Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Biol Res ; 57(1): 65, 2024 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-39261966

RESUMEN

Increasing evidences demonstrate the role of sensory innervation in bone metabolism, remodeling and repair, however neurovascular coupling in bone is rarely studied. Using microfluidic devices as an indirect co-culture model to mimic in vitro the physiological scenario of innervation, our group demonstrated that sensory neurons (SNs) were able to regulate the extracellular matrix remodeling by endothelial cells (ECs), in particular through sensory neuropeptides, i.e. calcitonin gene-related peptide (CGRP) and substance P (SP). Nonetheless, still little is known about the cell signaling pathways and mechanism of action in neurovascular coupling. Here, in order to characterize the communication between SNs and ECs at molecular level, we evaluated the effect of SNs and the neuropeptides CGRP and SP on ECs. We focused on different pathways known to play a role on endothelial functions: calcium signaling, p38 and Erk1/2; the control of signal propagation through Cx43; and endothelial functions through the production of nitric oxide (NO). The effect of SNs was evaluated on ECs Ca2+ influx, the expression of Cx43, endothelial nitric oxide synthase (eNOS) and nitric oxide (NO) production, p38, ERK1/2 as well as their phosphorylated forms. In addition, the role of CGRP and SP were either analyzed using respective antagonists in the co-culture model, or by adding directly on the ECs monocultures. We show that capsaicin-stimulated SNs induce increased Ca2+ influx in ECs. SNs stimulate the increase of NO production in ECs, probably involving a decrease in the inhibitory eNOS T495 phosphorylation site. The neuropeptide CGRP, produced by SNs, seems to be one of the mediators of this effect in ECs since NO production is decreased in the presence of CGRP antagonist in the co-culture of ECs and SNs, and increased when ECs are stimulated with synthetic CGRP. Taken together, our results suggest that SNs play an important role in the control of the endothelial cell functions through CGRP production and NO signaling pathway.


Asunto(s)
Péptido Relacionado con Gen de Calcitonina , Células Endoteliales , Óxido Nítrico , Células Receptoras Sensoriales , Transducción de Señal , Sustancia P , Péptido Relacionado con Gen de Calcitonina/metabolismo , Péptido Relacionado con Gen de Calcitonina/farmacología , Sustancia P/farmacología , Sustancia P/metabolismo , Transducción de Señal/fisiología , Transducción de Señal/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Receptoras Sensoriales/efectos de los fármacos , Células Receptoras Sensoriales/metabolismo , Animales , Óxido Nítrico/metabolismo , Técnicas de Cocultivo , Comunicación Celular/fisiología , Comunicación Celular/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo III/metabolismo , Células Cultivadas , Humanos , Ratas
2.
Cell Commun Signal ; 18(1): 162, 2020 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-33076927

RESUMEN

BACKGROUND: Recent physiological and experimental data highlight the role of the sensory nervous system in bone repair, but its precise role on angiogenesis in a bone regeneration context is still unknown. Our previous work demonstrated that sensory neurons (SNs) induce the osteoblastic differentiation of mesenchymal stem cells, but the influence of SNs on endothelial cells (ECs) was not studied. METHODS: Here, in order to study in vitro the interplay between SNs and ECs, we used microfluidic devices as an indirect co-culture model. Gene expression analysis of angiogenic markers, as well as measurements of metalloproteinases protein levels and enzymatic activity, were performed. RESULTS: We were able to demonstrate that two sensory neuropeptides, calcitonin gene-related peptide (CGRP) and substance P (SP), were involved in the transcriptional upregulation of angiogenic markers (vascular endothelial growth factor, angiopoietin 1, type 4 collagen, matrix metalloproteinase 2) in ECs. Co-cultures of ECs with SNs also increased the protein level and enzymatic activity of matrix metalloproteinases 2 and 9 (MMP2/MMP9) in ECs. CONCLUSIONS: Our results suggest a role of sensory neurons, and more specifically of CGRP and SP, in the remodelling of endothelial cells extracellular matrix, thus supporting and enhancing the angiogenesis process. Video abstract.


Asunto(s)
Células Endoteliales/metabolismo , Matriz Extracelular/metabolismo , Ganglios Espinales/metabolismo , Células Receptoras Sensoriales/metabolismo , Animales , Péptido Relacionado con Gen de Calcitonina/metabolismo , Células Endoteliales/ultraestructura , Matriz Extracelular/ultraestructura , Femenino , Ganglios Espinales/ultraestructura , Regulación de la Expresión Génica , Metaloproteinasas de la Matriz/biosíntesis , Microfluídica , Modelos Biológicos , Neuritas/metabolismo , Osteogénesis , Ratas Wistar , Células Receptoras Sensoriales/ultraestructura , Sustancia P/metabolismo
3.
BMC Cell Biol ; 17 Suppl 1: 19, 2016 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-27230612

RESUMEN

Electron micrographs revealed the presence of gap junctions in osteoblastic cells over 40 years ago. These intercellular channels formed from connexins are present in bone forming osteoblasts, bone resorbing osteoclasts, and osteocytes (mature osteoblasts embedded in the mineralized bone matrix). More recently, genetic and pharmacologic studies revealed the role of connexins, and in particular Cx43, in the differentiation and function of all bone types. Furthermore, mutations in the gene encoding Cx43 were found to be causally linked to oculodentodigital dysplasia, a condition that results in an abnormal skeleton. Pannexins, molecules with similar structure and single-membrane channel forming potential as connexins when organized as hemichannels, are also expressed in osteoblastic cells. The function of pannexins in bone and cartilage is beginning to be uncovered, but more research is needed to determine the role of pannexins in bone development, adult bone mass and skeletal homeostasis. We describe here the current knowledge on the role of connexins and pannexins on skeletal health and disease.


Asunto(s)
Regeneración Ósea/fisiología , Huesos/metabolismo , Huesos/patología , Conexinas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Animales , Conexinas/genética , Modelos Animales de Enfermedad , Humanos , Osteoartritis/metabolismo , Osteoartritis/patología
4.
J Cell Biochem ; 114(3): 650-7, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23059760

RESUMEN

Two dimensional (2D) co-cultures of human bone marrow stromal cells (HBMSCs) and human umbilical vein endothelial cells (HUVECs) stimulate osteoblastic differentiation of HBMSCs, induce the formation of self-assembled network and cell interactions between the two cell types involving many vascular molecules. Because of their strong activities on angiogenesis and tissue remodeling, urokinase plasminogen activator (uPA), plasminogen activator inhibitor-1 (PAI-1), matrix metalloproteinase-2 (MMP-2) as well tissue inhibitors of matrix metalloproteinase-2 (TIMP-2) were investigated in this 2D co-culture model. We found that the expression of uPA, MMP-2 in the co-cultured cells was significantly higher than those in mono-cultured cells. In opposite, PAI-1, expressed only by HUVECs is not regulated in the co-culture. Inhibition assays confirm that uPA played a critical role in the formation of self-assembled network as neutralization of uPA disturbed this network. In the same context, inhibition of MMP-2 prevented the formation of self-assembled network, while the inhibition of uPA abolished the over expression and the activity of MMP-2. This upregulation could initiate the uPA expression and proteolysis processes through the MMP-2 activity, and may contribute to endothelial cell migration and the formation of this self-assembled network observed in these 2D co-cultured cells.


Asunto(s)
Células de la Médula Ósea/metabolismo , Matriz Extracelular/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Células del Estroma/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Comunicación Celular , Diferenciación Celular , Movimiento Celular , Técnicas de Cocultivo , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Humanos , Inhibidores de la Metaloproteinasa de la Matriz , Neovascularización Fisiológica , Osteoblastos/metabolismo , Inhibidor 1 de Activador Plasminogénico/metabolismo , Proteolisis , Inhibidor Tisular de Metaloproteinasa-2/metabolismo , Regulación hacia Arriba , Activador de Plasminógeno de Tipo Uroquinasa/antagonistas & inhibidores
5.
Bioengineering (Basel) ; 10(11)2023 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-38002381

RESUMEN

INTRODUCTION: Guided bone regeneration (GBR) procedures require selecting suitable membranes for oral surgery. Pullulan and/or dextran-based polysaccharide materials have shown encouraging results in bone regeneration as bone substitutes but have not been used to produce barrier membranes. The present study aimed to develop and characterize pullulan/dextran-derived membranes for GBR. MATERIALS AND METHODS: Two pullulan/dextran-based membranes, containing or not hydroxyapatite (HA) particles, were developed. In vitro, cytotoxicity evaluation was performed using human bone marrow mesenchymal stem cells (hBMSCs). Biocompatibility was assessed on rats in a subcutaneous model for up to 16 weeks. In vivo, rat femoral defects were created on 36 rats to compare the two pullulan/dextran-based membranes with a commercial collagen membrane (Bio-Gide®). Bone repair was assessed radiologically and histologically. RESULTS: Both polysaccharide membranes demonstrated cytocompatibility and biocompatibility. Micro-computed tomography (micro-CT) analyses at two weeks revealed that the HA-containing membrane promoted a significant increase in bone formation compared to Bio-Gide®. At one month, similar effects were observed among the three membranes in terms of bone regeneration. CONCLUSION: The developed pullulan/dextran-based membranes evidenced biocompatibility without interfering with bone regeneration and maturation. The HA-containing membrane, which facilitated early bone regeneration and offered adequate mechanical support, showed promising potential for GBR procedures.

6.
Front Bioeng Biotechnol ; 10: 889481, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35845411

RESUMEN

Bone tissue engineering (BTE) strategies are increasingly investigated to overcome the limitations of currently used bone substitutes and to improve the bone regeneration process. Among the natural polymers used for tissue engineering, dextran and pullulan appear as natural hydrophilic polysaccharides that became promising biomaterials for BTE. This systematic review aimed to present the different published applications of pullulan and dextran-based biomaterials for BTE. An electronic search in Pubmed, Scopus, and Web of Science databases was conducted. Selection of articles was performed following PRISMA guidelines. This systematic review led to the inclusion of 28 articles on the use of pullulan and/or dextran-based biomaterials to promote bone regeneration in preclinical models. Sixteen studies focused on dextran-based materials for bone regeneration, six on pullulan substitutes and six on the combination of pullulan and dextran. Several strategies have been developed to provide bone regeneration capacity, mainly through their fabrication processes (functionalization methods, cross-linking process), or the addition of bioactive elements. We have summarized here the strategies employed to use the polysaccharide scaffolds (fabrication process, composition, application usages, route of administration), and we highlighted their relevance and limitations for BTE applications.

7.
Med Sci (Paris) ; 27(3): 289-96, 2011 Mar.
Artículo en Francés | MEDLINE | ID: mdl-21447302

RESUMEN

Tissue damages or loss of organs often result in structural and metabolic changes that can cause serious complications. The therapeutic objective of tissue engineering (TE) is to recreate, regenerate or restore function of damaged tissue. TE is based on the coalescence of three components: a scaffold or matrix from natural or synthetic origin biodegradable or not, reparative cells and signals (hypoxia, mechanical stress, morphogens…). Articular cartilage, bone and blood vessels are tissues for which TE has progressed significantly, from basic research to clinical trials. If biomaterials must exhibit different properties depending on the tissue to regenerate, the cellular component of TE is mostly represented by stem cells notably adult mesenchymal stem cells harvested from bone marrow or adipose tissue. In recent years, progress has been made in our understanding of the biological mechanisms that govern stem cell differentiation and in the development of materials with controlled physicochemical and biological properties. However, many technological barriers and regulations concerns have to be overcome before tissue engineering enters into the therapeutic arsenal of regenerative medicine. This review aims at highlighting the progress in the use of stem cells for engineering osteoarticular and vascular tissues.


Asunto(s)
Vasos Sanguíneos , Huesos , Cartílago Articular , Células Madre , Ingeniería de Tejidos/métodos , Humanos
8.
J Biomed Mater Res A ; 109(10): 1840-1848, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33797182

RESUMEN

Microbeads consisting of pullulan and dextran supplemented with hydroxyapatite have recently been developed for bone tissue engineering applications. Here, we evaluate the bone formation in two different preclinical models after injection of microbeads reconstituted with either saline buffer or autologous blood. Addition of saline solution or autologous blood to dried microbeads packaged into syringes allowed an easy injection. In the first rat bone defect model performed in the femoral condyle, microcomputed tomography performed after 30 and 60 days revealed an important mineralization process occurring around and within the core of the microbeads in both conditions. Bone volume/total volume measurements revealed no significant differences between the saline solution and the autologous blood groups. Histologically, osteoid tissue was evidenced around and in contact of the microbeads in both conditions. Using the sinus lift model performed in sheep, cone beam computed tomography revealed an important mineralization inside the sinus cavity for both groups after 3 months of implantation. Representative Masson trichrome staining images showed that bone formation occurs at the periphery and inside the microbeads in both conditions. Quantitative evaluation of the new bone formation displayed no significant differences between groups. In conclusion, reconstitution of microbeads with autologous blood did not enhance the regenerative capacity of these microbeads compared to the saline buffer group. This study is of particular interest for clinical applications in oral and maxillofacial surgery.


Asunto(s)
Sangre/metabolismo , Regeneración Ósea/fisiología , Huesos/patología , Huesos/fisiopatología , Durapatita/farmacología , Polímeros/farmacología , Solución Salina/farmacología , Animales , Modelos Animales de Enfermedad , Femenino , Implantes Experimentales , Microesferas , Ratas , Ovinos , Trasplante Autólogo , Microtomografía por Rayos X
9.
Am J Physiol Cell Physiol ; 299(2): C422-30, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20664068

RESUMEN

In our previous studies, roles of gap junction and vascular endothelial growth factor in the cross-talking of human bone marrow stromal cells (HBMSCs) and human umbilical vein endothelial cells (HUVECs) have been extensively studied. The present study focused on the investigation of the roles of neural (N)-cadherin in early differentiation of HBMSCs in direct-contact cocultures with HUVECs for 24 and 48 h. Quantitative real-time polymerase chain reaction, immunofluorescence, Western blot, as well as functional studies were applied to perform the studies at both protein and gene levels. Results showed that cocultured cells expressed much higher N-cadherin than monocultured cells after 24 and 48 h of culture. We observed that N-cadherin concentrated in the membrane of cocultured HBMSCs (co-HBMSCs) while distributed within the cytoplasm of monocultured HBMSCs, which indicated that the cell-cell adhesion was improved between cocultured cells. In addition, more beta-catenin was found to translocate into the cocultured cells nuclei and more T cell factor-1 (TCF-1) were detected in cocultured cells than in the monocultured cells. Moreover, mRNA levels of early osteoblastic markers including alkaline phosphatase (ALP) and type I collagen (Col-I) of co-HBMSCs were significantly upregulated, whereas neutralization of N-cadherin led to a downregulation of ALP and Col-I in both of the HBMSCs and co-HBMSCs compared with untreated cells. Taking our findings together it can be concluded that cocultures of HBMSCs with HUVECs increased N-cadherin expression and improved cell-cell adhesion. Whether this applies only to osteoprogenitor cells or to all the cell types in the culture will need to be determined by further studies. Subsequently, signaling transduction might be induced with the participation of beta-catenin and TCF-1. With the N-cadherin-mediated cell-cell adhesion and signaling transductions, the early osteoblastic differentiation of co-HBMSCs was significantly upregulated.


Asunto(s)
Antígenos CD/fisiología , Células de la Médula Ósea/fisiología , Cadherinas/fisiología , Diferenciación Celular/fisiología , Endotelio Vascular/fisiología , Osteoblastos/fisiología , Venas Umbilicales/fisiología , Adulto , Factores de Edad , Anciano , Células de la Médula Ósea/citología , Línea Celular , Técnicas de Cocultivo , Endotelio Vascular/citología , Humanos , Persona de Mediana Edad , Osteoblastos/citología , Osteogénesis/fisiología , Células del Estroma/citología , Células del Estroma/fisiología , Venas Umbilicales/citología , Adulto Joven
10.
J Cell Biochem ; 106(3): 390-8, 2009 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-19127540

RESUMEN

Proper bone remodeling requires an active process of angiogenesis which in turn supplies the necessary growth factors and stem cells. This tissue cooperation suggests a cross-talk between osteoblasts and endothelial cells. This work aims to identify the role of paracrine communication through vascular endothelial growth factor (VEGF) in co-culture between osteoblastic and endothelial cells. Through a well defined direct contact co-culture model between human osteoprogenitors (HOPs) and human umbilical vein endothelial cells (HUVECs), we observed that HUVECs were able to migrate along HOPs, inducing the formation of specific tubular-like structures. VEGF(165) gene expression was detected in the HOPs, was up-regulated in the co-cultured HOPs and both Flt-1 and KDR gene expression increased in co-cultured HUVECs. However, the cell rearrangement observed in co-culture was promoted by a combination of soluble chemoattractive factors and not by VEGF(165) alone. Despite having no observable effect on endothelial cell tubular-like formation, VEGF appeared to have a crucial role in osteoblastic differentiation since the inhibition of its receptors reduced the co-culture-stimulated osteoblastic phenotype. This co-culture system appears to enhance both primary angiogenesis events and osteoblastic differentiation, thus allowing for the development of new strategies in vascularized bone tissue engineering.


Asunto(s)
Comunicación Celular , Diferenciación Celular , Células Endoteliales/citología , Células Endoteliales/metabolismo , Osteoblastos/citología , Osteoblastos/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Movimiento Celular , Células Cultivadas , Técnicas de Cocultivo , Activación Enzimática , Regulación de la Expresión Génica/genética , Humanos , Microscopía Electrónica de Rastreo , ARN Mensajero/genética , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Cordón Umbilical/citología , Cordón Umbilical/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética
11.
Tissue Eng Part C Methods ; 25(7): 421-432, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31169074

RESUMEN

IMPACT STATEMENT: In this article, we first developed a new medium to culture together primary human osteoblastic, osteoclastic, and endothelial cells (ECs) chosen to represent the three major bone cell tissues. Indeed, no study has been conducted on primary human cells and on the phenotype/activity retention of these three primary human cell types. Thus, we established an original triculture model with osteoblastic, osteoclastic, and ECs, where not only both cell phenotype and cell activity were maintained but also cell culture homeostasis. These promising results will permit further investigations to create in vitro conditions to mimic the bone microenvironment and analyze cell interactions in ex vivo studies.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Células Endoteliales/citología , Modelos Biológicos , Osteoblastos/citología , Osteoclastos/citología , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Medios de Cultivo/farmacología , Células Endoteliales/efectos de los fármacos , Humanos , Osteoblastos/efectos de los fármacos , Osteoclastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Fenotipo , Fosfatasa Ácida Tartratorresistente/metabolismo
12.
Acta Biomater ; 99: 154-167, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31425892

RESUMEN

Despite significant progress in the field of biomaterials for bone repair, the lack of attention to the vascular and nervous networks within bone implants could be one of the main reasons for the delayed or impaired recovery of bone defects. The design of innovative biomaterials should improve the host capacity of healing to restore a functional tissue, taking into account that the nerve systems closely interact with blood vessels in the bone tissue. The aim of this work is to develop a cell-free and growth factor-free hydrogel capable to promote angiogenesis and innervation. To this end, we have used elastin-like polypeptides (ELPs), poly(ethylene glycol) (PEG) and increasing concentrations of the adhesion peptide IKVAV (25% (w/w) representing 1.7 mM and 50% (w/w) representing 4.1 mM) to formulate and produce hydrogels. When characterized in vitro, hydrogels have fine-tunable rheological properties, microporous structure and are biocompatible. At the biological level, 50% IKVAV composition up-regulated Runx2, Osx, Spp1, Vegfa and Bmp2 in mesenchymal stromal cells and Tek in endothelial cells, and sustained the formation of long neurites in sensory neurons. When implanted subcutaneously in mice, hydrogels induced no signals of major inflammation and the 50% IKVAV composition induced higher vessel density and formation of nervous terminations in the peripheral tissue. This novel composite has important features for tissue engineering, showing higher osteogenic, angiogenic and innervation potential in vitro, being not inflammatory in vivo, and inducing angiogenesis and innervation subcutaneously. STATEMENT OF SIGNIFICANCE: One of the main limitations in the field of tissue engineering remains the sufficient vascularization and innervation during tissue repair. In this scope, the development of advanced biomaterials that can support these processes is of crucial importance. Here, we formulated different compositions of Elastin-like polypeptide-based hydrogels bearing the IKVAV adhesion sequence. These compositions showed controlled mechanical properties, and were degradable in vitro. Additionally, we could identify in vitro a composition capable to promote neurite formation and to modulate endothelial and mesenchymal stromal cells gene expression, in view of angiogenesis and osteogenesis, respectively. When tested in vivo, it showed no signs of major inflammation and induced the formation of a highly vascularized and innervated neotissue. In this sense, our approach represents a potential advance in the development of new strategies to promote tissue regeneration, taking into account both angiogenesis and innervation.


Asunto(s)
Inductores de la Angiogénesis/química , Materiales Biocompatibles/química , Hidrogeles/química , Andamios del Tejido/química , Inductores de la Angiogénesis/metabolismo , Animales , Materiales Biocompatibles/metabolismo , Proteína Morfogenética Ósea 2/metabolismo , Proliferación Celular , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Elastina/química , Células Endoteliales/metabolismo , Regulación de la Expresión Génica , Humanos , Hidrogeles/metabolismo , Laminina/química , Células Madre Mesenquimatosas/metabolismo , Ratones , Neuronas/metabolismo , Osteogénesis/efectos de los fármacos , Osteopontina/genética , Osteopontina/metabolismo , Fragmentos de Péptidos/química , Péptidos/química , Polietilenglicoles/química , Porosidad , Implantación de Prótesis , Ratas Wistar , Reología , Factor de Transcripción Sp7/genética , Factor de Transcripción Sp7/metabolismo , Propiedades de Superficie , Ingeniería de Tejidos , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
13.
J Biotechnol ; 298: 35-44, 2019 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-30980843

RESUMEN

Elastin-like polypeptides (ELPs) are biocompatible-engineered polypeptides, with promising interest in tissue engineering due to their intrinsic biological and physical properties, and their ease of production. The IKVAV (Ile-Lys-Val-Ala-Val) laminin-1 sequence has been shown to sustain neuron attachment and growth. In this study, the IKVAV adhesion sequence, or a scrambled VKAIV sequence, were incorporated by genetic engineering in the structure of an ELP, expressed in Escherichia coli and purified. The transition temperatures of the ELP-IKVAV and ELP-VKAIV were determined to be 23 °C. Although the phase transition was fully reversible for ELP-VKAIV, we observed an irreversible aggregation for ELP-IKVAV. The corresponding aggregates shared some characteristics with amyloid-like polypeptides. The two ELPs were then reacted with functionalized polyethylene glycol (PEG) to form hydrogels. These hydrogels were characterized for rheological properties, tested with cultures of rat primary sensory neurons, and implanted subcutaneously in mice for 4 weeks. Sensory neurons cultured on high IKVAV concentration hydrogels (20%) formed longer neurite than those of neurons grown on hydrogels containing the scrambled IKVAV sequence. Finally, in vivo evaluation showed the absence of detectable inflammation. In conclusion, this functionalized ELP-IKVAV biomaterial shows interesting properties for tissue engineering requiring neurotization.


Asunto(s)
Elastina/química , Hidrogeles/química , Péptidos/química , Ingeniería de Tejidos , Secuencia de Aminoácidos/genética , Animales , Elastina/genética , Elastina/aislamiento & purificación , Elastina/farmacología , Hidrogeles/farmacología , Laminina/química , Laminina/genética , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Péptidos/genética , Péptidos/aislamiento & purificación , Péptidos/farmacología , Ratas , Reología , Células Receptoras Sensoriales/química , Células Receptoras Sensoriales/efectos de los fármacos
14.
Tissue Eng Part A ; 24(9-10): 703-710, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-28851250

RESUMEN

INTRODUCTION: Standard care for malignant tumors arising next to a bone structure is surgical removal with safety margins, followed by external beam radiotherapy (EBRT). Complete tumor removal can result in large bone defects. A two-step bone reconstruction technique using the induced membrane (IM) technique has proven its efficacy to bridge gap nonunion. During the first step, a spacer is placed in the bone gap. The spacer then is removed and the IM around it is filled with autologous cancellous bone graft. However, the feasibility of this technique with the addition of adjuvant EBRT between the two reconstruction steps has not yet been studied. Polymethyl methacrylate (PMMA) used to be the standard spacer material for the first step. Silicone spacers could replace them owing to their good behavior when submitted to EBRT and their easier removal from the surgical site during the second step. The aim of this study was to evaluate the influence of EBRT on the histological and biochemical properties of IM induced using PMMA or silicone as spacer. MATERIALS AND METHODS: The analyses were performed on PMMA- or silicone-IM with and without EBRT in a 6-mm bilateral femoral defect in 32 rats. Thickness and vessel content were measured in both groups. Bone morphogenetic protein 2 (BMP2) and vascular endothelial growth factor (VEGF) content in lysates of the crushed membranes were measured by enzyme immunoassay. Finally, alkaline phosphatase activity was analyzed in human bone marrow stromal cell cultures in contact with the same lysates. RESULTS: EBRT did not change the histological structure of the cellular internal layer or the fibrous outer layer. The nature of the spacer only influenced IM thickness, PMMA-IM with external radiotherapy being significantly thicker. EBRT decreased the vascular density of IM but was less effective on VEGF/BMP2 production. In vitro, IM could have an osteoinductive potential on human bone marrow stem cells. CONCLUSION: EBRT did not modify the histological properties of IMs but decreased their vascular density. VEGF and BMP2 production within IMs was not affected by EBRT. Silicone spacers are able to induce membranes with similar histological characteristics to PMMA-IM.


Asunto(s)
Huesos/metabolismo , Huesos/patología , Polimetil Metacrilato/química , Siliconas/química , Animales , Proteína Morfogenética Ósea 2/metabolismo , Línea Celular , Femenino , Humanos , Inmunohistoquímica , Cuidados Posoperatorios , Ratas , Factor A de Crecimiento Endotelial Vascular/metabolismo
15.
J Tissue Eng Regen Med ; 12(4): e1936-e1949, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29222846

RESUMEN

Because cell interactions play a fundamental role for cell differentiation, we investigated the expression of Pannexin 1 and Pannexin 3 in human bone marrow mesenchymal stromal cells (HBMSCs) in a three-dimensional (3D) microenvironment provided by a polysaccharide-based macroporous scaffold. The pannexin (Panx) family consists of three members, Panx1, Panx2, and Panx3. The roles of Panx large-pore ion and metabolite channels are recognized in many physiological and pathophysiological scenarios, but the role of these proteins in human physiological processes is still under investigation. Our study demonstrates that HBMSCs cultured within 3D scaffolds have induced Panx1 and Panx3 expression, compared with two-dimensional culture and that the Panx3 gene expression profile correlates with those of bone markers on mesenchymal stromal cells culture into the 3D scaffold. We showed that Panx1 is involved in the HBMSCs 3D cell-cell organization, as acting on the size of cellular aggregates, demonstrated by the use of Probenecid and the mimetic peptide 10panx1 as specific inhibitors. Inhibition of Panx3 using siRNA strategy shows to reduce the expression of osteocalcin as osteoblast-specific marker by HBMSCs cultured in 3D conditions, suggesting a role of this Panx in osteogenesis. Moreover, we evaluated Panx1 and Panx3 expression within the cellularized scaffolds upon subcutaneous implantation in NOG (NOD/Shi-scid/IL-2Rγnull ) mice, where we could observe a more intense expression in the constructs than in the surrounding tissues in vivo. This study provides new insights on the expression of pannexins in HBMSCs on a 3D microenvironment during the osteogenic differentiation, in vitro and in vivo.


Asunto(s)
Células de la Médula Ósea/metabolismo , Técnicas de Cultivo de Célula , Conexinas/biosíntesis , Dextranos/química , Glucanos/química , Células Madre Mesenquimatosas/metabolismo , Proteínas del Tejido Nervioso/biosíntesis , Andamios del Tejido/química , Animales , Células de la Médula Ósea/citología , Xenoinjertos , Humanos , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Ratones , Ratones Endogámicos NOD , Porosidad
16.
J Tissue Eng Regen Med ; 12(3): e1489-e1500, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-28875562

RESUMEN

Tissue engineering is a promising alternative to autografts, allografts, or biomaterials to address the treatment of severe and large bone lesions. Classically, tissue engineering products associate a scaffold and cells and are implanted or injected into the lesion. These cells must be embedded in an appropriate biocompatible scaffold, which offers a favourable environment for their survival and differentiation. Here, we designed a composite hydrogel composed of collagen I, an extracellular matrix protein widely used in several therapeutic applications, which we associated with a physical hydrogel generated from a synthetic small amphiphilic molecule. This composite showed improved mechanical and biological characteristics as compared with gels obtained from each separate compound. Incorporation of the physical hydrogel prevented shrinkage of collagen and cell diffusion out of the gel and yielded a gel with a higher elastic modulus than those of gels obtained with each component alone. The composite hydrogel allowed cell adhesion and proliferation in vitro and long-term cell survival in vivo. Moreover, it promoted the differentiation of human adipose-derived stem cells in the absence of any osteogenic factors. In vivo, cells embedded in the composite gel and injected subcutaneously in immunodeficient mice produced lamellar osteoid tissue and differentiated into osteoblasts. This study points this new composite hydrogel as a promising scaffold for bone tissue engineering applications.


Asunto(s)
Huesos/fisiología , Colágeno/farmacología , Hidrogeles/farmacología , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Tejido Adiposo/citología , Animales , Huesos/efectos de los fármacos , Carbono/química , Adhesión Celular/efectos de los fármacos , Técnicas de Cultivo de Célula , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Halogenación , Humanos , Inyecciones Subcutáneas , Ratas , Células Madre/citología , Células Madre/efectos de los fármacos
17.
Cell Death Dis ; 8(12): 3209, 2017 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-29238079

RESUMEN

Innervation by the sensory nervous system plays a key role in skeletal development and in orchestration of bone remodeling and regeneration. However, it is unclear how and in which bone cells can sensory nerves act to control these processes. Here, we show a microfluidic coculture system comprising dorsal root ganglion (DRG) neurons and mesenchymal stem cells (MSCs) that more faithfully represents the in vivo scenario of bone sensory innervation. We report that DRG neurons promote the osteogenic differentiation capacity of MSCs, by mediating the increase of alkaline phosphatase activity and the upregulation of osteoblast-specific genes. Furthermore, we show that DRG neurons have a positive impact on Cx43 levels in MSCs during osteoblastogenesis, especially at an early stage of this process. Conversely, we described a negative impact of DRG neurons on MSCs N-cadherin expression at a later stage. Finally, we demonstrate a cytoplasmic accumulation of ß-catenin translocation into the nucleus, and subsequently Lymphoid Enhancer Binding Factor 1-responsive transcriptional activation of downstream genes in cocultured MSCs. Together, our study provides a robust body of evidence that the direct interaction of DRG neurons with MSCs in a bone-like microenvironment leads to an enhancement of osteoblast differentiation potential of MSCs. The osteogenic effect of DRG neurons on MSCs is mediated through the regulation of Cx43 and N-cadherin expression and activation of the canonical/ß-catenin Wnt signaling pathway.


Asunto(s)
Cadherinas/genética , Conexina 43/genética , Ganglios Espinales/metabolismo , Células Madre Mesenquimatosas/metabolismo , Proteínas del Tejido Nervioso/genética , Osteoblastos/metabolismo , Células Receptoras Sensoriales/metabolismo , Vía de Señalización Wnt , Fosfatasa Alcalina/genética , Fosfatasa Alcalina/metabolismo , Animales , Huesos/citología , Huesos/metabolismo , Cadherinas/metabolismo , Diferenciación Celular , Proliferación Celular , Técnicas de Cocultivo , Conexina 43/metabolismo , Ganglios Espinales/citología , Regulación de la Expresión Génica , Dispositivos Laboratorio en un Chip , Factor de Unión 1 al Potenciador Linfoide/genética , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Células Madre Mesenquimatosas/citología , Proteínas del Tejido Nervioso/metabolismo , Osteoblastos/citología , Biosíntesis de Proteínas , Ratas , Ratas Wistar , Células Receptoras Sensoriales/citología , Ingeniería de Tejidos/métodos , Transcripción Genética , beta Catenina/genética , beta Catenina/metabolismo
18.
Sci Rep ; 7(1): 1778, 2017 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-28496103

RESUMEN

Bioprinting has emerged as a novel technological approach with the potential to address unsolved questions in the field of tissue engineering. We have recently shown that Laser Assisted Bioprinting (LAB), due to its unprecedented cell printing resolution and precision, is an attractive tool for the in situ printing of a bone substitute. Here, we show that LAB can be used for the in situ printing of mesenchymal stromal cells, associated with collagen and nano-hydroxyapatite, in order to favor bone regeneration, in a calvaria defect model in mice. Also, by testing different cell printing geometries, we show that different cellular arrangements impact on bone tissue regeneration. This work opens new avenues on the development of novel strategies, using in situ bioprinting, for the building of tissues, from the ground up.


Asunto(s)
Bioimpresión/métodos , Regeneración Ósea , Regeneración Tisular Dirigida , Rayos Láser , Células Madre Mesenquimatosas , Animales , Materiales Biocompatibles , Células Cultivadas , Colágeno/metabolismo , Femenino , Regeneración Tisular Dirigida/métodos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Ingeniería de Tejidos , Andamios del Tejido
19.
Acta Biomater ; 54: 377-385, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28242456

RESUMEN

Insufficient angiogenesis remains a major hurdle in current bone tissue engineering strategies. An extensive body of work has focused on the use of angiogenic factors or endothelial progenitor cells. However, these approaches are inherently complex, in terms of regulatory and methodologic implementation, and present a high cost. We have recently demonstrate the potential of electrospun poly(lactic acid) (PLA) fiber-based membranes, containing calcium phosphate (CaP) ormoglass particles, to elicit angiogenesis in vivo, in a subcutaneous model in mice. Here we have devised an injectable composite, containing CaP glass-ceramic particles, dispersed within a (Hydroxypropyl)methyl cellulose (HPMC) matrix, with the capacity to release calcium in a more sustained fashion. We show that by tuning the release of calcium in vivo, in a rat bone defect model, we could improve both bone formation and increase angiogenesis. The bone regeneration kinetics was dependent on the Ca2+ release rate, with the faster Ca2+ release composite gel showing improved bone repair at 3weeks, in relation to control. In the same line, improved angiogenesis could be observed for the same gel formulation at 6weeks post implantation. This methodology allows to integrate two fundamental processes for bone tissue regeneration while using a simple, cost effective, and safe approach. STATEMENT OF SIGNIFICANCE: In current bone tissue engineering approaches the achievement of sufficient angiogenesis, during tissue regeneration, is a major limitation in order to attain full tissue functionality. Recently, we have shown that calcium ions, released by the degradation of calcium phosphate ormoglasses (CaP), are effective angiogenic promoters, in both in vitro and in a subcutaneous implantation model. Here, we devised an injectable composite, containing CaP glass-ceramic particles, dispersed within a HPMC matrix, enabling the release of calcium in a more sustained fashion. We show that by tuning the release of calcium in vivo, in a rat bone defect model, we could improve both bone formation and increase angiogenesis. This simple and cost effective approach holds great promise to translate to the clinics.


Asunto(s)
Evaluación Preclínica de Medicamentos , Células Progenitoras Endoteliales , Neovascularización Fisiológica/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Animales , Calcio/química , Calcio/farmacología , Preparaciones de Acción Retardada/química , Preparaciones de Acción Retardada/farmacología , Células Progenitoras Endoteliales/citología , Células Progenitoras Endoteliales/metabolismo , Células Progenitoras Endoteliales/trasplante , Xenoinjertos , Humanos , Ratones , Poliésteres/química , Poliésteres/farmacología , Ratas , Ratas Wistar
20.
Med Sci (Paris) ; 33(1): 52-59, 2017 Jan.
Artículo en Francés | MEDLINE | ID: mdl-28120756

RESUMEN

Additive manufacturing covers a number of fashionable technologies that attract the interest of researchers in biomaterials and tissue engineering. Additive manufacturing applied to regenerative medicine covers two main areas: 3D printing and biofabrication. If 3D printing has penetrated the world of regenerative medicine, bioassembly and bioimprinting are still in their infancy. The objective of this paper is to make a non-exhaustive review of these different complementary aspects of additive manufacturing in restorative and regenerative medicine or for tissue engineering.


Asunto(s)
Bioimpresión , Impresión Tridimensional , Medicina Regenerativa , Ingeniería de Tejidos , Bioimpresión/tendencias , Humanos , Microtecnología/instrumentación , Microtecnología/métodos , Modelos Anatómicos , Medicina Regenerativa/instrumentación , Medicina Regenerativa/métodos , Ingeniería de Tejidos/instrumentación , Ingeniería de Tejidos/métodos , Andamios del Tejido/química
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda