Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
J Cell Biochem ; 120(3): 4057-4064, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30260015

RESUMO

OBJECTIVE: The current study was designed to investigate the effects and underlying mechanisms of adipose tissue-derived stem cells (ADSCs) on hypertrophic scar (HS) fibrosis. METHOD: Real-time quantitative polymerase chain reaction (qRT-PCR) and Western blot analysis were performed to detect the expression of collagen I (Col1), collagen III (Col3), and α-smooth muscle actin (α-SMA) after fibroblasts and cultured HS tissues were treated with ADSC medium. All data were analyzed by using SPSS17.0 software. Statistical analysis was performed by Student t tests. RESULTS: The in vitro study showed that ADSC medium decreased the expression of Col1, Col3, and α-SMA. In addition, the protein level of p-p38 was downregulated by ADSC medium treatment in a concentration dependent manner. CONCLUSION: The current study demonstrated that ADSC could decrease collagen deposition and scar formation in in vitro experiments. The regulation of the p38/MAPK signaling pathway might play an important role in the process.


Assuntos
Cicatriz Hipertrófica/genética , Fibrose/genética , Células-Tronco Mesenquimais/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Células Cultivadas , Cicatriz Hipertrófica/patologia , Colágeno Tipo I/genética , Colágeno Tipo III/genética , Fibroblastos/metabolismo , Fibrose/patologia , Regulação da Expressão Gênica/genética , Humanos , Transdução de Sinais/genética , Cicatrização/genética
2.
Biomedicines ; 10(8)2022 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-36009462

RESUMO

Large bone defects may develop fracture nonunion, leading to disability and psychosocial burdens. Bone grafting with anabolic agents is a good autografting alternative. Simvastatin, as a cholesterol-lowering agent worldwide, is proven to enhance osteogenesis. Considering its dose-dependent adverse effects, we developed a simvastatin derivative, named KMUHC-01, which has bone anabolic capacity and lower cytotoxicity than simvastatin. We hypothesize that KMUHC-01 could help bone formation in bone-defect animal models. We used rat models of critical calvarial and long-bone defects to evaluate the effects of KMUHC-01 and simvastatin on biological changes at the bone defect through histology, immunohistology, and mechanical testing using three-point bending and evaluated the new bone formation microstructure through microcomputed tomography analysis. The newly formed bone microstructure at the calvarial defect site showed a significantly improved trabecular bone volume in the KMUHC-01 1-µM group compared with that in the control and simvastatin groups. The biomechanical study revealed a significantly increased maximal strength in the KMUHC-01 1-µM group compared with that in the control group. KUMHC-01, as a simvastatin derivative, showed a great anabolic effect in promoting bone defect healing. However, further studies will be conducted to prove the bioavailability and bone-forming efficacy of KMUHC-01 via systemic administration.

3.
Biol Open ; 9(12)2020 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-33355167

RESUMO

Hypertrophic scar (HS) is a dermal fibro-proliferative disorder result from abnormal wound healing after skin injury. MicroRNA-9-5p (miR-9-5p) has been reported to be upregulated and closely related to collagen proteins in human dermal fibroblasts. However, the correlation and possible mechanism between miR-9-5p and HS require further investigation. The expressions of miR-9-5p in HS tissues and HS fibroblasts were detected by quantitative real-time PCR (RT-qPCR). The expression level of peroxisome proliferator-activated receptor ß (PPARß) was measured by RT-qPCR assay. The protein levels of PPARß, α-SMA, Vimentin, COL1A, cyclin D1, bcl-2, and bax were detected by western blot assay. The effect of miR-9-5p and PPARß on HS fibroblasts proliferation and apoptosis were detected by cell counting kit-8 (CCK-8) and flow cytometry assays. The interaction between miR-9-5p and PPARß was predicted by TargetScan, and then confirmed by dual-luciferase reporter assay. MiR-9-5p expression was downregulated in HS tissues and HS fibroblasts. MiR-9-5p inhibited the levels of extracellular matrix-associated genes (α-SMA, Vimentin, COL1A) in HS fibroblasts. MiR-9-5p repressed proliferation and induced apoptosis of HS fibroblasts. PPARß is a target gene of miR-9-5p. The silencing of PPARß expression hindered proliferation and expedited apoptosis of HS fibroblasts. MiR-9-5p suppressed proliferation and promoted apoptosis of HS fibroblasts by targeting PPARß. In this paper, we firstly disclosed that miR-9-5p hampered extracellular matrix deposition and proliferation, and induced apoptosis by targeting PPARß in HS fibroblasts. Our findings provided a new role of miR-9-5p/PPARß in the occurrence and development of HS fibroblasts, promising a new target for HS.


Assuntos
Apoptose/genética , Cicatriz Hipertrófica/genética , Cicatriz Hipertrófica/metabolismo , Fibroblastos/metabolismo , Regulação da Expressão Gênica , MicroRNAs/genética , PPAR beta/genética , Proliferação de Células/genética , Células Cultivadas , Cicatriz Hipertrófica/patologia , Matriz Extracelular/metabolismo , Humanos , PPAR beta/metabolismo , Interferência de RNA
4.
Mater Sci Eng C Mater Biol Appl ; 105: 109879, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31546456

RESUMO

In this study, a porous Ti-alloy based implant with an interconnected channel structure (MAO-CaP-BMP2) is fabricated using a method combining 3D printing, microarc oxidation (MAO) treatment, and co-precipitation of Ca,P layer with BMP-2 technique. The macroporous structure with pore size of 600 µm made by 3D printing not only enhances the ingrowth of cells but also allows the formation of blood vessels inside the implant. As a result, the new bond formation is promoted. In addition, the microporous dioxide layer formed on the implant surface by MAO provides the sites for co-precipitation of Ca,P layer with BMP-2. The microstructure allows the prolonged release of BMP-2. Our results show that a sustained release of BMP-2 over 35 days is achieved for MAO-CaP-BMP2 group longer than Ti without MAO modification group and without Ca,P electrochemical deposition group. The slow release of BMP-2 at the bone/implant interface for a long period of time leads to enhancement of the osseointegration between the implant and surrounding bones. This result indicates that MAO-CaP-BMP2 is a good candidate of growth factor carrier. Successful regeneration of bone requires the concomitant processes of osteogenesis and neovascularization. MAO-CaP-BMP2 modified Ti-alloy implant is both osteoinductive and osteoconductive which can create better osteogenesis and angiogenesis. As a result, it can enhance bone formation.


Assuntos
Proteína Morfogenética Óssea 2/farmacologia , Galvanoplastia , Osteogênese/efeitos dos fármacos , Titânio/química , Fator de Crescimento Transformador beta/farmacologia , Ligas/farmacologia , Animais , Fosfatos de Cálcio/farmacologia , Materiais Revestidos Biocompatíveis/farmacologia , Preparações de Ação Retardada/farmacologia , Liberação Controlada de Fármacos , Humanos , Camundongos , Neovascularização Fisiológica/efeitos dos fármacos , Oxirredução , Próteses e Implantes , Coelhos , Proteínas Recombinantes/farmacologia , Crânio/efeitos dos fármacos , Crânio/patologia
5.
Integr Cancer Ther ; 16(3): 397-405, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-27879376

RESUMO

BACKGROUND: Osteosarcoma is a malignant bone tumor prevalent in adolescents with poor prognosis. Toona sinensis showed potent antiproliferation effect on lung, melatonin, ovary, colon, and liver cancers. However, the effects of the species on osteosarcoma cells are rarely investigated. RESULTS: In this study, we found fraction 1 of Toona sinensis leaf (TSL-1) resulted in inhibition of cell viability in MG-63, Saos-2, and U2OS osteosarcoma cell lines, while it only caused a moderate suppressive effect on normal osteoblasts. In addition, TSL-1 significantly elevated lactate dehydrogenase leakage and induced apoptosis and necrosis in Saos-2 cells. TSL-1 increased mRNA expression of pro-apoptotic factor Bad. Most important, TSL-1 significantly suppressed Saos-2 xenograft tumor growth in nude mice by increasing caspase-3. The IC-50 of TSL-1 for the 3 tested osteosarcoma cells is around 1/9 of that for lung cancer cells. CONCLUSION: We demonstrated that TSL-1, a fractionated extract from TSL, caused significant cytotoxicity to osteosarcoma cells due to apoptosis. In vivo xenograft study showed that TSL-1 suppressed the growth of osteosarcoma cells at least in part by inducing apoptosis. Our results indicate that TSL-1 has potential to be a promising anti-osteosarcoma adjuvant functional plant extract.


Assuntos
Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Meliaceae/química , Osteossarcoma/tratamento farmacológico , Extratos Vegetais/farmacologia , Folhas de Planta/química , Animais , Caspase 3/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Masculino , Camundongos , Camundongos Nus , Osteossarcoma/metabolismo
6.
Int J Nanomedicine ; 10: 5881-94, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26451103

RESUMO

Recent studies have indicated that statins induce osteogenic differentiation both in vitro and in vivo. The molecular mechanism of statin-stimulated osteogenesis is unknown. Activation of RhoA signaling increases cytoskeletal tension, which plays a crucial role in the osteogenic differentiation of mesenchymal stem cells. We thus hypothesized that RhoA signaling is involved in simvastatin-induced osteogenesis in bone marrow mesenchymal stem cells. We found that although treatment with simvastatin shifts localization of RhoA protein from the membrane to the cytosol, the treatment still activates RhoA dose-dependently because it reduces the association with RhoGDIα. Simvastatin also increased the expression of osteogenic proteins, density of actin filament, the number of focal adhesions, and cellular tension. Furthermore, disrupting actin cytoskeleton or decreasing cell rigidity by using chemical agents reduced simvastatin-induced osteogenic differentiation. In vivo study also confirms that density of actin filament is increased in simvastatin-induced ectopic bone formation. Our study is the first to demonstrate that maintaining intact actin cytoskeletons and enhancing cell rigidity are crucial in simvastatin-induced osteogenesis. The results suggested that simvastatin, which is an osteoinductive factor and acts by increasing actin filament organization and cell rigidity combined with osteoconductive biomaterials, may benefit stem-cell-based bone regeneration.


Assuntos
Citoesqueleto de Actina/metabolismo , Células-Tronco Mesenquimais/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Sinvastatina/farmacologia , Proteína rhoA de Ligação ao GTP/metabolismo , Actinas/metabolismo , Fosfatase Alcalina/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Citoesqueleto/metabolismo , Masculino , Células-Tronco Mesenquimais/citologia , Camundongos , Microscopia de Força Atômica , Microscopia de Fluorescência , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais/efeitos dos fármacos , Sinvastatina/administração & dosagem
7.
Biochem Pharmacol ; 98(3): 453-64, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26410676

RESUMO

Simvastatin, an HMG-CoA reductase inhibitor, is known to promote osteogenic differentiation. However, the mechanism underlying simvastatin-induced osteogenesis is not well understood. In this study, we hypothesize that the estrogen receptor (ER) mediates simvastatin-induced osteogenic differentiation. ER antagonists and siRNA were used to determine the involvement of the ER in simvastatin-induced osteogenesis in mouse bone marrow mesenchymal stem cells (D1 cells). Osteogenesis was evaluated by mRNA expression, protein level/activity of osteogenic markers, and mineralization. The estrogen response element (ERE) promoter activity and the ER-simvastatin binding affinity were examined. Our results showed that the simvastatin-induced osteogenic effects were decreased by treatment with ERα antagonists and ERα siRNA but not by an antagonist specific for the G protein-coupled estrogen receptor (GPER-1). The simvastatin-induced osteogenic effects were further increased by E2 treatment and were reversed by ERα antagonists or siRNA treatment. Luciferase reporter gene assays demonstrated that simvastatin increase ERα-dependent transcriptional activity that was suppressed by ERα antagonists. Furthermore, the ERα-simvastatin binding assay showed that IC50 value of simvastatin is 7.85 µM and that of E2 is 32.8 nM, indicating that simvastatin is a weak ligand for ERα. These results suggest that simvastatin-stimulated osteogenesis is mediated by ERα but not GPER-1. Moreover, this is the first report to demonstrate that simvastatin acts as an ERα ligand and a co-activator to enhance ERα-dependent transcriptional activity and thus promotes osteogenesis. These results indicate that simvastatin-induced osteogenesis is mediated via an ERα-dependent pathway.


Assuntos
Células da Medula Óssea/efeitos dos fármacos , Estradiol/farmacologia , Receptor alfa de Estrogênio/fisiologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Sinvastatina/farmacologia , Animais , Células da Medula Óssea/citologia , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Antagonistas de Estrogênios/farmacologia , Humanos , Células-Tronco Mesenquimais/citologia , Camundongos
8.
PLoS One ; 9(3): e91581, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24632682

RESUMO

Pulsed electromagnetic field (PEMF) has been successfully applied to accelerate fracture repair since 1979. Recent studies suggest that PEMF might be used as a nonoperative treatment for the early stages of osteonecrosis. However, PEMF treatment requires a minimum of ten hours per day for the duration of the treatment. In this study, we modified the protocol of the single-pulsed electromagnetic field (SPEMF) that only requires a 3-minute daily treatment. In the in vitro study, cell proliferation and osteogenic differentiation was evaluated in the hBMSCs. In the in vivo study, new bone formation and revascularization were evaluated in the necrotic bone graft. Results from the in vitro study showed no significant cytotoxic effects on the hBMSCs after 5 days of SPEMF treatment (1 Tesla, 30 pulses per day). hBMSC proliferation was enhanced in the SPEMF-treated groups after 2 and 4 days of treatment. The osteogenic differentiation of hBMSCs was significantly increased in the SPEMF-treated groups after 3-7 days of treatment. Mineralization also increased after 10, 15, 20, and 25 days of treatment in SPEMF-treated groups compared to the control group. The 7-day short-course treatment achieved similar effects on proliferation and osteogenesis as the 25-day treatment. Results from the in vivo study also demonstrated that both the 7-day and 25-day treatments of SPEMF increased callus formation around the necrotic bone and also increased new vessel formation and osteocyte numbers in the grafted necrotic bone at the 2nd and 4th weeks after surgery. In conclusion, the newly developed SPEMF accelerates osteogenic differentiation of cultured hBMSCs and enhances bone repair, neo-vascularization, and cell growth in necrotic bone in mice. The potential clinical advantage of the SPEMF is the short daily application and the shorter treatment course. We suggest that SPEMF may be used to treat fractures and the early stages of osteonecrosis.


Assuntos
Regeneração Óssea , Diferenciação Celular , Campos Eletromagnéticos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos da radiação , Osteogênese/efeitos da radiação , Fosfatase Alcalina/metabolismo , Animais , Calcificação Fisiológica , Proliferação de Células/efeitos da radiação , Campos Eletromagnéticos/efeitos adversos , Ativação Enzimática , Feminino , Humanos , Imuno-Histoquímica , Masculino , Camundongos
9.
Int J Nanomedicine ; 8: 3895-904, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24143094

RESUMO

Statins are used clinically for reduction of cholesterol synthesis to prevent cardiovascular disease. Previous in vitro and in vivo studies have shown that statins stimulate bone formation. However, orally administered statins may be degraded during first-pass metabolism in the liver. This study aimed to prevent this degradation by developing a locally administered formulation of simvastatin that is encapsulated in poly(lactic-co-glycolic acid)/hydroxyapatite (SIM/PLGA/HAp) microspheres with controlled-release properties. The effect of this formulation of simvastatin on bone repair was tested using a mouse model of gap fracture bridging with a graft of necrotic bone. The simvastatin released over 12 days from 3 mg and 5 mg of SIM/PLGA/HAp was 0.03-1.6 µg/day and 0.05-2.6 µg/day, respectively. SIM/PLGA/HAp significantly stimulated callus formation around the repaired area and increased neovascularization and cell ingrowth in the grafted necrotic bone at week 2 after surgery. At week 4, both 3 mg and 5 mg of SIM/PLGA/HAp increased neovascularization, but only 5 mg SIM/PLGA/HAp enhanced cell ingrowth into the necrotic bone. The low dose of simvastatin released from SIM/PLGA/HAp enhanced initial callus formation, neovascularization, and cell ingrowth in the grafted bone, indicating that SIM/PLGA/HAp facilitates bone regeneration. We suggest that SIM/PLGA/HAp should be developed as an osteoinductive agent to treat osteonecrosis or in combination with an osteoconductive scaffold to treat severe bone defects.


Assuntos
Regeneração Óssea/efeitos dos fármacos , Preparações de Ação Retardada/administração & dosagem , Durapatita/química , Ácido Láctico/química , Osteonecrose/tratamento farmacológico , Osteonecrose/patologia , Ácido Poliglicólico/química , Sinvastatina/administração & dosagem , Animais , Cápsulas/administração & dosagem , Cápsulas/síntese química , Preparações de Ação Retardada/síntese química , Difusão , Camundongos , Camundongos Endogâmicos BALB C , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Sinvastatina/química , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA