Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 5.988
Filtrar
1.
Int J Implant Dent ; 10(1): 35, 2024 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-38967690

RESUMO

Considering the biological activity of osteoblasts is crucial when devising new approaches to enhance the osseointegration of implant surfaces, as their behavior profoundly influences clinical outcomes. An established inverse correlation exists between osteoblast proliferation and their functional differentiation, which constrains the rapid generation of a significant amount of bone. Examining the surface morphology of implants reveals that roughened titanium surfaces facilitate rapid but thin bone formation, whereas smooth, machined surfaces promote greater volumes of bone formation albeit at a slower pace. Consequently, osteoblasts differentiate faster on roughened surfaces but at the expense of proliferation speed. Moreover, the attachment and initial spreading behavior of osteoblasts are notably compromised on microrough surfaces. This review delves into our current understanding and recent advances in nanonodular texturing, meso-scale texturing, and UV photofunctionalization as potential strategies to address the "biological dilemma" of osteoblast kinetics, aiming to improve the quality and quantity of osseointegration. We discuss how these topographical and physicochemical strategies effectively mitigate and even overcome the dichotomy of osteoblast behavior and the biological challenges posed by microrough surfaces. Indeed, surfaces modified with these strategies exhibit enhanced recruitment, attachment, spread, and proliferation of osteoblasts compared to smooth surfaces, while maintaining or amplifying the inherent advantage of cell differentiation. These technology platforms suggest promising avenues for the development of future implants.


Assuntos
Implantes Dentários , Osseointegração , Osteoblastos , Propriedades de Superfície , Osteoblastos/fisiologia , Osteoblastos/citologia , Humanos , Diferenciação Celular , Proliferação de Células , Titânio/química , Osteogênese/fisiologia
2.
Biomater Adv ; 163: 213939, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-38954876

RESUMO

The bone turnover capability influences the acquisition and maintenance of osseointegration. The architectures of osteocyte three-dimensional (3D) networks determine the direction and activity of bone turnover through osteocyte intercellular crosstalk, which exchanges prostaglandins through gap junctions in response to mechanical loading. Titanium nanosurfaces with anisotropically patterned dense nanospikes promote the development of osteocyte lacunar-canalicular networks. We investigated the effects of titanium nanosurfaces on intercellular network development and regulatory capabilities of bone turnover in osteocytes under cyclic compressive loading. MLO-Y4 mouse osteocyte-like cell lines embedded in type I collagen 3D gels on titanium nanosurfaces promoted the formation of intercellular networks and gap junctions even under static culture conditions, in contrast to the poor intercellular connectivity in machined titanium surfaces. The osteocyte 3D network on the titanium nanosurfaces further enhanced gap junction formation after additional culturing under cyclic compressive loading simulating masticatory loading, beyond the degree observed on machined titanium surfaces. A prostaglandin synthesis inhibitor cancelled the dual effects of titanium nanosurfaces and cyclic compressive loading on the upregulation of gap junction-related genes in the osteocyte 3D culture. Supernatants from osteocyte monolayer culture on titanium nanosurfaces promoted osteocyte maturation and intercellular connections with gap junctions. With cyclic loading, titanium nanosurfaces induced expression of the regulatory factors of bone turnover in osteocyte 3D cultures, toward higher osteoblast activation than that observed on machined surfaces. Titanium nanosurfaces with anisotropically patterned dense nanospikes promoted intercellular 3D network development and regulatory function toward osteoblast activation in osteocytes activated by cyclic compressive loading, through intercellular crosstalk by prostaglandin.


Assuntos
Osteoblastos , Osteócitos , Titânio , Titânio/farmacologia , Titânio/química , Animais , Osteócitos/metabolismo , Osteócitos/fisiologia , Osteócitos/efeitos dos fármacos , Camundongos , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Osteoblastos/fisiologia , Linhagem Celular , Propriedades de Superfície , Junções Comunicantes/efeitos dos fármacos , Junções Comunicantes/fisiologia , Junções Comunicantes/metabolismo , Nanoestruturas
3.
Sensors (Basel) ; 24(11)2024 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-38894171

RESUMO

Adherent cells perceive mechanical feedback from the underlying matrix and convert it into biochemical signals through a process known as mechanotransduction. The response to changes in the microenvironment relies on the cell's mechanical properties, including elasticity, which was recently identified as a biomarker for various diseases. Here, we propose the design, development, and characterization of a new system for the measurement of adherent cells' strain drop, a parameter correlated with cells' elasticity. To consider the interplay between adherent cells and the host extracellular matrix, cell stretching was combined with adhesion on substrates with different stiffnesses. The technique is based on the linear stretching of silicone chambers, high-speed image acquisition, and feedback for image centering. The system was characterized in terms of the strain homogeneity, impact of collagen coating, centering capability, and sensitivity. Subsequently, it was employed to measure the strain drop of two osteosarcoma cell lines, low-aggressive osteoblast-like SaOS-2 and high-aggressive 143B, cultured on two different substrates to recall the stiffness of the bone and lung extracellular matrices. Results demonstrated good substrate homogeneity, a negligible effect of the collagen coating, and an accurate image centering. Finally, the experimental results showed an average strain drop that was lower in the 143B cells in comparison with the SaOS-2 cells in all the tested conditions.


Assuntos
Osteossarcoma , Osteossarcoma/patologia , Humanos , Linhagem Celular Tumoral , Matriz Extracelular/metabolismo , Mecanotransdução Celular/fisiologia , Adesão Celular/fisiologia , Elasticidade , Estresse Mecânico , Neoplasias Ósseas/patologia , Colágeno/química , Colágeno/metabolismo , Osteoblastos/citologia , Osteoblastos/fisiologia
4.
J Orthop Surg Res ; 19(1): 330, 2024 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-38825686

RESUMO

OBJECTIVE: The present study aimed to investigate the underlying mechanism of mechanical stimulation in regulating osteogenic differentiation. MATERIALS AND METHODS: Osteoblasts were exposed to compressive force (0-4 g/cm2) for 1-3 days or CGRP for 1 or 3 days. Expression of receptor activity modifying protein 1 (RAMP1), the transcription factor RUNX2, osteocalcin, p38 and p-p38 were analyzed by western blotting. Calcium mineralization was analyzed by alizarin red straining. RESULTS: Using compressive force treatments, low magnitudes (1 and 2 g/cm2) of compressive force for 24 h promoted osteoblast differentiation and mineral deposition whereas higher magnitudes (3 and 4 g/cm2) did not produce osteogenic effect. Through western blot assay, we observed that the receptor activity-modifying protein 1 (RAMP1) expression was upregulated, and p38 mitogen-activated protein kinase (MAPK) was phosphorylated during low magnitudes compressive force-promoted osteoblast differentiation. Further investigation of a calcitonin gene-related peptide (CGRP) peptide incubation, a ligand for RAMP1, showed that CGRP at concentration of 25 and 50 ng/ml could increase expression levels of RUNX2 and osteocalcin, and percentage of mineralization, suggesting its osteogenic potential. In addition, with the same conditions, CGRP also significantly upregulated RAMP1 and phosphorylated p38 expression levels. Also, the combination of compressive forces (1 and 2 g/cm2) with 50 ng/ml CGRP trended to increase RAMP1 expression, p38 activity, and osteogenic marker RUNX2 levels, as well as percentage of mineralization compared to compressive force alone. This suggest that RAMP1 possibly acts as an upstream regulator of p38 signaling during osteogenic differentiation. CONCLUSION: These findings suggest that CGRP-RAMP1/p38MAPK signaling implicates in osteoblast differentiation in response to optimal magnitude of compressive force. This study helps to define the underlying mechanism of compressive stimulation and may also enhance the application of compressive stimulation or CGRP peptide as an alternative approach for accelerating tooth movement in orthodontic treatment.


Assuntos
Diferenciação Celular , Osteoblastos , Osteogênese , Proteína 1 Modificadora da Atividade de Receptores , Proteínas Quinases p38 Ativadas por Mitógeno , Osteoblastos/fisiologia , Osteoblastos/metabolismo , Osteoblastos/citologia , Diferenciação Celular/fisiologia , Proteína 1 Modificadora da Atividade de Receptores/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Osteogênese/fisiologia , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Estresse Mecânico , Animais , Células Cultivadas , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Transdução de Sinais/fisiologia , Osteocalcina/metabolismo
5.
Eur J Oral Sci ; 132(4): e12992, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38771146

RESUMO

Finite element analysis (FEA) has been used to analyze the behavior of dental materials, mainly in implantology. However, FEA is a mechanical analysis and few studies have tried to simulate the biological characteristics of the healing process of loaded implants. This study used the rule of mixtures to simulate the biological healing process of immediate implants in an alveolus socket and bone-implant junction interface through FEA. Three-dimensional geometric models of the structures were obtained, and material properties were derived from the literature. The rule of mixtures was used to simulate the healing periods-immediate and early loading, in which the concentration of each cell type, based on in vivo studies, influenced the final elastic moduli. A 100 N occlusal load was simulated in axial and oblique directions. The models were evaluated for maximum and minimum principal strains, and the bone overload was assessed through Frost's mechanostat. There was a higher strain concentration in the healing regions and cortical bone tissue near the cervical portion. The bone overload was higher in the immediate load condition. The method used in this study may help to simulate the biological healing process and could be useful to relate FEA results to clinical practice.


Assuntos
Implantes Dentários , Módulo de Elasticidade , Análise de Elementos Finitos , Carga Imediata em Implante Dentário , Alvéolo Dental , Cicatrização , Humanos , Alvéolo Dental/fisiologia , Cicatrização/fisiologia , Fenômenos Biomecânicos , Simulação por Computador , Interface Osso-Implante/fisiologia , Estresse Mecânico , Processo Alveolar/fisiologia , Modelos Biológicos , Osseointegração/fisiologia , Força de Mordida , Análise do Estresse Dentário/métodos , Osteoblastos/fisiologia , Osso Cortical/fisiologia , Imageamento Tridimensional/métodos
6.
Ann Anat ; 255: 152286, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38810763

RESUMO

OBJECTIVES: The molecular mechanisms behind orthodontic tooth movements (OTM) were investigated by clarifying the role of chemical messengers released by cells. METHODS: Using the Cochrane library, Google scholar, and PubMed databases, a literature search was conducted, and studies published from 1984 to 2024 were considered. RESULTS: Both bone growth and remodeling may occur when a tooth is subjected to mechanical stress. These chemicals have a significant effect on the stimulation and regulation of osteoblasts, osteoclasts, and osteocytes during alveolar bone remodeling. This regulation can take place in pathological conditions, such as periodontal diseases, or during OTM alone. This comprehensive review outlines key molecular mechanisms underlying OTM and explores various clinical assumptions associated with specific molecules and their functional domains during this process. Furthermore, clinical applications of certain molecules such as relaxin, prostaglandin E (PGE), and interleukin-1ß (IL-1ß) in accelerating OTM have been reported. Our findings underscore the existing gap between OTM clinical applications and basic research investigations. CONCLUSION: A comprehensive understanding of orthodontic treatment is enriched by insights into biological systems. We reported the activation of osteoblasts, osteoclast precursor cells, osteoclasts, and osteocytes in response to mechanical stress, leading to targeted cellular and molecular interventions and facilitating rapid and regulated alveolar bone remodeling during tooth movement. Despite the shortcomings of clinical studies in accelerating OTM, this review highlights the crucial role of biological agents in this process and advocates for prioritizing high-quality human studies in future research to gain further insights from clinical trials.


Assuntos
Remodelação Óssea , Técnicas de Movimentação Dentária , Humanos , Remodelação Óssea/fisiologia , Osteoclastos/fisiologia , Estresse Mecânico , Osteócitos/fisiologia , Animais , Osteoblastos/fisiologia
7.
Bull Exp Biol Med ; 176(5): 620-625, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38733480

RESUMO

We studied the interaction of human buccal mesenchymal stem cells (MSCs) and osteoblasts differentiated from them with the surface of titanium samples. MSCs were isolated by enzymatic method from buccal fat pads. The obtained cell culture was presented by MSCs, which was confirmed by flow cytometry and differentiation into adipocytes and osteoblasts. Culturing of buccal MSCs on titanium samples was accompanied by an increase in the number of cells for 15 days and the formation of a developed network of F-actin fibers in the cells. The viability of buccal MSCs decreased by 8 days, but was restored by 15 days. Culturing of osteoblasts obtained as a result of buccal MSC differentiation on the surface of titanium samples was accompanied by a decrease in their viability and proliferation. Thus, MSCs from buccal fat pads can be used to coat implants to improve osseointegration during bone reconstruction in craniofacial surgery and dentistry. To improve the integration of osteoblasts, modification of the surface of titanium samples is required.


Assuntos
Diferenciação Celular , Células-Tronco Mesenquimais , Osseointegração , Osteoblastos , Titânio , Titânio/química , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Humanos , Osseointegração/fisiologia , Osteoblastos/citologia , Osteoblastos/fisiologia , Células Cultivadas , Proliferação de Células , Implantes Dentários , Sobrevivência Celular , Adipócitos/citologia , Adipócitos/fisiologia , Mucosa Bucal/citologia , Osteogênese/fisiologia
8.
Life Sci Space Res (Amst) ; 41: 127-135, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38670639

RESUMO

Understanding how skeletal tissues respond to microgravity is ever more important with the increased interest in human space travel. Here, we exposed larval Danio rerio at 3.5 dpf to simulated microgravity (SMG) using a 3D mode of rotation in a ground-based experiment and then studied different cellular, molecular, and morphological bone responses both immediately after exposure and one week later. Our results indicate an overall decrease in ossification in several developing skeletal elements immediately after SMG exposure with the exception of the otoliths, however ossification returns to normal levels seven days after exposure. Coincident with the reduction in overall ossification tnfsf11 (RANKL) expression is highly elevated after 24 h of SMG exposure and also returns to normal levels seven days after exposure. We also show that genes associated with osteoblasts are unaffected immediately after SMG exposure. Thus, the observed reduction in ossification is primarily the result of a high level of bone resorption. This study sheds insight into the nuances of how osteoblasts and osteoclasts in the skeleton of a vertebrate organism respond to an external environmental disturbance, in this case simulated microgravity.


Assuntos
Larva , Osteogênese , Simulação de Ausência de Peso , Peixe-Zebra , Animais , Larva/crescimento & desenvolvimento , Larva/fisiologia , Osteoblastos/fisiologia , Osteoclastos/fisiologia , Ligante RANK/metabolismo , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo , Ausência de Peso/efeitos adversos
9.
Theranostics ; 14(6): 2544-2559, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38646641

RESUMO

Background: Mechanical forces are indispensable for bone healing, disruption of which is recognized as a contributing cause to nonunion or delayed union. However, the underlying mechanism of mechanical regulation of fracture healing is elusive. Methods: We used the lineage-tracing mouse model, conditional knockout depletion mouse model, hindlimb unloading model and single-cell RNA sequencing to analyze the crucial roles of mechanosensitive protein polycystin-1 (PC1, Pkd1) promotes periosteal stem/progenitor cells (PSPCs) osteochondral differentiation in fracture healing. Results: Our results showed that cathepsin (Ctsk)-positive PSPCs are fracture-responsive and mechanosensitive and can differentiate into osteoblasts and chondrocytes during fracture repair. We found that polycystin-1 declines markedly in PSPCs with mechanical unloading while increasing in response to mechanical stimulus. Mice with conditional depletion of Pkd1 in Ctsk+ PSPCs show impaired osteochondrogenesis, reduced cortical bone formation, delayed fracture healing, and diminished responsiveness to mechanical unloading. Mechanistically, PC1 facilitates nuclear translocation of transcriptional coactivator TAZ via PC1 C-terminal tail cleavage, enhancing osteochondral differentiation potential of PSPCs. Pharmacological intervention of the PC1-TAZ axis and promotion of TAZ nuclear translocation using Zinc01442821 enhances fracture healing and alleviates delayed union or nonunion induced by mechanical unloading. Conclusion: Our study reveals that Ctsk+ PSPCs within the callus can sense mechanical forces through the PC1-TAZ axis, targeting which represents great therapeutic potential for delayed fracture union or nonunion.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Diferenciação Celular , Condrócitos , Consolidação da Fratura , Osteogênese , Células-Tronco , Canais de Cátion TRPP , Animais , Consolidação da Fratura/fisiologia , Camundongos , Canais de Cátion TRPP/metabolismo , Canais de Cátion TRPP/genética , Condrócitos/metabolismo , Células-Tronco/metabolismo , Osteogênese/fisiologia , Camundongos Knockout , Condrogênese/fisiologia , Periósteo/metabolismo , Osteoblastos/metabolismo , Osteoblastos/fisiologia , Modelos Animais de Doenças , Masculino
10.
J Dent Res ; 103(5): 467-476, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38616679

RESUMO

Implant osseointegration is reduced in patients with systemic conditions that compromise bone quality, such as osteoporosis, disuse syndrome, and type 2 diabetes. Studies using rodent models designed to mimic these compromised conditions demonstrated reduced bone-to-implant contact (BIC) or a decline in bone mineral density. These adverse effects are a consequence of disrupted intercellular communication. A variety of approaches have been developed to compensate for the altered microenvironment inherent in compromised conditions, including the use of biologics and implant surface modification. Chemical and physical modification of surface properties at the microscale, mesoscale, and nanoscale levels to closely resemble the surface topography of osteoclast resorption pits found in bone has proven to be a highly effective strategy for improving implant osseointegration. The addition of hydrophilicity to the surface further enhances osteoblast response at the bone-implant interface. These surface modifications, applied either alone or in combination, improve osseointegration by increasing proliferation and osteoblastic differentiation of osteoprogenitor cells and enhancing angiogenesis while modulating osteoclast activity to achieve net new bone formation, although the specific effects vary with surface treatment. In addition to direct effects on surface-attached cells, the communication between bone marrow stromal cells and immunomodulatory cells is sensitive to these surface properties. This article reports on the advances in titanium surface modifications, alone and in combination with novel therapeutics in animal models of human disease affecting bone quality. It offers clinically translatable perspectives for clinicians to consider when using different surface modification strategies to improve long-term implant performance in compromised patients. This review supports the use of surface modifications, bioactive coatings, and localized therapeutics as pragmatic approaches to improve BIC and enhance osteogenic activity from both structural and molecular standpoints.


Assuntos
Interface Osso-Implante , Implantes Dentários , Modelos Animais de Doenças , Osseointegração , Propriedades de Superfície , Osseointegração/fisiologia , Animais , Osteoblastos/fisiologia , Humanos , Osteogênese/fisiologia , Osteoclastos , Implantação Dentária Endóssea
11.
Int J Numer Method Biomed Eng ; 40(6): e3821, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38637289

RESUMO

Both cell migration and osteogenic differentiation are critical for successful bone regeneration. Therefore, understanding the mechanobiological aspects that govern these two processes is essential in designing effective scaffolds that promote faster bone regeneration. Studying these two factors at different locations is necessary to manage bone regeneration in various sections of a scaffold. Hence, a multiscale computational model was used to observe the mechanical responses of osteoblasts placed in different positions of the trabecular bone and gyroid scaffold. Fluid shear stresses in scaffolds at cell seeded locations (representing osteogenic differentiation) and strain energy densities in cells at cell substrate interface (representing cell migration) were observed as mechanical response parameters in this study. Comparison of these responses, as two critical factors for bone regeneration, between the trabecular bone and gyroid scaffold at different locations, is the overall goal of the study. This study reveals that the gyroid scaffold exhibits higher osteogenic differentiation and cell migration potential compared to the trabecular bone. However, the responses in the gyroid only mimic the trabecular bone in two out of nine positions. These findings can guide us in predicting the ideal cell seeded sites within a scaffold for better bone regeneration and in replicating a replaced bone condition by altering the physical parameters of a scaffold.


Assuntos
Regeneração Óssea , Osso Esponjoso , Diferenciação Celular , Movimento Celular , Osteoblastos , Osteogênese , Alicerces Teciduais , Regeneração Óssea/fisiologia , Osteoblastos/fisiologia , Osteoblastos/citologia , Diferenciação Celular/fisiologia , Alicerces Teciduais/química , Movimento Celular/fisiologia , Osso Esponjoso/fisiologia , Osteogênese/fisiologia , Humanos , Porosidade , Modelos Biológicos , Estresse Mecânico
12.
Nat Rev Endocrinol ; 20(7): 399-413, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38499689

RESUMO

Bone development and bone remodelling during adult life are highly anabolic processes requiring an adequate supply of oxygen and nutrients. Bone-forming osteoblasts and bone-resorbing osteoclasts interact closely to preserve bone mass and architecture and are often located close to blood vessels. Chondrocytes within the developing growth plate ensure that bone lengthening occurs before puberty, but these cells function in an avascular environment. With ageing, numerous bone marrow adipocytes appear, often with negative effects on bone properties. Many studies have now indicated that skeletal cells have specific metabolic profiles that correspond to the nutritional microenvironment and their stage-specific functions. These metabolic networks provide not only skeletal cells with sufficient energy, but also biosynthetic intermediates that are necessary for proliferation and extracellular matrix synthesis. Moreover, these metabolic pathways control redox homeostasis to avoid oxidative stress and safeguard cell survival. Finally, several intracellular metabolites regulate the activity of epigenetic enzymes and thus control the fate and function of skeletal cells. The metabolic profile of skeletal cells therefore not only reflects their cellular state, but can also drive cellular activity. Insight into skeletal cell metabolism will thus not only advance our understanding of skeletal development and homeostasis, but also of skeletal disorders, such as osteoarthritis, diabetic bone disease and bone malignancies.


Assuntos
Condrócitos , Osteoblastos , Humanos , Animais , Osteoblastos/metabolismo , Osteoblastos/fisiologia , Condrócitos/metabolismo , Condrócitos/fisiologia , Osso e Ossos/metabolismo , Osteoclastos/metabolismo , Osteoclastos/fisiologia , Remodelação Óssea/fisiologia , Desenvolvimento Ósseo/fisiologia , Diferenciação Celular/fisiologia , Homeostase/fisiologia , Adipócitos/metabolismo , Adipócitos/fisiologia
13.
Biomed Tech (Berl) ; 69(4): 367-382, 2024 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-38258440

RESUMO

OBJECTIVES: The biological performance of aluminum oxide-titanium (Al2O3-Ti) composites requires special attention to achieve improved osteoblastic differentiation, and subsequent osseointegration/strong anchorage with the surrounding bone. Therefore, the aim of this study was to improve them by providing calcium phosphate (Ca-P)/bovine serum albumin (BSA) coating on their surfaces. METHODS: Ca-P/BSA coatings were prepared on the surfaces of 75vol.%Ti composites (75Ti-BSA) and pure Ti (100Ti-BSA as a control). The surface characteristics, phase analysis, micro-hardness, BSA release profile and biological responses including cytotoxicity, cell viability, differentiation, mineralization, and cell adhesion were evaluated. RESULTS: The results showed that lower cytotoxicity% and higher mitochondrial activity or viability % were associated with the samples with Ca-P/BSA coatings (particularly 75Ti-BSA having 21.3% cytotoxicity, 111.4% and 288.6% viability at day 1 and 7, respectively). Furthermore, the Ca-P/BSA coating could highly enhance the differentiation of pre-osteoblast cells into osteoblasts in 75Ti-BSA group (ALP concentration of 4.8 ng/ml). However, its influence on cell differentiation in 100Ti-BSA group was negligible. Similar results were also obtained from mineralization assay. The results on cell adhesion revealed that the Ca-P/BSA coated samples differently interacted with MC3T3-E1 cells; enlarged flat cells on 75Ti-BSA vs more spindle-shaped cells on 100Ti-BSA. CONCLUSIONS: Ca-P/BSA coated Al2O3-Ti provided promising biological performance, superior to that of uncoated composites. Therefore, they have the potential to improve implant osseointegration.


Assuntos
Óxido de Alumínio , Fosfatos de Cálcio , Diferenciação Celular , Osteoblastos , Soroalbumina Bovina , Titânio , Fosfatos de Cálcio/química , Fosfatos de Cálcio/farmacologia , Osteoblastos/efeitos dos fármacos , Osteoblastos/fisiologia , Osteoblastos/citologia , Soroalbumina Bovina/química , Óxido de Alumínio/química , Óxido de Alumínio/farmacologia , Titânio/química , Titânio/farmacologia , Diferenciação Celular/efeitos dos fármacos , Animais , Adesão Celular/efeitos dos fármacos , Adesão Celular/fisiologia , Materiais Revestidos Biocompatíveis/química , Materiais Revestidos Biocompatíveis/farmacologia , Bovinos , Sobrevivência Celular/efeitos dos fármacos , Osseointegração/efeitos dos fármacos , Osseointegração/fisiologia , Propriedades de Superfície , Camundongos
14.
Endocr Rev ; 45(1): 95-124, 2024 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-37459436

RESUMO

The homeostasis of bone microenvironment is the foundation of bone health and comprises 2 concerted events: bone formation by osteoblasts and bone resorption by osteoclasts. In the early 21st century, leptin, an adipocytes-derived hormone, was found to affect bone homeostasis through hypothalamic relay and the sympathetic nervous system, involving neurotransmitters like serotonin and norepinephrine. This discovery has provided a new perspective regarding the synergistic effects of endocrine and nervous systems on skeletal homeostasis. Since then, more studies have been conducted, gradually uncovering the complex neuroendocrine regulation underlying bone homeostasis. Intriguingly, bone is also considered as an endocrine organ that can produce regulatory factors that in turn exert effects on neuroendocrine activities. After decades of exploration into bone regulation mechanisms, separate bioactive factors have been extensively investigated, whereas few studies have systematically shown a global view of bone homeostasis regulation. Therefore, we summarized the previously studied regulatory patterns from the nervous system and endocrine system to bone. This review will provide readers with a panoramic view of the intimate relationship between the neuroendocrine system and bone, compensating for the current understanding of the regulation patterns of bone homeostasis, and probably developing new therapeutic strategies for its related disorders.


Assuntos
Reabsorção Óssea , Osso e Ossos , Humanos , Osteoblastos/fisiologia , Sistemas Neurossecretores , Homeostase
15.
Cells ; 12(21)2023 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-37947654

RESUMO

This review focuses on understanding the macroscopic and microscopic characteristics of bone tissue and reviews current knowledge of its physiology. It explores how these features intricately collaborate to maintain the balance between osteoblast-mediated bone formation and osteoclast-mediated bone resorption, which plays a pivotal role in shaping not only our physical framework but also overall health. In this work, a comprehensive exploration of microscopic and macroscopic features of bone tissue is presented.


Assuntos
Reabsorção Óssea , Osteoclastos , Humanos , Osteoclastos/fisiologia , Osso e Ossos , Osteoblastos/fisiologia , Diferenciação Celular/fisiologia
16.
Anat Sci Int ; 98(4): 521-528, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37022568

RESUMO

Osteoblasts alignment and migration are involved in the directional formation of bone matrix and bone remodeling. Many studies have demonstrated that mechanical stretching controls osteoblast morphology and alignment. However, little is known about its effects on osteoblast migration. Here, we investigated changes in the morphology and migration of preosteoblastic MC3T3-E1 cells after the removal of continuous or cyclic stretching. Actin staining and time-lapse recording were performed after stretching removal. The continuous and cyclic groups showed parallel and perpendicular alignment to the stretch direction, respectively. A more elongated cell morphology was observed in the cyclic group than in the continuous group. In both stretch groups, the cells migrated in a direction roughly consistent with the cell alignment. Compared to the other groups, the cells in the cyclic group showed an increased migration velocity and were almost divided in the same direction as the alignment. To summarize, our study showed that mechanical stretching changed cell alignment and morphology in osteoblasts, which affected the direction of migration and cell division, and velocity of migration. These results suggest that mechanical stimulation may modulate the direction of bone tissue formation by inducing the directional migration and cell division of osteoblasts.


Assuntos
Actinas , Osteoblastos , Osteoblastos/fisiologia , Osso e Ossos , Divisão Celular
17.
Artigo em Inglês | MEDLINE | ID: mdl-37037204

RESUMO

Osteoclasts are the cells responsible for the bone resorption process during bone remodeling. In a healthy situation, this process results from an equilibrium between new matrix formation by osteoblast and matrix resorption by osteoclast. Osteoporosis (OP) is a systemic bone disease characterized by a decreased bone mass density and alterations in bone microarchitecture, increasing fracture predisposition. Despite the variety of available therapies for OP management there is a growing gap in its treatment associated to the low patients' adherence owing to concerns related with long-term efficacy or safety. This makes the development of new and safe treatments necessary. Among the newly developed strategies, the use of synthetic and natural nanoparticles to modulate osteoclasts differentiation, activity, apoptosis or crosstalk with osteoblasts have arisen. Synthetic nanoparticles exert their therapeutic effect either by loading antiresorptive drugs or including molecules for osteoclasts gene regulation. Moreover, this control over osteoclasts can be improved by their targeting to bone extracellular matrix or osteoclast membranes. Furthermore, natural nanoparticles, also known as extracellular vesicles, have been identified to play a key role in bone homeostasis. Consequently, these systems have been widely studied to control osteoblasts and osteoclasts under variable environments. Additionally, the ability to bioengineer extracellular vesicles has allowed to obtain biomimetic systems with desirable characteristics as drug carriers for osteoclasts. The analyzed information reveals the possibility of modulating osteoclasts by different mechanisms through nanoparticles decreasing bone resorption. These findings suggest that controlling osteoclast activity using nanoparticles has the potential to improve osteoporosis management. This article is categorized under: Implantable Materials and Surgical Technologies > Nanomaterials and Implants Implantable Materials and Surgical Technologies > Nanotechnology in Tissue Repair and Replacement Nanotechnology Approaches to Biology > Nanoscale Systems in Biology.


Assuntos
Reabsorção Óssea , Nanopartículas , Osteoporose , Humanos , Osteoclastos/fisiologia , Reabsorção Óssea/tratamento farmacológico , Osteoblastos/fisiologia , Osteoporose/tratamento farmacológico , Nanopartículas/uso terapêutico , Diferenciação Celular
18.
Cells Dev ; 174: 203836, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36972848

RESUMO

It is known that cellular events underlying the processes of bone maintenance, remodeling, and repair have their basis in the embryonic production of bone. Shh signaling is widely described developing important morphogenetic control in bone by modifying the activity of osteoblast. Furthermore, identifying whether it is associated with the modulation of nuclear control is very important to be the basis for further applications. Experimentally, osteoblasts were exposed with cyclopamine (CICLOP) considering up to 1 day and 7 days, here considered an acute and chronic responses respectively. Firstly, we have validated the osteogenic model in vitro by exposing the osteoblasts to classical differentiating solution up to 7 days to allow the analysis of alkaline phosphatase and mineralization. Conversely, our data shows that differentiating osteoblasts present higher activity of inflammasome-related genes, while Shh signaling members were lower, suggesting a negative feedback between them. Thereafter, to better know about the role of Shh signaling on this manner, functional assays using CICLOP (5 µM) were performed and the data validates the previously hypothesis that Shh represses inflammasome related genes activities. Altogether, our data supports the anti-inflammatory effect of Shh signaling by suppressing Tnfα, Tgfß and inflammasome related genes during osteoblast differentiation, and this comprehension might support the understanding the molecular and cellular mechanisms related in bone regeneration by reporting molecular-related osteoblast differentiation.


Assuntos
Ouriços , Inflamassomos , Animais , Inflamassomos/farmacologia , Osteogênese/genética , Osteoblastos/fisiologia
19.
JCI Insight ; 8(6)2023 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-36729662

RESUMO

The energetic costs of bone formation require osteoblasts to coordinate their activities with tissues, like adipose, that can supply energy-dense macronutrients. In the case of intermittent parathyroid hormone (PTH) treatment, a strategy used to reduce fracture risk, bone formation is preceded by a change in systemic lipid homeostasis. To investigate the requirement for fatty acid oxidation by osteoblasts during PTH-induced bone formation, we subjected mice with osteoblast-specific deficiency of mitochondrial long-chain ß-oxidation as well as mice with adipocyte-specific deficiency for the PTH receptor or adipose triglyceride lipase to an anabolic treatment regimen. PTH increased the release of fatty acids from adipocytes and ß-oxidation by osteoblasts, while the genetic mouse models were resistant to the hormone's anabolic effect. Collectively, these data suggest that PTH's anabolic actions require coordinated signaling between bone and adipose, wherein a lipolytic response liberates fatty acids that are oxidized by osteoblasts to fuel bone formation.


Assuntos
Osteogênese , Hormônio Paratireóideo , Camundongos , Animais , Osteoblastos/fisiologia , Osso e Ossos , Transdução de Sinais
20.
Prog Biophys Mol Biol ; 177: 168-180, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36462638

RESUMO

Static magnetic fields (SMFs), magnetic fields with constant intensity and orientation, have been extensively studied in the field of bone biology both fundamentally and clinically as a non-invasive physical factor. A large number of animal experiments and clinical studies have shown that SMFs have effective therapeutic effects on bone-related diseases such as non-healing fractures, bone non-union of bone implants, osteoporosis and osteoarthritis. The maintenance of bone health in adults depends on the basic functions of bone cells, such as bone formation by osteoblasts and bone resorption by osteoclasts. Numerous studies have revealed that SMFs can regulate the proliferation, differentiation, and function of bone tissue cells, including bone marrow mesenchymal stem cells (BMSCs), osteoblasts, bone marrow monocytes (BMMs), osteoclasts, and osteocytes. In this paper, the effects of SMFs on bone-related diseases and bone tissue cells are reviewed from both in vivo studies and in vitro studies, and the possible mechanisms are analyzed. In addition, some challenges that need to be further addressed in the research of SMF and bone are also discussed.


Assuntos
Osteoclastos , Osteócitos , Animais , Osteoblastos/fisiologia , Diferenciação Celular , Osteogênese , Campos Magnéticos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...