201
|
Di Felice V, Barone R, Nardone G, Forte G. Cardiac tissue engineering: a reflection after a decade of hurry. Front Physiol 2014; 5:365. [PMID: 25295012 PMCID: PMC4171986 DOI: 10.3389/fphys.2014.00365] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 09/05/2014] [Indexed: 11/13/2022] Open
Affiliation(s)
- Valentina Di Felice
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo Palermo, Italy
| | - Rosario Barone
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo Palermo, Italy ; Department of Stress Biology, Epigenetics and Biomarkers, Euro-Mediterranean Institute of Science and Technology (IEMEST) Palermo, Italy
| | - Giorgia Nardone
- Integrated Center for Cell Therapy and Regenerative Medicine (ICCT), International Clinical Research Center, St. Anne's University Hospital Brno, Czech Republic
| | - Giancarlo Forte
- Integrated Center for Cell Therapy and Regenerative Medicine (ICCT), International Clinical Research Center, St. Anne's University Hospital Brno, Czech Republic
| |
Collapse
|
202
|
Affiliation(s)
- James B Papizan
- From the Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas
| | - Eric N Olson
- From the Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas.
| |
Collapse
|
203
|
Primed 3D injectable microniches enabling low-dosage cell therapy for critical limb ischemia. Proc Natl Acad Sci U S A 2014; 111:13511-6. [PMID: 25197069 DOI: 10.1073/pnas.1411295111] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The promise of cell therapy for repair and restoration of damaged tissues or organs relies on administration of large dose of cells whose healing benefits are still limited and sometimes irreproducible due to uncontrollable cell loss and death at lesion sites. Using a large amount of therapeutic cells increases the costs for cell processing and the risks of side effects. Optimal cell delivery strategies are therefore in urgent need to enhance the specificity, efficacy, and reproducibility of cell therapy leading to minimized cell dosage and side effects. Here, we addressed this unmet need by developing injectable 3D microscale cellular niches (microniches) based on biodegradable gelatin microcryogels (GMs). The microniches are constituted by in vitro priming human adipose-derived mesenchymal stem cells (hMSCs) seeded within GMs resulting in tissue-like ensembles with enriched extracellular matrices and enhanced cell-cell interactions. The primed 3D microniches facilitated cell protection from mechanical insults during injection and in vivo cell retention, survival, and ultimate therapeutic functions in treatment of critical limb ischemia (CLI) in mouse models compared with free cell-based therapy. In particular, 3D microniche-based therapy with 10(5) hMSCs realized better ischemic limb salvage than treatment with 10(6) free-injected hMSCs, the minimum dosage with therapeutic effects for treating CLI in literature. To the best of our knowledge, this is the first convincing demonstration of injectable and primed cell delivery strategy realizing superior therapeutic efficacy for treating CLI with the lowest cell dosage in mouse models. This study offers a widely applicable cell delivery platform technology to boost the healing power of cell regenerative therapy.
Collapse
|
204
|
|
205
|
López-Ruiz E, Perán M, Picón-Ruiz M, García MA, Carrillo E, Jiménez-Navarro M, Hernández MC, Prat I, De Teresa E, Marchal JA. Cardiomyogenic differentiation potential of human endothelial progenitor cells isolated from patients with myocardial infarction. Cytotherapy 2014; 16:1229-37. [DOI: 10.1016/j.jcyt.2014.05.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Revised: 05/09/2014] [Accepted: 05/12/2014] [Indexed: 11/16/2022]
|
206
|
Huang CC, Liao ZX, Chen DY, Hsiao CW, Chang Y, Sung HW. Injectable cell constructs fabricated via culture on a thermoresponsive methylcellulose hydrogel system for the treatment of ischemic diseases. Adv Healthc Mater 2014; 3:1133-48. [PMID: 24470263 DOI: 10.1002/adhm.201300605] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Revised: 12/06/2013] [Indexed: 01/06/2023]
Abstract
Cell transplantation via direct intramuscular injection is a promising therapy for patients with ischemic diseases. However, following injections, retention of transplanted cells in engrafted areas remains problematic, and can be deleterious to cell-transplantation therapy. In this Progress Report, a thermoresponsive hydrogel system composed of aqueous methylcellulose (MC) blended with phosphate-buffered saline is constructed to grow cell sheet fragments and cell bodies for the treatment of ischemic diseases. The as-prepared MC hydrogel system undergoes a sol-gel reversible transition upon heating or cooling at ≈32 °C. Via this unique property, the grown cell sheet fragments (cell bodies) can be harvested without using proteolytic enzymes; consequently, their inherent extracellular matrices (ECMs) and integrative adhesive agents remain well preserved. In animal studies using rats and pigs with experimentally created myocardial infarction, the injected cell sheet fragments (cell bodies) become entrapped in the interstices of muscular tissues and adhere to engraftment sites, while a minimal number of cells exist in the group receiving dissociated cells. Moreover, transplantation of cell sheet fragments (cell bodies) significantly increases vascular density, thereby improving the function of an infarcted heart. These experimental results demonstrate that cell sheet fragments (cell bodies) function as a cell-delivery construct by providing a favorable ECM environment to retain transplanted cells locally and consequently, improving the efficacy of therapeutic cell transplantation.
Collapse
Affiliation(s)
- Chieh-Cheng Huang
- Department of Chemical Engineering and Institute of Biomedical Engineering; National Tsing Hua University; Hsinchu 30013 Taiwan (ROC)
| | - Zi-Xian Liao
- Department of Chemical Engineering and Institute of Biomedical Engineering; National Tsing Hua University; Hsinchu 30013 Taiwan (ROC)
| | - Ding-Yuan Chen
- Department of Chemical Engineering and Institute of Biomedical Engineering; National Tsing Hua University; Hsinchu 30013 Taiwan (ROC)
| | - Chun-Wen Hsiao
- Department of Chemical Engineering and Institute of Biomedical Engineering; National Tsing Hua University; Hsinchu 30013 Taiwan (ROC)
| | - Yen Chang
- Division of Cardiovascular Surgery; Veterans General Hospital at Taichung; Taichung 40705 Taiwan (ROC)
- College of Medicine, National Yang-Ming University; Taipei 11221 Taiwan (ROC)
| | - Hsing-Wen Sung
- Department of Chemical Engineering and Institute of Biomedical Engineering; National Tsing Hua University; Hsinchu 30013 Taiwan (ROC)
| |
Collapse
|
207
|
Reis LA, Chiu LLY, Feric N, Fu L, Radisic M. Biomaterials in myocardial tissue engineering. J Tissue Eng Regen Med 2014; 10:11-28. [PMID: 25066525 DOI: 10.1002/term.1944] [Citation(s) in RCA: 125] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Revised: 04/17/2014] [Accepted: 06/16/2014] [Indexed: 12/31/2022]
Abstract
Cardiovascular disease is the leading cause of death in the developed world, and as such there is a pressing need for treatment options. Cardiac tissue engineering emerged from the need to develop alternative sources and methods of replacing tissue damaged by cardiovascular diseases, as the ultimate treatment option for many who suffer from end-stage heart failure is a heart transplant. In this review we focus on biomaterial approaches to augmenting injured or impaired myocardium, with specific emphasis on: the design criteria for these biomaterials; the types of scaffolds - composed of natural or synthetic biomaterials or decellularized extracellular matrix - that have been used to develop cardiac patches and tissue models; methods to vascularize scaffolds and engineered tissue; and finally, injectable biomaterials (hydrogels) designed for endogenous repair, exogenous repair or as bulking agents to maintain ventricular geometry post-infarct. The challenges facing the field and obstacles that must be overcome to develop truly clinically viable cardiac therapies are also discussed.
Collapse
Affiliation(s)
- Lewis A Reis
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, ON, Canada
| | - Loraine L Y Chiu
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, ON, Canada
| | - Nicole Feric
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, ON, Canada
| | - Lara Fu
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, ON, Canada
| | - Milica Radisic
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, ON, Canada.,Department of Chemical Engineering and Applied Chemistry, University of Toronto, ON, Canada
| |
Collapse
|
208
|
In-vivo comparison of the acute retention of stem cell derivatives and fibroblasts after intramyocardial transplantation in the mouse model. Eur J Nucl Med Mol Imaging 2014; 41:2325-36. [DOI: 10.1007/s00259-014-2858-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Accepted: 07/04/2014] [Indexed: 01/15/2023]
|
209
|
Sommer P. Can stem cells really regenerate the human heart? Use your noggin, dickkopf! Lessons from developmental biology. Cardiovasc J Afr 2014; 24:189-93. [PMID: 24217168 PMCID: PMC3748454 DOI: 10.5830/cvja-2013-045] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 06/07/2013] [Indexed: 11/06/2022] Open
Abstract
The human heart is the first organ to develop and its development is fairly well characterised. In theory, the heart has the capacity to regenerate, as its cardiomyocytes may be capable of cell division and the adult heart contains a cardiac stem cell niche, presumably capable of differentiating into cardiomyocytes and other cardiac-associated cell types. However, as with most other organs, these mechanisms are not activated upon serious injury. Several experimental options to induce regeneration of the damaged heart tissue are available: activate the endogenous cardiomyocytes to divide, coax the endogenous population of stem cells to divide and differentiate, or add exogenous cell-based therapy to replace the lost cardiac tissue. This review is a summary of the recent research into all these avenues, discussing the reasons for the limited successes of clinical trials using stem cells after cardiac injury and explaining new advances in basic science. It concludes with a reiteration that chances of successful regeneration would be improved by understanding and implementing the basics of heart development and stem cell biology.
