Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Int J Mol Sci ; 24(4)2023 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-36835079

RESUMEN

The bone cancer osteosarcoma, found mainly in adolescents, routinely forms around the growth plate/metaphysis of long bones. Bone marrow composition changes with age, shifting from a more hematopoietic to an adipocyte-rich tissue. This conversion occurs in the metaphysis during adolescence, implicating a link between bone marrow conversion and osteosarcoma initiation. To assess this, the tri-lineage differentiation potential of human bone marrow stromal cells (HBMSCs) isolated from the femoral diaphysis/metaphysis (FD) and epiphysis (FE) was characterized and compared to two osteosarcoma cell lines, Saos-2 and MG63. Compared to FE-cells, FD-cells showed an increase in tri-lineage differentiation. Additionally, differences were found between the Saos-2 cells exhibiting higher levels of osteogenic differentiation, lower adipogenic differentiation, and a more developed chondrogenic phenotype than MG63, with the Saos-2 being more comparable to FD-derived HBMSCs. The differences found between the FD and FE derived cells are consistent with the FD region containing more hematopoietic tissue compared to the FE. This may be related to the similarities between FD-derived cells and Saos-2 cells during osteogenic and chondrogenic differentiation. These studies reveal distinct differences in the tri-lineage differentiations of 'hematopoietic' and 'adipocyte rich' bone marrow, which correlate with specific characteristics of the two osteosarcoma cell lines.


Asunto(s)
Células Madre Mesenquimatosas , Osteosarcoma , Adolescente , Humanos , Osteogénesis , Diferenciación Celular , Células Madre Mesenquimatosas/metabolismo , Células Cultivadas , Línea Celular , Células de la Médula Ósea , Osteosarcoma/metabolismo , Células del Estroma
2.
FASEB J ; 33(3): 3279-3290, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30403537

RESUMEN

The current study used an ex vivo [embryonic day (E)18] chick femur defect model to examine the bone regenerative capacity of implanted 3-dimensional (3D) skeletal-endothelial cell constructs. Human bone marrow stromal cell (HBMSC) and HUVEC spheroids were implanted within a bone defect site to determine the osteogenic potential of the skeletal-endothelial cell unit. Cells were pelleted as co- or monocell spheroids and placed within 1-mm-drill defects in the mid-diaphysis of E18 chick femurs and cultured organotypically for 10 d. Micro-computed tomography analysis revealed significantly ( P = 0.0001) increased levels of bone volume (BV) and BV/tissue volume ratio in all cell-pellet groups compared with the sham defect group. The highest increase was seen in BV in femurs containing the HUVEC and HBMSC monocell constructs. Type II collagen expression was particularly pronounced within the cell spheres containing HBMSCs and HUVECs, and CD31-positive cell clusters were prominent within HUVEC-implanted defects. These studies demonstrate the importance of the 3D osteogenic-endothelial niche interaction in bone regeneration. Elucidating the component cell interactions in the osteogenic-vascular niche and the role of exogenous factors in driving these osteogenic processes will aid the development of better bone reparative strategies.-Inglis, S., Kanczler, J. M., Oreffo, R. O. C. 3D human bone marrow stromal and endothelial cell spheres promote bone healing in an osteogenic niche.


Asunto(s)
Regeneración Ósea/fisiología , Células Endoteliales de la Vena Umbilical Humana/fisiología , Células Madre Mesenquimatosas/fisiología , Animales , Embrión de Pollo , Técnicas de Cocultivo , Fémur/embriología , Fémur/lesiones , Xenoinjertos , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/trasplante , Humanos , Imagenología Tridimensional , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Osteogénesis/fisiología , Esferoides Celulares/citología , Esferoides Celulares/fisiología , Nicho de Células Madre/fisiología , Microtomografía por Rayos X
3.
Int J Mol Sci ; 21(15)2020 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-32752092

RESUMEN

Treatment for osteosarcoma (OS) has been largely unchanged for several decades, with typical therapies being a mixture of chemotherapy and surgery. Although therapeutic targets and products against cancer are being continually developed, only a limited number have proved therapeutically active in OS. Thus, the understanding of the OS microenvironment and its interactions are becoming more important in developing new therapies. Three-dimensional (3D) models are important tools in increasing our understanding of complex mechanisms and interactions, such as in OS. In this review, in vivo animal models, in vitro 3D models and in ovo chorioallantoic membrane (CAM) models, are evaluated and discussed as to their contribution in understanding the progressive nature of OS, and cancer research. We aim to provide insight and prospective future directions into the potential translation of 3D models in OS.


Asunto(s)
Neoplasias Óseas/ultraestructura , Membrana Corioalantoides/ultraestructura , Modelos Teóricos , Osteosarcoma/ultraestructura , Animales , Biomarcadores de Tumor/genética , Neoplasias Óseas/genética , Membrana Corioalantoides/metabolismo , Humanos , Osteosarcoma/genética , Estudios Prospectivos , Microambiente Tumoral/genética
4.
Arthritis Rheum ; 65(9): 2290-300, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23740547

RESUMEN

OBJECTIVE: To generate doxycycline-inducible human tumor necrosis factor α (TNFα)-transgenic mice to overcome a major disadvantage of existing transgenic mice with constitutive expression of TNFα, which is the limitation in crossing them with various knockout or transgenic mice. METHODS: A transgenic mouse line that expresses the human TNFα cytokine exclusively after doxycycline administration was generated and analyzed for the onset of diseases. RESULTS: Doxycycline-inducible human TNFα-transgenic mice developed an inflammatory arthritis- and psoriasis-like phenotype, with fore and hind paws being prominently affected. The formation of "sausage digits" with characteristic involvement of the distal interphalangeal joints and nail malformation was observed. Synovial hyperplasia, enthesitis, cartilage and bone alterations, formation of pannus tissue, and inflammation of the skin epidermis and nail matrix appeared as early as 1 week after the treatment of mice with doxycycline and became aggravated over time. The abrogation of human TNFα expression by the removal of doxycycline 6 weeks after beginning stimulation resulted in fast resolution of the most advanced macroscopic and histologic disorders, and 3-6 weeks later, only minimal signs of disease were visible. CONCLUSION: Upon doxycycline administration, the doxycycline-inducible human TNFα-transgenic mouse displays the major features of inflammatory arthritis. It represents a unique animal model for studying the molecular mechanisms of arthritis, especially the early phases of disease genesis and tissue remodeling steps upon abrogation of TNFα expression. Furthermore, unlimited crossing of doxycycline-inducible human TNFα-transgenic mice with various knockout or transgenic mice opens new possibilities for unraveling the role of various signaling molecules acting in concert with TNFα.


Asunto(s)
Artritis Experimental/genética , Artritis Psoriásica/genética , Factor de Necrosis Tumoral alfa/genética , Animales , Artritis Experimental/metabolismo , Artritis Experimental/patología , Artritis Psoriásica/metabolismo , Artritis Psoriásica/patología , Cartílago/metabolismo , Cartílago/patología , Inflamación/patología , Articulaciones/metabolismo , Articulaciones/patología , Ratones , Ratones Transgénicos , Factor de Necrosis Tumoral alfa/metabolismo
5.
Biodes Manuf ; 7(2): 121-136, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38497056

RESUMEN

Autograft or metal implants are routinely used in skeletal repair. However, they fail to provide long-term clinical resolution, necessitating a functional biomimetic tissue engineering alternative. The use of native human bone tissue for synthesizing a biomimetic material ink for three-dimensional (3D) bioprinting of skeletal tissue is an attractive strategy for tissue regeneration. Thus, human bone extracellular matrix (bone-ECM) offers an exciting potential for the development of an appropriate microenvironment for human bone marrow stromal cells (HBMSCs) to proliferate and differentiate along the osteogenic lineage. In this study, we engineered a novel material ink (LAB) by blending human bone-ECM (B) with nanoclay (L, Laponite®) and alginate (A) polymers using extrusion-based deposition. The inclusion of the nanofiller and polymeric material increased the rheology, printability, and drug retention properties and, critically, the preservation of HBMSCs viability upon printing. The composite of human bone-ECM-based 3D constructs containing vascular endothelial growth factor (VEGF) enhanced vascularization after implantation in an ex vivo chick chorioallantoic membrane (CAM) model. The inclusion of bone morphogenetic protein-2 (BMP-2) with the HBMSCs further enhanced vascularization and mineralization after only seven days. This study demonstrates the synergistic combination of nanoclay with biomimetic materials (alginate and bone-ECM) to support the formation of osteogenic tissue both in vitro and ex vivo and offers a promising novel 3D bioprinting approach to personalized skeletal tissue repair. Supplementary Information: The online version contains supplementary material available at 10.1007/s42242-023-00265-z.

6.
Biomater Adv ; 164: 213959, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39083876

RESUMEN

Biomaterial-based approaches for bone regeneration seek to explore alternative strategies to repair non-healing fractures and critical-sized bone defects. Fracture non-union occurs due to a number of factors resulting in the formation of bone defects. Rigorous evaluation of the biomaterials in relevant models and assessment of their potential to translate towards clinical use is vital. Large animal experimentation can be used to model fracture non-union while scaling-up materials for clinical use. Growth factors modulate cell phenotype, behaviour and initiate signalling pathways leading to changes in matrix deposition and tissue formation. Bone morphogenetic protein-2 (BMP-2) is a potent osteogenic growth factor, with a rapid clearance time in vivo necessitating clinical use at a high dose, with potential deleterious side-effects. The current studies have examined the potential for Laponite® nanoclay coated poly(caprolactone) trimethacrylate (PCL-TMA900) scaffolds to bind BMP-2 for enhanced osteoinduction in a large animal critical-sized bone defect. An ovine femoral condyle defect model confirmed PCL-TMA900 scaffolds coated with Laponite®/BMP-2 produced significant bone formation compared to the uncoated PCL-TMA 900 scaffold in vivo, assessed by micro-computed tomography (µCT) and histology. This indicated the ability of Laponite® to deliver the bioactive BMP-2 on the PCL-TMA900 scaffold. Bone formed around the Laponite®/BMP-2 coated PCL-TMA900 scaffold, with no erroneous bone formation observed away from the scaffold material confirming localisation of BMP-2 delivery. The current studies demonstrate the ability of a nanoclay to localise and deliver bioactive BMP-2 within a tailored octet-truss scaffold for efficacious bone defect repair in a large animal model with significant implications for translation to the clinic.


Asunto(s)
Proteína Morfogenética Ósea 2 , Regeneración Ósea , Fémur , Impresión Tridimensional , Silicatos , Andamios del Tejido , Animales , Proteína Morfogenética Ósea 2/administración & dosificación , Proteína Morfogenética Ósea 2/farmacología , Regeneración Ósea/efectos de los fármacos , Silicatos/química , Silicatos/farmacología , Silicatos/administración & dosificación , Andamios del Tejido/química , Ovinos , Fémur/patología , Fémur/lesiones , Fémur/efectos de los fármacos , Materiales Biocompatibles Revestidos/química , Osteogénesis/efectos de los fármacos , Modelos Animales de Enfermedad
7.
Eur Cell Mater ; 26: 91-106; discussion 106, 2013 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-24027022

RESUMEN

Scientific research and progress, particularly in the drug discovery and regenerative medicine fields, is typically dependent on suitable animal models to develop new and improved clinical therapies for injuries and diseases. In vivo model systems are frequently utilised, but these models are expensive, highly complex and pose a number of ethical considerations leading to the development and use of a number of alternative ex vivo model systems. The ex vivo embryonic chick long bone and limb bud models have been utilised in the scientific research field as a model to understand skeletal development for over eighty years. The rapid development of avian skeletal tissues, coupled with the ease of experimental manipulation, availability of genome sequence and the presence of multiple cell and tissue types has seen such model systems gain significant research interest in the last few years in the tissue engineering field. The models have been explored both as systems for understanding the developmental bone niche and as potential testing tools for tissue engineering strategies for bone repair and regeneration. This review details the evolution of the chick limb organ culture system and presents recent innovative developments and emerging techniques and technologies applied to these models that are aiding our understanding of skeletal developmental and regenerative medicine research and application.


Asunto(s)
Regeneración Ósea , Fémur/fisiología , Técnicas de Cultivo de Órganos , Osteogénesis , Medicina Regenerativa/métodos , Animales , Pollos , Condrogénesis , Ingeniería de Tejidos/métodos
8.
Adv Mater ; 35(48): e2304461, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37658732

RESUMEN

Self-assembly, the spontaneous ordering of components into patterns, is widespread in nature and fundamental to generating function across length scales. Morphogen gradients in biological development are paradigmatic as both products and effectors of self-assembly and various attempts have been made to reproduce such gradients in biomaterial design. To date, approaches have typically utilized top-down fabrication techniques that, while allowing high-resolution control, are limited by scale and require chemical cross-linking steps to stabilize morphogen patterns in time. Here, a bottom-up approach to protein patterning is developed based on a novel binary reaction-diffusion process where proteins function as diffusive reactants to assemble a nanoclay-protein composite hydrogel. Using this approach, it is possible to generate scalable and highly stable 3D patterns of target proteins down to sub-cellular resolution through only physical interactions between clay nanoparticles and the proteins and ions present in blood. Patterned nanoclay gels are able to guide cell behavior to precisely template bone tissue formation in vivo. These results demonstrate the feasibility of stabilizing 3D gradients of biological signals through self-assembly processes and open up new possibilities for morphogen-based therapeutic strategies and models of biological development and repair.


Asunto(s)
Nanopartículas , Hidrogeles , Arcilla
9.
Methods Mol Biol ; 2206: 47-56, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32754810

RESUMEN

The development and maintenance of a functioning vascular system is a critical function for many aspects of tissue growth and regeneration. Vascular endothelial cell in vitro co-culture spheroids are self-organized cell composites that have the capacity to recapitulate the three-dimensional tissue microenvironment. These spheroid testing platforms aim to better understand the mechanisms of functional tissue and how new therapeutic agents can drive these 3D co-culture processes. Here we describe direct cell-cell 3D endothelial co-culture spheroid methods, to examine the physiological spatial growth and cell-cell interaction of vascular cells and surrounding native tissue cells in the formation of vascular networks within spheroids and the potential to regenerate tissue.


Asunto(s)
Técnicas de Cocultivo/métodos , Células Endoteliales de la Vena Umbilical Humana/citología , Esferoides Celulares/citología , Comunicación Celular/fisiología , Células Cultivadas , Humanos , Ingeniería de Tejidos/métodos
10.
ACS Appl Mater Interfaces ; 13(28): 32624-32639, 2021 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-34228435

RESUMEN

Organ dysfunction is a major cause of morbidity and mortality. Transplantation is typically the only definitive cure, challenged by the lack of sufficient donor organs. Tissue engineering encompasses the development of biomaterial scaffolds to support cell attachment, proliferation, and differentiation, leading to tissue regeneration. For efficient clinical translation, the forming technology utilized must be suitable for mass production. Herein, uniaxial polyhydroxyalkanoate scaffolds manufactured by pressurized gyration, a hybrid scalable spinning technique, are successfully used in bone, nerve, and cardiovascular applications. Chorioallantoic membrane and in vivo studies provided evidence of vascularization, collagen deposition, and cellular invasion for bone tissue engineering. Highly efficient axonal outgrowth was observed in dorsal root ganglion-based 3D ex vivo models. Human induced pluripotent stem cell derived cardiomyocytes exhibited a mature cardiomyocyte phenotype with optimal calcium handling. This study confirms that engineered polyhydroxyalkanoate-based gyrospun fibers provide an exciting and unique toolbox for the development of scalable scaffolds for both hard and soft tissue regeneration.


Asunto(s)
Células/metabolismo , Polihidroxialcanoatos/química , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Animales , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Pollos , Módulo de Elasticidad , Ganglios Espinales/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ratones , Miocitos Cardíacos/metabolismo , Porosidad , Presión , Ratas , Rotación , Células de Schwann/metabolismo
11.
J Tissue Eng ; 11: 2041731420942734, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33194169

RESUMEN

The chick chorioallantoic membrane model has been around for over a century, applied in angiogenic, oncology, dental and xenograft research. Despite its often perceived archaic, redolent history, the chorioallantoic membrane assay offers new and exciting opportunities for material and growth factor evaluation in bone tissue engineering. Currently, superior/improved experimental methodology for the chorioallantoic membrane assay are difficult to identify, given an absence of scientific consensus in defining experimental approaches, including timing of inoculation with materials and the analysis of results. In addition, critically, regulatory and welfare issues impact upon experimental designs. Given such disparate points, this review details recent research using the ex vivo chorioallantoic membrane assay and the ex vivo organotypic culture to advance the field of bone tissue engineering, and highlights potential areas of improvement for their application based on recent developments within our group and the tissue engineering field.

12.
Biofabrication ; 12(3): 035010, 2020 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-32259804

RESUMEN

Acellular soft hydrogels are not ideal for hard tissue engineering given their poor mechanical stability, however, in combination with cellular components offer significant promise for tissue regeneration. Indeed, nanocomposite bioinks provide an attractive platform to deliver human bone marrow stromal cells (HBMSCs) in three dimensions producing cell-laden constructs that aim to facilitate bone repair and functionality. Here we present the in vitro, ex vivo and in vivo investigation of bioprinted HBMSCs encapsulated in a nanoclay-based bioink to produce viable and functional three-dimensional constructs. HBMSC-laden constructs remained viable over 21 d in vitro and immediately functional when conditioned with osteogenic media. 3D scaffolds seeded with human umbilical vein endothelial cells (HUVECs) and loaded with vascular endothelial growth factor (VEGF) implanted ex vivo into a chick chorioallantoic membrane (CAM) model showed integration and vascularisation after 7 d of incubation. In a pre-clinical in vivo application of a nanoclay-based bioink to regenerate skeletal tissue, we demonstrated bone morphogenetic protein-2 (BMP-2) absorbed scaffolds produced extensive mineralisation after 4 weeks (p < 0.0001) compared to the drug-free and alginate controls. In addition, HBMSC-laden 3D printed scaffolds were found to significantly (p < 0.0001) support bone tissue formation in vivo compared to acellular and cast scaffolds. These studies illustrate the potential of nanoclay-based bioink, to produce viable and functional constructs for clinically relevant skeletal tissue regeneration.


Asunto(s)
Huesos/irrigación sanguínea , Arcilla/química , Minerales/metabolismo , Nanocompuestos/química , Neovascularización Fisiológica , Impresión Tridimensional , Ingeniería de Tejidos , Andamios del Tejido/química , Animales , Proteína Morfogenética Ósea 2/farmacología , Huesos/efectos de los fármacos , Calcificación Fisiológica/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Pollos , Membrana Corioalantoides/efectos de los fármacos , Humanos , Implantes Experimentales , Ratones , Modelos Animales , Neovascularización Fisiológica/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Silicatos/química , Tejido Subcutáneo/efectos de los fármacos
13.
J Control Release ; 325: 335-346, 2020 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-32629135

RESUMEN

Additive manufacturing processes used to create regenerative bone tissue engineered implants are not biocompatible, thereby restricting direct use with stem cells and usually require cell seeding post-fabrication. Combined delivery of stem cells with the controlled release of osteogenic factors, within a mechanically-strong biomaterial combined during manufacturing would replace injectable defect fillers (cements) and allow personalized implants to be rapidly prototyped by 3D bioprinting. Through the use of direct genetic programming via the sustained release of an exogenously delivered transcription factor RUNX2 (delivered as recombinant GET-RUNX2 protein) encapsulated in PLGA microparticles (MPs), we demonstrate that human mesenchymal stromal (stem) cells (hMSCs) can be directly fabricated into a thermo-sintered 3D bioprintable material and achieve effective osteogenic differentiation. Importantly we observed osteogenic programming of gene expression by released GET-RUNX2 (8.2-, 3.3- and 3.9-fold increases in OSX, RUNX2 and OPN expression, respectively) and calcification (von Kossa staining) in our scaffolds. The developed biodegradable PLGA/PEG paste formulation augments high-density bone development in a defect model (~2.4-fold increase in high density bone volume) and can be used to rapidly prototype clinically-sized hMSC-laden implants within minutes using mild, cytocompatible extrusion bioprinting. The ability to create mechanically strong 'cancellous bone-like' printable implants for tissue repair that contain stem cells and controlled-release of programming factors is innovative, and will facilitate the development of novel localized delivery approaches to direct cellular behaviour for many regenerative medicine applications including those for personalized bone repair.


Asunto(s)
Bioimpresión , Células Madre Mesenquimatosas , Diferenciación Celular , Humanos , Osteogénesis , Ingeniería de Tejidos , Andamios del Tejido
14.
Adv Healthc Mater ; 8(9): e1800088, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-29756272

RESUMEN

Decellularized matrices offer a beneficial substitute for biomimetic scaffolds in tissue engineering. The current study examines the potential of decellularized placental vessel sleeves (PVS) as a periosteal protective sleeve to enhance bone regeneration in embryonic day 18 chick femurs contained within the PVS and cultured organotypically over a 10 day period. The femurs are inserted into decellularized biocompatibility-tested PVS and maintained in an organotypic culture for a period of 10 days. In femurs containing decellularized PVS, a significant increase in bone volume (p < 0.001) is evident, demonstrated by microcomputed tomography (µCT) compared to femurs without PVS. Histological and immunohistological analyses reveal extensive integration of decellularized PVS with the bone periosteum, and enhanced conservation of bone architecture within the PVS. In addition, the expressions of hypoxia inducible factor-1 alpha (HIF-1α), type II collagen (COL-II), and proteoglycans are observed, indicating a possible repair mechanism via a cartilaginous stage of the bone tissue within the sleeve. The use of decellularized matrices like PVS offers a promising therapeutic strategy in surgical tissue replacement, promoting biocompatibility and architecture of the tissue as well as a factor-rich niche environment with negligible immunogenicity.


Asunto(s)
Células Endoteliales de la Vena Umbilical Humana/citología , Materiales Biocompatibles/química , Vasos Sanguíneos/citología , Vasos Sanguíneos/metabolismo , Regeneración Ósea/fisiología , Supervivencia Celular/fisiología , Células Cultivadas , Membrana Corioalantoides/citología , Membrana Corioalantoides/metabolismo , Femenino , Fémur/citología , Fémur/metabolismo , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Inmunohistoquímica , Técnicas In Vitro , Osteogénesis/genética , Osteogénesis/fisiología , Embarazo , Ingeniería de Tejidos/métodos , Cicatrización de Heridas/genética , Cicatrización de Heridas/fisiología
15.
Sci Rep ; 9(1): 17745, 2019 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-31780671

RESUMEN

Deficient bone vasculature is a key component in pathological conditions ranging from developmental skeletal abnormalities to impaired bone repair. Vascularisation is dependent upon vascular endothelial growth factor (VEGF), which drives both angiogenesis and osteogenesis. The aim of this study was to examine the efficacy of blood vessel and bone formation following transfection with VEGF RNA or delivery of recombinant human VEGF165 protein (rhVEGF165) across in vitro and in vivo model systems. To quantify blood vessels within bone, an innovative approach was developed using high-resolution X-ray computed tomography (XCT) to generate quantifiable three-dimensional reconstructions. Application of rhVEGF165 enhanced osteogenesis, as evidenced by increased human osteoblast-like MG-63 cell proliferation in vitro and calvarial bone thickness following in vivo administration. In contrast, transfection with VEGF RNA triggered angiogenic effects by promoting VEGF protein secretion from MG-63VEGF165 cells in vitro, which resulted in significantly increased angiogenesis in the chorioallantoic (CAM) assay in ovo. Furthermore, direct transfection of bone with VEGF RNA in vivo increased intraosseous vascular branching. This study demonstrates the importance of continuous supply as opposed to a single high dose of VEGF on angiogenesis and osteogenesis and, illustrates the potential of XCT in delineating in 3D, blood vessel connectivity in bone.


Asunto(s)
Neovascularización Fisiológica , Osteogénesis , ARN/administración & dosificación , Transfección , Factor A de Crecimiento Endotelial Vascular/administración & dosificación , Animales , Huesos/irrigación sanguínea , Huesos/efectos de los fármacos , Línea Celular , Pollos , Humanos , Ratones , Neovascularización Fisiológica/efectos de los fármacos , Osteogénesis/efectos de los fármacos , ARN/genética , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/genética , Factor A de Crecimiento Endotelial Vascular/genética
16.
J Bone Miner Res ; 34(11): 2117-2132, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31269275

RESUMEN

Osteoblast (OB) lineage cells are an important source of vascular endothelial growth factor (VEGF), which is critical for bone growth and repair. During bone development, pubertal differences in males and females exist, but little is known about whether VEGF signaling contributes to skeletal sexual dimorphism. We have found that in mice, conditional disruption of VEGF in osteocalcin-expressing cells (OcnVEGFKO) exerts a divergent influence on morphological, cellular, and whole bone properties between sexes. Furthermore, we describe an underlying sexual divergence in VEGF signaling in OB cultures in vitro independent of circulating sex hormones. High-resolution synchrotron computed tomography and backscattered scanning electron microscopy revealed, in males, extensive unmineralized osteoid encasing enlarged blood vessel canals and osteocyte lacunae in cortical bone after VEGF deletion, which contributed to increased porosity. VEGF was deleted in male and female long bone-derived OBs (OBVEGKO) in vitro and Raman spectroscopic analyses of mineral and matrix repertoires highlighted differences between male and female OBVEGFKO cells, with increased immature phosphate species prevalent in male OBVEGFKO cultures versus wild type (WT). Further sexual dimorphism was observed in bone marrow endothelial cell gene expression in vitro after VEGF deletion and in sclerostin protein expression, which was increased in male OcnVEGFKO bones versus WT. The impact of altered OB matrix composition after VEGF deletion on whole bone geometry was assessed between sexes, although significant differences between OcnVEGFKO and WT were identified only in females. Our results suggest that bone-derived VEGF regulates matrix mineralization and vascularization distinctly in males and females, which results in divergent physical bone traits.


Asunto(s)
Desarrollo Óseo , Células de la Médula Ósea/metabolismo , Huesos/irrigación sanguínea , Células Endoteliales/metabolismo , Caracteres Sexuales , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Huesos/metabolismo , Femenino , Masculino , Ratones , Ratones Noqueados , Factor A de Crecimiento Endotelial Vascular/genética
17.
Biomaterials ; 29(21): 3105-16, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18442852

RESUMEN

Type I Collagen matrices of defined porosity, incorporating carbonate substituted hydroxyapatite (HA) crystals, were assessed for their ability to support osteo- and chondrogenic differentiation of human bone marrow stromal cells (HBMSCs). Collagen-HA composite scaffolds supported the osteogenic differentiation of HBMSCs both in vitro and in vivo as demonstrated by histological and micro-CT analyses indicating the extensive penetration of alkaline phosphatase expressing cells and new matrix synthesis with localised areas immunologically positive for osteocalcin. In vivo, extensive new osteoid formation of implant origin was observed in the areas of vasculature. Chondrogenic matrix synthesis was evidenced in the peripheral regions of pure collagen systems by an abundance of Sox9 expressing chondrocytes embedded within a proteoglycan and collagen II rich ECM. The introduction of microchannels to the scaffold architecture was seen to enhance chondrogenesis. Tissue specific gene expression and corresponding matrix synthesis indicate that collagen matrices support the growth and differentiation of HBMSCs and suggest the potential of this platform for understanding the ECM cues necessary for osteogenesis and chondrogenesis.


Asunto(s)
Células de la Médula Ósea/citología , Colágeno/química , Células del Estroma/citología , Ingeniería de Tejidos/métodos , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Células de la Médula Ósea/ultraestructura , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Condrogénesis/efectos de los fármacos , Colágeno/farmacología , Humanos , Microscopía Electrónica de Rastreo , Osteogénesis/efectos de los fármacos , Células del Estroma/ultraestructura , Andamios del Tejido/química
18.
Biomaterials ; 29(22): 3221-7, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18456320

RESUMEN

Concerns over disease transmission, high costs and limited supply have led to interest in synthetic grafts in the field of impaction bone grafting (IBG). Poly(DL-lactic acid) (PLA) grafts are attractive alternatives due to their biocompatibility, established safety and versatile manufacturing process. This study examined the potential of PLA scaffolds augmented with human bone marrow stromal cells (HBMSCs) in IBG. In vitro and in vivo studies were performed on impacted morsellised PLA seeded with HBMSC and compared to PLA alone. In vitro samples were incubated under osteogenic conditions and in vivo samples were implanted subcutaneously into severely compromised immunodeficient mice, for 4 weeks. Biochemical, histological, mechanical and 3D micro-computed tomography analyses were performed. HBMSC viability, biochemical activity and histological evidence of osteogenic cellular differentiation, post-impaction were observed in vitro and in vivo in PLA/HBMSC samples compared to impacted PLA alone. In vitro PLA/HBMSC samples demonstrated evidence of mechanical enhancement over PLA alone. In vivo studies showed a significant increase in new bone and blood vessel formation in the PLA/HBMSC constructs compared to PLA alone. With alternatives to allograft being sought, these studies have demonstrated PLA/HBMSC living composites, to be a potential prospect as a biological bone graft extender for future use in the field of IBG.


Asunto(s)
Materiales Biocompatibles/química , Células de la Médula Ósea/citología , Ácido Láctico/química , Poliésteres/química , Polímeros/química , Células del Estroma/citología , Fosfatasa Alcalina/metabolismo , Animales , Materiales Biocompatibles/farmacología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Sustitutos de Huesos/química , Sustitutos de Huesos/farmacología , Huesos/citología , Huesos/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Humanos , Inmunohistoquímica , Ácido Láctico/farmacología , Masculino , Ratones , Ratones SCID , Poliésteres/farmacología , Polímeros/farmacología , Resistencia al Corte , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo , Ingeniería de Tejidos/métodos
19.
Biomaterials ; 29(12): 1892-900, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18234329

RESUMEN

The capacity to deliver, temporally, bioactive growth factors in combination with appropriate progenitor and stem cells to sites of tissue regeneration promoting angiogenesis and osteogenesis offers therapeutic opportunities in regenerative medicine. We have examined the bone regenerative potential of encapsulated vascular endothelial growth factor (VEGF(165)) biodegradable poly(DL-lactic acid) (PLA) scaffolds created using supercritical CO(2) fluid technology to encapsulate and release solvent-sensitive and thermolabile growth factors in combination with human bone marrow stromal cells (HBMSC) implanted in a mouse femur segmental defect (5 mm) for 4 weeks. HBMSC seeded on VEGF encapsulated PLA scaffolds showed significant bone regeneration in the femur segmental defect compared to the scaffold alone and scaffold seeded with HBMSC as analysed by indices of increased bone volume (BV mm(3)), trabecular number (Tb.N/mm) and reduced trabecular separation (Tb.Sp.mm) in the defect region using micro-computed tomography. Histological examination confirmed significant new bone matrix in the HBMSC seeded VEGF encapsulated scaffold group as evidenced by Sirius red/alcian blue and Goldner's trichrome staining and type I collagen immunocytochemistry expression in comparison to the other groups. These studies demonstrate the ability to deliver, temporally, a combination of VEGF released from scaffolds with seeded HBMSC to sites of bone defects, results in enhanced regeneration of a bone defect.


Asunto(s)
Implantes Absorbibles , Fracturas del Fémur/patología , Fracturas del Fémur/terapia , Ácido Láctico/química , Trasplante de Células Madre Mesenquimatosas/métodos , Osteogénesis/efectos de los fármacos , Polímeros/química , Factor A de Crecimiento Endotelial Vascular/administración & dosificación , Animales , Células Cultivadas , Terapia Combinada , Portadores de Fármacos/química , Humanos , Masculino , Ratones , Ratones Desnudos , Poliésteres , Resultado del Tratamiento , Factor A de Crecimiento Endotelial Vascular/química
20.
J Tissue Eng Regen Med ; 12(8): 1877-1890, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29893478

RESUMEN

Traditionally used as an angiogenic assay, the chorioallantoic membrane (CAM) assay of the chick embryo offers significant potential as an in vivo model for xenograft organ culture. Viable human bone can be cultivated on the CAM and increases in bone volume are evident; however, it remains unclear by what mechanism this change occurs and whether this reflects the physiological process of bone remodelling. In this study we tested the hypothesis that CAM-induced bone remodelling is a consequence of host and graft mediated processes. Bone cylinders harvested from femoral heads post surgery were placed on the CAM of green fluorescent protein (GFP)-chick embryos for 9 days, followed by micro computed tomography (µCT) and histological analysis. Three-dimensional registration of consecutive µCT-scans showed newly mineralised tissue in CAM-implanted bone cylinders, as well as new osteoid deposition histologically. Immunohistochemistry demonstrated the presence of bone resorption and formation markers (Cathepsin K, SOX9 and RUNX2) co-localising with GFP staining, expressed by avian cells only. To investigate the role of the human cells in the process of bone formation, decellularised bone cylinders were implanted on the CAM and comparable increases in bone volume were observed, indicating that avian cells were responsible for the bone mineralisation process. Finally, CAM-implantation of acellular collagen sponges, containing bone morphogenetic protein 2, resulted in the deposition of extracellular matrix and tissue mineralisation. These studies indicate that the CAM can respond to osteogenic stimuli and support formation or resorption of implanted human bone, providing a humanised CAM model for regenerative medicine research and a novel short-term in vivo model for tissue engineering and biomaterial testing.


Asunto(s)
Resorción Ósea , Membrana Corioalantoides/metabolismo , Osteogénesis , Anciano , Anciano de 80 o más Años , Animales , Embrión de Pollo , Femenino , Xenoinjertos , Humanos , Masculino , Persona de Mediana Edad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA