Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.896
Filtrar
1.
Development ; 151(17)2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-39250530

RESUMEN

Developing bones can adapt their shape in response to mechanical stresses from neighbouring growing organs. In a new study, Koichi Matsuo and colleagues examine how bone-forming osteoblasts and bone-resorbing osteoclasts coordinate growth in the mouse fibula. They describe the process called 'endo-forming trans-pairing', where bone resorption by osteoclasts in the outer periosteum is paired with bone formation by osteoblasts in the inner endosteum to shape the growing bone. To learn more about the story behind the paper, we caught up with first author Yukiko Kuroda and the corresponding author Koichi Matsuo, Professor at the School of Medicine, Keio University, Japan.


Asunto(s)
Huesos , Microscopía/métodos , Huesos/citología , Osteoclastos/citología , Osteoclastos/metabolismo , Osteoblastos/citología , Osteoblastos/metabolismo , Nervio Ciático , Desarrollo Óseo , Animales , Morfogénesis
2.
Biofabrication ; 16(4)2024 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-39121892

RESUMEN

The viscosity of gelatin methacryloyl (GelMA)-based bioinks generates shear stresses throughout the printing process that can affect cell integrity, reduce cell viability, cause morphological changes, and alter cell functionality. This study systematically investigated the impact of the viscosity of GelMA-gelatin bioinks on osteoblast-like cells in 2D and 3D culture conditions. Three bioinks with low, medium, and high viscosity prepared by supplementing a 5% GelMA solution with different concentrations of gelatin were evaluated. Cell responses were studied in a 2D environment after printing and incubation in non-cross-linked bioinks that caused the gelatin and GelMA to dissolve and release cells for attachment to tissue culture plates. The increased viscosity of the bioinks significantly affected cell area and aspect ratio. Cells printed using the bioink with medium viscosity exhibited greater metabolic activity and proliferation rate than those printed using the high viscosity bioink and even the unprinted control cells. Additionally, cells printed using the bioink with high viscosity demonstrated notably elevated expression levels of alkaline phosphatase and bone morphogenetic protein-2 genes. In the 3D condition, the printed cell-laden hydrogels were photo-cross-linked prior to incubation. The medium viscosity bioink supported greater cell proliferation compared to the high viscosity bioink. However, there were no significant differences in the expression of osteogenic markers between the medium and high viscosity bioinks. Therefore, the choice between medium and high viscosity bioinks should be based on the desired outcomes and objectives of the bone tissue engineering application. Furthermore, the bioprinting procedure with the medium viscosity bioink was used as an automated technique for efficiently seeding cells onto 3D printed porous titanium scaffolds for bone tissue engineering purposes.


Asunto(s)
Bioimpresión , Gelatina , Tinta , Metacrilatos , Gelatina/química , Viscosidad , Metacrilatos/química , Bioimpresión/métodos , Impresión Tridimensional , Osteoblastos/citología , Osteoblastos/metabolismo , Osteoblastos/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ingeniería de Tejidos , Línea Celular , Animales , Andamios del Tejido/química , Humanos , Supervivencia Celular/efectos de los fármacos , Huesos/citología
3.
Int J Mol Sci ; 25(16)2024 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-39201445
4.
Nature ; 632(8024): 357-365, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38987585

RESUMEN

In lactating mothers, the high calcium (Ca2+) demand for milk production triggers significant bone loss1. Although oestrogen normally counteracts excessive bone resorption by promoting bone formation, this sex steroid drops precipitously during this postpartum period. Here we report that brain-derived cellular communication network factor 3 (CCN3) secreted from KISS1 neurons of the arcuate nucleus (ARCKISS1) fills this void and functions as a potent osteoanabolic factor to build bone in lactating females. We began by showing that our previously reported female-specific, dense bone phenotype2 originates from a humoral factor that promotes bone mass and acts on skeletal stem cells to increase their frequency and osteochondrogenic potential. This circulatory factor was then identified as CCN3, a brain-derived hormone from ARCKISS1 neurons that is able to stimulate mouse and human skeletal stem cell activity, increase bone remodelling and accelerate fracture repair in young and old mice of both sexes. The role of CCN3 in normal female physiology was revealed after detecting a burst of CCN3 expression in ARCKISS1 neurons coincident with lactation. After reducing CCN3 in ARCKISS1 neurons, lactating mothers lost bone and failed to sustain their progeny when challenged with a low-calcium diet. Our findings establish CCN3 as a potentially new therapeutic osteoanabolic hormone for both sexes and define a new maternal brain hormone for ensuring species survival in mammals.


Asunto(s)
Densidad Ósea , Huesos , Encéfalo , Hormonas , Madres , Proteína Hiperexpresada del Nefroblastoma , Osteogénesis , Adolescente , Animales , Femenino , Humanos , Masculino , Ratones , Envejecimiento , Núcleo Arqueado del Hipotálamo/citología , Núcleo Arqueado del Hipotálamo/metabolismo , Huesos/citología , Huesos/metabolismo , Remodelación Ósea , Resorción Ósea/metabolismo , Encéfalo/citología , Encéfalo/metabolismo , Calcio/administración & dosificación , Calcio/metabolismo , Lactancia/metabolismo , Ratones Endogámicos C57BL , Neuronas/metabolismo , Células Madre/metabolismo , Células Madre/citología , Proteína Hiperexpresada del Nefroblastoma/metabolismo , Hormonas/metabolismo
5.
Tissue Cell ; 90: 102475, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39059134

RESUMEN

Decellularized cortical bone powder derived from adult animals has been shown to induce bone remodeling. Furthermore, it is increasingly evident that the extracellular matrix (ECM) within decellularized tissues differs depending on the source tissue and the age of the animal, leading to distinct effects on cells. In this study, we prepared powders from decellularized fetal and adult porcine bone tissues and conducted biological analyses to determine if the decellularized tissue could induce adipose-derived stem cell differentiation. Decellularized fetal tissues and adult cortical bone were converted into powder by cryomilling, but decellularized adult bone marrow and cartilage were not powdered through this process. In vitro assessments revealed that decellularized fetal tissues, decellularized adult cartilage extract, and decellularized fetal cartilage powder can induce osteoblast differentiation. This study suggests that decellularized fetal bone tissues and adult cartilage contain ECM components that can induce osteoblast differentiation. Additionally, it highlights the utility of decellularized fetal cartilage powder for bone reconstruction.


Asunto(s)
Cartílago , Diferenciación Celular , Matriz Extracelular , Feto , Osteogénesis , Animales , Cartílago/citología , Cartílago/metabolismo , Matriz Extracelular/metabolismo , Porcinos , Feto/citología , Huesos/citología , Osteoblastos/citología , Osteoblastos/metabolismo , Matriz Extracelular Descelularizada/farmacología
6.
J Biomater Sci Polym Ed ; 35(13): 2090-2114, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38953859

RESUMEN

Fe-Ca-SAPO-34/CS/PANI, a novel hybrid bio-composite scaffold with potential application in dental tissue engineering, was prepared by freeze drying technique. The scaffold was characterized using FT-IR and SEM methods. The effects of PANI on the physicochemical properties of the Fe-Ca-SAPO-34/CS scaffold were investigated, including changes in swelling ratio, mechanical behavior, density, porosity, biodegradation, and biomineralization. Compared to the Fe-Ca-SAPO-34/CS scaffold, adding PANI decreased the pore size, porosity, swelling ratio, and biodegradation, while increasing the mechanical strength and biomineralization. Cell viability, cytotoxicity, and adhesion of human dental pulp stem cells (hDPSCs) on the scaffolds were investigated by MTT assay and SEM. The Fe-Ca-SAPO-34/CS/PANI scaffold promoted hDPSC proliferation and osteogenic differentiation compared to the Fe-Ca-SAPO-34/CS scaffold. Alizarin red staining, alkaline phosphatase activity, and qRT-PCR results revealed that Fe-Ca-SAPO-34/CS/PANI triggered osteoblast/odontoblast differentiation in hDPSCs through the up-regulation of osteogenic marker genes BGLAP, RUNX2, and SPARC. The significance of this study lies in developing a novel scaffold that synergistically combines the beneficial properties of Fe-Ca-SAPO-34, chitosan, and PANI to create an optimized microenvironment for dental tissue regeneration. These findings highlight the potential of the Fe-Ca-SAPO-34/CS/PANI scaffold as a promising biomaterial for dental tissue engineering applications, paving the way for future research and clinical translation in regenerative dentistry.


Asunto(s)
Pulpa Dental , Osteogénesis , Células Madre , Ingeniería de Tejidos , Andamios del Tejido , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Humanos , Pulpa Dental/citología , Osteogénesis/efectos de los fármacos , Células Madre/citología , Porosidad , Proliferación Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Nanoestructuras/química , Huesos/citología , Supervivencia Celular/efectos de los fármacos , Quitosano/química , Ensayo de Materiales , Adhesión Celular/efectos de los fármacos , Hierro/química
7.
Sci Rep ; 14(1): 12721, 2024 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-38830871

RESUMEN

Surface structure plays a crucial role in determining cell behavior on biomaterials, influencing cell adhesion, proliferation, differentiation, as well as immune cells and macrophage polarization. While grooves and ridges stimulate M2 polarization and pits and bumps promote M1 polarization, these structures do not accurately mimic the real bone surface. Consequently, the impact of mimicking bone surface topography on macrophage polarization remains unknown. Understanding the synergistic sequential roles of M1 and M2 macrophages in osteoimmunomodulation is crucial for effective bone tissue engineering. Thus, exploring the impact of bone surface microstructure mimicking biomaterials on macrophage polarization is critical. In this study, we aimed to sequentially activate M1 and M2 macrophages using Poly-L-Lactic acid (PLA) membranes with bone surface topographical features mimicked through the soft lithography technique. To mimic the bone surface topography, a bovine femur was used as a model surface, and the membranes were further modified with collagen type-I and hydroxyapatite to mimic the bone surface microenvironment. To determine the effect of these biomaterials on macrophage polarization, we conducted experimental analysis that contained estimating cytokine release profiles and characterizing cell morphology. Our results demonstrated the potential of the hydroxyapatite-deposited bone surface-mimicked PLA membranes to trigger sequential and synergistic M1 and M2 macrophage polarizations, suggesting their ability to achieve osteoimmunomodulatory macrophage polarization for bone tissue engineering applications. Although further experimental studies are required to completely investigate the osteoimmunomodulatory effects of these biomaterials, our results provide valuable insights into the potential advantages of biomaterials that mimic the complex microenvironment of bone surfaces.


Asunto(s)
Macrófagos , Poliésteres , Propiedades de Superficie , Animales , Macrófagos/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Bovinos , Poliésteres/química , Ratones , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Ingeniería de Tejidos/métodos , Durapatita/química , Citocinas/metabolismo , Huesos/citología , Diferenciación Celular/efectos de los fármacos , Activación de Macrófagos/efectos de los fármacos , Adhesión Celular/efectos de los fármacos , Células RAW 264.7 , Polaridad Celular/efectos de los fármacos , Fémur , Colágeno Tipo I/metabolismo
8.
Stem Cell Res Ther ; 15(1): 183, 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38902814

RESUMEN

In the realm of studying joint-related diseases, there is a continuous quest for more accurate and representative models. Recently, regenerative medicine and tissue engineering have seen a growing interest in utilizing organoids as powerful tools for studying complex biological systems in vitro. Organoids, three-dimensional structures replicating the architecture and function of organs, provide a unique platform for investigating disease mechanisms, drug responses, and tissue regeneration. The surge in organoid research is fueled by the need for physiologically relevant models to bridge the gap between traditional cell cultures and in vivo studies. Osteochondral organoids have emerged as a promising avenue in this pursuit, offering a better platform to mimic the intricate biological interactions within bone and cartilage. This review explores the significance of osteochondral organoids and the need for their development in advancing our understanding and treatment of bone and cartilage-related diseases. It summarizes osteochondral organoids' insights and research progress, focusing on their composition, materials, cell sources, and cultivation methods, as well as the concept of organoids on chips and application scenarios. Additionally, we address the limitations and challenges these organoids face, emphasizing the necessity for further research to overcome these obstacles and facilitate orthopedic regeneration.


Asunto(s)
Organoides , Ingeniería de Tejidos , Organoides/citología , Organoides/metabolismo , Humanos , Ingeniería de Tejidos/métodos , Animales , Cartílago/citología , Medicina Regenerativa/métodos , Huesos/citología , Huesos/fisiología
9.
Stem Cell Res Ther ; 15(1): 156, 2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38816830

RESUMEN

Mesenchymal stem cells (MSCs) are multipotent cells that can differentiate into cells of different lineages to form mesenchymal tissues, which are promising in regard to treatment for bone diseases. Their osteogenic differentiation is under the tight regulation of intrinsic and extrinsic factors. Transforming growth factor ß (TGF-ß) is an essential growth factor in bone metabolism, which regulates the differentiation of MSCs. However, published studies differ in their views on whether TGF-ß signaling regulates the osteogenic differentiation of MSCs positively or negatively. The controversial results have not been summarized systematically and the related explanations are required. Therefore, we reviewed the basics of TGF-ß signaling and summarized how each of three isoforms regulates osteogenic differentiation. Three isoforms of TGF-ß (TGF-ß1/ß2/ß3) play distinct roles in regulating osteogenic differentiation of MSCs. Additionally, other possible sources of conflicts are summarized here. Further understanding of TGF-ß signaling regulation in MSCs may lead to new applications to promote bone regeneration and improve therapies for bone diseases.


Asunto(s)
Huesos , Diferenciación Celular , Células Madre Mesenquimatosas , Osteogénesis , Transducción de Señal , Factor de Crecimiento Transformador beta , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Humanos , Factor de Crecimiento Transformador beta/metabolismo , Huesos/metabolismo , Huesos/citología , Animales
10.
Int J Mol Sci ; 25(10)2024 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-38791362

RESUMEN

In the field of biomaterials for prosthetic reconstructive surgery, there is the lack of advanced innovative methods to investigate the potentialities of smart biomaterials before in vivo tests. Despite the complex osteointegration process being difficult to recreate in vitro, this study proposes an advanced in vitro tissue culture model of osteointegration using human bone. Cubic samples of trabecular bone were harvested, as waste material, from hip arthroplasty; inner cylindrical defects were created and assigned to the following groups: (1) empty defects (CTRneg); (2) defects implanted with a cytotoxic copper pin (CTRpos); (3) defects implanted with standard titanium pins (Ti). Tissues were dynamically cultured in mini rotating bioreactors and assessed weekly for viability and sterility. After 8 weeks, immunoenzymatic, microtomographic, histological, and histomorphometric analyses were performed. The model was able to simulate the effects of implantation of the materials, showing a drop in viability in CTR+, while Ti appears to have a trophic effect on bone. MicroCT and a histological analysis supported the results, with signs of matrix and bone deposition at the Ti implant site. Data suggest the reliability of the tested model in recreating the osteointegration process in vitro with the aim of reducing and refining in vivo preclinical models.


Asunto(s)
Oseointegración , Técnicas de Cultivo de Tejidos , Titanio , Humanos , Técnicas de Cultivo de Tejidos/métodos , Microtomografía por Rayos X , Huesos/citología , Materiales Biocompatibles , Prótesis e Implantes , Hueso Esponjoso/citología
11.
Biofabrication ; 16(3)2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38697073

RESUMEN

Osteochondral tissue (OC) repair remains a significant challenge in the field of musculoskeletal tissue engineering. OC tissue displays a gradient structure characterized by variations in both cell types and extracellular matrix components, from cartilage to the subchondral bone. These functional gradients observed in the native tissue have been replicated to engineer OC tissuein vitro. While diverse fabrication methods have been employed to create these microenvironments, emulating the natural gradients and effective regeneration of the tissue continues to present a significant challenge. In this study, we present the design and development of CMC-silk interpenetrating (IPN) hydrogel with opposing dual biochemical gradients similar to native tissue with the aim to regenerate the complete OC unit. The gradients of biochemical cues were generated using an in-house-built extrusion system. Firstly, we fabricated a hydrogel that exhibits a smooth transition of sulfated carboxymethyl cellulose (sCMC) and TGF-ß1 (SCT gradient hydrogel) from the upper to the lower region of the IPN hydrogel to regenerate the cartilage layer. Secondly, a hydrogel with a hydroxyapatite (HAp) gradient (HAp gradient hydrogel) from the lower to the upper region was fabricated to facilitate the regeneration of the subchondral bone layer. Subsequently, we developed a dual biochemical gradient hydrogel with a smooth transition of sCMC + TGF-ß1 and HAp gradients in opposing directions, along with a blend of both biochemical cues in the middle. The results showed that the dual biochemical gradient hydrogels with biochemical cues corresponding to the three zones (i.e. cartilage, interface and bone) of the OC tissue led to differentiation of bone-marrow-derived mesenchymal stem cells to zone-specific lineages, thereby demonstrating their efficacy in directing the fate of progenitor cells. In summary, our study provided a simple and innovative method for incorporating gradients of biochemical cues into hydrogels. The gradients of biochemical cues spatially guided the differentiation of stem cells and facilitated tissue growth, which would eventually lead to the regeneration of the entire OC tissue with a smooth transition from cartilage (soft) to bone (hard) tissues. This promising approach is translatable and has the potential to generate numerous biochemical and biophysical gradients for regeneration of other interface tissues, such as tendon-to-muscle and ligament-to-bone.


Asunto(s)
Hidrogeles , Ingeniería de Tejidos , Hidrogeles/química , Animales , Andamios del Tejido/química , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Condrogénesis/efectos de los fármacos , Cartílago/citología , Cartílago/fisiología , Diferenciación Celular/efectos de los fármacos , Huesos/citología , Durapatita/química , Durapatita/farmacología
13.
J Histochem Cytochem ; 72(6): 373-385, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38804525

RESUMEN

Osteoporosis poses a significant global health concern, affecting both the elderly and young individuals, including athletes. Despite the development of numerous antiosteoporotic drugs, addressing the unique needs of young osteoporosis patients remains challenging. This study focuses on young rats subjected to ovariectomy (OVX) to explore the impact of high-molecular-weight hyaluronan (HA) on preventing OVX-induced osteoporosis. Twenty-four rats underwent OVX, while 12 underwent sham procedures (sham control group). Among the OVX rats, half received subcutaneous injections of HA (MW: 2700 kDa) at 10 mg/kg/week into their backs (OVX-HA group), whereas the other half received saline injections (0.5 ml/week) at the same site (OVX-saline group). OVX-HA group exhibited significantly higher percentages of osteoclast surface (Oc. S/BS), osteoblast surface per bone surface (Ob. S/BS), and bone volume/tissue volume (BV/TV) compared with OVX-saline group at the same age. The proportions of Ob. S/BS and BV/TV in the OVX-HA group closely resembled those of the sham control group, whereas the proportion of Oc. S/BS in the OVX-HA group was notably higher than that in the sham control group. In summary, the administration of HA significantly mitigated bone resorption and enhanced bone formation, suggesting a crucial role for HA in the treatment of young adult osteoporosis.


Asunto(s)
Resorción Ósea , Ácido Hialurónico , Osteogénesis , Osteoporosis , Femenino , Ratas , Huesos/citología , Huesos/efectos de los fármacos , Huesos/patología , Conservadores de la Densidad Ósea/farmacología , Conservadores de la Densidad Ósea/uso terapéutico , Resorción Ósea/tratamiento farmacológico , Modelos Animales de Enfermedad , Ácido Hialurónico/farmacología , Ácido Hialurónico/uso terapéutico , Osteoblastos/efectos de los fármacos , Osteoclastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Osteoporosis/tratamiento farmacológico , Ovariectomía , Ratas Sprague-Dawley , Animales
14.
J Bone Miner Res ; 39(6): 633-654, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-38696703

RESUMEN

Bone development, growth, and repair are complex processes involving various cell types and interactions, with central roles played by skeletal stem and progenitor cells. Recent research brought new insights into the skeletal precursor populations that mediate intramembranous and endochondral bone development. Later in life, many of the cellular and molecular mechanisms determining development are reactivated upon fracture, with powerful trauma-induced signaling cues triggering a variety of postnatal skeletal stem/progenitor cells (SSPCs) residing near the bone defect. Interestingly, in this injury context, the current evidence suggests that the fates of both SSPCs and differentiated skeletal cells can be considerably flexible and dynamic, and that multiple cell sources can be activated to operate as functional progenitors generating chondrocytes and/or osteoblasts. The combined implementation of in vivo lineage tracing, cell surface marker-based cell selection, single-cell molecular analyses, and high-resolution in situ imaging has strongly improved our insights into the diversity and roles of developmental and reparative stem/progenitor subsets, while also unveiling the complexity of their dynamics, hierarchies, and relationships. Albeit incompletely understood at present, findings supporting lineage flexibility and possibly plasticity among sources of osteogenic cells challenge the classical dogma of a single primitive, self-renewing, multipotent stem cell driving bone tissue formation and regeneration from the apex of a hierarchical and strictly unidirectional differentiation tree. We here review the state of the field and the newest discoveries in the origin, identity, and fates of skeletal progenitor cells during bone development and growth, discuss the contributions of adult SSPC populations to fracture repair, and reflect on the dynamism and relationships among skeletal precursors and differentiated cell lineages. Further research directed at unraveling the heterogeneity and capacities of SSPCs, as well as the regulatory cues determining their fate and functioning, will offer vital new options for clinical translation toward compromised fracture healing and bone regenerative medicine.


Skeletal progenitor cells are crucial for bone development and growth, as they provide the cellular building blocks (chondrocytes and osteoblasts) that form the cartilage and bone tissues that the skeleton is composed of. In adult life, the occurrence of a bone fracture reactivates similar tissue-forming mechanisms, starting with the trauma triggering various postnatal skeletal stem/progenitor cells (SSPCs) residing near the bone defect to divide and migrate. These cells subsequently generate functional fracture-repairing cells by differentiating into mature chondrocytes and/or osteoblasts. In recent years, the combined use of various advanced research approaches and new techniques has strongly improved our insights into the origin, identity, fates, and roles of developmental and reparative skeletal stem cells and progenitor subsets. Concomitantly, this research also unveiled considerable complexity in their dynamics, diversity, hierarchies, and relationships, which is incompletely understood at present. In this review, we discuss the state of the field and the newest discoveries in the identity and roles of skeletal stem and progenitor cells mediating bone development, growth, and repair. Further research on these cell populations, including determining their exact nature, fate, and functioning, and how they can be harvested and regulated, is critical to develop new treatments for non-healing fractures.


Asunto(s)
Desarrollo Óseo , Células Madre , Humanos , Animales , Células Madre/metabolismo , Células Madre/citología , Huesos/fisiología , Huesos/citología , Regeneración Ósea , Diferenciación Celular , Osteogénesis
15.
Adv Mater ; 36(30): e2309875, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38642033

RESUMEN

Addressing large bone defects remains a significant challenge owing to the inherent limitations in self-healing capabilities, resulting in prolonged recovery and suboptimal regeneration. Although current clinical solutions are available, they have notable shortcomings, necessitating more efficacious approaches to bone regeneration. Organoids derived from stem cells show great potential in this field; however, the development of bone organoids has been hindered by specific demands, including the need for robust mechanical support provided by scaffolds and hybrid extracellular matrices (ECM). In this context, bioprinting technologies have emerged as powerful means of replicating the complex architecture of bone tissue. The research focused on the fabrication of a highly intricate bone ECM analog using a novel bioink composed of gelatin methacrylate/alginate methacrylate/hydroxyapatite (GelMA/AlgMA/HAP). Bioprinted scaffolds facilitate the long-term cultivation and progressive maturation of extensive bioprinted bone organoids, foster multicellular differentiation, and offer valuable insights into the initial stages of bone formation. The intrinsic self-mineralizing quality of the bioink closely emulates the properties of natural bone, empowering organoids with enhanced bone repair for both in vitro and in vivo applications. This trailblazing investigation propels the field of bone tissue engineering and holds significant promise for its translation into practical applications.


Asunto(s)
Bioimpresión , Durapatita , Organoides , Ingeniería de Tejidos , Andamios del Tejido , Durapatita/química , Organoides/citología , Organoides/metabolismo , Ingeniería de Tejidos/métodos , Humanos , Bioimpresión/métodos , Andamios del Tejido/química , Gelatina/química , Alginatos/química , Matriz Ósea/química , Matriz Ósea/metabolismo , Animales , Tinta , Osteogénesis , Metacrilatos/química , Regeneración Ósea , Huesos/citología , Calcificación Fisiológica
16.
Int J Mol Sci ; 25(7)2024 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-38612687

RESUMEN

Dimethyloxalylglycine (DMOG) has been found to stimulate osteogenesis and angiogenesis of stem cells, promoting neo-angiogenesis in bone tissue regeneration. In this review, we conducted a comprehensive search of the literature to investigate the effects of DMOG on osteogenesis and bone regeneration. We screened the studies based on specific inclusion criteria and extracted relevant information from both in vitro and in vivo experiments. The risk of bias in animal studies was evaluated using the SYRCLE tool. Out of the 174 studies retrieved, 34 studies met the inclusion criteria (34 studies were analyzed in vitro and 20 studies were analyzed in vivo). The findings of the included studies revealed that DMOG stimulated stem cells' differentiation toward osteogenic, angiogenic, and chondrogenic lineages, leading to vascularized bone and cartilage regeneration. Addtionally, DMOG demonstrated therapeutic effects on bone loss caused by bone-related diseases. However, the culture environment in vitro is notably distinct from that in vivo, and the animal models used in vivo experiments differ significantly from humans. In summary, DMOG has the ability to enhance the osteogenic and angiogenic differentiation potential of stem cells, thereby improving bone regeneration in cases of bone defects. This highlights DMOG as a potential focus for research in the field of bone tissue regeneration engineering.


Asunto(s)
Aminoácidos Dicarboxílicos , Regeneración Ósea , Diferenciación Celular , Osteogénesis , Células Madre , Regeneración Ósea/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Humanos , Diferenciación Celular/efectos de los fármacos , Animales , Aminoácidos Dicarboxílicos/farmacología , Células Madre/efectos de los fármacos , Células Madre/citología , Células Madre/metabolismo , Ingeniería de Tejidos/métodos , Huesos/efectos de los fármacos , Huesos/citología , Glicina/análogos & derivados
17.
Int J Mol Sci ; 25(8)2024 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-38673812

RESUMEN

Here, we report on the development of a cost-effective, well-characterized three-dimensional (3D) model of bone homeostasis derived from commonly available stocks of immortalized murine cell lines and laboratory reagents. This 3D murine-cell-derived bone organoid model (3D-mcBOM) is adaptable to a range of contexts and can be used in conjunction with surrogates of osteoblast and osteoclast function to study cellular and molecular mechanisms that affect bone homeostasis in vitro or to augment in vivo models of physiology or disease. The 3D-mcBOM was established using a pre-osteoblast murine cell line, which was seeded into a hydrogel extracellular matrix (ECM) and differentiated into functional osteoblasts (OBs). The OBs mineralized the hydrogel ECM, leading to the deposition and consolidation of hydroxyapatite into bone-like organoids. Fourier-transform infrared (FTIR) spectroscopy confirmed that the mineralized matrix formed in the 3D-mcBOM was bone. The histological staining of 3D-mcBOM samples indicated a consistent rate of ECM mineralization. Type I collagen C-telopeptide (CTX1) analysis was used to evaluate the dynamics of OC differentiation and activity. Reliable 3D models of bone formation and homeostasis align with current ethical trends to reduce the use of animal models. This functional model of bone homeostasis provides a cost-effective model system using immortalized cell lines and easily procured supplemental compounds, which can be assessed by measuring surrogates of OB and OC function to study the effects of various stimuli in future experimental evaluations of bone homeostasis.


Asunto(s)
Diferenciación Celular , Matriz Extracelular , Organoides , Osteoblastos , Osteogénesis , Animales , Ratones , Organoides/citología , Organoides/metabolismo , Osteoblastos/citología , Osteoblastos/metabolismo , Matriz Extracelular/metabolismo , Huesos/citología , Huesos/metabolismo , Línea Celular , Colágeno Tipo I/metabolismo , Hidrogeles/química , Calcificación Fisiológica , Técnicas de Cultivo Tridimensional de Células/métodos , Modelos Biológicos
18.
Stem Cell Rev Rep ; 20(4): 1106-1120, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38472643

RESUMEN

The regenerative function of stem cells is compromised when the proportion of senescent stem cells increases with ageing advance. Therefore, combating stem cell senescence is of great importance for stem cell-based tissue engineering in the elderly, but remains largely unexplored. Osteopontin (OPN), a glycosylated phosphoprotein, is one of the key extracellular matrix molecules in bone tissue. OPN activates various signalling pathways and modulates cellular activities, including cell senescence. However, the role of OPN in stem cell senescence remains largely unknown. This study aims to investigate if OPN modulates cell senescence and bone regenerative function in human adipose-derived mesenchymal stem cells (ASCs), and to determine the underlying mechanisms. We first developed a senescent ASC model using serial passaging until passage 10 (P10), in which senescent cells were characterised by reduced proliferation and osteogenic differentiation capacity compared to P4 ASCs. The conditioned medium from P10 ASCs exhibited a diminished trophic effect on human osteoblasts (HOBs), compared to that from P4 ASCs. P10 ASCs on OPN-coated surface showed rejuvenated phenotype and enhanced osteogenic differentiation. The conditioned medium from P10 ASCs on OPN-coating improved trophic effects on HOBs. OPN regulated the morphology of senescent ASCs, transforming them from a more rounded and flattened cell shape to an elongated shape with a smaller area. These findings demonstrated the effects of OPN in restoring senescent ASCs functions, possibly through a mechanism that involves the modulation of cell morphology, indicating that OPN might hold a great potential for rejuvenating senescent stem cells and could potentially open a new venue for regenerating bone tissue in age-related diseases.


Asunto(s)
Tejido Adiposo , Regeneración Ósea , Células Madre Mesenquimatosas , Osteogénesis , Osteopontina , Humanos , Tejido Adiposo/citología , Huesos/citología , Huesos/metabolismo , Diferenciación Celular , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Senescencia Celular , Medios de Cultivo Condicionados/farmacología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Osteoblastos/metabolismo , Osteoblastos/citología , Osteogénesis/efectos de los fármacos , Osteopontina/metabolismo
20.
Protein Cell ; 15(8): 559-574, 2024 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-38442300

RESUMEN

Tissue-resident stem cells are essential for development and repair, and in the skeleton, this function is fulfilled by recently identified skeletal stem cells (SSCs). However, recent work has identified that SSCs are not monolithic, with long bones, craniofacial sites, and the spine being formed by distinct stem cells. Recent studies have utilized techniques such as fluorescence-activated cell sorting, lineage tracing, and single-cell sequencing to investigate the involvement of SSCs in bone development, homeostasis, and disease. These investigations have allowed researchers to map the lineage commitment trajectory of SSCs in different parts of the body and at different time points. Furthermore, recent studies have shed light on the characteristics of SSCs in both physiological and pathological conditions. This review focuses on discussing the spatiotemporal distribution of SSCs and enhancing our understanding of the diversity and plasticity of SSCs by summarizing recent discoveries.


Asunto(s)
Desarrollo Óseo , Homeostasis , Células Madre , Humanos , Animales , Células Madre/citología , Células Madre/metabolismo , Huesos/citología , Huesos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...