1
|
Son Y, Li P, Ortega D, Qiu H, Prachyl H, Yang M, Zhu W. Generation of a Human Induced Pluripotent Stem Cell Line Expressing a Magnetic Resonance Imaging Reporter Gene. SMALL METHODS 2024; 8:e2301764. [PMID: 38708688 PMCID: PMC11483194 DOI: 10.1002/smtd.202301764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 04/09/2024] [Indexed: 05/07/2024]
Abstract
The objective of the current study is to develop a new method for tracking transplanted human induced pluripotent stem cells-derived cardiomyocytes (hiPSC-CMs) using magnetic resonance imaging (MRI). The CRISPR/dCas9 activation system is employed to overexpress ferritin heavy chain (FHC) in hiPSC-CMs. The mRNA and protein expression of FHC in hiPSC and hiPSC-CMs significantly increased after transfection. Iron chloride does not affect the cell viability in a concentration range from 0 to 2000 µm. hiPSCs overexpressing FHC (hiPSC- FHCOE) and hiPSC-CMs overexpressing FHC (hiPSC-CM-FHCOE) significantly enhanced cellular uptake of iron chloride but with no changes in electrophysiological properties compared to hiPSC-CM-Control. Furthermore, hiPSC-CM-FHCOE presented robust contrast and lower T2* values, signifying their potential as highly effective candidates for cardiac MRI. Next, hiPSC-CM-FHCOE is injected into mouse hearts and after 3 days of transplantation, MR images are obtained. hiPSC-CM-FHCOE cells exhibited clear signals in the hearts with lower T2* and rapid signal decay. Collectively, data from this proof-of-concept study demonstrated that endogenous labeling with FHC in hiPSC-CMs can be a potent strategy for enhancing the accuracy of cardiac MRI. This technology represents a significant step forward in tracking the transplanted hiPSC-CMs in the hearts of live animals.
Collapse
Affiliation(s)
- Yura Son
- Department of Cardiovascular Medicine, Department of Physiology and Biomedical Engineering, and Center for Regenerative Biotherapeutics, Mayo Clinic Arizona, Scottsdale, AZ, 85259, USA
| | - Pengsheng Li
- Department of Cardiovascular Medicine, Department of Physiology and Biomedical Engineering, and Center for Regenerative Biotherapeutics, Mayo Clinic Arizona, Scottsdale, AZ, 85259, USA
| | - Dakota Ortega
- KE Biosciences Preclinical Imaging Center, Arizona State University, Tempe, AZ, 85281, USA
| | - Huiliang Qiu
- Department of Cardiovascular Medicine, Department of Physiology and Biomedical Engineering, and Center for Regenerative Biotherapeutics, Mayo Clinic Arizona, Scottsdale, AZ, 85259, USA
| | - Hannah Prachyl
- Department of Cardiovascular Medicine, Department of Physiology and Biomedical Engineering, and Center for Regenerative Biotherapeutics, Mayo Clinic Arizona, Scottsdale, AZ, 85259, USA
| | - Ming Yang
- Department of Radiology, Mayo Clinic Arizona, Scottsdale, AZ, 85259, USA
| | - Wuqiang Zhu
- Department of Cardiovascular Medicine, Department of Physiology and Biomedical Engineering, and Center for Regenerative Biotherapeutics, Mayo Clinic Arizona, Scottsdale, AZ, 85259, USA
| |
Collapse
|
2
|
Visualization of regenerating and repairing hearts. Clin Sci (Lond) 2022; 136:787-798. [PMID: 35621122 PMCID: PMC9886236 DOI: 10.1042/cs20211116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 05/03/2022] [Accepted: 05/05/2022] [Indexed: 02/01/2023]
Abstract
With heart failure continuing to become more prevalent, investigating the mechanisms of heart injury and repair holds much incentive. In contrast with adult mammals, other organisms such as teleost fish, urodele amphibians, and even neonatal mammals are capable of robust cardiac regeneration to replenish lost or damaged myocardial tissue. Long-term high-resolution intravital imaging of the behaviors and interactions of different cardiac cell types in their native environment could yield unprecedented insights into heart regeneration and repair. However, this task remains challenging for the heart due to its rhythmic contraction and anatomical location. Here, we summarize recent advances in live imaging of heart regeneration and repair, discuss the advantages and limitations of current systems, and suggest future directions for novel imaging technology development.
