1
|
Hashemi M, Finklea FB, Hammons H, Tian Y, Young N, Kim E, Halloin C, Triebert W, Zweigerdt R, Mitra AK, Lipke EA. Hydrogel microsphere stem cell encapsulation enhances cardiomyocyte differentiation and functionality in scalable suspension system. Bioact Mater 2025; 43:423-440. [PMID: 39399838 PMCID: PMC11471139 DOI: 10.1016/j.bioactmat.2024.08.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 08/30/2024] [Accepted: 08/31/2024] [Indexed: 10/15/2024] Open
Abstract
A reliable suspension-based platform for scaling engineered cardiac tissue (ECT) production from human induced pluripotent stem cells (hiPSCs) is crucial for regenerative therapies. Here, we compared the production and functionality of ECTs formed using our scaffold-based, engineered tissue microsphere differentiation approach with those formed using the prevalent scaffold-free aggregate platform. We utilized a microfluidic system for the rapid (1 million cells/min), high density (30, 40, 60 million cells/ml) encapsulation of hiPSCs within PEG-fibrinogen hydrogel microspheres. HiPSC-laden microspheres and aggregates underwent suspension-based cardiac differentiation in chemically defined media. In comparison to aggregates, microspheres maintained consistent size and shape initially, over time, and within and between batches. Initial size and shape coefficients of variation for microspheres were eight and three times lower, respectively, compared to aggregates. On day 10, microsphere cardiomyocyte (CM) content was 27 % higher and the number of CMs per initial hiPSC was 250 % higher than in aggregates. Contraction and relaxation velocities of microspheres were four and nine times higher than those of aggregates, respectively. Microsphere contractile functionality also improved with culture time, whereas aggregate functionality remained unchanged. Additionally, microspheres displayed improved β-adrenergic signaling responsiveness and uniform calcium transient propagation. Transcriptomic analysis revealed that while both microspheres and aggregates demonstrated similar gene regulation patterns associated with cardiomyocyte differentiation, heart development, cardiac muscle contraction, and sarcomere organization, the microspheres exhibited more pronounced transcriptional changes over time. Taken together, these results highlight the capability of the microsphere platform for scaling up biomanufacturing of ECTs in a suspension-based culture platform.
Collapse
Affiliation(s)
| | - Ferdous B. Finklea
- Department of Chemical Engineering, Auburn University, Auburn, AL, United States
| | - Hanna Hammons
- Department of Chemical Engineering, Auburn University, Auburn, AL, United States
| | - Yuan Tian
- Department of Chemical Engineering, Auburn University, Auburn, AL, United States
| | - Nathan Young
- Department of Chemical Engineering, Auburn University, Auburn, AL, United States
| | - Emma Kim
- Department of Chemical Engineering, Auburn University, Auburn, AL, United States
| | - Caroline Halloin
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hanover, Germany
| | - Wiebke Triebert
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hanover, Germany
| | - Robert Zweigerdt
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hanover, Germany
| | - Amit Kumar Mitra
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL, United States
| | - Elizabeth A. Lipke
- Department of Chemical Engineering, Auburn University, Auburn, AL, United States
| |
Collapse
|
2
|
Finklea FB, Hashemi M, Tian Y, Hammons H, Halloin C, Triebert W, Zweigerdt R, Lipke EA. Chemically defined production of engineered cardiac tissue microspheres from hydrogel-encapsulated pluripotent stem cells. Biotechnol Bioeng 2024; 121:3614-3628. [PMID: 39104025 DOI: 10.1002/bit.28818] [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: 07/11/2023] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 08/07/2024]
Abstract
Chemically defined, suspension culture conditions are a key requirement in realizing clinical translation of engineered cardiac tissues (ECTs). Building on our previous work producing functional ECT microspheres through differentiation of biomaterial encapsulated human induced pluripotent stem cells (hiPSCs), here we establish the ability to use chemically defined culture conditions, including stem cell media (E8) and cardiac differentiation media (chemically defined differentiation media with three components, CDM3). A custom microfluidic cell encapsulation system was used to encapsulate hiPSCs at a range of initial cell concentrations and diameters in the hybrid biomaterial, poly(ethylene glycol)-fibrinogen (PF), for the formation of highly spherical and uniform ECT microspheres for subsequent cardiac differentiation. Initial microsphere diameter could be tightly controlled, and microspheres could be produced with an initial diameter between 400 and 800 µm. Three days after encapsulation, cardiac differentiation was initiated through small molecule modulation of Wnt signaling in CDM3. Cardiac differentiation occurred resulting in in situ ECT formation; results showed that this differentiation protocol could be used to achieve cardiomyocyte (CM) contents greater than 90%, although there was relatively high variability in CM content and yield between differentiation batches. Spontaneous contraction of ECT microspheres initiated between Days 7 and 10 of differentiation and ECT microspheres responded to electrical pacing up to 1.5 Hz. Resulting CMs had well-defined sarcomeres and the gap junction protein, connexin 43, and had appropriate temporal changes in gene expression. In summary, this study demonstrated the proof-of-concept to produce functional ECT microspheres with chemically defined media in suspension culture in combination with biomaterial support of microsphere encapsulated hiPSCs.
Collapse
Affiliation(s)
- Ferdous B Finklea
- Department of Chemical Engineering, Auburn University, Auburn, Alabama, USA
| | | | - Yuan Tian
- Department of Chemical Engineering, Auburn University, Auburn, Alabama, USA
| | - Hanna Hammons
- Department of Chemical Engineering, Auburn University, Auburn, Alabama, USA
| | - Caroline Halloin
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hanover, Germany
| | - Wiebke Triebert
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hanover, Germany
| | - Robert Zweigerdt
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hanover, Germany
| | - Elizabeth A Lipke
- Department of Chemical Engineering, Auburn University, Auburn, Alabama, USA
| |
Collapse
|
3
|
Riebel LL, Wang ZJ, Martinez-Navarro H, Trovato C, Camps J, Berg LA, Zhou X, Doste R, Sachetto Oliveira R, Weber Dos Santos R, Biasetti J, Rodriguez B. In silico evaluation of cell therapy in acute versus chronic infarction: role of automaticity, heterogeneity and Purkinje in human. Sci Rep 2024; 14:21584. [PMID: 39284812 PMCID: PMC11405404 DOI: 10.1038/s41598-024-67951-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 07/17/2024] [Indexed: 09/22/2024] Open
Abstract
Human-based modelling and simulation offer an ideal testbed for novel medical therapies to guide experimental and clinical studies. Myocardial infarction (MI) is a common cause of heart failure and mortality, for which novel therapies are urgently needed. Although cell therapy offers promise, electrophysiological heterogeneity raises pro-arrhythmic safety concerns, where underlying complex spatio-temporal dynamics cannot be investigated experimentally. Here, after demonstrating credibility of the modelling and simulation framework, we investigate cell therapy in acute versus chronic MI and the role of cell heterogeneity, scar size and the Purkinje system. Simulations agreed with experimental and clinical recordings from ionic to ECG dynamics in acute and chronic infarction. Following cell delivery, spontaneous beats were facilitated by heterogeneity in cell populations, chronic MI due to tissue depolarisation and slow sinus rhythm. Subsequent re-entrant arrhythmias occurred, in some instances with Purkinje involvement and their susceptibility was enhanced by impaired Purkinje-myocardium coupling, large scars and acute infarction. We conclude that homogeneity in injected ventricular-like cell populations minimises their spontaneous beating, which is enhanced by chronic MI, whereas a healthy Purkinje-myocardium coupling is key to prevent subsequent re-entrant arrhythmias, particularly for large scars.
Collapse
Affiliation(s)
| | | | | | - Cristian Trovato
- Department of Computer Science, University of Oxford, Oxford, UK
- Systems Medicine, Clinical Pharmacology & Safety Science, R&D, AstraZeneca, Cambridge, UK
| | - Julia Camps
- Department of Computer Science, University of Oxford, Oxford, UK
| | - Lucas Arantes Berg
- Department of Computer Science, University of Oxford, Oxford, UK
- Universidade Federal de Juiz de Fora, Juiz de Fora, Brazil
| | - Xin Zhou
- Department of Computer Science, University of Oxford, Oxford, UK
| | - Ruben Doste
- Department of Computer Science, University of Oxford, Oxford, UK
| | | | | | - Jacopo Biasetti
- Systems Medicine, Clinical Pharmacology & Safety Science, R&D, AstraZeneca, Gothenburg, Sweden
| | - Blanca Rodriguez
- Department of Computer Science, University of Oxford, Oxford, UK.
| |
Collapse
|
4
|
Farboud SP, Fathi E, Valipour B, Farahzadi R. Toward the latest advancements in cardiac regeneration using induced pluripotent stem cells (iPSCs) technology: approaches and challenges. J Transl Med 2024; 22:783. [PMID: 39175068 PMCID: PMC11342568 DOI: 10.1186/s12967-024-05499-8] [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: 04/16/2024] [Accepted: 07/10/2024] [Indexed: 08/24/2024] Open
Abstract
A novel approach to treating heart failures was developed with the introduction of iPSC technology. Knowledge in regenerative medicine, developmental biology, and the identification of illnesses at the cellular level has exploded since the discovery of iPSCs. One of the most frequent causes of mortality associated with cardiovascular disease is the loss of cardiomyocytes (CMs), followed by heart failure. A possible treatment for heart failure involves restoring cardiac function and replacing damaged tissue with healthy, regenerated CMs. Significant strides in stem cell biology during the last ten years have transformed the in vitro study of human illness and enhanced our knowledge of the molecular pathways underlying human disease, regenerative medicine, and drug development. We seek to examine iPSC advancements in disease modeling, drug discovery, iPSC-Based cell treatments, and purification methods in this article.
Collapse
Affiliation(s)
- Seyedeh Parya Farboud
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Ezzatollah Fathi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran.
| | - Behnaz Valipour
- Department of Anatomical Sciences, Sarab Faculty of Medical Sciences, Sarab, Iran
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Raheleh Farahzadi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
5
|
Prondzynski M, Berkson P, Trembley MA, Tharani Y, Shani K, Bortolin RH, Sweat ME, Mayourian J, Yucel D, Cordoves AM, Gabbin B, Hou C, Anyanwu NJ, Nawar F, Cotton J, Milosh J, Walker D, Zhang Y, Lu F, Liu X, Parker KK, Bezzerides VJ, Pu WT. Efficient and reproducible generation of human iPSC-derived cardiomyocytes and cardiac organoids in stirred suspension systems. Nat Commun 2024; 15:5929. [PMID: 39009604 PMCID: PMC11251028 DOI: 10.1038/s41467-024-50224-0] [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: 02/13/2023] [Accepted: 07/01/2024] [Indexed: 07/17/2024] Open
Abstract
Human iPSC-derived cardiomyocytes (hiPSC-CMs) have proven invaluable for cardiac disease modeling and regeneration. Challenges with quality, inter-batch consistency, cryopreservation and scale remain, reducing experimental reproducibility and clinical translation. Here, we report a robust stirred suspension cardiac differentiation protocol, and we perform extensive morphological and functional characterization of the resulting bioreactor-differentiated iPSC-CMs (bCMs). Across multiple different iPSC lines, the protocol produces 1.2E6/mL bCMs with ~94% purity. bCMs have high viability after cryo-recovery (>90%) and predominantly ventricular identity. Compared to standard monolayer-differentiated CMs, bCMs are more reproducible across batches and have more mature functional properties. The protocol also works with magnetically stirred spinner flasks, which are more economical and scalable than bioreactors. Minor protocol modifications generate cardiac organoids fully in suspension culture. These reproducible, scalable, and resource-efficient approaches to generate iPSC-CMs and organoids will expand their applications, and our benchmark data will enable comparison to cells produced by other cardiac differentiation protocols.
Collapse
Affiliation(s)
| | - Paul Berkson
- Department of Cardiology, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Michael A Trembley
- Department of Cardiology, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Yashasvi Tharani
- Department of Cardiology, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Kevin Shani
- Disease Biophysics Group, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA, 02134, USA
| | - Raul H Bortolin
- Department of Cardiology, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Mason E Sweat
- Department of Cardiology, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Joshua Mayourian
- Department of Cardiology, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Dogacan Yucel
- Department of Cardiology, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Albert M Cordoves
- Disease Biophysics Group, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA, 02134, USA
| | - Beatrice Gabbin
- Department of Cardiology, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Cuilan Hou
- Department of Cardiology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Cardiology, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062, China
| | - Nnaemeka J Anyanwu
- Disease Biophysics Group, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA, 02134, USA
| | - Farina Nawar
- Department of Cardiology, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Justin Cotton
- Department of Cardiology, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Joseph Milosh
- Department of Cardiology, Boston Children's Hospital, Boston, MA, 02115, USA
| | - David Walker
- Department of Cardiology, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Yan Zhang
- Department of Cardiology, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Fujian Lu
- Department of Cardiology, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Xujie Liu
- Department of Cardiology, Boston Children's Hospital, Boston, MA, 02115, USA
- Fuwai Hospital, Chinese Academy of Medical Science, Shenzhen, Shenzhen, Guangdong Province, 518057, China
| | - Kevin Kit Parker
- Disease Biophysics Group, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA, 02134, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
- Harvard Stem Cell Institute, Cambridge, MA, 02138, USA
| | | | - William T Pu
- Department of Cardiology, Boston Children's Hospital, Boston, MA, 02115, USA.
