1
|
Raabe J, Wittig I, Laurette P, Stathopoulou K, Brand T, Schulze T, Klampe B, Orthey E, Cabrera-Orefice A, Meisterknecht J, Thiemann E, Laufer SD, Shibamiya A, Reinsch M, Fuchs S, Kaiser J, Yang J, Zehr S, Wrona KM, Lorenz K, Lukowski R, Hansen A, Gilsbach R, Brandes RP, Ulmer BM, Eschenhagen T, Cuello F. Physioxia rewires mitochondrial complex composition to protect stem cell viability. Redox Biol 2024; 77:103352. [PMID: 39341035 PMCID: PMC11466565 DOI: 10.1016/j.redox.2024.103352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) are an invaluable tool to study molecular mechanisms on a human background. Culturing stem cells at an oxygen level different from their microenvironmental niche impacts their viability. To understand this mechanistically, dermal skin fibroblasts of 52 probands were reprogrammed into hiPSCs, followed by either hyperoxic (20 % O2) or physioxic (5 % O2) culture and proteomic profiling. Analysis of chromosomal stability by Giemsa-banding revealed that physioxic -cultured hiPSC clones exhibited less pathological karyotypes than hyperoxic (e.g. 6 % vs. 32 % mosaicism), higher pluripotency as evidenced by higher Stage-Specific Embryonic Antigen 3 positivity, higher glucose consumption and lactate production. Global proteomic analysis demonstrated lower abundance of several subunits of NADH:ubiquinone oxidoreductase (complex I) and an underrepresentation of pathways linked to oxidative phosphorylation and cellular senescence. Accordingly, release of the pro-senescent factor IGFBP3 and β-galactosidase staining were lower in physioxic hiPSCs. RNA- and ATAC-seq profiling revealed a distinct hypoxic transcription factor-binding footprint, amongst others higher expression of the HIF1α-regulated target NDUFA4L2 along with increased chromatin accessibility of the NDUFA4L2 gene locus. While mitochondrial DNA content did not differ between groups, physioxic hiPSCs revealed lower polarized mitochondrial membrane potential, altered mitochondrial network appearance and reduced basal respiration and electron transfer capacity. Blue-native polyacrylamide gel electrophoresis coupled to mass spectrometry of the mitochondrial complexes detected higher abundance of NDUFA4L2 and ATP5IF1 and loss of incorporation into complex IV or V, respectively. Taken together, physioxic culture of hiPSCs improved chromosomal stability, which was associated with downregulation of oxidative phosphorylation and senescence and extensive re-wiring of mitochondrial complex composition.
Collapse
Affiliation(s)
- Janice Raabe
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Ilka Wittig
- Functional Proteomics Center, Institute for Cardiovascular Physiology, Goethe-University, 60590 Frankfurt am Main, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Rhein-Main, Frankfurt, Germany
| | - Patrick Laurette
- Institute of Experimental Cardiology, Heidelberg University Hospital, 69120 Heidelberg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Germany
| | - Konstantina Stathopoulou
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Theresa Brand
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany
| | - Thomas Schulze
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Birgit Klampe
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Ellen Orthey
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Alfredo Cabrera-Orefice
- Functional Proteomics Center, Institute for Cardiovascular Physiology, Goethe-University, 60590 Frankfurt am Main, Germany
| | - Jana Meisterknecht
- Functional Proteomics Center, Institute for Cardiovascular Physiology, Goethe-University, 60590 Frankfurt am Main, Germany
| | - Ellen Thiemann
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Sandra D Laufer
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Aya Shibamiya
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Marina Reinsch
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Sigrid Fuchs
- Institute for Human Genetics, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Jennifer Kaiser
- Institute for Human Genetics, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Jiaqi Yang
- Institute of Pharmacy, Experimental Pharmacology, University Tübingen, 72076 Tübingen, Germany
| | - Simonida Zehr
- DZHK (German Center for Cardiovascular Research), Partner Site Rhein-Main, Frankfurt, Germany; Institute for Cardiovascular Physiology, Goethe-University, 60590 Frankfurt am Main, Germany
| | - Kinga M Wrona
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Kristina Lorenz
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany; Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund, Germany
| | - Robert Lukowski
- Institute of Pharmacy, Experimental Pharmacology, University Tübingen, 72076 Tübingen, Germany
| | - Arne Hansen
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Ralf Gilsbach
- Institute of Experimental Cardiology, Heidelberg University Hospital, 69120 Heidelberg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Germany
| | - Ralf P Brandes
- DZHK (German Center for Cardiovascular Research), Partner Site Rhein-Main, Frankfurt, Germany; Institute for Cardiovascular Physiology, Goethe-University, 60590 Frankfurt am Main, Germany
| | - Bärbel M Ulmer
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Thomas Eschenhagen
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany.
| | - Friederike Cuello
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany.
| |
Collapse
|
2
|
Goecke T, Ius F, Ruhparwar A, Martin U. Unlocking the Future: Pluripotent Stem Cell-Based Lung Repair. Cells 2024; 13:635. [PMID: 38607074 PMCID: PMC11012168 DOI: 10.3390/cells13070635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 03/18/2024] [Accepted: 03/26/2024] [Indexed: 04/13/2024] Open
Abstract
The human respiratory system is susceptible to a variety of diseases, ranging from chronic obstructive pulmonary disease (COPD) and pulmonary fibrosis to acute respiratory distress syndrome (ARDS). Today, lung diseases represent one of the major challenges to the health care sector and represent one of the leading causes of death worldwide. Current treatment options often focus on managing symptoms rather than addressing the underlying cause of the disease. The limitations of conventional therapies highlight the urgent clinical need for innovative solutions capable of repairing damaged lung tissue at a fundamental level. Pluripotent stem cell technologies have now reached clinical maturity and hold immense potential to revolutionize the landscape of lung repair and regenerative medicine. Meanwhile, human embryonic (HESCs) and human-induced pluripotent stem cells (hiPSCs) can be coaxed to differentiate into lung-specific cell types such as bronchial and alveolar epithelial cells, or pulmonary endothelial cells. This holds the promise of regenerating damaged lung tissue and restoring normal respiratory function. While methods for targeted genetic engineering of hPSCs and lung cell differentiation have substantially advanced, the required GMP-grade clinical-scale production technologies as well as the development of suitable preclinical animal models and cell application strategies are less advanced. This review provides an overview of current perspectives on PSC-based therapies for lung repair, explores key advances, and envisions future directions in this dynamic field.
Collapse
Affiliation(s)
- Tobias Goecke
- Leibniz Research Laboratories for Biotechnology and Artificial Organs, Lower Saxony Center for Biomedical Engineering, Implant Research and Development /Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany; (F.I.); (A.R.)
- REBIRTH-Research Center for Translational and Regenerative Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
- Biomedical Research in End-stage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Fabio Ius
- Leibniz Research Laboratories for Biotechnology and Artificial Organs, Lower Saxony Center for Biomedical Engineering, Implant Research and Development /Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany; (F.I.); (A.R.)
- REBIRTH-Research Center for Translational and Regenerative Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
- Biomedical Research in End-stage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Arjang Ruhparwar
- Leibniz Research Laboratories for Biotechnology and Artificial Organs, Lower Saxony Center for Biomedical Engineering, Implant Research and Development /Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany; (F.I.); (A.R.)
- REBIRTH-Research Center for Translational and Regenerative Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
- Biomedical Research in End-stage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Ulrich Martin
- Leibniz Research Laboratories for Biotechnology and Artificial Organs, Lower Saxony Center for Biomedical Engineering, Implant Research and Development /Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany; (F.I.); (A.R.)
- REBIRTH-Research Center for Translational and Regenerative Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
- Biomedical Research in End-stage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| |
Collapse
|
3
|
Hsu CN, Lin YT, Chen YH, Tseng TY, Tsai HF, Hong SG, Yao CL. An Aligned Poly(3-hydroxybutyrate-co-3-hydroxyvalerate) Scaffold Fixed with Fibronectin to Enhance the Attachment and Growth of Human Endothelial Progenitor Cells. BIOTECHNOL BIOPROC E 2023. [DOI: 10.1007/s12257-022-0255-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
4
|
Human Induced Pluripotent Stem Cell-Derived Vascular Cells: Recent Progress and Future Directions. J Cardiovasc Dev Dis 2021; 8:jcdd8110148. [PMID: 34821701 PMCID: PMC8622843 DOI: 10.3390/jcdd8110148] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/01/2021] [Accepted: 11/02/2021] [Indexed: 12/12/2022] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) hold great promise for cardiovascular regeneration following ischemic injury. Considerable effort has been made toward the development and optimization of methods to differentiate hiPSCs into vascular cells, such as endothelial and smooth muscle cells (ECs and SMCs). In particular, hiPSC-derived ECs have shown robust potential for promoting neovascularization in animal models of cardiovascular diseases, potentially achieving significant and sustained therapeutic benefits. However, the use of hiPSC-derived SMCs that possess high therapeutic relevance is a relatively new area of investigation, still in the earlier investigational stages. In this review, we first discuss different methodologies to derive vascular cells from hiPSCs with a particular emphasis on the role of key developmental signals. Furthermore, we propose a standardized framework for assessing and defining the EC and SMC identity that might be suitable for inducing tissue repair and regeneration. We then highlight the regenerative effects of hiPSC-derived vascular cells on animal models of myocardial infarction and hindlimb ischemia. Finally, we address several obstacles that need to be overcome to fully implement the use of hiPSC-derived vascular cells for clinical application.
Collapse
|
5
|
Huang Y, Qian JY, Cheng H, Li XM. Effects of shear stress on differentiation of stem cells into endothelial cells. World J Stem Cells 2021; 13:894-913. [PMID: 34367483 PMCID: PMC8316872 DOI: 10.4252/wjsc.v13.i7.894] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/20/2021] [Accepted: 06/22/2021] [Indexed: 02/06/2023] Open
Abstract
Stem cell transplantation is an appealing potential therapy for vascular diseases and an indispensable key step in vascular tissue engineering. Substantial effort has been made to differentiate stem cells toward vascular cell phenotypes, including endothelial cells (ECs) and smooth muscle cells. The microenvironment of vascular cells not only contains biochemical factors that influence differentiation but also exerts hemodynamic forces, such as shear stress and cyclic strain. More recently, studies have shown that shear stress can influence the differentiation of stem cells toward ECs. A deep understanding of the responses and underlying mechanisms involved in this process is essential for clinical translation. This review highlights current data supporting the role of shear stress in stem cell differentiation into ECs. Potential mechanisms and signaling cascades for transducing shear stress into a biological signal are proposed. Further study of stem cell responses to shear stress will be necessary to apply stem cells for pharmacological applications and cardiovascular implants in the realm of regenerative medicine.
Collapse
Affiliation(s)
- Yan Huang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Jia-Yi Qian
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Hong Cheng
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Xiao-Ming Li
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| |
Collapse
|
6
|
Vasyutin I, Butnaru D, Lyundup A, Timashev P, Vinarov A, Kuznetsov S, Atala A, Zhang Y. Frontiers in urethra regeneration: current state and future perspective. Biomed Mater 2021; 16. [PMID: 32503009 DOI: 10.1088/1748-605x/ab99d2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 06/05/2020] [Indexed: 12/13/2022]
Abstract
Despite the positive achievements attained, the treatment of male urethral strictures and hypospadiases still remains a challenge, particularly in cases of severe urethral defects. Complications and the need for additional interventions in such cases are common. Also, shortage of autologous tissue for graft harvesting and significant morbidity in the location of harvesting present problems and often lead to staged treatment. Tissue engineering provides a promising alternative to the current sources of grafts for urethroplasty. Since the first experiments in urethral substitution with tissue engineered grafts, this topic in regenerative medicine has grown remarkably, as many different types of tissue-engineered grafts and approaches in graft design have been suggested and testedin vivo. However, there have been only a few clinical trials of tissue-engineered grafts in urethral substitution, involving hardly more than a hundred patients overall. This indicates that the topic is still in its inception, and the search for the best graft design is continuing. The current review focuses on the state of the art in urethral regeneration with tissue engineering technology. It gives a comprehensive overview of the components of the tissue-engineered graft and an overview of the steps in graft development. Different cell sources, types of scaffolds, assembling approaches, options for vascularization enhancement and preclinical models are considered.
Collapse
Affiliation(s)
- Igor Vasyutin
- Sechenov University, 8-2 Trubetskaya str., Moscow 119991, Russia
| | - Denis Butnaru
- Sechenov University, 8-2 Trubetskaya str., Moscow 119991, Russia
| | - Alexey Lyundup
- Sechenov University, 8-2 Trubetskaya str., Moscow 119991, Russia
| | - Peter Timashev
- Sechenov University, 8-2 Trubetskaya str., Moscow 119991, Russia
| | - Andrey Vinarov
- Sechenov University, 8-2 Trubetskaya str., Moscow 119991, Russia
| | - Sergey Kuznetsov
- Sechenov University, 8-2 Trubetskaya str., Moscow 119991, Russia
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, 391 Technology Way NE, Winston-Salem, NC 27101, United States of America
| | - Yuanyuan Zhang
- Sechenov University, 8-2 Trubetskaya str., Moscow 119991, Russia.,Wake Forest Institute for Regenerative Medicine, 391 Technology Way NE, Winston-Salem, NC 27101, United States of America
| |
Collapse
|
7
|
Park JJ, Kwon YW, Kim JW, Park GT, Yoon JW, Kim YS, Kim DS, Kwon SM, Bae SS, Ko K, Kim CS, Kim JH. Coadministration of endothelial and smooth muscle cells derived from human induced pluripotent stem cells as a therapy for critical limb ischemia. Stem Cells Transl Med 2020; 10:414-426. [PMID: 33174379 PMCID: PMC7900584 DOI: 10.1002/sctm.20-0132] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 09/24/2020] [Accepted: 10/09/2020] [Indexed: 12/19/2022] Open
Abstract
Critical limb ischemia is a condition in which tissue necrosis occurs due to arterial occlusion, resulting in limb amputation in severe cases. Both endothelial cells (ECs) and vascular smooth muscle cells (SMCs) are needed for the regeneration of peripheral arteries in ischemic tissues. However, it is difficult to isolate and cultivate primary EC and SMC from patients for therapeutic angiogenesis. Induced pluripotent stem cells (iPSCs) are regarded as useful stem cells due to their pluripotent differentiation potential. In this study, we explored the therapeutic efficacy of human iPSC‐derived EC and iPSC‐derived SMC in peripheral artery disease model. After the induction of mesodermal differentiation of iPSC, CD34+ progenitor cells were isolated by magnetic‐activated cell sorting. Cultivation of the CD34+ progenitor cells in endothelial culture medium induced the expression of endothelial markers and phenotypes. Moreover, the CD34+ cells could be differentiated into SMC by cultivation in SMC culture medium. In a murine hindlimb ischemia model, cotransplantation of EC with SMC improved blood perfusion and increased the limb salvage rate in ischemic limbs compared to transplantation of either EC or SMC alone. Moreover, cotransplantation of EC and SMC stimulated angiogenesis and led to the formation of capillaries and arteries/arterioles in vivo. Conditioned medium derived from SMC stimulated the migration, proliferation, and tubulation of EC in vitro, and these effects were recapitulated by exosomes isolated from the SMC‐conditioned medium. Together, these results suggest that iPSC‐derived SMC enhance the therapeutic efficacy of iPSC‐derived EC in peripheral artery disease via an exosome‐mediated paracrine mechanism.
Collapse
Affiliation(s)
- Jin Ju Park
- Department of Physiology, College of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Yang Woo Kwon
- Department of Physiology, College of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Jeong Won Kim
- Department of Cogno-Mechatronics Engineering, Pusan National University, Busan, Republic of Korea
| | - Gyu Tae Park
- Department of Physiology, College of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Jung Won Yoon
- Department of Physiology, College of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Ye Seul Kim
- Department of Physiology, College of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Da Sol Kim
- Department of Physiology, College of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Sang Mo Kwon
- Department of Physiology, College of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Sun Sik Bae
- Department of Physiology, College of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Kinarm Ko
- Department of Stem Cell Biology, Konkuk University School of Medicine, Seoul, Republic of Korea
| | - Chang-Seok Kim
- Department of Cogno-Mechatronics Engineering, Pusan National University, Busan, Republic of Korea
| | - Jae Ho Kim
- Department of Physiology, College of Medicine, Pusan National University, Yangsan, Republic of Korea.,Research Institute of Convergence Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| |
Collapse
|
8
|
Floy ME, Mateyka TD, Foreman KL, Palecek SP. Human pluripotent stem cell-derived cardiac stromal cells and their applications in regenerative medicine. Stem Cell Res 2020; 45:101831. [PMID: 32446219 PMCID: PMC7931507 DOI: 10.1016/j.scr.2020.101831] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 03/16/2020] [Accepted: 04/15/2020] [Indexed: 02/06/2023] Open
Abstract
Coronary heart disease is one of the leading causes of death in the United States. Recent advances in stem cell biology have led to the development and engineering of human pluripotent stem cell (hPSC)-derived cardiac cells and tissues for application in cellular therapy and cardiotoxicity studies. Initial studies in this area have largely focused on improving differentiation efficiency and maturation states of cardiomyocytes. However, other cell types in the heart, including endothelial and stromal cells, play crucial roles in cardiac development, injury response, and cardiomyocyte function. This review discusses recent advances in differentiation of hPSCs to cardiac stromal cells, identification and classification of cardiac stromal cell types, and application of hPSC-derived cardiac stromal cells and tissues containing these cells in regenerative and drug development applications.
Collapse
Affiliation(s)
- Martha E Floy
- Department of Chemical and Biological Engineering, University of Wisconsin - Madison, Madison, WI, USA
| | - Taylor D Mateyka
- Department of Chemical and Biological Engineering, University of Wisconsin - Madison, Madison, WI, USA
| | - Koji L Foreman
- Department of Chemical and Biological Engineering, University of Wisconsin - Madison, Madison, WI, USA
| | - Sean P Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin - Madison, Madison, WI, USA.
| |
Collapse
|
9
|
Park SY, Lee H, Kwon YW, Park MR, Kim JH, Kim JB. Etv2- and Fli1-Induced Vascular Progenitor Cells Enhance Functional Recovery in Ischemic Vascular Disease Model-Brief Report. Arterioscler Thromb Vasc Biol 2020; 40:e105-e113. [PMID: 32075417 DOI: 10.1161/atvbaha.119.313684] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVE Vascular progenitor cells (VPCs), which are able to differentiate into both endothelial cells and smooth muscle cells, have the potential for treatment of ischemic diseases. Generated by pluripotent stem cells, VPCs carry the risk of tumorigenicity in clinical application. This issue could be resolved by direct lineage conversion, the induction of functional cells from another lineage by using only lineage-restricted transcription factors. Here, we show that induced VPCs (iVPCs) can be generated from fibroblasts by ETS (E-twenty six) transcription factors, Etv2 and Fli1. Approach and Results: Mouse fibroblasts were infected with lentivirus encoding Etv2 and Fli1. Cell colonies appeared in Fli1- and Etv2/Fli1-infected groups and were mechanically picked. The identity of cell colonies was confirmed by proliferation assay and reverse-transcription polymerase chain reaction with vascular markers. Etv2/Fli1- infected cell colonies were sorted by CD144 (also known as CDH5, VE-cadherin). We defined that CD144-positive iVPCs maintained its own population and expanded stably at multiple passages. iVPCs could differentiate into functional endothelial cells and smooth muscle cells by a defined medium. The functionalities of iVPC-derived endothelial cells and smooth muscle cells were confirmed by analyzing LDL (low-density lipoprotein) uptake, carbachol-induced contraction, and tube formation in vitro. Transplantation of iVPCs into the ischemic hindlimb model enhanced blood flow without tumor formation in vivo. Human iVPCs were generated by human ETS transcription factors ETV2 and FLI1. CONCLUSIONS We demonstrate that ischemic disease curable iVPCs, which have self-renewal and bipotency, can be generated from mouse fibroblasts by enforced ETS family transcription factors, Etv2 and Fli1 expression. Our simple strategy opens insights into stem cell-based ischemic disease therapy.
Collapse
Affiliation(s)
- Soo Yong Park
- From the Hans Schöler Stem Cell Research Center, School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), South Korea (S.Y.P., H.L., M.R.P., J.B.K.)
| | - Hyunah Lee
- From the Hans Schöler Stem Cell Research Center, School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), South Korea (S.Y.P., H.L., M.R.P., J.B.K.)
| | - Yang Woo Kwon
- Department of Physiology, Pusan National University School of Medicine, Yangsan, South Korea (Y.W.K., J.H.K.)
| | - Myung Rae Park
- From the Hans Schöler Stem Cell Research Center, School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), South Korea (S.Y.P., H.L., M.R.P., J.B.K.)
| | - Jae Ho Kim
- Department of Physiology, Pusan National University School of Medicine, Yangsan, South Korea (Y.W.K., J.H.K.)
| | - Jeong Beom Kim
- From the Hans Schöler Stem Cell Research Center, School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), South Korea (S.Y.P., H.L., M.R.P., J.B.K.)
| |
Collapse
|
10
|
Shear Stress Promotes Arterial Endothelium-Oriented Differentiation of Mouse-Induced Pluripotent Stem Cells. Stem Cells Int 2019; 2019:1847098. [PMID: 31827524 PMCID: PMC6881757 DOI: 10.1155/2019/1847098] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 09/05/2019] [Accepted: 10/17/2019] [Indexed: 12/29/2022] Open
Abstract
Establishment of a functional vascular network, which is required in tissue repair and regeneration, needs large-scale production of specific arterial or venous endothelial cells (ECs) from stem cells. Previous in vitro studies by us and others revealed that shear stress induces EC differentiation of bone marrow-derived mesenchymal stem cells and embryonic stem cells. In this study, we focused on the impact of different magnitudes of shear stress on the differentiation of mouse-induced pluripotent stem cells (iPSCs) towards arterial or venous ECs. When iPSCs were exposed to shear stress (5, 10, and 15 dyne/cm2) with 50 ng/mL vascular endothelial growth factor and 10 ng/mL fibroblast growth factor, the expression levels of the general EC markers and the arterial markers increased, and the stress amplitude of 10 dyne/cm2 could be regarded as a proper promoter, whereas the venous and lymphatic markers had little or no expression. Further, shear stress caused cells to align parallel to the direction of the flow, induced cells forming functional tubes, and increased the secretion of nitric oxide. In addition, Notch1 was significantly upregulated, and the Notch ligand Delta-like 4 was activated in response to shear stress, while inhibition of Notch signaling by DAPT remarkably abolished the shear stress-induced arterial epithelium differentiation. Taken together, our results indicate that exposure to appropriate shear stress facilitated the differentiation of mouse iPSCs towards arterial ECs via Notch signaling pathways, which have potential applications for both disease modeling and regenerative medicine.
Collapse
|
11
|
Titmarsh DM, Nurcombe V, Cheung C, Cool SM. Vascular Cells and Tissue Constructs Derived from Human Pluripotent Stem Cells for Toxicological Screening. Stem Cells Dev 2019; 28:1347-1364. [PMID: 31397206 DOI: 10.1089/scd.2018.0246] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The ability of human stem cells to generate somatic cell lineages makes them ideal candidates for use in toxicological testing and eventually, preclinical drug development. Such resources would support an evolution away from human primary cells or research animal models, which suffer from variability and poor predictability, toward off-the-shelf assays of chemical toxicity and drug efficacy using human cells and tissues. To this end, we generated vascular cell populations (smooth muscle cells and endothelial cells) from human pluripotent stem cells (hPSCs), arranged them into 3D co-cultures within supportive gel matrices, and directed their propensity for self-organization resembling microvasculature. The resulting vascular cell populations and co-cultured constructs were then arrayed in high throughput and used for screening a library of environmental and clinical chemical agents for immunological and toxicological responses. The screen effectively stratified the chemicals into various levels of toxicity, with both cell type-specific and co-culture-dependent responses observed. Thus, hPSC-derived vascular cells and constructs could be progressed further toward use in toxicant and drug screening.
Collapse
Affiliation(s)
- Drew M Titmarsh
- Institute of Medical Biology and Agency for Science Technology and Research (A*STAR), Singapore, Singapore
| | - Victor Nurcombe
- Institute of Medical Biology and Agency for Science Technology and Research (A*STAR), Singapore, Singapore.,Lee Kong Chian School of Medicine, Nanyang Technological University-Imperial College London, Singapore, Singapore
| | - Christine Cheung
- Lee Kong Chian School of Medicine, Nanyang Technological University-Imperial College London, Singapore, Singapore.,Institute of Molecular and Cell Biology, Agency for Science Technology and Research (A*STAR), Singapore, Singapore
| | - Simon M Cool
- Institute of Medical Biology and Agency for Science Technology and Research (A*STAR), Singapore, Singapore.,Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
12
|
Kwong G, Marquez HA, Yang C, Wong JY, Kotton DN. Generation of a Purified iPSC-Derived Smooth Muscle-like Population for Cell Sheet Engineering. Stem Cell Reports 2019; 13:499-514. [PMID: 31422908 PMCID: PMC6739689 DOI: 10.1016/j.stemcr.2019.07.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 07/18/2019] [Accepted: 07/19/2019] [Indexed: 10/31/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) provide a potential source for the derivation of smooth muscle cells (SMCs); however, current approaches are limited by the production of heterogeneous cell types and a paucity of tools or markers for tracking and purifying candidate SMCs. Here, we develop murine and human iPSC lines carrying fluorochrome reporters (Acta2hrGFP and ACTA2eGFP, respectively) that identify Acta2+/ACTA2+ cells as they emerge in vitro in real time during iPSC-directed differentiation. We find that Acta2hrGFP+ and ACTA2eGFP+ cells can be sorted to purity and are enriched in markers characteristic of an immature or synthetic SMC. We characterize the resulting GFP+ populations through global transcriptomic profiling and functional studies, including the capacity to form engineered cell sheets. We conclude that these reporter lines allow for generation of sortable, live iPSC-derived Acta2+/ACTA2+ cells highly enriched in smooth muscle lineages for basic developmental studies, tissue engineering, or future clinical regenerative applications.
Collapse
Affiliation(s)
- George Kwong
- Center for Regenerative Medicine, Boston University and Boston Medical Center, 670 Albany Street, 2(nd) Floor, Boston, MA 02118, USA; Department of Biomedical Engineering, Boston University, 44 Cummington Mall, Boston, MA 02215, USA
| | - Hector A Marquez
- Center for Regenerative Medicine, Boston University and Boston Medical Center, 670 Albany Street, 2(nd) Floor, Boston, MA 02118, USA; The Pulmonary Center and Department of Medicine, Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118, USA
| | - Chian Yang
- Center for Regenerative Medicine, Boston University and Boston Medical Center, 670 Albany Street, 2(nd) Floor, Boston, MA 02118, USA; Department of Biomedical Engineering, Boston University, 44 Cummington Mall, Boston, MA 02215, USA
| | - Joyce Y Wong
- Department of Biomedical Engineering, Boston University, 44 Cummington Mall, Boston, MA 02215, USA.
| | - Darrell N Kotton
- Center for Regenerative Medicine, Boston University and Boston Medical Center, 670 Albany Street, 2(nd) Floor, Boston, MA 02118, USA; The Pulmonary Center and Department of Medicine, Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118, USA.
| |
Collapse
|
13
|
The Application of Induced Pluripotent Stem Cells in Pathogenesis Study and Gene Therapy for Vascular Disorders: Current Progress and Future Challenges. Stem Cells Int 2019; 2019:9613258. [PMID: 31281393 PMCID: PMC6594248 DOI: 10.1155/2019/9613258] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 03/05/2019] [Accepted: 03/27/2019] [Indexed: 12/27/2022] Open
Abstract
Vascular disorders are complex diseases with high morbidity and mortality. Among them, the dilated macrovascular diseases (MVD), such as aortic aneurysm and aortic dissection, have presented a huge threat to human health. The pathogenesis of vascular diseases is mostly associated with property alteration of vascular endothelial cells (VECs) and vascular smooth muscle cells (VSMCs). Studies have confirmed that induced pluripotent stem cells (iPSCs) can be proliferated and differentiated into other somatic cells, such as VECs and VSMCs. And patient-specific cells could provide detailed human-associated information in regard to pathogenesis or drug responses. In addition, differentiated ECs from iPSC have been widely used in disease modeling as a cell therapy. In this review, we mainly discussed the application of hiPSCs in investigating the pathological mechanism of different inherited vascular diseases and provide a comprehensive understanding of hiPSCs in the field of clinical diagnosis and gene therapy.
Collapse
|
14
|
Lin H, Du Q, Li Q, Wang O, Wang Z, Liu K, Akert L, Zhang C, Chung S, Duan B, Lei Y. Differentiating human pluripotent stem cells into vascular smooth muscle cells in three dimensional thermoreversible hydrogels. Biomater Sci 2019; 7:347-361. [PMID: 30483691 DOI: 10.1039/c8bm01128a] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Vascular smooth muscle cells (VSMCs) are of great value and are needed in large quantities for tissue engineering, drug screening, disease modeling and cell-based therapies. However, getting high quantity VSMCs remains a challenge. Here, we report a method for the scalable manufacturing of VSMCs from human pluripotent stem cells (hPSCs). hPSCs are expanded and differentiated into VSMCs in a three dimensional (3D) thermoreversible hydrogel. The hydrogel not only acts as a 3D scaffold for cells to grow, but also protects cells from hydrodynamic stresses in the culture vessel and prevents cells from excessive aggregation. Together, the hydrogel creates a cell-friendly microenvironment, leading to high culture efficiency. We show that VSMCs can be generated in 10 days with high viability (>90%), high purity (>80%) and high yield (∼2.0 × 107 cells per mL hydrogel) in the hydrogel scaffold. The generated VSMCs have normal functions. Genome-wide gene expression analysis shows VSMCs made in the hydrogel (i.e. 3D-VSMCs) have higher expression of genes related to vasculature development and glycolysis compared to VSMCs made in the conventional 2D cultures (i.e. 2D-VSMCs), while 2D-VSMCs have higher expression of genes related to cell proliferation. This simple, defined and efficient method is scalable for manufacturing hPSC-VSMCs for various biomedical applications.
Collapse
Affiliation(s)
- Haishuang Lin
- Department of Chemical and Biomolecular Engineering, University of Nebraska-Lincoln, Nebraska, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Colunga T, Hayworth M, Kreß S, Reynolds DM, Chen L, Nazor KL, Baur J, Singh AM, Loring JF, Metzger M, Dalton S. Human Pluripotent Stem Cell-Derived Multipotent Vascular Progenitors of the Mesothelium Lineage Have Utility in Tissue Engineering and Repair. Cell Rep 2019; 26:2566-2579.e10. [PMID: 30840882 PMCID: PMC6585464 DOI: 10.1016/j.celrep.2019.02.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 11/30/2018] [Accepted: 02/02/2019] [Indexed: 01/01/2023] Open
Abstract
In this report we describe a human pluripotent stem cell-derived vascular progenitor (MesoT) cell of the mesothelium lineage. MesoT cells are multipotent and generate smooth muscle cells, endothelial cells, and pericytes and self-assemble into vessel-like networks in vitro. MesoT cells transplanted into mechanically damaged neonatal mouse heart migrate into the injured tissue and contribute to nascent coronary vessels in the repair zone. When seeded onto decellularized vascular scaffolds, MesoT cells differentiate into the major vascular lineages and self-assemble into vasculature capable of supporting peripheral blood flow following transplantation. These findings demonstrate in vivo functionality and the potential utility of MesoT cells in vascular engineering applications.
