51
|
Ziegler M, Haigh K, Nguyen T, Wang X, Lim B, Yap ML, Eddy EM, Haigh JJ, Peter K. The pulmonary microvasculature entraps induced vascular progenitor cells (iVPCs) systemically delivered after cardiac ischemia-reperfusion injury: Indication for preservation of heart function via paracrine effects beyond engraftment. Microcirculation 2018; 26:e12493. [PMID: 30030876 DOI: 10.1111/micc.12493] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 07/05/2018] [Accepted: 07/17/2018] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Stem cell-based regenerative therapies have been intensively studied with the aim to define an ideal cell type for the treatment of myocardial infarction. We tested systemically delivered, platelet-targeted induced vascular progenitor cells (iVPCs) to study their potential to salvage damaged myocardium after ischemia-reperfusion injury. METHODS Using a mouse model of ischemia-reperfusion injury, we tested the potential of platelet-targeted iVPCs (1 × 106 targ-iVPCs) compared to non-targ-iVPCs and a saline control. Bioluminescence imaging, echocardiography, and histological analyses were performed. RESULTS Four weeks after ischemia-reperfusion injury, systemic delivery of targ-iVPCs led to reduced fibrosis and infarct size (PBS: 25.7 ± 3.9 vs targ-iVPC: 18.4 ± 6.6 vs non-targ-iVPC: 25.1 ± 3.7%I/LV, P < 0.05), increased neovascularization, and restored cardiac function (PBS: 44.0 ± 4.2 vs targ-iVPC: 54.3 ± 4.5 vs non-targ-iVPC: 46.4 ± 3.8%EF, P < 0.01). Cell tracking experiments revealed entrapment of intravenously injected iVPCs in the pulmonary microvasculature in both cell-treated groups. CONCLUSIONS Systemic delivery of iVPCs after cardiac ischemia-reperfusion injury is limited by pulmonary entrapment of the cells. Nevertheless, targ-iVPCs reduced infarct size, fibrosis, increased neovascularization, and most importantly retained cardiac function. These findings contribute to the mechanistic discussion of cell-based therapy and ultimately identify activated platelet-targeted iVPCs as candidates for cell therapy and also describe cell therapy benefits without the necessity of engrafting.
Collapse
Affiliation(s)
- Melanie Ziegler
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, Melbourne, Vic., Australia
| | - Katharina Haigh
- Mammalian Functional Genetics Laboratory, Division of Blood Cancers, Australian Centre for Blood Diseases, Monash University, Melbourne, Vic., Australia
| | - Thao Nguyen
- Mammalian Functional Genetics Laboratory, Division of Blood Cancers, Australian Centre for Blood Diseases, Monash University, Melbourne, Vic., Australia
| | - Xiaowei Wang
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, Melbourne, Vic., Australia.,Department of Medicine, Monash University, Melbourne, Vic., Australia
| | - Bock Lim
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, Melbourne, Vic., Australia
| | - May Lin Yap
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, Melbourne, Vic., Australia
| | - Eleanor M Eddy
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, Melbourne, Vic., Australia
| | - Jody J Haigh
- Mammalian Functional Genetics Laboratory, Division of Blood Cancers, Australian Centre for Blood Diseases, Monash University, Melbourne, Vic., Australia
| | - Karlheinz Peter
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, Melbourne, Vic., Australia.,Department of Medicine, Monash University, Melbourne, Vic., Australia
| |
Collapse
|
52
|
Lazar E, Benedek T, Korodi S, Rat N, Lo J, Benedek I. Stem cell-derived exosomes - an emerging tool for myocardial regeneration. World J Stem Cells 2018; 10:106-115. [PMID: 30190780 PMCID: PMC6121000 DOI: 10.4252/wjsc.v10.i8.106] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 07/11/2018] [Accepted: 07/16/2018] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular diseases (CVDs) continue to represent the number one cause of death and disability in industrialized countries. The most severe form of CVD is acute myocardial infarction (AMI), a devastating disease associated with high mortality and disability. In a substantial proportion of patients who survive AMI, loss of functional cardiomyocytes as a result of ischaemic injury leads to ventricular failure, resulting in significant alteration to quality of life and increased mortality. Therefore, many attempts have been made in recent years to identify new tools for the regeneration of functional cardiomyocytes. Regenerative therapy currently represents the ultimate goal for restoring the function of damaged myocardium by stimulating the regeneration of the infarcted tissue or by providing cells that can generate new myocardial tissue to replace the damaged tissue. Stem cells (SCs) have been proposed as a viable therapy option in these cases. However, despite the great enthusiasm at the beginning of the SC era, justified by promising initial results, this therapy has failed to demonstrate a significant benefit in large clinical trials. One interesting finding of SC studies is that exosomes released by mesenchymal SCs (MSCs) are able to enhance the viability of cardiomyocytes after ischaemia/reperfusion injury, suggesting that the beneficial effects of MSCs in the recovery of functional myocardium could be related to their capacity to secrete exosomes. Ten years ago, it was discovered that exosomes have the unique property of transferring miRNA between cells, acting as miRNA nanocarriers. Therefore, exosome-based therapy has recently been proposed as an emerging tool for cardiac regeneration as an alternative to SC therapy in the post-infarction period. This review aims to discuss the emerging role of exosomes in developing innovative therapies for cardiac regeneration as well as their potential role as candidate biomarkers or for developing new diagnostic tools.
Collapse
Affiliation(s)
- Erzsebet Lazar
- Department of Internal Medicine, Clinic of Haematology and Bone Marrow Transplantation, University of Medicine and Pharmacy Tirgu Mures, Tirgu Mures 540042, Romania
| | - Theodora Benedek
- Department of Internal Medicine, Clinic of Cardiology, University of Medicine and Pharmacy Tirgu Mures, Tirgu Mures 540136, Romania
- Department of Advanced Research in Multimodality Cardiac Imaging, Cardio Med Medical Center, Tirgu Mures 540124, Romania
| | - Szilamer Korodi
- Department of Internal Medicine, Clinic of Cardiology, University of Medicine and Pharmacy Tirgu Mures, Tirgu Mures 540136, Romania
- Department of Advanced Research in Multimodality Cardiac Imaging, Cardio Med Medical Center, Tirgu Mures 540124, Romania
| | - Nora Rat
- Department of Internal Medicine, Clinic of Cardiology, University of Medicine and Pharmacy Tirgu Mures, Tirgu Mures 540136, Romania
- Department of Advanced Research in Multimodality Cardiac Imaging, Cardio Med Medical Center, Tirgu Mures 540124, Romania
| | - Jocelyn Lo
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, United States
| | - Imre Benedek
- Department of Internal Medicine, Clinic of Cardiology, University of Medicine and Pharmacy Tirgu Mures, Tirgu Mures 540136, Romania
- Department of Advanced Research in Multimodality Cardiac Imaging, Cardio Med Medical Center, Tirgu Mures 540124, Romania
| |
Collapse
|
53
|
Baio J, Martinez AF, Silva I, Hoehn CV, Countryman S, Bailey L, Hasaniya N, Pecaut MJ, Kearns-Jonker M. Cardiovascular progenitor cells cultured aboard the International Space Station exhibit altered developmental and functional properties. NPJ Microgravity 2018; 4:13. [PMID: 30062101 PMCID: PMC6062551 DOI: 10.1038/s41526-018-0048-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 06/18/2018] [Accepted: 06/25/2018] [Indexed: 12/15/2022] Open
Abstract
The heart and its cellular components are profoundly altered by missions to space and injury on Earth. Further research, however, is needed to characterize and address the molecular substrates of such changes. For this reason, neonatal and adult human cardiovascular progenitor cells (CPCs) were cultured aboard the International Space Station. Upon return to Earth, we measured changes in the expression of microRNAs and of genes related to mechanotransduction, cardiogenesis, cell cycling, DNA repair, and paracrine signaling. We additionally assessed endothelial-like tube formation, cell cycling, and migratory capacity of CPCs. Changes in microRNA expression were predicted to target extracellular matrix interactions and Hippo signaling in both neonatal and adult CPCs. Genes related to mechanotransduction (YAP1, RHOA) were downregulated, while the expression of cytoskeletal genes (VIM, NES, DES, LMNB2, LMNA), non-canonical Wnt ligands (WNT5A, WNT9A), and Wnt/calcium signaling molecules (PLCG1, PRKCA) was significantly elevated in neonatal CPCs. Increased mesendodermal gene expression along with decreased expression of mesodermal derivative markers (TNNT2, VWF, and RUNX2), reduced readiness to form endothelial-like tubes, and elevated expression of Bmp and Tbx genes, were observed in neonatal CPCs. Both neonatal and adult CPCs exhibited increased expression of DNA repair genes and paracrine factors, which was supported by enhanced migration. While spaceflight affects cytoskeletal organization and migration in neonatal and adult CPCs, only neonatal CPCs experienced increased expression of early developmental markers and an enhanced proliferative potential. Efforts to recapitulate the effects of spaceflight on Earth by regulating processes described herein may be a promising avenue for cardiac repair.
Collapse
Affiliation(s)
- Jonathan Baio
- 1Department of Pathology and Human Anatomy, Loma Linda University, Loma Linda, CA USA
| | - Aida F Martinez
- 1Department of Pathology and Human Anatomy, Loma Linda University, Loma Linda, CA USA
| | - Ivan Silva
- 1Department of Pathology and Human Anatomy, Loma Linda University, Loma Linda, CA USA
| | - Carla V Hoehn
- 2BioServe Space Technologies, University of Colorado Boulder, Boulder, CO USA
| | | | - Leonard Bailey
- 3Department of Cardiovascular and Thoracic Surgery, Loma Linda University, Loma Linda, CA USA
| | - Nahidh Hasaniya
- 3Department of Cardiovascular and Thoracic Surgery, Loma Linda University, Loma Linda, CA USA
| | - Michael J Pecaut
- 4Division of Biomedical Engineering Sciences, Department of Basic Sciences, Loma Linda University, Loma Linda, CA USA
| | - Mary Kearns-Jonker
- 1Department of Pathology and Human Anatomy, Loma Linda University, Loma Linda, CA USA
| |
Collapse
|
54
|
Abstract
Death of adult cardiac myocytes and supportive tissues resulting from cardiovascular diseases such as myocardial infarction is the proximal driver of pathological ventricular remodeling that often culminates in heart failure. Unfortunately, no currently available therapeutic barring heart transplantation can directly replenish myocytes lost from the injured heart. For decades, the field has struggled to define the intrinsic capacity and cellular sources for endogenous myocyte turnover in pursuing more innovative therapeutic strategies aimed at regenerating the injured heart. Although controversy persists to this day as to the best therapeutic regenerative strategy to use, a growing consensus has been reached that the very limited capacity for new myocyte formation in the adult mammalian heart is because of proliferation of existing cardiac myocytes but not because of the activity of an endogenous progenitor cell source of some sort. Hence, future therapeutic approaches should take into consideration the fundamental biology of myocyte renewal in designing strategies to potentially replenish these cells in the injured heart.
Collapse
Affiliation(s)
| | - Jeffery D Molkentin
- From the Department of Pediatrics (R.J.V., J.D.M.)
- Howard Hughes Medical Institute (J.D.M.)
| | - Steven R Houser
- Cincinnati Children's Hospital Medical Center, OH; and the Lewis Katz School of Medicine, Cardiovascular Research Center, Temple University, Philadelphia, PA (S.R.H.)
| |
Collapse
|
55
|
Lewandowski J, Rozwadowska N, Kolanowski TJ, Malcher A, Zimna A, Rugowska A, Fiedorowicz K, Łabędź W, Kubaszewski Ł, Chojnacka K, Bednarek-Rajewska K, Majewski P, Kurpisz M. The impact of in vitro cell culture duration on the maturation of human cardiomyocytes derived from induced pluripotent stem cells of myogenic origin. Cell Transplant 2018; 27:1047-1067. [PMID: 29947252 PMCID: PMC6158549 DOI: 10.1177/0963689718779346] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Ischemic heart disease, also known as coronary artery disease (CAD), poses a challenge
for regenerative medicine. iPSC technology might lead to a breakthrough due to the
possibility of directed cell differentiation delivering a new powerful source of human
autologous cardiomyocytes. One of the factors supporting proper cell maturation is in
vitro culture duration. In this study, primary human skeletal muscle myoblasts were
selected as a myogenic cell type reservoir for genetic iPSC reprogramming. Skeletal muscle
myoblasts have similar ontogeny embryogenetic pathways (myoblasts vs. cardiomyocytes), and
thus, a greater chance of myocardial development might be expected, with maintenance of
acquired myogenic cardiac cell characteristics, from the differentiation process when
iPSCs of myoblastoid origin are obtained. Analyses of cell morphological and structural
changes, gene expression (cardiac markers), and functional tests (intracellular calcium
transients) performed at two in vitro culture time points spanning the early stages of
cardiac development (day 20 versus 40 of cell in vitro culture) confirmed the ability of
the obtained myogenic cells to acquire adult features of differentiated cardiomyocytes.
Prolonged 40-day iPSC-derived cardiomyocytes (iPSC-CMs) revealed progressive cellular
hypertrophy; a better-developed contractile apparatus; expression of marker genes similar
to human myocardial ventricular cells, including a statistically significant
CX43 increase, an MHC isoform switch, and a troponin I isoform
transition; more efficient intercellular calcium handling; and a stronger response to
β-adrenergic stimulation.
Collapse
Affiliation(s)
- Jarosław Lewandowski
- 1 Institute of Human Genetics, Polish Academy of Sciences, Strzeszynska, Poznan, Poland
| | - Natalia Rozwadowska
- 1 Institute of Human Genetics, Polish Academy of Sciences, Strzeszynska, Poznan, Poland
| | - Tomasz J Kolanowski
- 1 Institute of Human Genetics, Polish Academy of Sciences, Strzeszynska, Poznan, Poland
| | - Agnieszka Malcher
- 1 Institute of Human Genetics, Polish Academy of Sciences, Strzeszynska, Poznan, Poland
| | - Agnieszka Zimna
- 1 Institute of Human Genetics, Polish Academy of Sciences, Strzeszynska, Poznan, Poland
| | - Anna Rugowska
- 1 Institute of Human Genetics, Polish Academy of Sciences, Strzeszynska, Poznan, Poland
| | - Katarzyna Fiedorowicz
- 1 Institute of Human Genetics, Polish Academy of Sciences, Strzeszynska, Poznan, Poland
| | - Wojciech Łabędź
- 2 Department of Orthopaedics and Traumatology, W. Dega University Hospital, Poznan University of Medical Sciences, Poznan, Poland.,3 Department of Spondyloorthopaedics and Biomechanics of the Spine, W. Dega University Hospital, Poznan University of Medical Sciences, Poznan, Poland
| | - Łukasz Kubaszewski
- 2 Department of Orthopaedics and Traumatology, W. Dega University Hospital, Poznan University of Medical Sciences, Poznan, Poland.,3 Department of Spondyloorthopaedics and Biomechanics of the Spine, W. Dega University Hospital, Poznan University of Medical Sciences, Poznan, Poland
| | - Katarzyna Chojnacka
- 4 Department of Clinical Pathology, Heliodor Swiecicki Clinical Hospital No. 2 of the Poznan University of Medical Sciences, Poznan, Poland
| | | | - Przemysław Majewski
- 5 Department of Clinical Pathology, Poznan University of Medical Sciences, Poznan, Poland
| | - Maciej Kurpisz
- 1 Institute of Human Genetics, Polish Academy of Sciences, Strzeszynska, Poznan, Poland
| |
Collapse
|
56
|
Protective effects of human induced pluripotent stem cell-derived exosomes on high glucose-induced injury in human endothelial cells. Exp Ther Med 2018; 15:4791-4797. [PMID: 29805497 PMCID: PMC5958753 DOI: 10.3892/etm.2018.6059] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 08/25/2017] [Indexed: 11/05/2022] Open
Abstract
Exosomes are a family of extracellular vesicles that are secreted from almost all types of cells and are associated with cell-to-cell communication. The present study was performed to investigate the effects of human induced pluripotent stem cell-derived exosomes (hiPSC-exo) on cell viability, capillary-like structure formation and senescence in endothelial cells exposed to high glucose. Exosomes were isolated from the conditional medium of hiPSCs and confirmed by transmission electron microscopy, nanoparticle tracking analysis and western blot analysis using Alix and cluster of differentiation-63 as markers. hiPSC-exo were labeled with PKH26 for tracking, and it was determined that spherical exosomes, with a typical cup-shape, were absorbed by human umbilical vascular endothelial cells (HUVECs). Cultured HUVECs were treated with high glucose (33 mM) with or without hiPSC-exo (20 µg/ml) for 48 h, and cell viability, capillary tube formation and senescence were assessed. When exposed to high glucose, viability and tube formation in HUVECs was significantly reduced (P<0.0001), whereas the proportion of senescent cells was higher compared with that in control HUVECs (P<0.0001). Furthermore, hiPSC-exo restored cell viability and capillary-like structure formation, and reduced senescence in HUVECs exposed to high glucose (P<0.0001). However, hiPSC-exo had minimal effects on normal HUVECs. These findings suggest that stem cell-derived exosomes are able to promote cell proliferation, enhance capillary-like structure formation and reduce senescence in endothelial cells exposed to high glucose.
Collapse
|
57
|
Madigan M, Atoui R. Therapeutic Use of Stem Cells for Myocardial Infarction. Bioengineering (Basel) 2018; 5:bioengineering5020028. [PMID: 29642402 PMCID: PMC6027340 DOI: 10.3390/bioengineering5020028] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 03/29/2018] [Accepted: 04/04/2018] [Indexed: 12/15/2022] Open
Abstract
Myocardial infarction is a leading cause of morbidity and mortality worldwide. Although medical and surgical treatments can significantly improve patient outcomes, no treatment currently available is able to generate new contractile tissue or reverse ischemic myocardium. Driven by the recent/novel understanding that regenerative processes do exist in the myocardium—tissue previously thought not to possess regenerative properties—the use of stem cells has emerged as a promising therapeutic approach with high expectations. The literature describes the use of cells from various sources, categorizing them as either embryonic, induced pluripotent, or adult/tissue stem cells (mesenchymal, hematopoietic, skeletal myoblasts, cardiac stem cells). Many publications show the successful use of these cells to regenerate damaged myocardium in both animal and human models; however, more studies are needed to directly compare cells of various origins in efforts to draw conclusions on the ideal source. Although numerous challenges exist in this developing area of research and clinical practice, prospects are encouraging. The following aims to provide a concise review outlining the different types of stem cells used in patients after myocardial infarction.
Collapse
Affiliation(s)
- Mariah Madigan
- Northern Ontario School of Medicine, Sudbury, ON P3E 2C6, Canada.
| | - Rony Atoui
- Health Sciences North, Sudbury, ON P3E 5J1, Canada.
| |
Collapse
|
58
|
|
59
|
Baio J, Martinez AF, Bailey L, Hasaniya N, Pecaut MJ, Kearns-Jonker M. Spaceflight Activates Protein Kinase C Alpha Signaling and Modifies the Developmental Stage of Human Neonatal Cardiovascular Progenitor Cells. Stem Cells Dev 2018; 27:805-818. [PMID: 29320953 DOI: 10.1089/scd.2017.0263] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Spaceflight impacts cardiovascular function in astronauts; however, its impact on cardiac development and the stem cells that form the basis for cardiac repair is unknown. Accordingly, further research is needed to uncover the potential relevance of such changes to human health. Using simulated microgravity (SMG) generated by two-dimensional clinorotation and culture aboard the International Space Station (ISS), we assessed the effects of mechanical unloading on human neonatal cardiovascular progenitor cell (CPC) developmental properties and signaling. Following 6-7 days of SMG and 12 days of ISS culture, we analyzed changes in gene expression. Both environments induced the expression of genes that are typically associated with an earlier state of cardiovascular development. To understand the mechanism by which such changes occurred, we assessed the expression of mechanosensitive small RhoGTPases in SMG-cultured CPCs and observed decreased levels of RHOA and CDC42. Given the effect of these molecules on intracellular calcium levels, we evaluated changes in noncanonical Wnt/calcium signaling. After 6-7 days under SMG, CPCs exhibited elevated levels of WNT5A and PRKCA. Similarly, ISS-cultured CPCs exhibited elevated levels of calcium handling and signaling genes, which corresponded to protein kinase C alpha (PKCα), a calcium-dependent protein kinase, activation after 30 days. Akt was activated, whereas phosphorylated extracellular signal-regulated kinase levels were unchanged. To explore the effect of calcium induction in neonatal CPCs, we activated PKCα using hWnt5a treatment on Earth. Subsequently, early cardiovascular developmental marker levels were elevated. Transcripts induced by SMG and hWnt5a-treatment are expressed within the sinoatrial node, which may represent embryonic myocardium maintained in its primitive state. Calcium signaling is sensitive to mechanical unloading and directs CPC developmental properties. Further research both in space and on Earth may help refine the use of CPCs in stem cell-based therapies and highlight the molecular events of development.
Collapse
Affiliation(s)
- Jonathan Baio
- 1 Department of Pathology and Human Anatomy, Loma Linda University , Loma Linda, California
| | - Aida F Martinez
- 1 Department of Pathology and Human Anatomy, Loma Linda University , Loma Linda, California
| | - Leonard Bailey
- 2 Department of Cardiovascular and Thoracic Surgery, Loma Linda University , Loma Linda, California
| | - Nahidh Hasaniya
- 2 Department of Cardiovascular and Thoracic Surgery, Loma Linda University , Loma Linda, California
| | - Michael J Pecaut
- 3 Division of Biomedical Engineering Sciences, Department of Basic Sciences, Loma Linda University , Loma Linda, California
| | - Mary Kearns-Jonker
- 1 Department of Pathology and Human Anatomy, Loma Linda University , Loma Linda, California
| |
Collapse
|
60
|
Zhu K, Wu Q, Ni C, Zhang P, Zhong Z, Wu Y, Wang Y, Xu Y, Kong M, Cheng H, Tao Z, Yang Q, Liang H, Jiang Y, Li Q, Zhao J, Huang J, Zhang F, Chen Q, Li Y, Chen J, Zhu W, Yu H, Zhang J, Yang HT, Hu X, Wang J. Lack of Remuscularization Following Transplantation of Human Embryonic Stem Cell-Derived Cardiovascular Progenitor Cells in Infarcted Nonhuman Primates. Circ Res 2018; 122:958-969. [PMID: 29343525 DOI: 10.1161/circresaha.117.311578] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 01/09/2018] [Accepted: 01/12/2018] [Indexed: 01/13/2023]
Abstract
RATIONALE Human pluripotent stem cell-derived cardiovascular progenitor cells (hPSC-CVPCs) should be thoroughly investigated in large animal studies before testing in clinical trials. OBJECTIVE The main of this study is to clarify whether hPSC-CVPCs can engraft for long time in the heart of primates after myocardial infarction (MI) and compare the effectiveness and safety of immunosuppression with cyclosporine alone or multiple-drug regimen (MDR) containing cyclosporine, methylprednisolone, and basiliximab in cynomolgus monkeys that had received intramyocardial injections of 1×107 EGFP (enhanced green fluorescent protein)-expressing hPSC-CVPCs after MI. A third group of animals received the immunosuppression MDR but without cell therapy after MI (MI+MDR group). METHODS AND RESULTS Measurements of EGFP gene levels and EGFP immunofluorescence staining indicated that the hPSC-CVPC engraftment rate was greater in the MI+MDR+CVPC group than that in the MI+cyclosporine+CVPC group. However, even in the MI+MDR+CVPC group, no transplanted cells could be detected at 140 days after transplantation. Concomitantly, immunofluorescent analysis of CD3, CD4, and CD8 expression indicated that T-lymphocyte infiltration in the CVPC-transplanted hearts was less in the MDR-treated animals than in the cyclosporine-alone-treated animals. The recovery of left ventricular function on day 28 post-MI in the MI+MDR+CVPC group was better than that in the MI+MDR group. Apoptotic cardiac cells were also less common in the MI+MDR+CVPC group than in the MI+MDR group, although both immunosuppression regimens were associated with transient hepatic dysfunction. CONCLUSIONS This is the largest study of hPSCs in nonhuman primates in cardiovascular field to date (n=32). Compared with cyclosporine alone, MDR attenuates immune rejection and improves survival of hPSC-CVPCs in primates; this is associated with less apoptosis of native cardiac cells and better recovery of left ventricular function at 28 days. However, even with MDR, transplanted hPSC-CVPCs do not engraft and do not survive at 140 days after transplantation, thereby excluding remuscularization as a mechanism for the functional effect.
Collapse
Affiliation(s)
- Keyang Zhu
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang)
| | - Qiang Wu
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang)
| | - Cheng Ni
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang)
| | - Peng Zhang
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang)
| | - Zhiwei Zhong
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang)
| | - Yan Wu
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang)
| | - Yingchao Wang
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang)
| | - Yinchuan Xu
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang)
| | - Minjian Kong
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang)
| | - Haifeng Cheng
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang)
| | - Zhihua Tao
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang)
| | - Qian Yang
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang)
| | - He Liang
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang)
| | - Yun Jiang
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang)
| | - Qingju Li
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang)
| | - Jing Zhao
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang)
| | - Jijun Huang
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang)
| | - Fengjiang Zhang
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang)
| | - Qi Chen
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang)
| | - Yi Li
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang)
| | - Jinghai Chen
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang)
| | - Wei Zhu
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang)
| | - Hong Yu
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang)
| | - Jianyi Zhang
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang)
| | - Huang-Tian Yang
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang).
| | - Xinyang Hu
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang).
| | - Jian'an Wang
- From the Department of Cardiology (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.Y., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., J.W.), Department of Cardiovascular Surgery (M.K., H.C.), Department of Laboratory Medicine (Z.T.), and Department of Anesthesiology (F.Z., Q.C., Y.L.), Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China; Key Laboratory of Stem Cell Biology and Laboratory of Molecular Cardiology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS) Shanghai, University of CAS, Beijing, PR China (Q.W., P.Z., H.L., Y.J., J.H., H.-T.Y.); Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, PR China (K.Z., C.N., Z.Z., Y. Wu, Y. Wang, Y.X., Q.L., J. Zhao, J.C., W.Z., H.Y., X.H., Y.W.); and Department of Biomedical Engineering, University of Alabama at Birmingham (J. Zhang).
| |
Collapse
|
61
|
Abstract
Bioengineering cardiac tissue constructs with adult cardiomyocytes may help treat adult heart defects and injury. In this study, we fabricated cardiac tissue constructs by seeding adult rat cardiomyocytes on a fibrin gel matrix and analyzed the electromechanical properties of the formed cardiac tissue constructs. Adult rat cardiomyocytes were isolated with a collagenase type II buffer using an optimized Langendorff perfusion system. Cardiac tissue constructs were fabricated using either indirect plating with cardiomyocytes that were cultured for 1 week and dedifferentiated or with freshly isolated cardiomyocytes. The current protocol generated (3.1 ± 0.5) × 10 (n = 5 hearts) fresh cardiomyocytes from a single heart. Tissue constructs obtained by both types of plating contracted up to 30 days, and electrogram (ECG) signals and contractile twitch forces were detected. The constructs bioengineered by indirect plating of dedifferentiated cardiomyocytes produced an ECG R wave amplitude of 15.1 ± 5.2 µV (n = 7 constructs), a twitch force of 70-110 µN, and a spontaneous contraction rate of about 390 bpm. The constructs bioengineered by direct plating of fresh cardiomyocytes generated an ECG R wave amplitude of 6.3 ± 2.5 µV (n = 8 constructs), a twitch force of 40-60 µN, and a spontaneous contraction rate of about 230 bpm. This study successfully bioengineered three-dimensional cardiac tissue constructs using primary adult cardiomyocytes.
Collapse
|
62
|
Domingues S, Masson Y, Marteyn A, Allouche J, Perrier AL, Peschanski M, Martinat C, Baldeschi C, Lemaître G. Differentiation of nonhuman primate pluripotent stem cells into functional keratinocytes. Stem Cell Res Ther 2017; 8:285. [PMID: 29258610 PMCID: PMC5738144 DOI: 10.1186/s13287-017-0741-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 11/16/2017] [Accepted: 12/01/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Epidermal grafting using cells derived from pluripotent stem cells will change the face of this side of regenerative cutaneous medicine. To date, the safety of the graft would be the major unmet deal in order to implement long-term skin grafting. In this context, experiments on large animals appear unavoidable to assess this question and possible rejection. Cellular tools for large animal models should be constructed. METHODS In this study, we generated monkey pluripotent stem cell-derived keratinocytes and evaluated their capacities to reconstruct an epidermis, in vitro as well as in vivo. RESULTS Monkey pluripotent stem cells were differentiated efficiently into keratinocytes able to reconstruct fully epidermis presenting a low level of major histocompatibility complex class-I antigens, opening the way for autologous or allogeneic epidermal long-term grafting. CONCLUSIONS Functional keratinocytes generated from nonhuman primate embryonic stem cells and induced pluripotent stem cells reproduce an in-vitro and in-vivo stratified epidermis. These monkey skin grafts will be considered to model autologous or allogeneic epidermal grafting using either embryonic stem cells or induced pluripotent stem cells. This graft model will allow us to further investigate the safety, efficacy and immunogenicity of nonhuman primate PSC-derived epidermis in the perspective of human skin cell therapy.
Collapse
Affiliation(s)
- Sophie Domingues
- INSERM U-861, Institut des cellules Souches pour le Traitement et l'Etude des Maladies monogéniques (I-Stem), Association Française contre les Myopathies (AFM), 91100, Corbeil Essonnes, France.,Centre d'Etude des Cellules Souches (CECS), I-Stem, AFM, 91100, Corbeil Essonnes, France
| | - Yolande Masson
- Centre d'Etude des Cellules Souches (CECS), I-Stem, AFM, 91100, Corbeil Essonnes, France
| | - Aurore Marteyn
- Centre d'Etude des Cellules Souches (CECS), I-Stem, AFM, 91100, Corbeil Essonnes, France
| | - Jennifer Allouche
- INSERM U-861, Institut des cellules Souches pour le Traitement et l'Etude des Maladies monogéniques (I-Stem), Association Française contre les Myopathies (AFM), 91100, Corbeil Essonnes, France.,Université Evry Val d'Essonne (UEVE), Paris Saclay, U861, I-Stem, AFM, 91030, Evry Cedex, France
| | - Anselme L Perrier
- INSERM U-861, Institut des cellules Souches pour le Traitement et l'Etude des Maladies monogéniques (I-Stem), Association Française contre les Myopathies (AFM), 91100, Corbeil Essonnes, France
| | - Marc Peschanski
- INSERM U-861, Institut des cellules Souches pour le Traitement et l'Etude des Maladies monogéniques (I-Stem), Association Française contre les Myopathies (AFM), 91100, Corbeil Essonnes, France.,Université Evry Val d'Essonne (UEVE), Paris Saclay, U861, I-Stem, AFM, 91030, Evry Cedex, France
| | - Cecile Martinat
- INSERM U-861, Institut des cellules Souches pour le Traitement et l'Etude des Maladies monogéniques (I-Stem), Association Française contre les Myopathies (AFM), 91100, Corbeil Essonnes, France.,Université Evry Val d'Essonne (UEVE), Paris Saclay, U861, I-Stem, AFM, 91030, Evry Cedex, France
| | - Christine Baldeschi
- INSERM U-861, Institut des cellules Souches pour le Traitement et l'Etude des Maladies monogéniques (I-Stem), Association Française contre les Myopathies (AFM), 91100, Corbeil Essonnes, France.,Université Evry Val d'Essonne (UEVE), Paris Saclay, U861, I-Stem, AFM, 91030, Evry Cedex, France
| | - Gilles Lemaître
- INSERM U-861, Institut des cellules Souches pour le Traitement et l'Etude des Maladies monogéniques (I-Stem), Association Française contre les Myopathies (AFM), 91100, Corbeil Essonnes, France. .,Université Evry Val d'Essonne (UEVE), Paris Saclay, U861, I-Stem, AFM, 91030, Evry Cedex, France.
| |
Collapse
|
63
|
Martin U. Therapeutic Application of Pluripotent Stem Cells: Challenges and Risks. Front Med (Lausanne) 2017; 4:229. [PMID: 29312943 PMCID: PMC5735065 DOI: 10.3389/fmed.2017.00229] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 11/30/2017] [Indexed: 12/30/2022] Open
Abstract
Stem-cell-based therapies are considered to be promising and innovative but complex approaches. Induced pluripotent stem cells (iPSCs) combine the advantages of adult stem cells with the hitherto unique characteristics of embryonic stem cells (ESCs). Major progress has already been achieved with regard to reprogramming technology, but also regarding targeted genome editing and scalable expansion and differentiation of iPSCs and ESCs, in some cases yielding highly enriched preparations of well-defined cell lineages at clinically required dimensions. It is noteworthy, however, that for many applications critical requirements such as the targeted specification into distinct cellular subpopulations and a proper cell maturation remain to be achieved. Moreover, current hurdles such as low survival rates and insufficient functional integration of cellular transplants remain to be overcome. Nevertheless, PSC technologies obviously have come of age and matured to a stage where various clinical applications of PSC-based cellular therapies have been initiated and are conducted.
Collapse
Affiliation(s)
- Ulrich Martin
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, REBIRTH Cluster of Excellence, German Center for Lung Research, Hannover Medical School, Hannover, Germany
| |
Collapse
|
64
|
Abstract
PURPOSE OF REVIEW This review provides an overview of the current state of tissue-engineered heart repair with a special focus on the anticipated modes of action of tissue-engineered therapy candidates and particular implications as to transplant immunology. RECENT FINDINGS Myocardial tissue engineering technologies have made tremendous advances in recent years. Numerous different strategies are under investigation and have reached different stages on their way to clinical translation. Studies in animal models demonstrated that heart repair requires either remuscularization by delivery of bona fide cardiomyocytes or paracrine support for the activation of endogenous repair mechanisms. Tissue engineering approaches result in enhanced cardiomyocyte retention and sustained remuscularization, but may also be explored for targeted paracrine or mechanical support. Some of the more advanced tissue engineering approaches are already tested clinically; others are at late stages of pre-clinical development. Process optimization towards cGMP compatibility and clinical scalability of contractile engineered human myocardium is an essential step towards clinical translation. Long-term allograft retention can be achieved under immune suppression. HLA matching may be an option to enhance graft retention and reduce the need for comprehensive immune suppression. Tissue-engineered heart repair is entering the clinical stage of the translational pipeline. Like in any effective therapy, side effects must be anticipated and carefully controlled. Allograft implantation under immune suppression is the most likely clinical scenario. Strategies to overcome transplant rejection are evolving and may further boost the clinical acceptance of tissue-engineered heart repair.
Collapse
Affiliation(s)
- Buntaro Fujita
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany.,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Göttingen, Germany.,Department of Thoracic and Cardiovascular Surgery, Heart and Diabetes Center NRW, Ruhr-University Bochum, Bad Oeynhausen, Germany
| | - Wolfram-Hubertus Zimmermann
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany. .,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Göttingen, Germany.
| |
Collapse
|
65
|
Functional expression of the Ca 2+ signaling machinery in human embryonic stem cells. Acta Pharmacol Sin 2017; 38:1663-1672. [PMID: 28713161 DOI: 10.1038/aps.2017.29] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 03/13/2017] [Indexed: 12/28/2022] Open
Abstract
Emerging evidence suggests that Ca2+ signals are important for the self-renewal and differentiation of human embryonic stem cells (hESCs). However, little is known about the physiological and pharmacological properties of the Ca2+-handling machinery in hESCs. In this study we used RT-PCR and Western blotting to analyze the expression profiles of genes encoding Ca2+-handling proteins; we also used confocal Ca2+ imaging and pharmacological approaches to determine the contribution of the Ca2+-handling machinery to the regulation of Ca2+ signaling in hESCs. We revealed that hESCs expressed pluripotent markers and various Ca2+-handling-related genes. ATP-induced Ca2+ transients in almost all hESCs were inhibited by the inositol-1,4,5-triphosphate receptor (IP3R) blocker 2-APB or xestospongin C. In addition, Ca2+ transients were induced by a ryanodine receptor (RyR) activator, caffeine, in 10%-15% of hESCs and were blocked by ryanodine, whereas caffeine and ATP did not have additive effects. Moreover, store-operated Ca2+ entry (SOCE) but not voltage-operated Ca2+ channel-mediated Ca2+ entry was observed. Inhibition of sarco/endoplasmic reticulum (ER) Ca2+-ATPase (SERCA) by thapsigargin induced a significant increase in the cytosolic free Ca2+ concentration ([Ca2+]i). For the Ca2+ extrusion pathway, inhibition of plasma membrane Ca2+ pumps (PMCAs) by carboxyeosin induced a slow increase in [Ca2+]i, whereas the Na+/Ca2+ exchanger (NCX) inhibitor KBR7943 induced a rapid increase in [Ca2+]i. Taken together, increased [Ca2+]i is mainly mediated by Ca2+ release from intracellular stores via IP3Rs. In addition, RyRs function in a portion of hESCs, thus indicating heterogeneity of the Ca2+-signaling machinery in hESCs; maintenance of low [Ca2+]i is mediated by uptake of cytosolic Ca2+ into the ER via SERCA and extrusion of Ca2+ out of cells via NCX and PMCA in hESCs.
Collapse
|
66
|
Myocardial Regeneration via Progenitor Cell-Derived Exosomes. Stem Cells Int 2017; 2017:7849851. [PMID: 29333167 PMCID: PMC5733225 DOI: 10.1155/2017/7849851] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 10/11/2017] [Indexed: 12/16/2022] Open
Abstract
In the past 20 years, a variety of cell products has been evaluated in terms of their capacity to treat patients with acute myocardial infarction and chronic heart failure. Despite initial enthusiasm, therapeutic efficacy has overall been disappointing, and clinical application is costly and complex. Recently, a subset of small extracellular vesicles (EVs), commonly referred to as "exosomes," was shown to confer cardioprotective and regenerative signals at a magnitude similar to that of their donor cells. The conceptual advantage is that they may be produced in industrial quantities and stored at the point-of-care for off-the-shelf application, ideally without eliciting a relevant recipient immune response or other adverse effects associated with viable cells. The body of evidence on beneficial exosome-mediated effects in animal models of heart diseases is rapidly growing. However, there is significant heterogeneity in terms of exosome source cells, isolation process, therapeutic dosage, and delivery mode. This review summarizes the current state of research on exosomes as experimental therapy of heart diseases and seeks to identify roadblocks that need to be overcome prior to clinical application.
Collapse
|
67
|
Sommese L, Zullo A, Schiano C, Mancini FP, Napoli C. Possible Muscle Repair in the Human Cardiovascular System. Stem Cell Rev Rep 2017; 13:170-191. [PMID: 28058671 DOI: 10.1007/s12015-016-9711-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The regenerative potential of tissues and organs could promote survival, extended lifespan and healthy life in multicellular organisms. Niches of adult stemness are widely distributed and lead to the anatomical and functional regeneration of the damaged organ. Conversely, muscular regeneration in mammals, and humans in particular, is very limited and not a single piece of muscle can fully regrow after a severe injury. Therefore, muscle repair after myocardial infarction is still a chimera. Recently, it has been recognized that epigenetics could play a role in tissue regrowth since it guarantees the maintenance of cellular identity in differentiated cells and, therefore, the stability of organs and tissues. The removal of these locks can shift a specific cell identity back to the stem-like one. Given the gradual loss of tissue renewal potential in the course of evolution, in the last few years many different attempts to retrieve such potential by means of cell therapy approaches have been performed in experimental models. Here we review pathways and mechanisms involved in the in vivo repair of cardiovascular muscle tissues in humans. Moreover, we address the ongoing research on mammalian cardiac muscle repair based on adult stem cell transplantation and pro-regenerative factor delivery. This latter issue, involving genetic manipulations of adult cells, paves the way for developing possible therapeutic strategies in the field of cardiovascular muscle repair.
Collapse
Affiliation(s)
- Linda Sommese
- Department of Internal and Specialty Medicine, U.O.C. Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology, Regional Reference Laboratory of Transplant Immunology, Azienda Ospedaliera Universitaria, Università degli Studi della Campania "Luigi Vanvitelli", Piazza Miraglia 2, 80138, Naples, Italy.
| | - Alberto Zullo
- Department of Sciences and Technologies, University of Sannio, Benevento, Italy.,CEINGE Advanced Biotechnologies, s.c.ar.l, Naples, Italy
| | | | - Francesco P Mancini
- Department of Sciences and Technologies, University of Sannio, Benevento, Italy
| | - Claudio Napoli
- Department of Internal and Specialty Medicine, U.O.C. Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology, Regional Reference Laboratory of Transplant Immunology, Azienda Ospedaliera Universitaria, Università degli Studi della Campania "Luigi Vanvitelli", Piazza Miraglia 2, 80138, Naples, Italy.,IRCCS Foundation SDN, Naples, Italy
| |
Collapse
|
68
|
Steinhoff G, Nesteruk J, Wolfien M, Große J, Ruch U, Vasudevan P, Müller P. Stem cells and heart disease - Brake or accelerator? Adv Drug Deliv Rev 2017; 120:2-24. [PMID: 29054357 DOI: 10.1016/j.addr.2017.10.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 10/12/2017] [Accepted: 10/13/2017] [Indexed: 12/11/2022]
Abstract
After two decades of intensive research and attempts of clinical translation, stem cell based therapies for cardiac diseases are not getting closer to clinical success. This review tries to unravel the obstacles and focuses on underlying mechanisms as the target for regenerative therapies. At present, the principal outcome in clinical therapy does not reflect experimental evidence. It seems that the scientific obstacle is a lack of integration of knowledge from tissue repair and disease mechanisms. Recent insights from clinical trials delineate mechanisms of stem cell dysfunction and gene defects in repair mechanisms as cause of atherosclerosis and heart disease. These findings require a redirection of current practice of stem cell therapy and a reset using more detailed analysis of stem cell function interfering with disease mechanisms. To accelerate scientific development the authors suggest intensifying unified computational data analysis and shared data knowledge by using open-access data platforms.
Collapse
Affiliation(s)
- Gustav Steinhoff
- University Medicine Rostock, Department of Cardiac Surgery, Reference and Translation Center for Cardiac Stem Cell Therapy, University Medical Center Rostock, Schillingallee 35, 18055 Rostock, Germany.
| | - Julia Nesteruk
- University Medicine Rostock, Department of Cardiac Surgery, Reference and Translation Center for Cardiac Stem Cell Therapy, University Medical Center Rostock, Schillingallee 35, 18055 Rostock, Germany.
| | - Markus Wolfien
- University Rostock, Institute of Computer Science, Department of Systems Biology and Bioinformatics, Ulmenstraße 69, 18057 Rostock, Germany.
| | - Jana Große
- University Medicine Rostock, Department of Cardiac Surgery, Reference and Translation Center for Cardiac Stem Cell Therapy, University Medical Center Rostock, Schillingallee 35, 18055 Rostock, Germany.
| | - Ulrike Ruch
- University Medicine Rostock, Department of Cardiac Surgery, Reference and Translation Center for Cardiac Stem Cell Therapy, University Medical Center Rostock, Schillingallee 35, 18055 Rostock, Germany.
| | - Praveen Vasudevan
- University Medicine Rostock, Department of Cardiac Surgery, Reference and Translation Center for Cardiac Stem Cell Therapy, University Medical Center Rostock, Schillingallee 35, 18055 Rostock, Germany.
| | - Paula Müller
- University Medicine Rostock, Department of Cardiac Surgery, Reference and Translation Center for Cardiac Stem Cell Therapy, University Medical Center Rostock, Schillingallee 35, 18055 Rostock, Germany.
| |
Collapse
|
69
|
Bei Y, Das S, Rodosthenous RS, Holvoet P, Vanhaverbeke M, Monteiro MC, Monteiro VVS, Radosinska J, Bartekova M, Jansen F, Li Q, Rajasingh J, Xiao J. Extracellular Vesicles in Cardiovascular Theranostics. Am J Cancer Res 2017; 7:4168-4182. [PMID: 29158817 PMCID: PMC5695004 DOI: 10.7150/thno.21274] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 08/19/2017] [Indexed: 12/17/2022] Open
Abstract
Extracellular vesicles (EVs) are small bilayer lipid membrane vesicles that can be released by most cell types and detected in most body fluids. EVs exert key functions for intercellular communication via transferring their bioactive cargos to recipient cells or activating signaling pathways in target cells. Increasing evidence has shown the important regulatory effects of EVs in cardiovascular diseases (CVDs). EVs secreted by cardiomyocytes, endothelial cells, fibroblasts, and stem cells play essential roles in pathophysiological processes such as cardiac hypertrophy, cardiomyocyte survival and apoptosis, cardiac fibrosis, and angiogenesis in relation to CVDs. In this review, we will first outline the current knowledge about the physical characteristics, biological contents, and isolation methods of EVs. We will then focus on the functional roles of cardiovascular EVs and their pathophysiological effects in CVDs, as well as summarize the potential of EVs as therapeutic agents and biomarkers for CVDs. Finally, we will discuss the specific application of EVs as a novel drug delivery system and the utility of EVs in the field of regenerative medicine.
Collapse
|
70
|
Han B, Nia HT, Wang C, Chandrasekaran P, Li Q, Chery DR, Li H, Grodzinsky AJ, Han L. AFM-Nanomechanical Test: An Interdisciplinary Tool That Links the Understanding of Cartilage and Meniscus Biomechanics, Osteoarthritis Degeneration, and Tissue Engineering. ACS Biomater Sci Eng 2017; 3:2033-2049. [PMID: 31423463 PMCID: PMC6697429 DOI: 10.1021/acsbiomaterials.7b00307] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Our objective is to provide an in-depth review of the recent technical advances of atomic force microscopy (AFM)-based nanomechanical tests and their contribution to a better understanding and diagnosis of osteoarthritis (OA), as well as the repair of tissues undergoing degeneration during OA progression. We first summarize a range of technical approaches for AFM-based nanoindentation, including considerations in both experimental design and data analysis. We then provide a more detailed description of two recently developed modes of AFM-nanoindentation, a high-bandwidth nanorheometer system for studying poroviscoelasticity and an immunofluorescence-guided nanomechanical mapping technique for delineating the pericellular matrix (PCM) and territorial/interterritorial matrix (T/IT-ECM) of surrounding cells in connective tissues. Next, we summarize recent applications of these approaches to three aspects of joint-related healthcare and disease: cartilage aging and OA, developmental biology and OA pathogenesis in murine models, and nanomechanics of the meniscus. These studies were performed over a hierarchy of length scales, from the molecular, cellular to the whole tissue level. The advances described here have contributed greatly to advancing the fundamental knowledge base for improved understanding, detection, and treatment of OA.
Collapse
Affiliation(s)
- Biao Han
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania 19104, United States
| | - Hadi T. Nia
- Department of Radiation Oncology, Massachusetts General Hospital Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Chao Wang
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania 19104, United States
| | - Prashant Chandrasekaran
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania 19104, United States
| | - Qing Li
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania 19104, United States
| | - Daphney R. Chery
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania 19104, United States
| | - Hao Li
- College of Architecture and the Built Environment, Philadelphia University, Philadelphia, Pennsylvania 19144, United States
| | - Alan J. Grodzinsky
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Lin Han
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
71
|
Fernández-Avilés F, Sanz-Ruiz R, Climent AM, Badimon L, Bolli R, Charron D, Fuster V, Janssens S, Kastrup J, Kim HS, Lüscher TF, Martin JF, Menasché P, Simari RD, Stone GW, Terzic A, Willerson JT, Wu JC. Global position paper on cardiovascular regenerative medicine. Eur Heart J 2017; 38:2532-2546. [PMID: 28575280 PMCID: PMC5837698 DOI: 10.1093/eurheartj/ehx248] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 04/13/2017] [Accepted: 04/20/2017] [Indexed: 12/11/2022] Open
Affiliation(s)
- Francisco Fernández-Avilés
- Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain
- CIBERCV, ISCIII, Madrid, Spain
| | - Ricardo Sanz-Ruiz
- Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain
- CIBERCV, ISCIII, Madrid, Spain
| | - Andreu M Climent
- Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain
- CIBERCV, ISCIII, Madrid, Spain
| | - Lina Badimon
- CIBERCV, ISCIII, Madrid, Spain
- Cardiovascular Research Center (CSIC-ICCC), Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain
| | - Roberto Bolli
- Institute of Molecular Cardiology, Diabetes and Obesity Center, University of Louisville School of Medicine, Louisville, Kentucky
| | - Dominique Charron
- LabEx TRANSPLANTEX; HLA & Médecine "Jean Dausset" Laboratory Network, Hôpital Saint-Louis AP-HP, Université Paris Diderot, 75013, France
| | - Valentin Fuster
- CIBERCV, ISCIII, Madrid, Spain
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of medicine at Mount Sinai, New York, NY, USA
| | - Stefan Janssens
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Jens Kastrup
- Department of Cardiology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Hyo-Soo Kim
- National Research Laboratory for Stem Cell Niche, Center for Medical Innovation, Seoul National University Hospital, Seoul, Korea; Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea
| | - Thomas F Lüscher
- Department of Cardiology, University Heart Center Zurich, Zurich, Switzerland; Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
| | | | - Philippe Menasché
- Department of Cardiovascular Surgery Hôpital Européen Georges Pompidou; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Robert D Simari
- School of Medicine, University of Kansas, 3901 Rainbow Boulevard, Kansas City, KS, USA
| | - Gregg W Stone
- Center for Clinical Trials, Cardiovascular Research Foundation, New York, New York; Center for Clinical Trials, NewYork-Presbyterian Hospital, Columbia University Medical Center, New York, NY, USA
| | - Andre Terzic
- Center for Regenerative Medicine, Department of Cardiovascular Diseases, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, NY, USA
| | - James T Willerson
- Department of Regenerative Medicine Research, Texas Heart Institute, Houston, TX, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Division of Cardiovascular Medicine, Department of Medicine and Department of Radiology, Stanford University School of Medicine, CA, USA
| |
Collapse
|
72
|
Rotini A, Martínez-Sarrà E, Pozzo E, Sampaolesi M. Interactions between microRNAs and long non-coding RNAs in cardiac development and repair. Pharmacol Res 2017. [PMID: 28629929 DOI: 10.1016/j.phrs.2017.05.029] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Non-coding RNAs (ncRNAs) are emerging players in muscle regulation. Based on their length and differences in molecular structure, ncRNAs are subdivided into several categories including small interfering RNAs, stable non-coding RNAs, microRNAs (miRs), long non-coding RNAs (lncRNAs), and circular RNAs. miRs and lncRNAs are able to post-transcriptionally regulate many genes and bring into play several traits simultaneously due to a myriad of different targets. Recent studies have emphasized their importance in cardiac regeneration and repair. As their altered expression affects cardiac function, miRs and lncRNAs could be potential targets for therapeutic intervention. In this context, miR- and lncRNA-based gene therapies are an interesting field for harnessing the complexity of ncRNA-based therapeutic approaches in cardiac diseases. In this review we will focus on lncRNA- and miR-driven regulations of cardiac development and repair. Finally, we will summarize miRs and lncRNAs as promising candidates for the treatment of heart diseases.
Collapse
Affiliation(s)
- Alessio Rotini
- Translational Cardiomyology, Stem Cell Research Institute, Stem Cell Biology and Embryology Unit, Department of Development and Regeneration, KU Leuven, Herestraat 49 B-3000 Leuven, Belgium; Department of Neuroscience, Imaging and Clinical Sciences, University "G. d'Annunzio" Chieti-Pescara, Chieti, Italy; Interuniversity Institute of Myology, Italy
| | - Ester Martínez-Sarrà
- Translational Cardiomyology, Stem Cell Research Institute, Stem Cell Biology and Embryology Unit, Department of Development and Regeneration, KU Leuven, Herestraat 49 B-3000 Leuven, Belgium; Regenerative Medicine Research Institute, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Enrico Pozzo
- Translational Cardiomyology, Stem Cell Research Institute, Stem Cell Biology and Embryology Unit, Department of Development and Regeneration, KU Leuven, Herestraat 49 B-3000 Leuven, Belgium
| | - Maurilio Sampaolesi
- Translational Cardiomyology, Stem Cell Research Institute, Stem Cell Biology and Embryology Unit, Department of Development and Regeneration, KU Leuven, Herestraat 49 B-3000 Leuven, Belgium; Human Anatomy Unit, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Via Forlanini 8, 27100 Pavia, Italy.
| |
Collapse
|
73
|
Tan Y, Han P, Gu Q, Chen G, Wang L, Ma R, Wu J, Feng C, Zhang Y, Wang L, Hu B, Li W, Hao J, Zhou Q. Generation of clinical-grade functional cardiomyocytes from human embryonic stem cells in chemically defined conditions. J Tissue Eng Regen Med 2017; 12:153-163. [PMID: 27943600 DOI: 10.1002/term.2381] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 11/21/2016] [Accepted: 12/06/2016] [Indexed: 01/26/2023]
Abstract
A highly efficient cardiac differentiation from human pluripotent stem cells (hPSCs) is achievable using existing methods, especially with the standard B27 induction system. However, bovine serum albumin (BSA), one of the essential ingredients in B27, may pose significant complications for clinical studies owing to its animal origin and potential risks of virus contamination. Furthermore, the high cost of the B27 induction system also limits the applications of hPSCs-derived cardiomyocytes. Here, a BSA-free and chemically defined medium has been developed for differentiating hPSCs to clinical-grade cardiomyocytes, which generated over 80% cardiac troponin T (cTNT)-positive cardiomyocytes with high yield. When engrafting the cardiomyocytes into the hearts of myocardial infarction model rats, the rats survived with significantly improved heart functions in Δ ejection fraction and Δ fractional shortening. Importantly, the human embryonic stem cell (hESC) line (Q-CTS-hESC-2) chosen for differentiation was of a clinical-grade maintained in defined xeno-free conditions. Compliant with the biological safety requirements, the Q-CTS-hESC-2-derived cardiomyocytes have passed the sterility and pathogen criteria tests for clinical applications. This study reports, for the first time, the generation of clinical-grade and functional cardiomyocytes from hPSCs where BSA-free and chemically defined conditions were maintained throughout the whole process. This provides the possibility of future therapeutic use of clinical-grade hPSCs-derived cardiomyocytes in treating heart diseases. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Yuanqing Tan
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China.,Beijing Stem Cell Bank, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100192, China
| | - Pengcheng Han
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Qi Gu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,Beijing Stem Cell Bank, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100192, China
| | - Gang Chen
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,Beijing Stem Cell Bank, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100192, China
| | - Lei Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China.,Beijing Stem Cell Bank, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100192, China
| | - Ruoyu Ma
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,Beijing Stem Cell Bank, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100192, China
| | - Jun Wu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,Beijing Stem Cell Bank, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100192, China
| | - Chunjing Feng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China.,Beijing Stem Cell Bank, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100192, China
| | - Ying Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Liu Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Baoyang Hu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jie Hao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,Beijing Stem Cell Bank, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100192, China
| | - Qi Zhou
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China.,Beijing Stem Cell Bank, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100192, China
| |
Collapse
|
74
|
Skelton RJP, Kamp TJ, Elliott DA, Ardehali R. Biomarkers of Human Pluripotent Stem Cell-Derived Cardiac Lineages. Trends Mol Med 2017; 23:651-668. [PMID: 28576602 DOI: 10.1016/j.molmed.2017.05.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 04/24/2017] [Accepted: 05/04/2017] [Indexed: 02/07/2023]
Abstract
Human pluripotent stem cells (hPSCs) offer a practical source for the de novo generation of cardiac tissues and a unique opportunity to investigate cardiovascular lineage commitment. Numerous strategies have focused on the in vitro production of cardiomyocytes, smooth muscle, and endothelium from hPSCs. However, these differentiation protocols often yield undesired cell types. Thus, establishing a set of stage-specific markers for pure cardiac subpopulations will assist in defining the hierarchy of cardiac differentiation, aid in the development of cellular therapy, and facilitate drug screening and disease modeling. The recent characterization of many such markers is enabling the isolation of major cardiac lineages and subpopulations from differentiating hPSCs. We provide here a comprehensive review detailing the suite of biomarkers used to differentiate cardiac lineages from mixed hPSC-derived populations.
Collapse
Affiliation(s)
- Rhys J P Skelton
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; Eli and Edythe Broad Stem Cell Research Center, University of California, Los Angeles, CA 90095, USA
| | - Timothy J Kamp
- Department of Medicine, Division of Cardiovascular Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - David A Elliott
- Murdoch Childrens Research Institute, The Royal Children's Hospital, Parkville, Victoria 3052, Australia
| | - Reza Ardehali
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; Eli and Edythe Broad Stem Cell Research Center, University of California, Los Angeles, CA 90095, USA.
| |
Collapse
|
75
|
Lu J, Kaestle K, Huang J, Liu Q, Zhang P, Gao L, Gardiner J, Thissen H, Yang HT. Interactions of human embryonic stem cell-derived cardiovascular progenitor cells with immobilized extracellular matrix proteins. J Biomed Mater Res A 2017; 105:1094-1104. [PMID: 28085215 DOI: 10.1002/jbm.a.36005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 12/19/2016] [Accepted: 01/10/2017] [Indexed: 11/11/2022]
Abstract
Human embryonic stem cell-derived cardiovascular progenitor cells (hESC-CVPCs) hold great promise for cell-based therapies of heart diseases. However, little is known about their niche microenvironment and in particular the required extracellular matrix (ECM) components. Here we screened combinations of surface-immobilized ECM proteins to identify substrates that support the attachment and survival of hESC-CVPCs. Covalent immobilization of ECM proteins laminin (Lm), fibronectin (Fn), collagen I (CI), collagen III (CIII), and collagen IV (CIV) in multiple combinations and concentrations was achieved by reductive amination on transparent acetaldehyde plasma polymer (AAPP) interlayer coatings. We identified that CI, CIII, CIV, and Fn and their combinations were important for hESC-CVPC attachment and survival, while Lm was dispensable. Moreover, for coatings displaying single ECM proteins, CI and CIII performed better than CIV and Fn, while coatings displaying the combined ECM proteins CIII + CIV and Fn + CIII + CIV at 100 µg/mL were comparable to Matrigel in regard to supporting hESC-CVPC attachment and viability. Our results identify ECM proteins required for hESC-CVPCs and demonstrate that coatings displaying multiple immobilized ECM proteins offer a suitable microenvironment for the attachment and survival of hESC-CVPCs. This knowledge contributes to the development of approaches for maintaining hESC-CVPCs and therefore to advances in cardiovascular regeneration. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 1094-1104, 2017.
Collapse
Affiliation(s)
- Jizhen Lu
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences & Shanghai Jiao Tong University School of Medicine, Biological Research Building A, 320 Yueyang Road, Shanghai 200031, China.,Second Affiliated Hospital, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009, China
| | - Katrin Kaestle
- CSIRO Manufacturing, Bayview Avenue, Clayton, Victoria, 3168, Australia
| | - Jijun Huang
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences & Shanghai Jiao Tong University School of Medicine, Biological Research Building A, 320 Yueyang Road, Shanghai 200031, China.,Second Affiliated Hospital, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009, China
| | - Qiao Liu
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences & Shanghai Jiao Tong University School of Medicine, Biological Research Building A, 320 Yueyang Road, Shanghai 200031, China.,Second Affiliated Hospital, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009, China
| | - Peng Zhang
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences & Shanghai Jiao Tong University School of Medicine, Biological Research Building A, 320 Yueyang Road, Shanghai 200031, China.,Second Affiliated Hospital, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009, China
| | - Ling Gao
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences & Shanghai Jiao Tong University School of Medicine, Biological Research Building A, 320 Yueyang Road, Shanghai 200031, China.,Second Affiliated Hospital, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009, China
| | - James Gardiner
- CSIRO Manufacturing, Bayview Avenue, Clayton, Victoria, 3168, Australia
| | - Helmut Thissen
- CSIRO Manufacturing, Bayview Avenue, Clayton, Victoria, 3168, Australia
| | - Huang-Tian Yang
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences & Shanghai Jiao Tong University School of Medicine, Biological Research Building A, 320 Yueyang Road, Shanghai 200031, China.,Second Affiliated Hospital, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009, China
| |
Collapse
|
76
|
Stoltz JF, Bensoussan D, De Isla N, Zhang L, Han Z, Magdalou J, Huselstein C, Ye J, Leballe B, Decot V, Reppel L. Stem cells and vascular regenerative medicine: A mini review. Clin Hemorheol Microcirc 2017; 64:613-633. [DOI: 10.3233/ch-168036] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- J.-F. Stoltz
- CNRS, UMR 7365, Biopole, Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
- CNRS – GDRI France-Chine « Stem cells and Regenerative medicine », Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
- CHRU de Nancy, Unité de Thérapie Cellulaire et Tissulaire (UTCT) (FR CNRS-INSERM-UHP-CHU), Vandoeuvre-Lès-Nancy, France
| | - D. Bensoussan
- CNRS, UMR 7365, Biopole, Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
- CNRS – GDRI France-Chine « Stem cells and Regenerative medicine », Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
- CHRU de Nancy, Unité de Thérapie Cellulaire et Tissulaire (UTCT) (FR CNRS-INSERM-UHP-CHU), Vandoeuvre-Lès-Nancy, France
| | - N. De Isla
- CNRS, UMR 7365, Biopole, Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
- CNRS – GDRI France-Chine « Stem cells and Regenerative medicine », Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
| | - L. Zhang
- CNRS – GDRI France-Chine « Stem cells and Regenerative medicine », Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
- Centre de Recherche, Calmette Hospital, Kunming, China
| | - Z. Han
- CNRS – GDRI France-Chine « Stem cells and Regenerative medicine », Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
- Centre de Recvherche sur les cellules souches, Beijing et Tianjin, China
| | - J. Magdalou
- CNRS, UMR 7365, Biopole, Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
- CNRS – GDRI France-Chine « Stem cells and Regenerative medicine », Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
| | - C. Huselstein
- CNRS, UMR 7365, Biopole, Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
- CNRS – GDRI France-Chine « Stem cells and Regenerative medicine », Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
| | - J.S. Ye
- CNRS – GDRI France-Chine « Stem cells and Regenerative medicine », Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
- Centre de Recherche, Calmette Hospital, Kunming, China
| | | | - V. Decot
- CNRS, UMR 7365, Biopole, Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
- CNRS – GDRI France-Chine « Stem cells and Regenerative medicine », Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
- CHRU de Nancy, Unité de Thérapie Cellulaire et Tissulaire (UTCT) (FR CNRS-INSERM-UHP-CHU), Vandoeuvre-Lès-Nancy, France
| | - L. Reppel
- CNRS, UMR 7365, Biopole, Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
- CNRS – GDRI France-Chine « Stem cells and Regenerative medicine », Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
- CHRU de Nancy, Unité de Thérapie Cellulaire et Tissulaire (UTCT) (FR CNRS-INSERM-UHP-CHU), Vandoeuvre-Lès-Nancy, France
| |
Collapse
|
77
|
Jung JH, Fu X, Yang PC. Exosomes Generated From iPSC-Derivatives: New Direction for Stem Cell Therapy in Human Heart Diseases. Circ Res 2017; 120:407-417. [PMID: 28104773 PMCID: PMC5260934 DOI: 10.1161/circresaha.116.309307] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 12/07/2016] [Accepted: 12/13/2016] [Indexed: 12/15/2022]
Abstract
Cardiovascular disease (CVD) is the leading cause of death in modern society. The adult heart innately lacks the capacity to repair and regenerate the damaged myocardium from ischemic injury. Limited understanding of cardiac tissue repair process hampers the development of effective therapeutic solutions to treat CVD such as ischemic cardiomyopathy. In recent years, rapid emergence of induced pluripotent stem cells (iPSC) and iPSC-derived cardiomyocytes presents a valuable opportunity to replenish the functional cells to the heart. The therapeutic effects of iPSC-derived cells have been investigated in many preclinical studies. However, the underlying mechanisms of iPSC-derived cell therapy are still unclear, and limited engraftment of iPSC-derived cardiomyocytes is well known. One facet of their mechanism is the paracrine effect of the transplanted cells. Microvesicles such as exosomes secreted from the iPSC-derived cardiomyocytes exert protective effects by transferring the endogenous molecules to salvage the injured neighboring cells by regulating apoptosis, inflammation, fibrosis, and angiogenesis. In this review, we will focus on the current advances in the exosomes from iPSC derivatives and discuss their therapeutic potential in the treatment of CVD.
Collapse
Affiliation(s)
- Ji-Hye Jung
- From the Stanford Cardiovascular Institute, Division of Cardiovascular Medicine, Stanford University School of Medicine, CA
| | - Xuebin Fu
- From the Stanford Cardiovascular Institute, Division of Cardiovascular Medicine, Stanford University School of Medicine, CA
| | - Phillip C Yang
- From the Stanford Cardiovascular Institute, Division of Cardiovascular Medicine, Stanford University School of Medicine, CA.
| |
Collapse
|
78
|
Zhang Y, Cao N, Huang Y, Spencer CI, Fu JD, Yu C, Liu K, Nie B, Xu T, Li K, Xu S, Bruneau BG, Srivastava D, Ding S. Expandable Cardiovascular Progenitor Cells Reprogrammed from Fibroblasts. Cell Stem Cell 2016; 18:368-81. [PMID: 26942852 DOI: 10.1016/j.stem.2016.02.001] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Revised: 12/11/2015] [Accepted: 02/09/2016] [Indexed: 12/31/2022]
Abstract
Stem cell-based approaches to cardiac regeneration are increasingly viable strategies for treating heart failure. Generating abundant and functional autologous cells for transplantation in such a setting, however, remains a significant challenge. Here, we isolated a cell population with extensive proliferation capacity and restricted cardiovascular differentiation potentials during cardiac transdifferentiation of mouse fibroblasts. These induced expandable cardiovascular progenitor cells (ieCPCs) proliferated extensively for more than 18 passages in chemically defined conditions, with 10(5) starting fibroblasts robustly producing 10(16) ieCPCs. ieCPCs expressed cardiac signature genes and readily differentiated into functional cardiomyocytes (CMs), endothelial cells (ECs), and smooth muscle cells (SMCs) in vitro, even after long-term expansion. When transplanted into mouse hearts following myocardial infarction, ieCPCs spontaneously differentiated into CMs, ECs, and SMCs and improved cardiac function for up to 12 weeks after transplantation. Thus, ieCPCs are a powerful system to study cardiovascular specification and provide strategies for regenerative medicine in the heart.
Collapse
Affiliation(s)
- Yu Zhang
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA; Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Nan Cao
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA; Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Yu Huang
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA
| | - C Ian Spencer
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA
| | - Ji-Dong Fu
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA; Department of Medicine, Heart and Vascular Research Center, MetroHealth Campus, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Chen Yu
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA; Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Kai Liu
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA; Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Baoming Nie
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA; Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Tao Xu
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA; Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Ke Li
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA; Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Shaohua Xu
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA; Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Benoit G Bruneau
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA; Roddenberry Center for Stem Cell Biology and Medicine, Gladstone Institutes, San Francisco, CA 94158, USA; Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94143, USA; Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Deepak Srivastava
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA; Roddenberry Center for Stem Cell Biology and Medicine, Gladstone Institutes, San Francisco, CA 94158, USA; Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Sheng Ding
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA; Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
79
|
Abstract
Pluripotent stem cells (PSCs) can differentiate into virtually any cell type in the body, making them attractive for both regenerative medicine and drug discovery. Over the past 10 years, technological advances and innovative platforms have yielded first-in-man PSC-based clinical trials and opened up new approaches for disease modeling and drug development. Induced PSCs have become the foremost alternative to embryonic stem cells and accelerated the development of disease-in-a-dish models. Over the years and with each new discovery, PSCs have proven to be extremely versatile. This review article highlights key advancements in PSC research, from 2006 to 2016, and how they will guide the direction of the field over the next decade.
Collapse
Affiliation(s)
- Erin A Kimbrel
- Astellas Institute for Regenerative Medicine, 33 Locke Drive, Marlborough, MA 01752, USA
| | - Robert Lanza
- Astellas Institute for Regenerative Medicine, 33 Locke Drive, Marlborough, MA 01752, USA
| |
Collapse
|
80
|
Pacheco-Leyva I, Matias AC, Oliveira DV, Santos JMA, Nascimento R, Guerreiro E, Michell AC, van De Vrugt AM, Machado-Oliveira G, Ferreira G, Domian I, Bragança J. CITED2 Cooperates with ISL1 and Promotes Cardiac Differentiation of Mouse Embryonic Stem Cells. Stem Cell Reports 2016; 7:1037-1049. [PMID: 27818139 PMCID: PMC5161512 DOI: 10.1016/j.stemcr.2016.10.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 10/05/2016] [Accepted: 10/06/2016] [Indexed: 01/07/2023] Open
Abstract
The transcriptional regulator CITED2 is essential for heart development. Here, we investigated the role of CITED2 in the specification of cardiac cell fate from mouse embryonic stem cells (ESC). The overexpression of CITED2 in undifferentiated ESC was sufficient to promote cardiac cell emergence upon differentiation. Conversely, the depletion of Cited2 at the onset of differentiation resulted in a decline of ESC ability to generate cardiac cells. Moreover, loss of Cited2 expression impairs the expression of early mesoderm markers and cardiogenic transcription factors (Isl1, Gata4, Tbx5). The cardiogenic defects in Cited2-depleted cells were rescued by treatment with recombinant CITED2 protein. We showed that Cited2 expression is enriched in cardiac progenitors either derived from ESC or mouse embryonic hearts. Finally, we demonstrated that CITED2 and ISL1 proteins interact physically and cooperate to promote ESC differentiation toward cardiomyocytes. Collectively, our results show that Cited2 plays a pivotal role in cardiac commitment of ESC. Overexpression of CITED2 in ESC promotes cardiogenesis upon differentiation Cited2 depletion reduces ESC ability to generate cardiac cells Cited2 expression is enriched in cardiac progenitors CITED2 and ISL1 cooperate to promote ESC differentiation toward cardiomyocytes
Collapse
Affiliation(s)
- Ivette Pacheco-Leyva
- Regenerative Medicine Program, Department of Biomedical Sciences and Medicine, University of Algarve, 8005-139 Faro, Portugal; Centre for Biomedical Research - CBMR, University of Algarve, Campus of Gambelas, Building 8, Room 2.22, 8005-139 Faro, Portugal
| | - Ana Catarina Matias
- Regenerative Medicine Program, Department of Biomedical Sciences and Medicine, University of Algarve, 8005-139 Faro, Portugal; Centre for Biomedical Research - CBMR, University of Algarve, Campus of Gambelas, Building 8, Room 2.22, 8005-139 Faro, Portugal
| | - Daniel V Oliveira
- Regenerative Medicine Program, Department of Biomedical Sciences and Medicine, University of Algarve, 8005-139 Faro, Portugal; Centre for Biomedical Research - CBMR, University of Algarve, Campus of Gambelas, Building 8, Room 2.22, 8005-139 Faro, Portugal
| | - João M A Santos
- Regenerative Medicine Program, Department of Biomedical Sciences and Medicine, University of Algarve, 8005-139 Faro, Portugal; Centre for Biomedical Research - CBMR, University of Algarve, Campus of Gambelas, Building 8, Room 2.22, 8005-139 Faro, Portugal
| | - Rita Nascimento
- Regenerative Medicine Program, Department of Biomedical Sciences and Medicine, University of Algarve, 8005-139 Faro, Portugal; Centre for Biomedical Research - CBMR, University of Algarve, Campus of Gambelas, Building 8, Room 2.22, 8005-139 Faro, Portugal
| | - Eduarda Guerreiro
- Regenerative Medicine Program, Department of Biomedical Sciences and Medicine, University of Algarve, 8005-139 Faro, Portugal; Centre for Biomedical Research - CBMR, University of Algarve, Campus of Gambelas, Building 8, Room 2.22, 8005-139 Faro, Portugal
| | - Anna C Michell
- Division of Cardiovascular Medicine, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Annebel M van De Vrugt
- Cardiovascular Research Center, Massachusetts General Hospital, Charles River Plaza/CPZN 3200, 185 Cambridge Street, Boston, MA 02114-2790, USA; Harvard Medical School, 250 Longwood Avenue, Boston, MA 02115, USA; Department of Cardiology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands
| | - Gisela Machado-Oliveira
- Regenerative Medicine Program, Department of Biomedical Sciences and Medicine, University of Algarve, 8005-139 Faro, Portugal; Centre for Biomedical Research - CBMR, University of Algarve, Campus of Gambelas, Building 8, Room 2.22, 8005-139 Faro, Portugal
| | - Guilherme Ferreira
- DSM Biotechnology Center, Alexander Fleminglaan 1, 2613 AX Delft, the Netherlands
| | - Ibrahim Domian
- Cardiovascular Research Center, Massachusetts General Hospital, Charles River Plaza/CPZN 3200, 185 Cambridge Street, Boston, MA 02114-2790, USA; Harvard Medical School, 250 Longwood Avenue, Boston, MA 02115, USA
| | - José Bragança
- Regenerative Medicine Program, Department of Biomedical Sciences and Medicine, University of Algarve, 8005-139 Faro, Portugal; Centre for Biomedical Research - CBMR, University of Algarve, Campus of Gambelas, Building 8, Room 2.22, 8005-139 Faro, Portugal; ABC - Algarve Biomedical Centre, 8005-139 Faro, Portugal.
| |
Collapse
|
81
|
Miyagawa S, Fukushima S, Imanishi Y, Kawamura T, Mochizuki-Oda N, Masuda S, Sawa Y. Building A New Treatment For Heart Failure-Transplantation of Induced Pluripotent Stem Cell-derived Cells into the Heart. Curr Gene Ther 2016; 16:5-13. [PMID: 26785736 PMCID: PMC4997929 DOI: 10.2174/1566523216666160119094143] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 01/07/2016] [Accepted: 01/08/2016] [Indexed: 02/08/2023]
Abstract
Advanced cardiac failure is a progressive intractable disease and is the main cause of mortality and morbidity worldwide. Since this pathology is represented by a definite decrease in cardiomyocyte number, supplementation of functional cardiomyocytes into the heart would hypothetically be an ideal therapeutic option. Recently, unlimited in vitro production of human functional cardiomyocytes was established by using induced pluripotent stem cell (iPSC) technology, which avoids the use of human embryos. A number of basic studies including ours have shown that transplantation of iPSC-derived cardiomyocytes (iPSC-CMs) into the damaged heart leads to recovery of cardiac function, thereby establishing “proof-of-concept” of this iPSC-transplantation therapy. However, considering clinical application of this therapy, its feasibility, safety, and therapeutic efficacy need to be further investigated in the pre-clinical stage. This review summarizes up-to-date important topics related to safety and efficacy of iPSC-CMs transplantation therapy for cardiac disease and discusses the prospects for this treatment in clinical studies.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yoshiki Sawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
82
|
Skelton RJP, Brady B, Khoja S, Sahoo D, Engel J, Arasaratnam D, Saleh KK, Abilez OJ, Zhao P, Stanley EG, Elefanty AG, Kwon M, Elliott DA, Ardehali R. CD13 and ROR2 Permit Isolation of Highly Enriched Cardiac Mesoderm from Differentiating Human Embryonic Stem Cells. Stem Cell Reports 2016; 6:95-108. [PMID: 26771355 PMCID: PMC4720015 DOI: 10.1016/j.stemcr.2015.11.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 11/14/2015] [Accepted: 11/18/2015] [Indexed: 01/17/2023] Open
Abstract
The generation of tissue-specific cell types from human embryonic stem cells (hESCs) is critical for the development of future stem cell-based regenerative therapies. Here, we identify CD13 and ROR2 as cell-surface markers capable of selecting early cardiac mesoderm emerging during hESC differentiation. We demonstrate that the CD13+/ROR2+ population encompasses pre-cardiac mesoderm, which efficiently differentiates to all major cardiovascular lineages. We determined the engraftment potential of CD13+/ROR2+ in small (murine) and large (porcine) animal models, and demonstrated that CD13+/ROR2+ progenitors have the capacity to differentiate toward cardiomyocytes, fibroblasts, smooth muscle, and endothelial cells in vivo. Collectively, our data show that CD13 and ROR2 identify a cardiac lineage precursor pool that is capable of successful engraftment into the porcine heart. These markers represent valuable tools for further dissection of early human cardiac differentiation, and will enable a detailed assessment of human pluripotent stem cell-derived cardiac lineage cells for potential clinical applications. CD13 and ROR2 separate hESC-derived MIXL1+ mesoderm from MIXL1+ endoderm CD13 and ROR2 select for a population of highly enriched pre-cardiac mesoderm CD13+/ROR2+ cells derived from hESCs engraft into porcine, but not murine hearts CD13+/ROR2+ cells differentiate to all major cardiac lineages in the pig heart
Collapse
Affiliation(s)
- Rhys J P Skelton
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine at UCLA, 675 Charles E Young Drive South, Room 3645, Los Angeles, CA 90095, USA; Eli and Edythe Broad Stem Cell Research Center, University of California, Los Angeles, CA 90095, USA; Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, VIC 3052, Australia
| | - Bevin Brady
- Bio-X Program, Cardiovascular Medicine, Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Suhail Khoja
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine at UCLA, 675 Charles E Young Drive South, Room 3645, Los Angeles, CA 90095, USA; Eli and Edythe Broad Stem Cell Research Center, University of California, Los Angeles, CA 90095, USA
| | - Debashis Sahoo
- Bio-X Program, Cardiovascular Medicine, Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - James Engel
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine at UCLA, 675 Charles E Young Drive South, Room 3645, Los Angeles, CA 90095, USA; Eli and Edythe Broad Stem Cell Research Center, University of California, Los Angeles, CA 90095, USA
| | - Deevina Arasaratnam
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, VIC 3052, Australia
| | - Kholoud K Saleh
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine at UCLA, 675 Charles E Young Drive South, Room 3645, Los Angeles, CA 90095, USA; Eli and Edythe Broad Stem Cell Research Center, University of California, Los Angeles, CA 90095, USA
| | - Oscar J Abilez
- Bio-X Program, Cardiovascular Medicine, Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Peng Zhao
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine at UCLA, 675 Charles E Young Drive South, Room 3645, Los Angeles, CA 90095, USA; Eli and Edythe Broad Stem Cell Research Center, University of California, Los Angeles, CA 90095, USA
| | - Edouard G Stanley
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, VIC 3052, Australia
| | - Andrew G Elefanty
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, VIC 3052, Australia
| | - Murray Kwon
- Division of Cardiothoracic Surgery, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - David A Elliott
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, VIC 3052, Australia
| | - Reza Ardehali
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine at UCLA, 675 Charles E Young Drive South, Room 3645, Los Angeles, CA 90095, USA; Eli and Edythe Broad Stem Cell Research Center, University of California, Los Angeles, CA 90095, USA.
| |
Collapse
|
83
|
Choudhury GR, Kim J, Frost PA, Bastarrachea RA, Daadi MM. Nonhuman primate model in clinical modeling of diseases for stem cell therapy. Brain Circ 2016; 2:141-145. [PMID: 30276291 PMCID: PMC6126269 DOI: 10.4103/2394-8108.192524] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Revised: 08/16/2016] [Accepted: 09/06/2016] [Indexed: 01/11/2023] Open
Abstract
Nonhuman primates (NHPs) are alike humans in size, behavior, physiology, biochemistry, and immunology. Given close similarities to humans, the NHP model offers exceptional opportunities to understand the biological mechanisms and translational applications with direct relevance to human conditions. Here, we evaluate the opportunities and limitations of NHPs as animal models for translational regenerative medicine. NHP models of human disease propose exceptional opportunities to advance stem cell-based therapy by addressing pertinent translational concerns related to this research. Nonetheless, the value of these primates must be carefully assessed, taking into account the expense of specialized equipment and requirement of highly specialized staff. Well-designed initial fundamental studies in small animal models are essential before translating research into NHP models and eventually into human trials. In addition, we suggest that applying a directed and collaborative approach, as seen in the evolution of stroke NHP models, will greatly benefit the translation of stem cell therapy in other NHP disease models.
Collapse
Affiliation(s)
- Gourav R Choudhury
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Jeffrey Kim
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Patrice A Frost
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Raul A Bastarrachea
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Marcel M Daadi
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas, USA.,Department of Cellular and Structural Biology, UT Health Science Center, San Antonio, Texas, USA.,Department of Radiology, Medical School, UT Health Science Center, San Antonio, Texas, USA
| |
Collapse
|
84
|
Cardiomyogenesis of periodontal ligament-derived stem cells by dynamic tensile strain. Cell Tissue Res 2016; 367:229-241. [DOI: 10.1007/s00441-016-2503-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 09/06/2016] [Indexed: 12/25/2022]
|
85
|
Huethorst E, Hortigon M, Zamora-Rodriguez V, Reynolds PM, Burton F, Smith G, Gadegaard N. Enhanced Human-Induced Pluripotent Stem Cell Derived Cardiomyocyte Maturation Using a Dual Microgradient Substrate. ACS Biomater Sci Eng 2016; 2:2231-2239. [PMID: 27990488 PMCID: PMC5155309 DOI: 10.1021/acsbiomaterials.6b00426] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 10/17/2016] [Indexed: 12/29/2022]
Abstract
Human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM) raise many possibilities for cardiac research but they exhibit an immature phenotype, which influences experimental outcomes. The aim of our research is to investigate the effects of a topographical gradient substrate on the morphology and function of commercially available hiPSC-CM. The lateral dimensions the microgrooves on the substrate varied from 8 to 100 μm space between the 8 μm grooves on one axis and from ∼5 nm to ∼1 μm in depth on the other axis. Cells were seeded homogeneously across the substrate and according to the manufacturers protocols. At days 4 and 10, measures of eccentricity, elongation, orientation, sarcomere length (SL), and contractility of the hiPSC-CM were taken. Only the deepest and widest region (8-30 μm wide and 0.85-1 μm deep) showed a significantly higher percentage of hiPSC-CM with an increased eccentricity (31.3 ± 6.4%), elongation (10.4 ± 4.3%), and orientation (<10°) (32.1 ± 2.7%) when compared with the control (flat substrate) (15.8 ± 5.0%, 3.4 ± 2.7%, and 10.6 ± 1.1%, respectively). Additionally, during stimulus-induced contraction, the relaxation phase of the twitch was prolonged (400 ms) compared to nonelongated cells (200 ms). These findings support the potential use of dual microgradient substrates to investigate substrate topographies that stimulate migration and/or maturation of hiPSC-CM.
Collapse
Affiliation(s)
- E Huethorst
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom; Division of Biomedical Engineering, School of Engineering, University of Glasgow, Glasgow G12 8LT, United Kingdom
| | - M Hortigon
- Institute of Cardiovascular and Medical Sciences, University of Glasgow , Glasgow G12 8QQ, United Kingdom
| | - V Zamora-Rodriguez
- Institute of Cardiovascular and Medical Sciences, University of Glasgow , Glasgow G12 8QQ, United Kingdom
| | - P M Reynolds
- Division of Biomedical Engineering, School of Engineering, University of Glasgow , Glasgow G12 8LT, United Kingdom
| | - F Burton
- Institute of Cardiovascular and Medical Sciences, University of Glasgow , Glasgow G12 8QQ, United Kingdom
| | - G Smith
- Institute of Cardiovascular and Medical Sciences, University of Glasgow , Glasgow G12 8QQ, United Kingdom
| | - N Gadegaard
- Division of Biomedical Engineering, School of Engineering, University of Glasgow , Glasgow G12 8LT, United Kingdom
| |
Collapse
|
86
|
Burridge PW, Sharma A, Wu JC. Genetic and Epigenetic Regulation of Human Cardiac Reprogramming and Differentiation in Regenerative Medicine. Annu Rev Genet 2016; 49:461-84. [PMID: 26631515 DOI: 10.1146/annurev-genet-112414-054911] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Regeneration or replacement of lost cardiomyocytes within the heart has the potential to revolutionize cardiovascular medicine. Numerous methodologies have been used to achieve this aim, including the engraftment of bone marrow- and heart-derived cells as well as the identification of modulators of adult cardiomyocyte proliferation. Recently, the conversion of human somatic cells into induced pluripotent stem cells and induced cardiomyocyte-like cells has transformed potential approaches toward this goal, and the engraftment of cardiac progenitors derived from human embryonic stem cells into patients is now feasible. Here we review recent advances in our understanding of the genetic and epigenetic control of human cardiogenesis, cardiac differentiation, and the induced reprogramming of somatic cells to cardiomyocytes. We also cover genetic programs for inducing the proliferation of endogenous cardiomyocytes and discuss the genetic state of cells used in cardiac regenerative medicine.
Collapse
Affiliation(s)
- Paul W Burridge
- Stanford Cardiovascular Institute.,Institute for Stem Cell Biology and Regenerative Medicine.,Department of Medicine, Division of Cardiology, Stanford University School of Medicine, Stanford, California 94305.,Department of Pharmacology.,Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611; ,
| | - Arun Sharma
- Stanford Cardiovascular Institute.,Institute for Stem Cell Biology and Regenerative Medicine.,Department of Medicine, Division of Cardiology, Stanford University School of Medicine, Stanford, California 94305
| | - Joseph C Wu
- Stanford Cardiovascular Institute.,Institute for Stem Cell Biology and Regenerative Medicine.,Department of Medicine, Division of Cardiology, Stanford University School of Medicine, Stanford, California 94305
| |
Collapse
|
87
|
Liu Y, Chen L, Diaz AD, Benham A, Xu X, Wijaya CS, Fa'ak F, Luo W, Soibam B, Azares A, Yu W, Lyu Q, Stewart MD, Gunaratne P, Cooney A, McConnell BK, Schwartz RJ. Mesp1 Marked Cardiac Progenitor Cells Repair Infarcted Mouse Hearts. Sci Rep 2016; 6:31457. [PMID: 27538477 PMCID: PMC4990963 DOI: 10.1038/srep31457] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 07/18/2016] [Indexed: 12/15/2022] Open
Abstract
Mesp1 directs multipotential cardiovascular cell fates, even though it's transiently induced prior to the appearance of the cardiac progenitor program. Tracing Mesp1-expressing cells and their progeny allows isolation and characterization of the earliest cardiovascular progenitor cells. Studying the biology of Mesp1-CPCs in cell culture and ischemic disease models is an important initial step toward using them for heart disease treatment. Because of Mesp1's transitory nature, Mesp1-CPC lineages were traced by following EYFP expression in murine Mesp1(Cre/+); Rosa26(EYFP/+) ES cells. We captured EYFP+ cells that strongly expressed cardiac mesoderm markers and cardiac transcription factors, but not pluripotent or nascent mesoderm markers. BMP2/4 treatment led to the expansion of EYFP+ cells, while Wnt3a and Activin were marginally effective. BMP2/4 exposure readily led EYFP+ cells to endothelial and smooth muscle cells, but inhibition of the canonical Wnt signaling was required to enter the cardiomyocyte fate. Injected mouse pre-contractile Mesp1-EYFP+ CPCs improved the survivability of injured mice and restored the functional performance of infarcted hearts for at least 3 months. Mesp1-EYFP+ cells are bona fide CPCs and they integrated well in infarcted hearts and emerged de novo into terminally differentiated cardiac myocytes, smooth muscle and vascular endothelial cells.
Collapse
Affiliation(s)
- Yu Liu
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Li Chen
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Andrea Diaz Diaz
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, TX 77204, USA
| | - Ashley Benham
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Xueping Xu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Cori S Wijaya
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, TX 77204, USA
| | - Faisal Fa'ak
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, TX 77204, USA
| | - Weijia Luo
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Benjamin Soibam
- Department of Computer Science and Engineering Technology, University of Houston-Downtown, Houston, 77002, USA
| | - Alon Azares
- Stem Cell Engineering, Texas Heart Institute at St. Luke's Episcopal Hospital, Houston, TX 77030, USA
| | - Wei Yu
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Qiongying Lyu
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - M David Stewart
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA.,Stem Cell Engineering, Texas Heart Institute at St. Luke's Episcopal Hospital, Houston, TX 77030, USA
| | - Preethi Gunaratne
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Austin Cooney
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Bradley K McConnell
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, TX 77204, USA
| | - Robert J Schwartz
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA.,Stem Cell Engineering, Texas Heart Institute at St. Luke's Episcopal Hospital, Houston, TX 77030, USA
| |
Collapse
|
88
|
Jha R, Wu Q, Singh M, Preininger MK, Han P, Ding G, Cho HC, Jo H, Maher KO, Wagner MB, Xu C. Simulated Microgravity and 3D Culture Enhance Induction, Viability, Proliferation and Differentiation of Cardiac Progenitors from Human Pluripotent Stem Cells. Sci Rep 2016; 6:30956. [PMID: 27492371 PMCID: PMC4974658 DOI: 10.1038/srep30956] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 07/12/2016] [Indexed: 12/21/2022] Open
Abstract
Efficient generation of cardiomyocytes from human pluripotent stem cells is critical for their regenerative applications. Microgravity and 3D culture can profoundly modulate cell proliferation and survival. Here, we engineered microscale progenitor cardiac spheres from human pluripotent stem cells and exposed the spheres to simulated microgravity using a random positioning machine for 3 days during their differentiation to cardiomyocytes. This process resulted in the production of highly enriched cardiomyocytes (99% purity) with high viability (90%) and expected functional properties, with a 1.5 to 4-fold higher yield of cardiomyocytes from each undifferentiated stem cell as compared with 3D-standard gravity culture. Increased induction, proliferation and viability of cardiac progenitors as well as up-regulation of genes associated with proliferation and survival at the early stage of differentiation were observed in the 3D culture under simulated microgravity. Therefore, a combination of 3D culture and simulated microgravity can be used to efficiently generate highly enriched cardiomyocytes.
Collapse
Affiliation(s)
- Rajneesh Jha
- Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Qingling Wu
- Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA.,Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Monalisa Singh
- Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Marcela K Preininger
- Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA.,Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Pengcheng Han
- Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Gouliang Ding
- Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Hee Cheol Cho
- Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA.,Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Hanjoong Jo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.,Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Kevin O Maher
- Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Mary B Wagner
- Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA.,Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Chunhui Xu
- Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA.,Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| |
Collapse
|
89
|
Furtado MB, Nim HT, Boyd SE, Rosenthal NA. View from the heart: cardiac fibroblasts in development, scarring and regeneration. Development 2016; 143:387-97. [PMID: 26839342 DOI: 10.1242/dev.120576] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In the adult, tissue repair after injury is generally compromised by fibrosis, which maintains tissue integrity with scar formation but does not restore normal architecture and function. The process of regeneration is necessary to replace the scar and rebuild normal functioning tissue. Here, we address this problem in the context of heart disease, and discuss the origins and characteristics of cardiac fibroblasts, as well as the crucial role that they play in cardiac development and disease. We discuss the dual nature of cardiac fibroblasts, which can lead to scarring, pathological remodelling and functional deficit, but can also promote heart function in some contexts. Finally, we review current and proposed approaches whereby regeneration could be fostered by interventions that limit scar formation.
Collapse
Affiliation(s)
- Milena B Furtado
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | - Hieu T Nim
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia Systems Biology Institute (SBI) Australia, Monash University, Clayton, Victoria 3800, Australia
| | - Sarah E Boyd
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia Systems Biology Institute (SBI) Australia, Monash University, Clayton, Victoria 3800, Australia
| | - Nadia A Rosenthal
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia Systems Biology Institute (SBI) Australia, Monash University, Clayton, Victoria 3800, Australia National Heart and Lung Institute, Imperial College London, London SW3 6LY, UK The Jackson Laboratory, Bar Harbor, ME 04609, USA
| |
Collapse
|
90
|
Bartulos O, Zhuang ZW, Huang Y, Mikush N, Suh C, Bregasi A, Wang L, Chang W, Krause DS, Young LH, Pober JS, Qyang Y. ISL1 cardiovascular progenitor cells for cardiac repair after myocardial infarction. JCI Insight 2016; 1:80920. [PMID: 27525311 DOI: 10.1172/jci.insight.80920] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Cardiovascular progenitor cells (CPCs) expressing the ISL1-LIM-homeodomain transcription factor contribute developmentally to cardiomyocytes in all 4 chambers of the heart. Here, we show that ISL1-CPCs can be applied to myocardial regeneration following injury. We used a rapid 3D methylcellulose approach to form murine and human ISL1-CPC spheroids that engrafted after myocardial infarction in murine hearts, where they differentiated into cardiomyocytes and endothelial cells, integrating into the myocardium and forming new blood vessels. ISL1-CPC spheroid-treated mice exhibited reduced infarct area and increased blood vessel formation compared with control animals. Moreover, left ventricular (LV) contractile function was significantly better in mice transplanted with ISL1-CPCs 4 weeks after injury than that in control animals. These results provide proof-of-concept of a cardiac repair strategy employing ISL1-CPCs that, based on our previous lineage-tracing studies, are committed to forming heart tissue, in combination with a robust methylcellulose spheroid-based delivery approach.
Collapse
Affiliation(s)
- Oscar Bartulos
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine.,Yale Stem Cell Center
| | - Zhen Wu Zhuang
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine
| | - Yan Huang
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine
| | - Nicole Mikush
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine
| | - Carol Suh
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine.,Yale Stem Cell Center
| | - Alda Bregasi
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine
| | - Lin Wang
- Yale Stem Cell Center.,Department of Laboratory Medicine
| | - William Chang
- Department of Internal Medicine, Section of Nephrology
| | - Diane S Krause
- Yale Stem Cell Center.,Department of Laboratory Medicine.,Department of Cell Biology.,Pathology
| | - Lawrence H Young
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine.,Cellular and Molecular Physiology
| | - Jordan S Pober
- Pathology.,Immunobiology, and.,Vascular Biology and Therapeutics Program, Yale University, New Haven, Connecticut, USA
| | - Yibing Qyang
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine.,Yale Stem Cell Center.,Pathology.,Vascular Biology and Therapeutics Program, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
91
|
Cao N, Huang Y, Zheng J, Spencer CI, Zhang Y, Fu JD, Nie B, Xie M, Zhang M, Wang H, Ma T, Xu T, Shi G, Srivastava D, Ding S. Conversion of human fibroblasts into functional cardiomyocytes by small molecules. Science 2016. [DOI: 10.1126/science.aaf1502\] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Making cardiac cells from fibroblasts
Reprogramming noncardiac cells into functional cardiomyocytes without any genetic manipulation could open up new avenues for cardiac regenerative therapies. Cao
et al.
identified a combination of nine small molecules that could epigenetically activate human fibroblasts, efficiently reprogramming them into chemically induced cardiomyocytes (ciCMs). The ciCMs contracted uniformly and resembled human cardiomyocytes. This method may be adapted for reprogramming multiple cell types and have important implications in regenerative medicine.
Science
, this issue p.
1216
Collapse
Affiliation(s)
- Nan Cao
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA
- Department of Pharmaceutical Chemistry, University of California–San Francisco, San Francisco, CA 94158, USA
| | - Yu Huang
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA
| | - Jiashun Zheng
- Department of Biochemistry and Biophysics, University of California–San Francisco, San Francisco, CA 94158, USA
- California Institute for Quantitative Biosciences, University of California–San Francisco, San Francisco, CA 94158, USA
| | - C. Ian Spencer
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA
| | - Yu Zhang
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA
- Department of Pharmaceutical Chemistry, University of California–San Francisco, San Francisco, CA 94158, USA
| | - Ji-Dong Fu
- Department of Medicine, Heart and Vascular Research Center, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Baoming Nie
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA
- Department of Pharmaceutical Chemistry, University of California–San Francisco, San Francisco, CA 94158, USA
| | - Min Xie
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA
- Department of Pharmaceutical Chemistry, University of California–San Francisco, San Francisco, CA 94158, USA
| | - Mingliang Zhang
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA
- Department of Pharmaceutical Chemistry, University of California–San Francisco, San Francisco, CA 94158, USA
| | - Haixia Wang
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA
- Department of Pharmaceutical Chemistry, University of California–San Francisco, San Francisco, CA 94158, USA
| | - Tianhua Ma
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA
- Department of Pharmaceutical Chemistry, University of California–San Francisco, San Francisco, CA 94158, USA
| | - Tao Xu
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA
- Department of Pharmaceutical Chemistry, University of California–San Francisco, San Francisco, CA 94158, USA
| | - Guilai Shi
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA
- Department of Pharmaceutical Chemistry, University of California–San Francisco, San Francisco, CA 94158, USA
| | - Deepak Srivastava
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA
- Department of Pediatrics, University of California–San Francisco, San Francisco, CA 94158, USA
- Department of Biochemistry and Biophysics, University of California–San Francisco, San Francisco, CA 94158, USA
| | - Sheng Ding
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA
- Department of Pharmaceutical Chemistry, University of California–San Francisco, San Francisco, CA 94158, USA
| |
Collapse
|
92
|
Menasché P, Vanneaux V. Stem cells for the treatment of heart failure. Curr Res Transl Med 2016; 64:97-106. [PMID: 27316393 DOI: 10.1016/j.retram.2016.04.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Revised: 04/02/2016] [Accepted: 04/08/2016] [Indexed: 12/17/2022]
Abstract
Stem cell-based therapy is currently tested in several trials of chronic heart failure. The main question is to determine how its implementation could be extended to standard clinical practice. To answer this question, it is helpful to capitalize on the three main lessons drawn from the accumulated experience, both in the laboratory and in the clinics. Regarding the cell type, the best outcomes seem to be achieved by cells the phenotype of which closely matches that of the target tissue. This argues in favor of the use of cardiac-committed cells among which the pluripotent stem cell-derived cardiac progeny is particularly attractive. Regarding the mechanism of action, there has been a major paradigm shift whereby cells are no longer expected to structurally integrate within the recipient myocardium but rather to release biomolecules that foster endogenous repair processes. This implies to focus on early cell retention, rather than on sustained cell survival, so that the cells reside in the target tissue long enough and in sufficient amounts to deliver the factors underpinning their action. Biomaterials are here critical adjuncts to optimize this residency time. Furthermore, the paracrine hypothesis gives more flexibility for using allogeneic cells in that targeting an only transient engraftment requires to delay, and no longer to avoid, rejection, which, in turn, should simplify immunomodulation regimens. Regarding manufacturing, a broad dissemination of cardiac cell therapy requires the development of automated systems allowing to yield highly reproducible cell products. This further emphasizes the interest of allogeneic cells because of their suitability for industrially-relevant and cost-effective scale-up and quality control procedures. At the end, definite confirmation that the effects of cells can be recapitulated by the factors they secrete could lead to acellular therapies whereby factors alone (possibly clustered in extracellular vesicles) would be delivered to the patient. The production process of these cell-derived biologics would then be closer to that of a pharmaceutical compound, which could streamline the manufacturing and regulatory paths and thereby facilitate an expended clinical use.
Collapse
Affiliation(s)
- P Menasché
- Department of Cardiovascular Surgery, Hôpital Européen Georges Pompidou, 20, rue Leblanc, 75015 Paris, France; Université Paris Descartes, Sorbonne Paris Cité, 75010 Paris, France; INSERM U 970, 75010 Paris, France.
| | - V Vanneaux
- INSERM UMR1160, Institut Universitaire d'Hématologie, 75475 Paris cedex 10, France; Assistance publique-Hôpitaux de Paris, Unité de thérapie cellulaire et CIC de Biothérapies, Hôpital Saint-Louis, 75475 Paris cedex 10, France
| |
Collapse
|
93
|
Cao N, Huang Y, Zheng J, Spencer CI, Zhang Y, Fu JD, Nie B, Xie M, Zhang M, Wang H, Ma T, Xu T, Shi G, Srivastava D, Ding S. Conversion of human fibroblasts into functional cardiomyocytes by small molecules. Science 2016; 352:1216-20. [PMID: 27127239 DOI: 10.1126/science.aaf1502] [Citation(s) in RCA: 265] [Impact Index Per Article: 33.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 04/15/2016] [Indexed: 12/11/2022]
Abstract
Reprogramming somatic fibroblasts into alternative lineages would provide a promising source of cells for regenerative therapy. However, transdifferentiating human cells into specific homogeneous, functional cell types is challenging. Here we show that cardiomyocyte-like cells can be generated by treating human fibroblasts with a combination of nine compounds that we term 9C. The chemically induced cardiomyocyte-like cells uniformly contracted and resembled human cardiomyocytes in their transcriptome, epigenetic, and electrophysiological properties. 9C treatment of human fibroblasts resulted in a more open-chromatin conformation at key heart developmental genes, enabling their promoters and enhancers to bind effectors of major cardiogenic signals. When transplanted into infarcted mouse hearts, 9C-treated fibroblasts were efficiently converted to chemically induced cardiomyocyte-like cells. This pharmacological approach to lineage-specific reprogramming may have many important therapeutic implications after further optimization to generate mature cardiac cells.
Collapse
Affiliation(s)
- Nan Cao
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA. Department of Pharmaceutical Chemistry, University of California-San Francisco, San Francisco, CA 94158, USA
| | - Yu Huang
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA
| | - Jiashun Zheng
- Department of Biochemistry and Biophysics, University of California-San Francisco, San Francisco, CA 94158, USA. California Institute for Quantitative Biosciences, University of California-San Francisco, San Francisco, CA 94158, USA
| | - C Ian Spencer
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA
| | - Yu Zhang
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA. Department of Pharmaceutical Chemistry, University of California-San Francisco, San Francisco, CA 94158, USA
| | - Ji-Dong Fu
- Department of Medicine, Heart and Vascular Research Center, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Baoming Nie
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA. Department of Pharmaceutical Chemistry, University of California-San Francisco, San Francisco, CA 94158, USA
| | - Min Xie
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA. Department of Pharmaceutical Chemistry, University of California-San Francisco, San Francisco, CA 94158, USA
| | - Mingliang Zhang
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA. Department of Pharmaceutical Chemistry, University of California-San Francisco, San Francisco, CA 94158, USA
| | - Haixia Wang
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA. Department of Pharmaceutical Chemistry, University of California-San Francisco, San Francisco, CA 94158, USA
| | - Tianhua Ma
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA. Department of Pharmaceutical Chemistry, University of California-San Francisco, San Francisco, CA 94158, USA
| | - Tao Xu
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA. Department of Pharmaceutical Chemistry, University of California-San Francisco, San Francisco, CA 94158, USA
| | - Guilai Shi
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA. Department of Pharmaceutical Chemistry, University of California-San Francisco, San Francisco, CA 94158, USA
| | - Deepak Srivastava
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA. Department of Pediatrics, University of California-San Francisco, San Francisco, CA 94158, USA. Department of Biochemistry and Biophysics, University of California-San Francisco, San Francisco, CA 94158, USA
| | - Sheng Ding
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA. Department of Pharmaceutical Chemistry, University of California-San Francisco, San Francisco, CA 94158, USA
| |
Collapse
|
94
|
Huang J, Zhang M, Zhang P, Liang H, Ouyang K, Yang HT. Coupling switch of P2Y-IP3 receptors mediates differential Ca(2+) signaling in human embryonic stem cells and derived cardiovascular progenitor cells. Purinergic Signal 2016; 12:465-78. [PMID: 27098757 DOI: 10.1007/s11302-016-9512-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 04/04/2016] [Indexed: 12/18/2022] Open
Abstract
Purinergic signaling mediated by P2 receptors (P2Rs) plays important roles in embryonic and stem cell development. However, how it mediates Ca(2+) signals in human embryonic stem cells (hESCs) and derived cardiovascular progenitor cells (CVPCs) remains unclear. Here, we aimed to determine the role of P2Rs in mediating Ca(2+) mobilizations of these cells. hESCs were induced to differentiate into CVPCs by our recently established methods. Gene expression of P2Rs and inositol 1,4,5-trisphosphate receptors (IP3Rs) was analyzed by quantitative/RT-PCR. IP3R3 knockdown (KD) or IP3R2 knockout (KO) hESCs were established by shRNA- or TALEN-mediated gene manipulations, respectively. Confocal imaging revealed that Ca(2+) responses in CVPCs to ATP and UTP were more sensitive and stronger than those in hESCs. Consistently, the gene expression levels of most P2YRs except P2Y1 were increased in CVPCs. Suramin or PPADS blocked ATP-induced Ca(2+) transients in hESCs but only partially inhibited those in CVPCs. Moreover, the P2Y1 receptor-specific antagonist MRS2279 abolished most ATP-induced Ca(2+) signals in hESCs but not in CVPCs. P2Y1 receptor-specific agonist MRS2365 induced Ca(2+) transients only in hESCs but not in CVPCs. Furthermore, IP3R2KO but not IP3R3KD decreased the proportion of hESCs responding to MRS2365. In contrast, both IP3R2 and IP3R3 contributed to UTP-induced Ca(2+) responses while ATP-induced Ca(2+) responses were more dependent on IP3R2 in the CVPCs. In conclusion, a predominant role of P2Y1 receptors in hESCs and a transition of P2Y-IP3R coupling in derived CVPCs are responsible for the differential Ca(2+) mobilization between these cells.
Collapse
Affiliation(s)
- Jijun Huang
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, Shanghai, 200031, China.,Second Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, 310009, China
| | - Min Zhang
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, Shanghai, 200031, China
| | - Peng Zhang
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, Shanghai, 200031, China
| | - He Liang
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, Shanghai, 200031, China.,Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Kunfu Ouyang
- Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Huang-Tian Yang
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, Shanghai, 200031, China. .,Second Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, 310009, China. .,Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
| |
Collapse
|
95
|
Madonna R, Van Laake LW, Davidson SM, Engel FB, Hausenloy DJ, Lecour S, Leor J, Perrino C, Schulz R, Ytrehus K, Landmesser U, Mummery CL, Janssens S, Willerson J, Eschenhagen T, Ferdinandy P, Sluijter JPG. Position Paper of the European Society of Cardiology Working Group Cellular Biology of the Heart: cell-based therapies for myocardial repair and regeneration in ischemic heart disease and heart failure. Eur Heart J 2016; 37:1789-98. [PMID: 27055812 DOI: 10.1093/eurheartj/ehw113] [Citation(s) in RCA: 179] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 02/01/2016] [Indexed: 12/27/2022] Open
Abstract
Despite improvements in modern cardiovascular therapy, the morbidity and mortality of ischaemic heart disease (IHD) and heart failure (HF) remain significant in Europe and worldwide. Patients with IHD may benefit from therapies that would accelerate natural processes of postnatal collateral vessel formation and/or muscle regeneration. Here, we discuss the use of cells in the context of heart repair, and the most relevant results and current limitations from clinical trials using cell-based therapies to treat IHD and HF. We identify and discuss promising potential new therapeutic strategies that include ex vivo cell-mediated gene therapy, the use of biomaterials and cell-free therapies aimed at increasing the success rates of therapy for IHD and HF. The overall aim of this Position Paper of the ESC Working Group Cellular Biology of the Heart is to provide recommendations on how to improve the therapeutic application of cell-based therapies for cardiac regeneration and repair.
Collapse
Affiliation(s)
- Rosalinda Madonna
- Institute of Cardiology and Center of Excellence on Aging, 'G. d'Annunzio' University - Chieti, Chieti, Italy Texas Heart Institute, Houston, USA
| | - Linda W Van Laake
- Hubrecht Institute, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, London, UK
| | - Felix B Engel
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Derek J Hausenloy
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore, National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore The National Institute of Health Research University College London Hospitals Biomedical Research Centre, London, UK
| | - Sandrine Lecour
- MRC Cape Heart Unit, Hatter Cardiovascular Research Institute, University of Cape Town, Cape Town, South Africa
| | - Jonathan Leor
- Neufeld Cardiac Research Institute, Tel-Aviv University, Tel Aviv-Yafo, Israel Tamman Cardiovascular Research Institute, Sheba Medical Center, Tel HaShomer, Israel Sheba Center for Regenerative Medicine, Stem Cell, and Tissue Engineering, Tel Hashomer, Israel
| | - Cinzia Perrino
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Rainer Schulz
- Institute of Physiology, Justus-Liebig Giessen University of Giessen, Gießen, Germany
| | - Kirsti Ytrehus
- Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| | - Ulf Landmesser
- Department of Cardiology, Charite Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | | | - Stefan Janssens
- Department of Cardiovascular Sciences, Clinical Cardiology, KU Leuven, Leuven, Belgium
| | - James Willerson
- Department of Cardiology, Texas Heart Institute, Houston, TX, USA
| | - Thomas Eschenhagen
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, Hamburg 20246, Germany
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary Pharmahungary Group, Szeged, Hungary
| | | |
Collapse
|
96
|
Månsson-Broberg A, Rodin S, Bulatovic I, Ibarra C, Löfling M, Genead R, Wärdell E, Felldin U, Granath C, Alici E, Le Blanc K, Smith CIE, Salašová A, Westgren M, Sundström E, Uhlén P, Arenas E, Sylvén C, Tryggvason K, Corbascio M, Simonson OE, Österholm C, Grinnemo KH. Wnt/β-Catenin Stimulation and Laminins Support Cardiovascular Cell Progenitor Expansion from Human Fetal Cardiac Mesenchymal Stromal Cells. Stem Cell Reports 2016; 6:607-617. [PMID: 27052314 PMCID: PMC4834052 DOI: 10.1016/j.stemcr.2016.02.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 02/19/2016] [Accepted: 02/22/2016] [Indexed: 11/29/2022] Open
Abstract
The intrinsic regenerative capacity of human fetal cardiac mesenchymal stromal cells (MSCs) has not been fully characterized. Here we demonstrate that we can expand cells with characteristics of cardiovascular progenitor cells from the MSC population of human fetal hearts. Cells cultured on cardiac muscle laminin (LN)-based substrata in combination with stimulation of the canonical Wnt/β-catenin pathway showed increased gene expression of ISL1, OCT4, KDR, and NKX2.5. The majority of cells stained positive for PDGFR-α, ISL1, and NKX2.5, and subpopulations also expressed the progenitor markers TBX18, KDR, c-KIT, and SSEA-1. Upon culture of the cardiac MSCs in differentiation media and on relevant LNs, portions of the cells differentiated into spontaneously beating cardiomyocytes, and endothelial and smooth muscle-like cells. Our protocol for large-scale culture of human fetal cardiac MSCs enables future exploration of the regenerative functions of these cells in the context of myocardial injury in vitro and in vivo. Cells with progenitor properties can be expanded from human fetal cardiac MSCs Specific LNs support expansion and differentiation of cardiac MSCs The fetal cardiac MSCs express ISL1, PDGFR-α, and NKX2.5 Subpopulations express the progenitor markers KDR, SSEA-1, c-KIT, and TBX18
Collapse
Affiliation(s)
- Agneta Månsson-Broberg
- Division of Cardiology, Department of Medicine, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Sergey Rodin
- Division of Matrix Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Ivana Bulatovic
- Division of Cardiothoracic Surgery and Anesthesiology, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Cristián Ibarra
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden; Cardiovascular & Metabolic Diseases, Innovative Medicines and Early Development, AstraZeneca R&D, 43150 Mölndal, Sweden
| | - Marie Löfling
- Division of Cardiothoracic Surgery and Anesthesiology, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Rami Genead
- Division of Cardiothoracic Surgery and Anesthesiology, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Eva Wärdell
- Division of Cardiology, Department of Medicine, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Ulrika Felldin
- Division of Cardiothoracic Surgery and Anesthesiology, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Carl Granath
- Division of Cardiothoracic Surgery and Anesthesiology, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Evren Alici
- Division of Hematology, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, 17177 Stockholm, Sweden; Cell Therapy Institute, Nova Southeastern University, Fort Lauderdale, FL 33314, USA
| | - Katarina Le Blanc
- Division of Hematology, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, 17177 Stockholm, Sweden; Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital, 17177 Stockholm, Sweden
| | - C I Edvard Smith
- Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital, 17177 Stockholm, Sweden
| | - Alena Salašová
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Magnus Westgren
- CLINTEC, Division of Obstetrics and Gynecology, Karolinska Institutet, Karolinska University Hospital, 17177 Stockholm, Sweden
| | - Erik Sundström
- Division of Neurodegeneration, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Karolinska University Hospital, 17177 Stockholm, Sweden
| | - Per Uhlén
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Ernest Arenas
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Christer Sylvén
- Division of Cardiology, Department of Medicine, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Karl Tryggvason
- Division of Matrix Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden; Duke-NUS Graduate Medical School, Durham, NC 27710, USA
| | - Matthias Corbascio
- Division of Cardiothoracic Surgery and Anesthesiology, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Oscar E Simonson
- Division of Cardiothoracic Surgery and Anesthesiology, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Cecilia Österholm
- Division of Cardiothoracic Surgery and Anesthesiology, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17177 Stockholm, Sweden; Cell Therapy Institute, Nova Southeastern University, Fort Lauderdale, FL 33314, USA
| | - Karl-Henrik Grinnemo
- Division of Cardiothoracic Surgery and Anesthesiology, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17177 Stockholm, Sweden; Cell Therapy Institute, Nova Southeastern University, Fort Lauderdale, FL 33314, USA.
| |
Collapse
|
97
|
Techniques of Human Embryonic Stem Cell and Induced Pluripotent Stem Cell Derivation. Arch Immunol Ther Exp (Warsz) 2016; 64:349-70. [PMID: 26939778 PMCID: PMC5021740 DOI: 10.1007/s00005-016-0385-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 11/17/2015] [Indexed: 12/22/2022]
Abstract
Developing procedures for the derivation of human pluripotent stem cells (PSCs) gave rise to novel pathways into regenerative medicine research. For many years, stem cells have attracted attention as a potentially unlimited cell source for cellular therapy in neurodegenerative disorders, cardiovascular diseases, and spinal cord injuries, for example. In these studies, adult stem cells were insufficient; therefore, many attempts were made to obtain PSCs by other means. This review discusses key issues concerning the techniques of pluripotent cell acquisition. Technical and ethical issues hindered the medical use of somatic cell nuclear transfer and embryonic stem cells. Therefore, induced PSCs (iPSCs) emerged as a powerful technique with great potential for clinical applications, patient-specific disease modelling and pharmaceutical studies. The replacement of viral vectors or the administration of analogous proteins or chemical compounds during cell reprogramming are modifications designed to reduce tumorigenesis risk and to augment the procedure efficiency. Intensified analysis of new PSC lines revealed other barriers to overcome, such as epigenetic memory, disparity between human and mouse pluripotency, and variable response to differentiation of some iPSC lines. Thus, multidimensional verification must be conducted to fulfil strict clinical-grade requirements. Nevertheless, the first clinical trials in patients with spinal cord injury and macular dystrophy were recently carried out with differentiated iPSCs, encouraging alternative strategies for potential autologous cellular therapies.
Collapse
|
98
|
Focus on Extracellular Vesicles: Development of Extracellular Vesicle-Based Therapeutic Systems. Int J Mol Sci 2016; 17:172. [PMID: 26861303 PMCID: PMC4783906 DOI: 10.3390/ijms17020172] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2015] [Accepted: 01/29/2016] [Indexed: 01/01/2023] Open
Abstract
Many types of cells release phospholipid membrane vesicles thought to play key roles in cell-cell communication, antigen presentation, and the spread of infectious agents. Extracellular vesicles (EVs) carry various proteins, messenger RNAs (mRNAs), and microRNAs (miRNAs), like a “message in a bottle” to cells in remote locations. The encapsulated molecules are protected from multiple types of degradative enzymes in body fluids, making EVs ideal for delivering drugs. This review presents an overview of the potential roles of EVs as natural drugs and novel drug-delivery systems.
Collapse
|
99
|
Kervadec A, Bellamy V, El Harane N, Arakélian L, Vanneaux V, Cacciapuoti I, Nemetalla H, Périer MC, Toeg HD, Richart A, Lemitre M, Yin M, Loyer X, Larghero J, Hagège A, Ruel M, Boulanger CM, Silvestre JS, Menasché P, Renault NKE. Cardiovascular progenitor-derived extracellular vesicles recapitulate the beneficial effects of their parent cells in the treatment of chronic heart failure. J Heart Lung Transplant 2016; 35:795-807. [PMID: 27041495 DOI: 10.1016/j.healun.2016.01.013] [Citation(s) in RCA: 145] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 12/23/2015] [Accepted: 01/10/2016] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Cell-based therapies are being explored as a therapeutic option for patients with chronic heart failure following myocardial infarction. Extracellular vesicles (EV), including exosomes and microparticles, secreted by transplanted cells may orchestrate their paracrine therapeutic effects. We assessed whether post-infarction administration of EV released by human embryonic stem cell-derived cardiovascular progenitors (hESC-Pg) can provide equivalent benefits to administered hESC-Pg and whether hESC-Pg and EV treatments activate similar endogenous pathways. METHODS Mice underwent surgical occlusion of their left coronary arteries. After 2-3 weeks, 95 mice included in the study were treated with hESC-Pg, EV, or Minimal Essential Medium Alpha Medium (alpha-MEM; vehicle control) delivered by percutaneous injections under echocardiographic guidance into the peri-infarct myocardium. functional and histologic end-points were blindly assessed 6 weeks later, and hearts were processed for gene profiling. Genes differentially expressed between control hearts and hESC-Pg-treated and EV-treated hearts were clustered into functionally relevant pathways. RESULTS At 6 weeks after hESC-Pg administration, treated mice had significantly reduced left ventricular end-systolic (-4.20 ± 0.96 µl or -7.5%, p = 0.0007) and end-diastolic (-4.48 ± 1.47 µl or -4.4%, p = 0.009) volumes compared with baseline values despite the absence of any transplanted hESC-Pg or human embryonic stem cell-derived cardiomyocytes in the treated mouse hearts. Equal benefits were seen with the injection of hESC-Pg-derived EV, whereas animals injected with alpha-MEM (vehicle control) did not improve significantly. Histologic examination suggested a slight reduction in infarct size in hESC-Pg-treated animals and EV-treated animals compared with alpha-MEM-treated control animals. In the hESC-Pg-treated and EV-treated groups, heart gene profiling identified 927 genes that were similarly upregulated compared with the control group. Among the 49 enriched pathways associated with these up-regulated genes that could be related to cardiac function or regeneration, 78% were predicted to improve cardiac function through increased cell survival and/or proliferation or DNA repair as well as pathways related to decreased fibrosis and heart failure. CONCLUSIONS In this post-infarct heart failure model, either hESC-Pg or their secreted EV enhance recovery of cardiac function and similarly affect cardiac gene expression patterns that could be related to this recovery. Although the mechanisms by which EV improve cardiac function remain to be determined, these results support the idea that a paracrine mechanism is sufficient to effect functional recovery in cell-based therapies for post-infarction-related chronic heart failure.
Collapse
Affiliation(s)
- Anaïs Kervadec
- INSERM U970, Hôpital Européen Georges Pompidou, Paris Centre de Recherche Cardiovasculaire, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, UMR-S970, Paris, France
| | - Valérie Bellamy
- INSERM U970, Hôpital Européen Georges Pompidou, Paris Centre de Recherche Cardiovasculaire, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, UMR-S970, Paris, France
| | - Nadia El Harane
- INSERM U970, Hôpital Européen Georges Pompidou, Paris Centre de Recherche Cardiovasculaire, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, UMR-S970, Paris, France
| | - Lousineh Arakélian
- Cell Therapy Unit, Assistance Publique-Hôpitaux de Paris, Hôpital Saint-Louis, Paris, France; INSERM, CIC de Biothérapies (CBT-501) and U1160, Institut Universitaire d'Hématologie, Hôpital Saint-Louis, Paris, France
| | - Valérie Vanneaux
- Cell Therapy Unit, Assistance Publique-Hôpitaux de Paris, Hôpital Saint-Louis, Paris, France; INSERM, CIC de Biothérapies (CBT-501) and U1160, Institut Universitaire d'Hématologie, Hôpital Saint-Louis, Paris, France
| | - Isabelle Cacciapuoti
- Cell Therapy Unit, Assistance Publique-Hôpitaux de Paris, Hôpital Saint-Louis, Paris, France; INSERM, CIC de Biothérapies (CBT-501) and U1160, Institut Universitaire d'Hématologie, Hôpital Saint-Louis, Paris, France
| | - Hany Nemetalla
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Department of Cardiology, Paris, France
| | - Marie-Cécile Périer
- INSERM U970, Hôpital Européen Georges Pompidou, Paris Centre de Recherche Cardiovasculaire, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, UMR-S970, Paris, France
| | - Hadi D Toeg
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Adèle Richart
- INSERM U970, Hôpital Européen Georges Pompidou, Paris Centre de Recherche Cardiovasculaire, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, UMR-S970, Paris, France
| | - Mathilde Lemitre
- INSERM U970, Hôpital Européen Georges Pompidou, Paris Centre de Recherche Cardiovasculaire, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, UMR-S970, Paris, France
| | - Min Yin
- INSERM U970, Hôpital Européen Georges Pompidou, Paris Centre de Recherche Cardiovasculaire, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, UMR-S970, Paris, France; University Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Xavier Loyer
- INSERM U970, Hôpital Européen Georges Pompidou, Paris Centre de Recherche Cardiovasculaire, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, UMR-S970, Paris, France
| | - Jérôme Larghero
- Cell Therapy Unit, Assistance Publique-Hôpitaux de Paris, Hôpital Saint-Louis, Paris, France; INSERM, CIC de Biothérapies (CBT-501) and U1160, Institut Universitaire d'Hématologie, Hôpital Saint-Louis, Paris, France; University Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Albert Hagège
- INSERM U970, Hôpital Européen Georges Pompidou, Paris Centre de Recherche Cardiovasculaire, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, UMR-S970, Paris, France; Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Department of Cardiology, Paris, France
| | - Marc Ruel
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Chantal M Boulanger
- INSERM U970, Hôpital Européen Georges Pompidou, Paris Centre de Recherche Cardiovasculaire, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, UMR-S970, Paris, France
| | - Jean-Sébastien Silvestre
- INSERM U970, Hôpital Européen Georges Pompidou, Paris Centre de Recherche Cardiovasculaire, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, UMR-S970, Paris, France
| | - Philippe Menasché
- INSERM U970, Hôpital Européen Georges Pompidou, Paris Centre de Recherche Cardiovasculaire, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, UMR-S970, Paris, France; Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Department of Cardiovascular Surgery, Paris, France.
| | - Nisa K E Renault
- INSERM U970, Hôpital Européen Georges Pompidou, Paris Centre de Recherche Cardiovasculaire, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, UMR-S970, Paris, France
| |
Collapse
|
100
|
Lewandowski J, Kolanowski TJ, Kurpisz M. Techniques for the induction of human pluripotent stem cell differentiation towards cardiomyocytes. J Tissue Eng Regen Med 2016; 11:1658-1674. [PMID: 26777594 DOI: 10.1002/term.2117] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 09/16/2015] [Accepted: 11/18/2015] [Indexed: 01/04/2023]
Abstract
The derivation of pluripotent stem cells from human embryos and the generation of induced pluripotent stem cells (iPSCs) from somatic cells opened a new chapter in studies on the regeneration of the post-infarction heart and regenerative medicine as a whole. Thus, protocols for obtaining iPSCs were enthusiastically adopted and widely used for further experiments on cardiac differentiation. iPSC-mediated cardiomyocytes (iPSC-CMs) under in vitro culture conditions are generated by simulating natural cardiomyogenesis and involve the wingless-type mouse mammary tumour virus integration site family (WNT), transforming growth factor beta (TGF-β) and fibroblast growth factor (FGF) signalling pathways. New strategies have been proposed to take advantage of small chemical molecules, organic compounds and even electric or mechanical stimulation. There are three main approaches to support cardiac commitment in vitro: embryoid bodis (EBs), monolayer in vitro cultures and inductive co-cultures with visceral endoderm-like (END2) cells. In EB technique initial uniform size of pluripotent stem cell (PSC) colonies has a pivotal significance. Hence, some methods were designed to support cells aggregation. Another well-suited procedure is based on culturing cells in monolayer conditions in order to improve accessibility of growth factors and nutrients. Other distinct tactics are using visceral endoderm-like cells to culture them with PSCs due to secretion of procardiac cytokines. Finally, the appropriate purification of the obtained cardiomyocytes is required prior to their administration to a patient under the prospective cellular therapy strategy. This goal can be achieved using non-genetic methods, such as the application of surface markers and fluorescent dyes. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Jarosław Lewandowski
- Department of Reproductive Biology and Stem Cells, Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| | - Tomasz J Kolanowski
- Department of Reproductive Biology and Stem Cells, Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| | - Maciej Kurpisz
- Department of Reproductive Biology and Stem Cells, Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| |
Collapse
|