1
|
Lin Z, Lin B, Hang C, Lu R, Xiong H, Liu J, Wang S, Gong Z, Zhang M, Li D, Fang G, Ding J, Su X, Guo H, Shi D, Xie D, Liu Y, Liang D, Yang J, Chen YH. A new paradigm for generating high-quality cardiac pacemaker cells from mouse pluripotent stem cells. Signal Transduct Target Ther 2024; 9:230. [PMID: 39237509 PMCID: PMC11377569 DOI: 10.1038/s41392-024-01942-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 07/23/2024] [Accepted: 07/31/2024] [Indexed: 09/07/2024] Open
Abstract
Cardiac biological pacing (BP) is one of the future directions for bradyarrhythmias intervention. Currently, cardiac pacemaker cells (PCs) used for cardiac BP are mainly derived from pluripotent stem cells (PSCs). However, the production of high-quality cardiac PCs from PSCs remains a challenge. Here, we developed a cardiac PC differentiation strategy by adopting dual PC markers and simulating the developmental route of PCs. First, two PC markers, Shox2 and Hcn4, were selected to establish Shox2:EGFP; Hcn4:mCherry mouse PSC reporter line. Then, by stepwise guiding naïve PSCs to cardiac PCs following naïve to formative pluripotency transition and manipulating signaling pathways during cardiac PCs differentiation, we designed the FSK method that increased the yield of SHOX2+; HCN4+ cells with typical PC characteristics, which was 12 and 42 folds higher than that of the embryoid body (EB) and the monolayer M10 methods respectively. In addition, the in vitro cardiac PCs differentiation trajectory was mapped by single-cell RNA sequencing (scRNA-seq), which resembled in vivo PCs development, and ZFP503 was verified as a key regulator of cardiac PCs differentiation. These PSC-derived cardiac PCs have the potential to drive advances in cardiac BP technology, help with the understanding of PCs (patho)physiology, and benefit drug discovery for PC-related diseases as well.
Collapse
Affiliation(s)
- Zheyi Lin
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Frontiers Center of Nanocatalytic Medicine, Shanghai, 200092, China
- Department of Pathology and Pathophysiology, School of Medicine, Tongji University, Shanghai, 200092, China
- Clinical Center for Heart Disease Research, Tongji University, Shanghai, 200092, China
| | - Bowen Lin
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Frontiers Center of Nanocatalytic Medicine, Shanghai, 200092, China
- Department of Pathology and Pathophysiology, School of Medicine, Tongji University, Shanghai, 200092, China
- Clinical Center for Heart Disease Research, Tongji University, Shanghai, 200092, China
| | - Chengwen Hang
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Frontiers Center of Nanocatalytic Medicine, Shanghai, 200092, China
- Department of Pathology and Pathophysiology, School of Medicine, Tongji University, Shanghai, 200092, China
- Clinical Center for Heart Disease Research, Tongji University, Shanghai, 200092, China
| | - Renhong Lu
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Frontiers Center of Nanocatalytic Medicine, Shanghai, 200092, China
- Department of Pathology and Pathophysiology, School of Medicine, Tongji University, Shanghai, 200092, China
- Clinical Center for Heart Disease Research, Tongji University, Shanghai, 200092, China
| | - Hui Xiong
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Frontiers Center of Nanocatalytic Medicine, Shanghai, 200092, China
- Clinical Center for Heart Disease Research, Tongji University, Shanghai, 200092, China
- Department of Cell Biology, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Junyang Liu
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Frontiers Center of Nanocatalytic Medicine, Shanghai, 200092, China
- Clinical Center for Heart Disease Research, Tongji University, Shanghai, 200092, China
- Department of Cell Biology, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Siyu Wang
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Zheng Gong
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Mingshuai Zhang
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Frontiers Center of Nanocatalytic Medicine, Shanghai, 200092, China
- Clinical Center for Heart Disease Research, Tongji University, Shanghai, 200092, China
- Department of Cell Biology, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Desheng Li
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Frontiers Center of Nanocatalytic Medicine, Shanghai, 200092, China
- Clinical Center for Heart Disease Research, Tongji University, Shanghai, 200092, China
| | - Guojian Fang
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Frontiers Center of Nanocatalytic Medicine, Shanghai, 200092, China
- Department of Pathology and Pathophysiology, School of Medicine, Tongji University, Shanghai, 200092, China
- Clinical Center for Heart Disease Research, Tongji University, Shanghai, 200092, China
| | - Jie Ding
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Frontiers Center of Nanocatalytic Medicine, Shanghai, 200092, China
- Department of Pathology and Pathophysiology, School of Medicine, Tongji University, Shanghai, 200092, China
- Clinical Center for Heart Disease Research, Tongji University, Shanghai, 200092, China
| | - Xuling Su
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Huixin Guo
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Dan Shi
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Frontiers Center of Nanocatalytic Medicine, Shanghai, 200092, China
- Clinical Center for Heart Disease Research, Tongji University, Shanghai, 200092, China
| | - Duanyang Xie
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Frontiers Center of Nanocatalytic Medicine, Shanghai, 200092, China
- Department of Pathology and Pathophysiology, School of Medicine, Tongji University, Shanghai, 200092, China
- Clinical Center for Heart Disease Research, Tongji University, Shanghai, 200092, China
| | - Yi Liu
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Frontiers Center of Nanocatalytic Medicine, Shanghai, 200092, China
- Department of Pathology and Pathophysiology, School of Medicine, Tongji University, Shanghai, 200092, China
- Clinical Center for Heart Disease Research, Tongji University, Shanghai, 200092, China
| | - Dandan Liang
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Frontiers Center of Nanocatalytic Medicine, Shanghai, 200092, China
- Department of Pathology and Pathophysiology, School of Medicine, Tongji University, Shanghai, 200092, China
- Clinical Center for Heart Disease Research, Tongji University, Shanghai, 200092, China
- Research Units of Origin and Regulation of Heart Rhythm, Chinese Academy of Medical Sciences, Shanghai, 200092, China
| | - Jian Yang
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
- Shanghai Frontiers Center of Nanocatalytic Medicine, Shanghai, 200092, China.
- Clinical Center for Heart Disease Research, Tongji University, Shanghai, 200092, China.
- Department of Cell Biology, School of Medicine, Tongji University, Shanghai, 200092, China.
- Research Units of Origin and Regulation of Heart Rhythm, Chinese Academy of Medical Sciences, Shanghai, 200092, China.
| | - Yi-Han Chen
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
- Shanghai Frontiers Center of Nanocatalytic Medicine, Shanghai, 200092, China.
- Department of Pathology and Pathophysiology, School of Medicine, Tongji University, Shanghai, 200092, China.
- Clinical Center for Heart Disease Research, Tongji University, Shanghai, 200092, China.
- Research Units of Origin and Regulation of Heart Rhythm, Chinese Academy of Medical Sciences, Shanghai, 200092, China.
| |
Collapse
|
2
|
Liu CM, Chen YC, Hu YF. Harnessing cell reprogramming for cardiac biological pacing. J Biomed Sci 2023; 30:74. [PMID: 37633890 PMCID: PMC10463311 DOI: 10.1186/s12929-023-00970-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 08/22/2023] [Indexed: 08/28/2023] Open
Abstract
Electrical impulses from cardiac pacemaker cardiomyocytes initiate cardiac contraction and blood pumping and maintain life. Abnormal electrical impulses bring patients with low heart rates to cardiac arrest. The current therapy is to implant electronic devices to generate backup electricity. However, complications inherent to electronic devices remain unbearable suffering. Therefore, cardiac biological pacing has been developed as a hardware-free alternative. The approaches to generating biological pacing have evolved recently using cell reprogramming technology to generate pacemaker cardiomyocytes in-vivo or in-vitro. Different from conventional methods by electrical re-engineering, reprogramming-based biological pacing recapitulates various phenotypes of de novo pacemaker cardiomyocytes and is more physiological, efficient, and easy for clinical implementation. This article reviews the present state of the art in reprogramming-based biological pacing. We begin with the rationale for this new approach and review its advances in creating a biological pacemaker to treat bradyarrhythmia.
Collapse
Affiliation(s)
- Chih-Min Liu
- Division of Cardiology, Department of Medicine, Heart Rhythm Center, Taipei Veterans General Hospital, Taipei, Taiwan
- Faculty of Medicine and Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-Chun Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yu-Feng Hu
- Division of Cardiology, Department of Medicine, Heart Rhythm Center, Taipei Veterans General Hospital, Taipei, Taiwan.
- Faculty of Medicine and Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
3
|
Rybak‐d'obyrn J, Machoń NJ, Lewandowska JA, Owczarczyk‐Saczonek A, Placek W. Abscess after cardiac pacemaker implantation: A case report. Clin Case Rep 2023; 11:e7639. [PMID: 37554568 PMCID: PMC10405241 DOI: 10.1002/ccr3.7639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 08/10/2023] Open
Abstract
This is a report of one of the most serious complications of the cardiac pacemaker implantation - infection of the implanted system. We present the case, which was misdiagnosed at the beginning and after cardiological consultation it was decided to immediately remove the peacemaker and transfer the patient to the Cardiological Department.
Collapse
Affiliation(s)
- Joanna Rybak‐d'obyrn
- Department of Dermatology, Sexually Transmitted Diseases and Clinical ImmunologyUniversity of Warmia and MazuryOlsztynPoland
| | | | | | - Agnieszka Owczarczyk‐Saczonek
- Department of Dermatology, Sexually Transmitted Diseases and Clinical ImmunologyUniversity of Warmia and MazuryOlsztynPoland
| | - Waldemar Placek
- Department of Dermatology, Sexually Transmitted Diseases and Clinical ImmunologyUniversity of Warmia and MazuryOlsztynPoland
| |
Collapse
|
4
|
Cavero I, Holzgrefe H. Remembering the canonical discoverers of the core components of the mammalian cardiac conduction system: Keith and Flack, Aschoff and Tawara, His, and Purkinje. ADVANCES IN PHYSIOLOGY EDUCATION 2022; 46:549-579. [PMID: 35924782 DOI: 10.1152/advan.00072.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 07/12/2022] [Accepted: 07/25/2022] [Indexed: 03/08/2025]
Abstract
The mammalian cardiac conduction system (CCS) is a multifaceted continuum of electrically distinct, interconnected constructs within the myocardial mass. The key components are the sinus node (SAN), the atrioventricular node (AVN), the His bundle (HB), and the Purkinje fiber network (PF), the latter serendipitously discovered by Jan Evangelista Purkinje in 1839 in the sheep ventricle. In 1893, Wilhelm His, Jr. described a ventricular muscular tract conveying SAN-generated action potentials from the AVN (discovered by Sunao Tawara and Karl Albert Aschoff in 1906) to the PF. In 1906, Keith and Flack completed these explorations by localizing the SAN, the primum movens of CCS, which functions as a biologic oscillator emitting cadenced impulses that travel via the Bachmann bundle to the atrial myocytes and, via internodal pathways, to the AVN. Here these impulses are briefly delayed, enabling atrial systole before continuing via the AVN and the high-speed His-Purkinje conduction axis to signal ventricular contraction. The CCS canonical discoverers (Keith and Flack, Aschoff and Tawara, His, and Purkinje), historical controversies, fundamental notions of anatomy, physiology, and pathology, and therapeutic interventions pertaining to the CCS are the main themes of this review. Any scientist mentioned or unmentioned in this report who contributed directly or indirectly, with correct or inaccurate hypotheses, to the characterization of the CCS deserves our deepest gratitude for the long and painstaking hours spent microscopically scrutinizing heart specimens from multiple mammalian species, including humans.NEW & NOTEWORTHY This report presents the first comprehensive summary of the factors enabling the discovery of the cardiac conduction system (CCS). Biographical highlights and achievements of the CCS canonical discoverers, hypotheses concerning mechanisms underlying sinus node (SAN) automaticity, use of eponyms to denominate a discovery, famous controversies, possible reproachable behavior (e.g., intellectual support for eugenics in post-World War I Germany) by two of the discoverers, and examples of historical mentor-pupil relationships are discussed.
Collapse
|
5
|
Zhang X, Zhao Y, Zhou Y, Lv J, Peng J, Zhu H, Liu R. Trends in research on sick sinus syndrome: A bibliometric analysis from 2000 to 2022. Front Cardiovasc Med 2022; 9:991503. [DOI: 10.3389/fcvm.2022.991503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 10/19/2022] [Indexed: 11/11/2022] Open
Abstract
Sick sinus syndrome (SSS) is a refractory arrhythmia disease caused by the pathological changes of sinoatrial node and its adjacent tissues. 2,251 publications related to SSS were retrieved from Web of Science database from 2000 to 2022 and analyzed by using VOS viewer and CiteSpace software. The results showed the United States dominated the field, followed by Japan, Germany, and China. SSS was closely related to risk factors such as atrial fibrillation and aging. Sick sinus syndrome, atrial fibrillation and sinus node dysfunction were the top three keywords that had the strongest correlation with the study. Pacemaker implantation, differentiation and mutation are research hotspots currently. Clinical studies on SSS found that sick sinus syndrome, atrial fibrillation, and pacemakers were the top three keywords that had the largest nodes and the highest frequency. In the field of basic applied research and basic research, atrial fibrillation and pacemaker cells were the focus of research. In conclusion, bibliometric analysis provided valuable information for the prevention, treatment and future research trends of SSS.
Collapse
|
6
|
Li J, Wiesinger A, Fokkert L, Boukens BJ, Verkerk AO, Christoffels VM, Boink GJ, Devalla HD. Molecular and electrophysiological evaluation of human cardiomyocyte subtypes to facilitate generation of composite cardiac models. J Tissue Eng 2022; 13:20417314221127908. [PMID: 36277058 PMCID: PMC9583221 DOI: 10.1177/20417314221127908] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 09/06/2022] [Indexed: 11/06/2022] Open
Abstract
Paucity of physiologically relevant cardiac models has limited the widespread application of human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes in drug development. Here, we performed comprehensive characterization of hiPSC-derived cardiomyocyte subtypes from 2D and 3D cultures and established a novel 3D model to study impulse initiation and propagation. Directed differentiation approaches were used to generate sinoatrial nodal (SANCM), atrial (ACM) and ventricular cardiomyocytes (VCM). Single cell RNA sequencing established that the protocols yield distinct cell populations in line with expected identities, which was also confirmed by electrophysiological characterization. In 3D EHT cultures of all subtypes, we observed prominent expression of stretch-responsive genes such as NPPA. Response to rate modulating drugs noradrenaline, carbachol and ivabradine were comparable in single cells and EHTs. Differences in the speed of impulse propagation between the subtypes were more pronounced in EHTs compared with 2D monolayers owing to a progressive increase in conduction velocities in atrial and ventricular cardiomyocytes, in line with a more mature phenotype. In a novel binary EHT model of pacemaker-atrial interface, the SANCM end of the tissue consistently paced the EHTs under baseline conditions, which was inhibited by ivabradine. Taken together, our data provide comprehensive insights into molecular and electrophysiological properties of hiPSC-derived cardiomyocyte subtypes, facilitating the creation of next generation composite cardiac models for drug discovery, disease modeling and cell-based regenerative therapies.
Collapse
Affiliation(s)
- Jiuru Li
- Department of Medical Biology,
Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The
Netherlands
| | - Alexandra Wiesinger
- Department of Medical Biology,
Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The
Netherlands
| | - Lianne Fokkert
- Department of Medical Biology,
Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The
Netherlands
| | - Bastiaan J. Boukens
- Department of Medical Biology,
Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The
Netherlands
| | - Arie O. Verkerk
- Department of Medical Biology,
Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The
Netherlands,Department of Experimental Cardiology,
Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The
Netherlands
| | - Vincent M. Christoffels
- Department of Medical Biology,
Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The
Netherlands
| | - Gerard J.J. Boink
- Department of Medical Biology,
Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The
Netherlands,Department of Cardiology, Amsterdam
University Medical Centers, University of Amsterdam, Amsterdam, The
Netherlands
| | - Harsha D. Devalla
- Department of Medical Biology,
Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The
Netherlands,Harsha D Devalla, Department of Medical
Biology, Amsterdam University Medical Centers, University of Amsterdam,
Meibergdreef 9, Amsterdam 1105 AZ, The Netherlands.
| |
Collapse
|
7
|
Wiesinger A, Li J, Fokkert L, Bakker P, Verkerk AO, Christoffels VM, Boink GJJ, Devalla HD. A single cell transcriptional roadmap of human pacemaker cell differentiation. eLife 2022; 11:76781. [PMID: 36217819 PMCID: PMC9553210 DOI: 10.7554/elife.76781] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 08/16/2022] [Indexed: 12/26/2022] Open
Abstract
Each heartbeat is triggered by the sinoatrial node (SAN), the primary pacemaker of the heart. Studies in animal models have revealed that pacemaker cells share a common progenitor with the (pro)epicardium, and that the pacemaker cardiomyocytes further diversify into ‘transitional’, ‘tail’, and ‘head’ subtypes. However, the underlying molecular mechanisms, especially of human pacemaker cell development, are poorly understood. Here, we performed single cell RNA sequencing (scRNA-seq) and trajectory inference on human induced pluripotent stem cells (hiPSCs) differentiating to SAN-like cardiomyocytes (SANCMs) to construct a roadmap of transcriptional changes and lineage decisions. In differentiated SANCM, we identified distinct clusters that closely resemble different subpopulations of the in vivo SAN. Moreover, the presence of a side population of proepicardial cells suggested their shared ontogeny with SANCM, as also reported in vivo. Our results demonstrate that the divergence of SANCM and proepicardial lineages is determined by WNT signaling. Furthermore, we uncovered roles for TGFβ and WNT signaling in the branching of transitional and head SANCM subtypes, respectively. These findings provide new insights into the molecular processes involved in human pacemaker cell differentiation, opening new avenues for complex disease modeling in vitro and inform approaches for cell therapy-based regeneration of the SAN.
