Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Actual. osteol ; 19(1): 18-29, ago. 2023. tab
Article in English | LILACS, UNISALUD, BINACIS | ID: biblio-1511400

ABSTRACT

MicroRNAs (miRNAs) are small non-coding RNA molecules that play critical roles in post-transcriptional gene regulation. They function by binding to target messenger RNA (mRNA) molecules, leading to their degradation or inhibiting their translation into proteins. In the context of skeletal diseases, such as osteoporosis, osteoarthritis, and bone metastasis, there is growing evidence osteoblastic miRNAs, are involved in the regulation of bone formation and maintenance.Osteoblasts are bone-forming cells responsible for synthesizing and depositing the extracellular matrix, which ultimately mineralizes to form bone tissue. Osteoblastic miRNAs modulate various aspects of osteoblast function, including proliferation, differentiation, mineralization, and apoptosis. Dysregulation of these miRNAs can disrupt the balance between bone formation and resorption, leading to skeletal diseases.The therapeutic implications of targeting osteoblastic miRNAs in skeletal diseases are significant. Modulating the expression levels of specific miRNAs holds promise for developing novel therapeutic strategies to enhance bone formation, prevent bone loss, and promote bone regeneration. Potential therapeutic approaches include the use of synthetic miRNA mimics to restore miRNA expression in diseases associated with miRNA downregulation or the use of anti-miRNA oligonucleotides to inhibit miRNA function in diseases associated with miRNA upregulation.miRNA-based therapies are still in the early stages of development, and further research is needed to fully understand the complexity of miRNA networks. Additionally, the delivery of miRNAs to specific target tissues and cells remains a challenge that needs to be addressed for effective clinical translation. Nonetheless, targeting osteoblastic miRNAs represents a promising avenue for future therapeutic interventions in skeletal diseases. (AU)


Los micro-ARNs (miARNss) son pequeños ARN no codificantes que desempeñan un papel fundamental en la regulación génica postranscripcional. Ejercen su función al unir-se a moléculas de ARN mensajero (ARNm), promoviendo su degradación e inhibiendo su traducción en proteínas. En el contexto de las enfermedades esqueléticas, como la osteoporosis, la osteoartritis y la metástasis ósea existe evidencia de que los miARNs osteoblásticos están involucrados en la regulación de la formación y del mantenimiento óseo. Los osteoblastos son células formadoras de hueso responsables de sintetizar y depositar la matriz extracelular, que finalmente se mineraliza para formar el hueso. Los miARNs derivados de osteoblastos modulan varios aspectos de la función de estas células, incluida la proliferación, diferenciación, mineralización y la apoptosis. La desregulación de estos miARNs puede alterar el equilibrio entre la formación y la resorción ósea, lo que lleva a enfermedades óseas. Las implicaciones terapéuticas de los miARNs osteoblásticos en enfermedades esqueléticas son significativas. La modulación de los niveles de expresión de miARNs específicos es prometedora para desarrollar nuevas estrate-gias terapéuticas a fin de mejorar la formación, prevenir la pérdida y promover la regeneración ósea. Los enfoques terapéuticos potenciales incluyen el uso de miméticos de miARNs para restaurar la expresión de miARNs o el uso de oligonucleótidos anti-miARNs para inhibir su función. Las terapias basadas en miARNs aún se encuentran en las primeras etapas de desarrollo. La administración de miARNs a las células y los tejidos específicos sigue siendo un desafío para lograr una aplicación clínica eficaz. (AU)


Subject(s)
Humans , Osteoblasts/cytology , Osteogenesis/genetics , MicroRNAs/genetics , Osteoclasts/cytology , Bone Diseases/prevention & control , Signal Transduction , Gene Expression Regulation , MicroRNAs/biosynthesis , MicroRNAs/physiology , MicroRNAs/therapeutic use
2.
J. appl. oral sci ; 28: e20200242, 2020. tab, graf
Article in English | LILACS, BBO | ID: biblio-1134786

ABSTRACT

Abstract Heterogeneous cell populations of osteo/cementoblastic (O/C) or fibroblastic phenotypes constitute the periodontal dental ligament (PDL). A better understanding of these PDL cell subpopulations is essential to propose regenerative approaches based on a sound biological rationale. Objective Our study aimed to clarify the differential transcriptome profile of PDL cells poised to differentiate into the O/C cell lineage. Methodology To characterize periodontal-derived cells with distinct differentiation capacities, single-cell-derived clones were isolated from adult human PDL progenitor cells and their potential to differentiate into osteo/cementoblastic (O/C) phenotype (C-O clones) or fibroblastic phenotype (C-F clones) was assessed in vitro. The transcriptome profile of the clonal cell lines in standard medium cultivation was evaluated using next-generation sequencing technology (RNA-seq). Over 230 differentially expressed genes (DEG) were identified, in which C-O clones showed a higher number of upregulated genes (193) and 42 downregulated genes. Results The upregulated genes were associated with the Cadherin and Wnt signaling pathways as well as annotated biological processes, including "anatomical structure development" and "cell adhesion." Both transcriptome and RT-qPCR showed up-regulation of WNT2, WNT16, and WIF1 in C-O clones. Conclusions This comprehensive transcriptomic assessment of human PDL progenitor cells revealed that expression of transcripts related to the biological process "anatomical structure development," Cadherin signaling, and Wnt signaling can identify PDL cells with a higher potential to commit to the O/C phenotype. A better understanding of these pathways and their function in O/C differentiation will help to improve protocols for periodontal regenerative therapies.


Subject(s)
Humans , Adult , Osteoblasts/cytology , Periodontal Ligament/surgery , Dental Cementum/cytology , Cadherins/metabolism , Cell Differentiation , Cells, Cultured , Clone Cells , Transcriptome
3.
Braz. dent. j ; 29(2): 117-127, Mar.-Apr. 2018. tab, graf
Article in English | LILACS | ID: biblio-951529

ABSTRACT

Article This study evaluated comparatively two configurations (powder and putty) of a composite biomaterial based on PLGA (Poly(lactide-co-glycolide)/nanoescale hydroxyapatite (ReOss®, Intra-Lock International) through microscopic morphology, in vitro cytotoxicity, biocompatibility and in vivo response as a bone substitute. SEM and EDS characterized the biomaterials before/after grafting. Cytocompatibility was assessed with murine pre-osteoblasts. Osteoconductivity and biocompatibility were evaluated in White New Zealand rabbits. Both configurations were implanted in the calvaria of eighteen animals in non-critical size defects, with blood clot as the control group. After 30, 60 and 90 days, the animals were euthanized and the fragments containing the biomaterials and controls were harvested. Bone blocks were embedded in paraffin (n=15) aiming at histological and histomorphometric analysis, and in resin (n=3) aiming at SEM and EDS. Before implantation, the putty configuration showed both a porous and a fibrous morphological phase. Powder revealed porous particles with variable granulometry. EDS showed calcium, carbon, and oxygen in putty configuration, while powder also showed phosphorus. After implantation EDS revealed calcium, carbon, and oxygen in both configurations. The materials were considered cytotoxic by the XTT test. Histological analysis showed new bone formation and no inflammatory reaction at implant sites. However, the histomorphometric analysis indicated that the amount of newly formed bone was not statistically different between experimental groups. Although both materials presented in vitro cytotoxicity, they were biocompatible and osteoconductive. The configuration of ReOss® affected morphological characteristics and the in vitro cytocompatibility but did not impact on the in vivo biological response, as measured by the present model.


Resumo Este estudo avaliou comparativamente duas configurações (pó e massa) de um biomaterial composto com base de PLGA (Poli(láctico-co-glicólico)/hidroxiapatita em nanoescala (ReOss®, Intra-Lock International) através da morfologia microscópica, citotoxicidade in vitro, biocompatibilidade e resposta in vivo como substituto ósseo. MEV e EDS caracterizaram os biomateriais antes/após o enxerto. A citocompatibilidade foi avaliada em pré-osteoblastos murinos. A osteocondutividade e a biocompatibilidade foram avaliadas em coelhos Branco da Nova Zelândia. Ambas as configurações foram implantadas na calvária de dezoito animais em defeitos não-críticos, com coágulo sanguíneo como grupo controle. Após 30, 60 e 90 dias, os animais foram eutanasiados e os fragmentos contendo os biomateriais e controles coletados. Blocos ósseos foram embebidos em parafina (n=15) destinados às análises histológica e histomorfométrica, e em resina (n=3) destinadas à MEV e EDS. Antes da implantação, a configuração massa mostrou ambas fases morfológicas porosa e fibrosa. O pó revelou partículas porosas com granulometria variável. EDS mostrou cálcio, carbono e oxigênio na configuração massa, enquanto o pó mostrou também fósforo. Após a implantação a EDS revelou cálcio, carbono e oxigênio em ambas configurações. Os materiais foram considerados citotóxicos pelo teste XTT. A análise histológica mostrou nova formação óssea e nenhuma reação inflamatória nos sítios de implante. Entretanto, a análise histomorfométrica indicou que a quantidade de osso neoformado não foi estatisticamente diferente entre os grupos experimentais. Embora ambos os materiais tenham apresentado citotoxicidade in vitro, foram biocompatíveis e osteocondutores. A configuração do ReOss® afetou as características morfológicas e a citocompatibilidade in vitro, porém não impactou a resposta biológica in vivo, como medido pelo presente modelo.


Subject(s)
Animals , Male , Female , Rabbits , Bone Substitutes , Osteoblasts/cytology , Powders , Spectrometry, X-Ray Emission , Bone Regeneration , Materials Testing , Microscopy, Electron, Scanning , Durapatite/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry
4.
Braz. j. med. biol. res ; 51(9): e6948, 2018. tab, graf
Article in English | LILACS | ID: biblio-951763

ABSTRACT

It is increasingly evident that the microenvironment of bone can influence cancer phenotype in many ways that favor growth in bone. CD147, a transmembrane protein of the immunoglobulin (Ig) superfamily, was identified independently in different species and has many designations across different species. However, expression levels of CD147 mRNA in bone cancer have not been described. In this study, we have used real-time fluorescence quantification (RT-PCR) to demonstrate CD147 expression in malignant bone cancer and benign bone tumor tissues. The results suggested that the expression of CD147 gene was significantly up-regulated in malignant bone cancer. Moreover, we found that over-expressed RANKL progressively enhanced osteoclast formation up to 48 h, which suggested that RANKL could promote the formation of osteoclast, indicating that both CD147 and RANKL play important roles in the formation of osteoclasts. Furthermore, the expressions of four osteoclast specific expression genes, including TRACP, MMP-2, MMP-9 and c-Src, were analyzed using RT-PCR. The results indicated that four osteoclast-specific expression genes were detectable in all osteoclast with different treatments. However, the highest expression level of these four osteoclast-specific expression genes appears in the CD147+ RANKL group and the lowest expression level of these four osteoclast-specific expression genes appears with si-RANKL treatment. Characterization of the role of CD147 in the development of tumors should lead to a better understanding of the changes occurring at the molecular level during the development and progression of primary human bone cancer.


Subject(s)
Humans , Male , Female , Middle Aged , Aged , Osteoclasts/metabolism , Bone Neoplasms/genetics , Up-Regulation , Basigin/genetics , RANK Ligand/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Bone Neoplasms/secondary , Bone Neoplasms/therapy , Gene Expression Regulation, Neoplastic , Blotting, Western , Reverse Transcriptase Polymerase Chain Reaction
5.
Actual. osteol ; 13(3): 207-213, Sept - DIc. 2017. ilus, graf
Article in English | LILACS | ID: biblio-1117111

ABSTRACT

Osteocytes are the most abundant bone cell and are formed when osteoblasts become embedded in the bone matrix. Through changes in gene expression and paracrine effects, osteocytes regulate the number of osteoblasts, bone forming cells, and osteoclasts, bone resorbing cells, which are needed to maintain bone mass. MLO-Y4 is the better characterized osteocytic cell line; however, lacks expression of sclerostin, the product of the SOST gene, which is fundamental for osteocyte function and blocks bone formation. With the objective to isolate MLO-Y4 clones with different gene expression profiles, we performed cultures at very low density of MLO-Y4 cells stably transfected with nuclear green fluorescent protein (MLOnGFP). Cell morphology was visualized under a fluorescence microscope. Once the cells reached 80% confluency, RNA was extracted and quantitative real time PCR was performed. Clones exhibit different sizes and morphology, with some cells showing a spindle-like shape and others with abundant projections and a star-like shape. Gene expression also differed among clones. However, none of the clones examined expressed SOST. We conclude that the MLO-nGFP clones constitute a useful tool to study osteocyte differentiation and the role of osteocytes in the control of bone formation and resorption in vitro. (AU)


Los osteocitos son las células más abundantes del hueso y se forman cuando los osteoblastos se encuentran rodeados de matriz ósea. A través de cambios en la expresión génica y efectos paracrinos, los osteocitos controlan el número de osteoblastos que forman el hueso, y osteoclastos que resorben el hueso, células necesarias para mantener la masa ósea. Las células MLO-Y4 son la línea celular osteocítica más investigada; sin embargo, no expresan esclerostina, el pro esclerostina, el producto del gen SOST que bloquea la formación ósea y es indispensable para la función de los osteocitos. Con el objetivo de aislar clones de las células MLO-Y4 con diferentes perfiles de expresión génica, realizamos cultivos a muy baja densidad de las células transfectadas en forma estable con proteína verde fluorescente nuclear (MLO-nGFP). La morfología celular fue evaluada utilizando un microscopio de fluorescencia. Una vez que las células alcanzaron el 80% de confluencia, el ARN fue extraído y analizado por PCR cuantitativa en tiempo real. Las células de los diferentes clones tienen diferentes tamaños y morfología, algunas células son fusiformes y otras con proyecciones citoplasmáticas abundantes y en forma de estrella. La expresión de los genes también varió en los distintos clones. Sin embargo, ninguno de ellos expresó SOST. En conclusión, los clones de las células MLO-nGFP constituyen una herramienta útil para estudiar la diferenciación de los osteocitos y el rol de estas células en el control de la formación y resorción ósea in vitro. (AU)


Subject(s)
Humans , Male , Female , Osteoblasts/cytology , Osteoclasts/cytology , Osteocytes/cytology , Cell Line , Clone Cells/cytology , Osteoblasts/metabolism , Osteoclasts/metabolism , Osteocytes/metabolism , Osteogenesis/genetics , Bone Resorption/genetics , In Vitro Techniques , RNA/analysis , Gene Expression , Polymerase Chain Reaction , Collagen/genetics , Alkaline Phosphatase/metabolism , Fluorescence , Anti-Bacterial Agents/administration & dosage
6.
Braz. dent. j ; 28(3): 307-316, May-June 2017. tab, graf
Article in English | LILACS | ID: biblio-888646

ABSTRACT

Abstract This study aimed to investigate the influence of a three-dimensional cell culture model and bioactive glass (BG) particles on the expression of osteoblastic phenotypes in rat calvaria osteogenic cells culture. Cells were seeded on two-dimensional (2D) and three-dimensional (3D) collagen with BG particles for up to 14 days. Cell viability and alkaline phosphatase (ALP) activity was performed. Cell morphology and immunolabeling of noncollagenous bone matrix proteins were assessed by epifluorescence and confocal microscopy. The expressions of osteogenic markers were analyzed using RT-PCR. Mineralized bone-like nodule formation was visualized by microscopy and calcium content was assessed quantitatively by alizarin red assay. Experimental cultures produced a growing cell viability rate up to 14 days. Although ALP activity at 7 days was higher on BG cultures, cells on 3D and 3D+BG had an activity decrease of ALP at 14 days. Three-dimensional conditions favored the immunolabeling for OPN and BSP and the expression of ALP and COL I mRNAs. BG particles influenced positively the OC and OPN mRNAs expression and calcified nodule formation in vitro. The results indicated that the 3D cultures and BG particles contribute to the expression of osteoblastic phenotype and to differentiated and mineralized matrix formation.


Resumo O objetivo deste estudo foi investigar a influência do modelo de cultura celular tridimensional e das partículas de vidro bioativo (BG) sobre a expressão fenotípica de culturas de células osteogênicas da calvária de ratos. As células foram mantidas em culturas sobre superfícies colágenas bi-dimensionais (2D) e em géis de colágeno tridimensional (3D) com e sem partículas de BG até 14 dias. Foram avaliadas: viabilidade celular, atividade de fosfatase alcalina (ALP), morfologia celular e imunomarcação de proteínas da matriz não-colágena do osso através de epifluorescência e microscopia confocal. As expressões de marcadores osteogênicos foram analisadas utilizando RT-PCR. A formação de nódulos mineralizados foi visualizada através de microscopia e o conteúdo de cálcio foi avaliado quantitativamente pelo Alizarina Red. As culturas experimentais produziram uma taxa crescente de viabilidade até 14 dias. Embora a atividade ALP em 7 dias tenha sido maior em culturas com BG, as células em 3D e 3D+BG apresentaram uma diminuição da atividade ALP aos 14 dias. As condições tridimensionais favoreceram a imunomarcação para OPN e BSP e a expressão de mRNAs para ALP e COL I. As partículas de BG influenciaram positivamente a expressão do mRNAs para OPN e OC e a formação de nódulos calcificados in vitro. Os resultados indicaram que as culturas em 3D e partículas BG contribuíram para a expressão do fenótipo osteoblástico e para a diferenciação e formação de matriz mineralizada.


Subject(s)
Animals , Biocompatible Materials , Glass , Osteoblasts/cytology , Osteogenesis , Skull/cytology , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Biomarkers/metabolism , Calcium/metabolism , Cell Culture Techniques , Cell Survival , Collagen Type I/genetics , Collagen Type I/metabolism , Fluorescent Antibody Technique, Indirect , Gene Expression Profiling , Integrin-Binding Sialoprotein/metabolism , Microscopy, Confocal , Microscopy, Fluorescence , Osteoblasts/enzymology , Osteoblasts/metabolism , Osteopontin/metabolism , Rats, Wistar , Real-Time Polymerase Chain Reaction , RNA, Messenger/genetics , Skull/enzymology , Skull/metabolism , Tissue Scaffolds
7.
Biol. Res ; 50: 28, 2017. tab, graf
Article in English | LILACS | ID: biblio-950879

ABSTRACT

BACKGROUND: The Tridax procumbens extracts (TPE) are known for their ethno-medicinal properties to increase osteogenic functioning in mesenchymal stem cells. Recently, we found that the T. procumbens flavonoids (TPF) significantly suppressed the RANKL-induced osteoclasts differentiation and bone resorption. The TPF also promoted osteoblasts differentiation and bone formation demonstrated by increasing bone formation markers in cultured mouse primary osteoblasts. However, the effects of the TPF on in vivo bone formation remain unclear. In this study, we investigated the effects of the TPF on in vivo bone formation, injected the TPF (20 mg/kg) twice a day in the low calcium diet mice and killed them after 21 day. Radiographic and histomorphometric analyses were performed on the dissected bones to determine the anabolic effects of the TPF. RESULTS: Bone mineral density and bone mineral content of the TPF-treated mice were significantly increased compared to the control mice. Bone formation-related indices like osteoblast number, osteoblast surface, bone volume, mineralizing surface, mineral apposition rate and bone formation rate were significantly increased in the TPF-treated mice compared to the control mice. CONCLUSION: Our findings point towards the stimulation of bone formation by TPF, suggested that the TPF could be a potential natural anabolic agent to treat patients with bone loss-associated diseases such as osteoporosis.


Subject(s)
Animals , Male , Mice , Rats , Osteogenesis/drug effects , Flavonoids/pharmacology , Bone Resorption/drug therapy , Plant Extracts/pharmacology , Bone Density/drug effects , Cell Differentiation/drug effects , Asteraceae/chemistry , Osteoblasts/cytology , Osteoblasts/drug effects , Flavonoids/isolation & purification , Bone Resorption/pathology , Mice, Inbred C57BL
8.
Hist. ciênc. saúde-Manguinhos ; 22(3): 941-960, jul.-set. 2015. tab, ilus
Article in Spanish | LILACS | ID: lil-756452

ABSTRACT

La poliomielitis en forma epidémica despertó diversos desafíos, uno de los cuales fue la rehabilitación de las secuelas que había dejado esta dolencia en muchas de las personas que la padecieron. La parálisis y las formas en cómo se fue transformando, el concepto de rehabilitación física (donde el objetivo era solo restituir la movilidad de los músculos afectados), la rehabilitación integral que incluía los planos sociales, educativos y profesionales, en la Argentina de mediados del siglo XX, son los temas tratados en este artículo. Usa la metodología de historia institucional que dialoga de manera permanente con la historia de la salud y de la enfermedad.


Poliomyelitis on an epidemic scale gave rise to several challenges, one of which was the rehabilitation from the after-effects on many of the people who suffered from the disease. Paralysis and the ways it transformed the concept of physical rehabilitation (where the objective was only to restore the mobility of the affected muscles) and comprehensive rehabilitation that included social, educational and professional aspects in Argentina in the mid-twentieth century are the themes addressed in this article. It uses the methodology of institutional history that interacts in an ongoing manner with the history of health and disease.


Subject(s)
Humans , Biocompatible Materials/chemistry , Osteoblasts/cytology , Surface Properties , Tissue Scaffolds , Titanium/chemistry , Cell Differentiation , Cell Line, Tumor , Cell Survival , Tissue Engineering , Titanium/pharmacology
9.
Braz. j. med. biol. res ; 48(4): 299-307, 4/2015. graf
Article in English | LILACS | ID: lil-744357

ABSTRACT

Bone homeostasis seems to be controlled by delicate and subtle “cross talk” between the nervous system and “osteo-neuromediators” that control bone remodeling. The purpose of this study was to evaluate the effect of interactions between neuropeptides and human bone morphogenetic protein 2 (hBMP2) on human osteoblasts. We also investigated the effects of neuropeptides and hBMP2 on gap junction intercellular communication (GJIC). Osteoblasts were treated with neuropeptide Y (NPY), substance P (SP), or hBMP2 at three concentrations. At various intervals after treatment, cell viability was measured by the MTT assay. In addition, cellular alkaline phosphatase (ALP) activity and osteocalcin were determined by colorimetric assay and radioimmunoassay, respectively. The effects of NPY, SP and hBMP on GJIC were determined by laser scanning confocal microscopy. The viability of cells treated with neuropeptides and hBMP2 increased significantly in a time-dependent manner, but was inversely associated with the concentration of the treatments. ALP activity and osteocalcin were both reduced in osteoblasts exposed to the combination of neuropeptides and hBMP2. The GJIC of osteoblasts was significantly increased by the neuropeptides and hBMP2. These results suggest that osteoblast activity is increased by neuropeptides and hBMP2 through increased GJIC. Identification of the GJIC-mediated signal transduction capable of modulating the cellular activities of bone cells represents a novel approach to studying the biology of skeletal innervation.


Subject(s)
Humans , /pharmacology , Cell Communication/drug effects , Gap Junctions/drug effects , Neuropeptide Y/pharmacology , Osteoblasts/drug effects , Substance P/pharmacology , /administration & dosage , Cell Survival/drug effects , Cells, Cultured/drug effects , Enzyme-Linked Immunosorbent Assay , Neuropeptide Y/administration & dosage , Osteoblasts/cytology , Osteocalcin/analysis , Osteogenesis/drug effects , Substance P/administration & dosage
10.
Recife; s.n; 2015. 94 p. ilus, graf, tab.
Thesis in Portuguese | LILACS | ID: biblio-871418

ABSTRACT

O titânio e suas ligas são os materiais mais comumente utilizados na substituição de tecidos duros por possuírem resistência mecânica, biocompatibilidade, resistência à corrosão e fácil manipulação. Embora o titânio possua várias vantagens sobre outros biomateriais, seu uso em longo prazo pode ocasionar problemas de rejeição. A modificação da superfície do titânio a fim de criar microrrugosidades é uma estratégia efetiva para melhorar a adesão e proliferação celular sobre implantes. Quando um implante danifica ou invade as barreiras epitelial e das mucosas, pode servir como reservatório para microrganismos e desta forma predispor à infecção. Neste sentido, o objetivo deste trabalho foi modificar a superfície do titânio, utilizando nanopartículas de prata (Ag) e lectina, a fim de melhorar a sua biocompatibilidade e conferir propriedades antimicrobianas a este material. O racional por trás destas mudanças é que a criação de uma topografia em nanoescala pode contribuir para mimetizar o ambiente celular melhorando a osseointegração e diminuindo o risco de infecção. Em nosso estudo, nanotubos de dióxido de titânio (NTs-TiO2) com estrutura bem distribuída e organizada, com diâmetro em torno de 70-80nm, foram sintetizados por anodização eletroquímica e decorados com nanopartículas de Ag usando a técnica de layer-by-layer (LbL), enquanto a lectina do peixe Oreochromis niloticus (OniL) foi incorporada aos NTs-TiO2 por spin coating. Estas amostras foram caracterizadas e avaliadas quanto a sua citotoxidade, adesão celular, potencial osteogênico e atividade bactericida. Nossos resultados mostraram que tanto as nanopartículas de Ag, como a Onil foram incorporadas com sucesso à superfície dos NTs-TiO2. Entretanto nossas preparações de LbL não foram capazes de melhorar a biocompatibilidade ou inibir o crescimento de bactérias nos NTs-TiO2. Por outro lado, a funcionalização dos NTs-TiO2 com a OniL induziu eficientemente a adesão e proliferação dos osteoblastos. Nossos resultados apontam para o uso da lectina OniL para melhorar a qualidade dos implantes de NT-TiO2 existentes.


Subject(s)
Humans , Animals , Anti-Bacterial Agents/chemistry , Lectins , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/toxicity , Nanotubes , Metal Nanoparticles , Silver , Titanium , Anti-Bacterial Agents/toxicity , Cell Adhesion , Cells, Cultured , Candida albicans , Escherichia coli , Materials Testing , Coated Materials, Biocompatible/pharmacology , Osteoblasts/cytology , Osteoblasts/physiology , Staphylococcus aureus
11.
Biol. Res ; 48: 1-8, 2015. graf, tab
Article in English | LILACS | ID: biblio-950829

ABSTRACT

BACKGROUND: Tridaxprocumbens flavonoids (TPFs) are well known for their medicinal properties among local natives. Besides traditionally used for dropsy, anemia, arthritis, gout, asthma, ulcer, piles, and urinary problems, it is also used in treating gastric problems, body pain, and rheumatic pains of joints. TPFs have been reported to increase osteogenic functioning in mesenchymal stem cells. Our previous study showed that TPFs were significantly suppressed the RANKL-induced differentiation of osteoclasts and bone resorption. However, the effects of TPFs to promote osteoblasts differentiation and bone formation remain unclear. TPFs were isolated from Tridax procumbens and investigated for their effects on osteoblasts differentiation and bone formation by using primary mouse calvarial osteoblasts. RESULTS: TPFs promoted osteoblast differentiation in a dose-dependent manner demonstrated by up-regulation of alkaline phosphatase and osteocalcin. TPFs also upregulated osteoblast differentiation related genes, including osteocalcin, osterix, and Runx2 in primary osteoblasts. TPFs treated primary osteoblast cells showed significant upregulation of bone morphogenetic proteins (BMPs) including Bmp-2, Bmp-4, and Bmp-7. Addition of noggin, a BMP specific-antagonist, inhibited TPFs induced upregulation of the osteocalcin, osterix, and Runx2. CONCLUSION: Our findings point towards the induction of osteoblast differentiation by TPFs and suggested that TPFs could be a potential anabolic agent to treat patients with bone loss-associated diseases such as osteoporosis.


Subject(s)
Animals , Mice , Osteoblasts/drug effects , Osteogenesis/drug effects , Flavonoids/pharmacology , Cell Differentiation/drug effects , Asteraceae/chemistry , Osteoblasts/cytology , Osteoblasts/metabolism , Skull/cytology , Skull/drug effects , Transcription Factors/genetics , Flavonoids/analysis , Calcification, Physiologic/drug effects , Osteocalcin/drug effects , Osteocalcin/genetics , Up-Regulation/genetics , Bone Morphogenetic Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Alkaline Phosphatase/drug effects , Alkaline Phosphatase/metabolism , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Primary Cell Culture , Sp7 Transcription Factor , Medicine, Traditional , Mice, Inbred C57BL
12.
Experimental & Molecular Medicine ; : e128-2015.
Article in English | WPRIM | ID: wpr-220401

ABSTRACT

Fucoidan has attracted attention as a potential drug because of its biological activities, which include osteogenesis. However, the molecular mechanisms involved in the osteogenic activity of fucoidan in human alveolar bone marrow-derived mesenchymal stem cells (hABM-MSCs) remain largely unknown. We investigated the action of fucoidan on osteoblast differentiation in hABM-MSCs and its impact on signaling pathways. Its effect on proliferation was determined using the crystal violet staining assay. Osteoblast differentiation was evaluated based on alkaline phosphatase (ALP) activity and the mRNA expression of multiple osteoblast markers. Calcium accumulation was determined by Alizarin red S staining. We found that fucoidan induced hABM-MSC proliferation. It also significantly increased ALP activity, calcium accumulation and the expression of osteoblast-specific genes, such as ALP, runt-related transcription factor 2, type I collagen-alpha 1 and osteocalcin. Moreover, fucoidan induced the expression of bone morphogenetic protein 2 (BMP2) and stimulated the activation of extracellular signal-related kinase (ERK), c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase by increasing phosphorylation. However, the effect of fucoidan on osteogenic differentiation was inhibited by specific inhibitors of ERK (PD98059) and JNK (SP600125) but not p38 (SB203580). Fucoidan enhanced BMP2 expression and Smad 1/5/8, ERK and JNK phosphorylation. Moreover, the effect of fucoidan on osteoblast differentiation was diminished by BMP2 knockdown. These results indicate that fucoidan induces osteoblast differentiation through BMP2-Smad 1/5/8 signaling by activating ERK and JNK, elucidating the molecular basis of the osteogenic effects of fucoidan in hABM-MSCs.


Subject(s)
Humans , Bone Morphogenetic Protein 2/genetics , Calcium/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , JNK Mitogen-Activated Protein Kinases/metabolism , Mesenchymal Stem Cells/cytology , Osteoblasts/cytology , Osteogenesis/drug effects , Phosphorylation , Polysaccharides/pharmacology , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/genetics , Signal Transduction/drug effects , Smad Proteins/metabolism
13.
Experimental & Molecular Medicine ; : e152-2015.
Article in English | WPRIM | ID: wpr-57312

ABSTRACT

The Wnt/beta-catenin pathway has a role in osteoblast differentiation and bone formation. We screened 100 plant extracts and identified an extract from Euodia sutchuenensis Dode (ESD) leaf and young branch as an effective activator of the Wnt/beta-catenin pathway. ESD extract increased beta-catenin levels and beta-catenin nuclear accumulation in murine primary osteoblasts. The ESD extract also increased mRNA levels of osteoblast markers, including RUNX2, BMP2 and COL1A1, and enhanced alkaline phosphatase (ALP) activity in murine primary osteoblasts. Both ESD extract-induced beta-catenin increment and ALP activation were abolished by beta-catenin knockdown, confirming that the Wnt/beta-catenin pathway functions in osteoblast differentiation. ESD extract enhanced terminal osteoblast differentiation as shown by staining with Alizarin Red S and significantly increased murine calvarial bone thickness. This study shows that ESD extract stimulates osteoblast differentiation via the Wnt/beta-catenin pathway and enhances murine calvarial bone formation ex vivo.


Subject(s)
Animals , Humans , Mice , Cell Differentiation/drug effects , Evodia/chemistry , HEK293 Cells , Osteoblasts/cytology , Osteogenesis/drug effects , Plant Extracts/chemistry , Skull/anatomy & histology , Wnt Signaling Pathway/drug effects , beta Catenin/genetics
14.
Braz. j. med. biol. res ; 47(9): 759-765, 09/2014. tab, graf
Article in English | LILACS | ID: lil-719322

ABSTRACT

The monoamine serotonin (5-hydroxytryptamine, 5-HT), a well-known neurotransmitter, also has important functions outside the central nervous system. The objective of this study was to investigate the role of 5-HT in the proliferation, differentiation, and function of osteoblasts in vitro. We treated rat primary calvarial osteoblasts with various concentrations of 5-HT (1 nM to 10 µM) and assessed the rate of osteoblast proliferation, expression levels of osteoblast-specific proteins and genes, and the ability to form mineralized nodules. Next, we detected which 5-HT receptor subtypes were expressed in rat osteoblasts at different stages of osteoblast differentiation. We found that 5-HT could inhibit osteoblast proliferation, differentiation, and mineralization at low concentrations, but this inhibitory effect was mitigated at relatively high concentrations. Six of the 5-HT receptor subtypes (5-HT1A, 5-HT1B, 5-HT1D, 5-HT2A, 5-HT2B, and 5-HT2C) were found to exist in rat osteoblasts. Of these, 5-HT2A and 5-HT1B receptors had the highest expression levels, at both early and late stages of differentiation. Our results indicated that 5-HT can regulate osteoblast proliferation and function in vitro.


Subject(s)
Animals , Calcification, Physiologic/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Osteoblasts/drug effects , Serotonin/pharmacology , DNA Primers , Gene Expression , Osteoblasts/cytology , Osteoblasts/metabolism , Primary Cell Culture , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Receptors, Serotonin/metabolism , Serotonin/metabolism
15.
Experimental & Molecular Medicine ; : e107-2014.
Article in English | WPRIM | ID: wpr-175272

ABSTRACT

MicroRNAs (miRNAs) have recently been recognized to have a role in human orthopedic disorders. The objective of our study was to explore the expression profile and biological function of miRNA-17-5p (miR-17-5p), which is well known to be related to cancer cell proliferation and invasion, in osteoblastic differentiation and in cell proliferation. The expression levels of miR-17-5p in the femoral head mesenchymal stem cells of 20 patients with non-traumatic osteonecrosis (ON) and 10 patients with osteoarthritis (OA) were examined by quantitative reverse transcription-PCR (qRT-PCR). Furthermore, the interaction between miR-17-5p and SMAD7 was observed. We found that in non-traumatic ON samples the level of mature miR-17-5p was significantly lower than that of OA samples (P=0.0002). By targeting SMAD7, miR-17-5p promoted nuclear translocation of beta-catenin, enhanced expression of COL1A1 and finally facilitated the proliferation and differentiation of HMSC-bm cells. We also demonstrated that restoring expression of SMAD7 in HMSC-bm cells partially reversed the function of miR-17-5p. Together, our data suggested a theory that dysfunction of a network containing miR-17-5p, SMAD7 and beta-catenin could contribute to ON pathogenesis. The present study prompts the potential clinical value of miR-17-5p in non-traumatic ON.


Subject(s)
Adult , Female , Humans , Male , Middle Aged , Base Sequence , Bone Morphogenetic Protein 2/metabolism , Cell Differentiation , Cell Line , Cell Proliferation , Gene Expression Regulation , MicroRNAs/genetics , Osteoarthritis/genetics , Osteoblasts/cytology , Osteogenesis , Osteonecrosis/genetics , Signal Transduction , Smad7 Protein/genetics , beta Catenin/metabolism
16.
Braz. j. med. biol. res ; 46(9): 809-814, 19/set. 2013. graf
Article in English | LILACS | ID: lil-686578

ABSTRACT

Bone morphogenetic protein 2 (BMP2) and basic fibroblast growth factor (bFGF) have been shown to exhibit a synergistic effect to promote bone repair and healing. In this study, we constructed a novel adenovirus with high coexpression of BMP2 and bFGF and evaluated its effect on osteogenic differentiation of goat bone marrow progenitor cells (BMPCs). Recombinant adenovirus Ad-BMP2-bFGF was constructed by using the T2A sequence. BMPCs were isolated from goats by density gradient centrifugation and adherent cell culture, and were then infected with Ad-BMP2-bFGF or Ad-BMP2. Expression of BMP2 and bFGF was detected by ELISA, and alkaline phosphatase (ALP) activity was detected by an ALP assay kit. In addition, von Kossa staining and immunocytochemical staining of collagen II were performed on BMPCs 21 days after infection. There was a high coexpression of BMP2 and bFGF in BMPCs infected with Ad-BMP2-bFGF. Twenty-one days after infection, ALP activity was significantly higher in BMPCs infected with Ad-BMP2-bFGF than in those infected with Ad-BMP2. Larger and more mineralized calcium nodules, as well as stronger collagen II staining, were observed in BMPCs infected with Ad-BMP2-bFGF than in those infected with Ad-BMP2. In summary, we developed a novel adenovirus vector Ad-BMP2-bFGF for simultaneous high coexpression of BMP2 and bFGF, which could induce BMPCs to differentiate efficiently into osteoblasts.


Subject(s)
Animals , Adenoviridae/metabolism , Bone Marrow Cells/cytology , /metabolism , Cell Differentiation/physiology , /metabolism , Osteogenesis/physiology , Stem Cells/cytology , Analysis of Variance , Adenoviridae/genetics , Alkaline Phosphatase/metabolism , Base Sequence , Bone Marrow Cells/virology , /genetics , Centrifugation, Density Gradient , Enzyme-Linked Immunosorbent Assay , /genetics , Gene Transfer Techniques , Goats , Genetic Vectors/metabolism , Immunohistochemistry , Osteoblasts/cytology , Primary Cell Culture , Recombinant Proteins/genetics , Stem Cells/virology
17.
Braz. j. med. biol. res ; 46(10): 831-838, 24/set. 2013. tab, graf
Article in English | LILACS | ID: lil-688557

ABSTRACT

Wear particles are phagocytosed by macrophages and other inflammatory cells, resulting in cellular activation and release of proinflammatory factors, which cause periprosthetic osteolysis and subsequent aseptic loosening, the most common causes of total joint arthroplasty failure. During this pathological process, tumor necrosis factor-alpha (TNF-α) plays an important role in wear-particle-induced osteolysis. In this study, recombination adenovirus (Ad) vectors carrying both target genes [TNF-α small interfering RNA (TNF-α-siRNA) and bone morphogenetic protein 2 (BMP-2)] were synthesized and transfected into RAW264.7 macrophages and pro-osteoblastic MC3T3-E1 cells, respectively. The target gene BMP-2, expressed on pro-osteoblastic MC3T3-E1 cells and silenced by the TNF-α gene on cells, was treated with titanium (Ti) particles that were assessed by real-time PCR and Western blot. We showed that recombinant adenovirus (Ad-siTNFα-BMP-2) can induce osteoblast differentiation when treated with conditioned medium (CM) containing RAW264.7 macrophages challenged with a combination of Ti particles and Ad-siTNFα-BMP-2 (Ti-ad CM) assessed by alkaline phosphatase activity. The receptor activator of nuclear factor-κB ligand was downregulated in pro-osteoblastic MC3T3-E1 cells treated with Ti-ad CM in comparison with conditioned medium of RAW264.7 macrophages challenged with Ti particles (Ti CM). We suggest that Ad-siTNFα-BMP-2 induced osteoblast differentiation and inhibited osteoclastogenesis on a cell model of a Ti particle-induced inflammatory response, which may provide a novel approach for the treatment of periprosthetic osteolysis.


Subject(s)
Animals , /metabolism , Osteoblasts/metabolism , RNA, Small Interfering/metabolism , Titanium/adverse effects , Tumor Necrosis Factor-alpha/metabolism , Adenoviridae/genetics , /genetics , Bone Resorption/genetics , Cell Differentiation , Cell Line , Gene Expression , Genetic Vectors/genetics , Osteoblasts/cytology , Osteoblasts/drug effects , RNA, Small Interfering/genetics , Tumor Necrosis Factor-alpha/genetics
18.
Braz. j. med. biol. res ; 46(8): 676-680, ago. 2013. graf
Article in English | LILACS | ID: lil-684529

ABSTRACT

Multipotent mesenchymal stromal cells (MSCs) were first isolated from bone marrow and then from various adult tissues including placenta, cord blood, deciduous teeth, and amniotic fluid. MSCs are defined or characterized by their ability to adhere to plastic, to express specific surface antigens, and to differentiate into osteogenic, chondrogenic, adipogenic, and myogenic lineages. Although the molecular mechanisms that control MSC proliferation and differentiation are not well understood, the involvement of microRNAs has been reported. In the present study, we investigated the role of miR-125b during osteoblastic differentiation in humans. We found that miR-125b increased during osteoblastic differentiation, as well as Runx2 and ALPL genes. To study whether the gain or loss of miR-125b function influenced osteoblastic differentiation, we transfected MSCs with pre-miR-125b or anti-miR-125b and cultured the transfected cells in an osteoblastic differentiation medium. After transfection, no change was observed in osteoblastic differentiation, and Runx2, OPN, and ALPL gene expression were not changed. These results suggest that the gain or loss of miR-125b function does not influence levels of Runx2, OPN, and ALPL during osteoblastic differentiation.


Subject(s)
Female , Humans , Male , Alkaline Phosphatase/metabolism , Cell Differentiation/physiology , Core Binding Factor Alpha 1 Subunit/metabolism , MicroRNAs/metabolism , Osteoblasts/cytology , Osteopontin/metabolism , Alkaline Phosphatase/genetics , Antigens, Differentiation/isolation & purification , Bone Marrow Cells/cytology , Core Binding Factor Alpha 1 Subunit/genetics , Gene Expression/physiology , Leukocytes, Mononuclear/cytology , Mesenchymal Stem Cells/cytology , MicroRNAs/genetics , Osteoblasts/metabolism , Osteogenesis/physiology , Osteopontin/genetics , Primary Cell Culture , Reverse Transcriptase Polymerase Chain Reaction , Transfection
19.
Indian J Biochem Biophys ; 2013 Feb; 50(1): 19-25
Article in English | IMSEAR | ID: sea-147282

ABSTRACT

R-spondin (Rspo)s proteins are a new group of Wnt/beta-catenin signaling agonists. These signaling molecules are known to be involved in the developmental stages of skeletal system. Recent studies in various murine osteoblast models have proposed that Rspo1 may interact with Wnt signaling pathway to induce differentiation in osteoblasts. Though findings in murine osteoblasts implicate a synergestic role of Rspo1 with Wnt signaling, still no study has addressed the similar role in more clinically applicable osteoblast models i.e., human cell lines or primary cells. Therefore, in the present study, we investigated the possible role of Rspo1 during differentiation process of human in vitro osteoblast cell models like primary osteoblasts or human osteoprogenitor cell line hFOB1.19 along with murine preosteoblast cell line MC3T3 E-1. Our results showed increase in Rspo1 at transcript level during differentiating phase of human primary osteoblasts and human FOB1.19 cells. We also found that Rspo1 (100 ng/mL) acts additively with Wnt3a to activate Wnt signaling, as confirmed by luciferase activity after transfection of TOPFLASH construct to hFOB1.19 cells. Similar additive role of Rspo1 and Wnt3a was apparent in alkaline phosphatase (ALP) activity analysis of human primary cells. Moreover, a reduction in ALP activity was observed with knock-down of Rspo1 by transfected shRNA in hFOB1.19 cells. These results suggested the possibility of autocrine regulation by Rspo1 on the osteogenic activities in human in vitro osteoblast models. Furthermore, these results were corroborated in MC3T3-E1, murine osteoblast cell model. Osteoblastic differentiation was induced by transfection of Rspo1 which was confirmed by increased ALP staining and qRT-PCR analysis of osteogenic markers, such as Runx2 and osteocalcin. In conclusion, present study highlights the role of Rspo1 in bone remodeling where it activates Wnt signaling to induce differentiation, as shown in human as well murine in vitro osteoblast cell models.


Subject(s)
Animals , Cell Differentiation , Cell Line , Humans , Mice , Osteoblasts/cytology , Osteoblasts/metabolism , Osteogenesis/physiology , Thrombospondins/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway/physiology
20.
Experimental & Molecular Medicine ; : e9-2013.
Article in English | WPRIM | ID: wpr-199829

ABSTRACT

Activated protein C (APC) is a cytoprotective anticoagulant that can promote cutaneous healing. We examined the effect of APC on viability and differentiation of the osteoblastic line, MG63, in the presence and absence of bisphosphonates (BPs). Osteoblasts were cultured and treated for 24 or 48 h with Alendronate (Aln), Zoledronate (Zol) or Pamidronate (Pam) at concentrations ranging from 10-4 to 10-6 M. Cell differentiation was measured using type 1 collagen production, Alizarin red staining and alkaline phosphatase activity, whereas cell viability was assessed using MTT and crystal violet assays. All three BPs induced MG63 cell death in a dose- and time-dependent manner. Pam- and Zol-related cell death was prevented by APC treatment; however, cell death induced by Aln was accelerated by APC. APC induced MG63 cell differentiation that was enhanced by Aln, but inhibited by Pam or Zol. Endothelial protein C receptor (EPCR) was expressed by MG63 cells and mediated the protective effect of APC on Zol-induced viability. In summary, we have demonstrated that (1) APC favorably regulates MG63 viability and differentiation toward bone growth, (2) APC differentially regulates the effects of specific BPs and (3) at least part of the effects of APC is mediated through EPCR. These findings highlight the potential importance of the PC pathway in bone physiology and provide strong evidence that APC may influence bone cells and has potential to be a therapeutic drug for bone regeneration, depending on concurrent BP treatment.


Subject(s)
Humans , Antigens, CD/metabolism , Caspases/metabolism , Cell Differentiation/drug effects , Cell Survival/drug effects , Collagen Type I/metabolism , Diphosphonates/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Matrix Metalloproteinase 2/metabolism , NF-kappa B/metabolism , Osteoblasts/cytology , Protein C/pharmacology , Receptors, Cell Surface/metabolism , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL