Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Oral Dis ; 2024 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-38764359

RESUMO

OBJECTIVE: Hypertension disrupts the bone integrity and its repair ability. This study explores the efficiency of a therapy based on the application of mesenchymal stem cells (MSCs) to repair bone defects of spontaneously hypertensive rats (SHR). METHODS: First, we evaluated SHR in terms of bone morphometry and differentiation of MSCs into osteoblasts. Then, the effects of the interactions between MSCs from normotensive rats (NTR-MSCs) cocultured with SHR (SHR-MSCs) on the osteoblast differentiation of both cell populations were evaluated. Also, bone formation into calvarial defects of SHR treated with NTR-MSCs was analyzed. RESULTS: Hypertension induced bone loss evidenced by reduced bone morphometric parameters of femurs of SHR compared with NTR as well as decreased osteoblast differentiation of SHR-MSCs compared with NTR-MSCs. NTR-MSCs partially restored the capacity of SHR-MSCs to differentiate into osteoblasts, while SHR-MSCs exhibited a slight negative effect on NTR-MSCs. An enhanced bone repair was observed in defects treated with NTR-MSCs compared with control, stressing this cell therapy efficacy even in bones damaged by hypertension. CONCLUSION: The use of MSCs derived from a heathy environment can be in the near future a smart approach to treat bone loss in the context of regenerative dentistry for oral rehabilitation of hypertensive patients.

2.
Life Sci ; 340: 122463, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38286209

RESUMO

AIMS: Cell therapy utilizing mesenchymal stem cells (MSCs) from healthy donors (HE-MSCs) is a promising strategy for treating osteoporotic bone defects. This study investigated the effects of interaction between HE-MSCs and MSCs from osteoporotic donors (ORX-MSCs) on osteoblast differentiation of MSCs and of HE-MSCs on bone formation in calvarial defects of osteoporotic rats. MATERIALS AND METHODS: Osteoporosis was induced by orchiectomy (ORX) and its effects on the bone were evaluated by femur microtomography (µCT) and osteoblast differentiation of bone marrow MSCs. HE- and ORX-MSCs were cocultured, and osteoblast differentiation was evaluated using genotypic and phenotypic parameters. HE-MSCs were injected into the calvarial defects of osteoporotic rats, and bone formation was evaluated by µCT, histology, and gene expression of osteoblast markers. KEY FINDINGS: ORX-induced osteoporosis was revealed by reduced bone morphometric parameters and osteoblast differentiation in ORX-MSCs. HE-MSCs partially recovered the osteogenic potential of ORX-MSCs, whereas HE-MSCs were mildly affected by ORX-MSCs. Additionally, the bone morphogenetic protein and wingless-related integration site signaling pathway components were similarly modulated in cocultures involving ORX-MSCs. HE-MSCs induced meaningful bone formation, highlighting the effectiveness of cell therapy even in osteoporotic bones. SIGNIFICANCE: These results provide new perspectives on the development of cell-based therapies to regenerate bone defects in patients with disorders that affect bone tissue.


Assuntos
Células-Tronco Mesenquimais , Osteoporose , Humanos , Ratos , Animais , Osteogênese , Osso e Ossos/metabolismo , Diferenciação Celular/genética , Osteoporose/metabolismo , Osteoblastos/metabolismo , Células Cultivadas
3.
J Cell Physiol ; 238(11): 2625-2637, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37661654

RESUMO

The secretome present in the conditioned medium (CM) of mesenchymal stem cells (MSCs) is a promising tool to be used in therapies to promote bone regeneration. Considering the high osteogenic potential of the bone morphogenetic protein 9 (BMP-9), we hypothesized that the secretome of MSCs overexpressing BMP-9 (MSCsBMP-9 ) enhances the osteoblast differentiation of MSCs and the bone formation in calvarial defects. CM of either MSCsBMP-9 (CM-MSCsBMP-9 ) or MSCs without BMP-9 overexpression (CM-MSCsVPR ) were obtained at different periods. As the CM-MSCsBMP-9 generated after 1 h presented the highest BMP-9 concentration, CM-MSCsBMP-9 and CM-MSCsVPR were collected at this time point and used to culture MSCs and to be injected into mouse calvarial defects. The CM-MSCsBMP-9 enhanced the osteoblast differentiation of MSC by upregulating RUNX2, alkaline phosphatase (ALP) and osteopontin protein expression, and ALP activity, compared with CM-MSCsVPR . The CM-MSCsBMP-9 also enhanced the bone repair of mouse calvarial defects, increasing bone volume, bone volume/total volume, bone surface, and trabecular number compared with untreated defects and defects treated with CM-MSCsVPR or even with MSCsBMP-9 themselves. In conclusion, the potential of the MSCBMP-9 -secretome to induce osteoblast differentiation and bone formation shed lights on novel cell-free-based therapies to promote bone regeneration of challenging defects.


