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












Base de datos
Intervalo de año de publicación
1.
iScience ; 26(10): 108094, 2023 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-37860774

RESUMEN

This work describes a patient-derived tumoroid model (PDTs) to support precision medicine in lung oncology. The use of human adipose tissue-derived microvasculature and patient-derived peripheral blood mononuclear cells (PBMCs) permits to achieve a physiologically relevant tumor microenvironment. This study involved ten patients at various stages of tumor progression. The vascularized, immune-infiltrated PDT model could be obtained within two weeks, matching the requirements of the therapeutic decision. Histological and transcriptomic analyses confirmed that the main features from the original tumor were reproduced. The 3D tumor model could be used to determine the dynamics of response to antiangiogenic therapy and platinum-based chemotherapy. Antiangiogenic therapy showed a significant decrease in vascular endothelial growth factor (VEGF)-A expression, reflecting its therapeutic effect in the model. In an immune-infiltrated PDT model, chemotherapy showed the ability to decrease the levels of lymphocyte activation gene-3 protein (LAG-3), B and T lymphocyte attenuator (BTLA), and inhibitory receptors of T cells functions.

2.
Biomedicines ; 11(7)2023 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-37509464

RESUMEN

Radiation therapy and platinum-based chemotherapy are common treatments for lung cancer patients. Several factors are considered for the low overall survival rate of lung cancer, such as the patient's physical state and the complex heterogeneity of the tumor, which leads to resistance to the treatment. Consequently, precision medicines are needed for the patients to improve their survival and their quality of life. Until now, no patient-derived tumoroid model has been reported to predict the efficiency of radiation therapy in non-small-cell lung cancer. Using our patient-derived tumoroid model, we report that this model could be used to evaluate the efficiency of radiation therapy and cisplatin-based chemotherapy in non-small-cell lung cancer. In addition, these results can be correlated to clinical outcomes of patients, indicating that this patient-derived tumoroid model can predict the response to radiotherapy and chemotherapy in non-small-cell lung cancer.

3.
J Microencapsul ; 40(2): 106-123, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36749573

RESUMEN

The fundamental purpose of this study was to develop a stable lyophilised finasteride nanosystem (FNS-NS) for topical delivery. The FNS-NS was fabricated using an ultrasonication technique. The impact of two different cryoprotectants on the physicochemical characteristics of FNS-NS before and after lyophilisation was thoroughly investigated. The lyophilised FNS-NS had spherical shape with particle size lied between 188.6 nm ± 4.4 and 298.7 nm ± 4.7, low PDI values (0.26 ± 0.02 to 0.32 ± 0.02) and zeta potential ranging from -38.3 to +53.3 mV. The confocal laser microscopy depicted a comparatively higher cellular internalisation achieved for undecorated FNS-NS with respect to its chitosan-decorated counterpart. The lyophilised FNS-NS was stable for 90 days at proper storage conditions. The FNS-NS with 15% trehalose had appropriate physicochemical attributes that could be a promising carrier for topical delivery to treat androgenic alopecia.


Asunto(s)
Finasterida , Nanopartículas , Humanos , Finasterida/farmacología , Alopecia , Liofilización , Tamaño de la Partícula
4.
Biomedicines ; 10(5)2022 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-35625840

RESUMEN

Patient-derived tumoroid (PDT) has been developed and used for anti-drug screening in the last decade. As compared to other existing drug screening models, a PDT-based in vitro 3D cell culture model could preserve the histological and mutational characteristics of their corresponding tumors and mimic the tumor microenvironment. However, few studies have been carried out to improve the microvascular network connecting the PDT and its surrounding microenvironment, knowing that poor tumor-selective drug transport and delivery is one of the major reasons for both the failure of anti-cancer drug screens and resistance in clinical treatment. In this study, we formed vascularized PDTs in six days using multiple cell types which maintain the histopathological features of the original cancer tissue. Furthermore, our results demonstrated a vascular network connecting PDT and its surrounding microenvironment. This fast and promising PDT model opens new perspectives for personalized medicine: this model could easily be used to test all therapeutic treatments and could be connected with a microfluidic device for more accurate drug screening.

5.
Biomedicines ; 9(8)2021 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-34440156

RESUMEN

One major limitation for the vascularization of bone substitutes used for filling is the presence of mineral blocks. The newly-formed blood vessels are stopped or have to circumvent the mineral blocks, resulting in inefficient delivery of oxygen and nutrients to the implant. This leads to necrosis within the implant and to poor engraftment of the bone substitute. The aim of the present study is to provide a bone substitute currently used in the clinic with suitably guided vascularization properties. This therapeutic hybrid bone filling, containing a mineral and a polymeric component, is fortified with pro-angiogenic smart nano-therapeutics that allow the release of angiogenic molecules. Our data showed that the improved vasculature within the implant promoted new bone formation and that the newly-formed bone swapped the mineral blocks of the bone substitutes much more efficiently than in non-functionalized bone substitutes. Therefore, we demonstrated that our therapeutic bone substitute is an advanced therapeutical medicinal product, with great potential to recuperate and guide vascularization that is stopped by mineral blocks, and can improve the regeneration of critical-sized bone defects. We have also elucidated the mechanism to understand how the newly-formed vessels can no longer encounter mineral blocks and pursue their course of vasculature, giving our advanced therapeutical bone filling great potential to be used in many applications, by combining filling and nano-regenerative medicine that currently fall short because of problems related to the lack of oxygen and nutrients.

6.
PLoS One ; 15(10): e0240676, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33052979

RESUMEN

The impact of cancer on lifespan is significantly increasing worldwide. Enhanced activity of drug efflux pumps and the incidences of the tumor microenvironment such as the apparition of a hypoxic gradient inside of the bulk tumor, are the major causes of chemotherapy failure. For instance, expression of Hypoxia-inducible factor (HIF-1α) has been associated with metastasis, resistance to chemotherapy and reduced survival rate. One of the current challenges to fight against cancer is therefore to find new molecules with therapeutic potential that could overcome this chemoresistance. In the present study, we focused on the bioactive plant flavonoid quercetin, which has strong antioxidant and anti-proliferative properties. We examined the efficacy of combined treatments of quercetin and the anti-cancer drugs gemcitabine and doxorubicin, known to specifically act on human pancreatic and hepatic cancer cells, respectively. Moreover, our study aimed to investigate more in-depth the implication of the multidrug transporter MDR1 and HIF-1α n chemoresistance and if quercetin could act on the activity of the drug efflux pumps and the hypoxia-associated effects. We observed that the anti-cancer drugs, were more effective when administered in combination with quercetin, as shown by an increased percentage of dead cells up to 60% in both 2D and 3D cultures. In addition, our results indicated that the combination of anti-cancer drugs and quercetin down-regulated the expression of HIF-1α and increased the expression levels of the regulator of apoptosis p53. Moreover, we observed that quercetin could inhibit the efflux activity of MDR1. Finally, our in vitro study suggests that the efficiency of the chemotherapeutic activity of known anti-cancer drugs might be significantly increased upon combination with quercetin. This flavonoid may therefore be a promising pharmacological agent for novel combination therapy since it potentializes the cytotoxic activity of gemcitabine and doxorubicin on by targeting the chemoresistance developed by the pancreatic and liver cancer cells respectively.


Asunto(s)
Antineoplásicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Antioxidantes/farmacología , Desoxicitidina/análogos & derivados , Doxorrubicina/farmacología , Quercetina/farmacología , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Adenocarcinoma/tratamiento farmacológico , Hipoxia de la Célula/efectos de los fármacos , Desoxicitidina/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células Hep G2 , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Pancreáticas/tratamiento farmacológico , Microambiente Tumoral/efectos de los fármacos , Gemcitabina
7.
Sci Rep ; 10(1): 13750, 2020 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-32792639

RESUMEN

Glioblastoma (GBM) is one of the most aggressive types of cancer, which begins within the brain. It is the most invasive type of glioma developed from astrocytes. Until today, Temozolomide (TMZ) is the only standard chemotherapy for patients with GBM. Even though chemotherapy extends the survival of patients, there are many undesirable side effects, and most cases show resistance to TMZ. FL3 is a synthetic flavagline which displays potent anticancer activities, and is known to inhibit cell proliferation, by provoking cell cycle arrest, and leads to apoptosis in a lot of cancer cell lines. However, the effect of FL3 in glioblastoma cancer cells has not yet been examined. Hypoxia is a major problem for patients with GBM, resulting in tumor resistance and aggressiveness. In this study, we explore the effect of FL3 in glioblastoma cells under normoxia and hypoxia conditions. Our results clearly indicate that this synthetic flavagline inhibits cell proliferation and induced senescence in glioblastoma cells cultured under both conditions. In addition, FL3 treatment had no effect on human brain astrocytes. These findings support the notion that the FL3 molecule could be used in combination with other chemotherapeutic agents or other therapies in glioblastoma treatments.


Asunto(s)
Antineoplásicos/farmacología , Astrocitos/efectos de los fármacos , Benzofuranos/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Senescencia Celular/efectos de los fármacos , Glioblastoma/tratamiento farmacológico , Aglaia/química , Anaerobiosis/fisiología , Apoptosis/efectos de los fármacos , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Glioblastoma/patología , Humanos , Preparaciones de Plantas/farmacología
8.
Materials (Basel) ; 13(14)2020 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-32664278

RESUMEN

The treatment of osteochondral defects remains a challenge. Four scaffolds were produced using Food and Drug Administration (FDA)-approved polymers to investigate their therapeutic potential for the regeneration of the osteochondral unit. Polycaprolactone (PCL) and poly(vinyl-pyrrolidone) (PVP) scaffolds were made by electrohydrodynamic techniques. Hydroxyapatite (HAp) and/or sodium hyaluronate (HA) can be then loaded to PCL nanofibers and/or PVP particles. The purpose of adding hydroxyapatite and sodium hyaluronate into PCL/PVP scaffolds is to increase the regenerative ability for subchondral bone and joint cartilage, respectively. Human bone marrow-derived mesenchymal stem cells (hBM-MSCs) were seeded on these biomaterials. The biocompatibility of these biomaterials in vitro and in vivo, as well as their potential to support MSC differentiation under specific chondrogenic or osteogenic conditions, were evaluated. We show here that hBM-MSCs could proliferate and differentiate both in vitro and in vivo on these biomaterials. In addition, the PCL-HAp could effectively increase the mineralization and induce the differentiation of MSCs into osteoblasts in an osteogenic condition. These results indicate that PCL-HAp biomaterials combined with MSCs could be a beneficial candidate for subchondral bone regeneration.

9.
Nat Commun ; 10(1): 2156, 2019 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-31089136

RESUMEN

The extended life expectancy and the raise of accidental trauma call for an increase of osteoarticular surgical procedures. Arthroplasty, the main clinical option to treat osteoarticular lesions, has limitations and drawbacks. In this manuscript, we test the preclinical safety of the innovative implant ARTiCAR for the treatment of osteoarticular lesions. Thanks to the combination of two advanced therapy medicinal products, a polymeric nanofibrous bone wound dressing and bone marrow-derived mesenchymal stem cells, the ARTiCAR promotes both subchondral bone and cartilage regeneration. In this work, the ARTiCAR shows 1) the feasibility in treating osteochondral defects in a large animal model, 2) the possibility to monitor non-invasively the healing process and 3) the overall safety in two animal models under GLP preclinical standards. Our data indicate the preclinical safety of ARTiCAR according to the international regulatory guidelines; the ARTiCAR could therefore undergo phase I clinical trial.


Asunto(s)
Cartílago Articular/fisiopatología , Trasplante de Células Madre Mesenquimatosas/métodos , Nanofibras/química , Osteoartritis/terapia , Andamios del Tejido/química , Animales , Regeneración Ósea , Línea Celular , Terapia Combinada/métodos , Modelos Animales de Enfermedad , Estudios de Factibilidad , Femenino , Humanos , Masculino , Células Madre Mesenquimatosas , Osteoartritis/fisiopatología , Ratas , Ratas Desnudas , Ovinos , Ingeniería de Tejidos/métodos , Cicatrización de Heridas/fisiología
10.
Int J Pharm ; 549(1-2): 299-305, 2018 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-30075249

RESUMEN

This study investigates the formulation of surfactant-free Pickering nano-emulsions able to release a drug at specific pH, in order to enhance its oral bioavailability. The stabilizing nanoparticles composed of magnesium hydroxide, were obtained by nano-precipitation method. The oil-in-water Pickering nano-emulsions stabilized with Mg(OH)2 nanoparticles, and encapsulating a model of hydrophobic drug (ibuprofen) were formulated following a high-energy process, using a sonication probe. The experimental approach explored the impact of all formulation parameters, composition and size of Mg(OH)2 nanoparticles, on the physico-chemical properties of the Pickering nano-emulsions. The system was characterized by DLS and transmission electron microscopy. In addition, Mg(OH)2 has the advantage of being solubilized in an acid medium leading to the destabilization of the nano-emulsion and the release of the active ingredient orally. The acid release study (pH = 1.2) showed cumulative release as a function of initial nanodroplet loading and saturation concentration. In basic media (pH = 6.8), we found a significant release of ibuprofen from the nano-emulsions that already had saturation in an acid medium. These nano-emulsions can not only protect patients from the side effects of acid medicines through the basic properties of hydroxides but also can contribute to the increase of the bioavailability of these drugs. In addition, once in the stomach pH is increased by hydroxides and promotes the release of active ingredients such as ibuprofen whose solubility is strongly influenced by pH.


Asunto(s)
Inhibidores de la Ciclooxigenasa/química , Portadores de Fármacos , Ibuprofeno/química , Óxido de Magnesio/química , Nanopartículas , Nanotecnología , Tecnología Farmacéutica/métodos , Administración Oral , Inhibidores de la Ciclooxigenasa/administración & dosificación , Preparaciones de Acción Retardada , Composición de Medicamentos , Liberación de Fármacos , Estabilidad de Medicamentos , Emulsiones , Concentración de Iones de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Ibuprofeno/administración & dosificación , Cinética , Solubilidad
11.
J Tissue Eng ; 9: 2041731418776819, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29899969

RESUMEN

Bone replacement might have been practiced for centuries with various materials of natural origin, but had rarely met success until the late 19th century. Nowadays, many different bone substitutes can be used. They can be either derived from biological products such as demineralized bone matrix, platelet-rich plasma, hydroxyapatite, adjunction of growth factors (like bone morphogenetic protein) or synthetic such as calcium sulfate, tri-calcium phosphate ceramics, bioactive glasses, or polymer-based substitutes. All these substitutes are not suitable for every clinical use, and they have to be chosen selectively depending on their purpose. Thus, this review aims to highlight the principal characteristics of the most commonly used bone substitutes and to give some directions concerning their clinical use, as spine fusion, open-wedge tibial osteotomy, long bone fracture, oral and maxillofacial surgery, or periodontal treatments. However, the main limitations to bone substitutes use remain the management of large defects and the lack of vascularization in their central part, which is likely to appear following their utilization. In the field of bone tissue engineering, developing porous synthetic substitutes able to support a faster and a wider vascularization within their structure seems to be a promising way of research.

12.
Biomed Res Int ; 2018: 7380389, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29682553

RESUMEN

Current approaches of regenerative therapies constitute strategies for bone tissue reparation and engineering, especially in the context of genetical diseases with skeletal defects. Bone regeneration using electrospun nanofibers' implant has the following objectives: bone neoformation induction with rapid healing, reduced postoperative complications, and improvement of bone tissue quality. In vivo implantation of polycaprolactone (PCL) biomembrane functionalized with BMP-2/Ibuprofen in mouse maxillary defects was followed by bone neoformation kinetics evaluation using microcomputed tomography. Wild-Type (WT) and Tabby (Ta) mice were used to compare effects on a normal phenotype and on a mutant model of ectodermal dysplasia (ED). After 21 days, no effect on bone neoformation was observed in Ta treated lesion (4% neoformation compared to 13% in the control lesion). Between the 21st and the 30th days, the use of biomembrane functionalized with BMP-2/Ibuprofen in maxillary bone lesions allowed a significant increase in bone neoformation peaks (resp., +8% in mutant Ta and +13% in WT). Histological analyses revealed a neoformed bone with regular trabecular structure, areas of mineralized bone inside the membrane, and an improved neovascularization in the treated lesion with bifunctionalized membrane. In conclusion, PCL functionalized biomembrane promoted bone neoformation, this effect being modulated by the Ta bone phenotype responsible for an alteration of bone response.


Asunto(s)
Enfermedades Óseas/tratamiento farmacológico , Regeneración Ósea/efectos de los fármacos , Maxilares/efectos de los fármacos , Maxilar/efectos de los fármacos , Nanofibras/administración & dosificación , Osteogénesis/efectos de los fármacos , Poliésteres/farmacología , Animales , Enfermedades Óseas/metabolismo , Proteína Morfogenética Ósea 2/metabolismo , Calcificación Fisiológica/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Maxilares/metabolismo , Maxilar/metabolismo , Ratones , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Ingeniería de Tejidos/métodos , Andamios del Tejido , Microtomografía por Rayos X/métodos
13.
Int J Mol Sci ; 19(2)2018 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-29393880

RESUMEN

The temporomandibular joint (TMJ) is an articulation formed between the temporal bone and the mandibular condyle which is commonly affected. These affections are often so painful during fundamental oral activities that patients have lower quality of life. Limitations of therapeutics for severe TMJ diseases have led to increased interest in regenerative strategies combining stem cells, implantable scaffolds and well-targeting bioactive molecules. To succeed in functional and structural regeneration of TMJ is very challenging. Innovative strategies and biomaterials are absolutely crucial because TMJ can be considered as one of the most difficult tissues to regenerate due to its limited healing capacity, its unique histological and structural properties and the necessity for long-term prevention of its ossified or fibrous adhesions. The ideal approach for TMJ regeneration is a unique scaffold functionalized with an osteochondral molecular gradient containing a single stem cell population able to undergo osteogenic and chondrogenic differentiation such as BMSCs, ADSCs or DPSCs. The key for this complex regeneration is the functionalization with active molecules such as IGF-1, TGF-ß1 or bFGF. This regeneration can be optimized by nano/micro-assisted functionalization and by spatiotemporal drug delivery systems orchestrating the 3D formation of TMJ tissues.


Asunto(s)
Regeneración Ósea/efectos de los fármacos , Medicina Regenerativa/métodos , Fracturas Craneales/terapia , Trasplante de Células Madre , Células Madre/citología , Ingeniería de Tejidos/métodos , Tejido Adiposo/citología , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Diferenciación Celular/efectos de los fármacos , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Factor 2 de Crecimiento de Fibroblastos/farmacología , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/farmacología , Piel/citología , Piel/efectos de los fármacos , Piel/metabolismo , Fracturas Craneales/patología , Fracturas Craneales/cirugía , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Articulación Temporomandibular/lesiones , Articulación Temporomandibular/cirugía , Andamios del Tejido , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta1/farmacología
14.
J Tissue Eng Regen Med ; 12(4): e2151-e2161, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29430872

RESUMEN

The sensory innervation of the dental pulp is essential for tooth function and protection. It is mediated by axons originating from the trigeminal ganglia and is spatio-temporally regulated. We have previously shown that the innervation of bioengineered teeth can be achieved only under immunosuppressive conditions. The aim of this study was to develop a model to determine the role of Semaphorin 3A (Sema3A) in the innervation of bioengineered teeth. We first analysed innervation of the dental pulp of mandibular first molars in newborn (postnatal day 0: PN0) mice deficient for Sema3A (Sema3A-/- ), a strong inhibitor of axon growth. While at PN0, axons detected by immunostaining for peripherin and NF200 were restricted to the peridental mesenchyme in Sema3A+/+ mice, they entered the dental pulp in Sema3A-/- mice. Then, we have implanted cultured teeth obtained from embryonic day-14 (E14) molar germs of Sema3A-/- mice together with trigeminal ganglia. The dental pulps of E14 cultured and implanted Sema3A-/- teeth were innervated, whereas the axons did not enter the pulp of E14 Sema3A+/+ cultured and implanted teeth. A "Membrane Targeting Peptide NRP1," suppressing the inhibitory effect of Sema3A, has been previously identified. The injection of this peptide at the site of implantation allowed the innervation of the dental pulp of bioengineered teeth obtained from E14 dental dissociated mesenchymal and epithelial cells reassociations of ICR mice. In conclusion, these data show that inhibition of only one axon repellent molecule, Sema3A, allows for pulp innervation of bioengineered teeth.


Asunto(s)
Pulpa Dental , Diente Molar , Receptores de Superficie Celular/metabolismo , Semaforina-3A/metabolismo , Ingeniería de Tejidos , Ganglio del Trigémino , Animales , Pulpa Dental/inervación , Pulpa Dental/metabolismo , Pulpa Dental/patología , Mandíbula/inervación , Mandíbula/metabolismo , Mandíbula/patología , Ratones , Ratones Endogámicos ICR , Ratones Mutantes , Diente Molar/inervación , Diente Molar/metabolismo , Diente Molar/patología , Receptores de Superficie Celular/genética , Semaforina-3A/genética , Ganglio del Trigémino/metabolismo , Ganglio del Trigémino/patología
15.
Nanomedicine (Lond) ; 12(23): 2651-2674, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29094650

RESUMEN

AIM: We developed polymeric membranes for local administration of nonsoluble anti-inflammatory statin, as potential wound patch in rheumatic joint or periodontal lesions. METHODS: Electrospun polycaprolactone membranes were fitted with polysaccharide-atorvastatin nanoreservoirs by using complexes with poly-aminocyclodextrin. Characterization methods are UV-Visible and X-ray photoelectron spectroscopy, molecular dynamics, scanning and transmission electron microscopy. In vitro, membranes were seeded with macrophages, and inflammatory cytokine expression were monitored. RESULTS & CONCLUSION: Stable inclusion complexes were formed in solution (1:1 stability constant 368 M-1, -117.40 kJ mol-1), with supramolecular globular organization (100 nm, substructure 30 nm). Nanoreservoir technology leads to homogeneous distribution of atorvastatin calcium trihydrate complexes in the membrane. Quantity embedded was estimated (70-90 µg in 30 µm × 6 mm membrane). Anti-inflammatory effect by cell contact-dependent release reached 60% inhibition for TNF-α and 80% for IL-6. The novelty resides in the double protection offered by the cyclodextrins as drug molecular chaperones, with further embedding into biodegradable nanoreservoirs. The strategy is versatile and can target other diseases.


Asunto(s)
Antiinflamatorios/farmacología , Atorvastatina/farmacología , Nanofibras/química , Poliésteres/química , Antiinflamatorios/química , Atorvastatina/química , Ciclodextrinas/química , Liberación de Fármacos , Humanos , Interleucina-6/metabolismo , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Membranas Artificiales , Simulación de Dinámica Molecular , Nanoconjugados/química , Células THP-1 , Termodinámica , Factor de Necrosis Tumoral alfa/metabolismo , Infección de Heridas/prevención & control
16.
Exp Cell Res ; 360(2): 138-145, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-28867479

RESUMEN

Angiogenesis is now well known for being involved in tumor progression, aggressiveness, emergence of metastases, and also resistance to cancer therapies. In this study, to better mimic tumor angiogenesis encountered in vivo, we used 3D culture of osteosarcoma cells (MG-63) that we deposited on 2D endothelial cells (HUVEC) grown in monolayer. We report that endothelial cells combined with tumor cells were able to form a well-organized network, and that tubule-like structures corresponding to new vessels infiltrate tumor spheroids. These vessels presented a lumen and expressed specific markers as CD31 and collagen IV. The combination of 2D endothelial cells and 3D microtissues of tumor cells also increased expression of angiogenic factors as VEGF, CXCR4 and ICAM1. The cell environment is the key point to develop tumor vascularization in vitro and to be closer to tumor encountered in vivo.


Asunto(s)
Neoplasias Óseas/patología , Técnicas de Cultivo de Célula/métodos , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/fisiología , Neovascularización Patológica/patología , Osteosarcoma/patología , Neoplasias Óseas/irrigación sanguínea , Neoplasias Óseas/genética , Células Cultivadas , Regulación Neoplásica de la Expresión Génica , Humanos , Neovascularización Patológica/genética , Osteosarcoma/irrigación sanguínea , Osteosarcoma/genética , Andamios del Tejido/química
17.
Biomed Mater Eng ; 28(s1): S185-S192, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28372294

RESUMEN

The time needed to obtain functional regenerated bone tissue depends on the existence of a reliable vascular support. Current techniques used in clinic, for example after tooth extraction, do not allow regaining or preserving the same bone volume. Our aim is to develop a cellularized active implant of the third generation, equipped with human mesenchymal stem cells to improve the quality of implant vascularization. We seeded a commercialized collagen implant with human mesenchymal stem cells (hMSCs) and then with human umbilical vein endothelial cells (HUVECs). We analyzed the biocompatibility and the behavior of endothelial cells with this implant. We observed a biocompatibility of the active implant, and a re-organization of endothelial cells into clustered networks. This work shows the possibility to develop an implant of the third generation supporting vascularization, improving the medical care of patients.


Asunto(s)
Colágeno/química , Células Endoteliales de la Vena Umbilical Humana/citología , Células Madre Mesenquimatosas/citología , Neovascularización Fisiológica , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Animales , Materiales Biocompatibles/química , Bovinos , Humanos , Ensayo de Materiales
18.
Int J Nanomedicine ; 12: 447-457, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28138241

RESUMEN

In tissue engineering, it is still rare today to see clinically transferable strategies for tissue-engineered graft production that conclusively offer better tissue regeneration than the already existing technologies, decreased recovery times, and less risk of complications. Here a novel tissue-engineering concept is presented for the production of living bone implants combining 1) a nanofibrous and microporous implant as cell colonization matrix and 2) 3D bone cell spheroids. This combination, double 3D implants, shows clinical relevant thicknesses for the treatment of an early stage of bone lesions before the need of bone substitutes. The strategy presented here shows a complete closure of a defect in nude mice calvaria after only 31 days. As a novel strategy for bone regenerative nanomedicine, it holds great promises to enhance the therapeutic efficacy of living bone implants.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Nanomedicina/métodos , Nanotecnología/métodos , Osteoblastos/citología , Prótesis e Implantes , Medicina Regenerativa/métodos , Esferoides Celulares/citología , Animales , Regeneración Ósea/efectos de los fármacos , Calcificación Fisiológica/efectos de los fármacos , Humanos , Ratones Desnudos , Nanopartículas/química , Osteoblastos/efectos de los fármacos , Polímeros/farmacología , Porosidad , Ingeniería de Tejidos , Andamios del Tejido/química
19.
Nanomedicine (Lond) ; 11(18): 2419-30, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27529130

RESUMEN

AIM: Success of functional vascularized tissue repair depends on vascular support system supply and still remains challenging. Our objective was to develop a nanoactive implant enhancing endothelial cell activity, particularly for bone tissue engineering in the regenerative medicine field. MATERIALS & METHODS: We developed a new strategy of tridimensional implant based on cell-dependent sustained release of VEGF nanoparticles. These nanoparticles were homogeneously distributed within nanoreservoirs onto the porous scaffold, with quicker reorganization of endothelial cells. Moreover, the activity of this active smart implant on cells was also modulated by addition of osteoblastic cells. RESULTS & CONCLUSION: This sophisticated active strategy should potentiate efficiency of current therapeutic implants for bone repair, avoiding the need for bone substitutes.


Asunto(s)
Sustitutos de Huesos/química , Neovascularización Fisiológica/efectos de los fármacos , Medicina Regenerativa , Ingeniería de Tejidos , Animales , Desarrollo Óseo/efectos de los fármacos , Sustitutos de Huesos/administración & dosificación , Células Endoteliales/efectos de los fármacos , Humanos , Neovascularización Patológica , Prótesis e Implantes
20.
PLoS One ; 7(12): e53498, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23300933

RESUMEN

BACKGROUND: Morphogenesis results from the coordination of distinct cell signaling pathways controlling migration, differentiation, apoptosis, and proliferation, along stem/progenitor cell dynamics. To decipher this puzzle, we focused on epithelial-mesenchymal transition (EMT) "master genes". EMT has emerged as a unifying concept, involving cell-cell adhesion, migration and apoptotic pathways. EMT also appears to mingle with stemness. However, very little is known on the physiological role and relevance of EMT master-genes. We addressed this question during mammary morphogenesis. Recently, a link between Slug/Snai2 and stemness has been described in mammary epithelial cells, but EMT master genes actual localization, role and targets during mammary gland morphogenesis are not known and we focused on this basic question. METHODOLOGY/PRINCIPAL FINDINGS: Using a Slug-lacZ transgenic model and immunolocalization, we located Slug in a distinct subpopulation covering about 10-20% basal cap and duct cells, mostly cycling cells, coexpressed with basal markers P-cadherin, CK5 and CD49f. During puberty, Slug-deficient mammary epithelium exhibited a delayed development after transplantation, contained less cycling cells, and overexpressed CK8/18, ER, GATA3 and BMI1 genes, linked to luminal lineage. Other EMT master genes were overexpressed, suggesting compensation mechanisms. Gain/loss-of-function in vitro experiments confirmed Slug control of mammary epithelial cell luminal differentiation and proliferation. In addition, they showed that Slug enhances specifically clonal mammosphere emergence and growth, cell motility, and represses apoptosis. Strikingly, Slug-deprived mammary epithelial cells lost their potential to generate secondary clonal mammospheres. CONCLUSIONS/SIGNIFICANCE: We conclude that Slug pathway controls the growth dynamics of a subpopulation of cycling progenitor basal cells during mammary morphogenesis. Overall, our data better define a key mechanism coordinating cell lineage dynamics and morphogenesis, and provide physiological relevance to broadening EMT pathways.


Asunto(s)
Diferenciación Celular , Células Epiteliales/metabolismo , Glándulas Mamarias Animales/embriología , Factores de Transcripción/genética , Animales , Adhesión Celular , Movimiento Celular , Proliferación Celular , Células Epiteliales/citología , Transición Epitelial-Mesenquimal , Femenino , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/metabolismo , Ratones , Morfogénesis , Factores de Transcripción de la Familia Snail , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...