- -

In vitro development of bioimplants made up of elastomeric scaffolds with peptide gel filling seeded with human subcutaneous adipose tissue-derived progenitor cells

RiuNet: Repositorio Institucional de la Universidad Politécnica de Valencia

Compartir/Enviar a

Citas

Estadísticas

  • Estadisticas de Uso

In vitro development of bioimplants made up of elastomeric scaffolds with peptide gel filling seeded with human subcutaneous adipose tissue-derived progenitor cells

Mostrar el registro sencillo del ítem

Ficheros en el ítem

dc.contributor.author Castells-Sala, Cristina es_ES
dc.contributor.author Martínez Ramos, Cristina es_ES
dc.contributor.author Vallés Lluch, Ana es_ES
dc.contributor.author Monleón Pradas, Manuel es_ES
dc.contributor.author Semino, Carlos es_ES
dc.date.accessioned 2016-11-16T10:42:18Z
dc.date.available 2016-11-16T10:42:18Z
dc.date.issued 2015-11
dc.identifier.issn 1549-3296
dc.identifier.uri http://hdl.handle.net/10251/74183
dc.description.abstract [EN] Myocardial tissue lacks the ability to regenerate itself significantly following a myocardial infarction. Thus, new strategies that could compensate this lack are of high interest. Cardiac tissue engineering (CTE) strategies are a relatively new approach that aims to compensate the tissue loss using combination of biomaterials, cells and bioactive molecules. The goal of the present study was to evaluate cell survival and growth, seeding capacity and cellular phenotype maintenance of subcutaneous adipose tissue-derived progenitor cells in a new synthetic biomaterial scaffold platform. Specifically, here we tested the effect of the RAD16-I peptide gel in microporous poly(ethyl acrylate) polymers using two-dimensional PEA films as controls. Results showed optimal cell adhesion efficiency and growth in the polymers coated with the self-assembling peptide RAD16-I. Importantly, subATDPCs seeded into microporous PEA scaffolds coated with RAD16-I maintained its phenotype and were able to migrate outwards the bioactive patch, hopefully toward the infarcted area once implanted. These data suggest that this bioimplant (scaffold/RAD16-I/cells) can be suitable for further in vivo implantation with the aim to improve the function of affected tissue after myocardial infarction. (c) 2015 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 103A: 3419-3430, 2015. es_ES
dc.description.sponsorship Contract grant sponsor: European Union Seventh Framework Programme; contract grant number: 229239 en_EN
dc.language Inglés es_ES
dc.publisher Wiley es_ES
dc.relation.ispartof Journal of Biomedical Materials Research Part A es_ES
dc.rights Reserva de todos los derechos es_ES
dc.subject.classification MAQUINAS Y MOTORES TERMICOS es_ES
dc.title In vitro development of bioimplants made up of elastomeric scaffolds with peptide gel filling seeded with human subcutaneous adipose tissue-derived progenitor cells es_ES
dc.type Artículo es_ES
dc.identifier.doi 10.1002/jbm.a.35482
dc.relation.projectID info:eu-repo/grantAgreement/EC/FP7/229239/EU/Regeneration of Cardiac Tissue Assisted by Bioactive Implants/ es_ES
dc.rights.accessRights Abierto es_ES
dc.contributor.affiliation Universitat Politècnica de València. Escuela Técnica Superior de Ingenieros Industriales - Escola Tècnica Superior d'Enginyers Industrials es_ES
dc.contributor.affiliation Universitat Politècnica de València. Centro de Biomateriales e Ingeniería Tisular - Centre de Biomaterials i Enginyeria Tissular es_ES
dc.description.bibliographicCitation Castells-Sala, C.; Martínez Ramos, C.; Vallés Lluch, A.; Monleón Pradas, M.; Semino, C. (2015). In vitro development of bioimplants made up of elastomeric scaffolds with peptide gel filling seeded with human subcutaneous adipose tissue-derived progenitor cells. Journal of Biomedical Materials Research Part A. 103(11):3419-3430. https://doi.org/10.1002/jbm.a.35482 es_ES
dc.description.accrualMethod S es_ES
dc.relation.publisherversion http://dx.doi.org/10.1002/jbm.a.35482 es_ES
dc.description.upvformatpinicio 3419 es_ES
dc.description.upvformatpfin 3430 es_ES
dc.type.version info:eu-repo/semantics/publishedVersion es_ES
dc.description.volume 103 es_ES
dc.description.issue 11 es_ES
dc.relation.senia 295665 es_ES
dc.contributor.funder European Commission
dc.description.references Persidis, A. (1999). Tissue engineering. Nature Biotechnology, 17(5), 508-510. doi:10.1038/8700 es_ES
dc.description.references Venugopal, J. R., Prabhakaran, M. P., Mukherjee, S., Ravichandran, R., Dan, K., & Ramakrishna, S. (2011). Biomaterial strategies for alleviation of myocardial infarction. Journal of The Royal Society Interface, 9(66), 1-19. doi:10.1098/rsif.2011.0301 es_ES
dc.description.references Castells-Sala, C., & Semino, C. E. (2012). Biomaterials for stem cell culture and seeding for the generation and delivery of cardiac myocytes. Current Opinion in Organ Transplantation, 17(6), 681-687. doi:10.1097/mot.0b013e32835a34a6 es_ES
dc.description.references Nunes, S. S., Song, H., Chiang, C. K., & Radisic, M. (2011). Stem Cell-Based Cardiac Tissue Engineering. Journal of Cardiovascular Translational Research, 4(5), 592-602. doi:10.1007/s12265-011-9307-x es_ES
dc.description.references Fernandes, S., Kuklok, S., McGonigle, J., Reinecke, H., & Murry, C. E. (2012). Synthetic Matrices to Serve as Niches for Muscle Cell Transplantation. Cells Tissues Organs, 195(1-2), 48-59. doi:10.1159/000331414 es_ES
dc.description.references Murry, C. E., Field, L. J., & Menasché, P. (2005). Cell-Based Cardiac Repair. Circulation, 112(20), 3174-3183. doi:10.1161/circulationaha.105.546218 es_ES
dc.description.references Wang, F., & Guan, J. (2010). Cellular cardiomyoplasty and cardiac tissue engineering for myocardial therapy☆. Advanced Drug Delivery Reviews, 62(7-8), 784-797. doi:10.1016/j.addr.2010.03.001 es_ES
dc.description.references Patel, A. N., & Genovese, J. A. (2007). Stem cell therapy for the treatment of heart failure. Current Opinion in Cardiology, 22(5), 464-470. doi:10.1097/hco.0b013e3282c3cb2a es_ES
dc.description.references Pendyala, L., Goodchild, T., Gadesam, R., Chen, J., Robinson, K., Chronos, N., & Hou, D. (2008). Cellular Cardiomyoplasty and Cardiac Regeneration. Current Cardiology Reviews, 4(2), 72-80. doi:10.2174/157340308784245748 es_ES
dc.description.references Li, Z., Guo, X., & Guan, J. (2012). A Thermosensitive Hydrogel Capable of Releasing bFGF for Enhanced Differentiation of Mesenchymal Stem Cell into Cardiomyocyte-like Cells under Ischemic Conditions. Biomacromolecules, 13(6), 1956-1964. doi:10.1021/bm300574j es_ES
dc.description.references Huang, C.-C., Wei, H.-J., Yeh, Y.-C., Wang, J.-J., Lin, W.-W., Lee, T.-Y., … Sung, H.-W. (2012). Injectable PLGA porous beads cellularized by hAFSCs for cellular cardiomyoplasty. Biomaterials, 33(16), 4069-4077. doi:10.1016/j.biomaterials.2012.02.024 es_ES
dc.description.references Liu, Z., Wang, H., Wang, Y., Lin, Q., Yao, A., Cao, F., … Wang, C. (2012). The influence of chitosan hydrogel on stem cell engraftment, survival and homing in the ischemic myocardial microenvironment. Biomaterials, 33(11), 3093-3106. doi:10.1016/j.biomaterials.2011.12.044 es_ES
dc.description.references Kadner, K., Dobner, S., Franz, T., Bezuidenhout, D., Sirry, M. S., Zilla, P., & Davies, N. H. (2012). The beneficial effects of deferred delivery on the efficiency of hydrogel therapy post myocardial infarction. Biomaterials, 33(7), 2060-2066. doi:10.1016/j.biomaterials.2011.11.031 es_ES
dc.description.references Naderi, H., Matin, M. M., & Bahrami, A. R. (2011). Review paper: Critical Issues in Tissue Engineering: Biomaterials, Cell Sources, Angiogenesis, and Drug Delivery Systems. Journal of Biomaterials Applications, 26(4), 383-417. doi:10.1177/0885328211408946 es_ES
dc.description.references Ptaszek, L. M., Mansour, M., Ruskin, J. N., & Chien, K. R. (2012). Towards regenerative therapy for cardiac disease. The Lancet, 379(9819), 933-942. doi:10.1016/s0140-6736(12)60075-0 es_ES
dc.description.references Song, H., Yoon, C., Kattman, S. J., Dengler, J., Massé, S., Thavaratnam, T., … Zandstra, P. W. (2009). Interrogating functional integration between injected pluripotent stem cell-derived cells and surrogate cardiac tissue. Proceedings of the National Academy of Sciences, 107(8), 3329-3334. doi:10.1073/pnas.0905729106 es_ES
dc.description.references Liu, J., Zhang, Z., Liu, Y., Guo, C., Gong, Y., Yang, S., … He, Z. (2012). Generation, Characterization, and Potential Therapeutic Applications of Cardiomyocytes from Various Stem Cells. Stem Cells and Development, 21(12), 2095-2110. doi:10.1089/scd.2012.0031 es_ES
dc.description.references Abdelli, L. S., Merino, H., Rocher, C. M., & Singla, D. K. (2012). Cell therapy in the heart. Canadian Journal of Physiology and Pharmacology, 90(3), 307-315. doi:10.1139/y11-130 es_ES
dc.description.references Hoover-Plow, J., & Gong, Y. (2012). Challenges for heart disease stem cell therapy. Vascular Health and Risk Management, 99. doi:10.2147/vhrm.s25665 es_ES
dc.description.references Chachques, J. C., Trainini, J. C., Lago, N., Cortes-Morichetti, M., Schussler, O., & Carpentier, A. (2008). Myocardial Assistance by Grafting a New Bioartificial Upgraded Myocardium (MAGNUM Trial): Clinical Feasibility Study. The Annals of Thoracic Surgery, 85(3), 901-908. doi:10.1016/j.athoracsur.2007.10.052 es_ES
dc.description.references Karam, J.-P., Muscari, C., & Montero-Menei, C. N. (2012). Combining adult stem cells and polymeric devices for tissue engineering in infarcted myocardium. Biomaterials, 33(23), 5683-5695. doi:10.1016/j.biomaterials.2012.04.028 es_ES
dc.description.references Clifford, D. M., Fisher, S. A., Brunskill, S. J., Doree, C., Mathur, A., Clarke, M. J., … Martin-Rendon, E. (2012). Long-Term Effects of Autologous Bone Marrow Stem Cell Treatment in Acute Myocardial Infarction: Factors That May Influence Outcomes. PLoS ONE, 7(5), e37373. doi:10.1371/journal.pone.0037373 es_ES
dc.description.references Lee, R. H., Kim, B., Choi, I., Kim, H., Choi, H. S., Suh, K., … Jung, J. S. (2004). Characterization and Expression Analysis of Mesenchymal Stem Cells from Human Bone Marrow and Adipose Tissue. Cellular Physiology and Biochemistry, 14(4-6), 311-324. doi:10.1159/000080341 es_ES
dc.description.references Qayyum, A. A., Haack-Sørensen, M., Mathiasen, A. B., Jørgensen, E., Ekblond, A., & Kastrup, J. (2012). Adipose-derived mesenchymal stromal cells for chronic myocardial ischemia (MyStromalCell Trial): study design. Regenerative Medicine, 7(3), 421-428. doi:10.2217/rme.12.17 es_ES
dc.description.references Bai, X., Ma, J., Pan, Z., Song, Y.-H., Freyberg, S., Yan, Y., … Alt, E. (2007). Electrophysiological properties of human adipose tissue-derived stem cells. American Journal of Physiology-Cell Physiology, 293(5), C1539-C1550. doi:10.1152/ajpcell.00089.2007 es_ES
dc.description.references Rigol, M., Solanes, N., Farré, J., Roura, S., Roqué, M., Berruezo, A., … Heras, M. (2010). Effects of Adipose Tissue-Derived Stem Cell Therapy After Myocardial Infarction: Impact of the Route of Administration. Journal of Cardiac Failure, 16(4), 357-366. doi:10.1016/j.cardfail.2009.12.006 es_ES
dc.description.references Planat-Bénard, V., Menard, C., André, M., Puceat, M., Perez, A., Garcia-Verdugo, J.-M., … Casteilla, L. (2004). Spontaneous Cardiomyocyte Differentiation From Adipose Tissue Stroma Cells. Circulation Research, 94(2), 223-229. doi:10.1161/01.res.0000109792.43271.47 es_ES
dc.description.references Weber, B., Zeisberger, S. M., & Hoerstrup, S. P. (2011). Prenatally harvested cells for cardiovascular tissue engineering: Fabrication of autologous implants prior to birth. Placenta, 32, S316-S319. doi:10.1016/j.placenta.2011.04.001 es_ES
dc.description.references Cortes-Morichetti, M., Frati, G., Schussler, O., Van Huyen, J.-P. D., Lauret, E., Genovese, J. A., … Chachques, J. C. (2007). Association Between a Cell-Seeded Collagen Matrix and Cellular Cardiomyoplasty for Myocardial Support and Regeneration. Tissue Engineering, 13(11), 2681-2687. doi:10.1089/ten.2006.0447 es_ES
dc.description.references Chi, N.-H., Yang, M.-C., Chung, T.-W., Chen, J.-Y., Chou, N.-K., & Wang, S.-S. (2012). Cardiac repair achieved by bone marrow mesenchymal stem cells/silk fibroin/hyaluronic acid patches in a rat of myocardial infarction model. Biomaterials, 33(22), 5541-5551. doi:10.1016/j.biomaterials.2012.04.030 es_ES
dc.description.references Wang, T., Jiang, X.-J., Tang, Q.-Z., Li, X.-Y., Lin, T., Wu, D.-Q., … Okello, E. (2009). Bone marrow stem cells implantation with α-cyclodextrin/MPEG–PCL–MPEG hydrogel improves cardiac function after myocardial infarction. Acta Biomaterialia, 5(8), 2939-2944. doi:10.1016/j.actbio.2009.04.040 es_ES
dc.description.references Wu, J., Zeng, F., Huang, X.-P., Chung, J. C.-Y., Konecny, F., Weisel, R. D., & Li, R.-K. (2011). Infarct stabilization and cardiac repair with a VEGF-conjugated, injectable hydrogel. Biomaterials, 32(2), 579-586. doi:10.1016/j.biomaterials.2010.08.098 es_ES
dc.description.references Vunjak-Novakovic, G., Lui, K. O., Tandon, N., & Chien, K. R. (2011). Bioengineering Heart Muscle: A Paradigm for Regenerative Medicine. Annual Review of Biomedical Engineering, 13(1), 245-267. doi:10.1146/annurev-bioeng-071910-124701 es_ES
dc.description.references Dobner, S., Bezuidenhout, D., Govender, P., Zilla, P., & Davies, N. (2009). A Synthetic Non-degradable Polyethylene Glycol Hydrogel Retards Adverse Post-infarct Left Ventricular Remodeling. Journal of Cardiac Failure, 15(7), 629-636. doi:10.1016/j.cardfail.2009.03.003 es_ES
dc.description.references Blan, N. R., & Birla, R. K. (2008). Design and fabrication of heart muscle using scaffold-based tissue engineering. Journal of Biomedical Materials Research Part A, 86A(1), 195-208. doi:10.1002/jbm.a.31642 es_ES
dc.description.references Zhao, X., & Zhang, S. (s. f.). Self-Assembling Nanopeptides Become a New Type of Biomaterial. Advances in Polymer Science, 145-170. doi:10.1007/12_088 es_ES
dc.description.references Vallés-Lluch, A., Arnal-Pastor, M., Martínez-Ramos, C., Vilariño-Feltrer, G., Vikingsson, L., Castells-Sala, C., … Monleón Pradas, M. (2013). Combining self-assembling peptide gels with three-dimensional elastomer scaffolds. Acta Biomaterialia, 9(12), 9451-9460. doi:10.1016/j.actbio.2013.07.038 es_ES
dc.description.references Arnal-Pastor, M., Vallés-Lluch, A., Keicher, M., & Pradas, M. M. (2011). Coating typologies and constrained swelling of hyaluronic acid gels within scaffold pores. Journal of Colloid and Interface Science, 361(1), 361-369. doi:10.1016/j.jcis.2011.05.013 es_ES
dc.description.references Pérez Olmedilla, M., Garcia-Giralt, N., Pradas, M. M., Ruiz, P. B., Gómez Ribelles, J. L., Palou, E. C., & García, J. C. M. (2006). Response of human chondrocytes to a non-uniform distribution of hydrophilic domains on poly (ethyl acrylate-co-hydroxyethyl methacrylate) copolymers. Biomaterials, 27(7), 1003-1012. doi:10.1016/j.biomaterials.2005.07.030 es_ES
dc.description.references Soria, J. M., Martínez Ramos, C., Salmerón Sánchez, M., Benavent, V., Campillo Fernández, A., Gómez Ribelles, J. L., … Barcia, J. A. (2006). Survival and differentiation of embryonic neural explants on different biomaterials. Journal of Biomedical Materials Research Part A, 79A(3), 495-502. doi:10.1002/jbm.a.30803 es_ES
dc.description.references Campillo-Fernandez, A. J., Pastor, S., Abad-Collado, M., Bataille, L., Gomez-Ribelles, J. L., Meseguer-Dueñas, J. M., … Ruiz-Moreno, J. M. (2007). Future Design of a New Keratoprosthesis. Physical and Biological Analysis of Polymeric Substrates for Epithelial Cell Growth. Biomacromolecules, 8(8), 2429-2436. doi:10.1021/bm0703012 es_ES
dc.description.references Martínez-Ramos, C., Vallés-Lluch, A., Verdugo, J. M. G., Ribelles, J. L. G., Barcia Albacar, J. A., Orts, A. B., … Pradas, M. M. (2012). Channeled scaffolds implanted in adult rat brain. Journal of Biomedical Materials Research Part A, 100A(12), 3276-3286. doi:10.1002/jbm.a.34273 es_ES
dc.description.references Hernández, J. C. R., Salmerón Sánchez, M., Soria, J. M., Gómez Ribelles, J. L., & Monleón Pradas, M. (2007). Substrate Chemistry-Dependent Conformations of Single Laminin Molecules on Polymer Surfaces are Revealed by the Phase Signal of Atomic Force Microscopy. Biophysical Journal, 93(1), 202-207. doi:10.1529/biophysj.106.102491 es_ES
dc.description.references Kisiday, J., Jin, M., Kurz, B., Hung, H., Semino, C., Zhang, S., & Grodzinsky, A. J. (2002). Self-assembling peptide hydrogel fosters chondrocyte extracellular matrix production and cell division: Implications for cartilage tissue repair. Proceedings of the National Academy of Sciences, 99(15), 9996-10001. doi:10.1073/pnas.142309999 es_ES
dc.description.references Semino, C. E., Merok, J. R., Crane, G. G., Panagiotakos, G., & Zhang, S. (2003). Functional differentiation of hepatocyte-like spheroid structures from putative liver progenitor cells in three-dimensional peptide scaffolds. Differentiation, 71(4-5), 262-270. doi:10.1046/j.1432-0436.2003.7104503.x es_ES
dc.description.references Narmoneva, D. A., Vukmirovic, R., Davis, M. E., Kamm, R. D., & Lee, R. T. (2004). Endothelial Cells Promote Cardiac Myocyte Survival and Spatial Reorganization. Circulation, 110(8), 962-968. doi:10.1161/01.cir.0000140667.37070.07 es_ES
dc.description.references Genové, E., Shen, C., Zhang, S., & Semino, C. E. (2005). The effect of functionalized self-assembling peptide scaffolds on human aortic endothelial cell function. Biomaterials, 26(16), 3341-3351. doi:10.1016/j.biomaterials.2004.08.012 es_ES
dc.description.references Ellis-Behnke, R. G., Liang, Y.-X., You, S.-W., Tay, D. K. C., Zhang, S., So, K.-F., & Schneider, G. E. (2006). Nano neuro knitting: Peptide nanofiber scaffold for brain repair and axon regeneration with functional return of vision. Proceedings of the National Academy of Sciences, 103(13), 5054-5059. doi:10.1073/pnas.0600559103 es_ES
dc.description.references Garreta, E., Genové, E., Borrós,, S., & Semino, C. E. (2006). Osteogenic Differentiation of Mouse Embryonic Stem Cells and Mouse Embryonic Fibroblasts in a Three-Dimensional Self-Assembling Peptide Scaffold. Tissue Engineering, 12(8), 2215-2227. doi:10.1089/ten.2006.12.2215 es_ES
dc.description.references Martínez-Ramos, C., Rodríguez-Pérez, E., Garnes, M. P., Chachques, J. C., Moratal, D., Vallés-Lluch, A., & Monleón Pradas, M. (2014). Design and Assembly Procedures for Large-Sized Biohybrid Scaffolds as Patches for Myocardial Infarct. Tissue Engineering Part C: Methods, 20(10), 817-827. doi:10.1089/ten.tec.2013.0489 es_ES
dc.description.references Diego, R. B., Olmedilla, M. P., Aroca, A. S., Ribelles, J. L. G., Pradas, M. M., Ferrer, G. G., & Sánchez, M. S. (2005). Acrylic scaffolds with interconnected spherical pores and controlled hydrophilicity for tissue engineering. Journal of Materials Science: Materials in Medicine, 16(8), 693-698. doi:10.1007/s10856-005-2604-7 es_ES
dc.description.references Diego, R. B., Estellés, J. M., Sanz, J. A., García-Aznar, J. M., & Sánchez, M. S. (2007). Polymer scaffolds with interconnected spherical pores and controlled architecture for tissue engineering: Fabrication, mechanical properties, and finite element modeling. Journal of Biomedical Materials Research Part B: Applied Biomaterials, 81B(2), 448-455. doi:10.1002/jbm.b.30683 es_ES
dc.description.references Bayes-Genis, A., Soler-Botija, C., Farré, J., Sepúlveda, P., Raya, A., Roura, S., … Belmonte, J. C. I. (2010). Human progenitor cells derived from cardiac adipose tissue ameliorate myocardial infarction in rodents. Journal of Molecular and Cellular Cardiology, 49(5), 771-780. doi:10.1016/j.yjmcc.2010.08.010 es_ES
dc.description.references MARTINEZESTRADA, O., MUNOZSANTOS, Y., JULVE, J., REINA, M., & VILARO, S. (2005). Human adipose tissue as a source of Flk-1 cells: new method of differentiation and expansion. Cardiovascular Research, 65(2), 328-333. doi:10.1016/j.cardiores.2004.11.015 es_ES
dc.description.references Cyganek, L. (2013). Cardiac Progenitor Cells and their Therapeutic Application for Cardiac Repair. Journal of Clinical & Experimental Cardiology, 01(S11). doi:10.4172/2155-9880.s11-008 es_ES
dc.description.references Wang, C., Cao, D., Wang, Q., & Wang, D.-Z. (2011). Synergistic Activation of Cardiac Genes by Myocardin and Tbx5. PLoS ONE, 6(8), e24242. doi:10.1371/journal.pone.0024242 es_ES
dc.description.references Wilson-Rawls, J., Molkentin, J. D., Black, B. L., & Olson, E. N. (1999). Activated Notch Inhibits Myogenic Activity of the MADS-Box Transcription Factor Myocyte Enhancer Factor 2C. Molecular and Cellular Biology, 19(4), 2853-2862. doi:10.1128/mcb.19.4.2853 es_ES
dc.description.references Lockhart, M. M., Wirrig, E. E., Phelps, A. L., Ghatnekar, A. V., Barth, J. L., Norris, R. A., & Wessels, A. (2013). Mef2c Regulates Transcription of the Extracellular Matrix Protein Cartilage Link Protein 1 in the Developing Murine Heart. PLoS ONE, 8(2), e57073. doi:10.1371/journal.pone.0057073 es_ES
dc.description.references Jiang, W., Ma, A., Wang, T., Han, K., Liu, Y., Zhang, Y., … Zheng, X. (2006). Homing and differentiation of mesenchymal stem cells delivered intravenously to ischemic myocardium in vivo: a time-series study. Pflügers Archiv - European Journal of Physiology, 453(1), 43-52. doi:10.1007/s00424-006-0117-y es_ES
dc.description.references Prabhakaran, M. P., Venugopal, J., Kai, D., & Ramakrishna, S. (2011). Biomimetic material strategies for cardiac tissue engineering. Materials Science and Engineering: C, 31(3), 503-513. doi:10.1016/j.msec.2010.12.017 es_ES
dc.description.references Xu, B., Li, Y., Fang, X., Thouas, G. A., Cook, W. D., Newgreen, D. F., & Chen, Q. (2013). Mechanically tissue-like elastomeric polymers and their potential as a vehicle to deliver functional cardiomyocytes. Journal of the Mechanical Behavior of Biomedical Materials, 28, 354-365. doi:10.1016/j.jmbbm.2013.06.005 es_ES
dc.description.references Zhou, J., Shu, Y., Lü, S.-H., Li, J.-J., Sun, H.-Y., Tang, R.-Y., … Wang, C.-Y. (2013). The Spatiotemporal Development of Intercalated Disk in Three-Dimensional Engineered Heart Tissues Based on Collagen/Matrigel Matrix. PLoS ONE, 8(11), e81420. doi:10.1371/journal.pone.0081420 es_ES
dc.description.references Zhang, S., Gelain, F., & Zhao, X. (2005). Designer self-assembling peptide nanofiber scaffolds for 3D tissue cell cultures. Seminars in Cancer Biology, 15(5), 413-420. doi:10.1016/j.semcancer.2005.05.007 es_ES
dc.description.references Santos, E., Hernández, R. M., Pedraz, J. L., & Orive, G. (2012). Novel advances in the design of three-dimensional bio-scaffolds to control cell fate: translation from 2D to 3D. Trends in Biotechnology, 30(6), 331-341. doi:10.1016/j.tibtech.2012.03.005 es_ES
dc.description.references Dégano, I. R., Quintana, L., Vilalta, M., Horna, D., Rubio, N., Borrós, S., … Blanco, J. (2009). The effect of self-assembling peptide nanofiber scaffolds on mouse embryonic fibroblast implantation and proliferation. Biomaterials, 30(6), 1156-1165. doi:10.1016/j.biomaterials.2008.11.021 es_ES
dc.description.references Kanno, S., & Saffitz, J. E. (2001). The role of myocardial gap junctions in electrical conduction and arrhythmogenesis. Cardiovascular Pathology, 10(4), 169-177. doi:10.1016/s1054-8807(01)00078-3 es_ES
dc.description.references Center CHG 1996 es_ES
dc.description.references Messner, B., Baum, H., Fischer, P., Quasthoff, S., & Neumeier, D. (2000). Expression of Messenger RNA of the Cardiac Isoforms of Troponin T and I in Myopathic Skeletal Muscle. American Journal of Clinical Pathology, 114(4), 544-549. doi:10.1309/8kcl-uqrf-6eel-36xk es_ES
dc.description.references Gnecchi, M., Danieli, P., & Cervio, E. (2012). Mesenchymal stem cell therapy for heart disease. Vascular Pharmacology, 57(1), 48-55. doi:10.1016/j.vph.2012.04.002 es_ES


Este ítem aparece en la(s) siguiente(s) colección(ones)

Mostrar el registro sencillo del ítem