Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Nanobiotechnology ; 22(1): 185, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38627717

RESUMEN

Rare earth nanomaterials (RE NMs), which are based on rare earth elements, have emerged as remarkable biomaterials for use in bone regeneration. The effects of RE NMs on osteogenesis, such as promoting the osteogenic differentiation of mesenchymal stem cells, have been investigated. However, the contributions of the properties of RE NMs to bone regeneration and their interactions with various cell types during osteogenesis have not been reviewed. Here, we review the crucial roles of the physicochemical and biological properties of RE NMs and focus on their osteogenic mechanisms. RE NMs directly promote the proliferation, adhesion, migration, and osteogenic differentiation of mesenchymal stem cells. They also increase collagen secretion and mineralization to accelerate osteogenesis. Furthermore, RE NMs inhibit osteoclast formation and regulate the immune environment by modulating macrophages and promote angiogenesis by inducing hypoxia in endothelial cells. These effects create a microenvironment that is conducive to bone formation. This review will help researchers overcome current limitations to take full advantage of the osteogenic benefits of RE NMs and will suggest a potential approach for further osteogenesis research.


Asunto(s)
Nanoestructuras , Osteogénesis , Células Endoteliales , Regeneración Ósea , Osteoclastos/metabolismo , Diferenciación Celular
2.
BMC Oral Health ; 22(1): 541, 2022 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-36434576

RESUMEN

BACKGROUND: MicroRNAs (miRNAs) play an important role in gene regulation that controls stem cells differentiation. Periodontal ligament stem cells (PDLSCs) could differentiate into osteo-/cementoblast-like cells that secretes cementum-like matrix both in vitro and in vivo. Whether miRNAs play key roles in osteoblastic differentiation of PDLSCs triggered by a special microenviroment remains elusive. In this study, we aimed to investigate potential miRNA expression changes in osteoblastic differentiation of PDLSCs by the induction of apical tooth germ cell-conditioned medium (APTG-CM). METHODS AND RESULTS: First, we analyzed the ability of APTG-CM to osteogenically differentiate PDLSCs. The results exhibited an enhanced mineralization ability, higher ALP activity and increased expression of osteogenic genes in APTG-CM-induced PDLSCs. Second, we used miRNA sequencing to analyze the miRNA expression profile of PDLSCs derived from three donors under 21-day induction or non-induction of APTG-CM. MiR-146a-5p was found to be up-regulated miRNA in induced PDLSCs and validated by RT-qPCR. Third, we used lentivirus-up/down system to verify the role of miR-146a-5p in the regulation of osteoblastic differentiation of PDLSCs. CONCLUSIONS: In conclusion, our results demonstrated that miR-146a-5p was involved in the promotion effect of APTG-CM on osteoblastic differentiation of PDLSCs, and suggested that miR-146a-5p might be a novel way in deciding the direction of PDLSCs differentiation.


Asunto(s)
MicroARNs , Ligamento Periodontal , Humanos , Medios de Cultivo Condicionados/farmacología , Medios de Cultivo Condicionados/metabolismo , Células Madre/metabolismo , Germen Dentario/metabolismo , MicroARNs/genética , MicroARNs/metabolismo
3.
Inflammation ; 44(1): 278-296, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32939669

RESUMEN

The potential risk associated with ACP nanoparticles (ACP NPs) cultured with immune cells and their indirect effects on osteogenesis have not been studied deeply. This project aims to evaluate the safety of ACP NPs in macrophages, the responses of macrophages (macrophage polarization, the cytokine secretion pattern of macrophages and intracellular homeostasis) to ACP NPs and the effect of ACP NPs/macrophage-modulated environments on the osteogenic ability of BMSCs. The cell proliferation rate and apoptosis were detected by CCK-8 and Annexin V Apoptosis Detection kits. ROS and autophagy expression were evaluated by ROS test kits and Western blot (WB). Macrophage polarization and cytokine expression were determined by SEM, cytoskeletal staining, RT-PCR and ELISA. TMT™ quantitative protein analysis was used to evaluate protein expression. BMSC osteogenic differentiation was detected by ALP staining, Alizarin Red solution staining and RT-PCR. ACP NPs were safe to macrophages but promoted autophagy and induced ROS production at high concentrations. ACP NPs changed morphology of macrophages and induced polarization into M1 type, thus promoting the expression of inflammatory cytokines. ACP NPs/macrophage-modulated environments weakened the osteogenic ability of BMSCs. ACP NPs polarize macrophages into the M1 phenotype and change the cytokine secretion pattern. ACP NPs/macrophage-modulated environments weaken the osteogenic ability of BMSCs. ACP NPs may cause aseptic inflammation and attenuate osteogenesis.


Asunto(s)
Fosfatos de Calcio/farmacología , Macrófagos/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , Nanopartículas/administración & dosificación , Osteogénesis/efectos de los fármacos , Animales , Macrófagos/inmunología , Macrófagos/metabolismo , Células Madre Mesenquimatosas/inmunología , Células Madre Mesenquimatosas/metabolismo , Ratones , Osteogénesis/fisiología , Células RAW 264.7
4.
Int J Nanomedicine ; 15: 2419-2435, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32368035

RESUMEN

BACKGROUND: In recent years, nanomaterials have been increasingly developed and applied in the field of bone tissue engineering. However, there are few studies on the induction of bone regeneration by tantalum nanoparticles (Ta NPs) and no reports on the effects of Ta NPs on the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) and the underlying mechanisms. The main purpose of this study was to investigate the effects of Ta NPs on bone regeneration and BMSC osteogenic differentiation and the underlying mechanisms. MATERIALS AND METHODS: The effects of Ta NPs on bone regeneration were evaluated in an animal experiment, and the effects of Ta NPs on osteogenic differentiation of BMSCs and the underlying mechanisms were evaluated in cell experiments. In the animal experiment, hematoxylin-eosin (HE) staining and hard-tissue section analysis showed that Ta NPs promoted bone regeneration, and immunohistochemistry revealed elevated expression of BMP2 and Smad4 in cells cultured with Ta NPs. RESULTS: The results of the cell experiments showed that Ta NPs promoted BMSC proliferation, alkaline phosphatase (ALP) activity, BMP2 secretion and extracellular matrix (ECM) mineralization, and the expression of related osteogenic genes and proteins (especially BMP2, Smad4 and Runx2) was upregulated under culture with Ta NPs. Smad4 expression, ALP activity, ECM mineralization, and osteogenesis-related gene and protein expression decreased after inhibiting Smad4. CONCLUSION: These data suggest that Ta NPs have an osteogenic effect and induce bone regeneration by activating the BMP2/Smad4/Runx2 signaling pathway, which in turn causes BMSCs to undergo osteogenic differentiation. This study provides insight into the molecular mechanisms underlying the effects of Ta NPs in bone regeneration.

5.
Mol Med Rep ; 22(1): 135-144, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32377690

RESUMEN

The use of human bone marrow mesenchymal stem cells (hBMSCs) as a tissue engineering application for individuals affected by osteoporosis and other types of bone loss diseases has been well studied in recent years. The osteogenic differentiation of hBMSCs can be regulated by a number of cues. MicroRNAs (miRNAs/miRs) serve as the key regulators of various biological processes; however, to the best of our knowledge, no information exists with regards to the specific modulatory effects of miR­10a­5p on osteogenic differentiation of hBMSCs. The aim of the present study was to investigate the relationship between hBMSCs and miR­10a­5p and, ultimately, to determine how miR­10a­5p affects the osteogenic differentiation process of hBMSCs in vitro and in vivo. The hBMSCs used in the present study were transfected with mirVana™ miRNA inhibitors and mimics, and transfection efficiency was assessed by fluorescence microscopy and reverse transcription­quantitative PCR (RT­qPCR). Viability of hBMSCs following transfection was analyzed using a Cell Counting Kit­8 assay. The mRNA expression levels of specific osteoblast markers, including alkaline phosphatase (ALP) and runt­related transcription factor 2 (RUNX2) were measured using RT­qPCR and western blot analysis. New bone formation was evaluated by Goldner's trichrome staining and micro­CT analysis in vivo. No significant difference in cell viability was observed among the different groups 24 h post­transfection. Overexpression of miR­10a­5p inhibited the expression of osteoblast makers in hBMSCs, whereas inhibition of miR­10a­5p upregulated the expression of ALP and RUNX2 in vitro. Furthermore, miR­10a­5p acted as a suppressor during the process of new bone formation in vivo. In conclusion, the findings of the present study suggested that miR­10a­5p served as a negative regulatory factor during osteoblast differentiation of hBMSCs and may be utilized in a treatment approach for bone repair in osteogenic­related diseases.


Asunto(s)
Células Madre Mesenquimatosas/citología , MicroARNs/genética , Osteogénesis , Diferenciación Celular , Línea Celular , Humanos , Células Madre Mesenquimatosas/metabolismo , Osteoblastos/citología , Osteoblastos/metabolismo , Regulación hacia Arriba
6.
Biomater Sci ; 8(13): 3664-3677, 2020 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-32463418

RESUMEN

Dicalcium silicate microparticle (C2S)-based biomaterials have a potential for bone and dental tissue regenerative applications. The C2S-mediated transcriptome level mechanism in mesenchymal stem cells (MSCs) during bone-defect healing has not been investigated yet. In this study, we elucidated the differential expression pattern of messenger RNAs (mRNAs) and circular RNAs (circRNAs) in C2S-treated MSCs and their involvement in the osteogenesis process. C2S robustly enhanced the osteogenic differentiation of MSCs and cranial bone defect healing. C2S-treatment modulated the differential expression of mRNAs and circRNAs in MSCs. Differentially expressed circRNAs and mRNAs were involved in competing endogenous RNA (ceRNA-interaction networks). These ceRNA-interaction networks regulated the signaling pathways associated with osteogenesis, e.g., Wnt, PI3K-Akt, MAPK, and JAK/STAT signaling. C2S-treatment upregulated the expression of circ_1983, Gas7, and Runx2 in BMSCs. RNase R and luciferase activity assay confirmed the stability and miR-6931 sponging property of circ_1983, respectively. Knockdown of circ_1983 enhanced miR-6931 expression but inhibited Gas7 and Runx2 expression and osteogenic differentiation in C2S-treated MSCs. In conclusion, for the first time, we report the role of cicr_1983-miR-6931-Gas7 ceRNA-interaction in C2S-induced osteogenic differentiation of MSCs and bone defect healing. This study opens a new research stream "the role of circRNAs-mediated ceRNA-interaction in biomaterials and stem cell-based bone tissue engineering".


Asunto(s)
Materiales Biocompatibles/farmacología , Compuestos de Calcio/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Proteínas del Tejido Nervioso/metabolismo , Osteogénesis/efectos de los fármacos , ARN Circular/metabolismo , ARN Mensajero/metabolismo , Silicatos/farmacología , Animales , Materiales Biocompatibles/química , Compuestos de Calcio/química , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Masculino , Células Madre Mesenquimatosas/metabolismo , Proteínas del Tejido Nervioso/genética , ARN Circular/genética , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Silicatos/química
7.
Nanomedicine (Lond) ; 13(15): 1939-1962, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-30152253

RESUMEN

Silicon-based materials and their oxides are widely used in drug delivery, dietary supplements, implants and dental fillers. Silica nanoparticles (SiNPs) interact with immunocompetent cells and induce immunotoxicity. However, the toxic effects of SiNPs on the immune system have been inadequately reviewed. The toxicity of SiNPs to the immune system depends on their physicochemical properties and the cell type. Assessments of immunotoxicity include determining cell dysfunctions, cytotoxicity and genotoxicity. This review focuses on the immunotoxicity of SiNPs and investigates the underlying mechanisms. The main mechanisms were proinflammatory responses, oxidative stress and autophagy. Considering the toxicity of SiNPs, surface and shape modifications may mitigate the toxic effects of SiNPs, providing a new way to produce these nanomaterials with less toxic impaction.


Asunto(s)
Sistema Inmunológico/efectos de los fármacos , Nanopartículas/química , Dióxido de Silicio/química , Animales , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Humanos , Sistema Inmunológico/fisiología , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Nanopartículas/toxicidad , Estrés Oxidativo/efectos de los fármacos , Dióxido de Silicio/toxicidad , Receptores Toll-Like/metabolismo
8.
Mediators Inflamm ; 2018: 8167932, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29853794

RESUMEN

Proinflammatory responses are important aspects of the immune response to biomaterials, which may cause peri-implantitis and implant shedding. The purpose of this study was to test the cytotoxicity and proinflammatory effects of dicalcium silicate particles on RAW 264.7 macrophages and to investigate the proinflammatory response mechanism induced by C2S and tricalcium phosphate (TCP). C2S and TCP particles were characterized using scanning electron microscopy (SEM), energy spectrum analysis (EDS) and X-ray diffraction (XRD). Cytotoxicity and apoptosis assays with C2S and TCP in the murine RAW 264.7 cell line were tested using the cell counting kit-8 (CCK-8) assay and flow cytometry (FCM). The detection results showed that C2S and TCP particles had no obvious toxicity in RAW 264.7 cells and did not cause obvious apoptosis, although they both caused an oxidative stress response by producing ROS when the concentrations were at 100 µg/mL. C2S particles are likely to induce a proinflammatory response by inducing high TLR2, TNF-α mRNA, TNF-α proinflammatory cytokine, p-IκB, and p-JNK1 + JNK2 + JNK3 expression levels. When we added siRNA-TLR2-1, a significant reduction was observed. These findings support the theory that C2S particles induce proinflammatory responses through the TLR2-mediated NF-κB and JNK pathways in the murine RAW 264.7 macrophage cell line.


Asunto(s)
Compuestos de Calcio/toxicidad , Silicatos/toxicidad , Receptor Toll-Like 2/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ratones , Microscopía Electrónica de Rastreo , FN-kappa B/metabolismo , Células RAW 264.7 , Proteína de Dominio de Muerte Asociada a Receptor de TNF/metabolismo , Factor 2 Asociado a Receptor de TNF/metabolismo , Difracción de Rayos X
9.
Nan Fang Yi Ke Da Xue Xue Bao ; 38(1): 106-111, 2018 Jan 30.
Artículo en Chino | MEDLINE | ID: mdl-33177016

RESUMEN

OBJECTIVE: To investigate osteogenic effect of collagen/bioglass composites loaded with a small interfering RNA (siRNA) targeting noggin. METHODS: The collagen/bioglass composites loaded with the negative control siRNA or noggin siRNA were prepared by freeze-drying method. CCK8 test was used to evaluate the proliferation of MC3T3 cells exposed to the aqueous extracts of collagen/bioglass composites and the siRNA-loaded collagen/bioglass composites. ALP activity assay, quantitative real-time PCR and Alizarin Red staining were used to assess the effect of the 3 composites on mineralization in MC3T3 cells. RESULTS: MC3T3 cells cultured for 3 and 5 days in the presence of the extracts of the 3 composites all showed significantly more active proliferation than the blank control cells (P < 0.05). Compared with the cells seeded on the scaffold without siRNA, MC3T3 cells seeded on collagen/bioglass scaffold loaded with noggin siRNA showed a significantly higher ALP activity at 14 days after seeding (P < 0.05) with significantly increased expression of ALP, Runx2 and BSP mRNAs (P < 0.05). Alizarin Red staining showed that the cells seeded on the noggin siRNA-loading collagen/bioglass scaffold contained significantly more mineralized nodules than the other cells (P < 0.05). CONCLUSIONS: The collagen/bioglass composites loaded with noggin siRNA have a good biocompatibility, and the collagen/bioglass composites and noggin siRNA show a synergistic effect in promoting osteogenesis.

10.
Int J Nanomedicine ; 12: 8085-8099, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29138564

RESUMEN

Due to the widespread applications of zinc oxide nanoparticles (ZnO NPs), the potential exposure of workers, consumers, and scientists to these particles has increased. This potential for exposure has attracted extensive attention in the science community. Many studies have examined the toxicological profile of ZnO NPs in the immune system, digestive system, however, information regarding the toxicity of ZnO NPs in the nervous system is scarce. In this study, we detected the cytotoxicity of two types of ZnO NPs of various sizes - ZnOa NPs and ZnOb NPs - and we characterized the shedding ability of zinc ions within culture medium and the cytoplasm. We found that reactive oxygen species played a crucial role in ZnO NP-induced cytotoxicity, likely because zinc ions were leached from ZnO NPs. Apoptosis and cytoskeleton changes were also toxic responses induced by the ZnO NPs, and ZnOb NPs induced more significant toxic responses than ZnOa NPs in SHSY5Y cells. In conclusion, ZnO NPs induced toxic responses in SHSY5Y cells in a size-dependent manner, which can probably be attributed to their ion-shedding ability.


Asunto(s)
Apoptosis/efectos de los fármacos , Nanopartículas/toxicidad , Especies Reactivas de Oxígeno/metabolismo , Óxido de Zinc/toxicidad , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Humanos , Nanopartículas/química , Neuroblastoma/patología , Estrés Oxidativo/efectos de los fármacos , Tamaño de la Partícula
11.
Int J Nanomedicine ; 12: 4323-4333, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28652735

RESUMEN

Porous tantalum (Ta) implants are highly corrosion resistant and biocompatible, and they possess significantly better initial stability than that of conventional titanium (Ti) implants. During loading wear, Ta nanoparticles (Ta-NPs) that were deposited on the surface of a porous Ta implant are inevitably released and come into direct contact with peri-implant osteoblasts. The wear debris may influence cell behavior and implant stabilization. However, the interaction of Ta-NPs with osteoblasts has not been clearly investigated. This study aimed to investigate the effect of Ta-NPs on cell proliferation and their underlying mechanism. The Cell Counting Kit-8 (CCK-8) assay was used to measure the cell viability of MC3T3-E1 mouse osteoblasts and showed that Ta-NP treatment could increase cell viability. Then, confocal microscopy, Western blotting, and transmission electron microscopy were used to confirm the autophagy induced by Ta-NPs, and evidence of autophagy induction was observed as positive LC3 puncta, high-LC3-II expression, and autophagic vesicle ultrastructures. The CCK-8 assay revealed that the cell viability was further increased and decreased by the application of an autophagy inducer and inhibitor, respectively. In addition, pre-treatment with autophagy inhibitor 3-methyladenine (3-MA) inhibited the Ta-NP-induced autophagy. These results indicate that the Ta-NPs can promote cell proliferation, that an autophagy inducer can further strengthen this effect and that an autophagy inhibitor can weaken this effect. In conclusion, autophagy was involved in Ta-NP-induced cell proliferation and had a promoting effect.


Asunto(s)
Autofagia/efectos de los fármacos , Nanopartículas/química , Osteoblastos/efectos de los fármacos , Tantalio/farmacología , Adenina/análogos & derivados , Adenina/farmacología , Animales , Línea Celular , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ratones , Osteoblastos/citología , Porosidad , Prótesis e Implantes , Tantalio/química
12.
Crit Rev Toxicol ; 46(4): 348-84, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26963861

RESUMEN

Zinc oxide nanoparticles (ZnO NPs) are nanomaterials that are widely used in many fields. ZnO NPs are ion-shedding particles, and zinc ions produce important and potent effects that differ from those of other metal or metal oxide NPs. Several studies have reported the toxicological effects of ZnO NPs administered via several different routes, including orally, dermally, by pulmonary absorption, intraperitoneally, and intravenously. Some potential routes for human exposure have produced various toxic effects in animal models. Moreover, several in vitro studies using a range of cell lines have reported the mechanisms underlying ZnO NP toxicity. Zinc ions play a very important role in ZnO NP toxicity, although the effects of the particulate form cannot be excluded. A crucial determinant of toxicity is the solubility of ZnO NPs, which is influenced by various factors, including the pH of the environment in tissues, cells, and organelles. In addition to the inflammatory responses and oxidative stress known to be induced by ZnO NPs, these NPs also exhibit some positive anti-inflammatory, anti-diabetic, and pro-coagulant effects at sub-toxic doses; these effects are probably induced by zinc ions, which are an essential element in cell homeostasis. It is highly likely that there are additional distinct mechanisms at sub-toxic doses and concentrations, which may be concealed or altered by the toxic effects observed at higher levels of ZnO NPs. Furthermore, many signaling pathway molecules associated with necrosis and apoptosis can be activated, leading to cell death. This review presents the status of ZnO NP toxicology and highlights areas requiring further investigation.


Asunto(s)
Nanopartículas/toxicidad , Óxido de Zinc/toxicidad , Animales , Humanos , Nanopartículas/química , Nanopartículas/metabolismo , Nanopartículas/envenenamiento , Óxido de Zinc/química , Óxido de Zinc/farmacocinética , Óxido de Zinc/envenenamiento
13.
J Mater Sci Mater Med ; 27(3): 59, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26800690

RESUMEN

Inflammatory effects are significant elements of the immune response to biomaterials. Previously, we reported inflammatory effects in response to dicalcium silicate (Ca2SiO4, C2S) particles. However, the immunological effects of C2S coatings have not been studied. C2S often used as coatings materials in orthopedic and dentistry applications. It may have different effect from C2S particles. Further, it remains unclear whether C2S coating is equally biocompatible as 45S5 coating. The aim of this study was to test the cytotoxicity and pro-inflammatory effects of C2S coating on RAW 264.7 macrophages. C2S and 45S5 coatings were characterized using scanning electron microscopy (SEM), atomic force microscopy (AFM), energy dispersive analysis (EDS) and X-ray diffraction (XRD). inductively coupled plasma optical emission spectroscopy (ICP-OES) was used to detect ionic concentrations after soaking coated discs in medium. The cytotoxicity of C2S and 45S5 coatings against RAW 264.7 macrophages was measured using the LDH Cytotoxicity Assay Kit, Cell Counting Kit-8 (CCK-8) assays and flow cytometry for apoptosis assays. The gene and protein expression of TNF-α, IL-6 and IL-1ß were detected using RT-q PCR and ELISA, respectively. The tested coating materials are not cytotoxic to macrophages. The C2S-coated surface stimulated macrophages to express pro-inflammatory mediators, such as TNF-α, IL-6 and IL-1ß, and C2S coating caused less IL-6 but greater IL-1ß production than the 45S5 coating. C2S coating have no cytotoxicity when directly cultured with macrophages. C2S and 45S5 coatings both have the potential to induce pro-inflammatory effects, and the biocompatibility of C2S is similar to that of 45S5.


Asunto(s)
Compuestos de Calcio/toxicidad , Supervivencia Celular/efectos de los fármacos , Inflamación/inducido químicamente , Macrófagos/efectos de los fármacos , Silicatos/toxicidad , Animales , Compuestos de Calcio/química , Línea Celular , Citocinas/genética , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Regulación de la Expresión Génica/efectos de los fármacos , Inflamación/metabolismo , Ensayo de Materiales , Ratones , ARN Mensajero/genética , ARN Mensajero/metabolismo , Silicatos/química , Andamios del Tejido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...