Assuntos
Células-Tronco Mesenquimais , Osteogênese , Animais , Camundongos , Proteína Morfogenética Óssea 2/metabolismo , Diferenciação Celular , Células Cultivadas , Fator 2 de Diferenciação de Crescimento/genética , Células-Tronco Mesenquimais/metabolismo , Osteoblastos/metabolismo , Secretoma
4.
Cell Signal ; 109: 110773, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37331417

RESUMO

The Wnt/ß-catenin signaling pathway is associated with the regulation of cancer stem cells, and it can be driven by epigenetic modifications. Here, we aim to identify epigenetic modifications involved in the control of the Wnt/ß-catenin signaling and investigate the role of this pathway in the accumulation of cancer stem cells (CSC) and chemoresistance of Head and Neck Squamous Cell Carcinoma (HNSCC). Quantitative-PCR, western blot, shRNA assay, viability assay, flow cytometry assay, spheres formation, xenograft model, and chromatin immunoprecipitation were employed to evaluate the Wnt/ß-catenin pathway and EZH2 in wild-type and chemoresistant oral carcinoma cell lines, and in the populations of CSC and non-stem cells. We demonstrated that ß-catenin and EZH2 were accumulated in cisplatin-resistant and CSC population. The upstream genes of the Wnt/ß-catenin signaling (APC and GSK3ß) were decreased, and the downstream gene MMP7 was increased in the chemoresistant cell lines. The inhibition of ß-catenin and EZH2 combined effectively decreased the CSC population in vitro and reduced the tumor volume and CSC population in vivo. EZH2 inhibition increased APC and GSK3ß, and the Wnt/ß-catenin inhibition reduced MMP7 levels. In contrast, EZH2 overexpression decreased APC and GSK3ß and increased MMP7. EZH2 and ß-catenin inhibitors sensitized chemoresistant cells to cisplatin. EZH2 and H3K27me3 bounded the promoter of APC, leading to its repression. These results suggest that EZH2 regulates ß-catenin by inhibiting the upstream gene APC contributing to the accumulation of cancer stem cells and chemoresistance. Moreover, the pharmacological inhibition of the Wnt/ß-catenin combined with EZH2 can be an effective strategy for treating HNSCC.


Assuntos
Cisplatino , Neoplasias de Cabeça e Pescoço , Humanos , Carcinoma de Células Escamosas de Cabeça e Pescoço/tratamento farmacológico , Cisplatino/farmacologia , Cisplatino/uso terapêutico , beta Catenina/metabolismo , Glicogênio Sintase Quinase 3 beta/metabolismo , Metaloproteinase 7 da Matriz/genética , Metaloproteinase 7 da Matriz/metabolismo , Metaloproteinase 7 da Matriz/farmacologia , Linhagem Celular Tumoral , Via de Sinalização Wnt , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Neoplasias de Cabeça e Pescoço/patologia , Células-Tronco Neoplásicas/metabolismo , Regulação Neoplásica da Expressão Gênica , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo
5.
J Funct Biomater ; 14(6)2023 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-37367270

RESUMO

BACKGROUND: Tissue engineering and cell therapy have been the focus of investigations on how to treat challenging bone defects. This study aimed to produce and characterize a P(VDF-TrFE)/BaTiO3 scaffold and evaluate the effect of mesenchymal stem cells (MSCs) combined with this scaffold and photobiomodulation (PBM) on bone repair. METHODS AND RESULTS: P(VDF-TrFE)/BaTiO3 was synthesized using an electrospinning technique and presented physical and chemical properties suitable for bone tissue engineering. This scaffold was implanted in rat calvarial defects (unilateral, 5 mm in diameter) and, 2 weeks post-implantation, MSCs were locally injected into these defects (n = 12/group). Photobiomodulation was then applied immediately, and again 48 and 96 h post-injection. The µCT and histological analyses showed an increment in bone formation, which exhibited a positive correlation with the treatments combined with the scaffold, with MSCs and PBM inducing more bone repair, followed by the scaffold combined with PBM, the scaffold combined with MSCs, and finally the scaffold alone (ANOVA, p ≤ 0.05). CONCLUSIONS: The P(VDF-TrFE)/BaTiO3 scaffold acted synergistically with MSCs and PBM to induce bone repair in rat calvarial defects. These findings emphasize the need to combine a range of techniques to regenerate large bone defects and provide avenues for further investigations on innovative tissue engineering approaches.

