51
|
Mester A, Oltean-Péter B, Rodean I, Opincariu D, Stănescu A, Lázár E, Benedek I, Benedek I, Benedek I. Magnetic Resonance Imaging of Myocardial Function Following Intracoronary and Intramyocardial Stem Cell Injection. JOURNAL OF INTERDISCIPLINARY MEDICINE 2017. [DOI: 10.1515/jim-2017-0053] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Abstract
Stem cell-based therapy is a new therapeutic option that can be used in patients with cardiac diseases caused by myocardial injury. Cardiac magnetic resonance imaging (MRI) is a new noninvasive imaging method with an increasingly widespread indication. The aim of this review was to evaluate the role of cardiac MRI in patients with myocardial infarction undergoing stem cell therapy. We studied the role of MRI in the assessment of myocardial viability, stem cell tracking, assessment of cell survival rate, and monitoring of the long-term effects of stem cell therapy. Based on the current knowledge in this field, this noninvasive, in vivo cardiac imaging technique has a large indication in this group of patients and plays an important role in all stages of stem cell therapy, from the indication to the long-term follow-up of patients.
Collapse
Affiliation(s)
- András Mester
- University of Medicine and Pharmacy , Tîrgu Mureș , Romania
- Center of Advanced Research in Multimodality Cardiac Imaging , Cardio Med Medical Center , Tîrgu Mureș , Romania
| | - Balázs Oltean-Péter
- University of Medicine and Pharmacy , Tîrgu Mureș , Romania
- Center of Advanced Research in Multimodality Cardiac Imaging , Cardio Med Medical Center , Tîrgu Mureș , Romania
| | - Ioana Rodean
- Center of Advanced Research in Multimodality Cardiac Imaging , Cardio Med Medical Center , Tîrgu Mureș , Romania
| | - Diana Opincariu
- University of Medicine and Pharmacy , Tîrgu Mureș , Romania
- Center of Advanced Research in Multimodality Cardiac Imaging , Cardio Med Medical Center , Tîrgu Mureș , Romania
| | - Alexandra Stănescu
- University of Medicine and Pharmacy , Tîrgu Mureș , Romania
- Center of Advanced Research in Multimodality Cardiac Imaging , Cardio Med Medical Center , Tîrgu Mureș , Romania
| | - Erzsébet Lázár
- University of Medicine and Pharmacy , Tîrgu Mureș , Romania
- Clinic of Hematology and Bone Marrow Transplantation Unit , Tîrgu Mureș , Romania
| | - István Benedek
- University of Medicine and Pharmacy , Tîrgu Mureș , Romania
- Clinic of Hematology and Bone Marrow Transplantation Unit , Tîrgu Mureș , Romania
| | - Imre Benedek
- University of Medicine and Pharmacy , Tîrgu Mureș , Romania
- Center of Advanced Research in Multimodality Cardiac Imaging , Cardio Med Medical Center , Tîrgu Mureș , Romania
| | - István Benedek
- University of Medicine and Pharmacy , Tîrgu Mureș , Romania
- Clinic of Hematology and Bone Marrow Transplantation Unit , Tîrgu Mureș , Romania
| |
Collapse
|
52
|
Zhang Z, Yang C, Shen M, Yang M, Jin Z, Ding L, Jiang W, Yang J, Chen H, Cao F, Hu T. Autophagy mediates the beneficial effect of hypoxic preconditioning on bone marrow mesenchymal stem cells for the therapy of myocardial infarction. Stem Cell Res Ther 2017; 8:89. [PMID: 28420436 PMCID: PMC5395756 DOI: 10.1186/s13287-017-0543-0] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 03/04/2017] [Accepted: 03/23/2017] [Indexed: 12/12/2022] Open
Abstract
Background Stem cell therapy has emerged as a promising therapeutic strategy for myocardial infarction (MI). However, the poor viability of transplanted stem cells hampers their therapeutic efficacy. Hypoxic preconditioning (HPC) can effectively promote the survival of stem cells. The aim of this study was to investigate whether HPC improved the functional survival of bone marrow mesenchymal stem cells (BM-MSCs) and increased their cardiac protective effect. Methods BM-MSCs, isolated from Tg(Fluc-egfp) mice which constitutively express both firefly luciferase (Fluc) and enhanced green fluorescent protein (eGFP), were preconditioned with HPC (1% O2) for 12 h, 24 h, 36 h, and 48 h, respectively, followed by 24 h of hypoxia and serum deprivation (H/SD) injury. Results HPC dose-dependently increased the autophagy in BM-MSCs. However, the protective effects of HPC for 24 h are most pronounced. Moreover, hypoxic preconditioned BM-MSCs (HPCMSCs) and nonhypoxic preconditioned BM-MSCs (NPCMSCs) were transplanted into infarcted hearts. Longitudinal in vivo bioluminescence imaging (BLI) and immunofluorescent staining revealed that HPC enhanced the survival of engrafted BM-MSCs. Furthermore, HPCMSCs significantly reduced fibrosis, decreased apoptotic cardiomyocytes, and preserved heart function. However, the beneficial effect of HPC was abolished by autophagy inhibition with 3-methyladenine (3-MA) and Atg7siRNA. Conclusion This study demonstrates that HPC may improve the functional survival and the therapeutic efficiencies of engrafted BM-MSCs, at least in part through autophagy regulation. Hypoxic preconditioning may serve as a promising strategy for optimizing cell-based cardiac regenerative therapy. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0543-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zheng Zhang
- Department of Cardiology, The General Hospital of the PLA Rocket Force, Beijing, 100088, China
| | - Chao Yang
- Department of Blood Transfusion, The General Hospital of the PLA Rocket Force, Beijing, 100088, China
| | - Mingzhi Shen
- Department of Cardiology, Hainan Branch of PLA General Hospital, Sanya, 572013, China
| | - Ming Yang
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 201306, China.,School of Basic Medical Sciences, Taishan Medical University, Taian, Shandong, 271000, China
| | - Zhitao Jin
- Department of Cardiology, The General Hospital of the PLA Rocket Force, Beijing, 100088, China
| | - Liping Ding
- Department of Cardiology, The General Hospital of the PLA Rocket Force, Beijing, 100088, China
| | - Wei Jiang
- Department of Cardiology, The General Hospital of the PLA Rocket Force, Beijing, 100088, China
| | - Junke Yang
- Department of Cardiology, The General Hospital of the PLA Rocket Force, Beijing, 100088, China
| | - Haixu Chen
- Core Laboratory of Translational Medicine, Institute of Geriatrics, PLA general Hospital, Beijing, 100853, China
| | - Feng Cao
- Department of Cardiology, The General Hospital of Chinese People's Liberation Army, Beijing, 100853, China.
| | - Taohong Hu
- Department of Cardiology, The General Hospital of the PLA Rocket Force, Beijing, 100088, China.
| |
Collapse
|
53
|
Kang KT, Lin RZ, Kuppermann D, Melero-Martin JM, Bischoff J. Endothelial colony forming cells and mesenchymal progenitor cells form blood vessels and increase blood flow in ischemic muscle. Sci Rep 2017; 7:770. [PMID: 28396600 PMCID: PMC5429692 DOI: 10.1038/s41598-017-00809-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 03/16/2017] [Indexed: 11/22/2022] Open
Abstract
Here we investigated whether endothelial colony forming cells (ECFC) and mesenchymal progenitor cells (MPC) form vascular networks and restore blood flow in ischemic skeletal muscle, and whether host myeloid cells play a role. ECFC + MPC, ECFC alone, MPC alone, or vehicle alone were injected into the hind limb ischemic muscle one day after ligation of femoral artery and vein. At day 5, hind limbs injected with ECFC + MPC showed greater blood flow recovery compared with ECFC, MPC, or vehicle. Tail vein injection of human endothelial specific Ulex europaeus agglutinin-I demonstrated an increased number of perfused human vessels in ECFC + MPC compared with ECFC. In vivo bioluminescence imaging showed ECFC persisted for 14 days in ECFC + MPC-injected hind limbs. Flow cytometric analysis of ischemic muscles at day 2 revealed increased myeloid lineage cells in ECFC + MPC-injected muscles compared to vehicle-injected muscles. Neutrophils declined by day 7, while the number of myeloid cells, macrophages, and monocytes did not. Systemic myeloid cell depletion with anti-Gr-1 antibody blocked the improved blood flow observed with ECFC + MPC and reduced ECFC and MPC retention. Our data suggest that ECFC + MPC delivery could be used to reestablish blood flow in ischemic tissues, and this may be enhanced by coordinated recruitment of host myeloid cells.
Collapse
Affiliation(s)
- Kyu-Tae Kang
- Vascular Biology Program and Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.,College of Pharmacy and Innovative Drug Center, Duksung Women's University, Seoul, Republic of Korea.,College of Pharmacy and Innovative Drug Center, Duksung Women's University, Pharmacy building (Room 423), 33, Samyangro 144-gil, Dobong Gu, Seoul, South Korea
| | - Ruei-Zeng Lin
- Department of Cardiac Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - David Kuppermann
- Vascular Biology Program and Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.,Harvard Medical School, Boston, MA, USA
| | - Juan M Melero-Martin
- Department of Cardiac Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Joyce Bischoff
- Vascular Biology Program and Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
54
|
Saeed M, Liu H, Liang CH, Wilson MW. Magnetic resonance imaging for characterizing myocardial diseases. Int J Cardiovasc Imaging 2017; 33:1395-1414. [PMID: 28364177 DOI: 10.1007/s10554-017-1127-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 03/23/2017] [Indexed: 12/21/2022]
Abstract
The National Institute of Health defined cardiomyopathy as diseases of the heart muscle. These myocardial diseases have different etiology, structure and treatment. This review highlights the key imaging features of different myocardial diseases. It provides information on myocardial structure/orientation, perfusion, function and viability in diseases related to cardiomyopathy. The standard cardiac magnetic resonance imaging (MRI) sequences can reveal insight on left ventricular (LV) mass, volumes and regional contractile function in all types of cardiomyopathy diseases. Contrast enhanced MRI sequences allow visualization of different infarct patterns and sizes. Enhancement of myocardial inflammation and infarct (location, transmurality and pattern) on contrast enhanced MRI have been used to highlight the key differences in myocardial diseases, predict recovery of function and healing. The common feature in many forms of cardiomyopathy is the presence of diffuse-fibrosis. Currently, imaging sequences generating the most interest in cardiomyopathy include myocardial strain analysis, tissue mapping (T1, T2, T2*) and extracellular volume (ECV) estimation techniques. MRI sequences have the potential to decode the etiology by showing various patterns of infarct and diffuse fibrosis in myocarditis, amyloidosis, sarcoidosis, hypertrophic cardiomyopathy due to aortic stenosis, restrictive cardiomyopathy, arrythmogenic right ventricular dysplasia and hypertension. Integrated PET/MRI system may add in the future more information for the diagnosis and progression of cardiomyopathy diseases. With the promise of high spatial/temporal resolution and 3D coverage, MRI will be an indispensible tool in diagnosis and monitoring the benefits of new therapies designed to treat myocardial diseases.
Collapse
Affiliation(s)
- Maythem Saeed
- Department of Radiology and Biomedical Imaging, School of Medicine, University of California San Francisco, 185 Berry Street, Suite 350, Campus Box 0946, San Francisco, CA, 94107-5705, USA.
| | - Hui Liu
- Department of Radiology, Guangdong General Hospital, Guangzhou, China
| | - Chang-Hong Liang
- Department of Radiology, Guangdong General Hospital, Guangzhou, China
| | - Mark W Wilson
- Department of Radiology and Biomedical Imaging, School of Medicine, University of California San Francisco, 185 Berry Street, Suite 350, Campus Box 0946, San Francisco, CA, 94107-5705, USA
| |
Collapse
|
55
|
Chen HY, Strappe PM, Wang LX. Stem Cell Therapies for Cardiovascular Diseases: What Does the Future Hold? Heart Lung Circ 2017; 26:205-208. [DOI: 10.1016/j.hlc.2016.12.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
56
|
Huang W, Fan W, Wang Y, Han D, Li X, Li S, Li C, Xu B, Huang Y, Fu X, Cao F. Mesenchymal stem cells in alleviating sepsis-induced mice cardiac dysfunction via inhibition of mTORC1-p70S6K signal pathway. Cell Death Discov 2017; 3:16097. [PMID: 28250969 PMCID: PMC5327616 DOI: 10.1038/cddiscovery.2016.97] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 10/14/2016] [Accepted: 10/21/2016] [Indexed: 12/30/2022] Open
Abstract
Sepsis-induced cardiac dysfunction remains a major cause of morbidity and mortality in patients suffered from severe trauma. Mesenchymal stem cells (MSCs) -based treatment has been verified as a promising approach to mitigate the sepsis-induced cardiac dysfunction, but the mechanism is still ambiguous. Thus, our study was designed to explore the potential role of MSCs in sepsis-induced cardiac dysfunction. In vivo bioluminescence imaging revealed 80% acute donor cell death of bone marrow-derived MSCs (BM-MSCs) within 3 days after transplantation. However, echocardiography demonstrated that systolic function in wild-type mice group were reduced after sepsis, while the cardiac function was relatively well persevered in cardiac-conditional deletion of Raptor (component of mTORC1 complex) mice group. Raptor KO group treated with BM-MSCs appeared better cardiac function than other groups (P<0.05). In vitro cell study revealed that co-culture of H9C2 (Raptor-Knock down) and BM-MSC could attenuate the level of proinflammatory cytokines and promote the expression of anti-inflammatory cytokine accompanied by mTORC2-Akt activation (P<0.05). In contrast, co-culture H9C2 (Raptor-O.E) and BM-MSC could aggravate the inflammatory response accompanied by the activation of mTORC1-p70S6K and inhibition of mTORC2-Akt (P<0.05). The immunomodulatory property of MSC is related to the inhibition of mTORC1-p70S6K and activation of mTORC2-Akt signaling pathway. mTORC1-p70S6K and mTORC2-Akt pathways were involved in the therapeutic adjuncts of MSC. The possible mechanism due to MSC`s immunomodulatory property through activation of mTORC2-Akt and inhibition of mTORC1-p70S6K signal pathways which may lead to modulate the expression of inflammation cytokines.
Collapse
Affiliation(s)
- Wei Huang
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China; Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Shanxi, China
| | - Wensi Fan
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China; Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Shanxi, China
| | - Yabin Wang
- Department of Cardiology, Chinese PLA General Hospital , Beijing, China
| | - Dong Han
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University , Shanxi, China
| | - Xiujuan Li
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University , Shanxi, China
| | - Shuang Li
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University , Shanxi, China
| | - Congye Li
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University , Shanxi, China
| | - Bin Xu
- Department of Cardiology, Chinese PLA General Hospital , Beijing, China
| | - Yuesheng Huang
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, The Third Military Medical University , Chongqing, China
| | - Xiaobin Fu
- Department of Cardiology, Chinese PLA General Hospital , Beijing, China
| | - Feng Cao
- Department of Cardiology, Chinese PLA General Hospital , Beijing, China
| |
Collapse
|
57
|
|
58
|
Sheng L, Mao X, Yu Q, Yu D. Effect of the PI3K/AKT signaling pathway on hypoxia-induced proliferation and differentiation of bone marrow-derived mesenchymal stem cells. Exp Ther Med 2016; 13:55-62. [PMID: 28123468 PMCID: PMC5245145 DOI: 10.3892/etm.2016.3917] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 09/09/2016] [Indexed: 12/15/2022] Open
Abstract
Bone marrow-derived mesenchymal stem cell (BM-MSC) transplantation has been demonstrated to be an effective way of augmenting angiogenesis of ischemic tissue. The low oxygen conditions in ischemic tissue directly affect the biological behavior of engrafted cells. However, to date, the mechanism through which hypoxia regulates self-renewal, differentiation and paracrine function of BM-MSCs remains unclear. Clarification of this mechanism would be beneficial to the use of stem cell-based therapy. The PI3K/AKT pathway has been extensively investigated for its role in cell proliferation, cell transformation, paracrine function and angiogenesis. The present study aimed to analyze the role of PI3K/AKT pathway in hypoxia-induced proliferation of BM-MSCs and their differentiation into endothelial cells in vitro by the application of LY294002, a PI3K/AKT pathway inhibitor, with cells cultured in normoxia serving as a control. The results showed that rat BM-MSCs at passage 3 and 4 displayed only few phenotypical differences in the expression of surface antigens as detected by flow cytometry. When compared with the cells treated in normoxia, the proliferation of BM-MSCs in hypoxia was promoted, a greater number of cells expressed CD31 and a higher expression of vascular endothelial growth factor was observed after culture in hypoxic conditions. However, by inhibiting with LY294002, these changes induced by hypoxia were partly inhibited. In conclusion, the present study showed that the PI3K/AKT pathway served an important role in hypoxia-enhanced in vitro proliferation of BM-MSCs and their differentiation into endothelial cells and paracrine vascular endothelial growth factor.
Collapse
Affiliation(s)
- Lingling Sheng
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, P.R. China
| | - Xiyuan Mao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, P.R. China
| | - Qingxiong Yu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, P.R. China
| | - Dong Yu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, P.R. China
| |
Collapse
|
59
|
Momeni A, Neelamegham S, Parashurama N. Current challenges for the targeted delivery and molecular imaging of stem cells in animal models. Bioengineered 2016; 8:316-324. [PMID: 27813700 PMCID: PMC5553333 DOI: 10.1080/21655979.2016.1233090] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
In contrast to conventional, molecular medicine that focuses on targeting specific pathways, stem cell therapy aims to perturb many related mechanisms in order to derive therapeutic benefit. This emerging modality is inherently complex due to the variety of cell types that can be used, delivery approaches that need to be optimized in order to target the cellular therapeutic to specific sites in vivo, and non-invasive imaging methods that are needed to monitor cell fate. This review highlights advancements in the field, with focus on recent publications that use preclinical animal models for cardiovascular stem cell therapy. It highlights studies where cell adhesion engineering (CAE) has been used to functionalize stem cells to home them to sites of therapy, much like peripheral blood neutrophils. It also describes the current state of molecular imaging approaches that aim to non-invasively track the spatio-temporal pattern of stem cell delivery in living subjects.
Collapse
Affiliation(s)
- Arezoo Momeni
- a Department of Chemical and Biological Engineering , University at Buffalo (State University of New York) , Furnas Hall, Buffalo , NY , USA.,b Clinical and Translation Research Center (CTRC) , University at Buffalo (State University of New York) , NY , USA
| | - Sriram Neelamegham
- a Department of Chemical and Biological Engineering , University at Buffalo (State University of New York) , Furnas Hall, Buffalo , NY , USA.,b Clinical and Translation Research Center (CTRC) , University at Buffalo (State University of New York) , NY , USA
| | - Natesh Parashurama
- a Department of Chemical and Biological Engineering , University at Buffalo (State University of New York) , Furnas Hall, Buffalo , NY , USA.,b Clinical and Translation Research Center (CTRC) , University at Buffalo (State University of New York) , NY , USA
| |
Collapse
|
60
|
Xiong R, Joris F, Liang S, De Rycke R, Lippens S, Demeester J, Skirtach A, Raemdonck K, Himmelreich U, De Smedt SC, Braeckmans K. Cytosolic Delivery of Nanolabels Prevents Their Asymmetric Inheritance and Enables Extended Quantitative in Vivo Cell Imaging. NANO LETTERS 2016; 16:5975-5986. [PMID: 27684962 DOI: 10.1021/acs.nanolett.6b01411] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Long-term in vivo imaging of cells is crucial for the understanding of cellular fate in biological processes in cancer research, immunology, or in cell-based therapies such as beta cell transplantation in type I diabetes or stem cell therapy. Traditionally, cell labeling with the desired contrast agent occurs ex vivo via spontaneous endocytosis, which is a variable and slow process that requires optimization for each particular label-cell type combination. Following endocytic uptake, the contrast agents mostly remain entrapped in the endolysosomal compartment, which leads to signal instability, cytotoxicity, and asymmetric inheritance of the labels upon cell division. Here, we demonstrate that these disadvantages can be circumvented by delivering contrast agents directly into the cytoplasm via vapor nanobubble photoporation. Compared to classic endocytic uptake, photoporation resulted in 50 and 3 times higher loading of fluorescent dextrans and quantum dots, respectively, with improved signal stability and reduced cytotoxicity. Most interestingly, cytosolic delivery by photoporation prevented asymmetric inheritance of labels by daughter cells over subsequent cell generations. Instead, unequal inheritance of endocytosed labels resulted in a dramatic increase in polydispersity of the amount of labels per cell with each cell division, hindering accurate quantification of cell numbers in vivo over time. The combined benefits of cell labeling by photoporation resulted in a marked improvement in long-term cell visibility in vivo where an insulin producing cell line (INS-1E cell line) labeled with fluorescent dextrans could be tracked for up to two months in Swiss nude mice compared to 2 weeks for cells labeled by endocytosis.
Collapse
Affiliation(s)
- Ranhua Xiong
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University , 9000 Ghent, Belgium
- Centre for Nano- and Biophotonics, Ghent University , 9000 Ghent, Belgium
| | - Freya Joris
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University , 9000 Ghent, Belgium
| | - Sayuan Liang
- Biomedical NMR Unit, Faculty of Medicine, Katholieke Universiteit Leuven , 3000 Leuven, Belgium
| | - Riet De Rycke
- Inflammation Research Center, Image Core Facility, VIB , 9052 Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University , 9052 Ghent, Belgium
| | - Saskia Lippens
- Inflammation Research Center, Image Core Facility, VIB , 9052 Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University , 9052 Ghent, Belgium
| | - Jo Demeester
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University , 9000 Ghent, Belgium
| | - Andre Skirtach
- Department of Molecular Biotechnology, Ghent University , 9000 Ghent, Belgium
- Max-Planck Institute of Colloids and Interfaces , 14424 Potsdam, Germany
| | - Koen Raemdonck
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University , 9000 Ghent, Belgium
| | - Uwe Himmelreich
- Biomedical NMR Unit, Faculty of Medicine, Katholieke Universiteit Leuven , 3000 Leuven, Belgium
| | - Stefaan C De Smedt
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University , 9000 Ghent, Belgium
| | - Kevin Braeckmans
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University , 9000 Ghent, Belgium
- Centre for Nano- and Biophotonics, Ghent University , 9000 Ghent, Belgium
- Univ Lille 1, Univ Lille Nord France, IEMN, UMR 8520, 59652 Villeneuve Dascq, France
- Univ Lille 1, Univ Lille Nord France, Lab Phys Lasers Atomes & Mol, UMR 8523, 59655 Villeneuve Dascq, France
| |
Collapse
|
61
|
Bedel A, Beliveau F, Lamrissi‐Garcia I, Rousseau B, Moranvillier I, Rucheton B, Guyonnet‐Dupérat V, Cardinaud B, de Verneuil H, Moreau‐Gaudry F, Dabernat S. Preventing Pluripotent Cell Teratoma in Regenerative Medicine Applied to Hematology Disorders. Stem Cells Transl Med 2016; 6:382-393. [PMID: 28191782 PMCID: PMC5442801 DOI: 10.5966/sctm.2016-0201] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 06/27/2016] [Indexed: 12/27/2022] Open
Abstract
Iatrogenic tumorigenesis is a major limitation for the use of human induced pluripotent stem cells (hiPSCs) in hematology. The teratoma risk comes from the persistence of hiPSCs in differentiated cell populations. Our goal was to evaluate the best system to purge residual hiPSCs before graft without compromising hematopoietic repopulation capability. Teratoma risk after systemic injection of hiPSCs expressing the reporter gene luciferase was assessed for the first time. Teratoma formation in immune‐deficient mice was tracked by in vivo bioimaging. We observed that systemic injection of hiPSCs produced multisite teratoma as soon as 5 weeks after injection. To eliminate hiPSCs before grafting, we tested the embryonic‐specific expression of suicide genes under the control of the pmiR‐302/367 promoter. This promoter was highly active in hiPSCs but not in differentiated cells. The gene/prodrug inducible Caspase‐9 (iCaspase‐9)/AP20187 was more efficient and rapid than thymidine kinase/ganciclovir, fully specific, and without bystander effect. We observed that iCaspase‐9‐expressing hiPSCs died in a dose‐dependent manner with AP20187, without reaching full eradication in vitro. Unexpectedly, nonspecific toxicity of AP20187 on iCaspase‐9‐negative hiPSCs and on CD34+ cells was evidenced in vitro. This toxic effect strongly impaired CD34+‐derived human hematopoiesis in adoptive transfers. Survivin inhibition is an alternative to the suicide gene approach because hiPSCs fully rely on survivin for survival. Survivin inhibitor YM155 was more efficient than AP20187/iCaspase‐9 for killing hiPSCs, without toxicity on CD34+ cells, in vitro and in adoptive transfers. hiPSC purge by survivin inhibitor fully eradicated teratoma formation in immune‐deficient mice. This will be useful to improve the safety management for hiPSC‐based medicine. Stem Cells Translational Medicine2017;6:382–393
Collapse
MESH Headings
- Animals
- Caspase 9/genetics
- Caspase 9/metabolism
- Cell Line
- Cell Proliferation
- Cell Transformation, Neoplastic/drug effects
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/pathology
- Dose-Response Relationship, Drug
- Gene Expression Regulation, Neoplastic
- Genes, Transgenic, Suicide
- Hematologic Diseases/surgery
- Hematopoietic Stem Cell Transplantation/adverse effects
- Hematopoietic Stem Cell Transplantation/methods
- Hematopoietic Stem Cells/drug effects
- Hematopoietic Stem Cells/metabolism
- Hematopoietic Stem Cells/pathology
- Humans
- Imidazoles/pharmacology
- Induced Pluripotent Stem Cells/drug effects
- Induced Pluripotent Stem Cells/metabolism
- Induced Pluripotent Stem Cells/pathology
- Induced Pluripotent Stem Cells/transplantation
- Mice, Inbred NOD
- Mice, SCID
- Naphthoquinones/pharmacology
- Phenotype
- Regenerative Medicine/methods
- Risk Assessment
- Survivin/antagonists & inhibitors
- Survivin/metabolism
- Tacrolimus/analogs & derivatives
- Tacrolimus/pharmacology
- Teratoma/genetics
- Teratoma/metabolism
- Teratoma/pathology
- Teratoma/prevention & control
- Time Factors
- Tumor Burden
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Aurelie Bedel
- Université de Bordeaux, Bordeaux, France
- INSERM U1035, Bordeaux, France
- Bordeaux University Hospital, Bordeaux, France
| | - François Beliveau
- Université de Bordeaux, Bordeaux, France
- INSERM U1035, Bordeaux, France
| | | | - Benoit Rousseau
- Université de Bordeaux, Bordeaux, France
- Bordeaux University Hospital, Bordeaux, France
| | | | - Benoit Rucheton
- Université de Bordeaux, Bordeaux, France
- INSERM U1035, Bordeaux, France
| | | | - Bruno Cardinaud
- Université de Bordeaux, Bordeaux, France
- INSERM U1035, Bordeaux, France
| | - Hubert de Verneuil
- Université de Bordeaux, Bordeaux, France
- INSERM U1035, Bordeaux, France
- Bordeaux University Hospital, Bordeaux, France
| | - François Moreau‐Gaudry
- Université de Bordeaux, Bordeaux, France
- INSERM U1035, Bordeaux, France
- Bordeaux University Hospital, Bordeaux, France
- Plateforme de Vectorologie, Université de Bordeaux, Bordeaux, France
| | - Sandrine Dabernat
- Université de Bordeaux, Bordeaux, France
- INSERM U1035, Bordeaux, France
- Bordeaux University Hospital, Bordeaux, France
| |
Collapse
|
62
|
Wu S, Zhu Y, Liu H, Tang L, Du R, Shen Y, Feng J, Zhang K, Xu C, Zhang S, Chen Y, Song F, Zhu Y, Gu W, Liang P, Carrió I, Zhang H, Tian M. In Vivo Dynamic Metabolic Changes After Transplantation of Induced Pluripotent Stem Cells for Ischemic Injury. J Nucl Med 2016; 57:2012-2015. [PMID: 27561881 DOI: 10.2967/jnumed.115.171124] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 08/03/2016] [Indexed: 11/16/2022] Open
Abstract
This study aimed to investigate in vivo dynamic metabolic changes after transplantation of induced pluripotent stem cells (iPSCs) and iPSC-derived enriched cardiomyocytes (iPSC-CMs) in a rat model of ischemic injury. METHODS Serial 18F-FDG PET, echocardiographic, immunohistochemical, and immunofluorescence studies were performed after transplantation of iPSCs and iPSC-CMs and compared with embryonic stem cells (ESCs), ESC-CMs, and a phosphate-buffered saline control group of rats with myocardial infarction. RESULTS Increased glucose metabolism in periinfarct areas and improved myocardial function were observed in the stem cell transplantation groups compared with the control group, and serial immunofluorescence and immunohistochemical results exhibited the survival and migration of stem cells during the study period. CONCLUSION Serial 18F-FDG PET and echocardiographic imaging studies demonstrated the dynamic metabolic changes and recovery of myocardial function after stem cell transplantation. 18F-FDG PET could be a potential approach to evaluating spatiotemporal dynamic metabolic changes in vivo after transplantation of iPSCs or iPSC-CMs for ischemic injury.