Collapse
Affiliation(s)
- Paula Sommer
- School of Life Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
210
|
Abstract
Human heart failure (HF) is one of the leading causes of morbidity and mortality worldwide. Currently, heart transplantation and implantation of mechanical devices represent the only available treatments for advanced HF. Two alternative strategies have emerged to treat patients with HF. One approach relies on transplantation of exogenous stem cells (SCs) of non-cardiac or cardiac origin to induce cardiac regeneration and improve ventricular function. Another complementary strategy relies on stimulation of the endogenous regenerative capacity of uninjured cardiac progenitor cells to rebuild cardiac muscle and restore ventricular function. Various SC types and delivery strategies have been examined in the experimental and clinical settings; however, neither the ideal cell type nor the cell delivery method for cardiac cell therapy has yet emerged. Although the use of bone marrow (BM)-derived cells, most frequently exploited in clinical trials, appears to be safe, the results are controversial. Two recent randomized trials have failed to document any beneficial effects of intracardiac delivery of autologous BM mononuclear cells on cardiac function of patients with HF. The remarkable discovery that various populations of cardiac progenitor cells (CPCs) are present in the adult human heart and that it possesses limited regeneration capacity has opened a new era in cardiac repair. Importantly, unlike BM-derived SCs, autologous CPCs from myocardial biopsies cultured and subsequently delivered by coronary injection to patients have given positive results. Although these data are promising, a better understanding of how to control proliferation and differentiation of CPCs, to enhance their recruitment and survival, is required before CPCs become clinically applicable therapeutics.
Collapse
Affiliation(s)
- Alexander T Akhmedov
- The Molecular Cardiology and Neuromuscular Institute, 75 Raritan Ave., Highland Park, NJ, 08904, USA
| | | |
Collapse
|
211
|
Kang BJ, Kim H, Lee SK, Kim J, Shen Y, Jung S, Kang KS, Im SG, Lee SY, Choi M, Hwang NS, Cho JY. Umbilical-cord-blood-derived mesenchymal stem cells seeded onto fibronectin-immobilized polycaprolactone nanofiber improve cardiac function. Acta Biomater 2014; 10:3007-17. [PMID: 24657671 DOI: 10.1016/j.actbio.2014.03.013] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 03/08/2014] [Accepted: 03/10/2014] [Indexed: 12/28/2022]
Abstract
Stem cells seeded onto biofunctional materials have greater potency for therapeutic applications. We investigated whether umbilical-cord-blood-derived mesenchymal stem cell (UCB-MSC)-seeded fibronectin (FN)-immobilized polycaprolactone (PCL) nanofibers could improve cardiac function and inhibit left ventricle (LV) remodeling in a rat model of myocardial infarction (MI). Aligned nanofibers were uniformly coated with poly(glycidyl methacrylate) by initiated chemical vapor deposition followed by covalent immobilization of FN proteins. The degree of cell elongation and adhesion efficacy were improved by FN immobilization. Furthermore, genes related to angiogenesis and mesenchymal differentiations were up-regulated in the FN-immobilized PCL nanofibers in comparison to control PCL nanofibers in vitro. 4 weeks after the transplantation in the rat MI model, the echocardiogram showed that the UCB-MSC-seeded FN-immobilized PCL nanofiber group increased LV ejection fraction and fraction shortening as compared to the non-treated control and acellular FN-immobilized PCL nanofiber groups. Histological analysis indicated that the implantation of UCB-MSCs with FN-immobilized PCL nanofibers induced a decrease in MI size and fibrosis, and an increase in scar thickness. This study indicates that FN-immobilized biofunctional PCL nanofibers could be an effective carrier for UCB-MSC transplantation for the treatment of MI.
Collapse
Affiliation(s)
- Byung-Jae Kang
- Department of Veterinary Biochemistry, BK21 Plus and Research Institute of Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Hwan Kim
- School of Chemical and Biological Engineering, BioMAX Institute, Seoul National University, Seoul, Republic of Korea
| | - Seul Ki Lee
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Joohyun Kim
- Department of Veterinary Biochemistry, BK21 Plus and Research Institute of Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Yiming Shen
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Sunyoung Jung
- Department of Veterinary Medical Imaging, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Kyung-Sun Kang
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Sung Gap Im
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Technology, Daejeon, Republic of Korea
| | - So Yeong Lee
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Mincheol Choi
- Department of Veterinary Medical Imaging, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Nathaniel S Hwang
- School of Chemical and Biological Engineering, BioMAX Institute, Seoul National University, Seoul, Republic of Korea.
| | - Je-Yoel Cho
- Department of Veterinary Biochemistry, BK21 Plus and Research Institute of Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
212
|
Smiley D, Smith MA, Carreira V, Jiang M, Koch SE, Kelley M, Rubinstein J, Jones WK, Tranter M. Increased fibrosis and progression to heart failure in MRL mice following ischemia/reperfusion injury. Cardiovasc Pathol 2014; 23:327-34. [PMID: 25035060 DOI: 10.1016/j.carpath.2014.06.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 06/05/2014] [Accepted: 06/05/2014] [Indexed: 11/25/2022] Open
Abstract
The cardiac regenerative capacity of MRL/MpJ mouse remains a controversy. Although the MRL mouse has been reported to exhibit minimal scarring and subsequent cardiac regeneration after cryoinjury of the right ventricle, multiple studies have been unable to replicate this cardiac regenerative capacity after both cryogenic and coronary ligation cardiac injury. Therefore, we evaluated the cardiac regenerative wound-healing response and functional recovery of MRL mice compared to C57 mice, in response to a clinically relevant left ventricular (LV) coronary ligation. Male MRL/MpJ+/+ and C57BL/6 mice underwent left coronary artery ligation followed by reperfusion. Cardiac function was evaluated by echocardiography [LV ejection fraction (LVEF), LV end-diastolic volume (LVEDV), LV mass, wall thickness] at 24 hours post-ischemia and weekly for 13 weeks thereafter. Hearts were also analyzed histologically for individual cardiomyocyte hypertrophy and cardiac fibrosis. Our results show that contrary to prior reports of cardiac regenerations, MRL mice progress to heart failure more rapidly following I/R injury as marked by a significant decrease in LVEF, increase in LVEDV, LV mass, individual myocyte size, and fibrosis in the post-ischemic myocardium. Therefore, we conclude that MRL mice do not exhibit regeneration of the LV or enhanced functional improvement in response to coronary ligation. However, unlike prior studies, we matched initial infarct size in MRL and C57 mice, used high frequency echocardiography, and histological analysis to reach this conclusion. The prospect of cardiac regeneration after ischemia in MRL mice seems to have attenuated interest, given the multiple negative studies and the promise of stem cell cardiac regeneration. However, our novel observation that MRL may possess an impaired compensated hypertrophy response makes the MRL mouse strain an interesting model in the study of cardiac hypertrophy.
Collapse
Affiliation(s)
- Dia Smiley
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, College of Medicine, Cincinnati, OH
| | - Margaret A Smith
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, College of Medicine, Cincinnati, OH
| | - Vinicius Carreira
- Department of Environmental Health, University of Cincinnati, College of Medicine, Cincinnati, OH
| | - Min Jiang
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, College of Medicine, Cincinnati, OH
| | - Sheryl E Koch
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, College of Medicine, Cincinnati, OH
| | - Melissa Kelley
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, College of Medicine, Cincinnati, OH
| | - Jack Rubinstein
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, College of Medicine, Cincinnati, OH
| | - W Keith Jones
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, College of Medicine, Cincinnati, OH; Department of Pharmacology & Cell Biophysics, University of Cincinnati, College of Medicine, Cincinnati, OH
| | - Michael Tranter
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, College of Medicine, Cincinnati, OH.
| |
Collapse
|
213
|
Liu W, Li Y, Zeng Y, Zhang X, Wang J, Xie L, Li X, Du Y. Microcryogels as injectable 3-D cellular microniches for site-directed and augmented cell delivery. Acta Biomater 2014; 10:1864-75. [PMID: 24342043 DOI: 10.1016/j.actbio.2013.12.008] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Revised: 11/25/2013] [Accepted: 12/09/2013] [Indexed: 01/12/2023]
Abstract
The success of cell therapy for tissue repair and regeneration demands efficient and reliable cell delivery methods. Here we established a novel microengineered cryogel (microcryogel) array chip containing microcryogels with predefined size and shape as injectable cell delivery vehicles. The microscale macroporous cryogels enabled automatic and homogeneous loading of tailored cellular niches (e.g. cells, matrices, bioactive factors) and could be easily harvested from the ready-to-use array chip. In contrast to microscale hydrogels, microcryogels exhibited excellent elasticity and could retain their shape and integrity after injection through the microsyringe routinely used for cell therapy. Human mesenchymal stromal cells loaded within microcryogels could be shielded from the mechanical insult and necrosis caused by during direct cell injection. After subcutaneous injection to the mice, cell-loaded microcryogels exhibited concentrated localization and enhanced retention at the injection site compared to dissociated cells. To demonstrate the potential therapeutic application for ischemic diseases, site-directed induction of angiogenesis was achieved subcutaneously in mice 2weeks after injection of NIH/3T3 fibroblast-loaded microcryogels, indicating long-term engraftment, accumulative paracrine stimulation and augmented host tissue integration. Our results convincingly showed the great promise of microcryogels as 3-D cellular microniches and injectable cell delivery vehicles to tackle major challenges faced by cell therapy-based regenerative medicine including shear-induced damages, uncontrolled localization, poor retention, limited cellular survival and functionalities in vivo.
Collapse
|
214
|
Lei Y, Jeong D, Xiao J, Schaffer DV. Developing Defined and Scalable 3D Culture Systems for Culturing Human Pluripotent Stem Cells at High Densities. Cell Mol Bioeng 2014; 7:172-183. [PMID: 25419247 DOI: 10.1007/s12195-014-0333-z] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Human pluripotent stem cells (hPSCs) - including embryonic stem cells (hESCs) and induced pluripotent stem cells (hiPSCs) - are very promising candidates for cell therapies, tissue engineering, high throughput pharmacology screens, and toxicity testing. These applications require large numbers of high quality cells; however, scalable production of human pluripotent stem cells and their derivatives at a high density and under well-defined conditions has been a challenge. We recently reported a simple, efficient, fully defined, scalable, and good manufacturing practice (GMP) compatible 3D culture system based on a thermoreversible hydrogel for hPSC expansion and differentiation. Here, we describe additional design rationale and characterization of this system. For instance, we have determined that culturing hPSCs as a suspension in a liquid medium can exhibit lower volumetric yields due to cell agglomeration and possible shear force-induced cell loss. By contrast, using hydrogels as 3D scaffolds for culturing hPSCs reduces aggregation and may insulate from shear forces. Additionally, hydrogel-based 3D culture systems can support efficient hPSC expansion and differentiation at a high density if compatible with hPSC biology. Finally, there are considerable opportunities for future development to further enhance hydrogel-based 3D culture systems for producing hPSCs and their progeny.
Collapse
Affiliation(s)
- Yuguo Lei
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, California, 94720, USA ; Department of Bioengineering, University of California, Berkeley, Berkeley, California, 94720, USA ; California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, California, 94720, USA ; Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, California, 94720, USA
| | - Daeun Jeong
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California, 94720, USA
| | - Jifang Xiao
- Department of Bioengineering, University of California, Berkeley, Berkeley, California, 94720, USA
| | - David V Schaffer
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, California, 94720, USA ; Department of Bioengineering, University of California, Berkeley, Berkeley, California, 94720, USA ; California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, California, 94720, USA ; Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, California, 94720, USA
| |
Collapse
|
215
|
Sachlos E, Kerstetter-Fogle A. Highlights from the latest articles in regenerative medicine. Regen Med 2014; 9:145-7. [PMID: 24750055 DOI: 10.2217/rme.14.6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
216
|
Xiong JW, Chang NN. Recent advances in heart regeneration. ACTA ACUST UNITED AC 2014; 99:160-9. [PMID: 24078494 DOI: 10.1002/bdrc.21039] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2013] [Revised: 07/27/2013] [Accepted: 07/27/2013] [Indexed: 12/25/2022]
Abstract
Although cardiac stem cells (CSCs) and tissue engineering are very promising for cardiac regenerative medicine, studies with model organisms for heart regeneration will provide alternative therapeutic targets and opportunities. Here, we present a review on heart regeneration, with a particular focus on the most recent work in mouse and zebrafish. We attempt to summarize the recent progresses and bottlenecks of CSCs and tissue engineering for heart regeneration; and emphasize what we have learned from mouse and zebrafish regenerative models on discovering crucial genetic and epigenetic factors for stimulating heart regeneration; and speculate the potential application of these regenerative factors for heart failure. A brief perspective highlights several important and promising research directions in this exciting field.
Collapse
Affiliation(s)
- Jing-Wei Xiong
- are from Institute of Molecular Medicine, Peking University, Beijing, 100871, China and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100871, China
| | | |
Collapse
|
217
|
Murray IR, West CC, Hardy WR, James AW, Park TS, Nguyen A, Tawonsawatruk T, Lazzari L, Soo C, Péault B. Natural history of mesenchymal stem cells, from vessel walls to culture vessels. Cell Mol Life Sci 2014; 71:1353-74. [PMID: 24158496 PMCID: PMC11113613 DOI: 10.1007/s00018-013-1462-6] [Citation(s) in RCA: 188] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2013] [Revised: 08/17/2013] [Accepted: 08/23/2013] [Indexed: 02/06/2023]
Abstract
Mesenchymal stem/stromal cells (MSCs) can regenerate tissues by direct differentiation or indirectly by stimulating angiogenesis, limiting inflammation, and recruiting tissue-specific progenitor cells. MSCs emerge and multiply in long-term cultures of total cells from the bone marrow or multiple other organs. Such a derivation in vitro is simple and convenient, hence popular, but has long precluded understanding of the native identity, tissue distribution, frequency, and natural role of MSCs, which have been defined and validated exclusively in terms of surface marker expression and developmental potential in culture into bone, cartilage, and fat. Such simple, widely accepted criteria uniformly typify MSCs, even though some differences in potential exist, depending on tissue sources. Combined immunohistochemistry, flow cytometry, and cell culture have allowed tracking the artifactual cultured mesenchymal stem/stromal cells back to perivascular anatomical regions. Presently, both pericytes enveloping microvessels and adventitial cells surrounding larger arteries and veins have been described as possible MSC forerunners. While such a vascular association would explain why MSCs have been isolated from virtually all tissues tested, the origin of the MSCs grown from umbilical cord blood remains unknown. In fact, most aspects of the biology of perivascular MSCs are still obscure, from the emergence of these cells in the embryo to the molecular control of their activity in adult tissues. Such dark areas have not compromised intents to use these cells in clinical settings though, in which purified perivascular cells already exhibit decisive advantages over conventional MSCs, including purity, thorough characterization and, principally, total independence from in vitro culture. A growing body of experimental data is currently paving the way to the medical usage of autologous sorted perivascular cells for indications in which MSCs have been previously contemplated or actually used, such as bone regeneration and cardiovascular tissue repair.
Collapse
Affiliation(s)
- Iain R. Murray
- MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
- BHF Center for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Orthopedic Hospital Research Center and Broad Stem Cell Center, David Geffen School of Medicine, University of California, Los Angeles, USA
| | - Christopher C. West
- MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
- BHF Center for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Winters R. Hardy
- Orthopedic Hospital Research Center and Broad Stem Cell Center, David Geffen School of Medicine, University of California, Los Angeles, USA
- Indiana Center for Vascular Biology and Medicine, Indianapolis, USA
| | - Aaron W. James
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, USA
| | - Tea Soon Park
- Institute for Cell Engineering, Johns Hopkins School of Medicine, Baltimore, USA
| | - Alan Nguyen
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, USA
| | - Tulyapruek Tawonsawatruk
- MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
- BHF Center for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Lorenza Lazzari
- Cell Factory, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Chia Soo
- Division of Plastic and Reconstructive Surgery, Departments of Surgery and Orthopedic Surgery, David Geffen School of Medicine, University of California, Los Angeles, USA
| | - Bruno Péault
- MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
- BHF Center for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Orthopedic Hospital Research Center and Broad Stem Cell Center, David Geffen School of Medicine, University of California, Los Angeles, USA
| |
Collapse
|
218
|
Pfister O, Della Verde G, Liao R, Kuster GM. Regenerative therapy for cardiovascular disease. Transl Res 2014; 163:307-20. [PMID: 24378637 DOI: 10.1016/j.trsl.2013.12.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Revised: 11/04/2013] [Accepted: 12/05/2013] [Indexed: 11/25/2022]
Abstract
Recent insights into myocardial biology uncovered a hereto unknown regenerative capacity of the adult heart. The discovery of dividing cardiomyocytes and the identification and characterization of cardiac stem and progenitor cells with myogenic and angiogenic potential have generated new hopes that cardiac regeneration and repair might become a therapeutic option. During the past decade, multiple candidate cells have been proposed for cardiac regeneration, and their mechanisms of action in the myocardium have been explored. Initial clinical trials have focused on the use of bone marrow-derived cells to promote myocardial regeneration in ischemic heart disease and have yielded very mixed results, with no clear signs of clinically meaningful functional improvement. Although the efficiency of bona fide cardiomyocyte generation is generally low, stem cells delivered into the myocardium act mainly via paracrine mechanisms. More recent studies taking advantage of cardiac committed cells (eg, resident cardiac progenitor cells or primed cardiogenic mesenchymal stem cells) showed promising results in first clinical pilot trials. Also, transplantation of cardiomyogenic cells generated by induced pluripotent stem cells and genetic reprogramming of dividing nonmyocytes into cardiomyocytes may constitute attractive new regenerative approaches in cardiovascular medicine in the future. We discuss advantages and limitations of specific cell types proposed for cell-based therapy in cardiology and give an overview of the first clinical trials using this novel therapeutic approach in patients with cardiovascular disease.
Collapse
Affiliation(s)
- Otmar Pfister
- Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland; Division of Cardiology, University Hospital Basel, Basel, Switzerland.
| | - Giacomo Della Verde
- Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Ronglih Liao
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass
| | - Gabriela M Kuster
- Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland; Division of Cardiology, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
219
|
Abstract
Myocardial infarction triggers an intense inflammatory response that is essential for cardiac repair, but which is also implicated in the pathogenesis of postinfarction remodelling and heart failure. Signals in the infarcted myocardium activate toll-like receptor signalling, while complement activation and generation of reactive oxygen species induce cytokine and chemokine upregulation. Leukocytes recruited to the infarcted area, remove dead cells and matrix debris by phagocytosis, while preparing the area for scar formation. Timely repression of the inflammatory response is critical for effective healing, and is followed by activation of myofibroblasts that secrete matrix proteins in the infarcted area. Members of the transforming growth factor β family are critically involved in suppression of inflammation and activation of a profibrotic programme. Translation of these concepts to the clinic requires an understanding of the pathophysiological complexity and heterogeneity of postinfarction remodelling in patients with myocardial infarction. Individuals with an overactive and prolonged postinfarction inflammatory response might exhibit left ventricular dilatation and systolic dysfunction and might benefit from targeted anti-IL-1 or anti-chemokine therapies, whereas patients with an exaggerated fibrogenic reaction can develop heart failure with preserved ejection fraction and might require inhibition of the Smad3 (mothers against decapentaplegic homolog 3) cascade. Biomarker-based approaches are needed to identify patients with distinct pathophysiologic responses and to rationally implement inflammation-modulating strategies.