Collapse
Affiliation(s)
- Thomas Colunga
- Department of Biochemistry and Molecular Biology and Center for Molecular Medicine, University of Georgia, 325 Riverbend Road, Athens, GA 30605, USA
| | - Miranda Hayworth
- Department of Biochemistry and Molecular Biology and Center for Molecular Medicine, University of Georgia, 325 Riverbend Road, Athens, GA 30605, USA
| | - Sebastian Kreß
- Department of Tissue Engineering & Regenerative Medicine, University Hospital Würzburg, 97070 Würzburg, Germany
| | - David M Reynolds
- Department of Biochemistry and Molecular Biology and Center for Molecular Medicine, University of Georgia, 325 Riverbend Road, Athens, GA 30605, USA
| | - Luoman Chen
- Department of Biochemistry and Molecular Biology and Center for Molecular Medicine, University of Georgia, 325 Riverbend Road, Athens, GA 30605, USA
| | - Kristopher L Nazor
- The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Johannes Baur
- Department of General, Visceral, Vascular and Pediatric Surgery, University Hospital of Würzburg, 97080 Würzburg, Germany
| | - Amar M Singh
- Department of Biochemistry and Molecular Biology and Center for Molecular Medicine, University of Georgia, 325 Riverbend Road, Athens, GA 30605, USA
| | - Jeanne F Loring
- The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Marco Metzger
- Translational Centre for Regenerative Therapies TLZ-RT, Fraunhofer Institute for Silicate Research ISC, Röntgenring 11, 97070 Würzburg, Germany
| | - Stephen Dalton
- Department of Biochemistry and Molecular Biology and Center for Molecular Medicine, University of Georgia, 325 Riverbend Road, Athens, GA 30605, USA.
| |
Collapse
|
16
|
Lin H, Qiu X, Du Q, Li Q, Wang O, Akert L, Wang Z, Anderson D, Liu K, Gu L, Zhang C, Lei Y. Engineered Microenvironment for Manufacturing Human Pluripotent Stem Cell-Derived Vascular Smooth Muscle Cells. Stem Cell Reports 2019; 12:84-97. [PMID: 30527760 PMCID: PMC6335449 DOI: 10.1016/j.stemcr.2018.11.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 11/12/2018] [Accepted: 11/13/2018] [Indexed: 12/18/2022] Open
Abstract
Human pluripotent stem cell-derived vascular smooth muscle cells (hPSC-VSMCs) are of great value for disease modeling, drug screening, cell therapies, and tissue engineering. However, producing a high quantity of hPSC-VSMCs with current cell culture technologies remains very challenging. Here, we report a scalable method for manufacturing hPSC-VSMCs in alginate hydrogel microtubes (i.e., AlgTubes), which protect cells from hydrodynamic stresses and limit cell mass to <400 μm to ensure efficient mass transport. The tubes provide cells a friendly microenvironment, leading to extremely high culture efficiency. We have shown that hPSC-VSMCs can be generated in 10 days with high viability, high purity, and high yield (∼5.0 × 108 cells/mL). Phenotype and gene expression showed that VSMCs made in AlgTubes and VSMCs made in 2D cultures were similar overall. However, AlgTube-VSMCs had higher expression of genes related to vasculature development and angiogenesis, and 2D-VSMCs had higher expression of genes related to cell death and biosynthetic processes.
Collapse
Affiliation(s)
- Haishuang Lin
- Department of Chemical and Biomolecular Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Xuefeng Qiu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Qian Du
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Qiang Li
- Department of Chemical and Biomolecular Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA; Biomedical Engineering Program, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Ou Wang
- Department of Chemical and Biomolecular Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA; Biomedical Engineering Program, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Leonard Akert
- Department of Chemical and Biomolecular Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Zhanqi Wang
- Department of Vascular Surgery, Beijing Anzhen Hospital of Capital Medical University, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing 100029, China
| | - Dirk Anderson
- Center for Biotechnology, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Kan Liu
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Linxia Gu
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Chi Zhang
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Yuguo Lei
- Department of Chemical and Biomolecular Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA; Biomedical Engineering Program, University of Nebraska-Lincoln, Lincoln, NE 68588, USA; Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE 68198, USA; Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
17
|
Lau E, Paik DT, Wu JC. Systems-Wide Approaches in Induced Pluripotent Stem Cell Models. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2018; 14:395-419. [PMID: 30379619 DOI: 10.1146/annurev-pathmechdis-012418-013046] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Human induced pluripotent stem cells (iPSCs) provide a renewable supply of patient-specific and tissue-specific cells for cellular and molecular studies of disease mechanisms. Combined with advances in various omics technologies, iPSC models can be used to profile the expression of genes, transcripts, proteins, and metabolites in relevant tissues. In the past 2 years, large panels of iPSC lines have been derived from hundreds of genetically heterogeneous individuals, further enabling genome-wide mapping to identify coexpression networks and elucidate gene regulatory networks. Here, we review recent developments in omics profiling of various molecular phenotypes and the emergence of human iPSCs as a systems biology model of human diseases.
Collapse
Affiliation(s)
- Edward Lau
- Stanford Cardiovascular Institute, and Department of Medicine, Division of Cardiology, Stanford University, Stanford, California 94305, USA;
| | - David T Paik
- Stanford Cardiovascular Institute, and Department of Medicine, Division of Cardiology, Stanford University, Stanford, California 94305, USA;
| | - Joseph C Wu
- Stanford Cardiovascular Institute, and Department of Medicine, Division of Cardiology, Stanford University, Stanford, California 94305, USA; .,Department of Radiology, Stanford University, Stanford, California 94305, USA
| |
Collapse
|
18
|
Human Pluripotent Stem Cells to Engineer Blood Vessels. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2018; 163:147-168. [PMID: 29090328 DOI: 10.1007/10_2017_28] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Development of pluripotent stem cells (PSCs) is a remarkable scientific advancement that allows scientists to harness the power of regenerative medicine for potential treatment of disease using unaffected cells. PSCs provide a unique opportunity to study and combat cardiovascular diseases, which continue to claim the lives of thousands each day. Here, we discuss the differentiation of PSCs into vascular cells, investigation of the functional capabilities of the derived cells, and their utilization to engineer microvascular beds or vascular grafts for clinical application. Graphical Abstract Human iPSCs generated from patients are differentiated toward ECs and perivascular cells for use in disease modeling, microvascular bed development, or vascular graft fabrication.
Collapse
|
19
|
Dorsey TB, Kim D, Grath A, James D, Dai G. Multivalent biomaterial platform to control the distinct arterial venous differentiation of pluripotent stem cells. Biomaterials 2018; 185:1-12. [PMID: 30216805 DOI: 10.1016/j.biomaterials.2018.09.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 08/30/2018] [Accepted: 09/02/2018] [Indexed: 11/25/2022]
Abstract
Vascular endothelial cells (ECs) differentiated from pluripotent stem cells have enormous potential to be used in a variety of therapeutic areas such as tissue engineering of vascular grafts and re-vascularization of ischemic tissues. To date, various protocols have been developed to differentiate stem cells toward vascular ECs. However, current methods are still not sufficient to drive the distinct arterial venous differentiation. Therefore, developing refined method of arterial-venous differentiation is critically needed to address this gap. Here, we developed a biomaterial platform to mimic multivalent ephrin-B2/EphB4 signaling and investigated its role in the early arterial and venous specification of pluripotent stem cells. Our results show immobilized ephrinB2 or EphB4 on hydrogel substrates have a distinct effect on arterial venous differentiation by regulating several arterial venous markers. When in combination with Wnt pathway agonist or BMP4 signaling, the ephrin-B2/EphB4 biomaterial platform can create diverging EC progenitor populations, demonstrating differential gene expression pattern across a wide range of arterial and venous markers, as well as phenotypic markers such as anti-thrombotic, pro-atherogenic and osteogenic genes, that are consistent with the in vivo expression patterns of arterial and venous ECs. Importantly, this distinct EC progenitor population cannot be achieved by current methods of applying soluble factors or hemodynamic stimuli alone, illustrating that fine-tuning of developmental signals using the biomaterial platform offers a new approach to better control the arterial venous differentiation of stem cells.
Collapse
Affiliation(s)
- Taylor B Dorsey
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th St., Troy, NY 12180, United States; Rensselaer Polytechnic Institute, Center for Biotechnology and Interdisciplinary Studies, 1623 15th, St, Troy, NY 12180, United States; Department of Bioengineering, Northeastern University, Boston, MA 02115, United States
| | - Diana Kim
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th St., Troy, NY 12180, United States; Rensselaer Polytechnic Institute, Center for Biotechnology and Interdisciplinary Studies, 1623 15th, St, Troy, NY 12180, United States; Department of Bioengineering, Northeastern University, Boston, MA 02115, United States
| | - Alexander Grath
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th St., Troy, NY 12180, United States; Rensselaer Polytechnic Institute, Center for Biotechnology and Interdisciplinary Studies, 1623 15th, St, Troy, NY 12180, United States; Department of Bioengineering, Northeastern University, Boston, MA 02115, United States
| | - Daylon James
- Center for Reproductive Medicine and Infertility, Weill Cornell Medical College, New York, NY, 10065, United States
| | - Guohao Dai
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th St., Troy, NY 12180, United States; Rensselaer Polytechnic Institute, Center for Biotechnology and Interdisciplinary Studies, 1623 15th, St, Troy, NY 12180, United States; Department of Bioengineering, Northeastern University, Boston, MA 02115, United States.
| |
Collapse
|
20
|
Zhou Y, Kang G, Wen Y, Briggs M, Sebastiano V, Pederson R, Chen B. Do Induced Pluripotent Stem Cell Characteristics Correlate with Efficient In Vitro Smooth Muscle Cell Differentiation? A Comparison of Three Patient-Derived Induced Pluripotent Stem Cell Lines. Stem Cells Dev 2018; 27:1438-1448. [PMID: 30153084 DOI: 10.1089/scd.2018.0031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Human induced pluripotent stem cells (iPSCs) have the potential to repair/regenerate smooth muscle cells (SMCs) in different organs. However, there are many challenges in their translation to clinical therapies. In this study, we describe our observations of in vitro SMC differentiation in three iPSC lines derived from human fibroblasts using retroviral, episomal, and mRNA/miRNA reprogramming methods. We sought to elucidate correlations between differentiation characteristics and efficiencies that can facilitate large-scale production of differentiated cells for clinical applications, and to report differences in pluripotency marker expression in differentiated cells from different iPSC lines. A standardized SMC differentiation protocol was used to induce the CD31+/CD34+ vascular progenitor cell phenotype. These were sorted by magnetic-activated (MACS) and fluorescence-activated cell sorting (FACS), and then treated with PDGF-BB and smooth muscle growth medium for further differentiation into smooth muscle progenitor cells (pSMCs). The expression of SMC and pluripotency markers in early- and late-passage (P1 and P4) pSMCs was analyzed. A total of 36 differentiation runs was performed on the three patient iPSC lines. All pSMC populations expressed SMC markers and Ki67 consistent with the progenitor phenotype. Initial iPSC density correlated positively with the sorted cell FACS efficiency, and this correlation could be fit to a quadratic equation. We also observed that a specific "honeycomb" pattern of the starting cultured iPSCs cultured correlated with higher efficiency in all three iPSC lines. Pluripotency marker expression decreased significantly to nearly undetectable levels in all three lines. There was no significant change in SMC and pluripotent marker expression between passage 1 and 4. In summary, our observations suggest that the method of iPSC reprogramming does not affect iPSC differentiation into pSMCs. Protocol efficiency can be modeled mathematically and coupled with the initial "honeycomb" cell pattern to optimize production of large cell numbers for clinical therapies.
Collapse
Affiliation(s)
- Yingying Zhou
- 1 Department of Obstetrics/Gynecology, Stanford University School of Medicine , Stanford, California.,2 Department of Obstetrics/Gynecology, Shengjing Hospital, China Medical University , Shenyang, People's Republic of China
| | - Gugene Kang
- 3 Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine , Stanford, California
| | - Yan Wen
- 1 Department of Obstetrics/Gynecology, Stanford University School of Medicine , Stanford, California
| | - Mason Briggs
- 1 Department of Obstetrics/Gynecology, Stanford University School of Medicine , Stanford, California
| | - Vittorio Sebastiano
- 3 Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine , Stanford, California
| | - Roger Pederson
- 1 Department of Obstetrics/Gynecology, Stanford University School of Medicine , Stanford, California
| | - Bertha Chen
- 1 Department of Obstetrics/Gynecology, Stanford University School of Medicine , Stanford, California
| |
Collapse
|
21
|
Colunga T, Dalton S. Building Blood Vessels with Vascular Progenitor Cells. Trends Mol Med 2018; 24:630-641. [PMID: 29802036 PMCID: PMC6050017 DOI: 10.1016/j.molmed.2018.05.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 05/02/2018] [Accepted: 05/03/2018] [Indexed: 12/20/2022]
Abstract
Vascular progenitor cells have been identified from perivascular cell fractions and peripheral blood and bone marrow mononuclear fractions. These vascular progenitors share the ability to generate some of the vascular lineages, including endothelial cells, smooth muscle cells, and pericytes. The potential therapeutic uses for vascular progenitor cells are broad and relate to stroke, ischemic disease, and to the engineering of whole organs and tissues that require a vascular component. This review summarizes the best-characterized sources of vascular progenitor cells and discusses advances in 3D printing and electrospinning using blended polymers for the creation of biomimetic vascular grafts. These advances are pushing the field of regenerative medicine closer to the creation of small-diameter vascular grafts with long-term clinical utility.
Collapse
Affiliation(s)
- Thomas Colunga
- Center for Molecular Medicine, University of Georgia, 325 Riverbend Road, Athens, GA 30605, USA; Department of Biochemistry and Molecular Biology, University of Georgia, 325 Riverbend Road, Athens, GA 30605, USA
| | - Stephen Dalton
- Center for Molecular Medicine, University of Georgia, 325 Riverbend Road, Athens, GA 30605, USA; Department of Biochemistry and Molecular Biology, University of Georgia, 325 Riverbend Road, Athens, GA 30605, USA.
| |
Collapse
|
22
|
Ayoubi S, Sheikh SP, Eskildsen TV. Human induced pluripotent stem cell-derived vascular smooth muscle cells: differentiation and therapeutic potential. Cardiovasc Res 2018; 113:1282-1293. [PMID: 28859296 DOI: 10.1093/cvr/cvx125] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 07/12/2017] [Indexed: 12/18/2022] Open
Abstract
Cardiovascular diseases remain the leading cause of death worldwide and current treatment strategies have limited effect of disease progression. It would be desirable to have better models to study developmental and pathological processes and model vascular diseases in laboratory settings. To this end, human induced pluripotent stem cells (hiPSCs) have generated great enthusiasm, and have been a driving force for development of novel strategies in drug discovery and regenerative cell-therapy for the last decade. Hence, investigating the mechanisms underlying the differentiation of hiPSCs into specialized cell types such as cardiomyocytes, endothelial cells, and vascular smooth muscle cells (VSMCs) may lead to a better understanding of developmental cardiovascular processes and potentiate progress of safe autologous regenerative therapies in pathological conditions. In this review, we summarize the latest trends on differentiation protocols of hiPSC-derived VSMCs and their potential application in vascular research and regenerative therapy.
Collapse
Affiliation(s)
- Sohrab Ayoubi
- Department of Cardiovascular and Renal Research, University of Southern Denmark, J.B. Winslowvej 21 3, DK-5000 Odense, Denmark.,Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Sdr. Boulevard 29, DK-5000 Odense, Denmark
| | - Søren P Sheikh
- Department of Cardiovascular and Renal Research, University of Southern Denmark, J.B. Winslowvej 21 3, DK-5000 Odense, Denmark.,Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Sdr. Boulevard 29, DK-5000 Odense, Denmark
| | - Tilde V Eskildsen
- Department of Cardiovascular and Renal Research, University of Southern Denmark, J.B. Winslowvej 21 3, DK-5000 Odense, Denmark.,Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Sdr. Boulevard 29, DK-5000 Odense, Denmark
| |
Collapse
|
23
|
Smolar J, Horst M, Sulser T, Eberli D. Bladder regeneration through stem cell therapy. Expert Opin Biol Ther 2018; 18:525-544. [DOI: 10.1080/14712598.2018.1439013] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Jakub Smolar
- Department of Urology, University Hospital Zurich, Schlieren, Switzerland
| | - Maya Horst
- Department of Urology, University Children’s Hospital Zurich, Zurich, Switzerland
| | - Tulio Sulser
- Department of Urology, University Hospital Zurich, Zurich, Switzerland
| | - Daniel Eberli
- Department of Urology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
24
|
Maguire EM, Xiao Q, Xu Q. Differentiation and Application of Induced Pluripotent Stem Cell–Derived Vascular Smooth Muscle Cells. Arterioscler Thromb Vasc Biol 2017; 37:2026-2037. [DOI: 10.1161/atvbaha.117.309196] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 08/21/2017] [Indexed: 02/06/2023]
Abstract
Vascular smooth muscle cells (VSMCs) play a role in the development of vascular disease, for example, neointimal formation, arterial aneurysm, and Marfan syndrome caused by genetic mutations in VSMCs, but little is known about the mechanisms of the disease process. Advances in induced pluripotent stem cell technology have now made it possible to derive VSMCs from several different somatic cells using a selection of protocols. As such, researchers have set out to delineate key signaling processes involved in triggering VSMC gene expression to grasp the extent of gene regulatory networks involved in phenotype commitment. This technology has also paved the way for investigations into diseases affecting VSMC behavior and function, which may be treatable once an identifiable culprit molecule or gene has been repaired. Moreover, induced pluripotent stem cell–derived VSMCs are also being considered for their use in tissue-engineered blood vessels as they may prove more beneficial than using autologous vessels. Finally, while several issues remains to be clarified before induced pluripotent stem cell–derived VSMCs can become used in regenerative medicine, they do offer both clinicians and researchers hope for both treating and understanding vascular disease. In this review, we aim to update the recent progress on VSMC generation from stem cells and the underlying molecular mechanisms of VSMC differentiation. We will also explore how the use of induced pluripotent stem cell–derived VSMCs has changed the game for regenerative medicine by offering new therapeutic avenues to clinicians, as well as providing researchers with a new platform for modeling of vascular disease.
Collapse
Affiliation(s)
- Eithne Margaret Maguire
- From the Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom (E.M.M., Q. Xiao); and Cardiovascular Division, King’s College London BHF Centre, United Kingdom (Q. Xu)
| | - Qingzhong Xiao
- From the Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom (E.M.M., Q. Xiao); and Cardiovascular Division, King’s College London BHF Centre, United Kingdom (Q. Xu)
| | - Qingbo Xu
- From the Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom (E.M.M., Q. Xiao); and Cardiovascular Division, King’s College London BHF Centre, United Kingdom (Q. Xu)
| |
Collapse
|
25
|
Effects of various monomers and micro-structure of polyhydroxyalkanoates on the behavior of endothelial progenitor cells and endothelial cells for vascular tissue engineering. JOURNAL OF POLYMER RESEARCH 2017. [DOI: 10.1007/s10965-017-1341-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
26
|
Li Y, Wen Y, Green M, Cabral EK, Wani P, Zhang F, Wei Y, Baer TM, Chen B. Cell sex affects extracellular matrix protein expression and proliferation of smooth muscle progenitor cells derived from human pluripotent stem cells. Stem Cell Res Ther 2017; 8:156. [PMID: 28676082 PMCID: PMC5496346 DOI: 10.1186/s13287-017-0606-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 06/01/2017] [Accepted: 06/07/2017] [Indexed: 12/18/2022] Open
Abstract
Background Smooth muscle progenitor cells (pSMCs) differentiated from human pluripotent stem cells (hPSCs) hold great promise for treating diseases or degenerative conditions involving smooth muscle pathologies. However, the therapeutic potential of pSMCs derived from men and women may be very different. Cell sex can exert a profound impact on the differentiation process of stem cells into somatic cells. In spite of advances in translation of stem cell technologies, the role of cell sex and the effect of sex hormones on the differentiation towards mesenchymal lineage pSMCs remain largely unexplored. Methods Using a standard differentiation protocol, two human embryonic stem cell lines (one male line and one female line) and three induced pluripotent stem cell lines (one male line and two female lines) were differentiated into pSMCs. We examined differences in the differentiation of male and female hPSCs into pSMCs, and investigated the effect of 17β-estradiol (E2) on the extracellular matrix (ECM) metabolisms and cell proliferation rates of the pSMCs. Statistical analyses were performed by using Student’s t test or two-way ANOVA, p < 0.05. Results Male and female hPSCs had similar differentiation efficiencies and generated morphologically comparable pSMCs under a standard differentiation protocol, but the derived pSMCs showed sex differences in expression of ECM proteins, such as MMP-2 and TIMP-1, and cell proliferation rates. E2 treatment induced the expression of myogenic gene markers and suppressed ECM degradation activities through reduction of MMP activity and increased expression of TIMP-1 in female pSMCs, but not in male pSMCs. Conclusions hPSC-derived pSMCs from different sexes show differential expression of ECM proteins and proliferation rates. Estrogen appears to promote maturation and ECM protein expression in female pSMCs, but not in male pSMCs. These data suggest that intrinsic cell-sex differences may influence progenitor cell biology. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0606-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yanhui Li
- Department of Obstetrics/Gynecology, Stanford University School of Medicine, 300 Pasteur Drive HH-333, Stanford, CA, 94305, USA.,Department of Obstetrics/Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Yan Wen
- Department of Obstetrics/Gynecology, Stanford University School of Medicine, 300 Pasteur Drive HH-333, Stanford, CA, 94305, USA.
| | - Morgaine Green
- Department of Obstetrics/Gynecology, Stanford University School of Medicine, 300 Pasteur Drive HH-333, Stanford, CA, 94305, USA
| | - Elise K Cabral
- Department of Obstetrics/Gynecology, Stanford University School of Medicine, 300 Pasteur Drive HH-333, Stanford, CA, 94305, USA
| | - Prachi Wani
- Department of Obstetrics/Gynecology, Stanford University School of Medicine, 300 Pasteur Drive HH-333, Stanford, CA, 94305, USA
| | - Fan Zhang
- Department of Obstetrics/Gynecology, Stanford University School of Medicine, 300 Pasteur Drive HH-333, Stanford, CA, 94305, USA
| | - Yi Wei
- Department of Obstetrics/Gynecology, Stanford University School of Medicine, 300 Pasteur Drive HH-333, Stanford, CA, 94305, USA
| | - Thomas M Baer
- Stanford Photonics Research Center, Department of Applied Physics, Stanford University, Stanford, CA, USA
| | - Bertha Chen
- Department of Obstetrics/Gynecology, Stanford University School of Medicine, 300 Pasteur Drive HH-333, Stanford, CA, 94305, USA
| |
Collapse
|
27
|
Shi X, Zhang W, Yin L, Chilian WM, Krieger J, Zhang P. Vascular precursor cells in tissue injury repair. Transl Res 2017; 184:77-100. [PMID: 28284670 PMCID: PMC5429880 DOI: 10.1016/j.trsl.2017.02.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 12/25/2016] [Accepted: 02/14/2017] [Indexed: 12/22/2022]
Abstract
Vascular precursor cells include stem cells and progenitor cells giving rise to all mature cell types in the wall of blood vessels. When tissue injury occurs, local hypoxia and inflammation result in the generation of vasculogenic mediators which orchestrate migration of vascular precursor cells from their niche environment to the site of tissue injury. The intricate crosstalk among signaling pathways coordinates vascular precursor cell proliferation and differentiation during neovascularization. Establishment of normal blood perfusion plays an essential role in the effective repair of the injured tissue. In recent years, studies on molecular mechanisms underlying the regulation of vascular precursor cell function have achieved substantial progress, which promotes exploration of vascular precursor cell-based approaches to treat chronic wounds and ischemic diseases in vital organ systems. Verification of safety and establishment of specific guidelines for the clinical application of vascular precursor cell-based therapy remain major challenges in the field.
Collapse
Affiliation(s)
- Xin Shi
- Department of Integrative Medical Sciences, College of Medicine, Northeast Ohio Medical University, Rootstown, Ohio
| | - Weihong Zhang
- Department of Basic Medicine, School of Nursing, Zhengzhou University, Zhengzhou, Henan Province, PR China
| | - Liya Yin
- Department of Integrative Medical Sciences, College of Medicine, Northeast Ohio Medical University, Rootstown, Ohio
| | - William M Chilian
- Department of Integrative Medical Sciences, College of Medicine, Northeast Ohio Medical University, Rootstown, Ohio
| | - Jessica Krieger
- Department of Integrative Medical Sciences, College of Medicine, Northeast Ohio Medical University, Rootstown, Ohio
| | - Ping Zhang
- Department of Integrative Medical Sciences, College of Medicine, Northeast Ohio Medical University, Rootstown, Ohio.
| |
Collapse
|
28
|
Li Y, Green M, Wen Y, Wei Y, Wani P, Wang Z, Reijo Pera R, Chen B. Efficacy and Safety of Immuno-Magnetically Sorted Smooth Muscle Progenitor Cells Derived from Human-Induced Pluripotent Stem Cells for Restoring Urethral Sphincter Function. Stem Cells Transl Med 2017; 6:1158-1167. [PMID: 28213970 PMCID: PMC5442833 DOI: 10.1002/sctm.16-0160] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 10/31/2016] [Indexed: 12/21/2022] Open
Abstract
Human-induced pluripotent stem cells (hiPSCs)-based cell therapy holds promise for treating stress urinary incontinence (SUI). However, safety concerns, especially tumorgenic potential of residual undifferentiated cells in hiPSC derivatives, are major barriers for its clinical translation. An efficient, fast and clinical-scale strategy for purifying committed cells is also required. Our previous studies demonstrated the regenerative effects of hiPSC-derived smooth muscle progenitor cells (pSMCs) on the injured urethral sphincter in SUI, but the differentiation protocol required fluorescence-activated cell sorting (FACS) which is not practical for autologous clinical applications. In this study, we examined the efficacy and safety of hiPSC-derived pSMC populations sorted by FDA-approved magnetic-activated cell sorting (MACS) using cell-surface marker CD34 for restoring urethral sphincter function. Although the heterogeneity of MACS-sorted pSMCs was higher than that of FACS-sorted pSMCs, the percentage of undifferentiated cells dramatically decreased after directed differentiation in vitro. In vivo studies demonstrated long-term cell integration and no tumor formation of MACS-sorted pSMCs after transplantation. Furthermore, transplantation of MACS-sorted pSMCs into immunodeficient SUI rats was comparable to transplantation with FACS-sorted pSMCs for restoration of the extracellular matrix metabolism and function of the urethral sphincter. In summary, purification of hiPSC derivatives using MACS sorting for CD34 expression represent an efficient approach for production of clinical-scale pSMCs for autologous stem cell therapy for regeneration of smooth muscle tissues. Stem Cells Translational Medicine 2017;6:1158-1167.
Collapse
Affiliation(s)
- Yanhui Li
- Department of Obstetrics/GynecologyStanford University School of MedicineCaliforniaUSA
- Department of Obstetrics/GynecologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyThe People's Republic of China
| | - Morgaine Green
- Department of Obstetrics/GynecologyStanford University School of MedicineCaliforniaUSA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford UniversityCaliforniaUSA
| | - Yan Wen
- Department of Obstetrics/GynecologyStanford University School of MedicineCaliforniaUSA
| | - Yi Wei
- Department of Obstetrics/GynecologyStanford University School of MedicineCaliforniaUSA
| | - Prachi Wani
- Department of Obstetrics/GynecologyStanford University School of MedicineCaliforniaUSA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford UniversityCaliforniaUSA
| | - Zhe Wang
- Department of Obstetrics/GynecologyStanford University School of MedicineCaliforniaUSA
- Department of Obstetrics/GynecologyNanFang Hospital, Southern Medical UniversityGuangzhouGuangdongThe People's Republic of China
| | - Renee Reijo Pera
- Department of Cell Biology & Neuroscience
- Department of Chemistry and BiochemistryMontana State UniversityBozemanMontanaUSA
| | - Bertha Chen
- Department of Obstetrics/GynecologyStanford University School of MedicineCaliforniaUSA
| |
Collapse
|
29
|
Wang Z, Wen Y, Li YH, Wei Y, Green M, Wani P, Zhang P, Pera RR, Chen B. Smooth Muscle Precursor Cells Derived from Human Pluripotent Stem Cells for Treatment of Stress Urinary Incontinence. Stem Cells Dev 2016; 25:453-61. [PMID: 26785911 DOI: 10.1089/scd.2015.0343] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
There is great interest in using stem cells (SC) to regenerate a deficient urethral sphincter in patients with urinary incontinence. The smooth muscle component of the sphincter is a significant contributor to sphincter function. However, current translational efforts for sphincter muscle restoration focus only on skeletal muscle regeneration because they rely on adult mesenchymal SC as cell source. These adult SC do not yield sufficient smooth muscle cells (SMCs) for transplantation. We may be able to overcome this limitation by using pluripotent stem cell (PSC) to derive SMCs. Hence, we sought to investigate whether smooth muscle precursor cells (pSMCs) derived from human PSCs can restore urethral function in an animal model generated by surgical urethrolysis and ovariectomy. Rats were divided into four groups: control (no intervention), sham saline (surgery + saline injection), bladder SMC (surgery + human bladder SMC injection), and treatment (surgery + pSMC injection, which includes human embryonic stem cell (hESC) H9-derived pSMC, episomal reprogrammed induced pluripotent stem cells (iPSCs)-derived pSMC, or viral reprogrammed iPSC-derived pSMC). pSMCs (2 × 10(6) cells/rat) were injected periurethrally 3 weeks postsurgery. Leak point pressure (LPP) and baseline external urethral sphincter electromyography were measured 5 weeks postinjection. Both iPSC-derived pSMC treatment groups showed significantly higher LPP compared to the sham saline group, consistent with restoration of urethral sphincter function. While the difference between the H9-derived pSMC treatment and sham saline group was not significant, it did show a trend toward restoration of the LPP to the level of intact controls. Our data indicate that pSMCs derived from human PSCs (hESC and iPSC) can restore sphincter function.
Collapse
Affiliation(s)
- Zhe Wang
- 1 Department of OB/GYN, Stanford University School of Medicine , Stanford, California.,2 The Center for Reproductive Medicine, OB/GYN, Nanfang Hospital, Southern Medical University , Guangzhou, Guangdong, People's Republic of China
| | - Yan Wen
- 1 Department of OB/GYN, Stanford University School of Medicine , Stanford, California
| | - Yan Hui Li
- 1 Department of OB/GYN, Stanford University School of Medicine , Stanford, California.,3 The Department of OB/GYN, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan, People's Republic of China
| | - Yi Wei
- 1 Department of OB/GYN, Stanford University School of Medicine , Stanford, California
| | - Morgaine Green
- 1 Department of OB/GYN, Stanford University School of Medicine , Stanford, California
| | - Prachi Wani
- 1 Department of OB/GYN, Stanford University School of Medicine , Stanford, California
| | - Pengbo Zhang
- 4 Department of Pathology, Stanford University School of Medicine , Stanford, California
| | - Renee Reijo Pera
- 5 Department of Cell Biology and Neurosciences and Department of Chemistry and Biochemistry, Montana State University , Bozeman, Montana
| | - Bertha Chen
- 1 Department of OB/GYN, Stanford University School of Medicine , Stanford, California
| |
Collapse
|
30
|
Gao L, Cui W, Zhang P, Jang A, Zhu W, Zhang J. 31P NMR 2D Mapping of Creatine Kinase Forward Flux Rate in Hearts with Postinfarction Left Ventricular Remodeling in Response to Cell Therapy. PLoS One 2016; 11:e0162149. [PMID: 27606901 PMCID: PMC5015918 DOI: 10.1371/journal.pone.0162149] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Accepted: 07/15/2016] [Indexed: 11/19/2022] Open
Abstract
Utilizing a fast 31P magnetic resonance spectroscopy (MRS) 2-dimensional chemical shift imaging (2D-CSI) method, this study examined the heterogeneity of creatine kinase (CK) forward flux rate of hearts with postinfarction left ventricular (LV) remodeling. Immunosuppressed Yorkshire pigs were assigned to 4 groups: 1) A sham-operated normal group (SHAM, n = 6); 2) A 60 minutes distal left anterior descending coronary artery ligation and reperfusion (MI, n = 6); 3) Open patch group; ligation injury plus open fibrin patch over the site of injury (Patch, n = 6); and 4) Cell group, hiPSCs-cardiomyocytes, -endothelial cells, and -smooth muscle cells (2 million, each) were injected into the injured myocardium pass through a fibrin patch (Cell+Patch, n = 5). At 4 weeks, the creatine phosphate (PCr)/ATP ratio, CK forward flux rate (Flux PCr→ATP), and k constant of CK forward flux rate (kPCr→ATP) were severely decreased at border zone myocardium (BZ) adjacent to MI. Cell treatment results in significantly increase of PCr/ATP ratio and improve the value of kPCr→ATP and Flux PCr→ATP in BZ myocardium. Moreover, the BZ myocardial CK total activity and protein expression of CK mitochondria isozyme and CK myocardial isozyme were significantly reduced, but recovered in response to cell treatment. Thus, cell therapy results in improvement of BZ bioenergetic abnormality in hearts with postinfarction LV remodeling, which is accompanied by significantly improvements in BZ CK activity and CK isozyme expression. The fast 2D 31P MR CSI mapping can reliably measure the heterogeneity of bioenergetics in hearts with post infarction LV remodeling.
Collapse
Affiliation(s)
- Ling Gao
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, Alabama, 35233, United States of America
| | - Weina Cui
- Department of Medicine, Cardiology Division, University of Minnesota—Twin Cities, Minneapolis, Minnesota, 55455, United States of America
| | - Pengyuan Zhang
- Department of Medicine, Cardiology Division, University of Minnesota—Twin Cities, Minneapolis, Minnesota, 55455, United States of America
| | - Albert Jang
- Department of Medicine, Cardiology Division, University of Minnesota—Twin Cities, Minneapolis, Minnesota, 55455, United States of America
| | - Wuqiang Zhu
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, Alabama, 35233, United States of America
| | - Jianyi Zhang
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, Alabama, 35233, United States of America
- * E-mail:
| |
Collapse
|
31
|
Smolar J, Salemi S, Horst M, Sulser T, Eberli D. Stem Cells in Functional Bladder Engineering. Transfus Med Hemother 2016; 43:328-335. [PMID: 27781020 PMCID: PMC5073506 DOI: 10.1159/000447977] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 06/23/2016] [Indexed: 12/24/2022] Open
Abstract
Conditions impairing bladder function in children and adults, such as myelomeningocele, posterior urethral valves, bladder exstrophy or spinal cord injury, often need urinary diversion or augmentation cystoplasty as when untreated they may cause severe bladder dysfunction and kidney failure. Currently, the gold standard therapy of end-stage bladder disease refractory to conservative management is enterocystoplasty, a surgical enlargement of the bladder with intestinal tissue. Despite providing functional improvement, enterocystoplasty is associated with significant long-term complications, such as recurrent urinary tract infections, metabolic abnormalities, stone formation, and malignancies. Therefore, there is a strong clinical need for alternative therapies for these reconstructive procedures, of which stem cell-based tissue engineering (TE) is considered to be the most promising future strategy. This review is focused on the recent progress in bladder stem cell research and therapy and the challenges that remain for the development of a functional bladder wall.
Collapse
Affiliation(s)
- Jakub Smolar
- Laboratory for Tissue Engineering and Stem Cell Therapy, Department of Urology, University Hospital Zurich, Zurich, Switzerland
| | - Souzan Salemi
- Laboratory for Tissue Engineering and Stem Cell Therapy, Department of Urology, University Hospital Zurich, Zurich, Switzerland
| | - Maya Horst
- Division of Pediatric Urology, Department of Pediatric Surgery, University Children's Hospital, Zurich, Switzerland
| | - Tullio Sulser
- Laboratory for Tissue Engineering and Stem Cell Therapy, Department of Urology, University Hospital Zurich, Zurich, Switzerland
| | - Daniel Eberli
- Laboratory for Tissue Engineering and Stem Cell Therapy, Department of Urology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
32
|
Hsia K, Yao CL, Chen WM, Chen JH, Lee H, Lu JH. Scaffolds and Cell-Based Tissue Engineering for Blood Vessel Therapy. Cells Tissues Organs 2016; 202:281-295. [PMID: 27548610 DOI: 10.1159/000448169] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/04/2016] [Indexed: 11/19/2022] Open
Abstract
The increasing morbidity of cardiovascular diseases in modern society has made it crucial to develop a small-caliber blood vessel. In the absence of appropriate autologous vascular grafts, an alternative prosthesis must be constructed for cardiovascular disease patients. The aim of this article is to describe the advances in making cell-seeded cardiovascular prostheses. It also discusses the combinations of types of scaffolds and cells, especially autologous stem cells, which are suitable for application in tissue-engineered vessels with the favorable properties of mechanical strength, antithrombogenicity, biocompliance, anti-inflammation, fatigue resistance and long-term durability. This article highlights the advancements in cellular tissue-engineered vessels in recent years.
Collapse
|
33
|
Li Y, Wen Y, Wang Z, Wei Y, Wani P, Green M, Swaminathan G, Ramamurthi A, Pera RR, Chen B. Smooth Muscle Progenitor Cells Derived From Human Pluripotent Stem Cells Induce Histologic Changes in Injured Urethral Sphincter. Stem Cells Transl Med 2016; 5:1719-1729. [PMID: 27460854 DOI: 10.5966/sctm.2016-0035] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 05/13/2016] [Indexed: 12/21/2022] Open
Abstract
: Data suggest that myoblasts from various sources, including bone marrow, skeletal muscle, and adipose tissue, can restore muscle function in patients with urinary incontinence. Animal data have indicated that these progenitor cells exert mostly a paracrine effect on the native tissues rather than cell regeneration. Limited knowledge is available on the in vivo effect of human stem cells or muscle progenitors on injured muscles. We examined in vivo integration of smooth muscle progenitor cells (pSMCs) derived from human pluripotent stem cells (hPSCs). pSMCs were derived from a human embryonic stem cell line (H9-ESCs) and two induced pluripotent stem cell (iPSC) lines. pSMCs were injected periurethrally into urethral injury rat models (2 × 106 cells per rat) or intramuscularly into severe combined immunodeficiency mice. Histologic and quantitative image analysis revealed that the urethras in pSMC-treated rats contained abundant elastic fibers and thicker muscle layers compared with the control rats. Western blot confirmed increased elastin/collagen III content in the urethra and bladder of the H9-pSMC-treated rats compared with controls. iPSC-pSMC treatment also showed similar trends in elastin and collagen III. Human elastin gene expression was not detectable in rodent tissues, suggesting that the extracellular matrix synthesis resulted from the native rodent tissues rather than from the implanted human cells. Immunofluorescence staining and in vivo bioluminescence imaging confirmed long-term engraftment of pSMCs into the host urethra and the persistence of the smooth muscle phenotype. Taken together, the data suggest that hPSC-derived pSMCs facilitate restoration of urethral sphincter function by direct smooth muscle cell regeneration and by inducing native tissue elastin/collagen III remodeling. SIGNIFICANCE The present study provides evidence that a pure population of human smooth muscle progenitor cells (pSMCs) derived from human pluripotent stem cells (hPSCs) (human embryonic stem cells and patient induced pluripotent stem cells) restores urethral sphincter function by two mechanisms: modulation of extracellular matrix protein metabolism in vivo and pSMC proliferation and differentiation into smooth muscle cells to regenerate the muscle layer in the lower urinary tract. These findings on the in vivo effects of human pSMCs should aid in optimizing regenerative therapies using human myoblasts.
Collapse
Affiliation(s)
- Yanhui Li
- Department of Obstetrics/Gynecology, Stanford University School of Medicine, Stanford, California, USA
- Department of Obstetrics/Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, People's Republic of China
| | - Yan Wen
- Department of Obstetrics/Gynecology, Stanford University School of Medicine, Stanford, California, USA
| | - Zhe Wang
- Department of Obstetrics/Gynecology, Stanford University School of Medicine, Stanford, California, USA
- Department of Obstetrics/Gynecology, NanFang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Yi Wei
- Department of Obstetrics/Gynecology, Stanford University School of Medicine, Stanford, California, USA
| | - Prachi Wani
- Department of Obstetrics/Gynecology, Stanford University School of Medicine, Stanford, California, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California, USA
| | - Morgaine Green
- Department of Obstetrics/Gynecology, Stanford University School of Medicine, Stanford, California, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California, USA
| | - Ganesh Swaminathan
- Department of Biomedical Engineering, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio, USA
| | - Anand Ramamurthi
- Department of Biomedical Engineering, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio, USA
| | - Renee Reijo Pera
- Department of Cell Biology and Neuroscience, Montana State University, Bozeman, Montana, USA
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, Montana, USA
| | - Bertha Chen
- Department of Obstetrics/Gynecology, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
34
|
Chen IY, Matsa E, Wu JC. Induced pluripotent stem cells: at the heart of cardiovascular precision medicine. Nat Rev Cardiol 2016; 13:333-49. [PMID: 27009425 DOI: 10.1038/nrcardio.2016.36] [Citation(s) in RCA: 133] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The advent of human induced pluripotent stem cell (hiPSC) technology has revitalized the efforts in the past decade to realize more fully the potential of human embryonic stem cells for scientific research. Adding to the possibility of generating an unlimited amount of any cell type of interest, hiPSC technology now enables the derivation of cells with patient-specific phenotypes. Given the introduction and implementation of the large-scale Precision Medicine Initiative, hiPSC technology will undoubtedly have a vital role in the advancement of cardiovascular research and medicine. In this Review, we summarize the progress that has been made in the field of hiPSC technology, with particular emphasis on cardiovascular disease modelling and drug development. The growing roles of hiPSC technology in the practice of precision medicine will also be discussed.
Collapse
Affiliation(s)
- Ian Y Chen
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California 94305, USA.,Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Elena Matsa
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Joseph C Wu
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California 94305, USA.,Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, USA.,Department of Radiology, Stanford University School of Medicine, Stanford, California 94305, USA
| |
Collapse
|
35
|
Yang L, Geng Z, Nickel T, Johnson C, Gao L, Dutton J, Hou C, Zhang J. Differentiation of Human Induced-Pluripotent Stem Cells into Smooth-Muscle Cells: Two Novel Protocols. PLoS One 2016; 11:e0147155. [PMID: 26771193 PMCID: PMC4714916 DOI: 10.1371/journal.pone.0147155] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 12/29/2015] [Indexed: 01/18/2023] Open
Abstract
Conventional protocols for differentiating human induced-pluripotent stem cells (hiPSCs) into smooth-muscle cells (SMCs) can be inefficient and generally fail to yield cells with a specific SMC phenotype (i.e., contractile or synthetic SMCs). Here, we present two novel hiPSC-SMC differentiation protocols that yield SMCs with predominantly contractile or synthetic phenotypes. Flow cytometry analyses of smooth-muscle actin (SMA) expression indicated that ~45% of the cells obtained with each protocol assumed an SMC phenotype, and that the populations could be purified to ~95% via metabolic selection. Assessments of cellular mRNA and/or protein levels indicated that SMA, myosin heavy chain II, collagen 1, calponin, transgelin, connexin 43, and vimentin expression in the SMCs obtained via the Contractile SMC protocol and in SMCs differentiated via a traditional protocol were similar, while SMCs produced via the Sythetic SMC protocol expressed less calponin, more collagen 1, and more connexin 43. Differences were also observed in functional assessments of the two SMC populations: the two-dimensional surface area of Contractile SMCs declined more extensively (to 12% versus 44% of original size) in response to carbachol treatment, while quantification of cell migration and proliferation were greater in Synthetic SMCs. Collectively, these data demonstrate that our novel differentiation protocols can efficiently generate SMCs from hiPSCs.
Collapse
Affiliation(s)
- Libang Yang
- Division of Cardiology, Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Zhaohui Geng
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Thomas Nickel
- Division of Cardiology, Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Caitlin Johnson
- Division of Cardiology, Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Lin Gao
- Division of Cardiology, Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - James Dutton
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Cody Hou
- Division of Cardiology, Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Jianyi Zhang
- Division of Cardiology, Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota, United States of America
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota, United States of America
- Department of Biomedical Engineering, School of Medicine, School of Engineering, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| |
Collapse
|
36
|
Biel NM, Santostefano KE, DiVita BB, El Rouby N, Carrasquilla SD, Simmons C, Nakanishi M, Cooper-DeHoff RM, Johnson JA, Terada N. Vascular Smooth Muscle Cells From Hypertensive Patient-Derived Induced Pluripotent Stem Cells to Advance Hypertension Pharmacogenomics. Stem Cells Transl Med 2015; 4:1380-90. [PMID: 26494780 PMCID: PMC4675511 DOI: 10.5966/sctm.2015-0126] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 08/31/2015] [Indexed: 12/12/2022] Open
Abstract
UNLABELLED Studies in hypertension (HTN) pharmacogenomics seek to identify genetic sources of variable antihypertensive drug response. Genetic association studies have detected single-nucleotide polymorphisms (SNPs) that link to drug responses; however, to understand mechanisms underlying how genetic traits alter drug responses, a biological interface is needed. Patient-derived induced pluripotent stem cells (iPSCs) provide a potential source for studying otherwise inaccessible tissues that may be important to antihypertensive drug response. The present study established multiple iPSC lines from an HTN pharmacogenomics cohort. We demonstrated that established HTN iPSCs can robustly and reproducibly differentiate into functional vascular smooth muscle cells (VSMCs), a cell type most relevant to vasculature tone control. Moreover, a sensitive traction force microscopy assay demonstrated that iPSC-derived VSMCs show a quantitative contractile response on physiological stimulus of endothelin-1. Furthermore, the inflammatory chemokine tumor necrosis factor α induced a typical VSMC response in iPSC-derived VSMCs. These studies pave the way for a large research initiative to decode biological significance of identified SNPs in hypertension pharmacogenomics. SIGNIFICANCE Treatment of hypertension remains suboptimal, and a pharmacogenomics approach seeks to identify genetic biomarkers that could be used to guide treatment decisions; however, it is important to understand the biological underpinnings of genetic associations. Mouse models do not accurately recapitulate individual patient responses based on their genetics, and hypertension-relevant cells are difficult to obtain from patients. Induced pluripotent stem cell (iPSC) technology provides a great interface to bring patient cells with their genomic data into the laboratory and to study hypertensive responses. As an initial step, the present study established an iPSC bank from patients with primary hypertension and demonstrated an effective and reproducible method of generating functional vascular smooth muscle cells.
Collapse
Affiliation(s)
- Nikolett M. Biel
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
- Center for Cellular Reprogramming, University of Florida, Gainesville, Florida, USA
| | - Katherine E. Santostefano
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
- Center for Cellular Reprogramming, University of Florida, Gainesville, Florida, USA
| | - Bayli B. DiVita
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
- Center for Cellular Reprogramming, University of Florida, Gainesville, Florida, USA
| | - Nihal El Rouby
- Department of Pharmacotherapy and Translational Research, College of Pharmacy and Center for Pharmacogenomics, University of Florida, Gainesville, Florida, USA
| | - Santiago D. Carrasquilla
- J. Crayton Pruitt Family Department of Biomedical Engineering, College of Engineering, University of Florida, Gainesville, Florida, USA
| | - Chelsey Simmons
- Mechanical and Aerospace Engineering, College of Engineering, University of Florida, Gainesville, Florida, USA
- Division of Cardiovascular Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Mahito Nakanishi
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| | - Rhonda M. Cooper-DeHoff
- Department of Pharmacotherapy and Translational Research, College of Pharmacy and Center for Pharmacogenomics, University of Florida, Gainesville, Florida, USA
- Division of Cardiovascular Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Julie A. Johnson
- Department of Pharmacotherapy and Translational Research, College of Pharmacy and Center for Pharmacogenomics, University of Florida, Gainesville, Florida, USA
- Division of Cardiovascular Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Naohiro Terada
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
- Center for Cellular Reprogramming, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
37
|
Nakayama KH, Joshi PA, Lai ES, Gujar P, Joubert LM, Chen B, Huang NF. Bilayered vascular graft derived from human induced pluripotent stem cells with biomimetic structure and function. Regen Med 2015; 10:745-55. [PMID: 26440211 DOI: 10.2217/rme.15.45] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND We developed an aligned bi-layered vascular graft derived from human induced pluripotent stem cells (iPSCs) that recapitulates the cellular composition, orientation, and anti-inflammatory function of blood vessels. MATERIALS & METHODS The luminal layer consisted of longitudinal-aligned nanofibrillar collagen containing primary endothelial cells (ECs) or iPSC-derived ECs (iPSC-ECs). The outer layer contained circumferentially oriented nanofibrillar collagen with primary smooth muscle cells (SMCs) or iPSC-derived SMCs(iPSC-SMCs). RESULTS On the aligned scaffolds, cells organized F-actin assembly within 8º from the direction of nanofibrils. When compared to randomly-oriented scaffolds, EC-seeded aligned scaffolds had significant reduced inflammatory response, based on adhesivity to monocytes. CONCLUSION This study highlights the importance of anisotropic scaffolds in directing cell form and function, and has therapeutic significance as physiologically relevant blood vessels.
Collapse
Affiliation(s)
- Karina H Nakayama
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305, USA.,Department of Cardiothoracic Surgery, Stanford University, 300 Pasteur Drive, Stanford, CA 94305-5407, USA.,Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Prajakta A Joshi
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304, USA.,Department of Biological Sciences, San Jose State University, San Jose, CA 95112, USA
| | - Edwina S Lai
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Prachi Gujar
- Department of Obstetrics & Gynecology, Stanford University, Stanford, CA 94305, USA
| | - Lydia-M Joubert
- Cell Sciences Imaging Facility, Stanford University, Stanford, CA 94305, USA
| | - Bertha Chen
- Department of Obstetrics & Gynecology, Stanford University, Stanford, CA 94305, USA
| | - Ngan F Huang
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305, USA.,Department of Cardiothoracic Surgery, Stanford University, 300 Pasteur Drive, Stanford, CA 94305-5407, USA.,Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304, USA
| |
Collapse
|
38
|
Lowenthal J, Gerecht S. Stem cell-derived vasculature: A potent and multidimensional technology for basic research, disease modeling, and tissue engineering. Biochem Biophys Res Commun 2015; 473:733-42. [PMID: 26427871 DOI: 10.1016/j.bbrc.2015.09.127] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 09/23/2015] [Indexed: 02/08/2023]
Abstract
Proper blood vessel networks are necessary for constructing and re-constructing tissues, promoting wound healing, and delivering metabolic necessities throughout the body. Conversely, an understanding of vascular dysfunction has provided insight into the pathogenesis and progression of diseases both common and rare. Recent advances in stem cell-based regenerative medicine - including advances in stem cell technologies and related progress in bioscaffold design and complex tissue engineering - have allowed rapid advances in the field of vascular biology, leading in turn to more advanced modeling of vascular pathophysiology and improved engineering of vascularized tissue constructs. In this review we examine recent advances in the field of stem cell-derived vasculature, providing an overview of stem cell technologies as a source for vascular cell types and then focusing on their use in three primary areas: studies of vascular development and angiogenesis, improved disease modeling, and the engineering of vascularized constructs for tissue-level modeling and cell-based therapies.
Collapse
Affiliation(s)
- Justin Lowenthal
- Medical Scientist Training Program, School of Medicine, Johns Hopkins University, Baltimore, MD, United States; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Sharon Gerecht
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, United States; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, United States; Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, United States.
| |
Collapse
|
39
|
Martin U. New Muscle for Old Hearts: Engineering Tissue from Pluripotent Stem Cells. Hum Gene Ther 2015; 26:305-11. [DOI: 10.1089/hum.2015.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Ulrich Martin
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, REBIRTH Cluster of Excellence, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
40
|
A practical guide to induced pluripotent stem cell research using patient samples. J Transl Med 2015; 95:4-13. [PMID: 25089770 DOI: 10.1038/labinvest.2014.104] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 06/24/2014] [Accepted: 06/27/2014] [Indexed: 12/13/2022] Open
Abstract
Approximately 3 years ago, we assessed how patient induced pluripotent stem cell (iPSC) research could potentially impact human pathobiology studies in the future. Since then, the field has grown considerably with numerous technical developments, and the idea of modeling diseases 'in a dish' is becoming increasingly popular in biomedical research. Likely, it is even acceptable to include patient iPSCs as one of the standard research tools for disease mechanism studies, just like knockout mice. However, as the field matures, we acknowledge there remain many practical limitations and obstacles for their genuine application to understand diseases, and accept that it has not been as straightforward to model disorders as initially proposed. A major practical challenge has been efficient direction of iPSC differentiation into desired lineages and preparation of the large numbers of specific cell types required for study. Another even larger obstacle is the limited value of in vitro outcomes, which often do not closely represent disease conditions. To overcome the latter issue, many new approaches are underway, including three-dimensional organoid cultures from iPSCs, xenotransplantation of human cells to animal models and in vitro interaction of multiple cell types derived from isogenic iPSCs. Here we summarize the areas where patient iPSC studies have provided truly valuable information beyond existing skepticism, discuss the desired technologies to overcome current limitations and include practical guidance for how to utilize the resources. Undoubtedly, these human patient cells are an asset for experimental pathology studies. The future rests on how wisely we use them.
Collapse
|
41
|
Lian X, Bao X, Al-Ahmad A, Liu J, Wu Y, Dong W, Dunn KK, Shusta EV, Palecek SP. Efficient differentiation of human pluripotent stem cells to endothelial progenitors via small-molecule activation of WNT signaling. Stem Cell Reports 2014; 3:804-16. [PMID: 25418725 PMCID: PMC4235141 DOI: 10.1016/j.stemcr.2014.09.005] [Citation(s) in RCA: 241] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2014] [Revised: 09/05/2014] [Accepted: 09/08/2014] [Indexed: 12/13/2022] Open
Abstract
Human pluripotent stem cell (hPSC)-derived endothelial cells and their progenitors may provide the means for vascularization of tissue-engineered constructs and can serve as models to study vascular development and disease. Here, we report a method to efficiently produce endothelial cells from hPSCs via GSK3 inhibition and culture in defined media to direct hPSC differentiation to CD34+CD31+ endothelial progenitors. Exogenous vascular endothelial growth factor (VEGF) treatment was dispensable, and endothelial progenitor differentiation was β-catenin dependent. Furthermore, by clonal analysis, we showed that CD34+CD31+CD117+TIE-2+ endothelial progenitors were multipotent, capable of differentiating into calponin-expressing smooth muscle cells and CD31+CD144+vWF+I-CAM1+ endothelial cells. These endothelial cells were capable of 20 population doublings, formed tube-like structures, imported acetylated low-density lipoprotein, and maintained a dynamic barrier function. This study provides a rapid and efficient method for production of hPSC-derived endothelial progenitors and endothelial cells and identifies WNT/β-catenin signaling as a primary regulator for generating vascular cells from hPSCs. WNT pathway activation directs hPSC differentiation to endothelial progenitors hPSC-derived endothelial progenitors can differentiate to endothelial cells Purified hPSC-derived endothelial cells are capable of 20 population doublings WNT pathway activation permits defined production of endothelial cells from hPSCs
Collapse
Affiliation(s)
- Xiaojun Lian
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Xiaoping Bao
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Abraham Al-Ahmad
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Jialu Liu
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Yue Wu
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Wentao Dong
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Kaitlin K Dunn
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Eric V Shusta
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Sean P Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA.
| |
Collapse
|