Collapse
Affiliation(s)
- Alexandra Wiesinger
- Department of Medical Biology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Jiuru Li
- Department of Medical Biology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Lianne Fokkert
- Department of Medical Biology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Priscilla Bakker
- Department of Medical Biology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Arie O Verkerk
- Department of Medical Biology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands.,Department of Experimental Cardiology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Vincent M Christoffels
- Department of Medical Biology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Gerard J J Boink
- Department of Medical Biology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands.,Department of Cardiology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Harsha D Devalla
- Department of Medical Biology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
8
|
Buja LM, Schoen FJ. The pathology of cardiovascular interventions and devices for coronary artery disease, vascular disease, heart failure, and arrhythmias. Cardiovasc Pathol 2022. [DOI: 10.1016/b978-0-12-822224-9.00024-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
9
|
Mehek R, Iqbal N, Noor T, Amjad MZB, Ali G, Vignarooban K, Khan MA. Metal-organic framework based electrode materials for lithium-ion batteries: a review. RSC Adv 2021; 11:29247-29266. [PMID: 35479575 PMCID: PMC9040901 DOI: 10.1039/d1ra05073g] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/20/2021] [Indexed: 12/25/2022] Open
Abstract
Metal-organic frameworks (MOFs) with efficient surface and structural properties have risen as a distinctive class of porous materials through the last few decades, which has enabled MOFs to gain attention in a wide range of applications like drug delivery, gas separation and storage, catalysis and sensors. Likewise, they have also emerged as efficient active materials in energy storage devices owing to their remarkable conducting properties. Metal-organic frameworks (MOFs) have garnered great interest in high-energy-density rechargeable batteries and super-capacitors. Herein the study presents their expanding diversity, structures and chemical compositions which can be tuned at the molecular level. It also aims to evaluate their inherently porous framework and how it facilitates electronic and ionic transportation through the charging and discharging cycles of lithium-ion batteries. In this review we have summarized the various synthesis paths to achieve a particular metal-organic framework. This study focuses mainly on the implementation of metal-organic frameworks as efficient anode and cathode materials for lithium-ion batteries (LIBs) with an evaluation of their influence on cyclic stability and discharge capacity. For this purpose, a brief assessment is made of recent developments in metal-organic frameworks as anode or cathode materials for lithium-ion batteries which would provide enlightenment in optimizing the reaction conditions for designing a MOF structure for the battery community and electrochemical energy storage applications.
Collapse
Affiliation(s)
- Rimsha Mehek
- U.S.-Pakistan Center for Advanced Studies in Energy (USPCAS-E), National University of Sciences and Technology (NUST) H-12 Campus Islamabad 44000 Pakistan +92 51 9085 5281
| | - Naseem Iqbal
- U.S.-Pakistan Center for Advanced Studies in Energy (USPCAS-E), National University of Sciences and Technology (NUST) H-12 Campus Islamabad 44000 Pakistan +92 51 9085 5281
| | - Tayyaba Noor
- School of Chemical and Materials Engineering (SCME), National University of Sciences and Technology (NUST) Islamabad Pakistan
| | - M Zain Bin Amjad
- U.S.-Pakistan Center for Advanced Studies in Energy (USPCAS-E), National University of Sciences and Technology (NUST) H-12 Campus Islamabad 44000 Pakistan +92 51 9085 5281
| | - Ghulam Ali
- U.S.-Pakistan Center for Advanced Studies in Energy (USPCAS-E), National University of Sciences and Technology (NUST) H-12 Campus Islamabad 44000 Pakistan +92 51 9085 5281
| | - K Vignarooban
- Department of Physics, Faculty of Science, University of Jaffna Jaffna 40000 Sri Lanka
| | - M Abdullah Khan
- Renewable Energy Advancement Laboratory (REAL), Department of Environmental Sciences, Quaid-i-Azam University Islamabad 45320 Pakistan
| |
Collapse
|
10
|
Lopez-Lopez D, Gestal Vazquez M, Rama-Maceiras P, Gonzalez Pereira A, Casas Reza P. Perioperative management of epicardial pacemakers: A case report. J Clin Anesth 2021; 75:110478. [PMID: 34358851 DOI: 10.1016/j.jclinane.2021.110478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 07/25/2021] [Accepted: 07/27/2021] [Indexed: 10/20/2022]
Affiliation(s)
- David Lopez-Lopez
- Anesthesiology, Perioperative Care and Pain Medicine, Complexo Hospitalario Universitario de A Coruña, A Coruña, Spain.
| | - MarIa Gestal Vazquez
- Anesthesiology, Perioperative Care and Pain Medicine, Complexo Hospitalario Universitario de A Coruña, A Coruña, Spain.
| | - Pablo Rama-Maceiras
- Anesthesiology, Perioperative Care and Pain Medicine, Complexo Hospitalario Universitario de A Coruña, A Coruña, Spain.
| | - Almudena Gonzalez Pereira
- Anesthesiology, Perioperative Care and Pain Medicine, Complexo Hospitalario Universitario de A Coruña, A Coruña, Spain.
| | - Pablo Casas Reza
- Anesthesiology, Perioperative Care and Pain Medicine, Complexo Hospitalario Universitario de A Coruña, A Coruña, Spain.
| |
Collapse
|
11
|
Zhu Y, You J, Wei W, Gu J, Xu C, Gu X. Downregulated lncRNA RCPCD promotes differentiation of embryonic stem cells into cardiac pacemaker-like cells by suppressing HCN4 promoter methylation. Cell Death Dis 2021; 12:667. [PMID: 34215719 PMCID: PMC8253811 DOI: 10.1038/s41419-021-03949-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 06/16/2021] [Accepted: 06/16/2021] [Indexed: 12/25/2022]
Abstract
Long non-coding RNA (lncRNA) is receiving increasing attention in embryonic stem cells (ESCs) research. However, the roles of lncRNA in the differentiation of ESCs into pacemaker-like cells are still unclear. Therefore, the present study aims to explore the roles and mechanisms of lncRNA in the differentiation of ESCs into pacemaker-like cells. ESCs were cultured and induced differentiation to pacemaker-like cells. RNA sequencing was used to identify the differential expression lncRNAs during the differentiation of ESCs into pacemaker-like cells. Cell morphology observation, flow cytometry, quantitative real-time polymerase chain reaction, western blot, and immunofluorescence were used to detect the differentiation of ESCs into pacemaker-like cells. LncRNA and genes overexpression or knockdown through transfected adenovirus in the differentiation process. The fluorescence in situ hybridization (FISH) detected the lncRNA location in the differentiated ESCs. Luciferase reporter gene assay, methylation-specific PCR, chromatin immunoprecipitation assay, and RNA immunoprecipitation assay were performed to reveal the mechanism of lncRNA-regulating HCN4 expression. Rescue experiments were used to confirm that lncRNA regulates the differentiation of ESCs into pacemaker-like cells through HCN4. We cultured the ESCs and induced the differentiation of ESCs into pacemaker-like cells successfully. The expression of lncRNA RCPCD was significantly decreased in the differentiation of ESCs into pacemaker-like cells. Overexpression of RCPCD inhibited the differentiation of ESCs into pacemaker-like cells. RCPCD inhibited the expression of HCN4 by increasing HCN4 methylation at the promoter region through DNMT1, DNMT2, and DNMT3. RCPCD inhibited the differentiation of ESCs into pacemaker-like cells by inhibiting the expression of HCN4. Our results confirm the roles and mechanism of lncRNA RCPCD in the differentiation of ESCs into pacemaker-like cells, which could pave the path for the development of a cell-based biological pacemaker.
Collapse
Affiliation(s)
- Ye Zhu
- Clinical Medical College of Yangzhou University, Yangzhou, China. .,Department of Cardiology, Northern Jiangsu People's Hospital, Yangzhou, China.
| | - Jia You
- Department of Internal Medicine, Yangzhou Maternal and Child Health Care Hospital, Yangzhou, Jiangsu, 225001, China
| | - Wei Wei
- Clinical Medical College of Yangzhou University, Yangzhou, China.,Department of Cardiology, Northern Jiangsu People's Hospital, Yangzhou, China
| | - Jianjun Gu
- Clinical Medical College of Yangzhou University, Yangzhou, China.,Department of Cardiology, Northern Jiangsu People's Hospital, Yangzhou, China
| | - Chao Xu
- Department of Biostatistics and Epidemiology, University of Oklahoma Health Science Center, Oklahoma City, OK, 73104, US
| | - Xiang Gu
- Clinical Medical College of Yangzhou University, Yangzhou, China.,Department of Cardiology, Northern Jiangsu People's Hospital, Yangzhou, China
| |
Collapse
|
12
|
Boink GJJ, Coronel R. Towards Molecular Therapy of Atrioventricular Nodal Dysfunction. Circ Res 2021; 129:6-8. [PMID: 34166074 DOI: 10.1161/circresaha.121.319450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
| | - Ruben Coronel
- Amsterdam University Medical Centers, the Netherlands
| |
Collapse
|
13
|
Ismail MAM, Ibrahim NM, Che Hasan MK. What Do Our Nurses Know about Managing Patient with Permanent Pacemakers? JURNAL NERS 2020. [DOI: 10.20473/jn.v15i2.18925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Introduction: The number of patients with pacemaker implant is increasing in the health services sector in Malaysia, which requires nurses to have expertise in patient care with pacemaker implantation. Therefore, this study was conducted to analyse the level of knowledge among nurses regarding the management of patients with pacemaker implantation.Methods: A cross-sectional study was conducted through purposive sampling among all nurses working at the critical care unit, intensive care unit, cardiac rehabilitation ward, investigation clinical laboratory, and non-invasive clinical laboratory in a public hospital in Kelantan. A questionnaire consisted of demographic data and nurses’ knowledge was distributed. Data were analysed for descriptive analysis and using Pearson correlation test.Results: Results from all respondents (n=70), show 48.6 % of the respondents had moderate knowledge about patient management with pacemaker implantation, 32.9 % had a low level of knowledge and only 13.6% had high knowledge regarding management of patient with pacemaker implantation. There is a significant difference between the level of knowledge and demographic data, that is between the level of education (p=0.027), age (p=0.011) and length of service (p=0.015). There is no significant relationship between knowledge and demographic data, such as gender (p=0.481), marital status (p=0.315), and post-basic (p=0.067).Conclusion: Level of knowledge among nurses about the management of patient with pacemaker implantation is low to moderate. Additional education and exposure among nurses are needed to enhance the knowledge of nurses and improve the quality of care among patients with pacemaker implant.
Collapse
|
14
|
Takayama I, Kondo N, Kalies S, Heisterkamp A, Terakawa M. Myoblast adhesion and proliferation on biodegradable polymer films with femtosecond laser-fabricated micro through-holes. JOURNAL OF BIOPHOTONICS 2020; 13:e202000037. [PMID: 32250039 DOI: 10.1002/jbio.202000037] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/17/2020] [Accepted: 03/23/2020] [Indexed: 06/11/2023]
Abstract
Controlling cell adhesion and cell differentiation is necessary to fabricate a tissue with arbitrary properties for tissue engineering applications. A substrate with a porous structure as a cell scaffold allows the diffusion of the cell culture medium through the scaffold. In this work, we show that the femtosecond laser fabricated micro through-holes in biodegradable polymer films, enhance myoblast adhesion, and accelerates proliferation and differentiation. ChR2-C2C12 and UT-C2C12 cells were seeded on the films with micro through-holes each fabricated by a single femtosecond laser pulse. Cell adhesion was enhanced on films with holes fabricated by laser irradiation. In addition, cell proliferation was accelerated on films with micro through-holes that penetrate the film, compared to on films with micro craters that do not penetrate the film. On films with arrays consisting of micro through-holes, cells aligned along the arrays and cell fusion was enhanced, indicating the acceleration of cell differentiation.
Collapse
Affiliation(s)
- Izumi Takayama
- School of Integrated Design Engineering, Keio University, Yokohama-shi, Japan
| | - Naonari Kondo
- School of Integrated Design Engineering, Keio University, Yokohama-shi, Japan
| | - Stefan Kalies
- Institut fuer Quantenoptik, Gottfried Wilhelm Leibniz University Hannover, Hannover, Germany
| | - Alexander Heisterkamp
- Institut fuer Quantenoptik, Gottfried Wilhelm Leibniz University Hannover, Hannover, Germany
- Industrial and Biomedical Optics Department, Laser Zentrum Hannover e.V., Hannover, Germany
| | - Mitsuhiro Terakawa
- School of Integrated Design Engineering, Keio University, Yokohama-shi, Japan
- Department of Electronics and Electrical Engineering, Keio University, Yokohama-shi, Japan
| |
Collapse
|
15
|
Zhao H, Wang F, Tang Y, Wang X, Wang T, Zhao Q, Huang C. HCN2 and TBX3 Reprogram Human-Induced Pluripotent Stem Cells-Derived Cardiomyocytes into Pacemaker-Like Cells. DNA Cell Biol 2020; 39:289-298. [PMID: 31916853 DOI: 10.1089/dna.2019.5135] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
TBX3 reprograms cardiac myocytes into cells that possess sinoatrial node phenotype, but no specific funny current (If) was detected. We explore whether overexpression of TBX3 alone or combined with HCN2 can reprogram human-induced pluripotent stem cells-derived cardiomyocytes (hiPSC-CMs) into pacemaker-like cells. HiPSC-CMs were transfected with TBX3 and/or HCN2 in this study. Expression analysis showed that overexpression of TBX3 induces a reduced reduction expression profile of working cardiomyocytes into that of pacemaker cells. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and electrophysiological analyses showed a reduced expression of connexins subunits (CX40, CX43), the sodium current (SCN5A, INa), the inward rectified potassium channels (Kir2.1, IK1), and an increased expression of connexins subunits (CX30.2, CX45). No If was detected. The reduction of IK1 resulted in a more depolarized maximum diastolic potential together with an expression of If (generated by HCN2), which they work in synergy to generate spontaneous diastolic depolarization that was the most typical characteristic of pacemaker cells. In conclusion, overexpression of TBX3 and HCN2 could reprogram hiPSC-CMs into pacemaker-like cells. The ability to enable diastolic depolarization formation provides a new strategy for the construction of a biological pacemaker.
Collapse
Affiliation(s)
- Hongyi Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China.,Cardiovascular Research Institute, Wuhan University, Wuhan, P.R. China.,Hubei Key Laboratory of Cardiology, Wuhan, P.R. China
| | - Fengyuan Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China.,Cardiovascular Research Institute, Wuhan University, Wuhan, P.R. China.,Hubei Key Laboratory of Cardiology, Wuhan, P.R. China
| | - Yanhong Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China.,Cardiovascular Research Institute, Wuhan University, Wuhan, P.R. China.,Hubei Key Laboratory of Cardiology, Wuhan, P.R. China
| | - Xi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China.,Cardiovascular Research Institute, Wuhan University, Wuhan, P.R. China.,Hubei Key Laboratory of Cardiology, Wuhan, P.R. China
| | - Teng Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China.,Cardiovascular Research Institute, Wuhan University, Wuhan, P.R. China.,Hubei Key Laboratory of Cardiology, Wuhan, P.R. China
| | - Qingyan Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China.,Cardiovascular Research Institute, Wuhan University, Wuhan, P.R. China.,Hubei Key Laboratory of Cardiology, Wuhan, P.R. China
| | - Congxin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China.,Cardiovascular Research Institute, Wuhan University, Wuhan, P.R. China.,Hubei Key Laboratory of Cardiology, Wuhan, P.R. China
| |
Collapse
|
16
|
Wang F, Zhao H, Yin L, Tang Y, Wang X, Zhao Q, Wang T, Huang C. Transcription Factor TBX18 Reprograms Vascular Smooth Muscle Cells of Ascending Aorta to Pacemaker-Like Cells. DNA Cell Biol 2019; 38:1470-1479. [PMID: 31633376 DOI: 10.1089/dna.2019.4940] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Vascular smooth muscle cells (VSMCs) of ascending aorta and TBX18+ sinus node both originated from the second heart field. The study explored whether ascending aortic smooth muscle cells in vitro could be reprogrammed into pacemaker-like cells with human TBX18. In the study, VSMCs were infected with TBX18, and then cocultured with neonatal rat ventricular cardiomyocytes (NRVMs) in vitro. By overexpressing TBX18, the transfected VSMCs expressed high levels of hyperpolarization-activated cyclic nucleotide-gated channel 4 (HCN4), insulin gene enhancer binding protein 1, and human dwarf homeobox gene SHOX2, cardiac troponin I, and low level of connexin 43. In addition, funny current (If) was recorded by patch clamp appeared the time and voltage dependence in TBX18 group, which the amplitude of If density was from -5.164 ± 0.662 pA/pF to -0.765 ± 0.358 pA/pF (n = 14). Furthermore, TBX18-transfected VSMCs coupled with NRVMs showed typical action potential of pacemaker-like cells and the beating rate was faster (178.00 ± 7.55 bpm, p < 0.05) compared with other groups. In conclusion, our study indicated that transcription factor TBX18 could reprogram VSMCs into pacemaker-like cells in vitro.
Collapse
Affiliation(s)
- Fengyuan Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China.,Hubei Key Laboratory of Cardiology, Wuhan, People's Republic of China
| | - Hongyi Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China.,Hubei Key Laboratory of Cardiology, Wuhan, People's Republic of China
| | - Lin Yin
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China.,Hubei Key Laboratory of Cardiology, Wuhan, People's Republic of China
| | - Yanhong Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China.,Hubei Key Laboratory of Cardiology, Wuhan, People's Republic of China
| | - Xi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China.,Hubei Key Laboratory of Cardiology, Wuhan, People's Republic of China
| | - Qingyan Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China.,Hubei Key Laboratory of Cardiology, Wuhan, People's Republic of China
| | - Teng Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China.,Hubei Key Laboratory of Cardiology, Wuhan, People's Republic of China
| | - Congxin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China.,Hubei Key Laboratory of Cardiology, Wuhan, People's Republic of China
| |
Collapse
|
17
|
Abstract
The rate and rhythm of heart muscle contractions are coordinated by the cardiac conduction system (CCS), a generic term for a collection of different specialized muscular tissues within the heart. The CCS components initiate the electrical impulse at the sinoatrial node, propagate it from atria to ventricles via the atrioventricular node and bundle branches, and distribute it to the ventricular muscle mass via the Purkinje fibre network. The CCS thereby controls the rate and rhythm of alternating contractions of the atria and ventricles. CCS function is well conserved across vertebrates from fish to mammals, although particular specialized aspects of CCS function are found only in endotherms (mammals and birds). The development and homeostasis of the CCS involves transcriptional and regulatory networks that act in an embryonic-stage-dependent, tissue-dependent, and dose-dependent manner. This Review describes emerging data from animal studies, stem cell models, and genome-wide association studies that have provided novel insights into the transcriptional networks underlying CCS formation and function. How these insights can be applied to develop disease models and therapies is also discussed.
Collapse
|
18
|
Schulze ML, Lemoine MD, Fischer AW, Scherschel K, David R, Riecken K, Hansen A, Eschenhagen T, Ulmer BM. Dissecting hiPSC-CM pacemaker function in a cardiac organoid model. Biomaterials 2019; 206:133-145. [DOI: 10.1016/j.biomaterials.2019.03.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 03/15/2019] [Accepted: 03/17/2019] [Indexed: 12/21/2022]
|
19
|
van Eif VWW, Stefanovic S, van Duijvenboden K, Bakker M, Wakker V, de Gier-de Vries C, Zaffran S, Verkerk AO, Boukens BJ, Christoffels VM. Transcriptome analysis of mouse and human sinoatrial node cells reveals a conserved genetic program. Development 2019; 146:dev.173161. [PMID: 30936179 DOI: 10.1242/dev.173161] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 03/20/2019] [Indexed: 02/03/2023]
Abstract
The rate of contraction of the heart relies on proper development and function of the sinoatrial node, which consists of a small heterogeneous cell population, including Tbx3+ pacemaker cells. Here, we have isolated and characterized the Tbx3+ cells from Tbx3 +/Venus knock-in mice. We studied electrophysiological parameters during development and found that Venus-labeled cells are genuine Tbx3+ pacemaker cells. We analyzed the transcriptomes of late fetal FACS-purified Tbx3+ sinoatrial nodal cells and Nppb-Katushka+ atrial and ventricular chamber cardiomyocytes, and identified a sinoatrial node-enriched gene program, including key nodal transcription factors, BMP signaling and Smoc2, the disruption of which in mice did not affect heart rhythm. We also obtained the transcriptomes of the sinoatrial node region, including pacemaker and other cell types, and right atrium of human fetuses, and found a gene program including TBX3, SHOX2, ISL1 and HOX family members, and BMP and NOTCH signaling components conserved between human and mouse. We conclude that a conserved gene program characterizes the sinoatrial node region and that the Tbx3 +/Venus allele provides a reliable tool for visualizing the sinoatrial node, and studying its development and function.
Collapse
Affiliation(s)
- Vincent W W van Eif
- Department of Medical Biology, University of Amsterdam, Amsterdam University Medical Centers, Amsterdam 1105 AZ, The Netherlands
| | - Sonia Stefanovic
- Aix-Marseille University - INSERM U1251, Marseille Medical Genetics, Marseille 13005, France
| | - Karel van Duijvenboden
- Department of Medical Biology, University of Amsterdam, Amsterdam University Medical Centers, Amsterdam 1105 AZ, The Netherlands
| | - Martijn Bakker
- Department of Medical Biology, University of Amsterdam, Amsterdam University Medical Centers, Amsterdam 1105 AZ, The Netherlands
| | - Vincent Wakker
- Department of Medical Biology, University of Amsterdam, Amsterdam University Medical Centers, Amsterdam 1105 AZ, The Netherlands
| | - Corrie de Gier-de Vries
- Department of Medical Biology, University of Amsterdam, Amsterdam University Medical Centers, Amsterdam 1105 AZ, The Netherlands
| | - Stéphane Zaffran
- Aix-Marseille University - INSERM U1251, Marseille Medical Genetics, Marseille 13005, France
| | - Arie O Verkerk
- Department of Medical Biology, University of Amsterdam, Amsterdam University Medical Centers, Amsterdam 1105 AZ, The Netherlands
| | - Bas J Boukens
- Department of Medical Biology, University of Amsterdam, Amsterdam University Medical Centers, Amsterdam 1105 AZ, The Netherlands
| | - Vincent M Christoffels
- Department of Medical Biology, University of Amsterdam, Amsterdam University Medical Centers, Amsterdam 1105 AZ, The Netherlands
| |
Collapse
|
20
|
Zhang J, Huang C. A new combination of transcription factors increases the harvesting efficiency of pacemaker‑like cells. Mol Med Rep 2019; 19:3584-3592. [PMID: 30864738 PMCID: PMC6472109 DOI: 10.3892/mmr.2019.10012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 02/01/2019] [Indexed: 01/22/2023] Open
Abstract
Biological pacemakers that combine cell-based and gene-based therapies are a promising treatment for sick sinus syndrome or severe atrioventricular block. The current study aimed to induce differentiation of adipose tissue-derived stem cells (ADSCs) into cardiac pacemaker cells through co-expression of the transcription factors insulin gene enhancer binding protein 1 (ISL-1) and T-box18 (Tbx18). ADSCs were transfected with green fluorescent protein, ISL-1, Tbx18 or ISL-1+Tbx18 fluorescent protein lentiviral vectors, and subsequently co-cultured with neonatal rat ventricular cardiomyocytes in vitro for 7 days. The potential for regulating the differentiation of ADSCs into pacemaker-like cells was evaluated by cell morphology, beating rate, reverse transcription-quantitative polymerase chain reaction, western blotting, immunofluorescence and electrophysiological activity. ADSCs were successfully transformed into spontaneously beating cells that exhibited a behavior similar to that of co-cultured pacemaker cells. This effect was significantly increased in the combined ISL-1 and Tbx18 group. These results provide a potential strategy for enriching the cardiac pacemaker cell population from ADSCs.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Congxin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
21
|
Cardiomyocyte Progenitor Cells as a Functional Gene Delivery Vehicle for Long-Term Biological Pacing. Molecules 2019; 24:molecules24010181. [PMID: 30621310 PMCID: PMC6337610 DOI: 10.3390/molecules24010181] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 12/28/2018] [Accepted: 12/31/2018] [Indexed: 01/16/2023] Open
Abstract
Sustained pacemaker function is a challenge in biological pacemaker engineering. Human cardiomyocyte progenitor cells (CMPCs) have exhibited extended survival in the heart after transplantation. We studied whether lentivirally transduced CMPCs that express the pacemaker current If (encoded by HCN4) can be used as functional gene delivery vehicle in biological pacing. Human CMPCs were isolated from fetal hearts using magnetic beads coated with Sca-1 antibody, cultured in nondifferentiating conditions, and transduced with a green fluorescent protein (GFP)- or HCN4-GFP-expressing lentivirus. A patch-clamp analysis showed a large hyperpolarization-activated, time-dependent inward current (−20 pA/pF at −140 mV, n = 14) with properties typical of If in HCN4-GFP-expressing CMPCs. Gap-junctional coupling between CMPCs and neonatal rat ventricular myocytes (NRVMs) was demonstrated by efficient dye transfer and changes in spontaneous beating activity. In organ explant cultures, the number of preparations showing spontaneous beating activity increased from 6.3% in CMPC/GFP-injected preparations to 68.2% in CMPC/HCN4-GFP-injected preparations (P < 0.05). Furthermore, in CMPC/HCN4-GFP-injected preparations, isoproterenol induced a significant reduction in cycle lengths from 648 ± 169 to 392 ± 71 ms (P < 0.05). In sum, CMPCs expressing HCN4-GFP functionally couple to NRVMs and induce physiologically controlled pacemaker activity and may therefore provide an attractive delivery platform for sustained pacemaker function.
Collapse
|
22
|
Zhang J, Yang M, Yang AK, Wang X, Tang YH, Zhao QY, Wang T, Chen YT, Huang CX. Insulin gene enhancer binding protein 1 induces adipose tissue‑derived stem cells to differentiate into pacemaker‑like cells. Int J Mol Med 2018; 43:879-889. [PMID: 30483766 PMCID: PMC6317671 DOI: 10.3892/ijmm.2018.4002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 11/20/2018] [Indexed: 01/22/2023] Open
Abstract
Hybrid approaches combining gene- and cell-based therapies to make biological pacemakers are a promising therapeutic avenue for bradyarrhythmia. The present study aimed to direct adipose tissue-derived stem cells (ADSCs) to differentiate specifically into cardiac pacemaker cells by overexpressing a single transcription factor, insulin gene enhancer binding protein 1 (ISL-1). In the present study, the ADSCs were transfected with ISL‑1 or mCherry fluorescent protein lentiviral vectors and co-cultured with neonatal rat ventricular cardiomyocytes (NRVMs) in vitro for 5-7 days. The feasibility of regulating the differentiation of ADSCs into pacemaker-like cells by overexpressing ISL-1 was evaluated by observation of cell morphology and beating rate, reverse transcription-quantitative polymerase chain reaction analysis, western blotting, immunofluorescence and analysis of electrophysiological activity. In conclusion, these data indicated that the overexpression of ISL-1 in ADSCs may enhance the pacemaker phenotype and automaticity in vitro, features which were significantly increased following co‑culture induction.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Mei Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - An-Kang Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Xi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yan-Hong Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qing-Yan Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Teng Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yu-Ting Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Cong-Xin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
23
|
Mohan RA, Mommersteeg MTM, Domínguez JN, Choquet C, Wakker V, de Gier-de Vries C, Boink GJJ, Boukens BJ, Miquerol L, Verkerk AO, Christoffels VM. Embryonic Tbx3 + cardiomyocytes form the mature cardiac conduction system by progressive fate restriction. Development 2018; 145:dev167361. [PMID: 30042181 DOI: 10.1242/dev.167361] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 07/09/2018] [Indexed: 12/21/2022]
Abstract
A small network of spontaneously active Tbx3+ cardiomyocytes forms the cardiac conduction system (CCS) in adults. Understanding the origin and mechanism of development of the CCS network are important steps towards disease modeling and the development of biological pacemakers to treat arrhythmias. We found that Tbx3 expression in the embryonic mouse heart is associated with automaticity. Genetic inducible fate mapping revealed that Tbx3+ cells in the early heart tube are fated to form the definitive CCS components, except the Purkinje fiber network. At mid-fetal stages, contribution of Tbx3+ cells was restricted to the definitive CCS. We identified a Tbx3+ population in the outflow tract of the early heart tube that formed the atrioventricular bundle. Whereas Tbx3+ cardiomyocytes also contributed to the adjacent Gja5+ atrial and ventricular chamber myocardium, embryonic Gja5+ chamber cardiomyocytes did not contribute to the Tbx3+ sinus node or to atrioventricular ring bundles. In conclusion, the CCS is established by progressive fate restriction of a Tbx3+ cell population in the early developing heart, which implicates Tbx3 as a useful tool for developing strategies to study and treat CCS diseases.
Collapse
Affiliation(s)
- Rajiv A Mohan
- Department of Medical Biology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam 1105 AZ, The Netherlands
- Department of Cardiology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam 1105 AZ, The Netherlands
| | - Mathilda T M Mommersteeg
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Jorge N Domínguez
- Department of Experimental Biology, University of Jaén, Jaén 23071, Spain
| | - Caroline Choquet
- Aix Marseille University, CNRS UMR 7288, IBDM, Marseille 13288, France
| | - Vincent Wakker
- Department of Medical Biology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam 1105 AZ, The Netherlands
| | - Corrie de Gier-de Vries
- Department of Medical Biology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam 1105 AZ, The Netherlands
| | - Gerard J J Boink
- Department of Medical Biology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam 1105 AZ, The Netherlands
- Department of Cardiology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam 1105 AZ, The Netherlands
| | - Bastiaan J Boukens
- Department of Medical Biology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam 1105 AZ, The Netherlands
| | - Lucile Miquerol
- Aix Marseille University, CNRS UMR 7288, IBDM, Marseille 13288, France
| | - Arie O Verkerk
- Department of Medical Biology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam 1105 AZ, The Netherlands
| | - Vincent M Christoffels
- Department of Medical Biology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam 1105 AZ, The Netherlands
| |
Collapse
|
24
|
Direct Reprograming to Regenerate Myocardium and Repair Its Pacemaker and Conduction System. MEDICINES 2018; 5:medicines5020048. [PMID: 29867004 PMCID: PMC6023490 DOI: 10.3390/medicines5020048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 05/29/2018] [Accepted: 05/30/2018] [Indexed: 01/14/2023]
Abstract
The regenerative medicine field has been revolutionized by the direct conversion of one cell type to another by ectopic expression of lineage-specific transcription factors. The direct reprogramming of fibroblasts to induced cardiac myocytes (iCMs) by core cardiac transcription factors (Gata4, Mef2c, Tbx5) both in vitro and in vivo has paved the way in cardiac regeneration and repair. Several independent research groups have successfully reported the direct reprogramming of fibroblasts in injured myocardium to cardiac myocytes employing a variety of approaches that rely on transcription factors, small molecules, and micro RNAs (miRNAs). Recently, this technology has been considered for local repair of the pacemaker and the cardiac conduction system. To address this, we will first discuss the direct reprograming advancements in the setting of working myocardium regeneration, and then elaborate on how this technology can be applied to repair the cardiac pacemaker and the conduction system.
Collapse
|
25
|
Duverger JE, Jacquemet V, Vinet A, Comtois P. In silico study of multicellular automaticity of heterogeneous cardiac cell monolayers: Effects of automaticity strength and structural linear anisotropy. PLoS Comput Biol 2018. [PMID: 29529023 PMCID: PMC5877903 DOI: 10.1371/journal.pcbi.1005978] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
The biological pacemaker approach is an alternative to cardiac electronic pacemakers. Its main objective is to create pacemaking activity from added or modified distribution of spontaneous cells in the myocardium. This paper aims to assess how automaticity strength of pacemaker cells (i.e. their ability to maintain robust spontaneous activity with fast rate and to drive neighboring quiescent cells) and structural linear anisotropy, combined with density and spatial distribution of pacemaker cells, may affect the macroscopic behavior of the biological pacemaker. A stochastic algorithm was used to randomly distribute pacemaker cells, with various densities and spatial distributions, in a semi-continuous mathematical model. Simulations of the model showed that stronger automaticity allows onset of spontaneous activity for lower densities and more homogeneous spatial distributions, displayed more central foci, less variability in cycle lengths and synchronization of electrical activation for similar spatial patterns, but more variability in those same variables for dissimilar spatial patterns. Compared to their isotropic counterparts, in silico anisotropic monolayers had less central foci and displayed more variability in cycle lengths and synchronization of electrical activation for both similar and dissimilar spatial patterns. The present study established a link between microscopic structure and macroscopic behavior of the biological pacemaker, and may provide crucial information for optimized biological pacemaker therapies. Implantation of electronic pacemakers is a standard treatment to pathologically slow heart rhythm. Despite improving quality of life, those devices display many shortcomings. Bioengineered tissue pacemakers may be a therapeutic alternative, but associated design methods usually lack control of the way cells with spontaneous activity are scattered throughout the tissue. Our study is the first to use a mathematical model to rigorously define and thoroughly characterize how pacemaker cells scattering at the microscopic level may affect macroscopic behaviors of the bioengineered tissue pacemaker. Automaticity strength (ability of pacemaker cell to drive its non-pacemaker neighbors) and anisotropy (preferential orientation of cell shape) are also implemented and give unparalleled insights on how effects of uncontrollable scattered pacemaker cells may be modulated by available experimental techniques. Our model is a powerful tool to aid in optimized bioengineered pacemaker therapies.
Collapse
Affiliation(s)
- James Elber Duverger
- Research Centre, Montreal Heart Institute, Montreal, Quebec, Canada
- Department of Pharmacology and Physiology / Institute of Biomedical Engineering, Université de Montréal, Montreal, Quebec, Canada
| | - Vincent Jacquemet
- Department of Pharmacology and Physiology / Institute of Biomedical Engineering, Université de Montréal, Montreal, Quebec, Canada
- Research Centre, Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada
| | - Alain Vinet
- Department of Pharmacology and Physiology / Institute of Biomedical Engineering, Université de Montréal, Montreal, Quebec, Canada
- Research Centre, Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada
| | - Philippe Comtois
- Research Centre, Montreal Heart Institute, Montreal, Quebec, Canada
- Department of Pharmacology and Physiology / Institute of Biomedical Engineering, Université de Montréal, Montreal, Quebec, Canada
- * E-mail:
| |
Collapse
|
26
|
Antanavičiūtė I, Šimatonis L, Ulčinas O, Gadeikytė A, Abakevičienė B, Tamulevičius S, Mikalayeva V, Skeberdis VA, Stankevičius E, Tamulevičius T. Femtosecond laser micro-machined polyimide films for cell scaffold applications. J Tissue Eng Regen Med 2017; 12:e760-e773. [PMID: 27943611 DOI: 10.1002/term.2376] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 10/07/2016] [Accepted: 12/06/2016] [Indexed: 01/08/2023]
Abstract
Engineering of sophisticated synthetic 3D scaffolds that allow controlling behaviour and location of the cells requires advanced micro/nano-fabrication techniques. Ultrafast laser micro-machining employing a 1030-nm wavelength Yb:KGW femtosecond laser and a micro-fabrication workstation for micro-machining of commercially available 12.7 and 25.4 μm thickness polyimide (PI) film was applied. Mechanical properties of the fabricated scaffolds, i.e. arrays of differently spaced holes, were examined via custom-built uniaxial micro-tensile testing and finite element method simulations. We demonstrate that experimental micro-tensile testing results could be numerically simulated and explained by two-material model, assuming that 2-6 μm width rings around the holes possessed up to five times higher Young's modulus and yield stress compared with the rest of the laser intacted PI film areas of 'dog-bone'-shaped specimens. That was attributed to material modification around the micro-machined holes in the vicinity of the position of the focused laser beam track during trepanning drilling. We demonstrate that virgin PI films provide a suitable environment for the mobility, proliferation and intercellular communication of human bone marrow mesenchymal stem cells, and discuss how cell behaviour varies on the micro-machined PI films with holes of different diameters (3.1, 8.4 and 16.7 μm) and hole spacing (30, 35, 40 and 45 μm). We conclude that the holes of 3.1 μm diameter were sufficient for metabolic and genetic communication through membranous tunneling tubes between cells residing on the opposite sides of PI film, but prevented the trans-migration of cells through the holes. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Ieva Antanavičiūtė
- Institute of Cardiology of Lithuanian University of Health Sciences, Sukilėlių Ave. 17, LT-50009, Lithuania
| | - Linas Šimatonis
- Institute of Materials Science of Kaunas University of Technology, K. Baršausko, Str. 59, LT-51423, Kaunas, Lithuania
| | - Orestas Ulčinas
- Institute of Materials Science of Kaunas University of Technology, K. Baršausko, Str. 59, LT-51423, Kaunas, Lithuania
| | - Aušra Gadeikytė
- Institute of Materials Science of Kaunas University of Technology, K. Baršausko, Str. 59, LT-51423, Kaunas, Lithuania
| | - Brigita Abakevičienė
- Institute of Materials Science of Kaunas University of Technology, K. Baršausko, Str. 59, LT-51423, Kaunas, Lithuania
| | - Sigitas Tamulevičius
- Institute of Materials Science of Kaunas University of Technology, K. Baršausko, Str. 59, LT-51423, Kaunas, Lithuania
| | - Valeryia Mikalayeva
- Institute of Cardiology of Lithuanian University of Health Sciences, Sukilėlių Ave. 17, LT-50009, Lithuania
| | - Vytenis Arvydas Skeberdis
- Institute of Cardiology of Lithuanian University of Health Sciences, Sukilėlių Ave. 17, LT-50009, Lithuania
| | - Edgaras Stankevičius
- Institute of Physiology and Pharmacology of Lithuanian University of Health Sciences, A. Mickevičiaus Str. 9, LT-44307, Kaunas, Lithuania
| | - Tomas Tamulevičius
- Institute of Materials Science of Kaunas University of Technology, K. Baršausko, Str. 59, LT-51423, Kaunas, Lithuania
| |
Collapse
|
27
|
Driessen HE, van Veen TAB, Boink GJJ. Emerging molecular therapies targeting myocardial infarction-related arrhythmias. Europace 2017; 19:518-528. [PMID: 28431070 DOI: 10.1093/europace/euw198] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 06/06/2016] [Indexed: 12/18/2022] Open
Abstract
Cardiac disease is the leading cause of death in the developed world. Ventricular arrhythmias associated with myocardial ischaemia and/or infarction are a major contributor to cardiovascular mortality, and require improved prevention and treatment. Drugs, devices, and radiofrequency catheter ablation have made important inroads, but have significant limitations ranging from incomplete success to undesired toxicities and major side effects. These limitations derive from the nature of the intervention. Drugs are frequently ineffective, target the entire heart, and often do not deal with the specific arrhythmia trigger or substrate. Devices can terminate rapid rhythms but at best indirectly affect the underlying disease, while ablation, even when appropriately targeted, induces additional tissue damage. In contrast, exploration of gene and cell therapies are expected to provide a targeted, non-destructive, and potentially regenerative approach to ischaemia- and infarction-related arrhythmias. Although these approaches are in the early stages of development, they carry substantial potential to advance arrhythmia prevention and treatment.
Collapse
Affiliation(s)
- Helen E Driessen
- Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Toon A B van Veen
- Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Gerard J J Boink
- Heart Center, Department of Clinical and Experimental Cardiology, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands.,Netherlands Heart Institute, Utrecht, The Netherlands
| |
Collapse
|
28
|
Stefanovic S, Zaffran S. Mechanisms of retinoic acid signaling during cardiogenesis. Mech Dev 2016; 143:9-19. [PMID: 28007475 DOI: 10.1016/j.mod.2016.12.002] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Revised: 11/29/2016] [Accepted: 12/12/2016] [Indexed: 10/20/2022]
Abstract
Substantial experimental and epidemiological data have highlighted the interplay between nutritional and genetic factors in the development of congenital heart defects. Retinoic acid (RA), a derivative of vitamin A, plays a key role during vertebrate development including the formation of the heart. Retinoids bind to RA and retinoid X receptors (RARs and RXRs) which then regulate tissue-specific genes. Here, we will focus on the roles of RA signaling and receptors in gene regulation during cardiogenesis, and the consequence of deregulated retinoid signaling on heart formation and congenital heart defects.
Collapse
|
29
|
Greulich F, Trowe MO, Leffler A, Stoetzer C, Farin HF, Kispert A. Misexpression of Tbx18 in cardiac chambers of fetal mice interferes with chamber-specific developmental programs but does not induce a pacemaker-like gene signature. J Mol Cell Cardiol 2016; 97:140-9. [PMID: 27180262 DOI: 10.1016/j.yjmcc.2016.05.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 04/13/2016] [Accepted: 05/09/2016] [Indexed: 11/20/2022]
Abstract
Initiation of cardiac excitation depends on a specialized group of cardiomyocytes at the venous pole of the heart, the sinoatrial node (SAN). The T-box transcription factor gene Tbx18 is expressed in the SAN myocardium and is required for formation of a large portion of the pacemaker. Previous studies suggested that Tbx18 is also sufficient to reprogram ventricular cardiomyocytes into SAN cells in rat, guinea-pig and pig hearts. To evaluate the consequences of misexpression of Tbx18 for imposing a nodal phenotype onto chamber myocardial cells in fetal mice, we used two independent conditional approaches with chamber-specific cre driver lines and an Hprt(Tbx18) misexpression allele. Myh6-Cre/+;Hprt(Tbx18/y) mice developed dilated atria with thickened walls, reduced right ventricles and septal defects that resulted in reduced embryonic and post-natal survival. Tagln-Cre/+;Hprt(Tbx18/y) mice exhibited slightly smaller hearts with rounded trabeculae that supported normal embryonic survival. Molecular analyses showed that the SAN gap junction and ion channel profile was not ectopically induced in chamber myocardium but the working myocardial gene program was partially inhibited in atria and ventricles of both misexpression models. Left atrial expression of Pitx2 was strongly repressed in Myh6-Cre/+;Hprt(Tbx18/y) embryos. We conclude that exclusion of Tbx18 expression from the developing atria and (right) ventricle is important to achieve normal cardiac left-right patterning and myocardial differentiation, and that Tbx18 is not sufficient to induce full SAN differentiation of chamber cardiomyocytes in fetal mice.
Collapse
Affiliation(s)
- Franziska Greulich
- Institut für Molekularbiologie, OE5250, Medizinische Hochschule Hannover, Carl-Neuberg-Str.1, D-30625 Hannover, Germany
| | - Mark-Oliver Trowe
- Institut für Molekularbiologie, OE5250, Medizinische Hochschule Hannover, Carl-Neuberg-Str.1, D-30625 Hannover, Germany
| | - Andreas Leffler
- Klinik für Anästhesiologie und Intensivmedizin, OE8050, Medizinische Hochschule Hannover, Carl-Neuberg-Str.1, D-30625 Hannover, Germany
| | - Carsten Stoetzer
- Klinik für Anästhesiologie und Intensivmedizin, OE8050, Medizinische Hochschule Hannover, Carl-Neuberg-Str.1, D-30625 Hannover, Germany
| | - Henner F Farin
- Institut für Molekularbiologie, OE5250, Medizinische Hochschule Hannover, Carl-Neuberg-Str.1, D-30625 Hannover, Germany
| | - Andreas Kispert
- Institut für Molekularbiologie, OE5250, Medizinische Hochschule Hannover, Carl-Neuberg-Str.1, D-30625 Hannover, Germany.
| |
Collapse
|
30
|
Buja L, Schoen F. The Pathology of Cardiovascular Interventions and Devices for Coronary Artery Disease, Vascular Disease, Heart Failure, and Arrhythmias. Cardiovasc Pathol 2016. [DOI: 10.1016/b978-0-12-420219-1.00032-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
31
|
Husse B, Franz WM. Generation of cardiac pacemaker cells by programming and differentiation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1863:1948-52. [PMID: 26681531 DOI: 10.1016/j.bbamcr.2015.12.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 11/30/2015] [Accepted: 12/07/2015] [Indexed: 12/22/2022]
Abstract
A number of diseases are caused by faulty function of the cardiac pacemaker and described as "sick sinus syndrome". The medical treatment of sick sinus syndrome with electrical pacemaker implants in the diseased heart includes risks. These problems may be overcome via "biological pacemaker" derived from different adult cardiac cells or pluripotent stem cells. The generation of cardiac pacemaker cells requires the understanding of the pacing automaticity. Two characteristic phenomena the "membrane-clock" and the "Ca(2+)-clock" are responsible for the modulation of the pacemaker activity. Processes in the "membrane-clock" generating the spontaneous pacemaker firing are based on the voltage-sensitive membrane ion channel activity starting with slow diastolic depolarization and discharging in the action potential. The influence of the intracellular Ca(2+) modulating the pacemaker activity is characterized by the "Ca(2+)-clock". The generation of pacemaker cells started with the reprogramming of adult cardiac cells by targeted induction of one pacemaker function like HCN1-4 overexpression and enclosed in an activation of single pacemaker specific transcription factors. Reprogramming of adult cardiac cells with the transcription factor Tbx18 created cardiac cells with characteristic features of cardiac pacemaker cells. Another key transcription factor is Tbx3 specifically expressed in the cardiac conduction system including the sinoatrial node and sufficient for the induction of the cardiac pacemaker gene program. For a successful cell therapeutic practice, the generated cells should have all regulating mechanisms of cardiac pacemaker cells. Otherwise, the generated pacemaker cells serve only as investigating model for the fundamental research or as drug testing model for new antiarrhythmics. This article is part of a Special Issue entitled: Cardiomyocyte Biology: Integration of Developmental and Environmental Cues in the Heart edited by Marcus Schaub and Hughes Abriel.
Collapse
Affiliation(s)
- Britta Husse
- Medical University Innsbruck, Department of Internal Medicine III, Cardiology and Angiology, Anichstr. 35, A-6020 Innsbruck, Austria.
| | - Wolfgang-Michael Franz
- Medical University Innsbruck, Department of Internal Medicine III, Cardiology and Angiology, Anichstr. 35, A-6020 Innsbruck, Austria.
| |
Collapse
|
32
|
Vedantham V. New Approaches to Biological Pacemakers: Links to Sinoatrial Node Development. Trends Mol Med 2015; 21:749-761. [PMID: 26611337 DOI: 10.1016/j.molmed.2015.10.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 10/08/2015] [Accepted: 10/13/2015] [Indexed: 12/30/2022]
Abstract
Irreversible degeneration of the cardiac conduction system is a common disease that can cause activity intolerance, fainting, and death. While electronic pacemakers provide effective treatment, alternative approaches are needed when long-term indwelling hardware is undesirable. Biological pacemakers comprise electrically active cells that functionally integrate with the heart. Recent findings on cardiac pacemaker cells (PCs) within the sinoatrial node (SAN), along with developments in stem cell technology, have opened a new era in biological pacing. Recent experiments that have derived PC-like cells from non-PCs have brought the field closer than ever before to biological pacemakers that can faithfully recapitulate SAN activity. In this review, I discuss these approaches in the context of SAN biology and address the potential for clinical translation.
Collapse
Affiliation(s)
- Vasanth Vedantham
- Department of Medicine, Cardiology Division, University of California, San Francisco, CA, USA; Gladstone Institute of Cardiovascular Disease, San Francisco, CA, USA.
| |
Collapse
|
33
|
Cingolani E, Marbán E. Recreación del nódulo sinusal mediante reprogramación somática: ¿un sueño hecho realidad? Rev Esp Cardiol 2015. [DOI: 10.1016/j.recesp.2015.04.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
34
|
Cingolani E, Marbán E. Recreating the Sinus Node by Somatic Reprogramming: A Dream Come True? ACTA ACUST UNITED AC 2015; 68:743-5. [PMID: 26183662 DOI: 10.1016/j.rec.2015.04.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 04/29/2015] [Indexed: 11/25/2022]
Affiliation(s)
- Eugenio Cingolani
- Cedars-Sinai Heart Institute, Los Angeles, California, United States
| | - Eduardo Marbán
- Cedars-Sinai Heart Institute, Los Angeles, California, United States.
| |
Collapse
|
35
|
Affiliation(s)
- Eugenio Cingolani
- Familial Arrhythmia Clinic, Cedars-Sinai Heart Institute, Los Angeles, CA.
| |
Collapse
|