Collapse
Affiliation(s)
- Shuang Wu
- Department of Nuclear Medicine and Medical PET Center, Second Hospital of Zhejiang University School of Medicine, Hangzhou, China.,Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Yuankai Zhu
- Department of Nuclear Medicine and Medical PET Center, Second Hospital of Zhejiang University School of Medicine, Hangzhou, China.,Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Hao Liu
- Department of Nuclear Medicine and Medical PET Center, Second Hospital of Zhejiang University School of Medicine, Hangzhou, China.,Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Ling Tang
- First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Translational Medicine, Zhejiang University, Hangzhou, China
| | - Ruili Du
- Department of Nuclear Medicine and Medical PET Center, Second Hospital of Zhejiang University School of Medicine, Hangzhou, China.,Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Yehua Shen
- Department of Nuclear Medicine and Medical PET Center, Second Hospital of Zhejiang University School of Medicine, Hangzhou, China.,Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Jin Feng
- Department of Nuclear Medicine and Medical PET Center, Second Hospital of Zhejiang University School of Medicine, Hangzhou, China.,Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Kai Zhang
- Department of Nuclear Medicine and Medical PET Center, Second Hospital of Zhejiang University School of Medicine, Hangzhou, China.,Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Caiyun Xu
- Department of Nuclear Medicine and Medical PET Center, Second Hospital of Zhejiang University School of Medicine, Hangzhou, China.,Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Shouhong Zhang
- Department of Nuclear Medicine and Medical PET Center, Second Hospital of Zhejiang University School of Medicine, Hangzhou, China.,Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Yao Chen
- Department of Nuclear Medicine and Medical PET Center, Second Hospital of Zhejiang University School of Medicine, Hangzhou, China.,Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Fahuan Song
- Department of Nuclear Medicine and Medical PET Center, Second Hospital of Zhejiang University School of Medicine, Hangzhou, China.,Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Yunqi Zhu
- Department of Nuclear Medicine and Medical PET Center, Second Hospital of Zhejiang University School of Medicine, Hangzhou, China.,Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Weizhong Gu
- Department of Pathology, Children's Hospital of Zhejiang University School of Medicine, Hangzhou, China; and
| | - Ping Liang
- First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Translational Medicine, Zhejiang University, Hangzhou, China
| | - Ignasi Carrió
- Nuclear Medicine Department, Hospital Sant Pau, Autonomous University of Barcelona, Barcelona, Spain
| | - Hong Zhang
- Department of Nuclear Medicine and Medical PET Center, Second Hospital of Zhejiang University School of Medicine, Hangzhou, China.,Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Mei Tian
- Department of Nuclear Medicine and Medical PET Center, Second Hospital of Zhejiang University School of Medicine, Hangzhou, China .,Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| |
Collapse
|
63
|
Wu N, Wang Y, Yang L, Cho KS. Signaling Networks of Retinal Ganglion Cell Formation and the Potential Application of Stem Cell–Based Therapy in Retinal Degenerative Diseases. Hum Gene Ther 2016; 27:609-20. [PMID: 27466076 DOI: 10.1089/hum.2016.083] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Affiliation(s)
- Nan Wu
- 1 Department of Ophthalmology, Southwest Eye Hospital, Southwest Hospital, Third Military Medical University , Chongqing, China
| | - Yi Wang
- 1 Department of Ophthalmology, Southwest Eye Hospital, Southwest Hospital, Third Military Medical University , Chongqing, China
| | - Lanbo Yang
- 2 Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Harvard Medical School , Boston, Massachusetts
| | - Kin-Sang Cho
- 2 Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Harvard Medical School , Boston, Massachusetts
| |
Collapse
|
64
|
Kalimuthu S, Gangadaran P, Li XJ, Oh JM, Lee HW, Jeong SY, Lee SW, Lee J, Ahn BC. In Vivo therapeutic potential of mesenchymal stem cell-derived extracellular vesicles with optical imaging reporter in tumor mice model. Sci Rep 2016; 6:30418. [PMID: 27452924 PMCID: PMC4958922 DOI: 10.1038/srep30418] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 07/05/2016] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs) can be used as a therapeutic armor for cancer. Extracellular vesicles (EVs) from MSCs have been evaluated for anticancer effects. In vivo targeting of EVs to the tumor is an essential requirement for successful therapy. Therefore, non-invasive methods of monitoring EVs in animal models are crucial for developing EV-based cancer therapies. The present study to develop bioluminescent EVs using Renilla luciferase (Rluc)-expressing MSCs. The EVs from MSC/Rluc cells (EV-MSC/Rluc) were visualized in a murine lung cancer model. The anticancer effects of EVs on Lewis lung carcinoma (LLC) and other cancer cells were assessed. EV-MSC/Rluc were visualized in vivo in the LLC-efffuc tumor model using optical imaging. The induction of apoptosis was confirmed with Annexin-V and propidium iodide staining. EV-MSC/Rluc and EV-MSCs showed a significant cytotoxic effect against LLC-effluc cells and 4T1; however, no significant effect on CT26, B16F10, TC1 cells. Moreover, EV-MSC/Rluc inhibited LLC tumor growth in vivo. EV-MSC/Rluc-mediated LLC tumor inhibitory mechanism revealed the decreased pERK and increased cleaved caspase 3 and cleaved PARP. We successfully developed luminescent EV-MSC/Rluc that have a therapeutic effect on LLC cells in both in vitro and in vivo. This bioluminescent EV system can be used to optimize EV-based therapy.
Collapse
Affiliation(s)
- Senthilkumar Kalimuthu
- Department of Nuclear Medicine, Kyungpook National University School of Medicine/Hospital, Daegu 700-721, Republic of Korea
| | - Prakash Gangadaran
- Department of Nuclear Medicine, Kyungpook National University School of Medicine/Hospital, Daegu 700-721, Republic of Korea
| | - Xiu Juan Li
- Department of Nuclear Medicine, Kyungpook National University School of Medicine/Hospital, Daegu 700-721, Republic of Korea
| | - Ji Min Oh
- Department of Nuclear Medicine, Kyungpook National University School of Medicine/Hospital, Daegu 700-721, Republic of Korea
| | - Ho Won Lee
- Department of Nuclear Medicine, Kyungpook National University School of Medicine/Hospital, Daegu 700-721, Republic of Korea
| | - Shin Young Jeong
- Department of Nuclear Medicine, Kyungpook National University School of Medicine/Hospital, Daegu 700-721, Republic of Korea
| | - Sang-Woo Lee
- Department of Nuclear Medicine, Kyungpook National University School of Medicine/Hospital, Daegu 700-721, Republic of Korea
| | - Jaetae Lee
- Department of Nuclear Medicine, Kyungpook National University School of Medicine/Hospital, Daegu 700-721, Republic of Korea
- Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), 80 Cheombok-ro, Dong-gu, Daegu 701-310, Republic of Korea
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, Kyungpook National University School of Medicine/Hospital, Daegu 700-721, Republic of Korea
| |
Collapse
|
65
|
Faivre L, Chaussard M, Vercellino L, Vanneaux V, Hosten B, Teixera K, Parietti V, Merlet P, Sarda-Mantel L, Rizzo-Padoin N, Larghero J. 18F-FDG labelling of hematopoietic stem cells: Dynamic study of bone marrow homing by PET–CT imaging and impact on cell functionality. Curr Res Transl Med 2016; 64:141-148. [DOI: 10.1016/j.retram.2016.06.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 06/02/2016] [Accepted: 06/03/2016] [Indexed: 01/21/2023]
|
66
|
Molecular Imaging for Comparison of Different Growth Factors on Bone Marrow-Derived Mesenchymal Stromal Cells' Survival and Proliferation In Vivo. BIOMED RESEARCH INTERNATIONAL 2016; 2016:1363902. [PMID: 27419126 PMCID: PMC4932172 DOI: 10.1155/2016/1363902] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 01/19/2016] [Accepted: 02/16/2016] [Indexed: 12/24/2022]
Abstract
Introduction. Bone marrow-derived mesenchymal stromal cells (BMSCs) have emerged as promising cell candidates but with poor survival after transplantation. This study was designed to investigate the efficacy of VEGF, bFGF, and IGF-1 on BMSCs' viability and proliferation both in vivo and in vitro using bioluminescence imaging (BLI). Methods. BMSCs were isolated from β-actin-Fluc+ transgenic FVB mice, which constitutively express firefly luciferase. Apoptosis was induced by hypoxia preconditioning for up to 24 h followed by flow cytometry and TUNEL assay. 106 BMSCs with/without growth factors were injected subcutaneously into wild type FVB mice's backs. Survival of BMSCs was longitudinally monitored using bioluminescence imaging (BLI) for 5 weeks. Protein expression of Akt, p-Akt, PARP, and caspase-3 was detected by Western blot. Results. Hypoxia-induced apoptosis was significantly attenuated by bFGF and IGF-1 compared with VEGF and control group in vitro (P < 0.05). When combined with matrigel, IGF-1 showed the most beneficial effects in protecting BMSCs from apoptosis in vivo. The phosphorylation of Akt had a higher ratio in the cells from IGF-1 group. Conclusion. IGF-1 could protect BMSCs from hypoxia-induced apoptosis through activation of p-Akt/Akt pathway.