- Harvard Stem Cell Institute, Cambridge, MA, 02138, USA.
| |
Collapse
|
6
|
Prondzynski M, Bortolin RH, Berkson P, Trembley MA, Shani K, Sweat ME, Mayourian J, Cordoves AM, Anyanwu NJ, Tharani Y, Cotton J, Milosh JB, Walker D, Zhang Y, Liu F, Liu X, Parker KK, Bezzerides VJ, Pu WT. Efficient and reproducible generation of human iPSC-derived cardiomyocytes using a stirred bioreactor. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.24.581789. [PMID: 38464269 PMCID: PMC10925150 DOI: 10.1101/2024.02.24.581789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
In the last decade human iPSC-derived cardiomyocytes (hiPSC-CMs) proved to be valuable for cardiac disease modeling and cardiac regeneration, yet challenges with scale, quality, inter-batch consistency, and cryopreservation remain, reducing experimental reproducibility and limiting clinical translation. Here, we report a robust cardiac differentiation protocol that uses Wnt modulation and a stirred suspension bioreactor to produce on average 124 million hiPSC-CMs with >90% purity using a variety of hiPSC lines (19 differentiations; 10 iPSC lines). After controlled freeze and thaw, bioreactor-derived CMs (bCMs) showed high viability (>90%), interbatch reproducibility in cellular morphology, function, drug response and ventricular identity, which was further supported by single cell transcriptomes. bCMs on microcontact printed substrates revealed a higher degree of sarcomere maturation and viability during long-term culture compared to monolayer-derived CMs (mCMs). Moreover, functional investigation of bCMs in 3D engineered heart tissues showed earlier and stronger force production during long-term culture, and robust pacing capture up to 4 Hz when compared to mCMs. bCMs derived from this differentiation protocol will expand the applications of hiPSC-CMs by providing a reproducible, scalable, and resource efficient method to generate cardiac cells with well-characterized structural and functional properties superior to standard mCMs.
Collapse
Affiliation(s)
| | - Raul H Bortolin
- Department of Cardiology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Paul Berkson
- Department of Cardiology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Michael A Trembley
- Department of Cardiology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Kevin Shani
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences
| | - Mason E Sweat
- Department of Cardiology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Joshua Mayourian
- Department of Cardiology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Albert M Cordoves
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences
| | - Nnaemeka J Anyanwu
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences
| | - Yashasvi Tharani
- Department of Cardiology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Justin Cotton
- Department of Cardiology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Joseph B Milosh
- Department of Cardiology, Boston Children's Hospital, Boston, MA 02115, USA
| | - David Walker
- Department of Cardiology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Yan Zhang
- Department of Cardiology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Fujian Liu
- Department of Cardiology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Xujie Liu
- Department of Cardiology, Boston Children's Hospital, Boston, MA 02115, USA
- Fuwai Hospital, Chinese Academy of Medical Science, Shenzhen. Shenzhen, Guangdong Province, 518057, China
| | - Kevin K Parker
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences
- Harvard Stem Cell Institute, Cambridge, USA
| | | | - William T Pu
- Department of Cardiology, Boston Children's Hospital, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Cambridge, USA
| |
Collapse
|
7
|
Kamali-Dolat Abadi M, Yousefi G, Dehghani F, Alizadeh AA, Jangholi A, Moadab MA, Naseh M, Parsa S, Nasiri G, Azarpira N, Dianatpour M. The Effect of Mesenchymal Stem Cells Derived-Conditioned Media in Combination with Oral Anti-Androgenic Drugs on Male Pattern Baldness: An Animal Study. CELL JOURNAL 2023; 25:790-800. [PMID: 38071411 PMCID: PMC10711289 DOI: 10.22074/cellj.2023.2008138.1377] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 09/16/2023] [Accepted: 10/07/2023] [Indexed: 01/06/2025]
Abstract
OBJECTIVE Androgenetic alopecia (AGA) is a prevalent form of hair loss, mainly caused by follicular sensitivity to androgens. Despite developing different anti-androgen treatment options, the success rate of these treatments has been limited. Using animal models, this study evaluated the therapeutic effects of umbilical cord (UC) stem cell conditioned media (CM) combined with oral anti-androgens for hair regeneration. MATERIALS AND METHODS In this experimental study, Poloxamer 407 (P407) was used as a drug carrier for subcutaneous testosterone injection. AGA models were treated with oral finasteride, oral flutamide, and CM injections. Samples were thoroughly evaluated and compared using histological, stereological, and molecular analyses. RESULTS Injecting CM-loaded hydrogel alone or combined with oral intake of anti-androgens improved hair regeneration. These treatments could promote hair growth by inducing hair follicles in the anagen stage and shortening the telogen and catagen phases. Furthermore, the combination treatment led to an upregulation of hair induction gene expression with a downregulation of inflammation genes. CONCLUSION Through a reduction in inflammation, injection of CM-loaded hydrogel alone or combined with oral intake of anti-androgens induces the hair cell cycle with regeneration in damaged follicles. Hence, this could be a promising therapeutic method for AGA patients.
Collapse
Affiliation(s)
- Majid Kamali-Dolat Abadi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advance Medical Science and Technology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Gholamhossein Yousefi
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Farzaneh Dehghani
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Anatomical Sciences, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Akbar Alizadeh
- Department of Tissue Engineering and applied cell science, School of Advanced Medical Sciences & Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Abolfazl Jangholi
- Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran
| | | | - Maryam Naseh
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shima Parsa
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Golara Nasiri
- Department of Tissue Engineering and Cell Therapy, School of Advanced Technologies in Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mehdi Dianatpour
- Stem Cell Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Medical Genetics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
8
|
Oh MS, Lee SG, Lee GH, Kim CY, Song JH, Yu BY, Chung HM. Verification of Therapeutic Effect through Accelerator Mass Spectrometry-Based Single Cell Level Quantification of hESC-Endothelial Cells Distributed into an Ischemic Model. Adv Healthc Mater 2023; 12:e2300476. [PMID: 37068221 DOI: 10.1002/adhm.202300476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/01/2023] [Indexed: 04/19/2023]
Abstract
As the potential of pluripotent stem cell-derived differentiated cells has been demonstrated in regenerative medicine, differentiated vascular endothelial cells (ECs) are emerging as a therapeutic agent for the cardiovascular system. To verify the therapeutic efficacy of differentiated ECs in an ischemic model, human embryonic stem cells (hESCs) are induced as EC lineage and produce high-purity ECs through fluorescence-activated cell sorting (FACS). When hESC-ECs are transplanted into a hindlimb ischemic model, it is confirmed that blood flow and muscle regeneration are further improved by creating new blood vessels together with autologous ECs than the primary cell as cord blood endothelial progenitor cells (CB-EPCs). In addition, previously reported studies show the detection of transplanted cells engrafted in blood vessels through various tracking methods, but fail to provide accurate quantitative values over time. In this study, it is demonstrated that hESC-ECs are engrafted approximately sevenfold more than CB-EPCs by using an accelerator mass spectrometry (AMS)-based cell tracking technology that can perform quantification at the single cell level. An accurate quantification index is suggested. It has never been reported in in vivo kinetics of hESC-ECs that can act as therapeutic agents.
Collapse
Affiliation(s)
- Min-Seok Oh
- Department of Stem Cell Biology, School of Medicine, Konkuk University, 120 Neungdong-Ro, Gwangjin-Gu, Seoul, 05029, Republic of Korea
- Advanced Analysis and Data Center, Korea Institute of Science and Technology, Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Seul-Gi Lee
- Department of Stem Cell Biology, School of Medicine, Konkuk University, 120 Neungdong-Ro, Gwangjin-Gu, Seoul, 05029, Republic of Korea
| | - Gwan-Ho Lee
- Advanced Analysis and Data Center, Korea Institute of Science and Technology, Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - C-Yoon Kim
- College of Veterinary Medicine, Konkuk University, Gwangjin-Gu, Seoul, 05029, Republic of Korea
| | - Jong Han Song
- Advanced Analysis and Data Center, Korea Institute of Science and Technology, Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Byung-Yong Yu
- Advanced Analysis and Data Center, Korea Institute of Science and Technology, Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Hyung Min Chung
- Department of Stem Cell Biology, School of Medicine, Konkuk University, 120 Neungdong-Ro, Gwangjin-Gu, Seoul, 05029, Republic of Korea
- Mirae Cell Bio Co. Ltd, Seoul, 04795, Republic of Korea
| |
Collapse
|
9
|
Sridharan D, Pracha N, Rana SJ, Ahmed S, Dewani AJ, Alvi SB, Mergaye M, Ahmed U, Khan M. Preclinical Large Animal Porcine Models for Cardiac Regeneration and Its Clinical Translation: Role of hiPSC-Derived Cardiomyocytes. Cells 2023; 12:cells12071090. [PMID: 37048163 PMCID: PMC10093073 DOI: 10.3390/cells12071090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 03/29/2023] [Accepted: 03/30/2023] [Indexed: 04/14/2023] Open
Abstract
Myocardial Infarction (MI) occurs due to a blockage in the coronary artery resulting in ischemia and necrosis of cardiomyocytes in the left ventricular heart muscle. The dying cardiac tissue is replaced with fibrous scar tissue, causing a decrease in myocardial contractility and thus affecting the functional capacity of the myocardium. Treatments, such as stent placements, cardiac bypasses, or transplants are beneficial but with many limitations, and may decrease the overall life expectancy due to related complications. In recent years, with the advent of human induced pluripotent stem cells (hiPSCs), newer avenues using cell-based approaches for the treatment of MI have emerged as a potential for cardiac regeneration. While hiPSCs and their derived differentiated cells are promising candidates, their translatability for clinical applications has been hindered due to poor preclinical reproducibility. Various preclinical animal models for MI, ranging from mice to non-human primates, have been adopted in cardiovascular research to mimic MI in humans. Therefore, a comprehensive literature review was essential to elucidate the factors affecting the reproducibility and translatability of large animal models. In this review article, we have discussed different animal models available for studying stem-cell transplantation in cardiovascular applications, mainly focusing on the highly translatable porcine MI model.
Collapse
Affiliation(s)
- Divya Sridharan
- Department of Emergency Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Nooruddin Pracha
- Department of Emergency Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Schaza Javed Rana
- Department of Emergency Medicine, The Ohio State University, Columbus, OH 43210, USA
- Department of Internal Medicine, Northeast Georgia Medical Center, Gainesville, GA 30501, USA
| | - Salmman Ahmed
- Department of Emergency Medicine, The Ohio State University, Columbus, OH 43210, USA
- Lake Erie College of Osteopathic Medicine (LECOM), Erie, PA 16509, USA
| | - Anam J Dewani
- Department of Emergency Medicine, The Ohio State University, Columbus, OH 43210, USA
- Department of Chemistry & Biochemistry, The University of Toledo, Toledo, OH 43606, USA
| | - Syed Baseeruddin Alvi
- Department of Emergency Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Muhamad Mergaye
- Department of Emergency Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Uzair Ahmed
- Department of Emergency Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Mahmood Khan
- Department of Emergency Medicine, The Ohio State University, Columbus, OH 43210, USA
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
10
|
GAO Z, ZHOU F, MU J. Research Progress towards the Effects of Fatty Acids on the Differentiation and Maturation of Human Induced Pluripotent Stem Cells into Cardiomyocytes. Rev Cardiovasc Med 2023; 24:69. [PMID: 39077493 PMCID: PMC11264038 DOI: 10.31083/j.rcm2403069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 11/18/2022] [Accepted: 11/21/2022] [Indexed: 07/31/2024] Open
Abstract
The incidence of cardiovascular disease has been continuously increasing. Because cardiomyocytes (CM) are non-renewable cells, it is difficult to find appropriate CM sources to repair injured hearts. Research of human induced pluripotent stem cell (hiPSC) differentiation and maturation into CM has been invaluable for the treatment of heart diseases. The use of hiPSCs as regenerative therapy allows for the treatment of many diseases that cannot be cured, including progressive heart failure. This review contributes to the study of cardiac repair and targeted treatment of cardiovascular diseases at the cytological level. Recent studies have shown that for differentiation and maturation of hiPSCs into CMs, fatty acids have a strong influence on cellular metabolism, organelle development, expression of specific genes, and functional performance. This review describes the recent research progress on how fatty acids affect the differentiation of hiPSCs into CMs and their maturation.
Collapse
Affiliation(s)
- Zhen GAO
- Department of Cardiac Surgery, Capital Medical University Affiliated Beijing Anzhen Hospital, Beijing Institute of Heart Lung and Blood Vessel Diseases, 100029 Beijing, China
| | - Fan ZHOU
- Department of Ultrasound, The Third Medical Center of PLA General Hospital, 100039 Beijing, China
| | - Junsheng MU
- Department of Cardiac Surgery, Capital Medical University Affiliated Beijing Anzhen Hospital, Beijing Institute of Heart Lung and Blood Vessel Diseases, 100029 Beijing, China
| |
Collapse
|
11
|
Frljak S, Gozdowska R, Klimczak-Tomaniak D, Kucia M, Kuch M, Jadczyk T, Vrtovec B, Sanz-Ruiz R. Stem Cells in Heart Failure: Future Perspective. CARDIOVASCULAR APPLICATIONS OF STEM CELLS 2023:491-514. [DOI: 10.1007/978-981-99-0722-9_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
12
|
Advances in Cardiac Tissue Engineering. Bioengineering (Basel) 2022; 9:bioengineering9110696. [DOI: 10.3390/bioengineering9110696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/09/2022] [Accepted: 11/15/2022] [Indexed: 11/17/2022] Open
Abstract
Tissue engineering has paved the way for the development of artificial human cardiac muscle patches (hCMPs) and cardiac tissue analogs, especially for treating Myocardial infarction (MI), often by increasing its regenerative abilities. Low engraftment rates, insufficient clinical application scalability, and the creation of a functional vascular system remain obstacles to hCMP implementation in clinical settings. This paper will address some of these challenges, present a broad variety of heart cell types and sources that can be applied to hCMP biomanufacturing, and describe some new innovative methods for engineering such treatments. It is also important to note the injection/transplantation of cells in cardiac tissue engineering.
Collapse
|
13
|
Leowattana W, Leowattana T, Leowattana P. Human-induced pluripotent stem cell-atrial-specific cardiomyocytes and atrial fibrillation. World J Clin Cases 2022; 10:9588-9601. [PMID: 36186184 PMCID: PMC9516943 DOI: 10.12998/wjcc.v10.i27.9588] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/22/2022] [Accepted: 08/16/2022] [Indexed: 02/05/2023] Open
Abstract
Patient-specific human-induced pluripotent stem cell-derived atrial cardiomyocytes (hiPSC-aCMs) may be produced, genome-edited, and differentiated into multiple cell types for regenerative medicine, disease modeling, drug testing, toxicity screening, and three-dimensional tissue fabrication. There is presently no complete model of atrial fibrillation (AF) available for studying human pharmacological responses and evaluating the toxicity of potential medication candidates. It has been demonstrated that hiPSC-aCMs can replicate the electrophysiological disease phenotype and genotype of AF. The hiPSC-aCMs, however, are immature and do not reflect the maturity of aCMs in the native myocardium. Numerous laboratories utilize a variety of methodologies and procedures to improve and promote aCM maturation, including electrical stimulation, culture duration, biophysical signals, and changes in metabolic variables. This review covers the current methods being explored for use in the maturation of patient-specific hiPSC-aCMs and their application towards a personalized approach to the pharmacologic therapy of AF.
Collapse
Affiliation(s)
- Wattana Leowattana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Tawithep Leowattana
- Department of Medicine, Faculty of Medicine, Srinakharinwirot University, Bangkok 10110, Thailand
| | - Pathomthep Leowattana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| |
Collapse
|
14
|
Tsoi C, Deng R, Kwok M, Yan B, Lee C, Li HS, Ma CHY, Luo R, Leung KT, Chan GCF, Chow LMC, Poon EN. Temporal Control of the WNT Signaling Pathway During Cardiac Differentiation Impacts Upon the Maturation State of Human Pluripotent Stem Cell Derived Cardiomyocytes. Front Mol Biosci 2022; 9:714008. [PMID: 35402504 PMCID: PMC8987729 DOI: 10.3389/fmolb.2022.714008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 02/25/2022] [Indexed: 11/18/2022] Open
Abstract
Inefficient differentiation and insufficient maturation are barriers to the application of human pluripotent stem cell (hPSC)-derived cardiomyocytes (CMs) for research and therapy. Great strides have been made to the former, and multiple groups have reported cardiac differentiation protocol that can generate hPSC-CMs at high efficiency. Although many such protocols are based on the modulation of the WNT signaling pathway, they differ in their timing and in the WNT inhibitors used. Little is currently known about whether and how conditions of differentiation affect cardiac maturation. Here we adapted multiple cardiac differentiation protocols to improve cost-effectiveness and consistency, and compared the properties of the hPSC-CMs generated. Our results showed that the schedule of differentiation, but not the choice of WNT inhibitors, was a critical determinant not only of differentiation efficiency, which was expected, but also CM maturation. Among cultures with comparable purity, hPSC-CMs generated with different differentiation schedules vary in the expression of genes which are important for developmental maturation, and in their structural, metabolic, calcium transient and proliferative properties. In summary, we demonstrated that simple changes in the schedule of cardiac differentiation could promote maturation. To this end, we have optimized a cardiac differentiation protocol that can simultaneously achieve high differentiation efficiency and enhanced developmental maturation. Our findings would advance the production of hPSC-CMs for research and therapy.
Collapse
Affiliation(s)
- Chantelle Tsoi
- Centre for Cardiovascular Genomics and Medicine, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong (CUHK), Shatin, Hong Kong SAR, China
- Hong Kong Hub of Paediatric Excellence (HK HOPE), CUHK, Shatin, Hong Kong SAR, China
| | - Ruixia Deng
- Centre for Cardiovascular Genomics and Medicine, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong (CUHK), Shatin, Hong Kong SAR, China
- Hong Kong Hub of Paediatric Excellence (HK HOPE), CUHK, Shatin, Hong Kong SAR, China
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, China
| | - Maxwell Kwok
- Hong Kong Hub of Paediatric Excellence (HK HOPE), CUHK, Shatin, Hong Kong SAR, China
- Department of Medicine and Therapeutics, CUHK, Shatin, Hong Kong SAR, China
| | - Bin Yan
- Department of Computer Science, Faculty of Engineering, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Carrie Lee
- Centre for Cardiovascular Genomics and Medicine, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong (CUHK), Shatin, Hong Kong SAR, China
- Hong Kong Hub of Paediatric Excellence (HK HOPE), CUHK, Shatin, Hong Kong SAR, China
| | - Hung Sing Li
- Centre for Cardiovascular Genomics and Medicine, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong (CUHK), Shatin, Hong Kong SAR, China
- Hong Kong Hub of Paediatric Excellence (HK HOPE), CUHK, Shatin, Hong Kong SAR, China
- Department of Paediatrics, CUHK, Shatin, Hong Kong SAR, China
| | - Chloe Ho Yi Ma
- Hong Kong Hub of Paediatric Excellence (HK HOPE), CUHK, Shatin, Hong Kong SAR, China
- The School of Biomedical Sciences, CUHK, Shatin, Hong Kong SAR, China
| | - Ruibang Luo
- Department of Computer Science, Faculty of Engineering, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Kam Tong Leung
- Hong Kong Hub of Paediatric Excellence (HK HOPE), CUHK, Shatin, Hong Kong SAR, China
- Department of Paediatrics, CUHK, Shatin, Hong Kong SAR, China
| | - Godfrey Chi-Fung Chan
- Department of Pediatrics and Adolescent Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Larry Ming-cheung Chow
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, China
| | - Ellen N. Poon
- Centre for Cardiovascular Genomics and Medicine, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong (CUHK), Shatin, Hong Kong SAR, China
- Hong Kong Hub of Paediatric Excellence (HK HOPE), CUHK, Shatin, Hong Kong SAR, China
- Department of Medicine and Therapeutics, CUHK, Shatin, Hong Kong SAR, China
- The School of Biomedical Sciences, CUHK, Shatin, Hong Kong SAR, China
- *Correspondence: Ellen N. Poon,
| |
Collapse
|
15
|
Conditional immortalization of human atrial myocytes for the generation of in vitro models of atrial fibrillation. Nat Biomed Eng 2022; 6:389-402. [PMID: 34992271 DOI: 10.1038/s41551-021-00827-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 10/29/2021] [Indexed: 12/12/2022]
Abstract
The lack of a scalable and robust source of well-differentiated human atrial myocytes constrains the development of in vitro models of atrial fibrillation (AF). Here we show that fully functional atrial myocytes can be generated and expanded one-quadrillion-fold via a conditional cell-immortalization method relying on lentiviral vectors and the doxycycline-controlled expression of a recombinant viral oncogene in human foetal atrial myocytes, and that the immortalized cells can be used to generate in vitro models of AF. The method generated 15 monoclonal cell lines with molecular, cellular and electrophysiological properties resembling those of primary atrial myocytes. Multicellular in vitro models of AF generated using the immortalized atrial myocytes displayed fibrillatory activity (with activation frequencies of 6-8 Hz, consistent with the clinical manifestation of AF), which could be terminated by the administration of clinically approved antiarrhythmic drugs. The conditional cell-immortalization method could be used to generate functional cell lines from other human parenchymal cells, for the development of in vitro models of human disease.
Collapse
|
16
|
PGC-1α promotes mitochondrial respiration and biogenesis during the differentiation of hiPSCs into cardiomyocytes. Genes Dis 2021; 8:891-906. [PMID: 34522716 PMCID: PMC8427271 DOI: 10.1016/j.gendis.2020.12.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/04/2020] [Accepted: 12/16/2020] [Indexed: 02/07/2023] Open
Abstract
Although it is widely accepted that human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are readily available, robustly reproducible, and physiologically appropriate human cells for clinical applications and research in the cardiovascular field, hiPSC-CMs cultured in vitro retain an immature metabolic phenotype that limits their application, and little is known about the underlying molecular mechanism controlling mitochondrial metabolic maturation during human induced pluripotent stem cells (hiPSCs ) differentiation into cardiomyocytes. In this study, we found that peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α) played an important role in inducing mitochondrial biogenesis and establishing oxidative phosphorylation (OXPHOS) during the cardiac differentiation of hiPSCs. Knocking down PGC-1α by siRNA impaired mitochondrial respiration, while upregulating PGC-1α by ZLN005 promoted mitochondrial biosynthesis and function by regulating the expression of downstream genes involved in mitochondrial dynamics and oxidative metabolism in hiPSC-CMs. Furthermore, we found that estrogen-related receptor α (ERRα) was required for the induction of PGC-1α stimulatory effects in hiPSC-CMs. These findings provide key insights into the molecular control of mitochondrial metabolism during cardiac differentiation and may be used to generate more metabolically mature cardiomyocytes for application.
Collapse
|
17
|
Ho V, Tong G, Lam A, Reuveny S, Oh S. Integrating Human-Induced Pluripotent Stem Cell Expansion Capability and Cardiomyocyte Differentiation Potential in a Microcarrier Suspension Culture. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2021; 2436:67-81. [PMID: 34519977 DOI: 10.1007/7651_2021_423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Human-induced pluripotent stem cells are known for their high proliferation capacity as well as their ability to differentiate to different lineages (Ban et al., Theranostics 7(7):2067-2077, 2017; Chen et al., Stem Cell Res 15(2):365-375, 2015; Serra et al., Trends Biotechnol 30(6):350-359, 2012). For stem-cell-derived cardiomyocytes to evolve into a scalable therapeutic source, a large quantity of highly pure cardiomyocytes is needed. Thus, lies the challenge of defining an efficient cardiomyocyte differentiation process. This chapter describes a method to evaluate multiple human-induced pluripotent stem cell lines for their cardiac differentiation potentials before evaluating their integrated proliferation and differentiation abilities in microcarrier cultures in a spinner culture format.
Collapse
Affiliation(s)
- Valerie Ho
- Bioprocessing Technology Institute, A*STAR Research Entities, Singapore, Singapore
| | - Gerine Tong
- Bioprocessing Technology Institute, A*STAR Research Entities, Singapore, Singapore
| | - Alan Lam
- Bioprocessing Technology Institute, A*STAR Research Entities, Singapore, Singapore
| | - Shaul Reuveny
- Bioprocessing Technology Institute, A*STAR Research Entities, Singapore, Singapore
| | - Steve Oh
- Bioprocessing Technology Institute, A*STAR Research Entities, Singapore, Singapore.
| |
Collapse
|
18
|
Salem T, Frankman Z, Churko J. Tissue engineering techniques for iPSC derived three-dimensional cardiac constructs. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:891-911. [PMID: 34476988 PMCID: PMC9419978 DOI: 10.1089/ten.teb.2021.0088] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Recent developments in applied developmental physiology have provided well-defined methodologies for producing human stem cell derived cardiomyocytes. Cardiomyocytes produced in this way have become commonplace as cardiac physiology research models. This accessibility has also allowed for the development of tissue engineered human heart constructs for drug screening, surgical intervention, and investigating cardiac pathogenesis. However, cardiac tissue engineering is an interdisciplinary field that involves complex engineering and physiological concepts, which limits its accessibility. This review provides a readable, broad reaching, and thorough discussion of major factors to consider for the development of cardiovascular tissues from stem cell derived cardiomyocytes. This review will examine important considerations in undertaking a cardiovascular tissue engineering project, and will present, interpret, and summarize some of the recent advancements in this field. This includes reviewing different forms of tissue engineered constructs, a discussion on cardiomyocyte sources, and an in-depth discussion of the fabrication and maturation procedures for tissue engineered heart constructs.
Collapse
Affiliation(s)
- Tori Salem
- University of Arizona Medical Center - University Campus, 22165, Cellular and Molecular Medicine, Tucson, Arizona, United States;
| | - Zachary Frankman
- University of Arizona Medical Center - University Campus, 22165, Biomedical Engineering, Tucson, Arizona, United States;
| | - Jared Churko
- University of Arizona Medical Center - University Campus, 22165, 1501 N Campbell RD, SHC 6143, Tucson, Arizona, United States, 85724-5128;
| |
Collapse
|
19
|
McKnight CL, Low YC, Elliott DA, Thorburn DR, Frazier AE. Modelling Mitochondrial Disease in Human Pluripotent Stem Cells: What Have We Learned? Int J Mol Sci 2021; 22:7730. [PMID: 34299348 PMCID: PMC8306397 DOI: 10.3390/ijms22147730] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/16/2021] [Accepted: 07/16/2021] [Indexed: 02/06/2023] Open
Abstract
Mitochondrial diseases disrupt cellular energy production and are among the most complex group of inherited genetic disorders. Affecting approximately 1 in 5000 live births, they are both clinically and genetically heterogeneous, and can be highly tissue specific, but most often affect cell types with high energy demands in the brain, heart, and kidneys. There are currently no clinically validated treatment options available, despite several agents showing therapeutic promise. However, modelling these disorders is challenging as many non-human models of mitochondrial disease do not completely recapitulate human phenotypes for known disease genes. Additionally, access to disease-relevant cell or tissue types from patients is often limited. To overcome these difficulties, many groups have turned to human pluripotent stem cells (hPSCs) to model mitochondrial disease for both nuclear-DNA (nDNA) and mitochondrial-DNA (mtDNA) contexts. Leveraging the capacity of hPSCs to differentiate into clinically relevant cell types, these models permit both detailed investigation of cellular pathomechanisms and validation of promising treatment options. Here we catalogue hPSC models of mitochondrial disease that have been generated to date, summarise approaches and key outcomes of phenotypic profiling using these models, and discuss key criteria to guide future investigations using hPSC models of mitochondrial disease.
Collapse
Affiliation(s)
- Cameron L. McKnight
- Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, VIC 3052, Australia; (C.L.M.); (Y.C.L.); (D.A.E.); (D.R.T.)
- Department of Paediatrics, University of Melbourne, Parkville, VIC 3052, Australia
| | - Yau Chung Low
- Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, VIC 3052, Australia; (C.L.M.); (Y.C.L.); (D.A.E.); (D.R.T.)
- Department of Paediatrics, University of Melbourne, Parkville, VIC 3052, Australia
| | - David A. Elliott
- Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, VIC 3052, Australia; (C.L.M.); (Y.C.L.); (D.A.E.); (D.R.T.)
- Department of Paediatrics, University of Melbourne, Parkville, VIC 3052, Australia
| | - David R. Thorburn
- Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, VIC 3052, Australia; (C.L.M.); (Y.C.L.); (D.A.E.); (D.R.T.)
- Department of Paediatrics, University of Melbourne, Parkville, VIC 3052, Australia
- Victorian Clinical Genetics Services, Royal Children’s Hospital, Parkville, VIC 3052, Australia
| | - Ann E. Frazier
- Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, VIC 3052, Australia; (C.L.M.); (Y.C.L.); (D.A.E.); (D.R.T.)
- Department of Paediatrics, University of Melbourne, Parkville, VIC 3052, Australia
| |
Collapse
|
20
|
Andrysiak K, Stępniewski J, Dulak J. Human-induced pluripotent stem cell-derived cardiomyocytes, 3D cardiac structures, and heart-on-a-chip as tools for drug research. Pflugers Arch 2021; 473:1061-1085. [PMID: 33629131 PMCID: PMC8245367 DOI: 10.1007/s00424-021-02536-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 02/01/2021] [Accepted: 02/03/2021] [Indexed: 12/13/2022]
Abstract
Development of new drugs is of high interest for the field of cardiac and cardiovascular diseases, which are a dominant cause of death worldwide. Before being allowed to be used and distributed, every new potentially therapeutic compound must be strictly validated during preclinical and clinical trials. The preclinical studies usually involve the in vitro and in vivo evaluation. Due to the increasing reporting of discrepancy in drug effects in animal and humans and the requirement to reduce the number of animals used in research, improvement of in vitro models based on human cells is indispensable. Primary cardiac cells are difficult to access and maintain in cell culture for extensive experiments; therefore, the human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) became an excellent alternative. This technology enables a production of high number of patient- and disease-specific cardiomyocytes and other cardiac cell types for a large-scale research. The drug effects can be extensively evaluated in the context of electrophysiological responses with a use of well-established tools, such as multielectrode array (MEA), patch clamp, or calcium ion oscillation measurements. Cardiotoxicity, which is a common reason for withdrawing drugs from marketing or rejection at final stages of clinical trials, can be easily verified with a use of hiPSC-CM model providing a prediction of human-specific responses and higher safety of clinical trials involving patient cohort. Abovementioned studies can be performed using two-dimensional cell culture providing a high-throughput and relatively lower costs. On the other hand, more complex structures, such as engineered heart tissue, organoids, or spheroids, frequently applied as co-culture systems, represent more physiological conditions and higher maturation rate of hiPSC-derived cells. Furthermore, heart-on-a-chip technology has recently become an increasingly popular tool, as it implements controllable culture conditions, application of various stimulations and continuous parameters read-out. This paper is an overview of possible use of cardiomyocytes and other cardiac cell types derived from hiPSC as in vitro models of heart in drug research area prepared on the basis of latest scientific reports and providing thorough discussion regarding their advantages and limitations.
Collapse
Affiliation(s)
- Kalina Andrysiak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Jacek Stępniewski
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Józef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland.
| |
Collapse
|
21
|
Gao Y, Pu J. Differentiation and Application of Human Pluripotent Stem Cells Derived Cardiovascular Cells for Treatment of Heart Diseases: Promises and Challenges. Front Cell Dev Biol 2021; 9:658088. [PMID: 34055788 PMCID: PMC8149736 DOI: 10.3389/fcell.2021.658088] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 03/25/2021] [Indexed: 12/15/2022] Open
Abstract
Human pluripotent stem cells (hPSCs) are derived from human embryos (human embryonic stem cells) or reprogrammed from human somatic cells (human induced pluripotent stem cells). They can differentiate into cardiovascular cells, which have great potential as exogenous cell resources for restoring cardiac structure and function in patients with heart disease or heart failure. A variety of protocols have been developed to generate and expand cardiovascular cells derived from hPSCs in vitro. Precisely and spatiotemporally activating or inhibiting various pathways in hPSCs is required to obtain cardiovascular lineages with high differentiation efficiency. In this concise review, we summarize the protocols of differentiating hPSCs into cardiovascular cells, highlight their therapeutic application for treatment of cardiac diseases in large animal models, and discuss the challenges and limitations in the use of cardiac cells generated from hPSCs for a better clinical application of hPSC-based cardiac cell therapy.
Collapse
Affiliation(s)
- Yu Gao
- Department of Cardiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jun Pu
- Department of Cardiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
22
|
Cell surface markers for immunophenotyping human pluripotent stem cell-derived cardiomyocytes. Pflugers Arch 2021; 473:1023-1039. [PMID: 33928456 DOI: 10.1007/s00424-021-02549-8] [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: 10/01/2020] [Revised: 02/09/2021] [Accepted: 02/25/2021] [Indexed: 02/08/2023]
Abstract
Human pluripotent stem cells (hPSC) self-renew and represent a potentially unlimited source for the production of cardiomyocytes (CMs) suitable for studies of human cardiac development, drug discovery, cardiotoxicity testing, and disease modelling and for cell-based therapies. However, most cardiac differentiation protocols yield mixed cultures of atrial-, ventricular-, and pacemaker-like cells at various stages of development, as well as non-CMs. The proportions and maturation states of these cell types result from disparities among differentiation protocols and time of cultivation, as well as hPSC reprogramming inconsistencies and genetic background variations. The reproducible use of hPSC-CMs for research and therapy is therefore limited by issues of cell population heterogeneity and functional states of maturation. A validated method that overcomes issues of cell heterogeneity is immunophenotyping coupled with live cell sorting, an approach that relies on accessible surface markers restricted to the desired cell type(s). Here we review current progress in unravelling heterogeneity in hPSC-cardiac cultures and in the identification of surface markers suitable for defining cardiac identity, subtype specificity, and maturation states.
Collapse
|
23
|
Emelyanova L, Bai X, Yan Y, Bosnjak ZJ, Kress D, Warner C, Kroboth S, Rudic T, Kaushik S, Stoeckl E, Ross GR, Rizvi F, Tajik AJ, Jahangir A. Biphasic effect of metformin on human cardiac energetics. Transl Res 2021; 229:5-23. [PMID: 33045408 PMCID: PMC10655614 DOI: 10.1016/j.trsl.2020.10.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 09/30/2020] [Accepted: 10/05/2020] [Indexed: 02/01/2023]
Abstract
Metformin is the first-line medication for treatment of type 2 diabetes and has been shown to reduce heart damage and death. However, mechanisms by which metformin protects human heart remain debated. The aim of the study was to evaluate the cardioprotective effect of metformin on cardiomyocytes derived from human-induced pluripotent stem cells (hiPSC-CMs) and mitochondria isolated from human cardiac tissue. At concentrations ≤2.5 mM, metformin significantly increased oxygen consumption rate (OCR) in the hiPSC-CMs by activating adenosine monophosphate activated protein kinase (AMPK)-dependent signaling and enhancing mitochondrial biogenesis. This effect was abrogated by compound C, an inhibitor of AMPK. At concentrations >5 mM, metformin inhibited the cellular OCR and triggered metabolic reprogramming by enhancing glycolysis and glutaminolysis in the cardiomyocytes. In isolated cardiac mitochondria, metformin did not increase the OCR at any concentrations but inhibited the OCR starting at 1 mM through direct inhibition of electron-transport chain complex I. This was associated with reduction of superoxide production and attenuation of Ca2+-induced mitochondrial permeability transition pore (mPTP) opening in the mitochondria. Thus, in human heart, metformin might improve cardioprotection due to its biphasic effect on mitochondria: at low concentrations, it activates mitochondrial biogenesis via AMPK signaling and increases the OCR; at high concentrations, it inhibits the respiration by directly affecting the activity of complex I, reduces oxidative stress and delays mPTP formation. Moreover, metformin at high concentrations causes metabolic reprogramming by enhancing glycolysis and glutaminolysis. These effects can be a beneficial adjunct to patients with impaired endogenous cardioprotective responses.
Collapse
Affiliation(s)
- Larisa Emelyanova
- Center for Integrative Research on Cardiovascular Aging, Advocate Aurora Research Institute, Milwaukee, Wisconsin.
| | - Xiaowen Bai
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Yasheng Yan
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Zeljko J Bosnjak
- Departments of Medicine and Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - David Kress
- Aurora Cardiovascular and Thoracic Services, St. Luke's Medical Center, Advocate Aurora Health Care, Milwaukee, Wisconsin
| | - Catherine Warner
- Center for Integrative Research on Cardiovascular Aging, Advocate Aurora Research Institute, Milwaukee, Wisconsin
| | - Stacie Kroboth
- Aurora Cardiovascular and Thoracic Services, St. Luke's Medical Center, Advocate Aurora Health Care, Milwaukee, Wisconsin
| | - Teodore Rudic
- Center for Integrative Research on Cardiovascular Aging, Advocate Aurora Research Institute, Milwaukee, Wisconsin
| | - Sirisha Kaushik
- Center for Integrative Research on Cardiovascular Aging, Advocate Aurora Research Institute, Milwaukee, Wisconsin
| | - Elizabeth Stoeckl
- Center for Integrative Research on Cardiovascular Aging, Advocate Aurora Research Institute, Milwaukee, Wisconsin
| | - Gracious R Ross
- Center for Integrative Research on Cardiovascular Aging, Advocate Aurora Research Institute, Milwaukee, Wisconsin
| | - Farhan Rizvi
- Center for Integrative Research on Cardiovascular Aging, Advocate Aurora Research Institute, Milwaukee, Wisconsin
| | - A Jamil Tajik
- Aurora Cardiovascular and Thoracic Services, St. Luke's Medical Center, Advocate Aurora Health Care, Milwaukee, Wisconsin
| | - Arshad Jahangir
- Aurora Cardiovascular and Thoracic Services, St. Luke's Medical Center, Advocate Aurora Health Care, Milwaukee, Wisconsin
| |
Collapse
|
24
|
Hang C, Song Y, Li Y, Zhang S, Chang Y, Bai R, Saleem A, Jiang M, Lu W, Lan F, Cui M. Knockout of MYOM1 in human cardiomyocytes leads to myocardial atrophy via impairing calcium homeostasis. J Cell Mol Med 2021; 25:1661-1676. [PMID: 33452765 PMCID: PMC7875908 DOI: 10.1111/jcmm.16268] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 12/14/2020] [Accepted: 12/22/2020] [Indexed: 12/27/2022] Open
Abstract
Myomesin-1 (encoded by MYOM1 gene) is expressed in almost all cross-striated muscles, whose family (together with myomesin-2 and myomesin-3) helps to cross-link adjacent myosin to form the M-line in myofibrils. However, little is known about its biological function, causal relationship and mechanisms underlying the MYOM1-related myopathies (especially in the heart). Regrettably, there is no MYMO1 knockout model for its study so far. A better and further understanding of MYOM1 biology is urgently needed. Here, we used CRISPR/Cas9 gene-editing technology to establish an MYOM1 knockout human embryonic stem cell line (MYOM1-/- hESC), which was then differentiated into myomesin-1 deficient cardiomyocytes (MYOM1-/- hESC-CMs) in vitro. We found that myomesin-1 plays an important role in sarcomere assembly, contractility regulation and cardiomyocytes development. Moreover, myomesin-1-deficient hESC-CMs can recapitulate myocardial atrophy phenotype in vitro. Based on this model, not only the biological function of MYOM1, but also the aetiology, pathogenesis, and potential treatments of myocardial atrophy caused by myomesin-1 deficiency can be studied.
