Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.234
Filtrar
1.
Int J Biol Macromol ; 264(Pt 1): 130608, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38447840

RESUMO

Bone defects pose significant challenges in orthopedic surgery, often leading to suboptimal outcomes and complications. Addressing these challenges, we employed a three-electrode electrochemical system to fabricate surface-controlled polyaniline nano-tulips (PANINTs) decorated polycaprolactone (PCL) reinforced chitosan functionalized iron oxide nanoparticles (CS-f-Fe2O3) scaffolds. These structures were designed to emulate the natural extracellular matrix (ECM) and promote enhanced osseointegration by establishing a continuous interface between host bone and graft, thereby improving both biological processes and mechanical stability. In vitro experiments demonstrated that PANINTs-PCL/CS-f-Fe2O3 substrates significantly promoted the proliferation, differentiation, and spontaneous outgrowth and extension of MC3T3-E1 cell activity. The nanomaterials exhibited increased cell viability and osteogenic differentiation, as evidenced by elevated expression of bone-related markers such as ALP, ARS, COL-I, RUNX2, and SPP-I, as determined by qRT-PCR. Our findings underscore the regenerative potential of in situ cell culture systems for bone defects, emphasizing the targeted stimulation of essential cell subpopulations to facilitate rapid bone tissue regeneration.


Assuntos
Compostos de Anilina , Quitosana , Quitosana/química , Osteogênese , Tecidos Suporte/química , Regeneração Óssea/fisiologia , Técnicas Eletroquímicas , Engenharia Tecidual/métodos , Diferenciação Celular , Proliferação de Células , Poliésteres/química
2.
Colloids Surf B Biointerfaces ; 237: 113859, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38547794

RESUMO

The main goal of bone tissue engineering research is to replace the allogenic and autologous bone graft substitutes that can promote bone repair. Owing to excellent biocompatibility and osteoconductivity, hydroxyapatite is in extensive research and high demand for both medical and non-medical applications. Although various methods have been developed for the synthesis of hydroxyapatite, in the present study we have shown the use of nanosecond laser energy in the wet precipitation method of nano-hydroxyapatite (nHAP) synthesis without using ammonium solution or any other chemicals for pH maintenance. Here, the present study aimed to fabricate the nanohydroxyapatite using a nanosecond laser. The X-ray diffraction and Fourier transform infrared spectroscopy have confirmed the hydroxyapatite formation under laser irradiation in less time without aging. A transmission electron microscopy confirmed the nano size of synthesized nHAP, which is comparable to conventional nHAP. The length and width of the laser-assisted nHAP were found to be in the range of 50-200 nm and 15-20 nm, respectively, at various laser parameters. The crystallite size obtained by Debye Scherrer formulae was found to be in the range of ∼ 16-36 nm. In addition, laser-assisted nHAP based composite cryogel (nanohydroxyapatite/gelatin/collagen I) was synthesized and impregnated with bioactive molecules (bone morphogenic protein and zoledronic acid) that demonstrated significant osteogenic potential both in vitro in cell experiment and in vivo rat muscle pouch model (abdomen and tibia muscles). Dual-energy X-ray analysis, micro-CT, and histological analysis confirmed ectopic bone regeneration. Micro-CT based histomorphometry showed a higher amount (more than 10-fold) of mineralization for animal groups implanted with composite cryogels loaded with bioactive molecules compared to only composite cryogels groups. Our findings thus demonstrate a controlled and rapid synthetic method for the synthesis of nHAP with various physical, chemical, and biological properties exhibited as comparable to conventionally synthesized nHAP.


Assuntos
Criogéis , Durapatita , Pirenos , Ratos , Animais , Durapatita/farmacologia , Durapatita/química , Regeneração Óssea/fisiologia , Osso e Ossos , Tecidos Suporte/química
3.
Int J Mol Sci ; 25(6)2024 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-38542453

RESUMO

Promoting the efficiency of bone regeneration in bone loss diseases is a significant clinical challenge. Traditional therapies often fail to achieve better therapeutic outcomes and shorter treatment times. However, in recent years, extracellular vesicles (EVs) have gained significant attention due to their exceptional osteogenic function in bone regeneration and superior therapeutic effects compared to traditional cell therapy. EVs have emerged as a promising therapy for tissue defect regeneration due to their various physiological functions, such as regulating the immune response and promoting tissue repair and regeneration. Moreover, EVs have good biocompatibility, low immunogenicity, and long-term stability, and can be improved through pretreatment and other methods. Studies investigating the mechanisms by which extracellular vesicles promote bone regeneration and applying EVs from different sources using various methods to animal models of bone defects have increased. Therefore, this paper reviews the types of EVs used for bone regeneration, their sources, roles, delivery pathways, scaffold biomaterials, and applications.


Assuntos
Doenças Ósseas , Vesículas Extracelulares , Animais , Regeneração Óssea/fisiologia , Osteogênese , Vesículas Extracelulares/metabolismo , Materiais Biocompatíveis/metabolismo , Terapia Baseada em Transplante de Células e Tecidos , Doenças Ósseas/terapia , Doenças Ósseas/metabolismo
4.
Stem Cell Res Ther ; 15(1): 33, 2024 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-38321490

RESUMO

BACKGROUND: There is growing evidence that extracellular vesicles (EVs) play a crucial role in the paracrine mechanisms of transplanted human mesenchymal stem cells (hMSCs). Little is known, however, about the influence of microenvironmental stimuli on the osteogenic effects of EVs. This study aimed to investigate the properties and functions of EVs derived from undifferentiated hMSC (Naïve-EVs) and hMSC during the early stage of osteogenesis (Osteo-EVs). A further aim was to assess the osteoinductive potential of Osteo-EVs for bone regeneration in rat calvarial defects. METHODS: EVs from both groups were isolated using size-exclusion chromatography and characterized by size distribution, morphology, flow cytometry analysis and proteome profiling. The effects of EVs (10 µg/ml) on the proliferation, migration, and osteogenic differentiation of cultured hMSC were evaluated. Osteo-EVs (50 µg) or serum-free medium (SFM, control) were combined with collagen membrane scaffold (MEM) to repair critical-sized calvarial bone defects in male Lewis rats and the efficacy was assessed using µCT, histology and histomorphometry. RESULTS: Although Osteo- and Naïve-EVs have similar characteristics, proteomic analysis revealed an enrichment of bone-related proteins in Osteo-EVs. Both groups enhance cultured hMSC proliferation and migration, but Osteo-EVs demonstrate greater efficacy in promoting in vitro osteogenic differentiation, as evidenced by increased expression of osteogenesis-related genes, and higher calcium deposition. In rat calvarial defects, MEM with Osteo-EVs led to greater and more consistent bone regeneration than MEM loaded with SFM. CONCLUSIONS: This study discloses differences in the protein profile and functional effects of EVs obtained from naïve hMSC and hMSC during the early stage of osteogenesis, using different methods. The significant protein profile and cellular function of EVs derived from hMSC during the early stage of osteogenesis were further verified by a calvarial bone defect model, emphasizing the importance of using differentiated MSC to produce EVs for bone therapeutics.


Assuntos
Vesículas Extracelulares , Células-Tronco Mesenquimais , Humanos , Ratos , Masculino , Animais , Osteogênese/genética , Proteômica , Células-Tronco Mesenquimais/metabolismo , Ratos Endogâmicos Lew , Regeneração Óssea/fisiologia , Diferenciação Celular , Vesículas Extracelulares/metabolismo
5.
ACS Nano ; 18(10): 7504-7520, 2024 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-38412232

RESUMO

The essential role of the neural network in enhancing bone regeneration has often been overlooked in biomaterial design, leading to delayed or compromised bone healing. Engineered mesenchymal stem cells (MSCs)-derived exosomes are becoming increasingly recognized as potent cell-free agents for manipulating cellular behavior and improving therapeutic effectiveness. Herein, MSCs are stimulated with nerve growth factor (NGF) to regulate exosomal cargoes to improve neuro-promotive potential and facilitate innervated bone regeneration. In vitro cell experiments showed that the NGF-stimulated MSCs-derived exosomes (N-Exos) obviously improved the cellular function and neurotrophic effects of the neural cells, and consequently, the osteogenic potential of the osteo-reparative cells. Bioinformatic analysis by miRNA sequencing and pathway enrichment revealed that the beneficial effects of N-Exos may partly be ascribed to the NGF-elicited multicomponent exosomal miRNAs and the subsequent regulation and activation of the MAPK and PI3K-Akt signaling pathways. On this basis, N-Exos were delivered on the micropores of the 3D-printed hierarchical porous scaffold to accomplish the sustained release profile and extended bioavailability. In a rat model with a distal femoral defect, the N-Exos-functionalized hierarchical porous scaffold significantly induced neurovascular structure formation and innervated bone regeneration. This study provided a feasible strategy to modulate the functional cargoes of MSCs-derived exosomes to acquire desirable neuro-promotive and osteogenic potential. Furthermore, the developed N-Exos-functionalized hierarchical porous scaffold may represent a promising neurovascular-promotive bone reparative scaffold for clinical translation.


Assuntos
Exossomos , Células-Tronco Mesenquimais , Ratos , Animais , Exossomos/metabolismo , Diferenciação Celular/genética , Porosidade , Fosfatidilinositol 3-Quinases , Fator de Crescimento Neural/análise , Fator de Crescimento Neural/metabolismo , Fator de Crescimento Neural/farmacologia , Regeneração Óssea/fisiologia , Osteogênese , Impressão Tridimensional
6.
Biochem Biophys Res Commun ; 704: 149699, 2024 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-38412668

RESUMO

With an increasing understanding of the mechanisms of fracture healing, it has been found that nerve injury plays a crucial role in the process, but the specific mechanism is yet to be completely revealed. To address this issue and provide novel insights for fracture treatment, we compiled this review. This review aims to study the impact of nerve injury on fracture healing, exploring the role of neurotrophic factors in the healing process. We first revisited the effects of the central nervous system (CNS) and the peripheral nervous system (PNS) on the skeletal system, and further explained the phenomenon of significantly accelerated fracture healing under nerve injury conditions. Then, from the perspective of neurotrophic factors, we delved into the physiological functions and mechanisms of neurotrophic factors, such as nerve growth factor (NGF), Neuropeptides (NPs), and Brain-derived neurotrophic factor (BDNF), in bone metabolism. These effects include direct actions on bone cells, improvement of local blood supply, regulation of bone growth factors, control of cellular signaling pathways, promotion of callus formation and bone regeneration, and synergistic or antagonistic effects with other endocrine factors, such as Sema3A and Transforming Growth Factor ß (TGF-ß). Finally, we discussed the treatments of fractures with nerve injuries and the future research directions in this review, suggesting that the relationship between nerve injury and fracture healing, as well as the role of nerve injury in other skeletal diseases.


Assuntos
Fraturas Ósseas , Neuropeptídeos , Doenças do Sistema Nervoso Periférico , Humanos , Consolidação da Fratura/fisiologia , Regeneração Óssea/fisiologia
7.
ACS Biomater Sci Eng ; 10(3): 1302-1322, 2024 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-38346448

RESUMO

The treatment of bone defects has been a long-standing challenge in clinical practice. Among the various bone tissue engineering approaches, there has been substantial progress in the development of drug delivery systems based on functional drugs and appropriate carrier materials owing to technological advances in recent years. A large number of materials based on functional nanocarriers have been developed and applied to improve the complex osteogenic microenvironment, including for promoting osteogenic activity, inhibiting osteoclast activity, and exerting certain antibacterial effects. This Review discusses the physicochemical properties, drug loading mechanisms, advantages and disadvantages of nanoparticles (NPs) used for constructing drug delivery systems. In addition, we provide an overview of the osteogenic microenvironment regulation mechanism of drug delivery systems based on nanoparticle (NP) carriers and the construction strategies of drug delivery systems. Finally, the advantages and disadvantages of NP carriers are summarized along with their prospects and future research trends in bone tissue engineering. This Review thus provides advanced strategies for the design and application of drug delivery systems based on NPs in the treatment of bone defects.


Assuntos
Sistemas de Liberação de Fármacos por Nanopartículas , Nanopartículas , Sistemas de Liberação de Medicamentos , Regeneração Óssea/fisiologia , Nanopartículas/uso terapêutico , Nanopartículas/química , Engenharia Tecidual
8.
ACS Appl Mater Interfaces ; 16(8): 9768-9786, 2024 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-38349802

RESUMO

This study aims to overcome the drawbacks associated with hydroxyapatite (HAP) dense structures after sintering, which often result in undesirable features such as large grain size, reduced porosity, high crystallinity, and low specific surface area. These characteristics hinder osseointegration and limit the clinical applicability of the material. To address these issues, a new method involving the preparation of hollow hydroxyapatite (hHAP) microspheres has been proposed. These microspheres exhibit distinctive traits including weak crystallization, high specific surface area, and increased porosity. The weak crystallization aligns more closely with early mineralization products found in the human body and animals. Moreover, the microspheres' high specific surface area and porosity offer advantages for protein loading and facilitating osteoblast attachment. This innovative approach not only mitigates the limitations of conventional HAP structures but also holds the potential for improving the effectiveness of hydroxyapatite in biomedical applications, particularly in enhancing osseointegration. Three-dimensional printed hHAP/chitosan (CS) scaffolds with different hHAP concentration gradients were manufactured, and the physical and biological properties of each group were systematically evaluated. In vitro and in vivo experiments show that the hHAP/CS scaffold has excellent performance in bone remodeling. Furthermore, in-scaffold components, hHAP and CS were cocultured with bone marrow mesenchymal stem cells to explore the regulatory role of hHAP and CS in the process of bone healing and to reveal the cell-level specific regulatory network activated by hHAP. Enrichment analysis showed that hHAP can promote bone regeneration and reconstruction by recruiting calcium ions and regulating inflammatory reactions.


Assuntos
Quitosana , Durapatita , Animais , Humanos , Durapatita/farmacologia , Durapatita/química , Tecidos Suporte/química , Cálcio , Osteogênese , Regeneração Óssea/fisiologia , Quitosana/química , Impressão Tridimensional , Porosidade , Íons
9.
Sci Adv ; 10(6): eadk6722, 2024 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-38324693

RESUMO

Reconstructing extensive cranial defects represents a persistent clinical challenge. Here, we reported a hybrid three-dimensional (3D) printed scaffold with modification of QK peptide and KP peptide for effectively promoting endogenous cranial bone regeneration. The hybrid 3D printed scaffold consists of vertically aligned cryogel fibers that guide and promote cell penetration into the defect area in the early stages of bone repair. Then, the conjugated QK peptide and KP peptide further regulate the function of the recruited cells to promote vascularization and osteogenic differentiation in the defect area. The regenerated bone volume and surface coverage of the dual peptide-modified hybrid scaffold were significantly higher than the positive control group. In addition, the dual peptide-modified hybrid scaffold demonstrated sustained enhancement of bone regeneration and avoidance of bone resorption compared to the collagen sponge group. We expect that the design of dual peptide-modified hybrid scaffold will provide a promising strategy for bone regeneration.


Assuntos
Osteogênese , Tecidos Suporte , Criogéis , Regeneração Óssea/fisiologia , Peptídeos , Impressão Tridimensional
10.
Int J Mol Sci ; 25(4)2024 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-38396834

RESUMO

The periosteum is known as the thin connective tissue covering most bone surfaces. Its extrusive bone regeneration capacity was confirmed from the very first century-old studies. Recently, pluripotent stem cells in the periosteum with unique physiological properties were unveiled. Existing in dynamic contexts and regulated by complex molecular networks, periosteal stem cells emerge as having strong capabilities of proliferation and multipotential differentiation. Through continuous exploration of studies, we are now starting to acquire more insight into the great potential of the periosteum in bone formation and repair in situ or ectopically. It is undeniable that the periosteum is developing further into a more promising strategy to be harnessed in bone tissue regeneration. Here, we summarized the development and structure of the periosteum, cell markers, and the biological features of periosteal stem cells. Then, we reviewed their pivotal role in bone repair and the underlying molecular regulation. The understanding of periosteum-related cellular and molecular content will help enhance future research efforts and application transformation of the periosteum.


Assuntos
Regeneração Óssea , Periósteo , Regeneração Óssea/fisiologia , Osteogênese/fisiologia , Células-Tronco , Diferenciação Celular , Engenharia Tecidual
11.
Tissue Eng Part C Methods ; 30(4): 159-169, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38368556

RESUMO

Considerable research is being undertaken to develop novel biomaterials-based approaches for surgical reconstruction of bone defects. This extends to three-dimensional (3D) printed materials that provide stable, structural, and functional support in vivo. However, few preclinical models can simulate in vivo human biological conditions for clinically relevant testing. In this study we describe a novel ovine model that allows evaluation of in vivo osteogenesis via contact with bone and/or periosteum interfaced with printed polymer bioreactors loaded with biomaterial bone substitutes. The infraspinous scapular region of 14 Dorset cross sheep was exposed. Vascularized periosteum was elevated either attached to the infraspinatus muscle or separately. In both cases, the periosteum was supplied by the periosteal branch of the circumflex scapular vessels. In eight sheep, a 3D printed 4-chambered polyetheretherketone bioreactor was wrapped circumferentially in vascularized periosteum. In 6 sheep, 12 double-sided 3D printed 2-chambered polyetherketone bioreactors were secured to the underlying bone allowing direct contact with the bone on one side and periosteum on the other. Our model enabled simultaneous testing of up to 24 (12 double-sided) 10 × 10 × 5 mm bioreactors per scapula in the flat contact approach or a single 40 × 10 mm four-chambered bioreactor per scapula using the periosteal wrap. De novo bone growth was evaluated using histological and radiological analysis. Of importance, the experimental model was well tolerated by the animals and provides a versatile approach for comparing the osteogenic potential of cambium on the bone surface and elevated with periosteum. Furthermore, the periosteal flaps were sufficiently large for encasing bioreactors containing biomaterial bone substitutes for applications such as segmental mandibular reconstruction.


Assuntos
Substitutos Ósseos , Periósteo , Ovinos , Animais , Humanos , Periósteo/patologia , Periósteo/fisiologia , Periósteo/cirurgia , Regeneração Óssea/fisiologia , Osteogênese/fisiologia , Materiais Biocompatíveis , Reatores Biológicos
12.
Nat Commun ; 15(1): 119, 2024 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-38168072

RESUMO

The sophisticated hierarchical structure that precisely combines contradictory mechanical and biological characteristics is ideal for biomaterials, but it is challenging to achieve. Herein, we engineer a spatiotemporally hierarchical guided bone regeneration (GBR) membrane by rational bilayer integration of densely porous N-halamine functionalized bacterial cellulose nanonetwork facing the gingiva and loosely porous chitosan-hydroxyapatite composite micronetwork facing the alveolar bone. Our GBR membrane asymmetrically combine stiffness and flexibility, ingrowth barrier and ingrowth guiding, as well as anti-bacteria and cell-activation. The dense layer has a mechanically matched space maintenance capacity toward gingiva, continuously blocks fibroblasts, and prevents bacterial invasion with multiple mechanisms including release-killing, contact-killing, anti-adhesion, and nanopore-blocking; the loose layer is ultra-soft to conformally cover bone surfaces and defect cavity edges, enables ingrowth of osteogenesis-associated cells, and creates a favorable osteogenic microenvironment. As a result, our all-in-one porous membrane possesses full protective abilities in GBR.


Assuntos
Regeneração Óssea , Membranas Artificiais , Porosidade , Regeneração Óssea/fisiologia , Osteogênese , Materiais Biocompatíveis/química
13.
QJM ; 117(2): 95-108, 2024 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-37252831

RESUMO

Efficient regeneration of bone defects caused by disease or significant trauma is a major challenge in current medicine, which is particularly difficult yet significant under the emerging psychological stress in the modern society. Notably, the brain-bone axis has been proposed as a prominent new concept in recent years, among which autonomic nerves act as an essential and emerging skeletal pathophysiological factor related to psychological stress. Studies have established that sympathetic cues lead to impairment of bone homeostasis mainly through acting on mesenchymal stem cells (MSCs) and their derivatives with also affecting the hematopoietic stem cell (HSC)-lineage osteoclasts, and the autonomic neural regulation of stem cell lineages in bone is increasingly recognized to contribute to the bone degenerative disease, osteoporosis. This review summarizes the distribution characteristics of autonomic nerves in bone, introduces the regulatory effects and mechanisms of autonomic nerves on MSC and HSC lineages, and expounds the crucial role of autonomic neural regulation on bone physiology and pathology, which acts as a bridge between the brain and the bone. With the translational perspective, we further highlight the autonomic neural basis of psychological stress-induced bone loss and a series of pharmaceutical therapeutic strategies and implications toward bone regeneration. The summary of research progress in this field will add knowledge to the current landscape of inter-organ crosstalk and provide a medicinal basis for the achievement of clinical bone regeneration in the future.


Assuntos
Doenças Ósseas , Osso e Ossos , Humanos , Regeneração Óssea/fisiologia , Encéfalo , Estresse Psicológico
14.
Angiogenesis ; 27(1): 105-119, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38032405

RESUMO

The healing of calvarial bone defects is a pressing clinical problem that involves the dynamic interplay between angiogenesis and osteogenesis within the osteogenic niche. Although structural and functional vascular remodeling (i.e., angiogenic evolution) in the osteogenic niche is a crucial modulator of oxygenation, inflammatory and bone precursor cells, most clinical and pre-clinical investigations have been limited to characterizing structural changes in the vasculature and bone. Therefore, we developed a new multimodality imaging approach that for the first time enabled the longitudinal (i.e., over four weeks) and dynamic characterization of multiple in vivo functional parameters in the remodeled vasculature and its effects on de novo osteogenesis, in a preclinical calvarial defect model. We employed multi-wavelength intrinsic optical signal (IOS) imaging to assess microvascular remodeling, intravascular oxygenation (SO2), and osteogenesis; laser speckle contrast (LSC) imaging to assess concomitant changes in blood flow and vascular maturity; and micro-computed tomography (µCT) to validate volumetric changes in calvarial bone. We found that angiogenic evolution was tightly coupled with calvarial bone regeneration and corresponded to distinct phases of bone healing, such as injury, hematoma formation, revascularization, and remodeling. The first three phases occurred during the initial two weeks of bone healing and were characterized by significant in vivo changes in vascular morphology, blood flow, oxygenation, and maturity. Overall, angiogenic evolution preceded osteogenesis, which only plateaued toward the end of bone healing (i.e., four weeks). Collectively, these data indicate the crucial role of angiogenic evolution in osteogenesis. We believe that such multimodality imaging approaches have the potential to inform the design of more efficacious tissue-engineering calvarial defect treatments.


Assuntos
Regeneração Óssea , Crânio , Microtomografia por Raio-X , Crânio/diagnóstico por imagem , Crânio/irrigação sanguínea , Crânio/lesões , Regeneração Óssea/fisiologia , Osteogênese/fisiologia , Cicatrização
15.
Small ; 20(8): e2307062, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37824284

RESUMO

Vascularization and innervation play irreplaceable roles in bone regeneration and bone defect repair. However, the reconstruction of blood vessels and neural networks is often neglected in material design. This study aims to design a genetically functionalized matrix (GFM) and enable it to regulate angiogenesis and neurogenesis to accelerate the process of bone defect repair. The dual small interfering RNA (siRNA)-polyvinylimide (PEI) (siRP) complexes that locally knocked down soluble vascular endothelial growth factor receptor 1 (sFlt-1) and p75 neurotrophic factor receptor (p75NTR ) are prepared. The hybrid cell membrane (MM) loaded siRP is synthesized as siRNA@MMs to coat on polylactone (PCL) electrospun fibers for mimicking the natural bone matrix. The results indicates that siRNA@MMs could regulate the expression of vascular-related and neuro-related cytokines secreted by mesenchymal stem cells (MSCs). GFMs promote the expression of osteogenic differentiation through paracrine function in vitro. GFMs attenuates inflammation and promotes osseointegration by regulating the coupling of vascularization and innervation in vivo. This study uses the natural hybrid cell membrane to carry genetic material and assist in the vascularization and innervation function of two siRNA. The results present the significance of neuro-vascularized organoid bone and may provide a promising choice for the design of bone tissue engineering scaffold.


Assuntos
Osteogênese , Fator A de Crescimento do Endotélio Vascular , Osteogênese/genética , RNA Interferente Pequeno/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Regeneração Óssea/fisiologia , Tecidos Suporte , Diferenciação Celular , Neurogênese , Membrana Celular/metabolismo
16.
Stomatologiia (Mosk) ; 102(6. Vyp. 2): 8-14, 2023.
Artigo em Russo | MEDLINE | ID: mdl-38096388

RESUMO

Gene therapy is one of the most promising approaches in regenerative medicine for the restoration of extensive bone defects in dentistry and maxillofacial surgery. Matrices obtained using three-dimensional printing from bioresorbable polymers, impregnated with adenoviral constructs with genes for osteoinductive factors, can ensure safe and effective formation of bone tissue. OBJECTIVE: To study the properties of three-dimensional matrices based on polylactic-co-glycolic acid and adenoviral constructs with the GFP gene in vitro. MATERIALS AND METHODS: The matrices were obtained by antisolvent three-dimensional printing. Transduction efficiency was assessed by fluorescence microscopy and flow cytometry. The cytocompatibility of the matrices was assessed by the MTT test and by staining cells with fluorescent dyes. RESULTS: Matrices based on polylactic-co-glycolic acid have high cytocompatibility on adipose tissue-derived mesenchymal stem cells. Impregnation of adenoviral vectors with the green fluorescent protein gene in 3D matrices ensures the release of viral particles within a week, maintaining their high transducing ability. CONCLUSION: The developed method for obtaining gene-activated matrices can serve as the basis for the creation of effective osteoplastic materials for bone regeneration.


Assuntos
Células-Tronco Mesenquimais , Humanos , Polímeros/metabolismo , Regeneração Óssea/fisiologia , Osso e Ossos , Impressão Tridimensional , Tecidos Suporte
17.
Nat Commun ; 14(1): 7687, 2023 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-38001080

RESUMO

Guided bone regeneration aided by the application of occlusive membranes is a promising therapy for diverse inflammatory periodontal diseases. Symbiosis, homeostasis between the host microbiome and cells, occurs in the oral environment under normal, but not pathologic, conditions. Here, we develop a symbiotically integrating occlusive membrane by mimicking the tooth enamel growth or multiple nucleation biomineralization processes. We perform human saliva and in vivo canine experiments to confirm that the symbiotically integrating occlusive membrane induces a symbiotic healing environment. Moreover, we show that the membrane exhibits tractability and enzymatic stability, maintaining the healing space during the entire guided bone regeneration therapy period. We apply the symbiotically integrating occlusive membrane to treat inflammatory-challenged cases in vivo, namely, the open and closed healing of canine premolars with severe periodontitis. We find that the membrane promotes symbiosis, prevents negative inflammatory responses, and improves cellular integration. Finally, we show that guided bone regeneration therapy with the symbiotically integrating occlusive membrane achieves fast healing of gingival soft tissue and alveolar bone.


Assuntos
Perda do Osso Alveolar , Periodontite , Humanos , Regeneração Tecidual Guiada Periodontal , Cicatrização/fisiologia , Gengiva , Membranas Artificiais , Regeneração Óssea/fisiologia
18.
Int J Oral Maxillofac Implants ; 38(5): 923-932, 2023 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-37847834

RESUMO

PURPOSE: To compare bone substitutes composed of glycated collagen with synthetic micro-sized (1 to 10 µm) hydroxyapatite (OB) vs non-cross-linked collagen matrix with large-particle (250 to 1,000 µm) bovine-derived hydroxyapatite (BOC). MATERIALS AND METHODS: The P1 to P4 premolars were bilaterally extracted from the mandibles of 19 Beagle dogs. After 21 days, osteotomies were created in each dog that received OB or BOC and were covered with a collagen membrane or were left untreated. The animals were randomly divided into three groups based on sacrifice time (4, 12, or 24 weeks). The right and left hemimandibles were trimmed to facilitate imaging and histology, and all tissues were placed in 10% neutral-buffered formalin. Microcomputed tomography (MicroCT 40 Scanner, Scanco) was used to analyze bone sections. Bone volume, residual material volume, and bone mineral density were determined for each treatment site (OB and BOC) based on a volume of interest that encompassed the original defect. Additionally, blinded histopathologic assessment (based on the ISO 10993-6 scoring system) and histomorphometry were performed on sections ground to < 100 µm thick and stained with Stevenel's blue. RESULTS: No clinical side effects were noted. No statistical differences were observed for OB vs BOC regarding the mineral volume percentage. Compared to OB, BOC had significantly higher mean mineralization densities at 12 weeks (P < .01), but this difference did not extend to 24 weeks. For residual grafting material, bone maturation, alveolar ridge restoration, and inflammatory response, OB showed a residual amount of bone graft and no statistical differences compared to BOC. CONCLUSION: Both OB and BOC represent valid treatment options for critically sized bone defects. Both bone fillers outperformed the sham-operated, ungrafted (empty) control, demonstrating statistically improved bone growth and ridge restoration.


Assuntos
Aumento do Rebordo Alveolar , Substitutos Ósseos , Cães , Animais , Bovinos , Durapatita/uso terapêutico , Osteogênese , Microtomografia por Raio-X , Regeneração Tecidual Guiada Periodontal/métodos , Regeneração Óssea/fisiologia , Substitutos Ósseos/uso terapêutico , Colágeno/uso terapêutico , Aumento do Rebordo Alveolar/métodos
19.
Adv Healthc Mater ; 12(32): e2301724, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37767893

RESUMO

The bone matrix has distinct architecture and biochemistry which present a barrier to synthesizing bone-mimetic regenerative scaffolds. To mimic the natural structures and components of bone, biomimetic structural decellularized extracellular matrix (ECM)/regenerated silk fibroin (RSF) scaffolds incorporated with magnetic nanoparticles (MNP) are prepared using a facile synthetic methodology. The ECM/RSF/MNP scaffold is a hierarchically organized and interconnected porous structure with silk fibroin twined on the collagen nanofibers. The scaffold demonstrates saturation magnetization due to the presence of MNP, along with good cytocompatibility. Moreover, the ß-sheet crystalline domain of RSF and the chelated MNP could mimic the deposition of hydroxyapatite and enhance compressive modulus of the scaffold by ≈20%. The results indicate that an external static magnetic field (SMF) with a magnetic responsive scaffold effectively promotes cell migration, osteogenic differentiation, neogenesis of endotheliocytes in vitro, and new bone formation in a critical-size femur defect rat model. RNA sequencing reveals that the molecular mechanisms underlying this osteogenic effect involve calsequestrin-2-mediated Ca2+ release from the endoplasmic reticulum to activate Ca2+ /calmodulin/calmodulin-dependent kinase II signaling axis. Collectively, bionic magnetic scaffolds with SMF stimulation provide a potent strategy for bone regeneration through internal structural cues, biochemical composition, and external physical stimulation on intracellular Ca2+ homeostasis.


Assuntos
Fibroínas , Tecidos Suporte , Ratos , Animais , Tecidos Suporte/química , Fibroínas/química , Osteogênese , Cálcio , Biomimética , Calmodulina , Regeneração Óssea/fisiologia , Fenômenos Magnéticos , Engenharia Tecidual/métodos
20.
Artigo em Inglês | MEDLINE | ID: mdl-37612166

RESUMO

OBJECTIVE: Because of the anatomical complexity of the oral and maxillofacial sites, repairing bone defects in these regions is very difficult. This review article aims to consider the application of biocomposites-based strategies for dental bone regeneration. STUDY DESIGN: Research papers related to the topic, published over the last 20 years, were selected using the Web of Science, Pubmed, Scopus, and Google Scholar databases. RESULTS: The strategies of monophasic, biphasic/multiphasic scaffolds, and biopolymer-based nanocomposite scaffolds containing nanomaterials compared with traditional methods used for bone regeneration, such as autografts, allografts, xenografts, and alloplasts are found to be superior because of their ability to overcome the issues (e.g., limited bone sources, pain, immune responses, high cost) related to the applications of the traditional methods. CONCLUSIONS: In addition, additive manufacturing technologies were found to be highly advantageous for improving the efficacy of biocomposite scaffolds for treating dental bone defects.


Assuntos
Regeneração Óssea , Humanos , Regeneração Óssea/fisiologia , Transplante Autólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...