1
|
Kotani K, Ngako Kadji FM, Mandai Y, Hiraoka Y. Backflow reduction in local injection therapy with gelatin formulations. Drug Deliv 2024; 31:2329100. [PMID: 38515401 PMCID: PMC10962293 DOI: 10.1080/10717544.2024.2329100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 02/29/2024] [Indexed: 03/23/2024] Open
Abstract
The local injection of therapeutic drugs, including cells, oncolytic viruses and nucleic acids, into different organs is an administrative route used to achieve high drug exposure at the site of action. However, after local injection, material backflow and side effect reactions can occur. Hence, this study was carried out to investigate the effect of gelatin on backflow reduction in local injection. Gelatin particles (GPs) and hydrolyzed gelatin (HG) were injected into tissue models, including versatile training tissue (VTT), versatile training tissue tumor-in type (VTT-T), and broiler chicken muscles (BCM), using needle gauges between 23 G and 33 G. The backflow material fluid was collected with filter paper, and the backflow fluid rate was determined. The backflow rate was significantly reduced with 35 μm GPs (p value < .0001) at different concentrations up to 5% and with 75 μm GPs (p value < .01) up to 2% in the tissue models. The reduction in backflow with HG of different molecular weights showed that lower-molecular-weight HG required a higher-concentration dose (5% to 30%) and that higher-molecular-weight HG required a lower-concentration dose (7% to 8%). The backflow rate was significantly reduced with the gelatin-based formulation, in regard to the injection volumes, which varied from 10 μL to 100 μL with VTT or VTT-T and from 10 μL to 200 μL with BCM. The 35 μm GPs were injectable with needles of small gauges, which included 33 G, and the 75 μm GPs and HG were injectable with 27 G needles. The backflow rate was dependent on an optimal viscosity of the gelatin solutions. An optimal concentration of GPs or HG can prevent material backflow in local injection, and further studies with active drugs are necessary to investigate the applicability in tumor and organ injections.
Collapse
Affiliation(s)
- Kazuki Kotani
- Department of Biomedical, R&D C-enter, Nitta Gelatin, Inc, Yao City, Osaka, Japan
| | | | - Yoshinobu Mandai
- Department of Biomedical, R&D C-enter, Nitta Gelatin, Inc, Yao City, Osaka, Japan
| | - Yosuke Hiraoka
- Department of Biomedical, R&D C-enter, Nitta Gelatin, Inc, Yao City, Osaka, Japan
| |
Collapse
|
2
|
Brebu M, Pamfil D, Stoica I, Aflori M, Voicu G, Stoleru E. Photo-crosslinked chitosan-gelatin xerogel-like coating onto "cold" plasma functionalized poly(lactic acid) film as cell culture support. Carbohydr Polym 2024; 339:122288. [PMID: 38823936 DOI: 10.1016/j.carbpol.2024.122288] [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: 03/16/2024] [Revised: 05/13/2024] [Accepted: 05/15/2024] [Indexed: 06/03/2024]
Abstract
This paper reports on biofunctionalisation of a poly(lactic acid) (PLA) film by surface activation through cold plasma treatment followed by coating with a chitosan-gelatin xerogel. The UV cross-linking of the xerogel precursor was simultaneously performed with the fixation onto the PLA support. This has a strong effect on surface properties, in terms of wettability, surface free energy, morphology and micromechanical features. The hydrophilic - hydrophobic character of the surface, determined by contact angle measurements, was tuned along the process, passing from moderate hydrophobic PLA to enhanced hydrophilic plasma activated surface, which favors coating adhesion, then to moderate hydrophobic chitosan-gelatin coating. The coating has a Lewis amphoteric surface, with a porous xerogel-like morphology, as revealed by scanning electron microscopy images. By riboflavin mediated UV cross-linking the chitosan-gelatin coating becomes high adhesive and with a more pronounced plasticity, as shown by AFM force-distance spectroscopy. Thus prepared surface-coated PLA supports were successfully tested for growth of dermal fibroblasts, which are known for their induction potential of chondrogenic cells, which is very important in cartilage tissue engineering.