Collapse
Affiliation(s)
- Chengwen Hang
- Department of CardiologyPeking University Third HospitalBeijingChina
| | - Yuanxiu Song
- Department of CardiologyPeking University Third HospitalBeijingChina
| | - Ya’nan Li
- Beijing Lab for Cardiovascular Precision MedicineAnzhen HospitalCapital Medical UniversityBeijingChina
| | - Siyao Zhang
- Beijing Lab for Cardiovascular Precision MedicineAnzhen HospitalCapital Medical UniversityBeijingChina
| | - Yun Chang
- Beijing Lab for Cardiovascular Precision MedicineAnzhen HospitalCapital Medical UniversityBeijingChina
| | - Rui Bai
- Beijing Lab for Cardiovascular Precision MedicineAnzhen HospitalCapital Medical UniversityBeijingChina
| | - Amina Saleem
- Beijing Lab for Cardiovascular Precision MedicineAnzhen HospitalCapital Medical UniversityBeijingChina
| | - Mengqi Jiang
- Department of CardiologyPeking University Third HospitalBeijingChina
| | - Wenjing Lu
- Beijing Lab for Cardiovascular Precision MedicineAnzhen HospitalCapital Medical UniversityBeijingChina
| | - Feng Lan
- Beijing Lab for Cardiovascular Precision MedicineAnzhen HospitalCapital Medical UniversityBeijingChina
| | - Ming Cui
- Department of CardiologyPeking University Third HospitalBeijingChina
| |
Collapse
|
25
|
Venkatachalapathy H, Azarin SM, Sarkar CA. Trajectory-based energy landscapes of gene regulatory networks. Biophys J 2021; 120:687-698. [PMID: 33453275 DOI: 10.1016/j.bpj.2020.11.2279] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 10/31/2020] [Accepted: 11/11/2020] [Indexed: 12/31/2022] Open
Abstract
Multistability and natural biological variability can result in significant heterogeneity within a cell population, leading to challenges in understanding and modulating cell behavior. Energy landscapes can offer qualitatively intuitive visualizations of cell phenotype and facilitate a more quantitative understanding of cellular dynamics, but current methods for landscape generation are mathematically involved and often require specific system properties (e.g., ergodicity or independent gene/protein probability distributions) that do not always hold. Here, we present a simple kinetic Monte Carlo-based method for landscape generation from a system of ordinary differential equations using only simulation trajectories initialized throughout the phase space of interest. The resulting landscape produces three quantitative features relevant to understanding cell behavior: stability (reflected by the depth or potential of landscape valleys), velocity (representing average directional movement on the landscape), and variance in velocity (indicative of landscape positions with heterogeneous movements). We applied this method to a genetic toggle switch, a core decision-making network in binary cellular responses, to elucidate effects of biologically relevant intrinsic and extrinsic cues. Intrinsic noise, such as stochasticity in transcription-translation and differences in cell cycle position, manifests through changes in valley width and position, reflecting increased population heterogeneity and more probabilistic cell fate transitions. The landscapes also capture the effect of an external inducer, revealing a quantitative correlation between the rate of cell fate transition and the energy barrier above a threshold inducer concentration determined by the permissivity of the valley. Further, in tracking dynamically changing landscapes under time-varying external cues, we unexpectedly found that an oscillatory inducer input can modulate cell fate heterogeneity and lead to periodic cell fate transitions entrained to the input frequency, depending on the intrinsic degradation rate of the switch. The landscape generation approach outlined herein is generalizable to other network topologies and may provide new quantitative insights into their dynamics.
Collapse
Affiliation(s)
- Harish Venkatachalapathy
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota
| | - Samira M Azarin
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota
| | - Casim A Sarkar
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
26
|
Budharaju H, Subramanian A, Sethuraman S. Recent advancements in cardiovascular bioprinting and bioprinted cardiac constructs. Biomater Sci 2021; 9:1974-1994. [DOI: 10.1039/d0bm01428a] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Three-dimensionally bioprinted cardiac constructs with biomimetic bioink helps to create native-equivalent cardiac tissues to treat patients with myocardial infarction.
Collapse
Affiliation(s)
- Harshavardhan Budharaju
- Tissue Engineering & Additive Manufacturing (TEAM) Lab
- Centre for Nanotechnology & Advanced Biomaterials
- ACBDE Innovation Centre
- School of Chemical & Biotechnology
- SASTRA Deemed to be University
| | - Anuradha Subramanian
- Tissue Engineering & Additive Manufacturing (TEAM) Lab
- Centre for Nanotechnology & Advanced Biomaterials
- ACBDE Innovation Centre
- School of Chemical & Biotechnology
- SASTRA Deemed to be University
| | - Swaminathan Sethuraman
- Tissue Engineering & Additive Manufacturing (TEAM) Lab
- Centre for Nanotechnology & Advanced Biomaterials
- ACBDE Innovation Centre
- School of Chemical & Biotechnology
- SASTRA Deemed to be University
| |
Collapse
|
27
|
Song Y, Wang H, Yue F, Lv Q, Cai B, Dong N, Wang Z, Wang L. Silk-Based Biomaterials for Cardiac Tissue Engineering. Adv Healthc Mater 2020; 9:e2000735. [PMID: 32939999 DOI: 10.1002/adhm.202000735] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 08/29/2020] [Indexed: 12/18/2022]
Abstract
Cardiovascular diseases are one of the leading causes of death globally. Among various cardiovascular diseases, myocardial infarction is an important one. Compared with conventional treatments, cardiac tissue engineering provides an alternative to repair and regenerate the injured tissue. Among various types of materials used for tissue engineering applications, silk biomaterials have been increasingly utilized due to their biocompatibility, biological functions, and many favorable physical/chemical properties. Silk biomaterials are often used alone or in combination with other materials in the forms of patches or hydrogels, and serve as promising delivery systems for bioactive compounds in tissue engineering repair scenarios. This review focuses primarily on the promising characteristics of silk biomaterials and their recent advances in cardiac tissue engineering.
Collapse
Affiliation(s)
- Yu Song
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Huifang Wang
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Feifei Yue
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qiying Lv
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Bo Cai
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Nianguo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zheng Wang
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Lin Wang
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| |
Collapse
|
28
|
Menasché P. Cell Therapy With Human ESC-Derived Cardiac Cells: Clinical Perspectives. Front Bioeng Biotechnol 2020; 8:601560. [PMID: 33195177 PMCID: PMC7649799 DOI: 10.3389/fbioe.2020.601560] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 10/02/2020] [Indexed: 12/12/2022] Open
Abstract
In the ongoing quest for the “ideal” cell type for heart repair, pluripotent stem cells (PSC) derived from either embryonic or reprogrammed somatic cells have emerged as attractive candidates because of their unique ability to give rise to lineage-specific cells and to transplant them at the desired stage of differentiation. The technical obstacles which have initially hindered their clinical use have now been largely overcome and several trials are under way which encompass several different diseases, including heart failure. So far, there have been no safety warning but it is still too early to draw definite conclusions regarding efficacy. In parallel, mechanistic studies suggest that the primary objective of “remuscularizing” the heart with PSC-derived cardiac cells can be challenged by their alternate use as ex vivo sources of a biologically active extracellular vesicle-enriched secretome equally able to improve heart function through harnessing endogenous repair pathways. The exclusive use of this secretome would combine the advantages of a large-scale production more akin to that of a biological medication, the likely avoidance of cell-associated immune and tumorigenicity risks and the possibility of intravenous infusions compatible with repeated dosing.
Collapse
Affiliation(s)
- Philippe Menasché
- Department of Cardiovascular Surgery, Hôpital Européen Georges Pompidou, Paris, France.,PARCC, INSERM, University of Paris, Paris, France
| |
Collapse
|
29
|
Veldhuizen J, Cutts J, Brafman DA, Migrino RQ, Nikkhah M. Engineering anisotropic human stem cell-derived three-dimensional cardiac tissue on-a-chip. Biomaterials 2020; 256:120195. [DOI: 10.1016/j.biomaterials.2020.120195] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 05/15/2020] [Accepted: 06/09/2020] [Indexed: 01/02/2023]
|
30
|
Ravindran D, Kok C, Farraha M, Selvakumar D, Clayton ZE, Kumar S, Chong J, Kizana E. Gene and Cell Therapy for Cardiac Arrhythmias. Clin Ther 2020; 42:1911-1922. [PMID: 32988632 DOI: 10.1016/j.clinthera.2020.09.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/19/2020] [Accepted: 09/01/2020] [Indexed: 12/21/2022]
Abstract
PURPOSE In the last decade, interest in gene therapy as a therapeutic technology has increased, largely driven by an exciting yet modest number of successful applications for monogenic diseases. Setbacks in the use of gene therapy for cardiac disease have motivated efforts to develop vectors with enhanced tropism for the heart and more efficient delivery methods. Although monogenic diseases are the logical target, cardiac arrhythmias represent a group of conditions amenable to gene therapy because of focal targets (biological pacemakers, nodal conduction, or stem cell-related arrhythmias) or bystander effects on cells not directly transduced because of electrical coupling. METHODS This review provides a contemporary narrative of the field of gene therapy for experimental cardiac arrhythmias, including those associated with stem cell transplant. Recent articles published in the English language and available through the PubMed database and other prominent literature are discussed. FINDINGS The promise of gene therapy has been realized for a handful of monogenic diseases and is actively being pursued for cardiac applications in preclinical models. With improved vectors, it is likely that cardiac disease will also benefit from this technology. Cardiac arrhythmias, whether inherited or acquired, are a group of conditions with a potentially lower threshold for phenotypic correction and as such hold unique potential as targets for cardiac gene therapy. IMPLICATIONS There has been a proliferation of research on the potential of gene therapy for cardiac arrhythmias. This body of investigation forms a strong basis on which further developments, particularly with viral vectors, are likely to help this technology progress along its translational trajectory.
Collapse
Affiliation(s)
- Dhanya Ravindran
- Centre for Heart Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia; Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Cindy Kok
- Centre for Heart Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia; Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Melad Farraha
- Centre for Heart Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
| | - Dinesh Selvakumar
- Centre for Heart Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia; Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia; Department of Cardiology, Westmead Hospital, Westmead, New South Wales, Australia
| | - Zoe E Clayton
- Centre for Heart Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia; Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Saurabh Kumar
- Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia; Department of Cardiology, Westmead Hospital, Westmead, New South Wales, Australia
| | - James Chong
- Centre for Heart Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia; Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia; Department of Cardiology, Westmead Hospital, Westmead, New South Wales, Australia
| | - Eddy Kizana
- Centre for Heart Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia; Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia; Department of Cardiology, Westmead Hospital, Westmead, New South Wales, Australia.
| |
Collapse
|
31
|
"Betwixt Mine Eye and Heart a League Is Took": The Progress of Induced Pluripotent Stem-Cell-Based Models of Dystrophin-Associated Cardiomyopathy. Int J Mol Sci 2020; 21:ijms21196997. [PMID: 32977524 PMCID: PMC7582534 DOI: 10.3390/ijms21196997] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/17/2020] [Accepted: 09/21/2020] [Indexed: 12/19/2022] Open
Abstract
The ultimate goal of precision disease modeling is to artificially recreate the disease of affected people in a highly controllable and adaptable external environment. This field has rapidly advanced which is evident from the application of patient-specific pluripotent stem-cell-derived precision therapies in numerous clinical trials aimed at a diverse set of diseases such as macular degeneration, heart disease, spinal cord injury, graft-versus-host disease, and muscular dystrophy. Despite the existence of semi-adequate treatments for tempering skeletal muscle degeneration in dystrophic patients, nonischemic cardiomyopathy remains one of the primary causes of death. Therefore, cardiovascular cells derived from muscular dystrophy patients' induced pluripotent stem cells are well suited to mimic dystrophin-associated cardiomyopathy and hold great promise for the development of future fully effective therapies. The purpose of this article is to convey the realities of employing precision disease models of dystrophin-associated cardiomyopathy. This is achieved by discussing, as suggested in the title echoing William Shakespeare's words, the settlements (or "leagues") made by researchers to manage the constraints ("betwixt mine eye and heart") distancing them from achieving a perfect precision disease model.
Collapse
|
32
|
Selvakumar D, Clayton ZE, Chong JJH. Robust Cardiac Regeneration: Fulfilling the Promise of Cardiac Cell Therapy. Clin Ther 2020; 42:1857-1879. [PMID: 32943195 DOI: 10.1016/j.clinthera.2020.08.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/12/2020] [Accepted: 08/14/2020] [Indexed: 12/16/2022]
Abstract
PURPOSE We review the history of cardiac cell therapy, highlighting lessons learned from initial adult stem cell (ASC) clinical trials. We present pluripotent stem cell-derived cardiomyocytes (PSC-CMs) as a leading candidate for robust regeneration of infarcted myocardium but identify several issues that must be addressed before successful clinical translation. METHODS We conducted an unstructured literature review of PubMed-listed articles, selecting the most comprehensive and relevant research articles, review articles, clinical trials, and basic or translation articles in the field of cardiac cell therapy. Articles were identified using the search terms adult stem cells, pluripotent stem cells, cardiac stem cell, and cardiac regeneration or from references of relevant articles, Articles were prioritized and selected based on their impact, originality, or potential clinical applicability. FINDINGS Since its inception, the ASC therapy field has been troubled by conflicting preclinical data, academic controversies, and inconsistent trial designs. These issues have damaged perceptions of cardiac cell therapy among investors, the academic community, health care professionals, and, importantly, patients. In hindsight, the key issue underpinning these problems was the inability of these cell types to differentiate directly into genuine cardiomyocytes, rendering them unable to replace damaged myocardium. Despite this, beneficial effects through indirect paracrine or immunomodulatory effects remain possible and continue to be investigated. However, in preclinical models, PSC-CMs have robustly remuscularized infarcted myocardium with functional, force-generating cardiomyocytes. Hence, PSC-CMs have now emerged as a leading candidate for cardiac regeneration, and unpublished reports of first-in-human delivery of these cells have recently surfaced. However, the cardiac cell therapy field's history should serve as a cautionary tale, and we identify several translational hurdles that still remain. Preclinical solutions to issues such as arrhythmogenicity, immunogenicity, and poor engraftment rates are needed, and next-generation clinical trials must draw on robust knowledge of mechanistic principles of the therapy. IMPLICATIONS The clinical transplantation of functional stem cell-derived heart tissue with seamless integration into native myocardium is a lofty goal. However, considerable advances have been made during the past 2 decades. Currently, PSC-CMs appear to be the best prospect to reach this goal, but several hurdles remain. The history of adult stem cell trials has taught us that shortcuts cannot be taken without dire consequences, and it is essential that progress not be hurried and that a worldwide, cross-disciplinary approach be used to ensure safe and effective clinical translation.
Collapse
Affiliation(s)
- Dinesh Selvakumar
- Centre for Heart Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia; Department of Cardiology, Westmead Hospital, Westmead, New South Wales, Australia
| | - Zoe E Clayton
- Centre for Heart Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
| | - James J H Chong
- Centre for Heart Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia; Department of Cardiology, Westmead Hospital, Westmead, New South Wales, Australia.
| |
Collapse
|
33
|
Patino-Guerrero A, Veldhuizen J, Zhu W, Migrino RQ, Nikkhah M. Three-dimensional scaffold-free microtissues engineered for cardiac repair. J Mater Chem B 2020; 8:7571-7590. [PMID: 32724973 PMCID: PMC8314954 DOI: 10.1039/d0tb01528h] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cardiovascular diseases, including myocardial infarction (MI), persist as the leading cause of mortality and morbidity worldwide. The limited regenerative capacity of the myocardium presents significant challenges specifically for the treatment of MI and, subsequently, heart failure (HF). Traditional therapeutic approaches mainly rely on limiting the induced damage or the stress on the remaining viable myocardium through pharmacological regulation of remodeling mechanisms, rather than replacement or regeneration of the injured tissue. The emerging alternative regenerative medicine-based approaches have focused on restoring the damaged myocardial tissue with newly engineered functional and bioinspired tissue units. Cardiac regenerative medicine approaches can be broadly categorized into three groups: cell-based therapies, scaffold-based cardiac tissue engineering, and scaffold-free cardiac tissue engineering. Despite significant advancements, however, the clinical translation of these approaches has been critically hindered by two key obstacles for successful structural and functional replacement of the damaged myocardium, namely: poor engraftment of engineered tissue into the damaged cardiac muscle and weak electromechanical coupling of transplanted cells with the native tissue. To that end, the integration of micro- and nanoscale technologies along with recent advancements in stem cell technologies have opened new avenues for engineering of structurally mature and highly functional scaffold-based (SB-CMTs) and scaffold-free cardiac microtissues (SF-CMTs) with enhanced cellular organization and electromechanical coupling for the treatment of MI and HF. In this review article, we will present the state-of-the-art approaches and recent advancements in the engineering of SF-CMTs for myocardial repair.
Collapse
|
34
|
Desgres M, Menasché P. Clinical Translation of Pluripotent Stem Cell Therapies: Challenges and Considerations. Cell Stem Cell 2020; 25:594-606. [PMID: 31703770 DOI: 10.1016/j.stem.2019.10.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Although the clinical outcomes of cell therapy trials have not met initial expectations, emerging evidence suggests that injury-mediated tissue damage might benefit from the delivery of cells or their secreted products. Pluripotent stem cells (PSCs) are promising cell sources primarily because of their capacity to generate stage- and lineage-specific differentiated derivatives. However, they carry inherent challenges for safe and efficacious clinical translation. This Review describes completed or ongoing trials of PSCs, discusses their potential mechanisms of action, and considers how to address the challenges required for them to become a major therapy, using heart repair as a case study.
Collapse
Affiliation(s)
- Manon Desgres
- Université de Paris, PARCC, INSERM, 75015 Paris, France
| | - Philippe Menasché
- Université de Paris, PARCC, INSERM, 75015 Paris, France; Department of Cardiovascular Surgery, Hôpital Européen Georges Pompidou 20, rue Leblanc, 75015 Paris, France.
| |
Collapse
|
35
|
Optimizing the Use of iPSC-CMs for Cardiac Regeneration in Animal Models. Animals (Basel) 2020; 10:ani10091561. [PMID: 32887495 PMCID: PMC7552322 DOI: 10.3390/ani10091561] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/29/2020] [Accepted: 08/31/2020] [Indexed: 12/29/2022] Open
Abstract
Simple Summary In 2006, the first induced pluripotent stem cells were generated by reprogramming skin cells. Induced pluripotent stem cells undergo fast cell division, can differentiate into many different cell types, can be patient-specific, and do not raise ethical issues. Thus, they offer great promise as in vitro disease models, drug toxicity testing platforms, and for autologous tissue regeneration. Heart failure is one of the major causes of death worldwide. It occurs when the heart cannot meet the body’s metabolic demands. Induced pluripotent stem cells can be differentiated into cardiac myocytes, can form patches resembling native cardiac tissue, and can engraft to the damaged heart. However, despite correct host/graft coupling, most animal studies demonstrate an arrhythmogenicity of the engrafted tissue and variable survival. This is partially because of the heterogeneity and immaturity of the cells. New evidence suggests that by modulating induced pluripotent stem cells-cardiac myocytes (iPSC-CM) metabolism by switching substrates and changing metabolic pathways, you can decrease iPSC-CM heterogeneity and arrhythmogenicity. Novel culture methods and tissue engineering along with animal models of heart failure are needed to fully unlock the potential of cardiac myocytes derived from induced pluripotent stem cells for cardiac regeneration. Abstract Heart failure (HF) is a common disease in which the heart cannot meet the metabolic demands of the body. It mostly occurs in individuals 65 years or older. Cardiac transplantation is the best option for patients with advanced HF. High numbers of patient-specific cardiac myocytes (CMs) can be generated from induced pluripotent stem cells (iPSCs) and can possibly be used to treat HF. While some studies found iPSC-CMS can couple efficiently to the damaged heart and restore cardiac contractility, almost all found iPSC-CM transplantation is arrhythmogenic, thus hampering the use of iPSC-CMs for cardiac regeneration. Studies show that iPSC-CM cultures are highly heterogeneous containing atrial-, ventricular- and nodal-like CMs. Furthermore, they have an immature phenotype, resembling more fetal than adult CMs. There is an urgent need to overcome these issues. To this end, a novel and interesting avenue to increase CM maturation consists of modulating their metabolism. Combined with careful engineering and animal models of HF, iPSC-CMs can be assessed for their potential for cardiac regeneration and a cure for HF.
Collapse
|
36
|
Choi SW, Cho YW, Kim JG, Kim YJ, Kim E, Chung HM, Kang SW. Effect of Cell Labeling on the Function of Human Pluripotent Stem Cell-Derived Cardiomyocytes. Int J Stem Cells 2020; 13:287-294. [PMID: 32323512 PMCID: PMC7378900 DOI: 10.15283/ijsc19138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 03/01/2020] [Accepted: 03/03/2020] [Indexed: 11/09/2022] Open
Abstract
Cell labeling technologies are required to monitor the fate of transplanted cells in vivo and to select target cells for the observation of certain changes in vitro. Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have been transplanted for the treatment of heart injuries or used in vitro for preclinical cardiac safety assessments. Cardiomyocyte (CM) labeling has been used in these processes to facilitate target cell monitoring. However, the functional effect of the labeling agent on hiPSC-CMs has not been studied. Therefore, we investigated the effects of labeling agents on CM cellular functions. 3'-Dioctadecyloxacarbocyanine perchlorate (DiO), quantum dots (QDs), and a DNA plasmid expressing EGFP using Lipo2K were used to label hiPSC-CMs. We conclude that the hiPSC-CM labeling with DiO and QDs does not induce arrhythmogenic effects but rather improves the mRNA expression of cardiac ion channels and Ca2+ influx by L-type Ca2+ channels. Thus, DiO and QD labeling agents may be useful tools to monitor transplanted CMs, and further in vivo influences of the labeling agents should be investigated in the future.
Collapse
Affiliation(s)
- Seong Woo Choi
- Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Young-Woo Cho
- Department of Pharmacy, Chungbuk National University College of Pharmacy, Cheongju, Korea.,Division of Drug Evaluation, NDDC, Oseong Medical Innovation Foundation, Cheongju, Korea
| | - Jae Gon Kim
- Research Group for Biomimetic Advanced Technology, Korea Institute of Toxicology, Daejeon, Korea
| | - Yong-Jin Kim
- R&D Unit, Amorepacific Corporation, Yongin, Korea
| | - Eunmi Kim
- R&D Unit, Amorepacific Corporation, Yongin, Korea
| | - Hyung-Min Chung
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Korea
| | - Sun-Woong Kang
- Department of Human and Environmental Toxicology, University of Science and Technology, Daejeon, Korea
| |
Collapse
|
37
|
Puluca N, Durmus NG, Lee S, Belbachir N, Galdos FX, Ogut MG, Gupta R, Hirano KI, Krane M, Lange R, Wu JC, Wu SM, Demirci U. Levitating Cells to Sort the Fit and the Fat. ACTA ACUST UNITED AC 2020; 4:e1900300. [PMID: 32352239 DOI: 10.1002/adbi.201900300] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 03/12/2020] [Accepted: 03/30/2020] [Indexed: 01/22/2023]
Abstract
Density is a core material property and varies between different cell types, mainly based on differences in their lipid content. Sorting based on density enables various biomedical applications such as multi-omics in precision medicine and regenerative repair in medicine. However, a significant challenge is sorting cells of the same type based on density differences. Here, a new method for real-time monitoring and sorting of single cells based on their inherent levitation profiles driven by their lipid content is reported. As a model system, human-induced pluripotent stem cell (hiPSC)-derived cardiomyocytes (CMs) from a patient with neutral lipid storage disease (NLSD) due to loss of function of adipose triglyceride lipase (ATGL) resulting in abnormal lipid storage in cardiac muscle are used. This levitation-based strategy detects subpopulations within ATGL-deficient hiPSC-CMs with heterogenous lipid content, equilibrating at different levitation heights due to small density differences. In addition, sorting of these differentially levitating subpopulations are monitored in real time. Using this approach, sorted healthy and diseased hiPSC-CMs maintain viability and function. Pixel-tracking technologies show differences in contraction between NLSD and healthy hiPSC-CMs. Overall, this is a unique approach to separate diseased cell populations based on their intracellular lipid content that cannot be achieved using traditional flow cytometry techniques.
Collapse
Affiliation(s)
- Nazan Puluca
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA.,Department of Cardiovascular Surgery, German Heart Center Munich, Technische Universität München, München, 80333, Germany.,Insure (Institute for Translational Cardiac Surgery), Department of Cardiovascular Surgery, German Heart Center Munich, Technische Universität München, München, 80333, Germany.,Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Naside Gözde Durmus
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, Palo Alto, CA, 94304, USA
| | - Soah Lee
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA.,Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Nadjet Belbachir
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA.,Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Francisco X Galdos
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA.,Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Mehmet Giray Ogut
- Canary Center for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, Stanford University, Palo Alto, CA, 94304, USA
| | - Rakhi Gupta
- Canary Center for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, Stanford University, Palo Alto, CA, 94304, USA
| | - Ken-Ichi Hirano
- Department of Cardiovascular Medicine, Osaka University School of Medicine, Osaka, 565-0871, Japan
| | - Markus Krane
- Department of Cardiovascular Surgery, German Heart Center Munich, Technische Universität München, München, 80333, Germany.,Insure (Institute for Translational Cardiac Surgery), Department of Cardiovascular Surgery, German Heart Center Munich, Technische Universität München, München, 80333, Germany.,German Heart Center Munich-DZHK Partner Site Munich Heart Alliance, Munich, Germany
| | - Rüdiger Lange
- Department of Cardiovascular Surgery, German Heart Center Munich, Technische Universität München, München, 80333, Germany.,Insure (Institute for Translational Cardiac Surgery), Department of Cardiovascular Surgery, German Heart Center Munich, Technische Universität München, München, 80333, Germany.,German Heart Center Munich-DZHK Partner Site Munich Heart Alliance, Munich, Germany
| | - Joseph C Wu
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA.,Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Sean M Wu
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA.,Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Utkan Demirci
- Canary Center for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, Stanford University, Palo Alto, CA, 94304, USA
| |
Collapse
|
38
|
Wang C, Koo S, Park M, Vangelatos Z, Hoang P, Conklin B, Grigoropoulos CP, Healy KE, Ma Z. Maladaptive Contractility of 3D Human Cardiac Microtissues to Mechanical Nonuniformity. Adv Healthc Mater 2020; 9:e1901373. [PMID: 32090507 PMCID: PMC7274862 DOI: 10.1002/adhm.201901373] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 02/01/2020] [Indexed: 01/29/2023]
Abstract
Cardiac tissues are able to adjust their contractile behavior to adapt to the local mechanical environment. Nonuniformity of the native tissue mechanical properties contributes to the development of heart dysfunctions, yet the current in vitro cardiac tissue models often fail to recapitulate the mechanical nonuniformity. To address this issue, a 3D cardiac microtissue model is developed with engineered mechanical nonuniformity, enabled by 3D-printed hybrid matrices composed of fibers with different diameters. When escalating the complexity of tissue mechanical environments, cardiac microtissues start to develop maladaptive hypercontractile phenotypes, demonstrated in both contractile motion analysis and force-power analysis. This novel hybrid system could potentially facilitate the establishment of "pathologically-inspired" cardiac microtissue models for deeper understanding of heart pathology due to nonuniformity of the tissue mechanical environment.
Collapse
Affiliation(s)
- Chenyan Wang
- Department of Biomedical & Chemical Engineering, Syracuse Biomaterials Institute, Syracuse University
| | - Sangmo Koo
- Department of Mechanical Engineering, University of California, Berkeley
| | - Minok Park
- Department of Mechanical Engineering, University of California, Berkeley
| | | | - Plansky Hoang
- Department of Biomedical & Chemical Engineering, Syracuse Biomaterials Institute, Syracuse University
| | - Bruce Conklin
- Gladstone Institute of Cardiovascular Diseases, University of California, San Francisco
| | | | - Kevin E. Healy
- Department of Bioengineering, University of California, Berkeley
- Department of Material Science & Engineering, University of California, Berkeley
| | - Zhen Ma
- Department of Biomedical & Chemical Engineering, Syracuse Biomaterials Institute, Syracuse University
| |
Collapse
|
39
|
van Gorp PRR, Trines SA, Pijnappels DA, de Vries AAF. Multicellular In vitro Models of Cardiac Arrhythmias: Focus on Atrial Fibrillation. Front Cardiovasc Med 2020; 7:43. [PMID: 32296716 PMCID: PMC7138102 DOI: 10.3389/fcvm.2020.00043] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 03/06/2020] [Indexed: 12/13/2022] Open
Abstract
Atrial fibrillation (AF) is the most common cardiac arrhythmia in clinical practice with a large socioeconomic impact due to its associated morbidity, mortality, reduction in quality of life and health care costs. Currently, antiarrhythmic drug therapy is the first line of treatment for most symptomatic AF patients, despite its limited efficacy, the risk of inducing potentially life-threating ventricular tachyarrhythmias as well as other side effects. Alternative, in-hospital treatment modalities consisting of electrical cardioversion and invasive catheter ablation improve patients' symptoms, but often have to be repeated and are still associated with serious complications and only suitable for specific subgroups of AF patients. The development and progression of AF generally results from the interplay of multiple disease pathways and is accompanied by structural and functional (e.g., electrical) tissue remodeling. Rational development of novel treatment modalities for AF, with its many different etiologies, requires a comprehensive insight into the complex pathophysiological mechanisms. Monolayers of atrial cells represent a simplified surrogate of atrial tissue well-suited to investigate atrial arrhythmia mechanisms, since they can easily be used in a standardized, systematic and controllable manner to study the role of specific pathways and processes in the genesis, perpetuation and termination of atrial arrhythmias. In this review, we provide an overview of the currently available two- and three-dimensional multicellular in vitro systems for investigating the initiation, maintenance and termination of atrial arrhythmias and AF. This encompasses cultures of primary (animal-derived) atrial cardiomyocytes (CMs), pluripotent stem cell-derived atrial-like CMs and (conditionally) immortalized atrial CMs. The strengths and weaknesses of each of these model systems for studying atrial arrhythmias will be discussed as well as their implications for future studies.
Collapse
Affiliation(s)
| | | | | | - Antoine A. F. de Vries
- Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
40
|
Laco F, Lam ATL, Woo TL, Tong G, Ho V, Soong PL, Grishina E, Lin KH, Reuveny S, Oh SKW. Selection of human induced pluripotent stem cells lines optimization of cardiomyocytes differentiation in an integrated suspension microcarrier bioreactor. Stem Cell Res Ther 2020; 11:118. [PMID: 32183888 PMCID: PMC7076930 DOI: 10.1186/s13287-020-01618-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 02/11/2020] [Accepted: 02/24/2020] [Indexed: 01/13/2023] Open
Abstract
Background The production of large quantities of cardiomyocyte is essential for the needs of cellular therapies. This study describes the selection of a human-induced pluripotent cell (hiPSC) line suitable for production of cardiomyocytes in a fully integrated bioprocess of stem cell expansion and differentiation in microcarrier stirred tank reactor. Methods Five hiPSC lines were evaluated first for their cardiac differentiation efficiency in monolayer cultures followed by their expansion and differentiation compatibility in microcarrier (MC) cultures under continuous stirring conditions. Results Three cell lines were highly cardiogenic but only one (FR202) of them was successfully expanded on continuous stirring MC cultures. FR202 was thus selected for cardiac differentiation in a 22-day integrated bioprocess under continuous stirring in a stirred tank bioreactor. In summary, we integrated a MC-based hiPSC expansion (phase 1), CHIR99021-induced cardiomyocyte differentiation step (phase 2), purification using the lactate-based treatment (phase 3) and cell recovery step (phase 4) into one process in one bioreactor, under restricted oxygen control (< 30% DO) and continuous stirring with periodic batch-type media exchanges. High density of undifferentiated hiPSC (2 ± 0.4 × 106 cells/mL) was achieved in the expansion phase. By controlling the stirring speed and DO levels in the bioreactor cultures, 7.36 ± 1.2 × 106 cells/mL cardiomyocytes with > 80% Troponin T were generated in the CHIR99021-induced differentiation phase. By adding lactate in glucose-free purification media, the purity of cardiomyocytes was enhanced (> 90% Troponin T), with minor cell loss as indicated by the increase in sub-G1 phase and the decrease of aggregate sizes. Lastly, we found that the recovery period is important for generating purer and functional cardiomyocytes (> 96% Troponin T). Three independent runs in a 300-ml working volume confirmed the robustness of this process. Conclusion A streamlined and controllable platform for large quantity manufacturing of pure functional atrial, ventricular and nodal cardiomyocytes on MCs in conventional-type stirred tank bioreactors was established, which can be further scaled up and translated to a good manufacturing practice-compliant production process, to fulfill the quantity requirements of the cellular therapeutic industry. Supplementary information The online version of this article (10.1186/s13287-020-01618-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Filip Laco
- Bioprocessing Technology Institute, 20 Biopolis Way, Centros #06-01, Singapore, 138668, Singapore
| | - Alan Tin-Lun Lam
- Bioprocessing Technology Institute, 20 Biopolis Way, Centros #06-01, Singapore, 138668, Singapore.
| | - Tsung-Liang Woo
- Bioprocessing Technology Institute, 20 Biopolis Way, Centros #06-01, Singapore, 138668, Singapore
| | - Gerine Tong
- Bioprocessing Technology Institute, 20 Biopolis Way, Centros #06-01, Singapore, 138668, Singapore
| | - Valerie Ho
- Bioprocessing Technology Institute, 20 Biopolis Way, Centros #06-01, Singapore, 138668, Singapore
| | - Poh-Loong Soong
- Ternion Biosciences, National Heart Centre of Singapore, Singapore, Singapore
| | - Elina Grishina
- Ternion Biosciences, National Heart Centre of Singapore, Singapore, Singapore
| | - Kun-Han Lin
- Ternion Biosciences, National Heart Centre of Singapore, Singapore, Singapore
| | - Shaul Reuveny
- Bioprocessing Technology Institute, 20 Biopolis Way, Centros #06-01, Singapore, 138668, Singapore
| | - Steve Kah-Weng Oh
- Bioprocessing Technology Institute, 20 Biopolis Way, Centros #06-01, Singapore, 138668, Singapore.
| |
Collapse
|
41
|
Nemade H, Acharya A, Chaudhari U, Nembo E, Nguemo F, Riet N, Abken H, Hescheler J, Papadopoulos S, Sachinidis A. Cyclooxygenases Inhibitors Efficiently Induce Cardiomyogenesis in Human Pluripotent Stem Cells. Cells 2020; 9:cells9030554. [PMID: 32120775 PMCID: PMC7140528 DOI: 10.3390/cells9030554] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 01/28/2020] [Accepted: 02/26/2020] [Indexed: 12/12/2022] Open
Abstract
Application of human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) is limited by the challenges in their efficient differentiation. Recently, the Wingless (Wnt) signaling pathway has emerged as the key regulator of cardiomyogenesis. In this study, we evaluated the effects of cyclooxygenase inhibitors on cardiac differentiation of hPSCs. Cardiac differentiation was performed by adherent monolayer based method using 4 hPSC lines (HES3, H9, IMR90, and ES4SKIN). The efficiency of cardiac differentiation was evaluated by flow cytometry and RT-qPCR. Generated hPSC-CMs were characterised using immunocytochemistry, electrophysiology, electron microscopy, and calcium transient measurements. Our data show that the COX inhibitors Sulindac and Diclofenac in combination with CHIR99021 (GSK-3 inhibitor) efficiently induce cardiac differentiation of hPSCs. In addition, inhibition of COX using siRNAs targeted towards COX-1 and/or COX-2 showed that inhibition of COX-2 alone or COX-1 and COX-2 in combination induce cardiomyogenesis in hPSCs within 12 days. Using IMR90-Wnt reporter line, we showed that inhibition of COX-2 led to downregulation of Wnt signalling activity in hPSCs. In conclusion, this study demonstrates that COX inhibition efficiently induced cardiogenesis via modulation of COX and Wnt pathway and the generated cardiomyocytes express cardiac-specific structural markers as well as exhibit typical calcium transients and action potentials. These cardiomyocytes also responded to cardiotoxicants and can be relevant as an in vitro cardiotoxicity screening model.
Collapse
Affiliation(s)
- Harshal Nemade
- Institute of Neurophysiology, Faculty of Medicine, University of Cologne, Robert-Koch-Str. 39, 50931 Cologne, Germany; (H.N.); (A.A.); (U.C.); (E.N.); (F.N.); (J.H.); (S.P.)
| | - Aviseka Acharya
- Institute of Neurophysiology, Faculty of Medicine, University of Cologne, Robert-Koch-Str. 39, 50931 Cologne, Germany; (H.N.); (A.A.); (U.C.); (E.N.); (F.N.); (J.H.); (S.P.)
| | - Umesh Chaudhari
- Institute of Neurophysiology, Faculty of Medicine, University of Cologne, Robert-Koch-Str. 39, 50931 Cologne, Germany; (H.N.); (A.A.); (U.C.); (E.N.); (F.N.); (J.H.); (S.P.)
| | - Erastus Nembo
- Institute of Neurophysiology, Faculty of Medicine, University of Cologne, Robert-Koch-Str. 39, 50931 Cologne, Germany; (H.N.); (A.A.); (U.C.); (E.N.); (F.N.); (J.H.); (S.P.)
| | - Filomain Nguemo
- Institute of Neurophysiology, Faculty of Medicine, University of Cologne, Robert-Koch-Str. 39, 50931 Cologne, Germany; (H.N.); (A.A.); (U.C.); (E.N.); (F.N.); (J.H.); (S.P.)
| | - Nicole Riet
- Department I Internal Medicine and Center for Molecular Medicine Cologne (CMMC), University of Cologne (UKK), Robert-Koch-Str. 21, 50931 Cologne, Germany;
| | - Hinrich Abken
- Regensburg Centre for Interventional Immunology (RCI), Deptartment Genetic Immunotherapy, University Hospital Regensburg, 93053 Regensburg, Germany;
| | - Jürgen Hescheler
- Institute of Neurophysiology, Faculty of Medicine, University of Cologne, Robert-Koch-Str. 39, 50931 Cologne, Germany; (H.N.); (A.A.); (U.C.); (E.N.); (F.N.); (J.H.); (S.P.)
| | - Symeon Papadopoulos
- Institute of Neurophysiology, Faculty of Medicine, University of Cologne, Robert-Koch-Str. 39, 50931 Cologne, Germany; (H.N.); (A.A.); (U.C.); (E.N.); (F.N.); (J.H.); (S.P.)
| | - Agapios Sachinidis
- Institute of Neurophysiology, Faculty of Medicine, University of Cologne, Robert-Koch-Str. 39, 50931 Cologne, Germany; (H.N.); (A.A.); (U.C.); (E.N.); (F.N.); (J.H.); (S.P.)
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Robert-Koch-Str. 21, 50931 Cologne, Germany
- Correspondence: ; Tel.: +49-0221-4787373
| |
Collapse
|
42
|
Garbern JC, Escalante GO, Lee RT. Pluripotent stem cell-derived cardiomyocytes for treatment of cardiomyopathic damage: Current concepts and future directions. Trends Cardiovasc Med 2020; 31:85-90. [PMID: 31983535 DOI: 10.1016/j.tcm.2020.01.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 12/31/2019] [Accepted: 01/01/2020] [Indexed: 02/06/2023]
Abstract
Today, cell replacement therapy using pluripotent stem cell-derived cardiomyocytes (PSC-CMs) remains a research endeavor, with several hurdles that must be overcome before delivery of PSC-CMs can become a therapeutic reality. In this review, we highlight major findings to date from pre-clinical studies involving delivery of PSC-CMs and consider remaining challenges that must be addressed for successful clinical translation. Our goal is to provide an overview of the current status of cardiomyocyte replacement therapy and what challenges must be addressed before successful clinical translation of such therapies will be possible.
Collapse
Affiliation(s)
- Jessica C Garbern
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, 7 Divinity Ave, Cambridge, MA 02138, United States; Department of Cardiology, Boston Children's Hospital, 300 Longwood Ave, Boston, MA 02115, United States
| | - Gabriela O Escalante
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, 7 Divinity Ave, Cambridge, MA 02138, United States
| | - Richard T Lee
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, 7 Divinity Ave, Cambridge, MA 02138, United States; Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 75 Francis St, Boston, MA 02115, United States.
| |
Collapse
|
43
|
Biendarra-Tiegs SM, Clemens DJ, Secreto FJ, Nelson TJ. Human Induced Pluripotent Stem Cell-Derived Non-Cardiomyocytes Modulate Cardiac Electrophysiological Maturation Through Connexin 43-Mediated Cell-Cell Interactions. Stem Cells Dev 2019; 29:75-89. [PMID: 31744402 PMCID: PMC6978788 DOI: 10.1089/scd.2019.0098] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The functional maturation status of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) has a notable impact upon their use in pharmacological studies, disease modeling, and therapeutic applications. Non-cardiomyocytes (non-CMs) produced in the differentiation process have previously been identified as having an extrinsic influence upon hiPSC-CM development, yet the underlying mechanisms are not fully understood. Herein, we aimed to modulate electrophysiological properties of hiPSC-CMs within co-cultures containing varied proportions of non-CMs and investigate the nature of interactions between these different cell types. Therefore, we sorted cardiac differentiations on day 10 and subsequently replated the cells at ratios of 7:3, 1:1, 3:7, and 1:9 non-CMs to CMs. After a month of co-culture, we evaluated electrophysiological properties through the genetically encoded voltage indicator ArcLight. We ultimately identified that co-cultures with approximately 70%–90% CM purity demonstrated the highest action potential (AP) amplitude and maximum upstroke velocity by day 40 of differentiation, indicative of enhanced electrophysiological maturation, as well as more ventricular-like AP morphologies. Notably, these findings were distinct from those observed for co-cultures of hiPSC-CMs and dermal fibroblasts. We determined that the co-culture phenotypes could not be attributed to paracrine effects of non-CMs due to the inability of conditioned media to recapitulate the observed effects. This led to the further observation of a distinctive expression pattern of connexin 43 (Cx43) at cell-cell interfaces between both CMs and non-CMs. Depletion of Cx43 by short hairpin RNA (shRNA) specifically in the non-CM population within a co-culture environment was able to recapitulate electrophysiological phenotypes of a purer hiPSC-CM population. Collectively, our data demonstrate that abundant non-CM content exerts a significant negative influence upon the electrophysiological maturation of hiPSC-CMs through Cx43-mediated cell-cell-contacts, and thus should be considered regarding the future production of purpose-built hiPSC-CM systems.
Collapse
Affiliation(s)
- Sherri M Biendarra-Tiegs
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota.,Center for Regenerative Medicine, Mayo Clinic, Rochester, Minnesota
| | - Daniel J Clemens
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - Frank J Secreto
- Center for Regenerative Medicine, Mayo Clinic, Rochester, Minnesota.,Division of General Internal Medicine, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Timothy J Nelson
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota.,Center for Regenerative Medicine, Mayo Clinic, Rochester, Minnesota.,Division of General Internal Medicine, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota.,Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota.,Division of Pediatric Cardiology, Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
44
|
Li P, Liang M, Lu X, Chow JJM, Ramachandra CJA, Ai Y. Sheathless Acoustic Fluorescence Activated Cell Sorting (aFACS) with High Cell Viability. Anal Chem 2019; 91:15425-15435. [DOI: 10.1021/acs.analchem.9b03021] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Peixian Li
- Pillar of Engineering Product Development, Singapore University of Technology and Design, Singapore 487372, Singapore
| | - Minhui Liang
- Pillar of Engineering Product Development, Singapore University of Technology and Design, Singapore 487372, Singapore
| | - Xiaoguang Lu
- Pillar of Engineering Product Development, Singapore University of Technology and Design, Singapore 487372, Singapore
| | - Joycelyn Jia Ming Chow
- Pillar of Engineering Product Development, Singapore University of Technology and Design, Singapore 487372, Singapore
| | - Chrishan J. A. Ramachandra
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore 169609, Singapore
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
| | - Ye Ai
- Pillar of Engineering Product Development, Singapore University of Technology and Design, Singapore 487372, Singapore
| |
Collapse
|
45
|
Quantification of Contractile Dynamic Complexities Exhibited by Human Stem Cell-Derived Cardiomyocytes Using Nonlinear Dimensional Analysis. Sci Rep 2019; 9:14714. [PMID: 31604988 PMCID: PMC6789143 DOI: 10.1038/s41598-019-51197-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 09/26/2019] [Indexed: 12/22/2022] Open
Abstract
Understanding the complexity of biological signals has been gaining widespread attention due to increasing knowledge on the nonlinearity that exists in these systems. Cardiac signals are known to exhibit highly complex dynamics, consisting of high degrees of interdependency that regulate the cardiac contractile functions. These regulatory mechanisms are important to understand for the development of novel in vitro cardiac systems, especially with the exponential growth in deriving cardiac tissue directly from human induced pluripotent stem cells (hiPSCs). This work describes a unique analytical approach that integrates linear amplitude and frequency analysis of physical cardiac contraction, with nonlinear analysis of the contraction signals to measure the signals’ complexity. We generated contraction motion waveforms reflecting the physical contraction of hiPSC-derived cardiomyocytes (hiPSC-CMs) and implemented these signals to nonlinear analysis to compute the capacity and correlation dimensions. These parameters allowed us to characterize the dynamics of the cardiac signals when reconstructed into a phase space and provided a measure of signal complexity to supplement contractile physiology data. Thus, we applied this approach to evaluate drug response and observed that relationships between contractile physiology and dynamic complexity were unique to each tested drug. This illustrated the applicability of this approach in not only characterization of cardiac signals, but also monitoring and diagnostics of cardiac health in response to external stress.
Collapse
|
46
|
Horikoshi Y, Yan Y, Terashvili M, Wells C, Horikoshi H, Fujita S, Bosnjak ZJ, Bai X. Fatty Acid-Treated Induced Pluripotent Stem Cell-Derived Human Cardiomyocytes Exhibit Adult Cardiomyocyte-Like Energy Metabolism Phenotypes. Cells 2019; 8:cells8091095. [PMID: 31533262 PMCID: PMC6769886 DOI: 10.3390/cells8091095] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 09/12/2019] [Accepted: 09/14/2019] [Indexed: 12/17/2022] Open
Abstract
Human induced pluripotent stem cell (iPSC)-derived cardiomyocytes (CMs) (iPSC-CMs) are a promising cell source for myocardial regeneration, disease modeling and drug assessment. However, iPSC-CMs exhibit immature fetal CM-like characteristics that are different from adult CMs in several aspects, including cellular structure and metabolism. As an example, glycolysis is a major energy source for immature CMs. As CMs mature, the mitochondrial oxidative capacity increases, with fatty acid β-oxidation becoming a key energy source to meet the heart’s high energy demand. The immaturity of iPSC-CMs thereby limits their applications. The aim of this study was to investigate whether the energy substrate fatty acid-treated iPSC-CMs exhibit adult CM-like metabolic properties. After 20 days of differentiation from human iPSCs, iPSC-CMs were sequentially cultured with CM purification medium (lactate+/glucose-) for 7 days and maturation medium (fatty acids+/glucose-) for 3–7 days by mimicking the adult CM’s preference of utilizing fatty acids as a major metabolic substrate. The purity and maturity of iPSC-CMs were characterized via the analysis of: (1) Expression of CM-specific markers (e.g., troponin T, and sodium and potassium channels) using RT-qPCR, Western blot or immunofluorescence staining and electron microscopy imaging; and (2) cell energy metabolic profiles using the XF96 Extracellular Flux Analyzer. iPSCs-CMs (98% purity) cultured in maturation medium exhibited enhanced elongation, increased mitochondrial numbers with more aligned Z-lines, and increased expression of matured CM-related genes, suggesting that fatty acid-contained medium promotes iPSC-CMs to undergo maturation. In addition, the oxygen consumption rate (OCR) linked to basal respiration, ATP production, and maximal respiration and spare respiratory capacity (representing mitochondrial function) was increased in matured iPSC-CMs. Mature iPSC-CMs also displayed a larger change in basal and maximum respirations due to the utilization of exogenous fatty acids (palmitate) compared with non-matured control iPSC-CMs. Etomoxir (a carnitine palmitoyltransferase 1 inhibitor) but not 2-deoxyglucose (an inhibitor of glycolysis) abolished the palmitate pretreatment-mediated OCR increases in mature iPSC-CMs. Collectively, our data demonstrate for the first time that fatty acid treatment promotes metabolic maturation of iPSC-CMs (as evidenced by enhanced mitochondrial oxidative function and strong capacity of utilizing fatty acids as energy source). These matured iPSC-CMs might be a promising human CM source for broad biomedical application.
Collapse
Affiliation(s)
- Yuichi Horikoshi
- Department of Emergency Medicine, Asahikawa Medical University, Asahikawa, Hokkaido 078-8510, Japan.
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| | - Yasheng Yan
- Department of Cell Biology, Neuroscience & Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| | - Maia Terashvili
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| | - Clive Wells
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| | - Hisako Horikoshi
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
- Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, Tokyo 113-8421, Japan.
| | - Satoshi Fujita
- Department of Emergency Medicine, Asahikawa Medical University, Asahikawa, Hokkaido 078-8510, Japan.
| | - Zeljko J Bosnjak
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| | - Xiaowen Bai
- Department of Cell Biology, Neuroscience & Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| |
Collapse
|
47
|
Addressing Variability and Heterogeneity of Induced Pluripotent Stem Cell-Derived Cardiomyocytes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1212:1-29. [DOI: 10.1007/5584_2019_350] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
48
|
Ting S, Lam A, Tong G, Chen A, Wei H, Wu J, Lam YN, Reuveny S, Oh S. Meticulous optimization of cardiomyocyte yields in a 3-stage continuous integrated agitation bioprocess. Stem Cell Res 2018; 31:161-173. [DOI: 10.1016/j.scr.2018.07.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 07/12/2018] [Accepted: 07/23/2018] [Indexed: 01/15/2023] Open
|
49
|
Abstract
During the past decades, stem cell-based therapy has acquired a promising role in regenerative medicine. The application of novel cell therapeutics for the treatment of cardiovascular diseases could potentially achieve the ambitious aim of effective cardiac regeneration. Despite the highly positive results from preclinical studies, data from phase I/II clinical trials are inconsistent and the improvement of cardiac remodeling and heart performance was found to be quite limited. The major issues which cardiac stem cell therapy is facing include inefficient cell delivery to the site of injury, accompanied by low cell retention and weak effectiveness of remaining stem cells in tissue regeneration. According to preclinical and clinical studies, various stem cells (adult stem cells, embryonic stem cells, and induced pluripotent stem cells) represent the most promising cell types so far. Beside the selection of the appropriate cell type, researchers have developed several strategies to produce “second-generation” stem cell products with improved regenerative capacity. Genetic and nongenetic modifications, chemical and physical preconditioning, and the application of biomaterials were found to significantly enhance the regenerative capacity of transplanted stem cells. In this review, we will give an overview of the recent developments in stem cell engineering with the goal to facilitate stem cell delivery and to promote their cardiac regenerative activity.
Collapse
|
50
|
Park M, Yoon YS. Cardiac Regeneration with Human Pluripotent Stem Cell-Derived Cardiomyocytes. Korean Circ J 2018; 48:974-988. [PMID: 30334384 PMCID: PMC6196153 DOI: 10.4070/kcj.2018.0312] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 09/27/2018] [Indexed: 12/29/2022] Open
Abstract
Embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), which are collectively called pluripotent stem cells (PSCs), have emerged as a promising source for regenerative medicine. Particularly, human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) have shown robust potential for regenerating injured heart. Over the past two decades, protocols to differentiate hPSCs into CMs at high efficiency have been developed, opening the door for clinical application. Studies further demonstrated therapeutic effects of hPSC-CMs in small and large animal models and the underlying mechanisms of cardiac repair. However, gaps remain in explanations of the therapeutic effects of engrafted hPSC-CMs. In addition, bioengineering technologies improved survival and therapeutic effects of hPSC-CMs in vivo. While most of the original concerns associated with the use of hPSCs have been addressed, several issues remain to be resolved such as immaturity of transplanted cells, lack of electrical integration leading to arrhythmogenic risk, and tumorigenicity. Cell therapy with hPSC-CMs has shown great potential for biological therapy of injured heart; however, more studies are needed to ensure the therapeutic effects, underlying mechanisms, and safety, before this technology can be applied clinically.
Collapse
Affiliation(s)
- Misun Park
- Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Young Sup Yoon
- Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea.,Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|