6.
J Cell Biochem ; 124(7): 1050-1063, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37293736

RESUMO

Type 1 (T1DM) and type 2 (T2DM) diabetes mellitus are characterized by changes in glucose metabolism and cause bone damage via a variety of mechanisms, including effects on osteoblasts. We aimed to evaluate the osteoblast differentiation of mesenchymal stem cells (MSCs) from rats with T1DM or T2DM and the effects of removing the hyperglycemic stimulus on the osteogenic potential of these cells. MSCs from healthy rats were cultured in normoglycemic medium, whereas MSCs from rats with T1DM or T2DM were cultured in hyperglycemic or normoglycemic medium. T1DM and T2DM reduced osteoblast differentiation of MSCs grown in hyperglycemic media, with T1DM having a more pronounced effect, as evidenced by alkaline phosphatase activity, RUNX2 protein expression, and extracellular matrix mineralization, and modulated the gene expression of several components of the bone morphogenetic protein signaling pathway. The restoration of the normoglycemic environment partially recovers the osteogenic potential of MSCs from rats with T1DM but not with T2DM. Our findings highlight the need for specific therapies to treat T1DM- or T2DM-induced bone loss, as both disrupt osteoblast differentiation at distinct levels and likely through different mechanisms.


Assuntos
Diabetes Mellitus Tipo 1 , Diabetes Mellitus Tipo 2 , Células-Tronco Mesenquimais , Ratos , Animais , Diabetes Mellitus Tipo 1/metabolismo , Células Cultivadas , Osteogênese/genética , Diferenciação Celular , Osteoblastos/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Células-Tronco Mesenquimais/metabolismo
7.
Regen Med ; 18(5): 377-387, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37125511

RESUMO

Aim: We evaluated the bone repair induced by MSCs from adipose tissue (AT-MSCs) and bone marrow (BM-MSCs) injected into rat calvarial defects at two time points. Methods & results: Both cell populations expressed MSC surface markers and differentiated into adipocytes and osteoblasts. µCT showed that the combination of cells from distinct sources exhibited synergistic effects to increase bone repair with an advantage when BM-MSCs were injected prior to AT-MSCs. The higher osteogenic potential of these MSC combinations was demonstrated using an in vitro coculture system where BM-MSCs and AT-MSCs association induced higher ALP activity in MC3T3-E1 cells. Conclusion: Our findings may drive new approaches to treat bone defects and shed light on the complexity of the mechanisms involved in bone regeneration.


We evaluated the bone repair induced by cells that can develop into different types of cells (stem cells) derived from fat and spongy tissue inside the large bones and injected into defects created in rat skulls. Cells derived from both tissues developed into fat cells and bone-forming cells. The combination of cells from fat and spongy tissue exhibited cooperative effects to increase bone repair with an advantage when cells from spongy tissue were injected prior to cells from fat. Our findings may contribute to stablish new therapies based on the use of cells to treat large bone defects.


Assuntos
Medula Óssea , Células-Tronco Mesenquimais , Ratos , Animais , Tecido Adiposo , Osteogênese , Regeneração Óssea , Diferenciação Celular , Células da Medula Óssea , Células Cultivadas
8.
RGO (Porto Alegre) ; 71: e20230011, 2023. graf
Artigo em Inglês | LILACS-Express | LILACS, BBO - odontologia (Brasil) | ID: biblio-1431160

RESUMO

ABSTRACT The traumatic bone cyst is an uncommon nonneoplastic lesion of the jaws that is considered as a "pseudocyst" because of the lack of an epithelial lining. This lesion is particularly asymptomatic and, therefore, is diagnosed by routine dental radiographic examination as a unilocular radiolucency with scalloped borders, mainly in the posterior mandibular region. The exact etiopathogenesis of the lesion remains uncertain, though it is often associated with trauma. The objective of this paper is to report one case of atypical traumatic bone cyst involving impacted lower third molar, addressing its clinical and radiographic characteristics, differential diagnosis, treatment through surgical exploration and case follow-up.


RESUMO O cisto ósseo traumático é uma lesão não neoplásica incomum dos maxilares, considerada um "pseudocisto" devido à ausência de um revestimento epitelial. Esta lesão é particularmente assintomática e, portanto, é diagnosticada pelo exame radiográfico odontológico de rotina como uma radioluscência unilocular com bordas recortadas, principalmente na região mandibular posterior. A etiopatogenia exata da lesão permanece incerta, embora esteja frequentemente associada a trauma. O objetivo deste trabalho é relatar um caso de cisto ósseo traumático atípico envolvendo terceiro molar inferior impactado abordando suas características clínicas, radiográficas, diagnóstico diferencial, tratamento por meio de exploração cirúrgica e proservação do caso.