Collapse
Affiliation(s)
- Mihai Brebu
- "Petru Poni" Institute of Macromolecular Chemistry, Grigore Ghica Voda Alley, 41A, 700487, Iasi, Romania
| | - Daniela Pamfil
- "Petru Poni" Institute of Macromolecular Chemistry, Grigore Ghica Voda Alley, 41A, 700487, Iasi, Romania
| | - Iuliana Stoica
- "Petru Poni" Institute of Macromolecular Chemistry, Grigore Ghica Voda Alley, 41A, 700487, Iasi, Romania
| | - Magdalena Aflori
- "Petru Poni" Institute of Macromolecular Chemistry, Grigore Ghica Voda Alley, 41A, 700487, Iasi, Romania
| | - Geanina Voicu
- "Medical and Pharmaceutical BioNanoTechnologies" Laboratory (BioNanoMed) Institute of Cellular Biology and Pathology, "Nicolae Simionescu" 8, BP Hasdeu Street, 050568 Bucharest, Romania
| | - Elena Stoleru
- "Petru Poni" Institute of Macromolecular Chemistry, Grigore Ghica Voda Alley, 41A, 700487, Iasi, Romania.
| |
Collapse
|
3
|
Heima D, Takeda M, Tabata Y, Minatoya K, Yamashita JK, Masumoto H. Therapeutic potential of human induced pluripotent stem cell-derived cardiac tissue in an ischemic model with unloaded condition mimicking left ventricular assist device. J Thorac Cardiovasc Surg 2024; 168:e72-e88. [PMID: 37981100 DOI: 10.1016/j.jtcvs.2023.11.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 10/28/2023] [Accepted: 11/07/2023] [Indexed: 11/21/2023]
Abstract
OBJECTIVE This study aimed to explore the therapeutic potential of human induced pluripotent stem cell (hiPSC)-derived cardiac tissues (HiCTs) in the emerging approach of bridge to recovery for severe heart failure with ventricular assist devices. We used a rat model of heterotopic heart transplantation (HTx) to mimic ventricular assist device support and heart unloading. METHODS HiCTs were created by inserting gelatin hydrogel microspheres between cell sheets made from hiPSC-derived cardiovascular cells. Male athymic nude rats underwent myocardial infarction (MI) and were divided into the following groups: MI (loaded, untreated control), MI + HTx (unloaded, untreated control), MI + HTx + HiCT (unloaded, treated), and MI + HiCT (loaded, treated). HiCTs were placed on the epicardium of the heart in treated groups. We evaluated HiCT engraftment, fibrosis, and neovascularization using histologic analysis. RESULTS After 4 weeks, HiCTs successfully engrafted in 5 of 6 rats in the MI + HTx + HiCT group (83.3%). The engrafted HiCT area was greater under unloaded conditions (MI + HTx + HiCT) than loaded conditions (MI + HiCT) (P < .05). MI + HTx + HiCT had a significantly smaller infarct area compared with MI and MI + HTx. The MI + HTx + MiCT group exhibited greater vascular density in the border zone than MI and MI + HTx. HiCT treatment suppressed cardiomyocyte atrophy due to left ventricular unloading (P = .001). The protein level of muscle-specific RING finger 1, an atrophy-related ubiquitin ligase, was lower in the MI + HTx + HiCT group than in MI + HTx (P = .036). CONCLUSIONS Transplanting HiCTs into ischemic hearts under unloaded conditions promoted engraftment, neovascularization, attenuated infarct remodeling, and suppressed myocyte atrophy. These results suggest that HiCT treatment could contribute to future advancements in bridge to recovery.
Collapse
Affiliation(s)
- Daisuke Heima
- Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Masafumi Takeda
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Yasuhiko Tabata
- Department of Biomaterials, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Kenji Minatoya
- Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Jun K Yamashita
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan.
| | - Hidetoshi Masumoto
- Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Clinical Translational Research Program, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan.
| |
Collapse
|
4
|
Donmazov S, Saruhan EN, Pekkan K, Piskin S. Review of Machine Learning Techniques in Soft Tissue Biomechanics and Biomaterials. Cardiovasc Eng Technol 2024:10.1007/s13239-024-00737-y. [PMID: 38956008 DOI: 10.1007/s13239-024-00737-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 05/28/2024] [Indexed: 07/04/2024]
Abstract
BACKGROUND AND OBJECTIVE Advanced material models and material characterization of soft biological tissues play an essential role in pre-surgical planning for vascular surgeries and transcatheter interventions. Recent advances in heart valve engineering, medical device and patch design are built upon these models. Furthermore, understanding vascular growth and remodeling in native and tissue-engineered vascular biomaterials, as well as designing and testing drugs on soft tissue, are crucial aspects of predictive regenerative medicine. Traditional nonlinear optimization methods and finite element (FE) simulations have served as biomaterial characterization tools combined with soft tissue mechanics and tensile testing for decades. However, results obtained through nonlinear optimization methods are reliable only to a certain extent due to mathematical limitations, and FE simulations may require substantial computing time and resources, which might not be justified for patient-specific simulations. To a significant extent, machine learning (ML) techniques have gained increasing prominence in the field of soft tissue mechanics in recent years, offering notable advantages over conventional methods. This review article presents an in-depth examination of emerging ML algorithms utilized for estimating the mechanical characteristics of soft biological tissues and biomaterials. These algorithms are employed to analyze crucial properties such as stress-strain curves and pressure-volume loops. The focus of the review is on applications in cardiovascular engineering, and the fundamental mathematical basis of each approach is also discussed. METHODS The review effort employed two strategies. First, the recent studies of major research groups actively engaged in cardiovascular soft tissue mechanics are compiled, and research papers utilizing ML and deep learning (DL) techniques were included in our review. The second strategy involved a standard keyword search across major databases. This approach provided 11 relevant ML articles, meticulously selected from reputable sources including ScienceDirect, Springer, PubMed, and Google Scholar. The selection process involved using specific keywords such as "machine learning" or "deep learning" in conjunction with "soft biological tissues", "cardiovascular", "patient-specific," "strain energy", "vascular" or "biomaterials". Initially, a total of 25 articles were selected. However, 14 of these articles were excluded as they did not align with the criteria of focusing on biomaterials specifically employed for soft tissue repair and regeneration. As a result, the remaining 11 articles were categorized based on the ML techniques employed and the training data utilized. RESULTS ML techniques utilized for assessing the mechanical characteristics of soft biological tissues and biomaterials are broadly classified into two categories: standard ML algorithms and physics-informed ML algorithms. The standard ML models are then organized based on their tasks, being grouped into Regression and Classification subcategories. Within these categories, studies employ various supervised learning models, including support vector machines (SVMs), bagged decision trees (BDTs), artificial neural networks (ANNs) or deep neural networks (DNNs), and convolutional neural networks (CNNs). Additionally, the utilization of unsupervised learning approaches, such as autoencoders incorporating principal component analysis (PCA) and/or low-rank approximation (LRA), is based on the specific characteristics of the training data. The training data predominantly consists of three types: experimental mechanical data, including uniaxial or biaxial stress-strain data; synthetic mechanical data generated through non-linear fitting and/or FE simulations; and image data such as 3D second harmonic generation (SHG) images or computed tomography (CT) images. The evaluation of performance for physics-informed ML models primarily relies on the coefficient of determinationR 2 . In contrast, various metrics and error measures are utilized to assess the performance of standard ML models. Furthermore, our review includes an extensive examination of prevalent biomaterial models that can serve as physical laws for physics-informed ML models. CONCLUSION ML models offer an accurate, fast, and reliable approach for evaluating the mechanical characteristics of diseased soft tissue segments and selecting optimal biomaterials for time-critical soft tissue surgeries. Among the various ML models examined in this review, physics-informed neural network models exhibit the capability to forecast the mechanical response of soft biological tissues accurately, even with limited training samples. These models achieve highR 2 values ranging from 0.90 to 1.00. This is particularly significant considering the challenges associated with obtaining a large number of living tissue samples for experimental purposes, which can be time-consuming and impractical. Additionally, the review not only discusses the advantages identified in the current literature but also sheds light on the limitations and offers insights into future perspectives.
