Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Stem Cells ; 33(9): 2773-84, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26013357

RESUMEN

Tissue engineering using mesenchymal stem cells (MSCs) holds great promise for regenerating critically sized bone defects. While the bone marrow-derived MSC is the most widely studied stromal/stem cell type for this application, its rarity within bone marrow and painful isolation procedure have motivated investigation of alternative cell sources. Adipose-derived stromal/stem cells (ASCs) are more abundant and more easily procured; furthermore, they also possess robust osteogenic potency. While these two cell types are widely considered very similar, there is a growing appreciation of possible innate differences in their biology and response to growth factors. In particular, reports indicate that their osteogenic response to platelet-derived growth factor BB (PDGF-BB) is markedly different: MSCs responded negatively or not at all to PDGF-BB while ASCs exhibited enhanced mineralization in response to physiological concentrations of PDGF-BB. In this study, we directly tested whether a fundamental difference existed between the osteogenic responses of MSCs and ASCs to PDGF-BB. MSCs and ASCs cultured under identical osteogenic conditions responded disparately to 20 ng/ml of PDGF-BB: MSCs exhibited no difference in mineralization while ASCs produced more calcium per cell. siRNA-mediated knockdown of PDGFRß within ASCs abolished their ability to respond to PDGF-BB. Gene expression was also different; MSCs generally downregulated and ASCs generally upregulated osteogenic genes in response to PDGF-BB. ASCs transduced to produce PDGF-BB resulted in more regenerated bone within a critically sized murine calvarial defect compared to control ASCs, indicating PDGF-BB used specifically in conjunction with ASCs might enhance tissue engineering approaches for bone regeneration.


Asunto(s)
Tejido Adiposo/citología , Tejido Adiposo/fisiología , Médula Ósea/fisiología , Células Madre Mesenquimatosas/fisiología , Osteogénesis/fisiología , Proteínas Proto-Oncogénicas c-sis/farmacología , Tejido Adiposo/efectos de los fármacos , Adulto , Animales , Becaplermina , Médula Ósea/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Células Cultivadas , Femenino , Humanos , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Ratones Noqueados , Persona de Mediana Edad , Osteogénesis/efectos de los fármacos , Cráneo/citología , Cráneo/efectos de los fármacos , Cráneo/fisiología , Ingeniería de Tejidos/métodos
2.
Acta Biomater ; 113: 279-288, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32623098

RESUMEN

Stable and mature vascular formation is a current challenge in engineering functional tissues. Transient, non-viral gene delivery presents a unique platform for delivering genetic information to cells for tissue engineering purposes and to restore blood flow to ischemic tissue. The formation of new blood vessels can be induced by upregulation of hypoxia-inducible factor-1α (HIF-1), among other factors. We hypothesized that biodegradable polymers could be used to efficiently deliver the HIF-1α gene to human adipose-derived stromal/stem cells (hASCs) and that this treatment could recruit an existing endogenous endothelial cell population to induce angiogenesis in a 3D cell construct in vitro. In this study, end-modified poly(ß-amino ester) (PBAE) nanocomplexes were first optimized for transfection of hASCs and a new biodegradable polymer with increased hydrophobicity and secondary amine structures, N'-(3-aminopropyl)-N,N-dimethylpropane-1,3-diamine end-modified poly(1,4-butanediol diacrylate-co-4-amino-1-butanol), was found to be most effective. Optimal PBAE nanocomplexes had a hydrodynamic diameter of approximately 140 nm and had a zeta potential of 30 mV. The PBAE polymer self-assembled with HIF-1α plasmid DNA and treatment of hASCs with these nanocomplexes induced 3D vascularization. Cells transfected with this polymer-DNA complex were found to have 106-fold upregulation HIF-1α expression, an approximately 2-fold increase in secreted VEGF, and caused the formation of vessel tubules compared to an untransfected control. These gene therapy biomaterials may be useful for regenerative medicine. STATEMENT OF SIGNIFICANCE: Not only is the formation of stable vasculature a challenge for engineering human tissues in vitro, but it is also of valuable interest to clinical applications such as peripheral artery disease. Previous studies using HIF-1α to induce vascular formation have been limited by the necessity of hypoxic chambers. It would be advantageous to simulate endogenous responses to hypoxia without the need for physical hypoxia. In this study, 3D vascular formation was shown to be inducible through non-viral gene delivery of HIF-1α with new polymeric nanocomplexes. A biodegradable polymer N'-(3-aminopropyl)-N,N-dimethylpropane-1,3-diamine end-modified poly(1,4-butanediol diacrylate-co-4-amino-1-butanol) demonstrates improved transfection of human adipose-derived stem cells. This nanobiotechnology could be a promising strategy for the creation of vasculature for tissue engineering and clinical applications.


Asunto(s)
Técnicas de Transferencia de Gen , Terapia Genética , Subunidad alfa del Factor 1 Inducible por Hipoxia , Tejido Adiposo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Células Madre , Transfección
3.
Tissue Eng Part A ; 22(1-2): 161-9, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26481655

RESUMEN

Vascularization is critical for cell survival within tissue-engineered grafts. Adipose-derived stromal/stem cells (ASCs) are widely used in tissue engineering applications as they are a clinically relevant source of stem cells and endothelial progenitor cells. ASCs have previously been shown to self-assemble into pericyte-stabilized vascular networks in normoxic (20% O2) cultures. This capacity for de novo vascular assembly may accelerate graft vascularization in vivo rather than relying solely on angiogenic ingrowth. However, oxygen depletion within large cell-seeded grafts will be rapid, and it is unclear how this worsening hypoxic environment will impact the vascular assembly of the transplanted cells. The objectives of this study were to determine whether ASC-derived vessels could grow in hypoxia and to assess whether the vessel maturity (i.e., individual cells vs. preformed vessels) influenced this hypoxic response. Utilizing an in vitro vascularization model, ASCs were encapsulated within fibrin gels and cultured in vitro for up to 6 days in either normoxia (20% O2) or hypoxia (0.2% or 2% O2). In a subsequent experiment, vessels were allowed to preform in normoxia for 6 days before an additional 6 days of either normoxia or hypoxia. Viability, vessel growth, pericyte coverage, proliferation, metabolism, and angiogenic factor expression were assessed for each experimental approach. Vessel growth was dramatically inhibited in both moderate and severe hypoxia (47% and 11% total vessel length vs. normoxia, respectively), despite maintaining high cell viability and upregulating endogenous expression of vascular endothelial growth factor in hypoxia. Bromodeoxyuridine labeling indicated significantly reduced proliferation of endothelial cells in hypoxia. In contrast, when vascular networks were allowed to preform for 6 days in normoxia, vessels not only survived but also continued to grow more in hypoxia than those maintained in normoxia. These findings demonstrate that vascular assembly and growth are tightly regulated by oxygen tension and may be differentially affected by hypoxic conditions based on the maturity of the vessels. Understanding this relationship is critical to developing effective approaches to engineer viable tissue-engineered grafts in vivo.


Asunto(s)
Tejido Adiposo/metabolismo , Proliferación Celular , Neovascularización Fisiológica , Células Madre/metabolismo , Tejido Adiposo/citología , Hipoxia de la Célula , Femenino , Humanos , Masculino , Pericitos/citología , Pericitos/metabolismo , Células Madre/citología , Células del Estroma/citología , Células del Estroma/metabolismo
4.
PLoS One ; 9(9): e107199, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25248109

RESUMEN

The innate immune response following bone injury plays an important role in promoting cellular recruitment, revascularization, and other repair mechanisms. Tumor necrosis factor-α (TNF) is a prominent pro-inflammatory cytokine in this cascade, and has been previously shown to improve bone formation and angiogenesis in a dose- and timing-dependent manner. This ability to positively impact both osteogenesis and vascular growth may benefit bone tissue engineering, as vasculature is essential to maintaining cell viability in large grafts after implantation. Here, we investigated the effects of exogenous TNF on the induction of adipose-derived stem/stromal cells (ASCs) to engineer pre-vascularized osteogenic tissue in vitro with respect to dose, timing, and co-stimulation with other inflammatory mediators. We found that acute (2-day), low-dose exposure to TNF promoted vascularization, whereas higher doses and continuous exposure inhibited vascular growth. Co-stimulation with platelet-derived growth factor (PDGF), another key factor released following bone injury, increased vascular network formation synergistically with TNF. ASC-seeded grafts were then cultured within polycaprolactone-fibrin composite scaffolds and implanted in nude rats for 2 weeks, resulting in further tissue maturation and increased angiogenic ingrowth in TNF-treated grafts. VEGF-A expression levels were significantly higher in TNF-treated grafts immediately prior to implantation, indicating a long-term pro-angiogenic effect. These findings demonstrate that TNF has the potential to promote vasculogenesis in engineered osteogenic grafts both in vitro and in vivo. Thus, modulation and/or recapitulation of the immune response following bone injury may be a beneficial strategy for bone tissue engineering.


Asunto(s)
Osteogénesis/efectos de los fármacos , Células Madre/efectos de los fármacos , Células del Estroma/efectos de los fármacos , Grasa Subcutánea/citología , Ingeniería de Tejidos/métodos , Factor de Necrosis Tumoral alfa/farmacología , Animales , Trasplante Óseo/métodos , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Células Cultivadas , Femenino , Humanos , Masculino , Neovascularización Fisiológica/efectos de los fármacos , Ratas , Ratas Desnudas , Trasplante de Células Madre/métodos , Células Madre/citología , Grasa Subcutánea/efectos de los fármacos , Andamios del Tejido
5.
Biomed Mater ; 9(4): 045003, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24945873

RESUMEN

Trabecular bone has an interconnected porous structure, which influences cellular responses, biochemical transport and mechanical strength. Appropriately mimicking this structural organization in biomaterial scaffolds can facilitate more robust bone tissue regeneration and integration by providing a native microenvironment to the cells. This study examined the effect of pore size on human adipose-derived stem/stromal cell (ASC) osteogenesis within poly(ε-caprolactone) (PCL) scaffolds. Scaffold pore size was controlled by porogen leaching of custom-made paraffin particles with three different size ranges: P200 (< 500 µm), P500 (500-1000 µm), and P1000 (1000-1500 µm). Scaffolds produced by leaching these particles exhibited highly interconnected pores and rough surface structures that were favorable for cell attachment and ingrowth. The osteogenic response of ASCs was evaluated following 3 weeks of in vitro culture using biochemical (ALP, Ca(2+)/DNA content), mechanical (compression test) and histological (H&E and von Kossa staining) analyses. It was observed that while the total number of cells was similar for all scaffolds, the cell distributions and osteogenic properties were affected by the scaffold pore size. ASCs were able to bridge smaller pores and grow uniformly within these scaffolds (P200) while they grew as a layer along the periphery of the largest pores (P1000). The cell-biomaterial interactions specific to the latter case led to enhanced osteogenic responses. The ALP activity and Ca(2+) deposition were doubled in P1000 scaffolds as compared to P200 scaffolds. A significant difference was observed between the compressive strength of unseeded and seeded P1000 scaffolds. Therefore, we demonstrated that the use of scaffolds with pores that are in the range of 1 mm enhances in vitro ASC osteogenesis, which may improve their performance in engineered bone substitutes.


Asunto(s)
Adipocitos/citología , Tejido Adiposo/citología , Osteogénesis/fisiología , Células Madre/citología , Células del Estroma/citología , Andamios del Tejido , Materiales Biocompatibles/química , Huesos/metabolismo , Técnicas de Cultivo de Célula , Supervivencia Celular , Células Cultivadas , Fuerza Compresiva , Humanos , Ensayo de Materiales , Oxazinas/química , Parafina/química , Poliésteres/química , Porosidad , Estrés Mecánico , Xantenos/química
6.
J Biomed Mater Res A ; 102(12): 4317-25, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24510413

RESUMEN

The treatment of large craniomaxillofacial bone defects is clinically challenging due to the limited availability of transplantable autologous bone grafts and the complex geometry of the bones. The ability to regenerate new bone tissues that faithfully replicate the anatomy would revolutionize treatment options. Advances in the field of bone tissue engineering over the past few decades offer promising new treatment alternatives using biocompatible scaffold materials and autologous cells. This approach combined with recent advances in three-dimensional (3D) printing technologies may soon allow the generation of large, bioartificial bone grafts with custom, patient-specific architecture. In this study, we use a custom-built 3D printer to develop anatomically shaped polycaprolactone (PCL) scaffolds with varying internal porosities. These scaffolds are assessed for their ability to support induction of human adipose-derived stem cells (hASCs) to form vasculature and bone, two essential components of functional bone tissue. The development of functional tissues is assessed in vitro and in vivo. Finally, we demonstrate the ability to print large mandibular and maxillary bone scaffolds that replicate fine details extracted from patient's computed tomography scans. The findings of this study illustrate the capabilities and potential of 3D printed scaffolds to be used for engineering autologous, anatomically shaped, vascularized bone grafts.


Asunto(s)
Sustitutos de Huesos/química , Impresión Tridimensional , Células Madre/metabolismo , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Tejido Adiposo/citología , Células Cultivadas , Humanos , Anomalías Maxilofaciales/patología , Anomalías Maxilofaciales/terapia , Células Madre/citología
7.
Biomaterials ; 35(10): 3243-51, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24439410

RESUMEN

Hydrogels have been widely used for 3-dimensional (3D) cell culture and tissue regeneration due to their tunable biochemical and physicochemical properties as well as their high water content, which resembles the aqueous microenvironment of the natural extracellular matrix. While many properties of natural hydrogel matrices are modifiable, their intrinsic isotropic structure limits the control over cellular organization, which is critical to restore tissue function. Here we report a generic approach to incorporate alignment topography inside the hydrogel matrix using a combination of electrical and mechanical stretching. Hydrogel fibres with uniaxial alignment were prepared from aqueous solutions of natural polymers such as alginate, fibrin, gelatin, and hyaluronic acid under ambient conditions. The unique internal alignment feature drastically enhances the mechanical properties of the hydrogel microfibres. Furthermore, the facile, organic solvent-free processing conditions are amenable to the incorporation of live cells within the hydrogel fibre or on the fibre surface; both approaches effectively induce cellular alignment. This work demonstrates a versatile and scalable strategy to create aligned hydrogel microfibres from various natural polymers.


Asunto(s)
Hidrogeles/química , Polímeros/química , Materiales Biocompatibles , Microambiente Celular , Microscopía Confocal , Microscopía Electrónica de Rastreo , Microscopía Fluorescente , Dispersión del Ángulo Pequeño
8.
Stem Cell Res Ther ; 4(1): 10, 2013 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-23369796

RESUMEN

Bone is a load-bearing tissue and physical forces play key roles in the development and maintenance of its structure. Mechanical cues can stimulate the expression of an osteogenic phenotype, enhance matrix and mineral deposition, and influence tissue organization to improve the functional outcome of engineered bone grafts. In recent years, a number of studies have investigated the effects of biophysical forces on the bone formation properties of osteoprogenitor cells. The application of physiologically relevant stimuli to tissue-engineered bone may be determined through observation and understanding of forces to which osteoblasts, osteoclasts, and osteocytes are exposed in native bone. Subsequently, these cues may be parameterized and their effects studied in well-defined in vitro systems. The osteo-inductive effects of three specific mechanical cues - shear stress, substrate rigidity, and nanotopography - on cells cultured in monolayer or in three-dimensional biomaterial scaffolds in vitro are reviewed. Additionally, we address the time-dependent effects of mechanical cues on vascular infiltration and de novo bone formation in acellular scaffolds implanted into load-bearing sites in vivo. Recent studies employing cutting-edge advances in biomaterial fabrication and bioreactor design have provided key insights into the role of mechanical cues on cellular fate and tissue properties of engineered bone grafts. By providing mechanistic understanding, future studies may go beyond empirical approaches to rational design of engineering systems to control tissue development.


Asunto(s)
Huesos/fisiología , Osteogénesis/fisiología , Estrés Mecánico , Ingeniería de Tejidos/métodos , Animales , Materiales Biocompatibles/farmacología , Materiales Biocompatibles/uso terapéutico , Reactores Biológicos , Huesos/efectos de los fármacos , Humanos , Osteogénesis/efectos de los fármacos
9.
Tissue Eng Part A ; 19(17-18): 2076-86, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23582144

RESUMEN

Vasculature is essential to the functional integration of a tissue-engineered bone graft to enable sufficient nutrient delivery and viability after implantation. Native bone and vasculature develop through intimately coupled, tightly regulated spatiotemporal cell-cell signaling. The complexity of these developmental processes has been a challenge for tissue engineers to recapitulate, resulting in poor codevelopment of both bone and vasculature within a unified graft. To address this, we cultured adipose-derived stromal/stem cells (ASCs), a clinically relevant, single cell source that has been previously investigated for its ability to give rise to vascularized bone grafts, and studied the effects of initial spatial organization of cells, the temporal addition of growth factors, and the presence of exogenous platelet-derived growth factor-BB (PDGF-BB) on the codevelopment of bone and vascular tissue structures. Human ASCs were aggregated into multicellular spheroids via the hanging drop method before encapsulation and subsequent outgrowth in fibrin gels. Cellular aggregation substantially increased vascular network density, interconnectivity, and pericyte coverage compared to monodispersed cultures. To form robust vessel networks, it was essential to culture ASCs in a purely vasculogenic medium for at least 8 days before the addition of osteogenic cues. Physiologically relevant concentrations of exogenous PDGF-BB (20 ng/mL) substantially enhanced both vascular network stability and osteogenic differentiation. Comparisons with the bone morphogenetic protein-2, another pro-osteogenic and proangiogenic growth factor, indicated that this potential to couple the formation of both lineages might be unique to PDGF-BB. Furthermore, the resulting tissue structure demonstrated the close association of mineral deposits with pre-existing vascular structures that have been described for developing tissues. This combination of a single cell source with a potent induction factor used at physiological concentrations can provide a clinically relevant approach to engineering highly vascularized bone grafts.


Asunto(s)
Adipocitos/citología , Osteogénesis/efectos de los fármacos , Factor de Crecimiento Derivado de Plaquetas/farmacología , Células Madre/citología , Células Madre/efectos de los fármacos , Ingeniería de Tejidos/métodos , Becaplermina , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Femenino , Humanos , Proteínas Proto-Oncogénicas c-sis/farmacología
10.
Stem Cells Dev ; 21(10): 1726-35, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22034957

RESUMEN

Pluripotent and multipotent stem cells adopt an osteoblastic phenotype when cultured in environments that enhance their osteogenic potential. Embryonic stem cells differentiated as embryoid bodies (EBs) in osteogenic medium containing ß-glycerophosphate exhibit increased expression of bone markers, indicating that cells are osteoblastic. Interestingly, 1α,25-dihydroxyvitaminD3 (1,25D) enhances the osteogenic phenotype not just in EBs but also in multipotent adult mesenchymal stem cells (MSCs). 1,25D acts on osteoblasts via classical vitamin D receptors (VDR) and via a membrane 1,25D-binding protein [protein disulfide isomerase family A, member 3 (PDIA3)], which activates protein kinase C-signaling. The aims of this study were to determine whether these receptors are regulated during osteogenic differentiation of stem cells and if stem cells and differentiated progeny are responsive to 1,25D. mRNA and protein levels for VDR, PDIA3, and osteoblast-associated proteins were measured in undifferentiated cells and in cells treated with osteogenic medium. Mouse EBs expressed both VDR and PDIA3, but VDR increased as cells underwent osteogenic differentiation. Human MSCs expressed Pdia3 at constant levels throughout differentiation, but VDR increased in cells treated with osteogenic medium. These results suggest that both 1,25D signaling mechanisms are important, with PDIA3 playing a greater role during early events and VDR playing a greater role in later stages of differentiation. Understanding these coordinated events provide a powerful tool to control pluripotent and multipotent stem cell differentiation through induction medium.


Asunto(s)
Diferenciación Celular , Cuerpos Embrioides/fisiología , Células Madre Mesenquimatosas/fisiología , Osteogénesis , Receptores de Calcitriol/metabolismo , Animales , Antígenos de Diferenciación/metabolismo , Calcitriol/fisiología , Cuerpos Embrioides/metabolismo , Regulación de la Expresión Génica , Glicerofosfatos/fisiología , Humanos , Células Madre Mesenquimatosas/metabolismo , Ratones , Fenotipo , Proteína Disulfuro Isomerasas/genética , Proteína Disulfuro Isomerasas/metabolismo , Receptores de Calcitriol/genética
11.
Tissue Eng Part A ; 18(15-16): 1729-40, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22462659

RESUMEN

Adipose-derived stromal/stem cells (ASCs) are a promising cell source for vascular-based approaches to clinical therapeutics, as they have been shown to give rise to both endothelial and perivascular cells. While it is well known that ASCs can present a heterogeneous phenotypic profile, spontaneous interactions among these subpopulations that result in the formation of complex tissue structures have not been rigorously demonstrated. Our study reports the novel finding that ASCs grown in monolayers in the presence of angiogenic cues are capable of self-assembling into complex, three-dimensional vascular structures. This phenomenon is only apparent when the ASCs are seeded at a high density (20,000 cells/cm(2)) and occur through orchestrated interactions among three distinct subpopulations: CD31-positive cells (CD31+), α-smooth muscle actin-positive cells (αSMA+), and cells that are unstained for both these markers (CD31-/αSMA-). Investigations into the kinetics of the process revealed that endothelial vessel-like structures initially arose from individual CD31+ cells through proliferation and their interactions with CD31-/αSMA- cells. During this period, αSMA+ cells proliferated and appeared to migrate toward the vessel structures, eventually engaging in cell-cell contact with them after 1 week. By 2 weeks, the lumen-containing CD31+ vessels grew greater than a millimeter in length, were lined with vascular basement membrane proteins, and were encased within a dense, three-dimensional cluster of αSMA+ and CD31-/αSMA- cells. The recruitment of αSMA+ cells was largely due to platelet-derived growth factor (PDGF) signaling, as the inhibition of PDGF receptors substantially reduced αSMA+ cell growth and vessel coverage. Additionally, we found that while hypoxia increased endothelial gene expression and vessel width, it also inhibited the growth of the αSMA+ population. Together, these findings underscore the potential use of ASCs in forming mature vessels in vitro as well as the need for a further understanding of the heterotypic interactions among ASC subpopulations.


Asunto(s)
Tejido Adiposo/citología , Vasos Sanguíneos/crecimiento & desarrollo , Comunicación Celular , Morfogénesis , Neovascularización Fisiológica , Células Madre/citología , Vasos Sanguíneos/citología , Recuento de Células , Diferenciación Celular , Hipoxia de la Célula , Proliferación Celular , Células Endoteliales/citología , Femenino , Citometría de Flujo , Humanos , Persona de Mediana Edad , Modelos Biológicos , Pericitos/citología , Pericitos/metabolismo , Fenotipo , Receptores del Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal , Factores de Tiempo
12.
J Bone Miner Res ; 27(8): 1773-83, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22492532

RESUMEN

The surface properties of materials contribute to host cellular response and play a significant role in determining the overall success or failure of an implanted biomaterial. Rough titanium (Ti) surface microtopography and high surface free energy have been shown to enhance osteoblast maturation in vitro and increase bone formation in vivo. Whereas the surface properties of Ti are known to affect osteoblast response, host bone quality also plays a significant role in determining successful osseointegration. One factor affecting host bone quality is patient age. We examined both in vitro and in vivo whether response to Ti surface features was affected by animal age. Calvarial osteoblasts isolated from 1-, 3-, and 11-month-old rats all displayed a reduction in cell number and increases in alkaline phosphatase-specific activity and osteocalcin in response to increasing Ti surface microtopography and surface energy. Further, osteoblasts from the three ages examined displayed increased production of osteocalcin and local factors osteoprotegerin, vascular endothelial growth factor (VEGF)-A, and active transforming growth factor (TGF)-ß1 in response to increasing Ti surface roughness and surface energy. Latent TGF-ß1 only increased in cultures of osteoblasts from 1- and 3-month-old rats. Treatment with the systemic osteotropic hormone 1α,25(OH)(2)D(3) further enhanced the response of osteoblasts to Ti surface features for all three age groups. However, osteoblasts derived from 11-month-old animals had a reduced response to 1α,25(OH)(2)D(3) compared to osteoblasts derived from 1- or 3-month-old animals. These results were confirmed in vivo. Ti implants placed in the femoral intramedullary canal of old (9-month-old) mice yielded lower bone-to-implant contact and neovascularization in response to Ti surface roughness and energy compared to younger (2-month-old) mice. These results show that rodent osteoblast maturation in vitro as well as new bone formation in vivo is reduced with age. Whether comparable age differences exist in humans needs to be determined.


Asunto(s)
Envejecimiento/fisiología , Diferenciación Celular/efectos de los fármacos , Implantes Experimentales , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Titanio/farmacología , Animales , Regulación de la Expresión Génica/efectos de los fármacos , Interacciones Hidrofóbicas e Hidrofílicas/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Osteoblastos/metabolismo , Ratas , Ratas Sprague-Dawley , Propiedades de Superficie
13.
Acta Biomater ; 7(6): 2740-50, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21352958

RESUMEN

The Wnt signaling pathway inhibitor Dickkopf-2 (Dkk2) regulates osteoblast differentiation on microstructured titanium (Ti) surfaces, suggesting involvement of Wnt signaling in this process. To test this, human osteoblast-like MG63 cells were cultured on tissue culture polystyrene or Ti (smooth PT (Ra=0.2 µm), sand-blasted and acid-etched SLA (Ra=3.22 µm), modSLA (hydrophilic SLA)). Expression of Wnt pathway receptors, activators and inhibitors was measured by qPCR. Non-canonical pathway ligands, receptors and intracellular signaling molecules, as well as bone morphogenetic proteins BMP2 and BMP4, were upregulated on SLA and modSLA, whereas canonical pathway members were downregulated. To confirm that non-canonical signaling was involved, cells were cultured daily with exogenous Wnt3a (canonical pathway) or Wnt5a (non-canonical pathway). Alternatively, cells were cultured with antibodies to Wnt3a or Wnt5a to validate that Wnt proteins secreted by the cells were mediating cell responses to the surface. Wnt5a, but not Wnt3a, increased MG63 cell differentiation and BMP2 and BMP4 proteins, suggesting Wnt5a promotes osteogenic differentiation through production of BMPs. Effects of exogenous and endogenous Wnt5a were synergistic with surface microstructure, suggesting the response also depends on cell maturation state. These results indicate a major role for the non-canonical, calcium-dependent Wnt pathway in differentiation of osteoblasts on microstructured titanium surfaces during implant osseointegration.


Asunto(s)
Osteoblastos/citología , Transducción de Señal , Titanio , Proteínas Wnt/metabolismo , Secuencia de Bases , Línea Celular , Cartilla de ADN , Humanos , Reacción en Cadena de la Polimerasa , Propiedades de Superficie
14.
Biomaterials ; 31(10): 2728-35, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20053436

RESUMEN

Microstructured and high surface energy titanium substrates increase osseointegration in vivo. In vitro, osteoblast differentiation is increased, but effects of the surface directly on multipotent mesenchymal stem cells (MSCs) and consequences for MSCs in the peri-implant environment are not known. We evaluated responses of human MSCs to substrate surface properties and examined the underlying mechanisms involved. MSCs exhibited osteoblast characteristics (alkaline phosphatase, RUNX2, and osteocalcin) when grown on microstructured Ti; this effect was more robust with increased hydrophilicity. Factors produced by osteoblasts grown on microstructured Ti were sufficient to induce co-cultured MSC differentiation to osteoblasts. Silencing studies showed that this was due to signaling via alpha2beta1 integrins in osteoblasts on the substrate surface and paracrine action of secreted Dkk2. Thus, human MSCs are sensitive to substrate properties that induce osteoblastic differentiation; osteoblasts interact with these surface properties via alpha2beta1 and secrete Dkk2, which acts on distal MSCs.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Células Madre Mesenquimatosas/citología , Osteoblastos/citología , Titanio/química , Titanio/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Propiedades de Superficie/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA