65 results on '"Pelacho B"'
Search Results
2. Exosomes/EVs: REAL-TIME BIOPROCESS MONITORING FOR LARGE-SCALE PRODUCTION OF MESENCHYMAL STROMAL CELL-DERIVED EXTRACELLULAR VESICLES
- Author
-
Costa, M., primary, Painho, B., additional, Costa, M., additional, Sousa, C., additional, Carrondo, I., additional, Oltra, E., additional, Pelacho, B., additional, Prosper, F., additional, Isidro, I., additional, Alves, P., additional, and Serra, M., additional
- Published
- 2023
- Full Text
- View/download PDF
3. Topical Administration of a Marine Oil Rich in Pro-Resolving Lipid Mediators Accelerates Wound Healing in Diabetic db/db Mice through Angiogenesis and Macrophage Polarization
- Author
-
Ontoria-Oviedo I, Amaro-Prellezo E, Castellano D, Venegas-Venegas E, Gonzalez-Santos F, Ruiz-Sauri A, Pelacho B, Prosper F, del Caz M, and Sepulveda P
- Subjects
pro-resolving lipid mediators ,angiogenesis ,SPMs ,macrophage polarization ,wound healing ,omega-3 ,diabetic ulcer - Abstract
Impaired wound healing in patients with type 2 diabetes (DM2) is characterized by chronic inflammation, which delays wound closure. Specialized pro-resolving lipid mediators (SPMs) are bioactive molecules produced from essential polyunsaturated fatty acids (PUFAs), principally omega-3 docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA). SPMs are potent regulators of inflammation and have been used to suppress chronic inflammation in peripheral artery disease, non-alcoholic fatty liver disease, and central nervous system syndromes. LIPINOVA(R) is a commercially available safe-grade nutritional supplement made from a fractionated marine lipid concentrate derived from anchovy and sardine oil that is rich in SPMs and EPA, as well as DHA precursors. Here, we assessed the effect of LIPINOVA(R) in wound dressing applications. LIPINOVA(R) showed biocompatibility with keratinocytes and fibroblasts, reduced the abundance of pro-inflammatory macrophages (M phi 1), and promoted in vitro wound closure. Daily application of the marine oil to open wounds made by punch biopsy in db/db mice promoted wound closure by accelerating the resolution of inflammation, inducing neoangiogenesis and M phi 1/M phi 2 macrophage polarization. In conclusion, LIPINOVA(R) displays pro-resolutive properties and could be exploited as a therapeutic agent for the treatment of diabetic ulcers.
- Published
- 2022
4. Targeted delivery controlled release of hepatic growth factor and insulin-like growth factor-1 improves left ventricular repair in a porcine model of myocardial ischemia reperfusion injury
- Author
-
Wu, M, primary, Claus, P, additional, De Buck, S, additional, Veltman, D, additional, Gillijns, H, additional, Holemans, P, additional, Pokreisz, P, additional, Caluwe, E, additional, Estefania, E, additional, Cohen, S, additional, Prosper, F, additional, Pelacho, B, additional, and Janssens, S, additional
- Published
- 2021
- Full Text
- View/download PDF
5. Development of cellularized and functionalized collagen scaffolds for the treatment of myocardial infarction
- Author
-
Pérez-Estenaga, I. (Iñigo) and Pelacho, B. (Beatriz)
- Subjects
Ciencias de la vida::Citología, biología celular [Materias Investigacion] ,Ciencias de la vida::Inmunología [Materias Investigacion] ,Ciencias de la Salud::Microbiología y biología molecular [Materias Investigacion] - Abstract
Cardiovascular diseases (CVD) are the leading cause of death in the developed countries. According to the las report of the European Society of Cardiology, CVD remain the most common cause of death within Europe, accounting for 2.2 million deaths in females and 1.9 million deaths in males. These equate to 47% and 39% of all deaths in females and males, respectively, and almost half of them are caused by Myocardial Infarction (MI). In the last 20 years, in order to overcome the limitations of the conventional treatments, cell therapy, has emerged as a promising therapeutic option for the treatment of MI. In this thesis we have employed Adipose Derived Mesenchymal Stem Cells (ADSC) seeded in a collagen scaffold fabricated under GMP-conditions, to treat the infarcted heart in preclinical MI animal models. First, we assessed the patch safety in rodent models, proving a safe profile of the system, established by tumorigenicity, toxicity and biodistribution studies. Secondly, the collagen scaffold was cellularized with allogeneic ADSC and its immunological effect was analysed both in vitro and in vivo, showing no alloreactive response by the immune cells towards the allogeneic patch. Moreover in vitro, in a human context, the cellularized patch elicited a potent immunomodulatory effect towards lymphocytes by inhibiting their proliferation, phenotypical activation and pro-inflammatory cytokine production. Finally, the functionalization of the collagen membrane with the angiogenic factor SDF-1 was achieved, by the creation of a bilayer collagen scaffold, which confirmed to be biocompatible in vivo and exerted a therapeutic effect when tested in a rat MI model in the shape of cardiac function recovery.
- Published
- 2021
6. P347Epicardial-derived interstitial fibroblasts and bone marrow-derived cell interaction determines post-infarction ventricular remodeling
- Author
-
Perez-Pomares, J M, Ruiz-Villalba, A, Simon, AM, Pogontke, C, Abizanda, G, Castillo, MI, Cano, S, Pelacho, B, Prosper, F, and Segovia, JC
- Published
- 2014
- Full Text
- View/download PDF
7. Delivery of cardiovascular progenitors with biomimetic microcarriers reduces adverse ventricular remodeling in a rat model of chronic myocardial infarction
- Author
-
Garbayo, E., primary, Ruiz-Villalba, A., additional, Hernandez, S.C., additional, Saludas, L., additional, Abizanda, G., additional, Pelacho, B., additional, Roncal, C., additional, Sanchez, B., additional, Palacios, I., additional, Prósper, F., additional, and Blanco-Prieto, M.J., additional
- Published
- 2021
- Full Text
- View/download PDF
8. Nanoparticles loaded with hepatic growth factor and insulin-like growth factor-1 improve left ventricular repair in a porcine model of myocardial Ischemia reperfusion injury
- Author
-
Wu, M, primary, Claus, P, additional, De Buck, S, additional, Veltman, D, additional, Gillijns, H, additional, Holemans, P, additional, Pokreisz, P, additional, Caluwe, E, additional, Colino, E, additional, Cohen, S, additional, Prosper, F, additional, Pelacho, B, additional, and Janssens, S, additional
- Published
- 2020
- Full Text
- View/download PDF
9. Mechanical & biological characterization of collagen membranes cellularized with adipose-derived stem cells for tissue engineering in a rat model of chronic myocardial infarction: OP097
- Author
-
Araña, M, Lanana, S, Pelacho, B, Peña, E, Ochoa, I, Mazo, M, Longo, C, Falkenburg, D, Doblare, M, and Prosper, F
- Published
- 2009
10. Analysis of AAV9 biodistribution, transduction efficiency and AAV-miR-935 cardio-specific overexpression in a murine model of myocardial infarction
- Author
-
García-Olloqui, P. (Paula) and Pelacho, B. (Beatriz)
- Subjects
Ciencias de la vida::Citología, biología celular [Materias Investigacion] ,Ciencias de la Salud::Microbiología y biología molecular [Materias Investigacion] ,Cultivo celular ,Ciencias de la Salud [Materias Investigacion] ,Patología cardiovascular - Abstract
Myocardial infarction (MI) leads to an irreversible loss of cardiac myocytes, which compromises cardiac function. The cellular and molecular mechanisms involved in the ischemic event remain under study, being the cardiac exosomes transfer of non-coding RNAs a key process in this pathology. In this context, a comparative analysis of the exosomal compartment of human cardiac progenitor cells (CPC) was performed in comparison with human bone marrow-mesenchymal stem cells (MSC) and human dermal fibroblasts (HDF). A total of 481 differentially expressed microRNAs (miR) were found, being miR-935 the most differentially expressed in CPC. Analysis of miR-935 in vitro overexpression promoted a positive trend to increment cardiomyocyte survival, in response to oxidative stress. Furthermore, when miR-935 regulation was analyzed in a mouse model of MI disease, miR downregulation was shown five days post-infarct in different cardiac subpopulations. In order to study the role of miR-935 in MI and to achieve an optimal miR-935 overexpression in our mouse model, we developed a robust viral-based method for cardiac-specific miR overexpression. Cardiotropic Serotype 9-Adeno-Associated Virus (AAV9) were used for this purpose and their biodistribution was first determined in mice. Ubiquitous EF1α or the cardiac-specific TnT promoters were combined with Luciferase (Luc) or GFP reporters and administered by intramyocardial or intravenous injection, either in healthy or myocardial-infarcted mice. High transgene expression levels were found in the heart, but not in the liver, of mice receiving AAV-TnT, which was significantly higher after intramyocardial injection regardless of ischemia-induction. On the contrary, high hepatic transgene expression levels were detected with the EF1α-promoter, independently of the administration route and heart damage. Luc expression increased with both promoters in a time-dependent manner, reaching a peak by day 3-7 that was stable for at least 60 days. Moreover, tissue-specific GFP expression was found in cardiomyocytes with the TnT-vector, while minimal cardiac expression was detected with the ubiquitous one. Interestingly, we found that MI greatly increased the transcriptional activity of AAV genomes. Thus, we found AAV9-TnT as a robust and stable vector for cardiac-specific delivery after intramyocardial injection. Finally, the therapeutic potential of this vector in combination with miR-935 was evaluated in a mouse model of myocardial ischemia. Intramyocardial injection of AAV9-TnT-miR935 vector did not significantly improve heart function 60 days post-infarct. However, a slight positive trend in the ejection fraction and a decreased adverse cardiac remodeling are observed, suggesting miR-935 putative cardioprotective role.
- Published
- 2019
11. Electrospun poly(hydroxybutyrate) scaffolds promote engraftment of human skin equivalents via macrophage M2 polarization and angiogenesis
- Author
-
Castellano, D, Sanchis, A, Blanes, M, Pérez del Caz, MD, Ruiz-Sauri, A, Pelacho, B, and Ontoria-Oviedo, I
- Subjects
skin equivalents ,technology, industry, and agriculture ,human skin xenograft ,poly(hydroxybutyrate) ,electrospinning - Abstract
Human dermo-epidermal skin equivalents (DE) comprising in vitro expanded autologous keratinocytes and fibroblasts are a good option for massive burn treatment. However, the lengthy expansion time required to obtain sufficient surface to cover an extensive burn together with the challenging surgical procedure limits their clinical use. The integration of DE and biodegradable scaffolds has been proposed in an effort to enhance their mechanical properties. Here, it is shown that poly(hydroxybutyrate) electrospun scaffolds (PHB) present good biocompatibility both in vitro and in vivo and are superior to poly-epsilon-caprolactone electrospun scaffolds as a substrate for skin reconstruction. Implantation of PHB scaffolds in healthy rats polarized macrophages to an M2-type that promoted constructive in vivo remodelling. Moreover, implantation of DE-PHB composites in a NOD/SCID mouse xenograft model resulted in engraftment accompanied by an increase in angiogenesis that favoured the survival of the human graft. Thus, PHB scaffolds are an attractive substrate for further exploration in skin reconstruction procedures, probably due in part to their greater angiogenic and M2 macrophage polarization properties. Copyright (c) 2017 John Wiley & Sons, Ltd.
- Published
- 2018
12. P5676Safety and immunomodulatory action of epicardial patches combined with allogeneic adipose-derived mesenchymal stem cells in a rodent and porcine model of myocardial infarction
- Author
-
Pelacho, B, primary, Lopez-Diaz De Cerio, A, additional, Inoges, S, additional, Perez-Astenaga, I, additional, Gavira, J J, additional, Abizanda, G, additional, Andreu, E, additional, Crisostomo, V, additional, Bermejo, J, additional, Huss, A, additional, Gil, A G, additional, Koblizek, T, additional, Quintana, L L, additional, Fernandez-Aviles, F, additional, and Prosper, F, additional
- Published
- 2018
- Full Text
- View/download PDF
13. Global position paper on cardiovascular regenerative medicine
- Author
-
Fernández-Avilés, F. (Francisco), Sanz-Ruiz, R. (Ricardo), Climent, A.M. (Andreu M.), Badimon, L. (Lina), Bolli, R. (Roberto), Charron, D. (Dominique), Fuster, V. (Valentin), Janssens, S. (Stefan), Kastrup, J. (Jens), Kim, H.-S. (Hyo-Soo), Lüscher, T.F., Martin, J.F. (John F.), Menasche, P. (Philippe), Simari, P. (Patricio), Stone, G.W. (Gregg), Terzic, A. (Andre), Willerson, J.T. (James), Wu, J.C. (Joseph C.), Joseph, C.W. (C. Wu), Broughton, K. (Kathleen), DiFede, D.L. (Darcy L.), Dimmeler, S. (Stefanie), Madonna, R. (Rosalinda), Penn, M.S. (Marc S.), Sussman, M.A. (Mark A.), Sluijter, J.P.G., Woller, K.C. (Kai C.), Balkan, W. (Wayne), Chamuleau, S.A.J. (Steven), Fernández-Santos, M.E. (Maria Eugenia), Goliasch, G. (Georg), Gyöngyösi, M. (Mariann), Hare, J.M. (Joshua M.), Tompkins, B.A. (Bryon A.), Winkler, J. (Johannes), Bayés-Genis, A. (Antoni), Henry, T.D. (Timothy), Taylor, D.A. (Doris), Lerman, A. (Amir), Pelacho, B. (Beatriz), Prosper, F. (Felipe), Perin, E.C. (Emerson ), Pompilio, G. (Giulio), Gersh, B.J. (Bernard), Bartúnek, J. (Jozef), Duckers, E. (Eric), Ferdinandy, P. (Péter), Losordo, D.W. (Douglas W.), Čanchez, P.L. (Pedro L.), Sherman, W. (Warren), Wojakowski, W. (Wojtek), Zeiher, A.M. (Andreas), Roncalli, J. (Jérôme), Mathur, A. (Anthony), Crea, F. (Filippo), D'Amario, D. (Domenico), Povsic, T.J. (Thomas J.), Traverse, J.H. (Jay), Ylä-Herttuala, S. (Seppo), Fernández-Avilés, F. (Francisco), Sanz-Ruiz, R. (Ricardo), Climent, A.M. (Andreu M.), Badimon, L. (Lina), Bolli, R. (Roberto), Charron, D. (Dominique), Fuster, V. (Valentin), Janssens, S. (Stefan), Kastrup, J. (Jens), Kim, H.-S. (Hyo-Soo), Lüscher, T.F., Martin, J.F. (John F.), Menasche, P. (Philippe), Simari, P. (Patricio), Stone, G.W. (Gregg), Terzic, A. (Andre), Willerson, J.T. (James), Wu, J.C. (Joseph C.), Joseph, C.W. (C. Wu), Broughton, K. (Kathleen), DiFede, D.L. (Darcy L.), Dimmeler, S. (Stefanie), Madonna, R. (Rosalinda), Penn, M.S. (Marc S.), Sussman, M.A. (Mark A.), Sluijter, J.P.G., Woller, K.C. (Kai C.), Balkan, W. (Wayne), Chamuleau, S.A.J. (Steven), Fernández-Santos, M.E. (Maria Eugenia), Goliasch, G. (Georg), Gyöngyösi, M. (Mariann), Hare, J.M. (Joshua M.), Tompkins, B.A. (Bryon A.), Winkler, J. (Johannes), Bayés-Genis, A. (Antoni), Henry, T.D. (Timothy), Taylor, D.A. (Doris), Lerman, A. (Amir), Pelacho, B. (Beatriz), Prosper, F. (Felipe), Perin, E.C. (Emerson ), Pompilio, G. (Giulio), Gersh, B.J. (Bernard), Bartúnek, J. (Jozef), Duckers, E. (Eric), Ferdinandy, P. (Péter), Losordo, D.W. (Douglas W.), Čanchez, P.L. (Pedro L.), Sherman, W. (Warren), Wojakowski, W. (Wojtek), Zeiher, A.M. (Andreas), Roncalli, J. (Jérôme), Mathur, A. (Anthony), Crea, F. (Filippo), D'Amario, D. (Domenico), Povsic, T.J. (Thomas J.), Traverse, J.H. (Jay), and Ylä-Herttuala, S. (Seppo)
- Published
- 2017
- Full Text
- View/download PDF
14. Global position paper on cardiovascular regenerative medicine
- Author
-
Fernandez-Aviles, F, Sanz-Ruiz, R, Climent, AM, Badimon, L, Bolli, R, Charron, D, Fuster, V, Janssens, S, Kastrup, J, Kim, HS, Luscher, TF, Martin, JF, Menasche, P, Simari, RD, Stone, GW, Terzic, A, Willerson, JT, Wu, JC, Broughton, K, DiFede, DL, Dimmeler, S, Madonna, R, Penn, MS, Sussman, MA, Sluijter, JPG, Wollert, KC, Balkan, W, Chamuleau, S, Fernandez-Santos, ME, Goliasch, G, Gyongyosi, M, Hare, JM, Tompkins, BA, Winkler, J, Bayes-Genis, A, Henry, TD, Taylor, DA, Lerman, A, Pelacho, B, Prosper, F, Perin, EC, Pompilio, G, Gersh, B, Bartunek, J, Duckers, Eric, Ferdinandy, P, Losordo, DW, Sanchez, PL, Sherman, W, Wojakowski, W, Zeiher, A, Roncalli, J, Mathur, A, Crea, F, D'Amario, D, Povsic, TJ, Traverse, J, Yla-Herttuala, S, Fernandez-Aviles, F, Sanz-Ruiz, R, Climent, AM, Badimon, L, Bolli, R, Charron, D, Fuster, V, Janssens, S, Kastrup, J, Kim, HS, Luscher, TF, Martin, JF, Menasche, P, Simari, RD, Stone, GW, Terzic, A, Willerson, JT, Wu, JC, Broughton, K, DiFede, DL, Dimmeler, S, Madonna, R, Penn, MS, Sussman, MA, Sluijter, JPG, Wollert, KC, Balkan, W, Chamuleau, S, Fernandez-Santos, ME, Goliasch, G, Gyongyosi, M, Hare, JM, Tompkins, BA, Winkler, J, Bayes-Genis, A, Henry, TD, Taylor, DA, Lerman, A, Pelacho, B, Prosper, F, Perin, EC, Pompilio, G, Gersh, B, Bartunek, J, Duckers, Eric, Ferdinandy, P, Losordo, DW, Sanchez, PL, Sherman, W, Wojakowski, W, Zeiher, A, Roncalli, J, Mathur, A, Crea, F, D'Amario, D, Povsic, TJ, Traverse, J, and Yla-Herttuala, S
- Published
- 2017
15. Analysis of the regenerative potential of the induced pluripotent stem cells in a model of acute myocardial infarction in mice
- Author
-
Iglesias-García, O. (Olalla), Pelacho, B. (Beatriz), and Prosper, F. (Felipe)
- Subjects
Ciencias de la Vida [Materias Investigacion] ,Biología molecular ,Biología celular ,Cultivo celular - Abstract
Principal limitation for generating cardiomyocytes from stem cells is that differentiated cells generally present electrophysiological immature and heterogeneous phenotype, which may hamper their in vitro and in vivo application. The purpose of this study was to examine the effect of NRG-1b and DMSO in the in vitro generation of mature working-type cardiomyocytes from induced pluripotent stem (iPS) cells and to determine their contribution to the cardiac tissue regeneration after acute myocardial infarction (AMI). iPS cells were derived from α-MHC-GFP mice fibroblasts and were in vitro differentiated towards cardiomyocytes by DMSO and/or NRG-1b treatment. iPS cardiac specification and maturation was analyzed by Q-RT-PCR, immunofluorescence, electronic microscopy and patch-clamp techniques. The iPS-derived cardiomyocytes (n=15) or culture-medium as control (n=13) were injected into the peri-infarct region of mice hearts following coronary artery ligation. Echocardiography and histology assessments were performed from 1-8 weeks post-transplantation. iPS cells showed early and robust in vitro cardiogenesis with cardiac gene and protein expression in all cases. Electrophysiological studies demonstrated a more mature ventricular-like cardiac phenotype when cells were treated with NRG-1b and DMSO than with DMSO-treatment alone. In vivo studies in the AMI mouse model demonstrated that iPS-derived CMs preserved cardiac function and induced a positive heart tissue remodeling. Moreover, iPS-CMs engrafted and electromechanically couple into the heart tissue. The combination of NRG-1b and DMSO induced iPS cells differentiation towards mature ventricular-like cardiac cells which, when transplanted in a model of AMI, contributed to preserve the cardiac function and tissue viability.
- Published
- 2015
16. Substrate Stiffness and Composition Specifically Direct Differentiation of Induced Pluripotent Stem Cells
- Author
-
Macrí-Pellizzeri L, Pelacho B, Sancho A, Iglesias-García O, Simón-Yarza AM, Soriano-Navarro M, González-Granero S, García-Verdugo JM, De-Juan-Pardo EM, and Prosper F
- Abstract
Substrate stiffness, biochemical composition, and matrix topography deeply influence cell behavior, guiding motility, proliferation, and differentiation responses. The aim of this work was to determine the effect that the stiffness and protein composition of the underlying substrate has on the differentiation of induced pluripotent stem (iPS) cells and the potential synergy with specific soluble cues. With that purpose, murine iPS-derived embryoid bodies (iPS-EBs) were seeded on fibronectin- or collagen I-coated polyacrylamide (pAA) gels of tunable stiffness (0.6, 14, and 50 kPa) in the presence of basal medium; tissue culture polystyrene plates were employed as control. Specification of iPS cells toward the three germ layers was analyzed, detecting an increase of tissue-specific gene markers in the pAA matrices. Interestingly, soft matrix (0.6 kPa) coated with fibronectin favored differentiation toward cardiac and neural lineages and, in the case of neural differentiation, the effect was potentiated by the addition of specific soluble factors. The generation of mature astrocytes, neural cells, and cardiomyocytes was further proven by immunofluorescence and transmission electron microscopy. In summary, this work emphasizes the importance of using biomimetic matrices to accomplish a more specific and mature differentiation of stem cells for future therapeutic applications.
- Published
- 2015
17. Hematopoietic reconstitution by multipotent adult progenitor cells: Precursors to long-term hematopoietic stem cells
- Author
-
Serafini, M, Dylla, S, Oki, M, Heremans, Y, Tolar, J, Jiang, Y, Buckley, S, Pelacho, B, Burns, T, Frommer, S, Rossi, D, Bryder, D, Panoskaltsis-Mortari, A, O'Shaughnessy, M, Nelson-Holte, M, Fine, G, Weissman, I, Blazar, B, Verfaillie, C, Serafini M, Dylla SJ, Oki M, Heremans Y, Tolar J, Jiang YH, Buckley SM, Pelacho B, Burns TC, Frommer S, Rossi DJ, Bryder D, Panoskaltsis-Mortari A, O'Shaughnessy MJ, Nelson-Holte M, Fine GC, Weissman IL, Blazar BR, Verfaillie CM, Serafini, M, Dylla, S, Oki, M, Heremans, Y, Tolar, J, Jiang, Y, Buckley, S, Pelacho, B, Burns, T, Frommer, S, Rossi, D, Bryder, D, Panoskaltsis-Mortari, A, O'Shaughnessy, M, Nelson-Holte, M, Fine, G, Weissman, I, Blazar, B, Verfaillie, C, Serafini M, Dylla SJ, Oki M, Heremans Y, Tolar J, Jiang YH, Buckley SM, Pelacho B, Burns TC, Frommer S, Rossi DJ, Bryder D, Panoskaltsis-Mortari A, O'Shaughnessy MJ, Nelson-Holte M, Fine GC, Weissman IL, Blazar BR, and Verfaillie CM
- Abstract
For decades, in vitro expansion of transplantable hematopoietic stem cells (HSCs) has been an elusive goal. Here, we demonstrate that multipotent adult progenitor cells (MAPCs), isolated from green fluorescent protein (GFP)-transgenic mice and expanded in vitro for >40-80 population doublings, are capable of multilineage hematopoietic engraftment of immunodeficient mice. Among MAPC-derived GFP+CD45.2+ cells in the bone marrow of engrafted mice, HSCs were present that could radioprotect and reconstitute multilineage hematopoiesis in secondary and tertiary recipients, as well as myeloid and lymphoid hematopoietic progenitor subsets and functional GFP + MAPC-derived lymphocytes that were functional. Although hematopoietic contribution by MAPCs was comparable to control KTLS HSCs, approximately 103-fold more MAPCs were required for efficient engraftment. Because GFP+ host-derived CD45.1+ cells were not observed, fusion is not likely to account for the generation of HSCs by MAPCs. JEM
- Published
- 2007
18. Aplicación terapéutica de la bioingeniería mediante la combinación de células madre y matrices extracelulares en un modelo de infarto de miocardio en rata
- Author
-
Araña, M. (Miriam), Prosper, F. (Felipe), and Pelacho, B. (Beatriz)
- Subjects
Corazón ,Ciencias de la vida [Materias Investigacion] ,Rata Sprague‐Dawley ,Infartos de miocardio ,Células madres - Abstract
Los objetivos del presente trabajo han sido los siguientes: 1. Aislar y caracterizar la población de células madre derivadas del tejido adiposo (ADSC), a partir de grasa de rata Sprague‐Dawley. 2. Caracterizar el comportamiento biológico y mecánico de distintos tipos de membranas de colágeno y determinar su biocompatibilidad in vivo. 3. Analizar de forma comparativa, el potencial terapéutico de las ADSC en el corazón, al ser trasplantadas como parche celular (mediante su previa adhesión a una membrana de colágeno) o inyectadas sin soporte, en un modelo de infarto crónico de miocardio en rata. 4. Determinar los mecanismos implicados en la posible acción terapéutica de las membranas celularizadas con ADSC.
- Published
- 2014
19. Vascular endothelial growth factor-delivery systems for cardiac repair: An overview
- Author
-
Simon-Yarza, T. (Teresa), Formiga, F.R. (Fabio R.), Tamayo, E. (Esther), Pelacho, B. (Beatriz), Prosper, F. (Felipe), and Blanco-Prieto, M.J. (María José)
- Subjects
Protein delivery ,Tissue engineering ,Angiogenesis ,Cardiovascular disease ,VEGF - Abstract
Since the discovery of the Vascular Endothelial Growth Factor (VEGF) and its leading role in the angiogenic process, this has been seen as a promising molecule for promoting neovascularization in the infarcted heart. However, even though several clinical trials were initiated, no therapeutic effects were observed, due in part to the short half life of this factor when administered directly to the tissue. In this context, drug delivery systems appear to offer a promising strategy to overcome limitations in clinical trials of VEGF. The aim of this paper is to review the principal drug delivery systems that have been developed to administer VEGF in cardiovascular disease. Studies published in the last 5 years are reviewed and the main features of these systems are explained. The tissue engineering concept is introduced as a therapeutic alternative that holds promise for the near future.
- Published
- 2012
20. Growth factor loaded-microparticles as a tool for cardiac repair
- Author
-
Rocha, F. (Fabio), Pelacho, B. (Beatriz), and Blanco-Prieto, M.J. (María José)
- Subjects
Farmacia [Materias Investigacion] - Abstract
The clinical trials performed in patients with myocardial infarction and based on the intravascular injection of growth factors have failed, owing, among other reasons, to protein instability after injection. The hypothesis of this research is that a local controlled release of the growth factors by using a polymeric delivery system could protect the growth factors from degradation and stimulate cardiac repair. To test this, the following specific objectives were proposed: 1. Design, development and physico-chemical characterization of PLGA microparticles intended for intramyocardial administration. In vivo compatibility assessment of the developed microparticles with the cardiac tissue. 2. Development of VEGF165 loaded PLGA microparticles, in vitro characterization and assessment of the potential benefit of the VEGF165-microparticles in an acute rat myocardial ischemia– reperfusion model. 3. Development of FGF-1 and NRG-1 into PLGA microparticles, in vitro characterization and evaluation of the therapeutic potential of FGF-1 and/or NRG-1 cytokines delivered from PLGA microparticles in a rat myocardial infarction model.
- Published
- 2011
21. Controlled delivery of fibroblast growth factor-1 and neuregulin-1 from biodegradable microparticles promotes cardiac repair in a rat myocardial infarction model through activation of endogenous regeneration
- Author
-
Formiga, F.R. (Fabio R.), Pelacho, B. (Beatriz), Garbayo, E. (Elisa), Imbuluzqueta, I. (Izaskun), Diaz-Herraez, P. (Paula), Abizanda, G. (Gloria), Gavira, J.J. (Juan José), Simon-Yarza, T. (Teresa), Albiasu, E. (Edurne), Tamayo, E. (Esther), Prosper, F. (Felipe), Blanco-Prieto, M.J. (María José), Formiga, F.R. (Fabio R.), Pelacho, B. (Beatriz), Garbayo, E. (Elisa), Imbuluzqueta, I. (Izaskun), Diaz-Herraez, P. (Paula), Abizanda, G. (Gloria), Gavira, J.J. (Juan José), Simon-Yarza, T. (Teresa), Albiasu, E. (Edurne), Tamayo, E. (Esther), Prosper, F. (Felipe), and Blanco-Prieto, M.J. (María José)
- Abstract
Acidic fibroblast growth factor (FGF1) and neuregulin-1 (NRG1) are growth factors involved in cardiac development and regeneration. Microparticles (MPs) mediate cytokine sustained release, and can be utilized to overcome issues related to the limited therapeutic protein stability during systemic administration. We sought to examine whether the administration of microparticles (MPs) containing FGF1 and NRG1 could promote cardiac regeneration in a myocardial infarction (MI) rat model. We investigated the possible underlying mechanisms contributing to the beneficial effects of this therapy, especially those linked to endogenous regeneration. FGF1- and NRG1-loaded MPs were prepared using a multiple emulsion solvent evaporation technique. Seventy-three female Sprague-Dawley rats underwent permanent left anterior descending coronary artery occlusion, and MPs were intramyocardially injected in the peri-infarcted zone four days later. Cardiac function, heart tissue remodeling, revascularization, apoptosis, cardiomyocyte proliferation, and stem cell homing were evaluated one week and three months after treatment. MPs were shown to efficiently encapsulate FGF1 and NRG1, releasing the bioactive proteins in a sustained manner. Three months after treatment, a statistically significant improvement in cardiac function was detected in rats treated with growth factor-loaded MPs (FGF1, NRG1, or FGF1/NRG1). The therapy led to inhibition of cardiac remodeling with smaller infarct size, a lower fibrosis degree and induction of tissue revascularization. Cardiomyocyte proliferation and progenitor cell recruitment was detected. Our data support the therapeutic benefit of NRG1 and FGF1 when combined with protein delivery systems for cardiac regeneration. This approach could be scaled up for use in pre-clinical and clinical studies.
- Published
- 2014
22. New strategies for echocardiographic evaluation of left ventricular function in a mouse model of long-term myocardial infarction
- Author
-
Benavides, C. (Carolina), Corbacho, D. (David), Iglesias, O. (Olalla), Pelacho, B. (Beatriz), Albiasu, E. (Edurne), Castaño, S. (Sara), Muñoz-Barrutia, A. (Arrate), Prosper, F. (Felipe), Ortiz-de-Solorzano, C. (Carlos), Benavides, C. (Carolina), Corbacho, D. (David), Iglesias, O. (Olalla), Pelacho, B. (Beatriz), Albiasu, E. (Edurne), Castaño, S. (Sara), Muñoz-Barrutia, A. (Arrate), Prosper, F. (Felipe), and Ortiz-de-Solorzano, C. (Carlos)
- Abstract
In summary, we have performed a complete characterization of LV post-infarction remodeling in a DBA/2J mouse model of MI, using parameters adapted to the particular characteristics of the model In the future, this well characterized model will be used in both investigative and pharmacological studies that require accurate quantitative monitoring of cardiac recovery after myocardial infarction.
- Published
- 2014
23. Can bone marrow-derived multipotent adult progenitor cells regenerate infarcted myocardium?
- Author
-
AGBULUT, O, primary, MAZO, M, additional, BRESSOLLE, C, additional, GUTIERREZ, M, additional, AZARNOUSH, K, additional, SABBAH, L, additional, NIEDERLANDER, N, additional, ABIZANDA, G, additional, ANDREU, E, additional, and PELACHO, B, additional
- Published
- 2006
- Full Text
- View/download PDF
24. Pemphigus vulgaris autoantibodies induce apoptosis in HaCaT keratinocytes
- Author
-
Pelacho, B., primary, Natal, C., additional, España, A., additional, Sánchez-Carpintero, I., additional, Iraburu, M.J., additional, and López-Zabalza, M.J., additional
- Published
- 2004
- Full Text
- View/download PDF
25. P347 Epicardial-derived interstitial fibroblasts and bone marrow-derived cell interaction determines post-infarction ventricular remodeling.
- Author
-
Perez-Pomares, J M, Ruiz-Villalba, A, Simon, AM, Pogontke, C, Abizanda, G, Castillo, MI, Cano, S, Pelacho, B, Prosper, F, and Segovia, JC
- Subjects
MYOCARDIAL infarction ,FIBROBLASTS ,BONE marrow cells ,CELL communication ,VENTRICULAR remodeling ,HEART failure - Abstract
Myocardial infarction is a prevalent cardiovascular disease. Mechanisms of repair in the post-infarcted heart include a progressive fibrosis that severely affects cardiac performance, eventually leading to cardiac failure. Cardiac fibrosis in the context of ventricular remodeling after infarction depends on fibroblasts of the cardiac interstitium (cardiac fibroblasts), a heterogeneous population of cells which, upon interaction with other interstitial cell types, initiates a massive deposition of extracellular matrix promoting the formation of a characteristic scar. In this work we have studied the cellular components of the cardiac interstitium from the embryo to the adult. Our results show that Wilms tumor supresor (Wt1) positive epicardial-derived mesenchymal cells pioneer the formation of the cardiac interstitium along embryogenesis, followed by the peri- and post-natal incorporation of bone-marrow derived cells. Adult epicardial-derived cells robustly differentiate into cardiac fibroblasts under normal and pathologic conditions, and become the predominant fibroblast type in the post-infarction scar. Furthermore, epicardial-derived cardiac fibroblasts are shown to display stromal properties respect to bone marrow-derived cells, critically contributing to the homing and persistence of circulating cells after infarction. [ABSTRACT FROM PUBLISHER]
- Published
- 2014
- Full Text
- View/download PDF
26. 307 - Exosomes/EVs: REAL-TIME BIOPROCESS MONITORING FOR LARGE-SCALE PRODUCTION OF MESENCHYMAL STROMAL CELL-DERIVED EXTRACELLULAR VESICLES.
- Author
-
Costa, M., Painho, B., Sousa, C., Carrondo, I., Oltra, E., Pelacho, B., Prosper, F., Isidro, I., Alves, P., and Serra, M.
- Subjects
- *
BIOTECHNOLOGICAL process monitoring , *EXTRACELLULAR vesicles , *EXOSOMES , *VESICLES (Cytology) , *COATED vesicles - Published
- 2023
- Full Text
- View/download PDF
27. 190 - Mesenchymal Stem/Stromal Cells: SCALABLE AND TRANSIENT GENE EXPRESSION IN MESENCHYMAL STROMAL CELLS EXPANDED IN STIRRED-TANK BIOREACTORS TOWARDS THE TREATMENT OF MYOCARDIAL INFARCTION.
- Author
-
Painho, B., Costa, M., Sousa, C., Oltra, E., Arderiu, G., Badimon, L., Pelacho, B., Prosper, F., Alves, P., and Serra, M.
- Subjects
- *
STROMAL cells , *MYOCARDIAL infarction , *GENE expression , *BIOREACTORS - Published
- 2023
- Full Text
- View/download PDF
28. Fibroblast Diversity and Epigenetic Regulation in Cardiac Fibrosis.
- Author
-
Aguado-Alvaro LP, Garitano N, and Pelacho B
- Subjects
- Humans, Animals, Myocardium metabolism, Myocardium pathology, DNA Methylation, Epigenesis, Genetic, Fibrosis, Fibroblasts metabolism, Fibroblasts pathology
- Abstract
Cardiac fibrosis, a process characterized by excessive extracellular matrix (ECM) deposition, is a common pathological consequence of many cardiovascular diseases (CVDs) normally resulting in organ failure and death. Cardiac fibroblasts (CFs) play an essential role in deleterious cardiac remodeling and dysfunction. In response to injury, quiescent CFs become activated and adopt a collagen-secreting phenotype highly contributing to cardiac fibrosis. In recent years, studies have been focused on the exploration of molecular and cellular mechanisms implicated in the activation process of CFs, which allow the development of novel therapeutic approaches for the treatment of cardiac fibrosis. Transcriptomic analyses using single-cell RNA sequencing (RNA-seq) have helped to elucidate the high cellular diversity and complex intercellular communication networks that CFs establish in the mammalian heart. Furthermore, a significant body of work supports the critical role of epigenetic regulation on the expression of genes involved in the pathogenesis of cardiac fibrosis. The study of epigenetic mechanisms, including DNA methylation, histone modification, and chromatin remodeling, has provided more insights into CF activation and fibrotic processes. Targeting epigenetic regulators, especially DNA methyltransferases (DNMT), histone acetylases (HAT), or histone deacetylases (HDAC), has emerged as a promising approach for the development of novel anti-fibrotic therapies. This review focuses on recent transcriptomic advances regarding CF diversity and molecular and epigenetic mechanisms that modulate the activation process of CFs and their possible clinical applications for the treatment of cardiac fibrosis.
- Published
- 2024
- Full Text
- View/download PDF
29. Generation of heart and vascular system in rodents by blastocyst complementation.
- Author
-
Coppiello G, Barlabé P, Moya-Jódar M, Abizanda G, Pogontke C, Barreda C, Iglesias E, Linares J, Arellano-Viera E, Larequi E, San Martín-Úriz P, Carvajal-Vergara X, Pelacho B, Mazo MM, Pérez-Pomares JM, Ruiz-Villalba A, Ullate-Agote A, Prósper F, and Aranguren XL
- Subjects
- Animals, Mice, Rats, Rodentia, Endothelial Cells, Blastocyst, Myocytes, Cardiac, Pluripotent Stem Cells
- Abstract
Generating organs from stem cells through blastocyst complementation is a promising approach to meet the clinical need for transplants. In order to generate rejection-free organs, complementation of both parenchymal and vascular cells must be achieved, as endothelial cells play a key role in graft rejection. Here, we used a lineage-specific cell ablation system to produce mouse embryos unable to form both the cardiac and vascular systems. By mouse intraspecies blastocyst complementation, we rescued heart and vascular system development separately and in combination, obtaining complemented hearts with cardiomyocytes and endothelial cells of exogenous origin. Complemented chimeras were viable and reached adult stage, showing normal cardiac function and no signs of histopathological defects in the heart. Furthermore, we implemented the cell ablation system for rat-to-mouse blastocyst complementation, obtaining xenogeneic hearts whose cardiomyocytes were completely of rat origin. These results represent an advance in the experimentation towards the in vivo generation of transplantable organs., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
30. Compartmentalized drug localization studies in extracellular vesicles for anticancer therapy.
- Author
-
Pitchaimani A, Ferreira M, Palange A, Pannuzzo M, De Mei C, Spano R, Marotta R, Pelacho B, Prosper F, and Decuzzi P
- Abstract
In the development of therapeutic extracellular vesicles (EVs), drug encapsulation efficiencies are significantly lower when compared with synthetic nanomedicines. This is due to the hierarchical structure of the EV membrane and the physicochemical properties of the candidate drug (molecular weight, hydrophilicity, lipophilicity, and so on). As a proof of concept, here we demonstrated the importance of drug compartmentalization in EVs as an additional parameter affecting the therapeutic potential of drug-loaded EVs. In human adipose mesenchymal stem cell (hADSC) derived EVs, we performed a comparative drug loading analysis using two formulations of the same chemotherapeutic molecule - free doxorubicin (DOX) and 1,2-distearoyl- sn-glycero -3-phosphoethanolamine (DSPE) lipid-conjugated doxorubicin (L-DOX) - to enhance the intracellular uptake and therapeutic efficacy. By nano surface energy transfer (NSET) and molecular simulation techniques, along with cryo-TEM analysis, we confirmed the differential compartmentalization of these two molecules in hADSC EVs. L-DOX was preferentially adsorbed onto the surface of the EV, due to its higher lipophilicity, whereas free DOX was mostly encapsulated within the EV core. Also, the L-DOX loaded EV (LDOX@EV) returned an almost three-fold higher DOX content as compared to the free DOX loaded EV (DOX@EV), for a given input mass of drug. Based on the cellular investigations, L-DOX@EV showed higher cell internalization than DOX@EV. Also, in comparison with free L-DOX, the magnitude of therapeutic potential enhancement displayed by the surface compartmentalized L-DOX@EV is highly promising and can be exploited to overcome the sensitivity of many potential drugs, which are impermeable in nature. Overall, this study illustrates the significance of drug compartmentalization in EVs and how this could affect intracellular delivery, loading efficiency, and therapeutic effect. This will further lay the foundation for the future systematic investigation of EV-based biotherapeutic delivery platforms for personalized medicine., Competing Interests: All authors declared no conflict of interest., (This journal is © The Royal Society of Chemistry.)
- Published
- 2023
- Full Text
- View/download PDF
31. Cardiac Progenitor Cell Exosomal miR-935 Protects against Oxidative Stress.
- Author
-
Aguilar S, García-Olloqui P, Amigo-Morán L, Torán JL, López JA, Albericio G, Abizanda G, Herrero D, Vales Á, Rodríguez-Diaz S, Higuera M, García-Martín R, Vázquez J, Mora C, González-Aseguinolaza G, Prosper F, Pelacho B, and Bernad A
- Abstract
Oxidative stress-induced myocardial apoptosis and necrosis are critically involved in ischemic infarction, and several sources of extracellular vesicles appear to be enriched in therapeutic activities. The central objective was to identify and validate the differential exosome miRNA repertoire in human cardiac progenitor cells (CPC). CPC exosomes were first analyzed by LC-MS/MS and compared by RNAseq with exomes of human mesenchymal stromal cells and human fibroblasts to define their differential exosome miRNA repertoire (exo-miR
SEL ). Proteomics demonstrated a highly significant representation of cardiovascular development functions and angiogenesis in CPC exosomes, and RNAseq analysis yielded about 350 different miRNAs; among the exo-miRSEL population, miR-935 was confirmed as the miRNA most significantly up-regulated; interestingly, miR-935 was also found to be preferentially expressed in mouse primary cardiac Bmi1+high CPC, a population highly enriched in progenitors. Furthermore, it was found that transfection of an miR-935 antagomiR combined with oxidative stress treatment provoked a significant increment both in apoptotic and necrotic populations, whereas transfection of a miR-935 mimic did not modify the response. Conclusion. miR-935 is a highly differentially expressed miRNA in exo-miRSEL , and its expression reduction promotes oxidative stress-associated apoptosis. MiR-935, together with other exosomal miRNA members, could counteract oxidative stress-related apoptosis, at least in CPC surroundings.- Published
- 2023
- Full Text
- View/download PDF
32. Microstructured Polymeric Fabrics Modulating the Paracrine Activity of Adipose-Derived Stem Cells.
- Author
-
Grilli F, Albanesi E, Pelacho B, Prosper F, Decuzzi P, and Di Mascolo D
- Subjects
- Humans, Polylactic Acid-Polyglycolic Acid Copolymer, Lactic Acid chemistry, Tissue Engineering, Cells, Cultured, Collagen chemistry, Stem Cells ultrastructure, Tissue Scaffolds chemistry, Polyglycolic Acid chemistry
- Abstract
The deposition of stem cells at sites of injury is a clinically relevant approach to facilitate tissue repair and angiogenesis. However, insufficient cell engraftment and survival require the engineering of novel scaffolds. Here, a regular network of microscopic poly(lactic-co-glycolic acid) (PLGA) filaments was investigated as a promising biodegradable scaffold for human Adipose-Derived Stem Cell (hADSC) tissue integration. Via soft lithography, three different microstructured fabrics were realized where 5 × 5 and 5 × 3 μm PLGA 'warp' and 'weft' filaments crossed perpendicularly with pitch distances of 5, 10 and 20 μm. After hADSC seeding, cell viability, actin cytoskeleton, spatial organization and the secretome were characterized and compared to conventional substrates, including collagen layers. On the PLGA fabric, hADSC re-assembled to form spheroidal-like structures, preserving cell viability and favoring a nonlinear actin organization. Moreover, the secretion of specific factors involved in angiogenesis, the remodeling of the extracellular matrix and stem cell homing was favored on the PLGA fabric as compared to that which occurred on conventional substrates. The paracrine activity of hADSC was microstructure-dependent, with 5 μm PLGA fabric enhancing the expression of factors involved in all three processes. Although more studies are needed, the proposed PLGA fabric would represent a promising alternative to conventional collagen substrates for stem cell implantation and angiogenesis induction.
- Published
- 2023
- Full Text
- View/download PDF
33. Comparative Evaluation of Inducible Cre Mouse Models for Fibroblast Targeting in the Healthy and Infarcted Myocardium.
- Author
-
Aguado-Alvaro LP, Garitano N, Abizanda G, Larequi E, Prosper F, and Pelacho B
- Abstract
Several Cre recombinase transgenic mouse models have been generated for cardiac fibroblast (CF) tracking and heart regulation. However, there is still no consensus on the ideal mouse model to optimally identify and/or regulate these cells. Here, a comparative evaluation of the efficiency and specificity of the indirect reporter Cre-loxP system was carried out in three of the most commonly used fibroblast reporter transgenic mice (Pdgfr a -CreERT2, Col1a1-CreERT2 and PostnMCM) under healthy and ischemic conditions, to determine their suitability in in vivo studies of cardiac fibrosis. We demonstrate optimal Cre recombinase activity in CF (but also, although moderate, in endothelial cells (ECs)) derived from healthy and infarcted hearts in the PDGFR a -creERT2 mouse strain. In contrast, no positive reporter signal was found in CF derived from the Col1a1-CreERT2 mice. Finally, in the PostnMCM line, fluorescent reporter expression was specifically detected in activated CF but not in EC, which leads us to conclude that it may be the most reliable model for future studies on cardiovascular disease. Importantly, no lethality or cardiac fibrosis were induced after tamoxifen administration at the established doses, either in healthy or infarcted mice of the three fibroblast reporter lineages. This study lays the groundwork for future efficient in vivo CF tracking and functional analyses.
- Published
- 2022
- Full Text
- View/download PDF
34. Preclinical Evaluation of the Safety and Immunological Action of Allogeneic ADSC-Collagen Scaffolds in the Treatment of Chronic Ischemic Cardiomyopathy.
- Author
-
López-Díaz de Cerio A, Perez-Estenaga I, Inoges S, Abizanda G, Gavira JJ, Larequi E, Andreu E, Rodriguez S, Gil AG, Crisostomo V, Sanchez-Margallo FM, Bermejo J, Jauregui B, Quintana L, Fernández-Avilés F, Pelacho B, and Prósper F
- Abstract
The use of allogeneic adipose-derived mesenchymal stromal cells (alloADSCs) represents an attractive approach for treating myocardial infarction (MI). Furthermore, adding a natural support improves alloADSCs engraftment and survival in heart tissues, leading to a greater therapeutic effect. We aimed to examine the safety and immunological reaction induced by epicardial implantation of a clinical-grade collagen scaffold (CS) seeded with alloADSCs for its future application in humans. Thus, cellularized scaffolds were myocardially or subcutaneously implanted in immunosuppressed rodent models. The toxicological parameters were not significantly altered, and tumor formation was not found over the short or long term. Furthermore, biodistribution analyses in the infarcted immunocompetent rats displayed cell engraftment in the myocardium but no migration to other organs. The immunogenicity of alloADSC-CS was also evaluated in a preclinical porcine model of chronic MI; no significant humoral or cellular alloreactive responses were found. Moreover, CS cellularized with human ADSCs cocultured with human allogeneic immune cells produced no alloreactive response. Interestingly, alloADSC-CS significantly inhibited lymphocyte responses, confirming its immunomodulatory action. Thus, alloADSC-CS is likely safe and does not elicit any alloreactive immunological response in the host. Moreover, it exerts an immunomodulatory action, which supports its translation to a clinical setting.
- Published
- 2021
- Full Text
- View/download PDF
35. Single-Cell RNA Sequencing Analysis Reveals a Crucial Role for CTHRC1 (Collagen Triple Helix Repeat Containing 1) Cardiac Fibroblasts After Myocardial Infarction.
- Author
-
Ruiz-Villalba A, Romero JP, Hernández SC, Vilas-Zornoza A, Fortelny N, Castro-Labrador L, San Martin-Uriz P, Lorenzo-Vivas E, García-Olloqui P, Palacio M, Gavira JJ, Bastarrika G, Janssens S, Wu M, Iglesias E, Abizanda G, de Morentin XM, Lasaga M, Planell N, Bock C, Alignani D, Medal G, Prudovsky I, Jin YR, Ryzhov S, Yin H, Pelacho B, Gomez-Cabrero D, Lindner V, Lara-Astiaso D, and Prósper F
- Subjects
- Animals, Cardiomyopathy, Dilated genetics, Cardiomyopathy, Dilated metabolism, Cardiomyopathy, Dilated pathology, Disease Models, Animal, Extracellular Matrix Proteins genetics, Fibroblasts pathology, Humans, Mice, Myocardial Infarction genetics, Myocardial Infarction pathology, Myocardium pathology, Extracellular Matrix Proteins metabolism, Fibroblasts metabolism, Myocardial Infarction metabolism, Myocardium metabolism, RNA-Seq, Single-Cell Analysis
- Abstract
Background: Cardiac fibroblasts (CFs) have a central role in the ventricular remodeling process associated with different types of fibrosis. Recent studies have shown that fibroblasts do not respond homogeneously to heart injury. Because of the limited set of bona fide fibroblast markers, a proper characterization of fibroblast population heterogeneity in response to cardiac damage is lacking. The purpose of this study was to define CF heterogeneity during ventricular remodeling and the underlying mechanisms that regulate CF function., Methods: Collagen1α1-GFP (green fluorescent protein)-positive CFs were characterized after myocardial infarction (MI) by single-cell and bulk RNA sequencing, assay for transposase-accessible chromatin sequencing, and functional assays. Swine and patient samples were studied using bulk RNA sequencing., Results: We identified and characterized a unique CF subpopulation that emerges after MI in mice. These activated fibroblasts exhibit a clear profibrotic signature, express high levels of Cthrc1 (collagen triple helix repeat containing 1), and localize into the scar. Noncanonical transforming growth factor-β signaling and different transcription factors including SOX9 are important regulators mediating their response to cardiac injury. Absence of CTHRC1 results in pronounced lethality attributable to ventricular rupture. A population of CFs with a similar transcriptome was identified in a swine model of MI and in heart tissue from patients with MI and dilated cardiomyopathy., Conclusions: We report CF heterogeneity and their dynamics during the course of MI and redefine the CFs that respond to cardiac injury and participate in myocardial remodeling. Our study identifies CTHRC1 as a novel regulator of the healing scar process and a target for future translational studies.
- Published
- 2020
- Full Text
- View/download PDF
36. Long-Term Engraftment of Human Cardiomyocytes Combined with Biodegradable Microparticles Induces Heart Repair.
- Author
-
Saludas L, Garbayo E, Mazo M, Pelacho B, Abizanda G, Iglesias-Garcia O, Raya A, Prósper F, and Blanco-Prieto MJ
- Subjects
- Animals, Cell Differentiation, Cell Survival drug effects, Drug Delivery Systems, Heart Diseases pathology, Heart Function Tests, Humans, Male, Mice, Mice, Inbred BALB C, Myocardial Infarction therapy, Ventricular Remodeling, Biodegradable Plastics therapeutic use, Heart Diseases therapy, Induced Pluripotent Stem Cells transplantation, Myocytes, Cardiac transplantation, Nanoparticles therapeutic use, Stem Cell Transplantation methods
- Abstract
Cardiomyocytes derived from human induced pluripotent stem cells (hiPSC-CMs) are a promising cell source for cardiac repair after myocardial infarction (MI) because they offer several advantages such as potential to remuscularize infarcted tissue, integration in the host myocardium, and paracrine therapeutic effects. However, cell delivery issues have limited their potential application in clinical practice, showing poor survival and engraftment after transplantation. In this work, we hypothesized that the combination of hiPSC-CMs with microparticles (MPs) could enhance long-term cell survival and retention in the heart and consequently improve cardiac repair. CMs were obtained by differentiation of hiPSCs by small-molecule manipulation of the Wnt pathway and adhered to biomimetic poly(lactic-co-glycolic acid) MPs covered with collagen and poly(d-lysine). The potential of the system to support cell survival was analyzed in vitro, demonstrating a 1.70-fold and 1.99-fold increase in cell survival after 1 and 4 days, respectively. The efficacy of the system was tested in a mouse MI model. Interestingly, 2 months after administration, transplanted hiPSC-CMs could be detected in the peri-infarct area. These cells not only maintained the cardiac phenotype but also showed in vivo maturation and signs of electrical coupling. Importantly, cardiac function was significantly improved, which could be attributed to a paracrine effect of cells. These findings suggest that MPs represent an excellent platform for cell delivery in the field of cardiac repair, which could also be translated into an enhancement of the potential of cell-based therapies in other medical applications., (Copyright © 2019 by The American Society for Pharmacology and Experimental Therapeutics.)
- Published
- 2019
- Full Text
- View/download PDF
37. Allogeneic Mesenchymal Stem Cells and Biomaterials: The Perfect Match for Cardiac Repair?
- Author
-
Perez-Estenaga I, Prosper F, and Pelacho B
- Subjects
- Animals, Humans, Transplantation, Homologous, Biocompatible Materials chemistry, Guided Tissue Regeneration methods, Heart Diseases therapy, Mesenchymal Stem Cell Transplantation methods, Tissue Scaffolds chemistry
- Abstract
Coronary heart disease is the leading cause of death worldwide with huge socio-economic consequences. Cell therapy, and particularly mesenchymal stem cells (MSC), are considered a promising option to treat this disorder, due to their robust trophic and immunomodulatory properties. However, limitations such as their low rate of engraftment and poor survival after administration into the heart have precluded their large-scale clinical use. Nevertheless, the combination of MSC with polymer-made scaffolds or hydrogels has proven to enhance their retention and, therefore, their efficacy. Additionally, their allogeneic use could permit the creation of ready-to-use cell patches able to improve their feasibility and promote their application in clinical settings. In this review, the experimental and clinical results derived from the use of MSC in cardiac pathology, as well as advances in the bioengineering field to improve the potential of therapeutic cells, are extensively discussed. Additionally, the current understanding of the heart response to the allogeneic MSC transplants is addressed.
- Published
- 2018
- Full Text
- View/download PDF
38. Bmi1-Progenitor Cell Ablation Impairs the Angiogenic Response to Myocardial Infarction.
- Author
-
Herrero D, Cañón S, Pelacho B, Salvador-Bernáldez M, Aguilar S, Pogontke C, Carmona RM, Salvador JM, Perez-Pomares JM, Klein OD, Prósper F, Jimenez-Borreguero LJ, and Bernad A
- Subjects
- Animals, Cells, Cultured, Disease Models, Animal, Female, Humans, Male, Mice, Mice, Transgenic, Proto-Oncogene Proteins c-kit metabolism, Transcription Factors metabolism, Myocardial Infarction pathology, Myocardial Infarction physiopathology, Myocytes, Cardiac pathology, Myocytes, Cardiac physiology, Neovascularization, Pathologic, Polycomb Repressive Complex 1 physiology, Stem Cells pathology, Stem Cells physiology, Ventricular Remodeling
- Abstract
Objective- Cardiac progenitor cells reside in the heart in adulthood, although their physiological relevance remains unknown. Here, we demonstrate that after myocardial infarction, adult Bmi1
+ (B lymphoma Mo-MLV insertion region 1 homolog [PCGF4]) cardiac cells are a key progenitor-like population in cardiac neovascularization during ventricular remodeling. Approach and Results- These cells, which have a strong in vivo differentiation bias, are a mixture of endothelial- and mesenchymal-related cells with in vitro spontaneous endothelial cell differentiation capacity. Genetic lineage tracing analysis showed that heart-resident Bmi1+ progenitor cells proliferate after acute myocardial infarction and differentiate to generate de novo cardiac vasculature. In a mouse model of induced myocardial infarction, genetic ablation of these cells substantially deteriorated both heart angiogenesis and the ejection fraction, resulting in an ischemic-dilated cardiac phenotype. Conclusions- These findings imply that endothelial-related Bmi1+ progenitor cells are necessary for injury-induced neovascularization in adult mouse heart and highlight these cells as a suitable therapeutic target for preventing dysfunctional left ventricular remodeling after injury.- Published
- 2018
- Full Text
- View/download PDF
39. Non-invasive in vivo imaging of cardiac stem/progenitor cell biodistribution and retention after intracoronary and intramyocardial delivery in a swine model of chronic ischemia reperfusion injury.
- Author
-
Collantes M, Pelacho B, García-Velloso MJ, Gavira JJ, Abizanda G, Palacios I, Rodriguez-Borlado L, Álvarez V, Prieto E, Ecay M, Larequi E, Peñuelas I, and Prósper F
- Subjects
- Animals, Cell Separation, Disease Models, Animal, Female, Fluorodeoxyglucose F18 chemistry, Glucosamine analogs & derivatives, Glucosamine chemistry, Myocardial Reperfusion Injury diagnostic imaging, Myocardial Reperfusion Injury pathology, Positron Emission Tomography Computed Tomography, Sus scrofa, Tissue Distribution, Diagnostic Imaging methods, Injections, Myocardial Reperfusion Injury therapy, Myocardium pathology, Stem Cell Transplantation, Stem Cells cytology
- Abstract
Background: The safety and efficacy of cardiac stem/progenitor cells (CSC) have been demonstrated in previous preclinical and clinical assays for heart failure. However, their optimal delivery route to the ischemic heart has not yet been assessed. This study was designed to determine by a non-invasive imaging technique (PET/CT) the biodistribution and acute retention of allogeneic pig CSC implanted by two different delivery routes, intracoronary (IC) and intramyocardial (IM), in a swine preclinical model of chronic ischemia-reperfusion., Methods: Ischemia-reperfusion was induced in six Goettingen hybrid minipigs by 90 min coronary artery occlusion followed by reperfusion. Thirty days later, animals were allocated to receive IC (n = 3) or NOGA
® -guided IM injection (n = 3) of 50 million of18 F-FDG/GFP-labeled allogeneic pig CSC. Acute retention was quantified by PET/CT 4 h after injection and cell engraftment assessed by immunohistochemical quantification of GFP+ cells three days post-injection., Results: Biodistribution of18 F-FDG-labeled CSC was clearly visualized by PET/CT imaging and quantified. No statistical differences in acute cell retention (percentage of injected dose, %ID) were found in the heart when cells were administered by NOGA® -guided IM (13.4 ± 3.4%ID) or IC injections (17.4 ± 4.1%ID). Interestingly, engrafted CSC were histologically detected only after IM injection., Conclusion: PET/CT imaging of18 F-FDG-labeled CSC allows quantifying biodistribution and acute retention of implanted cells in a clinically relevant pig model of chronic myocardial infarction. Similar levels of acute retention are achieved when cells are IM or IC administered. However, acute cell retention does not correlate with cell engraftment, which is improved by IM injection.- Published
- 2017
- Full Text
- View/download PDF
40. Catheter-based Intramyocardial Injection of FGF1 or NRG1-loaded MPs Improves Cardiac Function in a Preclinical Model of Ischemia-Reperfusion.
- Author
-
Garbayo E, Gavira JJ, de Yebenes MG, Pelacho B, Abizanda G, Lana H, Blanco-Prieto MJ, and Prosper F
- Subjects
- Administration, Cutaneous, Animals, Cell Line, Disease Models, Animal, Female, Heart Function Tests drug effects, Injections, Lactic Acid, Male, Mice, Polyglycolic Acid, Polylactic Acid-Polyglycolic Acid Copolymer, Swine, Swine, Miniature, Treatment Outcome, Fibroblast Growth Factor 1 administration & dosage, Myocardial Reperfusion Injury drug therapy, Myocardial Reperfusion Injury physiopathology, Neuregulin-1 administration & dosage
- Abstract
Cardiovascular protein therapeutics such as neuregulin (NRG1) and acidic-fibroblast growth factor (FGF1) requires new formulation strategies that allow for sustained bioavailability of the drug in the infarcted myocardium. However, there is no FDA-approved injectable protein delivery platform due to translational concerns about biomaterial administration through cardiac catheters. We therefore sought to evaluate the efficacy of percutaneous intramyocardial injection of poly(lactic-co-glycolic acid) microparticles (MPs) loaded with NRG1 and FGF1 using the NOGA MYOSTAR injection catheter in a porcine model of ischemia-reperfusion. NRG1- and FGF1-loaded MPs were prepared using a multiple emulsion solvent-evaporation technique. Infarcted pigs were treated one week after ischemia-reperfusion with MPs containing NRG1, FGF1 or non-loaded MPs delivered via clinically-translatable percutaneous transendocardial-injection. Three months post-treatment, echocardiography indicated a significant improvement in systolic and diastolic cardiac function. Moreover, improvement in bipolar voltage and decrease in transmural infarct progression was demonstrated by electromechanical NOGA-mapping. Functional benefit was associated with an increase in myocardial vascularization and remodeling. These findings in a large animal model of ischemia-reperfusion demonstrate the feasibility and efficacy of using MPs as a delivery system for growth factors and provide strong evidence to move forward with clinical studies using therapeutic proteins combined with catheter-compatible biomaterials.
- Published
- 2016
- Full Text
- View/download PDF
41. Neovascularization Potential of Blood Outgrowth Endothelial Cells From Patients With Stable Ischemic Heart Failure Is Preserved.
- Author
-
Dauwe D, Pelacho B, Wibowo A, Walravens AS, Verdonck K, Gillijns H, Caluwe E, Pokreisz P, van Gastel N, Carmeliet G, Depypere M, Maes F, Vanden Driessche N, Droogne W, Van Cleemput J, Vanhaecke J, Prosper F, Verfaillie C, Luttun A, and Janssens S
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Animals, Case-Control Studies, Cell Proliferation physiology, Cells, Cultured, Endothelium, Vascular cytology, Endothelium, Vascular transplantation, Female, Humans, Male, Mice, Nude, Middle Aged, Oxidative Stress physiology, Young Adult, Endothelium, Vascular physiopathology, Myocardial Ischemia physiopathology, Neovascularization, Physiologic physiology
- Abstract
Background: Blood outgrowth endothelial cells (BOECs) mediate therapeutic neovascularization in experimental models, but outgrowth characteristics and functionality of BOECs from patients with ischemic cardiomyopathy (ICMP) are unknown. We compared outgrowth efficiency and in vitro and in vivo functionality of BOECs derived from ICMP with BOECs from age-matched (ACON) and healthy young (CON) controls., Methods and Results: We isolated 3.6±0.6 BOEC colonies/100×10(6) mononuclear cells (MNCs) from 60-mL blood samples of ICMP patients (n=45; age: 66±1 years; LVEF: 31±2%) versus 3.5±0.9 colonies/100×10(6) MNCs in ACON (n=32; age: 60±1 years) and 2.6±0.4 colonies/100×10(6) MNCs in CON (n=55; age: 34±1 years), P=0.29. Endothelial lineage (VEGFR2(+)/CD31(+)/CD146(+)) and progenitor (CD34(+)/CD133(-)) marker expression was comparable in ICMP and CON. Growth kinetics were similar between groups (P=0.38) and not affected by left ventricular systolic dysfunction, maladaptive remodeling, or presence of cardiovascular risk factors in ICMP patients. In vitro neovascularization potential, assessed by network remodeling on Matrigel and three-dimensional spheroid sprouting, did not differ in ICMP from (A)CON. Secretome analysis showed a marked proangiogenic profile, with highest release of angiopoietin-2 (1.4±0.3×10(5) pg/10(6) ICMP-BOECs) and placental growth factor (5.8±1.5×10(3) pg/10(6) ICMP BOECs), independent of age or ischemic disease. Senescence-associated β-galactosidase staining showed comparable senescence in BOECs from ICMP (5.8±2.1%; n=17), ACON (3.9±1.1%; n=7), and CON (9.0±2.8%; n=13), P=0.19. High-resolution microcomputed tomography analysis in the ischemic hindlimb of nude mice confirmed increased arteriogenesis in the thigh region after intramuscular injections of BOECs from ICMP (P=0.025; n=8) and CON (P=0.048; n=5) over vehicle control (n=8), both to a similar extent (P=0.831)., Conclusions: BOECs can be successfully culture-expanded from patients with ICMP. In contrast to impaired functionality of ICMP-derived bone marrow MNCs, BOECs retain a robust proangiogenic profile, both in vitro and in vivo, with therapeutic potential for targeting ischemic disease., (© 2016 The Authors. Published on behalf of the American Heart Association, Inc., by Wiley Blackwell.)
- Published
- 2016
- Full Text
- View/download PDF
42. Interacting resident epicardium-derived fibroblasts and recruited bone marrow cells form myocardial infarction scar.
- Author
-
Ruiz-Villalba A, Simón AM, Pogontke C, Castillo MI, Abizanda G, Pelacho B, Sánchez-Domínguez R, Segovia JC, Prósper F, and Pérez-Pomares JM
- Subjects
- Animals, Cell Differentiation, Cells, Cultured, Cicatrix etiology, Cicatrix physiopathology, Disease Models, Animal, Mice, Mice, Inbred C57BL, Myocardial Infarction complications, Myocardial Infarction physiopathology, Pericardium pathology, Bone Marrow Cells pathology, Cicatrix pathology, Fibroblasts pathology, Myocardial Infarction pathology, Myocytes, Cardiac pathology, Ventricular Remodeling
- Abstract
Background: Although efforts continue to find new therapies to regenerate infarcted heart tissue, knowledge of the cellular and molecular mechanisms involved remains poor., Objectives: This study sought to identify the origin of cardiac fibroblasts (CFs) in the infarcted heart to better understand the pathophysiology of ventricular remodeling following myocardial infarction (MI)., Methods: Permanent genetic tracing of epicardium-derived cell (EPDC) and bone marrow-derived blood cell (BMC) lineages was established using Cre/LoxP technology. In vivo gene and protein expression studies, as well as in vitro cell culture assays, were developed to characterize EPDC and BMC interaction and properties., Results: EPDCs, which colonize the cardiac interstitium during embryogenesis, massively differentiate into CFs after MI. This response is disease-specific, because angiotensin II-induced pressure overload does not trigger significant EPDC fibroblastic differentiation. The expansion of epicardial-derived CFs follows BMC infiltration into the infarct site; the number of EPDCs equals that of BMCs 1 week post-infarction. BMC-EPDC interaction leads to cell polarization, packing, massive collagen deposition, and scar formation. Moreover, epicardium-derived CFs display stromal properties with respect to BMCs, contributing to the sustained recruitment of circulating cells to the damaged zone and the cardiac persistence of hematopoietic progenitors/stem cells after MI., Conclusions: EPDCs, but not BMCs, are the main origin of CFs in the ischemic heart. Adult resident EPDC contribution to the CF compartment is time- and disease-dependent. Our findings are relevant to the understanding of post-MI ventricular remodeling and may contribute to the development of new therapies to treat this disease., (Copyright © 2015 American College of Cardiology Foundation. Published by Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
43. Heterogeneous micromechanical properties of the extracellular matrix in healthy and infarcted hearts.
- Author
-
Andreu I, Luque T, Sancho A, Pelacho B, Iglesias-García O, Melo E, Farré R, Prósper F, Elizalde MR, and Navajas D
- Subjects
- Animals, Male, Mice, Mice, Inbred C57BL, Microscopy, Atomic Force, Rheology, Extracellular Matrix physiology, Myocardial Infarction physiopathology
- Abstract
Infarcted hearts are macroscopically stiffer than healthy organs. Nevertheless, although cell behavior is mediated by the physical features of the cell niche, the intrinsic micromechanical properties of healthy and infarcted heart extracellular matrix (ECM) remain poorly characterized. Using atomic force microscopy, we studied ECM micromechanics of different histological regions of the left ventricle wall of healthy and infarcted mice. Hearts excised from healthy (n=8) and infarcted mice (n=8) were decellularized with sodium dodecyl sulfate and cut into 12 μm thick slices. Healthy ventricular ECM revealed marked mechanical heterogeneity across histological regions of the ventricular wall with the effective Young's modulus ranging from 30.2 ± 2.8 to 74.5 ± 8.7 kPa in collagen- and elastin-rich regions of the myocardium, respectively. Infarcted ECM showed a predominant collagen composition and was 3-fold stiffer than collagen-rich regions of the healthy myocardium. ECM of both healthy and infarcted hearts exhibited a solid-like viscoelastic behavior that conforms to two power-law rheology. Knowledge of intrinsic micromechanical properties of the ECM at the length scale at which cells sense their environment will provide further insight into the cell-scaffold interplay in healthy and infarcted hearts., (Copyright © 2014 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
44. Epicardial delivery of collagen patches with adipose-derived stem cells in rat and minipig models of chronic myocardial infarction.
- Author
-
Araña M, Gavira JJ, Peña E, González A, Abizanda G, Cilla M, Pérez MM, Albiasu E, Aguado N, Casado M, López B, González S, Soriano M, Moreno C, Merino J, García-Verdugo JM, Díez J, Doblaré M, Pelacho B, and Prosper F
- Subjects
- Animals, Chronic Disease, Heart physiopathology, Myocardial Infarction physiopathology, Rats, Rats, Sprague-Dawley, Swine, Swine, Miniature, Tissue Scaffolds, Adipose Tissue cytology, Collagen administration & dosage, Disease Models, Animal, Myocardial Infarction surgery, Pericardium, Stem Cell Transplantation
- Abstract
Although transplantation of adipose-derived stem cells (ADSC) in chronic myocardial infarction (MI) models is associated with functional improvement, its therapeutic value is limited due to poor long-term cell engraftment and survival. Thus, the objective of this study was to examine whether transplantation of collagen patches seeded with ADSC could enhance cell engraftment and improve cardiac function in models of chronic MI. With that purpose, chronically infarcted Sprague-Dawley rats (n = 58) were divided into four groups and transplanted with media, collagen scaffold (CS), rat ADSC, or CS seeded with rat ADSC (CS-rADSC). Cell engraftment, histological changes, and cardiac function were assessed 4 months after transplantation. In addition, Göttingen minipigs (n = 18) were subjected to MI and then transplanted 2 months later with CS or CS seeded with autologous minipig ADSC (CS-pADSC). Functional and histological assessments were performed 3 months post-transplantation. Transplantation of CS-rADSC was associated with increased cell engraftment, significant improvement in cardiac function, myocardial remodeling, and revascularization. Moreover, transplantation of CS-pADSC in the pre-clinical swine model improved cardiac function and was associated with decreased fibrosis and increased vasculogenesis. In summary, transplantation of CS-ADSC resulted in enhanced cell engraftment and was associated with a significant improvement in cardiac function and myocardial remodeling., (Copyright © 2013 Elsevier Ltd. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
45. Preparation and characterization of collagen-based ADSC-carrier sheets for cardiovascular application.
- Author
-
Araña M, Peña E, Abizanda G, Cilla M, Ochoa I, Gavira JJ, Espinosa G, Doblaré M, Pelacho B, and Prosper F
- Subjects
- Animals, Cell Differentiation, Cells, Cultured, Equipment Design, Equipment Failure Analysis, Female, Materials Testing, Myocardial Infarction pathology, Rats, Rats, Sprague-Dawley, Tissue Engineering instrumentation, Treatment Outcome, Absorbable Implants, Adipocytes cytology, Collagen Type I chemistry, Myocardial Infarction surgery, Stem Cell Transplantation methods, Stem Cells cytology, Tissue Scaffolds
- Abstract
The use of scaffolds composed of natural biodegradable matrices represents an attractive strategy to circumvent the lack of cell engraftment, a major limitation of stem cell therapy in cardiovascular diseases. Bovine-derived non-porous collagen scaffolds with different degrees of cross-linking (C0, C2, C5 and C10) were produced and tested for their mechanical behavior, in vitro biocompatibility with adipose-derived stem cells (ADSCs) and tissue adhesion and inflammatory reaction. Uniaxial tensile tests revealed an anisotropic behavior of collagen scaffolds (2×0.5cm) and statistically significant differences in the mechanical behavior between cross-linked and non-cross-linked scaffolds (n=5). In vitro, ADSCs adhered homogenously and showed a similar degree of proliferation on all four types of scaffolds (cells×10(3)cm(-2) at day 7: C0: 94.7±37.1; C2: 91.7±25.6; C5: 88.2±6.8; C10: 72.8±10.7; P=n.s.; n=3). In order to test the in vivo biocompatibility, a chronic myocardial infarction model was performed in rats and 1.2×1.2cm size collagen scaffolds implanted onto the heart 1month post-infarction. Six animals per group were killed 2, 7 and 30days after transplant. Complete and long-lasting adhesion to the heart was only observed with the non-cross-linked scaffolds with almost total degradation 1month post-transplantation. After 7 and 30days post-implantation, the degree of inflammation was significantly lower in the hearts treated with non-cross-linked scaffolds (day 7: C0: 10.2±2.1%; C2: 16.3±2.9%; C5: 15.9±4.8%; C10: 17.4±4.1%; P<0.05 vs. C0; day 30: C0: 1.3±1.3%; C2: 9.4±3.0%; C5: 7.0±2.1%; C10: 9.8±2.5%; P<0.01 vs. C0). In view of the results, the non-cross-linked scaffold (C0) was chosen as an ADSC-carrier sheet and tested in vivo. One week post-implantation, 25.3±7.0% of the cells transplanted were detected in those animals receiving the cell-carrier sheet whereas no cells were found in animals receiving cells alone (n=3 animals/group). We conclude that the biocompatibility and mechanical properties of the non-cross-linked collagen scaffolds make them a useful cell carrier that greatly favors tissue cell engraftment and may be exploited for cell transplantation in models of cardiac disease., (Copyright © 2012 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
46. New strategies for echocardiographic evaluation of left ventricular function in a mouse model of long-term myocardial infarction.
- Author
-
Benavides-Vallve C, Corbacho D, Iglesias-Garcia O, Pelacho B, Albiasu E, Castaño S, Muñoz-Barrutia A, Prosper F, and Ortiz-de-Solorzano C
- Subjects
- Animals, Disease Models, Animal, Female, Image Processing, Computer-Assisted, Mice, Myocardial Infarction pathology, Time Factors, Ventricular Remodeling, Echocardiography methods, Myocardial Infarction diagnostic imaging, Myocardial Infarction physiopathology, Ventricular Function, Left physiology
- Abstract
Background: The aim of this article is to present an optimized acquisition and analysis protocol for the echocardiographic evaluation of left ventricle (LV) remodeling in a mouse model of myocardial infarction (MI)., Methodology: 13 female DBA/2J mice underwent permanent occlusion of the left anterior descending (LAD) coronary artery leading to MI. Mice echocardiography was performed using a Vevo 770 (Visualsonics, Canada) before infarction, and 7, 14, 30, 60, 90 and 120 days after LAD ligation. LV systolic function was evaluated using different parameters, including the fractional area change (FAC%) computed in four high-temporal resolution B-mode short axis images taken at different ventricular levels, and in one parasternal long axis. Pulsed wave and tissue Doppler modes were used to evaluate the diastolic function and Tei Index for global cardiac function. The echocardiographic measurements of infarct size were validated histologically using collagen deposition labeled by Sirius red staining. All data was analyzed using Shapiro-Wilk and Student's t-tests., Principal Findings: Our results reveal LV dilation resulting in marked remodeling an severe systolic dysfunction, starting seven days after MI (LV internal apical diameter, basal = 2.82±0.24, 7d = 3.49±0.42; p<0.001. End-diastolic area, basal = 18.98±1.81, 7d = 22.04±2.11; p<0.001). A strong statistically significant negative correlation exists between the infarct size and long-axis FAC% (r = -0.946; R(2) = 0.90; p<0.05). Moreover, the measured Tei Index values confirmed significant post-infarction impairment of the global cardiac function (basal = 0.46±0.07, 7d = 0.55±0.08, 14 d = 0.57±0.06, 30 d = 0.54±0.06, 60 d = 0.54±0.07, 90 d = 0.57±0.08; p<0.01)., Conclusions/significance: In summary, we have performed a complete characterization of LV post-infarction remodeling in a DBA/2J mouse model of MI, using parameters adapted to the particular characteristics of the model In the future, this well characterized model will be used in both investigative and pharmacological studies that require accurate quantitative monitoring of cardiac recovery after myocardial infarction.
- Published
- 2012
- Full Text
- View/download PDF
47. Mesenchymal stem cells and cardiovascular disease: a bench to bedside roadmap.
- Author
-
Mazo M, Araña M, Pelacho B, and Prosper F
- Abstract
In recent years, the incredible boost in stem cell research has kindled the expectations of both patients and physicians. Mesenchymal progenitors, owing to their availability, ease of manipulation, and therapeutic potential, have become one of the most attractive options for the treatment of a wide range of diseases, from cartilage defects to cardiac disorders. Moreover, their immunomodulatory capacity has opened up their allogenic use, consequently broadening the possibilities for their application. In this review, we will focus on their use in the therapy of myocardial infarction, looking at their characteristics, in vitro and in vivo mechanisms of action, as well as clinical trials.
- Published
- 2012
- Full Text
- View/download PDF
48. Vascular endothelial growth factor-delivery systems for cardiac repair: an overview.
- Author
-
Simón-Yarza T, Formiga FR, Tamayo E, Pelacho B, Prosper F, and Blanco-Prieto MJ
- Abstract
Since the discovery of the Vascular Endothelial Growth Factor (VEGF) and its leading role in the angiogenic process, this has been seen as a promising molecule for promoting neovascularization in the infarcted heart. However, even though several clinical trials were initiated, no therapeutic effects were observed, due in part to the short half life of this factor when administered directly to the tissue. In this context, drug delivery systems appear to offer a promising strategy to overcome limitations in clinical trials of VEGF.The aim of this paper is to review the principal drug delivery systems that have been developed to administer VEGF in cardiovascular disease. Studies published in the last 5 years are reviewed and the main features of these systems are explained. The tissue engineering concept is introduced as a therapeutic alternative that holds promise for the near future.
- Published
- 2012
- Full Text
- View/download PDF
49. Repeated implantation of skeletal myoblast in a swine model of chronic myocardial infarction.
- Author
-
Gavira JJ, Nasarre E, Abizanda G, Pérez-Ilzarbe M, de Martino-Rodriguez A, García de Jalón JA, Mazo M, Macias A, García-Bolao I, Pelacho B, Martínez-Caro D, and Prósper F
- Subjects
- Animals, Arrhythmias, Cardiac etiology, Cell Differentiation, Chronic Disease, Fibrosis, Graft Survival, Immunohistochemistry, Myoblasts, Skeletal cytology, Myocardium pathology, Neovascularization, Physiologic physiology, Swine, Swine, Miniature, Ventricular Dysfunction, Left etiology, Ventricular Remodeling physiology, Myoblasts, Skeletal transplantation, Myocardial Infarction therapy
- Abstract
Aims: Although transplantation of skeletal myoblast (SkM) in models of chronic myocardial infarction (MI) induces an improvement in cardiac function, the limited engraftment remains a major limitation. We analyse in a pre-clinical model whether the sequential transplantation of autologous SkM by percutaneous delivery was associated with increased cell engraftment and functional benefit., Methods and Results: Chronically infarcted Goettingen minipigs (n = 20) were divided in four groups that received either media control or one, two, or three doses of SkM (mean of 329.6 x 10(6) cells per dose) at intervals of 6 weeks and were followed for a total of 7 months. At the time of sacrifice, cardiac function was significantly better in animals treated with SkM in comparison with the control group. A significantly greater increase in the DeltaLVEF was detected in animals that received three doses vs. a single dose of SkM. A correlation between the total number of transplanted cells and the improvement in LVEF and DeltaLVEF was found (P < 0.05). Skeletal myoblast transplant was associated with an increase in tissue vasculogenesis and decreased fibrosis (collagen vascular fraction) and these effects were greater in animals receiving three doses of cells., Conclusion: Repeated injection of SkM in a model of chronic MI is feasible and safe and induces a significant improvement in cardiac function.
- Published
- 2010
- Full Text
- View/download PDF
50. Characterization of the paracrine effects of human skeletal myoblasts transplanted in infarcted myocardium.
- Author
-
Perez-Ilzarbe M, Agbulut O, Pelacho B, Ciorba C, San Jose-Eneriz E, Desnos M, Hagège AA, Aranda P, Andreu EJ, Menasché P, and Prósper F
- Subjects
- Animals, Apoptosis, Biopsy, Cell Proliferation, Cells, Cultured, DNA-Binding Proteins biosynthesis, DNA-Binding Proteins genetics, Endothelium, Vascular metabolism, Endothelium, Vascular pathology, Enzyme-Linked Immunosorbent Assay, Gene Expression Regulation, Glycosylphosphatidylinositols, Humans, Matrix Metalloproteinase 2 biosynthesis, Matrix Metalloproteinase 2 genetics, Matrix Metalloproteinase 7 biosynthesis, Matrix Metalloproteinase 7 genetics, Membrane Proteins biosynthesis, Membrane Proteins genetics, Myoblasts, Skeletal cytology, Myoblasts, Skeletal metabolism, Myocardial Infarction metabolism, Myocardial Infarction pathology, Myocytes, Cardiac metabolism, Myocytes, Cardiac pathology, Neovascularization, Physiologic genetics, Oligonucleotide Array Sequence Analysis, Polymerase Chain Reaction, RNA genetics, Rats, Rats, Nude, Transcription Factors biosynthesis, Transcription Factors genetics, Treatment Outcome, Troponin I biosynthesis, Troponin I genetics, Cell Transplantation methods, Myoblasts, Skeletal transplantation, Myocardial Infarction surgery, Paracrine Communication physiology
- Abstract
Background: The discrepancy between the functional improvements yielded experimentally by skeletal myoblasts (SM) transplanted in infarcted myocardium and the paucity of their long-term engraftment has raised the hypothesis of cell-mediated paracrine mechanisms., Methods and Results: We analyzed gene expression and growth factors released by undifferentiated human SM (CD56(+)), myotubes (SM cultured until confluence) and fibroblasts-like cells (CD56(-)). Gene expression revealed up-regulation of pro-angiogenic (PGF), anti-apoptotics (BAG-1, BCL-2), heart development (TNNT2, TNNC1) and extracellular matrix remodelling (MMP-2, MMP-7) genes in SM. In line with the gene expression profile, the analysis of culture supernatants of SM by ELISA identified the release of growth factors involved in angiogenesis (VEGF, PIGF, angiogenin, angiopoietin, HGF and PDGF-BB) as well as proteases involved in matrix remodelling (MMP2, MMP9 and MMP10) and their inhibitors (TIMPs). Culture of smooth muscle cells (SMC), cardiomyocytes (HL-1) and human umbilical vein endothelial cells (HUVECs) with SM-released conditioned media demonstrated an increased proliferation of HUVEC, SMC and cardiomyocytes (p<0.05) and a decrease in apoptosis of cardiomyocytes (p<0.05). Analysis of nude rats transplanted with human SM demonstrated expression of human-specific MMP-2, TNNI3, CNN3, PGF, TNNT2, PAX7, TGF-beta, and IGF-1 1 month after transplant., Conclusions: Our data support the paracrine hypothesis whereby myoblast-secreted factors may contribute to the beneficial effects of myogenic cell transplantation in infarcted myocardium.
- Published
- 2008
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.