Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 22(11)2021 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-34071329

RESUMEN

Avascular transplantation of frozen-thawed testicular tissue fragments represents a potential future technique for fertility restoration in boys with cancer. A significant loss of spermatogonia was observed in xeno-transplants of human tissue most likely due to the hypoxic period before revascularization. To reduce the effect of hypoxia-reoxygenation injuries, several options have already been explored, like encapsulation in alginate hydrogel and supplementation with nanoparticles delivering a necrosis inhibitor (NECINH) or VEGF. While these approaches improved short-term (5 days) vascular surfaces in grafts, neovessels were not maintained up to 21 days; i.e., the time needed for achieving vessel stabilization. To better support tissue grafts, nanoparticles loaded with VEGF, PDGF and NECINH were developed. Testicular tissue fragments from 4-5-week-old mice were encapsulated in calcium-alginate hydrogels, either non-supplemented (control) or supplemented with drug-loaded nanoparticles (VEGF-nanoparticles; VEGF-nanoparticles + PDGF-nanoparticles; NECINH-nanoparticles; VEGF-nanoparticles + NECINH-nanoparticles; and VEGF-nanoparticles + PDGF-nanoparticles + NECINH-nanoparticles) before auto-transplantation. Grafts were recovered after 5 or 21 days for analyses of tissue integrity (hematoxylin-eosin staining), spermatogonial survival (immuno-histo-chemistry for promyelocytic leukemia zinc finger) and vascularization (immuno-histo-chemistry for α-smooth muscle actin and CD-31). Our results showed that a combination of VEGF and PDGF nanoparticles increased vascular maturity and induced a faster maturation of vascular structures in grafts.


Asunto(s)
Hidrogeles/química , Nanopartículas/administración & dosificación , Neovascularización Fisiológica/efectos de los fármacos , Factor de Crecimiento Derivado de Plaquetas/administración & dosificación , Testículo/trasplante , Factor A de Crecimiento Endotelial Vascular/administración & dosificación , Alginatos/química , Animales , Liberación de Fármacos , Preservación de la Fertilidad/métodos , Humanos , Masculino , Ratones Endogámicos , Nanopartículas/química , Factor de Crecimiento Derivado de Plaquetas/química , Factor de Crecimiento Derivado de Plaquetas/farmacocinética , Espermatogonias/efectos de los fármacos , Testículo/irrigación sanguínea , Factor A de Crecimiento Endotelial Vascular/química , Factor A de Crecimiento Endotelial Vascular/farmacocinética
2.
J Periodontal Res ; 52(4): 772-786, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28261803

RESUMEN

BACKGROUND AND OBJECTIVE: In regenerative dentistry, platelet preparations are applied to stimulate bone healing and periodontal regeneration. Here, we pursue a strategy where bone substitutes are used as carriers for platelet-released supernatants. The mitogenic capacity and release kinetics of loaded bone substitutes were assessed. MATERIAL AND METHODS: Platelet-released supernatants of washed platelets (washed PRS) and platelet-released supernatants of unwashed platelets (unwashed PRS) were lyophilized onto the bone substitutes deproteinized bovine bone mineral, hydroxyapatite and ß-tricalcium phosphate. Scanning electron microscopy images were taken. Supernatants of bone substitutes were collected at hours 1, 3, 6, 24, and 48 and medium was replaced. We evaluated the protein content with the bicinchoninic acid assay and the effect on proliferation using bioassays with human periodontal fibroblasts. Release of growth factors from the loaded bone substitutes was measured based on the platelet-derived growth factor isoform (PDGF-BB) and thrombin immunoassays. Furthermore, we assessed DNA and RNA content of washed PRS and unwashed PRS. RESULTS: Unwashed PRS showed higher total protein concentrations than washed PRS, while the concentration of PDGF-BB, thrombin, DNA, RNA and their mitogenic effect was not significantly different. The bone substitute materials adsorbed protein over time but no significant changes in overall appearance was found. Supernatants collected from unwashed PRS-loaded bone substitute after 1 h induced a potent mitogenic response in periodontal fibroblasts. This pro-mitogenic capacity of the supernatants decreased over the observation period. Supernatants of washed PRS-loaded bone substitutes did not induce a substantial mitogenic effect. Levels of PDGF-BB, thrombin and protein were higher in supernatants of unwashed PRS-loaded bone substitutes than of washed PRS-loaded bone substitutes. CONCLUSION: Bone substitutes loaded with unwashed PRS, but not bone substitutes loaded with washed PRS show continuously declining release kinetics. These data suggest that plasma components in platelet preparations can modify the release kinetics profile.


Asunto(s)
Plaquetas/fisiología , Sustitutos de Huesos/farmacocinética , Minerales/farmacocinética , Animales , Fosfatos de Calcio/farmacocinética , Bovinos , Durapatita/farmacocinética , Fibroblastos/metabolismo , Humanos , Microscopía Electrónica de Rastreo , Factor de Crecimiento Derivado de Plaquetas/farmacocinética
3.
J Nanosci Nanotechnol ; 17(4): 2363-369, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29641166

RESUMEN

Topical administration of growth factors has been suggested as a promising strategy for promoting the healing process and skin regeneration in wound management. However, several restrictions hinder their successful clinical use; specifically, limited percutaneous absorption causes inconsistent efficacy, and various growth factors with specific functionalities are required at different stages of healing. To overcome these shortcomings, previously we have constructed highly skin-permeable analogues of epidermal growth factor (EGF), insulin-like growth factor-I (IGF-I), and platelet-derived growth factor-A (PDGF-A) (LMWP-EGF, LMWP-IGF-I and LMWP-PDGF-A) by genetically conjugating the low-molecular-weight protamine (LMWP) to their N-terminus. In the present study, we determined the optimal concentration ratio of these growth factors by investigating In Vitro cell proliferation and the scratch wound repairing assay. After confirming synergetic effects of growth factors in combinations, we developed a topical delivery system consisting of a nanoemulsion (NE)-dispersed polyvinylpyrrolidone hydrogel loaded with all three growth factors. In Vitro permeability studies were also performed to assess whether the LMWP-conjugated growth factors in the formulation enhanced their skin permeation compared to native growth factors. Combinations of native or LMWP-fused growth factors significantly promoted fibroblast proliferation and scratch wound recovery, and the synergy of LMWP-EGF, LMWP-IGF-I and LMWP-PDGF-A was optimal at a ratio of 100:100:10 by concentration. The growth factor combination-loaded NE appeared to be spherical under cryo-transmission electron microscopy and the average droplet diameter was 127±4.30 nm. The LMWP-conjugated growth factors allowed significantly higher skin permeation than native growth factors from the NE-dispersed hydrogel. Thus, the LMWP-conjugated growth factor combination-loaded NE-dispersed hydrogel is expected to induce more rapid and prolonged wound healing.


Asunto(s)
Sistemas de Liberación de Medicamentos , Factor de Crecimiento Epidérmico , Hidrogel de Polietilenoglicol-Dimetacrilato/química , Nanoestructuras/química , Factor de Crecimiento Derivado de Plaquetas , Administración Tópica , Animales , Emulsiones/química , Factor de Crecimiento Epidérmico/administración & dosificación , Factor de Crecimiento Epidérmico/química , Factor de Crecimiento Epidérmico/farmacocinética , Factor de Crecimiento Epidérmico/farmacología , Hidrogel de Polietilenoglicol-Dimetacrilato/farmacología , Ratones , Modelos Biológicos , Células 3T3 NIH , Factor de Crecimiento Derivado de Plaquetas/administración & dosificación , Factor de Crecimiento Derivado de Plaquetas/química , Factor de Crecimiento Derivado de Plaquetas/farmacocinética , Factor de Crecimiento Derivado de Plaquetas/farmacología , Piel/química , Piel/metabolismo , Absorción Cutánea/efectos de los fármacos , Cicatrización de Heridas/efectos de los fármacos
4.
Acta Orthop Traumatol Turc ; 54(4): 453-460, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32812878

RESUMEN

OBJECTIVE: This study aimed to determine the effects of a novel biodegradable implant releasing platelet-derived growth factor (PDGF) at the fracture site on fracture healing in a rat tibia fracture model. METHODS: In this study, 35 male Sprague-Dawley rats weighing between 300 and 350 g were used. The rats were divided into four groups: Group A (control group without any treatment, n=10), Group B (spacer without PDGF Group, n=10), Group C (spacer with PDGF group, n=10), and Group D (healthy rat Group, n=5). Standardized fractures were created in the right tibias of rats, and then biodegradable implants made of poly-ß-hydroxybutyrate-co-3-hydroxy valerate were implanted at the fracture sites in Groups B and C. In Group C, implants were loaded with 600 ng of PDGF. Animals were sacrificed 30 days after the operation, and fracture healing in each group was assessed radiologically based on the Goldberg score. Furthermore, the anteroposterior (AP) and mediolateral (ML) callus diameters were measured macroscopically, and fracture sites were mechanically tested. RESULTS: In the radiological assessment, Group C showed higher fracture healing rate than Groups A and B (p=0.001), whereas no significant difference was found between group C and Group D (p>0.05). In the macroscopic assessment, while Group C exhibited the thickest AP callus diameter (p=0.02), no significant differences in ML callus diameters existed among the groups (p>0.05). Mechanical testing revealed that Group C had higher torsional strength (p=0.001) and stiffness than Groups A and B (p=0.001) while there was no significant difference between Groups C and D (p>0.05). CONCLUSION: Biodegradable implant releasing PDGF may have positive effects on fracture healing.


Asunto(s)
Implantes Absorbibles , Curación de Fractura/efectos de los fármacos , Factor de Crecimiento Derivado de Plaquetas/farmacocinética , Fracturas de la Tibia/terapia , Animales , Liberación de Fármacos , Masculino , Modelos Anatómicos , Ratas , Ratas Sprague-Dawley , Resultado del Tratamiento
5.
J Cosmet Dermatol ; 19(3): 705-713, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31397540

RESUMEN

BACKGROUND: As skin ages, a functional decrement occurs. To avoid future vulnerability to dermatologic diseases, an optimal cutaneous regeneration is mandatory. Biological therapies based on blood-derived autologous proteins are gaining attention of scientists and dermatologists. OBJECTIVES: A novel 100% autologous topical serum has been developed using plasma rich in growth factors technology. The physicochemical characterization and the biologic potential of the novel formulation have been studied. METHODS: Rheological and mechanical properties and the biological capacity of the formulation were characterized. Human dermal fibroblast culture and 3D organotypic skin explants were used as in vitro and ex vivo cutaneous models, respectively. RESULTS: The autologous topical serum presented an optimal spreadability index and appropriate shear thinning behavior that allowed an easy handling and rapid integration within the cutaneous tissue. The formulation has a high growth factor load with the ability to progressively penetrate into the dermal/epidermal layers of the skin. It is biocompatible and promotes cell proliferation and chemotactic activity. The autologous topical serum promotes the biosynthetic activity of cells by the stimulation of collagen and hyaluronic acid expression. CONCLUSIONS: These findings present an in situ and easy to prepare autologous topical serum based on the patient's own blood with physicochemical and bioactive properties that may be used for skin regeneration purposes.


Asunto(s)
Factores Biológicos/administración & dosificación , Transfusión de Sangre Autóloga/métodos , Factor de Crecimiento Derivado de Plaquetas/administración & dosificación , Regeneración/efectos de los fármacos , Envejecimiento de la Piel/efectos de los fármacos , Administración Cutánea , Factores Biológicos/farmacocinética , Células Cultivadas , Colágeno/metabolismo , Evaluación Preclínica de Medicamentos , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Ácido Hialurónico/metabolismo , Factor de Crecimiento Derivado de Plaquetas/farmacocinética , Cultivo Primario de Células , Piel/citología , Piel/efectos de los fármacos , Absorción Cutánea , Técnicas de Cultivo de Tejidos
6.
J Hand Surg Am ; 33(9): 1548-57, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18984337

RESUMEN

PURPOSE: Surgically repaired intrasynovial tendons are at greatest risk of failure in the first 3 weeks after surgery. Attempts to improve the strength of repair by modifying rehabilitation parameters have not always been successful. Manipulation of the biological environment of the sutured tendon holds great promise for accelerating the repair process. The goals of this study were to examine (1) the range of conditions (eg, dosage, delivery system formulation, presence of cells) over which delivery of platelet-derived growth factor-BB (PDGF-BB) can be sustained from fibrin matrices using a heparin-binding delivery system (HBDS) and (2) the biological activity of the PDGF-BB released from this system on canine tendon fibroblasts in vitro. METHODS: We examined in vitro release kinetics from cellular and acellular fibrin matrices using enzyme-linked immunosorbent assays. We examined the biologic activity of the PDGF-BB in vitro by measuring cell proliferation (ie, total DNA) and collagen synthesis (ie, proline incorporation). RESULTS: The acellular release kinetics of PDGF-BB was modulated by varying the ratio of PDGF-BB to heparin (PDGF-binding sites) or the dose of PDGF-BB in the presence of the delivery system. In the presence of canine tendon fibroblasts, the delivery system prolonged the duration of PDGF-BB release from fibrin matrices, thus demonstrating that cells are able to liberate PDGF-BB retained by the HBDS. Sustained delivery of PDGF-BB promoted increased cell proliferation at doses of 0.125 microg/mL and 1.25 microg/mL compared to fibrin without delivery system. Collagen synthesis was enhanced by PDGF-BB at doses of 0.125 microg/mL and 1.25 microg/mL; however, there was an enhancement over fibrin without the delivery system only at the lower dose. CONCLUSIONS: These results demonstrate that the PDGF-BB released from fibrin matrices containing an HBDS is biologically active and can modulate both cell proliferation and extracellular matrix synthesis, both of which are key factors in the process of tendon repair.


Asunto(s)
Inductores de la Angiogénesis/farmacocinética , Factor de Crecimiento Derivado de Plaquetas/farmacocinética , Traumatismos de los Tendones/terapia , Cicatrización de Heridas/efectos de los fármacos , Animales , Becaplermina , Proliferación Celular/efectos de los fármacos , Colágeno/biosíntesis , Perros , Relación Dosis-Respuesta a Droga , Sistemas de Liberación de Medicamentos/métodos , Ensayo de Inmunoadsorción Enzimática , Proteínas de la Matriz Extracelular/metabolismo , Fibrina/metabolismo , Fibroblastos/efectos de los fármacos , Técnicas In Vitro , Modelos Animales , Proteínas Proto-Oncogénicas c-sis
7.
Acta Biomater ; 76: 126-134, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29908335

RESUMEN

The aim of this study was to examine the potential of platelet-derived growth factor (PDGF)-coated decellularized meniscus scaffold in mediating integrative healing of meniscus tears by inducing endogenous cell migration. Fresh bovine meniscus was chemically decellularized and covalently conjugated with heparin and PDGF-BB. In vitro PDGF release kinetics was measured. The scaffold was transplanted into experimental tears in avascular bovine meniscus explants and cultured for 2 and 4 weeks. The number migrating and proliferating cells at the borderline between the scaffold and injured explant and PDGF receptor-ß (PDGFRß) expressing cells were counted. The alignment of the newly produced ECM and collagen was analyzed by Safranin-O, picrosirius red staining, and differential interference contrast (DIC). Tensile testing of the explants was performed after culture for 2 and 4 weeks. Heparin conjugated scaffold showed immobilization of high levels of PDGF-BB, with sustained release over 2 weeks. Insertion of the PDGF-BB treated scaffold in defects in avascular meniscus led to increased PDGFRß expression, cell migration and proliferation into the defect zone. Safranin-O, picrosirius red staining and DIC showed tissue integration between the scaffold and injured explants. Tensile properties of injured explants treated with PDGF-BB coated scaffold were significantly higher than in the scaffold without PDGF. In conclusion, PDGF-BB-coated scaffold increased PDGFRß expression and promoted migration of endogenous meniscus cells to the defect area. New matrix was formed that bridged the space between the native meniscus and the scaffold and this was associated with improved biomechanical properties. The PDGF-BB-coated scaffold will be promising for clinical translation to healing of meniscus tears. STATEMENT OF SIGNIFICANCE: Meniscus tears are the most common injury of the knee joint. The most prevalent forms that occur in the inner third typically do not spontaneously heal and represent a major risk factor for the development of knee osteoarthritis. The goal of this project was to develop an approach that is readily applicable for clinical use. We selected a natural and readily available decellularized meniscus scaffold and conjugated it with PDGF, which we had previously found to have strong chemotactic activity for chondrocytes and progenitor cells. The present results show that insertion of the PDGF-conjugated scaffold in defects in avascular meniscus led to endogenous cell migration and proliferation into the defect zone with tissue integration between the scaffold and injured explants and improved tensile properties. This PDGF-conjugated scaffold will be promising for a translational approach to healing of meniscus tears.


Asunto(s)
Materiales Biocompatibles Revestidos , Traumatismos de la Rodilla , Menisco , Factor de Crecimiento Derivado de Plaquetas , Ingeniería de Tejidos , Andamios del Tejido/química , Animales , Bovinos , Materiales Biocompatibles Revestidos/química , Materiales Biocompatibles Revestidos/farmacocinética , Materiales Biocompatibles Revestidos/farmacología , Humanos , Traumatismos de la Rodilla/metabolismo , Traumatismos de la Rodilla/terapia , Factor de Crecimiento Derivado de Plaquetas/química , Factor de Crecimiento Derivado de Plaquetas/farmacocinética , Factor de Crecimiento Derivado de Plaquetas/farmacología
8.
Biomaterials ; 28(8): 1515-22, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17169421

RESUMEN

In recent years, functional biomaterial research has been directed towards the development of improved scaffolds and new drug delivery systems. The objective of this study was to develop growth-factor gene releasing coral composites as a regenerative material for periodontal regeneration. In this study, porous chitosan/coral composites combined with plasmid encoding platelet-derived growth factor B (PDGFB) gene were prepared through a freeze-drying process. These scaffolds were evaluated in vitro by analysis of microscopic structure and cytocompatibility. The expression of PDGFB and type-I collagen were detected with RT-PCR after human periodontal ligament cells (HPLCs) were seeded in this scaffold. Then these scaffolds were implanted subcutaneously into athymic mice. Results indicated that HPLCs showed much better proliferation properties on the gene-activated scaffolds than on the pure coral scaffolds, and the expression of PDGFB and type-I collagen up-regulated in gene-activated scaffold. After implanted in vivo, HPLCs not only proliferate but also increased the expression of PDGFB. This study demonstrated the potential of coral scaffold combined PDGFB gene as a good substrate candidate in periodontal tissue regeneration.


Asunto(s)
Antozoos , Materiales Biocompatibles , Quitosano , Periodoncio , Factor de Crecimiento Derivado de Plaquetas/farmacocinética , Ingeniería de Tejidos , Animales , Regeneración Tisular Dirigida , Humanos , Ratones
9.
Int J Nanomedicine ; 12: 5525-5535, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28831251

RESUMEN

This study aimed to develop a functionally graded membrane (FGM) to prevent infection and promote tissue regeneration. Poly(l-lactide-co-d,l-lactide) encapsulating platelet-derived growth factor (PDLLA-PDGF) or metronidazole (PDLLA-MTZ) was electrospun to form a nanofibrous layer on the inner or outer surface of a clinically available collagen membrane, respectively. The membrane was characterized for the morphology, molecule release profile, in vitro and in vivo biocompatibility, and preclinical efficiency for alveolar ridge regeneration. The PDLLA-MTZ and PDLLA-PDGF nanofibers were 800-900 nm in diameter, and the thicknesses of the functional layers were 20-30 µm, with sustained molecule release over 28 days. All of the membranes tested were compatible with cell survival in vitro and showed good tissue integration with minimal fibrous capsule formation or inflammation. Cell proliferation was especially prominent on the PDLLA-PDGF layer in vivo. On the alveolar ridge, all FGMs reduced wound dehiscence compared with the control collagen membrane, and the FGM with PDLLA-PDGF promoted osteogenesis significantly. In conclusion, the FGMs with PDLLA-PDGF and PDLLA-MTZ showed high biocompatibility and facilitated wound healing compared with conventional membrane, and the FGM with PDLLA-PDGF enhanced alveolar ridge regeneration in vivo. The design represents a beneficial modification, which may be easily adapted for future clinical use.


Asunto(s)
Proceso Alveolar/fisiología , Metronidazol/farmacología , Nanofibras/química , Factor de Crecimiento Derivado de Plaquetas/farmacología , Regeneración/fisiología , Proceso Alveolar/citología , Proceso Alveolar/efectos de los fármacos , Animales , Proliferación Celular , Colágeno/química , Liberación de Fármacos , Masculino , Ensayo de Materiales , Metronidazol/química , Metronidazol/farmacocinética , Ratones Endogámicos C57BL , Nanofibras/administración & dosificación , Osteogénesis/efectos de los fármacos , Factor de Crecimiento Derivado de Plaquetas/química , Factor de Crecimiento Derivado de Plaquetas/farmacocinética , Poliésteres/química , Ratas Sprague-Dawley , Regeneración/efectos de los fármacos , Andamios del Tejido , Cicatrización de Heridas
10.
J Control Release ; 114(2): 223-9, 2006 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-16859798

RESUMEN

The therapeutic benefit of local administration of growth factors using controlled release systems for bone regeneration is under study. In the present work, a PDGF release profile from chitosan granules administered into bone defect produced in the femur of Wistar rats was obtained using 125I-PDGF as tracer and a probe-type gamma counter with a suitable collimator for detection. The measurement method was validated by the radioactivity values obtained from the isolated bones using a well-type gamma counter. Both the invasive and the non-invasive measurement methods were linear in the analyzed range, with coefficients of variation around 3% and 15%, respectively. A good correlation between the two methods was found for the 125I-PDGF release profile from the chitosan granules. These results confirm that a reliable release profile can be obtained for a drug incorporated into delivery systems for local bone therapy using this non-invasive measurement method.


Asunto(s)
Huesos/metabolismo , Factor de Crecimiento Derivado de Plaquetas/administración & dosificación , Factor de Crecimiento Derivado de Plaquetas/análisis , Animales , Enfermedades Óseas/tratamiento farmacológico , Enfermedades Óseas/patología , Huesos/química , Quitosano , Sistemas de Liberación de Medicamentos , Excipientes , Fémur/patología , Radioisótopos de Yodo , Masculino , Tamaño de la Partícula , Factor de Crecimiento Derivado de Plaquetas/farmacocinética , Polvos , Ratas , Ratas Wistar , Reproducibilidad de los Resultados
11.
Acta Biomater ; 29: 11-20, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26497625

RESUMEN

Sustained, local, low dose growth factor stimulus of target tissues/cells is believed to be of imminent importance in tissue regeneration and engineering. Recently, a technology was developed to bind growth factors to a fibrin matrix using the transglutaminase (TG) activity of factor XIIIa, thus allowing prolonged release through enzymatic cleavage. In this study we aimed to determine whether TG-PDGF.AB in fibrin could improve tissue regeneration in a standard ischemic flap model. In vitro determination of binding and release kinetics of TG-PDGF.AB allowed proof of concept of the developed binding technology. A single spray application of TG-PDGF.AB in fibrin matrix at a concentration of 10 and 100ng/ml significantly reduced ischemia-induced flap tissue necrosis in vivo on day 7 after ischemic impact compared to controls. TG-PDGF.AB at a concentration of 100ng/ml fibrin induced distinct angiogenesis as reflected by significantly improved tissue perfusion assessed by laser Doppler imaging as well as enhanced von Willebrand factor (vWF) protein expression determined by immunohistochemical means. In addition, significantly more mature microvessels were observed with 100ng/ml TG-PDGF.AB in fibrin compared to control and vehicle groups as evidenced by an improved smooth muscle actin (sma)/vWF protein ratio. In conclusion, PDGF.AB in a conjugated fibrin matrix effectively reduced ischemia-induced tissue necrosis, increased tissue perfusion and induced the growth of a mature and functional neovasculature. The sealing properties of the fibrin matrix in conjunction with the prolonged growth factor stimulus enabled by the TG-hook binding technology may present an innovative and suitable tool in tissue regeneration. STATEMENT OF SIGNIFICANCE: In our experimental study we elucidated recombinant platelet derived growth factor (PDGF) as a potential candidate in inducing angiogenesis. To avoid preterm growth factor degradation in vivo PDGF.AB was covalently linked to a fibrin scaffold using a bi-domain functionalized peptide (FXIII substrate site and plasmin cleavage site). This allowed PDGF binding to fibrin during spray application to the donor site and subsequent prolonged release via endogenous plasmin. This resulted in a mature vascular network thus enhancing tissue perfusion and consequently improved clinical outcome. With our present work we could certainly provide researchers and clinicians with an innovative versatile and reproducible technology not only to induce functional vascularity but also to improve attempts in tissue engineering in general by e.g. using different growth factors. Hence, we believe that this approach studied in the present work may provide a valuable input in an effort to drive the aim forward bringing experimental work in tissue engineering to clinic by using a clinically well characterized and used fibrin scaffold in combination with a human recombinant growth factor (fibrin scaffold linked with the specific binding technology).


Asunto(s)
Fibrina , Isquemia/tratamiento farmacológico , Neovascularización Fisiológica/efectos de los fármacos , Factor de Crecimiento Derivado de Plaquetas , Animales , Preparaciones de Acción Retardada/farmacocinética , Preparaciones de Acción Retardada/farmacología , Fibrina/farmacocinética , Fibrina/farmacología , Humanos , Factor de Crecimiento Derivado de Plaquetas/farmacocinética , Factor de Crecimiento Derivado de Plaquetas/farmacología , Ratas , Ratas Sprague-Dawley
12.
Nanomedicine (Lond) ; 11(24): 3237-3259, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27854552

RESUMEN

AIM: Cardiac tissue engineering aims to develop engineered constructs for myocardial infarction repair, where a challenge is the control of growth factor (GF) sequential release. Herein, bilayer polymeric nanoparticles composed of a GF-encapsulating core surrounded by rate-regulating shell were developed for sequential GF release. MATERIALS & METHODS: Single and bilayer polymeric nanoparticles were fabricated, characterized and biologically assessed. A novel 'Geno-Neural model' was developed and validated for rate-programming of the nanoparticles. RESULTS: The bilayer nanoparticles featured low burst effect and time-delayed release, and allowed for sequential release of PDGF following co-release of VEGFand bFGF, which promoted angiogenesis. CONCLUSION: The nanoparticulate delivery system, along with the Geno-Neural model, offers great potential for spatiotemporal control of GF release for cardiovascular regenerative medicine.


Asunto(s)
Sistemas de Liberación de Medicamentos/instrumentación , Factor 2 de Crecimiento de Fibroblastos/administración & dosificación , Nanopartículas/administración & dosificación , Factor de Crecimiento Derivado de Plaquetas/administración & dosificación , Polímeros/química , Ingeniería de Tejidos/métodos , Factor A de Crecimiento Endotelial Vascular/administración & dosificación , Algoritmos , Animales , Aorta/efectos de los fármacos , Aorta/fisiología , Sistemas de Liberación de Medicamentos/métodos , Liberación de Fármacos , Factor 2 de Crecimiento de Fibroblastos/farmacocinética , Humanos , Nanopartículas/química , Neovascularización Fisiológica/efectos de los fármacos , Redes Neurales de la Computación , Factor de Crecimiento Derivado de Plaquetas/farmacocinética , Ratas , Distribución Tisular , Factor A de Crecimiento Endotelial Vascular/farmacocinética
13.
J Tissue Eng Regen Med ; 10(5): E275-85, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-23723146

RESUMEN

Controlled delivery of growth factors from biodegradable biomatrices could accelerate and improve impaired wound healing. The study aim was to determine whether platelet-derived growth factor AB (PDGF.AB) with a transglutaminase (TG) crosslinking substrate site released from a fibrin biomatrix improves wound healing in severe thermal injury. The binding and release kinetics of TG-PDGF.AB were determined in vitro. Third-degree contact burns (dorsum of Yorkshire pigs) underwent epifascial necrosectomy 24 h post-burn. Wound sites were covered with autologous meshed (3:1) split-thickness skin autografts and either secured with staples or attached with sprayed fibrin sealant (FS; n = 8/group). TG-PDGF.AB binds to the fibrin biomatrix using the TG activity of factor XIIIa, and is subsequently released through enzymatic cleavage. Three doses of TG-PDGF.AB in FS (100 ng, 1 µg and 11 µg/ml FS) were tested. TG-PDGF.AB was bound to the fibrin biomatrix as evidenced by western blot analysis and subsequently released by enzymatic cleavage. A significantly accelerated and improved wound healing was achieved using sprayed FS containing TG-PDGF.AB compared to staples alone. Low concentrations (100 ng-1 µg TG-PDGF.AB/ml final FS clot) demonstrated to be sufficient to attain a nearly complete closure of mesh interstices 14 days after grafting. TG-PDGF.AB incorporated in FS via a specific binding technology was shown to be effective in grafted third-degree burn wounds. The adhesive properties of the fibrin matrix in conjunction with the prolonged growth factor stimulus enabled by this binding technology could be favourable in many pathological situations associated with wound-healing disturbances. Copyright © 2013 John Wiley & Sons, Ltd.


Asunto(s)
Quemaduras/tratamiento farmacológico , Matriz Extracelular/química , Fibrina , Factor de Crecimiento Derivado de Plaquetas , Proteínas Proto-Oncogénicas c-sis , Cicatrización de Heridas/efectos de los fármacos , Animales , Preparaciones de Acción Retardada/química , Preparaciones de Acción Retardada/farmacocinética , Preparaciones de Acción Retardada/farmacología , Fibrina/química , Fibrina/farmacocinética , Fibrina/farmacología , Factor de Crecimiento Derivado de Plaquetas/química , Factor de Crecimiento Derivado de Plaquetas/farmacocinética , Factor de Crecimiento Derivado de Plaquetas/farmacología , Proteínas Proto-Oncogénicas c-sis/química , Proteínas Proto-Oncogénicas c-sis/farmacocinética , Proteínas Proto-Oncogénicas c-sis/farmacología , Porcinos
14.
J Neurosci ; 19(22): 10014-25, 1999 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-10559409

RESUMEN

Platelet-derived growth factor (PDGF) is a critical regulator of cell proliferation. Because ethanol inhibits cell proliferation in vivo and in vitro, we hypothesize that ethanol-induced inhibition results from differential interference with signal transduction pathways activated by PDGF. Cultured cortical astrocytes were used to examine the effects of ethanol on PDGF-mediated signal transduction, on the expression of two PDGF monomers (A- and B-chains), and on the expression of two PDGF receptor subunits (PDGFalphar and PDGFbetar). PDGF-B chain homodimer (PDGF-BB), and to a lesser extent PDGF-A chain homodimer (PDGF-AA), stimulated the proliferation of astrocytes raised in a serum-free medium. Ethanol attenuated these actions in a concentration-dependent manner. Ethanol inhibited both PDGF-AA- and PDGF-BB-mediated phosphorylation of PDGFalphar, but it had little effect on PDGFbetar autophosphorylation. Likewise, ethanol abolished the association of PDGFalphar to Ras GTPase-activating protein (Ras-GAP), but it did not affect the binding of Ras-GAP to PDGFbetar. PDGF stimulated the activities of mitogen-activated protein kinase (MAPK) in protein kinase C (PKC) independent and dependent manners. Ethanol inhibited the PKC-independent, acute activation of MAPK; however, it stimulated the PKC-dependent, sustained activation of MAPK. The expression of neither ligand was altered by exposure to ethanol for 3 d. Moreover, such treatment specifically upregulated PDGFalphar expression in a concentration-dependent manner. It did not, however, affect the binding affinity of either receptor. Thus, the signal transduction pathways initiated by PDGF-AA and PDGF-BB were differentially affected by ethanol. This differential vulnerability resulted from the preferential effects of ethanol on PDGFalphar autophosphorylation. Hence, ethanol-induced alterations are transduced through specific receptors of mitogenic growth factors.


Asunto(s)
Astrocitos/citología , Astrocitos/fisiología , Etanol/farmacología , Factor de Crecimiento Derivado de Plaquetas/farmacología , Animales , Animales Recién Nacidos , Astrocitos/efectos de los fármacos , Becaplermina , División Celular/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Modelos Neurológicos , Fosforilación , Factor de Crecimiento Derivado de Plaquetas/farmacocinética , Proteína Quinasa C/metabolismo , Proteínas Proto-Oncogénicas c-sis , Ratas , Receptores del Factor de Crecimiento Derivado de Plaquetas/genética , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
15.
J Periodontol ; 76(11): 1833-41, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16274301

RESUMEN

BACKGROUND: The use of alloplastic matrices that mimic the mineral phase of bone has become a viable alternative to current mainstream therapies in dentistry such as allografts and autogenous grafts. Because alloplastic bone substitutes generally have relatively poor osteogenic properties, analyzing their potential as vehicles to deliver growth factors is an important step in assessing methods to enhance their clinical efficacy. The aim of these studies was to treat beta-tricalcium phosphate (beta-TCP) and calcium sulfate (CaSO(4)) with platelet-derived growth factor (PDGF)-BB to enhance the osteogenic capabilities of these materials. METHODS: In the beta-TCP studies, PDGF-BB adsorption and release were accomplished using (125)I radiolabeled growth factor and non-radioactive human recombinant PDGF at a ratio of 1:300 M. For the adsorption studies, the radiolabeled PDGF-BB/ non-radioactive PDGF solutions with resultant PDGF concentrations of 10(7) and 10(8) M were incubated with beta-TCP from 1 to 120 minutes, and the amount of adsorbed (125)I-PDGF-BB was measured using a gamma counter. Similar adsorption studies were conducted with a 30-minute incubation of beta-TCP with various PDGF concentrations. In vitro release studies were conducted with beta-TCP to which radiolabeled PDGF had been adsorbed as above. Release studies were also conducted with CaSO(4) that was hydrated with the radioactive PDGF solution described above for the TCP studies. In vivo PDGF-BB release from beta-TCP and CaSO(4) was evaluated in a mouse model, where the radioactive PDGF/non-radioactive PDGF-BB treated beta-TCP or CaSO(4) sample was inserted subcutaneously and later removed for radioactive measurement. Proliferation of human osteoblastic cells in the presence of PDGF- treated beta-TCP or CaSO(4) was assessed by (3)H thymidine incorporation. RESULTS: The absorption studies revealed that PDGF-BB was absorbed in a concentration and time-dependent manner to beta-TCP. In the in vitro release studies, approximately 45% of the adsorbed PDGF-BB was released after 10 days. In vivo release from both materials occurred faster than in vitro release. Osteoblastic cells incubated with PDGF-BB-treated matrices showed significantly (P <0.05, ANOVA) greater proliferation than with control matrices alone. CONCLUSION: These experiments demonstrate the feasibility of using PDGF-BB in combination with alloplastic materials such as beta-TCP or CaSO(4) to serve as more effective bone graft materials with enhanced osteogenic properties.


Asunto(s)
Sustitutos de Huesos/química , Fosfatos de Calcio/química , Sulfato de Calcio/química , Factor de Crecimiento Derivado de Plaquetas/química , Adolescente , Adsorción , Adulto , Anciano , Animales , Becaplermina , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Difusión , Estudios de Factibilidad , Femenino , Humanos , Masculino , Ratones , Ratones Desnudos , Persona de Mediana Edad , Modelos Animales , Osteoblastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Factor de Crecimiento Derivado de Plaquetas/farmacocinética , Factor de Crecimiento Derivado de Plaquetas/farmacología , Proteínas Proto-Oncogénicas c-sis , Piel/metabolismo , Factores de Tiempo
16.
AJNR Am J Neuroradiol ; 24(6): 1214-21, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12812957

RESUMEN

BACKGROUND AND PURPOSE: Coil embolization is safe and effective but may be followed by aneurysm recurrence. Our purpose was to explore the use of alginate as a new embolic agent that could deliver growth factors and improve results of endovascular treatment of aneurysms. METHODS: We first assessed the potential of alginate as a vector for growth factor delivery by using in vitro binding and elution studies. Lateral wall (n = 68) and bifurcation (n = 4) aneurysms were then constructed in six pigs and 36 dogs. We explored iodine-125 transforming growth factor-beta(1) in vivo alginate delivery in 16 canine aneurysms. We next assessed the effects of adding alginate to gelatin sponges on angiographic and pathologic results at 3 weeks (n = 4 each) in an established model used for the study of recanalization and recurrence. We then explored techniques to control endovascular alginate delivery without protection (n = 4), with the protection of a balloon (n = 4), and with the protection of a single coil (n = 12) at the aneurysm neck in 12 porcine aneurysms, four canine lateral wall aneurysms, and four canine bifurcation aneurysms. The stability of cross-linked alginate was studied after intraoperative injections in eight aneurysms. Finally, to determine the value of the material with or without growth factor in promoting aneurysm healing, we compared angiographic results and neointima formation 3 weeks after intraoperative embolization of canine lateral wall aneurysms with alginate blocks with or without platelet-derived growth factor-BB or transforming growth factor-beta(1) (n = 5 each). RESULTS: Growth factors rapidly eluted from alginate in vitro and in vivo. Alginate coating of sponges led to improved angiographic results and thick neointima formation. Intraoperative alginate block embolization did not lead to recurrence, and growth factors delivered with alginate did not show added benefits. Endovascular alginate embolization was complicated by carotid emboli, and the polymer was unstable once injected, causing delayed neurologic deficits. CONCLUSION: Growth factor delivery can be performed with alginate, but formulation changes and improved endovascular control are necessary before contemplating its use in intracranial aneurysms.


Asunto(s)
Alginatos , Oclusión con Balón/métodos , Materiales Biocompatibles , Portadores de Fármacos , Embolización Terapéutica/métodos , Aneurisma Intracraneal/terapia , Factor de Crecimiento Derivado de Plaquetas/administración & dosificación , Factor de Crecimiento Transformador beta/administración & dosificación , Animales , Becaplermina , Disponibilidad Biológica , Angiografía Cerebral , Terapia Combinada , Modelos Animales de Enfermedad , Perros , Ácido Glucurónico , Ácidos Hexurónicos , Inyecciones , Aneurisma Intracraneal/diagnóstico por imagen , Aneurisma Intracraneal/patología , Factor de Crecimiento Derivado de Plaquetas/farmacocinética , Proteínas Proto-Oncogénicas c-sis , Porcinos , Factor de Crecimiento Transformador beta/farmacocinética , Factor de Crecimiento Transformador beta1 , Túnica Íntima/diagnóstico por imagen , Túnica Íntima/patología , Cicatrización de Heridas/efectos de los fármacos
17.
Life Sci ; 62(21): 1911-8, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-9619839

RESUMEN

The kinetics of exogenously given 125I-platelet-derived growth factor-AB (PDGF-AB) was studied in mice. 125I-PDGF-AB was injected either intraperitoneally, intramuscularly or subcutaneously and the resulting concentrations of 125I-radioactivity monitored in the blood at different times. The serum levels of 125I-radioactivity rose to a maximum 2-4 hours after injection, before decreasing. Precipitation of serum with trichloroacetic acid demonstrated that 50 per cent or more of the 125I remained in macromolecular form. Further, gel chromatography studies showed that the molecular size of the labelled material in serum, three hours after injection, was the same as that of the original 125I-PDGF-AB. In addition, a low-molecular weight fraction was observed, indicating the presence of degradation products. The largest proportion of degraded material was obtained after subcutaneous administration. The absence of partially degraded 125I-labelled fragments, e.g. 125I oligopeptides, indicates complete rather than limited degradation and suggests that the 125I-PDGF-AB had been processed by cellular uptake. It is concluded that extra-vascularly given PDGF-AB in mice is taken up into the blood in intact macromolecular form. This finding suggests that it is possible to administer PDGF extravascularly to obtain a prolonged increase in the concentration of intact PDGF in the blood.


Asunto(s)
Radioisótopos de Yodo , Factor de Crecimiento Derivado de Plaquetas/farmacocinética , Animales , Precipitación Química , Cromatografía en Gel , Inyecciones Intramusculares , Inyecciones Intraperitoneales , Inyecciones Subcutáneas , Cinética , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Factor de Crecimiento Derivado de Plaquetas/administración & dosificación , Ácido Tricloroacético
18.
Am J Surg ; 176(2A Suppl): 68S-73S, 1998 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9777975

RESUMEN

This overview of the safety of becaplermin gel is based on six well-controlled clinical studies that included patients with lower extremity diabetic neuropathic ulcers. Patients receiving becaplermin gel (n = 538), placebo gel (n = 278), or good ulcer care alone (n = 190) had a similar incidence of ulcer-related adverse events, such as infection, cellulitis, or osteomyelitis. Erythematous rash occurred in 2% of patients with suspected wound infections treated with becaplermin gel and 1% of patients treated with placebo gel. No rashes were observed in patients treated with good ulcer care alone. The incidence of cardiovascular, respiratory, musculoskeletal, and central and peripheral nervous system disorders were similar across all treatment groups. Mortality rates were also similar across all treatment groups. Patients treated with becaplermin gel did not develop neutralizing antibodies against becaplermin. Therefore, becaplermin gel appears to be a safe therapy for the treatment of lower extremity diabetic ulcers.


Asunto(s)
Anticoagulantes/efectos adversos , Pie Diabético/tratamiento farmacológico , Factor de Crecimiento Derivado de Plaquetas/efectos adversos , Absorción , Adulto , Anciano , Anticoagulantes/farmacocinética , Anticoagulantes/uso terapéutico , Becaplermina , Método Doble Ciego , Femenino , Geles , Humanos , Masculino , Persona de Mediana Edad , Factor de Crecimiento Derivado de Plaquetas/farmacocinética , Factor de Crecimiento Derivado de Plaquetas/uso terapéutico , Proteínas Proto-Oncogénicas c-sis , Proteínas Recombinantes/efectos adversos , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/uso terapéutico , Enfermedades de la Piel/inducido químicamente
19.
J Periodontol ; 67(10): 981-5, 1996 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-8910837

RESUMEN

The purpose of this study was to investigate if the treatment of porous polysulfone (PPSF) with various concentrations of platelet-derived growth factor-BB (PDGF-BB) would stimulate the proliferation of the adherent human periodontal ligament fibroblasts (HPDLF) in culture. Sterilized PPSF cylinders were immersed in an Eagle's minimum essential medium supplemented with 0.5% fetal bovine serum and 1% penicillin/streptomycin containing either 0, 10, 20, or 50 ng/ml of PDGF-BB for 24 hours. After 24 hours, the PPSF cylinders were removed and allowed to dry then placed in culture plates for each time point. Pooled HPDLF (8 x 10(4)) and 3H-thymidine in medium were pipetted into each well to cover the treated and control PPSF cylinders and plates were then incubated. At 1, 4, and 10 days the PPSF cylinders were removed and macromolecular precipitation was performed. Incorporation of 3H-thymidine was measured and a 2-way ANOVA with repeated measures was performed. In addition, determination of binding and release was performed using I125-PDGF-BB treated PPSF at 0, 2, 12, and 24 hours, and at 4 and 10 days. Results showed that the effects on HPDLF were significant for dose (P = 0.0012; F = 5.74) and time (P = 0.0001; F = 40.83). At 4 days, the percent increases above the control for the doses 10, 20, and 50 ng/ml were 192%, 310%, and 162% respectively. In conclusion, our findings suggest that treating PPSF with PDGF-BB results in a significant increase in the proliferation of HPDLF cells adherent to PPSF.


Asunto(s)
Anticoagulantes/farmacología , Materiales Biocompatibles/química , Fibroblastos/efectos de los fármacos , Membranas Artificiales , Ligamento Periodontal/efectos de los fármacos , Factor de Crecimiento Derivado de Plaquetas/farmacología , Polímeros/química , Sulfonas/química , Análisis de Varianza , Animales , Anticoagulantes/administración & dosificación , Anticoagulantes/farmacocinética , Becaplermina , Bovinos , Adhesión Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Células Cultivadas , Medios de Cultivo , Preparaciones de Acción Retardada , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Radioisótopos de Yodo , Ligamento Periodontal/citología , Ligamento Periodontal/metabolismo , Factor de Crecimiento Derivado de Plaquetas/administración & dosificación , Factor de Crecimiento Derivado de Plaquetas/farmacocinética , Proteínas Proto-Oncogénicas c-sis , Radiofármacos , Proteínas Recombinantes , Propiedades de Superficie , Timidina/metabolismo , Tritio
20.
J Periodontol ; 62(7): 458-67, 1991 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-1920013

RESUMEN

Polypeptide growth factors are a class of potent natural biologic mediators which regulate many of the activities of wound healing including cell proliferation, migration, and metabolism. Platelet-derived growth factor (PDGF) and insulin-like growth factor-I (IGF-I) have been shown to regulate DNA and protein synthesis in bone cells in vitro and to interact synergistically to enhance soft tissue wound healing in vivo. We have hypothesized that the combination of PDGF and IGF-I may, therefore, enhance regeneration of both the soft and hard tissue components of the periodontium. To test this hypothesis we performed conventional periodontal surgery on all 4 quadrants of the mouth of 13 beagle dogs with naturally occurring periodontal disease. Following flap reflection, degranulation, and root planing, all premolar teeth in 2 quadrants of each dog received a combination of 3 micrograms of recombinant PDGF-B and IGF-I in a methylcellulose gel, while the premolar teeth in the contralateral quadrants received the gel alone. Teeth in 4 additional animals also received 125I-PDGF or 125I-IGF-I in the treated sites. The clearance rate of the 125I-labeled protein, changes in local bone metabolism, and amount of new bone and cementum with inserting collagen fibers were measured. The clearance studies revealed that the half-life of the factors at the site of application was 3.0 hours for IGF-I and to 4.2 hours for PDGF-B. Greater than 96% of the radio-labeled proteins was cleared by 96 hours and no radioactivity was detected 2 weeks after application. There was a significant (P less than 0.01) 2-fold increase in uptake of the bone-seeking radiopharmaceutical Technetium 99-MDP at 2 and 4 weeks in growth factor treated sites compared to controls, indicating that there was increased metabolic activity within the bone at these sites. Computer-aided histologic analyses of biopsies obtained at 2 and 5 weeks post-operatively revealed a significant (P less than 0.01), 5 to 10 fold increase in new bone and cementum in PDGF-B/IGF-I treated sites at both time points compared to controls receiving the placebo gel. The height and total area of new bone continued to increase from 2 to 5 weeks. The new bone underwent a normal maturation process as judged by histologic appearance. A physiologic periodontal ligament space was also formed between the new bone and new cementum. There was no increase in ankylosis in the treated sites.(ABSTRACT TRUNCATED AT 400 WORDS)


Asunto(s)
Factor I del Crecimiento Similar a la Insulina/uso terapéutico , Enfermedades Periodontales/tratamiento farmacológico , Factor de Crecimiento Derivado de Plaquetas/uso terapéutico , Pérdida de Hueso Alveolar/tratamiento farmacológico , Pérdida de Hueso Alveolar/patología , Pérdida de Hueso Alveolar/cirugía , Proceso Alveolar/metabolismo , Proceso Alveolar/patología , Animales , Tejido Conectivo/patología , Perros , Combinación de Medicamentos , Femenino , Gingivitis/tratamiento farmacológico , Gingivitis/patología , Gingivitis/cirugía , Factor I del Crecimiento Similar a la Insulina/administración & dosificación , Factor I del Crecimiento Similar a la Insulina/farmacocinética , Radioisótopos de Yodo , Tasa de Depuración Metabólica , Osteoblastos/patología , Osteocitos/patología , Enfermedades Periodontales/patología , Enfermedades Periodontales/cirugía , Ligamento Periodontal/patología , Factor de Crecimiento Derivado de Plaquetas/administración & dosificación , Factor de Crecimiento Derivado de Plaquetas/farmacocinética , Proteínas Recombinantes , Regeneración/efectos de los fármacos , Colgajos Quirúrgicos , Medronato de Tecnecio Tc 99m , Factores de Tiempo , Cicatrización de Heridas/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA