Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.469
Filtrar
1.
J Oral Implantol ; 50(3): 141-152, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38839071

RESUMEN

After dental extraction, a physiological phenomenon of reabsorption of the dentoalveolar process is triggered, especially if periradicular lesions are present, which can sometimes be associated with oroantral communication in the upper posterior maxilla. To investigate a minimally invasive approach, 19 patients undergoing tooth extraction in the posterosuperior maxilla were recruited. All cases presented an oroantral communication with a diameter of 2-5 mm after tooth extraction and the alveolar process and, in some cases, with a partial defect of 1 or more bony walls. In these cases, a single surgical procedure was used to preserve the alveolar ridge using an open barrier technique with an exposed dense polytetrafluoroethylene membrane. The bottom of the extraction socket was filled with a collagen fleece. The residual bone process was reconstructed using a biomaterial based on carbonate-apatite derived from porcine cancellous bone. After 6 months, all patients were recalled and subjected to radiographic control associated with an implant-prosthetic rehabilitation plan. Data relating to the sinus health status and the average height and thickness of the regenerated bone were collected. Radiographic evaluation verified the integrity of the maxillary sinus floor with new bone formation, detecting a vertical bone dimension between 3.1 mm and 7.4 mm (average 5.13 ± 1.15 mm) and a horizontal thickness between 4.2 mm and 9.6 mm (average 6.86 ± 1.55 mm). The goal of this study was to highlight the advantage of managing an oroantral communication and, simultaneously, obtain the preservation and regeneration of the alveolar bone crest. The open barrier technique appears to be effective for the minimally invasive management of oroantral communication up to 5 mm in diameter in postextraction sites, with a good regeneration of hard and soft tissue.


Asunto(s)
Membranas Artificiales , Fístula Oroantral , Politetrafluoroetileno , Extracción Dental , Humanos , Estudios Retrospectivos , Fístula Oroantral/cirugía , Persona de Mediana Edad , Masculino , Femenino , Proceso Alveolar/cirugía , Proceso Alveolar/diagnóstico por imagen , Alveolo Dental/cirugía , Anciano , Adulto , Maxilar/cirugía , Regeneración Ósea/fisiología , Aumento de la Cresta Alveolar/métodos , Colágeno/uso terapéutico
2.
Nat Commun ; 15(1): 4575, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38834586

RESUMEN

Bone regeneration requires a well-orchestrated cellular and molecular response including robust vascularization and recruitment of mesenchymal and osteogenic cells. In femoral fractures, angiogenesis and osteogenesis are closely coupled during the complex healing process. Here, we show with advanced longitudinal intravital multiphoton microscopy that early vascular sprouting is not directly coupled to osteoprogenitor invasion during calvarial bone regeneration. Early osteoprogenitors emerging from the periosteum give rise to bone-forming osteoblasts at the injured calvarial bone edge. Microvessels growing inside the lesions are not associated with osteoprogenitors. Subsequently, osteogenic cells collectively invade the vascularized and perfused lesion as a multicellular layer, thereby advancing regenerative ossification. Vascular sprouting and remodeling result in dynamic blood flow alterations to accommodate the growing bone. Single cell profiling of injured calvarial bones demonstrates mesenchymal stromal cell heterogeneity comparable to femoral fractures with increase in cell types promoting bone regeneration. Expression of angiogenesis and hypoxia-related genes are slightly elevated reflecting ossification of a vascularized lesion site. Endothelial Notch and VEGF signaling alter vascular growth in calvarial bone repair without affecting the ossification progress. Our findings may have clinical implications for bone regeneration and bioengineering approaches.


Asunto(s)
Regeneración Ósea , Células Madre Mesenquimatosas , Neovascularización Fisiológica , Osteogénesis , Cráneo , Animales , Regeneración Ósea/fisiología , Ratones , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Osteoblastos/citología , Osteoblastos/metabolismo , Masculino , Receptores Notch/metabolismo , Receptores Notch/genética , Ratones Endogámicos C57BL , Transducción de Señal , Femenino , Angiogénesis
3.
J Vis Exp ; (207)2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38801271

RESUMEN

Large-scale calvarial defects often coincide with cranial suture disruption, leading to impairments in calvarial defect restoration and skull development (the latter occurs in the developing cranium). However, the lack of a standardized model hinders progress in investigating suture-regenerative therapies and poses challenges for conducting comparative analyses across distinct studies. To address this issue, the current protocol describes the detailed modeling process of calvarial suture-bony composite defects in rats. The model was generated by drilling full-thickness rectangular holes measuring 4.5 mm × 2 mm across the coronal sutures. The rats were euthanized, and the cranium samples were harvested postoperatively at day 0, week 2, week 6, and week 12. µCT results from samples collected immediately post-surgery confirmed the successful establishment of the suture-bony composite defect, involving the removal of the coronal suture and the adjacent bone tissues. Data from the 6th and 12th postoperative weeks demonstrated a natural healing tendency for the defect to close. Histological staining further validated this trend by showing increased mineralized fibers and new bone at the defect center. These findings indicate progressive suture fusion over time following calvarial defects, underscoring the significance of therapeutic interventions for suture regeneration. We anticipate that this protocol will facilitate the development of suture-regenerative therapies, offering fresh insights into the functional restoration of calvarial defects and reducing adverse outcomes associated with suture loss.


Asunto(s)
Suturas Craneales , Cráneo , Animales , Ratas , Cráneo/cirugía , Suturas Craneales/cirugía , Modelos Animales de Enfermedad , Microtomografía por Rayos X/métodos , Masculino , Ratas Sprague-Dawley , Regeneración Ósea/fisiología
4.
Swiss Dent J ; 134(1): 144-157, 2024 Apr 05.
Artículo en Alemán | MEDLINE | ID: mdl-38741457

RESUMEN

The clinical impact of platelet-rich fibrin (PRF) and plasma rich in growth factors (PRGF®) respectively has been studied extensively in the field of regenerative dentistry during the last two decades. Literature supports evidence for additional benefits in regenerative periodontal therapy, alveolar ridge preservation, management of extraction sockets, implantology including guided bone regeneration as well as defect management in oral surgery. Regarding gingival wound healing and soft tissue regeneration, there is sufficient evidence for their positive effects which have been confirmed in several systematic reviews. The effects seem less clear in conjunction with osseous regenerative treatments, where the inter-study heterogenity in terms of different PRF-protocols, indications and application forms might hinder a systematic comparison. Nevertheless there is evidence that PRF might have beneficial effects on hard-tissue or its regeneration respectively.For being able to facilitate conclusions in systematic reviews, precise reporting of the used PRF-protocols is mandatory for future (clinical) research in the field of autologous platelet concentrates.


Asunto(s)
Fibrina Rica en Plaquetas , Plasma Rico en Plaquetas , Humanos , Regeneración Tisular Guiada Periodontal/métodos , Plaquetas/fisiología , Regeneración Ósea/fisiología , Regeneración Ósea/efectos de los fármacos , Cicatrización de Heridas/fisiología , Cicatrización de Heridas/efectos de los fármacos , Medicina Regenerativa/métodos
5.
Int J Implant Dent ; 10(1): 25, 2024 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-38760582

RESUMEN

PURPOSE: This retrospective cohort study evaluates the influence of connective tissue grafts (CTG) on bone regeneration at implant sites with total loss of the buccal bone wall treated with flapless immediate implant placement (IIP) and reconstruction with autogenous bone chips (AB) within a follow-up of up to 13 years. METHODS: Sixty implants were inserted in 55 patients in sites with total loss of the buccal bone wall between 2008 and 2021. The implants were inserted and the buccal gaps were grafted by AB. A subgroup of 34 sites was grafted additionally with CTG using tunnel technique. Primary outcome was the vertical bone regeneration in height and thickness. Secondary outcome parameters were interproximal marginal bone level, recession, soft tissue esthetics (PES), width of keratinized mucosa (KMW) and probing depths (PPD). RESULTS: Mean follow-up period was 60.8 months. In 55 sites a complete vertical bone regeneration was documented. The mean buccal bone level increased by 10.6 mm significantly. The thickness of the buccal bone wall ranged between 1.7 and 1.9 mm, and was significantly thicker in sites without CTG. Interproximal marginal bone level was at implant shoulder level. The mean recession improved significantly by 1.2 mm. In sites with CTG, recessions and PES improved significantly more. CONCLUSIONS: Additional CTG in extraction sites with total buccal bone loss followed by IIP with simultaneous AB grafting led to improved PES and recession, but also to a thinner buccal bone wall compared to sites grafted just with AB.


Asunto(s)
Tejido Conectivo , Carga Inmediata del Implante Dental , Humanos , Estudios Retrospectivos , Tejido Conectivo/trasplante , Femenino , Masculino , Persona de Mediana Edad , Carga Inmediata del Implante Dental/métodos , Adulto , Trasplante Óseo/métodos , Anciano , Regeneración Ósea/fisiología , Aumento de la Cresta Alveolar/métodos , Pérdida de Hueso Alveolar/cirugía
6.
J Orthop Surg Res ; 19(1): 299, 2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-38755635

RESUMEN

BACKGROUND: This study aims to evaluate the optimal ratio of synthetic bone graft (SBG) material and platelet rich fibrin (PRF) mixed in a metal 3D-printed implant to enhance bone regeneration. METHODS: Specialized titanium hollow implants (5 mm in diameter and 6 mm in height for rabbit; 6 mm in diameter and 5 mm in height for pig) were designed and manufactured using 3D printing technology. The implants were divided into three groups and filled with different bone graft combinations, namely (1) SBG alone; (2) PRF to SBG in 1:1 ratio; (3) PRF to SBG in 2:1 ratio. These three groups were replicated tightly into each bone defect in distal femurs of rabbits (nine implants, n = 3) and femoral shafts of pigs (fifteen implants, n = 5). Animal tissue sections were obtained after euthanasia at the 8th postoperative week. The rabbit specimens were stained with analine blue, while the pig specimens were stained with Masson-Goldner's trichrome stain to perform histologically examination. All titanium hollow implants were well anchored, except in fracture specimens (three in the rabbit and one fracture in the pig). RESULT: Rabbit specimens under analine blue staining showed that collagen tissue increased by about 20% and 40% in the 1:1 ratio group and the 2:1 ratio group, respectively. Masson-Goldner's trichrome stain results showed that new bone growth increased by 32% in the 1:1 ratio PRF to SBG, while - 8% in the 2:1 ratio group. CONCLUSION: This study demonstrated that placing a 1:1 ratio combination of PRF and SBG in a stabilized titanium 3D printed implant resulted in an optimal increase in bone growth.


Asunto(s)
Regeneración Ósea , Fibrina Rica en Plaquetas , Impresión Tridimensional , Titanio , Animales , Conejos , Regeneración Ósea/efectos de los fármacos , Regeneración Ósea/fisiología , Porcinos , Fémur/cirugía , Sustitutos de Huesos , Trasplante Óseo/métodos , Prótesis e Implantes
7.
J Am Acad Orthop Surg ; 32(10): e476-e481, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38700858

RESUMEN

With an aging population, and an anticipated increase in overall fracture incidence, a sound understanding of bone healing and how technology can optimize this process is crucial. Concentrated bone marrow aspirate (cBMA) is a technology that capitalizes on skeletal stem and progenitor cells (SSPCs) to enhance the regenerative capacity of bone. This overview highlights the science behind cBMA, discusses the role of SSPCs in bone homeostasis and fracture repair, and briefly details the clinical evidence supporting the use of cBMA in fracture healing. Despite promising early clinical results, a lack of standardization in harvest and processing techniques, coupled with patient variability, presents challenges in optimizing the use of cBMA. However, cBMA remains an emerging technology that may certainly play a crucial role in the future of fracture healing augmentation.


Asunto(s)
Curación de Fractura , Humanos , Curación de Fractura/fisiología , Trasplante de Médula Ósea/métodos , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Regeneración Ósea/fisiología , Fracturas Óseas/terapia , Células de la Médula Ósea/citología
8.
J Orthop Traumatol ; 25(1): 28, 2024 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-38789881

RESUMEN

Mesenchymal stem cells are core to bone homeostasis and repair. They both provide the progenitor cells from which bone cells are formed and regulate the local cytokine environment to create a pro-osteogenic environment. Dysregulation of these cells is often seen in orthopaedic pathology and can be manipulated by the physician treating the patient. This narrative review aims to describe the common applications of cell therapies to bone healing whilst also suggesting the future direction of these techniques.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Humanos , Trasplante de Células Madre Mesenquimatosas/métodos , Regeneración Ósea/fisiología , Células Madre Mesenquimatosas , Curación de Fractura/fisiología , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Osteogénesis/fisiología
9.
Int J Periodontics Restorative Dent ; 44(3): 287-297, 2024 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-38787705

RESUMEN

Titanium has been proposed as a mesh material for guided bone regeneration (GBR) since the 1990s. To overcome difficulties in shaping and adapting meshes to the defect, digital techniques were introduced to digitally print meshes capable of fitting the bone perfectly, reproduced through the patient's CT scan. Five patients were included in this case series, and their CBCT data were acquired and sent to the producer of the titanium meshes. 3D regenerative surgery was performed with titanium meshes and a mix of demineralized bovine bone matrix (DBBM) and autogenous bone (1:1 ratio). Radiographic measures were evaluated on paraxial sections of the CBCT through a dedicated software. When possible, regenerated bone samples were obtained at implant insertion. Four out of five regenerated areas healed without local or systemic complications. One mesh was removed after 2 months and 2 weeks due to exposure. The mean vertical bone gain was 4.3 ± 1.5 mm (range: 2.5 to 7 mm). Two histologic samples were obtained. In sample 1, bone tissue area and graft material area were 44.4% and 12.5%, respectively; in sample 2, the same parameters were 15.6% and 16.9%, respectively.


Asunto(s)
Diseño Asistido por Computadora , Tomografía Computarizada de Haz Cónico , Mallas Quirúrgicas , Titanio , Humanos , Persona de Mediana Edad , Masculino , Femenino , Adulto , Regeneración Tisular Guiada Periodontal/métodos , Regeneración Ósea/fisiología , Animales , Bovinos , Implantación Dental Endoósea/métodos , Trasplante Óseo/métodos , Anciano , Matriz Ósea/trasplante
10.
Exp Cell Res ; 439(1): 114096, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38768700

RESUMEN

Early vascularization plays an essential role during the whole process in bone regeneration because of the function of secreting cytokines, transporting nutrients and metabolic wastes. As the preliminary basis of bone repair, angiogenesis is regulated by immune cells represented by macrophages to a great extent. However, with the discovery of the endolymphatic circulation system inside bone tissue, the role of vascularization became complicated and confusing. Herein, we developed a macrophage/lymphatic endothelial cells (LECs)/human umbilical vein endothelial cells (HUVECs) co-culture system to evaluate the effect of macrophage treated lymphatic endothelial cells on angiogenesis in vitro and in vivo. In this study, we collected the medium from macrophage (CM) for LECs culture. We found that CM2 could promote the expression of LECs markers and migration ability, which indicated the enhanced lymphogenesis. In addition, the medium from LECs was collected for culturing HUVECs. The CM2-treated LECs showed superior angiogenesis property including the migration capacity and expression of angiogenetic markers, which suggested the superior vascularization. Rat femoral condyle defect model was applied to confirm the hypothesis in vivo. Generally, M2-macrophage treated LECs showed prominent angiogenetic potential coupling with osteogenesis.


Asunto(s)
Técnicas de Cocultivo , Células Endoteliales de la Vena Umbilical Humana , Macrófagos , Neovascularización Fisiológica , Osteogénesis , Humanos , Animales , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Macrófagos/metabolismo , Ratas , Células Endoteliales/metabolismo , Movimiento Celular , Ratas Sprague-Dawley , Regeneración Ósea/fisiología , Ratones , Células Cultivadas , Masculino , Angiogénesis
11.
Int J Oral Sci ; 16(1): 33, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38654018

RESUMEN

Precise orchestration of cell fate determination underlies the success of scaffold-based skeletal regeneration. Despite extensive studies on mineralized parenchymal tissue rebuilding, regenerating and maintaining undifferentiated mesenchyme within calvarial bone remain very challenging with limited advances yet. Current knowledge has evidenced the indispensability of rebuilding suture mesenchymal stem cell niches to avoid severe brain or even systematic damage. But to date, the absence of promising therapeutic biomaterials/scaffolds remains. The reason lies in the shortage of fundamental knowledge and methodological evidence to understand the cellular fate regulations of scaffolds. To address these issues, in this study, we systematically investigated the cellular fate determinations and transcriptomic mechanisms by distinct types of commonly used calvarial scaffolds. Our data elucidated the natural processes without scaffold transplantation and demonstrated how different scaffolds altered in vivo cellular responses. A feasible scaffold, polylactic acid electrospinning membrane (PLA), was next identified to precisely control mesenchymal ingrowth and self-renewal to rebuild non-osteogenic suture-like tissue at the defect center, meanwhile supporting proper osteointegration with defect bony edges. Especially, transcriptome analysis and cellular mechanisms underlying the well-orchestrated cell fate determination of PLA were deciphered. This study for the first time cellularly decoded the fate regulations of scaffolds in suture-bony composite defect healing, offering clinicians potential choices for regenerating such complicated injuries.


Asunto(s)
Regeneración Ósea , Andamios del Tejido , Transcriptoma , Animales , Regeneración Ósea/fisiología , Poliésteres , Cráneo/cirugía , Células Madre Mesenquimatosas , Mesodermo/citología , Diferenciación Celular , Ingeniería de Tejidos/métodos , Suturas Craneales , Materiales Biocompatibles
12.
Foot Ankle Clin ; 29(2): 281-290, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38679439

RESUMEN

Bone Marrow Stimulation of osteochondral lesions of the talus has been shown to be a successful way to treat cartilage injuries. Newer data suggest that Bone Marrow Stimulation is best reserved for osteochondral lesions of the talus Sizes Less Than 107.4 mm2 in area. Additionally, newer smaller and deeper techniques to perform bone marrow stimulation have resulted in less subchondral bone damage, less cancellous compaction, and superior bone marrow access with multiple trabecular access channels. Biologic adjuvants such as platelet-rich plasma (PRP), hyaluronic acid (HA), and bone marrow aspirate concentrate (BMAC) may lead to better functional outcomes when used concomitant to bone marrow stimulation.


Asunto(s)
Astrágalo , Humanos , Astrágalo/lesiones , Astrágalo/cirugía , Cartílago Articular/lesiones , Cartílago Articular/cirugía , Cartílago Articular/fisiología , Plasma Rico en Plaquetas , Médula Ósea , Regeneración Ósea/fisiología
13.
Biomater Adv ; 161: 213867, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38669824

RESUMEN

Graphene and graphene oxide (GO), due to their unique chemical and physical properties, possess biochemical characteristics that can trigger intercellular signals promoting tissue regeneration. Clinical applications of thin GO-derived sheets have inspired the development of various tissue regeneration and repair approaches. In this study, we demonstrate that ultrathin sheets of plasma-functionalized and reduced GO, with the oxygen content ranging from 3.2 % to 22 % and the nitrogen content from 0 % to 8.3 %, retain their essential mechanical and molecular integrity, and exhibit robust potential for regenerating bone tissue and blood vessels across multiple cellular and animal models. Initially, we observed the growth of blood vessels and bone tissue in vitro using these functionalized GO sheets on human adipose-derived mesenchymal stem cells and umbilical vein endothelial cells. Remarkably, our study indicates a 2.5-fold increase in mineralization and two-fold increase in tubule formation even in media lacking osteogenic and angiogenic supplements. Subsequently, we observed the initiation, conduction, and formation of bone and blood vessels in a rat tibial osteotomy model, evident from a marked 4-fold increase in the volume of low radio-opacity bone tissue and a significant elevation in connectivity density, all without the use of stem cells or growth factors. Finally, we validated these findings in a mouse critical-size calvarial defect model (33 % higher healing rate) and a rat skin lesion model (up to 2.5-fold increase in the number of blood vessels, and 35 % increase in blood vessels diameter). This study elucidates the pro-osteogenic and pro-angiogenic properties of both pristine and plasma-treated GO ultrathin films. These properties suggest their significant potential for clinical applications, and as valuable biomaterials for investigating fundamental aspects of bone and blood vessel regeneration.


Asunto(s)
Regeneración Ósea , Grafito , Células Endoteliales de la Vena Umbilical Humana , Células Madre Mesenquimatosas , Animales , Grafito/química , Humanos , Ratas , Regeneración Ósea/efectos de los fármacos , Regeneración Ósea/fisiología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Osteogénesis/efectos de los fármacos , Osteogénesis/fisiología , Ratones , Vasos Sanguíneos , Ratas Sprague-Dawley , Huesos/irrigación sanguínea , Huesos/efectos de los fármacos , Gases em Plasma/farmacología , Gases em Plasma/química , Tibia/irrigación sanguínea , Neovascularización Fisiológica/efectos de los fármacos , Ingeniería de Tejidos/métodos
14.
Calcif Tissue Int ; 114(6): 625-637, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38643416

RESUMEN

Loss of p21 leads to increased bone formation post-injury; however, the mechanism(s) by which this occurs remains undetermined. E2f1 is downstream of p21 and as a transcription factor can act directly on gene expression; yet it is unknown if E2f1 plays a role in the osteogenic effects observed when p21 is differentially regulated. In this study we aimed to investigate the interplay between p21 and E2f1 and determine if the pro-regenerative osteogenic effects observed with the loss of p21 are E2f1 dependent. To accomplish this, we employed knockout p21 and E2f1 mice and additionally generated a p21/E2f1 double knockout. These mice underwent burr-hole injuries to their proximal tibiae and healing was assessed over 7 days via microCT imaging. We found that p21 and E2f1 play distinct roles in bone regeneration where the loss of p21 increased trabecular bone formation and loss of E2f1 increased cortical bone formation, yet loss of E2f1 led to poorer bone repair overall. Furthermore, when E2f1 was absent, either individually or simultaneously with p21, there was a dramatic decrease of the number of osteoblasts, osteoclasts, and chondrocytes at the site of injury compared to p21-/- and C57BL/6 mice. Together, these results suggest that E2f1 regulates the cell populations required for bone repair and has a distinct role in bone formation/repair compared to p21-/-E2f1-/-. These results highlight the possibility of cell cycle and/or p21/E2f1 being potential druggable targets that could be leveraged in clinical therapies to improve bone healing in pathologies such as osteoporosis.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Factor de Transcripción E2F1 , Ratones Endogámicos C57BL , Ratones Noqueados , Osteogénesis , Animales , Factor de Transcripción E2F1/metabolismo , Factor de Transcripción E2F1/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Osteogénesis/fisiología , Ratones , Regeneración Ósea/fisiología , Osteoblastos/metabolismo
15.
Int J Biol Macromol ; 264(Pt 1): 130608, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38447840

RESUMEN

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.


Asunto(s)
Compuestos de Anilina , Quitosano , Quitosano/química , Osteogénesis , Andamios del Tejido/química , Regeneración Ósea/fisiología , Técnicas Electroquímicas , Ingeniería de Tejidos/métodos , Diferenciación Celular , Proliferación Celular , Poliésteres/química
16.
Adv Sci (Weinh) ; 11(16): e2304861, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38355304

RESUMEN

An ideal hydrogel for stem cell therapy would be injectable and efficiently promote stem cell proliferation and differentiation in body. Herein, an injectable, single-component hydrogel with hyaluronic acid (HA) modified with phenylboronic acid (PBA) and spermidine (SM) is introduced. The resulting HAps (HA-PBA-SM) hydrogel is based on the reversible crosslinking between the diol and the ionized PBA, which is stabilized by the SM. It has a shear-thinning property, enabling its injection through a syringe to form a stable hydrogel inside the body. In addition, HAps hydrogel undergoes a post-injection "self-curing," which stiffens the hydrogel over time. This property allows the HAps hydrogel to meet the physical requirements for stem cell therapy in rigid tissues, such as bone, while maintaining injectability. The hydrogel enabled favorable proliferation of human mesenchymal stem cells (hMSCs) and promoted their differentiation and mineralization. After the injection of hMSCs-containing HAps into a rat femoral defect model, efficient osteogenic differentiation of hMSCs and bone regeneration is observed. The study demonstrates that simple cationic modification of PBA-based hydrogel enabled efficient gelation with shear-thinning and self-curing properties, and it would be highly useful for stem cell therapy and in vivo bone regeneration.


Asunto(s)
Regeneración Ósea , Ácidos Borónicos , Diferenciación Celular , Hidrogeles , Células Madre Mesenquimatosas , Animales , Regeneración Ósea/fisiología , Ratas , Hidrogeles/química , Células Madre Mesenquimatosas/citología , Humanos , Ácido Hialurónico/química , Ratas Sprague-Dawley , Encapsulación Celular/métodos , Proliferación Celular , Osteogénesis/fisiología , Modelos Animales de Enfermedad , Espermidina/farmacología , Espermidina/química
17.
Adv Sci (Weinh) ; 11(17): e2309491, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38380490

RESUMEN

The regeneration of bone defects in diabetic patients still faces challenges, as the intrinsic healing process is impaired by hyperglycemia. Inspired by the discovery that the endoplasmic reticulum (ER) is in a state of excessive stress and dysfunction under hyperglycemia, leading to osteogenic disorder, a novel engineered exosome is proposed to modulate ER homeostasis for restoring the function of mesenchymal stem cells (MSCs). The results indicate that the constructed engineered exosomes efficiently regulate ER homeostasis and dramatically facilitate the function of MSCs in the hyperglycemic niche. Additionally, the underlying therapeutic mechanism of exosomes is elucidated. The results reveal that exosomes can directly provide recipient cells with SHP2 for the activation of mitophagy and elimination of mtROS, which is the immediate cause of ER dysfunction. To maximize the therapeutic effect of engineered exosomes, a high-performance hydrogel with self-healing, bioadhesive, and exosome-conjugating properties is applied to encapsulate the engineered exosomes for in vivo application. In vivo, evaluation in diabetic bone defect repair models demonstrates that the engineered exosomes delivering hydrogel system intensively enhance osteogenesis. These findings provide crucial insight into the design and biological mechanism of ER homeostasis-based tissue-engineering strategies for diabetic bone regeneration.


Asunto(s)
Regeneración Ósea , Retículo Endoplásmico , Exosomas , Homeostasis , Hidrogeles , Células Madre Mesenquimatosas , Exosomas/metabolismo , Regeneración Ósea/fisiología , Regeneración Ósea/genética , Animales , Homeostasis/fisiología , Hidrogeles/química , Ratones , Retículo Endoplásmico/metabolismo , Células Madre Mesenquimatosas/metabolismo , Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Experimental/metabolismo , Osteogénesis/fisiología , Modelos Animales de Enfermedad , Ingeniería de Tejidos/métodos , Masculino , Humanos
18.
Adv Healthc Mater ; 13(12): e2303600, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38303119

RESUMEN

Bone regenerative scaffolds with a bionic natural bone hierarchical porous structure provide a suitable microenvironment for cell migration and proliferation. Here, a bionic scaffold (DP-PLGA/HAp) with directional microchannels is prepared by combining 3D printing and directional freezing technology. The 3D printed framework provides structural support for new bone tissue growth, while the directional pore embedded in the scaffolds provides an express lane for cell migration and nutrition transport, facilitating cell growth and differentiation. The hierarchical porous scaffolds achieve rapid infiltration and adhesion of bone marrow mesenchymal stem cells (BMSCs) and improve the expression of osteogenesis-related genes. The rabbit cranial defect experiment presents significant new bone formation, demonstrating that DP-PLGA/HAp offers an effective means to guide cranial bone regeneration. The combination of 3D printing and directional freezing technology might be a promising strategy for developing bone regenerative biomaterials.


Asunto(s)
Regeneración Ósea , Células Madre Mesenquimatosas , Osteogénesis , Impresión Tridimensional , Andamios del Tejido , Regeneración Ósea/fisiología , Animales , Conejos , Andamios del Tejido/química , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Osteogénesis/efectos de los fármacos , Osteogénesis/fisiología , Porosidad , Diferenciación Celular , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Ingeniería de Tejidos/métodos , Proliferación Celular , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Durapatita/química
19.
Stem Cell Res Ther ; 15(1): 33, 2024 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-38321490

RESUMEN

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.


Asunto(s)
Vesículas Extracelulares , Células Madre Mesenquimatosas , Humanos , Ratas , Masculino , Animales , Osteogénesis/genética , Proteómica , Células Madre Mesenquimatosas/metabolismo , Ratas Endogámicas Lew , Regeneración Ósea/fisiología , Diferenciación Celular , Vesículas Extracelulares/metabolismo
20.
ACS Nano ; 18(10): 7504-7520, 2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38412232

RESUMEN

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.


Asunto(s)
Exosomas , Células Madre Mesenquimatosas , Ratas , Animales , Exosomas/metabolismo , Diferenciación Celular/genética , Porosidad , Fosfatidilinositol 3-Quinasas , Factor de Crecimiento Nervioso/análisis , Factor de Crecimiento Nervioso/metabolismo , Factor de Crecimiento Nervioso/farmacología , Regeneración Ósea/fisiología , Osteogénesis , Impresión Tridimensional
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA