Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
1.
Biomater Adv ; 158: 213761, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38281321

RESUMEN

Laminins are essential in basement membrane architecture and critical in re-epithelialization and angiogenesis. These processes and collagen deposition are vital in skin wound healing. The role of angiogenic peptides in accelerating the wound-healing process has been known. The bioactive peptides could be a potential approach due to their similar effects as growth factors and inherent biocompatible and biodegradable nature with lower cost. They can also recognize ligand-receptor interaction and mimic the extracellular matrix. Here, we report novel angiogenic DYVRLAI, CDYVRLAI, angiogenic-collagen PGPIKVAV, and Ac-PGPIKVAV peptides conjugated sodium carboxymethyl cellulose hydrogel, which was designed from laminin. The designed peptide exhibits a better binding with the α3ß1, αvß3, and α5ß1 integrins and CXCR2 receptor, indicating their angiogenic and collagen binding efficiency. The peptides were evaluated to stimulate wound healing in full-thickness excision wounds in normal and diabetic mice (type II). They demonstrated their efficacy in terms of angiogenesis (CD31), re-epithelialization through regeneration of the epidermis (H&E), and collagen deposition (MT). The synthesized peptide hydrogel (DYVRLAI and CDYVRLAI) showed enhanced wound contraction up to 10.1 % and 12.3 % on day 7th compared to standard becaplermin gel (49 %) in a normal wound model. The encouraging results were also observed with the diabetic model, where these peptides showed a significant decrease of 5.20 and 5.17 % in wound size on day 10th compared to the commercial gel (9.27 %). These outcomes signify that the modified angiogenic peptide is a cost effective, novel peptide motif to promote dermal wound healing in both models.


Asunto(s)
Diabetes Mellitus Experimental , Laminina , Animales , Ratones , Laminina/farmacología , Hidrogeles/farmacología , Colágeno/farmacología , Péptidos/farmacología , Péptidos/uso terapéutico , Cicatrización de Heridas , Proteínas Angiogénicas/farmacología , Integrina alfa5beta1
2.
Daru ; 30(2): 273-288, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35925539

RESUMEN

BACKGROUND: Breast cancer is currently the world's most predominant malignancy. In cancer progression, angiogenesis is a requirement for tumor growth and metastasis.Alpinumisoflavone (AIF), a bioactive isoflavonoid, exhibited good binding affinity with the angiogenesis pathway's druggable target through molecular docking. OBJECTIVES: To confirm AIF's angiogenesis inhibitory activity, cytotoxic potential toward breast cancer cells, and druggability. METHODS: Antiangiogenic activity was evaluated in six pro-angiogenic proteins in vitro, duck chorioallantoic membrane (CAM) in ovo, molecular docking and druggability in silico. RESULTS: Findings showed that AIF significantly inhibited (p = < 0.001) the HER2(IC50 = 2.96 µM), VEGFR-2(IC50 = 4.80 µM), MMP-9(IC50 = 23.00 µM), FGFR4(IC50 = 57.65 µM), EGFR(IC50 = 92.06 µM) and RET(IC50 = > 200 µM) activity in vitro.AIF at 25 µM-200 µM significantly inhibited (p = < 0.001) the total number of branch points (IC50 = 14.25 µM) and mean length of tubule complexes (IC50 = 3.52 µM) of duck CAM comparable (p = > 0.001) with the positive control 200 µM celecoxib on both parameters.AIF inhibited the growth of the estrogen-receptor-positive (ER +) human breast cancer cells (MCF-7) by 44.92 ± 1.79% at 100 µM while presenting less toxicity to human dermal fibroblast neonatal (HDFn) normal cells.The positive control 100 µM doxorubicin showed 86.66 ± 0.93% and 92.97 ± 1.27% inhibition with MCF-7 (IC50 = 3.62 µM) and HDFn, (IC50 = 27.16 µM) respectively.In docking, AIF has the greatest in silico binding affinity on HER2 (-10.9 kcal/mol) among the key angiogenic molecules tested. In silico rat oral LD50 calculation indicates that AIF is moderate to slightly toxic at 146.4 mg/kg with 1.1 g/kg and 20.1 mg/kg upper and lower 95% confidence limits. Lastly, it sufficiently complies with Lipinski's, Veber's, Egan's, Ghose's, and Muegge's Rule, supporting its oral drug-like property. CONCLUSION: This study revealed that AIF possesses characteristics of a phytoestrogen compound with significant binding affinity, inhibitory activity against pro-angiogenic proteins, and cytotoxic potential against ER + breast cancer cells.The acceptable and considerable safety and drug-likeness profiles of AIF are worthy of further confirmation in vivo and advanced pre-clinical studies so that AIF can be elevated as a promising molecule for breast cancer therapy.


Asunto(s)
Antineoplásicos , Neoplasias de la Mama , Humanos , Ratas , Animales , Femenino , Simulación del Acoplamiento Molecular , Proliferación Celular , Antineoplásicos/química , Neoplasias de la Mama/tratamiento farmacológico , Proteínas Angiogénicas/farmacología , Relación Estructura-Actividad , Estructura Molecular , Ensayos de Selección de Medicamentos Antitumorales , Inhibidores de Proteínas Quinasas/química , Relación Dosis-Respuesta a Droga
3.
Arterioscler Thromb Vasc Biol ; 41(11): 2756-2769, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34551592

RESUMEN

Objective: Angiogenic factor AGGF1 (angiogenic factor with G-patch and FHA [Forkhead-associated] domain 1) promotes angiogenesis as potently as VEGFA (vascular endothelial growth factor A) and regulates endothelial cell (EC) proliferation, migration, specification of multipotent hemangioblasts and venous ECs, hematopoiesis, and vascular development and causes vascular disease Klippel-Trenaunay syndrome when mutated. However, the receptor for AGGF1 and the underlying molecular mechanisms remain to be defined. Approach and Results: Using functional blocking studies with neutralizing antibodies, we identified [alpha]5[beta]1 as the receptor for AGGF1 on ECs. AGGF1 interacts with [alpha]5[beta]1 and activates FAK (focal adhesion kinase), Src (proto-oncogene tyrosine-protein kinase), and AKT (protein kinase B). Functional analysis of 12 serial N-terminal deletions and 13 C-terminal deletions by every 50 amino acids mapped the angiogenic domain of AGGF1 to a domain between amino acids 604-613 (FQRDDAPAS). The angiogenic domain is required for EC adhesion and migration, capillary tube formation, and AKT activation. The deletion of the angiogenic domain eliminated the effects of AGGF1 on therapeutic angiogenesis and increased blood flow in a mouse model for peripheral artery disease. A 40-mer or 15-mer peptide containing the angiogenic domain blocks AGGF1 function, however, a 15-mer peptide containing a single amino acid mutation from -RDD- to -RGD- (a classical RGD integrin-binding motif) failed to block AGGF1 function. Conclusions: We have identified integrin [alpha]5[beta]1 as an EC receptor for AGGF1 and a novel AGGF1-mediated signaling pathway of [alpha]5[beta]1-FAK-Src-AKT for angiogenesis. Our results identify an FQRDDAPAS angiogenic domain of AGGF1 crucial for its interaction with [alpha]5[beta]1 and signaling.


Asunto(s)
Proteínas Angiogénicas/metabolismo , Células Endoteliales/metabolismo , Miembro Posterior/irrigación sanguínea , Integrina alfa5beta1/metabolismo , Isquemia/metabolismo , Neovascularización Fisiológica , Células 3T3-L1 , Inductores de la Angiogénesis/farmacología , Proteínas Angiogénicas/genética , Proteínas Angiogénicas/farmacología , Animales , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Femenino , Quinasa 1 de Adhesión Focal/metabolismo , Células HEK293 , Células HeLa , Células Hep G2 , Humanos , Integrina alfa5beta1/genética , Isquemia/tratamiento farmacológico , Isquemia/genética , Isquemia/fisiopatología , Ligandos , Masculino , Ratones , Ratones Endogámicos C57BL , Neovascularización Fisiológica/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Fosforilación , Dominios y Motivos de Interacción de Proteínas , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Transducción de Señal , Familia-src Quinasas/metabolismo
4.
Theranostics ; 11(11): 5107-5126, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33859737

RESUMEN

Background: Pathological angiogenesis is the hallmark of many vision-threatening diseases. Anti-VEGF is a primary treatment with substantial beneficial effects. However, such agents require frequent intravitreal injections. Our previous work established a method for effectively modifying exosomes (EXOs) for loading therapeutic peptides. Here, we used this system to load the anti-angiogenic peptide KV11, aiming to establish an EXO-based therapy strategy to suppress neovascularization in the retina. Methods: Using an anchoring peptide, CP05, we linked KV11 to endothelial cell (EC) derived EXOs, yielding EXOKV11. We tested the delivery efficiency of EXOKV11 via two commonly used ocular injection methods: retro-orbital injection and intravitreal injection. Deploying an oxygen-induced retinopathy (OIR) model and a VEGF injection model, we tested the effects of EXOKV11 on neovascular formation, EC proliferation, and vascular permeability. In vitro experiments were used to test the mechanism and to analyze the effects of EXOKV11 on EC proliferation, migration, and sprouting. Results: By using the EXO loading system, KV11 was more efficiently delivered to the blood vessels of the mouse retina via retro-orbital injection. In both OIR model and VEGF injection model, EXOKV11 was more effective than KV11 alone in inhibiting neovascularization and vessel leakage. The therapeutic effect of retro-orbital injection of EXOKV11 was comparable to the intravitreal injection of VEGF-trap. Mechanistically, KV11 alone inhibited VEGF-downstream signaling, while EXOKV11 showed a stronger effect. Conclusions: We used EXOs as a carrier for intraocular delivery of KV11. We showed that KV11 itself has an anti-angiogenic effect through retro-orbital injection, but that this effect was greatly enhanced when delivered with EXOs. Thus, this system has the potential to treat proliferative retinopathy via retro-orbital injection which is a less invasive manner compared with intravitreal injection.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Proteínas Angiogénicas/farmacología , Exosomas/efectos de los fármacos , Retina/efectos de los fármacos , Neovascularización Retiniana/tratamiento farmacológico , Animales , Permeabilidad Capilar/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Inyecciones Intravítreas/métodos , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Oxígeno/farmacología , Fragmentos de Péptidos/metabolismo , Ratas , Ratas Sprague-Dawley , Retina/metabolismo , Neovascularización Retiniana/metabolismo , Transducción de Señal/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo
5.
Arterioscler Thromb Vasc Biol ; 40(11): 2632-2648, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32814441

RESUMEN

OBJECTIVE: We sought to identify and investigate the functional role of the major endothelial cell (EC)-derived factors that control pericyte recruitment to EC tubes and pericyte-induced tube maturation during capillary network formation. Approach and Results: We identify PDGF (platelet-derived growth factor)-BB, PDGF-DD, ET (endothelin)-1, TGF (transforming growth factor)-ß, and HB-EGF (heparin-binding epidermal growth factor), as the key individual and combined regulators of pericyte assembly around EC tubes. Using novel pericyte only assays, we demonstrate that PDGF-BB, PDGF-DD, and ET-1 are the primary direct drivers of pericyte invasion. Their addition to pericytes induces invasion as if ECs were present. In contrast, TGF-ß and HB-EGF have minimal ability to directly stimulate pericyte invasion. In contrast, TGF-ß1 can act as an upstream pericyte primer to stimulate invasion in response to PDGFs and ET-1. HB-EGF stimulates pericyte proliferation along with PDGFs and ET-1. Using EC-pericyte cocultures, individual, or combined blockade of these EC-derived factors, or their pericyte receptors, using neutralizing antibodies or chemical inhibitors, respectively, interferes with pericyte recruitment and proliferation. As individual factors, PDGF-BB and ET-1 have the strongest impact on these events. However, when the blocking reagents are combined to interfere with each of the above factors or their receptors, more dramatic and profound blockade of pericyte recruitment, proliferation, and pericyte-induced basement membrane deposition occurs. Under these conditions, ECs form tubes that become much wider and less elongated as if pericytes were absent. CONCLUSIONS: Overall, these new studies define and characterize a functional role for key EC-derived factors controlling pericyte recruitment, proliferation, and pericyte-induced basement membrane deposition during capillary network assembly.


Asunto(s)
Proteínas Angiogénicas/metabolismo , Encéfalo/irrigación sanguínea , Capilares/metabolismo , Movimiento Celular , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Neovascularización Fisiológica , Comunicación Paracrina , Pericitos/metabolismo , Proteínas Angiogénicas/farmacología , Becaplermina/metabolismo , Capilares/citología , Capilares/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Endotelina-1/metabolismo , Factor de Crecimiento Similar a EGF de Unión a Heparina/metabolismo , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Linfocinas/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Comunicación Paracrina/efectos de los fármacos , Pericitos/efectos de los fármacos , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo
6.
Esophagus ; 17(3): 289-297, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-31980976

RESUMEN

BACKGROUND: Vasohibins (VASH), which are angiogenesis regulators, consist of Vasohibin-1 (VASH1) and Vasohibin-2 (VASH2). VASH1 is an angiogenesis inhibitor, while VASH2 is a proangiogenic factor. Patients with esophageal squamous cell carcinoma (ESCC) with high tumor expression levels of VASH1 and VASH2 have been reported to show a poor prognosis. The clinical significance of VASH concentrations in the blood of patients with ESCC has not yet been investigated. METHODS: Plasma samples from 89 patients with ESCC were analyzed, and the relationships between the plasma VASH concentrations and the clinicopathological factors of the patients were evaluated. Immunohistochemical examination (IHC) of the resected tumor specimens for VASH was performed in 56 patients, and the correlation between the plasma VASH concentrations and tumor expression levels of VASH was analyzed. RESULTS: The patient group with high plasma concentrations of VASH1 showed a higher frequency of lymph node metastasis (P = 0.01) and an invasive growth pattern (P = 0.05). Furthermore, poorly differentiated cancer occurred at a higher frequency in the patient group with high plasma concentrations of VASH2 (P < 0.01). High tumor expression levels of VASH1 were encountered more frequently in the patient group with high plasma concentrations of VASH1 (P = 0.03), and high tumor expression levels of VASH2 were encountered more frequently in the patient group with high plasma concentrations of VASH2 (P = 0.04). CONCLUSIONS: In patients with ESCC, high plasma concentrations were associated with poor clinical outcomes for both VASH1 and VASH2. We propose that results indicate that plasma VASH1 and VASH2 are useful biomarkers in patients with ESCC.


Asunto(s)
Proteínas Angiogénicas/sangre , Proteínas de Ciclo Celular/sangre , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago/metabolismo , Carcinoma de Células Escamosas de Esófago/mortalidad , Anciano , Inductores de la Angiogénesis/sangre , Inductores de la Angiogénesis/farmacología , Inhibidores de la Angiogénesis/sangre , Inhibidores de la Angiogénesis/farmacología , Proteínas Angiogénicas/farmacología , Biomarcadores/sangre , Estudios de Casos y Controles , Proteínas de Ciclo Celular/farmacología , Diferenciación Celular , Carcinoma de Células Escamosas de Esófago/diagnóstico , Carcinoma de Células Escamosas de Esófago/cirugía , Femenino , Humanos , Inmunohistoquímica/métodos , Metástasis Linfática/patología , Masculino , Persona de Mediana Edad , Invasividad Neoplásica/patología , Estadificación de Neoplasias/métodos , Pronóstico , Estudios Prospectivos
7.
J Biomed Mater Res A ; 107(12): 2764-2773, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31408258

RESUMEN

Angiogenesis plays a pivotal role in tissue regeneration following bone-grafting procedures; however, nonautogenous graft materials typically lack critical angiogenic growth factors. While much research has focused on modifying grafts with angiogenic factors, controlled delivery of these molecules remains a challenge. The current study describes a method for sustained delivery of an angiogenic peptide from hydroxyapatite (HA), a common alloplast material. Specifically, VEGF-derived "QK" peptides were synthesized with polyglutamate domains containing varying numbers of glutamates. The rate of peptide release from HA inversely correlated with glutamate number, with diglutamate-QK (E2-QK) released first, followed by tetraglutamate-QK (E4-QK), and finally, heptaglutamate-QK (E7-QK). By coating HA with a mixture of these peptides, termed, PGM-QK (polyglutamate-modified mixture), sequential peptide release was achieved, enabling gradient QK delivery. To evaluate bioactivity, HA disks were coated with PGM-QK and then placed in fresh media for 6 days. Media containing the released peptides was collected at varying time intervals and placed on human umbilical vein endothelial cells (HUVECs). Cells were evaluated for activation of angiogenic signaling pathways (ERK and Akt) and cell migration. Results showed that QK peptides were continuously released over the 6-day interval, and maintained their capacity to activate HUVECs. These findings point to a new approach for gradient delivery of an angiogenic stimulus.


Asunto(s)
Inductores de la Angiogénesis/administración & dosificación , Proteínas Angiogénicas/administración & dosificación , Sustitutos de Huesos/química , Preparaciones de Acción Retardada/química , Durapatita/química , Ácido Poliglutámico/administración & dosificación , Inductores de la Angiogénesis/química , Inductores de la Angiogénesis/farmacología , Proteínas Angiogénicas/química , Proteínas Angiogénicas/farmacología , Liberación de Fármacos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Neovascularización Fisiológica/efectos de los fármacos , Ácido Poliglutámico/química , Ácido Poliglutámico/farmacología , Factor A de Crecimiento Endotelial Vascular/administración & dosificación , Factor A de Crecimiento Endotelial Vascular/química , Factor A de Crecimiento Endotelial Vascular/farmacología
8.
Int J Mol Sci ; 20(12)2019 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-31212918

RESUMEN

Cancer remains one of the major causes of death worldwide. Angiogenesis is crucial for the pathogenesis of various human diseases, especially solid tumors. The discovery of anti-angiogenic peptides is a promising therapeutic route for cancer treatment. Thus, reliably identifying anti-angiogenic peptides is extremely important for understanding their biophysical and biochemical properties that serve as the basis for the discovery of new anti-cancer drugs. This study aims to develop an efficient and interpretable computational model called TargetAntiAngio for predicting and characterizing anti-angiogenic peptides. TargetAntiAngio was developed using the random forest classifier in conjunction with various classes of peptide features. It was observed via an independent validation test that TargetAntiAngio can identify anti-angiogenic peptides with an average accuracy of 77.50% on an objective benchmark dataset. Comparisons demonstrated that TargetAntiAngio is superior to other existing methods. In addition, results revealed the following important characteristics of anti-angiogenic peptides: (i) disulfide bond forming Cys residues play an important role for inhibiting blood vessel proliferation; (ii) Cys located at the C-terminal domain can decrease endothelial formatting activity and suppress tumor growth; and (iii) Cyclic disulfide-rich peptides contribute to the inhibition of angiogenesis and cell migration, selectivity and stability. Finally, for the convenience of experimental scientists, the TargetAntiAngio web server was established and made freely available online.


Asunto(s)
Inhibidores de la Angiogénesis/química , Inhibidores de la Angiogénesis/farmacología , Proteínas Angiogénicas/química , Proteínas Angiogénicas/farmacología , Biología Computacional/métodos , Diseño de Fármacos , Programas Informáticos , Secuencia de Aminoácidos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Humanos , Aprendizaje Automático , Conformación Molecular , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Reproducibilidad de los Resultados , Relación Estructura-Actividad
9.
Stem Cell Res Ther ; 8(1): 219, 2017 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-28974256

RESUMEN

BACKGROUND: The therapeutic potential of mesenchymal stem cells (MSCs) may be attributed partly to humoral factors such as growth factors, cytokines, and chemokines. Human term placental tissue-derived MSCs (PlaMSCs), or conditioned medium left over from cultures of these cells, have been reported to enhance angiogenesis. Recently, the exosome, which can transport a diverse suite of macromolecules, has gained attention as a novel intercellular communication tool. However, the potential role of the exosome in PlaMSC therapeutic action is not well understood. The purpose of this study was to evaluate PlaMSC-derived exosome angiogenesis promotion in vitro and in vivo. METHODS: MSCs were isolated from human term placental tissue by enzymatic digestion. Conditioned medium was collected after 48-h incubation in serum-free medium (PlaMSC-CM). Angiogenic factors present in PlaMSC-CM were screened by a growth factor array. Exosomes were prepared by ultracentrifugation of PlaMSC-CM, and confirmed by transmission electron microscopy, dynamic light scattering, and western blot analyses. The proangiogenic activity of PlaMSC-derived exosomes (PlaMSC-exo) was assessed using an endothelial tube formation assay, a cell migration assay, and reverse transcription-PCR analysis. The in-vivo angiogenic activity of PlaMSC-exo was evaluated using a murine auricle ischemic injury model. RESULTS: PlaMSC-CM contained both angiogenic and angiostatic factors, which enhanced endothelial tube formation. PlaMSC-exo were incorporated into endothelial cells; these exosomes stimulated both endothelial tube formation and migration, and enhanced angiogenesis-related gene expression. Laser Doppler blood flow analysis showed that PlaMSC-exo infusion also enhanced angiogenesis in an in-vivo murine auricle ischemic injury model. CONCLUSIONS: PlaMSC-exo enhanced angiogenesis in vitro and in vivo, suggesting that exosomes play a role in the proangiogenic activity of PlaMSCs. PlaMSC-exo may be a novel therapeutic approach for treating ischemic diseases.


Asunto(s)
Proteínas Angiogénicas/farmacología , Pabellón Auricular/efectos de los fármacos , Exosomas/trasplante , Neovascularización Fisiológica/efectos de los fármacos , Placenta/citología , Daño por Reperfusión/terapia , Proteínas Angiogénicas/aislamiento & purificación , Animales , Bioensayo , Movimiento Celular , Medios de Cultivo Condicionados/química , Medio de Cultivo Libre de Suero , Pabellón Auricular/irrigación sanguínea , Pabellón Auricular/lesiones , Pabellón Auricular/patología , Exosomas/química , Femenino , Humanos , Péptidos y Proteínas de Señalización Intercelular/farmacología , Masculino , Células Madre Mesenquimatosas/química , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Desnudos , Placenta/metabolismo , Embarazo , Cultivo Primario de Células , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología
10.
Peptides ; 97: 70-78, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28987278

RESUMEN

In this study, we hydrolyzed rice endosperm protein (REP) with pepsin and generated 20 fractions containing multifunctional cationic peptides with varying isoelectric point (pI) values using ampholyte-free isoelectric focusing (autofocusing). Subsequently, we determined antimicrobial activities of each fraction against the pathogens Prophyromonas gingivalis, Propionibacterium acnes, Streptocossus mutans, and Candida albicans. Fractions 18, 19, and 20 had pI values greater than 12 and exhibited antimicrobial activity against P. gingivalis, P. acnes, and C. albicans, but not against S. mutans. In further experiments, we purified and identified cationic peptides from fractions 18, 19, and 20 using reversed-phase high-performance liquid chromatography and matrix-assisted laser/desorption ionization-time-of-flight mass spectroscopy. We also chemically synthesized five identified peptides (RSVSKSR, RRVIEPR, ERFQPMFRRPG, RVRQNIDNPNRADTYNPRAG, and VVRRVIEPRGLL) with pI values greater than 10.5 and evaluated antimicrobial, lipopolysaccharide (LPS)-neutralizing, and angiogenic activities. Among these synthetic peptides, only VVRRVIEPRGLL exhibited antimicrobial activity against P. gingivalis, with an IC50 value of 87µM. However, all five cationic peptides exhibited LPS-neutralizing and angiogenic activities with little or no hemolytic activity against mammalian red blood cells at functional concentrations. These present data show dual or multiple functions of the five identified cationic peptides with little or no hemolytic activity. Therefore, fractions containing cationic peptides from REP hydrolysates have the potential to be used as dietary supplements and functional ingredients in food products.


Asunto(s)
Proteínas Angiogénicas/química , Antiinfecciosos/química , Péptidos Catiónicos Antimicrobianos/química , Endospermo/química , Oryza/química , Proteínas de Plantas/química , Proteínas Angiogénicas/farmacología , Animales , Antiinfecciosos/farmacología , Antifúngicos/farmacología , Péptidos Catiónicos Antimicrobianos/síntesis química , Bacterias/efectos de los fármacos , Hongos/efectos de los fármacos , Hemólisis/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Hidrólisis , Concentración 50 Inhibidora , Lipopolisacáridos
11.
Stem Cell Res Ther ; 8(1): 212, 2017 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-28969687

RESUMEN

BACKGROUND: The main goal of bone tissue engineering has been the generation of healthy bone in order to replace affected tissue. Therefore, optimized biomaterials are needed which allow the survival and growth of mesenchymal stem cells. Until now the key challenge in the clinical application of cell-based tissue engineering bone implants was poor diffusion of oxygen into the tissue, making functional blood vessel networks a necessity. With their ability to evolve into different cell types, to expand extensively in vitro, and to release paracrine soluble factors, bone marrow stromal cells (BMSC) are highly attractive for tissue engineering. During the last years hypoxia became a proven method to control proliferation, differentiation, and pluripotency of BMSC. Here we applied different methods to characterize metabolically conditioned media (MCM) in comparison to hypoxia conditioned media (HCM) and evaluated their ability to attract BMSC in 2-D migration assays. METHODS: BMSC and fibroblasts of human origin were isolated and cultivated to obtain HCM and MCM. Both media were characterized by angiogenesis arrays, cytokine arrays, and ELISA for selected factors. 2-D migration tests were performed with Corning Transwell®-96 permeable support chambers with porous polyester membranes with a pore size of 8.0 µm. RESULTS: Characterization of HCM and MCM revealed that the concentration of angiogenic factors was higher in MCM than in HCM. However, the chemoattractive capacity of MCM for BMSC was equivalent to that of HCM. HCM and MCM produced by human skin fibroblasts attracted human BMSC as efficiently as HCM and MCM produced by human BMSC. CONCLUSIONS: HCM and MCM have a high chemoattractive capacity for BMSC. Both conditioned media harbor high concentrations of angiogenic factors which are important for angiogenesis and cell migration. Both chemoattracting conditioned media can also be derived from skin fibroblasts which can easily be obtained from patients in individualized therapy approaches.


Asunto(s)
Proteínas Angiogénicas/farmacología , Células de la Médula Ósea/metabolismo , Factores Quimiotácticos/farmacología , Medios de Cultivo Condicionados/química , Fibroblastos/metabolismo , Células Madre Mesenquimatosas/efectos de los fármacos , Proteínas Angiogénicas/biosíntesis , Proteínas Angiogénicas/aislamiento & purificación , Proteínas Angiogénicas/metabolismo , Bioensayo , Células de la Médula Ósea/citología , Hipoxia de la Célula , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Factores Quimiotácticos/biosíntesis , Factores Quimiotácticos/aislamiento & purificación , Factores Quimiotácticos/metabolismo , Quimiotaxis/efectos de los fármacos , Quimiotaxis/fisiología , Medios de Cultivo Condicionados/farmacología , Cámaras de Difusión de Cultivos , Fibroblastos/citología , Prepucio/citología , Prepucio/metabolismo , Humanos , Masculino , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/fisiología , Neovascularización Fisiológica/efectos de los fármacos , Cultivo Primario de Células
12.
Adv Gerontol ; 30(5): 671-675, 2017.
Artículo en Ruso | MEDLINE | ID: mdl-29322732

RESUMEN

We investigated the influence of vascular peptide bioregulator on microcirculation in the brain cortex of spontaneously hypertensive rats of different ages and figured out whether there is functional cumulation during two-time application of the drug «Slavinorm¼ by above-mentioned animals. It was shown that a single course treatment with vascular peptide bioregulator had increased the density of microvascular network of the pia mater in young animals ca. 1,2-fold and had not affected the perfusion and oxygen saturation of sensorimotor cortex. The second course treatment with «Slavinorm¼ was provided in a 6 months. Functional cumulation was revealed in 12 month-aged rats which had 2 course treatments with vascular peptide bioregulation: the density of microvascular network of the pia mater was increased ca. 1,6-fold; level of perfusion was increased ca. 15% in comparison with intact animals of the same age. These animals were more tolerant to cerebral vasospasm (the application of vasoconstrictor on the surface of the brain): the highest level of tissue oxygen saturation was remained at fairly constant perfusion in comparison with other animals.


Asunto(s)
Proteínas Angiogénicas/farmacología , Corteza Cerebral/irrigación sanguínea , Microcirculación/fisiología , Microvasos/efectos de los fármacos , Factores de Edad , Animales , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY
13.
PLoS Biol ; 14(8): e1002529, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27513923

RESUMEN

AGGF1 is an angiogenic factor with therapeutic potential to treat coronary artery disease (CAD) and myocardial infarction (MI). However, the underlying mechanism for AGGF1-mediated therapeutic angiogenesis is unknown. Here, we show for the first time that AGGF1 activates autophagy, a housekeeping catabolic cellular process, in endothelial cells (ECs), HL1, H9C2, and vascular smooth muscle cells. Studies with Atg5 small interfering RNA (siRNA) and the autophagy inhibitors bafilomycin A1 (Baf) and chloroquine demonstrate that autophagy is required for AGGF1-mediated EC proliferation, migration, capillary tube formation, and aortic ring-based angiogenesis. Aggf1+/- knockout (KO) mice show reduced autophagy, which was associated with inhibition of angiogenesis, larger infarct areas, and contractile dysfunction after MI. Protein therapy with AGGF1 leads to robust recovery of myocardial function and contraction with increased survival, increased ejection fraction, reduction of infarct areas, and inhibition of cardiac apoptosis and fibrosis by promoting therapeutic angiogenesis in mice with MI. Inhibition of autophagy in mice by bafilomycin A1 or in Becn1+/- and Atg5 KO mice eliminates AGGF1-mediated angiogenesis and therapeutic actions, indicating that autophagy acts upstream of and is essential for angiogenesis. Mechanistically, AGGF1 initiates autophagy by activating JNK, which leads to activation of Vps34 lipid kinase and the assembly of Becn1-Vps34-Atg14 complex involved in the initiation of autophagy. Our data demonstrate that (1) autophagy is essential for effective therapeutic angiogenesis to treat CAD and MI; (2) AGGF1 is critical to induction of autophagy; and (3) AGGF1 is a novel agent for treatment of CAD and MI. Our data suggest that maintaining or increasing autophagy is a highly innovative strategy to robustly boost the efficacy of therapeutic angiogenesis.


Asunto(s)
Proteínas Angiogénicas/metabolismo , Autofagia/fisiología , Cardiopatías/metabolismo , Neovascularización Patológica/metabolismo , Proteínas Angiogénicas/genética , Proteínas Angiogénicas/farmacología , Animales , Autofagia/efectos de los fármacos , Autofagia/genética , Proteína 5 Relacionada con la Autofagia/genética , Proteína 5 Relacionada con la Autofagia/metabolismo , Beclina-1/genética , Beclina-1/metabolismo , Western Blotting , Línea Celular , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Cardiopatías/tratamiento farmacológico , Cardiopatías/genética , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Macrólidos/farmacología , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/citología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/genética , Neovascularización Fisiológica/efectos de los fármacos , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología
14.
Nucl Med Biol ; 43(9): 552-558, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27395415

RESUMEN

INTRODUCTION: Although liposomes hold promise for cancer therapy, the effectiveness of treating myocardial ischemia by promoting angiogenesis has yet to be proved. Nanoliposomes loaded with therapeutic agents can effectively target ischemic myocardium via enhanced permeability and retention. Surface polyethylene glycol (PEG) modification can further facilitate effective targeting by prolonging liposomal circulation. This study aimed to determine whether PEGylated nanoliposomes are effective in facilitating targeted drug delivery and treating myocardial ischemia. METHODS: Rats subjected to 30min of myocardial ischemia were given (99m)Tc-hexamethylpropyleneamine oxime- or (99m)Tc-diethylenetriamine pentaacetate-labeled liposomes with mean diameters of ~100nm or ~600nm with or without PEG modifications to determine the extent of myocardial uptake in the different conditions. Therapeutic effectiveness was assessed by studying changes in myocardial perfusion defects with (99m)Tc-tetrofosmin autoradiography and vascular density with immunohistochemistry at 7days post-treatment. RESULTS: The liver and spleen showed the largest capacity for liposome uptake. Uptake by the liver and spleen was more pronounced when the liposomes were larger. Conversely, myocardial liposome uptake was significantly greater when the liposomes were ~100nm rather than ~600nm in diameter. Surface modification with PEG significantly augmented myocardial uptake of ~100nm liposomes. PEG modification did not affect the size dependence. To investigate therapeutic efficacy, hearts subjected to ischemia received PEGylated nanoliposomes encapsulated with angiogenic peptides. Our data demonstrated that PEGylated nanoliposomes loaded with angiogenic peptides improved myocardial perfusion defects and increased vascular density. A 10-fold increase in liposomal concentration did not further benefit myocardial ischemia. CONCLUSIONS: Liposomal angiogenic formulation with size control and PEG modification may be effective treatment strategy for myocardial ischemia. Increasing the concentration of liposomes does not necessarily benefit myocardial ischemia.


Asunto(s)
Proteínas Angiogénicas/administración & dosificación , Proteínas Angiogénicas/farmacología , Circulación Coronaria/efectos de los fármacos , Isquemia Miocárdica/diagnóstico por imagen , Isquemia Miocárdica/fisiopatología , Polietilenglicoles/química , Proteínas Angiogénicas/uso terapéutico , Animales , Cápsulas , Relación Dosis-Respuesta a Droga , Liposomas , Masculino , Isquemia Miocárdica/tratamiento farmacológico , Isquemia Miocárdica/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Compuestos Organofosforados/metabolismo , Compuestos Organofosforados/farmacocinética , Compuestos de Organotecnecio/metabolismo , Compuestos de Organotecnecio/farmacocinética , Cintigrafía , Ratas , Ratas Sprague-Dawley , Distribución Tisular
15.
Adv Gerontol ; 29(2): 291-296, 2016.
Artículo en Ruso | MEDLINE | ID: mdl-28514548

RESUMEN

Using a TV device to study brain microcirculation, we found that after a course of vascular peptide bioregulator the density of microvascular network of pia matter of old rats (22-24 months) sensomotor cortex increased about 2,5-2,8 times compared to control old rates; and noradrenaline-induced constriction reactions and acetylcholine-induced dilative reactions of the pial arterioles increased significantly. This perfusion in the tissue of the cerebral cortex is not increased, but the degree of blood oxygen saturation in the microvasculature of this tissue region raised.


Asunto(s)
Proteínas Angiogénicas/farmacología , Circulación Cerebrovascular , Microcirculación , Piamadre/irrigación sanguínea , Corteza Sensoriomotora , Animales , Factores Biológicos/farmacología , Circulación Cerebrovascular/efectos de los fármacos , Circulación Cerebrovascular/fisiología , Monitoreo de Drogas/métodos , Microcirculación/efectos de los fármacos , Microcirculación/fisiología , Microvasos/efectos de los fármacos , Microvasos/fisiología , Consumo de Oxígeno/efectos de los fármacos , Imagen de Perfusión/métodos , Sustancias Protectoras/farmacología , Ratas , Corteza Sensoriomotora/efectos de los fármacos , Corteza Sensoriomotora/metabolismo , Resultado del Tratamiento
16.
Arch Dermatol Res ; 307(9): 803-17, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26338700

RESUMEN

The injured skin produces a number of mediators that directly or indirectly modulate cell chemotaxis, migration, proliferation, and angiogenesis. Components of the kinin pathway including the kinin B1 receptor (B1R) have been found to occur in the human skin, but information about its role on keratinocyte biology is still scarce. Our aim was to determine whether stimulation of B1R causes the secretion of IL-4 and/or VEGF from human keratinocytes and to evaluate the role of the B1R agonist Lys-des[Arg(9)]bradykinin and IL-4 on various stages of angiogenesis, such as cell migration, proliferation, and release of metalloproteases. By using ELISA and Western blotting, we showed that HaCaT keratinocytes stimulated with the B1R agonist release IL-4 and VEGF. Stimulation of B1R also caused transient c-JunN-terminal kinase phosphorylation and JunB nuclear translocation, transcription factor that regulates IL-4 expression. The 3D-angiogenesis assay, performed on spheroids of EA.hy923 endothelial cells embedded in a collagen matrix, showed that their cumulative sprout area increased significantly following stimulation with either IL-4 or B1R agonist. Furthermore, these ligands produced significant endothelial cell migration and release of metalloproteases 2 and 9, but did not increase endothelial cell proliferation as measured by 5-bromo-2'-deoxyuridine incorporation. Our results provide experimental evidence that establishes IL-4 and B1R agonist as important angiogenic factors of relevance for skin repair.


Asunto(s)
Células Endoteliales/metabolismo , Interleucina-4/metabolismo , Interleucina-4/farmacología , Queratinocitos/metabolismo , Neovascularización Fisiológica/fisiología , Receptor de Bradiquinina B1/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteínas Angiogénicas/farmacología , Línea Celular , Movimiento Celular , Proliferación Celular , Medios de Cultivo Condicionados/farmacología , Activación Enzimática , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Calidina/análogos & derivados , Calidina/farmacología , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-jun , Interferencia de ARN , ARN Interferente Pequeño/genética , Receptor de Bradiquinina B1/agonistas , Receptor de Bradiquinina B1/genética , Transducción de Señal/fisiología , Piel/citología , Piel/metabolismo , Esferoides Celulares
17.
J Control Release ; 217: 191-201, 2015 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-26365781

RESUMEN

Therapeutic angiogenesis holds great potential for a myriad of tissue engineering and regenerative medicine approaches. While a number of peptides have been identified with pro-angiogenic behaviors, therapeutic efficacy is limited by poor tissue localization and persistence. Therefore, poly(ethylene glycol) hydrogels providing sustained, enzymatically-responsive peptide release were exploited for peptide delivery. Two pro-angiogenic peptide drugs, SPARC113 and SPARC118, from the Secreted Protein Acidic and Rich in Cysteine, were incorporated into hydrogels as crosslinking peptides flanked by matrix metalloproteinase (MMP) degradable substrates. In vitro testing confirmed peptide drug bioactivity requires sustained delivery. Furthermore, peptides retain bioactivity with residual MMP substrates present after hydrogel release. Incorporation into hydrogels achieved enzymatically-responsive bulk degradation, with peptide release in close agreement with hydrogel mass loss and released peptides retaining bioactivity. Interestingly, SPARC113 and SPARC118-releasing hydrogels had significantly different degradation time constants in vitro (1.16 and 8.77×10(-2) h(-1), respectively), despite identical MMP degradable substrates. However, upon subcutaneous implantation, both SPARC113 and SPARC118 hydrogels exhibited similar degradation constants of ~1.45×10(-2) h(-1), and resulted in significant ~1.65-fold increases in angiogenesis in vivo compared to controls. Thus, these hydrogels represent a promising pro-angiogenic approach for applications such as tissue engineering and ischemic tissue disorders.


Asunto(s)
Proteínas Angiogénicas/administración & dosificación , Portadores de Fármacos/administración & dosificación , Hidrogeles/administración & dosificación , Neovascularización Fisiológica/efectos de los fármacos , Oligopéptidos/administración & dosificación , Osteonectina/química , Proteínas Angiogénicas/farmacología , Animales , Compuestos Bicíclicos con Puentes/química , Células Cultivadas , Preparaciones de Acción Retardada/administración & dosificación , Preparaciones de Acción Retardada/farmacología , Portadores de Fármacos/farmacología , Femenino , Heptanos/química , Células Endoteliales de la Vena Umbilical Humana , Hidrogeles/farmacología , Metaloproteinasa 2 de la Matriz/química , Metaloproteinasa 2 de la Matriz/metabolismo , Ratones Endogámicos BALB C , Oligopéptidos/farmacología , Polietilenglicoles/química
18.
BMC Oral Health ; 15: 60, 2015 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-25981588

RESUMEN

BACKGROUND: Rapid wound healing of oral soft tissue may reduce the opportunity of infection and discomfort of patients. Previous studies have demonstrated that enhancement of angiogenesis is an effective way to accelerate wound repair. In this study, to enhance angiogenesis and healing of palatal wounds, dimethyloxalylglycine (DMOG) was applied to a rat palatal wound model. DMOG is known to inhibit oxygen-dependent degradation of hypoxia inducible factor-1 alpha (HIF-1α), which can lead to up-regulation of angiogenesis markers, favoring wound repair. We also evaluated the effects of DMOG on cell migration and HIF-1α expression of rat palatal (RP) cells. Furthermore, mRNA and protein expression of vascular endothelial growth factor (VEGF) were analyzed in DMOG-treated RP cells. METHODS: Primary cultures of rat palatal (RP) cells were obtained from Sprague-Dawley (SD) rats. Effects of DMOG on cell viability and migration of RP cells were evaluated by using a formazan and culture insert, respectively. VEGF mRNA was observed by real-time PCR, and VEGF and HIF-1α proteins were detected by Western blotting. For the animal study, excisional wounds, 3 mm in diameter, were made at the central part of the palate of SD rats. DMOG with hyaluronic acid ointment was topically applied three times during 1 week, and then wound closures were quantitated photographically and histologically. RESULTS: DMOG was cytotoxic to RP cells at concentrations higher than 2 mM and did not affect cell migration at non-cytotoxic concentrations. mRNA and protein expression of VEGF were significantly stimulated by DMOG treatment. The protein level of HIF-1α was also stabilized in RP cells by DMOG. In the animal study, groups treated with 1 mg/ml DMOG showed an increase of rat palatal wound contractures. CONCLUSIONS: DMOG enhanced wound healing of rat palatal mucosa, which was likely due to the angiogenic effect of the agent.


Asunto(s)
Aminoácidos Dicarboxílicos/uso terapéutico , Mucosa Bucal/lesiones , Hueso Paladar/lesiones , Proteínas Angiogénicas/farmacología , Animales , Técnicas de Cultivo de Célula , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Ácido Hialurónico/farmacología , Subunidad alfa del Factor 1 Inducible por Hipoxia/efectos de los fármacos , Masculino , Modelos Animales , Mucosa Bucal/efectos de los fármacos , Hueso Paladar/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Factor A de Crecimiento Endotelial Vascular/efectos de los fármacos , Cicatrización de Heridas/efectos de los fármacos
19.
Curr Pharm Des ; 21(21): 2993-3001, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26004411

RESUMEN

Ulcers in the stomach, duodenum, ileum/jejunum and colon may look alike grossly and microscopically, but they have very different etiologies and pathogenesis. Unfortunately, there is virtually no etiologic treatment for any of these lesions which are also accompanied by limited or extensive inflammation. This article reviews four groups of new antiulcer drugs discovered and patented in our lab in Boston and Long Beach/Irvine (Table 1). Actually, the first group, pyrazole and its derivatives can be used for prevention, i.e., long lasting protection of gastric mucosa against alcohol- or NSAID-induced erosions. Dopamine seems to be a new etiologic treatment for both upper and lower GI tract ulcers. Angiogenic growth factors like bFGF or PDGF (daily administration as peptides orally or by rectal enemas, or as single or double-dose of gene therapy) accelerated the healing of gastroduodenal ulcers and UC, while VEGF seems to be effective only for upper GI tract ulcers. Last but not least, a novel group of angiogenic steroids which not only stimulate new blood vessel formation and granulation tissue production (essential elements of healing of ulcer types) but may also exert mild or prominent antiinflammatory action and seem to be ideal drugs for the treatment of IBD.


Asunto(s)
Antiulcerosos/farmacología , Úlcera Duodenal/tratamiento farmacológico , Úlcera Péptica/tratamiento farmacológico , Proteínas Angiogénicas/farmacología , Animales , Dopamina/análogos & derivados , Dopamina/farmacología , Úlcera Duodenal/prevención & control , Humanos , Úlcera Péptica/prevención & control , Pirazoles/uso terapéutico , Úlcera Gástrica/prevención & control
20.
J Vis Exp ; (93): e51525, 2014 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-25490071

RESUMEN

The subcutaneous matrigel plug assay in mice is a method of choice for the in vivo evaluation of pro- and anti-angiogenic factors. In this method, desired factors are introduced into cold-liquid ECM-mimic gel which, after subcutaneous injection, solidifies to form an environment mimicking the cancer milieu. This matrix permits the penetration of host cells, such as endothelial cells, and therefore, the formation of vasculature. Herein we propose a new modified matrigel plug assay, which can be exploited to illustrate the angiogenic potential of a pool of factors secreted by cancer cells, as opposed to a specific factor (e.g., bFGF and VEGF) or agent. The plug containing ECM-mimic gel is utilized to introduce the host (i.e., mouse) with a pool of factors secreted to the C.M. of fast-growing tumor-generating glioblastoma cells. We have previously described an extensive comparison of the angiogenic potential of U-87 MG human glioblastoma and its dormant-derived clone, in this system model, showing induced angiogenesis in the U-87 MG parental cells. The C.M. is prepared by filtering collected media from confluent tissue culture plates of either cell line following 48 hr incubation. Hence, it contains only factors secreted by the cells, without the cells themselves. Described here is the combination of two imaging modalities, microbubbles contrast-enhanced ultrasound imaging and intravital fibered-confocal endomicroscopy, for an accurate, real-time characterization of the extent, morphology and functionality of newly-formed blood vessels within the plugs.


Asunto(s)
Células Endoteliales/diagnóstico por imagen , Células Endoteliales/patología , Glioblastoma/irrigación sanguínea , Microscopía Confocal/métodos , Ultrasonografía/métodos , Proteínas Angiogénicas/metabolismo , Proteínas Angiogénicas/farmacología , Animales , Línea Celular Tumoral , Colágeno , Medios de Cultivo Condicionados , Combinación de Medicamentos , Células Endoteliales/efectos de los fármacos , Glioblastoma/diagnóstico por imagen , Glioblastoma/metabolismo , Humanos , Aumento de la Imagen , Laminina , Ratones , Ratones Endogámicos BALB C , Microburbujas , Neovascularización Patológica/diagnóstico por imagen , Neovascularización Patológica/patología , Proteoglicanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...