Collapse
|
67
|
Parashurama N, Ahn BC, Ziv K, Ito K, Paulmurugan R, Willmann JK, Chung J, Ikeno F, Swanson JC, Merk DR, Lyons JK, Yerushalmi D, Teramoto T, Kosuge H, Dao CN, Ray P, Patel M, Chang YF, Mahmoudi M, Cohen JE, Goldstone AB, Habte F, Bhaumik S, Yaghoubi S, Robbins RC, Dash R, Yang PC, Brinton TJ, Yock PG, McConnell MV, Gambhir SS. Multimodality Molecular Imaging of Cardiac Cell Transplantation: Part I. Reporter Gene Design, Characterization, and Optical in Vivo Imaging of Bone Marrow Stromal Cells after Myocardial Infarction. Radiology 2016; 280:815-25. [PMID: 27308957 DOI: 10.1148/radiol.2016140049] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Purpose To use multimodality reporter-gene imaging to assess the serial survival of marrow stromal cells (MSC) after therapy for myocardial infarction (MI) and to determine if the requisite preclinical imaging end point was met prior to a follow-up large-animal MSC imaging study. Materials and Methods Animal studies were approved by the Institutional Administrative Panel on Laboratory Animal Care. Mice (n = 19) that had experienced MI were injected with bone marrow-derived MSC that expressed a multimodality triple fusion (TF) reporter gene. The TF reporter gene (fluc2-egfp-sr39ttk) consisted of a human promoter, ubiquitin, driving firefly luciferase 2 (fluc2), enhanced green fluorescent protein (egfp), and the sr39tk positron emission tomography reporter gene. Serial bioluminescence imaging of MSC-TF and ex vivo luciferase assays were performed. Correlations were analyzed with the Pearson product-moment correlation, and serial imaging results were analyzed with a mixed-effects regression model. Results Analysis of the MSC-TF after cardiac cell therapy showed significantly lower signal on days 8 and 14 than on day 2 (P = .011 and P = .001, respectively). MSC-TF with MI demonstrated significantly higher signal than MSC-TF without MI at days 4, 8, and 14 (P = .016). Ex vivo luciferase activity assay confirmed the presence of MSC-TF on days 8 and 14 after MI. Conclusion Multimodality reporter-gene imaging was successfully used to assess serial MSC survival after therapy for MI, and it was determined that the requisite preclinical imaging end point, 14 days of MSC survival, was met prior to a follow-up large-animal MSC study. (©) RSNA, 2016 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Natesh Parashurama
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Byeong-Cheol Ahn
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Keren Ziv
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Ken Ito
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Ramasamy Paulmurugan
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Jürgen K Willmann
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Jaehoon Chung
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Fumiaki Ikeno
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Julia C Swanson
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Denis R Merk
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Jennifer K Lyons
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - David Yerushalmi
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Tomohiko Teramoto
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Hisanori Kosuge
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Catherine N Dao
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Pritha Ray
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Manishkumar Patel
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Ya-Fang Chang
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Morteza Mahmoudi
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Jeff Eric Cohen
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Andrew Brooks Goldstone
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Frezghi Habte
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Srabani Bhaumik
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Shahriar Yaghoubi
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Robert C Robbins
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Rajesh Dash
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Phillip C Yang
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Todd J Brinton
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Paul G Yock
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Michael V McConnell
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Sanjiv S Gambhir
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| |
Collapse
|
68
|
Zhang Z, Yang M, Wang Y, Wang L, Jin Z, Ding L, Zhang L, Zhang L, Jiang W, Gao G, Yang J, Lu B, Cao F, Hu T. Autophagy regulates the apoptosis of bone marrow-derived mesenchymal stem cells under hypoxic condition via AMP-activated protein kinase/mammalian target of rapamycin pathway. Cell Biol Int 2016; 40:671-85. [PMID: 27005844 DOI: 10.1002/cbin.10604] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 03/19/2016] [Indexed: 12/19/2022]
Abstract
Bone marrow-derived mesenchymal stem cells (BM-MSCs) have been demonstrated as an ideal autologous stem cells source for cell-based therapy for myocardial infarction (MI). However, poor viability of donor stem cells after transplantation limits their therapeutic efficiency, whereas the underlying mechanism is still poorly understood. Autophagy, a highly conserved process of cellular degradation, is required for maintaining homeostasis and normal function. Here, we investigated the potential role of autophagy on apoptosis in BM-MSCs induced by hypoxic injury. BM-MSCs, isolated from male C57BL/6 mice, were subjected to hypoxia and serum deprivation (H/SD) injury for 6, 12, and 24 h, respectively. The autophagy state was regulated by 3-methyladenine (3MA) and rapamycin administration. Furthermore, compound C was administrated to inhibit AMPK. The apoptosis induced by H/SD was determined by TUNEL assays. Meanwhile, autophagy was measured by GFP-LC3 plasmids transfection and transmission electron microscope. Moreover, protein expressions were evaluated by Western blot assay. In the present study, we found that hypoxic stress increased autophagy and apoptosis in BM-MSCs time dependently. Meanwhile, hypoxia increased the activity of AMPK/mTOR signal pathway. Moreover, increased apoptosis in BM-MSCs under hypoxia was abolished by 3-MA, whereas was aggravated by rapamycin. Furthermore, the increased autophagy and apoptosis in BM-MSCs induced by hypoxia were abolished by AMPK inhibitor compound C. These data provide evidence that hypoxia induced AMPK/mTOR signal pathway activation which regulated the apoptosis and autophagy in BM-MSCs. Furthermore, the apoptosis of BM-MSCs under hypoxic condition was regulated by autophagy via AMPK/mTOR pathway.
Collapse
Affiliation(s)
- Zheng Zhang
- Department of Cardiology, The General Hospital of the PLA Rocket Force, Beijing, 100088, China
| | - Ming Yang
- School of Basic Medical Sciences, Taishan Medical University, Taian, Shandong, 271000, China
| | - Yabin Wang
- Department of Cardiology, The General Hospital of Chinese People's Liberation Army, Beijing, 100853, China
| | - Le Wang
- Institute of Orthopaedics and Traumatology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510000, China
| | - Zhitao Jin
- Department of Cardiology, The General Hospital of the PLA Rocket Force, Beijing, 100088, China
| | - Liping Ding
- Department of Cardiology, The General Hospital of the PLA Rocket Force, Beijing, 100088, China
| | - Lijuan Zhang
- Department of Cardiology, The General Hospital of the PLA Rocket Force, Beijing, 100088, China
| | - Lina Zhang
- Department of Cardiology, The General Hospital of the PLA Rocket Force, Beijing, 100088, China
| | - Wei Jiang
- Department of Cardiology, The General Hospital of the PLA Rocket Force, Beijing, 100088, China
| | - Guojie Gao
- Department of Cardiology, The General Hospital of the PLA Rocket Force, Beijing, 100088, China
| | - Junke Yang
- Department of Cardiology, The General Hospital of the PLA Rocket Force, Beijing, 100088, China
| | - Bingwei Lu
- Department of Cardiology, The General Hospital of the PLA Rocket Force, Beijing, 100088, China
| | - Feng Cao
- Department of Cardiology, The General Hospital of Chinese People's Liberation Army, Beijing, 100853, China
| | - Taohong Hu
- Department of Cardiology, The General Hospital of the PLA Rocket Force, Beijing, 100088, China
| |
Collapse
|
69
|
Ding J, Zhao Z, Wang C, Wang CX, Li PC, Qian C, Teng GJ. Bioluminescence imaging of transplanted human endothelial colony-forming cells in an ischemic mouse model. Brain Res 2016; 1642:209-218. [PMID: 27038754 DOI: 10.1016/j.brainres.2016.03.045] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 02/12/2016] [Accepted: 03/28/2016] [Indexed: 01/09/2023]
Abstract
Ischemic strokes are devastating events responsible for high mortality and morbidity worldwide each year. Endothelial colony-forming cell (ECFC) therapy holds promise for stroke treatment; however, grafted ECFCs need to be monitored better understand their biological behavior in vivo, so as to evaluate their safety and successful delivery. The objectives of this study are to visualize the fate of infused human cord blood derived ECFCs via bioluminescence imaging (BLI) in an ischemic stroke mouse model and to determine the therapeutic effects of ECFC transplantation. ECFCs derived from human umbilical cord blood were infected with lentivirus carrying enhanced green fluorescent protein (eGFP) and firefly luciferase (Luc2) double fusion reporter gene. Labeled ECFCs were grafted into a photothrombotic ischemic stroke mouse model via intra-arterial injection though the left cardiac ventricle. The homing of infused cells and functional recovery of stroke mice were evaluated using BLI, neurological scoring, and immunohistochemistry. Significantly, BLI signals were highest in the brain on day 1 and decreased steadily until day 14. GFP-positive cells were also found surrounding infarct border zones in brain sections using immunohistochemical staining, suggesting that ECFCs properly homed to the ischemic brain tissue. Using a modified neurological severity score assay and histological analysis of brain slices with CD31 immunostaining in brain tissue, double cortin analysis, and the TdT-mediated dUTP nick end labeling (TUNEL) assay, we demonstrated functional restoration, improved angiogenesis, neurogenesis, and decreased apoptosis in ischemic mice after ECFC infusion. Collectively, our data support that ECFCs may be a promising therapeutic agent for stroke.
Collapse
Affiliation(s)
- Jie Ding
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Zhen Zhao
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Chao Wang
- Education Ministry's Key Laboratory of Developmental Genes and Human Diseases, Institute of Life Sciences, Southeast University, Nanjing, China
| | - Cong-Xiao Wang
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Pei-Cheng Li
- Department of Interventional Radiology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Cheng Qian
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Gao-Jun Teng
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China.
| |
Collapse
|
70
|
Zhou Y, Zhu Y, Zhang L, Wu T, Wu T, Zhang W, Decker AM, He J, Liu J, Wu Y, Jiang X, Zhang Z, Liang C, Zou D. Human Stem Cells Overexpressing miR-21 Promote Angiogenesis in Critical Limb Ischemia by Targeting CHIP to Enhance HIF-1α Activity. Stem Cells 2016; 34:924-34. [PMID: 26841045 DOI: 10.1002/stem.2321] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 10/12/2015] [Accepted: 11/05/2015] [Indexed: 12/21/2022]
Abstract
Critical limb ischemia (CLI) is a severe blockage in the arteries of the lower extremities. However, the effective and optimal treatment for CLI remains to be elucidated. Previous therapeutic research is mainly focused on proangiogenic growth factors administrations. Recently, miR-21 has been revealed to play a crucial role in angiogenesis. Thus, we hypothesize that miR-21 over-expression in human umbilical cord blood-derived mesenchymal stem cells (UCBMSCs) can effectively treat CLI. Herein, UCBMSCs were transduced with lentivirus-miR-21-Luciferase (Lenti-miR-21) or lentivirus- LacZ-Luciferase (Lenti-LacZ). The results indicated that miR-21 induced UCBMSCs proliferation, migration, and angiogenesis in vitro. Subsequently, general observation and laser Doppler perfusion imaging were introduced to detect perfusion in muscles of CLI-nude mice on 1, 4, 7, 14, and 28 day postoperation. There was a significant improvement in blood vessels of the ischemic limb in Lenti-miR-21 group at 7 day compared with the saline or Lenti-LacZ groups. At 28 day, histological analysis confirmed that UCBMSCs over-expressing miR-21 increased neovascularization in CLI. Furthermore, carboxyl terminus of Hsc70-interacting protein (CHIP) was found to be the target gene for miR-21-mediated activation of hypoxia-inducible factor 1α (HIF-1α) in UCBMSCs. In summary, our study demonstrated that over-expressing miR-21 in UCBMSCs could improve neovascularization in CLI through enhancing HIF-1α activity by targeting CHIP, which may hold great therapeutic promise in treating CLI.
Collapse
Affiliation(s)
- Yong Zhou
- Department of Dental Implant Center, Stomatologic Hospital & College, Anhui Medical University, Key Laboratory of Oral Diseases Research of Anhui Province, Hefei, PR, China
| | - Youming Zhu
- Department of Dental Implant Center, Stomatologic Hospital & College, Anhui Medical University, Key Laboratory of Oral Diseases Research of Anhui Province, Hefei, PR, China
| | - Li Zhang
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Hefei, PR, China
| | - Tao Wu
- Department of Dental Implant Center, Stomatologic Hospital & College, Anhui Medical University, Key Laboratory of Oral Diseases Research of Anhui Province, Hefei, PR, China
| | - Tingting Wu
- Department of Dental Implant Center, Stomatologic Hospital & College, Anhui Medical University, Key Laboratory of Oral Diseases Research of Anhui Province, Hefei, PR, China
| | - Wenjie Zhang
- Department of Oral and Maxillofacial Surgery, Oral Implant, and Prosthodontics, School of Medicine, Ninth People's Hospital Affiliated with Shanghai Jiao Tong University, Shanghai, PR, China
| | - Ann Marie Decker
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry 1210 Eisenhower Place, Ann Arbor, Michigan, USA
| | - Jiacai He
- Department of Dental Implant Center, Stomatologic Hospital & College, Anhui Medical University, Key Laboratory of Oral Diseases Research of Anhui Province, Hefei, PR, China
| | - Jie Liu
- Translational Center for Stem Cell Research, Tongji Hospital, Stem Cell Research Center, Tongji University School of Medicine, Shanghai, PR, China
| | - Yiqun Wu
- Department of Oral and Maxillofacial Surgery, Oral Implant, and Prosthodontics, School of Medicine, Ninth People's Hospital Affiliated with Shanghai Jiao Tong University, Shanghai, PR, China
| | - Xinqun Jiang
- Department of Oral and Maxillofacial Surgery, Oral Implant, and Prosthodontics, School of Medicine, Ninth People's Hospital Affiliated with Shanghai Jiao Tong University, Shanghai, PR, China
| | - Zhiyuan Zhang
- Department of Oral and Maxillofacial Surgery, Oral Implant, and Prosthodontics, School of Medicine, Ninth People's Hospital Affiliated with Shanghai Jiao Tong University, Shanghai, PR, China
| | - Chaozhao Liang
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Hefei, PR, China
| | - Duohong Zou
- Department of Dental Implant Center, Stomatologic Hospital & College, Anhui Medical University, Key Laboratory of Oral Diseases Research of Anhui Province, Hefei, PR, China
| |
Collapse
|
71
|
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
|
72
|
Stem Cell Imaging: Tools to Improve Cell Delivery and Viability. Stem Cells Int 2016; 2016:9240652. [PMID: 26880997 PMCID: PMC4736428 DOI: 10.1155/2016/9240652] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 10/06/2015] [Accepted: 10/15/2015] [Indexed: 01/01/2023] Open
Abstract
Stem cell therapy (SCT) has shown very promising preclinical results in a variety of regenerative medicine applications. Nevertheless, the complete utility of this technology remains unrealized. Imaging is a potent tool used in multiple stages of SCT and this review describes the role that imaging plays in cell harvest, cell purification, and cell implantation, as well as a discussion of how imaging can be used to assess outcome in SCT. We close with some perspective on potential growth in the field.
Collapse
|
73
|
Ong SG, Huber BC, Lee WH, Kodo K, Ebert AD, Ma Y, Nguyen PK, Diecke S, Chen WY, Wu JC. Microfluidic Single-Cell Analysis of Transplanted Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes After Acute Myocardial Infarction. Circulation 2015; 132:762-771. [PMID: 26304668 DOI: 10.1161/circulationaha.114.015231] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Human induced pluripotent stem cells (iPSCs) are attractive candidates for therapeutic use, with the potential to replace deficient cells and to improve functional recovery in injury or disease settings. Here, we test the hypothesis that human iPSC-derived cardiomyocytes (iPSC-CMs) can secrete cytokines as a molecular basis to attenuate adverse cardiac remodeling after myocardial infarction. METHODS AND RESULTS Human iPSCs were generated from skin fibroblasts and differentiated in vitro with a small molecule-based protocol. Troponin(+) iPSC-CMs were confirmed by immunohistochemistry, quantitative polymerase chain reaction, fluorescence-activated cell sorting, and electrophysiological measurements. Afterward, 2×10(6) iPSC-CMs derived from a cell line transduced with a vector expressing firefly luciferase and green fluorescent protein were transplanted into adult NOD/SCID mice with acute left anterior descending artery ligation. Control animals received PBS injection. Bioluminescence imaging showed limited engraftment on transplantation into ischemic myocardium. However, magnetic resonance imaging of animals transplanted with iPSC-CMs showed significant functional improvement and attenuated cardiac remodeling compared with PBS-treated control animals. To understand the underlying molecular mechanism, microfluidic single-cell profiling of harvested iPSC-CMs, laser capture microdissection of host myocardium, and in vitro ischemia stimulation were used to demonstrate that the iPSC-CMs could release significant levels of proangiogenic and antiapoptotic factors in the ischemic microenvironment. CONCLUSIONS Transplantation of human iPSC-CMs into an acute mouse myocardial infarction model can improve left ventricular function and attenuate cardiac remodeling. Because of limited engraftment, most of the effects are possibly explained by paracrine activity of these cells.
Collapse
Affiliation(s)
- Sang-Ging Ong
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA.,Depts of Medicine and Radiology, Stanford University School of Medicine, Stanford, CA.,Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA
| | - Bruno C Huber
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA.,Depts of Medicine and Radiology, Stanford University School of Medicine, Stanford, CA.,Ludwig-Maximilians-University, Medical Department I, Campus Grosshadern, Munich, Germany
| | - Won Hee Lee
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA.,Depts of Medicine and Radiology, Stanford University School of Medicine, Stanford, CA
| | - Kazuki Kodo
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA.,Depts of Medicine and Radiology, Stanford University School of Medicine, Stanford, CA
| | - Antje D Ebert
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA.,Depts of Medicine and Radiology, Stanford University School of Medicine, Stanford, CA
| | - Yu Ma
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA.,Depts of Medicine and Radiology, Stanford University School of Medicine, Stanford, CA
| | - Patricia K Nguyen
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA.,Depts of Medicine and Radiology, Stanford University School of Medicine, Stanford, CA
| | - Sebastian Diecke
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA.,Depts of Medicine and Radiology, Stanford University School of Medicine, Stanford, CA
| | - Wen-Yi Chen
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA.,Depts of Medicine and Radiology, Stanford University School of Medicine, Stanford, CA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA.,Depts of Medicine and Radiology, Stanford University School of Medicine, Stanford, CA.,Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
74
|
Stem cell therapy for heart failure: Out with the new and in with the old? J Thorac Cardiovasc Surg 2015; 150:1035-7. [DOI: 10.1016/j.jtcvs.2015.09.035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 09/11/2015] [Indexed: 12/18/2022]
|
75
|
Den Hartogh SC, Passier R. Concise Review: Fluorescent Reporters in Human Pluripotent Stem Cells: Contributions to Cardiac Differentiation and Their Applications in Cardiac Disease and Toxicity. Stem Cells 2015; 34:13-26. [DOI: 10.1002/stem.2196] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Revised: 07/14/2015] [Accepted: 07/28/2015] [Indexed: 12/14/2022]
Affiliation(s)
- Sabine C. Den Hartogh
- Department of Anatomy and Embryology; Leiden University Medical Centre; Leiden The Netherlands
| | - Robert Passier
- Department of Anatomy and Embryology; Leiden University Medical Centre; Leiden The Netherlands
- Department of Applied Stem cell Technologies. MIRA Institute for Biomedical Technology and Technical Medicine; University of Twente, P.O.Box 217; Enschede The Netherlands
| |
Collapse
|
76
|
Affiliation(s)
- Sang-Ging Ong
- From the Stanford Cardiovascular Institute (S.-G.O., J.C.W.) and Division of Cardiology, Department of Medicine, Stanford University School of Medicine, CA (S.-G.O., J.C.W.)
| | - Joseph C Wu
- From the Stanford Cardiovascular Institute (S.-G.O., J.C.W.) and Division of Cardiology, Department of Medicine, Stanford University School of Medicine, CA (S.-G.O., J.C.W.).
| |
Collapse
|
77
|
Riegler J, Tiburcy M, Ebert A, Tzatzalos E, Raaz U, Abilez OJ, Shen Q, Kooreman NG, Neofytou E, Chen VC, Wang M, Meyer T, Tsao PS, Connolly AJ, Couture LA, Gold JD, Zimmermann WH, Wu JC. Human Engineered Heart Muscles Engraft and Survive Long Term in a Rodent Myocardial Infarction Model. Circ Res 2015; 117:720-30. [PMID: 26291556 DOI: 10.1161/circresaha.115.306985] [Citation(s) in RCA: 164] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 08/19/2015] [Indexed: 01/17/2023]
Abstract
RATIONALE Tissue engineering approaches may improve survival and functional benefits from human embryonic stem cell-derived cardiomyocyte transplantation, thereby potentially preventing dilative remodeling and progression to heart failure. OBJECTIVE Assessment of transport stability, long-term survival, structural organization, functional benefits, and teratoma risk of engineered heart muscle (EHM) in a chronic myocardial infarction model. METHODS AND RESULTS We constructed EHMs from human embryonic stem cell-derived cardiomyocytes and released them for transatlantic shipping following predefined quality control criteria. Two days of shipment did not lead to adverse effects on cell viability or contractile performance of EHMs (n=3, P=0.83, P=0.87). One month after ischemia/reperfusion injury, EHMs were implanted onto immunocompromised rat hearts to simulate chronic ischemia. Bioluminescence imaging showed stable engraftment with no significant cell loss between week 2 and 12 (n=6, P=0.67), preserving ≤25% of the transplanted cells. Despite high engraftment rates and attenuated disease progression (change in ejection fraction for EHMs, -6.7±1.4% versus control, -10.9±1.5%; n>12; P=0.05), we observed no difference between EHMs containing viable and nonviable human cardiomyocytes in this chronic xenotransplantation model (n>12; P=0.41). Grafted cardiomyocytes showed enhanced sarcomere alignment and increased connexin 43 expression at 220 days after transplantation. No teratomas or tumors were found in any of the animals (n=14) used for long-term monitoring. CONCLUSIONS EHM transplantation led to high engraftment rates, long-term survival, and progressive maturation of human cardiomyocytes. However, cell engraftment was not correlated with functional improvements in this chronic myocardial infarction model. Most importantly, the safety of this approach was demonstrated by the lack of tumor or teratoma formation.
Collapse
Affiliation(s)
- Johannes Riegler
- From the Division of Cardiology, Department of Medicine, Stanford Cardiovascular Institute (J.R., A.E., E.T., U.R., O.J.A., O.S., N.G.K., E.N., M.W., P.S.T., J.D.G., J.C.W.) and Department of Pathology (A.J.C.), Stanford University School of Medicine, CA; Department for Research and Development, Veterans Administration Palo Alto Health Care System, CA (P.S.T.); Institute of Pharmacology, Heart Research Center, University Medical Center, Georg-August-University and German Center for Cardiovascular Research, Göttingen, Germany (M.T., T.M., W.H.Z.); and Center for Biomedicine and Genetics (V.C.C., L.A.C.) and Center for Applied Technology Development, Beckman Research Institute (A.J.C.), City of Hope, Duarte, CA
| | - Malte Tiburcy
- From the Division of Cardiology, Department of Medicine, Stanford Cardiovascular Institute (J.R., A.E., E.T., U.R., O.J.A., O.S., N.G.K., E.N., M.W., P.S.T., J.D.G., J.C.W.) and Department of Pathology (A.J.C.), Stanford University School of Medicine, CA; Department for Research and Development, Veterans Administration Palo Alto Health Care System, CA (P.S.T.); Institute of Pharmacology, Heart Research Center, University Medical Center, Georg-August-University and German Center for Cardiovascular Research, Göttingen, Germany (M.T., T.M., W.H.Z.); and Center for Biomedicine and Genetics (V.C.C., L.A.C.) and Center for Applied Technology Development, Beckman Research Institute (A.J.C.), City of Hope, Duarte, CA
| | - Antje Ebert
- From the Division of Cardiology, Department of Medicine, Stanford Cardiovascular Institute (J.R., A.E., E.T., U.R., O.J.A., O.S., N.G.K., E.N., M.W., P.S.T., J.D.G., J.C.W.) and Department of Pathology (A.J.C.), Stanford University School of Medicine, CA; Department for Research and Development, Veterans Administration Palo Alto Health Care System, CA (P.S.T.); Institute of Pharmacology, Heart Research Center, University Medical Center, Georg-August-University and German Center for Cardiovascular Research, Göttingen, Germany (M.T., T.M., W.H.Z.); and Center for Biomedicine and Genetics (V.C.C., L.A.C.) and Center for Applied Technology Development, Beckman Research Institute (A.J.C.), City of Hope, Duarte, CA
| | - Evangeline Tzatzalos
- From the Division of Cardiology, Department of Medicine, Stanford Cardiovascular Institute (J.R., A.E., E.T., U.R., O.J.A., O.S., N.G.K., E.N., M.W., P.S.T., J.D.G., J.C.W.) and Department of Pathology (A.J.C.), Stanford University School of Medicine, CA; Department for Research and Development, Veterans Administration Palo Alto Health Care System, CA (P.S.T.); Institute of Pharmacology, Heart Research Center, University Medical Center, Georg-August-University and German Center for Cardiovascular Research, Göttingen, Germany (M.T., T.M., W.H.Z.); and Center for Biomedicine and Genetics (V.C.C., L.A.C.) and Center for Applied Technology Development, Beckman Research Institute (A.J.C.), City of Hope, Duarte, CA
| | - Uwe Raaz
- From the Division of Cardiology, Department of Medicine, Stanford Cardiovascular Institute (J.R., A.E., E.T., U.R., O.J.A., O.S., N.G.K., E.N., M.W., P.S.T., J.D.G., J.C.W.) and Department of Pathology (A.J.C.), Stanford University School of Medicine, CA; Department for Research and Development, Veterans Administration Palo Alto Health Care System, CA (P.S.T.); Institute of Pharmacology, Heart Research Center, University Medical Center, Georg-August-University and German Center for Cardiovascular Research, Göttingen, Germany (M.T., T.M., W.H.Z.); and Center for Biomedicine and Genetics (V.C.C., L.A.C.) and Center for Applied Technology Development, Beckman Research Institute (A.J.C.), City of Hope, Duarte, CA
| | - Oscar J Abilez
- From the Division of Cardiology, Department of Medicine, Stanford Cardiovascular Institute (J.R., A.E., E.T., U.R., O.J.A., O.S., N.G.K., E.N., M.W., P.S.T., J.D.G., J.C.W.) and Department of Pathology (A.J.C.), Stanford University School of Medicine, CA; Department for Research and Development, Veterans Administration Palo Alto Health Care System, CA (P.S.T.); Institute of Pharmacology, Heart Research Center, University Medical Center, Georg-August-University and German Center for Cardiovascular Research, Göttingen, Germany (M.T., T.M., W.H.Z.); and Center for Biomedicine and Genetics (V.C.C., L.A.C.) and Center for Applied Technology Development, Beckman Research Institute (A.J.C.), City of Hope, Duarte, CA
| | - Qi Shen
- From the Division of Cardiology, Department of Medicine, Stanford Cardiovascular Institute (J.R., A.E., E.T., U.R., O.J.A., O.S., N.G.K., E.N., M.W., P.S.T., J.D.G., J.C.W.) and Department of Pathology (A.J.C.), Stanford University School of Medicine, CA; Department for Research and Development, Veterans Administration Palo Alto Health Care System, CA (P.S.T.); Institute of Pharmacology, Heart Research Center, University Medical Center, Georg-August-University and German Center for Cardiovascular Research, Göttingen, Germany (M.T., T.M., W.H.Z.); and Center for Biomedicine and Genetics (V.C.C., L.A.C.) and Center for Applied Technology Development, Beckman Research Institute (A.J.C.), City of Hope, Duarte, CA
| | - Nigel G Kooreman
- From the Division of Cardiology, Department of Medicine, Stanford Cardiovascular Institute (J.R., A.E., E.T., U.R., O.J.A., O.S., N.G.K., E.N., M.W., P.S.T., J.D.G., J.C.W.) and Department of Pathology (A.J.C.), Stanford University School of Medicine, CA; Department for Research and Development, Veterans Administration Palo Alto Health Care System, CA (P.S.T.); Institute of Pharmacology, Heart Research Center, University Medical Center, Georg-August-University and German Center for Cardiovascular Research, Göttingen, Germany (M.T., T.M., W.H.Z.); and Center for Biomedicine and Genetics (V.C.C., L.A.C.) and Center for Applied Technology Development, Beckman Research Institute (A.J.C.), City of Hope, Duarte, CA
| | - Evgenios Neofytou
- From the Division of Cardiology, Department of Medicine, Stanford Cardiovascular Institute (J.R., A.E., E.T., U.R., O.J.A., O.S., N.G.K., E.N., M.W., P.S.T., J.D.G., J.C.W.) and Department of Pathology (A.J.C.), Stanford University School of Medicine, CA; Department for Research and Development, Veterans Administration Palo Alto Health Care System, CA (P.S.T.); Institute of Pharmacology, Heart Research Center, University Medical Center, Georg-August-University and German Center for Cardiovascular Research, Göttingen, Germany (M.T., T.M., W.H.Z.); and Center for Biomedicine and Genetics (V.C.C., L.A.C.) and Center for Applied Technology Development, Beckman Research Institute (A.J.C.), City of Hope, Duarte, CA
| | - Vincent C Chen
- From the Division of Cardiology, Department of Medicine, Stanford Cardiovascular Institute (J.R., A.E., E.T., U.R., O.J.A., O.S., N.G.K., E.N., M.W., P.S.T., J.D.G., J.C.W.) and Department of Pathology (A.J.C.), Stanford University School of Medicine, CA; Department for Research and Development, Veterans Administration Palo Alto Health Care System, CA (P.S.T.); Institute of Pharmacology, Heart Research Center, University Medical Center, Georg-August-University and German Center for Cardiovascular Research, Göttingen, Germany (M.T., T.M., W.H.Z.); and Center for Biomedicine and Genetics (V.C.C., L.A.C.) and Center for Applied Technology Development, Beckman Research Institute (A.J.C.), City of Hope, Duarte, CA
| | - Mouer Wang
- From the Division of Cardiology, Department of Medicine, Stanford Cardiovascular Institute (J.R., A.E., E.T., U.R., O.J.A., O.S., N.G.K., E.N., M.W., P.S.T., J.D.G., J.C.W.) and Department of Pathology (A.J.C.), Stanford University School of Medicine, CA; Department for Research and Development, Veterans Administration Palo Alto Health Care System, CA (P.S.T.); Institute of Pharmacology, Heart Research Center, University Medical Center, Georg-August-University and German Center for Cardiovascular Research, Göttingen, Germany (M.T., T.M., W.H.Z.); and Center for Biomedicine and Genetics (V.C.C., L.A.C.) and Center for Applied Technology Development, Beckman Research Institute (A.J.C.), City of Hope, Duarte, CA
