Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Front Oncol ; 13: 1252014, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37909014

RESUMEN

Radiation treatment is one of the most frequently used therapies in patients with cancer, employed in approximately half of all patients. However, the use of radiation therapy is limited by acute or chronic adverse effects and the failure to consider the tumor microenvironment. Blood vessels substantially contribute to radiation responses in both normal and tumor tissues. The present study employed a three-dimensional (3D) microvasculature-on-a-chip that mimics physiological blood vessels to determine the effect of radiation on blood vessels. This model represents radiation-induced pathophysiological effects on blood vessels in terms of cellular damage and structural and functional changes. DNA double-strand breaks (DSBs), apoptosis, and cell viability indicate cellular damage. Radiation-induced damage leads to a reduction in vascular structures, such as vascular area, branch length, branch number, junction number, and branch diameter; this phenomenon occurs in the mature vascular network and during neovascularization. Additionally, vasculature regression was demonstrated by staining the basement membrane and microfilaments. Radiation exposure could increase the blockage and permeability of the vascular network, indicating that radiation alters the function of blood vessels. Radiation suppressed blood vessel recovery and induced a loss of angiogenic ability, resulting in a network of irradiated vessels that failed to recover, deteriorating gradually. These findings demonstrate that this model is valuable for assessing radiation-induced vascular dysfunction and acute and chronic effects and can potentially improve radiotherapy efficiency.

2.
Tissue Eng Regen Med ; 20(4): 593-605, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37195569

RESUMEN

BACKGROUND: Tissue engineering, including 3D bioprinting, holds great promise as a therapeutic tool for repairing cartilage defects. Mesenchymal stem cells have the potential to treat various fields due to their ability to differentiate into different cell types. The biomimetic substrate, such as scaffolds and hydrogels, is a crucial factor that affects cell behavior, and the mechanical properties of the substrate have been shown to impact differentiation during incubation. In this study, we examine the effect of the mechanical properties of the 3D printed scaffolds, made using different concentrations of cross-linker, on hMSCs differentiation towards chondrogenesis. METHODS: The 3D scaffold was fabricated using 3D bioprinting technology with gelatin/hyaluronic acid (HyA) biomaterial ink. Crosslinking was achieved by using different concentrations of 4-(4,6-dimethoxy-1,3,5-triazin-2-yl)-4-methlymorpholinium chloride n-hydrate (DMTMM), allowing for control of the scaffold's mechanical properties. The printability and stability were also evaluated based on the concentration of DMTMM used. The effects of the gelatin/HyA scaffold on chondrogenic differentiation was analyzed by utilizing various concentrations of DMTMM. RESULTS: The addition of HyA was found to improve the printability and stability of 3D printed gelatin/HyA scaffolds. The mechanical properties of the 3D gelatin/HyA scaffold could be regulated through the use of different concentrations of DMTMM cross-linker. In particular, the use of 0.25 mM DMTMM for crosslinking the 3D gelatin/HyA scaffold resulted in enhanced chondrocyte differentiation. CONCLUSION: The mechanical properties of 3D printed gelatin/HyA scaffolds cross-linked using various concentrations of DMTMM can influence the differentiation of hMSCs into chondrocytes.


Asunto(s)
Células Madre Mesenquimatosas , Andamios del Tejido , Andamios del Tejido/química , Gelatina/química , Ácido Hialurónico/farmacología , Condrogénesis , Diferenciación Celular , Células Madre Mesenquimatosas/metabolismo , Impresión Tridimensional
3.
Anticancer Res ; 43(5): 1973-1980, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-37097659

RESUMEN

BACKGROUND/AIM: The fibroblast growth factor receptor (FGFR) signaling pathway is abnormally activated in human cancers, including breast cancer. Therefore, targeting the FGFR signaling pathway is a potent strategy to treat breast cancer. The purpose of this study was to find drugs that could increase sensitivity to FGFR inhibitor effects in BT-474 breast cancer cells, and to investigate the combined effects and underlying mechanisms of these combinations for BT-474 breast cancer cell survival. MATERIALS AND METHODS: Cell viability was measured by MTT assay. Protein expression was determined by western blot analysis. mRNA expression was detected by Real-time PCR. Drug synergy effect was determined by isobologram analysis. RESULTS: Nebivolol, a third generation ß1-blocker, synergistically increased the sensitivity of BT-474 breast cancer cells to the potent and selective FGFR inhibitors erdafitinib (JNJ-42756493) and AZD4547. A combination of nebivolol and erdafitinib markedly reduced AKT activation. Suppression of AKT activation using specific siRNA and a selective inhibitor further enhanced cell sensitivity to combined treatment with nebivolol and erdafitinib, whereas SC79, a potent activator of AKT, reduced cell sensitivity to nebivolol and erdafitinib. CONCLUSION: Enhanced sensitivity of BT-474 breast cancer cells to nebivolol and erdafitinib was probably associated with down-regulation of AKT activation. Combined treatment with nebivolol and erdafitinib is a promising strategy for breast cancer treatment.


Asunto(s)
Neoplasias de la Mama , Humanos , Femenino , Nebivolol/farmacología , Nebivolol/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal , Inhibidores de Proteínas Quinasas/farmacología , Línea Celular Tumoral
4.
Cancers (Basel) ; 14(23)2022 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-36497331

RESUMEN

G protein-coupled receptors (GPCRs) are a diverse family of cell surface receptors implicated in various physiological functions, making them common targets for approved drugs. Many GPCRs are abnormally activated in cancers and have emerged as therapeutic targets for cancer. Neuropeptide FF receptor 2 (NPFFR2) is a GPCR that helps regulate pain and modulates the opioid system; however, its function remains unknown in cancers. Here, we found that NPFFR2 is significantly up-regulated in liver cancer and its expression is related to poor prognosis. Silencing of NPFFR2 reduced the malignancy of liver cancer cells by decreasing cell survival, invasion, and migration, while its overexpression increased invasion, migration, and anchorage-independent cell growth. Moreover, we found that the malignant function of NPFFR2 depends on RhoA and YAP signaling. Inhibition of Rho kinase activity completely restored the phenotypes induced by NPFFR2, and RhoA/F-Actin/YAP signaling was controlled by NPFFR2. These findings demonstrate that NPFFR2 may be a potential target for the treatment of hepatocellular carcinoma.

5.
Anticancer Res ; 42(7): 3475-3481, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35790270

RESUMEN

BACKGROUND/AIM: Metformin is a widely used drug for type 2 diabetes mellitus and has recently attracted broad attention for its therapeutic effects on many cancers. This study aimed to investigate the molecular mechanism of metformin's anticancer activity. MATERIALS AND METHODS: Cell viability was measured by MTT assay. Gene and protein expression levels were determined by reverse transcription-polymerase chain reaction and western blot analyses, respectively. RESULTS: Metformin and phenformin markedly induced NUPR1 expression in a dose- and time-dependent manner in H1299 non-small-cell lung cancer (NSCLC) cells. The silencing of NUPR1 in H1299 NSCLC cells enhanced cell sensitivity to metformin or ionizing radiation. Our previous report showed that metformin induces AKT serine/threonine kinase (AKT) activation in an activating transcription factor 4 (ATF4)-dependent manner and that the inhibition of AKT promotes cell sensitivity to metformin in H1299 NSCLC cells. Interestingly, ATF4-induced AKT activation in H1299 NSCLC cells treated with metformin was suppressed by the knockdown of NUPR1. CONCLUSION: Targeting NUPR1 could enhance the sensitivity of H1299 NSCLC cells to metformin by AKT inhibition.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Diabetes Mellitus Tipo 2 , Neoplasias Pulmonares , Metformina , Factor de Transcripción Activador 4 , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Metformina/farmacología , Proteínas Proto-Oncogénicas c-akt/genética
6.
Biochem Biophys Res Commun ; 601: 73-78, 2022 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-35231654

RESUMEN

Although endocrine therapy with tamoxifen has improved survival in breast cancer patients, resistance to this therapy remains one of the major causes of breast cancer mortality. In the present study, we found that the expression level of YAP/TAZ in tamoxifen-resistant MCF7 (MCF7-TR) breast cancer cells was significantly increased compared with that in MCF7 cells. Knockdown of YAP/TAZ with siRNA sensitized MCF7-TR cells to tamoxifen. Furthermore, siRNA targeting PSAT1, a downstream effector of YAP/TAZ, enhanced sensitivity to tamoxifen in MCF7-TR cells. Additionally, mTORC1 activity and survivin expression were significantly decreased during cell death induced by combination treatment with YAP/TAZ or PSAT1 siRNA and tamoxifen. In conclusion, targeting the YAP/TAZ-PSAT1 axis could sensitize tamoxifen-resistant MCF7 breast cancer cells by modulating the mTORC1-survivin axis.


Asunto(s)
Neoplasias de la Mama , Tamoxifeno , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ , Proteínas Señalizadoras YAP , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Resistencia a Antineoplásicos , Femenino , Humanos , Células MCF-7 , Diana Mecanicista del Complejo 1 de la Rapamicina , ARN Interferente Pequeño , Survivin/genética , Tamoxifeno/farmacología , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ/metabolismo , Proteínas Señalizadoras YAP/metabolismo
7.
Colloids Surf B Biointerfaces ; 189: 110829, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32036332

RESUMEN

Chlorin e6 (Ce6), with its high phototoxic potential, has wide applications in photodynamic therapy (PDT) for many human diseases. However, poor cancer cell localization of Ce6 has limited its direct application for PDT. Here, we developed cancer-targeting peptide p 18-4/chlorin e6 (Ce6)-conjugated polyhedral oligomeric silsesquioxane (PPC) nanoparticles for improving the targeting ability of Ce6 to breast cancer cells, thereby enhancing PDT efficacy. The synthesized PPC nanoparticles exhibited a spherical shape with an average diameter of 127.2 ± 11.3 nm in aqueous solution. Compared with free Ce6, the immobilization of p 18-4 enhanced the in vitro cellular uptake and targeting ability of PPC nanoparticles in breast cancer cell line MDA-MB-231. In addition, the intracellular uptake of PPC nanoparticles in MDA-MB-231 cells was dramatically increased compared with other cancer cells, indicating an obvious targeting ability of PPC nanoparticles on breast cancer cells. Upon light irradiation, PPC nanoparticles revealed significantly improved phototoxicity to MDA-MB-231 cells, mainly due to apoptotic cell death. In vivo PDT study suggested that PPC nanoparticles exhibited increased retention in tumor tissues and effectively inhibited the growth of MDA-MB-231 tumors in a target-specific manner. Overall, these results indicate that PPC nanoparticles are highly effective PDT agents for breast cancer therapy.


Asunto(s)
Antineoplásicos/farmacología , Nanopartículas/química , Compuestos de Organosilicio/farmacología , Péptidos/farmacología , Fotoquimioterapia , Fármacos Fotosensibilizantes/farmacología , Porfirinas/farmacología , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Clorofilidas , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Estructura Molecular , Compuestos de Organosilicio/química , Tamaño de la Partícula , Péptidos/química , Fármacos Fotosensibilizantes/síntesis química , Fármacos Fotosensibilizantes/química , Porfirinas/química , Propiedades de Superficie , Células Tumorales Cultivadas
8.
Nanomedicine ; 24: 102141, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31830613

RESUMEN

Lysyl oxidase (LOX) is a cell-secreted amine oxidase that crosslinks collagen and elastin in extracellular microenvironment. LOX-traceable nanoparticles (LOXab-NPs) consisting of LOX antibodies (LOXab) and paclitaxel, can accumulate at high concentrations at radiation-treated target sites, as a tumor-targeting drug carrier for chemotherapy. Tumor-targeting and anticancer effects of PLGA based LOXab-NPs in vitro and in vivo were evaluated at radiation-targeted site. In the in vivo A549 lung carcinoma xenograft model, we showed highly specific tumor targeting (above 7.0 times higher) of LOXab-NPs on irradiated tumors. Notably, systemically administered NPs delayed tumor growth, reducing tumor volumes by more than 2 times compared with non-irradiated groups (222% vs. >500%) over 2 weeks. Radiotropic LOXab-NPs can serve as chemotherapeutic vehicles for combined targeted chemo-radiotherapy in clinical oncology.


Asunto(s)
Apoptosis/efectos de la radiación , Nanopartículas/química , Nanopartículas/uso terapéutico , Proteína-Lisina 6-Oxidasa/metabolismo , Radiación Ionizante , Células A549 , Animales , Western Blotting , Células Cultivadas , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Tamaño de la Partícula , Proteína-Lisina 6-Oxidasa/química , Ensayos Antitumor por Modelo de Xenoinjerto
9.
J Tissue Eng Regen Med ; 12(4): 890-896, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-28478656

RESUMEN

Clinical irradiation therapy for cancer could increase the risk of localized wound complications. This study was conducted to evaluate the potential use of a chitosan microparticle-pluronic F127 (CSMP-PF) hydrogel complex containing bioactive molecules, substance P and transforming growth factor-ß1, to regeneratively repair skin damaged by local ionizing radiation (IR). The BALB/c/bkl mice were locally irradiated to their limbs with a single 40 Gy dose of Co-60 γ rays to induce a skin injury. The morphological characteristics of the chitosan microparticles were analysed by scanning electron microscopy. The amounts of bioactive molecules taken up and released by the CSMP-PF hydrogel complex were measured. Haematoxylin and eosin staining of IR-damaged skin showed acanthosis and hyperkeratosis in the epidermis; and damage to hair follicles/skin appendages and adipose tissue, as well as panniculus carnosus, in the dermis. Injection of the CSMP-PF hydrogel complex into IR-damaged skin resulted in skin repair, suggesting that the complex has potential for use in the regenerative repair of IR-damaged skin.


Asunto(s)
Quitosano , Rayos gamma/efectos adversos , Hidrogeles , Traumatismos Experimentales por Radiación , Sustancia P , Factor de Crecimiento Transformador beta , Cicatrización de Heridas/efectos de los fármacos , Animales , Quitosano/química , Quitosano/farmacología , Hidrogeles/química , Hidrogeles/farmacología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Traumatismos Experimentales por Radiación/tratamiento farmacológico , Traumatismos Experimentales por Radiación/patología , Piel/lesiones , Piel/metabolismo , Piel/patología , Sustancia P/química , Sustancia P/farmacología , Factor de Crecimiento Transformador beta/química , Factor de Crecimiento Transformador beta/farmacología
10.
Int J Oncol ; 50(4): 1279-1288, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28350116

RESUMEN

Epithelial ovarian cancer is the most aggressive and lethal among the gynecological malignancies, which is often found disseminated to peritoneal cavity at the time of diagnosis. There is accumulating evidence on the existence of genetic alteration and amplification of fibroblast growth factor receptor (FGFR) in various cancers. Also the aberrated FGFR/FGF signaling has been implicated in cancer development and tumor microenvironment. However, the antitumor activity of BGJ398, a selective inhibitor of FGFR 1/2/3 against ovarian cancer still remains unknown. The aim of the present study is to evaluate the antitumoral activity of BGJ398 on ovarian cancer cell line SKOV3ip1 using 3-dimensional (3D) sphere culture system which has been accepted as a better mimic in vivo microenvironment than conventional 2-dimensional (2D) monolayer culture system. We examined the differential expression features of key signaling molecules which have a role in cell survival and proliferation between sphere-cultured SKOV3ip1 cells and monolayer-cultured SKOV3ip1 cells. The phosphorylation of AKT and signal transducer and activator of transcription 3 (STAT3) known as survival signaling molecules were upregulated in sphere-cultured SKOV3ip1 cells compared to in monolayer-cultured SKOV3ip1 cells. Next, we evaluated the antitumor activity of BGJ398 in monolayer-cultured SKOV3ip1 cells or sphere-cultured SKOV3ip1 cells. Treatment of BGJ398 did not affect the SKOV3ip1 cell viability in monolayer culture system, but, the cell viability of sphere-cultured SKOV3ip1 cells was markedly reduced by BGJ398. The phosphorylation of AKT and STAT3 was downregulated by BGJ398 in sphere-cultured SKOV3ip1 cells, but not in monolayer cultured-SKOV3ip1 cells. Moreover, combination treatment with BGJ398 and paclitaxel in sphere-cultured SKOV3ip1 showed synergistic inhibitory effect on cell viability. Collectively, our report reveals the BGJ398 is a potent antitumor agent against ovarian cancer and FGFR is a promising therapeutic target to anticancer therapy considering ovarian cancer metastatic microenvironment.

11.
Tissue Eng Regen Med ; 14(4): 421-432, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30603498

RESUMEN

Mesenchymal stem cells (MSCs), which are multipotent and have self-renewal ability, support the regeneration of damaged normal tissue. A number of external stimuli promote migration of MSCs into peripheral blood and support their participation in wound healing. In an attempt to harness the potential beneficial effects of such external stimuli, we exposed human MSCs (hMSCs) to one such stimulus-low-dose ionizing radiation (LDIR)-and examined their biological properties. To this end, we evaluated differences in proliferation, cell cycle, DNA damage, expression of surface markers (CD29, CD34, CD90, and CD105), and differentiation potential of hMSCs before and after irradiation with γ-rays generated using a 137CS irradiator. At doses less than 50 mGy, LDIR had no significant effect on the viability or apoptosis of hMSCs. Interestingly, 10 mGy of LDIR increased hMSC viability by 8% (p < 0.001) compared with non-irradiated hMSCs. At doses less than 50 mGy, LDIR did not induce DNA damage, including DNA strand breaks, or cause cellular senescence or cell-cycle arrest. Surface marker expression and in vitro differentiation potential of hMSCs were maintained after two exposures to LDIR at 10 mGy per dose. In conclusion, a two-dose exposure to LDIR at 10 mGy per dose not only facilitates proliferation of hMSCs, it also maintains the stem cell characteristics of hMSCs without affecting their viability. These results provide evidence for the potential of LDIR as an external stimulus for in vitro expansion of hMSCs and application in tissue engineering and regenerative medicine.

12.
Mol Med Rep ; 13(5): 4135-42, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27053172

RESUMEN

Lung fibrosis is a major complication in radiation­induced lung damage following thoracic radiotherapy, while the underlying mechanism has remained to be elucidated. The present study performed immunofluorescence and immunoblot assays on irradiated human pulmonary artery endothelial cells (HPAECs) with or without pre­treatment with VAS2870, a novel NADPH oxidase (NOX) inhibitor, or small hairpin (sh)RNA against NOX1, ­2 or ­4. VAS2870 reduced the cellular reactive oxygen species content induced by 5 Gy radiation in HPAECs and inhibited phenotypic changes in fibrotic cells, including increased alpha smooth muscle actin and vimentin, and decreased CD31 and vascular endothelial cadherin expression. These fibrotic changes were significantly inhibited by treatment with NOX1 shRNA, but not by NOX2 or NOX4 shRNA. Next, the role of NOX1 in pulmonary fibrosis development was assessed in the lung tissues of C57BL/6J mice following thoracic irradiation using trichrome staining. Administration of an NOX1­specific inhibitor suppressed radiation­induced collagen deposition and fibroblastic changes in the endothelial cells (ECs) of these mice. The results suggested that radiation­induced pulmonary fibrosis may be efficiently reduced by specific inhibition of NOX1, an effect mediated by reduction of fibrotic changes of ECs.


Asunto(s)
Células Endoteliales/enzimología , Rayos gamma/efectos adversos , NADH NADPH Oxidorreductasas/metabolismo , NADPH Oxidasas/metabolismo , Fibrosis Pulmonar/enzimología , Traumatismos por Radiación/enzimología , Animales , Benzoxazoles/farmacología , Línea Celular , Células Endoteliales/patología , Humanos , Ratones , NADH NADPH Oxidorreductasas/antagonistas & inhibidores , NADPH Oxidasa 1 , NADPH Oxidasas/antagonistas & inhibidores , Fibrosis Pulmonar/patología , Traumatismos por Radiación/patología , Triazoles/farmacología
13.
Oncotarget ; 7(9): 9634-44, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26909594

RESUMEN

Growth differentiation factor 15 (GDF15) is an emerging biomarker of cardiovascular risk and disease. Microarray analyses revealed that GDF15 levels were increased during cellular senescence induced by ionizing radiation (IR) in human aortic endothelial cells (HAECs). However, the role of GDF15 in HAEC cellular senescence remains unclear. This study demonstrated that downregulation of GDF15 in HAECs partially prevented cellular senescence triggered by IR, which was confirmed by recovery of cell proliferation and reverse senescence-associated ß-galactosidase (SA-ß-gal) staining. Conversely, upregulation of GDF15-induced cellular senescence in HAECs, confirmed by G0/G1 cell cycle arrest, decreased during cell proliferation and increased SA-ß-gal staining. GDF15-induced cellular senescence was observed in p16-knockdown cells but not in p53-knockdown cells. GDF15 expression in endothelial cells also generated reactive oxygen species (ROS), which led to activation of extracellular signal-regulated kinases (ERKs) and induction of senescence by oxidative stress. These results suggested that GDF15 might play an important role in cellular senescence through a ROS-mediated p16 pathway and contribute to the pathogenesis of atherosclerosis via pro-senescent activity.


Asunto(s)
Aorta/efectos de la radiación , Aterosclerosis/patología , Senescencia Celular/efectos de la radiación , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Células Endoteliales/metabolismo , Factor 15 de Diferenciación de Crecimiento/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Aorta/citología , Línea Celular , Proliferación Celular/efectos de la radiación , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de la radiación , Factor 15 de Diferenciación de Crecimiento/genética , Humanos , Estrés Oxidativo , Interferencia de ARN , ARN Interferente Pequeño/genética , Radiación Ionizante , Proteína p53 Supresora de Tumor/genética , beta-Galactosidasa/metabolismo
14.
Int J Mol Sci ; 17(1)2016 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-26751441

RESUMEN

The goal to successful wound healing is essentially to immobilize and recruit appropriate numbers of host stem or progenitor cells to the wound area. In this study, we developed a chitosan nanofiber-immobilized neuropeptide substance-P (SP), which mediates stem cell mobilization and migration, onto the surfaces of nanofibers using a peptide-coupling agent, and evaluated its biological effects on stem cells. The amount of immobilized SP on chitosan nanofibers was modulated over the range of 5.89 ± 3.27 to 75.29 ± 24.31 ng when reacted with 10 to 500 ng SP. In vitro migration assays showed that SP-incorporated nanofibers induced more rapid migration of human mesenchymal stem cells on nanofibers compared to pristine samples. Finally, the conjugated SP evoked a minimal foreign body reaction and recruited a larger number of CD29- and CD44-positive stem cells into nanofibers in a mouse subcutaneous pocket model.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Quitosano/química , Células Madre Mesenquimatosas/efectos de los fármacos , Nanofibras/química , Neurotransmisores/farmacología , Sustancia P/farmacología , Andamios del Tejido/química , Animales , Femenino , Humanos , Células Madre Mesenquimatosas/citología , Ratones Endogámicos BALB C , Ratones Desnudos , Nanofibras/ultraestructura , Neurotransmisores/administración & dosificación , Neurotransmisores/química , Sustancia P/administración & dosificación , Sustancia P/química , Ingeniería de Tejidos/métodos , Cicatrización de Heridas/efectos de los fármacos
15.
Macromol Biosci ; 15(11): 1586-94, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26183918

RESUMEN

For electrical stimulation of hMSCs, gold nanoparticles were coated onto polyethyleneimine coated glass cover slips. The effects of pulsed or constant electrical stimulation upon cytotoxicity and differentiation of hMSCs were examined. The effects of co culturing hMSCs with neuronal cells were also tested. The neuronal differentiation of the stem cells was evaluated by determining the expression of neuron-specific genes and proteins using RT-PCR and Western blotting. Morphological changes were evaluated by scanning electron microscopy. The hMSCs co-cultured with mature neuronal cells and stimulated with electrical shock showed the greatest level of neurite outgrowth (>150 mm) and smaller cell body sizes.


Asunto(s)
Regulación de la Expresión Génica , Células Madre Mesenquimatosas/metabolismo , Neuritas/metabolismo , Neurogénesis , Técnicas de Cocultivo , Estimulación Eléctrica , Humanos , Células Madre Mesenquimatosas/ultraestructura , Neuritas/ultraestructura
16.
Int J Mol Med ; 35(3): 731-8, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25543718

RESUMEN

Radiation-induced heart disease (RIHD) is becoming an increasing concern for patients and clinicians alike due to the use of radiotherapy for the treatment of breast cancer, Hodgkin's lymphoma, pediatric cancer and tumors of the thorax. However, the mechanisms underlying this phenomenon remain largely unknown. As the senescent cell fraction following irradiation is known to increase, in the present study, we investigated whether ionizing radiation (IR) causes the onset of heart disease by inducing cellular senescence in cardiomyocytes. In the present study, we evaluated the effects of IR on HL-1 and H9C2 cells, cells predominantly used in in vitro myocardial cell models. We found that the exposure of the HL-1 and H9C2 cells to IR induced reactive oxygen species (ROS)-mediated cellular senescence, as shown by staining of senescence-associated ß-galactosidase (SA-ß-gal). The levels of ROS in irradiated cells were determined using the fluorescent dye, 2', 7'-dichlorodihydrofluorescein diacetate (DCF-DA). Notably, the expression of corin, a cardiac protease that is essential for the proteolytic cleavage of natriuretic peptides, was significantly decreased following the exposure of the cells to IR. Importantly, the knockdown of corin by RNA interference enhanced IR-induced senescence. On the contrary, the overexpression of natriuretic peptides reversed the IR-induced senescence. Taken together, our data suggest that defects in corin function and the inhibition of natriuretic peptides following exposure to IR may contribute to the development of RIHD through the acceleration of cellular senescence.


Asunto(s)
Senescencia Celular/genética , Senescencia Celular/efectos de la radiación , Expresión Génica , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/efectos de la radiación , Radiación Ionizante , Serina Endopeptidasas/genética , Regulación hacia Abajo , Técnicas de Silenciamiento del Gen , Péptidos Natriuréticos/genética , Especies Reactivas de Oxígeno/metabolismo , Serina Endopeptidasas/deficiencia
17.
PLoS One ; 9(1): e85509, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24465581

RESUMEN

Matrix metalloproteinases regulate pathophysiological events by processing matrix proteins and secreted proteins. Previously, we demonstrated that soluble heat shock protein B1 (HSPB1) is released primarily from endothelial cells (ECs) and regulates angiogenesis via direct interaction with vascular endothelial growth factor (VEGF). Here we report that MMP9 can cleave HSPB1 and release anti-angiogenic fragments, which play a key role in tumorprogression. We mapped the cleavage sites and explored their physiological relevance during these processing events. HSPB1 cleavage by MMP9 inhibited VEGF-induced ECs activation and the C-terminal HSPB1 fragment exhibited more interaction with VEGF than did full-length HSPB1. HSPB1 cleavage occurs during B16F10 lung progression in wild-type mice. Also, intact HSPB1 was more detected on tumor endothelium of MMP9 null mice than wild type mice. Finally, we confirmed that secretion of C-terminal HSPB1 fragment was significantly inhibited lung and liver tumor progression of B16F10 melanoma cells and lung tumor progression of CT26 colon carcinoma cells, compared to full-length HSPB1. These data suggest that in vivo MMP9-mediated processing of HSPB1 acts to regulate VEGF-induced ECs activation for tumor progression, releasing anti-angiogenic HSPB1 fragments. Moreover, these findings potentially explain an anti-target effect for the failure of MMP inhibitors in clinical trials, suggesting that MMP inhibitors may have pro-tumorigenic effects by reducing HSPB1 fragmentation.


Asunto(s)
Carcinogénesis/patología , Proteínas de Choque Térmico HSP27/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Animales , Carcinogénesis/genética , Carcinogénesis/metabolismo , Movimiento Celular , Proliferación Celular , Progresión de la Enfermedad , Células Endoteliales/metabolismo , Proteínas de Choque Térmico HSP27/genética , Proteínas de Choque Térmico , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Metaloproteinasa 9 de la Matriz/genética , Ratones , Chaperonas Moleculares , Fosforilación , Receptores de Factores de Crecimiento Endotelial Vascular/genética , Sarcoma/genética , Sarcoma/metabolismo , Sarcoma/patología , Neoplasias de los Tejidos Blandos/genética , Neoplasias de los Tejidos Blandos/metabolismo , Neoplasias de los Tejidos Blandos/patología
18.
Cancer Lett ; 346(2): 249-56, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24434653

RESUMEN

DNA lesion-induced centrosomal abnormalities during the replication phase are relatively unknown. Here, we report that RNAi-mediated depletion of RRM1 induces cell-cycle arrest at the replication phase, along with severe DNA damage and centrosomal amplification. Interestingly, CHK1 depletion synergistically increased RRM1-depletion-induced centrosomal amplification. In response to hydroxyurea, CHK1 was delocalized from the centrosome by RRM1 depletion. Moreover, CDK1, which functions in centrosome separation and is inhibited by CHK1, was found to be essential for RRMI1-depletion-induced centrosomal amplification. Thus, we herein demonstrate that RRM1 preserves chromosomal stability via the CHK1- and CDK1-dependent stabilization of the centrosomal integrity at the replication stage.


Asunto(s)
Proteína Quinasa CDC2/metabolismo , Centrosoma/fisiología , Replicación del ADN/fisiología , Proteínas Quinasas/metabolismo , Proteínas Supresoras de Tumor/genética , Proteína Quinasa CDC2/genética , Línea Celular Tumoral , Centrosoma/metabolismo , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Ciclina A/metabolismo , Ciclina B/metabolismo , Daño del ADN , Replicación del ADN/genética , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Proteínas Quinasas/genética , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Ribonucleósido Difosfato Reductasa , Transducción de Señal , Transfección , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/metabolismo
19.
J Nanosci Nanotechnol ; 14(10): 7402-9, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25942801

RESUMEN

Microenvironment of the extracellular matrix can influence cellular responses through alternation of initial attachment and induce production of new tissue. To study the effect of such microenvironment on the relationship of cell cytoskeletal shape and its biological behaviors such as adhesion, proliferation and differentiation, we designed a patterned strip line of fibronectin on self assembled monolayers via microcontact printing. The physiological behavior of human mesenchymal stem cell (hMSC) on defined micro-patterns of fibronectin was evaluated after 4 h and 2 days of culture. Initial adhesion of hMSCs on a substrate with pattern spacing of 11 µm was stabilized faster than that on other substrates. Ratio of proliferating hMSC on 5 and 11 µm substrate constantly maintained a high rate. hMSCs on 5 and 11 µm substrate could adhere to substrate as spreading from fibronectin pattern line to several and lateral fibronectin pattern line. Their nucleus area could represent artificial increase by widely spreading on several fibronectin pattern lines. On the contrary to this, ratio of proliferating hMSC on 20 µm substrate constantly maintained a low rate less than even control and 0 µm substrate without fibronectin pattern. Tiny nucleus caused narrow and elongated hMSC morphology on 20 µm substrate gave the negative effect on the cell adhesion and proliferation. However, hMSCs on 20 µm substrate possessed not only slightly increased value of GO/G1 phase but also down regulation of CD marker expression compared with other groups. These results show initial adhesion and morphology of hMSC could regulate specific cellular behavior of hMSC.


Asunto(s)
Fibronectinas/química , Fibronectinas/farmacología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Microtecnología/métodos , Adhesión Celular/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Humanos , Impresión , Propiedades de Superficie
20.
J Nanosci Nanotechnol ; 14(10): 7621-6, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25942837

RESUMEN

Natural and synthetic polymers, in particular those that are conductive, are of great interest in the field of tissue engineering and the pursuit of biomimetic extracellular matrix (ECM) structures for adhesion, proliferation, and differentiation of cells. In the present study, natural chitin and conductive polyaniline (PANi) blended solutions were electrospun to produce biodegradable and conductive biomimetic nanostructured scaffolds. The chitin/PANi (Chi-PANi) nanofibrous materials were characterized using field emission scanning electron microscopy, Fourier transform-infrared spectroscopy, wettability analysis, mechanical testing, and electrical conductivity measurements using a 4-point probe method. The calculated electrical conductivities of the PANi-containing nanofiber scaffolds significantly increased as the amount of PANi increased, reaching 5.21 ± 0.28 x 10(-3) S/cm for 0.3 wt% content of the conducting polymer. In addition, the viability of human mesenchymal stem cells (hMSCs) cultured on the Chi-PANi nanofiber scaffolds in vitro was found to be excellent. These results suggest that the Chi-PANi nanofiber scaffolds have great potential for use in tissue engineering applications that involve electrical stimulation.


Asunto(s)
Compuestos de Anilina/química , Materiales Biocompatibles/química , Conductividad Eléctrica , Nanofibras/química , Nanotecnología/métodos , Ingeniería de Tejidos , Andamios del Tejido/química , Materiales Biocompatibles/farmacología , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Quitina/química , Humanos , Fenómenos Mecánicos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Neuritas/efectos de los fármacos , Neuritas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA