Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 317
Filtrar
1.
ACS Appl Mater Interfaces ; 16(22): 28209-28221, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38778020

RESUMEN

Diabetic chronic wounds are notoriously difficult to heal as a result of their susceptibility to infection. To address this issue, we constructed an innovated and adaptable solution in the form of injectable chitosan (CS) hydrogel, denoted as CCOD, with enhanced antibacterial and anti-inflammatory properties. This hydrogel is created through a Schiff base reaction that combines chitosan-grafted chlorogenic acid (CS-CGA) and oxidized hyaluronic acid (OHA) with deferoxamine (DFO) as a model drug. The combination of CS and CGA has demonstrated excellent antibacterial and anti-inflammatory properties, while grafting played a pivotal role in making these positive effects stable. These unique features make it possible to customize injectable hydrogel and fit any wound shape, allowing for more effective and personalized treatment of complex bacterial infections. Furthermore, the hydrogel system is not only effective against inflammation and bacterial infections but also possesses antioxidant and angiogenic abilities, making it an ideal solution for the repair of chronic wounds that have been previously thought of as unmanageable.


Asunto(s)
Antibacterianos , Antiinflamatorios , Quitosano , Ácido Clorogénico , Deferoxamina , Ácido Hialurónico , Hidrogeles , Cicatrización de Heridas , Quitosano/química , Quitosano/farmacología , Ácido Hialurónico/química , Ácido Hialurónico/farmacología , Antibacterianos/química , Antibacterianos/farmacología , Deferoxamina/química , Deferoxamina/farmacología , Cicatrización de Heridas/efectos de los fármacos , Hidrogeles/química , Hidrogeles/farmacología , Animales , Ácido Clorogénico/química , Ácido Clorogénico/farmacología , Ácido Clorogénico/administración & dosificación , Antiinflamatorios/química , Antiinflamatorios/farmacología , Antiinflamatorios/administración & dosificación , Ratones , Humanos , Oxidación-Reducción , Inductores de la Angiogénesis/farmacología , Inductores de la Angiogénesis/química , Inductores de la Angiogénesis/administración & dosificación , Inductores de la Angiogénesis/uso terapéutico , Neovascularización Fisiológica/efectos de los fármacos , Staphylococcus aureus/efectos de los fármacos , Angiogénesis
2.
J Colloid Interface Sci ; 667: 54-63, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38615623

RESUMEN

Type 1 diabetes mellitus (T1DM) is a chronic disease affecting millions worldwide. Insulin therapy is currently the golden standard for treating T1DM; however, it does not restore the normal glycaemic balance entirely, which increases the risk of secondary complications. Beta-cell therapy may be a possible way of curing T1DM and has already shown promising results in the clinic. However, low retention rates, poor cell survival, and limited therapeutic potential are ongoing challenges, thus increasing the need for better cell encapsulation devices. This study aimed to develop a mechanically reinforced vascular endothelial growth factor (VEGF)-delivering encapsulation device suitable for beta cell encapsulation and transplantation. Poly(l-lactide-co-ε-caprolactone) (PLCL)/gelatin methacryloyl (GelMA)/alginate coaxial nanofibres were produced using electrospinning and embedded in an alginate hydrogel. The encapsulation device was physically and biologically characterised and was found to be suitable for INS-1E beta cell encapsulation, vascularization, and transplantation in terms of its biocompatibility, porosity, swelling ratio and mechanical properties. Lastly, VEGF was incorporated into the hydrogel and the release kinetics and functional studies revealed a sustained release of bioactive VEGF for at least 14 days, making the modified alginate system a promising candidate for improving the beta cell survival after transplantation.


Asunto(s)
Alginatos , Gelatina , Hidrogeles , Células Secretoras de Insulina , Factor A de Crecimiento Endotelial Vascular , Hidrogeles/química , Alginatos/química , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/citología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/administración & dosificación , Gelatina/química , Animales , Poliésteres/química , Ratas , Supervivencia Celular/efectos de los fármacos , Humanos , Diabetes Mellitus Tipo 1/terapia , Metacrilatos/química , Inductores de la Angiogénesis/química , Inductores de la Angiogénesis/farmacología , Inductores de la Angiogénesis/administración & dosificación , Propiedades de Superficie
3.
Naunyn Schmiedebergs Arch Pharmacol ; 397(7): 4677-4692, 2024 07.
Artículo en Inglés | MEDLINE | ID: mdl-38112730

RESUMEN

Ischemic heart diseases such as myocardial infarction (MI) are a global health problem and a leading cause of mortality worldwide. Angiogenesis is an important approach for myocardial healing following ischemia. Thus, this study aimed to explore the potential cardiac angiogenic effects of selenium (Se), alone and in combination with the tumor necrosis factor-alpha inhibitor, pentoxifylline (PTXF), via Akt/HIF-1α signaling. MI was induced in rats using two subcutaneous doses of isoprenaline (ISP) at a 24-h interval (150 mg/kg). One week later, rats were orally given Se (150 µg/kg/day), PTXF (50 mg/kg/day), or Se/PTXF combination. ISP-induced myocardial damage was evident by increased HW/TL ratios, ST segment elevation, and increased serum levels of CK-MB, LDH, and troponin-I. ISP increased the cardiac levels of the lipid peroxidation marker MDA; the pro-inflammatory cytokines IL-6, IL-1ß, and TNF-α; and the pro-apoptotic protein Bax and caspase-3. In contrast, the cardiac levels of the antioxidant markers GSH and SOD and the anti-apoptotic marker Bcl-2 were reduced. Furthermore, ISP markedly increased the cardiac levels of p-Akt and HIF-1α proteins and the cardiac gene expression of ANGPT-1, VEGF, and FGF-2. Treatment with Se both alone and in combination with PTXF ameliorated the ISP-induced myocardial damage and further increased cardiac angiogenesis via Akt/HIF-1α signaling. Se/PTXF combined therapy was more beneficial than individual treatments. Our study revealed for the first time the cardiac angiogenic effects of Se both alone and in combination with PTXF in myocardial infarction, suggesting that both may be promising candidates for clinical studies.


Asunto(s)
Modelos Animales de Enfermedad , Subunidad alfa del Factor 1 Inducible por Hipoxia , Infarto del Miocardio , Neovascularización Fisiológica , Pentoxifilina , Proteínas Proto-Oncogénicas c-akt , Selenio , Transducción de Señal , Animales , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/patología , Infarto del Miocardio/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Selenio/farmacología , Selenio/administración & dosificación , Masculino , Transducción de Señal/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Pentoxifilina/farmacología , Pentoxifilina/administración & dosificación , Pentoxifilina/uso terapéutico , Ratas Wistar , Ratas , Miocardio/metabolismo , Miocardio/patología , Quimioterapia Combinada , Inductores de la Angiogénesis/farmacología , Inductores de la Angiogénesis/administración & dosificación , Isoproterenol , Factor de Necrosis Tumoral alfa/metabolismo , Angiogénesis
4.
Pharm Biol ; 60(1): 65-74, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34913414

RESUMEN

CONTEXT: Concanavalin A (Con A) exhibited multiple roles in cancer cells. However, the role of Con A in endothelial cells was not reported. OBJECTIVE: Our present study investigated the potential angiogenic role of Con A in endothelial cells and ischaemic hind-limb mice. MATERIALS AND METHODS: Human umbilical vein endothelial cells and Ea.hy926 cells were employed to determine the effect of Con A (0.3, 1, and 3 µg/mL) or vehicle on angiogenesis and cell proliferation with tube formation, ELISA, flow cytometry, EdU, and western blot. Hind-limb ischaemic mice were conducted to determine the pro-angiogenic effect of Con A (10 mg/kg) for 7 days. RESULTS: Con A promoted tube formation to about three-fold higher than the control group and increased the secretion of VEGFa, PDGFaa, and bFGF in the medium. The cell viability was promoted to 1.3-fold by Con A 3 µg/mL, and cell cycle progression of G0G1 phase was decreased from 77% in the vehicle group to 70% in Con A 3 µg/mL, G2M was promoted from 15 to 19%, and S-phase was from 7 to 10%. Con A significantly stimulated phosphorylation of Akt and ERK1/2 and expression of cyclin D1 and decreased the expression of p27. These effects of Con A were antagonised by the PI3K inhibitor LY294002 (10 µM) and MEK pathway antagonist PD98059 (10 µM). Moreover, Con A (10 mg/kg) exhibited a repair effect in ischaemic hind-limb mice. DISCUSSION AND CONCLUSIONS: This study will provide a new option for treating ischaemic disease by local injection with Con A.


Asunto(s)
Inductores de la Angiogénesis/farmacología , Proliferación Celular/efectos de los fármacos , Concanavalina A/farmacología , Neovascularización Fisiológica/efectos de los fármacos , Inductores de la Angiogénesis/administración & dosificación , Animales , Supervivencia Celular/efectos de los fármacos , Cromonas/farmacología , Concanavalina A/administración & dosificación , Ciclina D1/metabolismo , Relación Dosis-Respuesta a Droga , Flavonoides/farmacología , Miembro Posterior , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Isquemia/tratamiento farmacológico , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Morfolinas/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo
5.
Gynecol Oncol ; 161(2): 496-501, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33637348

RESUMEN

OBJECTIVE: Report the results from a preplanned interim analysis of a phase III, double blind, randomized controlled study of ofranergene obadenovec (VB-111), a targeted anti-cancer gene therapy, in combination with paclitaxel in patients with platinum resistant ovarian cancer (PROC). METHODS: The OVAL (NCT03398655) study is an on-going study where patients are randomly assigned in a 1:1 ratio to weekly paclitaxel 80 mg/m2 with VB-111 or placebo. The protocol specifies a pre-planned unblinded futility interim analysis of CA-125 response per GCIG criteria in the first 60 evaluable patients. The futility rule determined for this analysis was that the response rate of VB-111 must be greater than the response rate of placebo by at least 10% in order to continue the study. Coincident with the interim analysis, the blinded CA-125 response rate was estimated as a proportion of the first 60 evaluable patients with CA-125 response per GCIG criteria. Post-treatment fever is provided as a possible surrogate marker of VB-111 therapy activity. RESULTS: The median age of the evaluable patients was 62 years (range 41-82); 97% had high-grade serous cancer; 58% had been treated with 3 or more previous lines of therapy, 70% received prior anti-angiogenic treatment, 43% received prior PARP inhibitors. CA-125 response in the VB-111 and weekly paclitaxel treated arm met the pre-specified interim criterion of an absolute advantage of 10% or higher compared to the control. Blinded results show a 53% CA-125 response rate (32/60) with 15% complete response (n=9). Assuming balanced randomization and an absolute advantage of 10% or higher to the VB-111 arm, it may be deducted that the response in the VB-111 treatment arm is 58% or higher. Among patients with post-treatment fever, the CA-125 response rate was 69%. CONCLUSIONS: At the time of the interim analysis, response rate findings are comparable to the responses seen in a similar patient population in the phase I/II study. The independent data and safety monitoring committee (iDSMC) recommended continuing the OVAL trial as planned. No new safety signals were identified.


Asunto(s)
Terapia Genética/métodos , Neoplasias Ováricas/terapia , Paclitaxel/administración & dosificación , Adenoviridae/genética , Adulto , Anciano , Anciano de 80 o más Años , Inductores de la Angiogénesis/administración & dosificación , Terapia Combinada , Método Doble Ciego , Esquema de Medicación , Femenino , Humanos , Persona de Mediana Edad , Neoplasias Ováricas/tratamiento farmacológico , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Transgenes , Receptor fas/genética
6.
Am J Physiol Heart Circ Physiol ; 320(3): H1066-H1079, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33356962

RESUMEN

Butyrate is a short-chain fatty acid (SCFA) derived from microbiota and is involved in a range of cell processes in a concentration-dependent manner. Low concentrations of sodium butyrate (NaBu) were shown to be proangiogenic. However, the mechanisms associated with these effects are not yet fully known. Here, we investigated the contribution of the SCFA receptor GPR43 in the proangiogenic effects of local treatment with NaBu and its effects on matrix remodeling using the sponge-induced fibrovascular tissue model in mice lacking the Gpr43 gene (Gpr43-KO) and the wild-type (WT) mice. We demonstrated that NaBu (0.2 mM intraimplant) treatment enhanced the neovascularization process, blood flow, and VEGF levels in a GPR43-dependent manner in the implants. Moreover, NaBu was able to modulate matrix remodeling aspects of the granulation tissue such as proteoglycan production, collagen deposition, and α-smooth muscle actin (α-SMA) expression in vivo, besides increasing transforming growth factor (TGF)-ß1 levels in the fibrovascular tissue, in a GPR43-dependent manner. Interestingly, NaBu directly stimulated L929 murine fibroblast migration and TGF-ß1 and collagen production in vitro. GPR43 was found to be expressed in human dermal fibroblasts, myofibroblasts, and endothelial cells. Overall, our findings evidence that the metabolite-sensing receptor GPR43 contributes to the effects of low dose of NaBu in inducing angiogenesis and matrix remodeling during granulation tissue formation. These data provide important insights for the proposition of new therapeutic approaches based on NaBu, beyond the highly explored intestinal, anti-inflammatory, and anticancer purposes, as a local treatment to improve tissue repair, particularly, by modulating granulation tissue components.NEW & NOTEWORTHY Our data show the contribution of the metabolite-sensing receptor GPR43 in the effects of low dose of sodium butyrate (NaBu) on stimulating angiogenesis and extracellular matrix remodeling in a model of granulation tissue formation in mice. We also show that human dermal fibroblasts, myofibroblasts, and endothelial cells express the receptor GPR43. These data provide important insights for the use of NaBu in local therapeutic approaches applicable to tissue repair in sites other than the intestine.


Asunto(s)
Inductores de la Angiogénesis/administración & dosificación , Ácido Butírico/administración & dosificación , Matriz Extracelular/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Tejido de Granulación/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Receptores Acoplados a Proteínas G/metabolismo , Animales , Línea Celular , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Colágeno/metabolismo , Modelos Animales de Enfermedad , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Fibroblastos/metabolismo , Fibroblastos/patología , Tejido de Granulación/metabolismo , Tejido de Granulación/patología , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Acoplados a Proteínas G/deficiencia , Receptores Acoplados a Proteínas G/genética , Tapones Quirúrgicos de Gaza , Factor de Crecimiento Transformador beta1/metabolismo
7.
Eur J Ophthalmol ; 31(2): 754-758, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32507032

RESUMEN

PURPOSE: To compare the efficacy of intravitreal injections (IVI) of ranibizumab (Lucentis®, Novartis, Basel, Switzerland; RAN), aflibercept (Eylea®, Bayer, Leverkusen, Germany; AFL) and dexamethasone implant (Ozurdex®, Allergan, Irvine, California; DXI) in the treatment of naive diabetic macular oedema (DME) during a 12-month follow-up, in real life. METHODS: Nineteen eyes treated with RAN, 20 with AFL and 21 with DXI were analysed from inclusion up to 12 months (M12) with intermediate analysis at M6. Best corrected visual acuity (BCVA), fundus and central retinal thickness (CRT) using spectral-domain optical coherence tomography (SD-OCT; Spectralis/HRA, Heidelberg Engineering, Germany) were performed at inclusion, M3, M6 and M12. RESULTS: BCVA improved until 67.9 letters ±13.3 SD (+5.5 letters) at M6 and 69.6 letters ±12 SD (+7.2 letters) at 12 months for RAN group (p = 0.036). For the AFL group it improved until 63.6 letters ±15.2 SD (+6.6 letters) at M6 and 67.5 letters ±12.2 SD (+8.5 letters) at 12 months (p = 0.014). Lastly DXI group improved by 66.9 letters ±15.1 SD (+7.9 letters) at M6 and 68.4 letters ±11.2 SD (+9.4 letters) at 12 months (p = 0.0023). CRT decreased by 124.4 µm at M6 and 99.3 µm at M12 in RAN group, 144.3 µm and 101.5 µm in AFL group and finally 95.6 µm and 162.7 µm in DXI group. CONCLUSION: In summary, these three drugs provide an efficient treatment option with an acceptable benefit-risk ratio for the treatment of naive patients with DME, whether on BCVA or CRT on the first year of treatment.


Asunto(s)
Inductores de la Angiogénesis/administración & dosificación , Dexametasona/administración & dosificación , Retinopatía Diabética/tratamiento farmacológico , Edema Macular/tratamiento farmacológico , Agudeza Visual , Anciano , Retinopatía Diabética/complicaciones , Retinopatía Diabética/diagnóstico , Angiografía con Fluoresceína/métodos , Estudios de Seguimiento , Fondo de Ojo , Glucocorticoides/administración & dosificación , Humanos , Inyecciones Intravítreas , Edema Macular/diagnóstico , Edema Macular/etiología , Masculino , Estudios Prospectivos , Retina/diagnóstico por imagen , Factores de Tiempo , Tomografía de Coherencia Óptica/métodos , Resultado del Tratamiento , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores
8.
Mol Pharm ; 18(2): 550-575, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-32519875

RESUMEN

The poor healing associated with chronic wounds affects millions of people worldwide through high mortality rates and associated costs. Chronic wounds present three main problems: First, the absence of a suitable environment to facilitate cell migration, proliferation, and angiogenesis; second, bacterial infection; and third, unbalanced and prolonged inflammation. Unfortunately, current therapeutic approaches have not been able to overcome these main issues and, therefore, have limited clinical success. Over the past decade, incorporating the unique advantages of nanomedicine into wound healing approaches has yielded promising outcomes. Nanomedicine is capable of stimulating various cellular and molecular mechanisms involved in the wound microenvironment via antibacterial, anti-inflammatory, and angiogenetic effects, potentially reversing the wound microenvironment from nonhealing to healing. This review briefly discusses wound healing mechanisms and pathophysiology and then highlights recent findings regarding the opportunities and challenges of using nanomedicine in chronic wound management.


Asunto(s)
Portadores de Fármacos/química , Nanopartículas/química , Piel/lesiones , Nanomedicina Teranóstica/métodos , Cicatrización de Heridas/efectos de los fármacos , Actinobacteria , Inductores de la Angiogénesis/administración & dosificación , Inductores de la Angiogénesis/farmacocinética , Animales , Antibacterianos/administración & dosificación , Antibacterianos/farmacocinética , Antiinflamatorios/administración & dosificación , Antiinflamatorios/farmacocinética , Vendajes , Enfermedad Crónica/tratamiento farmacológico , Modelos Animales de Enfermedad , Composición de Medicamentos/métodos , Humanos , Hidrogeles/química , Nanopartículas/administración & dosificación , Neovascularización Fisiológica/efectos de los fármacos , Fármacos Fotosensibilizantes/administración & dosificación , Terapia Fototérmica/métodos , Piel/efectos de los fármacos , Piel/inmunología , Piel/microbiología , Cicatrización de Heridas/fisiología
9.
Retin Cases Brief Rep ; 15(1): 24-26, 2021 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-29757814

RESUMEN

PURPOSE: To report early formation and spontaneous closure of a full-thickness macular hole that developed after successful pneumatic retinopexy in a patient who had been undergoing treatment for diabetic macular edema. METHODS: Case report of a 68-year-old man with bilateral nonproliferative diabetic retinopathy who was currently undergoing anti-vascular endothelial growth factor treatment for bilateral diabetic macular edema. RESULTS: On presentation, visual acuity was 20/200 in the left eye, and examination revealed a bullous, macula-off retinal detachment with a single horseshoe tear at 12 o'clock in the left eye. Pneumatic retinopexy was performed followed by laser augmentation 3 days later. Three weeks postoperatively, he returned with visual acuity of 20/50 and a full-thickness macular hole in the left eye. Although he elected for initial observation, he returned 2 weeks later with visual acuity of 20/50 in both eyes and a retinal detachment with a single break at 10 o'clock in the right eye. The macular hole in the left eye had spontaneously resolved. Pneumatic retinopexy was performed to the right eye. Over 1 year after bilateral pneumatic retinopexy, his retina remains without recurrence of a macular hole in the left eye. CONCLUSION: In the early postoperative period after pneumatic retinopexy to repair a retinal detachment, a macular hole can form and spontaneously close.


Asunto(s)
Inductores de la Angiogénesis/efectos adversos , Retinopatía Diabética/tratamiento farmacológico , Mácula Lútea/patología , Edema Macular/tratamiento farmacológico , Perforaciones de la Retina/cirugía , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Vitrectomía/métodos , Anciano , Inductores de la Angiogénesis/administración & dosificación , Retinopatía Diabética/complicaciones , Retinopatía Diabética/diagnóstico , Humanos , Inyecciones Intravítreas , Edema Macular/complicaciones , Edema Macular/diagnóstico , Masculino , Perforaciones de la Retina/diagnóstico , Perforaciones de la Retina/etiología , Tomografía de Coherencia Óptica , Agudeza Visual
10.
Eur J Pharm Biopharm ; 158: 1-10, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33152482

RESUMEN

Active self-encapsulation (ASE) is a recently developed post-loading method based on absorption of (positively charged) proteins in microporous PLGA microspheres loaded with negatively charged polysaccharides (trapping agents). The aim of this study was to investigate ASE for simultaneous loading and controlled release of multiple growth factors. For this purpose, vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF) and insulin-like growth factor (IGF) were loaded in microspheres containing high molecular weight dextran sulfate (HDS) as trapping agent; loading was performed in a concentrated growth factor solution of low ionic strength and of pH 5 under conditions at which the proteins are positively charged. Subsequent pore closure was induced by incubation of the growth factor-loaded microspheres at 42.5 °C, i.e. above the Tg of (hydrated) PLGA (~30 °C). A 1:1:1 combination of VEGF, FGF and IGF was loaded with high loading (4.3%) and loading efficiency (91%). The in vitro release kinetics and bioactivity of loaded growth factors were studied for 4 weeks using ELISA and an endothelial cell proliferation assay, respectively. While IGF was released quickly, VEGF and FGF were continuously released for 4 weeks in their bioactive form, whereby a growth factor combination had a synergistic angiogenic effect. Therefore, ASE is a suitable method for co-loading growth factors which can provide sustained release profiles of bioactive growth factors, which is attractive for vascularization of biomaterial implants.


Asunto(s)
Inductores de la Angiogénesis/administración & dosificación , Materiales Biocompatibles/administración & dosificación , Portadores de Fármacos/química , Microesferas , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Inductores de la Angiogénesis/farmacocinética , Materiales Biocompatibles/farmacocinética , Preparaciones de Acción Retardada/administración & dosificación , Preparaciones de Acción Retardada/farmacocinética , Composición de Medicamentos/métodos , Liberación de Fármacos , Factores de Crecimiento de Fibroblastos/administración & dosificación , Factores de Crecimiento de Fibroblastos/farmacocinética , Humanos , Neovascularización Fisiológica/efectos de los fármacos , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacocinética , Somatomedinas/administración & dosificación , Somatomedinas/farmacocinética , Factores de Crecimiento Endotelial Vascular/administración & dosificación , Factores de Crecimiento Endotelial Vascular/farmacocinética
11.
Inflammopharmacology ; 29(1): 153-166, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33201349

RESUMEN

Donepezil has proven to be an effective drug to reduce neuronal death and subsequently injury in neurodegenerative diseases. The current study evaluated the neuroprotective effects of donepezil in a rat model of ischaemic stroke and explored possible mechanisms which by this drug may reduce cell death. Temporary middle cerebral artery occlusion (tMCAO) was exerted for 45 min to induce ischaemic stroke. The animals were assigned into five groups: sham, control, and three groups treated with different doses of donepezil. Donepezil was intraperitoneally (IP) injected 4 h after reperfusion for 10 consecutive days. Infarct size was determined using TTC staining. The expression of proteins was evaluated using immunohistochemistry assays. Compared with the control group, infarct size was significantly reduced in tMCAO rats treated with different doses of donepezil. Moreover, our results showed significant decreased expression levels of apoptotic markers and pro-inflammatory mediators after treatment with different doses of donepezil for 10 days (P < 0.05). Likewise, significant increase of brain-derived neurotrophic factor (BDNF) and peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) proteins were found in tMCAO rats treated with donepezil compared with the control group (P < 0.05). Collectively, our findings show the validity of donepezil as a new therapeutic agent for attenuation of injury following ischaemic stroke through attenuation of inflammation and improvement of mitochondrial function, neurogenesis, and angiogenesis.


Asunto(s)
Donepezilo/farmacología , Inflamación/tratamiento farmacológico , Accidente Cerebrovascular Isquémico/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Inductores de la Angiogénesis/administración & dosificación , Inductores de la Angiogénesis/farmacología , Animales , Apoptosis/efectos de los fármacos , Donepezilo/administración & dosificación , Relación Dosis-Respuesta a Droga , Infarto de la Arteria Cerebral Media , Inflamación/fisiopatología , Accidente Cerebrovascular Isquémico/fisiopatología , Masculino , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Neurogénesis/efectos de los fármacos , Fármacos Neuroprotectores/administración & dosificación , Ratas , Ratas Wistar
12.
J Tradit Chin Med ; 40(6): 928-937, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33258344

RESUMEN

OBJECTIVE: To investigate the efficacy of Lichong decoction (LD) from Traditional Chinese Medicine, on micro-angiogenesis in a mouse model of hysteromyoma. METHODS: A mouse model of hysteromyoma was developed by orthotopic intrauterine injection of primary human myoma cells isolated from patients from the Beijing Obstetrics and Gynecology Hospital into CB-17 Scid mice. Mice were administered high-dose LD, low-dose LD, mifepristone or water (control) daily by gavage for 4 weeks. Uterine diameter and coefficient (uterine weight/body weight) were measured. Uterine morphology was assessed by light microscopy (hematoxylin and eosin) and transmission electron microscopy. Serum levels of estradiol, progesterone, follicle-stimulating hormone and luteinizing hormone (LH) were measured by enzyme-linked immunosorbent assay. Uterine protein expression of hypoxia inducible factor (HIF)-1α, CD31 and proliferating cell nuclear antigen (PCNA) was detected by immunohistochemistry. VEGF and HIF-1α mRNAs were quantified by RT-PCR. RESULTS: High-dose LD, low-dose LD and mifepristone reduced uterine diameter and coefficient, and attenuated the morphologic abnormalities associated with hysteromyoma. High-dose LD, low-dose LD and mifepristone inhibited hysteromyoma-induced micro-angiogenesis, as evidenced by a decrease in the number of new microvessels co-immunostaining for CD31 and PCNA (P < 0.01). High-dose LD and mifepristone lowered serum levels of estradiol, progesterone and LH (P < 0.05). High-dose LD, low-dose LD and mifepristone down-regulated HIF-1α mRNA and protein expressions and VEGF mRNA expression (P < 0.01). CONCLUSION: The inhibition of hysteromyoma by LD may involve reductions in HIF-1α and VEGF expression and suppression of micro-angiogenesis.


Asunto(s)
Inductores de la Angiogénesis/administración & dosificación , Medicamentos Herbarios Chinos/administración & dosificación , Mioma/tratamiento farmacológico , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Mioma/genética , Mioma/metabolismo , Mioma/fisiopatología , Neovascularización Patológica , Factor A de Crecimiento Endotelial Vascular/genética , Factores de Crecimiento Endotelial Vascular/genética
13.
Arterioscler Thromb Vasc Biol ; 40(12): 2922-2940, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32998518

RESUMEN

OBJECTIVE: In patients with peripheral artery disease, blockages in arterioles <1 mm cannot be treated surgically, and there are currently few effective medicines. Studies have shown that inflammation in ischemic tissue is related to injury recovery and angiogenesis, but insufficient attention has been paid to this area. Studies have suggested that HMGB1 (high mobility group protein 1), which is released by ischemic tissue, promotes angiogenesis, but the mechanism is not entirely clear. In this study, we tested the internalization of HMGB1 in endothelial cells and investigated a novel proangiogenic pathway. Approach and Results: Using green fluorescent protein-tagged HMGB1 to stimulate endothelial cells, we demonstrated HMGB1 internalization via dynamin and RAGE (receptor for advanced glycation end products)-dependent signaling. Using a fluorescence assay, we detected internalized protein fusion to lysosomes, followed by activation of CatB (cathepsin B) and CatL (cathepsin L). The latter promoted the release of VEGF (vascular endothelial growth factor)-A and endoglin and upregulated the capacities of cell migration, proliferation, and tube formation in endothelial cells. We identified that the cytokine-induced fragment-a key functional domain in HMGB1-mediates the internalization and angiogenic function of HMGB1. We further confirmed that HMGB1 internalization also occurs in vivo in endothelial cells and promotes angiogenesis in mouse femoral artery ligation. CONCLUSIONS: In this study, we identified a novel pathway of HMGB1 internalization-induced angiogenesis in endothelial cells. This finding sheds light on the regulatory role of inflammatory factors in angiogenesis through cell internalization and opens a new door to understand the relationship between inflammation and angiogenesis in ischemic diseases.


Asunto(s)
Inductores de la Angiogénesis/administración & dosificación , Células Progenitoras Endoteliales/metabolismo , Proteína HMGB1/administración & dosificación , Isquemia/tratamiento farmacológico , Músculo Esquelético/irrigación sanguínea , Neovascularización Fisiológica/efectos de los fármacos , Inductores de la Angiogénesis/metabolismo , Animales , Transporte Biológico , Células Cultivadas , Modelos Animales de Enfermedad , Dinaminas/metabolismo , Proteína HMGB1/metabolismo , Miembro Posterior , Inyecciones Intramusculares , Isquemia/genética , Isquemia/metabolismo , Isquemia/fisiopatología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor para Productos Finales de Glicación Avanzada/genética , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Flujo Sanguíneo Regional , Transducción de Señal , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
14.
Mater Sci Eng C Mater Biol Appl ; 115: 111123, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32600722

RESUMEN

Anti-inflammation and angiogenesis play an essential role in wound healing. In this study, we developed a composite hydrogel dressing with stepwise delivery of diclofenac sodium (DS) and basic fibroblast growth factor (bFGF) in the inflammation stage and new tissue formation stage respectively for wound repair. Sodium alginate (SA) crosslinked by calcium ion acted as the continuous phase, and thermosensitive bFGF-loaded poly(N-isopropylacrylamide) nanogels (pNIPAM NGs, LCST1 ~33 °C) and DS-loaded p(N-isopropylacrylamide-co-acrylic acid) nanogels [p(NIPAM-co-AA) NGs, LCST2 ~40 °C] acted as the dispersed phase. The synthesized SA/bFGF@pNIPAM/DS@p(NIPAM-co-AA) hydrogel presented a desirable storage modulus of ~4500 Pa, a high water equilibrium swelling ratio of ~90, an appropriate water vapor transmission rate of ~2300 g/m2/day, and nontoxicity to human skin fibroblasts. The in vitro thermosensitive cargo delivery of this hydrogel showed that 92% of DS was sustainably delivered at 37 °C within the early three days mimicking the inflammation stage, while 80% of bFGF was controlled released at 25 °C within the later eight days mimicking new tissue formation stage. The in vivo wound healing of rats showed that this composite hydrogel presented a better healing effect with a wound contraction of 96% at 14 d, less inflammation and higher angiogenesis, than all control groups. These findings indicate SA/bFGF@pNIPAM/DS@p(NIPAM-co-AA) composite hydrogel is a potential dressing for wound repair.


Asunto(s)
Inductores de la Angiogénesis/administración & dosificación , Antiinflamatorios/administración & dosificación , Diclofenaco/administración & dosificación , Factor 2 de Crecimiento de Fibroblastos/administración & dosificación , Cicatrización de Heridas/efectos de los fármacos , Resinas Acrílicas/química , Alginatos/química , Inductores de la Angiogénesis/química , Inductores de la Angiogénesis/farmacología , Animales , Antiinflamatorios/química , Antiinflamatorios/farmacología , Vendajes , Diclofenaco/química , Diclofenaco/farmacología , Modelos Animales de Enfermedad , Factor 2 de Crecimiento de Fibroblastos/química , Factor 2 de Crecimiento de Fibroblastos/farmacología , Humanos , Masculino , Nanogeles , Ratas
15.
Neurol Res ; 42(8): 683-692, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32584207

RESUMEN

OBJECTIVES: Following stroke, angiogenic strategy has been proposed to alleviate ischemia-induced injury by promoting angiogenesis and improving cerebrovascular function in the ischemic regions. Ligustilide (LGSL) is known to have neuroprotection against ischemic stroke-induced injury. But the effect of LGSL on promoting angiogenesis after ischemic stroke is unknown. The purpose of this study was to determine the effects of LGSL on neuroprotection and angiogenesis after stroke. METHODS: BrdU cell proliferation assay, transwell, wound healing, and tube-formation experiments were performed to evaluate the effects of LGSL on mouse microvascular endothelial cells (bEnd.3). Male adult C57/BL6 mice were subjected to focal transient cerebral ischemia followed by intragastric LGSL administration. Immunostaining was performed to assess angiogenesis. VEGF level was assayed by ELISA. eNOS expression was performed by Western blot. RESULTS: LGSL significantly improved neurological function. LGSL promoted angiogenesis in vitro and in vivo. Neurological improvement conferred by LGSL correlated with increased cerebral vessels number. We also found an increase of VEGF and eNOS activation in mice ischemic hemisphere following LGSL administration. CONCLUSION: LGSL promoted focal angiogenesis and attenuated ischemia-induced brain injury, suggesting that LGSL is a potential agent that improves angiogenesis and promotes recovery from ischemic stroke.


Asunto(s)
4-Butirolactona/análogos & derivados , Inductores de la Angiogénesis/administración & dosificación , Isquemia Encefálica/fisiopatología , Neovascularización Fisiológica/efectos de los fármacos , Fármacos Neuroprotectores/administración & dosificación , 4-Butirolactona/administración & dosificación , Animales , Conducta Animal/efectos de los fármacos , Isquemia Encefálica/prevención & control , Línea Celular , Masculino , Ratones , Ratones Endogámicos C57BL
16.
Am J Respir Crit Care Med ; 202(1): 100-111, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32240596

RESUMEN

Rationale: Advances in neonatal critical care have greatly improved the survival of preterm infants, but the long-term complications of prematurity, including bronchopulmonary dysplasia (BPD), cause mortality and morbidity later in life. Although VEGF (vascular endothelial growth factor) improves lung structure and function in rodent BPD models, severe side effects of VEGF therapy prevent its use in patients with BPD.Objectives: To test whether nanoparticle delivery of proangiogenic transcription factor FOXM1 (forkhead box M1) or FOXF1 (forkhead box F1), both downstream targets of VEGF, can improve lung structure and function after neonatal hyperoxic injury.Methods: Newborn mice were exposed to 75% O2 for the first 7 days of life before being returned to a room air environment. On Postnatal Day 2, polyethylenimine-(5) myristic acid/polyethylene glycol-oleic acid/cholesterol nanoparticles containing nonintegrating expression plasmids with Foxm1 or Foxf1 cDNAs were injected intravenously. The effects of the nanoparticles on lung structure and function were evaluated using confocal microscopy, flow cytometry, and the flexiVent small-animal ventilator.Measurements and Main Results: The nanoparticles efficiently targeted endothelial cells and myofibroblasts in the alveolar region. Nanoparticle delivery of either FOXM1 or FOXF1 did not protect endothelial cells from apoptosis caused by hyperoxia but increased endothelial proliferation and lung angiogenesis after the injury. FOXM1 and FOXF1 improved elastin fiber organization, decreased alveolar simplification, and preserved lung function in mice reaching adulthood.Conclusions: Nanoparticle delivery of FOXM1 or FOXF1 stimulates lung angiogenesis and alveolarization during recovery from neonatal hyperoxic injury. Delivery of proangiogenic transcription factors has promise as a therapy for BPD in preterm infants.


Asunto(s)
Inductores de la Angiogénesis/administración & dosificación , Sistemas de Liberación de Medicamentos , Proteína Forkhead Box M1/administración & dosificación , Factores de Transcripción Forkhead/administración & dosificación , Hiperoxia/tratamiento farmacológico , Nanopartículas , Alveolos Pulmonares/efectos de los fármacos , Inductores de la Angiogénesis/farmacología , Inductores de la Angiogénesis/uso terapéutico , Animales , Animales Recién Nacidos , Western Blotting , Femenino , Citometría de Flujo , Proteína Forkhead Box M1/farmacología , Proteína Forkhead Box M1/uso terapéutico , Factores de Transcripción Forkhead/farmacología , Factores de Transcripción Forkhead/uso terapéutico , Hiperoxia/patología , Hiperoxia/fisiopatología , Inyecciones Intravenosas , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Alveolos Pulmonares/irrigación sanguínea , Alveolos Pulmonares/patología , Alveolos Pulmonares/fisiopatología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Resultado del Tratamiento
17.
Int J Mol Sci ; 21(5)2020 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-32120998

RESUMEN

Biomaterials that promote angiogenesis are required for repair and regeneration of bone. In-situ formed injectable hydrogels functionalised with bioactive agents, facilitating angiogenesis have high demand for bone regeneration. In this study, pH and thermosensitive hydrogels based on chitosan (CS) and hydroxyapatite (HA) composite materials loaded with heparin (Hep) were investigated for their pro-angiogenic potential. Hydrogel formulations with varying Hep concentrations were prepared by sol-gel technique for these homogeneous solutions were neutralised with sodium bicarbonate (NaHCO3) at 4 °C. Solutions (CS/HA/Hep) constituted hydrogels setting at 37 °C which was initiated from surface in 5-10 minutes. Hydrogels were characterised by performing injectability, gelation, rheology, morphology, chemical and biological analyses. Hydrogel solutions facilitated manual dropwise injection from 21 Gauge which is highly used for orthopaedic and dental administrations, and the maximum injection force measured through 19 G needle (17.191 ± 2.296N) was convenient for manual injections. Angiogenesis tests were performed by an ex-ovo chick chorioallantoic membrane (CAM) assay by applying injectable solutions on CAM, which produced in situ hydrogels. Hydrogels induced microvascularity in CAM assay this was confirmed by histology analyses. Hydrogels with lower concentration of Hep showed more efficiency in pro-angiogenic response. Thereof, novel injectable hydrogels inducing angiogenesis (CS/HA/Hep) are potential candidates for bone regeneration and drug delivery applications.


Asunto(s)
Inductores de la Angiogénesis/administración & dosificación , Portadores de Fármacos/química , Heparina/administración & dosificación , Hidrogeles/química , Neovascularización Fisiológica/efectos de los fármacos , Animales , Regeneración Ósea/efectos de los fármacos , Embrión de Pollo , Quitosano/química , Membrana Corioalantoides/citología , Membrana Corioalantoides/efectos de los fármacos , Durapatita/química , Hidrogeles/síntesis química , Concentración de Iones de Hidrógeno , Microscopía Electrónica de Rastreo , Reología , Espectroscopía Infrarroja por Transformada de Fourier , Espectrometría Raman , Temperatura
18.
ACS Appl Mater Interfaces ; 12(10): 11375-11387, 2020 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-32068386

RESUMEN

Adipose-derived stem cell (ASC) spheroids exhibit enhanced angiogenic efficacy toward ischemia treatment. Thus, it is necessary to develop an all-in-one platform that enables efficient spheroid production, collection, and injectable implantation in vivo. The present study fabricated a poly(l-glutamic acid) (PLGA)-based porous hydrogel that can not only produce ASC spheroids but also conveniently collect spheroids for in vivo implantation via minimally invasive injection to treat hind limb ischemia. PLGA was cross-linked with cystamine (Cys), which contains disulfide bonds, to form a porous hydrogel that could realize "gel-sol" transition by the reduction effect of glutathione (GSH). For one thing, it was found that the introduction of the disulfide bond in the PLGA hydrogel promoted cellular adhesion via combining fibronectin, preventing the formation of spheroids, while the introduction of polyethylene glycol monomethyl ether (mPEG) could disturb the effect of the disulfide bond on cellular adhesion, supporting spheroid formation inside the porous hydrogel. For another, the porous hydrogel transferred into a syringe could turn into liquid polymer solution within about 40 min for collection of the produced spheroids and in vivo injection. In addition, because of the lubrication of polymer solution, the spheroids were protected during the injection of the spheroids/polymer suspensoid through a 25G syringe needle, avoiding damages from shearing. After the in vivo injection, the enhanced paracrine secretion of ASC spheroids resulted in promoted angiogenesis and muscle regeneration, exhibiting obvious therapeutic effect on limb ischemia in mice after 21 days. At the same time, PLGA-based material exhibited well-performed biocompatibility in vivo.


Asunto(s)
Inductores de la Angiogénesis , Hidrogeles , Isquemia/metabolismo , Células Madre Mesenquimatosas , Esferoides Celulares , Adipatos , Tejido Adiposo/citología , Inductores de la Angiogénesis/administración & dosificación , Inductores de la Angiogénesis/química , Inductores de la Angiogénesis/farmacología , Animales , Células Cultivadas , Cistamina , Disulfuros , Miembro Posterior/irrigación sanguínea , Humanos , Hidrogeles/administración & dosificación , Hidrogeles/química , Hidrogeles/farmacología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Neovascularización Fisiológica/efectos de los fármacos , Transición de Fase , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/metabolismo
19.
Aging (Albany NY) ; 12(6): 4727-4741, 2020 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-32045366

RESUMEN

In this study, we aimed to identify a candidate drug that can activate endogenous Angiopoietin 1 (Ang1) expression via drug repositioning as a pharmacological treatment for avascular osteonecrosis. After incubation with 821 drugs from the Food and Drug Administration (FDA)-approved drug library, Ang1 expression in U2OS cell culture media was examined by ELISA. Metformin, the first-line medication for treatment of type 2 diabetes, was selected as a candidate for in vitro and in vivo experimental evaluation. Ang1 was induced, and alkaline phosphatase activity was increased by metformin treatment in U2OS and MG63 cells. Wound healing and migration assay showed increased osteoblastic cell mobility by metformin treatment in U2OS and MG63 cells. Metformin upregulated expression of protein markers for osteoblastic differentiation in U2OS and MG63 cells but inhibited osteoclastic differentiation in Raw264.7 cells. Metformin (25 mg/kg) protected against ischemic necrosis in the epiphysis of the rat femoral head by maintaining osteoblast/osteocyte function and vascular density but inhibiting osteoclast activity in the necrotic femoral head. These findings provide novel insight into the specific biomarkers that are targeted and regulated by metformin in osteoblast differentiation and contribute to understanding the effects of these FDA-approved small-molecule drugs as novel therapeutics for ischemic osteonecrosis.


Asunto(s)
Inductores de la Angiogénesis/administración & dosificación , Diferenciación Celular/efectos de los fármacos , Isquemia/fisiopatología , Metformina/administración & dosificación , Osteoblastos/efectos de los fármacos , Osteoclastos/efectos de los fármacos , Osteonecrosis/fisiopatología , Angiopoyetina 1/metabolismo , Angiopoyetina 1/fisiología , Animales , Línea Celular Tumoral , Cabeza Femoral/irrigación sanguínea , Cabeza Femoral/fisiopatología , Humanos , Isquemia/complicaciones , Masculino , Osteonecrosis/complicaciones , Ratas Sprague-Dawley
20.
Int J Pharm ; 576: 118997, 2020 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-31893542

RESUMEN

Cardiovascular diseases (CVD) are the leading cause of death worldwide. Growth factor therapy has emerged as novel therapeutic strategy under investigation for CVD. In this sense, adrenomedullin-2 (ADM-2) has been recently identified as a new angiogenic factor able to regulate the regional blood flow and cardiovascular function. However, the therapeutic value of ADM-2 is limited by its short biological half-life and low plasma stability. Poly (lactic-co-glycolic acid) (PLGA) micro- and nanoparticles have been investigated as growth factor delivery systems for cardiac repair. In this study, we aimed to develop PLGA nanoparticles containing ADM-2 intended for therapeutic angiogenesis. PLGA nanoparticles containing ADM-2 were prepared by a double emulsion modified method, resulting in 300 nm-sized stable particles with zeta potential around - 30 mV. Electron microscopy analysis by SEM and TEM revealed spherical particles with a smooth surface. High encapsulation efficiency was reached (ca.70%), as quantified by ELISA. ADM-2 associated to polymer nanoparticles was also determined by EDS elemental composition analysis, SDS-PAGE and LC-MS/MS for peptide identification. In vitro release assays showed the sustained release of ADM-2 from polymer nanoparticles for 21 days. Cell viability experiments were performed in J774 macrophages and H9c2 cardiomyocyte cells, about which PLGA nanoparticles loaded with ADM-2 did not cause toxicity in the range 0.01-1 mg/ml. Of note, encapsulated ADM-2 significantly induced cell proliferation in EA.hy926 endothelial cells, indicating the ADM-2 bioactivity was preserved after the encapsulation process. Collectively, these results demonstrate the feasibility of using PLGA nanoparticles as delivery systems for the angiogenic peptide ADM-2, which could represent a novel approach for therapeutic angiogenesis in CVD using growth factor therapy.


Asunto(s)
Inductores de la Angiogénesis/administración & dosificación , Proliferación Celular/efectos de los fármacos , Portadores de Fármacos , Células Endoteliales/efectos de los fármacos , Hormonas Peptídicas/administración & dosificación , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Inductores de la Angiogénesis/química , Inductores de la Angiogénesis/toxicidad , Animales , Línea Celular , Preparaciones de Acción Retardada , Composición de Medicamentos , Liberación de Fármacos , Humanos , Cinética , Ratones , Nanopartículas , Hormonas Peptídicas/química , Hormonas Peptídicas/toxicidad , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/toxicidad , Ratas , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/química , Solubilidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...