1
|
Bayes-Genis A, Gastelurrutia P, Monguió-Tortajada M, Cámara ML, Prat-Vidal C, Cediel G, Rodríguez-Gómez L, Teis A, Revuelta-López E, Ferrer-Curriu G, Roura S, Gálvez-Montón C, Bisbal F, Vives J, Vilarrodona A, Muñoz-Guijosa C, Querol S. Implantation of a double allogeneic human engineered tissue graft on damaged heart: insights from the PERISCOPE phase I clinical trial. EBioMedicine 2024; 102:105060. [PMID: 38490102 PMCID: PMC10955661 DOI: 10.1016/j.ebiom.2024.105060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 02/29/2024] [Accepted: 03/01/2024] [Indexed: 03/17/2024] Open
Abstract
BACKGROUND In preclinical studies, the use of double allogeneic grafts has shown promising results in promoting tissue revascularization, reducing infarct size, preventing adverse remodelling and fibrosis, and ultimately enhancing cardiac function. Building upon these findings, the safety of PeriCord, an engineered tissue graft consisting of a decellularised pericardial matrix and umbilical cord Wharton's jelly mesenchymal stromal cells, was evaluated in the PERISCOPE Phase I clinical trial (NCT03798353), marking its first application in human subjects. METHODS This was a double-blind, single-centre trial that enrolled patients with non-acute myocardial infarction eligible for surgical revascularization. Seven patients were implanted with PeriCord while five served as controls. FINDINGS Patients who received PeriCord showed no adverse effects during post-operative phase and one-year follow-up. No significant changes in secondary outcomes, such as quality of life or cardiac function, were found in patients who received PeriCord. However, PeriCord did modulate the kinetics of circulating monocytes involved in post-infarction myocardial repair towards non-classical inflammation-resolving macrophages, as well as levels of monocyte chemoattractants and the prognostic marker Meteorin-like in plasma following treatment. INTERPRETATION In summary, the PeriCord graft has exhibited a safe profile and notable immunomodulatory properties. Nevertheless, further research is required to fully unlock its potential as a platform for managing inflammatory-related pathologies. FUNDING This work was supported in part by grants from MICINN (SAF2017-84324-C2-1-R); Instituto de Salud Carlos III (ICI19/00039 and Red RICORS-TERAV RD21/0017/0022, and CIBER Cardiovascular CB16/11/00403) as a part of the Plan Nacional de I + D + I, and co-funded by ISCIII-Subdirección General de Evaluación y el Fondo Europeo de Desarrollo Regional (FEDER) and AGAUR (2021-SGR-01437).
Collapse
Affiliation(s)
- Antoni Bayes-Genis
- Heart Institute and Heart Failure Unit (iCor), Germans Trias i Pujol University Hospital (HUGTiP), Badalona, Spain; CIBER Cardiovascular, Instituto de Salud Carlos III, Madrid, Spain; Department of Medicine, Autonomous University of Barcelona, Barcelona, Spain; ICREC Research Program, Germans Trias i Pujol Health Science Research Institute (IGTP), Spain.
| | - Paloma Gastelurrutia
- Heart Institute and Heart Failure Unit (iCor), Germans Trias i Pujol University Hospital (HUGTiP), Badalona, Spain; CIBER Cardiovascular, Instituto de Salud Carlos III, Madrid, Spain; ICREC Research Program, Germans Trias i Pujol Health Science Research Institute (IGTP), Spain
| | - Marta Monguió-Tortajada
- Heart Institute and Heart Failure Unit (iCor), Germans Trias i Pujol University Hospital (HUGTiP), Badalona, Spain; ICREC Research Program, Germans Trias i Pujol Health Science Research Institute (IGTP), Spain
| | - Maria Luisa Cámara
- Heart Institute and Heart Failure Unit (iCor), Germans Trias i Pujol University Hospital (HUGTiP), Badalona, Spain
| | | | - German Cediel
- Heart Institute and Heart Failure Unit (iCor), Germans Trias i Pujol University Hospital (HUGTiP), Badalona, Spain; CIBER Cardiovascular, Instituto de Salud Carlos III, Madrid, Spain
| | | | - Albert Teis
- Heart Institute and Heart Failure Unit (iCor), Germans Trias i Pujol University Hospital (HUGTiP), Badalona, Spain
| | - Elena Revuelta-López
- Heart Institute and Heart Failure Unit (iCor), Germans Trias i Pujol University Hospital (HUGTiP), Badalona, Spain; CIBER Cardiovascular, Instituto de Salud Carlos III, Madrid, Spain; ICREC Research Program, Germans Trias i Pujol Health Science Research Institute (IGTP), Spain
| | | | - Santiago Roura
- Heart Institute and Heart Failure Unit (iCor), Germans Trias i Pujol University Hospital (HUGTiP), Badalona, Spain; CIBER Cardiovascular, Instituto de Salud Carlos III, Madrid, Spain; ICREC Research Program, Germans Trias i Pujol Health Science Research Institute (IGTP), Spain; Faculty of Medicine, University of Vic-Central University of Catalonia (UVic-UCC), Victoria, Spain
| | - Carolina Gálvez-Montón
- Heart Institute and Heart Failure Unit (iCor), Germans Trias i Pujol University Hospital (HUGTiP), Badalona, Spain; CIBER Cardiovascular, Instituto de Salud Carlos III, Madrid, Spain; ICREC Research Program, Germans Trias i Pujol Health Science Research Institute (IGTP), Spain
| | - Felipe Bisbal
- Heart Institute and Heart Failure Unit (iCor), Germans Trias i Pujol University Hospital (HUGTiP), Badalona, Spain; CIBER Cardiovascular, Instituto de Salud Carlos III, Madrid, Spain
| | - Joaquim Vives
- Department of Medicine, Autonomous University of Barcelona, Barcelona, Spain; Cell Therapy Service, Banc de Sang i Teixits (BST), Barcelona, Spain; Vall d'Hebron Institute of Research (VHIR), Autonomous University of Barcelona, Barcelona, Spain
| | - Anna Vilarrodona
- Barcelona Tissue Bank (BTB), Banc de Sang i Teixits (BST), Barcelona, Spain
| | - Christian Muñoz-Guijosa
- Heart Institute and Heart Failure Unit (iCor), Germans Trias i Pujol University Hospital (HUGTiP), Badalona, Spain; CIBER Cardiovascular, Instituto de Salud Carlos III, Madrid, Spain
| | - Sergi Querol
- Cell Therapy Service, Banc de Sang i Teixits (BST), Barcelona, Spain.
| |
Collapse
|
2
|
Zhang J, Tu Y, Wei J, Zheng R, Shao J, Chen Q, Liang G, Ying H, Han X, Shi Q. Dectin1 contributes to hypertensive vascular injury by promoting macrophage infiltration through activating the Syk/NF-κB pathway. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166911. [PMID: 37813169 DOI: 10.1016/j.bbadis.2023.166911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 09/13/2023] [Accepted: 10/02/2023] [Indexed: 10/11/2023]
Abstract
Vascular injury is an early manifestation leading to end-organ damage in hypertension pathogenesis, which involves a macrophage-associated immune response. Dendritic cell-associated C-type lectin-1 (Dectin1) is a pivotal player in regulating inflammation-mediated cardiovascular disease. However, its role in hypertension-induced vascular damage and the underlying mechanisms remain unclear. We hypothesized that Dectin1 might accelerate angiotensin II (Ang II)- or deoxycorticosterone acetate-salt (DOCA-salt)-induced vascular injury through proinflammatory actions in macrophages. Macrophage Dectin1 was upregulated in mouse aortic tissues stimulated with Ang II. In the peripheral blood, Ang II also increased CD11b+F4/80+ macrophages in mice. In our constructed Dectin1 knockout mice, Dectin1 deletion protected against Ang II-induced EB extravasation and aortic wall thickness. Deficiency of Dectin1 or its pharmacological inhibition considerably improved fibrosis and inflammation responses, accompanied by a reduction in M1 macrophage polarization as well as proinflammatory cytokines and chemokines induced by Ang II or DOCA-salt. Through the bone marrow (BM) transplantation assay, these effects were verified in the wild type mice reconstituted with Dectin1-deficient BM cells. Mechanistically, Ang II promoted Dectin1 homodimerization, thereby triggering the spleen tyrosine kinase/nuclear factor kappa B pro-inflammatory cascade to induce the expression of inflammatory factors and chemokines in vivo and in vitro. In conclusion, Dectin1 has an essential role in the pathogenic procedure of Ang II-stimulated or DOCA-salt-induced vascular damage in mice and represents a promising therapeutic target for cardiovascular diseases.
Collapse
Affiliation(s)
- Jiajia Zhang
- Zhejiang Provincial Key Laboratory of Laboratory Animals and Safety Research, Hangzhou Medical College, Hangzhou 310013, China; Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, Hangzhou Medical College, Hangzhou 310013, China; Engineering Research Center of Novel Vaccine of Zhejiang Province, Hangzhou Medical College, Hangzhou 310013, China
| | - Yu Tu
- Zhejiang Provincial Key Laboratory of Laboratory Animals and Safety Research, Hangzhou Medical College, Hangzhou 310013, China; Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, Hangzhou Medical College, Hangzhou 310013, China; Engineering Research Center of Novel Vaccine of Zhejiang Province, Hangzhou Medical College, Hangzhou 310013, China
| | - Jiajia Wei
- Zhejiang Provincial Key Laboratory of Laboratory Animals and Safety Research, Hangzhou Medical College, Hangzhou 310013, China
| | - Ruyi Zheng
- Zhejiang Provincial Key Laboratory of Laboratory Animals and Safety Research, Hangzhou Medical College, Hangzhou 310013, China
| | - Ji Shao
- Zhejiang Provincial Key Laboratory of Laboratory Animals and Safety Research, Hangzhou Medical College, Hangzhou 310013, China
| | - Qinhua Chen
- Key Laboratory of TCM Clinical Pharmacy, Shenzhen Baoan Authentic TCM Therapy Hospital, Guangzhou University of Chinese Medicine, Shenzhen 518101, China
| | - Guang Liang
- Zhejiang Provincial Key Laboratory of Laboratory Animals and Safety Research, Hangzhou Medical College, Hangzhou 310013, China; Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, Hangzhou Medical College, Hangzhou 310013, China
| | - Huazhong Ying
- Zhejiang Provincial Key Laboratory of Laboratory Animals and Safety Research, Hangzhou Medical College, Hangzhou 310013, China; Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, Hangzhou Medical College, Hangzhou 310013, China; Engineering Research Center of Novel Vaccine of Zhejiang Province, Hangzhou Medical College, Hangzhou 310013, China.
| | - Xue Han
- Zhejiang Provincial Key Laboratory of Laboratory Animals and Safety Research, Hangzhou Medical College, Hangzhou 310013, China; Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, Hangzhou Medical College, Hangzhou 310013, China.
| | - Qiaojuan Shi
- Zhejiang Provincial Key Laboratory of Laboratory Animals and Safety Research, Hangzhou Medical College, Hangzhou 310013, China; Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, Hangzhou Medical College, Hangzhou 310013, China; Engineering Research Center of Novel Vaccine of Zhejiang Province, Hangzhou Medical College, Hangzhou 310013, China.
| |
Collapse
|
3
|
Monguió-Tortajada M, Prat-Vidal C, Martínez-Falguera D, Teis A, Soler-Botija C, Courageux Y, Munizaga-Larroudé M, Moron-Font M, Bayes-Genis A, Borràs FE, Roura S, Gálvez-Montón C. Acellular cardiac scaffolds enriched with MSC-derived extracellular vesicles limit ventricular remodelling and exert local and systemic immunomodulation in a myocardial infarction porcine model. Theranostics 2022; 12:4656-4670. [PMID: 35832072 PMCID: PMC9254233 DOI: 10.7150/thno.72289] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 05/13/2022] [Indexed: 11/05/2022] Open
Abstract
Rationale: Extracellular vesicles (EVs) from mesenchymal stromal cell (MSC) are a potential therapy for cardiac healing after myocardial infarction (MI). Nevertheless, neither their efficient administration nor therapeutic mechanisms are fully elucidated. Here, we evaluate the preclinical efficacy of a tissue engineering approach to locally deliver porcine cardiac adipose tissue MSC-EV (cATMSC-EV) in an acute MI pig model. Methods: After MI by permanent ligation of the coronary artery, pigs (n = 24) were randomized to Untreated or treated groups with a decellularised pericardial scaffold filled with peptide hydrogel and cATMSC-EV purified by size exclusion chromatography (EV-Treated group) or buffer (Control group), placed over the post-infarcted myocardium. Results: After 30 days, cardiac MRI showed an improved cardiac function in EV-Treated animals, with significantly higher right ventricle ejection fraction (+20.8% in EV-Treated; p = 0.026), and less ventricle dilatation, indicating less myocardial remodelling. Scar size was reduced, with less fibrosis in the distal myocardium (-42.6% Col I in EV-Treated vs Untreated; p = 0.03), a 2-fold increase in vascular density (EV-Treated; p = 0.019) and less CCL2 transcription in the infarct core. EV-treated animals had less macrophage infiltration in the infarct core (-31.7% of CD163+ cells/field in EV-Treated; p = 0.026), but 5.8 times more expressing anti-inflammatory CD73 (p = 0.015). Systemically, locally delivered cATMSC-EV also triggered a systemic effect, doubling the circulating IL-1ra (p = 0.01), and reducing the PBMC rush 2d post-MI, the TNFα and GM-CSF levels at 30d post-MI, and modulating the CD73+ and CCR2+ monocyte populations, related to immunomodulation and fibrosis modulation. Conclusions: These results highlight the potential of cATMSC-EV in modulating hallmarks of ischemic injury for cardiac repair after MI.
Collapse
Affiliation(s)
- Marta Monguió-Tortajada
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain
- Heart Institute (iCor), Cardiology Department, Germans Trias i Pujol University Hospital, Badalona, Spain
| | - Cristina Prat-Vidal
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain
- Cell Therapy Service, Banc de Sang i Teixits (BST), Barcelona, Spain
| | - Daina Martínez-Falguera
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain
- Faculty of Medicine, Universitat de Barcelona (UB), Barcelona, Spain
| | - Albert Teis
- Heart Institute (iCor), Cardiology Department, Germans Trias i Pujol University Hospital, Badalona, Spain
| | - Carolina Soler-Botija
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain
- CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Yvan Courageux
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain
- Department of Biochemistry, Molecular Biology and Biomedicine, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Micaela Munizaga-Larroudé
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain
| | - Miriam Moron-Font
- REMAR-IVECAT Group, Health Science Research Institute Germans Trias i Pujol and Nephrology Service, Germans Trias i Pujol University Hospital, Can Ruti Campus, Badalona, Spain
| | - Antoni Bayes-Genis
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain
- Heart Institute (iCor), Cardiology Department, Germans Trias i Pujol University Hospital, Badalona, Spain
- CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
- Department of Medicine, UAB, Barcelona, Spain
| | - Francesc E. Borràs
- REMAR-IVECAT Group, Health Science Research Institute Germans Trias i Pujol and Nephrology Service, Germans Trias i Pujol University Hospital, Can Ruti Campus, Badalona, Spain
- Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Spain
| | - Santiago Roura
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain
- Heart Institute (iCor), Cardiology Department, Germans Trias i Pujol University Hospital, Badalona, Spain
- CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
- Faculty of Medicine, University of Vic-Central University of Catalonia (UVic-UCC), Vic, Barcelona 08500, Spain
- Institut d'Investigació Biomèdica de Bellvitge-IDIBELL, L´Hospitalet de Llobregat, Spain
| | - Carolina Gálvez-Montón
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain
- Heart Institute (iCor), Cardiology Department, Germans Trias i Pujol University Hospital, Badalona, Spain
- CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
- Institut d'Investigació Biomèdica de Bellvitge-IDIBELL, L´Hospitalet de Llobregat, Spain
| |
Collapse
|
4
|
Miloradovic D, Miloradovic D, Ljujic B, Jankovic MG. Optimal Delivery Route of Mesenchymal Stem Cells for Cardiac Repair: The Path to Good Clinical Practice. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022:83-100. [PMID: 35389200 DOI: 10.1007/5584_2022_709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Research has shown that mesenchymal stem cells (MSCs) could be a promising therapy for treating progressive heart disease. However, translation into clinics efficiently and successfully has proven to be much more complicated. Many questions remain for optimizing treatment. Application method influences destiny of MSCs and afterwards impacts results of procedure, yet there is no general agreement about most suitable method of MSC delivery in the clinical setting. Herein, we explain principle of most-frequent MSCs delivery techniques in cardiology. This chapter summarizes crucial translational obstacles of clinical employment of MSCs for cardiac repair when analysed trough a prism of latest research centred on different techniques of MSCs application.
Collapse
Affiliation(s)
- Dragica Miloradovic
- Faculty of Medical Sciences, Department of Genetics, University of Kragujevac, Kragujevac, Serbia
| | - Dragana Miloradovic
- Faculty of Medical Sciences, Department of Genetics, University of Kragujevac, Kragujevac, Serbia
| | - Biljana Ljujic
- Faculty of Medical Sciences, Department of Genetics, University of Kragujevac, Kragujevac, Serbia
| | - Marina Gazdic Jankovic
- Faculty of Medical Sciences, Department of Genetics, University of Kragujevac, Kragujevac, Serbia.
| |
Collapse
|
5
|
Tajabadi M, Goran Orimi H, Ramzgouyan MR, Nemati A, Deravi N, Beheshtizadeh N, Azami M. Regenerative strategies for the consequences of myocardial infarction: Chronological indication and upcoming visions. Biomed Pharmacother 2021; 146:112584. [PMID: 34968921 DOI: 10.1016/j.biopha.2021.112584] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 12/13/2022] Open
Abstract
Heart muscle injury and an elevated troponin level signify myocardial infarction (MI), which may result in defective and uncoordinated segments, reduced cardiac output, and ultimately, death. Physicians apply thrombolytic therapy, coronary artery bypass graft (CABG) surgery, or percutaneous coronary intervention (PCI) to recanalize and restore blood flow to the coronary arteries, albeit they were not convincingly able to solve the heart problems. Thus, researchers aim to introduce novel substitutional therapies for regenerating and functionalizing damaged cardiac tissue based on engineering concepts. Cell-based engineering approaches, utilizing biomaterials, gene, drug, growth factor delivery systems, and tissue engineering are the most leading studies in the field of heart regeneration. Also, understanding the primary cause of MI and thus selecting the most efficient treatment method can be enhanced by preparing microdevices so-called heart-on-a-chip. In this regard, microfluidic approaches can be used as diagnostic platforms or drug screening in cardiac disease treatment. Additionally, bioprinting technique with whole organ 3D printing of human heart with major vessels, cardiomyocytes and endothelial cells can be an ideal goal for cardiac tissue engineering and remarkable achievement in near future. Consequently, this review discusses the different aspects, advancements, and challenges of the mentioned methods with presenting the advantages and disadvantages, chronological indications, and application prospects of various novel therapeutic approaches.
Collapse
Affiliation(s)
- Maryam Tajabadi
- School of Metallurgy and Materials Engineering, Iran University of Science and Technology (IUST), Narmak, Tehran 16844, Iran
| | - Hanif Goran Orimi
- School of Metallurgy and Materials Engineering, Iran University of Science and Technology (IUST), Narmak, Tehran 16844, Iran; Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Maryam Roya Ramzgouyan
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Iran; Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Alireza Nemati
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran; Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Niloofar Deravi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Nima Beheshtizadeh
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Iran; Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mahmoud Azami
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Iran; Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| |
Collapse
|
6
|
Wang L, Liu Y, Ye G, He Y, Li B, Guan Y, Gong B, Mequanint K, Xing MMQ, Qiu X. Injectable and conductive cardiac patches repair infarcted myocardium in rats and minipigs. Nat Biomed Eng 2021; 5:1157-1173. [PMID: 34593988 DOI: 10.1038/s41551-021-00796-9] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 08/17/2021] [Indexed: 02/07/2023]
Abstract
Cardiac patches can help to restore the electrophysiological properties of the heart after myocardial infarction. However, scaffolds for the repair of heart muscle typically require surgical implantation or, if they are injectable, they are not electrically conductive or do not maintain their shape or function. Here, we report the performance, as demonstrated for the repair of infarcted heart muscle in rats and minipigs, of injectable and conductive scaffolds consisting of methacrylated elastin and gelatin, and carbon nanotubes that display shape-memory behaviour, a hierarchical porous structure and a negligible Poisson's ratio. In rats, the implantation of cell-free patches or patches seeded with rat cardiomyocytes onto the myocardium after ligation of the left anterior descending coronary artery led to functional repair after 4 weeks, as indicated by increases in fractional shortening and the ejection fraction, and by a decrease in the infarcted area. We also observed measures of functional recovery in minipigs with infarcted hearts after the delivery of cell-free patches or patches incorporating cardiomyocytes differentiated from human pluripotent stem cells.
Collapse
Affiliation(s)
- Leyu Wang
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Science; Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, China.,Department of Mechanical Engineering, University of Manitoba and Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - Yuqing Liu
- Department of Mechanical Engineering, University of Manitoba and Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - Genlan Ye
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Science; Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, China
| | - Yutong He
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Science; Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, China
| | - Bingyun Li
- Department of Orthopaedics, School of Medicine, West Virginia University, Morgantown, WV, USA
| | - Yezhi Guan
- Guangdong Laboratory Animals Monitoring Institute, Guangdong Provincial Key Laboratory of Laboratory Animals, Guangzhou, China
| | - Baoyong Gong
- Guangdong Laboratory Animals Monitoring Institute, Guangdong Provincial Key Laboratory of Laboratory Animals, Guangzhou, China
| | - Kibret Mequanint
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, Ontario, Canada.,School of Biomedical Engineering, The University of Western Ontario, London, Ontario, Canada
| | - Malcolm M Q Xing
- Department of Mechanical Engineering, University of Manitoba and Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada.
| | - Xiaozhong Qiu
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Science; Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, China.
| |
Collapse
|
7
|
Wharton's Jelly Mesenchymal Stromal Cells and Derived Extracellular Vesicles as Post-Myocardial Infarction Therapeutic Toolkit: An Experienced View. Pharmaceutics 2021; 13:pharmaceutics13091336. [PMID: 34575412 PMCID: PMC8471243 DOI: 10.3390/pharmaceutics13091336] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/18/2021] [Accepted: 08/23/2021] [Indexed: 12/18/2022] Open
Abstract
Outstanding progress has been achieved in developing therapeutic options for reasonably alleviating symptoms and prolonging the lifespan of patients suffering from myocardial infarction (MI). Current treatments, however, only partially address the functional recovery of post-infarcted myocardium, which is in fact the major goal for effective primary care. In this context, we largely investigated novel cell and TE tissue engineering therapeutic approaches for cardiac repair, particularly using multipotent mesenchymal stromal cells (MSC) and natural extracellular matrices, from pre-clinical studies to clinical application. A further step in this field is offered by MSC-derived extracellular vesicles (EV), which are naturally released nanosized lipid bilayer-delimited particles with a key role in cell-to-cell communication. Herein, in this review, we further describe and discuss the rationale, outcomes and challenges of our evidence-based therapy approaches using Wharton's jelly MSC and derived EV in post-MI management.
Collapse
|
8
|
Hemalatha T, Aarthy M, Pandurangan S, Kamini NR, Ayyadurai N. A deep dive into the darning effects of biomaterials in infarct myocardium: current advances and future perspectives. Heart Fail Rev 2021; 27:1443-1467. [PMID: 34342769 DOI: 10.1007/s10741-021-10144-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/07/2021] [Indexed: 12/21/2022]
Abstract
Myocardial infarction (MI) occurs due to the obstruction of coronary arteries, a major crux that restricts blood flow and thereby oxygen to the distal part of the myocardium, leading to loss of cardiomyocytes and eventually, if left untreated, leads to heart failure. MI, a potent cardiovascular disorder, requires intense therapeutic interventions and thereby presents towering challenges. Despite the concerted efforts, the treatment strategies for MI are still demanding, which has paved the way for the genesis of biomaterial applications. Biomaterials exhibit immense potentials for cardiac repair and regeneration, wherein they act as extracellular matrix replacing scaffolds or as delivery vehicles for stem cells, protein, plasmids, etc. This review concentrates on natural, synthetic, and hybrid biomaterials; their function; and interaction with the body, mechanisms of repair by which they are able to improve cardiac function in a MI milieu. We also provide focus on future perspectives that need attention. The cognizance provided by the research results certainly indicates that biomaterials could revolutionize the treatment paradigms for MI with a positive impact on clinical translation.
Collapse
Affiliation(s)
- Thiagarajan Hemalatha
- Department of Biochemistry and Biotechnology, CSIR- Central Leather Research Institute, Chennai, 600020, India
| | - Mayilvahanan Aarthy
- Department of Biochemistry and Biotechnology, CSIR- Central Leather Research Institute, Chennai, 600020, India
| | - Suryalakshmi Pandurangan
- Department of Biochemistry and Biotechnology, CSIR- Central Leather Research Institute, Chennai, 600020, India
| | - Numbi Ramudu Kamini
- Department of Biochemistry and Biotechnology, CSIR- Central Leather Research Institute, Chennai, 600020, India
| | - Niraikulam Ayyadurai
- Department of Biochemistry and Biotechnology, CSIR- Central Leather Research Institute, Chennai, 600020, India.
| |
Collapse
|
9
|
Litowczenko J, Woźniak-Budych MJ, Staszak K, Wieszczycka K, Jurga S, Tylkowski B. Milestones and current achievements in development of multifunctional bioscaffolds for medical application. Bioact Mater 2021; 6:2412-2438. [PMID: 33553825 PMCID: PMC7847813 DOI: 10.1016/j.bioactmat.2021.01.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/23/2020] [Accepted: 01/07/2021] [Indexed: 12/13/2022] Open
Abstract
Tissue engineering (TE) is a rapidly growing interdisciplinary field, which aims to restore or improve lost tissue function. Despite that TE was introduced more than 20 years ago, innovative and more sophisticated trends and technologies point to new challenges and development. Current challenges involve the demand for multifunctional bioscaffolds which can stimulate tissue regrowth by biochemical curves, biomimetic patterns, active agents and proper cell types. For those purposes especially promising are carefully chosen primary cells or stem cells due to its high proliferative and differentiation potential. This review summarized a variety of recently reported advanced bioscaffolds which present new functions by combining polymers, nanomaterials, bioactive agents and cells depending on its desired application. In particular necessity of study biomaterial-cell interactions with in vitro cell culture models, and studies using animals with in vivo systems were discuss to permit the analysis of full material biocompatibility. Although these bioscaffolds have shown a significant therapeutic effect in nervous, cardiovascular and muscle, tissue engineering, there are still many remaining unsolved challenges for scaffolds improvement.
Collapse
Affiliation(s)
- Jagoda Litowczenko
- NanoBioMedical Centre, Adam Mickiewicz University in Poznan, Wszechnicy Piastowskiej 3, Poznan, Poland
| | - Marta J. Woźniak-Budych
- NanoBioMedical Centre, Adam Mickiewicz University in Poznan, Wszechnicy Piastowskiej 3, Poznan, Poland
| | - Katarzyna Staszak
- Institute of Technology and Chemical Engineering, Poznan University of Technology, ul. Berdychowo 4, Poznan, Poland
| | - Karolina Wieszczycka
- Institute of Technology and Chemical Engineering, Poznan University of Technology, ul. Berdychowo 4, Poznan, Poland
| | - Stefan Jurga
- NanoBioMedical Centre, Adam Mickiewicz University in Poznan, Wszechnicy Piastowskiej 3, Poznan, Poland
| | - Bartosz Tylkowski
- Eurecat, Centre Tecnològic de Catalunya, Chemical Technologies Unit, Marcel·lí Domingo s/n, Tarragona, 43007, Spain
| |
Collapse
|
10
|
Tenreiro MF, Louro AF, Alves PM, Serra M. Next generation of heart regenerative therapies: progress and promise of cardiac tissue engineering. NPJ Regen Med 2021; 6:30. [PMID: 34075050 PMCID: PMC8169890 DOI: 10.1038/s41536-021-00140-4] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 05/10/2021] [Indexed: 02/04/2023] Open
Abstract
The adult heart is a vital and highly specialized organ of the human body, with limited capability of self-repair and regeneration in case of injury or disease. Engineering biomimetic cardiac tissue to regenerate the heart has been an ambition in the field of tissue engineering, tracing back to the 1990s. Increased understanding of human stem cell biology and advances in process engineering have provided an unlimited source of cells, particularly cardiomyocytes, for the development of functional cardiac muscle, even though pluripotent stem cell-derived cardiomyocytes poorly resemble those of the adult heart. This review outlines key biology-inspired strategies reported to improve cardiomyocyte maturation features and current biofabrication approaches developed to engineer clinically relevant cardiac tissues. It also highlights the potential use of this technology in drug discovery science and disease modeling as well as the current efforts to translate it into effective therapies that improve heart function and promote regeneration.
Collapse
Affiliation(s)
- Miguel F Tenreiro
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal
- Instituto de Tecnologia Química e Biologica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Ana F Louro
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal
- Instituto de Tecnologia Química e Biologica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Paula M Alves
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal
- Instituto de Tecnologia Química e Biologica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Margarida Serra
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal.
- Instituto de Tecnologia Química e Biologica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal.
| |
Collapse
|
11
|
Monguió-Tortajada M, Prat-Vidal C, Moron-Font M, Clos-Sansalvador M, Calle A, Gastelurrutia P, Cserkoova A, Morancho A, Ramírez MÁ, Rosell A, Bayes-Genis A, Gálvez-Montón C, Borràs FE, Roura S. Local administration of porcine immunomodulatory, chemotactic and angiogenic extracellular vesicles using engineered cardiac scaffolds for myocardial infarction. Bioact Mater 2021; 6:3314-3327. [PMID: 33778207 PMCID: PMC7973387 DOI: 10.1016/j.bioactmat.2021.02.026] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 02/16/2021] [Accepted: 02/17/2021] [Indexed: 02/06/2023] Open
Abstract
The administration of extracellular vesicles (EV) from mesenchymal stromal cells (MSC) is a promising cell-free nanotherapy for tissue repair after myocardial infarction (MI). However, the optimal EV delivery strategy remains undetermined. Here, we designed a novel MSC-EV delivery, using 3D scaffolds engineered from decellularised cardiac tissue as a cell-free product for cardiac repair. EV from porcine cardiac adipose tissue-derived MSC (cATMSC) were purified by size exclusion chromatography (SEC), functionally analysed and loaded to scaffolds. cATMSC-EV markedly reduced polyclonal proliferation and pro-inflammatory cytokines production (IFNγ, TNFα, IL12p40) of allogeneic PBMC. Moreover, cATMSC-EV recruited outgrowth endothelial cells (OEC) and allogeneic MSC, and promoted angiogenesis. Fluorescently labelled cATMSC-EV were mixed with peptide hydrogel, and were successfully retained in decellularised scaffolds. Then, cATMSC-EV-embedded pericardial scaffolds were administered in vivo over the ischemic myocardium in a pig model of MI. Six days from implantation, the engineered scaffold efficiently integrated into the post-infarcted myocardium. cATMSC-EV were detected within the construct and MI core, and promoted an increase in vascular density and reduction in macrophage and T cell infiltration within the damaged myocardium. The confined administration of multifunctional MSC-EV within an engineered pericardial scaffold ensures local EV dosage and release, and generates a vascularised bioactive niche for cell recruitment, engraftment and modulation of short-term post-ischemic inflammation.
Collapse
Affiliation(s)
- Marta Monguió-Tortajada
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain.,REMAR-IVECAT Group, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain.,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Cristina Prat-Vidal
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain.,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain.,Institut d'Investigació Biomèdica de Bellvitge-IDIBELL, L'Hospitalet de Llobregat, Spain
| | - Miriam Moron-Font
- REMAR-IVECAT Group, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain
| | - Marta Clos-Sansalvador
- REMAR-IVECAT Group, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Alexandra Calle
- Departamento de Reproducción Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Madrid, Spain
| | - Paloma Gastelurrutia
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain.,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain.,Institut d'Investigació Biomèdica de Bellvitge-IDIBELL, L'Hospitalet de Llobregat, Spain
| | - Adriana Cserkoova
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain
| | - Anna Morancho
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute (VHIR), UAB, Barcelona, Spain
| | - Miguel Ángel Ramírez
- Departamento de Reproducción Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Madrid, Spain
| | - Anna Rosell
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute (VHIR), UAB, Barcelona, Spain
| | - Antoni Bayes-Genis
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain.,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain.,Cardiology Service, Germans Trias i Pujol University Hospital, Badalona, Spain.,Department of Medicine, UAB, Barcelona, Spain
| | - Carolina Gálvez-Montón
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain.,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Francesc E Borràs
- REMAR-IVECAT Group, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain.,Nephrology Service, Germans Trias i Pujol University Hospital, Badalona, Spain
| | - Santiago Roura
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain.,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain.,Faculty of Medicine, University of Vic-Central University of Catalonia (UVic-UCC), Vic, Barcelona, 08500, Spain
| |
Collapse
|
12
|
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
|
13
|
Mazzola M, Di Pasquale E. Toward Cardiac Regeneration: Combination of Pluripotent Stem Cell-Based Therapies and Bioengineering Strategies. Front Bioeng Biotechnol 2020; 8:455. [PMID: 32528940 PMCID: PMC7266938 DOI: 10.3389/fbioe.2020.00455] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 04/21/2020] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases represent the major cause of morbidity and mortality worldwide. Multiple studies have been conducted so far in order to develop treatments able to prevent the progression of these pathologies. Despite progress made in the last decade, current therapies are still hampered by poor translation into actual clinical applications. The major drawback of such strategies is represented by the limited regenerative capacity of the cardiac tissue. Indeed, after an ischaemic insult, the formation of fibrotic scar takes place, interfering with mechanical and electrical functions of the heart. Hence, the ability of the heart to recover after ischaemic injury depends on several molecular and cellular pathways, and the imbalance between them results into adverse remodeling, culminating in heart failure. In this complex scenario, a new chapter of regenerative medicine has been opened over the past 20 years with the discovery of induced pluripotent stem cells (iPSCs). These cells share the same characteristic of embryonic stem cells (ESCs), but are generated from patient-specific somatic cells, overcoming the ethical limitations related to ESC use and providing an autologous source of human cells. Similarly to ESCs, iPSCs are able to efficiently differentiate into cardiomyocytes (CMs), and thus hold a real regenerative potential for future clinical applications. However, cell-based therapies are subjected to poor grafting and may cause adverse effects in the failing heart. Thus, over the last years, bioengineering technologies focused their attention on the improvement of both survival and functionality of iPSC-derived CMs. The combination of these two fields of study has burst the development of cell-based three-dimensional (3D) structures and organoids which mimic, more realistically, the in vivo cell behavior. Toward the same path, the possibility to directly induce conversion of fibroblasts into CMs has recently emerged as a promising area for in situ cardiac regeneration. In this review we provide an up-to-date overview of the latest advancements in the application of pluripotent stem cells and tissue-engineering for therapeutically relevant cardiac regenerative approaches, aiming to highlight outcomes, limitations and future perspectives for their clinical translation.
Collapse
Affiliation(s)
- Marta Mazzola
- Stem Cell Unit, Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy
| | - Elisa Di Pasquale
- Stem Cell Unit, Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy.,Institute of Genetic and Biomedical Research (IRGB) - UOS of Milan, National Research Council (CNR), Milan, Italy
| |
Collapse
|
14
|
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
|
15
|
Liu Z, Mikrani R, Zubair HM, Taleb A, Naveed M, Baig MMFA, Zhang Q, Li C, Habib M, Cui X, Sembatya KR, Lei H, Zhou X. Systemic and local delivery of mesenchymal stem cells for heart renovation: Challenges and innovations. Eur J Pharmacol 2020; 876:173049. [PMID: 32142771 DOI: 10.1016/j.ejphar.2020.173049] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 02/20/2020] [Accepted: 02/27/2020] [Indexed: 02/07/2023]
Abstract
In the beginning stage of heart disease, the blockage of blood flow frequently occurs due to the persistent damage and even death of myocardium. Cicatricial tissue developed after the death of myocardium can affect heart function, which ultimately leads to heart failure. In recent years, several studies carried out about the use of stem cells such as embryonic, pluripotent, cardiac and bone marrow-derived stem cells as well as myoblasts to repair injured myocardium. Current studies focus more on finding appropriate measures to enhance cell homing and survival in order to increase paracrine function. Until now, there is no universal delivery route for mesenchymal stem cells (MSCs) for different diseases. In this review, we summarize the advantages and challenges of the systemic and local pathways of MSC delivery. In addition, we also describe some advanced measures of cell delivery to improve the efficiency of transplantation. The combination of cells and therapeutic substances could be the most reliable method, which allows donor cells to deliver sufficient amounts of paracrine factors and provide long-lasting effects. The cardiac support devices or tissue engineering techniques have the potential to facilitate the controlled release of stem cells on local tissue for a sustained period. A novel promising epicardial drug delivery system is highlighted here, which not only provides MSCs with a favorable environment to promote retention but also increases the contact area and a number of cells recruited in the heart muscle.
Collapse
Affiliation(s)
- Ziwei Liu
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, 211198, PR China
| | - Reyaj Mikrani
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, 211198, PR China
| | | | - Abdoh Taleb
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, 211166, PR China
| | - Muhammad Naveed
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, 211166, PR China
| | - Mirza Muhammad Faran Asraf Baig
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, Jiangsu, 210023, PR China
| | - Qin Zhang
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, 211198, PR China
| | - Cuican Li
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, 211198, PR China
| | - Murad Habib
- Department of Surgery, Ayub Teaching Hospital, Abbottabad, Pakistan
| | - Xingxing Cui
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, 211198, PR China
| | - Kiganda Raymond Sembatya
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, 211198, PR China
| | - Han Lei
- Department of Pharmacy, Jiangsu Worker Medical University, Nanjing, Jiangsu Province, 211198, PR China
| | - Xiaohui Zhou
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, 211198, PR China; Department of Surgery, Zhongda Hospital Affiliated to Southeast University, Nanjing, Jiangsu Province, 210017, PR China; Department of Surgery, Nanjing Shuiximen Hospital, Nanjing, Jiangsu Province, 210017, PR China.
| |
Collapse
|
16
|
Spotlight on epigenetic reprogramming in cardiac regeneration. Semin Cell Dev Biol 2020; 97:26-37. [PMID: 31002867 DOI: 10.1016/j.semcdb.2019.04.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 04/02/2019] [Accepted: 04/15/2019] [Indexed: 02/06/2023]
|
17
|
KC P, Hong Y, Zhang G. Cardiac tissue-derived extracellular matrix scaffolds for myocardial repair: advantages and challenges. Regen Biomater 2019; 6:185-199. [PMID: 31404421 PMCID: PMC6683951 DOI: 10.1093/rb/rbz017] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Revised: 03/04/2019] [Accepted: 03/14/2019] [Indexed: 12/12/2022] Open
Abstract
Decellularized extracellular matrix (dECM) derived from myocardium has been widely explored as a nature scaffold for cardiac tissue engineering applications. Cardiac dECM offers many unique advantages such as preservation of organ-specific ECM microstructure and composition, demonstration of tissue-mimetic mechanical properties and retention of biochemical cues in favor of subsequent recellularization. However, current processes of dECM decellularization and recellularization still face many challenges including the need for balance between cell removal and extracellular matrix preservation, efficient recellularization of dECM for obtaining homogenous cell distribution, tailoring material properties of dECM for enhancing bioactivity and prevascularization of thick dECM. This review summarizes the recent progresses of using dECM scaffold for cardiac repair and discusses its major advantages and challenges for producing biomimetic cardiac patch.
Collapse
Affiliation(s)
- Pawan KC
- Department of Biomedical Engineering, The University of Akron, Olson Research Center, Room 301L, 260 S Forge Street, Akron, OH, USA
| | - Yi Hong
- Department of Bioengineering, University of Texas at Arlington, 500 UTA Blvd, Room 240, Arlington, TX, USA
| | - Ge Zhang
- Department of Biomedical Engineering, The University of Akron, Olson Research Center, Room 301L, 260 S Forge Street, Akron, OH, USA
| |
Collapse
|
18
|
Roura S, Gálvez-Montón C, Mirabel C, Vives J, Bayes-Genis A. Mesenchymal stem cells for cardiac repair: are the actors ready for the clinical scenario? Stem Cell Res Ther 2017; 8:238. [PMID: 29078809 PMCID: PMC5658929 DOI: 10.1186/s13287-017-0695-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
For years, sufficient progress has been made in treating heart failure following myocardial infarction; however, the social and economic burdens and the costs to world health systems remain high. Moreover, treatment advances have not resolved the underlying problem of functional heart tissue loss. In this field of research, for years we have actively explored innovative biotherapies for cardiac repair. Here, we present a general, critical overview of our experience in using mesenchymal stem cells, derived from cardiac adipose tissue and umbilical cord blood, in a variety of cell therapy and tissue engineering approaches. We also include the latest advances and future challenges, including good manufacturing practice and regulatory issues. Finally, we evaluate whether recent approaches hold potential for reliable translation to clinical trials.
Collapse
Affiliation(s)
- Santiago Roura
- ICREC Research Program, Germans Trias i Pujol Health Research Institute, Badalona, Spain. .,Center of Regenerative Medicine in Barcelona, Barcelona, Spain. .,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain. .,ICREC (Heart Failure and Cardiac Regeneration) Research Programme, Health Sciences Research Institute Germans Trias i Pujol (IGTP), Carretera de Can Ruti, Camí de les Escoles s/n, 08916, Badalona, Barcelona, Spain.
| | - Carolina Gálvez-Montón
- ICREC Research Program, Germans Trias i Pujol Health Research Institute, Badalona, Spain.,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Clémentine Mirabel
- Servei de Teràpia Cel∙lular, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005, Barcelona, Spain.,Musculoskeletal Tissue Engineering Group, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Passeig de la Vall d'Hebron 129-139, 08035, Barcelona, Spain
| | - Joaquim Vives
- Servei de Teràpia Cel∙lular, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005, Barcelona, Spain.,Musculoskeletal Tissue Engineering Group, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Passeig de la Vall d'Hebron 129-139, 08035, Barcelona, Spain.,Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Antoni Bayes-Genis
- ICREC Research Program, Germans Trias i Pujol Health Research Institute, Badalona, Spain. .,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain. .,Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain. .,Cardiology Service, Hospital Universitari Germans Trias i Pujol, Badalona, Barcelona, Spain. .,Heart Institute, Hospital Universitari Germans Trias i Pujol University Hospital, Carretera de Canyet s/n, 08916, Badalona, Barcelona, Spain.
| |
Collapse
|
19
|
Bobi J, Solanes N, Fernández-Jiménez R, Galán-Arriola C, Dantas AP, Fernández-Friera L, Gálvez-Montón C, Rigol-Monzó E, Agüero J, Ramírez J, Roqué M, Bayés-Genís A, Sánchez-González J, García-Álvarez A, Sabaté M, Roura S, Ibáñez B, Rigol M. Intracoronary Administration of Allogeneic Adipose Tissue-Derived Mesenchymal Stem Cells Improves Myocardial Perfusion But Not Left Ventricle Function, in a Translational Model of Acute Myocardial Infarction. J Am Heart Assoc 2017; 6:e005771. [PMID: 28468789 PMCID: PMC5524109 DOI: 10.1161/jaha.117.005771] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 03/30/2017] [Indexed: 11/16/2022]
Abstract
BACKGROUND Autologous adipose tissue-derived mesenchymal stem cells (ATMSCs) therapy is a promising strategy to improve post-myocardial infarction outcomes. In a porcine model of acute myocardial infarction, we studied the long-term effects and the mechanisms involved in allogeneic ATMSCs administration on myocardial performance. METHODS AND RESULTS Thirty-eight pigs underwent 50 minutes of coronary occlusion; the study was completed in 33 pigs. After reperfusion, allogeneic ATMSCs or culture medium (vehicle) were intracoronarily administered. Follow-ups were performed at short (2 days after acute myocardial infarction vehicle-treated, n=10; ATMSCs-treated, n=9) or long term (60 days after acute myocardial infarction vehicle-treated, n=7; ATMSCs-treated, n=7). At short term, infarcted myocardium analysis showed reduced apoptosis in the ATMSCs-treated animals (48.6±6% versus 55.9±5.7% in vehicle; P=0.017); enhancement of the reparative process with up-regulated vascular endothelial growth factor, granulocyte macrophage colony-stimulating factor, and stromal-derived factor-1α gene expression; and increased M2 macrophages (67.2±10% versus 54.7±10.2% in vehicle; P=0.016). In long-term groups, increase in myocardial perfusion at the anterior infarct border was observed both on day-7 and day-60 cardiac magnetic resonance studies in ATMSCs-treated animals, compared to vehicle (87.9±28.7 versus 57.4±17.7 mL/min per gram at 7 days; P=0.034 and 99±22.6 versus 43.3±14.7 22.6 mL/min per gram at 60 days; P=0.0001, respectively). At day 60, higher vascular density was detected at the border zone in the ATMSCs-treated animals (118±18 versus 92.4±24.3 vessels/mm2 in vehicle; P=0.045). Cardiac magnetic resonance-measured left ventricular ejection fraction of left ventricular volumes was not different between groups at any time point. CONCLUSIONS In this porcine acute myocardial infarction model, allogeneic ATMSCs-based therapy was associated with increased cardioprotective and reparative mechanisms and with better cardiac magnetic resonance-measured perfusion. No effect on left ventricular volumes or ejection fraction was observed.
Collapse
Affiliation(s)
- Joaquim Bobi
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Institut de Malalties Cardiovasculars, Hospital Clínic de Barcelona, Universitat de Barcelona, Spain
| | - Núria Solanes
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Institut de Malalties Cardiovasculars, Hospital Clínic de Barcelona, Universitat de Barcelona, Spain
| | - Rodrigo Fernández-Jiménez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, CNIC, Madrid, Spain
- Icahn School of Medicine at Mount Sinai, New York, NY
- CIBER de enfermedades CardioVasculares (CIBERCV), Madrid, Spain
| | - Carlos Galán-Arriola
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, CNIC, Madrid, Spain
- CIBER de enfermedades CardioVasculares (CIBERCV), Madrid, Spain
| | - Ana Paula Dantas
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Institut de Malalties Cardiovasculars, Hospital Clínic de Barcelona, Universitat de Barcelona, Spain
| | - Leticia Fernández-Friera
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, CNIC, Madrid, Spain
- Hospital Universitario HM Montepríncipe, Madrid, Spain
- CIBER de enfermedades CardioVasculares (CIBERCV), Madrid, Spain
| | - Carolina Gálvez-Montón
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol, Badalona, Spain
- CIBER de enfermedades CardioVasculares (CIBERCV), Madrid, Spain
| | | | - Jaume Agüero
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, CNIC, Madrid, Spain
- CIBER de enfermedades CardioVasculares (CIBERCV), Madrid, Spain
- Cardiology Department, Hospital Universitari i Politecnic La Fe, Valencia, Spain
| | - José Ramírez
- Servei d'Anatomia Patològica, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Mercè Roqué
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Institut de Malalties Cardiovasculars, Hospital Clínic de Barcelona, Universitat de Barcelona, Spain
| | - Antoni Bayés-Genís
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol, Badalona, Spain
- Cardiology Service, Germans Trias i Pujol University Hospital, Badalona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
- CIBER de enfermedades CardioVasculares (CIBERCV), Madrid, Spain
| | | | - Ana García-Álvarez
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Institut de Malalties Cardiovasculars, Hospital Clínic de Barcelona, Universitat de Barcelona, Spain
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, CNIC, Madrid, Spain
| | - Manel Sabaté
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Institut de Malalties Cardiovasculars, Hospital Clínic de Barcelona, Universitat de Barcelona, Spain
| | - Santiago Roura
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol, Badalona, Spain
- Center of Regenerative Medicine in Barcelona, Barcelona, Spain
- CIBER de enfermedades CardioVasculares (CIBERCV), Madrid, Spain
| | - Borja Ibáñez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, CNIC, Madrid, Spain
- IIS- Fundación Jiménez Díaz Hospital, Madrid, Spain
- CIBER de enfermedades CardioVasculares (CIBERCV), Madrid, Spain
| | - Montserrat Rigol
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Institut de Malalties Cardiovasculars, Hospital Clínic de Barcelona, Universitat de Barcelona, Spain
| |
Collapse
|