1
|
Wang J, Wu Y, Wang Y, Shuai Y, Xu Z, Wan Q, Chen Y, Yang M. Graphene Oxide-Coated Patterned Silk Fibroin Films Promote Cell Adhesion and Induce Cardiomyogenic Differentiation of Human Mesenchymal Stem Cells. Biomolecules 2023; 13:990. [PMID: 37371570 DOI: 10.3390/biom13060990] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/03/2023] [Accepted: 06/04/2023] [Indexed: 06/29/2023] Open
Abstract
Cardiac tissue engineering is a promising strategy for the treatment of myocardial damage. Mesenchymal stem cells (MSCs) are extensively used in tissue engineering. However, transformation of MSCs into cardiac myocytes is still a challenge. Furthermore, weak adhesion of MSCs to substrates often results in poor cell viability. Here, we designed a composite matrix based on silk fibroin (SF) and graphene oxide (GO) for improving the cell adhesion and directing the differentiation of MSCs into cardiac myocytes. Specifically, patterned SF films were first produced by soft lithographic. After being treated by air plasma, GO nanosheets could be successfully coated on the patterned SF films to construct the desired matrix (P-GSF). The resultant P-GSF films presented a nano-topographic surface characterized by linear grooves interlaced with GO ridges. The P-GSF films exhibited high protein absorption and suitable mechanical strength. Furthermore, the P-GSF films accelerated the early cell adhesion and directed the growth orientation of MSCs. RT-PCR results and immunofluorescence imaging demonstrated that the P-GSF films significantly improved the cardiomyogenic differentiation of MSCs. This work indicates that patterned SF films coated with GO are promising matrix in the field of myocardial repair tissue engineering.
Collapse
Affiliation(s)
- Jie Wang
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Yi Wu
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Yecheng Wang
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Yajun Shuai
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Zongpu Xu
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Quan Wan
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Yuyin Chen
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Mingying Yang
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| |
Collapse
|
2
|
Lee CS, Kim J, Cho HJ, Kim HS. Cardiovascular Regeneration via Stem Cells and Direct Reprogramming: A Review. Korean Circ J 2022; 52:341-353. [PMID: 35502566 PMCID: PMC9064703 DOI: 10.4070/kcj.2022.0005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/16/2022] [Accepted: 02/22/2022] [Indexed: 11/24/2022] Open
Abstract
Despite recent advancements in treatment strategies, cardiovascular disease such as heart failure remains a significant source of global mortality. Stem cell technology and cellular reprogramming are rapidly growing fields that will continue to prove useful in cardiac regenerative therapeutics. This review provides information on the role of human pluripotent stem cells (hPSCs) in cardiac regeneration and discusses the practical applications of hPSC-derived cardiomyocytes (CMCs). Moreover, we discuss the practical applications of hPSC-derived CMCs while outlining the relevance of directly-reprogrammed CMCs in regenerative medicine. This review critically summarizes the most recent advances in the field will help to guide future research in this developing area. Cardiovascular disease (CVD) is the leading causes of morbidity and death globally. In particular, a heart failure remains a major problem that contributes to global mortality. Considerable advancements have been made in conventional pharmacological therapies and coronary intervention surgery for cardiac disorder treatment. However, more than 15% of patients continuously progress to end-stage heart failure and eventually require heart transplantation. Over the past year, numerous numbers of protocols to generate cardiomyocytes (CMCs) from human pluripotent stem cells (hPSCs) have been developed and applied in clinical settings. Number of studies have described the therapeutic effects of hPSCs in animal models and revealed the underlying repair mechanisms of cardiac regeneration. In addition, biomedical engineering technologies have improved the therapeutic potential of hPSC-derived CMCs in vivo. Recently substantial progress has been made in driving the direct differentiation of somatic cells into mature CMCs, wherein an intermediate cellular reprogramming stage can be bypassed. This review provides information on the role of hPSCs in cardiac regeneration and discusses the practical applications of hPSC-derived CMCs; furthermore, it outlines the relevance of directly reprogrammed CMCs in regenerative medicine.
Collapse
Affiliation(s)
- Choon-Soo Lee
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
- Strategic Center of Cell & Bio Therapy, Seoul National University Hospital, Seoul, Korea
| | - Joonoh Kim
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
- Strategic Center of Cell & Bio Therapy, Seoul National University Hospital, Seoul, Korea
| | - Hyun-Jai Cho
- Division of Cardiology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Hyo-Soo Kim
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
- Strategic Center of Cell & Bio Therapy, Seoul National University Hospital, Seoul, Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, and College of Medicine or College of Pharmacy, Seoul National University, Seoul, Korea
| |
Collapse
|
3
|
Sun J, Wang R, Chao T, Wang C. Long Noncoding RNAs Involved in Cardiomyocyte Apoptosis Triggered by Different Stressors. J Cardiovasc Transl Res 2021; 15:588-603. [PMID: 34855148 DOI: 10.1007/s12265-021-10186-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 11/04/2021] [Indexed: 12/26/2022]
Abstract
Cardiomyocytes are essential to maintain the normal cardiac function. Ischemia, hypoxia, and drug stimulation can induce pathological apoptosis of cardiomyocytes which eventually leads to heart failure, arrhythmia, and other cardiovascular diseases. Understanding the molecular mechanisms that regulate cardiomyocyte apoptosis is of great significance for the prevention and treatment of cardiovascular diseases. In recent years, more and more evidences reveal that long noncoding RNAs (lncRNAs) play important regulatory roles in myocardial cell apoptosis. They can modulate the expression of apoptosis-related genes at post-transcriptional level by altering the translation efficacy of target mRNAs or functioning as a precursor for miRNAs or competing for miRNA-mediated inhibition. Moreover, reversing the abnormal expression of lncRNAs can attenuate and even reverse the pathological apoptosis of cardiomyocytes. Therefore, apoptosis-related lncRNAs may become a potential new field for studying cardiomyocyte apoptosis and provide new ideas for the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Jinghui Sun
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ru Wang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Tiantian Chao
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chenglong Wang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
4
|
Aoyama J, Homma K, Tanabe N, Usui S, Miyagi Y, Matsuura K, Kaneda M, Nitta T. Spatiotemporal imaging documented the maturation of the cardiomyocytes from human induced pluripotent stem cells. J Thorac Cardiovasc Surg 2019; 159:2260-2271.e7. [PMID: 31409490 DOI: 10.1016/j.jtcvs.2019.06.060] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 06/10/2019] [Accepted: 06/14/2019] [Indexed: 12/20/2022]
Abstract
OBJECTIVES Cardiomyocytes derived from human induced pluripotent stem cells are a promising source of cells for regenerative medicine. However, contractions in such derived cardiomyocytes are often irregular and asynchronous, especially at early stages of differentiation. This study aimed to determine the differentiation stage of initiation of synchronized and regular contractions, using spatiotemporal imaging and physiological and genetic analyses. METHODS Knock-in human induced pluripotent stem cell lines were established with clustered regularly interspaced short palindromic repeats/clustered regularly interspaced short palindromic repeats-associated protein 9 to analyze cardiac and pacemaker cell maturation. Time-frequency analysis and Ca2+ imaging were performed, and the expression of related proteins and specific cardiac/pacemaker mRNAs in contracting embryoid bodies was analyzed at various differentiation stages. RESULTS Time-frequency analysis and Ca2+ imaging revealed irregular, asynchronous contractions at the early stage of differentiation with altered electrophysiological properties upon differentiation. Genes associated with electrophysiological properties were upregulated after 70 days of culturing in differentiation media, whereas pacemaker genes were initially upregulated during the early stage and downregulated at the later stage. CONCLUSIONS A differentiation period >70 days is required for adequate development of cardiac elements including ion channels and gap junctions and for sarcomere maturation.
Collapse
Affiliation(s)
- Junya Aoyama
- Department of Cardiovascular Surgery, Nippon Medical School, Tokyo, Japan; Department of Physiology, Nippon Medical School, Tokyo, Japan
| | - Kohei Homma
- Department of Physiology, Nippon Medical School, Tokyo, Japan; Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Nari Tanabe
- SUWA, Tokyo University of Science, Nagano, Japan
| | - Sumiko Usui
- Department of Physiology, Nippon Medical School, Tokyo, Japan
| | - Yasuo Miyagi
- Department of Cardiovascular Surgery, Nippon Medical School, Tokyo, Japan
| | - Katsuhisa Matsuura
- Department of Cardiology, Tokyo Women's Medical University, Tokyo, Japan
| | - Makoto Kaneda
- Department of Physiology, Nippon Medical School, Tokyo, Japan
| | - Takashi Nitta
- Department of Cardiovascular Surgery, Nippon Medical School, Tokyo, Japan.
| |
Collapse
|
5
|
Yang SC, Liu JJ, Wang CK, Lin YT, Tsai SY, Chen WJ, Huang WK, Tu PWA, Lin YC, Chang CF, Cheng CL, Lin H, Lai CY, Lin CY, Lee YH, Chiu YC, Hsu CC, Hsu SC, Hsiao M, Schuyler SC, Lu FL, Lu J. Down-regulation of ATF1 leads to early neuroectoderm differentiation of human embryonic stem cells by increasing the expression level of SOX2. FASEB J 2019; 33:10577-10592. [PMID: 31242772 DOI: 10.1096/fj.201800220rr] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
We reveal by high-throughput screening that activating transcription factor 1 (ATF1) is a novel pluripotent regulator in human embryonic stem cells (hESCs). The knockdown of ATF1 expression significantly up-regulated neuroectoderm (NE) genes but not mesoderm, endoderm, and trophectoderm genes. Of note, down-regulation or knockout of ATF1 with short hairpin RNA (shRNA), small interfering RNA (siRNA), or clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) was sufficient to up-regulate sex-determining region Y-box (SOX)2 and paired box 6 (PAX6) expression under the undifferentiated or differentiated conditions, whereas overexpression of ATF1 suppressed NE differentiation. Endogenous ATF1 was spontaneously down-regulated after d 1-3 of neural induction. By double-knockdown experiments, up-regulation of SOX2 was critical for the increase of PAX6 and SOX1 expression in shRNA targeting Atf1 hESCs. Using the luciferase reporter assay, we identified ATF1 as a negative transcriptional regulator of Sox2 gene expression. A novel function of ATF1 was discovered, and these findings contribute to a broader understanding of the very first steps in regulating NE differentiation in hESCs.-Yang, S.-C., Liu, J.-J., Wang, C.-K., Lin, Y.-T., Tsai, S.-Y., Chen, W.-J., Huang, W.-K., Tu, P.-W. A., Lin, Y.-C., Chang, C.-F., Cheng, C.-L., Lin, H., Lai, C.-Y., Lin, C.-Y., Lee, Y.-H., Chiu, Y.-C., Hsu, C.-C., Hsu, S.-C., Hsiao, M., Schuyler, S. C., Lu, F. L., Lu, J. Down-regulation of ATF1 leads to early neuroectoderm differentiation of human embryonic stem cells by increasing the expression level of SOX2.
Collapse
Affiliation(s)
- Shang-Chih Yang
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan.,Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Jan-Jan Liu
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Cheng-Kai Wang
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan.,Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Yu-Tsen Lin
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Su-Yi Tsai
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - Wei-Ju Chen
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Wei-Kai Huang
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Po-Wen A Tu
- Department of Pediatrics, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Chen Lin
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | | | - Chih-Lun Cheng
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Hsuan Lin
- Genomics Research Center, Academia Sinica, Taipei, Taiwan.,Department of Pediatrics, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chien-Ying Lai
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Chun-Yu Lin
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Yi-Hsuan Lee
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Yen-Chun Chiu
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | | | - Shu-Ching Hsu
- National Institute of Infectious Diseases and Vaccinology, Zhunan, Taiwan.,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Scott C Schuyler
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan City, Taiwan.,Division of Head and Neck Surgery, Department of Otolaryngology, Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| | - Frank Leigh Lu
- Department of Pediatrics, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Jean Lu
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan.,Genomics Research Center, Academia Sinica, Taipei, Taiwan.,RNAi Core, National Core Facility, Academia Sinica, Taipei, Taiwan.,Department of Life Science, Tzu Chi University, Hualien, Taiwan.,Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
6
|
Abou-Saleh H, Zouein FA, El-Yazbi A, Sanoudou D, Raynaud C, Rao C, Pintus G, Dehaini H, Eid AH. The march of pluripotent stem cells in cardiovascular regenerative medicine. Stem Cell Res Ther 2018; 9:201. [PMID: 30053890 PMCID: PMC6062943 DOI: 10.1186/s13287-018-0947-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Cardiovascular disease (CVD) continues to be the leading cause of global morbidity and mortality. Heart failure remains a major contributor to this mortality. Despite major therapeutic advances over the past decades, a better understanding of molecular and cellular mechanisms of CVD as well as improved therapeutic strategies for the management or treatment of heart failure are increasingly needed. Loss of myocardium is a major driver of heart failure. An attractive approach that appears to provide promising results in reducing cardiac degeneration is stem cell therapy (SCT). In this review, we describe different types of stem cells, including embryonic and adult stem cells, and we provide a detailed discussion of the properties of induced pluripotent stem cells (iPSCs). We also present and critically discuss the key methods used for converting somatic cells to pluripotent cells and iPSCs to cardiomyocytes (CMs), along with their advantages and limitations. Integrating and non-integrating reprogramming methods as well as characterization of iPSCs and iPSC-derived CMs are discussed. Furthermore, we critically present various methods of differentiating iPSCs to CMs. The value of iPSC-CMs in regenerative medicine as well as myocardial disease modeling and cardiac regeneration are emphasized.
Collapse
Affiliation(s)
- Haissam Abou-Saleh
- Department of Biological and Environmental Sciences, Qatar University, Doha, Qatar
| | - Fouad A. Zouein
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Ahmed El-Yazbi
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Department of Pharmacology and Toxicology, Alexandria University, Alexandria, Egypt
| | - Despina Sanoudou
- Clinical Genomics and Pharmacogenomics Unit, 4th Department of Internal Medicine, “Attikon” Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Christopher Rao
- Department of Surgery, Queen Elizabeth Hospital, Woolwich, London, UK
| | - Gianfranco Pintus
- Department of Biomedical Sciences, College of Health Sciences, Qatar University, Doha, Qatar
| | - Hassan Dehaini
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Ali H. Eid
- Department of Biological and Environmental Sciences, Qatar University, Doha, Qatar
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Department of Biomedical Sciences, College of Health Sciences, Qatar University, Doha, Qatar
| |
Collapse
|
7
|
Regenerative Stem Cell Therapy Optimization via Tissue Engineering in Heart Failure with Reduced Ejection Fraction. Cardiovasc Eng Technol 2017; 8:515-526. [DOI: 10.1007/s13239-017-0325-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 07/31/2017] [Indexed: 12/30/2022]
|
8
|
Ban K, Bae S, Yoon YS. Current Strategies and Challenges for Purification of Cardiomyocytes Derived from Human Pluripotent Stem Cells. Theranostics 2017. [PMID: 28638487 PMCID: PMC5479288 DOI: 10.7150/thno.19427] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Cardiomyocytes (CMs) derived from human pluripotent stem cells (hPSCs) are considered a most promising option for cell-based cardiac repair. Hence, various protocols have been developed for differentiating hPSCs into CMs. Despite remarkable improvement in the generation of hPSC-CMs, without purification, these protocols can only generate mixed cell populations including undifferentiated hPSCs or non-CMs, which may elicit adverse outcomes. Therefore, one of the major challenges for clinical use of hPSC-CMs is the development of efficient isolation techniques that allow enrichment of hPSC-CMs. In this review, we will discuss diverse strategies that have been developed to enrich hPSC-CMs. We will describe major characteristics of individual hPSC-CM purification methods including their scientific principles, advantages, limitations, and needed improvements. Development of a comprehensive system which can enrich hPSC-CMs will be ultimately useful for cell therapy for diseased hearts, human cardiac disease modeling, cardiac toxicity screening, and cardiac tissue engineering.
Collapse
|
9
|
Shen X, Pan B, Zhou H, Liu L, Lv T, Zhu J, Huang X, Tian J. Differentiation of mesenchymal stem cells into cardiomyocytes is regulated by miRNA-1-2 via WNT signaling pathway. J Biomed Sci 2017; 24:29. [PMID: 28490365 PMCID: PMC5424345 DOI: 10.1186/s12929-017-0337-9] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 05/03/2017] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Bone marrow derived stem cells (BMSCs) have the potential to differentiate into cardiomyocytes, but the rate of differentiation is low and the mechanism of differentiation is unclear completely. Here, we aimed to investigate the role of miR1-2 in differentiation of mouse BMSCs into cardiomyocyte-like cells and reveal the involved signaling pathways in the procedure. METHODS Mouse BMSCs were treated with miR1-2 and 5-azacytine (5-aza). The expression of cardiac cell markers: NKx2.5, cTnI and GATA4 in BMSCs were examined by qPCR. The apoptosis rate was detected by flow cytometry and the activity of the Wnt/β-catenin signaling pathway was evaluated by measuring the upstream protein of this signaling pathway. RESULTS After over-expression of miR1-2 in mouse BMSCs, the apoptosis rate was significantly lower than the 5-aza group, while the expressions of cardiac-specific genes: such as Nkx2.5, cTnI and GATA4 were significantly increased compared to the control group and the 5-aza group. Meanwhile, over-expression of miR1-2 in mouse BMSCs enhanced the expression of wnt11, JNK, β-catenin and TCF in the Wnt/β-catenin signaling pathway. Use of LGK-974, an inhibitor of Wnt/β-catenin signaling pathway, significantly reduced the expression of cardiac-specific genes and partially blocked the role of the miR1-2. CONCLUSION Over-expression of miR1-2 in mouse BMSCs can induce them toward promoted cardiomyocyte differentiation via the activation of the Wnt/β-catenin signaling pathway. Compared to 5-aza, miR1-2 can induce differentiation of BMSCs into cardiomyocytes more effectively with a less cytotoxicity.
Collapse
Affiliation(s)
- Xing Shen
- Department of Cardiology, Heart Centre, The Children's Hospital of Chongqing Medical University, 136 Zhongshan Er Road, Chongqing, 400014, Yu Zhong District, China.,Department of Pediatrics, the Affiliated Hospital of Southwest Medical University, LuZhou, Sichuan, 646000, China
| | - Bo Pan
- Department of Cardiology, Heart Centre, The Children's Hospital of Chongqing Medical University, 136 Zhongshan Er Road, Chongqing, 400014, Yu Zhong District, China
| | - Huiming Zhou
- Department of Cardiology, Heart Centre, The Children's Hospital of Chongqing Medical University, 136 Zhongshan Er Road, Chongqing, 400014, Yu Zhong District, China
| | - Lingjuan Liu
- Department of Cardiology, Heart Centre, The Children's Hospital of Chongqing Medical University, 136 Zhongshan Er Road, Chongqing, 400014, Yu Zhong District, China
| | - Tiewei Lv
- Department of Cardiology, Heart Centre, The Children's Hospital of Chongqing Medical University, 136 Zhongshan Er Road, Chongqing, 400014, Yu Zhong District, China
| | - Jing Zhu
- Department of Cardiology, Heart Centre, The Children's Hospital of Chongqing Medical University, 136 Zhongshan Er Road, Chongqing, 400014, Yu Zhong District, China
| | - Xupei Huang
- Department of Biomedical Science, Charlie E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, 33431, USA
| | - Jie Tian
- Department of Cardiology, Heart Centre, The Children's Hospital of Chongqing Medical University, 136 Zhongshan Er Road, Chongqing, 400014, Yu Zhong District, China.
| |
Collapse
|
10
|
Zhang X, Cao H, Bai S, Huo W, Ma Y. Differentiation and characterization of rhesus monkey atrial and ventricular cardiomyocytes from induced pluripotent stem cells. Stem Cell Res 2017; 20:21-29. [PMID: 28249229 DOI: 10.1016/j.scr.2017.02.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Revised: 02/02/2017] [Accepted: 02/08/2017] [Indexed: 02/07/2023] Open
Abstract
The combination of non-human primate animals and their induced pluripotent stem cell derived cardiomyocytes (iPSC-CMs) provides not only transplantation models for cell-based therapy of heart diseases, but also opportunities for heart-related drug research on both cellular and animal levels. However, the subtypes and electrophysiology properties of non-human primate iPSC-CMs hadn't been detailed characterized. In this study, we generated rhesus monkey induced pluripotent stem cells (riPSCs), and efficiently differentiated them into ventricular or atrial cardiomyocytes by modulating retinoic acid (RA) pathways. Our results revealed that the electrophysiological characteristics and response to canonical drugs of riPSC-CMs were similar with those of human pluripotent stem cell derived CMs. Therefore, rhesus monkeys and their iPSC-CMs provide a powerful and practicable system for heart related biomedical research.
Collapse
Affiliation(s)
- Xiaoqian Zhang
- University of Chinese Academy of Sciences, Beijing 100049, China; National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing 100101, China
| | - Henghua Cao
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing 100101, China
| | - Shuyun Bai
- University of Chinese Academy of Sciences, Beijing 100049, China; National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing 100101, China
| | - Weibang Huo
- University of Chinese Academy of Sciences, Beijing 100049, China; National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing 100101, China
| | - Yue Ma
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing 100101, China.
| |
Collapse
|
11
|
Coyle R, Jia J, Mei Y. Polymer microarray technology for stem cell engineering. Acta Biomater 2016; 34:60-72. [PMID: 26497624 PMCID: PMC4811723 DOI: 10.1016/j.actbio.2015.10.030] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 09/10/2015] [Accepted: 10/19/2015] [Indexed: 12/12/2022]
Abstract
Stem cells hold remarkable promise for applications in tissue engineering and disease modeling. During the past decade, significant progress has been made in developing soluble factors (e.g., small molecules and growth factors) to direct stem cells into a desired phenotype. However, the current lack of suitable synthetic materials to regulate stem cell activity has limited the realization of the enormous potential of stem cells. This can be attributed to a large number of materials properties (e.g., chemical structures and physical properties of materials) that can affect stem cell fate. This makes it challenging to design biomaterials to direct stem cell behavior. To address this, polymer microarray technology has been developed to rapidly identify materials for a variety of stem cell applications. In this article, we summarize recent developments in polymer array technology and their applications in stem cell engineering. STATEMENT OF SIGNIFICANCE Stem cells hold remarkable promise for applications in tissue engineering and disease modeling. In the last decade, significant progress has been made in developing chemically defined media to direct stem cells into a desired phenotype. However, the current lack of the suitable synthetic materials to regulate stem cell activities has been limiting the realization of the potential of stem cells. This can be attributed to the number of variables in material properties (e.g., chemical structures and physical properties) that can affect stem cells. Polymer microarray technology has shown to be a powerful tool to rapidly identify materials for a variety of stem cell applications. Here we summarize recent developments in polymer array technology and their applications in stem cell engineering.
Collapse
Affiliation(s)
- Robert Coyle
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA; Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Jia Jia
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA; Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Ying Mei
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA; Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA.
| |
Collapse
|
12
|
Jiang B, Xiang Z, Ai Z, Wang H, Li Y, Ji W, Li T. Generation of cardiac spheres from primate pluripotent stem cells in a small molecule-based 3D system. Biomaterials 2015; 65:103-14. [DOI: 10.1016/j.biomaterials.2015.06.024] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 06/08/2015] [Accepted: 06/12/2015] [Indexed: 12/21/2022]
|
13
|
Szebényi K, Péntek A, Erdei Z, Várady G, Orbán TI, Sarkadi B, Apáti Á. Efficient generation of human embryonic stem cell-derived cardiac progenitors based on tissue-specific enhanced green fluorescence protein expression. Tissue Eng Part C Methods 2015; 21:35-45. [PMID: 24734786 DOI: 10.1089/ten.tec.2013.0646] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Cardiac progenitor cells (CPCs) are committed to the cardiac lineage but retain their proliferative capacity before becoming quiescent mature cardiomyocytes (CMs). In medical therapy and research, the use of human pluripotent stem cell-derived CPCs would have several advantages compared with mature CMs, as the progenitors show better engraftment into existing heart tissues, and provide unique potential for cardiovascular developmental as well as for pharmacological studies. Here, we demonstrate that the CAG promoter-driven enhanced green fluorescence protein (EGFP) reporter system enables the identification and isolation of embryonic stem cell-derived CPCs. Tracing of CPCs during differentiation confirmed up-regulation of surface markers, previously described to identify cardiac precursors and early CMs. Isolated CPCs express cardiac lineage-specific transcripts, still have proliferating capacity, and can be re-aggregated into embryoid body-like structures (CAG-EGFP(high) rEBs). Expression of troponin T and NKX2.5 mRNA is up-regulated in long-term cultured CAG-EGFP(high) rEBs, in which more than 90% of the cells become Troponin I positive mature CMs. Moreover, about one third of the CAG-EGFP(high) rEBs show spontaneous contractions. The method described here provides a powerful tool to generate expandable cultures of pure human CPCs that can be used for exploring early markers of the cardiac lineage, as well as for drug screening or tissue engineering applications.
Collapse
Affiliation(s)
- Kornélia Szebényi
- 1 Institute of Molecular Pharmacology , Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | | | | | | | | | | | | |
Collapse
|
14
|
Tan Y, Richards D, Xu R, Stewart-Clark S, Mani SK, Borg TK, Menick DR, Tian B, Mei Y. Silicon nanowire-induced maturation of cardiomyocytes derived from human induced pluripotent stem cells. NANO LETTERS 2015; 15:2765-72. [PMID: 25826336 PMCID: PMC4431939 DOI: 10.1021/nl502227a] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
The current inability to derive mature cardiomyocytes from human pluripotent stem cells has been the limiting step for transitioning this powerful technology into clinical therapies. To address this, scaffold-based tissue engineering approaches have been utilized to mimic heart development in vitro and promote maturation of cardiomyocytes derived from human pluripotent stem cells. While scaffolds can provide 3D microenvironments, current scaffolds lack the matched physical/chemical/biological properties of native extracellular environments. On the other hand, scaffold-free, 3D cardiac spheroids (i.e., spherical-shaped microtissues) prepared by seeding cardiomyocytes into agarose microwells were shown to improve cardiac functions. However, cardiomyocytes within the spheroids could not assemble in a controlled manner and led to compromised, unsynchronized contractions. Here, we show, for the first time, that incorporation of a trace amount (i.e., ∼0.004% w/v) of electrically conductive silicon nanowires (e-SiNWs) in otherwise scaffold-free cardiac spheroids can form an electrically conductive network, leading to synchronized and significantly enhanced contraction (i.e., >55% increase in average contraction amplitude), resulting in significantly more advanced cellular structural and contractile maturation.
Collapse
Affiliation(s)
- Yu Tan
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | - Dylan Richards
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | - Ruoyu Xu
- Department of Chemistry, the James Franck Institute and the Institute for Biophysical Dynamics, the University of Chicago, Chicago, IL 60637, USA
| | | | - Santhosh Kumar Mani
- Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute, Medical University of South Carolina, Charleston SC 29425, USA
| | - Thomas Keith Borg
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Donald R. Menick
- Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute, Medical University of South Carolina, Charleston SC 29425, USA
| | - Bozhi Tian
- Department of Chemistry, the James Franck Institute and the Institute for Biophysical Dynamics, the University of Chicago, Chicago, IL 60637, USA
| | - Ying Mei
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
15
|
Ban K, Park HJ, Kim S, Andukuri A, Cho KW, Hwang JW, Cha HJ, Kim SY, Kim WS, Jun HW, Yoon YS. Cell therapy with embryonic stem cell-derived cardiomyocytes encapsulated in injectable nanomatrix gel enhances cell engraftment and promotes cardiac repair. ACS NANO 2014; 8:10815-25. [PMID: 25210842 PMCID: PMC4212793 DOI: 10.1021/nn504617g] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 09/11/2014] [Indexed: 05/25/2023]
Abstract
A significant barrier to the therapeutic use of stem cells is poor cell retention in vivo. Here, we evaluate the therapeutic potential and long-term engraftment of cardiomyocytes (CMs) derived from mouse embryonic stem cells (mESCs) encapsulated in an injectable nanomatrix gel consisting of peptide amphiphiles incorporating cell adhesive ligand Arg-Gly-Asp-Ser (PA-RGDS) in experimental myocardial infarction (MI). We cultured rat neonatal CMs in PA-RGDS for 7 days and found that more than 90% of the CMs survived. Next, we intramyocardially injected mouse CM cell line HL-1 CMs with or without PA-RGDS into uninjured hearts. Histologic examination and flow cytometry analysis of digested heart tissues showed approximately 3-fold higher engraftment in the mice that received CMs with PA-RGDS compared to those without PA-RGDS. We further investigated the therapeutic effects and long-term engraftment of mESC-CMs with PA-RGDS on MI in comparison with PBS control, CM-only, and PA-RGDS only. Echocardiography demonstrated that the CM-only and CM+PA-RGDS groups showed higher cardiac function at week 2 compared to other groups. However, from 3 weeks, higher cardiac function was maintained only in the CM+PA-RGDS group; this was sustained for 12 weeks. Confocal microscopic examination of the cardiac tissues harvested at 14 weeks demonstrated sustained engraftment and integration of mESC-CMs into host myocardium in the CM+PA-RGDS group only. This study for the first time demonstrated that PA-RGDS encapsulation can enhance survival of mESC-derived CMs and improve cardiac function post-MI. This nanomatrix gel-mediated stem cell therapy can be a promising option for treating MI.
Collapse
Affiliation(s)
- Kiwon Ban
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Hun-Jun Park
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sangsung Kim
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Adinarayana Andukuri
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Kyu-Won Cho
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Jung Wook Hwang
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Ho Jin Cha
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Sang Yoon Kim
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Woan-Sang Kim
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Ho-Wook Jun
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama 35203, United States
| | - Young-Sup Yoon
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| |
Collapse
|
16
|
Lee TJ, Kang S, Jeong GJ, Yoon JK, Bhang SH, Oh J, Kim BS. Incorporation of gold-coated microspheres into embryoid body of human embryonic stem cells for cardiomyogenic differentiation. Tissue Eng Part A 2014; 21:374-81. [PMID: 25065511 DOI: 10.1089/ten.tea.2014.0015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Human embryonic stem cells (hESCs) are a useful cell source for cardiac regeneration by stem cell therapy. In this study, we show that incorporation of gold-coated microspheres into hESC-derived embryoid bodies (EBs) enhances the cardiomyogenic differentiation process of pluripotent embryonic stem cells. A polycaprolactone (PCL) microsphere surface was coated with gold. Either gold-coated PCL microspheres (AuMS) or PCL microspheres (MS) were incorporated into hESC-derived EBs. AuMS and MS were not cytotoxic. AuMS promoted the expression of genes for mesodermal and cardiac mesodermal lineage cells, both of which are intermediates in the process of cardiac differentiation of hESCs on day 4 and the expression of cardiomyogenic differentiation markers on day 14 compared to MS. AuMS also enhanced gene expression of cardiac-specific extracellular matrices. Incorporation of gold-coated MS into hESC-derived EBs may provide a new platform for inducing cardiomyogenic differentiation of pluripotent embryonic stem cells.
Collapse
Affiliation(s)
- Tae-Jin Lee
- 1 Engineering Research Institute, Seoul National University , Seoul, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
17
|
Lee TJ, Park S, Bhang SH, Yoon JK, Jo I, Jeong GJ, Hong BH, Kim BS. Graphene enhances the cardiomyogenic differentiation of human embryonic stem cells. Biochem Biophys Res Commun 2014; 452:174-80. [PMID: 25152405 DOI: 10.1016/j.bbrc.2014.08.062] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 08/15/2014] [Indexed: 12/11/2022]
Abstract
Graphene has drawn attention as a substrate for stem cell culture and has been reported to stimulate the differentiation of multipotent adult stem cells. Here, we report that graphene enhances the cardiomyogenic differentiation of human embryonic stem cells (hESCs) at least in part, due to nanoroughness of graphene. Large-area graphene on glass coverslips was prepared via the chemical vapor deposition method. The coating of the graphene with vitronectin (VN) was required to ensure high viability of the hESCs cultured on the graphene. hESCs were cultured on either VN-coated glass (glass group) or VN-coated graphene (graphene group) for 21 days. The cells were also cultured on glass coated with Matrigel (Matrigel group), which is a substrate used in conventional, directed cardiomyogenic differentiation systems. The culture of hESCs on graphene promoted the expression of genes involved in the stepwise differentiation into mesodermal and endodermal lineage cells and subsequently cardiomyogenic differentiation compared with the culture on glass or Matrigel. In addition, the culture on graphene enhanced the gene expression of cardiac-specific extracellular matrices. Culture on graphene may provide a new platform for the development of stem cell therapies for ischemic heart diseases by enhancing the cardiomyogenic differentiation of hESCs.
Collapse
Affiliation(s)
- Tae-Jin Lee
- Engineering Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Subeom Park
- Department of Chemistry, Seoul National University, Seoul, Republic of Korea
| | - Suk Ho Bhang
- School of Chemical Engineering, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - Jeong-Kee Yoon
- School of Chemical and Biological Engineering, Seoul National University, Seoul, Republic of Korea
| | - Insu Jo
- Department of Chemistry, Seoul National University, Seoul, Republic of Korea
| | - Gun-Jae Jeong
- School of Chemical and Biological Engineering, Seoul National University, Seoul, Republic of Korea
| | - Byung Hee Hong
- Department of Chemistry, Seoul National University, Seoul, Republic of Korea.
| | - Byung-Soo Kim
- Engineering Research Institute, Seoul National University, Seoul, Republic of Korea; School of Chemical and Biological Engineering, Seoul National University, Seoul, Republic of Korea; Institute of Bioengineering, Institute of Chemical Processes, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
18
|
Martens A, Rojas SV, Baraki H, Rathert C, Schecker N, Hernandez SR, Schwanke K, Zweigerdt R, Martin U, Saito S, Haverich A, Kutschka I. Macroscopic fluorescence imaging: a novel technique to monitor retention and distribution of injected microspheres in an experimental model of ischemic heart failure. PLoS One 2014; 9:e101775. [PMID: 25089764 PMCID: PMC4121070 DOI: 10.1371/journal.pone.0101775] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 06/11/2014] [Indexed: 11/19/2022] Open
Abstract
Background The limited effectiveness of cardiac cell therapy has generated concern regarding its clinical relevance. Experimental studies show that cell retention and engraftment are low after injection into ischemic myocardium, which may restrict therapy effectiveness significantly. Surgical aspects and mechanical loss are suspected to be the main culprits behind this phenomenon. As current techniques of monitoring intramyocardial injections are complex and time-consuming, the aim of the study was to develop a fast and simple model to study cardiac retention and distribution following intramyocardial injections. For this purpose, our main hypothesis was that macroscopic fluorescence imaging could adequately serve as a detection method for intramyocardial injections. Methods and Results A total of 20 mice underwent ligation of the left anterior descending artery (LAD) for myocardial infarction. Fluorescent microspheres with cellular dimensions were used as cell surrogates. Particles (5×105) were injected into the infarcted area of explanted resting hearts (Ex vivo myocardial injetions EVMI, n = 10) and in vivo into beating hearts (In vivo myocardial injections IVMI, n = 10). Microsphere quantification was performed by fluorescence imaging of explanted organs. Measurements were repeated after a reduction to homogenate dilutions. Cardiac microsphere retention was 2.78×105±0.31×105 in the EVMI group. In the IVMI group, cardiac retention of microspheres was significantly lower (0.74×105±0.18×105; p<0.05). Direct fluorescence imaging revealed venous drainage through the coronary sinus, resulting in a microsphere accumulation in the left (0.90×105±0.20×105) and the right (1.07×105±0.17×105) lung. Processing to homogenates involved further particle loss (p<0.05) in both groups. Conclusions We developed a fast and simple direct fluorescence imaging method for biodistribution analysis which enabled the quantification of fluorescent microspheres after intramyocardial delivery using macroscopic fluorescence imaging. This new technique showed massive early particle loss and venous drainage into the right atrium leading to substantial accumulation of graft particles in both lungs.
Collapse
Affiliation(s)
- Andreas Martens
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
- Leibniz Research Laboratories for Biotechnology and Artificial Organs, Hannover Medical School, Hannover, Germany
| | - Sebastian V. Rojas
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
- Leibniz Research Laboratories for Biotechnology and Artificial Organs, Hannover Medical School, Hannover, Germany
- * E-mail:
| | - Hassina Baraki
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Christian Rathert
- Leibniz Research Laboratories for Biotechnology and Artificial Organs, Hannover Medical School, Hannover, Germany
| | - Natalie Schecker
- Leibniz Research Laboratories for Biotechnology and Artificial Organs, Hannover Medical School, Hannover, Germany
| | - Sara Rojas Hernandez
- Department of Anaesthesiology and Intensive Care Medicine, Hannover Medical School, Hannover, Germany
| | - Kristin Schwanke
- Leibniz Research Laboratories for Biotechnology and Artificial Organs, Hannover Medical School, Hannover, Germany
| | - Robert Zweigerdt
- Leibniz Research Laboratories for Biotechnology and Artificial Organs, Hannover Medical School, Hannover, Germany
| | - Ulrich Martin
- Leibniz Research Laboratories for Biotechnology and Artificial Organs, Hannover Medical School, Hannover, Germany
| | - Shunsuke Saito
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Axel Haverich
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
- Leibniz Research Laboratories for Biotechnology and Artificial Organs, Hannover Medical School, Hannover, Germany
| | - Ingo Kutschka
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
- Leibniz Research Laboratories for Biotechnology and Artificial Organs, Hannover Medical School, Hannover, Germany
| |
Collapse
|
19
|
Wang Y, Qian DEJ, Zhong WY, Lu JH, Guo XK, Cao YL, Liu J. TGF-β1 induces the formation of vascular-like structures in embryoid bodies derived from human embryonic stem cells. Exp Ther Med 2014; 8:52-58. [PMID: 24944596 PMCID: PMC4061233 DOI: 10.3892/etm.2014.1721] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Accepted: 05/14/2014] [Indexed: 12/17/2022] Open
Abstract
Human embryonic stem cells (ESCs) can differentiate into endothelial cells in response to stimuli from extracellular cytokines. Transforming growth factor (TGF)-β1 signaling is involved in stem cell renewal and vascular development. Previously, human ESCs were isolated from inner cell mass and a stable ESC line was developed. In the present study, the effects of extracellular TGF-β1 were investigated on human ESC-derived embryoid bodies (EB) in suspension. The structures of the EBs were analyzed with light and electron microscopy, while the cellular composition of the EBs was examined via the expression levels of specific markers. Vascular-like tubular structures and cardiomyocyte-like beating cells were observed in the EBs at day 3 and 8, respectively. The frequencies of vascular-like structures and beating cells in the TGF-β1 treated group were significantly higher compared with the control group (84.31 vs. 12.77%; P<0.001; 37.25 vs. 8.51%; P<0.001, respectively). Electron microscopy revealed the presence of lumens and gap junctions in the sections of the tubular structures. Semiquantitative polymerase chain reaction revealed elevated expression levels of CD31 and fetal liver kinase-1 in EBs cultured with TGF-β1. In addition, extensive staining of von Willebrand factor was observed in the vascular-like structures of TGF-β1-treated EBs. Therefore, the results of the present study may aid the understanding of the underlying mechanisms of human ESC differentiation and improve the methods of propagating specific cell types for the clinical therapy of cardiovascular diseases.
Collapse
Affiliation(s)
- Yan Wang
- Department of Plastic Surgery, Provincial Qianfoshan Hospital Affiliated to Shandong University, Jinan, Shandong 250014, P.R. China
| | - DE-Jian Qian
- Department of Plastic Surgery, Provincial Qianfoshan Hospital Affiliated to Shandong University, Jinan, Shandong 250014, P.R. China
| | - Wen-Yu Zhong
- Department of Gynecology and Obstetrics, Jinan Central Hospital, Jinan, Shandong 250013, P.R. China
| | - Jun-Hong Lu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai 200011, P.R. China
| | - Xiang-Kai Guo
- Department of Plastic Surgery, Provincial Qianfoshan Hospital Affiliated to Shandong University, Jinan, Shandong 250014, P.R. China
| | - Yi-Lin Cao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai 200011, P.R. China
| | - Ju Liu
- Department of Plastic Surgery, Provincial Qianfoshan Hospital Affiliated to Shandong University, Jinan, Shandong 250014, P.R. China ; Medical Research Center, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
| |
Collapse
|
20
|
Ye D, Peramo A. Protocol for serial cultivation of epithelial cells without enzymes or chemical compounds. Methods Mol Biol 2014; 1195:23-32. [PMID: 24519003 DOI: 10.1007/7651_2013_41] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Deriving keratinocytes from epidermis or oral mucosa is a critical first step in the construction of cell-based tissue engineering and regenerative medicine applications. It would be advantageous to develop a methodology to grow adult somatic cells with maximum plasticity in a rapid fashion and in large numbers with minimal manipulation. With routine methods, keratinocytes are cultured in standard amount of medium and passaged with enzymes, and the confluence of the monolayer induces differentiation and eventual cell death. A protocol to expand keratinocytes in culture by growing keratinocyte in large numbers using a technique in which keratinocytes are released into the overlaying medium, effectively "popping-up" into suspension from the cell monolayer, is described in this chapter. This technique does not require the use of enzymes or chemical compounds for serial cultivation. The cells possess the ability of active cell proliferation at 100 % confluence over 1-2 months' time. Based on previous characterization reports, these are untransformed, normal keratinocytes that appear to be highly suitable for clinical applications.
Collapse
Affiliation(s)
- Dongxia Ye
- Shanghai Ninth People's Hospital, Medical School of Shanghai Jiaotong University, Shanghai, China,
| | | |
Collapse
|
21
|
Castro-Sesquen YE, Gilman RH, Paico H, Yauri V, Angulo N, Ccopa F, Bern C. Cell death and serum markers of collagen metabolism during cardiac remodeling in Cavia porcellus experimentally infected with Trypanosoma cruzi. PLoS Negl Trop Dis 2013; 7:e1996. [PMID: 23409197 PMCID: PMC3566988 DOI: 10.1371/journal.pntd.0001996] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2012] [Accepted: 11/21/2012] [Indexed: 01/19/2023] Open
Abstract
We studied cell death by apoptosis and necrosis in cardiac remodeling produced by Trypanosoma cruzi infection. In addition, we evaluated collagen I, III, IV (CI, CIII and CIV) deposition in cardiac tissue, and their relationship with serum levels of procollagen type I carboxy-terminal propeptide (PICP) and procollagen type III amino-terminal propeptide (PIIINP). Eight infected and two uninfected guinea pigs were necropsied at seven time points up to one year post-infection. Cell death by necrosis and apoptosis was determined by histopathological observation and terminal deoxynucleotidyl transferase dUTP nick end labeling, respectively. Deposition of cardiac collagen types was determined by immunohistochemistry and serum levels of PICP, PIIINP, and anti-T. cruzi IgG1 and IgG2 by ELISA. IgG2 (Th1 response) predominated throughout the course of infection; IgG1 (Th2 response) was detected during the chronic phase. Cardiac cell death by necrosis predominated over apoptosis during the acute phase; during the chronic phase, both apoptosis and necrosis were observed in cardiac cells. Apoptosis was also observed in lymphocytes, endothelial cells and epicardial adipose tissue, especially in the chronic phase. Cardiac levels of CI, CIII, CIV increased progressively, but the highest levels were seen in the chronic phase and were primarily due to increase in CIII and CIV. High serum levels of PICP and PIIINP were observed throughout the infection, and increased levels of both biomarkers were associated with cardiac fibrosis (p = 0.002 and p = 0.038, respectively). These results confirm the role of apoptosis in cell loss mainly during the chronic phase and the utility of PICP and PIIINP as biomarkers of fibrosis in cardiac remodeling during T. cruzi infection. Chronic Chagas heart disease (CHHD) caused by the infection with the parasite Trypanosoma cruzi is the most important infectious heart disease in the world. The typical manifestations are dilated cardiomyopathy and congestive heart failure; they result from death of cardiomyocytes and their replacement by collagen. Knowing the mechanisms of cardiomyocyte death is important for the development of therapies that prevent them. The contribution of apoptosis in cardiomyocyte death was evaluated in the guinea pig model of T. cruzi infection, and the detection of serum levels of collagen precursors were evaluated as biomarkers of cardiac fibrosis. We observed apoptosis of lymphocytes, cardiomyocytes, endothelial cells and epicardial adipose tissue in cardiac tissue of infected guinea pigs. The increase of serum levels of collagen precursors PICP and PIIINP were associated with cardiac fibrosis. Areas of inflammation and apoptosis of epicardial adipose tissue were associated with cardiac pathology, which suggests the importance of epicardial adipose tissue in CCHD. These results show that apoptosis is an important characteristic of cardiac cell death during CCHD and serum levels of PICP and PIIINP could be used as biomarkers of cardiac fibrosis.
Collapse
Affiliation(s)
- Yagahira E. Castro-Sesquen
- Laboratorio de Investigación en Enfermedades Infecciosas, Universidad Peruana Cayetano Heredia, Lima, Peru
- Asociación Benéfica PRISMA, Lima, Peru
| | - Robert H. Gilman
- Department of International Health, Johns Hopkins University, Bloomberg School of Hygiene and Public Health, Baltimore, Maryland, United States of America
- * E-mail:
| | - Henry Paico
- Laboratorio de Investigación en Enfermedades Infecciosas, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Verónica Yauri
- School of Veterinary Medicine, Universidad Nacional Mayor de San Marcos, Lima, Peru
| | - Noelia Angulo
- Laboratorio de Investigación en Enfermedades Infecciosas, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Fredy Ccopa
- Laboratorio de Investigación en Enfermedades Infecciosas, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Caryn Bern
- Global Health Sciences and Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California, United States of America
| |
Collapse
|
22
|
Parsons XH. Constraining the Pluripotent Fate of Human Embryonic Stem Cells for Tissue Engineering and Cell Therapy - The Turning Point of Cell-Based Regenerative Medicine. ACTA ACUST UNITED AC 2013. [PMID: 24926434 PMCID: PMC4051304 DOI: 10.9734/bbj/2013/4309] [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: 12/28/2022]
Abstract
To date, the lack of a clinically-suitable source of engraftable human stem/progenitor cells with adequate neurogenic potential has been the major setback in developing safe and effective cell-based therapies for regenerating the damaged or lost CNS structure and circuitry in a wide range of neurological disorders. Similarly, the lack of a clinically-suitable human cardiomyocyte source with adequate myocardium regenerative potential has been the major setback in regenerating the damaged human heart. Given the limited capacity of the CNS and heart for self-repair, there is a large unmet healthcare need to develop stem cell therapies to provide optimal regeneration and reconstruction treatment options to restore normal tissues and function. Derivation of human embryonic stem cells (hESCs) provides a powerful in vitro model system to investigate molecular controls in human embryogenesis as well as an unlimited source to generate the diversity of human somatic cell types for regenerative medicine. However, realizing the developmental and therapeutic potential of hESC derivatives has been hindered by the inefficiency and instability of generating clinically-relevant functional cells from pluripotent cells through conventional uncontrollable and incomplete multi-lineage differentiation. Recent advances and breakthroughs in hESC research have overcome some major obstacles in bringing hESC therapy derivatives towards clinical applications, including establishing defined culture systems for de novo derivation and maintenance of clinical-grade pluripotent hESCs and lineage-specific differentiation of pluripotent hESCs by small molecule induction. Retinoic acid was identified as sufficient to induce the specification of neuroectoderm direct from the pluripotent state of hESCs and trigger a cascade of neuronal lineage-specific progression to human neuronal progenitors and neurons of the developing CNS in high efficiency, purity, and neuronal lineage specificity by promoting nuclear translocation of the neuronal specific transcription factor Nurr-1. Similarly, nicotinamide was rendered sufficient to induce the specification of cardiomesoderm direct from the pluripotent state of hESCs by promoting the expression of the earliest cardiac-specific transcription factor Csx/Nkx2.5 and triggering progression to cardiac precursors and beating cardiomyocytes with high efficiency. This technology breakthrough enables direct conversion of pluripotent hESCs into a large supply of high purity neuronal cells or heart muscle cells with adequate capacity to regenerate CNS neurons and contractile heart muscles for developing safe and effective stem cell therapies. Transforming pluripotent hESCs into fate-restricted therapy derivatives dramatically increases the clinical efficacy of graft-dependent repair and safety of hESC-derived cellular products. Such milestone advances and medical innovations in hESC research allow generation of a large supply of clinical-grade hESC therapy derivatives targeting for major health problems, bringing cell-based regenerative medicine to a turning point.
Collapse
Affiliation(s)
- Xuejun H Parsons
- San Diego Regenerative Medicine Institute, San Diego, CA 92109, USA. ; Xcelthera, San Diego, CA 92109, USA
| |
Collapse
|
23
|
Parsons XH. Constraining the Pluripotent Fate of Human Embryonic Stem Cells for Tissue Engineering and Cell Therapy - The Turning Point of Cell-Based Regenerative Medicine. ACTA ACUST UNITED AC 2013; 3:424-457. [PMID: 24926434 DOI: 10.9734/bbj/2013/4309#sthash.6d8rulbv.dpuf] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
To date, the lack of a clinically-suitable source of engraftable human stem/progenitor cells with adequate neurogenic potential has been the major setback in developing safe and effective cell-based therapies for regenerating the damaged or lost CNS structure and circuitry in a wide range of neurological disorders. Similarly, the lack of a clinically-suitable human cardiomyocyte source with adequate myocardium regenerative potential has been the major setback in regenerating the damaged human heart. Given the limited capacity of the CNS and heart for self-repair, there is a large unmet healthcare need to develop stem cell therapies to provide optimal regeneration and reconstruction treatment options to restore normal tissues and function. Derivation of human embryonic stem cells (hESCs) provides a powerful in vitro model system to investigate molecular controls in human embryogenesis as well as an unlimited source to generate the diversity of human somatic cell types for regenerative medicine. However, realizing the developmental and therapeutic potential of hESC derivatives has been hindered by the inefficiency and instability of generating clinically-relevant functional cells from pluripotent cells through conventional uncontrollable and incomplete multi-lineage differentiation. Recent advances and breakthroughs in hESC research have overcome some major obstacles in bringing hESC therapy derivatives towards clinical applications, including establishing defined culture systems for de novo derivation and maintenance of clinical-grade pluripotent hESCs and lineage-specific differentiation of pluripotent hESCs by small molecule induction. Retinoic acid was identified as sufficient to induce the specification of neuroectoderm direct from the pluripotent state of hESCs and trigger a cascade of neuronal lineage-specific progression to human neuronal progenitors and neurons of the developing CNS in high efficiency, purity, and neuronal lineage specificity by promoting nuclear translocation of the neuronal specific transcription factor Nurr-1. Similarly, nicotinamide was rendered sufficient to induce the specification of cardiomesoderm direct from the pluripotent state of hESCs by promoting the expression of the earliest cardiac-specific transcription factor Csx/Nkx2.5 and triggering progression to cardiac precursors and beating cardiomyocytes with high efficiency. This technology breakthrough enables direct conversion of pluripotent hESCs into a large supply of high purity neuronal cells or heart muscle cells with adequate capacity to regenerate CNS neurons and contractile heart muscles for developing safe and effective stem cell therapies. Transforming pluripotent hESCs into fate-restricted therapy derivatives dramatically increases the clinical efficacy of graft-dependent repair and safety of hESC-derived cellular products. Such milestone advances and medical innovations in hESC research allow generation of a large supply of clinical-grade hESC therapy derivatives targeting for major health problems, bringing cell-based regenerative medicine to a turning point.
Collapse
Affiliation(s)
- Xuejun H Parsons
- San Diego Regenerative Medicine Institute, San Diego, CA 92109, USA. ; Xcelthera, San Diego, CA 92109, USA
| |
Collapse
|
24
|
Characterization of cultured epithelial cells using a novel technique not requiring enzymatic digestion for subculturing. Cell Tissue Bank 2012; 14:423-35. [PMID: 23149549 DOI: 10.1007/s10561-012-9343-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Accepted: 10/06/2012] [Indexed: 11/27/2022]
Abstract
Our laboratory had developed a methodology to expand epithelial cells in culture by growing keratinocyte monolayers, under large volumes of medium that produces large numbers of keratinocytes that leave the monolayer and move into suspension. The cells have been defined as epithelial Pop Up Keratinocytes or ePUKs cells and appear to be highly suitable for clinical applications. In this publication we extend the characterization of the cells with a detailed analysis of the capabilities of the monolayer of a single culture flask to produce, over time, ePUK cells. The cells were characterized using standard epithelial markers for proliferation and differentiation. Analysis of morphology of the monolayer formed and total number of cells produced is presented for a variety of human epithelial cell strains. These keratinocytes provide an additional controlled human cell system for investigation of the mechanisms regulating epithelia cell growth and differentiation and since they are produced in large numbers, they are highly suitable for use in epithelial cell banking.
Collapse
|
25
|
Minami I, Yamada K, Otsuji TG, Yamamoto T, Shen Y, Otsuka S, Kadota S, Morone N, Barve M, Asai Y, Tenkova-Heuser T, Heuser JE, Uesugi M, Aiba K, Nakatsuji N. A small molecule that promotes cardiac differentiation of human pluripotent stem cells under defined, cytokine- and xeno-free conditions. Cell Rep 2012; 2:1448-60. [PMID: 23103164 DOI: 10.1016/j.celrep.2012.09.015] [Citation(s) in RCA: 185] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Revised: 05/18/2012] [Accepted: 09/12/2012] [Indexed: 12/15/2022] Open
Abstract
Human pluripotent stem cells (hPSCs), including embryonic stem cells and induced pluripotent stem cells, are potentially useful in regenerative therapies for heart disease. For medical applications, clinical-grade cardiac cells must be produced from hPSCs in a defined, cost-effective manner. Cell-based screening led to the discovery of KY02111, a small molecule that promotes differentiation of hPSCs to cardiomyocytes. Although the direct target of KY02111 remains unknown, results of the present study suggest that KY02111 promotes differentiation by inhibiting WNT signaling in hPSCs but in a manner that is distinct from that of previously studied WNT inhibitors. Combined use of KY02111 and WNT signaling modulators produced robust cardiac differentiation of hPSCs in a xeno-free, defined medium, devoid of serum and any kind of recombinant cytokines and hormones, such as BMP4, Activin A, or insulin. The methodology has potential as a means for the practical production of human cardiomyocytes for regeneration therapies.
Collapse
Affiliation(s)
- Itsunari Minami
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto 606-8501, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Mummery CL, Zhang J, Ng ES, Elliott DA, Elefanty AG, Kamp TJ. Differentiation of human embryonic stem cells and induced pluripotent stem cells to cardiomyocytes: a methods overview. Circ Res 2012; 111:344-58. [PMID: 22821908 DOI: 10.1161/circresaha.110.227512] [Citation(s) in RCA: 548] [Impact Index Per Article: 42.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Since human embryonic stem cells were first differentiated to beating cardiomyocytes a decade ago, interest in their potential applications has increased exponentially. This has been further enhanced over recent years by the discovery of methods to induce pluripotency in somatic cells, including those derived from patients with hereditary cardiac diseases. Human pluripotent stem cells have been among the most challenging cell types to grow stably in culture, but advances in reagent development now mean that most laboratories can expand both embryonic and induced pluripotent stem cells robustly using commercially available products. However, differentiation protocols have lagged behind and in many cases only produce the cell types required with low efficiency. Cardiomyocyte differentiation techniques were also initially inefficient and not readily transferable across cell lines, but there are now a number of more robust protocols available. Here, we review the basic biology underlying the differentiation of pluripotent cells to cardiac lineages and describe current state-of-the-art protocols, as well as ongoing refinements. This should provide a useful entry for laboratories new to this area to start their research. Ultimately, efficient and reliable differentiation methodologies are essential to generate desired cardiac lineages to realize the full promise of human pluripotent stem cells for biomedical research, drug development, and clinical applications.
Collapse
Affiliation(s)
- Christine L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands
| | | | | | | | | | | |
Collapse
|
27
|
Mei Y. Microarrayed Materials for Stem Cells. MATERIALS TODAY (KIDLINGTON, ENGLAND) 2012; 15:10.1016/S1369-7021(12)70196-7. [PMID: 24311967 PMCID: PMC3848960 DOI: 10.1016/s1369-7021(12)70196-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Stem cells hold remarkable promise for applications in disease modeling, cancer therapy and regenerative medicine. Despite the significant progress made during the last decade, designing materials to control stem cell fate remains challenging. As an alternative, materials microarray technology has received great attention because it allows for high throughput materials synthesis and screening at a reasonable cost. Here, we discuss recent developments in materials microarray technology and their applications in stem cell engineering. Future opportunities in the field will also be reviewed.
Collapse
Affiliation(s)
- Ying Mei
- Clemson-MUSC Bioengineering Program, Department of Bioengineering, Clemson University, Charleston, SC 29425, USA
| |
Collapse
|
28
|
Marcelo CL, Peramo A, Ambati A, Feinberg SE. Characterization of a unique technique for culturing primary adult human epithelial progenitor/"stem cells". BMC DERMATOLOGY 2012; 12:8. [PMID: 22726819 PMCID: PMC3441704 DOI: 10.1186/1471-5945-12-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2011] [Accepted: 06/13/2012] [Indexed: 01/19/2023]
Abstract
Background Primary keratinocytes derived from epidermis, oral mucosa, and urothelium are used in construction of cell based wound healing devices and in regenerative medicine. This study presents in vitro technology that rapidly expands keratinocytes in culture by growing monolayers under large volumes of serum-free, essential fatty acid free, low calcium medium that is replaced every 24 hrs. Methods Primary cell cultures were produced from epidermal skin, oral mucosa and ureter by trypsinization of tissue. Cells were grown using Epilife medium with growth factors under high medium volumes. Once densely confluent, the keratinocyte monolayer produced cells in suspension in the overlying medium that can be harvested every 24 hrs. over a 7–10 day period. The cell suspension (approximately 8 X 105 cells/ml) is poured into a new flask to form another confluent monolayer over 2–4 days. This new culture, in turn produced additional cell suspensions that when serially passed expand the cell strain over 2–3 months, without the use of enzymes to split the cultures. The cell suspension, called epithelial Pop Up Keratinocytes (ePUKs) were analyzed for culture expansion, cell size and glucose utilization, attachment to carrier beads, micro-spheroid formation, induction of keratinocyte differentiation, and characterized by immunohistochemistry. Results The ePUKs expanded greatly in culture, attached to carrier beads, did not form micro-spheroids, used approximately 50% of medium glucose over 24 hrs., contained a greater portion of smaller diameter cells (8–10 microns), reverted to classical appearing cultures when returned to routine feeding schedules (48 hrs. and 15 ml/T-75 flask) and can be differentiated by either adding 1.2 mM medium calcium, or essential fatty acids. The ePUK cells are identified as cycling (Ki67 expressing) basal cells (p63, K14 expressing). Conclusions Using this primary culture technique, large quantities of epithelial cells can be generated without the use of the enzyme trypsin to split the cultures. The cells are small in diameter and have basal cell progenitor/”stem” (P/SC) cell characteristics induced by daily feeding with larger than normal medium volumes. The ePUK epithelial cells have the potential to be used in regenerative medicine and for basic studies of epithelia P/SC phenotype.
Collapse
Affiliation(s)
- Cynthia Luz Marcelo
- Department of Surgery, Section of Plastic and Reconstructive Surgery, University of Michigan, MSRBII, A560 1150 W, Medical Center Dr,, Ann Arbor, MI, 48109, USA.
| | | | | | | |
Collapse
|
29
|
Parsons JF, Smotrich DB, Gonzalez R, Snyder EY, Moore DA, Parsons XH. Defining Conditions for Sustaining Epiblast Pluripotence Enables Direct Induction of Clinically-Suitable Human Myocardial Grafts from Biologics-Free Human Embryonic Stem Cells. ACTA ACUST UNITED AC 2012; S9. [PMID: 22905333 DOI: 10.4172/2155-9880.s9-001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
To date, lacking of a clinically-suitable human cardiac cell source with adequate myocardium regenerative potential has been the major setback in regenerating the damaged human myocardium. Pluripotent Human Embryonic Stem Cells (hESCs) proffer unique revenue to generate a large supply of cardiac lineage-committed cells as human myocardial grafts for cell-based therapy. Due to the prevalence of heart disease worldwide and acute shortage of donor organs or human myocardial grafts, there is intense interest in developing hESC-based therapy for heart disease and failure. However, realizing the potential of hESCs has been hindered by the inefficiency and instability of generating cardiac cells from pluripotent cells through uncontrollable multi-lineage differentiation. In addition, the need for foreign biologics for derivation, maintenance, and differentiation of hESCs may make direct use of such cells and their derivatives in patients problematic. Understanding the requirements for sustaining pluripotentce and self-renewal of hESCs will provide the foundation for de novo derivation and long-term maintenance of biologics-free hESCs under optimal yet well-defined culture conditions from which they can be efficiently directed towards clinically-relevant lineages for therapies. We previously reported the resolving of the elements of a defined culture system, serving as a platform for effectively directing pluripotent hESCs uniformly towards a cardiac lineage-specific fate by small molecule induction. In this study, we found that, under the defined culture conditions, primitive endoderm-like (PEL) cells constitutively emerged and acted through the activin-A-SMAD pathway in a paracrine fashion to sustain the epiblast pluripotence of hESCs. Such defined conditions enable the spontaneous unfolding of inherent early embryogenesis processes that, in turn, aid efficient clonal propagation and de novo derivation of stable biologics-free hESCs from blastocysts that can be directly differentiated into a large supply of clinically-suitable human myocardial grafts across the spectrum of developmental stages using small molecule induction for cardiovascular repair.
Collapse
Affiliation(s)
- James F Parsons
- San Diego Regenerative Medicine Institute, San Diego, CA 92109, USA
| | | | | | | | | | | |
Collapse
|
30
|
Kodo K, Yamagishi H. A decade of advances in the molecular embryology and genetics underlying congenital heart defects. Circ J 2011; 75:2296-304. [PMID: 21914956 DOI: 10.1253/circj.cj-11-0636] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Congenital heart defects (CHD) are the most common type of human birth defect and result in significant mortality worldwide. Despite numerous epidemiologic studies in the past decades, few genetic causes have been identified until recently. CHD result from abnormal morphogenesis of the systematic cardiovascular construction during development. Recent advances in molecular embryology, including the discovery of a new source of cardiac progenitor cells termed the second heart field (SHF), have revealed that the heart arises from multiple distinct embryonic origins. Cells derived from the SHF contribute to the development of the cardiac outflow tract, together with the other progenitor cell lineage called cardiac neural crest cells. Numerous cardiac transcription factors regulate these progenitor cells during heart development. Elucidation of the transcriptional network for these cardiac progenitor cells is essential for further understanding cardiac development and providing new insights into the morphogenesis of CHD. This review outlines the recent discoveries of the molecular embryology of the normal heart and the genetic basis of CHD.
Collapse
Affiliation(s)
- Kazuki Kodo
- Department of Pediatrics, Division of Pediatric Cardiology, Keio University School of Medicine, Japan
| | | |
Collapse
|
31
|
Zhang WY, Ebert AD, Narula J, Wu JC. Imaging cardiac stem cell therapy: translations to human clinical studies. J Cardiovasc Transl Res 2011; 4:514-22. [PMID: 21538182 PMCID: PMC3657500 DOI: 10.1007/s12265-011-9281-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2011] [Accepted: 04/14/2011] [Indexed: 12/21/2022]
Abstract
Stem cell therapy promises to open exciting new options in the treatment of cardiovascular diseases. Although feasible and clinically safe, the in vivo behavior and integration of stem cell transplants still remain largely unknown. Thus, the development of innovative non-invasive imaging techniques capable of effectively tracking such therapy in vivo is vital for a more in-depth investigation into future clinical applications. Such imaging modalities will not only generate further insight into the mechanisms behind stem cell-based therapy, but also address some major concerns associated with translational cardiovascular stem cell therapy. In the present review, we summarize the principles underlying three major stem cell tracking methods: (1) radioactive labeling for positron emission tomography (PET) and single photon emission computed tomography (SPECT) imaging, (2) iron particle labeling for magnetic resonance imaging (MRI), and (3) reporter gene labeling for bioluminescence, fluorescence, MRI, SPECT, and PET imaging. We then discuss recent clinical studies that have utilized these modalities to gain biological insights into stem cell fate.
Collapse
Affiliation(s)
- Wendy Y. Zhang
- Department of Medicine (Division of Cardiology), Stanford University School of Medicine
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine
| | - Antje D. Ebert
- Department of Medicine (Division of Cardiology), Stanford University School of Medicine
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine
| | - Jagat Narula
- Division of Cardiology, UC Irvine Medical Center, Orange, California, USA
| | - Joseph C. Wu
- Department of Medicine (Division of Cardiology), Stanford University School of Medicine
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine
| |
Collapse
|
32
|
Chew SY, Low WC. Scaffold-based approach to direct stem cell neural and cardiovascular differentiation: An analysis of physical and biochemical effects. J Biomed Mater Res A 2011; 97:355-74. [DOI: 10.1002/jbm.a.33064] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2010] [Revised: 01/11/2011] [Accepted: 01/24/2011] [Indexed: 01/12/2023]
|
33
|
Forte E, Chimenti I, Barile L, Gaetani R, Angelini F, Ionta V, Messina E, Giacomello A. Cardiac Cell Therapy: The Next (Re)Generation. Stem Cell Rev Rep 2011; 7:1018-30. [DOI: 10.1007/s12015-011-9252-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
34
|
Fujita T, Ishikawa Y. Apoptosis in Heart Failure - The Role of the .BETA.-Adrenergic Receptor-Mediated Signaling Pathway and p53-Mediated Signaling Pathway in the Apoptosis of Cardiomyocytes -. Circ J 2011; 75:1811-1818. [DOI: 10.1253/circj.cj-11-0025] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Affiliation(s)
- Takayuki Fujita
- Cardiovascular Research Institute, Yokohama City University Graduate School of Medicine
| | - Yoshihiro Ishikawa
- Cardiovascular Research Institute, Yokohama City University Graduate School of Medicine
| |
Collapse
|
35
|
Yu L, Gao S, Nie L, Tang M, Huang W, Luo H, Hu X, Xi J, Zhu M, Zheng Y, Gao L, Zhang L, Song Y, Hescheler J, Liang H. Molecular and Functional Changes in Voltage-Gated Na+ Channels in Cardiomyocytes During Mouse Embryogenesis. Circ J 2011; 75:2071-9. [DOI: 10.1253/circj.cj-10-1212] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Liangzhu Yu
- Chinese-German Stem Cell Center, Department of Physiology, Tongji Medical College, Huazhong University of Science and Technology
| | - Shijun Gao
- Chinese-German Stem Cell Center, Department of Physiology, Tongji Medical College, Huazhong University of Science and Technology
| | - Li Nie
- Chinese-German Stem Cell Center, Department of Physiology, Tongji Medical College, Huazhong University of Science and Technology
| | - Ming Tang
- Chinese-German Stem Cell Center, Department of Physiology, Tongji Medical College, Huazhong University of Science and Technology
| | - Weifeng Huang
- Chinese-German Stem Cell Center, Department of Physiology, Tongji Medical College, Huazhong University of Science and Technology
| | - Hongyan Luo
- Chinese-German Stem Cell Center, Department of Physiology, Tongji Medical College, Huazhong University of Science and Technology
| | - Xinwu Hu
- Chinese-German Stem Cell Center, Department of Physiology, Tongji Medical College, Huazhong University of Science and Technology
| | - Jiaoya Xi
- Chinese-German Stem Cell Center, Department of Physiology, Tongji Medical College, Huazhong University of Science and Technology
| | - Minjie Zhu
- Chinese-German Stem Cell Center, Department of Physiology, Tongji Medical College, Huazhong University of Science and Technology
| | - Yunjie Zheng
- Chinese-German Stem Cell Center, Department of Physiology, Tongji Medical College, Huazhong University of Science and Technology
| | - Linlin Gao
- Chinese-German Stem Cell Center, Department of Physiology, Tongji Medical College, Huazhong University of Science and Technology
| | - Lanqiu Zhang
- Chinese-German Stem Cell Center, Department of Physiology, Tongji Medical College, Huazhong University of Science and Technology
| | - Yuanlong Song
- Chinese-German Stem Cell Center, Department of Physiology, Tongji Medical College, Huazhong University of Science and Technology
| | | | - Huamin Liang
- Chinese-German Stem Cell Center, Department of Physiology, Tongji Medical College, Huazhong University of Science and Technology
| |
Collapse
|