1
|
Paul SK, Oshima M, Patil A, Sone M, Kato H, Maezawa Y, Kaneko H, Fukuyo M, Rahmutulla B, Ouchi Y, Tsujimura K, Nakanishi M, Kaneda A, Iwama A, Yokote K, Eto K, Takayama N. Retrotransposons in Werner syndrome-derived macrophages trigger type I interferon-dependent inflammation in an atherosclerosis model. Nat Commun 2024; 15:4772. [PMID: 38858384 PMCID: PMC11164933 DOI: 10.1038/s41467-024-48663-w] [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: 05/07/2022] [Accepted: 05/07/2024] [Indexed: 06/12/2024] Open
Abstract
The underlying mechanisms of atherosclerosis, the second leading cause of death among Werner syndrome (WS) patients, are not fully understood. Here, we establish an in vitro co-culture system using macrophages (iMφs), vascular endothelial cells (iVECs), and vascular smooth muscle cells (iVSMCs) derived from induced pluripotent stem cells. In co-culture, WS-iMφs induces endothelial dysfunction in WS-iVECs and characteristics of the synthetic phenotype in WS-iVSMCs. Transcriptomics and open chromatin analysis reveal accelerated activation of type I interferon signaling and reduced chromatin accessibility of several transcriptional binding sites required for cellular homeostasis in WS-iMφs. Furthermore, the H3K9me3 levels show an inverse correlation with retrotransposable elements, and retrotransposable element-derived double-stranded RNA activates the DExH-box helicase 58 (DHX58)-dependent cytoplasmic RNA sensing pathway in WS-iMφs. Conversely, silencing type I interferon signaling in WS-iMφs rescues cell proliferation and suppresses cellular senescence and inflammation. These findings suggest that Mφ-specific inhibition of type I interferon signaling could be targeted to treat atherosclerosis in WS patients.
Collapse
Affiliation(s)
- Sudip Kumar Paul
- Department of Regenerative Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Motohiko Oshima
- Division of Stem Cell and Molecular Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | | | - Masamitsu Sone
- Department of Regenerative Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
- Hibernation Metabolism, Physiology and Development Group, Institute of Low Temperature Science, Hokkaido University, Sapporo, Japan
| | - Hisaya Kato
- Department of Endocrinology, Hematology and Gerontology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Yoshiro Maezawa
- Department of Endocrinology, Hematology and Gerontology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Hiyori Kaneko
- Department of Endocrinology, Hematology and Gerontology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Masaki Fukuyo
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Bahityar Rahmutulla
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Yasuo Ouchi
- Department of Regenerative Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Kyoko Tsujimura
- Department of Regenerative Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | | | - Atsushi Kaneda
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Atsushi Iwama
- Division of Stem Cell and Molecular Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Koutaro Yokote
- Department of Endocrinology, Hematology and Gerontology, Graduate School of Medicine, Chiba University, Chiba, Japan.
| | - Koji Eto
- Department of Regenerative Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan.
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan.
| | - Naoya Takayama
- Department of Regenerative Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan.
| |
Collapse
|
2
|
Holm A, Mulliken JB, Bischoff J. Infantile hemangioma: the common and enigmatic vascular tumor. J Clin Invest 2024; 134:e172836. [PMID: 38618963 PMCID: PMC11014660 DOI: 10.1172/jci172836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2024] Open
Abstract
Infantile hemangioma (IH) is a benign vascular tumor that occurs in 5% of newborns. The tumor follows a life cycle of rapid proliferation in infancy, followed by slow involution in childhood. This unique life cycle has attracted the interest of basic and clinical scientists alike as a paradigm for vasculogenesis, angiogenesis, and vascular regression. Unanswered questions persist about the genetic and molecular drivers of the proliferating and involuting phases. The beta blocker propranolol usually accelerates regression of problematic IHs, yet its mechanism of action on vascular proliferation and differentiation is unclear. Some IHs fail to respond to beta blockers and regrow after discontinuation. Side effects occur and long-term sequelae of propranolol treatment are unknown. This poses clinical challenges and raises novel questions about the mechanisms of vascular overgrowth in IH.
Collapse
Affiliation(s)
- Annegret Holm
- Vascular Biology Program and Department of Surgery, Boston Children’s Hospital, Boston, Massachusetts, USA
- Department of Surgery, Harvard Medical School, Boston, Massachusetts, USA
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, University Hospital Freiburg, VASCERN-VASCA European Reference Center, Freiburg, Germany
| | - John B. Mulliken
- Department of Surgery, Harvard Medical School, Boston, Massachusetts, USA
- Department of Plastic and Oral Surgery, Boston Children’s Hospital, Boston, Massachusetts, USA
| | - Joyce Bischoff
- Vascular Biology Program and Department of Surgery, Boston Children’s Hospital, Boston, Massachusetts, USA
- Department of Surgery, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
3
|
Meng Y, Du J, Liu N, Qiang Y, Xiao L, Lan X, Ma L, Yang J, Yu J, Lu G. Epigenetic modulation: Research progress on histone acetylation levels in major depressive disorders. J Drug Target 2023; 31:142-151. [PMID: 36112185 DOI: 10.1080/1061186x.2022.2125978] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Depression is a serious mental illness and a prevalent condition with multiple aetiologies. The impact of the current therapeutic strategies is limited and the pathogenesis of the illness is not well understood. According to previous studies, depression onset is influenced by a variety of environmental and genetic factors, including chronic stress, aberrant changes in gene expression, and hereditary predisposition. Transcriptional regulation in eukaryotes is closely related to chromosome packing and is controlled by histone post-translational modifications. The development of new antidepressants may proceed along a new path with medications that target epigenetics. Histone deacetylase inhibitors (HDACis) are a class of compounds that interfere with the function of histone deacetylases (HDACs). This review explores the relationship between HDACs and depression and focuses on the current knowledge on their regulatory mechanism in depression and the potential therapeutic use of HDACis with antidepressant efficacy in preclinical research. Future research on inhibitors is also proposed and discussed.
Collapse
Affiliation(s)
- Yuan Meng
- Ningxia Key Laboratory of Cerebrocranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, PR China.,Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, PR China
| | - Juan Du
- Ningxia Key Laboratory of Cerebrocranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, PR China.,Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, PR China
| | - Ning Liu
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, PR China
| | - Yuanyuan Qiang
- Ningxia Key Laboratory of Cerebrocranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, PR China
| | - Lifei Xiao
- Ningxia Key Laboratory of Cerebrocranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, PR China
| | - Xiaobing Lan
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, PR China
| | - Lin Ma
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, PR China
| | - Jiamei Yang
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, PR China
| | - Jianqiang Yu
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, PR China
| | - Guangyuan Lu
- Ningxia Key Laboratory of Cerebrocranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, PR China
| |
Collapse
|
4
|
Spampinato SF, Takeshita Y, Obermeier B. An In Vitro Model of the Blood-Brain Barrier to Study Alzheimer's Disease: The Role of β-Amyloid and Its Influence on PBMC Infiltration. Methods Mol Biol 2022; 2492:333-352. [PMID: 35733055 DOI: 10.1007/978-1-0716-2289-6_20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The blood-brain barrier (BBB) is a highly specialized structure, constituted by endothelial cells that together with astrocytes and pericytes provide a functional interface between the central nervous system and the periphery. Several pathological conditions may affect its functions, and lately BBB involvement in the pathogenesis of Alzheimer's disease has been demonstrated. Both endothelial cells and astrocytes can be differentially affected during the course of the disease. In vitro BBB models present a powerful tool in evaluating the effects that β-amyloid (Aβ), or other pathogenic stimuli, play on the BBB at cellular level. In vitro BBB models derived from human cell sources are rare and not easily implemented. We generated two conditionally immortalized human cell lines, brain microvascular endothelial cells (TY10), and astrocytes (hAST), that, when co-cultured under appropriate conditions, exhibit BBB-like characteristics. This model allowed us to evaluate the transmigration of peripheral blood mononuclear cells (PBMCs) through the in vitro barrier exposed to Aβ and the role played by astrocytes in the modulation of this phenomenon. We describe here the methodology used in our lab to set up our in vitro model of the BBB and to carry out a PBMC transmigration assay.
Collapse
Affiliation(s)
- Simona Federica Spampinato
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy.
- Departement of Scienza e Tecnologia del Farmaco, Universita' di Turin, Turin, Italy.
| | - Yukio Takeshita
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | | |
Collapse
|
5
|
Li X, Yu Y, Wei R, Li Y, Lv J, Liu Z, Zhang Y. In vitro and in vivo study on angiogenesis of porcine induced pluripotent stem cell-derived endothelial cells. Differentiation 2021; 120:10-18. [PMID: 34116291 DOI: 10.1016/j.diff.2021.05.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/16/2021] [Accepted: 05/29/2021] [Indexed: 10/21/2022]
Abstract
Pluripotent stem cells (PSCs) are a promising source of endothelial cells (ECs) for the treatment of cardiovascular diseases. Since clinical application of embryo stem cells (ESCs) involves issues of medical ethics and risk of immune rejection, induced pluripotent stem cells (iPSCs) will facilitate cell transplantation therapy for the cardiovascular diseases. Swine is identified as an ideal large-animal model for human, because of its similar organ size and physiological characteristics. However, there are very few studies on EC differentiation of porcine iPSCs (piPSCs). In recent study, we provided an efficient protocol to differentiate piPSCs into ECs with the purity of 19.76% CD31 positive cells within 16 days. Passaging of these cells yielded a nearly pure population, which also expressed other endothelial markers such as CD144, eNOS and vWF. Besides, these cells exhibited functions of ECs such as uptake of low-density lipoprotein and formation of tubes in vitro or blood vessels in vivo. Our study successfully obtained ECs from piPSCs via a feeder- and serum-free monolayer system and demonstrated their angiogenic function in vivo and in vitro. piPSC-ECs derivation is not only potential for the autologous cell transplantation and cardiovascular drug screening, but also for the mechanistic studies on EC differentiation and endothelial dysfunction.
Collapse
Affiliation(s)
- Xuechun Li
- College of Life Science, Northeast Agricultural University, Harbin, 150030, PR China
| | - Yang Yu
- College of Life Science, Northeast Agricultural University, Harbin, 150030, PR China
| | - Renyue Wei
- College of Life Science, Northeast Agricultural University, Harbin, 150030, PR China
| | - Yimei Li
- College of Life Science, Northeast Agricultural University, Harbin, 150030, PR China
| | - Jiawei Lv
- College of Life Science, Northeast Agricultural University, Harbin, 150030, PR China
| | - Zhonghua Liu
- College of Life Science, Northeast Agricultural University, Harbin, 150030, PR China.
| | - Yu Zhang
- College of Life Science, Northeast Agricultural University, Harbin, 150030, PR China.
| |
Collapse
|
6
|
Tien (田婷怡) TY, Wu (吳懿哲) YJ, Su (蘇正煌) CH, Wang (王學孝) HH, Hsieh (謝金玲) CL, Wang (王博正) BJ, Su (蘇瑀) Y, Yeh (葉宏一) HI. Reduction of Connexin 43 Attenuates Angiogenic Effects of Human Smooth Muscle Progenitor Cells via Inactivation of Akt and NF-κB Pathway. Arterioscler Thromb Vasc Biol 2021; 41:915-930. [PMID: 33356390 DOI: 10.1161/atvbaha.120.315650] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Circulating progenitor cells possess vasculogenesis property and participate in repair of vascular injury. Cx (connexin) 43-a transmembrane protein constituting gap junctions-is involved in vascular pathology. However, the role of Cx43 in smooth muscle progenitor cells (SPCs) remained unclear. Approach and Results: Human SPCs cultured from CD34+ peripheral blood mononuclear cells expressed smooth muscle cell markers, such as smooth muscle MHC (myosin heavy chain), nonmuscle MHC, calponin, and CD140B, and Cx43 was the most abundant Cx isoform. To evaluate the role of Cx43 in SPCs, short interference RNA was used to knock down Cx43 expression. Cellular activities of SPCs were reduced by Cx43 downregulation. In addition, Cx43 downregulation attenuated angiogenic potential of SPCs in hind limb ischemia mice. Protein array and ELISA of the supernatant from SPCs showed that IL (interleukin)-6, IL-8, and HGF (hepatocyte growth factor) were reduced by Cx43 downregulation. Simultaneously, Cx43 downregulation reduced the phosphorylation of NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) and Akt (protein kinase B) pathway and reactivation of NF-κB and Akt using betulinic acid, and SC79 could restore the secretion of growth factors and cytokines. Moreover, FAK (focal adhesion kinase)-Src (proto-oncogene tyrosine-protein kinase Src) activation was increased by Cx43 downregulation, and inactivation of Akt-NF-κB could be restored by Src inhibitor (PP2), indicating that Akt-NF-κB inactivated by Cx43 downregulation arose from FAK-Src activation. Finally, the depressed cellular activities and secretion of SPCs after Cx43 downregulation were restored by FAK inhibitor PF-562271 or PP2. CONCLUSIONS SPCs possess angiogenic potential to repair ischemic tissue mainly through paracrine effects. Gap junction protein Cx43 plays an important role in regulating cellular function and paracrine effects of SPCs through FAK-Src axis.
Collapse
Affiliation(s)
- Ting-Yi Tien (田婷怡)
- Department of Medical Research (T.-Y.T., C.-L.H., B.-J.W.), MacKay Memorial Hospital, Taipei, Taiwan
- Institute of Biopharmaceutical Science/National Yang-Ming University, Taipei, Taiwan (T.-Y.T., Y.S.)
| | - Yih-Jer Wu (吳懿哲)
- Department of Internal Medicine (Y.-J.W., C.-H.S., H.-I.Y.), MacKay Memorial Hospital, Taipei, Taiwan
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan (Y.-J.W., C.-H.S., H.-H.W.)
| | - Cheng-Huang Su (蘇正煌)
- Department of Internal Medicine (Y.-J.W., C.-H.S., H.-I.Y.), MacKay Memorial Hospital, Taipei, Taiwan
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan (Y.-J.W., C.-H.S., H.-H.W.)
| | - Hsueh-Hsiao Wang (王學孝)
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan (Y.-J.W., C.-H.S., H.-H.W.)
| | - Chin-Ling Hsieh (謝金玲)
- Department of Medical Research (T.-Y.T., C.-L.H., B.-J.W.), MacKay Memorial Hospital, Taipei, Taiwan
| | - Bo-Jeng Wang (王博正)
- Department of Medical Research (T.-Y.T., C.-L.H., B.-J.W.), MacKay Memorial Hospital, Taipei, Taiwan
| | - Yeu Su (蘇瑀)
- Institute of Biopharmaceutical Science/National Yang-Ming University, Taipei, Taiwan (T.-Y.T., Y.S.)
| | - Hung-I. Yeh (葉宏一)
- Department of Internal Medicine (Y.-J.W., C.-H.S., H.-I.Y.), MacKay Memorial Hospital, Taipei, Taiwan
| |
Collapse
|
7
|
Habu T, Harada KH. UBC13 is an RNF213-associated E2 ubiquitin-conjugating enzyme, and Lysine 63-linked ubiquitination by the RNF213-UBC13 axis is responsible for angiogenic activity. FASEB Bioadv 2021; 3:243-258. [PMID: 33842849 PMCID: PMC8019261 DOI: 10.1096/fba.2019-00092] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 12/26/2020] [Accepted: 12/30/2020] [Indexed: 12/13/2022] Open
Abstract
Moyamoya disease (MMD) is a cryptogenic vascular disorder in the intracranial arteries. RING protein 213 (RNF213) is the susceptibility gene for MMD, and encodes a RING domain and a Walker motif. Herein, we identified UBC13 (UBE2N) as an E2 ubiquitin‐conjugating enzyme for RNF213 E3 ubiquitin ligase by yeast two‐hybrid screening with a fragment containing RNF213 RING domain as bait, and the immunocomplex of RNF213‐UBC13 was detected in vivo. Analysis of the ubiquitin chain on RNF213 by monitoring autoubiquitination showed that RNF213 was autoubiquitinated in a K63 chain fashion, but not in a K48 chain fashion. Finally, this RNF213 ubiquitination in a UBC13‐dependent manner was required for cell mobility and invasion activity for HUVEC cells in UBC13 knock‐down and ubiquitination‐dead RNF213 mutant expressing experiments. These findings demonstrated that RNF213 is a K63‐linked E3 ubiquitin ligase, and UBC13 is responsible for RNF213 dependent ubiquitination. The RNF213‐UBC13 axis may be associated with angiogenic activity and MMD.
Collapse
Affiliation(s)
- Toshiyuki Habu
- Department of Food Sciences and Nutrition School of Food Sciences and Nutrition Mukogawa Women's University Nishinomiya Hyogo Japan
| | - Kouji H Harada
- Department of Health and Environmental Sciences Kyoto University Graduate School of Medicine Kyoto Japan
| |
Collapse
|
8
|
Modulating hESC-derived cardiomyocyte and endothelial cell function with triple-helical peptides for heart tissue engineering. Biomaterials 2020; 269:120612. [PMID: 33385684 PMCID: PMC7884910 DOI: 10.1016/j.biomaterials.2020.120612] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 12/14/2020] [Indexed: 11/21/2022]
Abstract
In this study, we investigated the role of cardiomyocyte (CM) and endothelial cell (EC) specific interactions with collagen in the assembly of an operational myocardium in vitro. Engineered cardiac patches represent valuable tools for myocardial repair following infarction and are generally constituted of a suitable biomaterial populated by CMs and supportive cell types. Among those, ECs are required for tissue vascularization and positively modulate CM function. To direct the function of human embryonic stem cell (hESC)-derived CM and EC seeded on biomaterials, we replicated cell-collagen interactions, which regulate cellular behaviour in the native myocardium, using triple-helical peptides (THPs) that are ligands for collagen-binding proteins. THPs enhanced proliferation and activity of CMs and ECs separately and in co-culture, drove CM maturation and enabled coordinated cellular contraction on collagen films. These results highlight the importance of collagen interactions on cellular response and establish THP-functionalized biomaterials as novel tools to produce engineered cardiac tissues.
Collapse
|
9
|
Taura D, Nakao K, Nakagawa Y, Kinoshita H, Sone M, Nakao K. C-type natriuretic peptide (CNP)/guanylate cyclase B (GC-B) system and endothelin-1(ET-1)/ET receptor A and B system in human vasculature. Can J Physiol Pharmacol 2020; 98:611-617. [PMID: 32268070 DOI: 10.1139/cjpp-2019-0686] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
To assess the physiological and clinical implications of the C-type natriuretic peptide (CNP)/guanylyl cyclase B (GC-B) system in the human vasculature, we have examined gene expressions of CNP and its receptor, GC-B, in human vascular endothelial cells (ECs) and smooth muscle cells (SMCs) and have also compared the endothelin-1(ET-1)/endothelin receptor-A (ETR-A) and endothelin receptor-B (ETR-B) system in human aortic ECs (HAECs) and vascular SMCs (HSMCs) in vitro. We also examined these gene expressions in human embryonic stem (ES)/induced pluripotent stem cell (iPS)-derived ECs and mural cells (MCs). A little but significant amount of mRNA encoding CNP was detected in both human ES-derived ECs and HAECs. A substantial amount of GC-B was expressed in both ECs (iPS-derived ECs and HAECs) and SMCs (iPS-derived MCs and HSMCs). ET-1 was expressed solely in ECs. ETR-A was expressed in SMCs, while ETR-B was expressed in ECs. These results indicate the existence of a vascular CNP/GC-B system in the human vascular wall, indicating the evidence for clinical implication of the CNP/GC-B system in concert with the ET-1/ETR-A and ETR-B system in the human vasculature.
Collapse
Affiliation(s)
- Daisuke Taura
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kazuhiro Nakao
- National Cardiovascular, Cerebrovascular Research Center Hospital, Suita, Japan
| | - Yasuaki Nakagawa
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hideyuki Kinoshita
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masakatsu Sone
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kazuwa Nakao
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
10
|
Arora S, Yim EKF, Toh YC. Environmental Specification of Pluripotent Stem Cell Derived Endothelial Cells Toward Arterial and Venous Subtypes. Front Bioeng Biotechnol 2019; 7:143. [PMID: 31259171 PMCID: PMC6587665 DOI: 10.3389/fbioe.2019.00143] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 05/28/2019] [Indexed: 12/25/2022] Open
Abstract
Endothelial cells (ECs) are required for a multitude of cardiovascular clinical applications, such as revascularization of ischemic tissues or endothelialization of tissue engineered grafts. Patient derived primary ECs are limited in number, have donor variabilities and their in vitro phenotypes and functions can deteriorate over time. This necessitates the exploration of alternative EC sources. Although there has been a recent surge in the use of pluripotent stem cell derived endothelial cells (PSC-ECs) for various cardiovascular clinical applications, current differentiation protocols yield a heterogeneous EC population, where their specification into arterial or venous subtypes is undefined. Since arterial and venous ECs are phenotypically and functionally different, inappropriate matching of exogenous ECs to host sites can potentially affect clinical efficacy, as exemplified by venous graft mismatch when placed into an arterial environment. Therefore, there is a need to design and employ environmental cues that can effectively modulate PSC-ECs into a more homogeneous arterial or venous phenotype for better adaptation to the host environment, which will in turn contribute to better application efficacy. In this review, we will first give an overview of the developmental and functional differences between arterial and venous ECs. This provides the foundation for our subsequent discussion on the different bioengineering strategies that have been investigated to varying extent in providing biochemical and biophysical environmental cues to mature PSC-ECs into arterial or venous subtypes. The ability to efficiently leverage on a combination of biochemical and biophysical environmental cues to modulate intrinsic arterio-venous specification programs in ECs will greatly facilitate future translational applications of PSC-ECs. Since the development and maintenance of arterial and venous ECs in vivo occur in disparate physio-chemical microenvironments, it is conceivable that the application of these environmental factors in customized combinations or magnitudes can be used to selectively mature PSC-ECs into an arterial or venous subtype.
Collapse
Affiliation(s)
- Seep Arora
- Department of Biomedical Engineering, National University of Singapore, Singapore, Singapore.,Singapore Institute for Neurotechnology (SINAPSE), National University of Singapore, Singapore, Singapore
| | - Evelyn K F Yim
- Department of Chemical Engineering, University of Waterloo, Waterloo, ON, Canada
| | - Yi-Chin Toh
- Department of Biomedical Engineering, National University of Singapore, Singapore, Singapore.,Singapore Institute for Neurotechnology (SINAPSE), National University of Singapore, Singapore, Singapore.,Biomedical Institute for Global Health Research and Technology (BIGHEART), National University of Singapore, Singapore, Singapore.,NUS Tissue Engineering Program, National University of Singapore, Singapore, Singapore
| |
Collapse
|
11
|
Williams IM, Wu JC. Generation of Endothelial Cells From Human Pluripotent Stem Cells. Arterioscler Thromb Vasc Biol 2019; 39:1317-1329. [PMID: 31242035 DOI: 10.1161/atvbaha.119.312265] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Endothelial cells (ECs) are critical for several aspects of cardiovascular disease therapy, including vascular regeneration, personalized drug development, and tissue engineering. Human pluripotent stem cells (hPSCs) afford us with an unprecedented opportunity to produce virtually unlimited quantities of human ECs. In this review, we highlight key developments and outstanding challenges in our ability to derive ECs de novo from hPSCs. Furthermore, we consider strategies for recapitulating the vessel- and tissue-specific functional heterogeneity of ECs in vitro. Finally, we discuss ongoing attempts to utilize hPSC-derived ECs and their progenitors for various therapeutic applications. Continued progress in generating hPSC-derived ECs will profoundly enhance our ability to discover novel drug targets, revascularize ischemic tissues, and engineer clinically relevant tissue constructs. Visual Overview- An online visual overview is available for this article.
Collapse
Affiliation(s)
- Ian M Williams
- From the Stanford Cardiovascular Institute, Division of Cardiovascular Medicine, Department of Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, CA
| | - Joseph C Wu
- From the Stanford Cardiovascular Institute, Division of Cardiovascular Medicine, Department of Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, CA
| |
Collapse
|
12
|
Ong SB, Lee WH, Shao NY, Ismail NI, Katwadi K, Lim MM, Kwek XY, Michel NA, Li J, Newson J, Tahmasebi S, Rehman J, Kodo K, Jang HR, Ong SG. Calpain Inhibition Restores Autophagy and Prevents Mitochondrial Fragmentation in a Human iPSC Model of Diabetic Endotheliopathy. Stem Cell Reports 2019; 12:597-610. [PMID: 30799273 PMCID: PMC6411483 DOI: 10.1016/j.stemcr.2019.01.017] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 01/17/2019] [Accepted: 01/17/2019] [Indexed: 02/09/2023] Open
Abstract
The relationship between diabetes and endothelial dysfunction remains unclear, particularly the association with pathological activation of calpain, an intracellular cysteine protease. Here, we used human induced pluripotent stem cells-derived endothelial cells (iPSC-ECs) to investigate the effects of diabetes on vascular health. Our results indicate that iPSC-ECs exposed to hyperglycemia had impaired autophagy, increased mitochondria fragmentation, and was associated with increased calpain activity. In addition, hyperglycemic iPSC-ECs had increased susceptibility to cell death when subjected to a secondary insult-simulated ischemia-reperfusion injury (sIRI). Importantly, calpain inhibition restored autophagy and reduced mitochondrial fragmentation, concurrent with maintenance of ATP production, normalized reactive oxygen species levels and reduced susceptibility to sIRI. Using a human iPSC model of diabetic endotheliopathy, we demonstrated that restoration of autophagy and prevention of mitochondrial fragmentation via calpain inhibition improves vascular integrity. Our human iPSC-EC model thus represents a valuable platform to explore biological mechanisms and new treatments for diabetes-induced endothelial dysfunction.
Collapse
Affiliation(s)
- Sang-Bing Ong
- Signature Research Program in Cardiovascular & Metabolic Disorders, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Won Hee Lee
- Department of Basic Medical Sciences, University of Arizona, Phoenix, AZ 85004, USA
| | - Ning-Yi Shao
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Nur Izzah Ismail
- Signature Research Program in Cardiovascular & Metabolic Disorders, Duke-NUS Medical School, Singapore 169857, Singapore; Department of Biomedical Engineering, Faculty of Engineering, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Khairunnisa Katwadi
- Signature Research Program in Cardiovascular & Metabolic Disorders, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Mim-Mim Lim
- Signature Research Program in Cardiovascular & Metabolic Disorders, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Xiu-Yi Kwek
- Signature Research Program in Cardiovascular & Metabolic Disorders, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Nathaly Anto Michel
- Signature Research Program in Cardiovascular & Metabolic Disorders, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Jiajun Li
- Department of Pharmacology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Jordan Newson
- Department of Pharmacology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Soroush Tahmasebi
- Department of Pharmacology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Jalees Rehman
- Department of Pharmacology, The University of Illinois College of Medicine, Chicago, IL 60612, USA; Division of Cardiology, Department of Medicine, The University of Illinois College of Medicine, 909 S Wolcott Avenue, Chicago, IL 60612, USA
| | - Kazuki Kodo
- Department of Pediatrics, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Hye Ryoun Jang
- Division of Nephrology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351 Republic of Korea.
| | - Sang-Ging Ong
- Department of Pharmacology, The University of Illinois College of Medicine, Chicago, IL 60612, USA; Division of Cardiology, Department of Medicine, The University of Illinois College of Medicine, 909 S Wolcott Avenue, Chicago, IL 60612, USA.
| |
Collapse
|
13
|
Masuda S, Matsuura K, Shimizu T. Preparation of iPS cell-derived CD31 + endothelial cells using three-dimensional suspension culture. Regen Ther 2018; 9:1-9. [PMID: 30525069 PMCID: PMC6222294 DOI: 10.1016/j.reth.2018.06.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 05/24/2018] [Accepted: 06/22/2018] [Indexed: 12/03/2022] Open
Abstract
A well-organised vascular network is essential for metabolic exchange to maintain homoeostasis in the body. Therefore, for progress in regenerative medicine, it is particularly important to establish methods of vascularization in bioengineered three-dimensional (3D) functional tissues. In addition, it is necessary to develop methods to supply a large number of iPS cell-derived endothelial cells for fabricating the vascular network structure. There are already many reports on the method of inducing the differentiation of endothelial cells from iPS cells using 2D culture. However, there are few reports on methods for preparing a large number of iPS cell-derived endothelial cells. Therefore, we developed methods for inducing vascular endothelial cells from human inducible pluripotent stem (hiPS) cells using 3D suspension culture. hiPS cell-derived CD31+ cells expressed several endothelial marker genes and formed endothelial cell network structures, similar to human umbilical vein endothelial cells. These results indicate that hiPS cell-derived CD31+ cells may be a useful cell source for pre-vascularised network structures in 3D functional tissues, and it is important to develop 3D mass culture system for preparing a large number of cells to fabricate bioengineered tissues.
Collapse
Affiliation(s)
- Shinako Masuda
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku, Tokyo 162-8666, Japan
| | - Katsuhisa Matsuura
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku, Tokyo 162-8666, Japan
- Department of Cardiology, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku, Tokyo 162-8666, Japan
| | - Tatsuya Shimizu
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku, Tokyo 162-8666, Japan
| |
Collapse
|
14
|
Human very Small Embryonic-like Cells Support Vascular Maturation and Therapeutic Revascularization Induced by Endothelial Progenitor Cells. Stem Cell Rev Rep 2018; 13:552-560. [PMID: 28303468 DOI: 10.1007/s12015-017-9731-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Very small embryonic-like stem cells (VSELs) are major pluripotent stem cells defined as cells of small size being Lineage- negative, CD133-positive, and CD45-negative. We previously described that human bone marrow VSELs were able to differentiate into endothelial cells and promoted post-ischemic revascularization in mice with surgically induced critical limb ischemia. In the present work, we isolated bone marrow VSELs from patients with critical limb ischemia and studied their ability to support endothelial progenitor cells therapeutic capacity and revascularization potential. Sorted bone marrow VSELs cultured in angiogenic media were co-injected with endothelial progenitor cells and have been show to trigger post-ischemic revascularization in immunodeficient mice, and support vessel formation in vivo in Matrigel implants better than human bone marrow mesenchymal stem cells. In conclusion, VSELs are a potential new source of therapeutic cells that may give rise to cells of the endothelial and perivascular lineage in humans. VSELs are the first real vasculogenic stem cells able to differentiate in endothelial and perivascular lineage in human adult described from now. Thus, because VSELs presence have been proposed in adult tissues, we think that VSELs are CD45 negative stem cells able to give rise to vascular regeneration in human tissues and vessels.
Collapse
|
15
|
Analysis of mitochondrial function in human induced pluripotent stem cells from patients with mitochondrial diabetes due to the A3243G mutation. Sci Rep 2018; 8:949. [PMID: 29343702 PMCID: PMC5772054 DOI: 10.1038/s41598-018-19264-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 12/16/2017] [Indexed: 01/19/2023] Open
Abstract
We previously established human induced pluripotent stem (iPS) cells in two diabetic patients from different families with the mitochondrial A3243G mutation and isolated isogenic iPS cell clones with either undetectable or high levels of the mutation in both patients. In the present study, we analyzed the mitochondrial functions of two mutation-undetectable and two mutation-high clones in each patient through four methods to assess complex I activity, mitochondrial membrane potential, mitochondrial respiration, and mitochondrial ATP production. In the first patient, complex I activity, mitochondrial respiration, and mitochondrial ATP production were decreased in the mutation-high clones compared with the mutation-undetectable clones, and mitochondrial membrane potential was decreased in a mutation-high clone compared with a mutation-undetectable clone. In the second patient, complex I activity was decreased in one mutation-high clone compared with the other clones. The other parameters showed no differences in any clones. In addition, the complex I activity and mitochondrial respiration of the mutation-undetectable clones from both patients were located in the range of those of iPS cells from healthy subjects. The present study suggests that the mitochondrial function of the mutation-undetectable iPS cell clones obtained from two patients with the A3243G mutation is comparable to the control iPS cells.
Collapse
|
16
|
Banno K, Yoder MC. Tissue regeneration using endothelial colony-forming cells: promising cells for vascular repair. Pediatr Res 2018; 83:283-290. [PMID: 28915234 DOI: 10.1038/pr.2017.231] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 07/07/2017] [Indexed: 12/24/2022]
Abstract
Repairing and rebuilding damaged tissue in diseased human subjects remains a daunting challenge for clinical medicine. Proper vascular formation that serves to deliver blood-borne nutrients and adequate levels of oxygen and to remove wastes is critical for successful tissue regeneration. Endothelial colony-forming cells (ECFC) represent a promising cell source for revascularization of damaged tissue. ECFCs are identified by displaying a hierarchy of clonal proliferative potential and by pronounced postnatal vascularization ability in vivo. In this review, we provide a brief overview of human ECFC isolation and characterization, a survey of a number of animal models of human disease in which ECFCs have been shown to have prominent roles in tissue repair, and a summary of current challenges that must be overcome before moving ECFC into human subjects as a cell therapy.
Collapse
Affiliation(s)
- Kimihiko Banno
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Mervin C Yoder
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
17
|
Combinatorial Extracellular Matrix Microenvironments for Probing Endothelial Differentiation of Human Pluripotent Stem Cells. Sci Rep 2017; 7:6551. [PMID: 28747756 PMCID: PMC5529516 DOI: 10.1038/s41598-017-06986-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 06/21/2017] [Indexed: 12/02/2022] Open
Abstract
Endothelial cells derived from human pluripotent stem cells are a promising cell type for enhancing angiogenesis in ischemic cardiovascular tissues. However, our understanding of microenvironmental factors that modulate the process of endothelial differentiation is limited. We examined the role of combinatorial extracellular matrix (ECM) proteins on endothelial differentiation systematically using an arrayed microscale platform. Human pluripotent stem cells were differentiated on the arrayed ECM microenvironments for 5 days. Combinatorial ECMs composed of collagen IV + heparan sulfate + laminin (CHL) or collagen IV + gelatin + heparan sulfate (CGH) demonstrated significantly higher expression of CD31, compared to single-factor ECMs. These results were corroborated by fluorescence activated cell sorting showing a 48% yield of CD31+/VE-cadherin+ cells on CHL, compared to 27% on matrigel. To elucidate the signaling mechanism, a gene expression time course revealed that VE-cadherin and FLK1 were upregulated in a dynamically similar manner as integrin subunit β3 (>50 fold). To demonstrate the functional importance of integrin β3 in promoting endothelial differentiation, the addition of neutralization antibody inhibited endothelial differentiation on CHL-modified dishes by >50%. These data suggest that optimal combinatorial ECMs enhance endothelial differentiation, compared to many single-factor ECMs, in part through an integrin β3-mediated pathway.
Collapse
|
18
|
Liang J, Huang W, Cai W, Wang L, Guo L, Paul C, Yu XY, Wang Y. Inhibition of microRNA-495 Enhances Therapeutic Angiogenesis of Human Induced Pluripotent Stem Cells. Stem Cells 2017; 35:337-350. [DOI: 10.1002/stem.2477] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Abstract
Therapeutic angiogenesis has emerged as a promising strategy to regenerate the damaged blood vessels resulting from ischemic diseases such as myocardial infarction (MI). However, the functional integration of implanted endothelial cells (ECs) in infarcted heart remains challenging. We herein develop an EC generation approach by inhibiting microRNA-495 (miR-495) in human induced pluripotent stem cells (hiPSCs) and assess the angiogenic potential for MI treatment. The anti-angiogenic miR-495 belonging to Dlk1-Dio3 miR cluster was identified through expression profiling and computational analysis. Loss-of-function experiments for miR-495 were performed using a lentiviral transfer of antisense sequence in hiPSCs. The pluripotency of hiPSCs was not impacted by the genetic modification. Induced with differentiation medium, miR-495 inhibition enhanced the expression of EC genes of hiPSCs, as well as the yield of ECs. Newly derived ECs displayed prominent angiogenic characteristics including tube formation, cell migration, and proliferation. Mechanistically, miR-495 mediated the expression of endothelial or angiogenic genes by directly targeting vascular endothelial zinc finger 1. After transplantation in immunodeficient MI mice, the derived ECs significantly increased neovascularization in the infarcted heart, prevented functional worsening, and attenuated expansion of infarct size. The functional integration of the implanted ECs into coronary networks was also enhanced by inhibiting miR-495. miR-495 represents a new target not only for promoting EC generation from hiPSCs but also for enhancing angiogenesis and engraftment of hiPSC-derived ECs in ischemic heart.
Collapse
Affiliation(s)
- Jialiang Liang
- a Departments of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio, USA
| | - Wei Huang
- a Departments of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio, USA
| | - Wenfeng Cai
- a Departments of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio, USA
| | - Lei Wang
- a Departments of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio, USA
| | - Linlin Guo
- a Departments of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio, USA
| | - Christian Paul
- a Departments of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio, USA
| | - Xi-Yong Yu
- b Institute of Molecular and Clinical Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yigang Wang
- a Departments of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
19
|
Yamaguchi S, Morizane R, Homma K, Monkawa T, Suzuki S, Fujii S, Koda M, Hiratsuka K, Yamashita M, Yoshida T, Wakino S, Hayashi K, Sasaki J, Hori S, Itoh H. Generation of kidney tubular organoids from human pluripotent stem cells. Sci Rep 2016; 6:38353. [PMID: 27982115 PMCID: PMC5159864 DOI: 10.1038/srep38353] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 11/08/2016] [Indexed: 12/12/2022] Open
Abstract
Recent advances in stem cell research have resulted in methods to generate kidney organoids from human pluripotent stem cells (hPSCs), which contain cells of multiple lineages including nephron epithelial cells. Methods to purify specific types of cells from differentiated hPSCs, however, have not been established well. For bioengineering, cell transplantation, and disease modeling, it would be useful to establish those methods to obtain pure populations of specific types of kidney cells. Here, we report a simple two-step differentiation protocol to generate kidney tubular organoids from hPSCs with direct purification of KSP (kidney specific protein)-positive cells using anti-KSP antibody. We first differentiated hPSCs into mesoderm cells using a glycogen synthase kinase-3β inhibitor for 3 days, then cultured cells in renal epithelial growth medium to induce KSP+ cells. We purified KSP+ cells using flow cytometry with anti-KSP antibody, which exhibited characteristics of all segments of kidney tubular cells and cultured KSP+ cells in 3D Matrigel, which formed tubular organoids in vitro. The formation of tubular organoids by KSP+ cells induced the acquisition of functional kidney tubules. KSP+ cells also allowed for the generation of chimeric kidney cultures in which human cells self-assembled into 3D tubular structures in combination with mouse embryonic kidney cells.
Collapse
Affiliation(s)
- Shintaro Yamaguchi
- Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Ryuji Morizane
- Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan.,Renal Division, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, USA.,Department of Medicine, Harvard Medical School, 25 Shattuck St, Boston, MA 02115, USA.,Harvard Stem Cell Institute, 7 Divinity Ave, Cambridge, MA 02138, USA
| | - Koichiro Homma
- Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan.,Emergency and Critical Care Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Toshiaki Monkawa
- Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan.,Medical Education Center, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Sayuri Suzuki
- Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Shizuka Fujii
- Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Muneaki Koda
- Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Ken Hiratsuka
- Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Maho Yamashita
- Apheresis and Dialysis Center, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Tadashi Yoshida
- Apheresis and Dialysis Center, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Shu Wakino
- Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Koichi Hayashi
- Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Junichi Sasaki
- Emergency and Critical Care Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Shingo Hori
- Emergency and Critical Care Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Hiroshi Itoh
- Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| |
Collapse
|
20
|
Glaser DE, Turner WS, Madfis N, Wong L, Zamora J, White N, Reyes S, Burns AB, Gopinathan A, McCloskey KE. Multifactorial Optimizations for Directing Endothelial Fate from Stem Cells. PLoS One 2016; 11:e0166663. [PMID: 27907001 PMCID: PMC5131944 DOI: 10.1371/journal.pone.0166663] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 11/01/2016] [Indexed: 01/08/2023] Open
Abstract
Embryonic stem cells (ESC) and induced pluripotent stem (iPS) cells are attractive in vitro models of vascular development, therapeutic angiogenesis, and tissue engineering. However, distinct ESC and iPS cell lines respond differentially to the same microenvironmental factors. Developing improved/optimized differentiation methodologies tailored/applicable in a number of distinct iPS and ESC lines remains a challenge in the field. Currently published methods for deriving endothelial cells (EC) robustly generate high numbers of endothlelial progenitor cells (EPC) within a week, but their maturation to definitive EC is much more difficult, taking up to 2 months and requiring additional purification. Therefore, we set out to examine combinations/levels of putative EC induction factors—utilizing our stage-specific chemically-defined derivation methodology in 4 ESC lines including: kinetics, cell seeding density, matrix signaling, as well as medium treatment with vascular endothelial growth factor (VEGF), and basic fibroblast growth factor (bFGF). The results indicate that temporal development in both early and late stages is the most significant factor generating the desired cells. The generation of early Flk-1+/KDR+ vascular progenitor cells (VPC) from pluripotent ESC is directed predominantly by high cell seeding density and matrix signaling from fibronectin, while VEGF supplementation was NOT statistically significant in more than one cell line, especially with fibronectin matrix which sequesters autocrine VEGF production by the differentiating stem cells. Although some groups have shown that the GSK3-kinase inhibitor (CHIR) can facilitate EPC fate, it hindered the generation of KDR+ cells in our preoptimized medium formulations. The methods summarized here significantly increased the production of mature vascular endothelial (VE)-cadherin+ EC, with up to 93% and 57% purity from mouse and human ESC, respectively, before VE-cadherin+ EC purification.
Collapse
Affiliation(s)
- Drew E. Glaser
- School of Engineering, University of California, Merced, United States of America
- Graduate Program in Biological Engineering and Small-scale Technologies, University of California, Merced, United States of America
| | - William S. Turner
- School of Engineering, University of California, Merced, United States of America
| | - Nicole Madfis
- Graduate Program in Quantitative and Systems Biology, University of California, Merced, United States of America
| | - Lian Wong
- School of Engineering, University of California, Merced, United States of America
- Graduate Program in Biological Engineering and Small-scale Technologies, University of California, Merced, United States of America
| | - Jose Zamora
- Department of Physics, University of California, Merced, United States of America
- Department of Molecular and Cellular Biology, University of California, Merced, United States of America
| | - Nicholas White
- School of Engineering, University of California, Merced, United States of America
| | - Samuel Reyes
- School of Engineering, University of California, Merced, United States of America
| | - Andrew B. Burns
- Department of Molecular and Cellular Biology, University of California, Merced, United States of America
| | - Ajay Gopinathan
- Department of Physics, University of California, Merced, United States of America
| | - Kara E. McCloskey
- School of Engineering, University of California, Merced, United States of America
- Graduate Program in Biological Engineering and Small-scale Technologies, University of California, Merced, United States of America
- * E-mail:
| |
Collapse
|
21
|
Hou L, Coller J, Natu V, Hastie TJ, Huang NF. Combinatorial extracellular matrix microenvironments promote survival and phenotype of human induced pluripotent stem cell-derived endothelial cells in hypoxia. Acta Biomater 2016; 44:188-99. [PMID: 27498178 DOI: 10.1016/j.actbio.2016.08.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Revised: 07/14/2016] [Accepted: 08/03/2016] [Indexed: 10/21/2022]
Abstract
UNLABELLED Recent developments in cell therapy using human induced pluripotent stem cell-derived endothelial cells (iPSC-ECs) hold great promise for treating ischemic cardiovascular tissues. However, poor post-transplantation viability largely limits the potential of stem cell therapy. Although the extracellular matrix (ECM) has become increasingly recognized as an important cell survival factor, conventional approaches primarily rely on single ECMs for in vivo co-delivery with cells, even though the endothelial basement membrane is comprised of a milieu of different ECMs. To address this limitation, we developed a combinatorial ECM microarray platform to simultaneously interrogate hundreds of micro-scale multi-component chemical compositions of ECMs on iPSC-EC response. After seeding iPSC-ECs onto ECM microarrays, we performed high-throughput analysis of the effects of combinatorial ECMs on iPSC-EC survival, endothelial phenotype, and nitric oxide production under conditions of hypoxia (1% O2) and reduced nutrients (1% fetal bovine serum), as is present in ischemic injury sites. Using automated image acquisition and analysis, we identified combinatorial ECMs such as collagen IV+gelatin+heparan sulfate+laminin and collagen IV+fibronectin+gelatin+heparan sulfate+laminin that significantly improved cell survival, nitric oxide production, and CD31 phenotypic expression, in comparison to single-component ECMs. These results were further validated in conventional cell culture platforms and within three-dimensional scaffolds. Furthermore, this approach revealed complex ECM interactions and non-intuitive cell behavior that otherwise could not be easily determined using conventional cell culture platforms. Together these data suggested that iPSC-EC delivery within optimal combinatorial ECMs may improve their survival and function under the condition of hypoxia with reduced nutrients. STATEMENT OF SIGNIFICANCE Human endothelial cells (ECs) derived from induced pluripotent stem cells (iPSC-ECs) are promising for treating diseases associated with reduced nutrient and oxygen supply like heart failure. However, diminished iPSC-EC survival after implantation into diseased environments limits their therapeutic potential. Since native ECs interact with numerous extracellular matrix (ECM) proteins for functional maintenance, we hypothesized that combinatorial ECMs may improve cell survival and function under conditions of reduced oxygen and nutrients. We developed a high-throughput system for simultaneous screening of iPSC-ECs cultured on multi-component ECM combinations under the condition of hypoxia and reduced serum. Using automated image acquisition and analytical algorithms, we identified combinatorial ECMs that significantly improved cell survival and function, in comparison to single ECMs. Furthermore, this approach revealed complex ECM interactions and non-intuitive cell behavior that otherwise could not be easily determined.
Collapse
|
22
|
Abstract
Stem cell therapy is a promising therapeutic option for severe cardiac diseases that are resistant to conventional therapies. To overcome the unsatisfactory results of most clinical researches on stem cell injections to an injured heart, we are developing bioengineered cardiac tissue grafts using pluripotent stem cell-derived cardiomyocytes and vascular cells. We have validated the functional benefits of mouse embryonic stem cell-derived and human induced pluripotent stem cell-derived cardiac tissue sheets (CTSs) in a rat myocardial infarction model. We further showed enhanced functional recovery and engraftment efficiency leading to de novo myocardium upon transplanting thick multi-layered CTSs that had gelatin hydrogel microspheres between the layers. We anticipate that the combination of pluripotent stem cell biology and tissue engineering will contribute to future stem cell therapies for severe heart diseases.
Collapse
|
23
|
Geng Y, Feng B. A small molecule-based strategy for endothelial differentiation and three-dimensional morphogenesis from human embryonic stem cells. Heliyon 2016; 2:e00133. [PMID: 27512727 PMCID: PMC4971129 DOI: 10.1016/j.heliyon.2016.e00133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Revised: 06/21/2016] [Accepted: 07/15/2016] [Indexed: 12/25/2022] Open
Abstract
The emerging models of human embryonic stem cell (hESC) self-organizing organoids provide a valuable in vitro platform for studying self-organizing processes that presumably mimic in vivo human developmental events. Here we report that through a chemical screen, we identified two novel and structurally similar small molecules BIR1 and BIR2 which robustly induced the self-organization of a balloon-shaped three-dimensional structure when applied to two-dimensional adherent hESC cultures in the absence of growth factors. Gene expression analyses and functional assays demonstrated an endothelial identity of this balloon-like structure, while cell surface marker analyses revealed a VE-cadherin(+)CD31(+)CD34(+)KDR(+)CD43(-) putative endothelial progenitor population. Furthermore, molecular marker labeling and morphological examinations characterized several other distinct DiI-Ac-LDL(+) multi-cellular modules and a VEGFR3(+) sprouting structure in the balloon cultures that likely represented intermediate structures of balloon-formation.
Collapse
Affiliation(s)
- Yijie Geng
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Bradley Feng
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
24
|
Identification of MMP1 as a novel risk factor for intracranial aneurysms in ADPKD using iPSC models. Sci Rep 2016; 6:30013. [PMID: 27418197 PMCID: PMC4945931 DOI: 10.1038/srep30013] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 06/29/2016] [Indexed: 11/08/2022] Open
Abstract
Cardiovascular complications are the leading cause of death in autosomal dominant polycystic kidney disease (ADPKD), and intracranial aneurysm (ICA) causing subarachnoid hemorrhage is among the most serious complications. The diagnostic and therapeutic strategies for ICAs in ADPKD have not been fully established. We here generated induced pluripotent stem cells (iPSCs) from seven ADPKD patients, including four with ICAs. The vascular cells differentiated from ADPKD-iPSCs showed altered Ca(2+) entry and gene expression profiles compared with those of iPSCs from non-ADPKD subjects. We found that the expression level of a metalloenzyme gene, matrix metalloproteinase (MMP) 1, was specifically elevated in iPSC-derived endothelia from ADPKD patients with ICAs. Furthermore, we confirmed the correlation between the serum MMP1 levels and the development of ICAs in 354 ADPKD patients, indicating that high serum MMP1 levels may be a novel risk factor. These results suggest that cellular disease models with ADPKD-specific iPSCs can be used to study the disease mechanisms and to identify novel disease-related molecules or risk factors.
Collapse
|
25
|
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: 38] [Impact Index Per Article: 3.8] [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
|
26
|
Rammal H, Harmouch C, Lataillade JJ, Laurent-Maquin D, Labrude P, Menu P, Kerdjoudj H. Stem cells: a promising source for vascular regenerative medicine. Stem Cells Dev 2015; 23:2931-49. [PMID: 25167472 DOI: 10.1089/scd.2014.0132] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The rising and diversity of many human vascular diseases pose urgent needs for the development of novel therapeutics. Stem cell therapy represents a challenge in the medicine of the twenty-first century, an area where tissue engineering and regenerative medicine gather to provide promising treatments for a wide variety of diseases. Indeed, with their extensive regeneration potential and functional multilineage differentiation capacity, stem cells are now highlighted as promising cell sources for regenerative medicine. Their multilineage differentiation involves environmental factors such as biochemical, extracellular matrix coating, oxygen tension, and mechanical forces. In this review, we will focus on human stem cell sources and their applications in vascular regeneration. We will also discuss the different strategies used for their differentiation into both mature and functional smooth muscle and endothelial cells.
Collapse
Affiliation(s)
- Hassan Rammal
- 1 UMR 7365, Biopôle, Faculté de Médecine, CNRS-Université de Lorraine , Vandœuvre-lès-Nancy, France
| | | | | | | | | | | | | |
Collapse
|
27
|
Shi N, Chen SY. Smooth Muscle Cell Differentiation: Model Systems, Regulatory Mechanisms, and Vascular Diseases. J Cell Physiol 2015; 231:777-87. [DOI: 10.1002/jcp.25208] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 09/29/2015] [Indexed: 02/06/2023]
Affiliation(s)
- Ning Shi
- Department of Physiology and Pharmacology; University of Georgia; Athens Georgia
| | - Shi-You Chen
- Department of Physiology and Pharmacology; University of Georgia; Athens Georgia
| |
Collapse
|
28
|
Xie N, Li Z, Adesanya TM, Guo W, Liu Y, Fu M, Kilic A, Tan T, Zhu H, Xie X. Transplantation of placenta-derived mesenchymal stem cells enhances angiogenesis after ischemic limb injury in mice. J Cell Mol Med 2015; 20:29-37. [PMID: 26282458 PMCID: PMC4717860 DOI: 10.1111/jcmm.12489] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 07/10/2014] [Indexed: 01/06/2023] Open
Abstract
Mesenchymal stem cell‐based therapy has emerged as a promising approach for the treatment of peripheral arterial disease. The purpose of this study was to examine the potential effects of human placenta‐derived mesenchymal stem cells (PMSCs) on mouse hindlimb ischemia. PMSCs were isolated from human placenta tissue and characterized by flow cytometry. An in vivo surgical ligation‐induced murine limb ischemia model was generated with fluorescent dye (CM‐DiI) labelled PMSCs delivered via intramuscular injection. Our data show that PMSCs treatment significantly enhanced microvessel density, improved blood perfusion and diminished pathologies in ischemic mouse hindlimbs as compared to those in the control group. Further immunostaining studies suggested that injected PMSCs can incorporate into the vasculature and differentiate into endothelial and smooth muscle cells to enhance angiogenesis in ischemic hind limbs. This may in part explain the beneficial effects of PMSCs treatment. Taken together, we found that PMSCs treatment might be an effective treatment modality for treatment of ischemia‐induced injury to mouse hind limbs by enhancement of angiogenesis.
Collapse
Affiliation(s)
- Nanzi Xie
- Division of Geriatrics, Tongji Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Zhihong Li
- Division of General Surgery, Chenzhou First People's Hospital, Chenzhou, Hunan, China
| | - Timothy M Adesanya
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Weixin Guo
- Guangdong Geriatrics Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yang Liu
- Division of Geriatrics, Tongji Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Minghuan Fu
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Ahmet Kilic
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Tao Tan
- Division of Geriatrics, Tongji Hospital, Tongji University, School of Medicine, Shanghai, China.,Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Hua Zhu
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Xiaoyun Xie
- Division of Geriatrics, Tongji Hospital, Tongji University, School of Medicine, Shanghai, China
| |
Collapse
|
29
|
Kobayashi H, Matsuda Y, Hitomi T, Okuda H, Shioi H, Matsuda T, Imai H, Sone M, Taura D, Harada KH, Habu T, Takagi Y, Miyamoto S, Koizumi A. Biochemical and Functional Characterization of RNF213 (Mysterin) R4810K, a Susceptibility Mutation of Moyamoya Disease, in Angiogenesis In Vitro and In Vivo. J Am Heart Assoc 2015; 4:JAHA.115.002146. [PMID: 26126547 PMCID: PMC4608092 DOI: 10.1161/jaha.115.002146] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Background P.R4810K of RNF213 (mysterin: rs112735431), which is an AAA+ ATPase, is the susceptibility polymorphism for moyamoya disease (MMD) in East Asians. However, the role of RNF213 R4810K in the etiology of MMD is unknown. Methods and Results To clarify the role of RNF213 in known angiogenic pathways, RNF213 expression was analyzed in endothelial cells (ECs) treated with several angiogenic and antiangiogenic factors, including interferons (IFNs). RNF213 was upregulated by IFN-β through signal transducer and activator of transcription x in the promoter and mediated antiangiogenic activity of IFN-β. RNF213 wild-type (WT) overexpression could not lower angiogenesis without IFN-β, but RNF213 R4810K overexpression could. To correlate biochemical function as ATPase and the role of RNF213 oligomer formation with antiangiogenic activity, we investigated the effects of mutations in the AAA+ module. A mutation of the Walker B motif (WEQ), which stabilizes oligomerization, inhibited angiogenesis, but AAA+ module deletion, which cannot initiate oligomerization, did not. Intriguingly, R4810K, similar to WEQ, decreased ATPase activity, suggesting its antiangiogenic activity through stabilizing oligomers. To confirm the antiangiogenic effect of RNF213 upregulation in vivo, vascular EC- or smooth muscle cell-specific Rnf213 R4757K (R4810K ortholog) or WT transgenic (Tg) mice were exposed to hypoxia. Cerebral angiogenesis by hypoxia was suppressed in EC-specific Rnf213 R4757K Tg mice, whereas it was not suppressed in other mice. Conclusions This study suggests the importance of inflammatory signals as environmental factors and R4810K carriers for susceptibility to cerebral hypoxia. A specific inhibitor of ATP binding to the first AAA+ could be a promising therapeutic candidate for MMD.
Collapse
Affiliation(s)
- Hatasu Kobayashi
- Department of Health and Environmental Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan (H.K., T.H., H.O., H.S., K.H.H., A.K.)
| | - Yoshiko Matsuda
- Department of Neurosurgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan (Y.M., Y.T., S.M.)
| | - Toshiaki Hitomi
- Department of Health and Environmental Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan (H.K., T.H., H.O., H.S., K.H.H., A.K.) Department of Preventive Medicine, St Marianna University School of Medicine, Kawasaki, Japan (T.H.)
| | - Hiroko Okuda
- Department of Health and Environmental Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan (H.K., T.H., H.O., H.S., K.H.H., A.K.)
| | - Hirotomo Shioi
- Department of Health and Environmental Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan (H.K., T.H., H.O., H.S., K.H.H., A.K.)
| | - Tetsuya Matsuda
- Department of Systems Science, Graduate School of Informatics, Kyoto University, Kyoto, Japan (T.M., H.I.)
| | - Hirohiko Imai
- Department of Systems Science, Graduate School of Informatics, Kyoto University, Kyoto, Japan (T.M., H.I.)
| | - Masakatsu Sone
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto, Japan (M.S., D.T.)
| | - Daisuke Taura
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto, Japan (M.S., D.T.)
| | - Kouji H Harada
- Department of Health and Environmental Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan (H.K., T.H., H.O., H.S., K.H.H., A.K.)
| | - Toshiyuki Habu
- Radiation Biology Center, Kyoto University, Kyoto, Japan (T.H.)
| | - Yasushi Takagi
- Department of Neurosurgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan (Y.M., Y.T., S.M.)
| | - Susumu Miyamoto
- Department of Neurosurgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan (Y.M., Y.T., S.M.)
| | - Akio Koizumi
- Department of Health and Environmental Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan (H.K., T.H., H.O., H.S., K.H.H., A.K.)
| |
Collapse
|
30
|
Lee S, Valmikinathan CM, Byun J, Kim S, Lee G, Mokarram N, Pai SB, Um E, Bellamkonda RV, Yoon YS. Enhanced therapeutic neovascularization by CD31-expressing cells and embryonic stem cell-derived endothelial cells engineered with chitosan hydrogel containing VEGF-releasing microtubes. Biomaterials 2015; 63:158-67. [PMID: 26102992 DOI: 10.1016/j.biomaterials.2015.06.009] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Revised: 06/07/2015] [Accepted: 06/10/2015] [Indexed: 12/14/2022]
Abstract
Various stem cells and their progeny have been used therapeutically for vascular regeneration. One of the major hurdles for cell-based therapy is low cell retention in vivo, and to improve cell survival several biomaterials have been used to encapsulate cells before transplantation. Vascular regeneration involves new blood vessel formation which consists of two processes, vasculogenesis and angiogenesis. While embryonic stem cell (ESC)-derived endothelial cells (ESC-ECs) have clearer vasculogenic potency, adult cells exert their effects mainly through paracrine angiogenic activities. While these two cells have seemingly complementary advantages, there have not been any studies to date combining these two cell types for vascular regeneration. We have developed a novel chitosan-based hydrogel construct that encapsulates both CD31-expressing BM-mononuclear cells (BM-CD31(+) cells) and ESC-ECs, and is loaded with VEGF-releasing microtubes. This cell construct showed high cell survival and minimal cytotoxicity in vitro. When implanted into a mouse model of hindlimb ischemia, it induced robust cell retention, neovascularization through vasculogenesis and angiogenesis, and efficiently induced recovery of blood flow in ischemic hindlimbs. This chitosan-based hydrogel encapsulating mixed adult and embryonic cell derivatives and containing VEGF can serve as a novel platform for treating various cardiovascular diseases.
Collapse
Affiliation(s)
- Sangho Lee
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, 101 Woodruff Circle, Atlanta, GA 30322, USA
| | - Chandra M Valmikinathan
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, 313 Ferst Drive, Atlanta, GA 30332, USA
| | - Jaemin Byun
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, 101 Woodruff Circle, Atlanta, GA 30322, USA
| | - Sangsung Kim
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, 101 Woodruff Circle, Atlanta, GA 30322, USA
| | - Geehee Lee
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, 101 Woodruff Circle, Atlanta, GA 30322, USA
| | - Nassir Mokarram
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, 313 Ferst Drive, Atlanta, GA 30332, USA
| | - S Balakrishna Pai
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, 313 Ferst Drive, Atlanta, GA 30332, USA
| | - Elisa Um
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, 101 Woodruff Circle, Atlanta, GA 30322, USA
| | - Ravi V Bellamkonda
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, 313 Ferst Drive, Atlanta, GA 30332, USA
| | - Young-sup Yoon
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, 101 Woodruff Circle, Atlanta, GA 30322, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, 313 Ferst Drive, Atlanta, GA 30332, USA; Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 120-752, South Korea.
| |
Collapse
|
31
|
Minami H, Tashiro K, Okada A, Hirata N, Yamaguchi T, Takayama K, Mizuguchi H, Kawabata K. Generation of Brain Microvascular Endothelial-Like Cells from Human Induced Pluripotent Stem Cells by Co-Culture with C6 Glioma Cells. PLoS One 2015; 10:e0128890. [PMID: 26061227 PMCID: PMC4464886 DOI: 10.1371/journal.pone.0128890] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 05/02/2015] [Indexed: 11/30/2022] Open
Abstract
The blood brain barrier (BBB) is formed by brain microvascular endothelial cells (BMECs) and tightly regulates the transport of molecules from blood to neural tissues. In vitro BBB models from human pluripotent stem cell (PSCs)-derived BMECs would be useful not only for the research on the BBB development and function but also for drug-screening for neurological diseases. However, little is known about the differentiation of human PSCs to BMECs. In the present study, human induced PSCs (iPSCs) were differentiated into endothelial cells (ECs), and further maturated to BMECs. Interestingly, C6 rat glioma cell-conditioned medium (C6CM), in addition to C6 co-culture, induced the differentiation of human iPSC-derived ECs (iPS-ECs) to BMEC-like cells, increase in the trans-endothelial electrical resistance, decreased in the dextran transport and up-regulation of gene expression of tight junction molecules in human iPS-ECs. Moreover, Wnt inhibitors attenuated the effects of C6CM. In summary, we have established a simple protocol of the generation of BMEC-like cells from human iPSCs, and have demonstrated that differentiation of iPS-ECs to BMEC-like cells is induced by C6CM-derived signals, including canonical Wnt signals.
Collapse
Affiliation(s)
- Haruka Minami
- Laboratory of Stem Cell Regulation, National Institute of Biomedical Innovation, 7-6-8, Saito-Asagi, Ibaraki, Osaka 567–0085, Japan
- Laboratory of Biomedical Innovation, Graduate School of Pharmaceutical Sciences, Osaka University, 1–6 Yamadaoka, Suita, Osaka 565–0871, Japan
| | - Katsuhisa Tashiro
- Laboratory of Stem Cell Regulation, National Institute of Biomedical Innovation, 7-6-8, Saito-Asagi, Ibaraki, Osaka 567–0085, Japan
| | - Atsumasa Okada
- Laboratory of Stem Cell Regulation, National Institute of Biomedical Innovation, 7-6-8, Saito-Asagi, Ibaraki, Osaka 567–0085, Japan
| | - Nobue Hirata
- Laboratory of Stem Cell Regulation, National Institute of Biomedical Innovation, 7-6-8, Saito-Asagi, Ibaraki, Osaka 567–0085, Japan
| | - Tomoko Yamaguchi
- Laboratory of Stem Cell Regulation, National Institute of Biomedical Innovation, 7-6-8, Saito-Asagi, Ibaraki, Osaka 567–0085, Japan
| | - Kazuo Takayama
- Laboratory of Hepatocyte Regulation, National Institute of Biomedical Innovation, 7-6-8, Saito-Asagi, Ibaraki, Osaka 567–0085, Japan
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1–6 Yamadaoka, Suita, Osaka 565–0871, Japan
- iPS Cell-based Research Project on Hepatic Toxicity and Metabolism, Graduate School of Pharmaceutical Sciences, Osaka University, 1–6 Yamadaoka, Suita, Osaka 565–0871, Japan
| | - Hiroyuki Mizuguchi
- Laboratory of Hepatocyte Regulation, National Institute of Biomedical Innovation, 7-6-8, Saito-Asagi, Ibaraki, Osaka 567–0085, Japan
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1–6 Yamadaoka, Suita, Osaka 565–0871, Japan
- iPS Cell-based Research Project on Hepatic Toxicity and Metabolism, Graduate School of Pharmaceutical Sciences, Osaka University, 1–6 Yamadaoka, Suita, Osaka 565–0871, Japan
- The Center for Advanced Medical Engineering and Informatics, Osaka University, 1–6, Yamadaoka, Suita, Osaka, 565–0871, Japan
| | - Kenji Kawabata
- Laboratory of Stem Cell Regulation, National Institute of Biomedical Innovation, 7-6-8, Saito-Asagi, Ibaraki, Osaka 567–0085, Japan
- Laboratory of Biomedical Innovation, Graduate School of Pharmaceutical Sciences, Osaka University, 1–6 Yamadaoka, Suita, Osaka 565–0871, Japan
- * E-mail:
| |
Collapse
|
32
|
Wang G, Jacquet L, Karamariti E, Xu Q. Origin and differentiation of vascular smooth muscle cells. J Physiol 2015; 593:3013-30. [PMID: 25952975 PMCID: PMC4532522 DOI: 10.1113/jp270033] [Citation(s) in RCA: 210] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 04/19/2015] [Indexed: 12/18/2022] Open
Abstract
Vascular smooth muscle cells (SMCs), a major structural component of the vessel wall, not only play a key role in maintaining vascular structure but also perform various functions. During embryogenesis, SMC recruitment from their progenitors is an important step in the formation of the embryonic vascular system. SMCs in the arterial wall are mostly quiescent but can display a contractile phenotype in adults. Under pathophysiological conditions, i.e. vascular remodelling after endothelial dysfunction or damage, contractile SMCs found in the media switch to a secretory type, which will facilitate their ability to migrate to the intima and proliferate to contribute to neointimal lesions. However, recent evidence suggests that the mobilization and recruitment of abundant stem/progenitor cells present in the vessel wall are largely responsible for SMC accumulation in the intima during vascular remodelling such as neointimal hyperplasia and arteriosclerosis. Therefore, understanding the regulatory mechanisms that control SMC differentiation from vascular progenitors is essential for exploring therapeutic targets for potential clinical applications. In this article, we review the origin and differentiation of SMCs from stem/progenitor cells during cardiovascular development and in the adult, highlighting the environmental cues and signalling pathways that control phenotypic modulation within the vasculature.
![]()
Collapse
Affiliation(s)
- Gang Wang
- Department of Emergency Medicine, the Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Laureen Jacquet
- Cardiovascular Division, King's College London BHF Centre, London, UK
| | - Eirini Karamariti
- Cardiovascular Division, King's College London BHF Centre, London, UK
| | - Qingbo Xu
- Cardiovascular Division, King's College London BHF Centre, London, UK
| |
Collapse
|
33
|
Kane NM, Thrasher AJ, Angelini GD, Emanueli C. Concise review: MicroRNAs as modulators of stem cells and angiogenesis. Stem Cells 2014; 32:1059-66. [PMID: 24449004 DOI: 10.1002/stem.1629] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 12/08/2013] [Indexed: 12/15/2022]
Abstract
MicroRNAs (miRs) are highly conserved, short noncoding RNA molecules that negatively regulate messenger RNA (mRNA) stability and/or translational efficiency. Since a given miR can control the expression of many mRNAs, their importance in governing gene expression in specific cell types including vascular cells and their progenitor cells has become increasingly clear. Understanding how the expression of miRs themselves is regulated and how miRs exert their influence on post-transcriptional gene control provides novel opportunities to dissect gene regulatory networks in clinically relevant cell types. A multitude of miRs have been identified with key roles in vascular development, homeostasis, function, disease, and regeneration. In this review, we will describe the impact of miRs on angiogenesis and their capacity to modulate the behavior of stem and progenitor cells which may be utilitarian for promoting vascular growth in ischemic tissue. Moreover, we summarize these strategies available for modulating miR expression and function and future therapeutic applications.
Collapse
Affiliation(s)
- Nicole M Kane
- Molecular Immunology Unit, Institute of Child Health, University College of London, London, United Kingdom
| | | | | | | |
Collapse
|
34
|
Differentiation of human pluripotent stem cells to cells similar to cord-blood endothelial colony-forming cells. Nat Biotechnol 2014; 32:1151-1157. [PMID: 25306246 DOI: 10.1038/nbt.3048] [Citation(s) in RCA: 167] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 07/11/2014] [Indexed: 02/06/2023]
Abstract
The ability to differentiate human pluripotent stem cells into endothelial cells with properties of cord-blood endothelial colony-forming cells (CB-ECFCs) may enable the derivation of clinically relevant numbers of highly proliferative blood vessel-forming cells to restore endothelial function in patients with vascular disease. We describe a protocol to convert human induced pluripotent stem cells (hiPSCs) or embryonic stem cells (hESCs) into cells similar to CB-ECFCs at an efficiency of >10(8) ECFCs produced from each starting pluripotent stem cell. The CB-ECFC-like cells display a stable endothelial phenotype with high clonal proliferative potential and the capacity to form human vessels in mice and to repair the ischemic mouse retina and limb, and they lack teratoma formation potential. We identify Neuropilin-1 (NRP-1)-mediated activation of KDR signaling through VEGF165 as a critical mechanism for the emergence and maintenance of CB-ECFC-like cells.
Collapse
|
35
|
Chen YE, Xie C, Yang B. Stem cells for vascular engineering. BIOMATERIALS AND REGENERATIVE MEDICINE 2014:621-639. [DOI: 10.1017/cbo9780511997839.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
36
|
Moon SH, Kim JM, Hong KS, Shin JM, Kim J, Chung HM. Differentiation of hESCs into Mesodermal Subtypes: Vascular-, Hematopoietic- and Mesenchymal-lineage Cells. Int J Stem Cells 2014; 4:24-34. [PMID: 24298331 DOI: 10.15283/ijsc.2011.4.1.24] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/03/2011] [Indexed: 12/30/2022] Open
Abstract
To date, studies on the application of mesodermally derived mesenchymal-, hematopoietic- and vascular-lineage cells for cell therapy have provided either poor or insufficient data. The results are equivocal with regard to therapeutic efficiency and yield. Since the establishment of human embryonic stem cells (hESCs) in 1998, the capacity of hESCs to differentiate into various mesodermal lineages has sparked considerable interest in the regenerative medicine community, a group interested in generating specialized cells to treat patients suffering from degenerative diseases. Even though hESCs are sensitive, effective methods for guiding the differentiation of hESCs into specific mesodermal cell types are still being developed. In addition, to understand the functional properties of hESC derivatives, numerous animal model studies have been performed by many research groups over the last decade. In this review, we describe and summarize the protocols currently used for differentiation of hESCs into multiple mesodermal lineages and their therapeutic efficiency in different animal models. Furthermore, we discuss the technical hurdles associated with each protocol and the safety of hESC derivatives for therapeutic applications. Technical improvement of the methods used to produce hESC derivatives for therapeutic use in patients with degenerative diseases should remain an objective of future studies, as should the development of effective and stable induction systems.
Collapse
|
37
|
Baldwin J, Antille M, Bonda U, De-Juan-Pardo EM, Khosrotehrani K, Ivanovski S, Petcu EB, Hutmacher DW. In vitro pre-vascularisation of tissue-engineered constructs A co-culture perspective. Vasc Cell 2014; 6:13. [PMID: 25071932 PMCID: PMC4112973 DOI: 10.1186/2045-824x-6-13] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Accepted: 06/12/2014] [Indexed: 12/29/2022] Open
Abstract
In vitro pre-vascularization is one of the main vascularization strategies in the tissue engineering field. Culturing cells within a tissue-engineered construct (TEC) prior to implantation provides researchers with a greater degree of control over the fate of the cells. However, balancing the diverse range of different cell culture parameters in vitro is seldom easy and in most cases, especially in highly vascularized tissues, more than one cell type will reside within the cell culture system. Culturing multiple cell types in the same construct presents its own unique challenges and pitfalls. The following review examines endothelial-driven vascularization and evaluates the direct and indirect role other cell types have in vessel and capillary formation. The article then analyses the different parameters researchers can modulate in a co-culture system in order to design optimal tissue-engineered constructs to match desired clinical applications.
Collapse
Affiliation(s)
- Jeremy Baldwin
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD 4059, Australia
| | - Mélanie Antille
- Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Ulrich Bonda
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD 4059, Australia
- Leibniz Institute of Polymer Research Dresden (IPF) & Max Bergmann Center of Biomaterials Dresden (MBC), Hohe Str. 6, 01069, Dresden, Germany
| | - Elena M De-Juan-Pardo
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD 4059, Australia
| | - Kiarash Khosrotehrani
- University of Queensland, UQ Centre for Clinical Research, Royal Brisbane & Women's Hospital Campus, Building 71/918, Herston, QLD 4029, Australian
- The University of Queensland, UQ Diamantina Institute, Translational Research Institute, Brisbane, QLD, Australia
| | - Saso Ivanovski
- Griffith Health Institute, Regenerative Medicine Centre, Gold Coast, QLD 4222, Australia
| | - Eugen Bogdan Petcu
- Griffith Health Institute, Regenerative Medicine Centre, Gold Coast, QLD 4222, Australia
| | - Dietmar Werner Hutmacher
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD 4059, Australia
| |
Collapse
|
38
|
Sahara M, Hansson EM, Wernet O, Lui KO, Später D, Chien KR. Manipulation of a VEGF-Notch signaling circuit drives formation of functional vascular endothelial progenitors from human pluripotent stem cells. Cell Res 2014; 24:820-41. [PMID: 24810299 PMCID: PMC4085760 DOI: 10.1038/cr.2014.59] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 03/10/2014] [Accepted: 03/31/2014] [Indexed: 12/13/2022] Open
Abstract
Human pluripotent stem cell (hPSC)-derived endothelial lineage cells constitutes a promising source for therapeutic revascularization, but progress in this arena has been hampered by a lack of clinically-scalable differentiation protocols and inefficient formation of a functional vessel network integrating with the host circulation upon transplantation. Using a human embryonic stem cell reporter cell line, where green fluorescent protein expression is driven by an endothelial cell-specific VE-cadherin (VEC) promoter, we screened for > 60 bioactive small molecules that would promote endothelial differentiation, and found that administration of BMP4 and a GSK-3β inhibitor in an early phase and treatment with VEGF-A and inhibition of the Notch signaling pathway in a later phase led to efficient differentiation of hPSCs to the endothelial lineage within six days. This sequential approach generated > 50% conversion of hPSCs to endothelial cells (ECs), specifically VEC+CD31+CD34+CD14−KDRhigh endothelial progenitors (EPs) that exhibited higher angiogenic and clonogenic proliferation potential among endothelial lineage cells. Pharmaceutical inhibition or genetical knockdown of Notch signaling, in combination with VEGF-A treatment, resulted in efficient formation of EPs via KDR+ mesodermal precursors and blockade of the conversion of EPs to mature ECs. The generated EPs successfully formed functional capillary vessels in vivo with anastomosis to the host vessels when transplanted into immunocompromised mice. Manipulation of this VEGF-A-Notch signaling circuit in our protocol leads to rapid large-scale production of the hPSC-derived EPs by 12- to 20-fold vs current methods, which may serve as an attractive cell population for regenerative vascularization with superior vessel forming capability compared to mature ECs.
Collapse
Affiliation(s)
- Makoto Sahara
- 1] Department of Stem Cell and Regenerative Biology, 7 Divinity Avenue, Cambridge, MA 02138, USA [2] Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA [3] Center for Regenerative Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 021141, USA [4] Department of Medicine-Cardiology/Cell and Molecular Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Emil M Hansson
- 1] Department of Stem Cell and Regenerative Biology, 7 Divinity Avenue, Cambridge, MA 02138, USA [2] Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | - Oliver Wernet
- Department of Anesthesiology and Intensive Care Medicine, Charité-University Medicine Berlin, Campus Charité Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Kathy O Lui
- 1] Department of Stem Cell and Regenerative Biology, 7 Divinity Avenue, Cambridge, MA 02138, USA [2] Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA [3] Center for Regenerative Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 021141, USA
| | - Daniela Später
- 1] Department of Stem Cell and Regenerative Biology, 7 Divinity Avenue, Cambridge, MA 02138, USA [2] Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA [3] Center for Regenerative Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 021141, USA
| | - Kenneth R Chien
- 1] Department of Stem Cell and Regenerative Biology, 7 Divinity Avenue, Cambridge, MA 02138, USA [2] Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA [3] Department of Medicine-Cardiology/Cell and Molecular Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| |
Collapse
|
39
|
Molecular pathways governing development of vascular endothelial cells from ES/iPS cells. Stem Cell Rev Rep 2014; 9:586-98. [PMID: 23765563 DOI: 10.1007/s12015-013-9450-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Assembly of complex vascular networks occurs in numerous biological systems through morphogenetic processes such as vasculogenesis, angiogenesis and vascular remodeling. Pluripotent stem cells such as embryonic stem (ES) and induced pluripotent stem (iPS) cells can differentiate into any cell type, including endothelial cells (ECs), and have been extensively used as in vitro models to analyze molecular mechanisms underlying EC generation and differentiation. The emergence of these promising new approaches suggests that ECs could be used in clinical therapy. Much evidence suggests that ES/iPS cell differentiation into ECs in vitro mimics the in vivo vascular morphogenic process. Through sequential steps of maturation, ECs derived from ES/iPS cells can be further differentiated into arterial, venous, capillary and lymphatic ECs, as well as smooth muscle cells. Here, we review EC development from ES/iPS cells with special attention to molecular pathways functioning in EC specification.
Collapse
|
40
|
Park HS, Choi GH, Hahn S, Yoo YS, Jung IM, Lee T. Tracking the Fate of Muscle-derived Stem Cells: an Insight into the Distribution and Mode of Action. Vasc Specialist Int 2014. [PMID: 26217610 PMCID: PMC4480300 DOI: 10.5758/vsi.2014.30.1.11] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Purpose: To examine the fate of muscle-derived stem cells (MDSC) after injection into different host conditions and provide an insight for their mechanism of action. Materials and Methods: MDSCs differentiated in vitro towards the endothelial lineage and transfected with lentivirus tagged with green fluorescent protein (GFP) were injected into two animal models mimicking vascular diseases: hindlimb ischemia and carotid injury models. Injected cells were tracked at the site of injection and in remote organs by harvesting the respective tissues at different time intervals and performing immunofluorescent histological analyses. Stem cell survival was quantified at the site of injection for up to 4 weeks. Results: MDSCs were successfully tagged with fluorescent material GFP and showed successful implantation into the respective injection sites. These cells showed a higher affinity to implant in blood vessel walls as shown by double fluorescent co-stain with CD31. Quantification of stem cell survival showed a timede pendent decrease from day 3 to 4 weeks (survival rate normalized against day 3 was 72.0% at 1 week, 26.8% at 2 weeks and 2.4% at 4 weeks). Stem cells were also found in distant organs, especially the kidneys and liver, which survived up to 4 weeks. Conclusion: MDSCs were successfully tracked in different vascular disease models, and their fate was assessed in terms of cell survival and distribution. Better understanding of the donor cell properties, including their interaction with the host conditions and their mechanism of action, are needed to enhance cell survival and achieve improved outcomes.
Collapse
Affiliation(s)
- Hyung Sub Park
- Department of Surgery, Seoul National University College of Medicine, Seoul ; Department of Surgery, Seoul National University Bundang Hospital, Seongnam
| | - Geum Hee Choi
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam
| | - Soli Hahn
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam
| | - Young Sun Yoo
- Department of Surgery, Chosun University Hospital, Chosun University School of Medicine, Gwangju
| | - In Mok Jung
- Department of Surgery, Seoul National University College of Medicine, Seoul ; Department of Surgery, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul, Korea
| | - Taeseung Lee
- Department of Surgery, Seoul National University College of Medicine, Seoul ; Department of Surgery, Seoul National University Bundang Hospital, Seongnam
| |
Collapse
|
41
|
Kusuma S, Peijnenburg E, Patel P, Gerecht S. Low oxygen tension enhances endothelial fate of human pluripotent stem cells. Arterioscler Thromb Vasc Biol 2014; 34:913-20. [PMID: 24526696 DOI: 10.1161/atvbaha.114.303274] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
OBJECTIVE A critical regulator of the developing or regenerating vasculature is low oxygen tension. Precise elucidation of the role of low oxygen environments on endothelial commitment from human pluripotent stem cells necessitates controlled in vitro differentiation environments. APPROACH AND RESULTS We used a feeder-free, 2-dimensional differentiation system in which we could monitor accurately dissolved oxygen levels during human pluripotent stem cell differentiation toward early vascular cells (EVCs). We found that oxygen uptake rate of differentiating human pluripotent stem cells is lower in 5% O2 compared with atmospheric conditions. EVCs differentiated in 5% O2 had an increased vascular endothelial cadherin expression with clusters of vascular endothelial cadherin+ cells surrounded by platelet-derived growth factor β+ cells. When we assessed the temporal effects of low oxygen differentiation environments, we determined that low oxygen environments during the early stages of EVC differentiation enhance endothelial lineage commitment. EVCs differentiated in 5% O2 exhibited an increased expression of vascular endothelial cadherin and CD31 along with their localization to the membrane, enhanced lectin binding and acetylated low-density lipoprotein uptake, rapid cord-like structure formation, and increased expression of arterial endothelial cell markers. Inhibition of reactive oxygen species generation during the early stages of differentiation abrogated the endothelial inductive effects of the low oxygen environments. CONCLUSIONS Low oxygen tension during early stages of EVC derivation induces endothelial commitment and maturation through the accumulation of reactive oxygen species, highlighting the importance of regulating oxygen tensions during human pluripotent stem cell-vascular differentiation.
Collapse
Affiliation(s)
- Sravanti Kusuma
- From the Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center, Institute for NanoBioTechnology (S.K., P.P., S.G.), Department of Biomedical Engineering (S.K.), Cellular and Molecular Biology (E.P.), and Department of Materials Science and Engineering (S.G.), Johns Hopkins University, Baltimore, MD
| | | | | | | |
Collapse
|
42
|
Sinha S, Iyer D, Granata A. Embryonic origins of human vascular smooth muscle cells: implications for in vitro modeling and clinical application. Cell Mol Life Sci 2014; 71:2271-88. [PMID: 24442477 PMCID: PMC4031394 DOI: 10.1007/s00018-013-1554-3] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 12/03/2013] [Accepted: 12/30/2013] [Indexed: 01/06/2023]
Abstract
Vascular smooth muscle cells (SMCs) arise from multiple origins during development, raising the possibility that differences in embryological origins between SMCs could contribute to site-specific localization of vascular diseases. In this review, we first examine the developmental pathways and embryological origins of vascular SMCs and then discuss in vitro strategies for deriving SMCs from human embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs). We then review in detail the potential for vascular disease modeling using iPSC-derived SMCs and consider the pathological implications of heterogeneous embryonic origins. Finally, we touch upon the role of human ESC-derived SMCs in therapeutic revascularization and the challenges remaining before regenerative medicine using ESC- or iPSC-derived cells comes of age.
Collapse
Affiliation(s)
- Sanjay Sinha
- Anne McLaren Laboratory for Regenerative Medicine, University of Cambridge, Cambridge, CB2 0SZ, UK,
| | | | | |
Collapse
|
43
|
Generation of robust vascular networks from cardiovascular blast populations derived from human induced pluripotent stem cells in vivo and ex vivo organ culture system. Biochem Biophys Res Commun 2013; 441:180-5. [DOI: 10.1016/j.bbrc.2013.10.035] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Accepted: 10/08/2013] [Indexed: 11/21/2022]
|
44
|
Cao N, Liang H, Huang J, Wang J, Chen Y, Chen Z, Yang HT. Highly efficient induction and long-term maintenance of multipotent cardiovascular progenitors from human pluripotent stem cells under defined conditions. Cell Res 2013; 23:1119-32. [PMID: 23896987 DOI: 10.1038/cr.2013.102] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 05/05/2013] [Accepted: 05/17/2013] [Indexed: 12/23/2022] Open
Abstract
Cardiovascular progenitor cells (CVPCs) derived from human pluripotent stem cells (hPSCs), including human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs), hold great promise for the study of cardiovascular development and cell-based therapy of heart diseases, but their applications are challenged by the difficulties in their efficient generation and stable maintenance. This study aims to develop chemically defined systems for robust generation and stable propagation of hPSC-derived CVPCs by modulating the key early developmental pathways involved in human cardiovascular specification and CVPC self-renewal. Herein we report that a combination of bone morphogenetic protein 4 (BMP4), glycogen synthase kinase 3 (GSK3) inhibitor CHIR99021 and ascorbic acid is sufficient to rapidly convert monolayer-cultured hPSCs, including hESCs and hiPSCs, into homogeneous CVPCs in a chemically defined medium under feeder- and serum-free culture conditions. These CVPCs stably self-renewed under feeder- and serum-free conditions and expanded over 10(7)-fold when the differentiation-inducing signals from BMP, GSK3 and Activin/Nodal pathways were simultaneously eliminated. Furthermore, these CVPCs exhibited expected genome-wide molecular features of CVPCs, retained potentials to generate major cardiovascular lineages including cardiomyocytes, smooth muscle cells and endothelial cells in vitro, and were non-tumorigenic in vivo. Altogether, the established systems reported here permit efficient generation and stable maintenance of hPSC-derived CVPCs, which represent a powerful tool to study early embryonic cardiovascular development and provide a potentially safe source of cells for myocardial regenerative medicine.
Collapse
Affiliation(s)
- Nan Cao
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences & Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200025, China
| | | | | | | | | | | | | |
Collapse
|
45
|
Azhdari M, Baghaban-Eslaminejad M, Baharvand H, Aghdami N. Therapeutic potential of human-induced pluripotent stem cell-derived endothelial cells in a bleomycin-induced scleroderma mouse model. Stem Cell Res 2013; 10:288-300. [DOI: 10.1016/j.scr.2012.12.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Revised: 11/15/2012] [Accepted: 12/12/2012] [Indexed: 10/27/2022] Open
|
46
|
Song SH, Jung W, Kim KL, Hong W, Kim HO, Lee KA, Lee KY, Suh W. Distinct transcriptional profiles of angioblasts derived from human embryonic stem cells. Exp Cell Res 2013; 319:1136-45. [PMID: 23458169 DOI: 10.1016/j.yexcr.2013.02.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Revised: 02/20/2013] [Accepted: 02/21/2013] [Indexed: 11/19/2022]
Abstract
Identification of differentially expressed genes in angioblasts derived from human embryonic stem cells (hESCs) is of great interest for elucidating the molecular mechanisms underlying human vasculogenesis. The aim of this study was to define hESC-derived angioblasts at the clonal level and to perform comparative transcriptional analysis to characterize their distinct gene expression profiles. In a clonal analysis performed in cell-specific differentiation media, hESC-derived CD34(+)CD31(+) cells were identified as angioblasts in that they exhibited a significantly higher ability to form endothelial cell (EC) and smooth muscle cell (SMC) colonies than CD34(+)CD31(-) and CD34(-) cell populations did. Microarray analysis showed that many genes involved in vascular development and signaling transduction were overexpressed in hESC-derived CD34(+)CD31(+) cells, whereas those related to mitosis, the DNA damage response, and translation were substantially downregulated. In addition, comparative gene expression profiling of hESC-derived CD34(+)CD31(+) cells and human somatic primary vascular cells demonstrated that hESC-derived CD34(+)CD31(+) cells expressed key genes involved in the EC and SMC differentiation processes, which supports the result that hESC-derived CD34(+)CD31(+) cells are bipotent angioblasts. Our results may provide insights into the identity and function of hESC-derived angioblasts and may also facilitate further investigation of the molecular mechanisms regulating human embryonic vasculogenesis.
Collapse
Affiliation(s)
- Sun-Hwa Song
- College of Pharmacy, School of Medicine, Ajou University, San 5, Woncheon-Dong, Yeongtong-Gu, Suwon, Gyeonggi-do 443-749, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Blancas AA, Wong LE, Glaser DE, McCloskey KE. Specialized tip/stalk-like and phalanx-like endothelial cells from embryonic stem cells. Stem Cells Dev 2013; 22:1398-407. [PMID: 23249281 DOI: 10.1089/scd.2012.0376] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Endothelial cells (EC) generated in vitro from stem cells are desirable for their potential in a variety of in vitro models and cell-based therapeutic approaches; however, EC can take on a number of functionally and phenotypically distinct specializations. Here, we show the generation of functionally distinct EC subpopulations, including (1) the pro-angiogenic migrating tip-like and proliferative stalk-like EC, and (2) the less migratory cobblestone-shaped phalanx-like EC. Both embryonic stem cell (ESC)-derived EC subpopulations are generated from outgrowths of Flk-1+ vascular progenitor cells with high levels of vascular endothelial growth factor treatment, while the phalanx-like ESC-derived EC (ESC-EC) are subsequently isolated by selecting for cobblestone shape. Compared with the ESC-derived angiogenic endothelial cells (named ESC-AEC) that contain only 14% Flt-1+ and 25% Tie-1+ cells, the selected phalanx-like ESC-EC express higher numbers of cells expressing the phalanx markers Flt-1+ and Tie-1+, 89% and 90%, respectively. The ESC-AEC also contain 35% CXCR4+ tip cells, higher expression levels of stalk marker Notch-1, and lower expression levels of Tie-2 compared with the phalanx-type ESC-EC that do not contain discernible numbers of CXCR4+ tip cells. Perhaps most notably, the ESC-AEC display increased cell migration, proliferation, and 3 times more vessel-like structures after 48 h on Matrigel compared with the phalanx-like ESC-EC. This work analyzes, for the first time, the presence of distinct EC subtypes (tip/stalk, and phalanx) generated in vitro from ESC, and shows that phalanx-like EC can be purified and maintained in culture separate from the tip/stalk-like containing EC.
Collapse
Affiliation(s)
- Alicia A Blancas
- Graduate Program in Quantitative and Systems Biology, University of California , Merced, California, USA
| | | | | | | |
Collapse
|
48
|
Serbo JV, Gerecht S. Vascular tissue engineering: biodegradable scaffold platforms to promote angiogenesis. Stem Cell Res Ther 2013; 4:8. [PMID: 23347554 PMCID: PMC3706776 DOI: 10.1186/scrt156] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The ability to understand and regulate human vasculature development and differentiation has the potential to benefit patients suffering from a variety of ailments, including cardiovascular disease, peripheral vascular disease, ischemia, and burn wounds. Current clinical treatments for vascular-related diseases commonly use the grafting from patients of autologous vessels, which are limited and often damaged due to disease. Considerable progress is being made through a tissue engineering strategy in the vascular field. Tissue engineering takes a multidisciplinary approach seeking to repair, improve, or replace biological tissue function in a controlled and predictable manner. To address the clinical need to perfuse and repair damaged, ischemic tissue, one approach of vascular engineering aims to understand and promote the growth and differentiation of vascular networks. Vascular tissue engineered constructs enable the close study of vascular network assembly and vessel interactions with the surrounding microenvironment. Scaffold platforms provide a method to control network development through the biophysical regulation of different scaffold properties, such as composition, mechanics, dimensionality, and so forth. Following a short description of vascular physiology and blood vessel biomechanics, the key principles in vascular tissue engineering are discussed. This review focuses on various biodegradable scaffold platforms and demonstrates how they are being used to regulate, promote, and understand angiogenesis and vascular network formation.
Collapse
Affiliation(s)
- Janna V Serbo
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Sharon Gerecht
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences - Oncology Center and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| |
Collapse
|
49
|
Guo X, Stice SL, Boyd NL, Chen SY. A novel in vitro model system for smooth muscle differentiation from human embryonic stem cell-derived mesenchymal cells. Am J Physiol Cell Physiol 2012; 304:C289-98. [PMID: 23220114 DOI: 10.1152/ajpcell.00298.2012] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The objective of this study was to develop a novel in vitro model for smooth muscle cell (SMC) differentiation from human embryonic stem cell-derived mesenchymal cells (hES-MCs). We found that hES-MCs were differentiated to SMCs by transforming growth factor-β (TGF-β) in a dose- and time-dependent manner as demonstrated by the expression of SMC-specific genes smooth muscle α-actin, calponin, and smooth muscle myosin heavy chain. Under normal growth conditions, however, the differentiation capacity of hES-MCs was very limited. hES-MC-derived SMCs had an elongated and spindle-shaped morphology and contracted in response to the induction of carbachol and KCl. KCl-induced calcium transient was also evident in these cells. Compared with the parental cells, TGF-β-treated hES-MCs sustained the endothelial tube formation for a longer time due to the sustained SMC phenotype. Mechanistically, TGF-β-induced differentiation was both Smad- and serum response factor/myocardin dependent. TGF-β regulated myocardin expression via multiple signaling pathways including Smad2/3, p38 MAPK, and PI3K. Importantly, we found that a low level of myocardin was present in mesoderm prior to SMC lineage determination, and a high level of myocardin was not induced until the differentiation process was initiated. Taken together, our study characterized a novel SMC differentiation model that can be used for studying human SMC differentiation from mesoderm during vascular development.
Collapse
Affiliation(s)
- Xia Guo
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA 30602, USA
| | | | | | | |
Collapse
|
50
|
El-Mounayri O, Mihic A, Shikatani EA, Gagliardi M, Steinbach SK, Dubois N, DaCosta R, Li RK, Keller G, Husain M. Serum-free differentiation of functional human coronary-like vascular smooth muscle cells from embryonic stem cells. Cardiovasc Res 2012; 98:125-35. [DOI: 10.1093/cvr/cvs357] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|