Collapse
|
220
|
Greiner JFW, Grunwald LM, Müller J, Sudhoff H, Widera D, Kaltschmidt C, Kaltschmidt B. Culture bag systems for clinical applications of adult human neural crest-derived stem cells. Stem Cell Res Ther 2014; 5:34. [PMID: 24629140 PMCID: PMC4055128 DOI: 10.1186/scrt422] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 03/11/2014] [Indexed: 11/15/2022] Open
Abstract
Introduction Facing the challenging treatment of neurodegenerative diseases as well as complex craniofacial injuries such as those common after cancer therapy, the field of regenerative medicine increasingly relies on stem cell transplantation strategies. Here, neural crest-derived stem cells (NCSCs) offer many promising applications, although scale up of clinical-grade processes prior to potential transplantations is currently limiting. In this study, we aimed to establish a clinical-grade, cost-reducing cultivation system for NCSCs isolated from the adult human nose using cGMP-grade Afc-FEP bags. Methods We cultivated human neural crest-derived stem cells from inferior turbinate (ITSCs) in a cell culture bag system using Afc-FEP bags in human blood plasma-supplemented medium. Investigations of viability, proliferation and expression profile of bag-cultured ITSCs were followed by DNA-content and telomerase activity determination. Cultivated ITSCs were introduced to directed in vitro differentiation assays to assess their potential for mesodermal and ectodermal differentiation. Mesodermal differentiation was determined using an enzyme activity assay (alkaline phosphatase, ALP), respective stainings (Alizarin Red S, Von Kossa and Oil Red O), and RT-PCR, while immunocytochemistry and synaptic vesicle recycling were applied to assay neuroectodermal differentiation of ITSCs. Results When cultivated within Afc-FEP bags, ITSCs grew three-dimensionally in a human blood plasma-derived matrix, thereby showing unchanged morphology, proliferation capability, viability and expression profile in comparison to three dimensionally-cultured ITSCs growing in standard cell culture plastics. Genetic stability of bag-cultured ITSCs was further accompanied by unchanged telomerase activity. Importantly, ITSCs retained their potential to differentiate into mesodermal cell types, particularly including ALP-active, Alizarin Red S-, and Von Kossa-positive osteogenic cell types, as well as adipocytes positive in Oil Red O assays. Bag culture further did not affect the potential of ITSCs to undergo differentiation into neuroectodermal cell types coexpressing β-III-tubulin and MAP2 and exhibiting the capability for synaptic vesicle recycling. Conclusions Here, we report for the first time the successful cultivation of human NCSCs within cGMP-grade Afc-FEP bags using a human blood plasma-supplemented medium. Our findings particularly demonstrate the unchanged differentiation capability and genetic stability of the cultivated NCSCs, suggesting the great potential of this culture system for future medical applications in the field of regenerative medicine.
Collapse
|
221
|
Budniatzky I, Gepstein L. Concise review: reprogramming strategies for cardiovascular regenerative medicine: from induced pluripotent stem cells to direct reprogramming. Stem Cells Transl Med 2014; 3:448-57. [PMID: 24591731 DOI: 10.5966/sctm.2013-0163] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Myocardial cell-replacement therapies are emerging as novel therapeutic paradigms for myocardial repair but are hampered by the lack of sources of autologous human cardiomyocytes. The recent advances in stem cell biology and in transcription factor-based reprogramming strategies may provide exciting solutions to this problem. In the current review, we describe the different reprogramming strategies that can give rise to cardiomyocytes for regenerative medicine purposes. Initially, we describe induced pluripotent stem cell technology, a method by which adult somatic cells can be reprogrammed to yield pluripotent stem cells that could later be coaxed ex vivo to differentiate into cardiomyocytes. The generated induced pluripotent stem cell-derived cardiomyocytes could then be used for myocardial cell transplantation and tissue engineering strategies. We also describe the more recent direct reprogramming approaches that aim to directly convert the phenotype of one mature cell type (fibroblast) to another (cardiomyocyte) without going through a pluripotent intermediate cell type. The advantages and shortcomings of each strategy for cardiac regeneration are discussed, along with the hurdles that need to be overcome on the road to clinical translation.
Collapse
Affiliation(s)
- Inbar Budniatzky
- Sohnis Research Laboratory for Cardiac Electrophysiology and Regenerative Medicine and Cardiology Department, Rambam Medical Center, Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel
| | | |
Collapse
|
222
|
Wang S, Zhou Y, Andreyev O, Hoyt RF, Singh A, Hunt T, Horvath KA. Overexpression of FABP3 inhibits human bone marrow derived mesenchymal stem cell proliferation but enhances their survival in hypoxia. Exp Cell Res 2014; 323:56-65. [PMID: 24583397 DOI: 10.1016/j.yexcr.2014.02.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Revised: 01/25/2014] [Accepted: 02/16/2014] [Indexed: 11/29/2022]
Abstract
Studying the proliferative ability of human bone marrow derived mesenchymal stem cells in hypoxic conditions can help us achieve the effective regeneration of ischemic injured myocardium. Cardiac-type fatty acid binding protein (FABP3) is a specific biomarker of muscle and heart tissue injury. This protein is purported to be involved in early myocardial development, adult myocardial tissue repair and responsible for the modulation of cell growth and proliferation. We have investigated the role of FABP3 in human bone marrow derived mesenchymal stem cells under ischemic conditions. MSCs from 12 donors were cultured either in standard normoxic or modified hypoxic conditions, and the differential expression of FABP3 was tested by quantitative (RT)PCR and western blot. We also established stable FABP3 expression in MSCs and searched for variation in cellular proliferation and differentiation bioprocesses affected by hypoxic conditions. We identified: (1) the FABP3 differential expression pattern in the MSCs under hypoxic conditions; (2) over-expression of FABP3 inhibited the growth and proliferation of the MSCs; however, improved their survival in low oxygen environments; (3) the cell growth factors and positive cell cycle regulation genes, such as PCNA, APC, CCNB1, CCNB2 and CDC6 were all down-regulated; while the key negative cell cycle regulation genes TP53, BRCA1, CASP3 and CDKN1A were significantly up-regulated in the cells with FABP3 overexpression. Our data suggested that FABP3 was up-regulated under hypoxia; also negatively regulated the cell metabolic process and the mitotic cell cycle. Overexpression of FABP3 inhibited cell growth and proliferation via negative regulation of the cell cycle and down-regulation of cell growth factors, but enhances cell survival in hypoxic or ischemic conditions.
Collapse
Affiliation(s)
- Suna Wang
- Cellular Biology Section, Cardiothoracic Surgery Research Program, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Yifu Zhou
- Cellular Biology Section, Cardiothoracic Surgery Research Program, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Oleg Andreyev
- Cellular Biology Section, Cardiothoracic Surgery Research Program, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Robert F Hoyt
- Cellular Biology Section, Cardiothoracic Surgery Research Program, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Avneesh Singh
- Cellular Biology Section, Cardiothoracic Surgery Research Program, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Timothy Hunt
- Cellular Biology Section, Cardiothoracic Surgery Research Program, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Keith A Horvath
- Cellular Biology Section, Cardiothoracic Surgery Research Program, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
223
|
Chen A, Ting S, Seow J, Reuveny S, Oh S. Considerations in designing systems for large scale production of human cardiomyocytes from pluripotent stem cells. Stem Cell Res Ther 2014; 5:12. [PMID: 24444355 PMCID: PMC4055057 DOI: 10.1186/scrt401] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Human pluripotent stem cell (hPSC)-derived cardiomyocytes have attracted attention as an unlimited source of cells for cardiac therapies. One of the factors to surmount to achieve this is the production of hPSC-derived cardiomyocytes at a commercial or clinical scale with economically and technically feasible platforms. Given the limited proliferation capacity of differentiated cardiomyocytes and the difficulties in isolating and culturing committed cardiac progenitors, the strategy for cardiomyocyte production would be biphasic, involving hPSC expansion to generate adequate cell numbers followed by differentiation to cardiomyocytes for specific applications. This review summarizes and discusses up-to-date two-dimensional cell culture, cell-aggregate and microcarrier-based platforms for hPSC expansion. Microcarrier-based platforms are shown to be the most suitable for up-scaled production of hPSCs. Subsequently, different platforms for directing hPSC differentiation to cardiomyocytes are discussed. Monolayer differentiation can be straightforward and highly efficient and embryoid body-based approaches are also yielding reasonable cardiomyocyte efficiencies, whereas microcarrier-based approaches are in their infancy but can also generate high cardiomyocyte yields. The optimal target is to establish an integrated scalable process that combines hPSC expansion and cardiomyocyte differentiation into a one unit operation. This review discuss key issues such as platform selection, bioprocess parameters, medium development, downstream processing and parameters that meet current good manufacturing practice standards.
Collapse
|
224
|
Rhett JM, Fann SA, Yost MJ. Purinergic signaling in early inflammatory events of the foreign body response: modulating extracellular ATP as an enabling technology for engineered implants and tissues. TISSUE ENGINEERING PART B-REVIEWS 2014; 20:392-402. [PMID: 24279914 DOI: 10.1089/ten.teb.2013.0554] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Purinergic signaling is a ubiquitous and vital aspect of mammalian biology in which purines--mainly adenosine triphosphate (ATP)--are released from cells through loss of membrane integrity (cell death), exocytosis, or transport/diffusion across membrane channels, and exert paracrine or autocrine signaling effects through three subclasses of well-characterized receptors: the P1 adenosine receptors, the P2X ionotropic nucleotide receptors, and the P2Y metabotropic receptors. ATP and its metabolites are released by damaged and stressed cells in injured tissues. The early events of wound healing, hemostasis, and inflammation are highly regulated by these signals through activation of purinergic receptors on platelets and neutrophils. Recent data have demonstrated that ATP signaling is of particular importance to targeting leukocytes to sites of injury. This is particularly relevant to the subject of implanted medical devices, engineered tissues, and grafts as all these technologies elicit a wound healing response with varying degrees of encapsulation, rejection, extrusion, or destruction of the tissue or device. Here, we review the biology of purinergic signaling and focus on ATP release and response mechanisms that pertain to the early inflammatory phase of wound healing. Finally, therapeutic options are explored, including a new class of peptidomimetic drugs based on the ATP-conductive channel connexin43.
Collapse
Affiliation(s)
- J Matthew Rhett
- Division of General Surgery, Department of Surgery, Medical University of South Carolina , Charleston, South Carolina
| | | | | |
Collapse
|
225
|
Abstract
Although amphibian and fish models of heart regeneration have existed for decades, a mammalian equivalent has long remained elusive. Our discovery of a brief postnatal window for heart regeneration in neonatal mice has led to the establishment of surgical models for cardiac regenerative studies in mammals for the first time. This protocol describes a 10-min surgical procedure to induce cardiac injury in 1-d-old neonatal mice. This allows for the analysis of cardiac regeneration after surgical amputation of the left ventricle (LV) (apical resection) and coronary artery occlusion (myocardial infarction (MI)). A comparative analysis of neonatal and adult responses to myocardial injury should enable identification of the key differences between regenerative and nonregenerative responses to cardiac injury. This protocol can also be adapted to the growing repertoire of genetic models available in the mouse, and it provides a valuable tool for unlocking the molecular mechanisms that guide mammalian heart regeneration during early postnatal life.
Collapse
|
226
|
Chen J, Guo R, Zhou Q, Wang T. Injection of composite with bone marrow-derived mesenchymal stem cells and a novel synthetic hydrogel after myocardial infarction: a protective role in left ventricle function. Kaohsiung J Med Sci 2014; 30:173-80. [PMID: 24656157 DOI: 10.1016/j.kjms.2013.12.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2013] [Accepted: 12/10/2013] [Indexed: 01/24/2023] Open
Abstract
A number of studies have shown that the transplantation of bone marrow-derived mesenchymal stem cells (BMSCs) into the thinned infarct wall improves regional wall motion. In this study, we hypothesized that the injection of biomaterials and MSCs into the infarcted myocardium can preserve left ventricular (LV) function. To test this hypothesis, anterior acute myocardial infarction (AMI) was induced in 34 rabbits and BMSCs with hydrogel composite were prepared. One week after inducing AMI, 28 of the 34 rabbits were divided into four groups (Groups A-D; three rabbits were used for bone marrow aspiration, and three rabbits died) and all received an epicardial injection. Group A received BMSCs with hydrogel composite marked by 5-bromodeoxyuridine (BrdU); Group B received BMSCs only marked by BrdU; Group C received hydrogel only marked by BrdU; and Group D was the control group, which received fetal bovine serum. Echocardiography was performed before AMI was induced, 1 week after AMI, and 4 weeks after the epicardial injection. The results were compared with those before AMI, and the rabbits of all the four groups had significantly larger LV end-diastolic diameter (LVDd), thinner anterior wall (AW), lower LV ejection fraction (LVEF), lower VS and VE (p<0.05) 1 week after AMI, which were more significant in Group A (p<0.05). Compared with 1 week after AMI, Group A and Group B had significantly smaller LVDd, thicker AW, larger LVEF, larger VS and VE (p<0.05) 4 weeks after the epicardial injection, which were more significant in Group A (p<0.05); however, there was no significant difference in Group C and Group D. These results suggested that BMSCs with hydrogel composite could serve as an injectable biomaterial that prevents LV remodeling and dilation, and improves local systolic and diastolic function after AMI.
Collapse
Affiliation(s)
- Jinling Chen
- Department of Echocardiography, Renmin Hospital, Wuhan University, Wuhan, Hubei Province, China.
| | - Ruiqiang Guo
- Department of Echocardiography, Renmin Hospital, Wuhan University, Wuhan, Hubei Province, China
| | - Qing Zhou
- Department of Echocardiography, Renmin Hospital, Wuhan University, Wuhan, Hubei Province, China
| | - Tao Wang
- Department of Echocardiography, Renmin Hospital, Wuhan University, Wuhan, Hubei Province, China
| |
Collapse
|
227
|
van Vliet P, Sluijter JPG, Doevendans PA, Goumans MJ. Isolation and expansion of resident cardiac progenitor cells. Expert Rev Cardiovasc Ther 2014; 5:33-43. [PMID: 17187455 DOI: 10.1586/14779072.5.1.33] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
After myocardial infarction, loss of viable cardiomyocytes severely impairs cardiac function. Recently, stem cell transplantation has been put forward as a promising approach to repair the damaged heart. Although several clinical trials have already been performed, the dominant beneficial effects are probably due to neoangiogenesis and arteriogenesis. However, replacement of cardiomyocytes is vital to improve cardiac function in the long term. Stem cells and progenitor cells, with the capacity to differentiate into cardiomyocytes, have been described in both embryonic and adult tissues. Upon stimulation, cardiac progenitor cells proliferate and differentiate into cardiomyocytes, vascular smooth muscle cells, and endothelial cells. Currently however, high proliferation rates and differentiation of cardiac progenitor cells beyond the fetal stage have not yet been achieved. Full differentiation into adult cardiomyocytes in vitro and in vivo might be important for efficient integration with the host environment and therefore more research is needed to study factors that influence proliferation and differentiation. Here we will discuss the isolation of cardiac progenitor cells, their potential to differentiate into various cell types needed for cardiac repair, the possible mechanisms behind these events, and how these cells may be implemented in future clinical settings.
Collapse
Affiliation(s)
- Patrick van Vliet
- The Interuniversity Cardiology Institute of The Netherlands, Utrecht, The Netherlands.
| | | | | | | |
Collapse
|
228
|
Abstract
Tissue engineering aims to create, repair and/or replace tissues and organs by using cells, scaffolds, biologically active molecules and physiologic signals. It is an interdisciplinary field that integrates aspects of engineering, chemistry, biology and medicine. One of the most challenging goals in the field of cardiovascular tissue engineering is the creation of a heart muscle patch. This review describes the principles, achievements and challenges of achieving this ambitious goal of creating contractile heart muscle. In addition, the new strategy of in situ and injectable tissue engineering for myocardial repair and regeneration is presented.
Collapse
Affiliation(s)
- Jonathan Leor
- Sheba-Medical Center, Neufeld Cardiac Research Institute, Tel-Aviv University, Tel-Hashomer 52621, Israel.
| | | | | |
Collapse
|
229
|
Lai CY, Wu PJ, Roffler SR, Lee ST, Hwang SM, Wang SS, Wang K, Hsieh PCH. Clearance kinetics of biomaterials affects stem cell retention and therapeutic efficacy. Biomacromolecules 2014; 15:564-73. [PMID: 24372561 DOI: 10.1021/bm401583b] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The use of biomaterial carriers to improve the therapeutic efficacy of stem cells is known to augment cell delivery, retention, and viability. However, the way that carrier clearance kinetics boosts stem cell delivery and impacts stem cell function remains poorly characterized. In this study, we designed a platform to simultaneously quantify carrier clearance and stem cell retention to evaluate the impact of carrier clearance kinetics on stem cell retention. Additionally, a murine model of hindlimb ischemia was employed to investigate the effects of various cell retention profiles on mitigating peripheral arterial disease. To image the in vivo behaviors of material and cells, we used biotinylated hyaluronan with fluorescently labeled streptavidin and Discosoma sp. Red (Ds-Red)-expressing human mesenchymal stem cells. We found that the retention of transplanted stem cells was closely related to the remaining biomaterial. Furthermore, therapeutic effectiveness was also affected by stem cell retention. These results demonstrate that low-molecular-weight hyaluronan had a slow clearance and high cell retention profile, improving the therapeutic efficacy of human stem cells.
Collapse
Affiliation(s)
- Chia Y Lai
- Institute of Biomedical Sciences, Academia Sinica , No. 128 Sec. 2, Academia Road, Taipei 115, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
230
|
Cha C, Oh J, Kim K, Qiu Y, Joh M, Shin SR, Wang X, Camci-Unal G, Wan KT, Liao R, Khademhosseini A. Microfluidics-assisted fabrication of gelatin-silica core-shell microgels for injectable tissue constructs. Biomacromolecules 2014; 15:283-90. [PMID: 24344625 PMCID: PMC3922064 DOI: 10.1021/bm401533y] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
![]()
Microfabrication
technology provides a highly versatile platform for engineering hydrogels
used in biomedical applications with high-resolution control and injectability.
Herein, we present a strategy of microfluidics-assisted fabrication
photo-cross-linkable gelatin microgels, coupled with providing protective
silica hydrogel layer on the microgel surface to ultimately generate
gelatin-silica core–shell microgels for applications as in
vitro cell culture platform and injectable tissue constructs. A microfluidic
device having flow-focusing channel geometry was utilized to generate
droplets containing methacrylated gelatin (GelMA), followed by a photo-cross-linking
step to synthesize GelMA microgels. The size of the microgels could
easily be controlled by varying the ratio of flow rates of aqueous
and oil phases. Then, the GelMA microgels were used as in vitro cell
culture platform to grow cardiac side population cells on the microgel
surface. The cells readily adhered on the microgel surface and proliferated
over time while maintaining high viability (∼90%). The cells
on the microgels were also able to migrate to their surrounding area.
In addition, the microgels eventually degraded over time. These results
demonstrate that cell-seeded GelMA microgels have a great potential
as injectable tissue constructs. Furthermore, we demonstrated that
coating the cells on GelMA microgels with biocompatible and biodegradable
silica hydrogels via sol–gel method provided significant protection
against oxidative stress which is often encountered during and after
injection into host tissues, and detrimental to the cells. Overall,
the microfluidic approach to generate cell-adhesive microgel core,
coupled with silica hydrogels as a protective shell, will be highly
useful as a cell culture platform to generate a wide range of injectable
tissue constructs.
Collapse
Affiliation(s)
- Chaenyung Cha
- Harvard-MIT Division of Health Sciences and Technology, Division of Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School , Cambridge, Massachusetts 02139, United States
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
231
|
Abstract
3D printing technology has recently gained substantial interest for potential applications in tissue engineering due to the ability of making a three-dimensional object of virtually any shape from a digital model. 3D-printed biopolymers, which combine the 3D printing technology and biopolymers, have shown great potential in tissue engineering applications and are receiving significant attention, which has resulted in the development of numerous research programs regarding the material systems which are available for 3D printing. This review focuses on recent advances in the development of biopolymer materials, including natural biopolymer-based materials and synthetic biopolymer-based materials prepared using 3D printing technology, and some future challenges and applications of this technology are discussed.
Collapse
|
232
|
Tabata Y, Horiguchi I, Lutolf MP, Sakai Y. Development of bioactive hydrogel capsules for the 3D expansion of pluripotent stem cells in bioreactors. Biomater Sci 2014; 2:176-183. [DOI: 10.1039/c3bm60183h] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
233
|
Majkut S, Idema T, Swift J, Krieger C, Liu A, Discher DE. Heart-specific stiffening in early embryos parallels matrix and myosin expression to optimize beating. Curr Biol 2013; 23:2434-9. [PMID: 24268417 PMCID: PMC4116639 DOI: 10.1016/j.cub.2013.10.057] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Revised: 09/05/2013] [Accepted: 10/22/2013] [Indexed: 01/08/2023]
Abstract
In development and differentiation, morphological changes often accompany mechanical changes [1], but it is unclear whether or when cells in embryos sense tissue elasticity. The earliest embryo is uniformly pliable, while adult tissues vary widely in mechanics from soft brain and stiff heart to rigid bone [2]. However, cell sensitivity to microenvironment elasticity is debated based in part on results from complex three-dimensional culture models [3]. Regenerative cardiology provides strong motivation to clarify any cell-level sensitivities to tissue elasticity because rigid postinfarct regions limit pumping by the adult heart [4]. Here, we focus on the spontaneously beating embryonic heart and sparsely cultured cardiomyocytes, including cells derived from pluripotent stem cells. Tissue elasticity, Et, increases daily for heart to 1-2 kPa by embryonic day 4 (E4), and although this is ~10-fold softer than adult heart, the beating contractions of E4 cardiomyocytes prove optimal at ~Et,E4 both in vivo and in vitro. Proteomics reveals daily increases in a small subset of proteins, namely collagen plus cardiac-specific excitation-contraction proteins. Rapid softening of the heart's matrix with collagenase or stiffening it with enzymatic crosslinking suppresses beating. Sparsely cultured E4 cardiomyocytes on collagen-coated gels likewise show maximal contraction on matrices with native E4 stiffness, highlighting cell-intrinsic mechanosensitivity. While an optimal elasticity for striation proves consistent with the mathematics of force-driven sarcomere registration, contraction wave speed is linear in Et as theorized for excitation-contraction coupled to matrix elasticity. Pluripotent stem cell-derived cardiomyocytes also prove to be mechanosensitive to matrix and thus generalize the main observation that myosin II organization and contractile function are optimally matched to the load contributed by matrix elasticity.
Collapse
Affiliation(s)
- Stephanie Majkut
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Physics and Astronomy, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Timon Idema
- Department of Physics and Astronomy, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Bionanoscience, Kavli Institute of Nanoscience, Delft University of Technology, Lorentzweg 1, 2628 CJ Delft, The Netherlands
| | - Joe Swift
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christine Krieger
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Andrea Liu
- Department of Physics and Astronomy, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Dennis E. Discher
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Physics and Astronomy, University of Pennsylvania, Philadelphia, PA 19104, USA
- Cell and Molecular Biology Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
234
|
Kimura W, Sadek HA. The cardiac hypoxic niche: emerging role of hypoxic microenvironment in cardiac progenitors. Cardiovasc Diagn Ther 2013; 2:278-89. [PMID: 24282728 DOI: 10.3978/j.issn.2223-3652.2012.12.02] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2012] [Accepted: 12/10/2012] [Indexed: 12/11/2022]
Abstract
Resident stem cells persist throughout the entire lifetime of an organism where they replenishing damaged cells. Numerous types of resident stem cells are housed in a low-oxygen tension (hypoxic) microenvironment, or niches, which seem to be critical for survival and maintenance of stem cells. Recently our group has identified the adult mammalian epicardium and subepicardium as a hypoxic niche for cardiac progenitor cells. Similar to hematopoietic stem cells (LT-HSCs), progenitor cells in the hypoxic epicardial niche utilize cytoplasmic glycolysis instead of mitochondrial oxidative phosphorylation, where hypoxia inducible factor 1α (Hif-1α) maintains them in glycolytic undifferentiated state. In this review we summarize the relationship between hypoxic signaling and stem cell function, and discuss potential roles of several cardiac stem/progenitor cells in cardiac homeostasis and regeneration.
Collapse
Affiliation(s)
- Wataru Kimura
- Department of Internal Medicine, Division of Cardiology, UT Southwestern Medical Center, Dallas, TX, USA
| | | |
Collapse
|
235
|
A fully defined and scalable 3D culture system for human pluripotent stem cell expansion and differentiation. Proc Natl Acad Sci U S A 2013; 110:E5039-48. [PMID: 24248365 DOI: 10.1073/pnas.1309408110] [Citation(s) in RCA: 225] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Human pluripotent stem cells (hPSCs), including human embryonic stem cells and induced pluripotent stem cells, are promising for numerous biomedical applications, such as cell replacement therapies, tissue and whole-organ engineering, and high-throughput pharmacology and toxicology screening. Each of these applications requires large numbers of cells of high quality; however, the scalable expansion and differentiation of hPSCs, especially for clinical utilization, remains a challenge. We report a simple, defined, efficient, scalable, and good manufacturing practice-compatible 3D culture system for hPSC expansion and differentiation. It employs a thermoresponsive hydrogel that combines easy manipulation and completely defined conditions, free of any human- or animal-derived factors, and entailing only recombinant protein factors. Under an optimized protocol, the 3D system enables long-term, serial expansion of multiple hPSCs lines with a high expansion rate (~20-fold per 5-d passage, for a 10(72)-fold expansion over 280 d), yield (~2.0 × 10(7) cells per mL of hydrogel), and purity (~95% Oct4+), even with single-cell inoculation, all of which offer considerable advantages relative to current approaches. Moreover, the system enabled 3D directed differentiation of hPSCs into multiple lineages, including dopaminergic neuron progenitors with a yield of ~8 × 10(7) dopaminergic progenitors per mL of hydrogel and ~80-fold expansion by the end of a 15-d derivation. This versatile system may be useful at numerous scales, from basic biological investigation to clinical development.
Collapse
|
236
|
Developing stem cell therapeutics for the heart also requires targeting non-myocytes. Heart Lung Circ 2013; 22:975-9. [PMID: 24231432 DOI: 10.1016/j.hlc.2013.10.091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
237
|
He WJ, Hou Q, Han QW, Han WD, Fu XB. Pluripotent reprogramming and lineage reprogramming: promises and challenges in cardiovascular regeneration. TISSUE ENGINEERING PART B-REVIEWS 2013; 20:304-13. [PMID: 24063625 DOI: 10.1089/ten.teb.2013.0393] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cardiovascular disease is a leading cause of death in industrialized countries. Scientists are trying to generate cardiomyocytes in vitro and in vivo to repair damaged heart tissue. Pluripotent reprogramming brings an alternative source of embryonic-like stem cells, and the possibility of regenerating mammalian tissues by first reverting somatic cells to induced pluripotent stem cells, followed by redifferentiating these cells into cardiomyocytes. More recently, lineage reprogramming of fibroblasts directly into functional cardiomyocytes has been reported. The procedure does not involve reverting cells back to a pluripotent stage, and, thus, would presumably reduce tumorigenic potential. Interestingly, lineage reprogramming could be used for in situ conversion of cell fate. Moreover, zebrafish-like regenerative mechanism in mammalian heart tissue, which was observed in mice within the first week of postpartum, should be further addressed. Here, we review the landmark progresses of the two major reprogramming strategies, compare their pros and cons in cardiovascular regeneration, and forecast the future directions of cardiac repair.
Collapse
Affiliation(s)
- Wen-Jun He
- 1 Tissue Repair and Regeneration Laboratory, College of Life Science, Chinese PLA General Hospital , Beijing, China
| | | | | | | | | |
Collapse
|
238
|
Abstract
Dilated cardiomyopathy is a serious and life-threatening disorder in children. It is the most common form of pediatric cardiomyopathy. Therapy for this condition has varied little over the last several decades and mortality continues to be high. Currently, children with dilated cardiomyopathy are treated with pharmacological agents and mechanical support, but most require heart transplantation and survival rates are not optimal. The lack of common treatment guidelines and inadequate survival rates after transplantation necessitates more therapeutic clinical trials. Stem cell and cell-based therapies offer an innovative approach to restore cardiac structure and function towards normal, possibly reducing the need for aggressive therapies and cardiac transplantation. Mesenchymal stem cells and cardiac stem cells may be the most promising cell types for treating children with dilated cardiomyopathy. The medical community must begin a systematic investigation of the benefits of current and novel treatments such as stem cell therapies for treating pediatric dilated cardiomyopathy.
Collapse
Affiliation(s)
- Sarah M Selem
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Biomedical Research Building/Room 908, PO Box 016960 (R-125), Miami, FL 33101, USA
| | | | | |
Collapse
|
239
|
The Biochemical and histopathological effects of estrogen replacement therapy on the heart of ovariectomized female rats subjected to myocardial infarction. ACTA ACUST UNITED AC 2013. [DOI: 10.1097/01.ebx.0000435382.84206.91] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
240
|
Chatterjee M, Gawaz M. Platelet-derived CXCL12 (SDF-1α): basic mechanisms and clinical implications. J Thromb Haemost 2013; 11:1954-67. [PMID: 24024928 DOI: 10.1111/jth.12404] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Indexed: 12/19/2022]
Abstract
Platelets are a major source of CXCL12 (stromal cell-derived factor -1α, SDF-1α) and store CXCL12 as part of their α-granule secretome. Platelet activation enhances surface expression and release of CXCL12. Platelets and megakaryocytes express CXCR4, the major receptor for CXCL12, and interaction of CXCL12 with CXCR4 regulates megakaryopoiesis and the function of circulating platelets. Platelet-derived CXCL12 also modulates paracrine mechanisms such as chemotaxis, adhesion, proliferation and differentiation of nucleated cells, including progenitor cells. Platelet-derived CXCL12 enhances peripheral recruitment of progenitor cells to the sites of vascular and tissue injury both in vitro and in vivo and thereby promotes repair mechanisms. CXCL12 expression on platelets is elevated in patients with acute myocardial infarction, correlates with the number of circulating progenitor cells, is associated with preservation of myocardial function and is an independent predictor of clinical outcome. Administration of recombinant CXCL12 reduces infarct size following transient ischemia in mice. The present review summarizes the role of platelet-derived CXCL12 in cardiovascular biology and its diagnostic and therapeutic implications.
Collapse
Affiliation(s)
- M Chatterjee
- Medizinische Klinik III, Kardiologie und Kreislauferkrankungen, Eberhard Karls Universität, Tübingen, Germany
| | | |
Collapse
|
241
|
Ma S, Liang S, Jiao H, Chi L, Shi X, Tian Y, Yang B, Guan F. Human umbilical cord mesenchymal stem cells inhibit C6 glioma growth via secretion of dickkopf-1 (DKK1). Mol Cell Biochem 2013; 385:277-86. [PMID: 24104453 DOI: 10.1007/s11010-013-1836-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 09/26/2013] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) represent a potential therapeutic target for glioma. We determined the molecular mechanism of inhibitory effect of human umbilical cord-derived MSCs (hUC-MSCs) on the growth of C6 glioma cells. We demonstrated that hUC-MSCs inhibited C6 cell growth and modulated the cell cycle to G0/G1 phase. The expression of β-catenin and c-Myc was downregulated in C6 cells by conditioned media from hUC-MSCs, and the levels of secreted DKK1 were positively correlated with concentrations of hUCMSCs-CM. The inhibitory effect of hUC-MSCs on C6 cell proliferation was enhanced as the concentration of DKK1 in hUCMSCs-CM increased. When DKK1 was neutralized by anti-DKK1 antibody, the inhibitory effect of hUC-MSCs on C6 cells was attenuated. Furthermore, we found that conditioned media from hUC-MSCs transfection with siRNA targeting DKK1 mRNA or pEGFPN1-DKK1 plasmid lost or enhanced the abilities to regulate the Wnt signaling in C6 cells. Therefore, hUC-MSCs inhibited C6 glioma cell growth via secreting DKK1, an inhibitor of Wnt pathway, may represent a novel therapeutic strategy for malignant glioma.
Collapse
Affiliation(s)
- Shanshan Ma
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | | | | | | | | | | | | | | |
Collapse
|
242
|
Kim YY, Ku SY, Huh Y, Liu HC, Kim SH, Choi YM, Moon SY. Anti-aging effects of vitamin C on human pluripotent stem cell-derived cardiomyocytes. AGE (DORDRECHT, NETHERLANDS) 2013; 35:1545-1557. [PMID: 22843416 PMCID: PMC3776090 DOI: 10.1007/s11357-012-9457-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Accepted: 07/06/2012] [Indexed: 06/01/2023]
Abstract
Human pluripotent stem cells (hPSCs) have arisen as a source of cells for biomedical research due to their developmental potential. Stem cells possess the promise of providing clinicians with novel treatments for disease as well as allowing researchers to generate human-specific cellular metabolism models. Aging is a natural process of living organisms, yet aging in human heart cells is difficult to study due to the ethical considerations regarding human experimentation as well as a current lack of alternative experimental models. hPSC-derived cardiomyocytes (CMs) bear a resemblance to human cardiac cells and thus hPSC-derived CMs are considered to be a viable alternative model to study human heart cell aging. In this study, we used hPSC-derived CMs as an in vitro aging model. We generated cardiomyocytes from hPSCs and demonstrated the process of aging in both human embryonic stem cell (hESC)- and induced pluripotent stem cell (hiPSC)-derived CMs. Aging in hESC-derived CMs correlated with reduced membrane potential in mitochondria, the accumulation of lipofuscin, a slower beating pattern, and the downregulation of human telomerase RNA (hTR) and cell cycle regulating genes. Interestingly, the expression of hTR in hiPSC-derived CMs was not significantly downregulated, unlike in hESC-derived CMs. In order to delay aging, vitamin C was added to the cultured CMs. When cells were treated with 100 μM of vitamin C for 48 h, anti-aging effects, specifically on the expression of telomere-related genes and their functionality in aging cells, were observed. Taken together, these results suggest that hPSC-derived CMs can be used as a unique human cardiomyocyte aging model in vitro and that vitamin C shows anti-aging effects in this model.
Collapse
Affiliation(s)
- Yoon Young Kim
- />Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, Seoul, South Korea
| | - Seung-Yup Ku
- />Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, Seoul, South Korea
- />Department of Obstetrics and Gynecology, College of Medicine, Seoul National University, Seoul, 110-744 South Korea
| | - Yul Huh
- />Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, Seoul, South Korea
| | - Hung-Ching Liu
- />Center for Reproductive Medicine and Infertility, Weill Cornell Medical College, New York, NY 10021 USA
| | - Seok Hyun Kim
- />Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, Seoul, South Korea
- />Department of Obstetrics and Gynecology, College of Medicine, Seoul National University, Seoul, 110-744 South Korea
| | - Young Min Choi
- />Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, Seoul, South Korea
- />Department of Obstetrics and Gynecology, College of Medicine, Seoul National University, Seoul, 110-744 South Korea
| | - Shin Yong Moon
- />Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, Seoul, South Korea
- />Department of Obstetrics and Gynecology, College of Medicine, Seoul National University, Seoul, 110-744 South Korea
| |
Collapse
|
243
|
|
244
|
Krenz M, Baines C, Kalogeris T, Korthuis R. Cell Survival Programs and Ischemia/Reperfusion: Hormesis, Preconditioning, and Cardioprotection. ACTA ACUST UNITED AC 2013. [DOI: 10.4199/c00090ed1v01y201309isp044] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
245
|
Boffito M, Sartori S, Ciardelli G. Polymeric scaffolds for cardiac tissue engineering: requirements and fabrication technologies. POLYM INT 2013. [DOI: 10.1002/pi.4608] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Monica Boffito
- Department of Mechanical and Aerospace Engineering; Politecnico di Torino; Corso Duca degli Abruzzi 24 10129 Turin Italy
| | - Susanna Sartori
- Department of Mechanical and Aerospace Engineering; Politecnico di Torino; Corso Duca degli Abruzzi 24 10129 Turin Italy
| | - Gianluca Ciardelli
- Department of Mechanical and Aerospace Engineering; Politecnico di Torino; Corso Duca degli Abruzzi 24 10129 Turin Italy
- CNR-IPCF UOS Pisa; Via Moruzzi 1 56124 Pisa Italy
| |
Collapse
|
246
|
Sreejit P, Verma RS. Natural ECM as biomaterial for scaffold based cardiac regeneration using adult bone marrow derived stem cells. Stem Cell Rev Rep 2013; 9:158-71. [PMID: 23319217 DOI: 10.1007/s12015-013-9427-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Cellular therapy using stem cells for cardiac diseases has recently gained much interest in the scientific community due to its potential in regenerating damaged and even dead tissue and thereby restoring the organ function. Stem cells from various sources and origin are being currently used for regeneration studies directly or along with differentiation inducing agents. Long term survival and minimal side effects can be attained by using autologous cells and reduced use of inducing agents. Cardiomyogenic differentiation of adult derived stem cells has been previously reported using various inducing agents but the use of a potentially harmful DNA demethylating agent 5-azacytidine (5-azaC) has been found to be critical in almost all studies. Alternate inducing factors and conditions/stimulant like physical condition including electrical stimulation, chemical inducers and biological agents have been attempted by numerous groups to induce cardiac differentiation. Biomaterials were initially used as artificial scaffold in in vitro studies and later as a delivery vehicle. Natural ECM is the ideal biological scaffold since it contains all the components of the tissue from which it was derived except for the living cells. Constructive remodeling can be performed using such natural ECM scaffolds and stem cells since, the cells can be delivered to the site of infraction and once delivered the cells adhere and are not "lost". Due to the niche like conditions of ECM, stem cells tend to differentiate into tissue specific cells and attain several characteristics similar to that of functional cells even in absence of any directed differentiation using external inducers. The development of niche mimicking biomaterials and hybrid biomaterial can further advance directed differentiation without specific induction. The mechanical and electrical integration of these materials to the functional tissue is a problem to be addressed. The search for the perfect extracellular matrix for therapeutic applications including engineering cardiac tissue structures for post ischemic cardiac tissue regeneration continues.
Collapse
Affiliation(s)
- P Sreejit
- Stem Cell and Molecular Biology Laboratory, Department of Biotechnology, Indian Institute of Technology Madras, Chennai, 600036, TN, India
| | | |
Collapse
|
247
|
Cardiac progenitor cells engineered with βARKct have enhanced β-adrenergic tolerance. Mol Ther 2013; 22:178-85. [PMID: 24002692 DOI: 10.1038/mt.2013.200] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Accepted: 08/21/2013] [Indexed: 01/08/2023] Open
Abstract
Stem cell survival and retention in myocardium after injury following adoptive transfer is low. Elevated catecholamine levels coinciding with myocardial injury adversely affect cardiac progenitor cell (CPC) survival. The G protein-coupled receptor kinase 2 (GRK2)-derived inhibitory peptide, βARKct, enhance myocyte contractility, survival, and normalize the neurohormonal axis in failing heart, however salutary effects of βARKct on CPC survival and proliferation are unknown. Herein, we investigated whether the protective effects of βARKct expression seen in the failing heart relate to CPCs. Modified CPCs expressing βARKct enhanced AKT/eNOS signaling through protective β2-adrenergic receptors (β2-ARs). In addition, to the actions of βARKct expression on β2- AR signaling, pharmacologic inhibition of GRK2 also increased β2-AR signaling in nonengineered CPCs (lacking βARKct) but had limited effects in βARKct engineered CPCs providing evidence for the strength of the βARKct in inhibiting GRK2 in these cells. Increased proliferation and metabolic activity were observed in βARKct-engineered CPCs following catecholamine stimulation indicating improved adrenergic tolerance. βARKct modification of CPCs increased survival and proliferation following adoptive transfer in an acute myocardial infarction model concomitant with increased expression of β-AR. Thus, βARKct engineering of CPCs promotes survival and proliferation of injected cells following myocardial infarction, which includes improved β-adrenergic tolerance essential for stem cell survival.
Collapse
|
248
|
Lakshmanan R, Krishnan UM, Sethuraman S. Polymeric scaffold aided stem cell therapeutics for cardiac muscle repair and regeneration. Macromol Biosci 2013; 13:1119-34. [PMID: 23982911 DOI: 10.1002/mabi.201300223] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 07/05/2013] [Indexed: 12/13/2022]
Abstract
The constantly expanding repository of novel polymers and stem cells has opened up new vistas in the field of cardiac tissue engineering. Successful regeneration of the complex cardiac tissue mainly centres on the appropriate scaffold material with topographical features that mimic the native environment. The integration of stem cells on these scaffolds is expected to enhance the regeneration potential. This review elaborates on the interplay of these vital factors in achieving the functional cardiac tissue. The recent advances in polymers, nanocomposites, and stem cells from different sources are highlighted. Special emphasis is laid on the clinical trials involving stem cells and the state-of-the-art materials to obtain a balanced perspective on the translational potential of this strategy.
Collapse
Affiliation(s)
- Rajesh Lakshmanan
- Centre for Nanotechnology & Advanced Biomaterials, School of Chemical and Biotechnology, SASTRA University, Thanjavur, 613 401, Tamil Nadu, India
| | | | | |
Collapse
|
249
|
Duran JM, Makarewich CA, Sharp TE, Starosta T, Fang Z, Hoffman NE, Chiba Y, Madesh M, Berretta RM, Kubo H, Houser SR. Bone-derived stem cells repair the heart after myocardial infarction through transdifferentiation and paracrine signaling mechanisms. Circ Res 2013; 113:539-52. [PMID: 23801066 PMCID: PMC3822430 DOI: 10.1161/circresaha.113.301202] [Citation(s) in RCA: 139] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2013] [Accepted: 06/25/2013] [Indexed: 12/21/2022]
Abstract
RATIONALE Autologous bone marrow-derived or cardiac-derived stem cell therapy for heart disease has demonstrated safety and efficacy in clinical trials, but functional improvements have been limited. Finding the optimal stem cell type best suited for cardiac regeneration is the key toward improving clinical outcomes. OBJECTIVE To determine the mechanism by which novel bone-derived stem cells support the injured heart. METHODS AND RESULTS Cortical bone-derived stem cells (CBSCs) and cardiac-derived stem cells were isolated from enhanced green fluorescent protein (EGFP+) transgenic mice and were shown to express c-kit and Sca-1 as well as 8 paracrine factors involved in cardioprotection, angiogenesis, and stem cell function. Wild-type C57BL/6 mice underwent sham operation (n=21) or myocardial infarction with injection of CBSCs (n=67), cardiac-derived stem cells (n=36), or saline (n=60). Cardiac function was monitored using echocardiography. Only 2/8 paracrine factors were detected in EGFP+ CBSCs in vivo (basic fibroblast growth factor and vascular endothelial growth factor), and this expression was associated with increased neovascularization of the infarct border zone. CBSC therapy improved survival, cardiac function, regional strain, attenuated remodeling, and decreased infarct size relative to cardiac-derived stem cells- or saline-treated myocardial infarction controls. By 6 weeks, EGFP+ cardiomyocytes, vascular smooth muscle, and endothelial cells could be identified in CBSC-treated, but not in cardiac-derived stem cells-treated, animals. EGFP+ CBSC-derived isolated myocytes were smaller and more frequently mononucleated, but were functionally indistinguishable from EGFP- myocytes. CONCLUSIONS CBSCs improve survival, cardiac function, and attenuate remodeling through the following 2 mechanisms: (1) secretion of proangiogenic factors that stimulate endogenous neovascularization, and (2) differentiation into functional adult myocytes and vascular cells.
Collapse
Affiliation(s)
- Jason M. Duran
- Temple University School of Medicine Cardiovascular Research Center, Philadelphia, PA
| | | | - Thomas E. Sharp
- Temple University School of Medicine Cardiovascular Research Center, Philadelphia, PA
| | - Timothy Starosta
- Temple University School of Medicine Cardiovascular Research Center, Philadelphia, PA
| | - Zhu Fang
- Fox Chase Cancer Center Biostatistics and Bioinformatics Facility, Philadelphia PA
| | - Nicholas E. Hoffman
- Temple University School of Medicine Center for Translational Medicine, Philadelphia, PA
| | - Yumi Chiba
- Temple University School of Medicine Cardiovascular Research Center, Philadelphia, PA
| | - Muniswamy Madesh
- Temple University School of Medicine Center for Translational Medicine, Philadelphia, PA
| | - Remus M. Berretta
- Temple University School of Medicine Cardiovascular Research Center, Philadelphia, PA
| | - Hajime Kubo
- Temple University School of Medicine Cardiovascular Research Center, Philadelphia, PA
| | - Steven R. Houser
- Temple University School of Medicine Cardiovascular Research Center, Philadelphia, PA
| |
Collapse
|
250
|
Anversa P, Leri A. Innate regeneration in the aging heart: healing from within. Mayo Clin Proc 2013; 88:871-83. [PMID: 23910414 PMCID: PMC3936323 DOI: 10.1016/j.mayocp.2013.04.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Revised: 03/29/2013] [Accepted: 04/01/2013] [Indexed: 12/31/2022]
Abstract
The concept of the heart as a terminally differentiated organ incapable of replacing damaged myocytes has been at the center of cardiovascular research and therapeutic development for the past 50 years. The progressive decline in myocyte number as a function of age and the formation of scarred tissue after myocardial infarction have been interpreted as irrefutable proofs of the postmitotic characteristic of the heart. However, emerging evidence supports a more dynamic view of the heart in which cell death and renewal are vital components of the remodeling process that governs cardiac homeostasis, aging, and disease. The identification of dividing myocytes in the adult and senescent heart raises the important question concerning the origin of these newly formed cells. In vitro and in vivo findings strongly suggest that replicating myocytes derive from lineage determination of resident primitive cells, supporting the notion that cardiomyogenesis is controlled by activation and differentiation of a stem cell compartment. It is the current view that the myocardium is an organ permissive of tissue regeneration mediated by exogenous and endogenous progenitor cells.
Collapse
Affiliation(s)
- Piero Anversa
- Department of Anesthesia, Department of Medicine, and Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.
| | | |
Collapse
|