Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 212
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Adv Healthc Mater ; 12(11): e2200976, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36808718

RESUMO

Bone autografts remain the gold standard for bone grafting surgeries despite having increased donor site morbidity and limited availability. Bone morphogenetic protein-loaded grafts represent another successful commercial alternative. However, the therapeutic use of recombinant growth factors has been associated with significant adverse clinical outcomes. This highlights the need to develop biomaterials that closely approximate the structure and composition of bone autografts, which are inherently osteoinductive and biologically active with embedded living cells, without the need for added supplements. Here, injectable growth factor-free bone-like tissue constructs are developed, that closely approximate the cellular, structural, and chemical composition of bone autografts. It is demonstrated that these micro-constructs are inherently osteogenic, and demonstrate the ability to stimulate mineralized tissue formation and regenerate bone in critical-sized defects in-vivo. Furthermore, the mechanisms that allow human mesenchymal stem cells (hMSCs) to be highly osteogenic in these constructs, despite the lack of osteoinductive supplements, are assessed, whereby Yes activated protein (YAP) nuclear localization and adenosine signaling appear to regulate osteogenic cell differentiation. The findings represent a step toward a new class of minimally invasive, injectable, and inherently osteoinductive scaffolds, which are regenerative by virtue of their ability to mimic the tissue cellular and extracellular microenvironment, thus showing promise for clinical applications in regenerative engineering.


Assuntos
Microgéis , Humanos , Regeneração Óssea/fisiologia , Osteogênese/fisiologia , Osso e Ossos , Materiais Biocompatíveis/química , Diferenciação Celular/fisiologia , Engenharia Tecidual , Alicerces Teciduais/química
2.
Int J Med Sci ; 18(16): 3692-3696, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34790041

RESUMO

Background: To evaluate hyperbaric oxygen therapy (HBOT) on infection rates and repair rates during the treatment of large jaw cysts. Methods: A prospective randomized, non-blinded, controlled clinical trial included 90 patients with jaw cysts, randomly divided into three groups. Patients were treated with enucleations and bone substitute was used in the experimental and control groups. The experimental group received HBOT. The primary predictor variable was HBOT. The infection rate, repair rate, preoperative volume of the jaw cysts, age, and sex were statistically analyzed. The Fisher exact test was used to compare the infection rate and postoperative complications. The repair rate of the bone defects was analyzed using the repeated-measures analysis of variance and the least significant difference tests. The Kendall's coefficient of concordance and Kappa statistics were calculated to evaluate the consistency between the two investigators. Results: The infection rate was 3.4% in the experimental group, 14.3% in the blank group, and 32.1% in the control group (P<0.05). The repair rate in the experimental group was significantly higher than in the control and blank groups at 1, 3 and 6 months after surgery (P<0.05). Conclusion: The results showed that HBOT reduced the postoperative infection rate following the enucleation of large jaw cysts with bone substitute filling, and it also improved the bone repair rate.


Assuntos
Oxigenoterapia Hiperbárica/métodos , Cistos Maxilomandibulares/terapia , Adolescente , Adulto , Idoso , Doenças Ósseas/complicações , Doenças Ósseas/terapia , Regeneração Óssea/fisiologia , Substitutos Ósseos/uso terapêutico , China , Feminino , Humanos , Infecções/complicações , Infecções/terapia , Cistos Maxilomandibulares/patologia , Masculino , Pessoa de Meia-Idade , Procedimentos de Cirurgia Plástica , Resultado do Tratamento , Carga Tumoral , Adulto Jovem
3.
Int J Mol Sci ; 22(17)2021 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-34502354

RESUMO

Oyster shells are rich in calcium, and thus, the potential use of waste shells is in the production of calcium phosphate (CaP) minerals for osteopathic biomedical applications, such as scaffolds for bone regeneration. Implanted scaffolds should stimulate the differentiation of induced pluripotent stem cells (iPSCs) into osteoblasts. In this study, oyster shells were used to produce nano-grade hydroxyapatite (HA) powder by the liquid-phase precipitation. Then, biphasic CaP (BCP) bioceramics with two different phase ratios were obtained by the foaming of HA nanopowders and sintering by two different two-stage heat treatment processes. The different sintering conditions yielded differences in structure and morphology of the BCPs, as determined by scanning electron microscopy (SEM), X-ray diffraction (XRD), and Brunauer-Emmett-Teller (BET) surface area analysis. We then set out to determine which of these materials were most biocompatible, by co-culturing with iPSCs and examining the gene expression in molecular pathways involved in self-renewal and differentiation of iPSCs. We found that sintering for a shorter time at higher temperatures gave higher expression levels of markers for proliferation and (early) differentiation of the osteoblast. The differences in biocompatibility may be related to a more hierarchical pore structure (micropores within macropores) obtained with briefer, high-temperature sintering.


