Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Intervalo de año de publicación
1.
J Control Release ; 370: 277-286, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38679161

RESUMEN

Addressing bone defects represents a significant challenge to public health. Localized delivery of growth factor has emerged as promising approach for bone regeneration. However, the clinical application of Platelet-Derived Growth Factor (PDGF) is hindered by its high cost and short half-life. In this work, we introduce the application of PDGF-mimicking peptide (PMP1) hydrogels for calvarial defect restoration, showcasing their remarkable effectiveness. Through osteogenic differentiation assays and q-PCR analyses, we demonstrate PMP1's substantial capacity to enhance osteogenic differentiation of bone marrow mesenchymal stem cell (BMSC), leading to increased expression of crucial osteogenic genes. Further molecular mechanistic investigations reveal PMP1's activation of the PI3K-AKT-mTOR signaling pathway, a key element of its osteogenic effect. In vivo experiments utilizing a rat calvaria critical-sized defect model underscore the hydrogels' exceptional ability to accelerate new bone formation, thereby significantly advancing the restoration of calvaria defects. This research provides a promising bioactive material for bone tissue regeneration.


Asunto(s)
Becaplermina , Regeneración Ósea , Diferenciación Celular , Hidrogeles , Células Madre Mesenquimatosas , Osteogénesis , Ratas Sprague-Dawley , Cráneo , Animales , Hidrogeles/química , Cráneo/efectos de los fármacos , Cráneo/lesiones , Osteogénesis/efectos de los fármacos , Becaplermina/administración & dosificación , Regeneración Ósea/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Masculino , Péptidos/química , Péptidos/administración & dosificación , Péptidos/farmacología , Células Cultivadas , Ratas
2.
Allergol. immunopatol ; 50(1): 17-24, ene 2, 2022. ilus, graf
Artículo en Inglés | IBECS | ID: ibc-203081

RESUMEN

Background Curcumol, possessing antiviral, antifungal, antimicrobial, anticancer, and anti-inflammatory properties, has been widely used in treating cancers and liver fibrosis. The aim of this study was to determine the effect of curcumol on the progression of asthma.Materials and methods Curcumol was administrated to platelet-derived growth factor (PDGF)-BB-stimulated airway smooth muscle cells (ASMCs). The proliferation of ASMCs was assessed by MTT and EdU incorporation assays. The apoptosis of ASMCs was measured by flow cytometry and Western blotting. The migration of ASMCs was evaluated by Transwell migration assay and Western blotting. The regulatory effects of curcumol on extracellular signal-regulated protein kinase (ERK)/cAMP response element-binding protein (CREB) pathway was evaluated by Western blotting.Results The proliferation and migration of ASMCs induced by PDGF-BB was suppressed, and the apoptosis of ASMCs was elevated by curcumol in a dose-dependent manner. The activation of ERK/CREB pathway induced by PDGF-BB was suppressed by curcumol.Conclusion Curcumol could suppress ERK/CREB pathway to inhibit proliferation and migration and promote apoptosis of PDGF-BB-stimulated ASMCs. These findings suggest that curcumol may act as a potential drug for asthma treatment (AU)


Asunto(s)
Humanos , Asma/metabolismo , Becaplermina/administración & dosificación , Proteína de Unión a Elemento de Respuesta al AMP Cíclico , Quinasas MAP Reguladas por Señal Extracelular , Miocitos del Músculo Liso , Movimiento Celular , Proliferación Celular , Sesquiterpenos , Transducción de Señal , Apoptosis
3.
CNS Neurosci Ther ; 27(11): 1300-1312, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34346167

RESUMEN

AIMS: The neurotropic growth factor PDGF-BB was shown to have vital neurorestorative functions in various animal models of Parkinson's disease (PD). Previous studies indicated that the regenerative property of PDGF-BB contributes to the increased intensity of tyrosine hydroxylase (TH) fibers in vivo. However, whether PDGF-BB directly modulates the expression of TH, and the underlying mechanism is still unknown. We will carefully examine this in our current study. METHOD: MPTP-lesion mice received PDGF-BB treatment via intracerebroventricular (i.c.v) administration, and the expression of TH in different brain regions was assessed by RT-PCR, Western blot, and immunohistochemistry staining. The molecular mechanisms of PDGF-BB-mediated TH upregulation were examined by RT-PCR, Western blot, ChIP assay, luciferase reporter assay, and immunocytochemistry. RESULTS: We validated a reversal expression of TH in MPTP-lesion mice upon i.c.v administration of PDGF-BB for seven days. Similar effects of PDGF-BB-mediated TH upregulation were also observed in MPP+ -treated primary neuronal culture and dopaminergic neuronal cell line SH-SY5Y cells. We next demonstrated that PDGF-BB rapidly activated the pro-survival PI3K/Akt and MAPK/ERK signaling pathways, as well as the downstream CREB in SH-SY5Y cells. We further confirmed the significant induction of p-CREB in PDGF-BB-treated animals in vivo. Using a genetic approach, we demonstrated that the transcription factor CREB is critical for PDGF-BB-mediated TH expression. The activation and nucleus translocation of CREB were promoted in PDGF-BB-treated SH-SY5Y cells, and the enrichment of CREB on the promoter region of TH gene was also increased upon PDGF-BB treatment. CONCLUSION: Our data demonstrated that PDGF-BB directly regulated the expression of TH via activating the downstream Akt/ERK/CREB signaling pathways. Our finding will further support the therapeutic potential of PDGF-BB in PD, and provide the possibility that targeting PDGF signaling can be harnessed as an adjunctive therapy in PD in the future.


