1
|
Báez-Díaz C, Torrescusa-Bermejo A, Sánchez-Margallo FM, Vázquez-López F, Pulido M, López E, Arenal Á, Crisóstomo V. Intracoronary-Cardiosphere-Derived Cell Secretome Therapy: Effects on Ventricular Tachycardia Inducibility and Cardiac Function in a Swine Model. Biomedicines 2025; 13:1043. [PMID: 40426870 PMCID: PMC12108644 DOI: 10.3390/biomedicines13051043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 04/22/2025] [Accepted: 04/23/2025] [Indexed: 05/29/2025] Open
Abstract
Background/Objectives: Ventricular tachycardia (VT) resulting in sudden cardiac death is common following a myocardial infarction (MI). Our objective was to evaluate the effects of an intracoronary (IC) administration of cardiosphere-derived cell secretome (S-CDCs) on VT inducibility and cardiac function in a swine model of MI. Methods: Fourteen pigs underwent endovascular MI model creation. At 4 weeks, saline (CON; 5 mL; n = 7) or S-CDCs (S-CDCs; 9.16 mg protein in 5 mL saline; n = 7) was blindly administered via the IC route. VT inducibility and magnetic resonance imaging (MRI) studies were performed both pre- and 4 months post-IC therapy, calculating left ventricular ejection fraction (LVEF), infarct size as a percentage of left ventricle (% MI), and left ventricular indexed end-diastolic and end-systolic volumes (LVEDVi, LVESVi). Results: While VT was inducible in 100% of the animals before IC therapy, at 4 months, the inducibility rate was lower in the S-CDCs group compared to the CON group (57% versus 100%, p = 0.05). Likewise, in the S-CDCs group, % MI was significantly lower than in the CON group (12 ± 3% versus 16 ± 3%, p = 0.03). LVEF (S-CDCs: 35 ± 10% versus CON: 29 ± 10%, p = NS), LVEDVi and LVESVi (S-CDCs: 83 ± 18 mL/m2 and 56 ± 20 mL/m2 versus CON: 88 ± 29 mL/m2 and 64 ± 20 mL/m2, p = NS) did not change. Conclusions: IC therapy with S-CDCs appears to reduce the development of post-MI VT. Furthermore, it suggests a beneficial effect on infarct size, reducing % MI in this experimental swine model.
Collapse
Affiliation(s)
- Claudia Báez-Díaz
- Fundación Centro de Cirugía de Mínima Invasión Jesús Usón, 10071 Cáceres, Spain; (A.T.-B.); (F.M.S.-M.); (F.V.-L.); (M.P.); (E.L.); (V.C.)
- Red Española de Terapias Avanzadas RICORS-TERAV, 28029 Madrid, Spain
| | - Axiel Torrescusa-Bermejo
- Fundación Centro de Cirugía de Mínima Invasión Jesús Usón, 10071 Cáceres, Spain; (A.T.-B.); (F.M.S.-M.); (F.V.-L.); (M.P.); (E.L.); (V.C.)
| | - Francisco Miguel Sánchez-Margallo
- Fundación Centro de Cirugía de Mínima Invasión Jesús Usón, 10071 Cáceres, Spain; (A.T.-B.); (F.M.S.-M.); (F.V.-L.); (M.P.); (E.L.); (V.C.)
- Red Española de Terapias Avanzadas RICORS-TERAV, 28029 Madrid, Spain
| | - Fátima Vázquez-López
- Fundación Centro de Cirugía de Mínima Invasión Jesús Usón, 10071 Cáceres, Spain; (A.T.-B.); (F.M.S.-M.); (F.V.-L.); (M.P.); (E.L.); (V.C.)
| | - María Pulido
- Fundación Centro de Cirugía de Mínima Invasión Jesús Usón, 10071 Cáceres, Spain; (A.T.-B.); (F.M.S.-M.); (F.V.-L.); (M.P.); (E.L.); (V.C.)
| | - Esther López
- Fundación Centro de Cirugía de Mínima Invasión Jesús Usón, 10071 Cáceres, Spain; (A.T.-B.); (F.M.S.-M.); (F.V.-L.); (M.P.); (E.L.); (V.C.)
- Red Española de Terapias Avanzadas RICORS-TERAV, 28029 Madrid, Spain
| | - Ángel Arenal
- Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain;
| | - Verónica Crisóstomo
- Fundación Centro de Cirugía de Mínima Invasión Jesús Usón, 10071 Cáceres, Spain; (A.T.-B.); (F.M.S.-M.); (F.V.-L.); (M.P.); (E.L.); (V.C.)
- Red Española de Terapias Avanzadas RICORS-TERAV, 28029 Madrid, Spain
| |
Collapse
|
2
|
Cau MF, Ferraresso F, Seadler M, Badior K, Zhang Y, Ketelboeter LM, Rodriguez GG, Chen T, Ferraresso M, Wietrzny A, Robertson M, Haugen A, Cullis PR, de Moya M, Dyer M, Kastrup CJ. siRNA-mediated reduction of a circulating protein in swine using lipid nanoparticles. Mol Ther Methods Clin Dev 2024; 32:101258. [PMID: 38779336 PMCID: PMC11109470 DOI: 10.1016/j.omtm.2024.101258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 04/22/2024] [Indexed: 05/25/2024]
Abstract
Genetic manipulation of animal models is a fundamental research tool in biology and medicine but is challenging in large animals. In rodents, models can be readily developed by knocking out genes in embryonic stem cells or by knocking down genes through in vivo delivery of nucleic acids. Swine are a preferred animal model for studying the cardiovascular and immune systems, but there are limited strategies for genetic manipulation. Lipid nanoparticles (LNPs) efficiently deliver small interfering RNA (siRNA) to knock down circulating proteins, but swine are sensitive to LNP-induced complement activation-related pseudoallergy (CARPA). We hypothesized that appropriately administering optimized siRNA-LNPs could knock down circulating levels of plasminogen, a blood protein synthesized in the liver. siRNA-LNPs against plasminogen (siPLG) reduced plasma plasminogen protein and hepatic plasminogen mRNA levels to below 5% of baseline values. Functional assays showed that reducing plasminogen levels modulated systemic blood coagulation. Clinical signs of CARPA were not observed, and occasional mild and transient hepatotoxicity was present in siPLG-treated animals at 5 h post-infusion, which returned to baseline by 7 days. These findings advance siRNA-LNPs in swine models, enabling genetic engineering of blood and hepatic proteins, which can likely expand to proteins in other tissues in the future.
Collapse
Affiliation(s)
- Massimo F. Cau
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Francesca Ferraresso
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Versiti Blood Research Institute, Milwaukee, WI 53226, USA
| | - Monica Seadler
- Versiti Blood Research Institute, Milwaukee, WI 53226, USA
- Department of Surgery, Division of Trauma and Acute Care Surgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | | | - Youjie Zhang
- Versiti Blood Research Institute, Milwaukee, WI 53226, USA
| | | | | | - Taylor Chen
- Versiti Blood Research Institute, Milwaukee, WI 53226, USA
| | | | | | - Madelaine Robertson
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Amber Haugen
- Versiti Blood Research Institute, Milwaukee, WI 53226, USA
| | - Pieter R. Cullis
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Marc de Moya
- Department of Surgery, Division of Trauma and Acute Care Surgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Mitchell Dyer
- Versiti Blood Research Institute, Milwaukee, WI 53226, USA
- Department of Surgery, Division of Vascular and Endovascular Surgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Christian J. Kastrup
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Versiti Blood Research Institute, Milwaukee, WI 53226, USA
- Department of Surgery, Division of Trauma and Acute Care Surgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Departments of Biochemistry, Biomedical Engineering, and Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
3
|
Jia H, Chang Y, Song J. The pig as an optimal animal model for cardiovascular research. Lab Anim (NY) 2024; 53:136-147. [PMID: 38773343 DOI: 10.1038/s41684-024-01377-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 04/22/2024] [Indexed: 05/23/2024]
Abstract
Cardiovascular disease is a worldwide health problem and a leading cause of morbidity and mortality. Preclinical cardiovascular research using animals is needed to explore potential targets and therapeutic options. Compared with rodents, pigs have many advantages, with their anatomy, physiology, metabolism and immune system being more similar to humans. Here we present an overview of the available pig models for cardiovascular diseases, discuss their advantages over other models and propose the concept of standardized models to improve translation to the clinical setting and control research costs.
Collapse
Affiliation(s)
- Hao Jia
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuan Chang
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiangping Song
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Sanya Institute of China Agricultural University, Sanya, China.
| |
Collapse
|
4
|
Stone CR, Harris DD, Broadwin M, Kanuparthy M, Sabe SA, Xu C, Feng J, Abid MR, Sellke FW. Crafting a Rigorous, Clinically Relevant Large Animal Model of Chronic Myocardial Ischemia: What Have We Learned in 20 Years? Methods Protoc 2024; 7:17. [PMID: 38392691 PMCID: PMC10891802 DOI: 10.3390/mps7010017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 02/10/2024] [Accepted: 02/17/2024] [Indexed: 02/24/2024] Open
Abstract
The past several decades have borne witness to several breakthroughs and paradigm shifts within the field of cardiovascular medicine, but one component that has remained constant throughout this time is the need for accurate animal models for the refinement and elaboration of the hypotheses and therapies crucial to our capacity to combat human disease. Numerous sophisticated and high-throughput molecular strategies have emerged, including rational drug design and the multi-omics approaches that allow extensive characterization of the host response to disease states and their prospective resolutions, but these technologies all require grounding within a faithful representation of their clinical context. Over this period, our lab has exhaustively tested, progressively refined, and extensively contributed to cardiovascular discovery on the basis of one such faithful representation. It is the purpose of this paper to review our porcine model of chronic myocardial ischemia using ameroid constriction and the subsequent myriad of physiological and molecular-biological insights it has allowed our lab to attain and describe. We hope that, by depicting our methods and the insight they have yielded clearly and completely-drawing for this purpose on comprehensive videographic illustration-other research teams will be empowered to carry our work forward, drawing on our experience to refine their own investigations into the pathogenesis and eradication of cardiovascular disease.