Assuntos
Exoesqueleto/química , Hidroxiapatitas/química , Células-Tronco Pluripotentes Induzidas/metabolismo , Exoesqueleto/metabolismo , Animais , Materiais Biocompatíveis/química , Regeneração Óssea/fisiologia , Fosfatos de Cálcio/química , Adesão Celular/fisiologia , Diferenciação Celular/efeitos dos fármacos , Cerâmica/química , Humanos , Hidroxiapatitas/síntese química , Hidroxiapatitas/metabolismo , Hidroxiapatitas/farmacologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Osteoblastos/citologia , Osteogênese/efeitos dos fármacos , Osteogênese/fisiologia , Ostreidae/metabolismo , Porosidade/efeitos dos fármacos , Engenharia Tecidual/métodos , Alicerces Teciduais/química
4.
Int J Biol Sci ; 17(7): 1821-1836, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33994865

RESUMO

Both osteoblasts and preosteoclasts contribute to the coupling of osteogenesis and angiogenesis, regulating bone regeneration. Astragaloside IV (AS-IV), a glycoside of cycloartane-type triterpene derived from the Chinese herb Astragalus membranaceus, exhibits various biological activities, including stimulating angiogenesis and attenuating ischemic-hypoxic injury. However, the effects and underlying mechanisms of AS-IV in osteogenesis, osteoclastogenesis, and bone regeneration remain poorly understood. In the present study, we found that AS-IV treatment inhibited osteoclastogenesis, preserved preosteoclasts, and enhanced platelet-derived growth factor-BB (PDGF-BB)-induced angiogenesis. Additionally, AS-IV promoted cell viability, osteogenic differentiation, and angiogenic gene expression in bone marrow mesenchymal stem cells (BMSCs). The activation of AKT/GSK-3ß/ß-catenin signaling was found to contribute to the effects of AS-IV on osteoclastogenesis and osteogenesis. Furthermore, AS-IV accelerated bone regeneration during distraction osteogenesis (DO), as evidenced from the improved radiological and histological manifestations and biomechanical parameters, accompanied by enhanced angiogenesis within the distraction zone. In summary, AS-IV accelerates bone regeneration during DO, by enhancing osteogenesis and preosteoclast-induced angiogenesis simultaneously, partially through AKT/GSK-3ß/ß-catenin signaling. These findings reveal that AS-IV may serve as a potential bioactive molecule for promoting the coupling of osteogenesis and angiogenesis, and imply that AKT/GSK-3ß/ß-catenin signaling may be a promising therapeutic target for patients during DO treatment.


Assuntos
Medula Óssea/metabolismo , Regeneração Óssea/fisiologia , Neovascularização Fisiológica/fisiologia , Osteoblastos/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Saponinas/farmacologia , Triterpenos/farmacologia , Animais , Medula Óssea/efeitos dos fármacos , Regeneração Óssea/efeitos dos fármacos , Proliferação de Células , Células Cultivadas , Medicamentos de Ervas Chinesas , Masculino , Modelos Animais , Osteoblastos/citologia , Osteogênese/fisiologia , Ratos , Ratos Sprague-Dawley
5.
J Biomed Mater Res A ; 109(10): 1840-1848, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33797182

RESUMO

Microbeads consisting of pullulan and dextran supplemented with hydroxyapatite have recently been developed for bone tissue engineering applications. Here, we evaluate the bone formation in two different preclinical models after injection of microbeads reconstituted with either saline buffer or autologous blood. Addition of saline solution or autologous blood to dried microbeads packaged into syringes allowed an easy injection. In the first rat bone defect model performed in the femoral condyle, microcomputed tomography performed after 30 and 60 days revealed an important mineralization process occurring around and within the core of the microbeads in both conditions. Bone volume/total volume measurements revealed no significant differences between the saline solution and the autologous blood groups. Histologically, osteoid tissue was evidenced around and in contact of the microbeads in both conditions. Using the sinus lift model performed in sheep, cone beam computed tomography revealed an important mineralization inside the sinus cavity for both groups after 3 months of implantation. Representative Masson trichrome staining images showed that bone formation occurs at the periphery and inside the microbeads in both conditions. Quantitative evaluation of the new bone formation displayed no significant differences between groups. In conclusion, reconstitution of microbeads with autologous blood did not enhance the regenerative capacity of these microbeads compared to the saline buffer group. This study is of particular interest for clinical applications in oral and maxillofacial surgery.


Assuntos
Sangue/metabolismo , Regeneração Óssea/fisiologia , Osso e Ossos/patologia , Osso e Ossos/fisiopatologia , Durapatita/farmacologia , Polímeros/farmacologia , Solução Salina/farmacologia , Animais , Modelos Animais de Doenças , Feminino , Implantes Experimentais , Microesferas , Ratos , Ovinos , Transplante Autólogo , Microtomografia por Raio-X
6.
J Biomed Mater Res A ; 109(9): 1765-1775, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33733598

RESUMO

In general, bone fractures are able of healing by itself. However, in critical situations such as large bone defects, poor blood supply or even infections, the biological capacity of repair can be impaired, resulting in a delay of the consolidation process or even in non-union fractures. Thus, technologies able of improving the process of bone regeneration are of high demand. In this context, ceramic biomaterials-based bone substitutes and photobiomodulation (PBM) have been emerging as promising alternatives. Thus, the present study performed a systematic review targeting to analyze studies in the literature which investigated the effects of the association of ceramic based bone substitutes and PBM in the process of bone healing using animal models of bone defects. The search was conducted from March and April of 2019 in PubMed, Web of Science and Scopus databases. After the eligibility analyses, 16 studies were included in this review. The results showed that the most common material used was hydroxyapatite (HA) followed by Biosilicate associated with infrared PBM. Furthermore, 75% of the studies demonstrated positive effects to stimulate bone regeneration from association of ceramic biomaterials and PBM. All studies used low-level laser therapy (LLLT) device and the most studies used LLLT infrared. The evidence synthesis was moderate for all experimental studies for the variable histological analysis demonstrating the efficacy of techniques on the process of bone repair stimulation. In conclusion, this review demonstrates that the association of ceramic biomaterials and PBM presented positive effects for bone repair in experimental models of bone defects.


Assuntos
Regeneração Óssea/fisiologia , Substitutos Ósseos/farmacologia , Terapia com Luz de Baixa Intensidade , Animais , Materiais Biocompatíveis/farmacologia , Regeneração Óssea/efeitos dos fármacos , Regeneração Óssea/efeitos da radiação
7.
J Biomed Mater Res B Appl Biomater ; 109(4): 538-547, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-32915522

RESUMO

Extracellular matrix (ECM) products have the potential to improve cellular attachment and promote tissue-specific development by mimicking the native cellular niche. In this study, the therapeutic efficacy of an ECM substratum produced by bone marrow stem cells (BM-MSCs) to promote bone regeneration in vitro and in vivo were evaluated. Fluorescence-activated cell sorting analysis and phenotypic expression were employed to characterize the in vitro BM-MSC response to bone marrow specific ECM (BM-ECM). BM-ECM encouraged cell proliferation and stemness maintenance. The efficacy of BM-ECM as an adjuvant in promoting bone regeneration was evaluated in an orthotopic, segmental critical-sized bone defect in the rat femur over 8 weeks. The groups evaluated were either untreated (negative control); packed with calcium phosphate granules or granules+BM-ECM free protein and stabilized by collagenous membrane. Bone regeneration in vivo was analyzed using microcomputed tomography and histology. in vivo results demonstrated improvements in mineralization, osteogenesis, and tissue infiltration (114 ± 15% increase) in the BM-ECM complex group from 4 to 8 weeks compared to mineral granules only (45 ± 21% increase). Histological observations suggested direct apposition of early bone after 4 weeks and mineral consolidation after 8 weeks implantation for the group supplemented with BM-ECM. Significant osteoid formation and greater functional bone formation (polar moment of inertia was 71 ± 0.2 mm4 with BM-ECM supplementation compared to 48 ± 0.2 mm4 in untreated defects) validated in vivo indicated support of osteoconductivity and increased defect site cellularity. In conclusion, these results suggest that BM-ECM free protein is potentially a therapeutic supplement for stemness maintenance and sustaining osteogenesis.


Assuntos
Regeneração Óssea/efeitos dos fármacos , Proteínas da Matriz Extracelular/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Animais , Regeneração Óssea/fisiologia , Calcificação Fisiológica/efeitos dos fármacos , Fosfatos de Cálcio/farmacologia , Colágeno/uso terapêutico , Fêmur/diagnóstico por imagem , Fêmur/lesões , Fêmur/fisiologia , Técnicas In Vitro , Teste de Materiais , Especificidade de Órgãos , Osteogênese/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Microtomografia por Raio-X
8.
Int J Biol Macromol ; 165(Pt A): 1346-1360, 2020 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-33038401

RESUMO

Biogenic bioceramics scaffolds are receiving considerable attention for bone restoration applications. Compared with scaffolds of chemical origin, biogenic scaffolds exhibit greater biocompatibility and enhanced bioactive features. In the present study, porous biogenic hydroxyapatite (bHA) was prepared via a polymeric infiltration route and was subsequently coated with alginate to produce alginate/biogenic hydroxyapatite (Alg/bHA) composites. Alginate was used to enhance the mechanical properties as well as the bioactivity and biodegradability of the HA scaffolds. A coating of 3%w/v alginate applied for 10 min was found to result in the best coating for the HA porous scaffolds. The in vitro study demonstrated that the prepared composites had acceptable bioactivity and biodegradability characteristics. The histological study in femur bone of rats indicated that the 3Alg/HA scaffolds capable of supporting both endochondral and intramembranous bone formation. The defect was fully regenerated and mostly filled with the mature lamellar bone after 6 months, with Ca/P atomic ratio similar to the rat's normal bone. The studied scaffolds provide a promising therapeutic option to enhance local bone healing because they do not damage liver or kidney functions and do not induce carcinogenic or inflammatory effects. Accordingly, 3Alg/HA scaffolds are recommended for the tissue engineering applications.


Assuntos
Óxido de Alumínio/farmacologia , Desenvolvimento Ósseo/efeitos dos fármacos , Regeneração Óssea/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Alginatos/farmacologia , Óxido de Alumínio/química , Animais , Regeneração Óssea/fisiologia , Osso e Ossos , Durapatita/farmacologia , Fêmur/efeitos dos fármacos , Fêmur/crescimento & desenvolvimento , Humanos , Polímeros/química , Polímeros/farmacologia , Porosidade , Ratos , Engenharia Tecidual , Alicerces Teciduais/química , Terapia Tecidual Histórica/métodos
9.
Theranostics ; 10(25): 11837-11861, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33052249

RESUMO

Recently, the rapid development of biomaterials has induced great interest in the precisely targeted treatment of bone-related diseases, including bone cancers, infections, and inflammation. Realizing noninvasive therapeutic effects, as well as improving bone tissue regeneration, is essential for the success of bone­related disease therapies. In recent years, researchers have focused on the development of stimuli-responsive strategies to treat bone-related diseases and to realize bone regeneration. Among the various external stimuli for targeted therapy, near infrared (NIR) light has attracted considerable interests due to its high tissue penetration capacity, minimal damage toward normal tissues, and easy remote control properties. The main objective of this systematic review was to reveal the current applications of NIR light-assisted phototherapy for bone-related disease treatment and bone tissue regeneration. Database collection was completed by June 1, 2020, and a total of 81 relevant studies were finally included. We outlined the various therapeutic applications of photothermal, photodynamic and photobiomodulation effects under NIR light irradiation for bone­related disease treatment and bone regeneration, based on the retrieved literatures. In addition, the advantages and promising applications of NIR light-responsive drug delivery systems for spatiotemporal-controlled therapy were summarized. These findings have revealed that NIR light-assisted phototherapy plays an important role in bone-related disease treatment and bone tissue regeneration, with significant promise for further biomedical and clinical applications.


Assuntos
Doenças Ósseas/terapia , Regeneração Óssea/efeitos da radiação , Raios Infravermelhos/uso terapêutico , Terapia com Luz de Baixa Intensidade/métodos , Fotoquimioterapia/métodos , Terapia Fototérmica/métodos , Animais , Doenças Ósseas/fisiopatologia , Regeneração Óssea/efeitos dos fármacos , Regeneração Óssea/fisiologia , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/fisiopatologia , Osso e Ossos/efeitos da radiação , Ensaios Clínicos como Assunto , Modelos Animais de Doenças , Sistemas de Liberação de Medicamentos/métodos , Humanos , Nanopartículas/administração & dosagem , Resultado do Tratamento
10.
Int J Mol Sci ; 21(12)2020 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-32630456

RESUMO

Jellyfish collagen, which can be defined as "collagen type 0" due to its homogeneity to the mammalian types I, II, III, V, and IX and its batch-to-batch consistent producibility, is of special interest for different medical applications related to (bone) tissue regeneration as an alternative to mammalian collagen-based biomaterials. However, no in vivo studies regarding the induction of M1- and M2-macrophages and their time-dependent ration as well as the analysis of the bone regeneration capacity of jellyfish collagen scaffolds have been conducted until now. Thus, the goal of this study was to determine the nature of the immune response to jellyfish collagen scaffolds and their bone healing capacities. Two in vivo studies using established implantation models, i.e., the subcutaneous and the calvarian implantation model in Wistar rats, were conducted. Furthermore, specialized histological, histopathological, and histomorphometrical methods have been used. As a control biomaterial, a collagen scaffold, originating from porcine pericardium, which has already been stated as biocompatible, was used for the subcutaneous study. The results of the present study show that jellyfish collagen scaffolds are nearly completely resorbed until day 60 post implantation by stepwise integration within the subcutaneous connective tissue mediated mainly by macrophages and single multinucleated giant cells. Interestingly, the degradation process ended in a vessel rich connective tissue that is understood to be an optimal basis for tissue regeneration. The study results showed an overall weaker immune response to jellyfish collagen than to porcine pericardium matrices by the induction of significantly lower numbers of macrophages together with a more balanced occurrence of M1- and M2-macrophages. However, both collagen-based biomaterials induced balanced numbers of both macrophage subtypes, which supports their good biocompatibility. Moreover, the histomorphometrical results for the calvarial implantation of the jellyfish scaffolds revealed an average of 46.20% de novo bone formation at day 60, which was significantly higher compared to the control group. Thereby, the jellyfish collagen scaffolds induced also significantly higher numbers of anti-inflammatory macrophages within the bony implantation beds. Altogether, the results show that the jellyfish collagen scaffolds allowed for a directed integration behavior, which is assumed to be in accordance with the concept of Guided Bone Regeneration (GBR). Furthermore, the jellyfish collagen scaffolds induced a long-term anti-inflammatory macrophage response and an optimal vascularization pattern within their implant beds, thus showing excellent biocompatibility and (bone) tissue healing properties.


Assuntos
Regeneração Óssea/fisiologia , Colágeno/metabolismo , Engenharia Tecidual/métodos , Animais , Materiais Biocompatíveis/metabolismo , Regeneração Óssea/genética , Osso e Ossos/imunologia , Osso e Ossos/metabolismo , Colágeno/imunologia , Imunidade , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Osteogênese/imunologia , Osteogênese/fisiologia , Ratos , Ratos Wistar , Cifozoários/metabolismo , Alicerces Teciduais , Cicatrização/fisiologia
11.
Int J Nanomedicine ; 15: 2045-2058, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32273701

RESUMO

Bone regeneration remains a great clinical challenge. Two-dimensional materials, especially graphene and its derivative graphene oxide, have been widely used for bone regeneration. Since its discovery in 2014, black phosphorus (BP) nanomaterials including BP nanosheets and BP quantum dots have attracted considerable scientific attention and are considered as prospective graphene substitutes. BP nanomaterials exhibit numerous advantages such as excellent optical and mechanical properties, electrical conductivity, excellent biocompatibility, and good biodegradation, all of which make them particularly attractive in biomedicine. In this review, we comprehensively summarize recent advances of BP-based nanomaterials in bone regeneration. The advantages are reviewed, the different synthesis methods of BP are summarized, and the applications to promote bone regeneration are highlighted. Finally, the existing challenges and perspectives of BP in bone regeneration are briefly discussed.


Assuntos
Regeneração Óssea/fisiologia , Nanoestruturas/química , Nanoestruturas/uso terapêutico , Fósforo/química , Animais , Materiais Biocompatíveis/química , Materiais Biocompatíveis/uso terapêutico , Regeneração Óssea/efeitos dos fármacos , Grafite/química , Humanos , Pontos Quânticos/química
12.
Int J Pharm ; 580: 119213, 2020 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-32165229

RESUMO

In-situ forming implants receive great attention for repairing serious bone injuries. The aim of the present study was to prepare novel chitosan in-situ forming implants (CIFI) loaded with bioactive glass nanoparticles and/or raloxifene hydrochloride (RLX). Incorporating raloxifene hydrochloride (RLX) as a selective estrogen receptor modulator was essential to make use of its anti-resorptive properties. The prepared formulae were tested for their in-vitro gelation time, drug release, injectability, rheological properties, erosion rate and morphological properties. Results revealed that the formulation composed of 1% (w/v) chitosan with 2% (w/v) NaHCO3 and 1% (w/v) bioactive glass nanoparticles (CIFI-BG) possessed the most sustained drug release profile which extended over four months with low burst release effect compared to the same formulation lacking bioactive glass nanoparticles (CIFI). Selected formulations were tested for their ability to enhance bone regeneration in induced puncture in rate tibia. Results declared that these formulations were able to enhance bone regeneration after 12 weeks in comparison to the untreated tibial punctures and that containing bioactive glass could be considered as novel approach for treatment of serious bone injuries which require long term treatment and internal mechanical bone support during healing.


Assuntos
Conservadores da Densidade Óssea/síntese química , Quitosana/síntese química , Composição de Medicamentos/métodos , Nanopartículas/química , Cloridrato de Raloxifeno/síntese química , Tíbia/efeitos dos fármacos , Animais , Conservadores da Densidade Óssea/administração & dosagem , Conservadores da Densidade Óssea/metabolismo , Regeneração Óssea/efeitos dos fármacos , Regeneração Óssea/fisiologia , Quitosana/administração & dosagem , Quitosana/metabolismo , Modelos Animais de Doenças , Portadores de Fármacos/administração & dosagem , Portadores de Fármacos/química , Portadores de Fármacos/metabolismo , Avaliação Pré-Clínica de Medicamentos/métodos , Implantes de Medicamento/administração & dosagem , Implantes de Medicamento/síntese química , Implantes de Medicamento/metabolismo , Vidro/química , Masculino , Nanopartículas/administração & dosagem , Nanopartículas/metabolismo , Cloridrato de Raloxifeno/administração & dosagem , Cloridrato de Raloxifeno/metabolismo , Ratos , Ratos Sprague-Dawley , Tíbia/lesões , Tíbia/metabolismo , Resultado do Tratamento
13.
Biomed Mater Eng ; 30(5-6): 475-486, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31771032

RESUMO

BACKGROUND: We have developed a technology to electrically polarize living bone. OBJECTIVE: The effects of stored electrical charge in electrical polarized bone on the facilitation of new bone formation were assayed. METHODS: Stimulated depolarized current measurement was performed in electrically polarized and nonpolarized femora of SD rats. These bone specimens were implanted into bone defects of the rat femora and fixed with a custom-made external fixator. X-ray imaging of the implant was performed every week. After 3 weeks, micro-CT scanning was performed to evaluate the displacement rate. Histological observation was performed, and the occupancy ratio of the newly formed bone was calculated from tissue specimens stained with Villanueva's Goldner method. RESULTS: There was a tendency for the displacement rate of the implant to be smaller and the occupancy ratio of the newly formed bone to be larger, especially at the distal end, in the polarized group compared with the nonpolarized group. The time of callus appearance was significantly earlier in the polarized group than in the nonpolarized group, and bridging callus grew from the distal to the proximal end. CONCLUSIONS: Bone specimens can be electrically polarized, and the stored electrical charge can work effectively to facilitate new bone formation.


Assuntos
Terapia por Estimulação Elétrica , Fixadores Externos , Fraturas do Fêmur/terapia , Implantes Experimentais , Animais , Temperatura Corporal/fisiologia , Regeneração Óssea/fisiologia , Modelos Animais de Doenças , Estimulação Elétrica , Terapia por Estimulação Elétrica/instrumentação , Terapia por Estimulação Elétrica/métodos , Eletricidade , Fraturas do Fêmur/patologia , Masculino , Osteogênese/fisiologia , Medicina de Precisão/instrumentação , Ratos , Ratos Sprague-Dawley , Resultado do Tratamento
14.
Injury ; 50(11): 1853-1867, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31585673

RESUMO

BACKGROUND: Photobiomodulation therapy (PBMT) using low-level laser influences the release of several growth factors involved in the formation of epithelial cells, fibroblasts, collagen and vascular proliferation, besides accelerating the synthesis of bone matrix due to the increased vascularization and lower inflammatory response, with significant increase of osteocytes in the irradiated bone. Considering its properties, beneficial effects and clinical relevance, the aim of this review was to analyze the scientific literature regarding the use of PBMT in the process of bone defect repair. METHODS: Electronic search was carried out in PubMed/MEDLINEⓇ and Web of Science databases with combination of the descriptors low-level laser therapy AND bone repair, considering the period of publication until the year 2018. RESULTS: The literature search identified 254 references in PubMed/MEDLINE and 204 in Web of Science, of which 33 and 4 were selected, respectively, in accordance with the eligibility requirements. The analysis of researches showed articles using PBMT in several places of experimentation in the subjects, different types of associated biomaterials, stimulatory effects on cell proliferation, besides variations in the parameters of use of laser therapy, mainly in relation to the wavelength and density of energy. Only four articles reported that the laser did not improve the osteogenic properties of a biomaterial. CONCLUSIONS: Many studies have shown that PBMT has positive photobiostimulatory effects on bone regeneration, accelerating its process regardless of parameters and the use of biomaterials. However, standardization of its use is still imperfect and should be better studied to allow correct application concerning the utilization protocols.


Assuntos
Regeneração Óssea/efeitos da radiação , Proliferação de Células/efeitos da radiação , Consolidação da Fratura/efeitos da radiação , Fraturas Ósseas/radioterapia , Terapia com Luz de Baixa Intensidade , Osteogênese/efeitos da radiação , Regeneração Óssea/fisiologia , Colágeno/metabolismo , Fibroblastos/metabolismo , Consolidação da Fratura/fisiologia , Fraturas Ósseas/fisiopatologia , Humanos , Terapia com Luz de Baixa Intensidade/métodos , Osteogênese/fisiologia
15.
J Mater Sci Mater Med ; 30(8): 87, 2019 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-31325047

RESUMO

Previous studies have demonstrated the significant roles of simvastatin (SVA) and oxysterols in the osteogenesis process. In this study, we evaluate the effect of a combination of SVA and 20(S)-hydroxycholesterol (20(S)OHC) on the cell viability and osteogenic differentiation of bone marrow stromal cells (BMSCs). After treatment with a control vehicle, SVA (0.025, 0.10, 0.25 or 1.0 µM), 20(S)OHC (5 µM), or a combination of both (0.25 µM SVA + 5 µM 20(S)OHC), the proliferation, apoptosis, ALP activity, mineralization, osteogenesis-related gene expression and Raf/MEK/ERK signaling activity in BMSCs were measured. Our results showed that high concentrations of SVA (0.25 and 1.0 µM) enhanced osteogenesis-related genes expression while attenuating cell viability. The addition of 5 µM 20(S)OHC induced significantly higher proliferative activity, which neutralized the inhibitory effect of SVA on the viability of BMSCs. Moreover, compared to supplementation with only one of the additives, combined supplementation with both SVA and 20(S)OHC induced significantly enhanced ALP activity, calcium sedimentation, osteogenesis-related genes (ALP, OCN and BMP-2) expression and Raf/MEK/ERK signaling activity in BMSCs; these enhancements were attenuated by treatment with the inhibitor U0126, indicating a significant role of Raf/MEK/ERK signaling in mediating the synergistically enhanced osteogenic differentiation of BMSCs by combined SVA and 20(S)OHC treatment. Additionally, histological examination confirmed a synergistic effect of SVA and 20(S)OHC on enhancing bone regeneration in a rabbit calvarial defect model. This newly developed SVA/20(S)OHC formulation may be used as an osteoinductive drug to enhance bone healing.


Assuntos
Regeneração Óssea/efeitos dos fármacos , Hidroxicolesteróis/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Sinvastatina/farmacologia , Fosfatase Alcalina/metabolismo , Animais , Regeneração Óssea/fisiologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Sinergismo Farmacológico , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Células-Tronco Mesenquimais/fisiologia , Osteogênese/genética , Coelhos , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Quinases raf/metabolismo
16.
ACS Appl Mater Interfaces ; 11(19): 17234-17246, 2019 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-31008576

RESUMO

Due to the great similarity to the natural extracellular matrix and minimally invasive surgeries, injectable hydrogels are appealing biomaterials in cartilage and bone tissue engineering. Nevertheless, undesirable mechanical properties and bioactivity greatly hamper their availability in clinic applications. Here, we developed an injectable nanocomposite hydrogel by in situ growth of CaP nanoparticles (ICPNs) during the free-radical polymerization of dimethylaminoethyl methacrylate (DMAEMA) and 2-hydroxyethyl methacrylate (HEMA) matrix (PDH) for bone regeneration. The ICPNs are self-assembled by incorporation of poly-l-glutamic acid (PGA) with abundant carboxyl functional groups during the formation of carboxyl-Ca2+ coordination and further CaP precipitation. Furthermore, the carboxyl groups of PGA could interact with the tertiary amines of DMAEMA fragments and thus improve the mechanical strength of hydrogels. Upon mixing solutions of DMAEMA and HEMA bearing PGA, Ca2+, and PO43-, this effective and dynamic coordination led to the rapid self-assembly of CaP NPs and PDH nanocomposite hydrogels (PDH/mICPN). The obtained optimal nanocomposite hydrogels exhibited suitable injectable time, an enhanced tensile strength of 321.1 kPa, and a fracture energy of 29.0 kJ/m2 and dramatically facilitated cell adhesion and upregulated osteodifferentiation compared to hydrogels prepared by blending ex situ prefabricated CaP NPs. In vivo experiments confirmed the promoted osteogenesis, which shows a striking contrast to pure PDH hydrogels. Additionally, the methacrylate groups on the monomers could easily be functionalized with aptamers and thereby facilitate recognition and capturing of bone marrow stromal cells both in vitro and in vivo and strengthen the bone regeneration. We believe that our conducted research about in situ self-assembled CaP nanoparticle-coordinated hydrogels will open a new avenue for bone regeneration in the future endeavors.


Assuntos
Regeneração Óssea/efeitos dos fármacos , Hidrogéis/farmacologia , Osteogênese/efeitos dos fármacos , Engenharia Tecidual , Animais , Materiais Biocompatíveis/síntese química , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Regeneração Óssea/fisiologia , Cálcio/química , Cartilagem/efeitos dos fármacos , Cartilagem/crescimento & desenvolvimento , Sobrevivência Celular/efeitos dos fármacos , Matriz Extracelular/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Humanos , Hidrogéis/síntese química , Hidrogéis/química , Células-Tronco Mesenquimais/efeitos dos fármacos , Metacrilatos/química , Metacrilatos/farmacologia , Nanopartículas/química , Fósforo/química , Ratos
17.
J Appl Oral Sci ; 27: e20180103, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30994771

RESUMO

OBJECTIVE: This study aimed to evaluate the inflammatory effect and bone formation in sterile surgical failures after implantation of a collagen sponge with mesenchymal stem cells from human dental pulp (hDPSCs) and Aloe vera. MATERIAL AND METHODS: Rattus norvegicus (n=75) were divided into five experimental groups according to treatment: G1) control (blood clot); G2) Hemospon®; G3) Hemospon® in a culture medium enriched with 8% Aloe vera; G4) Hemospon® in a culture medium containing hDPSCs and G5) Hemospon® in a culture medium enriched with 8% Aloe vera and hDPSCs. On days 7, 15 and 30, the animals were euthanized, and the tibia was dissected for histological, immunohistochemistry and immunofluorescence analyses. The results were analyzed using nonparametric Kruskal-Wallis test and Dunn's post-test. RESULTS: On days 7 and 15, the groups with Aloe vera had less average acute inflammatory infiltrate compared to the control group and the group with Hemospon® (p<0.05). No statistically significant difference was found between the groups regarding bone formation at the three experimental points in time. Osteopontin expression corroborated the intensity of bone formation. Fluorescence microscopy revealed positive labeling with Q-Tracker® in hDPSCs before transplantation and tissue repair. CONCLUSION: The results suggest that the combination of Hemospon®, Aloe vera and hDPSCs is a form of clinical treatment for the repair of non-critical bone defects that reduces the inflammatory cascade's effects.


Assuntos
Aloe/química , Regeneração Óssea/efeitos dos fármacos , Regeneração Óssea/fisiologia , Polpa Dentária/citologia , Transplante de Células-Tronco Mesenquimais/métodos , Extratos Vegetais/farmacologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Colágeno/farmacologia , Citometria de Fluxo , Hemostáticos/farmacologia , Humanos , Imuno-Histoquímica , Masculino , Microscopia de Fluorescência , Osteogênese/efeitos dos fármacos , Osteogênese/fisiologia , Osteopontina/análise , Ratos , Reprodutibilidade dos Testes , Tíbia/efeitos dos fármacos , Tíbia/patologia , Tíbia/fisiologia , Fatores de Tempo , Resultado do Tratamento
18.
Prog Biophys Mol Biol ; 149: 147-159, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31002851

RESUMO

Photobiomodulation (PBM) encompasses a light application aimed to increase healing process, tissue regeneration, and reducing inflammation and pain. PBM is specifically aimed to modify the expression of cellular molecules; however, PBM impacts on cellular and molecular pathways especially in bone regenerative medicine have been investigated in scattered different studies. The purpose of the current study is to systematically review evidence on molecular impact of PBM on bone regeneration. A comprehensive electronic search in Medline, Scopus, EMBASE, EBSCO, Cochrane library, web of science, and google scholar was conducted from January 1975 to October 2018 limited to English language publications on administrations of photobiomodulation for bone regeneration which evaluated biological factors. In addition, hand search of selected journals was done to retrieve all articles. This systematic review was performed based on PRISMA guideline. Among these studies, five articles reported in vitro results, twelve articles were in vivo, and three of them were clinical trials. The data tabulated according to the type of markers (osteogenic markers, angiogenic markers, growth factors, and inflammation mediators). PBM's effects depend on many parameters which energy density is more important than the others. PBM can significantly enhance expression of osteocalcin, collagen, RUNX-2, vascular endothelial growth factor, bone morphogenic proteins, and COX-2. Although since the heterogeneity of the studies and their limitations, an evidence-based decision for definite therapeutic application of PBM is still unattainable, the findings of our review can help other researchers to ameliorate their study design and elect more efficient approach for their investigation.


Assuntos
Regeneração Óssea/fisiologia , Terapia com Luz de Baixa Intensidade/métodos , Osteogênese/fisiologia , Biomarcadores/metabolismo , Proliferação de Células , Colágeno/metabolismo , Subunidades alfa de Fatores de Ligação ao Core/metabolismo , Células-Tronco Mesenquimais/metabolismo , Osteocalcina/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
19.
Int Orthop ; 43(3): 539-551, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30645684

RESUMO

INTRODUCTION: Biophysical stimulation is a non-invasive therapy used in orthopaedic practice to increase and enhance reparative and anabolic activities of tissue. METHODS: A sistematic web-based search for papers was conducted using the following titles: (1) pulsed electromagnetic field (PEMF), capacitively coupled electrical field (CCEF), low intensity pulsed ultrasound system (LIPUS) and biophysical stimulation; (2) bone cells, bone tissue, fracture, non-union, prosthesis and vertebral fracture; and (3) chondrocyte, synoviocytes, joint chondroprotection, arthroscopy and knee arthroplasty. RESULTS: Pre-clinical studies have shown that the site of interaction of biophysical stimuli is the cell membrane. Its effect on bone tissue is to increase proliferation, synthesis and release of growth factors. On articular cells, it creates a strong A2A and A3 adenosine-agonist effect inducing an anti-inflammatory and chondroprotective result. In treated animals, it has been shown that the mineralisation rate of newly formed bone is almost doubled, the progression of the osteoarthritic cartilage degeneration is inhibited and quality of cartilage is preserved. Biophysical stimulation has been used in the clinical setting to promote the healing of fractures and non-unions. It has been successfully used on joint pathologies for its beneficial effect on improving function in early OA and after knee surgery to limit the inflammation of periarticular tissues. DISCUSSION: The pooled result of the studies in this review revealed the efficacy of biophysical stimulation for bone healing and joint chondroprotection based on proven methodological quality. CONCLUSION: The orthopaedic community has played a central role in the development and understanding of the importance of the physical stimuli. Biophysical stimulation requires care and precision in use if it is to ensure the success expected of it by physicians and patients.


Assuntos
Doenças Ósseas/terapia , Doenças das Cartilagens/terapia , Terapia por Estimulação Elétrica/métodos , Fraturas Ósseas/terapia , Magnetoterapia/métodos , Animais , Doenças Ósseas/metabolismo , Doenças Ósseas/patologia , Regeneração Óssea/fisiologia , Regeneração Óssea/efeitos da radiação , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Osso e Ossos/efeitos da radiação , Cartilagem/metabolismo , Cartilagem/patologia , Cartilagem/efeitos da radiação , Doenças das Cartilagens/metabolismo , Doenças das Cartilagens/patologia , Condrócitos/metabolismo , Condrócitos/patologia , Condrócitos/efeitos da radiação , Terapia por Estimulação Elétrica/tendências , Fraturas Ósseas/metabolismo , Fraturas Ósseas/patologia , Humanos , Magnetoterapia/tendências
20.
Biomaterials ; 193: 1-11, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30550998

RESUMO

Mild heat stimulation can promote the restoration of bone defects but unfortunately, the delivery of exo-hyperthermy into human body is not efficient enough. In this study, mild heat-induced osteogenesis with high efficacy is demonstrated on an osteoimplant composed of black phosphorus nanosheets and poly(lactic-co-glycolic acid) (BPs@PLGA) with the participation of near-infrared (NIR) light irradiation. BPs@PLGA with only 0.2 wt% BPs show the highly-efficient NIR photothermal response even when being covered by a biological tissue as thick as 7 mm. In addition, this composite is completely biodegradable and the final degradation products are harmless H2O, CO2 and PO43- which can serve as necessary bone ingredient. The BPs@PLGA specimen mediated by low intensity and periodic NIR irradiation can effectively up-regulate the expressions of heat shock proteins and finally promote osteogenesis in vitro and in vivo. Boasting good biodegradability and NIR-mediated osteogenetic performances, the BPs@PLGA implant has great potential in orthopedic applications and this study provides new insights into the design and fabrication of new-style osteoimplants which can be remotely controlled.


Assuntos
Regeneração Óssea/fisiologia , Regeneração Óssea/efeitos da radiação , Raios Infravermelhos , Fósforo/química , Fototerapia/métodos , Animais , Proteínas de Choque Térmico/metabolismo , Humanos , Engenharia Tecidual/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA