Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Pharmacol Res ; 162: 105267, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33127556

RESUMO

Cystic fibrosis (CF) is a lethal autosomal recessive genetic disease, caused by a mutation in the cystic fibrosis transmembrane conductance regulator gene (CFTR), which primarily affects the lungs and digestive system. This gene encodes the CFTR protein, a distinctive membrane transporter of the ATP-binding cassette (ABC) superfamily. It functions as a chloride channel, allowing the balance and transport of chloride through the apical membrane of epithelial cells. Due to its ubiquitous location, mutations in the CFTR gene trigger multiple changes in ion transport and metabolic pathways, affecting various organs, as it will be herein explained. Pulmonary impairment is the most characteristic comorbidity of CF and respiratory failure is the main cause of death. This review presents the importance of an early diagnosis of CF to establish, as soon as possible, a primary therapy for symptomatic prevention and relief. It also mentions new therapeutic approaches that include CFTR modulators. They are correctors and/or potentiators of the deficient CFTR channel. In an attempt to overcome the disadvantages of CFTR modulators, the application of biotechnology techniques is addressed, such as gene therapy, gene editing, RNA therapy and therapeutic microRNAs. The potential of the intranasal administration route is another presented aspect.


Assuntos
Fibrose Cística , Animais , Biotecnologia , Fibrose Cística/diagnóstico , Fibrose Cística/tratamento farmacológico , Fibrose Cística/genética , Fibrose Cística/fisiopatologia , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/fisiologia , Humanos
2.
Cell Biol Int ; 40(10): 1123-8, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27460177

RESUMO

A study was conducted to examine the effect of phenamil on adipogenic differentiation and expression of key adipogenic transcripts in hen preadipocytes. Preadipocytes were isolated from 20-week old Single Comb White Leghorn hens (Gallas gallus, Lohman strain). The experiment lasted for 48 h and had six treatments. Non-treated control (C) cells, cells treated with dexamethasone, 3-isobutyl-1-methylxanthine, insulin, and oleic acid (DMIOA) (T1), DMIOA + 15 µM phenamil (T2), DMIOA + 30 µM phenamil (T3), 15 µM phenamil alone (T4), and 30 µM phenamil alone (T5). Neutral lipid accumulation and the mRNA expression of key adipogenic transcripts were measured in all treatments and compared. Lipid accumulation was detected in T1, T2, and T3 only. Expression of peroxisome proliferator receptor-activator gamma 2 (PPARγ2), the core enhancer binding protein α (C/EBPα), C/EBPß, fatty acid binding protein 4 (FABP4), and lipoprotein lipase (LPL) as well as ETS variant 4 (ETV4) and 5 was higher (P < 0.05) in T2, T3, T4, and T5 compared to C. Expression of these transcripts was higher (P < 0.05) in T2 and T3 compared to T4 and T5. The core enhancer binding protein α, C/EBPß, and FABP4 were highly expressed (P < 0.05) in T1 compared to C. However, the expression of PPARγ2, LPL, and ETV4 and ETV5 was not significantly different. Expression of C/EBPα, C/EBPß, and FABP4 was higher (P < 0.05) in T2 and T3 compared to T1. Expression of sterol regulatory element binding protein 1 (SREBP1) and leptin receptor (LEPR) was not significantly different among the treatments. In conclusion, phenamil enhances DMIOA-induced adipogenic differentiation of hen preadipocytes but does not induce adipogenesis by itself.


Assuntos
Adipócitos/efeitos dos fármacos , Adipogenia/efeitos dos fármacos , Amilorida/análogos & derivados , Amilorida/farmacologia , Animais , Proteína alfa Estimuladora de Ligação a CCAAT/metabolismo , Proteína beta Intensificadora de Ligação a CCAAT/genética , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Galinhas , Proteínas de Ligação a Ácido Graxo/metabolismo , Feminino , Ácido Oleico/metabolismo , PPAR gama/metabolismo
3.
Arch Insect Biochem Physiol ; 93(2): 67-85, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27357470

RESUMO

Larvae of Chironomus riparius respond to ion-poor and brackish water (IPW, BW) conditions by activating ion uptake mechanisms in the anal papillae and reducing ion absorption at the rectum, respectively. The role that the Malpighian tubules play in ion and osmoregulation under these conditions is not known in this species. This study examines rates of fluid secretion and major cation composition of secreted fluid from tubules of C. riparius reared in IPW, freshwater (FW) and BW. Fluid secretion of tubules from FW and BW larvae was similar but tubules from IPW larvae secrete fluid at higher rates, are more sensitive to serotonin stimulation, and the secreted fluid contains less Na(+) . Therefore in IPW, tubules work in concert with anal papillae to eliminate excess water while conserving Na(+) in the hemolymph. Tubules do not appear to play a significant role in ion/osmoregulation under BW. Serotonin immunoreactivity in the nervous system and gastrointestinal tract of larval C. riparius was similar to that seen in mosquito larvae with the exception that the hindgut was devoid of staining. Hemolymph serotonin titer was similar in FW and IPW; hence, serotonin is not responsible for the observed high rates of fluid secretion in IPW. Instead, it is suggested that serotonin may work in a synergistic manner with an unidentified hormonal factor in IPW. Ion transport mechanisms in the tubules of C. riparius are pharmacologically similar to those of other insects.


Assuntos
Chironomidae/fisiologia , Túbulos de Malpighi/metabolismo , Salinidade , Serotonina/metabolismo , Sódio/farmacologia , Animais , Chironomidae/efeitos dos fármacos , Chironomidae/crescimento & desenvolvimento , Transporte de Íons , Larva/crescimento & desenvolvimento , Larva/fisiologia
4.
Eur J Oral Sci ; 121(6): 559-65, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24102669

RESUMO

Dentin formation is preferred in the healing response of the pulp to pulp-capping agents during vital pulp therapy. Enhancement of the dentinogenic differentiation of dental pulp cells is thought to accelerate pulp repair. The aim of this study was to evaluate the dentinogenic activity of small molecules (three flavonoids and phenamil) that have been shown previously to induce osteoblast differentiation. Among the flavonoids (quercetin, genistein and baicalin), quercetin induced the highest alkaline phosphatase (ALP) activity of human dental pulp (HDP) cells. Phenamil, an amiloride derivative, elicited higher ALP activity than quercetin. However, increased expression of dentin sialophosphoprotein (DSPP) mRNA and mineral deposition were seen in cultures treated with quercetin compared with phenamil. This would seem to suggest that quercetin is the most dentinogenic agent among the tested chemicals. The increase in ALP activity in the quercetin-treated cells was not affected by ICI 182,780, an estrogen receptor inhibitor, and was partially blocked by PD98059, an extracellular signal-regulated kinase 1/2 (ERK1/2) inhibitor. This suggests that ERK1/2 is activated in the quercetin-induced differentiation of HDP cells without the mediation of estrogen receptors, which are known to be involved in osteoblast differentiation induced by quercetin.


Assuntos
Amilorida/análogos & derivados , Antioxidantes/farmacologia , Diferenciação Celular/efeitos dos fármacos , Polpa Dentária/efeitos dos fármacos , Dentinogênese/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Quercetina/farmacologia , Fosfatase Alcalina/sangue , Amilorida/farmacologia , Células Cultivadas , Polpa Dentária/citologia , Polpa Dentária/metabolismo , Proteínas da Matriz Extracelular/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Expressão Gênica/efeitos dos fármacos , Humanos , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Osteocalcina/metabolismo , Fosfoproteínas/metabolismo , RNA Mensageiro , Reação em Cadeia da Polimerase em Tempo Real , Sialoglicoproteínas/metabolismo
5.
Iran Biomed J ; 26(6): 463-74, 2022 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-36437797

RESUMO

Background: Background: Bone tissue engineering has shown to be a promising strategy for repairing bone defects without causing harmful side effects to the patient. Three main building blocks of tissue engineering, including seeding cells, scaffold, and signaling molecules, are required for adequate bone regeneration. The human amniotic membrane (hAM) is the innermost of the placental membranes. In addition to providing a source of stem cells and growth factors, hAM has several features that make it an appropriate scaffold containing stem cells for use in tissue engineering purposes. The present investigation aimed to assess the effect of bone morphogenetic protein-9 (BMP-9) combined with phenamil and simvastatin on osteogenic induction of hAM with its human amniotic membrane epithelial cells (hAECs). Method: Methods: Using six different osteogenic medium (OMs), we cultured hAM for 14 days. The basic OMs were chosen as the first group and other media were made by adding BMP-9, phenamil, simvastatin, BMP-9 alongside phenamil, and BMP-9 alongside simvastatin to the basic OMs. Finally, viability assay, tissue mineralization, calcium and phosphate content determination, and measurement of lactic acid dehydrogenase (LDH), and alkaline phosphatase (ALP) activity were performed. Results: Results: Among all study groups, groups containing simvastatin showed a significantly lower level of viability. Although all media could induce osteogenic features, the hAECs cultured in media containing BMP-9 and phenamil demonstrated a wider area of mineralization and a significantly higher level of calcium and phosphate content, LDH, and ALP activity. Conclusion: Conclusion: Our findings indicated that the use of phenamil together with BMP-9 could synergistically show in situ osteogenic induction in hAECs, which could be a new insight into translational medicine.


Assuntos
Fator 2 de Diferenciação de Crescimento , Osteogênese , Feminino , Gravidez , Humanos , Fator 2 de Diferenciação de Crescimento/farmacologia , Sinvastatina/farmacologia , Placenta , Diferenciação Celular , Células-Tronco , Células Cultivadas
6.
Biomater Adv ; 139: 213032, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35882123

RESUMO

For effective treatment of infected bone, it is essential to use local drug delivery systems with the ability to deliver both antibiotics and osteoinductive factors. Herein, a pH-sensitive silk fibroin (SF)/sodium alginate (SA) hydrogel scaffolds containing teicoplanin (TEC) and phenamil (PM) loaded SF nanoparticles (PMSFNPS) are introduced for treating chronic osteomyelitis. The TEC and PM showed a sustained- and pH-sensitive release behavior from SF/SA hydrogel. The higher release rate was seen in an alkaline pH in comparison to neutral and acidic pH during 10 days. The eluted TEC maintained its antibacterial activity of >75 % during 35 days and in three different pH values (5.5, 7.4, and 8.5). The cellular study indicated that the scaffolds containing PMSFNPs could promote the cell viability, ALP activity, and matrix mineralization. Moreover, the in vivo effectiveness of hydrogel scaffolds were analyzed with radiography, histological and Immunohistochemistry evaluations. The lower infection and higher regeneration were observed in methicillin-resistant Staphylococcus aureus (MRSA) infected rat bone treated with hydrogel scaffold containing PMSFNPs and TEC compared to other groups. Consequently, this dual-drug delivery system could be a hopeful approach for effective treatment of chronic bone infection.


Assuntos
Amilorida , Antibacterianos , Sistemas de Liberação de Medicamentos , Fibroínas , Staphylococcus aureus Resistente à Meticilina , Osteomielite , Teicoplanina , Alginatos/uso terapêutico , Amilorida/análogos & derivados , Animais , Antibacterianos/farmacologia , Fibroínas/uso terapêutico , Hidrogéis/uso terapêutico , Concentração de Íons de Hidrogênio , Osteomielite/tratamento farmacológico , Ratos , Teicoplanina/uso terapêutico
7.
J Tissue Eng Regen Med ; 14(3): 464-474, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31840422

RESUMO

Large bone defects represent a significant unmet medical challenge. Cost effectiveness and better stability make small molecule organic compounds a more promising alternative compared with biomacromolecules, for example, growth factors/hormones, in regenerative medicine. However, one common challenge for the application of these small compounds is their side-effect issue. Phenamil is emerging as an intriguing small molecule to promote bone repair by strongly activating bone morphogenetic protein signaling pathway. In addition to osteogenesis, phenamil also induces significant adipogenesis based on some in vitro studies, which is a concern that impedes it from potential clinical applications. Besides the soluble chemical signals, cellular differentiation is heavily dependent on the microenvironments provided by the 3D scaffolds. Therefore, we developed a 3D nanofibrous biomimetic scaffold-based strategy to harness the phenamil-induced stem cell lineage differentiation. Based on the gene expression, alkaline phosphatase activity, and mineralization data, we indicated that bone-matrix mimicking mineralized-gelatin nanofibrous scaffold effectively improved phenamil-induced osteoblastic differentiation, while mitigating the adipogenic differentiation in vitro. In addition to normal culture conditions, we also indicated that mineralized matrix can significantly improve phenamil-induced osteoblastic differentiation in simulated inflammatory condition. In viewing of the crucial role of mineralized matrix, we developed an innovative and facile mineral deposition-based strategy to sustain release of phenamil from 3D scaffolds for efficient local bone regeneration. Overall, our study demonstrated that biomaterials played a crucial role in modulating small molecule drug phenamil-induced osteoblastic differentiation by providing a bone-matrix mimicking mineralized gelatin nanofibrous scaffolds.


Assuntos
Adipogenia/efeitos dos fármacos , Amilorida/análogos & derivados , Diferenciação Celular/efeitos dos fármacos , Nanofibras/química , Osteoblastos/metabolismo , Alicerces Teciduais/química , Amilorida/química , Amilorida/farmacocinética , Amilorida/farmacologia , Animais , Linhagem Celular , Camundongos , Osteoblastos/citologia
8.
J Biomed Mater Res B Appl Biomater ; 108(6): 2699-2710, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32154997

RESUMO

Three-dimensional (3D) scaffolds with interconnected, hierarchically structured pores, and biomimetic nanostructures are desirable for tissue engineering, where preparation with a facile and biocompatible strategy remains challenging. In the present work, an innovative porous microspheres-aggregated 3D PCL scaffold with macropores, micropores, and nanofibrous-like structures was fabricated through a one-pot thermally induced phase separation (TIPS) method without the use of any porogen or specific instruments. Importantly, the porosity, pore size, and mechanical properties of our scaffolds were tailorable through tuning of the polymer concentration. Interestingly, the bioactivity of our 3D PCL scaffolds was significantly improved, as abundant apatite-like layers were formed on the 3D porous scaffolds, while no obvious apatite was observed on the 2D flat PCL film. Moreover, the high surface area attributed to the hierarchical macro/micro/nanostructure enabled our 3D porous scaffold to serve as a drug delivery depot for sustained release of both small molecule drug (phenamil) and protein (BMP2). In addition to sustained drug release, the hierarchical structure and high mechanical properties also contribute to significantly improving BMP2-induced osteogenic differentiation. In summary, we developed a novel PCL porous scaffold through a facile, one-pot TIPS method and demonstrated its promising potential application in large bone defect repair.


Assuntos
Regeneração Óssea , Microesferas , Engenharia Tecidual/métodos , Alicerces Teciduais , Amilorida/administração & dosagem , Amilorida/análogos & derivados , Amilorida/química , Apatitas/química , Materiais Biocompatíveis , Proteína Morfogenética Óssea 2 , Diferenciação Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Liberação Controlada de Fármacos , Expressão Gênica/efeitos dos fármacos , Humanos , Nanoestruturas , Poliésteres , Porosidade , Proteínas Recombinantes , Fator de Crescimento Transformador beta
9.
Int J Nanomedicine ; 14: 8221-8234, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31632029

RESUMO

BACKGROUND: Phenamil (PH) is a small molecule that induces bone formation through upregulation of the TRB3 gene in the bone-regeneration process. ß-Cyclodextrins (ßCDs) with hydrophilic surfaces and a relatively hydrophobic cavity can form inclusion complexes with primarily hydrophobic small molecules such as PH, and increase their apparent solubility and dissolution rate. The hydrophilic surface of ßCDs prevents their interaction with the hydrophobic lipids of the cell membrane for penetration. Therefore, binding of penetrative groups, such as lysine, arginine, and histidine (His), to ßCDs for cell penetration is required. AIM: The aim of this study was to investigate the effect of His-conjugated ßCD on cellular uptake of PH for bone differentiation. METHODS: In this study, His-ßCDs were synthesized and used to prepare an inclusion complex of His-ßCD-PH. A hydroxypropyl-ßCD-PH (HP-ßCD-PH) inclusion complex for increasing PH solubility without a penetrative group was prepared for comparison. 3-D geometry of ßCD derivatives and PH-inclusion complexes was investigated by Fourier-transform infrared spectroscopy and molecular docking. Alizarin red staining and real-time PCR were performed to compare bone differentiation of His-ßCD-PH and HP-ßCD-PH. RESULTS: The results suggested that the benzene ring of PH was inserted into the wide side of both His-ßCD and HP-ßCD. Alizarin red staining at 14 days postculture in the presence of His-ßCD-PH at total concentration of 50 µM for PH showed that bone-matrix mineralization increased significantly compared with free PH and HP-ßCD-PH. Real-time PCR confirmed this result, and showed gene expression increased significantly (OPN 1.84-fold, OCN 1.69-fold) when stem cells were cultured with His-ßCD-PH. CONCLUSION: The overall results indicated that His-ßCD-PH is a promising carrier for osteoinductive PH with possible penetration ability and sustained release that reduces BMP2 consumption for differentiation of mesenchymal stem cells to bone tissue.


Assuntos
2-Hidroxipropil-beta-Ciclodextrina/química , Amilorida/análogos & derivados , Endocitose , Histidina/química , Osseointegração , Células-Tronco/metabolismo , 2-Hidroxipropil-beta-Ciclodextrina/síntese química , Tecido Adiposo/citologia , Amilorida/farmacologia , Calcificação Fisiológica/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Humanos , Simulação de Acoplamento Molecular , Osseointegração/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Tamanho da Partícula , Espectroscopia de Prótons por Ressonância Magnética , Solubilidade , Espectroscopia de Infravermelho com Transformada de Fourier , Eletricidade Estática
10.
Appl Mater Today ; 10: 194-202, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29577064

RESUMO

Bone morphogenic protein 2 (BMP2) is a key growth factor for bone regeneration, possessing FDA approval for orthopedic applications. BMP2 is often required in supratherapeutic doses clinically, yielding adverse side effects and substantial treatment costs. Considering the crucial role of materials for BMPs delivery and cell osteogenic differentiation, we devote to engineering an innovative bone-matrix mimicking niche to improve low dose of BMP2-induced bone formation. Our previous work describes a novel technique, named thermally induced nanofiber self-agglomeration (TISA), for generating 3D electrospun nanofibrous (NF) polycaprolactone (PCL) scaffolds. TISA process could readily blend PCL with PLA, leading to increased osteogenic capabilities in vitro, however, these bio-inert synthetic polymers produced limited BMP2-induced bone formation in vivo. We therefore hypothesize that functionalization of NF 3D PCL scaffolds with bone-like hydroxyapatite (HA) and BMP2 signaling activator phenamil will provide a favorable osteogenic niche for bone formation at low doses of BMP2. Compared to PCL-3D scaffolds, PCL/HA-3D scaffolds demonstrated synergistically enhanced osteogenic differentiation capabilities of C2C12 cells with phenamil. Importantly, in vivo studies showed this synergism was able to generate significantly increased new bone in an ectopic mouse model, suggesting PCL/HA-3D scaffolds act as a favorable synthetic extracellular matrix for bone regeneration.

11.
Tissue Eng Part A ; 23(5-6): 195-207, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27771997

RESUMO

Growth factor-based therapeutics using bone morphogenetic protein 2 (BMP-2) presents a promising strategy to reconstruct craniofacial bone defects such as mandible. However, clinical applications require supraphysiological BMP doses that often increase inappropriate adipogenesis, resulting in well-documented, cyst-like bone formation. Here we reported a novel complementary strategy to enhance osteogenesis and mandibular bone repair by using small-molecule phenamil that has been shown to be a strong activator of BMP signaling. Phenamil synergistically induced osteogenic differentiation of human bone marrow mesenchymal stem cells with BMP-2 while suppressing their adipogenic differentiation induced by BMP-2 in vitro. The observed pro-osteogenic and antiadipogenic activity of phenamil was mediated by expression of tribbles homolog 3 (Trb3) that enhanced BMP-smad signaling and inhibited expression of peroxisome proliferator-activated receptor gamma (PPARγ), a master regulator of adipogenesis. The synergistic effect of BMP-2+phenamil on bone regeneration was further confirmed in a critical-sized rat mandibular bone defect by implanting polymer scaffolds designed to slowly release the therapeutic molecules. These findings indicate a new complementary osteoinductive strategy to improve clinical efficacy and safety of current BMP-based therapeutics.


Assuntos
Amilorida/análogos & derivados , Proteína Morfogenética Óssea 2 , Mandíbula/metabolismo , Traumatismos Mandibulares/tratamento farmacológico , Amilorida/farmacocinética , Amilorida/farmacologia , Animais , Proteína Morfogenética Óssea 2/farmacocinética , Proteína Morfogenética Óssea 2/farmacologia , Implantes de Medicamento/farmacocinética , Implantes de Medicamento/farmacologia , Humanos , Mandíbula/patologia , Traumatismos Mandibulares/metabolismo , Traumatismos Mandibulares/patologia , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/patologia , Ratos Sprague-Dawley
12.
Stem Cells Transl Med ; 5(4): 539-51, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26956209

RESUMO

UNLABELLED: Although adipose-derived stem cells (ASCs) are an attractive cell source for bone tissue engineering, direct use of ASCs alone has had limited success in the treatment of large bone defects. Although bone morphogenetic proteins (BMPs) are believed to be the most potent osteoinductive factors to promote osteogenic differentiation of ASCs, their clinical applications require supraphysiological dosage, leading to high medical burden and adverse side effects. In the present study, we demonstrated an alternative approach that can effectively complement the BMP activity to maximize the osteogenesis of ASCs without exogenous application of BMPs by regulating levels of antagonists and agonists to BMP signaling. Treatment of ASCs with the amiloride derivative phenamil, a positive regulator of BMP signaling, combined with gene manipulation to suppress the BMP antagonist noggin, significantly enhanced osteogenic differentiation of ASCs through increased BMP-Smad signaling in vitro. Furthermore, the combination approach of noggin suppression and phenamil stimulation enhanced the BMP signaling and bone repair in a mouse calvarial defect model by adding noggin knockdown ASCs to apatite-coated poly(lactic-coglycolic acid) scaffolds loaded with phenamil. These results suggest novel complementary osteoinductive strategies that could maximize activity of the BMP pathway in ASC bone repair while reducing potential adverse effects of current BMP-based therapeutics. SIGNIFICANCE: Although stem cell-based tissue engineering strategy offers a promising alternative to repair damaged bone, direct use of stem cells alone is not adequate for challenging healing environments such as in large bone defects. This study demonstrates a novel strategy to maximize bone formation pathways in osteogenic differentiation of mesenchymal stem cells and functional bone formation by combining gene manipulation with a small molecule activator toward osteogenesis. The findings indicate promising stem cell-based therapy for treating bone defects that can effectively complement or replace current osteoinductive therapeutics.


Assuntos
Tecido Adiposo/citologia , Células-Tronco Adultas/fisiologia , Proteínas Morfogenéticas Ósseas/agonistas , Proteínas Morfogenéticas Ósseas/antagonistas & inibidores , Osteogênese/fisiologia , Células-Tronco Adultas/efeitos dos fármacos , Amilorida/análogos & derivados , Amilorida/farmacologia , Animais , Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas de Transporte/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Osteogênese/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Proteínas Smad/fisiologia
13.
J Tissue Eng Regen Med ; 8(9): 728-36, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22815259

RESUMO

Osteoblast cell adhesion and differentiation on biomaterials are important achievements necessary for implants to be useful in bone regenerative engineering. Recombinant bone morphogenetic proteins (BMPs) have been shown to be important for these processes; however, there are many challenges associated with the widespread use of these proteins. A recent report demonstrated that the small molecule phenamil, a diuretic derivative, was able to induce osteoblast differentiation and mineralization in vitro via the canonical BMP signalling cascade (Park et al., 2009). In this study, the feasibility of using phenamil as a novel biofactor in conjunction with a biodegradable poly(lactide-co-glycolide acid) (PLAGA) polymeric scaffold for engineering bone tissue was evaluated. The in vitro cellular behaviour of osteoblast-like MC3T3-E1 cells cultured on PLAGA scaffolds in the presence of phenamil at 10 µM were characterized with regard to initial cell adhesion, proliferation, alkaline phosphatase (ALP) activity and matrix mineralization. The results demonstrate that phenamil supported cell proliferation, promoted ALP activity and facilitated matrix mineralization of osteoblast-like MC3T3-E1 cells. Moreover, in this study, we found that phenamil promoted integrin-mediated cell adhesion on PLAGA scaffolds. It was also shown that phenamil encapsulated within porous, microsphere PLAGA scaffolds retained its osteogenic activity upon release. Based on these findings, the small molecule phenamil has the potential to serve as a novel biofactor for the repair and regeneration of bone tissues.


Assuntos
Amilorida/análogos & derivados , Materiais Biocompatíveis/farmacologia , Regeneração Óssea/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Engenharia Tecidual/métodos , Fosfatase Alcalina/metabolismo , Amilorida/farmacologia , Animais , Matriz Óssea/efeitos dos fármacos , Matriz Óssea/fisiologia , Calcificação Fisiológica/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Estudos de Viabilidade , Ácido Láctico/farmacologia , Camundongos , Microesferas , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Osteoblastos/enzimologia , Ácido Poliglicólico/farmacologia , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Alicerces Teciduais/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA