Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 179
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Phys Rev E ; 108(4-1): 044602, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37978710

RESUMEN

We employ only the positions of colloidal particles and construct machine learning (ML) models to test the presence of structural order in glass transition for two kinds of two-dimensional (2D) colloids: 2D polydisperse colloids (PC) with medium-range crystalline order (MRCO) and 2D binary colloids (BC) without MRCO. ML models predict the glass transition of 2D colloids successfully without any information on MRCO. Even certain ML models trained with BC predict the glass transition of PC successfully, thus suggesting that universal structural characteristics would exist besides MRCO.

2.
Expert Rev Med Devices ; 19(3): 213-228, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35255755

RESUMEN

INTRODUCTION: Artificial Intelligence (AI) is becoming increasingly utilized in the medical device industry as it can address unmet demands in clinical sites and provide more patient treatment options. This study aims to analyze the FDA's Breakthrough Device Program and MFDS' Innovative Medical Device Program, which support regulatory science for innovative medical devices today. Through this study, it is intended to enable prediction of current development trends of Software as a Medical Device (SaMD) and Digital Therapeutics (DTx), which combine AI and technologies to be used in the clinical field soon. AREAS COVERED: A systematic search was conducted on the broad topics of 'FDA and MFDS Program's SaMD, DTx.' A parallel review and update of PubMed, and the official websites were conducted to investigate the regulator's databases, review official press releases of regulatory agencies, and provide detailed descriptions of researchers. EXPERT OPINION: The efforts of related stakeholders are needed to expand AI technology to diagnosis, prevention, and treatment technologies for diseases that are difficult to diagnose early or are classified as clinical challenges. It is important to prepare regulatory policies suitable for the rapid pace of technological development and to create an environment where regulatory science can be realized by developers.


Asunto(s)
Inteligencia Artificial , Programas Informáticos , Humanos , República de Corea , Tecnología , Estados Unidos , United States Food and Drug Administration
4.
Theranostics ; 8(11): 3087-3098, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29896304

RESUMEN

Rationale: Characterizing the regulation of bone-resorbing osteoclasts is central to the understanding of the pathogenesis and treatment of bone diseases, such as osteoporosis and periodontitis. 5-hydroxytryptamine (5-HT) has drawn considerable attention for its role in bone; however, it remains unknown whether the intracellular signaling of 5-HT receptors (5-HTRs) is linked to any of the regulatory mechanisms in osteoclasts. Herein, we report 5-HT6R to be a key regulatory receptor for osteoclastogenesis. Methods: In order to explore the critical role of 5-HT6R in bone-resorbing osteoclasts, in vitro experiments were performed using mouse whole bone marrow cells isolated from femora and tibiae and In vivo animal experiments were performed using 5-HT6R-deficient (5-HT6RKO-/-) mice, bone resorption mice model, and osteoporosis mice model. Results: Compared to other 5HTRs, activation of 5-HT6R relatively increased TRAP (tartrate-resistant acid phosphatase) activity during osteoclastogenesis. 5-HT6RKO(-/-) mice and 5-HT6RKO(-/-) osteoclast lineages presented with an abnormal phenotype and impaired osteoclastogenesis and impaired osteoclastogenesis. Activation of 5-HT6R increased the number of TRAP-positive multinuclear osteoclasts, actin ring formation, and expression of early osteoclast markers with osteoclast lineage commitment. Intracellular 5-HT6R signaling was found to be linked to RhoA GTPase activation and was involved in the maturation of osteoclasts. This signaling pathway also showed enhanced bone destruction after lipopolysaccharide (LPS) administration in mice. Furthermore, inhibition of 5-HT6R-mediated RhoA GTPase signaling protected against ovariectomy(OVX)-induced bone loss in mice. Conclusion: Taken together, our findings place the 5-HT6R system in a new context of osteoclast lineages in both an in vitro and in vivo system, and also it may offer a novel molecular target for the treatment of bone diseases.


Asunto(s)
Resorción Ósea/etiología , Osteogénesis , Osteoporosis/etiología , Receptores de Serotonina/metabolismo , Serotonina/metabolismo , Transducción de Señal , Animales , Resorción Ósea/inducido químicamente , Resorción Ósea/terapia , Huesos/patología , Modelos Animales de Enfermedad , Femenino , Lipopolisacáridos/efectos adversos , Ratones , Ratones Noqueados , Osteoclastos/fisiología , Osteoporosis/inducido químicamente , Osteoporosis/terapia , Ovariectomía/efectos adversos , Receptores de Serotonina/genética , Serotonina/genética , Fosfatasa Ácida Tartratorresistente/metabolismo
5.
Int J Mol Sci ; 19(6)2018 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-29895782

RESUMEN

The present study evaluated the protective effects of melatonin in ethanol (EtOH)-induced senescence and osteoclastic differentiation in human periodontal ligament cells (HPDLCs) and cementoblasts and the underlying mechanism. EtOH increased senescence activity, levels of reactive oxygen species (ROS) and the expression of cell cycle regulators (p53, p21 and p16) and senescence-associated secretory phenotype (SASP) genes (interleukin [IL]-1ß, IL-6, IL-8 and tumor necrosis factor-α) in HPDLCs and cementoblasts. Melatonin inhibited EtOH-induced senescence and the production of ROS as well as the increased expression of cell cycle regulators and SASP genes. However, it recovered EtOH-suppressed osteoblastic/cementoblastic differentiation, as evidenced by alkaline phosphatase activity, alizarin staining and mRNA expression levels of Runt-related transcription factor 2 (Runx2) and osteoblastic and cementoblastic markers (glucose transporter 1 and cementum-derived protein-32) in HPDLCs and cementoblasts. Moreover, it inhibited EtOH-induced osteoclastic differentiation in mouse bone marrow⁻derived macrophages (BMMs). Inhibition of protein never in mitosis gene A interacting-1 (PIN1) by juglone or small interfering RNA reversed the effects of melatonin on EtOH-mediated senescence as well as osteoblastic and osteoclastic differentiation. Melatonin blocked EtOH-induced activation of mammalian target of rapamycin (mTOR), AMP-activated protein kinase (AMPK), mitogen-activated protein kinase (MAPK) and Nuclear factor of activated T-cells (NFAT) c-1 pathways, which was reversed by inhibition of PIN1. This is the first study to show the protective effects of melatonin on senescence-like phenotypes and osteoclastic differentiation induced by oxidative stress in HPDLCs and cementoblasts through the PIN1 pathway.


Asunto(s)
Cemento Dental/citología , Etanol/farmacología , Melatonina/farmacología , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Ligamento Periodontal/citología , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Senescencia Celular/efectos de los fármacos , Cemento Dental/metabolismo , Humanos , Peptidilprolil Isomerasa de Interacción con NIMA/metabolismo , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Osteoclastos/citología , Ligamento Periodontal/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo
6.
Bioelectromagnetics ; 39(5): 394-404, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29709064

RESUMEN

Although we recently demonstrated that static magnetic fields (SMFs) of 3, 15, and 50 mT stimulate osteoblastic differentiation, the effects of SMFs on osteoclastogenesis are still poorly understood. This study focused on the suppressive effects of SMFs on receptor activator of nuclear factor κB ligand (RANKL)-induced osteoclastogenesis and bone resorption. Direct SMFs inhibit RANKL-induced multinucleated osteoclast formation, tartrate-resistant acid phosphatase activity, and bone resorption in mouse bone marrow-derived macrophage cells. The conditioned medium from osteoblasts treated with SMFs also resulted in the inhibition of osteoclast differentiation as well as resorption. The RANKL-induced expression of osteoclast-specific transcription factors, such as c-Fos and NFATc1, was remarkably downregulated by SMF at 15 mT. In addition, SMF inhibited RANKL-activated Akt, glycogen synthase kinase 3ß (GSK3ß), extracellular signal-regulated kinase, c-jun N-terminal protein kinase, mitogen-activated protein kinase (MAPK), and nuclear factor-κB (NF-κB) formation. These findings indicate that SMF-mediated attenuation of RANKL-induced Akt, GSK3ß, MAPK, and NF-κB pathways could contribute to the direct and indirect inhibition of osteoclast formation and bone resorption. Therefore, SMFs could be developed as a therapeutic agent against periprosthetic or peri-implant osteolysis. Additionally, these could be used against osteolytic diseases such as osteoporosis and rheumatoid arthritis. Bioelectromagnetics. 39:394-404, 2018. © 2018 Wiley Periodicals, Inc.


Asunto(s)
Células de la Médula Ósea/fisiología , Diferenciación Celular/fisiología , Campos Magnéticos , Osteoclastos/fisiología , Animales , Células de la Médula Ósea/citología , Resorción Ósea/patología , Resorción Ósea/fisiopatología , Células Cultivadas , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ratones Endogámicos ICR , FN-kappa B/metabolismo , Factores de Transcripción NFATC/metabolismo , Osteoclastos/citología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ligando RANK/metabolismo , Transducción de Señal
7.
Mol Cells ; 41(5): 476-485, 2018 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-29764006

RESUMEN

Although tectorigenin (TG), a major compound in the rhizome of Belamcanda chinensis, is conventionally used for the treatment of inflammatory diseases, its effects on osteogenesis and osteoclastogenesis have not been reported. The objective of this study was to investigate the effects and possible underlying mechanism of TG on in vitro osteoblastic differentiation and in vivo bone formation, as well as in vitro osteoclast differentiation and in vivo bone resorption. TG promoted the osteogenic differentiation of primary osteoblasts and periodontal ligament cells. Moreover, TG upregulated the expression of the BMP2, BMP4, and Smad-4 genes, and enhanced the expression of Runx2 and Osterix. In vivo studies involving mouse calvarial bone defects with µCT and histologic analysis revealed that TG significantly increased new bone formation. Furthermore, TG treatment inhibited osteoclast differentiation and the mRNA levels of osteoclast markers. In vivo studies of mice demonstrated that TG caused the marked attenuation of bone resorption. These results collectively demonstrated that TG stimulated osteogenic differentiation in vitro, increased in vivo bone regeneration, inhibited osteoclast differentiation in vitro, and suppressed inflammatory bone loss in vivo. These novel findings suggest that TG may be useful for bone regeneration and treatment of bone diseases.


Asunto(s)
Resorción Ósea/prevención & control , Curación de Fractura/efectos de los fármacos , Isoflavonas/farmacología , Osteoblastos/efectos de los fármacos , Osteoclastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Animales , Diferenciación Celular/efectos de los fármacos , Línea Celular Transformada , Células Cultivadas , Femenino , Humanos , Isoflavonas/toxicidad , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Ratones Endogámicos ICR , Ligamento Periodontal/citología , Cultivo Primario de Células , Ligando RANK/fisiología , Células RAW 264.7 , Cráneo/efectos de los fármacos , Cráneo/lesiones , Cráneo/cirugía , Factores de Transcripción/metabolismo
8.
Tissue Eng Regen Med ; 15(5): 615-627, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30603583

RESUMEN

BACKGROUND: We aimed to determine the effect of fibronectin (FN)-immobilized microgrooved titanium (Ti) on human gingival fibroblast proliferation, gene expression and protein expression. METHODS: Photolithography was used to fabricate the microgrooved Ti, and amine funtionalization (silanization) was used for FN immobilization on titanium surfaces. Cell proliferation, gene expression and protein expression were analyzed, followed by multiple regression analysis for determining the influential factors on cell proliferation. RESULTS: FN-immobilized microgrooved Ti significantly enhanced the fibroblast proliferation in various timelines of culture, among which a burst of fivefold increase is induced at 96 h of culture compared to that on the control smooth Ti. We suggest a presence of the synergistic promotion effect of microgrooves and FN immobilization on fibroblast proliferation. Through a series of analyses on the expression of various genes and proteins involved in cell adhesion and proliferation, cyclin-dependent kinase 6, cyclin D1, integrin α5, oncogene c-Src, osteonectin, paxillin and talin-2 were determined as influential factors on promoting fibroblast proliferation induced by FN-immobilized microgrooved Ti. CONCLUSION: FN-immobilized microgrooved Ti can act as an effective surface for enhancing fibroblast proliferation, and can be used for promoting soft tissue response on the connective tissue attachment zone of biomaterial surfaces.

9.
Clin Oral Investig ; 22(5): 1933-1943, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29188452

RESUMEN

OBJECTIVES: Although bisphosphonates (BPs) are known to be associated with osteonecrosis of the maxilla, the precise effects of BPs on bone metabolism in human maxillary sinus mucosal cells (HMSMCs) are not yet known. The purposes of this study were to examine the effects of the BPs zoledronate (ZOL) and alendronate (ALN) on osteoblastic and osteoclastic differentiation in HMSMCs and to investigate the signaling pathways involved. MATERIALS AND METHODS: The effects of ZOL and ALN were assessed for osteoblast differentiation by alkaline phosphatase (ALP) activity, alizarin red staining, and RT-PCR for genes encoding Runx2 and osterix. Receptor activator of nuclear factor-κB ligand (RANKL)-mediated osteoclast differentiation in bone marrow macrophages (BMMs) was also examined. RESULTS: ZOL and ALN both suppressed osteoblastic differentiation, as evidenced by their effects on ALP activity, mineralization nodule formation, and the mRNA expression levels of osteoblastic transcript factors. The RANKL/osteoprotegerin ratio in HMSMCs was increased by ALN, whereas ZOL had the opposite effect. Conditioned medium obtained from ALN-treated HMSMCs stimulated osteoclast formation and upregulated NFATc1 expression, whereas conditioned medium from ZOL-treated cells did not. ALN was more cytotoxic and stimulated apoptosis more strongly than ZOL. BPs decreased the protein levels of the non-canonical Wnt signaling protein Wnt5a and calmodulin-dependent kinase II. Moreover, recombinant human Wnt5a reversed the effects of BPs on osteoblastic and osteoclastic differentiation. CONCLUSION: This study is the first demonstration that BPs exert negative effects on osteoblastic and osteoclastic processes via the non-canonical Wnt pathway in HMSMSCs. CLINICAL RELEVANCE: It suggests that patients taking BPs during the period of maxillary sinus lifting and amentation should be given special attention.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Difosfonatos/farmacología , Seno Maxilar/citología , Osteoblastos/efectos de los fármacos , Osteoclastos/efectos de los fármacos , Células Madre/metabolismo , Adulto , Alendronato/farmacología , Fosfatasa Alcalina/metabolismo , Western Blotting , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Femenino , Humanos , Masculino , Ligando RANK/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor de Transcripción Sp7/metabolismo , Ácido Zoledrónico/farmacología
10.
J Periodontal Implant Sci ; 47(5): 273-291, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29093986

RESUMEN

PURPOSE: Although static magnetic fields (SMFs) have been used in dental prostheses and osseointegrated implants, their biological effects on osteoblastic and cementoblastic differentiation in cells involved in periodontal regeneration remain unknown. This study was undertaken to investigate the effects of SMFs (15 mT) on the osteoblastic and cementoblastic differentiation of human osteoblasts, periodontal ligament cells (PDLCs), and cementoblasts, and to explore the possible mechanisms underlying these effects. METHODS: Differentiation was evaluated by measuring alkaline phosphatase (ALP) activity, mineralized nodule formation based on Alizarin red staining, calcium content, and the expression of marker mRNAs assessed by reverse transcription polymerase chain reaction (RT-PCR). Signaling pathways were analyzed by western blotting and immunocytochemistry. RESULTS: The activities of the early marker ALP and the late markers matrix mineralization and calcium content, as well as osteoblast- and cementoblast-specific gene expression in osteoblasts, PDLCs, and cementoblasts were enhanced. SMFs upregulated the expression of Wnt proteins, and increased the phosphorylation of glycogen synthase kinase-3ß (GSK-3ß) and total ß-catenin protein expression. Furthermore, p38 and c-Jun N-terminal kinase (JNK) mitogen-activated protein kinase (MAPK), and nuclear factor-κB (NF-κB) pathways were activated. CONCLUSIONS: SMF treatment enhanced osteoblastic and/or cementoblastic differentiation in osteoblasts, cementoblasts, and PDLCs. These findings provide a molecular basis for the beneficial osteogenic and/or cementogenic effect of SMFs, which could have potential in stimulating bone or cementum formation during bone regeneration and in patients with periodontal disease.

11.
Arch Oral Biol ; 84: 94-99, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28965045

RESUMEN

OBJECTIVE: Caffeic acid phenethyl ester (CAPE), a natural honeybee product exhibits a spectrum of biological activities including antimicrobial, anti-inflammatory, antioxidant and antitumor actions. The purpose of this research was to investigate the anticancer potential of CAPE and its molecular mechanism in human oral cancer cell lines (YD15, HSC-4 and HN22 cells). DESIGN: To determine the apoptotic activity of CAPE and identify its molecular targets, trypan blue exclusion assay, soft agar assay, Western blot analysis, DAPI staining, and live/dead assay were performed. RESULTS: CAPE significantly suppressed transformation of neoplastic cells induced by epidermal growth factor (EGF) and 12-O-tetradecanoylphorbol 13-acetate (TPA) without inhibiting growth. CAPE treatment inhibited cell growth, increased the cleavages of caspase-3 and poly (ADP-ribose) polymerase (PARP), and augmented the number of fragmented nuclei in human oral cancer cell lines. CAPE activated Bax protein causing it to undergo a conformational change, translocate to the mitochondrial outer membrane, and oligomere. CAPE also significantly increased Puma expression and interestingly Puma and Bax were co-localized. CONCLUSION: Overall, these results suggest that CAPE is a potent apoptosis-inducing agent in human oral cancer cell lines. Its action is accompanied by up-regulation of Bax and Puma proteins.


Asunto(s)
Apoptosis/efectos de los fármacos , Ácidos Cafeicos/farmacología , Neoplasias de la Boca/tratamiento farmacológico , Alcohol Feniletílico/análogos & derivados , Proteínas Reguladoras de la Apoptosis/metabolismo , Biomarcadores de Tumor/metabolismo , Western Blotting , Línea Celular Tumoral , Transformación Celular Neoplásica/efectos de los fármacos , Humanos , Inmunohistoquímica , Alcohol Feniletílico/farmacología , Proteínas Proto-Oncogénicas/metabolismo , Coloración y Etiquetado , Proteína X Asociada a bcl-2/metabolismo
12.
Cell Transplant ; 26(8): 1405-1417, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28901183

RESUMEN

Osteoarthritis (OA) is an inflammatory joint disease characterized by degeneration of articular cartilage within synovial joints. An estimated 27 million Americans suffer from OA, and the population is expected to reach 67 million in the United States by 2030. Thus, it is urgent to find an effective treatment for OA. Traditional OA treatments have no disease-modifying effect, while regenerative OA therapies such as autologous chondrocyte implantation show some promise. Nonetheless, current regenerative therapies do not overcome synovial inflammation that suppresses the differentiation of mesenchymal stem cells (MSCs) to chondrocytes and the expression of type II collagen, the major constituent of functional cartilage. We discovered a synergistic combination that overcame synovial inflammation to form type II collagen-producing chondrocytes. The combination consists of peroxisome proliferator-activated receptor (PPAR) δ agonist, human bone marrow (hBM)-derived MSCs, and hyaluronic acid (HA) gel. Interestingly, those individual components showed their own strong enhancing effects on chondrogenesis. GW0742, a PPAR-δ agonist, greatly enhanced MSC chondrogenesis and the expression of type II collagen and glycosaminoglycan (GAG) in hBM-MSC-derived chondrocytes. GW0742 also increased the expression of transforming growth factor ß that enhances chondrogenesis and suppresses cartilage fibrillation, ossification, and inflammation. HA gel also increased MSC chondrogenesis and GAG production. However, neither GW0742 nor HA gel could enhance the formation of type II collagen-producing chondrocytes from hBM-MSCs within human OA synovial fluid. Our data demonstrated that the combination of hBM-MSCs, PPAR-δ agonist, and HA gel significantly enhanced the formation of type II collagen-producing chondrocytes within OA synovial fluid from 3 different donors. In other words, the novel combination of PPAR-δ agonist, hBM-MSCs, and HA gel can overcome synovial inflammation to form type II collagen cartilage within human OA synovial fluid. This novel articularly injectable formula could improve OA treatment in the future clinical application.


Asunto(s)
Condrocitos/metabolismo , Colágeno Tipo II/metabolismo , Células Madre Mesenquimatosas/metabolismo , PPAR delta/metabolismo , Células Cultivadas , Humanos , Células Madre Mesenquimatosas/citología , Líquido Sinovial/metabolismo
13.
Arch Pharm Res ; 40(8): 933-942, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28736799

RESUMEN

Magnolol, honokiol, and obovatol are well known bioactive constituents of the bark of Magnolia officinalis and have been reported to have beneficial effects in various diseases. We recently isolated a novel active compound, 4-O-methylhonokiol (4-O-MH) from the ethanol extract of M. officinalis, which was previously reported to have pharmacological effects including anti-inflammatory, anti-oxidative, and anti-aging activities. Here, we examined the pharmacological properties of 4-O-MH on osteoblast (bone-forming cells) and osteoclast (bone-resorbing cells) differentiation, and its underlying signaling pathways in primary cultured pre-osteoblasts and bone marrow macrophages. Our results showed that 4-O-MH did not affect cell viability in pre-osteoblasts and did not influence osteoblast differentiation and mineralized nodule formation, as assessed by alkaline phosphatase activity and Alizarin red staining. However, 4-O-MH significantly inhibited TRAP-positive multinuclear osteoclasts and F-actin ring formation during Receptor activator of NF-κB ligand (RANKL)-mediated osteoclastogenesis without cytotoxicity. In addition, 4-O-MH suppressed RANKL-induced critical factors (c-Fos, NF-ATc1, TRAP, and ITB3) for osteoclast differentiation and function. Furthermore, RANKL-mediated signaling, including ERK1/2, AKT, and NF-kB pathways was attenuated by 4-O-MH. Taken together, 4-O-MH has an inhibitory role in RANKL-mediated osteoclastogenesis but not osteoblast differentiation, and our findings also suggest that 4-O-MH is a potential therapeutic agent for bone-destructive diseases such as osteoporosis, alveolar bone resorption, and osteoarthritis.


Asunto(s)
Compuestos de Bifenilo/farmacología , Lignanos/farmacología , Macrófagos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Ligando RANK/metabolismo , Animales , Enfermedades Óseas/tratamiento farmacológico , Enfermedades Óseas/fisiopatología , Diferenciación Celular/efectos de los fármacos , Macrófagos/metabolismo , Magnolia/química , Ratones , Ratones Endogámicos ICR , FN-kappa B/metabolismo , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteoclastos/citología , Osteoclastos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
14.
Arch Pharm Res ; 40(5): 601-609, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28386742

RESUMEN

This study was performed to examine the anticancer and anti-metastatic effects of 4-parvifuran (PVN), a novel flavonoid isolated from the heartwood of Dalbergia odorifera, and to study its underlying signaling pathway in human osteosarcoma cells. In the present study, PVN was found to inhibit cell proliferation in a concentration- and time-dependent manner in the human osteosarcoma cell lines studied (MG-63 and U-2 OS) and induce apoptosis, as evidenced by Annexin V+ and TUNEL+ cells. Cleaved poly (ADP-ribose) polymerase (PARP) and caspase-3 were up-regulated while anti-apoptotic proteins including Bcl-2, Bcl-xL, and survivin were down-regulated after treatment with PVN. Matrigel cell migration assay, invasion assay, and soft agar assay were used to show that PVN effectively suppressed cell migration and invasion and colony formation in osteosarcoma cells. Protein and mRNA levels of MMP-2 and MMP-9 were reduced by PVN in a concentration-dependent manner. Furthermore, PVN inhibited Janus kinase 2 (JAK2)/signal transducers and activators of transcription 3 (STAT3), mitogen-activated protein kinases (MAPKs) including JNK, ERK, p38 kinase, and cAMP response element-binding protein (CREB). Therefore, this is the first study to demonstrate that PVN might be a novel anticancer and anti-metastatic agent for the treatment of osteosarcoma through the inhibition of JAK2/STAT3, MAPKs, and CREB signaling pathways.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Neoplasias Óseas/tratamiento farmacológico , Dalbergia/química , Flavonoides/farmacología , Janus Quinasa 2/antagonistas & inhibidores , Osteosarcoma/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Factor de Transcripción STAT3/antagonistas & inhibidores , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/aislamiento & purificación , Neoplasias Óseas/patología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Flavonoides/química , Flavonoides/aislamiento & purificación , Humanos , Janus Quinasa 2/metabolismo , Estructura Molecular , Osteosarcoma/patología , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/aislamiento & purificación , Factor de Transcripción STAT3/metabolismo , Relación Estructura-Actividad , Células Tumorales Cultivadas
15.
J Endod ; 43(3): 417-424, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28231980

RESUMEN

INTRODUCTION: The aims of this study were to examine the immunolocalization of protein phosphatase 1 (PP1) in developing mouse pulp tissue and to explore the role of PP1 in odontoblastic differentiation and in vitro angiogenesis in human dental pulp cells (HDPCs). METHODS: Immunolocalization of PP1 was assessed in developing mouse pulp tissue. Odontogenic differentiation was examined by alkaline phosphatase activity, alizarin red staining, and reverse transcriptase polymerase chain reaction. Angiogenesis was evaluated by endothelial cell migration and capillary tube formation. Signaling pathways were analyzed by Western blotting and confocal immunofluorescence. RESULTS: PP1 expression was detected in preodontoblasts, odontoblasts, dental pulp cells, and endothelial cells within pulp tissue during the crown formed, root formation, and root completion stages. PP1 messenger RNA (mRNA) and protein levels were up-regulated at the late mineralization stage during odontogenic differentiation of HDPCs. The PP1 activator C2 ceramide increased alkaline phosphatase activity, mineralized nodule formation, and mRNA expression of dentin matrix protein 1 and dentin sialophosphoprotein. In contrast, knockdown by PP1 small interfering RNA inhibited odontoblastic differentiation. Moreover, PP1 activator up-regulated mRNA expression of angiogenic genes in HDPCs and increased the migration and capillary tube formation of endothelial cells, whereas PP1 small interfering RNA showed opposite effects. C2 ceramide increased levels of bone morphogenetic protein 2, phosphorylation of Smad 1/5/8, and mRNA expression of runt-related transcription factor 2 and osterix. CONCLUSIONS: This study provides the first evidence that PP1 might be a potent regulator of developing pulp tissue in vivo and odontoblastic differentiation and angiogenesis in HDPCs in vitro and may have clinical implications for pulp/dentin regeneration or reparative dentinogenesis.


Asunto(s)
Diferenciación Celular , Pulpa Dental/citología , Neovascularización Fisiológica/fisiología , Proteína Fosfatasa 1/metabolismo , Animales , Movimiento Celular , Células Cultivadas , Pulpa Dental/irrigación sanguínea , Silenciador del Gen , Humanos , Ratones , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Regulación hacia Arriba
16.
Cell Transplant ; 26(6): 1001-1016, 2017 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-28105979

RESUMEN

We compared the therapeutic effects and mechanism of transplanted human dental pulp stem cells (hDPSCs) and human bone marrow-derived mesenchymal stem cells (hBM-MSCs) in a rat stroke model and an in vitro model of ischemia. Rats were intravenously injected with hDPSCs or hBM-MSCs 24 h after middle cerebral artery occlusion (MCAo), and both groups showed improved functional recovery and reduced infarct volume versus control rats, but the hDPSC group showed greater reduction in infarct volume than the hBM-MSC group. The positive area for the endothelial cell marker was greater in the lesion boundary areas in the hDPSC group than in the hBM-MSC group. Administration of hDPSCs to rats with stroke significantly decreased reactive gliosis, as evidenced by the attenuation of MCAo-induced GFAP+/nestin+ and GFAP+/Musashi-1+ cells, compared with hBM-MSCs. In vivo findings were confirmed by in vitro data illustrating that hDPSCs showed superior neuroprotective, migratory, and in vitro angiogenic effects in oxygen-glucose deprivation (OGD)-injured human astrocytes (hAs) versus hBM-MSCs. Comprehensive comparative bioinformatics analyses from hDPSC- and hBM-MSC-treated in vitro OGD-injured hAs were examined by RNA sequencing technology. In gene ontology and KEGG pathway analyses, significant pathways in the hDPSC-treated group were the MAPK and TGF-ß signaling pathways. Thus, hDPSCs may be a better cell therapy source for ischemic stroke than hBM-MSCs.


Asunto(s)
Células de la Médula Ósea/citología , Isquemia Encefálica/terapia , Pulpa Dental/citología , Células Madre Mesenquimatosas/citología , Células Madre/citología , Animales , Modelos Animales de Enfermedad , Rechazo de Injerto , Humanos , Inmunidad Innata/fisiología , Etiquetado Corte-Fin in Situ , Macrófagos/citología , Macrófagos/fisiología , Células Madre Mesenquimatosas/fisiología , Neutrófilos/citología , Neutrófilos/fisiología , Epitelio Pigmentado de la Retina/citología , Células Madre/fisiología , Linfocitos T/citología , Linfocitos T/fisiología
17.
Clin Oral Implants Res ; 28(4): 396-405, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26972335

RESUMEN

OBJECTIVES: The aim of this study was to investigate the effects of static magnetic fields (SMFs) on bone regeneration around titanium implants by µCT, histologic analysis, microarrays, and quantitative real-time PCR (qRT-PCR). MATERIALS AND METHODS: Neodymium magnets provided the source of SMFs, the specimens were grade 5 titanium implants, and the animals were twenty-seven adult male New Zealand white rabbits. These implants were divided into six groups according to the presence of a magnet and predetermined healing period (1, 4, and 8 weeks). Each group comprised six specimens for µCT (n = 6) and histologic examination, and three specimens (n = 3) for microarrays and qRT-PCR, yielding a total of 54 specimens. RESULTS: The µCT data showed that SMFs increased bone volume fraction (bone volume/total volume, BV/TV), trabecular number (Tb.N), and trabecular thickness (Tb.Th). Histologic observation indicated that SMFs promoted new bone formation and direct bony contact with implants. Microarray analysis identified 293 genes upregulated (>twofold) in response to SMFs. The upregulated genes included extracellular matrix (ECM)-related genes (COL10A1, COL9A1, and COL12A1) and growth factor (GF)-related genes (CTGF and PDGFD), and the upregulation was confirmed by qRT-PCR. Gene Ontology (GO) and pathway analysis revealed the involvement of the mitogen-activated protein kinase (MAPK), Wnt, and PPAR-gamma signaling pathways in implant healing. CONCLUSIONS: µCT, histology, microarrays, and real-time PCR indicate that SMFs could be an effective approach to improving bone regeneration around dental implants.


Asunto(s)
Regeneración Ósea/fisiología , Implantes Dentales , Magnetoterapia/métodos , Titanio , Animales , Masculino , Conejos , Reacción en Cadena en Tiempo Real de la Polimerasa , Análisis de Matrices Tisulares , Microtomografía por Rayos X
18.
Muscle Nerve ; 56(3): 510-518, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-27977864

RESUMEN

INTRODUCTION: N-methyl-d-aspartate (NMDA) is expressed in sensory neurons and plays important roles in peripheral pain mechanisms. The aim of this study was to examine the effects and molecular mechanisms of NMDA on C2C12 myoblast proliferation and differentiation. METHODS: Cytotoxicity and differentiation were examined by the MTT assay, reverse transcription-polymerase chain reaction, and immunofluorescence. RESULTS: NMDA had no cytotoxicity (10-500 µM) and inhibited myoblastic differentiation of C2C12 cells, as assessed by F-actin immunofluorescence and levels of mRNAs encoding myogenic markers such as myogenin and myosin heavy-chain 2. It inhibited phosphorylation of mammalian target of rapamycin (mTOR) by inactivating mitogen-activated protein kinases (MAPKs), including extracellular signal-regulated kinase, c-Jun N-terminal kinase, and p38. It induced reactive oxygen species production. Furthermore, NMDA-suppressed expression of F-actin was reversed by adding the antioxidant N-acetylcysteine. CONCLUSIONS: Collectively, these results indicate that NMDA impairs myogenesis or myogenic differentiation in C2C12 cells through the mTOR/MAPK signaling pathways and may lead to skeletal muscle degeneration. Muscle Nerve 56: 510-518, 2017.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Desarrollo de Músculos/efectos de los fármacos , Mioblastos/efectos de los fármacos , N-Metilaspartato/toxicidad , Animales , Diferenciación Celular/fisiología , Línea Celular , Proliferación Celular/fisiología , Ratones , Desarrollo de Músculos/fisiología , Mioblastos/metabolismo , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo
19.
J Biomed Nanotechnol ; 13(2): 180-91, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29377648

RESUMEN

A xerogel was interfaced onto biopolymer nanofibers though a core­shell electrospinning design for bone regeneration. The xerogel-interfaced biopolymer nanofibrous matrix was bioactive and highly hydrophilic, with a significant decrease in the water contact angle. The matrix showed excellent in vitro responses of primary osteoblasts in terms of adhesion, proliferation, and migration. Furthermore, the osteoblastic differentiation of cells, including alkaline phosphatase activity, mineralization, and gene expression, was significantly upregulated by the xerogel interface. In vivo animal tests in a critical-sized calvarial defect confirmed the new bone formation ability of the xerogel-surfaced nanofiber matrices. The underlying signaling mechanisms of the stimulation were implied to be integrin and bone morphogenetic protein (BMP) pathways, as demonstrated by the activation of integrin (α2ß1) and downstream signaling molecules (FAK, paxillin, RhoA, MAPK, and NF-κB), as well as the BMPs and the downstream transcription factor Smad1/5/8. Taking these findings together, the xerogel-surfaced biopolymer nanofibers are proposed to be a promising scaffold candidate for bone regeneration.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Regeneración Ósea/efectos de los fármacos , Geles/farmacología , Integrinas/metabolismo , Nanofibras/química , Animales , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Geles/química , Ratones , Ratones Endogámicos ICR , Osteoblastos/efectos de los fármacos , Andamios del Tejido/química
20.
J Nanosci Nanotechnol ; 17(1): 143-52, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29617095

RESUMEN

In the present study, we evaluated the potential of poly-l-lysine/hyaluronic acid (HA/PLL) hydrogels containing curcumin (CUR) and bone morphogenetic protein-2 (BMP-2) as bone tissue regeneration scaffolds. Hydrogels HP-1˜2 were formed by amide bonds via the condensation reactions between 0.02 µmol HA and 0.06­0.12 µmol poly-l-lysine · hydrobromide (PLL · HBr). Physical, chemical, and thermal analyses revealed that the amount of PLL · HBr significantly influenced hydrogel properties. Based on an In Vitro MG-63 cell proliferation test, HP-1˜2 were cytocompatible, and all hydrogels containing different amounts of CUR and BMP-2, except for HA0.02/PLL0.06/CUR20/BMP-2100 (HPCB-4), resulted in cell proliferation above 80%. An In Vitro release test showed that CUR and BMP-2 were consistently released from HA0.02/PLL0.06/CUR15 (HPC), HA0.02/PLL0.06/BMP-2100 (HPB), HA0.02/PLL0.06/CUR15/BMP-210 , 50 , or 100 (HPCB-1˜3), and HA0.02/PLL0.06/CUR10 or 20/BMP-2100 (HPCB-4˜5) for 7 and 28 days, respectively. In Vitro ALP activity and calcium deposition and In Vivo micro-computed tomography (micro-CT) tests demonstrated the potential application of HPCB-3 as bone tissue regeneration scaffolds, suggesting that bone tissue regeneration can be optimized by controlling the amounts of CUR and BMP-2.


Asunto(s)
Proteína Morfogenética Ósea 2/farmacología , Curcumina/farmacología , Osteogénesis/efectos de los fármacos , Ingeniería de Tejidos/métodos , Animales , Regeneración Ósea/efectos de los fármacos , Ácido Hialurónico/química , Hidrogeles/química , Ratones , Polilisina/química , Andamios del Tejido/química , Microtomografía por Rayos X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...