9.
J Biomed Mater Res B Appl Biomater ; 110(12): 2744-2750, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35857711

RESUMO

The aim of this study was to evaluate the in vivo performance of two different deproteinized bovine bone (DBB) grafting materials: DBBB (Bio-Oss®) and DBBL (Laddec®), for the regeneration of critically sized (8 mm) defects in rabbit's calvaria. Three round-shaped defects were surgically created in the calvaria of 13 New Zealand White rabbits proximal to the coronal suture in the parietal bone. Two of the defects were filled with one of the grafting materials while a third was left empty to serve as a negative control. Bone regeneration properties were evaluated at 4- and 8-weeks after implantation by means of histological and histomorphometrical analyses. Statistical analyses were performed through a mixed model analysis with fixed factors of time and material. Histological evaluation of the control group evidenced a lack of bridging bone formation across the defect sites at both evaluation time points. For the experimental groups, new bone formation was observed around the defect periphery and to progress radially inwards to the center of the defect site, regardless of the grafting material. Histomorphometric analyses at 4 weeks demonstrated higher amount of bone formation through the defect for DBBB group. However, at 8 weeks, DBBL and DBBB demonstrated osteoconductivity and low resorption rates with evidence of statistically similar bone regeneration through the complete boney defect. Finally, DBBB presented lower soft tissue migration within the defect when compared to DBBL at both evaluation time points. DBBB and DBBL presented similar bone regeneration performance and slow resorption rates. Although both materials promoted bone regeneration through the complete defect, DBBB presented lower soft tissue migration within the defects at 4- and 8-weeks.


Assuntos
Substitutos Ósseos , Animais , Regeneração Óssea , Substitutos Ósseos/farmacologia , Transplante Ósseo , Bovinos , Minerais , Coelhos , Crânio/cirurgia
10.
Regen Med ; 17(6): 341-353, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35291805

RESUMO

Aim: This study aimed to evaluate the ability of human periodontal ligament stem cells (PDLSCs) with high (HP-PDLSCs) and low (LP-PDLSCs) osteogenic potential, in addition to mixed cells, to repair bone tissue. Methods: Cell phenotype, proliferation and differentiation were evaluated. Undifferentiated PDLSCs were injected into rat calvarial defects and the new bone was evaluated by µCT, histology and real-time PCR. Results: PDLSCs exhibited a typical mesenchymal stem cell phenotype and HP-PDLSCs showed lower proliferative and higher osteogenic potential than LP-PDLSCs. PDLSCs induced similar bone formation and histological analysis suggests a remodeling process, confirmed by osteogenic and osteoclastogenic markers, especially in tissues derived from defects treated with HP-PDLSCs. Conclusion: PDLSCs induced similar bone formation irrespective of their in vitro osteogenic potential.


Bone is one of the most transplanted tissues worldwide and cell-based therapies has been investigated as an alternative for the treatment of bone defects. Dental tissues have been investigated as sources of stem cells and the periodontal ligament has been shown to be a viable source of these cells. Stem cells from periodontal ligament induce significant bone formation in rat calvaria defects and are safe for cell-based therapies, as the cells remain at the bone defect site for up to 4 weeks and do not migrate to vital organs, such as brain, heart, lungs, spleen, kidneys, and liver in the same period. In addition, immune responses were not detected. Considering that, stem cells from periodontal ligament can be useful in cell therapy strategies to induce bone regeneration.


Assuntos
Osteogênese , Ligamento Periodontal , Animais , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Humanos , Ratos , Crânio , Células-Tronco
11.
Materials (Basel) ; 15(3)2022 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-35161039

RESUMO

Chemical and topographical surface modifications on dental implants aim to increase the bone surface contact area of the implant and improve osseointegration. This study analyzed the cellular response of undifferentiated mesenchymal stem cells (MSC), derived from senile rats' femoral bone marrow, when cultured on a bioactive coating (by plasma electrolytic oxidation, PEO, with Ca2+ and P5+ ions), a sandblasting followed by acid-etching (SLA) surface, and a machined surface (MSU). A total of 102 Ti-6Al-4V discs were divided into three groups (n = 34). The surface chemistry was analyzed by energy dispersive spectroscopy (EDS). Cell viability assay, gene expression of osteoblastic markers, and mineralized matrix formation were investigated. The cell growth and viability results were higher for PEO vs. MSU surface (p = 0.001). An increase in cell proliferation from 3 to 7 days (p < 0.05) and from 7 to 10 days (p < 0.05) was noted for PEO and SLA surfaces. Gene expression for OSX, ALP, BSP, and OPN showed a statistical significance (p = 0.001) among groups. In addition, the PEO surface showed a higher mineralized matrix bone formation (p = 0.003). In conclusion, MSC from senile female rats cultured on SLA and PEO surfaces showed similar cellular responses and should be considered for future clinical investigations.

12.
Cell Tissue Res ; 386(2): 335-347, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34223979

RESUMO

The extracellular matrix protein Agrin has been detected in chondrocytes and endosteal osteoblasts but its function in osteoblast differentiation has not been investigated yet. Thus, it is possible that Agrin contributes to osteoblast differentiation and, due to Agrin and wingless-related integration site (Wnt) sharing the same receptor, transmembrane low-density lipoprotein receptor-related protein 4 (Lrp4), and the crosstalk between Wnt and bone morphogenetic protein (BMP) signalling, both pathways could be involved in this Agrin-mediated osteoblast differentiation. Confirming this, Agrin and its receptors Lrp4 and α-dystroglycan (Dag1) were expressed during differentiation of osteoblasts from three different sources. Moreover, the disruption of Agrin impaired the expression of its receptors and osteoblast differentiation, and the treatment with recombinant Agrin slightly increase this process. In addition, whilst Agrin knockdown downregulated the expression of genes related to Wnt and BMP signalling pathways, the addition of Agrin had no effect on these genes. Altogether, these data uncover the contribution of Agrin to osteoblast differentiation and suggest that, at least in part, an Agrin-Wnt-BMP circuit is involved in this process. This makes Agrin a candidate as target for developing new therapeutic strategies to treat bone-related diseases and injuries.


Assuntos
Agrina/análise , Osteoblastos/citologia , Células 3T3 , Agrina/genética , Animais , Diferenciação Celular , Células Cultivadas , Regulação da Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Osteoblastos/metabolismo , Osteogênese
13.
Gene Ther ; 28(12): 748-759, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-33686254

RESUMO

Cell therapy is a valuable strategy for the replacement of bone grafts and repair bone defects, and mesenchymal stem cells (MSCs) are the most frequently used cells. This study was designed to genetically edit MSCs to overexpress bone morphogenetic protein 9 (BMP-9) using Clustered Regularly Interspaced Short Palindromic Repeats/associated nuclease Cas9 (CRISPR-Cas9) technique to generate iMSCs-VPRBMP-9+, followed by in vitro evaluation of osteogenic potential and in vivo enhancement of bone formation in rat calvaria defects. Overexpression of BMP-9 was confirmed by its gene expression and protein expression, as well as its targets Hey-1, Bmpr1a, and Bmpr1b, Dlx-5, and Runx2 and  protein expression of SMAD1/5/8 and pSMAD1/5/8. iMSCs-VPRBMP-9+ displayed significant changes in the expression of a panel of genes involved in TGF-ß/BMP signaling pathway. As expected, overexpression of BMP-9 increased the osteogenic potential of MSCs indicated by increased gene expression of osteoblastic markers Runx2, Sp7, Alp, and Oc, higher ALP activity, and matrix mineralization. Rat calvarial bone defects treated with injection of iMSCs-VPRBMP-9+ exhibited increased bone formation and bone mineral density when compared with iMSCs-VPR- and phosphate buffered saline (PBS)-injected defects. This is the first study to confirm that CRISPR-edited MSCs overexpressing BMP-9 effectively enhance bone formation, providing novel options for exploring the capability of genetically edited cells to repair bone defects.


Assuntos
Fator 2 de Diferenciação de Crescimento , Células-Tronco Mesenquimais , Osteogênese , Animais , Sistemas CRISPR-Cas , Diferenciação Celular , Células Cultivadas , Fator 2 de Diferenciação de Crescimento/genética , Células-Tronco Mesenquimais/citologia , Osteogênese/genética , Ratos
14.
Mater Sci Eng C Mater Biol Appl ; 120: 111775, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33545905

RESUMO

The treatment of polytrauma patients represents a great challenge in the maxillofacial and orthopedic surgery fields. Therefore, this study tested the hypothesis that the use of a bioactive coating (by plasma electrolytic oxidation, PEO) on titanium microplates could improve the fracture healing of low bone mineral density (BMD) rats. Thirty female rats underwent bilateral ovariectomy surgery (OVX), and 35 rats underwent fake surgery (SHAM). Three months later, animals were subjected to femoral fracture simulation and were fixed with either non-coated (CONV) or coated (PEO) titanium miniplates. Eight weeks postoperatively, microplate/bone complexes were analyzed through computed microtomography, histometric, confocal microscopy, molecular, and biomechanical analysis. Bioactive elements (Ca and P) were incorporated on the PEO microplate and the surface was modified in a volcano-like structure. In the microCT analysis the OVX/PEO group had greater values for Tb.Th (bone trabecular thickness), Tb.Sp (separation of bone trabeculae) and Tb.N (number of trabeculae) parameters compared to the OVX/CONV group. According to histometric analysis, the OVX/PEO group showed significantly higher new bone formation than the OVX/CONV group (P < 0.05). For the fluorochrome area, the OVX groups (PEO and CONV) showed greater values for calcein precipitation (old bone) than alizarin red (new bone). Molecular results showed greater values for proteins related to the final phase of bone formation (P < 0.05) in the OVX/PEO group. The OVX/PEO group showed higher bone/miniplate system resilience compared to the others (P < 0.05). It was concluded that PEO coating optimizes bone healing on simulated femoral fractures in low bone mineral density rats. This sheds new light in the treatment of osteoporotic patients with bone fractures.


Assuntos
Doenças Ósseas Metabólicas , Fraturas do Fêmur , Osteoporose , Animais , Densidade Óssea , Feminino , Fraturas do Fêmur/diagnóstico por imagem , Fraturas do Fêmur/terapia , Humanos , Ovariectomia , Ratos
15.
J Bone Miner Metab ; 39(2): 163-173, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32889573

RESUMO

INTRODUCTION: The jabuticaba peel extract (JPE) contains bioactive compounds that regulate fat metabolism. Because the negative correlation between fat accumulation and bone formation in bone marrow, we hypothesized that JPE inhibits adipocyte as well as favors osteoblast differentiation of mesenchymal stromal cells (MSCs) under healthy and osteoporotic conditions, a disease that display an imbalance between adipocyte and osteoblast differentiation resulting in reduced bone mass. MATERIAL AND METHODS: To test these hypotheses, bone marrow MSCs were harvested from healthy and osteoporotic rats and cultured in adipogenic and osteogenic media with three concentrations of JPE, 0.25, 5 and 10 µg/ml, and vehicle (control). After selecting the most efficient concentrations of JPE, we used them to evaluate adipocyte and osteoblast differentiation of MSCs from both sources. RESULTS: We observed that, in general, JPE inhibited adipocyte differentiation of MSCs with more pronounced effects in cells from healthy than osteoporotic rats. In addition, JPE increased osteoblast differentiation, exhibiting a slightly higher osteogenic potential on MSCs from osteoporotic compared to healthy condition. CONCLUSION: Our results demonstrated that JPE drives MSCs to inhibit adipocyte differentiation and toward osteoblast differentiation under healthy and osteoporotic conditions. These findings pave the way for further translational studies to investigate the therapeutic possibilities of JPE in both prevention and treatment of osteoporosis.


Assuntos
Adipócitos/citologia , Células-Tronco Mesenquimais/citologia , Osteoblastos/citologia , Osteoporose/patologia , Extratos Vegetais/farmacologia , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Adipogenia/efeitos dos fármacos , Adipogenia/fisiologia , Animais , Medula Óssea/efeitos dos fármacos , Medula Óssea/metabolismo , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/patologia , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Feminino , Células-Tronco Mesenquimais/efeitos dos fármacos , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Osteogênese/efeitos dos fármacos , Osteoporose/metabolismo , Ovariectomia , Ratos Wistar
16.
J Bone Miner Metab ; 38(4): 481-490, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32078052

RESUMO

INTRODUCTION: The aim of this study was to evaluate the in vitro osteogenic potential of osteoblasts from neural crest-derived frontal bone (OB-NC) and mesoderm-derived parietal bone (OB-MS) and the bone formation induced by them when injected into calvarial defects. MATERIALS AND METHODS: Calvarial bones were collected from newborn Wistar rats (3-day old) and characterized as frontal and parietal prior to OB-NC and OB-MS harvesting. The cells were cultured, and several parameters of osteoblast differentiation were evaluated. These cells, or PBS without cells (control), were locally injected into 5-mm rat calvarial defects (5 × 106 cells/defect) and after 4 weeks bone formation was evaluated by morphometric and histological analyses. RESULTS: The characterization of frontal and parietal bones assured the different embryonic origin of both cell populations, OB-NC and OB-MS. The OB-NC presented higher proliferation while the OB-MS presented higher alkaline phosphatase (ALP) activity, extracellular matrix mineralization and gene expression of runt-related transcription factor 2, Alp, bone sialoprotein and osteocalcin revealing their high osteogenic potential. µCT analysis indicated that there was higher amount of bone formation in defects injected with both OB-NC and OB-MS compared to the control. Moreover, the bone tissue formed by both cells displayed the same histological characteristics. CONCLUSIONS: Despite the distinct in vitro osteogenic potential, OB-NC and OB-MS induced similar bone repair in a rat calvarial defect model. Thus, osteoblasts, irrespective of their in vitro osteogenic potential linked to embryonic origins, seem to be suitable for cell-based therapies aiming to repair bone defects.


Assuntos
Osteoblastos/citologia , Osteogênese , Crânio/embriologia , Cicatrização , Animais , Animais Recém-Nascidos , Biomarcadores/metabolismo , Diferenciação Celular , Proliferação de Células/genética , Células Cultivadas , Regulação da Expressão Gênica , Osteogênese/genética , Ratos Wistar , Cicatrização/genética , Microtomografia por Raio-X
17.
J Appl Oral Sci ; 28: e20190156, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32049134

RESUMO

OBJECTIVE: The present study aimed to investigate the participation of focal adhesion kinases (FAK) in interactions between osteoblastic cells and titanium (Ti) surfaces with three different topographies, namely, untreated (US), microstructured (MS), and nanostructured (NS). METHODOLOGY: Osteoblasts harvested from the calvarial bones of 3-day-old rats were cultured on US, MS and NS discs in the presence of PF-573228 (FAK inhibitor) to evaluate osteoblastic differentiation. After 24 h, we evaluated osteoblast morphology and vinculin expression, and on day 10, the following parameters: gene expression of osteoblastic markers and integrin signaling components, FAK protein expression and alkaline phosphatase (ALP) activity. A smooth surface, porosities at the microscale level, and nanocavities were observed in US, MS, and NS, respectively. RESULTS: FAK inhibition decreased the number of filopodia in cells grown on US and MS compared with that in NS. FAK inhibition decreased the gene expression of Alp, bone sialoprotein, osteocalcin, and ALP activity in cells grown on all evaluated surfaces. FAK inhibition did not affect the gene expression of Fak, integrin alpha 1 ( Itga1 ) and integrin beta 1 ( Itgb1 ) in cells grown on MS, increased the gene expression of Fak in cells grown on NS, and increased the gene expression of Itga1 and Itgb1 in cells grown on US and NS. Moreover, FAK protein expression decreased in cells cultured on US but increased in cells cultured on MS and NS after FAK inhibition; no difference in the expression of vinculin was observed among cells grown on all surfaces. CONCLUSIONS: Our data demonstrate the relevance of FAK in the interactions between osteoblastic cells and Ti surfaces regardless of surface topography. Nanotopography positively regulated FAK expression and integrin signaling pathway components during osteoblast differentiation. In this context, the development of Ti surfaces with the ability to upregulate FAK activity could positively impact the process of implant osseointegration.


Assuntos
Proteína-Tirosina Quinases de Adesão Focal/antagonistas & inibidores , Osteoblastos/efeitos dos fármacos , Quinolonas/farmacologia , Sulfonas/farmacologia , Titânio/química , Animais , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Proteína-Tirosina Quinases de Adesão Focal/análise , Proteína-Tirosina Quinases de Adesão Focal/química , Expressão Gênica , Integrinas/análise , Microscopia Eletrônica de Varredura , Osseointegração/efeitos dos fármacos , Osteoblastos/fisiologia , Quinolonas/química , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais , Sulfonas/química , Propriedades de Superfície
18.
Bio Protoc ; 10(4): e3534, 2020 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-33654758

RESUMO

Since their discovery, mesenchymal stromal cells (MSCs) have received a lot of attention, mainly due to their self-renewal potential and multilineage differentiation capacity. For these reasons, MSCs are a useful tool in cell biology and regenerative medicine. In this article, we describe protocols to isolate MSCs from bone marrow (BM-MSCs) and adipose tissues (AT-MSCs), and methods to culture, characterize, and differentiate MSCs into osteoblasts, adipocytes, and chondrocytes. After the harvesting of cells from bone marrow by flushing the femoral diaphysis and enzymatic digestion of abdominal and inguinal adipose tissues, MSCs are selected by their adherence to the plastic tissue culture dish. Within 7 days, MSCs reach 70% confluence and are ready to be used in subsequent experiments. The protocols described here are easy to perform, cost-efficient, require minimal time, and yield a cell population rich in MSCs.

19.
J. appl. oral sci ; 28: e20190156, 2020. graf
Artigo em Inglês | LILACS, BBO - odontologia (Brasil) | ID: biblio-1090765

RESUMO

Abstract Objective The present study aimed to investigate the participation of focal adhesion kinases (FAK) in interactions between osteoblastic cells and titanium (Ti) surfaces with three different topographies, namely, untreated (US), microstructured (MS), and nanostructured (NS). Methodology Osteoblasts harvested from the calvarial bones of 3-day-old rats were cultured on US, MS and NS discs in the presence of PF-573228 (FAK inhibitor) to evaluate osteoblastic differentiation. After 24 h, we evaluated osteoblast morphology and vinculin expression, and on day 10, the following parameters: gene expression of osteoblastic markers and integrin signaling components, FAK protein expression and alkaline phosphatase (ALP) activity. A smooth surface, porosities at the microscale level, and nanocavities were observed in US, MS, and NS, respectively. Results FAK inhibition decreased the number of filopodia in cells grown on US and MS compared with that in NS. FAK inhibition decreased the gene expression of Alp, bone sialoprotein, osteocalcin, and ALP activity in cells grown on all evaluated surfaces. FAK inhibition did not affect the gene expression of Fak, integrin alpha 1 ( Itga1 ) and integrin beta 1 ( Itgb1 ) in cells grown on MS, increased the gene expression of Fak in cells grown on NS, and increased the gene expression of Itga1 and Itgb1 in cells grown on US and NS. Moreover, FAK protein expression decreased in cells cultured on US but increased in cells cultured on MS and NS after FAK inhibition; no difference in the expression of vinculin was observed among cells grown on all surfaces. Conclusions Our data demonstrate the relevance of FAK in the interactions between osteoblastic cells and Ti surfaces regardless of surface topography. Nanotopography positively regulated FAK expression and integrin signaling pathway components during osteoblast differentiation. In this context, the development of Ti surfaces with the ability to upregulate FAK activity could positively impact the process of implant osseointegration.


Assuntos
Animais , Osteoblastos/efeitos dos fármacos , Sulfonas/farmacologia , Titânio/química , Quinolonas/farmacologia , Proteína-Tirosina Quinases de Adesão Focal/antagonistas & inibidores , Osteoblastos/fisiologia , Sulfonas/química , Propriedades de Superfície , Microscopia Eletrônica de Varredura , Transdução de Sinais , Expressão Gênica , Integrinas/análise , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Osseointegração/efeitos dos fármacos , Ratos Wistar , Quinolonas/química , Proliferação de Células/efeitos dos fármacos , Proteína-Tirosina Quinases de Adesão Focal/análise , Proteína-Tirosina Quinases de Adesão Focal/química , Reação em Cadeia da Polimerase em Tempo Real
20.
Sci Rep ; 9(1): 13476, 2019 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-31530883

RESUMO

Treatment of large bone defects is a challenging clinical situation that may be benefited from cell therapies based on regenerative medicine. This study was conducted to evaluate the effect of local injection of bone marrow-derived mesenchymal stromal cells (BM-MSCs) or adipose tissue-derived MSCs (AT-MSCs) on the regeneration of rat calvarial defects. BM-MSCs and AT-MSCs were characterized based on their expression of specific surface markers; cell viability was evaluated after injection with a 21-G needle. Defects measuring 5 mm that were created in rat calvaria were injected with BM-MSCs, AT-MSCs, or vehicle-phosphate-buffered saline (Control) 2 weeks post-defect creation. Cells were tracked by bioluminescence, and 4 weeks post-injection, the newly formed bone was evaluated by µCT, histology, nanoindentation, and gene expression of bone markers. BM-MSCs and AT-MSCs exhibited the characteristics of MSCs and maintained their viability after passing through the 21-G needle. Injection of both BM-MSCs and AT-MSCs resulted in increased bone formation compared to that in Control and with similar mechanical properties as those of native bone. The expression of genes associated with bone formation was higher in the newly formed bone induced by BM-MSCs, whereas the expression of genes involved in bone resorption was higher in the AT-MSC group. Cell therapy based on local injection of BM-MSCs or AT-MSCs is effective in delivering cells that induced a significant improvement in bone healing. Despite differences observed in molecular cues between BM-MSCs and AT-MSCs, both cells had the ability to induce bone tissue formation at comparable amounts and properties. These results may drive new cell therapy approaches toward complete bone regeneration.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Animais , Biomarcadores , Regeneração Óssea , Diferenciação Celular , Sobrevivência Celular , Rastreamento de Células , Terapia Baseada em Transplante de Células e Tecidos/métodos , Imuno-Histoquímica , Imunofenotipagem , Masculino , Transplante de Células-Tronco Mesenquimais/métodos , Imagem Molecular , Osteogênese , Ratos , Resultado do Tratamento , Microtomografia por Raio-X
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA