Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Regen Eng Transl Med ; 5(1): 30-41, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31008183

RESUMEN

Control of microvascular network growth is critical to treatment of ischemic tissue diseases and enhancing regenerative capacity of tissue engineering implants. Conventional therapeutic strategies for inducing angiogenesis aim to deliver one or more proangiogenic cytokines or to over-express known pro-angiogenic genes, but seldom address potential compensatory or cooperative effects between signals and the overarching signaling pathways that determine successful outcomes. An emerging grand challenge is harnessing the expanding knowledge base of angiogenic signaling pathways toward development of successful new therapies. We previously performed drug optimization studies by various substitutions of a 2-(2,6-dioxo-3-piperidyl)isoindole-1,3-dione scaffold to discover novel bioactive small molecules capable of inducing growth of microvascular networks, the most potent of which we termed phthalimide neovascularization factor 1 (PNF1, formerly known as SC-3-149). We then showed that PNF-1 regulates the transcription of signaling molecules that are associated with vascular initiation and maturation in a time-dependent manner through a novel pathway compendium analysis in which transcriptional regulatory networks of PNF-1-stimulated microvascular endothelial cells are overlaid with literature-derived angiogenic pathways. In this study, we generated three analogues (SC-3-143, SC-3-263, SC-3-13) through systematic transformations to PNF1 to evaluate the effects of electronic, steric, chiral, and hydrogen bonding changes on angiogenic signaling. We then expanded our compendium analysis toward these new compounds. Variables obtained from the compendium analysis were then used to construct a PLSR model to predict endothelial cell proliferation. Our combined approach suggests mechanisms of action involving suppression of VEGF pathways through TGF-ß andNR3C1 network activation.

2.
Cell Mol Bioeng ; 11(4): 241-253, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29983824

RESUMEN

INTRODUCTION: Mesenchymal stem and progenitor cells (MSCs), which normally reside in the bone marrow, are critical to bone health and can be recruited to sites of traumatic bone injury, contributing to new bone formation. The ability to control the trafficking of MSCs provides therapeutic potential for improving traumatic bone healing and therapy for genetic bone diseases such as hypophosphatasia. METHODS: In this study, we explored the sphingosine-1-phosphate (S1P) signaling axis as a means to control the mobilization of MSCs into blood and possibly to recruit MSCs enhancing bone growth. RESULTS: Loss of S1P receptor 3 (S1PR3) leads to an increase in circulating CD45-/CD29+/CD90+/Sca1 putative mesenchymal progenitor cells, suggesting that blocking S1PR3 may stimulate MSCs to leave the bone marrow. Antagonism of S1PR3 with the small molecule VPC01091 stimulated acute migration of CD45-/CD29+/CD90+/Sca1+ MSCs into the blood as early as 1.5 hours after treatment. VPC01091 administration also increased ectopic bone formation induced by BMP-2 and significantly increased new bone formation in critically sized rat cranial defects, suggesting that mobilized MSCs may home to injuries to contribute to healing. We also explored the possibility of combining S1P manipulation of endogenous host cell occupancy with exogenous MSC transplantation for potential use in combination therapies. Importantly, reducing niche occupancy of host MSCs with VPC01091 does not impede engraftment of exogenous MSCs. CONCLUSIONS: Our studies suggest that MSC mobilization through S1PR3 antagonism is a promising strategy for endogenous tissue engineering and improving MSC delivery to treat bone diseases.

3.
Stem Cells ; 35(4): 1040-1052, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28026131

RESUMEN

Hematopoietic stem and progenitor cells (HSPCs) egress from bone marrow (BM) during homeostasis and at increased rates during stress; however, the mechanisms regulating their trafficking remain incompletely understood. Here we describe a novel role for lipid receptor, sphingosine-1-phosphate receptor 3 (S1PR3), in HSPC residence within the BM niche. HSPCs expressed increased levels of S1PR3 compared to differentiated BM cells. Pharmacological antagonism or knockout (KO) of S1PR3 mobilized HSPCs into blood circulation, suggesting that S1PR3 influences niche localization. S1PR3 antagonism suppressed BM and plasma SDF-1, enabling HSPCs to migrate toward S1P-rich plasma. Mobilization synergized with AMD3100-mediated antagonism of CXCR4, which tethers HSPCs in the niche, and recovered homing deficits of AMD3100-treated grafts. S1PR3 antagonism combined with AMD3100 improved re-engraftment and survival in lethally irradiated recipients. Our studies indicate that S1PR3 and CXCR4 signaling cooperate to maintain HSPCs within the niche under homeostasis. These results highlight an important role for S1PR3 in HSPC niche occupancy and trafficking that can be harnessed for both rapid clinical stem cell mobilization and re-engraftment strategies, as well as the opportunity to design novel therapeutics for control of recruitment, homing, and localization through bioactive lipid signaling. Stem Cells 2017;35:1040-1052.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Animales , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Adhesión Celular/efectos de los fármacos , Microambiente Celular/efectos de los fármacos , Quimiotaxis/efectos de los fármacos , Movilización de Célula Madre Hematopoyética , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/efectos de los fármacos , Ligandos , Lisofosfolípidos/farmacología , Masculino , Ratones Endogámicos C57BL , Radiación Ionizante , Esfingosina/análogos & derivados , Esfingosina/farmacología , Nicho de Células Madre/efectos de los fármacos
5.
Drug Deliv Transl Res ; 6(2): 96-104, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26169381

RESUMEN

In this study, we used extracellular matrix (ECM) gels and human bone allograft as matrix vehicles to deliver the sphingolipid growth factor FTY720 to rodent models of tibial fracture and a critical-sized cranial defect. We show that FTY720 released from injectable ECM gels may accelerate callous formation and resolution and bone volume in a mouse tibial fracture model. We then show that FTY720 binds directly to human trabecular allograft bone and releases over 1 week in vitro. Rat critical-sized cranial defects treated with FTY720-coated grafts show increases in vascularization and bone deposition, with histological and micro-computed topography (microCT) evidence of enhanced bone formation within the graft and defect void. Immunohistochemical analysis suggests that osteogenesis within FTY720-coated grafts is associated with reduced CD68(+) macrophage infiltration and recruitment of CD29(+) bone progenitor cells. Matrix binding of FTY720 thus represents a promising and robust bone regeneration strategy with potential clinical translatability.


Asunto(s)
Hueso Esponjoso/citología , Matriz Extracelular/química , Clorhidrato de Fingolimod/administración & dosificación , Fracturas Óseas/terapia , Cráneo/lesiones , Tibia/lesiones , Animales , Regeneración Ósea , Trasplante Óseo/métodos , Hueso Esponjoso/efectos de los fármacos , Modelos Animales de Enfermedad , Clorhidrato de Fingolimod/farmacología , Humanos , Ratones , Ratas , Trasplante Homólogo
6.
Cell Tissue Res ; 364(1): 125-35, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26475719

RESUMEN

We have previously shown that the combined delivery of mesenchymal stem cells (MSCs), vascular endothelial growth factor (VEGF) and bone morphogenetic protein 6 (BMP-6) induces significantly more bone formation than that induced by the delivery of any single factor or a combination of any two factors. We now determine whether the exogenous addition of VEGF and BMP-6 is sufficient for bone healing when MSCs are not provided. Poly(lactic-co-glycolic acid) (PLAGA) microsphere-based three-dimensional scaffolds (P) were fabricated by thermal sintering of PLAGA microspheres. The scaffolds were chemically cross-linked with 200 ng recombinant human VEGF (P(VEGF)) or BMP-6 (P(BMP-6)) or both (P(VEGF+BMP-6)) by the EDC-NHS-MES method. Release of the proteins from the scaffolds was detected for 21 days in vitro which confirmed their comparable potential to supply the proteins in vivo. The scaffolds were delivered to a critical-sized mandibular defect created in 32 Sprague Dawley rats. Significant bone regeneration was observed only in rats with P(VEGF+BMP-6) scaffolds at weeks 2, 8 and 12 as revealed by micro-computer tomography. Vascular ingrowth was higher in the P(VEGF+BMP-6) group as seen by microfil imaging than in other groups. Trichrome staining revealed that a soft callus formed in P(VEGF), P(BMP-6) and P(VEGF+BMP-6) but not in P. MSCs isolated from rat femurs displayed expression of the bone-specific marker osteocalcin when cultured with P(VEGF), P(BMP-6), or P(VEGF+BMP-6) but not with P. Robust mineralization and increased alkaline phosphatase gene expression were seen in rat MSCs when cultured on P(VEGF+BMP-6) but not on P, P(VEGF), or P(BMP-6). Thus, unlike the delivery of VEGF or BMP-6 alone, the combined delivery of VEGF and BMP-6 to the bone defect significantly enhanced bone repair through the enhancement of angiogenesis and the differentiation of endogenously recruited MSCs into the bone repair site.


Asunto(s)
Proteína Morfogenética Ósea 6 , Ácido Láctico , Enfermedades Mandibulares/terapia , Células Madre Mesenquimatosas/metabolismo , Ácido Poliglicólico , Andamios del Tejido/química , Factor A de Crecimiento Endotelial Vascular , Animales , Proteína Morfogenética Ósea 6/química , Proteína Morfogenética Ósea 6/farmacología , Humanos , Ácido Láctico/química , Ácido Láctico/farmacología , Mandíbula/metabolismo , Mandíbula/patología , Enfermedades Mandibulares/patología , Células Madre Mesenquimatosas/patología , Ácido Poliglicólico/química , Ácido Poliglicólico/farmacología , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Ratas , Ratas Endogámicas F344 , Factor A de Crecimiento Endotelial Vascular/química , Factor A de Crecimiento Endotelial Vascular/farmacología
7.
Biomaterials ; 64: 98-107, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26125501

RESUMEN

Bone grafting procedures are performed to treat wounds incurred during wartime trauma, accidents, and tumor resections. Endogenous mechanisms of repair are often insufficient to ensure integration between host and donor bone and subsequent restoration of function. We investigated the role that bone marrow-derived cells play in bone regeneration and sought to increase their contributions by functionalizing bone allografts with bioactive lipid coatings. Polymer-coated allografts were used to locally deliver the immunomodulatory small molecule FTY720 in tibial defects created in rat bone marrow chimeras containing genetically-labeled bone marrow for monitoring cell origin and fate. Donor bone marrow contributed significantly to both myeloid and osteogenic cells in remodeling tissue surrounding allografts. FTY720 coatings altered the phenotype of immune cells two weeks post-injury, which was associated with increased vascularization and bone formation surrounding allografts. Consequently, degradable polymer coating strategies that deliver small molecule growth factors such as FTY720 represent a novel therapeutic strategy for harnessing endogenous bone marrow-derived progenitors and enhancing healing in load-bearing bone defects.


Asunto(s)
Trasplante Óseo , Clorhidrato de Fingolimod/administración & dosificación , Factores Inmunológicos/administración & dosificación , Aloinjertos , Animales , Células de la Médula Ósea/citología , Huesos/fisiología , Linaje de la Célula , Portadores de Fármacos/administración & dosificación , Sistemas de Liberación de Medicamentos , Femenino , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Ácido Láctico/administración & dosificación , Masculino , Mielopoyesis/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Ácido Poliglicólico/administración & dosificación , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Quimera por Radiación , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Regeneración , Estrés Mecánico , Tibia/lesiones , Tibia/cirugía , Microtomografía por Rayos X
9.
Craniomaxillofac Trauma Reconstr ; 8(1): 23-30, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25709750

RESUMEN

Craniofacial fractures and bony defects are common causes of morbidity and contribute to increasing health care costs. Successful regeneration of bone requires the concomitant processes of osteogenesis and neovascularization. Current methods of repair and reconstruction include rigid fixation, grafting, and free tissue transfer. However, these methods carry innate complications, including plate extrusion, nonunion, graft/flap failure, and donor site morbidity. Recent research efforts have focused on using stem cells and synthetic scaffolds to heal critical-sized bone defects similar to those sustained from traumatic injury or ablative oncologic surgery. Growth factors can be used to augment both osteogenesis and neovascularization across these defects. Many different growth factor delivery techniques and scaffold compositions have been explored yet none have emerged as the universally accepted standard. In this review, we will discuss the recent literature regarding the use of stem cells, growth factors, and synthetic scaffolds as alternative methods of craniofacial fracture repair.

10.
Tissue Eng Part A ; 21(1-2): 202-13, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25315888

RESUMEN

Local delivery of lipid mediators has become a promising new approach for therapeutic angiogenesis and regenerative medicine. In this study, we investigated how gradient stimulation (either abluminal/distal or luminal/proximal) of engineered microvessels with sphingosine 1-phosphate (S1P) receptor-subtype-targeted molecules affects endothelial sprout growth using a microfluidic device. Our studies show that distal stimulation of microvessels with FTY720, an S1P1/3 selective agonist, promotes both arterial and venular sprout growth, whereas proximal stimulation does not. Using novel pharmacological antagonists of S1P receptor subtypes, we further show that S1P3 functionality is necessary for VEGF-induced sprouting, and confirmed these findings ex vivo using a murine aortic ring assay from S1P3-deficient mice. S1P3 agonist stimulation enhanced vascular stability in both cell types via upregulation of the interendothelial junction protein VE-cadherin. Lastly, S1P3 activation under flow promoted endothelial sprouting and branching while decreasing migratory cell fate in the microfluidic device. We used an in vivo murine dorsal skinfold window chamber model to confirm S1P3's role in neovascular branching. Together, these data suggest that a distal transendothelial gradient of S1P1/3-targeted drugs is an effective technique for both enhancing and stabilizing capillary morphogenesis in angiogenic applications.


Asunto(s)
Células Endoteliales/metabolismo , Neovascularización Fisiológica , Receptores de Lisoesfingolípidos/metabolismo , Animales , Antígenos CD , Aorta/citología , Cadherinas , Movimiento Celular/efectos de los fármacos , Difusión , Células Endoteliales/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Técnicas In Vitro , Masculino , Ratones Endogámicos C57BL , Receptores de Lisoesfingolípidos/agonistas , Receptores de Lisoesfingolípidos/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/farmacología
11.
Plast Reconstr Surg ; 135(1): 124e-134e, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25539319

RESUMEN

BACKGROUND: Radiation therapy results in permanent damage to the microvasculature, leading to dermal damage and inelasticity in normal tissues. Deferoxamine is a U.S. Food and Drug Administration-approved iron-chelating medication that has also been shown to increase angiogenesis. The authors hypothesize that the application of deferoxamine will result in increased vascularity and improved tissue elasticity in a rat irradiated transverse rectus abdominis musculocutaneous flap model. METHODS: Fifteen rats underwent a transverse rectus abdominis myocutaneous flap and were randomized to three groups: control, radiation therapy, and radiation therapy plus deferoxamine. The flaps in the radiation therapy and radiation therapy plus deferoxamine groups were irradiated with 35 Gy in a single dose. Four weeks after irradiation, rats in the radiation therapy plus deferoxamine group were treated with deferoxamine. Flaps were imaged with micro-computed tomographic angiography. Flap creep and stress relaxation were assessed using a tensiometer. Hematoxylin and eosin, picrosirius red, and Verhoeff-van Gieson staining was performed. RESULTS: Irradiated flaps demonstrated gross stigmata of cutaneous radiation injury within 4 weeks. Histologically, the epidermis in the radiation therapy flaps was found to be thicker than in the radiation therapy plus deferoxamine and control flaps (p < 0.001). Micro-computed tomographic angiography demonstrated a statistically significant (p < 0.05) increase in vascularity in the radiation therapy plus deferoxamine flaps compared with radiation therapy alone. The creep curve was indicative of increased elasticity in the radiation therapy plus deferoxamine flaps compared with radiation therapy flaps. CONCLUSION: Deferoxamine appears to mitigate radiation-induced hypovascularity and improve tissue elasticity in a rat model of soft-tissue reconstruction.


Asunto(s)
Deferoxamina/uso terapéutico , Traumatismos por Radiación/tratamiento farmacológico , Colgajos Quirúrgicos , Animales , Modelos Animales de Enfermedad , Masculino , Ratas , Ratas Sprague-Dawley , Recto del Abdomen/trasplante
12.
ACS Nano ; 8(12): 12080-91, 2014 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-25426706

RESUMEN

Oxygenation in tissue scaffolds continues to be a limiting factor in regenerative medicine despite efforts to induce neovascularization or to use oxygen-generating materials. Unfortunately, many established methods to measure oxygen concentration, such as using electrodes, require mechanical disturbance of the tissue structure. To address the need for scaffold-based oxygen concentration monitoring, a single-component, self-referenced oxygen sensor was made into nanofibers. Electrospinning process parameters were tuned to produce a biomaterial scaffold with specific morphological features. The ratio of an oxygen sensitive phosphorescence signal to an oxygen insensitive fluorescence signal was calculated at each image pixel to determine an oxygenation value. A single component boron dye-polymer conjugate was chosen for additional investigation due to improved resistance to degradation in aqueous media compared to a boron dye polymer blend. Standardization curves show that in fully supplemented media, the fibers are responsive to dissolved oxygen concentrations less than 15 ppm. Spatial (millimeters) and temporal (minutes) ratiometric gradients were observed in vitro radiating outward from the center of a dense adherent cell grouping on scaffolds. Sensor activation in ischemia and cell transplant models in vivo show oxygenation decreases on the scale of minutes. The nanofiber construct offers a robust approach to biomaterial scaffold oxygen sensing.


Asunto(s)
Boro/química , Colorantes/química , Nanofibras/química , Nanotecnología/instrumentación , Oxígeno/metabolismo , Poliésteres/química , Animales , Línea Celular , Islotes Pancreáticos/citología , Ácido Láctico/química , Ratones , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Prótesis e Implantes , Análisis Espacio-Temporal , Ingeniería de Tejidos , Andamios del Tejido/química
13.
PLoS One ; 9(7): e101276, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25077607

RESUMEN

In this study, a microgel composed of chitosan and inorganic phosphates was used to deliver poly(lactic-co-glycolic acid) (PLAGA) microspheres loaded with sphingolipid growth factor FTY720 to critical size cranial defects in Sprague Dawley rats. We show that sustained release of FTY720 from injected microspheres used alone or in combination with recombinant human bone morphogenic protein-2 (rhBMP2) improves defect vascularization and bone formation in the presence and absence of rhBMP2 as evaluated by quantitative microCT and histological measurements. Moreover, sustained delivery of FTY720 from PLAGA and local targeting of sphingosine 1-phosphate (S1P) receptors reduces CD45+ inflammatory cell infiltration, promotes endogenous recruitment of CD29+CD90+ bone progenitor cells and enhances the efficacy of rhBMP2 from chitosan microgels. Companion in vitro studies suggest that selective activation of sphingosine receptor subtype-3 (S1P3) via FTY720 treatment induces smad-1 phosphorylation in bone-marrow stromal cells. Additionally, FTY720 enhances stromal cell-derived factor-1 (SDF-1) mediated chemotaxis of CD90+CD11B-CD45- bone progenitor cells in vitro after stimulation with rhBMP2. We believe that use of such small molecule delivery formulations to recruit endogenous bone progenitors may be an attractive alternative to exogenous cell-based therapy.


Asunto(s)
Regeneración Ósea , Geles , Lípidos/administración & dosificación , Microesferas , Células Madre/citología , Andamios del Tejido , Anomalías Múltiples , Animales , Catarata/congénito , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Córnea/anomalías , Femenino , Clorhidrato de Fingolimod , Hipogonadismo , Discapacidad Intelectual , Ratones , Ratones Endogámicos C57BL , Microcefalia , Atrofia Óptica , Glicoles de Propileno/farmacología , Ratas , Ratas Sprague-Dawley , Esfingosina/análogos & derivados , Esfingosina/farmacología
14.
J Biomed Mater Res A ; 102(4): 1210-8, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23640833

RESUMEN

Biodegradable polymer scaffolds can be used to deliver soluble factors to enhance osseous remodeling in bone defects. To this end, we designed a poly(lactic-co-glycolic acid) (PLAGA) microsphere scaffold to sustain the release of FTY720, a selective agonist for sphingosine 1-phosphate (S1P) receptors. The microsphere scaffolds were created from fast degrading 50:50 PLAGA and/or from slow-degrading 85:15 PLAGA. Temporal and spatial regulation of bone remodeling depended on the use of appropriate scaffolds for drug delivery. The release profiles from the scaffolds were used to design an optimal delivery system to treat critical size cranial defects in a rodent model. The ability of local FTY720 delivery to maximize bone regeneration was evaluated with micro-computed tomography (microCT) and histology. Following 4 weeks of defect healing, FTY720 delivery from 85:15 PLAGA scaffolds resulted in a significant increase in bone volumes in the defect region compared to the controls. A 85:15 microsphere scaffolds maintain their structural integrity over a longer period of time, and cause an initial burst release of FTY720 due to surface localization of the drug. This encourages cellular in-growth and an increase in new bone formation.


Asunto(s)
Regeneración Ósea/efectos de los fármacos , Sistemas de Liberación de Medicamentos , Ácido Láctico/química , Ácido Poliglicólico/química , Glicoles de Propileno/farmacología , Receptores de Lisoesfingolípidos/metabolismo , Cráneo/patología , Esfingosina/análogos & derivados , Animales , Clorhidrato de Fingolimod , Interacciones Hidrofóbicas e Hidrofílicas/efectos de los fármacos , Espectroscopía de Resonancia Magnética , Masculino , Microesferas , Osteogénesis/efectos de los fármacos , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Ratas , Ratas Sprague-Dawley , Cráneo/efectos de los fármacos , Esfingosina/farmacología , Factores de Tiempo , Andamios del Tejido/química
17.
Biomaterials ; 34(38): 9853-62, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24064148

RESUMEN

Endogenous signals originating at the site of injury are involved in the paracrine recruitment, proliferation, and differentiation of circulating progenitor and diverse inflammatory cell types. Here, we investigate a strategy to exploit endogenous cell recruitment mechanisms to regenerate injured bone by local targeting and activation of sphingosine-1-phosphate (S1P) receptors. A mandibular defect model was selected for evaluating regeneration of bone following trauma or congenital disease. The particular challenges of mandibular reconstruction are inherent in the complex anatomy and function of the bone given that the area is highly vascularized and in close proximity to muscle. Nanofibers composed of poly(DL-lactide-co-glycolide) (PLAGA) and polycaprolactone (PCL) were used to delivery FTY720, a targeted agonist of S1P receptors 1 and 3. In vitro culture of bone progenitor cells on drug-loaded constructs significantly enhanced SDF1α mediated chemotaxis of bone marrow mononuclear cells. In vivo results show that local delivery of FTY720 from composite nanofibers enhanced blood vessel ingrowth and increased recruitment of M2 alternatively activated macrophages, leading to significant osseous tissue ingrowth into critical sized defects after 12 weeks of treatment. These results demonstrate that local activation of S1P receptors is a regenerative cue resulting in recruitment of wound healing or anti-inflammatory macrophages and bone healing. Use of such small molecule therapy can provide an alternative to biological factors for the clinical treatment of critical size craniofacial defects.


Asunto(s)
Macrófagos/metabolismo , Mandíbula , Nanofibras/química , Receptores de Lisoesfingolípidos/metabolismo , Cicatrización de Heridas/fisiología , Animales , Clorhidrato de Fingolimod , Lisofosfolípidos/química , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Poliésteres/química , Glicoles de Propileno/química , Ratas , Esfingosina/análogos & derivados , Esfingosina/química
18.
World J Orthop ; 3(5): 49-57, 2012 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-22655222

RESUMEN

Osteonecrosis is a phenomenon involving disruption to the vascular supply to the femoral head, resulting in articular surface collapse and eventual osteoarthritis. Although alcoholism, steroid use, and hip trauma remain the most common causes, several other etiologies for osteonecrosis have been identified. Basic science research utilizing animal models and stem cell applications continue to further elucidate the pathophysiology of osteonecrosis and promise novel treatment options in the future. Clinical studies evaluating modern joint-sparing procedures have demonstrated significant improvements in outcomes, but hip arthroplasty is still the most common procedure performed in these affected younger adults. Further advances in joint-preserving procedures are required and will be widely studied in the coming decade.

19.
Cell Tissue Res ; 347(3): 553-66, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21863314

RESUMEN

Endogenous stem cell recruitment to the site of skeletal injury is key to enhanced osseous remodeling and neovascularization. To this end, this study utilized a novel bone allograft coating of poly(lactic-co-glycolic acid) (PLAGA) to sustain the release of FTY720, a selective agonist for sphingosine 1-phosphate (S1P) receptors, from calvarial allografts. Uncoated allografts, vehicle-coated, low dose FTY720 in PLAGA (1:200 w:w) and high dose FTY720 in PLAGA (1:40) were implanted into critical size calvarial bone defects. The ability of local FTY720 delivery to promote angiogenesis, maximize osteoinductivity and improve allograft incorporation by recruitment of bone progenitor cells from surrounding soft tissues and microcirculation was evaluated. FTY720 bioactivity after encapsulation and release was confirmed with sphingosine kinase 2 assays. HPLC-MS quantified about 50% loaded FTY720 release of the total encapsulated drug (4.5 µg) after 5 days. Following 2 weeks of defect healing, FTY720 delivery led to statistically significant increases in bone volumes compared to controls, with total bone volume increases for uncoated, coated, low FTY720 and high FTY720 of 5.98, 3.38, 7.2 and 8.9 mm(3), respectively. The rate and extent of enhanced bone growth persisted through week 4 but, by week 8, increases in bone formation in FTY720 groups were no longer statistically significant. However, micro-computed tomography (microCT) of contrast enhanced vascular ingrowth (MICROFIL®) and histological analysis showed enhanced integration as well as directed bone growth in both high and low dose FTY720 groups compared to controls.


Asunto(s)
Trasplante Óseo , Sistemas de Liberación de Medicamentos/métodos , Osteogénesis/efectos de los fármacos , Glicoles de Propileno/administración & dosificación , Glicoles de Propileno/farmacología , Cráneo/efectos de los fármacos , Esfingosina/análogos & derivados , Actinas/metabolismo , Animales , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/crecimiento & desarrollo , Remodelación Ósea/efectos de los fármacos , Materiales Biocompatibles Revestidos/química , Femenino , Clorhidrato de Fingolimod , Fluorescencia , Ácido Láctico/química , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Periostio/irrigación sanguínea , Periostio/diagnóstico por imagen , Periostio/efectos de los fármacos , Periostio/crecimiento & desarrollo , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Ratas , Ratas Sprague-Dawley , Cráneo/irrigación sanguínea , Cráneo/patología , Cráneo/cirugía , Esfingosina/administración & dosificación , Esfingosina/farmacología , Trasplante Homólogo , Microtomografía por Rayos X
20.
Tissue Eng Part B Rev ; 17(6): 403-14, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21902609

RESUMEN

Bone regeneration has long been a major focus for tissue engineers and the importance of vascularization to the bone regeneration process has been well documented. Over the past decade, technological advances in the areas of stem cell biology, scaffold fabrication, and protein engineering have significantly enhanced our understanding of the interplay between vascularization and bone growth. This review, therefore, describes the commonly used models for investigating the complex interactions between osteoblastic cells and endothelial cells, evaluates the different tools utilized to investigate the relationship between vascularization and bone growth in vivo, and finally, summarizes possible areas of research related to therapeutic development.


Asunto(s)
Neovascularización Fisiológica , Osteogénesis/fisiología , Animales , Regeneración Ósea/fisiología , Comunicación Celular , Células Endoteliales/citología , Humanos , Andamios del Tejido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...