Asunto(s)
Becaplermina/farmacología , Neuronas Dopaminérgicas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Transducción de Señal/efectos de los fármacos , Tirosina 3-Monooxigenasa/biosíntesis , Animales , Becaplermina/administración & dosificación , Línea Celular Tumoral , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/efectos de los fármacos , Femenino , Humanos , Inmunohistoquímica , Inyecciones Intraventriculares , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Intoxicación por MPTP/patología , Ratones , Ratones Endogámicos C57BL , Fármacos Neuroprotectores/administración & dosificación , Proteína Oncogénica v-akt/genética , Enfermedad de Parkinson Secundaria/inducido químicamente , Enfermedad de Parkinson Secundaria/patología , Embarazo
5.
Theranostics ; 10(26): 12144-12157, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33204334

RESUMEN

Rationale: Capillaries are composed of endothelial cells and the surrounding mural cells, pericytes. Microvascular repair after injury involves not only the proliferation of endothelial cells but also pericyte-based vessel stabilization. Exogenous bone marrow derived-putative endothelial progenitor cells (b-pEPCs) have the potential for vascular repair; however, their effect on vascular structure stabilization and pericyte-related pathobiological outcomes in the injured kidney has not been fully examined. Methods: We applied ischemia-reperfusion (IR) to induce renal vascular injury and renal fibrosis in mice. Platelet-derived growth factor receptor ß (PDGFR-ß)-DTR-positive mice were generated to deplete pericytes, and exogenous b-pEPCs and the PDGFR-ß ligand, PDGF chain B (PDGF-BB), were employed to explore the relationship among b-pEPCs, pericytes, vascular repair, and early renal fibrosis. Results: Administration of b-pEPCs reduced IR-induced pericyte-endothelial detachment, pericyte proliferation, and myofibroblast transition via a paracrine mode, which preserved not only vascular stabilization but also ameliorated IR-initiated renal fibrosis. PDGF-BB upregulated the expression of PDGFR-ß, exacerbated vascular abnormality, and pericyte-myofibroblast transition, which were ameliorated by b-pEPCs administration. The exogenous b-pEPCs and their culture medium (CM) induced vascular injury protection, and renal fibrosis was blocked by selective deletion of pericytes. Conclusion: Exogenous b-pEPCs directly protect against IR-induced vascular injury and prevent renal fibrosis by inhibiting the activation of PDGFR-ß-positive pericytes.


Asunto(s)
Lesión Renal Aguda/prevención & control , Células Progenitoras Endoteliales/trasplante , Riñón/patología , Pericitos/patología , Daño por Reperfusión/prevención & control , Lesión Renal Aguda/patología , Animales , Becaplermina/administración & dosificación , Becaplermina/metabolismo , Proliferación Celular , Modelos Animales de Enfermedad , Fibrosis , Humanos , Inyecciones Intraperitoneales , Riñón/irrigación sanguínea , Masculino , Ratones , Miofibroblastos/patología , Parabiosis , Comunicación Paracrina , Proteínas , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Daño por Reperfusión/patología
6.
Neurosci Lett ; 727: 134930, 2020 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-32251708

RESUMEN

Accumulated evidence indicates that platelet-derived growth factor (PDGF) contributes to various types of tissue regeneration. However, the effects and mechanisms of PDGF signaling for retina regeneration have not been sufficiently investigated. To clarify this, we investigated the role of PDGF signaling in retina regeneration process after needle puncture in zebrafish. Time-course analysis showed a spike peak of pdgf-a at 6 h after injury and a broad peak of pdgf-b during 6-96 h after injury. Inhibition of PDGF signaling with AG1295 suppressed BrdU-positive proliferative cell numbers at 4 days after injury. At the same time, retina regeneration-associated transcription factors, ascl1a and pax6b, were down-regulated by AG1295 treatment. Intravitreal injection of human recombinant PDGF-AA or -BB into intact zebrafish induced the cell proliferation. PDGF-BB injection induced the Müller glia-derived neurogenic cluster; PDGF-AA increased the 4C4-positive microglia. These findings indicate that PDGF signaling contributes to retina regeneration in zebrafish and causes different types of cell proliferation, depending on each subtype of PDGF. (160 words).


Asunto(s)
Becaplermina/administración & dosificación , Regeneración Nerviosa/fisiología , Factor de Crecimiento Derivado de Plaquetas/administración & dosificación , Retina/fisiología , Transducción de Señal/efectos de los fármacos , Animales , Animales Modificados Genéticamente , Becaplermina/metabolismo , Humanos , Inyecciones Intravítreas/métodos , Regeneración Nerviosa/efectos de los fármacos , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Retina/efectos de los fármacos , Transducción de Señal/fisiología , Pez Cebra , Proteínas de Pez Cebra/metabolismo
7.
Biomaterials ; 232: 119722, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31901689

RESUMEN

A major problem after tendon laceration is the low mechanical strength of the repaired tissue. One viable strategy for improving the functional and biomechanical properties of ruptured and repaired tendons is the delivery of growth factors at the injury site. Here, bioactive and reversibly expandable double-layered emulsion and coaxially electrospun tubes made from biodegradable DegraPol® (DP) (polyester urethane), delivering platelet-derived growth factor BB (PDGF-BB), are explored as implants to improve tendon healing in a rabbit Achilles tendon full laceration model. In vitro studies showed that both emulsion and coaxially electrospun scaffolds allow sustained delivery of bioactive PDGF-BB with similar release kinetics (150-190 pg PDGF-BB/mg of DP scaffold) over a period of 30 days. In vivo assessment after three weeks showed that PDGF-BB delivery through the bioactive DP tubes increased the tensile strength of the treated tendons 2-fold without additional pro-fibrotic effects, i.e., cell hyperproliferation or increase in α-smooth muscle actin expression at the wound site. While no major differences in ECM composition at the wound site were observed for ± PDGF-BB treated samples, collagen I and III were upregulated and fibronectin was downregulated compared to native tendons. In areas away from the wound, increased fibronectin expression was observed qualitatively in regions with lower collagen I and III expression. Both types of bioactive DP tubes provided surgeon-friendly and stable implants to deliver bioactive molecules and positively affected the strength of the repaired tendons after 3 weeks, thus presenting promising bioactive implants for clinical applications in the tendon repair field.


Asunto(s)
Tendón Calcáneo , Becaplermina/administración & dosificación , Traumatismos de los Tendones/terapia , Animales , Sistemas de Liberación de Medicamentos , Conejos , Rotura/terapia
8.
J Invest Surg ; 33(5): 476-488, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30430878

RESUMEN

Background and Objectives: Beta-tricalcium phosphate (beta-TCP) has been used for bone regeneration. The objective of this study was to assess longitudinally, the regeneration of critical sized segmental defects (CSSD) in rat femur using beta-TCP with or without recombinant platelet-derived growth factor (PDGF) through in vivo micro-computed tomography (micro-CT). Materials and Methods: Following ethical approval unilateral femoral CSSD measuring 5 mm was surgically created, under general anesthesia, in 30 male Wistar-Albino rats (aged 12-18 months; weighing 450-500 g). CSSD was stabilized using titanium mini-plate (4 holes, 1.0 mm thick with 8 mm bar). Depending upon biomaterial used for regeneration, the animals were randomly divided into: Control group (N = 10): CSSD covered with resorbable collagen membrane (RCM) only; Beta-TCP group (N = 10): CSSD filled with beta-TCP and covered by RCM; Beta-TCP + PDGF group (N = 10): CSSD filled with beta-TCP soaked in recombinant PDGF and covered by RCM. Longitudinal in vivo micro-CT analysis of the CSSD was done postoperatively at baseline, 2nd, 4th, 6th, and 8th weeks to assess volume and mineral density of newly formed bone (NFB) and beta-TCP. Results: Significant increase in NFB volume (NFBV) and mineral density (NFBMD) were observed from baseline to 8-weeks in all groups. Based on longitudinal in vivo micro-CT at 8-weeks, beta-TCP + PDGF group had significantly higher (p < 0.01) NFBV (38.98 ± 7.36 mm3) and NFBMD (3.72 ± 0.32 g/mm3) than the beta-TCP (NFBV-31.15 ± 6.68 mm3; NFBMD-2.28 ± 0.86g/mm3) and control (NFBV: 5.60 ± 1.06 mm3; NFBMD: 0.27 ± 0.02 g/mm3) groups. Significantly, higher reduction in beta-TCP volume (TCPV) and mineral density (TCPMD) were 1 observed in the beta-TCP + PDGF group when compared to the beta-TCP group. Conclusion: Addition of recombinant PDGF to beta-TCP enhanced bone regeneration within rat femoral CSSD and increased resorption rates of beta-TCP particles.


Asunto(s)
Becaplermina/administración & dosificación , Regeneración Ósea/efectos de los fármacos , Sustitutos de Huesos/administración & dosificación , Fosfatos de Calcio/administración & dosificación , Procedimientos Ortopédicos/métodos , Animales , Densidad Ósea/efectos de los fármacos , Placas Óseas , Modelos Animales de Enfermedad , Fémur/diagnóstico por imagen , Fémur/lesiones , Fémur/fisiología , Fémur/cirugía , Humanos , Estudios Longitudinales , Masculino , Procedimientos Ortopédicos/instrumentación , Ratas , Microtomografía por Rayos X
9.
J Cell Mol Med ; 24(1): 1128-1140, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31755222

RESUMEN

Increased immature neovessels contribute to plaque growth and instability. Here, we investigated a method to establish functional and stable neovessel networks to increase plaque stability. Rabbits underwent aortic balloon injury and were divided into six groups: sham, vector and lentiviral transfection with vascular endothelial growth factor-A (VEGF)-A, fibroblast growth factor (FGF)-2, platelet-derived growth factor (PDGF)-BB and FGF-2 + PDGF-BB. Lentivirus was percutaneously injected into the media-adventitia of the abdominal aorta by intravascular ultrasound guidance, and plaque-rupture rate, plaque-vulnerability index and plaque neovessel density at the injection site were evaluated. Confocal microscopy, Prussian Blue assay, Evans Blue, immunofluorescence and transmission electron microscopy were used to assess neovessel function and pericyte coverage. To evaluate the effect of FGF-2/PDGF-BB on pericyte migration, we used the mesenchymal progenitor cell line 10T1/2 as an in vitro model. VEGF-A- and FGF-2-overexpression increased the number of immature neovessels, which caused intraplaque haemorrhage and inflammatory cell infiltration, eventually resulting in the plaque vulnerability; however, FGF-2/PDGF-BB induced mature and functional neovessels, through increased neovessel pericyte coverage. Additionally, in vitro analysis of 10T1/2 cells revealed that FGF-2/PDGF-BB induced epsin-2 expression and enhanced the VEGF receptor-2 degradation, which negatively regulated pericyte function consistent with the in vivo data. These results showed that the combination of FGF-2 and PDGF-BB promoted the function and maturation of plaque neovessels, thereby representing a novel potential treatment strategy for vulnerable plaques.


Asunto(s)
Becaplermina/administración & dosificación , Factor 2 de Crecimiento de Fibroblastos/administración & dosificación , Vectores Genéticos/administración & dosificación , Lentivirus/genética , Neovascularización Patológica/prevención & control , Placa Aterosclerótica/terapia , Proteínas Adaptadoras del Transporte Vesicular , Animales , Becaplermina/genética , Becaplermina/metabolismo , Movimiento Celular , Factor 2 de Crecimiento de Fibroblastos/genética , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Vectores Genéticos/genética , Masculino , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Fosforilación , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patología , Conejos
10.
FEBS Open Bio ; 10(3): 327-337, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31571428

RESUMEN

Current methods for tendon rupture repair suffer from two main drawbacks: insufficient strength and adhesion formation, which lead to rerupture and impaired gliding. A novel polymer tube may help to overcome these problems by allowing growth factor delivery to the wound site and adhesion reduction, and by acting as a physical barrier to the surrounding tissue. In this study, we used a bilayered DegraPol® tube to deliver PDGF-BB to the wound site in a full-transection rabbit Achilles tendon model. We then performed histological and immunohistochemical analysis at 3 weeks postoperation. Sustained delivery of PDGF-BB to the healing Achilles tendon led to a significantly more homogenous cell distribution within the healing tissue. Lower cell densities next to the implant material were determined for +PDGF-BB samples compared to -PDGF-BB. PDGF-BB application increased proteoglycan content and reduced alpha-SMA+ areas, clusters of different sizes, mainly vessels. Finally, PDGF-BB reduced collagens I and III in the extracellular matrix. The sustained delivery of PDGF-BB via an electrospun DegraPol® tube accelerated tendon wound healing by causing a more uniform cell distribution with higher proteoglycan content and less fibrotic tissue. Moreover, the application of this growth factor reduced collagen III and alpha-SMA, indicating a faster and less fibrotic tendon healing.


Asunto(s)
Tendón Calcáneo/metabolismo , Becaplermina/farmacología , Sistemas de Liberación de Medicamentos/métodos , Tendón Calcáneo/cirugía , Animales , Becaplermina/administración & dosificación , Proliferación Celular , Colágeno/metabolismo , Matriz Extracelular/metabolismo , Femenino , Péptidos y Proteínas de Señalización Intercelular/farmacología , Conejos , Procedimientos de Cirugía Plástica/métodos , Rotura/tratamiento farmacológico , Rotura/patología , Rotura/cirugía , Traumatismos de los Tendones/tratamiento farmacológico , Traumatismos de los Tendones/patología , Traumatismos de los Tendones/cirugía , Adherencias Tisulares/patología , Cicatrización de Heridas/fisiología
11.
Sci Rep ; 9(1): 8073, 2019 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-31147568

RESUMEN

With the rapid development of tissue engineering therapies, there is a growing interest in the application of recombinant human growth factors (rhGFs) to regenerate human orofacial bones. However, despite reports of their ability to promote orofacial bone regeneration in animal experiments, their benefits in human clinical treatments remain unclear. Furthermore, the appropriate concentrations or indications of a specific rhGF remain ambiguous. Therefore it is essential to collect data from diverse clinical trials to evaluate their effects more precisely. Here we reviewed randomized clinical trials (RCT) that focused on the utilization of rhGFs in orofacial bone regeneration. Data from included studies were extracted, pooled and then quantitatively analyzed according to a pre-established protocol. Our results indicate that all current concentrations of rhBMP-2 produces insufficient effect on promoting either tooth extraction socket healing, sinus augmentation or reconstruction of alveolar clefts. However, 0.3 mg/ml rhPDGF-BB promotes the healing of tooth extraction sockets, though the effect does not reach a level of statistical significance. Summarily, we recommend concentrations of 0.3 mg/ml rhPDGF-BB only for the healing of tooth extraction sockets.


Asunto(s)
Becaplermina/administración & dosificación , Proteína Morfogenética Ósea 2/administración & dosificación , Regeneración Ósea/efectos de los fármacos , Huesos Faciales/efectos de los fármacos , Factor de Crecimiento Transformador beta/administración & dosificación , Injerto de Hueso Alveolar/métodos , Relación Dosis-Respuesta a Droga , Huesos Faciales/fisiología , Huesos Faciales/cirugía , Humanos , Ensayos Clínicos Controlados Aleatorios como Asunto , Proteínas Recombinantes/administración & dosificación , Elevación del Piso del Seno Maxilar/métodos , Resultado del Tratamiento , Cicatrización de Heridas/efectos de los fármacos
12.
Biochem Biophys Res Commun ; 516(2): 402-407, 2019 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-31217075

RESUMEN

Gestational diabetes mellitus (GDM) is often accompanied by the development of hyperinsulinemia as an adaptation to increased insulin demand, but this subsequently causes insulin resistance. Loss of function in pancreatic ß-cells further aggravates the development of GDM. The level of serum platelet-derived growth factor (PDGF) reportedly increases in GDM patients. The present study investigated whether enhanced PDGF signaling directly causes ß-cell dysfunction during gestation. Serum PDGF levels were negatively correlated with ß-cell function in GDM patients. Administration of PDGF-BB disrupted glucose tolerance and ß-cell function without inducing apoptosis in gestational mice but had no similar effect in non-gestational mice. The ß-cell-specific genes encoding insulin synthesis proteins were decreased in the islets of PDGF-BB-treated gestational mice. In vitro experiments using INS1 insulinoma cells showed that PDGF-BB promoted cell proliferation, whereas it downregulated ß-cell-specific genes. Taken together, these findings suggested that PDGF reduces ß-cell function during gestation possibly through ß-cell dedifferentiation.


Asunto(s)
Diabetes Gestacional/metabolismo , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal , Adulto , Animales , Becaplermina/administración & dosificación , Becaplermina/farmacología , Desdiferenciación Celular/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Diabetes Gestacional/sangre , Femenino , Prueba de Tolerancia a la Glucosa , Proteínas de Homeodominio/metabolismo , Humanos , Insulina/metabolismo , Secreción de Insulina/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Insulinoma/patología , Masculino , Ratones Endogámicos C57BL , Embarazo , Transporte de Proteínas/efectos de los fármacos , Ratas , Transactivadores/metabolismo
13.
Mar Drugs ; 17(6)2019 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-31226756

RESUMEN

Local administration of platelet-derived growth factor-BB (PGDF-BB) and bone morphogenetic protein-2 (BMP-2) in a sequential release manner could substantially promote bone healing. To achieve this goal, a delivery system that could sustain the release of PGDF-BB and BMP-2 by way of temporal separation was developed. One type of PGDF-BB-encapsulated alginate microsphere and another type of BMP-2-encapsulated microsphere with a core-shell structure were respectively produced using emulsification methods. These two types of microspheres were then embedded into chitosan/glycerophosphate hydrogel for constructing composite gels. Some of them were found to be injectable at ambient temperature and had thermo-sensitive features near physiological temperature and pH. The optimally formulated composite gels showed the ability to control the release of PGDF-BB and BMP-2 in a sequential fashion in which PDGF-BB was released earlier than BMP-2. In vitro release patterns indicated that the release rates could be significantly regulated by varying the embedded amount of the factor-encapsulated microspheres, which can in turn mediate the temporal separation release interval between PGDF-BB and BMP-2. The released PDGF-BB and BMP-2 were detected to be bioactive based on their respective effects on Balb/c 3T3 and C2C12 cells. These results suggest that the presently developed composite gels have the potential for bone repair by synergistically utilizing the early chemotactic effect of PDGF-BB and the subsequent osteogenic and angiogenic functions of PDGF-BB and BMP-2.


Asunto(s)
Becaplermina/administración & dosificación , Quitosano/química , Hidrogeles/química , Proteínas Proto-Oncogénicas c-sis/administración & dosificación , Alginatos/química , Animales , Células 3T3 BALB , Diferenciación Celular/efectos de los fármacos , Línea Celular , Ratones , Microesferas , Osteogénesis/efectos de los fármacos , Andamios del Tejido
14.
Phytomedicine ; 60: 152938, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31078367

RESUMEN

BACKGROUND: Tylophorine (TYL) is an alkaloid with antiproliferative action in cancer cells. Vascular smooth muscle cell (VSMC) proliferation and neointima formation contribute to restenosis after percutaneous coronary interventions. HYPOTHESIS/PURPOSE: Our goal was to examine the potential of TYL to inhibit VSMC proliferation and migration, and to dissect underlying signaling pathways. STUDY DESIGN AND METHODS: TYL was administered to platelet-derived growth factor (PDGF-BB)-stimulated, serum-stimulated, quiescent and unsynchronized VSMC of rat and human origin. BrdU incorporation and resazurin conversion were used to assess cell proliferation. Cell cycle progression was analyzed by flow cytometry of propidium iodide-stained nuclei. Expression profiles of proteins and mRNAs were determined using western blot analysis and RT-qPCR. The Click-iT OPP Alexa Fluor 488 assay was used to monitor protein biosynthesis. RESULTS: TYL inhibited PDGF-BB-induced proliferation of rat aortic VSMCs by arresting cells in G1 phase of the cell cycle with an IC50 of 0.13 µmol/l. The lack of retinoblastoma protein phosphorylation and cyclin D1 downregulation corroborated a G1 arrest. Inhibition of proliferation and cyclin D1 downregulation were species- and stimulus-independent. TYL also decreased levels of p21 and p27 proteins, although at later time points than observed for cyclin D1. Co-treatment of VSMC with TYL and MG132 or cycloheximide (CHX) excluded proteasome activation by TYL as the mechanism of action. Comparable time-dependent downregulation of cyclin D1, p21 and p27 in TYL- or CHX-treated cells, together with decreased protein synthesis observed in the Click-iT assay, suggests that TYL is a protein synthesis inhibitor. Besides proliferation, TYL also suppressed migration of PDGF-activated VSMC. In a human saphenous vein organ culture model for graft disease, TYL potently inhibited intimal hyperplasia. CONCLUSION: This unique activity profile renders TYL an interesting lead for the treatment of vasculo-proliferative disorders, such as restenosis.


Asunto(s)
Alcaloides/farmacología , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ciclina D1/efectos de los fármacos , Indolizinas/farmacología , Fenantrenos/farmacología , Biosíntesis de Proteínas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Alcaloides/administración & dosificación , Alcaloides/química , Animales , Becaplermina/administración & dosificación , Ciclina D1/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Indolizinas/administración & dosificación , Indolizinas/química , Miocitos del Músculo Liso/efectos de los fármacos , Técnicas de Cultivo de Órganos , Fenantrenos/administración & dosificación , Fenantrenos/química , Ratas , Ratas Sprague-Dawley , Venas Umbilicales
15.
Pulm Pharmacol Ther ; 56: 20-28, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30876906

RESUMEN

Airway smooth muscle cell (ASMC) hyperplasia causes airway wall remodelling, which is resisting to therapy. Long acting ß2-agonists (LABA) relax airway muscles, but their effect on remodelling is unclear. This study compared the anti-proliferative effect of LABA in human primary ASMC, in situations where LABA were applied before, together, or after platelet derived growth factor (PDGF-BB). Cells obtained from controls (n = 5), and asthma patients (n = 5) were stimulated by PDGF-BB (10 ng/ml) before or after the application of formoterol or salmeterol. Proliferation was determined by direct cell counts over three days, cell cycle control proteins p21(Waf1/Cip1), p27(Kip1), signalling proteins Erk1/2 and p38 mitogen activated protein kinase (MAPK) were detected by immuno-blotting. PDGF-BB induced proliferation was significantly stronger in asthmatic ASMC versus controls. Proliferation was prevented by 30 min pre-incubation with LABA. When LABA were applied together or after PDGF-BB, their anti-proliferative effect was no longer significant. In untreated ASMC, LABA increased the expression of p21(Waf1/Cip1) and p27(Kip1) through cAMP, and this mechanism was abolished by the presence of PDGF-BB. The data show that the anti-proliferative effect of cAMP signalling cannot overcome the mitogenic signalling cascade once it was activated. Therefore, remodelling in asthma cannot be reduced by LABA.


Asunto(s)
Agonistas de Receptores Adrenérgicos beta 2/farmacología , Asma/tratamiento farmacológico , AMP Cíclico/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Adulto , Anciano , Anciano de 80 o más Años , Remodelación de las Vías Aéreas (Respiratorias)/efectos de los fármacos , Asma/fisiopatología , Becaplermina/administración & dosificación , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Preparaciones de Acción Retardada , Femenino , Fumarato de Formoterol/farmacología , Humanos , Masculino , Persona de Mediana Edad , Mitógenos/metabolismo , Miocitos del Músculo Liso/metabolismo , Xinafoato de Salmeterol/farmacología
16.
J Ethnopharmacol ; 235: 155-163, 2019 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-30763696

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: The abnormal increase in vascular smooth muscle cell (VSMC) proliferation and migration are critical events in the pathogenesis of cardiovascular diseases (CVDs) including restenosis and atherosclerosis. The dried roots of Scutellaria baicalensis Georgi (common name: Huangqin in China) have been confirmed to possess beneficial effects on CVD by clinical and modern pharmacological studies. Flavonoids in Huangqin exert anti-proliferative and anti-migratory effects. Similar to Huangqin, Scutellaria strigillosa Hemsley (SSH) has been used to clear heat and damp and is especially rich in flavonoids including wogonin, wogonoside, baicalein, and baicalin. However, there have been few of reports about pharmacological activities of SSH. AIM OF THE STUDY: To investigate the anti-proliferative and anti-migratory properties of Scutellaria strigillosa Hemsley extract (SSHE) in vitro and in vivo and explore its possible mechanism of action. MATERIALS AND METHODS: The chemical constituents of SSHE were analyzed by ultra-high performance liquid chromatography coupled with triple time-of-flight mass spectrometry (UPLC-Triple-TOF-MS/MS). Cell proliferation and migration were investigated using BrdU incorporation assay and cell scratch test, respectively. The protein expression was determined by western blotting. In vivo, we established an artery ligation model of C57BL/6 mice and orally administered them with 50 or 100 mg/kg/day of SSHE. The carotid arteries were harvested and the intima-media thickness was examined 28 days post-ligation. RESULTS: Twelve compounds were identified and tentatively characterized. SSHE significantly inhibited the VSMC proliferation and migration stimulated by PDGF-BB and decreased the relative protein expression of regulatory signaling intermediates. Furthermore, the expression of SM22α was significantly elevated in SSHE-pretreated VSMCs, whereas knockdown of SM22α impaired the PDGF-BB-induced proliferation and migration arrest. Meanwhile, both ROS generation and the phosphorylation of ERK decreased in SSHE-pretreated VSMCs. In carotid artery ligation mice model, SSHE treatment significantly inhibited neointimal hyperplasia. CONCLUSIONS: SSHE significantly inhibited the PDGF-BB-induced VSMC proliferation, migration, and neointimal hyperplasia of carotid artery caused by ligation. Upregulation of SM22α expression, inhibition of ROS generation and ERK phosphorylation were, at least, partly responsible for the effects of SSHE on VSMCs.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Extractos Vegetales/farmacología , Scutellaria/química , Animales , Becaplermina/administración & dosificación , Grosor Intima-Media Carotídeo , Movimiento Celular/efectos de los fármacos , Cromatografía Líquida de Alta Presión , Relación Dosis-Respuesta a Droga , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Extractos Vegetales/administración & dosificación , Ratas , Espectrometría de Masas en Tándem
17.
Swiss Med Wkly ; 149: w20011, 2019 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-30685867

RESUMEN

Despite major advances in medical, catheter-based or surgical treatment, cardiovascular diseases such as peripheral artery disease and coronary artery disease still cause significant morbidity and mortality. Furthermore, many patients do not qualify for catheter-based treatment or bypass surgery because of advanced disease or surgical risk. There is therefore an urgent need for novel treatment strategies. Therapeutic angiogenesis aims to restore blood flow to ischaemic tissue by stimulating the growth of new blood vessels through the local delivery of angiogenic factors, and may thus be an attractive treatment alternative for these patients. Angiogenesis is a complex process and the growth of normal, stable and functional vasculature depends on the coordinated interplay of different cell types and growth factors. Vascular endothelial growth factor-A (VEGF) is the fundamental regulator of vascular growth and the key target of therapeutic angiogenesis approaches. However, first-generation clinical trials of VEGF gene therapy have been disappointing, and a clear clinical benefit has yet to be established. In particular, VEGF delivery (a) appears to have a very limited therapeutic window in vivo: low doses are safe but mostly inefficient, whereas higher doses become rapidly unsafe; and (b) requires a sustained expression in vivo of at least about four weeks to achieve stable vessels that persist after cessation of the angiogenic stimulus. Here we will review the current understanding of how VEGF induces the growth of normal or pathological blood vessels, what limitations for the controlled induction of safe and efficient angiogenesis are intrinsically linked to the biological properties of VEGF, and how this knowledge can guide the design of more effective strategies for therapeutic angiogenesis.


Asunto(s)
Isquemia/terapia , Neovascularización Fisiológica , Factor A de Crecimiento Endotelial Vascular/metabolismo , Inductores de la Angiogénesis/administración & dosificación , Animales , Becaplermina/administración & dosificación , Humanos
18.
PLoS One ; 13(9): e0203046, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30180189

RESUMEN

BACKGROUND: Pulmonary vascular remodeling of pulmonary arterial hypertension (PAH) is characterized by an inappropriate increase of vascular cells. The receptor for advanced glycation end products (RAGE) is a type I single-pass transmembrane protein belonging to the immunoglobulin superfamily and is involved in a broad range of hyperproliferative diseases. RAGE is also implicated in the etiology of PAH and is overexpressed in pulmonary artery smooth muscle cells (PASMCs) in patients with PAH. We examined the role of RAGE in the inappropriate increase of PASMCs in patients with PAH. METHODS AND RESULTS: PASMCs were obtained from 12 patients with PAH including 9 patients with idiopathic PAH (IPAH) and 3 patients with heritable PAH (HPAH) (2 patients with BMPR2 mutation and one patient with SMAD9 mutation) who underwent lung transplantation. Western blot analysis and immunofluorescence staining revealed that RAGE and S100A8 and A9, ligands of RAGE, were overexpressed in IPAH and HPAH-PASMCs in the absence of any external growth stimulus. PDGF-BB (10 ng/mL) up-regulated the expression of RAGE in IPAH and HPAH-PASMCs. PAH-PASMCs are hyperplastic in the absence of any external growth stimulus as assessed by 3H-thymidine incorporation. This result indicates overgrowth characterized by continued growth under a condition of no growth stimulation in PAH-PASMCs. PDGF-BB stimulation caused a higher growth rate of PAH-PASMCs than that of non-PAH-PASMCs. AS-1, an inhibitor of TIR domain-mediated RAGE signaling, significantly inhibited overgrowth characterized by continued growth under a condition of no growth stimulation in IPAH and HPAH-PASMCs (P<0.0001). Furthermore, AS-1 significantly inhibited PDGF-stimulated proliferation of IPAH and HPAH-PASMCs (P<0.0001). CONCLUSIONS: RAGE plays a crucial role in the inappropriate increase of PAH-PASMCs. Inhibition of RAGE signaling may be a new therapeutic strategy for PAH.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Hipertensión Pulmonar/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Arteria Pulmonar/metabolismo , Remodelación Vascular/fisiología , Adolescente , Adulto , Anciano , Becaplermina/administración & dosificación , Becaplermina/metabolismo , Calgranulina A/metabolismo , Calgranulina B/metabolismo , Proliferación Celular/fisiología , Células Cultivadas , Niño , Femenino , Humanos , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/patología , Hipertensión Pulmonar/cirugía , Masculino , Persona de Mediana Edad , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Arteria Pulmonar/patología , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA