Together, these results suggest that ST104P is a potent angiogenesis inhibitor and may hold potential for treatment of diseases due to excessive angiogenesis including cancer.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Endotelio Vascular/efectos de los fármacos , Compuestos Macrocíclicos/farmacología , Metaloproteinasa 2 de la Matriz/biosíntesis , Naftalenosulfonatos/farmacología , Neovascularización Fisiológica/efectos de los fármacos , Inhibidores de la Angiogénesis/química , Inhibidores de la Angiogénesis/uso terapéutico , Inhibidores de la Angiogénesis/toxicidad , Animales , Animales Modificados Genéticamente , Aorta , Carcinoma Pulmonar de Lewis/irrigación sanguínea , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Movimiento Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Embrión no Mamífero/irrigación sanguínea , Embrión no Mamífero/efectos de los fármacos , Endotelio Vascular/enzimología , Endotelio Vascular/metabolismo , Inducción Enzimática/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Compuestos Macrocíclicos/química , Compuestos Macrocíclicos/uso terapéutico , Compuestos Macrocíclicos/toxicidad , Metaloproteinasa 2 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Morfogénesis/efectos de los fármacos , Naftalenosulfonatos/química , Naftalenosulfonatos/uso terapéutico , Naftalenosulfonatos/toxicidad , Neovascularización Patológica/tratamiento farmacológico , Pez Cebra/embriología
8.
Oncotarget ; 5(6): 1475-90, 2014 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-24721996

RESUMEN

Celecoxib, a COX-2 inhibitor and non-steroidal anti-inflammatory drug, can prevent several types of cancer, including hepatocellular carcinoma (HCC). Here we show that celecoxib suppressed the self-renewal and drug-pumping functions in HCC cells. Besides, celecoxib depleted CD44+/CD133+ hepatic cancer stem cells (hCSC). Prostaglandin E2 (PGE2) and CD133 overexpression did not reverse the celecoxib-induced depletion of hCSC. Also, celecoxib inhibited progression of rat Novikoff hepatoma. Moreover, a 60-day celecoxib program increased the survival rate of rats with hepatoma. Histological analysis revealed that celecoxib therapy reduced the abundance of CD44+/CD133+ hCSCs in hepatoma tissues. Besides, the hCSCs depletion was associated with elevated apoptosis and blunted proliferation and angiogenesis in hepatoma. Celecoxib therapy activated peroxisome proliferator-activated receptor γ (PPARγ) and up-regulated PTEN, thereby inhibiting Akt and disrupting hCSC expansion. PTEN gene delivery by adenovirus reduced CD44/CD133 expression in vitro and hepatoma formation in vivo. This study suggests that celecoxib suppresses cancer stemness and progression of HCC via activation of PPARγ/PTEN signaling.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Inhibidores de la Ciclooxigenasa 2/farmacología , Neoplasias Hepáticas/tratamiento farmacológico , Fosfohidrolasa PTEN/metabolismo , Pirazoles/farmacología , Sulfonamidas/farmacología , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Celecoxib , Proliferación Celular/efectos de los fármacos , Ciclooxigenasa 2/química , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Dinoprostona/metabolismo , Progresión de la Enfermedad , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Receptores de Hialuranos/metabolismo , Técnicas para Inmunoenzimas , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Masculino , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , PPAR gamma/genética , PPAR gamma/metabolismo , Fosfohidrolasa PTEN/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Activación Transcripcional , Células Tumorales Cultivadas , Regulación hacia Arriba
9.
Biochim Biophys Acta ; 1840(6): 1850-60, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24530634

RESUMEN

BACKGROUND: Gene therapy of proopiomelanocortin, the precursor of α-melanocyte-stimulating hormone (α-MSH), suppresses the neovascularization in tumors. However, the roles of α-MSH in angiogenesis remain unclear. METHODS: The influence of α-MSH on angiogenesis was evaluated by ex vivo rat aorta and in vivo, including transgenic zebrafish and chicken chorioallantoic membrane (CAM) assays. The effect of α-MSH on proliferation, matrix metalloproteinase (MMP) secretion, migration and tube formation was examined using human umbilical vein endothelial cells (HUVECs). The expression of vascular endothelial growth factor (VEGF) and VEGF receptor 2 (VEGFR2) was investigated by quantitative RT-PCR, immunoblot and immunofluorescent analysis. Antibodies' neutralization was employed to dissect the receptor(s) transmitting α-MSH signaling. RESULTS: Application of α-MSH potently suppressed the microvessels sprouting in organotypic aortic rings. Besides, α-MSH perturbed the vessels development in zebrafish and chicken embryos. α-MSH (0.01-10nM) inhibited the MMP-2 secretion, migration and tube formation of HUVECs without affecting proliferation. Mechanistic studies unveiled α-MSH decreased the VEGF expression and release in HUVECs. Besides, α-MSH downregulated the VEGFR2 expression at transcriptional and translational levels. Importantly, α-MSH attenuated the Akt phosphorylation, but enhanced the expression of PTEN, endogenous antagonist of PI3K/Akt signaling. Expression analysis and antibody neutralization revealed that MC1-R and MC2-R participated in α-MSH-induced blockage of migration and VEGF/VEGFR2/Akt signaling. However, VEGF supply failed to reverse the anti-angiogenic function of α-MSH. CONCLUSIONS: α-MSH inhibits the physiological angiogenesis by attenuating VEGF/VEGFR2/Akt signaling in endothelial cells. GENERAL SIGNIFICANCE: α-MSH is a potent angiogenesis inhibitor targeting at endothelial VEGF/VEGFR2 signaling, which may have potential for therapeutic application.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Transducción de Señal/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/fisiología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/fisiología , alfa-MSH/farmacología , Animales , Células Cultivadas , Humanos , Masculino , Neovascularización Fisiológica/efectos de los fármacos , Fosfohidrolasa PTEN/análisis , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor de Melanocortina Tipo 1/fisiología , Receptor de Melanocortina Tipo 2/fisiología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Pez Cebra
10.
Biochem Biophys Res Commun ; 425(2): 169-76, 2012 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-22819846

RESUMEN

Hepatoma-derived growth factor (HDGF) stimulates the migration, invasion and metastasis in several types of cancer cells. However, the mechanism underlying HDGF-stimulated migration remains unclear. In this study, we investigated the influence of HDGF on cytoskeleton remodeling and phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway in non-transformed NIH/3T3 cells. Exogenous HDGF promoted the migration and the formation of dorsal ruffles and podosome rosettes. Besides, HDGF supply increased the PI3K expression and Akt phosphorylation in dose- and time-dependent manners. Application of LY294002, a PI3K inhibitor, attenuated the HDGF-induced migration, dorsal ruffles and podosome rosettes formation. Consistently, the HDGF-overexpressing NIH/3T3 transfectants exhibited significantly increased motility and elevated PI3K/Akt activities, which were repressed by LY294002 or adenovirus-mediated overexpression of endogenous PI3K antagonist, PTEN. In summary, HDGF elicits the activation of PI3K/Akt signaling cascade, thereby promoting cytoskeleton remodeling to stimulate cellular migration.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/fisiología , Fosfatidilinositol 3-Quinasa/biosíntesis , Proteínas Proto-Oncogénicas c-akt/biosíntesis , Formación de Roseta , Animales , Movimiento Celular , Citoesqueleto/genética , Citoesqueleto/metabolismo , Activación Enzimática , Péptidos y Proteínas de Señalización Intercelular/genética , Ratones , Células 3T3 NIH , Transfección
11.
Artif Organs ; 36(1): 78-86, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21752037

RESUMEN

Dialysis-related amyloidosis (DRA), caused by the accumulation of beta-2-microglobulin (ß-2M), remains a major concern in long-term renal replacement therapies. For years, we have developed an immunoadsorption wall (iWall) for the removal of ß-2M. In this study, we employed a new approach taking advantage of the melting of a buffer ice rod to improve the conditions associated with the manufacturing of an iWall and tested its performance with uremic serum and blood. The preliminary results reveal that the present iWalls thus prepared not only possess the superior properties of affinity and specificity but also show structural stability and acceptable hemocompatibility. We believe that this breakthrough might provide a promising path to successful treatment of DRA as well as establish a useful platform for studying removal of certain pathological toxins accumulated in the blood.


Asunto(s)
Amiloidosis/prevención & control , Diálisis/métodos , Técnicas de Inmunoadsorción , Desintoxicación por Sorción/métodos , Uremia/sangre , Microglobulina beta-2/metabolismo , Amiloidosis/sangre , Amiloidosis/etiología , Diálisis/efectos adversos , Diálisis/instrumentación , Humanos , Técnicas In Vitro , Ensayo de Materiales , Uremia/complicaciones , Microglobulina beta-2/química
12.
Mol Vis ; 16: 756-67, 2010 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-20454694

RESUMEN

PURPOSE: Vasostatin 48 (VS48) is a peptide of 48 amino acids derived from calreticulin. This study aimed to investigate the effects of topical application of VS48 eyedrops on experimental choroidal neovascularization (CNV). METHODS: Recombinant VS48 was expressed and purified as a thioredoxin (TRX)-fused protein, TRX-VS48. The anti-angiogenic effects of TRX-VS48 were validated by migration and tube formation assays performed on cultured endothelial cells, and by rat aorta ring assays. CNV lesions were created in Brown Norway rats by laser-induced photocoagulation at day 1. After topical TRX-VS48 application for 21 days, the CNV lesions were monitored via either choroidal flat mounts on day 21 or by fluorescent angiography on days 21, 28, 35, and 42. CNV lesions were evaluated by histological analysis. The retinal function of animals was examined by electroretinogram (ERG) to evaluate the safety and therapeutic efficacy of TRX-VS48. RESULTS: Application of TRX-VS48 inhibited the migration and tube formation of endothelial cells. TRX-VS48 inhibited the growth of sprouting vessels in aorta rings. ERG analysis revealed that topical TRX-VS48 application for 21 days had no effect on rat retinal functions. After CNV induction, topical TRX-VS48 application for 21 days significantly reduced the size of CNV, as assayed by flat mounts. Fluorescent angiography revealed that the CNV areas in TRX-VS48-treated eyes were significantly reduced compared with TRX-treated eyes on days 21, 28, 35, and 42. Histological analysis also revealed attenuated CNV lesions in TRX-VS48-treated eyes. Topical TRX-VS48 treatment significantly reversed the CNV-induced alterations in ERG parameters on day 35. CONCLUSIONS: Topical TRX-VS48 application suppressed laser-induced CNV in rats, thereby constituting a possible modality for ocular diseases due to excessive angiogenesis.


Asunto(s)
Inhibidores de la Angiogénesis/administración & dosificación , Calreticulina/administración & dosificación , Neovascularización Coroidal/diagnóstico , Neovascularización Coroidal/etiología , Rayos Láser , Fragmentos de Péptidos/administración & dosificación , Traumatismos por Radiación/complicaciones , Animales , Aorta/efectos de los fármacos , Aorta/patología , Calreticulina/efectos adversos , Línea Celular , Movimiento Celular/efectos de los fármacos , Neovascularización Coroidal/fisiopatología , Electrorretinografía , Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Angiografía con Fluoresceína , Humanos , Técnicas In Vitro , Masculino , Soluciones Oftálmicas , Fragmentos de Péptidos/efectos adversos , Ratas , Ratas Endogámicas BN , Ratas Sprague-Dawley , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/efectos adversos , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/efectos adversos , Retina/efectos de los fármacos , Retina/fisiopatología , Neovascularización Retiniana/diagnóstico , Tiorredoxinas/administración & dosificación , Tiorredoxinas/efectos adversos
13.
Mol Vis ; 15: 1897-905, 2009 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-19768130

RESUMEN

PURPOSE: Choroidal neovascularization (CNV) is the leading cause of blindness in patients with age-related macular degeneration (AMD). This study evaluated the inhibitory effect of vasostatin (VS), an endogenous angiogenesis inhibitor, on CNV. METHODS: Anti-angiogenic activity of VS was evaluated in vitro by migration and tube formation assays in human umbilical vein endothelial cells (HUVECs). CNV lesions were induced in Brown Norway rats by fundus argon laser photocoagulation. Beginning one day after CNV induction, rats were treated with eye drops containing 1 microg/ml VS in PBS buffer for three times daily for 20 days. The extent of CNV was examined by flat mount analysis on day 24 or by fundus fluorescein angiography (FAG) on days 21, 28, 35, and 42, respectively. CNV lesions and choroidal vascularity were evaluated by histological analysis. The spatial distribution of topically applied VS in rat eyes was evaluated by immunoblot analysis. RESULTS: VS inhibited migration and tube formation in HUVECs. Flat mount analysis revealed that, after laser-induced photocoagulation, topical VS application for 20 days significantly reduced CNV lesions. Moreover, serial FAG analysis indicated that a 20 day VS treatment significantly reduced CNV lesions on all subsequent days. Histological analysis revealed attenuated lesions, intact Bruch's membrane, and reduced choroidal vascularity in VS-treated eyes. Finally immunoblot analysis reveled VS expression in choroids. CONCLUSIONS: Topical VS application suppresses the progression of laser-induced CNV via angiogenesis inhibition and may constitute a therapeutic alternative for excessive neovascularization occurring with ocular diseases.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Calreticulina/farmacología , Neovascularización Coroidal/prevención & control , Fragmentos de Péptidos/farmacología , Administración Tópica , Animales , Movimiento Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Angiografía con Fluoresceína , Técnica del Anticuerpo Fluorescente , Humanos , Immunoblotting , Rayos Láser , Neovascularización Fisiológica/efectos de los fármacos , Ratas , Ratas Endogámicas BN , Retina/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...