Collapse
Affiliation(s)
- Christopher R. Stone
- Department of Cardiothoracic Surgery, The Warren Alpert School of Medicine at Brown University, Providence, RI 02903, USA; (D.D.H.); (M.B.); (M.K.); (S.A.S.); (C.X.); (J.F.); (M.R.A.); (F.W.S.)
| | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Hong X, Luo AC, Doulamis I, Oh N, Im GB, Lin CY, del Nido PJ, Lin RZ, Melero-Martin JM. Photopolymerizable Hydrogel for Enhanced Intramyocardial Vascular Progenitor Cell Delivery and Post-Myocardial Infarction Healing. Adv Healthc Mater 2023; 12:e2301581. [PMID: 37611321 PMCID: PMC10840685 DOI: 10.1002/adhm.202301581] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 08/08/2023] [Indexed: 08/25/2023]
Abstract
Cell transplantation success for myocardial infarction (MI) treatment is often hindered by low engraftment due to washout effects during myocardial contraction. A clinically viable biomaterial that enhances cell retention can optimize intramyocardial cell delivery. In this study, a therapeutic cell delivery method is developed for MI treatment utilizing a photocrosslinkable gelatin methacryloyl (GelMA) hydrogel. Human vascular progenitor cells, capable of forming functional vasculatures upon transplantation, are combined with an in situ photopolymerization approach and injected into the infarcted zones of mouse hearts. This strategy substantially improves acute cell retention and promotes long-term post-MI cardiac healing, including stabilized cardiac functions, preserved viable myocardium, and reduced cardiac fibrosis. Additionally, engrafted vascular cells polarize recruited bone marrow-derived neutrophils toward a non-inflammatory phenotype via transforming growth factor beta (TGFβ) signaling, fostering a pro-regenerative microenvironment. Neutrophil depletion negates the therapeutic benefits generated by cell delivery in ischemic hearts, highlighting the essential role of non-inflammatory, pro-regenerative neutrophils in cardiac remodeling. In conclusion, this GelMA hydrogel-based intramyocardial vascular cell delivery approach holds promise for enhancing the treatment of acute myocardial infarction.
Collapse
Affiliation(s)
- Xuechong Hong
- Department of Cardiac Surgery, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Allen Chilun Luo
- Department of Cardiac Surgery, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Ilias Doulamis
- Department of Cardiac Surgery, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Nicholas Oh
- Department of Cardiac Surgery, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Gwang-Bum Im
- Department of Cardiac Surgery, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Chun-Yen Lin
- Department of Lymphoma and Myeloma, The University of Texas, M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Pedro J. del Nido
- Department of Cardiac Surgery, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Ruei-Zeng Lin
- Department of Cardiac Surgery, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Juan M. Melero-Martin
- Department of Cardiac Surgery, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| |
Collapse
|
6
|
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, Bernad A. Cardiac Progenitor Cell Exosomal miR-935 Protects against Oxidative Stress. Cells 2023; 12:2300. [PMID: 37759522 PMCID: PMC10528297 DOI: 10.3390/cells12182300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 08/31/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
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-miRSEL). 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.
Collapse
Affiliation(s)
- Susana Aguilar
- Cardiac Stem Cells Lab, Centro Nacional de Biotecnología (CNB-CSIC), Department of Immunology and Oncology, Campus Universidad Autónoma de Madrid, 28049 Madrid, Spain; (S.A.); (L.A.-M.); (J.L.T.); (G.A.); (D.H.); (M.H.); (R.G.-M.); (C.M.)
| | - Paula García-Olloqui
- Center for Applied Medical Research (CIMA), Regenerative Medicine Department, University of Navarra, 31008 Pamplona, Spain; (P.G.-O.); (G.A.); (Á.V.); (S.R.-D.); (F.P.)
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain;
| | - Lidia Amigo-Morán
- Cardiac Stem Cells Lab, Centro Nacional de Biotecnología (CNB-CSIC), Department of Immunology and Oncology, Campus Universidad Autónoma de Madrid, 28049 Madrid, Spain; (S.A.); (L.A.-M.); (J.L.T.); (G.A.); (D.H.); (M.H.); (R.G.-M.); (C.M.)
| | - José Luis Torán
- Cardiac Stem Cells Lab, Centro Nacional de Biotecnología (CNB-CSIC), Department of Immunology and Oncology, Campus Universidad Autónoma de Madrid, 28049 Madrid, Spain; (S.A.); (L.A.-M.); (J.L.T.); (G.A.); (D.H.); (M.H.); (R.G.-M.); (C.M.)
| | - Juan Antonio López
- Cardiovascular Proteomics Laboratory, Spanish National Cardiovascular Research Center (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain; (J.A.L.); (J.V.)
- CIBER de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Guillermo Albericio
- Cardiac Stem Cells Lab, Centro Nacional de Biotecnología (CNB-CSIC), Department of Immunology and Oncology, Campus Universidad Autónoma de Madrid, 28049 Madrid, Spain; (S.A.); (L.A.-M.); (J.L.T.); (G.A.); (D.H.); (M.H.); (R.G.-M.); (C.M.)
| | - Gloria Abizanda
- Center for Applied Medical Research (CIMA), Regenerative Medicine Department, University of Navarra, 31008 Pamplona, Spain; (P.G.-O.); (G.A.); (Á.V.); (S.R.-D.); (F.P.)
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain;
| | - Diego Herrero
- Cardiac Stem Cells Lab, Centro Nacional de Biotecnología (CNB-CSIC), Department of Immunology and Oncology, Campus Universidad Autónoma de Madrid, 28049 Madrid, Spain; (S.A.); (L.A.-M.); (J.L.T.); (G.A.); (D.H.); (M.H.); (R.G.-M.); (C.M.)
| | - África Vales
- Center for Applied Medical Research (CIMA), Regenerative Medicine Department, University of Navarra, 31008 Pamplona, Spain; (P.G.-O.); (G.A.); (Á.V.); (S.R.-D.); (F.P.)
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain;
| | - Saray Rodríguez-Diaz
- Center for Applied Medical Research (CIMA), Regenerative Medicine Department, University of Navarra, 31008 Pamplona, Spain; (P.G.-O.); (G.A.); (Á.V.); (S.R.-D.); (F.P.)
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain;
| | - Marina Higuera
- Cardiac Stem Cells Lab, Centro Nacional de Biotecnología (CNB-CSIC), Department of Immunology and Oncology, Campus Universidad Autónoma de Madrid, 28049 Madrid, Spain; (S.A.); (L.A.-M.); (J.L.T.); (G.A.); (D.H.); (M.H.); (R.G.-M.); (C.M.)
| | - Rubén García-Martín
- Cardiac Stem Cells Lab, Centro Nacional de Biotecnología (CNB-CSIC), Department of Immunology and Oncology, Campus Universidad Autónoma de Madrid, 28049 Madrid, Spain; (S.A.); (L.A.-M.); (J.L.T.); (G.A.); (D.H.); (M.H.); (R.G.-M.); (C.M.)
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Jesús Vázquez
- Cardiovascular Proteomics Laboratory, Spanish National Cardiovascular Research Center (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain; (J.A.L.); (J.V.)
- CIBER de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Carmen Mora
- Cardiac Stem Cells Lab, Centro Nacional de Biotecnología (CNB-CSIC), Department of Immunology and Oncology, Campus Universidad Autónoma de Madrid, 28049 Madrid, Spain; (S.A.); (L.A.-M.); (J.L.T.); (G.A.); (D.H.); (M.H.); (R.G.-M.); (C.M.)
| | - Gloria González-Aseguinolaza
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain;
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Felipe Prosper
- Center for Applied Medical Research (CIMA), Regenerative Medicine Department, University of Navarra, 31008 Pamplona, Spain; (P.G.-O.); (G.A.); (Á.V.); (S.R.-D.); (F.P.)
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain;
- Program of Gene Therapy, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain
- Department of Hematology and Cell Therapy, Clínica Universidad de Navarra, 30008 Pamplona, Spain
| | - Beatriz Pelacho
- Center for Applied Medical Research (CIMA), Regenerative Medicine Department, University of Navarra, 31008 Pamplona, Spain; (P.G.-O.); (G.A.); (Á.V.); (S.R.-D.); (F.P.)
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain;
| | - Antonio Bernad
- Cardiac Stem Cells Lab, Centro Nacional de Biotecnología (CNB-CSIC), Department of Immunology and Oncology, Campus Universidad Autónoma de Madrid, 28049 Madrid, Spain; (S.A.); (L.A.-M.); (J.L.T.); (G.A.); (D.H.); (M.H.); (R.G.-M.); (C.M.)
| |
Collapse
|
7
|
Blanco-Blázquez V, Báez-Díaz C, Sánchez-Margallo FM, González-Bueno I, Martín H, Blázquez R, Casado JG, Usón A, Solares J, Palacios I, Steendam R, Crisóstomo V. Intracoronary Administration of Microencapsulated HGF in a Reperfused Myocardial Infarction Swine Model. J Cardiovasc Dev Dis 2023; 10:86. [PMID: 36826582 PMCID: PMC9960949 DOI: 10.3390/jcdd10020086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/09/2023] [Accepted: 02/15/2023] [Indexed: 02/19/2023] Open
Abstract
Therapy microencapsulation allows minimally invasive, safe, and effective administration. Hepatocyte growth factor (HGF) has angiogenic, anti-inflammatory, anti-apoptotic, and anti-fibrotic properties. Our objective was to evaluate the cardiac safety and effectiveness of intracoronary (IC) administration of HGF-loaded extended release microspheres in an acute myocardial infarction (AMI) swine model. An IC infusion of 5 × 106 HGF-loaded microspheres (MS+HGF, n = 7), 5 × 106 placebo microspheres (MS, n = 7), or saline (SAL, n = 7) was performed two days after AMI. TIMI flow and Troponin I (TnI) values were assessed pre- and post-treatment. Cardiac function was evaluated with magnetic resonance imaging (cMR) before injection and at 10 weeks. Plasma cytokines were determined to evaluate the inflammatory profile and hearts were subjected to histopathological evaluation. Post-treatment coronary flow was impaired in five animals (MS+HGF and MS group) without significant increases in TnI. One animal (MS group) died during treatment. There were no significant differences between groups in cMR parameters at any time (p > 0.05). No statistically significant changes were found between groups neither in cytokines nor in histological analyses. The IC administration of 5 × 106 HGF-loaded-microspheres 48 h post-AMI did not improve cardiac function, nor did it decrease inflammation or cardiac fibrosis in this experimental setting.
Collapse
Affiliation(s)
- Virginia Blanco-Blázquez
- Cardiovascular Area, Jesús Usón Minimally Invasive Surgery Centre, 10071 Cáceres, Spain
- Centro de Investigación Biomédica En Red de Enfermedades Cardiovasculares CIBERCV, 28029 Madrid, Spain
| | - Claudia Báez-Díaz
- Cardiovascular Area, Jesús Usón Minimally Invasive Surgery Centre, 10071 Cáceres, Spain
- Centro de Investigación Biomédica En Red de Enfermedades Cardiovasculares CIBERCV, 28029 Madrid, Spain
| | - Francisco Miguel Sánchez-Margallo
- Cardiovascular Area, Jesús Usón Minimally Invasive Surgery Centre, 10071 Cáceres, Spain
- Centro de Investigación Biomédica En Red de Enfermedades Cardiovasculares CIBERCV, 28029 Madrid, Spain
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre, 10071 Cáceres, Spain
| | - Irene González-Bueno
- Cardiovascular Area, Jesús Usón Minimally Invasive Surgery Centre, 10071 Cáceres, Spain
| | - Helena Martín
- Cardiovascular Area, Jesús Usón Minimally Invasive Surgery Centre, 10071 Cáceres, Spain
| | - Rebeca Blázquez
- Centro de Investigación Biomédica En Red de Enfermedades Cardiovasculares CIBERCV, 28029 Madrid, Spain
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre, 10071 Cáceres, Spain
| | - Javier G. Casado
- Centro de Investigación Biomédica En Red de Enfermedades Cardiovasculares CIBERCV, 28029 Madrid, Spain
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre, 10071 Cáceres, Spain
- Immunology Unit, University of Extremadura, 10003 Cáceres, Spain
| | - Alejandra Usón
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre, 10071 Cáceres, Spain
| | | | | | - Rob Steendam
- Innocore Pharmaceuticals, 9713 GX Groningen, The Netherlands
| | - Verónica Crisóstomo
- Cardiovascular Area, Jesús Usón Minimally Invasive Surgery Centre, 10071 Cáceres, Spain
- Centro de Investigación Biomédica En Red de Enfermedades Cardiovasculares CIBERCV, 28029 Madrid, Spain
| |
Collapse
|
8
|
Mabotuwana NS, Rech L, Lim J, Hardy SA, Murtha LA, Rainer PP, Boyle AJ. Paracrine Factors Released by Stem Cells of Mesenchymal Origin and their Effects in Cardiovascular Disease: A Systematic Review of Pre-clinical Studies. Stem Cell Rev Rep 2022; 18:2606-2628. [PMID: 35896860 PMCID: PMC9622561 DOI: 10.1007/s12015-022-10429-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/12/2022] [Indexed: 11/30/2022]
Abstract
Mesenchymal stem cell (MSC) therapy has gained significant traction in the context of cardiovascular repair, and have been proposed to exert their regenerative effects via the secretion of paracrine factors. In this systematic review, we examined the literature and consolidated available evidence for the "paracrine hypothesis". Two Ovid SP databases were searched using a strategy encompassing paracrine mediated MSC therapy in the context of ischemic heart disease. This yielded 86 articles which met the selection criteria for inclusion in this study. We found that the MSCs utilized in these articles were primarily derived from bone marrow, cardiac tissue, and adipose tissue. We identified 234 individual protective factors across these studies, including VEGF, HGF, and FGF2; which are proposed to exert their effects in a paracrine manner. The data collated in this systematic review identifies secreted paracrine factors that could decrease apoptosis, and increase angiogenesis, cell proliferation, and cell viability. These included studies have also demonstrated that the administration of MSCs and indirectly, their secreted factors can reduce infarct size, and improve left ventricular ejection fraction, contractility, compliance, and vessel density. Furthering our understanding of the way these factors mediate repair could lead to the identification of therapeutic targets for cardiac regeneration.
Collapse
Affiliation(s)
- Nishani S Mabotuwana
- College of Health, Medicine and Wellbeing, The University of Newcastle, Newcastle, NSW, Australia
- Hunter Medical Research Institute, Lot 1, Kookaburra Circuit, Newcastle, NSW, 2305, Australia
- Department of Internal Medicine, Division of Cardiology, Medical University of Graz, Graz, Austria
| | - Lavinia Rech
- Department of Internal Medicine, Division of Cardiology, Medical University of Graz, Graz, Austria
- Department of Cardiac Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Joyce Lim
- College of Health, Medicine and Wellbeing, The University of Newcastle, Newcastle, NSW, Australia
- Hunter Medical Research Institute, Lot 1, Kookaburra Circuit, Newcastle, NSW, 2305, Australia
- Department of Cardiovascular Medicine, John Hunter Hospital, Newcastle, NSW, Australia
| | - Sean A Hardy
- College of Health, Medicine and Wellbeing, The University of Newcastle, Newcastle, NSW, Australia
- Hunter Medical Research Institute, Lot 1, Kookaburra Circuit, Newcastle, NSW, 2305, Australia
- Department of Internal Medicine, Division of Cardiology, Medical University of Graz, Graz, Austria
| | - Lucy A Murtha
- College of Health, Medicine and Wellbeing, The University of Newcastle, Newcastle, NSW, Australia
- Hunter Medical Research Institute, Lot 1, Kookaburra Circuit, Newcastle, NSW, 2305, Australia
| | - Peter P Rainer
- Department of Internal Medicine, Division of Cardiology, Medical University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| | - Andrew J Boyle
- College of Health, Medicine and Wellbeing, The University of Newcastle, Newcastle, NSW, Australia.
- Hunter Medical Research Institute, Lot 1, Kookaburra Circuit, Newcastle, NSW, 2305, Australia.
- Department of Cardiovascular Medicine, John Hunter Hospital, Newcastle, NSW, Australia.
| |
Collapse
|
9
|
Im GB, Lin RZ. Bioengineering for vascularization: Trends and directions of photocrosslinkable gelatin methacrylate hydrogels. Front Bioeng Biotechnol 2022; 10:1053491. [PMID: 36466323 PMCID: PMC9713639 DOI: 10.3389/fbioe.2022.1053491] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 11/03/2022] [Indexed: 10/17/2023] Open
Abstract
Gelatin methacrylate (GelMA) hydrogels have been widely used in various biomedical applications, especially in tissue engineering and regenerative medicine, for their excellent biocompatibility and biodegradability. GelMA crosslinks to form a hydrogel when exposed to light irradiation in the presence of photoinitiators. The mechanical characteristics of GelMA hydrogels are highly tunable by changing the crosslinking conditions, including the GelMA polymer concentration, degree of methacrylation, light wavelength and intensity, and light exposure time et al. In this regard, GelMA hydrogels can be adjusted to closely resemble the native extracellular matrix (ECM) properties for the specific functions of target tissues. Therefore, this review focuses on the applications of GelMA hydrogels for bioengineering human vascular networks in vitro and in vivo. Since most tissues require vasculature to provide nutrients and oxygen to individual cells, timely vascularization is critical to the success of tissue- and cell-based therapies. Recent research has demonstrated the robust formation of human vascular networks by embedding human vascular endothelial cells and perivascular mesenchymal cells in GelMA hydrogels. Vascular cell-laden GelMA hydrogels can be microfabricated using different methodologies and integrated with microfluidic devices to generate a vasculature-on-a-chip system for disease modeling or drug screening. Bioengineered vascular networks can also serve as build-in vasculature to ensure the adequate oxygenation of thick tissue-engineered constructs. Meanwhile, several reports used GelMA hydrogels as implantable materials to deliver therapeutic cells aiming to rebuild the vasculature in ischemic wounds for repairing tissue injuries. Here, we intend to reveal present work trends and provide new insights into the development of clinically relevant applications based on vascularized GelMA hydrogels.
Collapse
Affiliation(s)
- Gwang-Bum Im
- Department of Cardiac Surgery, Boston Children’s Hospital, Boston, MA, United States
- Department of Surgery, Harvard Medical School, Boston, MA, United States
| | - Ruei-Zeng Lin
- Department of Cardiac Surgery, Boston Children’s Hospital, Boston, MA, United States
- Department of Surgery, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
10
|
Suhar RA, Doulames VM, Liu Y, Hefferon ME, Figueroa O, Buabbas H, Heilshorn SC. Hyaluronan and elastin-like protein (HELP) gels significantly improve microsphere retention in the myocardium. Biomater Sci 2022; 10:2590-2608. [PMID: 35411353 PMCID: PMC9123900 DOI: 10.1039/d1bm01890f] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2023]
Abstract
Heart disease is the leading cause of death globally, and delivery of therapeutic cargo (e.g., particles loaded with proteins, drugs, or genes and cells) through direct injection into the myocardium is a promising clinical intervention. However, retention of deliverables to the contracting myocardium is low, with as much as 60-90% of payload being lost within 24 hr. Commercially-available injectable hydrogels, including Matrigel, have been hypothesized to increase payload retention but have not yielded significant improvements in quantified analyses. Here, we assess a recombinant hydrogel composed of chemically modified hyaluronan and elastin-like protein (HELP) as an alternative injectable carrier to increase cargo retention. HELP is crosslinked using dynamic covalent bonds, and tuning the hyaluronan chemistry significantly alters hydrogel mechanical properties including stiffness, stress relaxation rate, and ease of injectability through a needle or catheter. These materials can be injected even after complete crosslinking, extending the time window for surgical delivery. We show that HELP gels significantly improve in vivo retention of microsphere cargo compared to Matrigel, both 1 day and 7 days post-injection directly into the rat myocardium. These data suggest that HELP gels may assist with the clinical translation of therapeutic cargo designed for delivery into the contracting myocardium by preventing acute cargo loss.
Collapse
Affiliation(s)
- Riley A Suhar
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, USA.
| | - Vanessa M Doulames
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, USA.
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Yueming Liu
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, USA.
| | - Meghan E Hefferon
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, USA.
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California 94305, USA
| | | | - Hana Buabbas
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, USA.
- Department of Biology, Stanford University, Stanford, California, 94305, USA
| | - Sarah C Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, USA.
| |
Collapse
|
11
|
Efficacy of Stem Cell Therapy in Large Animal Models of Ischemic Cardiomyopathies: A Systematic Review and Meta-Analysis. Animals (Basel) 2022; 12:ani12060749. [PMID: 35327146 PMCID: PMC8944644 DOI: 10.3390/ani12060749] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/09/2022] [Accepted: 03/14/2022] [Indexed: 12/13/2022] Open
Abstract
Stem-cell therapy provides a promising strategy for patients with ischemic heart disease. In recent years, numerous studies related to this therapeutic approach were performed; however, the results were often heterogeneous and contradictory. For this reason, we conducted a systematic review and meta-analysis of trials, reporting the use of stem-cell treatment against acute or chronic ischemic cardiomyopathies in large animal models with regard to Left Ventricular Ejection Fraction (LVEF). The defined research strategy was applied to the PubMed database to identify relevant studies published from January 2011 to July 2021. A random-effect meta-analysis was performed on LVEF mean data at follow-up between control and stem-cell-treated animals. In order to improve the definition of the effect measure and to analyze the factors that could influence the outcomes, a subgroup comparison was conducted. Sixty-six studies (n = 1183 animals) satisfied our inclusion criteria. Ischemia/reperfusion infarction was performed in 37 studies, and chronic occlusion in 29 studies; moreover, 58 studies were on a pig animal model. The meta-analysis showed that cell therapy increased LVEF by 7.41% (95% Confidence Interval 6.23−8.59%; p < 0.001) at follow-up, with significative heterogeneity and high inconsistency (I2 = 82%, p < 0.001). By subgroup comparison, the follow-up after 31−60 days (p = 0.025), the late cell injection (>7 days, p = 0.005) and the route of cellular delivery by surgical treatment (p < 0.001) were significant predictors of LVEF improvement. This meta-analysis showed that stem-cell therapy may improve heart function in large animal models and that the swine specie is confirmed as a relevant animal model in the cardiovascular field. Due to the significative heterogeneity and high inconsistency, future translational studies should be designed to take into account the evidenced predictors to allow for the reduction of the number of animals used.
Collapse
|
12
|
Albericio G, Aguilar S, Torán JL, Yañez R, López JA, Vázquez J, Mora C, Bernad A. Comparative proteomic analysis of nuclear and cytoplasmic compartments in human cardiac progenitor cells. Sci Rep 2022; 12:146. [PMID: 34997006 PMCID: PMC8742012 DOI: 10.1038/s41598-021-03956-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 12/08/2021] [Indexed: 11/16/2022] Open
Abstract
Clinical trials evaluating cardiac progenitor cells (CPC) demonstrated feasibility and safety, but no clear functional benefits. Therefore a deeper understanding of CPC biology is warranted to inform strategies capable to enhance their therapeutic potential. Here we have defined, using a label-free proteomic approach, the differential cytoplasmic and nuclear compartments of human CPC (hCPC). Global analysis of cytoplasmic repertoire in hCPC suggested an important hypoxia response capacity and active collagen metabolism. In addition, comparative analysis of the nuclear protein compartment identified a significant regulation of a small number of proteins in hCPC versus human mesenchymal stem cells (hMSC). Two proteins significantly upregulated in the hCPC nuclear compartment, IL1A and IMP3, showed also a parallel increase in mRNA expression in hCPC versus hMSC, and were studied further. IL1A, subjected to an important post-transcriptional regulation, was demonstrated to act as a dual-function cytokine with a plausible role in apoptosis regulation. The knockdown of the mRNA binding protein (IMP3) did not negatively impact hCPC viability, but reduced their proliferation and migration capacity. Analysis of a panel of putative candidate genes identified HMGA2 and PTPRF as IMP3 targets in hCPC. Therefore, they are potentially involved in hCPC proliferation/migration regulation.
Collapse
Affiliation(s)
- Guillermo Albericio
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), C/ Darwin 3, Campus Universidad Autónoma de Madrid, 28049, Madrid, Spain
| | - Susana Aguilar
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), C/ Darwin 3, Campus Universidad Autónoma de Madrid, 28049, Madrid, Spain
| | - Jose Luis Torán
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), C/ Darwin 3, Campus Universidad Autónoma de Madrid, 28049, Madrid, Spain.,Veterinary Faculty, Universidad Complutense de Madrid, Avda. Puerta de Hierro, s/n. Ciudad Universitaria, 28040, Madrid, Spain
| | - Rosa Yañez
- Hematopoietic Innovative Therapies Division, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras, Av Complutense, 40, 28040, Madrid, Spain.,Instituto de Investigaciones Sanitarias de la Fundación Jiménez Díaz, Madrid, Spain
| | - Juan Antonio López
- Laboratory of Cardiovascular Proteomics, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Jesús Vázquez
- Laboratory of Cardiovascular Proteomics, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Carmen Mora
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), C/ Darwin 3, Campus Universidad Autónoma de Madrid, 28049, Madrid, Spain
| | - Antonio Bernad
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), C/ Darwin 3, Campus Universidad Autónoma de Madrid, 28049, Madrid, Spain.
| |
Collapse
|
13
|
Crisóstomo V, Baéz-Diaz C, Blanco-Blázquez V, Álvarez V, López-Nieto E, Maestre J, Bayes-Genis A, Gálvez-Montón C, Casado JG, Sánchez-Margallo FM. The epicardial delivery of cardiosphere derived cells or their extracellular vesicles is safe but of limited value in experimental infarction. Sci Rep 2021; 11:22155. [PMID: 34772964 PMCID: PMC8590017 DOI: 10.1038/s41598-021-01728-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 11/02/2021] [Indexed: 02/08/2023] Open
Abstract
The epicardial administration of therapeutics via the pericardial sac offers an attractive route, since it is minimally invasive and carries no risks of coronary embolization. The aim of this study was to assess viability, safety and effectiveness of cardiosphere-derived cells (CDCs), their extracellular vesicles (EVs) or placebo administered via a mini-thoracotomy 72 h after experimental infarction in swine. The epicardial administration was completed successfully in all cases in a surgery time (knife-to-skin) below 30 min. No significant differences between groups were found in cardiac function parameters evaluated using magnetic resonance imaging before therapy and at the end of the study, despite a trend towards improved function in CDC-treated animals. Moreover, infarct size at 10 weeks was smaller in treated animals, albeit not significantly. Arrhythmia inducibility did not differ between groups. Pathological examination showed no differences, nor were there any pericardial adhesions evidenced in any case 10 weeks after surgery. These results show that the epicardial delivery of CDCs or their EVs is safe and technically easy 3 days after experimental myocardial infarction in swine, but it does not appear to have any beneficial effect on cardiac function. Our results do not support clinical translation of these therapies as implemented in this work.
Collapse
Affiliation(s)
- Verónica Crisóstomo
- Fundación Centro de Cirugía de Mínima Invasión Jesús Usón, Carretera N-521, km 41, 10071, Cáceres, Spain. .,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain.
| | - Claudia Baéz-Diaz
- Fundación Centro de Cirugía de Mínima Invasión Jesús Usón, Carretera N-521, km 41, 10071, Cáceres, Spain.,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Virginia Blanco-Blázquez
- Fundación Centro de Cirugía de Mínima Invasión Jesús Usón, Carretera N-521, km 41, 10071, Cáceres, Spain.,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Verónica Álvarez
- Fundación Centro de Cirugía de Mínima Invasión Jesús Usón, Carretera N-521, km 41, 10071, Cáceres, Spain
| | - Esther López-Nieto
- Fundación Centro de Cirugía de Mínima Invasión Jesús Usón, Carretera N-521, km 41, 10071, Cáceres, Spain
| | - Juan Maestre
- Fundación Centro de Cirugía de Mínima Invasión Jesús Usón, Carretera N-521, km 41, 10071, Cáceres, Spain.,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Antoni Bayes-Genis
- CIBERCV, Instituto de Salud Carlos III, Madrid, Spain.,ICREC Research Group (Insuficiència Cardíaca i REgeneració Cardíaca), Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
| | - Carolina Gálvez-Montón
- CIBERCV, Instituto de Salud Carlos III, Madrid, Spain.,ICREC Research Group (Insuficiència Cardíaca i REgeneració Cardíaca), Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
| | - Javier G Casado
- Fundación Centro de Cirugía de Mínima Invasión Jesús Usón, Carretera N-521, km 41, 10071, Cáceres, Spain.,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain.,Immunology Unit, University of Extremadura, Cáceres, Spain.,Institute of Molecular Pathology Biomarkers, University of Extremadura, Cáceres, Spain
| | - Francisco M Sánchez-Margallo
- Fundación Centro de Cirugía de Mínima Invasión Jesús Usón, Carretera N-521, km 41, 10071, Cáceres, Spain.,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
14
|
Báez-Díaz C, Blanco-Blázquez V, Sánchez-Margallo FM, López E, Martín H, Espona-Noguera A, Casado JG, Ciriza J, Pedraz JL, Crisóstomo V. Intrapericardial Delivery of APA-Microcapsules as Promising Stem Cell Therapy Carriers in an Experimental Acute Myocardial Infarction Model. Pharmaceutics 2021; 13:1824. [PMID: 34834235 PMCID: PMC8626005 DOI: 10.3390/pharmaceutics13111824] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 10/15/2021] [Accepted: 10/26/2021] [Indexed: 01/08/2023] Open
Abstract
The administration of cardiosphere-derived cells (CDCs) after acute myocardial infarction (AMI) is very promising. CDC encapsulation in alginate-poly-l-lysine-alginate (APA) could increase cell survival and adherence. The intrapericardial (IP) approach potentially achieves high concentrations of the therapeutic agent in the infarcted area. We aimed to evaluate IP therapy using a saline vehicle as a control (CON), a dose of 30 × 106 CDCs (CDCs) or APA microcapsules containing 30 × 106 CDCs (APA-CDCs) at 72 h in a porcine AMI model. Magnetic resonance imaging (MRI) was used to determine the left ventricular ejection fraction (LVEF), infarct size (IS), and indexed end diastolic and systolic volumes (EDVi; ESVi) pre- and 10 weeks post-injection. Programmed electrical stimulation (PES) was performed to test arrhythmia inducibility before euthanasia. Histopathological analysis was carried out afterwards. The IP infusion was successful in all animals. At 10 weeks, MRI revealed significantly higher LVEF in the APA-CDC group compared with CON. No significant differences were observed among groups in IS, EDVi, ESVi, PES and histopathological analyses. In conclusion, the IP injection of CDCs (microencapsulated or not) was feasible and safe 72 h post-AMI in the porcine model. Moreover, CDCs APA encapsulation could have a beneficial effect on cardiac function, reflected by a higher LVEF at 10 weeks.
Collapse
Affiliation(s)
- Claudia Báez-Díaz
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain; (V.B.-B.); (F.M.S.-M.); (V.C.)
- Fundación Centro de Cirugía de Mínima Invasión Jesús Usón, 10071 Cáceres, Spain; (E.L.); (H.M.)
| | - Virginia Blanco-Blázquez
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain; (V.B.-B.); (F.M.S.-M.); (V.C.)
- Fundación Centro de Cirugía de Mínima Invasión Jesús Usón, 10071 Cáceres, Spain; (E.L.); (H.M.)
| | - Francisco Miguel Sánchez-Margallo
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain; (V.B.-B.); (F.M.S.-M.); (V.C.)
- Fundación Centro de Cirugía de Mínima Invasión Jesús Usón, 10071 Cáceres, Spain; (E.L.); (H.M.)
| | - Esther López
- Fundación Centro de Cirugía de Mínima Invasión Jesús Usón, 10071 Cáceres, Spain; (E.L.); (H.M.)
| | - Helena Martín
- Fundación Centro de Cirugía de Mínima Invasión Jesús Usón, 10071 Cáceres, Spain; (E.L.); (H.M.)
| | - Albert Espona-Noguera
- Centro de Investigaciones y Estudios Avanzados Lucio Lascaray (CIEA), Laboratorio de Desarrollo y Evaluación de Medicamentos, 01006 Vitoria Gasteiz, Spain; (A.E.-N.); (J.L.P.)
- CIBER bbn, Instituto de Salud Carlos III, 28029 Madrid, Spain;
| | - Javier G. Casado
- Immunology Unit-Institute of Molecular Pathology Biomarkers, Veterinary Faculty, University of Extremadura, 10003 Cáceres, Spain;
| | - Jesús Ciriza
- CIBER bbn, Instituto de Salud Carlos III, 28029 Madrid, Spain;
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain
| | - José Luis Pedraz
- Centro de Investigaciones y Estudios Avanzados Lucio Lascaray (CIEA), Laboratorio de Desarrollo y Evaluación de Medicamentos, 01006 Vitoria Gasteiz, Spain; (A.E.-N.); (J.L.P.)
- CIBER bbn, Instituto de Salud Carlos III, 28029 Madrid, Spain;
| | - Verónica Crisóstomo
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain; (V.B.-B.); (F.M.S.-M.); (V.C.)
- Fundación Centro de Cirugía de Mínima Invasión Jesús Usón, 10071 Cáceres, Spain; (E.L.); (H.M.)
| |
Collapse
|
15
|
Prat-Vidal C, Crisóstomo V, Moscoso I, Báez-Díaz C, Blanco-Blázquez V, Gómez-Mauricio G, Albericio G, Aguilar S, Fernández-Santos ME, Fernández-Avilés F, Sánchez-Margallo FM, Bayes-Genis A, Bernad A. Intracoronary Delivery of Porcine Cardiac Progenitor Cells Overexpressing IGF-1 and HGF in a Pig Model of Sub-Acute Myocardial Infarction. Cells 2021; 10:cells10102571. [PMID: 34685551 PMCID: PMC8534140 DOI: 10.3390/cells10102571] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 09/17/2021] [Accepted: 09/24/2021] [Indexed: 12/26/2022] Open
Abstract
Human cardiac progenitor cells (hCPC) are considered a good candidate in cell therapy for ischemic heart disease, demonstrating capacity to improve functional recovery after myocardial infarction (MI), both in small and large preclinical animal models. However, improvements are required in terms of cell engraftment and efficacy. Based on previously published reports, insulin-growth factor 1 (IGF-1) and hepatocyte growth factor (HGF) have demonstrated substantial cardioprotective, repair and regeneration activities, so they are good candidates to be evaluated in large animal model of MI. We have validated porcine cardiac progenitor cells (pCPC) and lentiviral vectors to overexpress IGF-1 (co-expressing eGFP) and HGF (co-expressing mCherry). pCPC were transduced and IGF1-eGFPpos and HGF-mCherrypos populations were purified by cell sorting and further expanded. Overexpression of IGF-1 has a limited impact on pCPC expression profile, whereas results indicated that pCPC-HGF-mCherry cultures could be counter selecting high expresser cells. In addition, pCPC-IGF1-eGFP showed a higher cardiogenic response, evaluated in co-cultures with decellularized extracellular matrix, compared with native pCPC or pCPC-HGF-mCherry. In vivo intracoronary co-administration of pCPC-IGF1-eGFP and pCPC-HFG-mCherry (1:1; 40 × 106/animal), one week after the induction of an MI model in swine, revealed no significant improvement in cardiac function.
Collapse
Affiliation(s)
- Cristina Prat-Vidal
- ICREC Research Program, Germans Trias i Pujol Health Science Research Institute, Can Ruti Campus, Heart Institute (iCor), Germans Trias i Pujol University Hospital, 08916 Badalona, Spain; (C.P.-V.); (A.B.-G.)
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain; (V.C.); (I.M.); (C.B.-D.); (V.B.-B.); (M.-E.F.-S.); (F.F.-A.); (F.M.S.-M.)
- Institut d’Investigació Biomèdica de Bellvitge-IDIBELL, 08908 L’Hospitalet de Llobregat, Spain
| | - Verónica Crisóstomo
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain; (V.C.); (I.M.); (C.B.-D.); (V.B.-B.); (M.-E.F.-S.); (F.F.-A.); (F.M.S.-M.)
- Jesús Usón Minimally Invasive Surgery Center, 10071 Cáceres, Spain;
| | - Isabel Moscoso
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain; (V.C.); (I.M.); (C.B.-D.); (V.B.-B.); (M.-E.F.-S.); (F.F.-A.); (F.M.S.-M.)
- Cardiology Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela and Health Research Institute, University Clinical Hospital of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Claudia Báez-Díaz
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain; (V.C.); (I.M.); (C.B.-D.); (V.B.-B.); (M.-E.F.-S.); (F.F.-A.); (F.M.S.-M.)
- Jesús Usón Minimally Invasive Surgery Center, 10071 Cáceres, Spain;
| | - Virginia Blanco-Blázquez
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain; (V.C.); (I.M.); (C.B.-D.); (V.B.-B.); (M.-E.F.-S.); (F.F.-A.); (F.M.S.-M.)
- Jesús Usón Minimally Invasive Surgery Center, 10071 Cáceres, Spain;
| | | | - Guillermo Albericio
- Immunology and Oncology Department, National Center for Biotechnology, 28049 Madrid, Spain; (G.A.); (S.A.)
| | - Susana Aguilar
- Immunology and Oncology Department, National Center for Biotechnology, 28049 Madrid, Spain; (G.A.); (S.A.)
| | - María-Eugenia Fernández-Santos
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain; (V.C.); (I.M.); (C.B.-D.); (V.B.-B.); (M.-E.F.-S.); (F.F.-A.); (F.M.S.-M.)
- Servicio de Cardiología, Hospital General Universitario Gregorio Marañón, Laboratorio Investigación Traslacional en Cardiología (LITC), Unidad de Producción Celular-GMP (UPC-GMP), Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), TERCEL, 28007 Madrid, Spain
| | - Francisco Fernández-Avilés
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain; (V.C.); (I.M.); (C.B.-D.); (V.B.-B.); (M.-E.F.-S.); (F.F.-A.); (F.M.S.-M.)
- Servicio de Cardiología, Hospital General Universitario Gregorio Marañón, Laboratorio Investigación Traslacional en Cardiología (LITC), Unidad de Producción Celular-GMP (UPC-GMP), Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), TERCEL, 28007 Madrid, Spain
- Departamento de Medicina, Facultad de Medicina, Universidad Complutense de Madrid (UCM), 28040 Madrid, Spain
| | - Francisco M. Sánchez-Margallo
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain; (V.C.); (I.M.); (C.B.-D.); (V.B.-B.); (M.-E.F.-S.); (F.F.-A.); (F.M.S.-M.)
- Jesús Usón Minimally Invasive Surgery Center, 10071 Cáceres, Spain;
| | - Antoni Bayes-Genis
- ICREC Research Program, Germans Trias i Pujol Health Science Research Institute, Can Ruti Campus, Heart Institute (iCor), Germans Trias i Pujol University Hospital, 08916 Badalona, Spain; (C.P.-V.); (A.B.-G.)
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain; (V.C.); (I.M.); (C.B.-D.); (V.B.-B.); (M.-E.F.-S.); (F.F.-A.); (F.M.S.-M.)
- Cardiology Service, Germans Trias i Pujol University Hospital, 08916 Badalona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Antonio Bernad
- Immunology and Oncology Department, National Center for Biotechnology, 28049 Madrid, Spain; (G.A.); (S.A.)
- Correspondence: ; Tel.: +34-915-855-424
| |
Collapse
|
16
|
Naumova AV, Kicska G, Pimentel K, Neidig LE, Tsuchida H, Nakamura K, Murry CE. Quantitative Analyses of the Left Ventricle Volume and Cardiac Function in Normal and Infarcted Yucatan Minipigs. J Imaging 2021; 7:107. [PMID: 39080895 PMCID: PMC8321358 DOI: 10.3390/jimaging7070107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/18/2021] [Accepted: 06/28/2021] [Indexed: 11/16/2022] Open
Abstract
(1) Background: The accuracy of the left ventricular volume (LVV) and contractility measurements with cardiac magnetic resonance imaging (CMRI) is decreased if the papillary muscles are abnormally enlarged, such as in hypertrophic cardiomyopathy in human patients or in pig models of human diseases. The purpose of this work was to establish the best method of LVV quantification with CMRI in pigs. (2) Methods: The LVV in 29 Yucatan minipig hearts was measured using two different techniques: the "standard method", which uses smooth contouring along the endocardial surface and adds the papillary volume to the ventricular cavity volume, and the "detailed method", which traces the papillary muscles and trabeculations and adds them to the ventricular mass. (3) Results: Papillary muscles add 21% to the LV mass in normal and infarcted hearts of Yucatan minipigs. The inclusion or exclusion of these from the CMRI analysis significantly affected the study results. In the normal pig hearts, the biggest differences were found in measurements of the LVV, ejection fraction (EF), LV mass and indices derived from the LV mass (p < 0.001). The EF measurement in the normal pig heart was 11% higher with the detailed method, and 19% higher in the infarcted pig hearts (p < 0.0001). The detailed method of endocardium tracing with CMRI closely represented the LV mass measured ex vivo. (4) Conclusions: The detailed method, which accounts for the large volume of the papillary muscles in the pig heart, provides better accuracy and interobserver consistency in the assessment of LV mass and ejection fraction, and might therefore be preferable for these analyses.
Collapse
Affiliation(s)
- Anna V. Naumova
- Department of Radiology, University of Washington, Seattle, WA 98109, USA; (G.K.); (K.P.)
- Institute for Stem Cells and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA; (L.E.N.); (H.T.); (K.N.); (C.E.M.)
| | - Gregory Kicska
- Department of Radiology, University of Washington, Seattle, WA 98109, USA; (G.K.); (K.P.)
| | - Kiana Pimentel
- Department of Radiology, University of Washington, Seattle, WA 98109, USA; (G.K.); (K.P.)
| | - Lauren E. Neidig
- Institute for Stem Cells and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA; (L.E.N.); (H.T.); (K.N.); (C.E.M.)
- Department of Pathology, University of Washington, Seattle, WA 98109, USA
- Department of Comparative Medicine, University of Washington, Seattle, WA 98109, USA
| | - Hiroshi Tsuchida
- Institute for Stem Cells and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA; (L.E.N.); (H.T.); (K.N.); (C.E.M.)
- Department of Pathology, University of Washington, Seattle, WA 98109, USA
| | - Kenta Nakamura
- Institute for Stem Cells and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA; (L.E.N.); (H.T.); (K.N.); (C.E.M.)
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Charles E. Murry
- Institute for Stem Cells and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA; (L.E.N.); (H.T.); (K.N.); (C.E.M.)
- Department of Pathology, University of Washington, Seattle, WA 98109, USA
| |
Collapse
|
17
|
Shin HS, Shin HH, Shudo Y. Current Status and Limitations of Myocardial Infarction Large Animal Models in Cardiovascular Translational Research. Front Bioeng Biotechnol 2021; 9:673683. [PMID: 33996785 PMCID: PMC8116580 DOI: 10.3389/fbioe.2021.673683] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 04/06/2021] [Indexed: 01/16/2023] Open
Abstract
Establishing an appropriate disease model that mimics the complexities of human cardiovascular disease is critical for evaluating the clinical efficacy and translation success. The multifaceted and complex nature of human ischemic heart disease is difficult to recapitulate in animal models. This difficulty is often compounded by the methodological biases introduced in animal studies. Considerable variations across animal species, modifications made in surgical procedures, and inadequate randomization, sample size calculation, blinding, and heterogeneity of animal models used often produce preclinical cardiovascular research that looks promising but is irreproducible and not translatable. Moreover, many published papers are not transparent enough for other investigators to verify the feasibility of the studies and the therapeutics' efficacy. Unfortunately, successful translation of these innovative therapies in such a closed and biased research is difficult. This review discusses some challenges in current preclinical myocardial infarction research, focusing on the following three major inhibitors for its successful translation: Inappropriate disease model, frequent modifications to surgical procedures, and insufficient reporting transparency.
Collapse
Affiliation(s)
- Hye Sook Shin
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, United States
| | - Heather Hyeyoon Shin
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, United States
| | - Yasuhiro Shudo
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
18
|
Li J, Hu S, Zhu D, Huang K, Mei X, López de Juan Abad B, Cheng K. All Roads Lead to Rome (the Heart): Cell Retention and Outcomes From Various Delivery Routes of Cell Therapy Products to the Heart. J Am Heart Assoc 2021; 10:e020402. [PMID: 33821664 PMCID: PMC8174178 DOI: 10.1161/jaha.120.020402] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In the past decades, numerous preclinical studies and several clinical trials have evidenced the feasibility of cell transplantation in treating heart diseases. Over the years, different delivery routes of cell therapy have emerged and broadened the width of the field. However, a common hurdle is shared by all current delivery routes: low cell retention. A myriad of studies confirm that cell retention plays a crucial role in the success of cell-mediated cardiac repair. It is important for any delivery route to maintain donor cells in the recipient heart for enough time to not only proliferate by themselves, but also to send paracrine signals to surrounding damaged heart cells and repair them. In this review, we first undertake an in-depth study of primary theories of cell loss, including low efficiency in cell injection, "washout" effects, and cell death, and then organize the literature from the past decade that focuses on cell transplantation to the heart using various cell delivery routes, including intracoronary injection, systemic intravenous injection, retrograde coronary venous injection, and intramyocardial injection. In addition to a recapitulation of these approaches, we also clearly evaluate their strengths and weaknesses. Furthermore, we conduct comparative research on the cell retention rate and functional outcomes of these delivery routes. Finally, we extend our discussion to state-of-the-art bioengineering techniques that enhance cell retention, as well as alternative delivery routes, such as intrapericardial delivery. A combination of these novel strategies and more accurate assessment methods will help to address the hurdle of low cell retention and boost the efficacy of cell transplantation to the heart.
Collapse
Affiliation(s)
- Junlang Li
- Department of Molecular Biomedical SciencesNorth Carolina State UniversityRaleighNC
- Joint Department of Biomedical EngineeringNorth Carolina State University and University of North Carolina at Chapel HillRaleighNC
| | - Shiqi Hu
- Department of Molecular Biomedical SciencesNorth Carolina State UniversityRaleighNC
- Joint Department of Biomedical EngineeringNorth Carolina State University and University of North Carolina at Chapel HillRaleighNC
| | - Dashuai Zhu
- Department of Molecular Biomedical SciencesNorth Carolina State UniversityRaleighNC
- Joint Department of Biomedical EngineeringNorth Carolina State University and University of North Carolina at Chapel HillRaleighNC
| | - Ke Huang
- Department of Molecular Biomedical SciencesNorth Carolina State UniversityRaleighNC
- Joint Department of Biomedical EngineeringNorth Carolina State University and University of North Carolina at Chapel HillRaleighNC
| | - Xuan Mei
- Department of Molecular Biomedical SciencesNorth Carolina State UniversityRaleighNC
- Joint Department of Biomedical EngineeringNorth Carolina State University and University of North Carolina at Chapel HillRaleighNC
| | - Blanca López de Juan Abad
- Department of Molecular Biomedical SciencesNorth Carolina State UniversityRaleighNC
- Joint Department of Biomedical EngineeringNorth Carolina State University and University of North Carolina at Chapel HillRaleighNC
| | - Ke Cheng
- Department of Molecular Biomedical SciencesNorth Carolina State UniversityRaleighNC
- Joint Department of Biomedical EngineeringNorth Carolina State University and University of North Carolina at Chapel HillRaleighNC
| |
Collapse
|
19
|
Sousonis V, Sfakianaki T, Ntalianis A, Nanas I, Kontogiannis C, Aravantinos D, Kapelios C, Katsaros L, Nana M, Sampaziotis D, Sanoudou D, Papalois A, Malliaras K. Intracoronary Administration of Allogeneic Cardiosphere-Derived Cells Immediately Prior to Reperfusion in Pigs With Acute Myocardial Infarction Reduces Infarct Size and Attenuates Adverse Cardiac Remodeling. J Cardiovasc Pharmacol Ther 2021; 26:88-99. [PMID: 32677460 DOI: 10.1177/1074248420941672] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Allogeneic cardiosphere-derived cells (CDCs) exert cardioprotective effects when administered intracoronarily after reperfusion in animal models of acute myocardial infarction (AMI). The "no-reflow" phenomenon develops rapidly post-reperfusion and may undermine the efficacy of cell therapy, due to poor cell delivery in areas of microvascular obstruction (MVO). We hypothesized that CDC-induced cardioprotection would be enhanced by cell administration prior to reperfusion, when microvasculature is still relatively intact, to facilitate widespread cell delivery within the ischemic area. METHODS AND RESULTS We studied 81 farm pigs; 55 completed the specified protocols. A dose-optimization study in infarcted pigs demonstrated that the doses of 5 million and 10 million CDCs are the maximum safe doses that can be administered intracoronarily at 5 minutes prior to and at 5 minutes post-reperfusion, respectively, without aggravating MVO. Quantification of acute cell retention by polymerase chain reaction demonstrated that cell delivery prior to reperfusion resulted in higher cardiac cell retention compared to delivery post-reperfusion. We then performed a randomized, placebo-controlled study to assess the long-term efficacy of intracoronary infusion of 5 million allogeneic CDCs, delivered at 5 minutes prior to reperfusion, in a porcine model of AMI. The CDC therapy resulted in decreased scar size, improved regional systolic function, and attenuation of adverse cardiac remodeling (manifested as preserved global systolic function, preserved end-systolic volume, and decreased interstitial fibrosis) compared to placebo at 30 days post-MI. CONCLUSIONS Dose-optimized intracoronary infusion of allogeneic CDCs prior to reperfusion in a porcine model of AMI is feasible, safe and confers long-term benefits.
Collapse
Affiliation(s)
- Vasileios Sousonis
- Third Department of Cardiology, University of Athens School of Medicine, Athens, Greece
- Center for New Biotechnologies and Precision Medicine, University of Athens School of Medicine, Athens, Greece
| | - Titika Sfakianaki
- Third Department of Cardiology, University of Athens School of Medicine, Athens, Greece
| | - Argirios Ntalianis
- Third Department of Cardiology, University of Athens School of Medicine, Athens, Greece
| | - Ioannis Nanas
- Third Department of Cardiology, University of Athens School of Medicine, Athens, Greece
| | - Christos Kontogiannis
- Third Department of Cardiology, University of Athens School of Medicine, Athens, Greece
| | - Dionysios Aravantinos
- Third Department of Cardiology, University of Athens School of Medicine, Athens, Greece
| | - Chris Kapelios
- Third Department of Cardiology, University of Athens School of Medicine, Athens, Greece
| | - Lampros Katsaros
- Third Department of Cardiology, University of Athens School of Medicine, Athens, Greece
| | - Maria Nana
- Third Department of Cardiology, University of Athens School of Medicine, Athens, Greece
| | | | - Despina Sanoudou
- Center for New Biotechnologies and Precision Medicine, University of Athens School of Medicine, Athens, Greece
- Fouth Department of Internal Medicine, Clinical Genomics and Pharmacogenomics Unit, Attikon Hospital, University of Athens School of Medicine, Athens, Greece
| | - Apostolos Papalois
- Experimental Educational and Research Center, ELPEN Pharmaceuticals, Athens, Greece
| | | |
Collapse
|
20
|
Galow AM, Goldammer T, Hoeflich A. Xenogeneic and Stem Cell-Based Therapy for Cardiovascular Diseases: Genetic Engineering of Porcine Cells and Their Applications in Heart Regeneration. Int J Mol Sci 2020; 21:ijms21249686. [PMID: 33353186 PMCID: PMC7766969 DOI: 10.3390/ijms21249686] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/11/2020] [Accepted: 12/15/2020] [Indexed: 12/25/2022] Open
Abstract
Cardiovascular diseases represent a major health concern worldwide with few therapy options for ischemic injuries due to the limited regeneration potential of affected cardiomyocytes. Innovative cell replacement approaches could facilitate efficient regenerative therapy. However, despite extensive attempts to expand primary human cells in vitro, present technological limitations and the lack of human donors have so far prevented their broad clinical use. Cell xenotransplantation might provide an ethically acceptable unlimited source for cell replacement therapies and bridge the gap between waiting recipients and available donors. Pigs are considered the most suitable candidates as a source for xenogeneic cells and tissues due to their anatomical and physiological similarities with humans. The potential of porcine cells in the field of stem cell-based therapy and regenerative medicine is under intensive investigation. This review outlines the current progress and highlights the most promising approaches in xenogeneic cell therapy with a focus on the cardiovascular system.
Collapse
Affiliation(s)
- Anne-Marie Galow
- Institute of Genome Biology, Leibniz Institute for Farm Animal Biology, 18196 Dummerstorf, Germany; (T.G.); (A.H.)
- Correspondence: ; Tel.: +49-38208-68-723
| | - Tom Goldammer
- Institute of Genome Biology, Leibniz Institute for Farm Animal Biology, 18196 Dummerstorf, Germany; (T.G.); (A.H.)
- Molecular Biology and Fish Genetics Unit, Faculty of Agriculture and Environmental Sciences, University of Rostock, 18059 Rostock, Germany
| | - Andreas Hoeflich
- Institute of Genome Biology, Leibniz Institute for Farm Animal Biology, 18196 Dummerstorf, Germany; (T.G.); (A.H.)
| |
Collapse
|
21
|
Plasmatic Membrane Expression of Adhesion Molecules in Human Cardiac Progenitor/Stem Cells Might Explain Their Superior Cell Engraftment after Cell Transplantation. Stem Cells Int 2020; 2020:8872009. [PMID: 33101423 PMCID: PMC7569451 DOI: 10.1155/2020/8872009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/15/2020] [Accepted: 09/24/2020] [Indexed: 01/12/2023] Open
Abstract
Human bone marrow mesenchymal stem cells (BM-MSCs) and cardiac progenitor/stem cells (CPCs) have been extensively studied as a potential therapeutic treatment for myocardial infarction (MI). Previous reports suggest that lower doses of CPCs are needed to improve cardiac function relative to their bone marrow counterparts. Here, we confirmed this observations and investigated the surface protein expression profile that might explain this effect. Myocardial infarction was performed in nude rats by permanent ligation of the left coronary artery. Cardiac function and infarct size before and after cell transplantation were evaluated by echocardiography and morphometry, respectively. The CPC and BM-MSC receptome were analyzed by proteomic analysis of biotin-labeled surface proteins. Rats transplanted with CPCs showed a greater improvement in cardiac function after MI than those transplanted with BM-MSCs, and this was associated with a smaller infarct size. Analysis of the receptome of CPCs and BM-MSCs showed that gene ontology biological processes and KEGG pathways associated with adhesion mechanisms were upregulated in CPCs compared with BM-MSCs. Moreover, the membrane protein interactome in CPCs showed a strong relationship with biological processes related to cell adhesion whereas the BM-MSCs interactome was more related to immune regulation processes. We conclude that the stronger capacity of CPCs over BM-MSCs to engraft in the infarcted area is likely linked to a more pronounced cell adhesion expression program.
Collapse
|
22
|
Cobo AA, Margallo FMS, Díaz CB, Blázquez VB, Bueno IG, Crisóstomo V. Anesthesia Protocols used to Create Ischemia Reperfusion Myocardial Infarcts in Swine. JOURNAL OF THE AMERICAN ASSOCIATION FOR LABORATORY ANIMAL SCIENCE 2020; 59:478-487. [PMID: 32709259 DOI: 10.30802/aalas-jaalas-19-000137] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The porcine ischemia-reperfusion model is one of the most commonly used for cardiology research and for testing interventions for myocardial regeneration. In creating ischemic reperfusion injury, the anesthetic protocol is important for assuring hemodynamic stability of the animal during the induction of the experimental lesion and may affect its postoperative survival. This paper reviews the many drugs and anesthetic protocols used in recent studies involving porcine models of ischemiareperfusion injury. The paper also summarizes the most important characteristics of some commonly used anesthetic drugs. Literature was selected for inclusion in this review if the authors described the anesthetic protocol used and also reported the mortality rate attributed to the creation of the model. This information is an important consideration because the anesthetic protocol can influence hemodynamic stability during the experimental induction of an acute myocardial infarction, thereby impacting the survival rate and affecting the number of animals needed for each study.
Collapse
Affiliation(s)
- Ana Abad Cobo
- Jesús Usón Minimally Invasive Surgery Centre, Cáceres, Spain;,
| | | | - Claudia Báez Díaz
- Jesús Usón Minimally Invasive Surgery Centre, Cáceres, Spain; CIBERCV, Madrid, Spain
| | | | | | - Verónica Crisóstomo
- Jesús Usón Minimally Invasive Surgery Centre, Cáceres, Spain; CIBERCV, Madrid, Spain
| |
Collapse
|
23
|
Mancuso T, Barone A, Salatino A, Molinaro C, Marino F, Scalise M, Torella M, De Angelis A, Urbanek K, Torella D, Cianflone E. Unravelling the Biology of Adult Cardiac Stem Cell-Derived Exosomes to Foster Endogenous Cardiac Regeneration and Repair. Int J Mol Sci 2020; 21:E3725. [PMID: 32466282 PMCID: PMC7279257 DOI: 10.3390/ijms21103725] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 05/15/2020] [Accepted: 05/20/2020] [Indexed: 12/11/2022] Open
Abstract
Cardiac remuscularization has been the stated goal of the field of regenerative cardiology since its inception. Along with the refreshment of lost and dysfunctional cardiac muscle cells, the field of cell therapy has expanded in scope encompassing also the potential of the injected cells as cardioprotective and cardio-reparative agents for cardiovascular diseases. The latter has been the result of the findings that cell therapies so far tested in clinical trials exert their beneficial effects through paracrine mechanisms acting on the endogenous myocardial reparative/regenerative potential. The endogenous regenerative potential of the adult heart is still highly debated. While it has been widely accepted that adult cardiomyocytes (CMs) are renewed throughout life either in response to wear and tear and after injury, the rate and origin of this phenomenon are yet to be clarified. The adult heart harbors resident cardiac/stem progenitor cells (CSCs/CPCs), whose discovery and characterization were initially sufficient to explain CM renewal in response to physiological and pathological stresses, when also considering that adult CMs are terminally differentiated cells. The role of CSCs in CM formation in the adult heart has been however questioned by some recent genetic fate map studies, which have been proved to have serious limitations. Nevertheless, uncontested evidence shows that clonal CSCs are effective transplantable regenerative agents either for their direct myogenic differentiation and for their paracrine effects in the allogeneic setting. In particular, the paracrine potential of CSCs has been the focus of the recent investigation, whereby CSC-derived exosomes appear to harbor relevant regenerative and reparative signals underlying the beneficial effects of CSC transplantation. This review focuses on recent advances in our knowledge about the biological role of exosomes in heart tissue homeostasis and repair with the idea to use them as tools for new therapeutic biotechnologies for "cell-less" effective cardiac regeneration approaches.
Collapse
Affiliation(s)
- Teresa Mancuso
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (T.M.); (A.B.); (A.S.); (C.M.); (F.M.); (M.S.); (K.U.)
| | - Antonella Barone
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (T.M.); (A.B.); (A.S.); (C.M.); (F.M.); (M.S.); (K.U.)
| | - Alessandro Salatino
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (T.M.); (A.B.); (A.S.); (C.M.); (F.M.); (M.S.); (K.U.)
| | - Claudia Molinaro
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (T.M.); (A.B.); (A.S.); (C.M.); (F.M.); (M.S.); (K.U.)
| | - Fabiola Marino
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (T.M.); (A.B.); (A.S.); (C.M.); (F.M.); (M.S.); (K.U.)
| | - Mariangela Scalise
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (T.M.); (A.B.); (A.S.); (C.M.); (F.M.); (M.S.); (K.U.)
| | - Michele Torella
- Department of Translational Medical Sciences, AORN dei Colli/Monaldi Hospital, University of Campania “L. Vanvitelli”, Via Leonardo Bianchi, 80131 Naples, Italy;
| | - Antonella De Angelis
- Department of Experimental Medicine, Section of Pharmacology, University of Campania “L.Vanvitelli”, 80121 Naples, Italy;
| | - Konrad Urbanek
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (T.M.); (A.B.); (A.S.); (C.M.); (F.M.); (M.S.); (K.U.)
| | - Daniele Torella
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (T.M.); (A.B.); (A.S.); (C.M.); (F.M.); (M.S.); (K.U.)
| | - Eleonora Cianflone
- Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy;
| |
Collapse
|
24
|
Abstract
Ischemic heart disease (IHD) is one of the most common cardiovascular diseases and is the leading cause of death worldwide. Stem cell therapy is a promising strategy to promote cardiac regeneration and myocardial function recovery. Recently, the generation of human induced pluripotent cells (hiPSCs) and their differentiation into cardiomyocytes and vascular cells offer an unprecedented opportunity for the IHD treatment. This review briefly summarizes hiPSCs and their differentiation, and presents the recent advances in hiPSC injection, engineered cardiac patch fabrication, and the application of hiPSC derived extracellular vesicle. Current challenges and further perspectives are also discussed to understand current risks and concerns, identify potential solutions, and direct future clinical trials and applications.
Collapse
|
25
|
Báez-Díaz C, Blanco-Blázquez V, Sánchez-Margallo FM, Bayes-Genis A, González I, Abad A, Steendam R, Franssen O, Palacios I, Sánchez B, Gálvez-Montón C, Crisóstomo V. Microencapsulated Insulin-Like Growth Factor-1 therapy improves cardiac function and reduces fibrosis in a porcine acute myocardial infarction model. Sci Rep 2020; 10:7166. [PMID: 32346015 PMCID: PMC7188803 DOI: 10.1038/s41598-020-64097-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 04/03/2020] [Indexed: 12/12/2022] Open
Abstract
Insulin-like growth factor-1 (IGF-1) has demonstrated beneficial effects after myocardial infarction (MI). Microencapsulation of IGF-1 could potentially improve results. We aimed to test the effect of an intracoronary (IC) infusion of microencapsulated IGF-1 in a swine acute MI model. For that purpose IC injection of a 10 ml solution of 5 × 106 IGF-1 loaded microspheres (MSPs) (n = 8, IGF-1 MSPs), 5 × 106 unloaded MSPs (n = 9; MSPs) or saline (n = 7; CON) was performed 48 hours post-MI. Left ventricular ejection fraction (LVEF), indexed ventricular volumes and infarct size (IS) were determined by cardiac magnetic resonance at pre-injection and 10 weeks. Animals were euthanized at 10 weeks, and myocardial fibrosis and vascular density were analysed. End-study LVEF was significantly greater in IGF-1 MSPs compared to MSPs and CON, while ventricular volumes exhibited no significant differences between groups. IS decreased over time in all groups. Collagen volume fraction on the infarct area was significantly reduced in IGF-1 MSPs compared to CON and MSPs. Vascular density analysis of infarct and border zones showed no significant differences between groups. In conclusion, the IC injection of 5 × 106 IGF-1 loaded MSPs in a porcine acute MI model successfully improves cardiac function and limits myocardial fibrosis, which could be clinically relevant.
Collapse
Affiliation(s)
- Claudia Báez-Díaz
- Jesús Usón Minimally Invasive Surgery Centre, Cáceres, Spain.
- CIBERCV, Madrid, Spain.
| | | | | | - Antoni Bayes-Genis
- CIBERCV, Madrid, Spain
- ICREC (Heart Failure and Cardiac Regeneration) Research Programme, Health Sciences Research Institute Germans Trias i Pujol (IGTP), Badalona, Barcelona, Spain
| | - Irene González
- Jesús Usón Minimally Invasive Surgery Centre, Cáceres, Spain
| | - Ana Abad
- Jesús Usón Minimally Invasive Surgery Centre, Cáceres, Spain
| | - Rob Steendam
- Innocore Pharmaceuticals, Groningen, The Netherlands
| | | | | | | | - Carolina Gálvez-Montón
- CIBERCV, Madrid, Spain
- ICREC (Heart Failure and Cardiac Regeneration) Research Programme, Health Sciences Research Institute Germans Trias i Pujol (IGTP), Badalona, Barcelona, Spain
| | - Verónica Crisóstomo
- Jesús Usón Minimally Invasive Surgery Centre, Cáceres, Spain
- CIBERCV, Madrid, Spain
| |
Collapse
|