Collapse
|
3
|
Neef K, Drey F, Lepperhof V, Wahlers T, Hescheler J, Choi YH, Šarić T. Co-transplantation of Mesenchymal Stromal Cells and Induced Pluripotent Stem Cell-Derived Cardiomyocytes Improves Cardiac Function After Myocardial Damage. Front Cardiovasc Med 2022; 8:794690. [PMID: 35071360 PMCID: PMC8770928 DOI: 10.3389/fcvm.2021.794690] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 11/22/2021] [Indexed: 01/01/2023] Open
Abstract
Induced pluripotent stem cell-derived cardiomyocytes (iPS-CMs) represent an attractive resource for cardiac regeneration. However, survival and functional integration of transplanted iPS-CM is poor and remains a major challenge for the development of effective therapies. We hypothesized that paracrine effects of co-transplanted mesenchymal stromal cells (MSCs) augment the retention and therapeutic efficacy of iPS-CM in a mouse model of myocardial infarction (MI). To test this, either iPS-CM, MSC, or both cell types were transplanted into the cryoinfarction border zone of syngeneic mice immediately after injury. Bioluminescence imaging (BLI) of iPS-CM did not confirm enhanced retention by co-application of MSC during the 28-day follow-up period. However, histological analyses of hearts 28 days after cell transplantation showed that MSC increased the fraction of animals with detectable iPS-CM by 2-fold. Cardiac MRI analyses showed that from day 14 after transplantation on, the animals that have received cells had a significantly higher left ventricular ejection fraction (LVEF) compared to the placebo group. There was no statistically significant difference in LVEF between animals transplanted only with iPS-CM or only with MSC. However, combined iPS-CM and MSC transplantation resulted in higher LVEF compared to transplantation of single-cell populations during the whole observation period. Histological analyses revealed that MSC increased the capillarization in the myocardium when transplanted alone or with iPS-CM and decreased the infarct scar area only when transplanted in combination with iPS-CM. These results indicate that co-transplantation of iPS-CM and MSC improves cardiac regeneration after cardiac damage, demonstrating the potential of combining multiple cell types for increasing the efficacy of future cardiac cell therapies.
Collapse
Affiliation(s)
- Klaus Neef
- Department of Cardiac and Thoracic Surgery, Heart Center, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Florian Drey
- Department of Cardiac and Thoracic Surgery, Heart Center, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Vera Lepperhof
- Institute for Neurophysiology, Center for Physiology and Pathophysiology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Thorsten Wahlers
- Department of Cardiac and Thoracic Surgery, Heart Center, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Jürgen Hescheler
- Institute for Neurophysiology, Center for Physiology and Pathophysiology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Yeong-Hoon Choi
- Department of Cardiac and Thoracic Surgery, Heart Center, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- Clinic for Cardiac Surgery and Surgical Intensive Care Medicine, Kerckhoff Clinic Bad Nauheim, Kerckhoff Campus, Justus Liebig University Giessen, Giessen, Germany
| | - Tomo Šarić
- Institute for Neurophysiology, Center for Physiology and Pathophysiology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- *Correspondence: Tomo Šarić
| |
Collapse
|
4
|
Salicylic diamines selectively eliminate residual undifferentiated cells from pluripotent stem cell-derived cardiomyocyte preparations. Sci Rep 2021; 11:2391. [PMID: 33504837 PMCID: PMC7841182 DOI: 10.1038/s41598-021-81351-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 12/14/2020] [Indexed: 02/06/2023] Open
Abstract
Clinical translation of pluripotent stem cell (PSC) derivatives is hindered by the tumorigenic risk from residual undifferentiated cells. Here, we identified salicylic diamines as potent agents exhibiting toxicity to murine and human PSCs but not to cardiomyocytes (CMs) derived from them. Half maximal inhibitory concentrations (IC50) of small molecules SM2 and SM6 were, respectively, 9- and 18-fold higher for human than murine PSCs, while the IC50 of SM8 was comparable for both PSC groups. Treatment of murine embryoid bodies in suspension differentiation cultures with the most effective small molecule SM6 significantly reduced PSC and non-PSC contamination and enriched CM populations that would otherwise be eliminated in genetic selection approaches. All tested salicylic diamines exerted their toxicity by inhibiting the oxygen consumption rate (OCR) in PSCs. No or only minimal and reversible effects on OCR, sarcomeric integrity, DNA stability, apoptosis rate, ROS levels or beating frequency were observed in PSC-CMs, although effects on human PSC-CMs seemed to be more deleterious at higher SM-concentrations. Teratoma formation from SM6-treated murine PSC-CMs was abolished or delayed compared to untreated cells. We conclude that salicylic diamines represent promising compounds for PSC removal and enrichment of CMs without the need for other selection strategies.
Collapse
|
5
|
House A, Atalla I, Lee EJ, Guvendiren M. Designing Biomaterial Platforms for Cardiac Tissue and Disease Modeling. ADVANCED NANOBIOMED RESEARCH 2021; 1:2000022. [PMID: 33709087 PMCID: PMC7942203 DOI: 10.1002/anbr.202000022] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Heart disease is one of the leading causes of death in the world. There is a growing demand for in vitro cardiac models that can recapitulate the complex physiology of the cardiac tissue. These cardiac models can provide a platform to better understand the underlying mechanisms of cardiac development and disease and aid in developing novel treatment alternatives and platforms towards personalized medicine. In this review, a summary of engineered cardiac platforms is presented. Basic design considerations for replicating the heart's microenvironment are discussed considering the anatomy of the heart. This is followed by a detailed summary of the currently available biomaterial platforms for modeling the heart tissue in vitro. These in vitro models include 2D surface modified structures, 3D molded structures, porous scaffolds, electrospun scaffolds, bioprinted structures, and heart-on-a-chip devices. The challenges faced by current models and the future directions of in vitro cardiac models are also discussed. Engineered in vitro tissue models utilizing patients' own cells could potentially revolutionize the way we develop treatment and diagnostic alternatives.
Collapse
Affiliation(s)
- Andrew House
- Instructive Biomaterials and Additive Manufacturing Laboratory, Otto H. York Chemical and Materials Engineering, 138 York Center, University Heights, Newark, NJ 07102, USA
| | - Iren Atalla
- Instructive Biomaterials and Additive Manufacturing Laboratory, Otto H. York Chemical and Materials Engineering, 138 York Center, University Heights, Newark, NJ 07102, USA
| | - Eun Jung Lee
- Instructive Biomaterials and Additive Manufacturing Laboratory, Otto H. York Chemical and Materials Engineering, 138 York Center, University Heights, Newark, NJ 07102, USA
| | - Murat Guvendiren
- Instructive Biomaterials and Additive Manufacturing Laboratory, Otto H. York Chemical and Materials Engineering, 138 York Center, University Heights, Newark, NJ 07102, USA
| |
Collapse
|
6
|
Wanjare M, Kawamura M, Hu C, Alcazar C, Wang H, Woo YJ, Huang NF. Vascularization of Engineered Spatially Patterned Myocardial Tissue Derived From Human Pluripotent Stem Cells in vivo. Front Bioeng Biotechnol 2019; 7:208. [PMID: 31552234 PMCID: PMC6733921 DOI: 10.3389/fbioe.2019.00208] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Accepted: 08/19/2019] [Indexed: 12/28/2022] Open
Abstract
Tissue engineering approaches to regenerate myocardial tissue after disease or injury is promising. Integration with the host vasculature is critical to the survival and therapeutic efficacy of engineered myocardial tissues. To create more physiologically oriented engineered myocardial tissue with organized cellular arrangements and endothelial interactions, randomly oriented or parallel-aligned microfibrous polycaprolactone scaffolds were seeded with human pluripotent stem cell-derived cardiomyocytes (iCMs) and/or endothelial cells (iECs). The resultant engineered myocardial tissues were assessed in a subcutaneous transplantation model and in a myocardial injury model to evaluate the effect of scaffold anisotropy and endothelial interactions on vascular integration of the engineered myocardial tissue. Here we demonstrated that engineered myocardial tissue composed of randomly oriented scaffolds seeded with iECs promoted the survival of iECs for up to 14 days. However, engineered myocardial tissue composed of aligned scaffolds preferentially guided the organization of host capillaries along the direction of the microfibers. In a myocardial injury model, epicardially transplanted engineered myocardial tissues composed of randomly oriented scaffolds seeded with iCMs augmented microvessel formation leading to a significantly higher arteriole density after 4 weeks, compared to engineered tissues derived from aligned scaffolds. These findings that the scaffold microtopography imparts differential effect on revascularization, in which randomly oriented scaffolds promote pro-survival and pro-angiogenic effects, and aligned scaffolds direct the formation of anisotropic vessels. These findings suggest a dominant role of scaffold topography over endothelial co-culture in modulating cellular survival, vascularization, and microvessel architecture.
Collapse
Affiliation(s)
- Maureen Wanjare
- Center for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, United States.,Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States
| | - Masashi Kawamura
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, United States
| | - Caroline Hu
- Center for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, United States
| | - Cynthia Alcazar
- Center for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, United States
| | - Hanjay Wang
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, United States
| | - Y Joseph Woo
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States.,Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, United States.,Department of Bioengineering, Stanford University, Stanford, CA, United States
| | - Ngan F Huang
- Center for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, United States.,Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States.,Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, United States
| |
Collapse
|
7
|
Dubois VP, Zotova D, Parkins KM, Swick C, Hamilton AM, Kelly JJ, Ronald JA. Safe Harbor Targeted CRISPR-Cas9 Tools for Molecular-Genetic Imaging of Cells in Living Subjects. CRISPR J 2018; 1:440-449. [PMID: 31021241 DOI: 10.1089/crispr.2018.0030] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Noninvasive molecular-genetic imaging of cells expressing imaging reporter genes is an invaluable approach for longitudinal monitoring of the biodistribution and viability of cancer cells and cell-based therapies in preclinical models and patients. However, labeling cells with reporter genes often relies on using gene transfer methods that randomly integrate the reporter genes into the genome, which may cause unwanted and serious detrimental effects. To overcome this, we have developed CRISPR-Cas9 tools to edit cells at the adeno-associated virus site 1 (AAVS1) safe harbour with a large donor construct (∼6.3 kilobases) encoding an antibiotic resistance gene and reporter genes for bioluminescence (BLI) and fluorescence imaging. HEK293T cells were transfected with a dual plasmid system encoding the Cas9 endonuclease and an AAVS1-targeted guide RNA in one plasmid, and a donor plasmid encoding a puromycin resistance gene, tdTomato and firefly luciferase flanked by AAVS1 homology arms. Puromycin-resistant clonal cells were isolated and AAVS1 integration was confirmed via PCR and sequencing of the PCR product. In vitro BLI signal correlated well to cell number (R2 = 0.9988; p < 0.05) and was stable over multiple passages. Engineered cells (2.5 × 106) were injected into the left hind flank of nude mice and in vivo BLI was performed on days 0, 7, 14, 21, and 28. BLI signal trended down from day 0 to day 7, but significantly increased by day 28 due to cell growth (p < 0.05). This describes the first CRISPR-Cas9 system for AAVS1 integration of large gene constructs for molecular-genetic imaging of cells in vivo. With further development, including improving editing efficiency, use of clinically relevant reporters, and evaluation in other cell populations that can be readily expanded in culture (e.g., immortalized cells or T cells), this CRISPR-Cas9 reporter gene system could be broadly applied to a number of in vivo cell tracking studies.
Collapse
Affiliation(s)
- Veronica P Dubois
- 1 Department of Medical Biophysics, Western University , London, Ontario, Canada.,2 Imaging Research Laboratories, Robarts Research Institute , London, Ontario, Canada
| | - Darya Zotova
- 2 Imaging Research Laboratories, Robarts Research Institute , London, Ontario, Canada
| | - Katie M Parkins
- 1 Department of Medical Biophysics, Western University , London, Ontario, Canada
| | - Connor Swick
- 2 Imaging Research Laboratories, Robarts Research Institute , London, Ontario, Canada
| | - Amanda M Hamilton
- 2 Imaging Research Laboratories, Robarts Research Institute , London, Ontario, Canada
| | - John J Kelly
- 2 Imaging Research Laboratories, Robarts Research Institute , London, Ontario, Canada
| | - John A Ronald
- 1 Department of Medical Biophysics, Western University , London, Ontario, Canada.,2 Imaging Research Laboratories, Robarts Research Institute , London, Ontario, Canada.,3 Lawson Health Research Institute, London, Ontario, Canada
| |
Collapse
|
8
|
Genome Editing Redefines Precision Medicine in the Cardiovascular Field. Stem Cells Int 2018; 2018:4136473. [PMID: 29731778 PMCID: PMC5872631 DOI: 10.1155/2018/4136473] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 10/25/2017] [Indexed: 02/06/2023] Open
Abstract
Genome editing is a powerful tool to study the function of specific genes and proteins important for development or disease. Recent technologies, especially CRISPR/Cas9 which is characterized by convenient handling and high precision, revolutionized the field of genome editing. Such tools have enormous potential for basic science as well as for regenerative medicine. Nevertheless, there are still several hurdles that have to be overcome, but patient-tailored therapies, termed precision medicine, seem to be within reach. In this review, we focus on the achievements and limitations of genome editing in the cardiovascular field. We explore different areas of cardiac research and highlight the most important developments: (1) the potential of genome editing in human pluripotent stem cells in basic research for disease modelling, drug screening, or reprogramming approaches and (2) the potential and remaining challenges of genome editing for regenerative therapies. Finally, we discuss social and ethical implications of these new technologies.
Collapse
|
9
|
Wierzbinski KR, Szymanski T, Rozwadowska N, Rybka JD, Zimna A, Zalewski T, Nowicka-Bauer K, Malcher A, Nowaczyk M, Krupinski M, Fiedorowicz M, Bogorodzki P, Grieb P, Giersig M, Kurpisz MK. Potential use of superparamagnetic iron oxide nanoparticles for in vitro and in vivo bioimaging of human myoblasts. Sci Rep 2018; 8:3682. [PMID: 29487326 PMCID: PMC5829264 DOI: 10.1038/s41598-018-22018-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 02/12/2018] [Indexed: 02/07/2023] Open
Abstract
Myocardial infarction (MI) is one of the most frequent causes of death in industrialized countries. Stem cells therapy seems to be very promising for regenerative medicine. Skeletal myoblasts transplantation into postinfarction scar has been shown to be effective in the failing heart but shows limitations such, e.g. cell retention and survival. We synthesized and investigated superparamagnetic iron oxide nanoparticles (SPIONs) as an agent for direct cell labeling, which can be used for stem cells imaging. High quality, monodisperse and biocompatible DMSA-coated SPIONs were obtained with thermal decomposition and subsequent ligand exchange reaction. SPIONs' presence within myoblasts was confirmed by Prussian Blue staining and inductively coupled plasma mass spectrometry (ICP-MS). SPIONs' influence on tested cells was studied by their proliferation, ageing, differentiation potential and ROS production. Cytotoxicity of obtained nanoparticles and myoblast associated apoptosis were also tested, as well as iron-related and coating-related genes expression. We examined SPIONs' impact on overexpression of two pro-angiogenic factors introduced via myoblast electroporation method. Proposed SPION-labeling was sufficient to visualize firefly luciferase-modified and SPION-labeled cells with magnetic resonance imaging (MRI) combined with bioluminescence imaging (BLI) in vivo. The obtained results demonstrated a limited SPIONs' influence on treated skeletal myoblasts, not interfering with basic cell functions.
Collapse
Affiliation(s)
| | - Tomasz Szymanski
- Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland.,Faculty of Chemistry, Adam Mickiewicz University, Poznan, Poland.,Wielkopolska Centre of Advanced Technologies, Adam Mickiewicz University, Poznan, Poland
| | | | - Jakub D Rybka
- Faculty of Chemistry, Adam Mickiewicz University, Poznan, Poland.,Wielkopolska Centre of Advanced Technologies, Adam Mickiewicz University, Poznan, Poland
| | - Agnieszka Zimna
- Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| | - Tomasz Zalewski
- NanoBioMedical Centre, Adam Mickiewicz University, Poznan, Poland
| | | | - Agnieszka Malcher
- Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| | | | - Michal Krupinski
- The Henryk Niewodniczanski Institute, Institute of Nuclear Physics Polish Academy of Sciences, Cracow, Poland
| | - Michal Fiedorowicz
- Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Piotr Bogorodzki
- Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Pawel Grieb
- Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Michal Giersig
- Faculty of Chemistry, Adam Mickiewicz University, Poznan, Poland.,Wielkopolska Centre of Advanced Technologies, Adam Mickiewicz University, Poznan, Poland.,Institute of Experimental Physics, Freie Universität Berlin, Berlin, Germany
| | - Maciej K Kurpisz
- Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland.
| |
Collapse
|
10
|
Zhao X, Chen H, Xiao D, Yang H, Itzhaki I, Qin X, Chour T, Aguirre A, Lehmann K, Kim Y, Shukla P, Holmström A, Zhang JZ, Zhuge Y, Ndoye BC, Zhao M, Neofytou E, Zimmermann WH, Jain M, Wu JC. Comparison of Non-human Primate versus Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes for Treatment of Myocardial Infarction. Stem Cell Reports 2018; 10:422-435. [PMID: 29398480 PMCID: PMC5830958 DOI: 10.1016/j.stemcr.2018.01.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 01/03/2018] [Accepted: 01/04/2018] [Indexed: 12/21/2022] Open
Abstract
Non-human primates (NHPs) can serve as a human-like model to study cell therapy using induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs). However, whether the efficacy of NHP and human iPSC-CMs is mechanistically similar remains unknown. To examine this, RNU rats received intramyocardial injection of 1 × 107 NHP or human iPSC-CMs or the same number of respective fibroblasts or PBS control (n = 9-14/group) at 4 days after 60-min coronary artery occlusion-reperfusion. Cardiac function and left ventricular remodeling were similarly improved in both iPSC-CM-treated groups. To mimic the ischemic environment in the infarcted heart, both cultured NHP and human iPSC-CMs underwent 24-hr hypoxia in vitro. Both cells and media were collected, and similarities in transcriptomic as well as metabolomic profiles were noted between both groups. In conclusion, both NHP and human iPSC-CMs confer similar cardioprotection in a rodent myocardial infarction model through relatively similar mechanisms via promotion of cell survival, angiogenesis, and inhibition of hypertrophy and fibrosis.
Collapse
Affiliation(s)
- Xin Zhao
- Stanford Cardiovascular Institute, Stanford, CA 94305-5454, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA 94305, USA
| | - Haodong Chen
- Stanford Cardiovascular Institute, Stanford, CA 94305-5454, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA 94305, USA
| | - Dan Xiao
- Stanford Cardiovascular Institute, Stanford, CA 94305-5454, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA 94305, USA
| | - Huaxiao Yang
- Stanford Cardiovascular Institute, Stanford, CA 94305-5454, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA 94305, USA
| | - Ilanit Itzhaki
- Stanford Cardiovascular Institute, Stanford, CA 94305-5454, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA 94305, USA
| | - Xulei Qin
- Stanford Cardiovascular Institute, Stanford, CA 94305-5454, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA 94305, USA
| | - Tony Chour
- Stanford Cardiovascular Institute, Stanford, CA 94305-5454, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA 94305, USA
| | - Aitor Aguirre
- Departments of Medicine and Pharmacology, University of California, San Diego, CA 92093, USA
| | - Kim Lehmann
- Departments of Medicine and Pharmacology, University of California, San Diego, CA 92093, USA
| | - Youngkyun Kim
- Stanford Cardiovascular Institute, Stanford, CA 94305-5454, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA 94305, USA
| | - Praveen Shukla
- Stanford Cardiovascular Institute, Stanford, CA 94305-5454, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA 94305, USA
| | - Alexandra Holmström
- Stanford Cardiovascular Institute, Stanford, CA 94305-5454, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA 94305, USA
| | - Joe Z Zhang
- Stanford Cardiovascular Institute, Stanford, CA 94305-5454, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA 94305, USA
| | - Yan Zhuge
- Stanford Cardiovascular Institute, Stanford, CA 94305-5454, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA 94305, USA
| | - Babacar C Ndoye
- Stanford Cardiovascular Institute, Stanford, CA 94305-5454, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA 94305, USA
| | - Mingtao Zhao
- Stanford Cardiovascular Institute, Stanford, CA 94305-5454, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA 94305, USA
| | - Evgenios Neofytou
- Stanford Cardiovascular Institute, Stanford, CA 94305-5454, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA 94305, USA
| | - Wolfram-Hubertus Zimmermann
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, 37075 Goettingen, Germany; DZHK (German Center for Cardiovascular Research), Partner Site, Goettingen, Germany
| | - Mohit Jain
- Departments of Medicine and Pharmacology, University of California, San Diego, CA 92093, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford, CA 94305-5454, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
11
|
Zhou J, Sharkey J, Shukla R, Plagge A, Murray P. Assessing the Effectiveness of a Far-Red Fluorescent Reporter for Tracking Stem Cells In Vivo. Int J Mol Sci 2017; 19:E19. [PMID: 29271879 PMCID: PMC5795970 DOI: 10.3390/ijms19010019] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 12/15/2017] [Accepted: 12/20/2017] [Indexed: 12/13/2022] Open
Abstract
Far-red fluorescent reporter genes can be used for tracking cells non-invasively in vivo using fluorescence imaging. Here, we investigate the effectiveness of the far-red fluorescent protein, E2-Crimson (E2C), for tracking mouse embryonic cells (mESCs) in vivo following subcutaneous administration into mice. Using a knock-in strategy, we introduced E2C into the Rosa26 locus of an E14-Bra-GFP mESC line, and after confirming that the E2C had no obvious effect on the phenotype of the mESCs, we injected them into mice and imaged them over nine days. The results showed that fluorescence intensity was weak, and cells could only be detected when injected at high densities. Furthermore, intensity peaked on day 4 and then started to decrease, despite the fact that tumour volume continued to increase beyond day 4. Histopathological analysis showed that although E2C fluorescence could barely be detected in vivo at day 9, analysis of frozen sections indicated that all mESCs within the tumours continued to express E2C. We hypothesise that the decrease in fluorescence intensity in vivo was probably due to the fact that the mESC tumours became more vascular with time, thus leading to increased absorbance of E2C fluorescence by haemoglobin. We conclude that the E2C reporter has limited use for tracking cells in vivo, at least when introduced as a single copy into the Rosa26 locus.
Collapse
Affiliation(s)
- Jing Zhou
- Institute of Translational Medicine, University of Liverpool, Liverpool L69 3BX, UK.
| | - Jack Sharkey
- Institute of Translational Medicine, University of Liverpool, Liverpool L69 3BX, UK.
- Centre for Preclinical Imaging, University of Liverpool, Liverpool L69 3GE, UK.
| | - Rajeev Shukla
- Alder Hey Children's NHS Foundation Trust, Liverpool L12 2AP, UK.
| | - Antonius Plagge
- Institute of Translational Medicine, University of Liverpool, Liverpool L69 3BX, UK.
| | - Patricia Murray
- Institute of Translational Medicine, University of Liverpool, Liverpool L69 3BX, UK.
- Centre for Preclinical Imaging, University of Liverpool, Liverpool L69 3GE, UK.
| |
Collapse
|
12
|
Imaizumi M, Li-Jessen NY, Sato Y, Yang DT, Thibeault SL. Retention of Human-Induced Pluripotent Stem Cells (hiPS) With Injectable HA Hydrogels for Vocal Fold Engineering. Ann Otol Rhinol Laryngol 2017; 126:304-314. [DOI: 10.1177/0003489417691296] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Objective: One prospective treatment option for vocal fold scarring is regeneration with an engineered scaffold containing induced pluripotent stem cells (iPS). In the present study, we investigated the feasibility of utilizing an injectable hyaluronic acid (HA) scaffold encapsulated with human-iPS cell (hiPS) for regeneration of vocal folds. Methods: Thirty athymic nude rats underwent unilateral vocal fold injury. Contralateral vocal folds served as uninjured controls. Hyaluronic acid hydrogel scaffold, HA hydrogel scaffold containing hiPS, and HA hydrogel scaffold containing hiPS with epidermal growth factor (EGF) were injected in both vocal folds immediately after surgery. One and 2 weeks after injection, larynges were excised for histology, immunohistochemistry, and fluorescence in situ hybridization (FISH). Results: Presence of HA hydrogel was confirmed in vocal folds 1 and 2 weeks post injection. The FISH analysis confirmed the presence and viability of hiPS in the injected vocal folds. Histological results demonstrated that vocal folds injected with HA hydrogel scaffold containing EGF demonstrated less fibrosis than those with HA hydrogel only. Conclusions: Human-iPS survived in injured rat vocal folds. The HA hydrogel with hiPS and EGF ameliorated the fibrotic response. Additional work is necessary to optimize hiPS differentiation and further confirm the safety of hiPS for clinical applications.
Collapse
Affiliation(s)
- Mitsuyoshi Imaizumi
- Department of Otolaryngology, School of Medicine, Fukushima Medical University, Fukushima City, Japan
| | - Nicole Y.K. Li-Jessen
- School of Communication Sciences and Disorders, McGill University, Montreal, QC, Canada
| | - Yuka Sato
- Department of Otolaryngology, School of Medicine, Fukushima Medical University, Fukushima City, Japan
| | - David T. Yang
- Department of Pathology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Susan L. Thibeault
- Division of Otolaryngology-Head and Neck Surgery, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
13
|
Abstract
Genome-editing tools, which include zinc finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated 9 (Cas9) systems, have emerged as an invaluable technology to achieve somatic and germline genomic manipulation in cells and model organisms for multiple applications, including the creation of knockout alleles, introducing desired mutations into genomic DNA, and inserting novel transgenes. Genome editing is being rapidly adopted into all fields of biomedical research, including the cardiovascular field, where it has facilitated a greater understanding of lipid metabolism, electrophysiology, cardiomyopathies, and other cardiovascular disorders, has helped to create a wider variety of cellular and animal models, and has opened the door to a new class of therapies. In this Review, we discuss the applications of genome-editing technology throughout cardiovascular disease research and the prospect of in vivo genome-editing therapies in the future. We also describe some of the existing limitations of genome-editing tools that will need to be addressed if cardiovascular genome editing is to achieve its full scientific and therapeutic potential.
Collapse
|
14
|
Goodfellow F, Simchick GA, Mortensen LJ, Stice SL, Zhao Q. Tracking and Quantification of Magnetically Labeled Stem Cells using Magnetic Resonance Imaging. ADVANCED FUNCTIONAL MATERIALS 2016; 26:3899-3915. [PMID: 28751853 PMCID: PMC5526633 DOI: 10.1002/adfm.201504444] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Stem cell based therapies have critical impacts on treatments and cures of diseases such as neurodegenerative or cardiovascular disease. In vivo tracking of stem cells labeled with magnetic contrast agents is of particular interest and importance as it allows for monitoring of the cells' bio-distribution, viability, and physiological responses. Herein, recent advances are introduced in tracking and quantification of super-paramagnetic iron oxide (SPIO) nanoparticles-labeled cells with magnetic resonance imaging, a noninvasive approach that can longitudinally monitor transplanted cells. This is followed by recent translational research on human stem cells that are dual-labeled with green fluorescence protein (GFP) and SPIO nanoparticles, then transplanted and tracked in a chicken embryo model. Cell labeling efficiency, viability, and cell differentiation are also presented.
Collapse
Affiliation(s)
| | - Gregory A Simchick
- Bioimaging Research Center, Regenerative Bioscience Center, and Department of Physics University of Georgia, Athens, GA. 30602, USA
| | | | | | - Qun Zhao
- Bioimaging Research Center, Regenerative Bioscience Center, and Department of Physics University of Georgia, Athens, GA. 30602, USA
| |
Collapse
|
15
|
Wei ZZ, Lee JH, Zhang Y, Zhu YB, Deveau TC, Gu X, Winter MM, Li J, Wei L, Yu SP. Intracranial Transplantation of Hypoxia-Preconditioned iPSC-Derived Neural Progenitor Cells Alleviates Neuropsychiatric Defects After Traumatic Brain Injury in Juvenile Rats. Cell Transplant 2016; 25:797-809. [PMID: 26766038 DOI: 10.3727/096368916x690403] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Traumatic brain injury (TBI) is a common cause of mortality and long-term morbidity in children and adolescents. Posttraumatic stress disorder (PTSD) frequently develops in these patients, leading to a variety of neuropsychiatric syndromes. Currently, few therapeutic strategies are available to treat juveniles with PTSD and other developmental neuropsychiatric disorders. In the present investigation, postnatal day 14 (P14) Wistar rats were subjected to TBI induced by a controlled cortical impact (CCI) (velocity = 3 m/s, depth = 2.0 mm, contact time = 150 ms). This TBI injury resulted in not only cortical damages, but also posttrauma social behavior deficits. Three days after TBI, rats were treated with intracranial transplantation of either mouse iPSC-derived neural progenitor cells under normal culture conditions (N-iPSC-NPCs) or mouse iPSC-derived neural progenitor cells pretreated with hypoxic preconditioning (HP-iPSC-NPCs). Compared to TBI animals that received N-iPSC-NPCs or vehicle treatment, HP-iPSC-NPC-transplanted animals showed a unique benefit of improved performance in social interaction, social novelty, and social transmission of food preference tests. Western blotting showed that HP-iPSC-NPCs expressed significantly higher levels of the social behavior-related genes oxytocin and the oxytocin receptor. Overall, HP-iPSC-NPC transplantation exhibits a great potential as a regenerative therapy to improve neuropsychiatric outcomes after juvenile TBI.
Collapse
Affiliation(s)
- Zheng Zachory Wei
- Laboratories of Stem Cell Biology and Regenerative Medicine, Department of Neurology, Experimental Research Center and Neurological Disease Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Li X, Wang YN, Jin ZY. Molecular imaging of stem cells for the treatment of acute myocardial infarction. Int J Clin Exp Med 2015; 8:8938-8947. [PMID: 26309546 PMCID: PMC4538052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 06/10/2015] [Indexed: 06/04/2023]
Abstract
Stem cell therapy has a unique potential and promises hope for the treatment of acute myocardial infarction. Preclinical studies have identified barriers to clinical translation, one of which involves the monitoring of transplanted cells and the elucidation of their fates in vivo. Molecular imaging may help the solutions for these challenges. In this review, we illustrate the mechanisms by which molecular imaging enables insights into and the development of stem cell therapy.
Collapse
Affiliation(s)
- Xiao Li
- Department of Radiology, PUMC Hospital, CAMS and PUMC Beijing, China
| | - Yi-Ning Wang
- Department of Radiology, PUMC Hospital, CAMS and PUMC Beijing, China
| | - Zheng-Yu Jin
- Department of Radiology, PUMC Hospital, CAMS and PUMC Beijing, China
| |
Collapse
|
17
|
Perbellini F, Carr CA. Uterine cells-an immunoprivileged cell source for therapy-but are they for everyone? J Mol Cell Cardiol 2015; 85:127-30. [PMID: 26027783 DOI: 10.1016/j.yjmcc.2015.05.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 05/21/2015] [Accepted: 05/22/2015] [Indexed: 10/23/2022]
Affiliation(s)
| | - Carolyn A Carr
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford, UK.
| |
Collapse
|