| | - Tim Meyer
- From the Division of Cardiology, Department of Medicine, Stanford Cardiovascular Institute (J.R., A.E., E.T., U.R., O.J.A., O.S., N.G.K., E.N., M.W., P.S.T., J.D.G., J.C.W.) and Department of Pathology (A.J.C.), Stanford University School of Medicine, CA; Department for Research and Development, Veterans Administration Palo Alto Health Care System, CA (P.S.T.); Institute of Pharmacology, Heart Research Center, University Medical Center, Georg-August-University and German Center for Cardiovascular Research, Göttingen, Germany (M.T., T.M., W.H.Z.); and Center for Biomedicine and Genetics (V.C.C., L.A.C.) and Center for Applied Technology Development, Beckman Research Institute (A.J.C.), City of Hope, Duarte, CA
| | - Philip S Tsao
- From the Division of Cardiology, Department of Medicine, Stanford Cardiovascular Institute (J.R., A.E., E.T., U.R., O.J.A., O.S., N.G.K., E.N., M.W., P.S.T., J.D.G., J.C.W.) and Department of Pathology (A.J.C.), Stanford University School of Medicine, CA; Department for Research and Development, Veterans Administration Palo Alto Health Care System, CA (P.S.T.); Institute of Pharmacology, Heart Research Center, University Medical Center, Georg-August-University and German Center for Cardiovascular Research, Göttingen, Germany (M.T., T.M., W.H.Z.); and Center for Biomedicine and Genetics (V.C.C., L.A.C.) and Center for Applied Technology Development, Beckman Research Institute (A.J.C.), City of Hope, Duarte, CA
| | - Andrew J Connolly
- From the Division of Cardiology, Department of Medicine, Stanford Cardiovascular Institute (J.R., A.E., E.T., U.R., O.J.A., O.S., N.G.K., E.N., M.W., P.S.T., J.D.G., J.C.W.) and Department of Pathology (A.J.C.), Stanford University School of Medicine, CA; Department for Research and Development, Veterans Administration Palo Alto Health Care System, CA (P.S.T.); Institute of Pharmacology, Heart Research Center, University Medical Center, Georg-August-University and German Center for Cardiovascular Research, Göttingen, Germany (M.T., T.M., W.H.Z.); and Center for Biomedicine and Genetics (V.C.C., L.A.C.) and Center for Applied Technology Development, Beckman Research Institute (A.J.C.), City of Hope, Duarte, CA
| | - Larry A Couture
- From the Division of Cardiology, Department of Medicine, Stanford Cardiovascular Institute (J.R., A.E., E.T., U.R., O.J.A., O.S., N.G.K., E.N., M.W., P.S.T., J.D.G., J.C.W.) and Department of Pathology (A.J.C.), Stanford University School of Medicine, CA; Department for Research and Development, Veterans Administration Palo Alto Health Care System, CA (P.S.T.); Institute of Pharmacology, Heart Research Center, University Medical Center, Georg-August-University and German Center for Cardiovascular Research, Göttingen, Germany (M.T., T.M., W.H.Z.); and Center for Biomedicine and Genetics (V.C.C., L.A.C.) and Center for Applied Technology Development, Beckman Research Institute (A.J.C.), City of Hope, Duarte, CA
| | - Joseph D Gold
- From the Division of Cardiology, Department of Medicine, Stanford Cardiovascular Institute (J.R., A.E., E.T., U.R., O.J.A., O.S., N.G.K., E.N., M.W., P.S.T., J.D.G., J.C.W.) and Department of Pathology (A.J.C.), Stanford University School of Medicine, CA; Department for Research and Development, Veterans Administration Palo Alto Health Care System, CA (P.S.T.); Institute of Pharmacology, Heart Research Center, University Medical Center, Georg-August-University and German Center for Cardiovascular Research, Göttingen, Germany (M.T., T.M., W.H.Z.); and Center for Biomedicine and Genetics (V.C.C., L.A.C.) and Center for Applied Technology Development, Beckman Research Institute (A.J.C.), City of Hope, Duarte, CA
| | - Wolfram H Zimmermann
- From the Division of Cardiology, Department of Medicine, Stanford Cardiovascular Institute (J.R., A.E., E.T., U.R., O.J.A., O.S., N.G.K., E.N., M.W., P.S.T., J.D.G., J.C.W.) and Department of Pathology (A.J.C.), Stanford University School of Medicine, CA; Department for Research and Development, Veterans Administration Palo Alto Health Care System, CA (P.S.T.); Institute of Pharmacology, Heart Research Center, University Medical Center, Georg-August-University and German Center for Cardiovascular Research, Göttingen, Germany (M.T., T.M., W.H.Z.); and Center for Biomedicine and Genetics (V.C.C., L.A.C.) and Center for Applied Technology Development, Beckman Research Institute (A.J.C.), City of Hope, Duarte, CA.
| | - Joseph C Wu
- From the Division of Cardiology, Department of Medicine, Stanford Cardiovascular Institute (J.R., A.E., E.T., U.R., O.J.A., O.S., N.G.K., E.N., M.W., P.S.T., J.D.G., J.C.W.) and Department of Pathology (A.J.C.), Stanford University School of Medicine, CA; Department for Research and Development, Veterans Administration Palo Alto Health Care System, CA (P.S.T.); Institute of Pharmacology, Heart Research Center, University Medical Center, Georg-August-University and German Center for Cardiovascular Research, Göttingen, Germany (M.T., T.M., W.H.Z.); and Center for Biomedicine and Genetics (V.C.C., L.A.C.) and Center for Applied Technology Development, Beckman Research Institute (A.J.C.), City of Hope, Duarte, CA.
| |
Collapse
|
78
|
Guan X, Wang Z, Czerniecki S, Mack D, François V, Blouin V, Moullier P, Childers MK. Use of Adeno-Associated Virus to Enrich Cardiomyocytes Derived from Human Stem Cells. HUM GENE THER CL DEV 2015; 26:194-201. [PMID: 26252064 DOI: 10.1089/humc.2015.052] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Cardiomyocytes derived from human induced pluripotent stem cells (iPSCs) show great promise as autologous donor cells to treat heart disease. A major technical obstacle to this approach is that available induction methods often produce heterogeneous cell population with low percentage of cardiomyocytes. Here we describe a cardiac enrichment approach using nonintegrating adeno-associated virus (AAV). We first examined several AAV serotypes for their ability to selectively transduce iPSC-derived cardiomyocytes. Results showed that AAV1 demonstrated the highest in vitro transduction efficiency among seven widely used serotypes. Next, differentiated iPSC derivatives were transduced with drug-selectable AAV1 expressing neomycin resistance gene. Selection with G418 enriched the cardiac cell fraction from 27% to 57% in 2 weeks. Compared with other enrichment strategies such as integrative genetic selection, mitochondria labeling, or surface marker cell sorting, this simple AAV method described herein bypasses antibody or dye labeling. These findings provide proof of concept for large-scale cardiomyocyte enrichment by exploiting AAV's intrinsic tissue tropism.
Collapse
Affiliation(s)
- Xuan Guan
- 1 Department of Physiology and Pharmacology, School of Medicine, Wake Forest University Health Sciences , Winston-Salem, North Carolina.,2 Department of Rehabilitation Medicine, University of Washington , Seattle, Washington.,3 Institute for Stem Cell and Regenerative Medicine, University of Washington , Seattle, Washington
| | - Zejing Wang
- 4 Fred Hutchinson Cancer Research Center , Seattle, Washington.,5 Department of Medicine, University of Washington , Seattle, Washington
| | - Stefan Czerniecki
- 2 Department of Rehabilitation Medicine, University of Washington , Seattle, Washington.,3 Institute for Stem Cell and Regenerative Medicine, University of Washington , Seattle, Washington
| | - David Mack
- 2 Department of Rehabilitation Medicine, University of Washington , Seattle, Washington.,3 Institute for Stem Cell and Regenerative Medicine, University of Washington , Seattle, Washington
| | - Virginie François
- 6 INSERM UMR 1089 IRT 1, Institut de Recherche Thérapeutique, Université de Nantes , Nantes, France
| | - Veronique Blouin
- 6 INSERM UMR 1089 IRT 1, Institut de Recherche Thérapeutique, Université de Nantes , Nantes, France
| | - Philippe Moullier
- 6 INSERM UMR 1089 IRT 1, Institut de Recherche Thérapeutique, Université de Nantes , Nantes, France.,7 Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, Florida
| | - Martin K Childers
- 2 Department of Rehabilitation Medicine, University of Washington , Seattle, Washington.,3 Institute for Stem Cell and Regenerative Medicine, University of Washington , Seattle, Washington
| |
Collapse
|
79
|
Affiliation(s)
- David E Sosnovik
- Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA (D.E.S.) Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA (D.E.S.)
| |
Collapse
|
80
|
Guan X, Wang Z, Czerniecki S, Mack D, François V, Blouin V, Moullier P, Childers M. Use of adeno-associated virus to enrich cardiomyocytes derived from human stem cells. HUM GENE THER CL DEV 2015. [DOI: 10.1089/hum.2015.052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
81
|
Neofytou E, O'Brien CG, Couture LA, Wu JC. Hurdles to clinical translation of human induced pluripotent stem cells. J Clin Invest 2015; 125:2551-7. [PMID: 26132109 DOI: 10.1172/jci80575] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Human pluripotent stem cells are known to have the capacity to renew indefinitely, being intrinsically able to differentiate into many different cell types. These characteristics have generated tremendous enthusiasm about the potential applications of these cells in regenerative medicine. However, major challenges remain with the development and testing of novel experimental stem cell therapeutics in the field. In this Review, we focus on the nature of the preclinical challenges and discuss potential solutions that could help overcome them. Furthermore, we discuss the use of allogeneic versus autologous stem cell products, including a review of their respective advantages and disadvantages, major clinical requirements, quality standards, time lines, and costs of clinical grade development.
Collapse
|
82
|
Chen CH, Sereti KI, Wu BM, Ardehali R. Translational aspects of cardiac cell therapy. J Cell Mol Med 2015; 19:1757-72. [PMID: 26119413 PMCID: PMC4549027 DOI: 10.1111/jcmm.12632] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 05/13/2015] [Indexed: 02/06/2023] Open
Abstract
Cell therapy has been intensely studied for over a decade as a potential treatment for ischaemic heart disease. While initial trials using skeletal myoblasts, bone marrow cells and peripheral blood stem cells showed promise in improving cardiac function, benefits were found to be short-lived likely related to limited survival and engraftment of the delivered cells. The discovery of putative cardiac ‘progenitor’ cells as well as the creation of induced pluripotent stem cells has led to the delivery of cells potentially capable of electromechanical integration into existing tissue. An alternative strategy involving either direct reprogramming of endogenous cardiac fibroblasts or stimulation of resident cardiomyocytes to regenerate new myocytes can potentially overcome the limitations of exogenous cell delivery. Complimentary approaches utilizing combination cell therapy and bioengineering techniques may be necessary to provide the proper milieu for clinically significant regeneration. Clinical trials employing bone marrow cells, mesenchymal stem cells and cardiac progenitor cells have demonstrated safety of catheter based cell delivery, with suggestion of limited improvement in ventricular function and reduction in infarct size. Ongoing trials are investigating potential benefits to outcome such as morbidity and mortality. These and future trials will clarify the optimal cell types and delivery conditions for therapeutic effect.
Collapse
Affiliation(s)
- Cheng-Han Chen
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.,Department of Bioengineering, UCLA, Los Angeles, CA, USA
| | - Konstantina-Ioanna Sereti
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Benjamin M Wu
- Department of Bioengineering, UCLA, Los Angeles, CA, USA
| | - Reza Ardehali
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.,Eli and Edythe Broad Stem Cell Research Center, UCLA, Los Angeles, CA, USA
| |
Collapse
|
83
|
Bar-Shir A, Bulte JWM, Gilad AA. Molecular engineering of nonmetallic biosensors for CEST MRI. ACS Chem Biol 2015; 10:1160-70. [PMID: 25730583 PMCID: PMC11329289 DOI: 10.1021/cb500923v] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Recent advancements in molecular and synthetic biology, combined with synthetic chemistry and biotechnology, have opened up new opportunities to engineer novel platforms that can monitor complex biological processes with various noninvasive imaging modalities. After decades of using gadolinium- or iron-based metallic sensors for MRI, the recently developed chemical exchange saturation transfer (CEST) contrast mechanism has created an opportunity for rational design, in silico, of nonmetallic biosensors for MRI. These biomolecules are either naturally occurring compounds (amino acids, sugars, nucleosides, native proteins) or can be artificially engineered (synthetic probes or recombinant proteins). They can be administered either as exogenous agents or can be genetically (over)expressed. Moreover, they can be precisely engineered to achieve the desired biochemical properties for fine tuning optimized imaging schemes. The availability of these agents marks the dawn of a new scientific era for molecular and cellular MRI.
Collapse
Affiliation(s)
- Amnon Bar-Shir
- †Russell H. Morgan Department of Radiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States
- ‡Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Jeff W M Bulte
- †Russell H. Morgan Department of Radiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States
- ‡Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- §F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland 21205, United States
- ∥Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- #Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States
| | - Assaf A Gilad
- †Russell H. Morgan Department of Radiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States
- ‡Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- §F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland 21205, United States
| |
Collapse
|
84
|
Design of a functional cyclic HSV1-TK reporter and its application to PET imaging of apoptosis. Nat Protoc 2015; 10:807-21. [PMID: 25927390 DOI: 10.1038/nprot.2015.048] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Positron emission tomography (PET) is a sensitive and noninvasive imaging method that is widely used to explore molecular events in living subjects. PET can precisely and quantitatively evaluate cellular apoptosis, which has a crucial role in various physiological and pathological processes. In this protocol, we describe the design and use of an engineered cyclic herpes simplex virus 1-thymidine kinase (HSV1-TK) PET reporter whose kinase activity is specifically switched on by apoptosis. The expression of cyclic TK (cTK) in healthy cells leads to inactive product, whereas the activation of apoptosis through the caspase-3 pathway cleaves cTK, thus restoring its activity and enabling PET imaging. In addition to detailing the design and construction of the cTK plasmid in this protocol, we include assays for evaluating the function and specificity of the cTK reporter in apoptotic cells, such as assays for measuring the cell uptake of PET tracer in apoptotic cells, correlating doxorubicin (Dox)-induced cell apoptosis to cTK function recovery, and in vivo PET imaging of cancer cell apoptosis, and we also include corresponding data acquisition methods. The time to build the entire cTK reporter is ∼2-3 weeks. The selection of a stable cancer cell line takes ∼4-6 weeks. The time to implement assays regarding cTK function in apoptotic cells and the in vivo imaging varies depending on the experiment. The cyclization strategy described in this protocol can also be adapted to create other reporter systems for broad biomedical applications.
Collapse
|
85
|
124I-PET Assessment of Human Sodium Iodide Symporter Reporter Gene Activity for Highly Sensitive In Vivo Monitoring of Teratoma Formation in Mice. Mol Imaging Biol 2015; 17:874-83. [DOI: 10.1007/s11307-015-0857-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
86
|
Sanganeria P, Chandra S, Bahadur D, Khanna A. Effect of HSA coated iron oxide labeling on human umbilical cord derived mesenchymal stem cells. NANOTECHNOLOGY 2015; 26:125103. [PMID: 25744689 DOI: 10.1088/0957-4484/26/12/125103] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Human umbilical cord derived mesenchymal stem cells (hUC-MSCs) are known for self-renewal and differentiation into cells of various lineages like bone, cartilage and fat. They have been used in biomedical applications to treat degenerative disorders. However, to exploit the therapeutic potential of stem cells, there is a requirement of sensitive non-invasive imaging techniques which will offer the ability to track transplanted cells, bio-distribution, proliferation and differentiation. In this study, we have analyzed the efficacy of human serum albumin coated iron oxide nanoparticles (HSA-IONPs) on the differentiation of hUC-MSCs. The colloidal stability of the HSA-IONPs was tested over a long period of time (≥20 months) and the optimized concentration of HSA-IONPs for labeling the stem cells was 60 μg ml(-1). Detailed in vitro assays have been performed to ascertain the effect of the nanoparticles (NPs) on stem cells. Lactate dehydrogenase (LDH) assay showed minimum release of LDH depicting the least disruptions in cellular membrane. At the same time, mitochondrial impairment of the cells was also not observed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Flow cytometry analysis revealed lesser generation of reactive oxygen species in HSA-IONPs labeled hUC-MSCs in comparison to bare and commercial IONPs. Transmission electron microscopy showed endocytic engulfment of the NPs by the hUC-MSCs. During the process, the gross morphologies of the actin cytoskeleton were found to be intact as shown by immunofluorescence microscopy. Also, the engulfment of the HSA-IONPs did not show any detrimental effect on the differentiation potential of the stem cells into adipocytes, osteocytes and chondrocytes, thereby confirming that the inherent properties of stem cells were maintained.
Collapse
Affiliation(s)
- Purva Sanganeria
- Department of Biological Sciences, School of Science, NMIMS University, Vile Parle (West), Mumbai 400056, India
| | | | | | | |
Collapse
|
87
|
Adenovirus-mediated expression of human sodium-iodide symporter gene permits in vivo tracking of adipose tissue-derived stem cells in a canine myocardial infarction model. Nucl Med Biol 2015; 42:621-9. [PMID: 25899941 DOI: 10.1016/j.nucmedbio.2015.03.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Revised: 02/02/2015] [Accepted: 03/16/2015] [Indexed: 11/23/2022]
Abstract
INTRODUCTION In vivo tracking of the transplanted stem cells is important in pre-clinical research of stem cell therapy for myocardial infarction. We examined the feasibility of adenovirus-mediated sodium iodide symporter (NIS) gene to cell tracking imaging of transplanted stem cells in a canine infarcted myocardium by clinical single photon emission computed tomography (SPECT). METHODS Beagle dogs were injected intramyocardially with NIS-expressing adenovirus-transfected canine stem cells (Ad-hNIS-canine ADSCs) a week after myocardial infarction (MI) development. (99m)Tc-methoxyisobutylisonitrile ((99m)Tc-MIBI) and (99m)Tc-pertechnetate ((99m)TcO4(-)) SPECT imaging were performed for assessment of infarcted myocardium and viable stem cell tracking. Transthoracic echocardiography was performed to monitor any functional cardiac changes. RESULTS Left ventricular ejection fraction (LVEF) was decreased after LAD ligation. There was no significant difference in EF between the groups with the stem cell or saline injection. (125)I uptake was higher in Ad-hNIS-canine ADSCs than in non-transfected ADSCs. Cell proliferation and differentiation were not affected by hNIS-carrying adenovirus transfection. (99m)Tc-MIBI myocardial SPECT imaging showed decreased radiotracer uptake in the infarcted apex and mid-anterolateral regions. Ad-hNIS-canine ADSCs were identified as a region of focally increased (99m)TcO4(-) uptake at the lateral wall and around the apex of the left ventricle, peaked at 2 days and was observed until day 9. CONCLUSIONS Combination of adenovirus-mediated NIS gene transfection and clinical nuclear imaging modalities enables to trace the fate of transplanted stem cells in infarcted myocardium for translational in vivo cell tracking study for prolonged duration.
Collapse
|
88
|
Ghrelin improves functional survival of engrafted adipose-derived mesenchymal stem cells in ischemic heart through PI3K/Akt signaling pathway. BIOMED RESEARCH INTERNATIONAL 2015; 2015:858349. [PMID: 25879037 PMCID: PMC4387976 DOI: 10.1155/2015/858349] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 01/26/2015] [Accepted: 01/27/2015] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cells (MSCs) have been proposed as a promising cell population for cell therapy and regenerative medicine applications. However, the low retention and poor survival of engrafted cells hampered the therapeutic efficacy of engrafted MSCs. Ghrelin is a 28-amino-acid peptide hormone and is proved to exert a protective effect on the cardiovascular system. This study is designed to investigate the protective effects of ghrelin on engrafted adipose-derived mesenchymal stem cells (ADMSCs) and its beneficial effects with cellular therapy in mice myocardial infarction (MI). Results showed that intramyocardial injection of ADMSCs combining with ghrelin administration inhibited host cardiomyocyte apoptosis, reduced fibrosis, and improved cardiac function. To reveal possible mechanisms, ADMSCs were subjected to hypoxia/serum deprivation (H/SD) injury to simulate ischemic conditions in vivo. Ghrelin (10−8 M, 33712 pg/ml) improved ADMSCs survival under H/SD condition. Western blot assay revealed that ghrelin increased
AKT phosphorylation both in vivo and in vitro, decreased the proapoptotic protein Bax, and increased the antiapoptotic protein Bcl-2 in vitro, while these effects were abolished by PI3K inhibitor LY294002. These revealed that ghrelin may serve as a promising candidate for hormone-driven approaches to improve the efficacy of mesenchymal stem cell-based therapy for cardiac ischemic disease via PI3K/AKT pathway.
Collapse
|
89
|
Zhang Y, Kuraitis D, Burgon PG, Crowe S, Vulesevic B, Beanlands RS, deKemp RA, DaSilva JN, Ruel M, Suuronen EJ. Development of reporter gene imaging techniques for long-term assessment of human circulating angiogenic cells. ACTA ACUST UNITED AC 2015; 10:034104. [PMID: 25782444 DOI: 10.1088/1748-6041/10/3/034104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The use of biomaterials and tracking the long-term fate of the transplanted cells is expected to help improve the clinical translation of cell therapies for cardiac regeneration. To this end, reporter gene strategies are promising for monitoring the fate of cells transplanted with or without a delivery biomaterial; however, their application with primary adult progenitor cells (such as human circulating angiogenic cells (CACs)) has not been extensively evaluated. In this study, human CACs were transduced with reporter genes via one of two lentiviral (LV) vectors: LV-GFP-iresTK or LV-Fluc-RFP-tTK. The mean transduction efficiency was 15% (LV-GFP-iresTK) and 13% (LV-Fluc-RFP-tTK) at multiplicities of infection (MOI) of 10 and 50, respectively. Western blot analysis confirmed HSV1-tk protein expression in transduced CACs. There was no significant difference in viability between the transduced CACs and the untreated controls at a MOI of 50 or below. However, a reduction was observed in cell viability of CACs transduced with LV-Fluc-RFP-tTK at an MOI of 100. Cell migration and angiogenic potential were not affected by transduction protocol. After 4 weeks, 80.3 ± 8.4% of the labeled cells continued to express the reporters and could be visualized when embedded within a collagen matrix scaffold. Therefore, quiescent human CACs can be stably transduced to express reporter genes without affecting their function. This reporter gene technique is a promising approach to be further tested for tracking transplanted CACs (±delivery matrix) non-invasively and longitudinally.
Collapse
Affiliation(s)
- Yan Zhang
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, K1Y 4W7, Canada. Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, K1H 8M5, Canada. Division of Cardiology (Department of Medicine), Central Hospital Affiliated to Shenyang Medical College, Shenyang, 110024, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
90
|
Saleh IG, Ali Z, Hammad MA, Wilson FD, Hamada FM, Abd-Ellah MF, Walker LA, Khan IA, Ashfaq MK. Stem cell intervention ameliorates epigallocatechin-3-gallate/lipopolysaccharide-induced hepatotoxicity in mice. Hum Exp Toxicol 2015; 34:1180-94. [PMID: 25701483 DOI: 10.1177/0960327115572707] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Stem cells are identified as a novel cell therapy for regenerative medicine because of their ability to differentiate into many functional cell types. We have shown earlier a new model of hepatotoxicity in mice by administering (1500 mg/kg) epigallocatechin-3-gallate (EGCG) intragastric (IG) for 5 days after a single intraperitoneal dose (6 mg/kg) of lipopolysaccharide (LPS). In this study, we aimed to study the effect of intrahepatic (IH) injection of mouse embryonic stem cells (MESCs) on the hepatotoxicity induced by EGCG/LPS in mice. Mice were administered EGCG/LPS and rested for 3 days. MESCs were obtained from American Type Culture Collection and cultured in vitro for 4 days. Stem cells were injected IH. Seven days later, a single dose of LPS (6 mg/kg) followed by daily doses of IG administration of EGCG were re-administered for 5 days. At the end of the experiment, blood samples were collected for analysis of biochemical parameters associated with liver. Results showed that the group of mice that were administered MESCs prior to EGCG/LPS showed lower levels of alanine amino transferase, alkaline phosphatase, and bilirubin, higher albumin/globulin ratio, and less remarkable histopathological lesions. Also, that group of mice showed less expression of oxidative stress biomarkers (oxidized low-density lipoprotein Ox.LDL and chemokine CXCL16), less expression of nuclear protein receptors (retinoic acid receptor and retinoid X receptor), and less expression of inflammatory biomarkers (tumor necrosis factor α and transforming growth factor β1) compared with other groups of mice that were not given MESCs. In conclusion, MESCs can ameliorate EGCG/LPS-induced hepatotoxicity in mice.
Collapse
Affiliation(s)
- I G Saleh
- National Center for Natural Products Research, School of Pharmacy, University of Mississippi, Oxford, MS, USA Department of Pharmacology, Faculty of Pharmacy, Al-Azhar University, Nasr City, Cairo, Egypt
| | - Z Ali
- National Center for Natural Products Research, School of Pharmacy, University of Mississippi, Oxford, MS, USA
| | - M A Hammad
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - F D Wilson
- Mississippi Veterinary Research and Diagnostic Laboratory, College of Veterinary Medicine, Mississippi State University, Pearl, MS, USA
| | - F M Hamada
- Department of Pharmacology, Faculty of Pharmacy, Al-Azhar University, Nasr City, Cairo, Egypt
| | - M F Abd-Ellah
- Department of Pharmacology, Faculty of Pharmacy, Al-Azhar University, Nasr City, Cairo, Egypt
| | - L A Walker
- National Center for Natural Products Research, School of Pharmacy, University of Mississippi, Oxford, MS, USA Department of Pharmacology, School of Pharmacy, University of Mississippi, Oxford, MS, USA
| | - I A Khan
- National Center for Natural Products Research, School of Pharmacy, University of Mississippi, Oxford, MS, USA Department of Pharmacognosy, School of Pharmacy, University of Mississippi, Oxford, MS, USA Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - M K Ashfaq
- National Center for Natural Products Research, School of Pharmacy, University of Mississippi, Oxford, MS, USA
| |
Collapse
|
91
|
Tang YH, Ma YY, Zhang ZJ, Wang YT, Yang GY. Opportunities and challenges: stem cell-based therapy for the treatment of ischemic stroke. CNS Neurosci Ther 2015; 21:337-47. [PMID: 25676164 DOI: 10.1111/cns.12386] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Revised: 01/09/2015] [Accepted: 01/09/2015] [Indexed: 01/01/2023] Open
Abstract
Stem cell-based therapy for ischemic stroke has been widely explored in animal models and provides strong evidence of benefits. In this review, we summarize the types of stem cells, various delivery routes, and tracking tools for stem cell therapy of ischemic stroke. MSCs, EPCs, and NSCs are the most explored cell types for ischemic stroke treatment. Although the mechanisms of stem cell-based therapies are not fully understood, the most possible functions of the transplanted cells are releasing growth factors and regulating microenvironment through paracrine mechanism. Clinical application of stem cell-based therapy is still in its infancy. The next decade of stem cell research in stroke field needs to focus on combining different stem cells and different imaging modalities to fully explore the potential of this therapeutic avenue: from bench to bedside and vice versa.
Collapse
Affiliation(s)
- Yao-Hui Tang
- Neuroscience and Neuroengineering Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | | | | | | | | |
Collapse
|
92
|
Nelakanti RV, Kooreman NG, Wu JC. Teratoma formation: a tool for monitoring pluripotency in stem cell research. CURRENT PROTOCOLS IN STEM CELL BIOLOGY 2015; 32:4A.8.1-4A.8.17. [PMID: 25640819 PMCID: PMC4402211 DOI: 10.1002/9780470151808.sc04a08s32] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
This unit describes protocols for evaluating the pluripotency of embryonic and induced pluripotent stem cells using a teratoma formation assay. Cells are prepared for injection and transplanted into immunodeficient mice at the gastrocnemius muscle, a site well suited for teratoma growth and surgical access. Teratomas that form from the cell transplants are explanted, fixed in paraformaldehyde, and embedded in paraffin. These preserved samples are sectioned, stained, and analyzed. Pluripotency of a cell line is confirmed by whether the teratoma contains tissues derived from each of the embryonic germ layers: endoderm, mesoderm, and ectoderm. Alternatively, explanted and fixed teratomas can be cryopreserved for immunohistochemistry, which allows for more detailed identification of specific tissue types present in the samples.
Collapse
Affiliation(s)
- Raman V Nelakanti
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California
- Departments of Medicine and Radiology (Molecular Imaging Program), Stanford University School of Medicine, Stanford, California
- Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California
| | - Nigel G Kooreman
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California
- Departments of Medicine and Radiology (Molecular Imaging Program), Stanford University School of Medicine, Stanford, California
- Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California
- Departments of Medicine and Radiology (Molecular Imaging Program), Stanford University School of Medicine, Stanford, California
- Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
93
|
Lee SW, Jeon TJ, Biswal S. Effect of local treatment with adipose tissue-derived mesenchymal stem cells in the early tumorigenesis of osteosarcoma. Oncol Rep 2015; 33:1381-7. [PMID: 25572125 DOI: 10.3892/or.2015.3711] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 12/12/2014] [Indexed: 11/05/2022] Open
Abstract
There are conflicting data describing the effect of mesenchymal stem cells (MSCs) on tumorigenesis. The present study aimed to determine the survival rate and effect of adipose tissue-derived MSCs (ADMSCs) in tumor growth using bioluminescence imaging (BLI) and ultrasound (US) in an osteosarcoma xenograft model. Firefly luciferase-expressing ADMSCs combined with the osteosarcoma cell line UMR-106 in 4 different proportions (5, 10, 15 and 25%, named G1-G4, respectively) were xenografted into the right flanks of nude mice. The same number of UMR-106 cells was inoculated into the contralateral side of each mouse. Serial bioluminescence images were captured over 16 days to monitor the presence of ADMSCs in each group of 5 animals. The tumor volume was measured by ultra-high resolution US, and the tumor volume ratio (AMDSC mixed xenograft/control xenograft) was obtained to evaluate the effect of AMDSCs on tumor growth. Immunohistochemistry was performed to confirm the distribution of residual AMDSCs in the tumor. In G1, G2 and G3, the suppression of tumor growth by AMDSCs was noted in 2/5, 4/5 and 4/5 mice, respectively. However, accelerated tumor growth was noted in G4, which had the highest proportion of ADMSCs. The tumor volume ratio was significantly lower in G2 and G3 compared to G4, by Mann-Whitney U test (P=0.0159). Bioluminescence images demonstrated a serial decrement of the reporter gene for ADMSCs in the tumor mass without evidence of proliferation. Immunohistochemistry staining revealed minimal residual ADMSCs in the tumor periphery. Taken together, our data revealed that direct inoculation of ADMSCs into a tumor xenograft caused the death of the majority of ADMSCs in the tumor mass. Furthermore, relatively low proportions of ADMSCs suppressed the growth of osteosarcoma, while higher proportions showed a tumor-promoting effect.
Collapse
Affiliation(s)
- Sheen-Woo Lee
- Department of Radiology, Gil Hospital, Gachon University School of Medicine and Science, Incheon 406‑799, Republic of Korea
| | - Tae Joo Jeon
- Department of Nuclear Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 135-720, Republic of Korea
| | - Sandip Biswal
- Department of Radiology, Stanford University School of Medicine, Stanford, CA 94305-5105, USA
| |
Collapse
|
94
|
Liu J, Fan W, Liu M, Lin X, Wang Y, Wang F, Chen X, Cao F, Liang J. Spatial vascular volume fraction imaging for quantitative assessment of angiogenesis. Mol Imaging Biol 2015; 16:362-71. [PMID: 24158404 DOI: 10.1007/s11307-013-0694-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE The purpose of this study is to set up a quantitative imaging strategy for therapeutic angiogenesis and monitoring the spatial distribution and survival of the transplanted mesenchymal stem cells (MSCs) in vivo simultaneously. PROCEDURES Mouse adipose-derived MSCs (AD-MSCs) were isolated from firefly luciferase and enhanced green fluorescent protein positive transgenic mice, and implanted intramuscularly into hindlimbs of C57BL/6 mice. Serial spatial vascular volume fraction (SVVF) imaging was performed to quantitatively assess angiogenesis by calculating the spatially explicit vascular volume. The hybrid microcomputed tomography angiography/bioluminescence tomography (micro-CTA/BLT) was used to track the fate of AD-MSCs in vivo. Laser Doppler perfusion imaging (LDPI) was used to evaluate blood perfusion. Ex vivo conventional methods were performed to cross-validate the therapeutic angiogenesis. RESULTS There was a linear correlation relation between the cell number and Fluc/Fluc protein signal intensity in AD-MSCs via BLT. LDPI showed improved perfusion rate in mice treated with cytotherapy, compared to control mice. Furthermore, angiogenesis assessed by SVVF was 10.67 ± 0.41 %, 13.99 ± 0.28 %, and 23.50 ± 1.23 % on days 7, 14, and 28 post-transplantation of AD-MSCs, respectively. Vascular densities of the longitudinally monitored ischemic hindlimbs were significantly higher than those at early time points and controls, which was also confirmed by vascular corrosion casting, scanning electron microscopic imaging, and histological analysis. CONCLUSIONS Hybrid high-resolution micro-CTA/BLT enabled monitoring and quantitative assessment of cytotherapeutically induced angiogenesis in vivo.
Collapse
Affiliation(s)
- Junting Liu
- School of Life Science and Technology, Xidian University, P.O. Box 97, , No. 2 South Taibai Rd, Xi'an, 710071, China,
| | | | | | | | | | | | | | | | | |
Collapse
|
95
|
Abstract
Stem cell-based interventions aim to use special regenerative cells (stem cells) to facilitate neuronal function beyond the site of the injury. Many studies involving animal models of spinal cord injury (SCI) suggest that certain stem cell-based therapies may restore function after SCI. Currently, in case of spinal cord injuries, new discoveries with clinical implications have been continuously made in basic stem cell research, and stem cell-based approaches are advancing rapidly toward application in patients. There is a huge base of preclinical evidence in vitro and in animal models which suggests the safety and clinical efficacy of cellular therapies after SCI. Despite this, data from clinical studies is not very encouraging and at times confounding. Here, we have attempted to cover preclinical and clinical evidence base dealing with safety, feasibility and efficacy of cell based interventions after SCI. The limitations of preclinical data and the reasons underlying its failure to translate in a clinical setting are also discussed. Based on the evidence base, it is suggested that a multifactorial approach is required to address this situation. Need for standardized, stringently designed multi-centric clinical trials for obtaining validated proof of evidence is also highlighted.
Collapse
Affiliation(s)
- Harvinder Singh Chhabra
- Spine Service, Indian Spinal Injuries Centre, Vasant Kunj, New Delhi, India,Address for correspondence: Dr. Harvinder Singh Chhabra, Indian Spinal Injuries Centre, Sector C, Vasant Kunj, New Delhi - 110 070, India. E-mail:
| | - Kanchan Sarda
- Spine Service, Indian Spinal Injuries Centre, Vasant Kunj, New Delhi, India
| |
Collapse
|
96
|
Ding D, Mao D, Li K, Wang X, Qin W, Liu R, Chiam DS, Tomczak N, Yang Z, Tang BZ, Kong D, Liu B. Precise and long-term tracking of adipose-derived stem cells and their regenerative capacity via superb bright and stable organic nanodots. ACS NANO 2014; 8:12620-12631. [PMID: 25427294 DOI: 10.1021/nn505554y] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Monitoring and understanding long-term fate and regenerative therapy of administrated stem cells in vivo is of great importance. Herein we report organic nanodots with aggregation-induced emission characteristics (AIE dots) for long-term tracking of adipose-derived stem cells (ADSCs) and their regenerative capacity in living mice. The AIE dots possess high fluorescence (with a high quantum yield of 25±1%), excellent biological and photophysical stabilities, low in vivo toxicity, and superb retention in living ADSCs with negligible interference on their pluripotency and secretome. These AIE dots also exhibit superior in vitro cell tracking capability compared to the most popular commercial cell trackers, PKH26 and Qtracker 655. In vivo quantitative studies with bioluminescence and GFP labeling as the controls reveal that the AIE dots can precisely and quantitatively report the fate of ADSCs and their regenerative capacity for 42 days in an ischemic hind limb bearing mouse model.
Collapse
Affiliation(s)
- Dan Ding
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, and Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University , Tianjin 300071, P. R. China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
97
|
Decuzzi P. Patient-specific computational modeling and magnetic nanoconstructs: tools for maximizing the efficacy of stem cell-based therapies. Methodist Debakey Cardiovasc J 2014; 9:223-8. [PMID: 24298315 DOI: 10.14797/mdcj-9-4-223] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Stem cell transplantation has the potential to restore heart function following myocardial infarction. However, the success of any stem cell-based therapy is critically linked to the effective homing and early engraftment of the injected cells at the infarcted site. Here, a hierarchical multiscale computational model is proposed for predicting the patient-specific vascular transport and intratissue homing and migration of stem cells injected either systemically or locally. Starting with patient-specific data, such as the vascular geometry, blood flow, and location of the infarcted area, the computational model can be used to perform parametric analysis to identify optimal injection conditions in terms of administration route, injection site, catheter type, and infusion velocity. In addition to this, a new generation of magnetic nanoconstructs is introduced for labeling stem cells and monitoring their behavior in vivo via magnetic resonance imaging. These nanoconstructs also can be used for multimodal imaging, merging MRI and nuclear imaging, and the intracellular delivery of active agents to support stem cell differentiation. The convergence of computational modeling and novel nanoconstructs for stem cell labeling could improve our understanding in cell homing and early engraftment at the infarcted site and thus pave the way to more effective stem cell-based therapies.
Collapse
Affiliation(s)
- Paolo Decuzzi
- Houston Methodist Hospital Research Institute, Houston Methodist Hospital, Houston, Texas
| |
Collapse
|
98
|
Kim JE, Kalimuthu S, Ahn BC. In vivo cell tracking with bioluminescence imaging. Nucl Med Mol Imaging 2014; 49:3-10. [PMID: 25774232 DOI: 10.1007/s13139-014-0309-x] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 11/06/2014] [Accepted: 11/11/2014] [Indexed: 12/29/2022] Open
Abstract
Molecular imaging is a fast growing biomedical research that allows the visual representation, characterization and quantification of biological processes at the cellular and subcellular levels within intact living organisms. In vivo tracking of cells is an indispensable technology for development and optimization of cell therapy for replacement or renewal of damaged or diseased tissue using transplanted cells, often autologous cells. With outstanding advantages of bioluminescence imaging, the imaging approach is most commonly applied for in vivo monitoring of transplanted stem cells or immune cells in order to assess viability of administered cells with therapeutic efficacy in preclinical small animal models. In this review, a general overview of bioluminescence is provided and recent updates of in vivo cell tracking using the bioluminescence signal are discussed.
Collapse
Affiliation(s)
- Jung Eun Kim
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, 50, Samduk 2-ga, Jung Gu, Daegu, Republic of Korea 700-721
| | - Senthilkumar Kalimuthu
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, 50, Samduk 2-ga, Jung Gu, Daegu, Republic of Korea 700-721
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, 50, Samduk 2-ga, Jung Gu, Daegu, Republic of Korea 700-721
| |
Collapse
|
99
|
|
100
|
Moudgil R, Dick AJ. Regenerative Cell Imaging in Cardiac Repair. Can J Cardiol 2014; 30:1323-34. [DOI: 10.1016/j.cjca.2014.08.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 08/29/2014] [Accepted: 08/29/2014] [Indexed: 01/03/2023] Open
|