Collapse
Affiliation(s)
- Samir Donmazov
- Department of Mathematics, University of Kentucky, Lexington, KY, 40506, USA
| | - Eda Nur Saruhan
- Department of Computer Science and Engineering, Koc University, Sariyer, Istanbul, Turkey
| | - Kerem Pekkan
- Department of Mechanical Engineering, Koc University, Sariyer, Istanbul, Turkey
| | - Senol Piskin
- Department of Mechanical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Vadi Kampusu, Sariyer, 34396, Istanbul, Turkey.
- Modeling, Simulation and Extended Reality Laboratory, Faculty of Engineering and Natural Sciences, Istinye University, Vadi Kampusu, Sariyer, 34396, Istanbul, Turkey.
| |
Collapse
|
5
|
Cai S, Dai Q. Progress in preclinical research on induced pluripotent stem cell therapy for acute myocardial infarction. Zhejiang Da Xue Xue Bao Yi Xue Ban 2024; 53:244-253. [PMID: 38594961 PMCID: PMC11057988 DOI: 10.3724/zdxbyxb-2023-0402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 03/09/2024] [Indexed: 04/11/2024]
Abstract
Induced pluripotent stem cells (iPSCs) are obtained by introducing exogenous genes or adding chemicals to the culture medium to induce somatic cell differentiation. Similarly to embryonic stem cells, iPSCs have the ability to differentiate into all three embryonic cell lines. iPSCs can differentiate into cardiac muscle cells through two-dimensional differentiation methods such as monolayer cell culture and co-culture, or through embryoid body and scaffold-based three-dimensional differentiation methods. In addition, the process of iPSCs differentiation into cardiac muscle cells also requires activation or inhibition of specific signaling pathways,such as Wnt, BMP, Notch signaling pathways to mimic the development of the heart in vivo. In recent years, suspension culturing in bioreactors has been shown to produce large number of iPSCs derived cardiac muscle cells (iPSC-CMs). Before transplantation, it is necessary to purify iPSC-CMs through metabolic regulation or cell sorting to eliminate undifferentiated iPSCs, which may lead to teratoma formation. The transplantation methods for iPSC-CMs are mainly injection of cell suspension and transplantation of cell patches into the infarcted myocardium. Animal studies have shown that transplantation of iPSC-CMs into the infarcted myocardium can improve cardiac function. This article reviews the progress in preclinical studies on iPSC-CMs therapy for acute myocardial infarction and discusses the limitations and challenges of its clinical application to provide references for further clinical research and application.
Collapse
Affiliation(s)
- Songyan Cai
- Department of Cardiology, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China.
| | - Qingyuan Dai
- Department of Cardiology, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China.
| |
Collapse
|
6
|
Kitahata S, Mandai M, Ichikawa H, Tanaka Y, Senba T, Kajita K, Sugita S, Kadonosono K, Takahashi M. Investigation of the effectiveness of gelatin hydrolysate in human iPS-RPE cell suspension transplantation. Regen Ther 2024; 25:238-249. [PMID: 38293586 PMCID: PMC10825282 DOI: 10.1016/j.reth.2023.12.016] [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: 10/13/2023] [Revised: 12/21/2023] [Accepted: 12/29/2023] [Indexed: 02/01/2024] Open
Abstract
Introduction The retinal pigment epithelium (RPE) plays essential roles in maintaining retinal functions as well as choroidal capillaries and can lead to visual disorders if dysfunctional. Transplantation of human-induced pluripotent stem cell-derived RPE (hiPSC-RPE) is a promising therapy for such RPE impaired conditions including age-related macular degeneration. The challenge with cell suspension transplantation is targeted delivery of graft cells and undesired cell reflux. Gelatin hydrolysate, a soluble variant with specific molecular weight distribution, is examined in this study for its potential use in hiPSC-RPE suspension transplantation, particularly in reducing cell reflux and enhancing RPE engraftment. Methods A retinal bleb model was created using polydimethylsiloxane (PDMS) soft lithography to quantify cellular reflux. We examined the effects of gelatin hydrolysate on the hiPSC-RPE of various aspects of cell behavior and performance such as cell viability, hypoxia reaction, morphology, induction of inflammation and immune responses. Results Gelatin hydrolysate at 5 % concentration effectively mitigated cell reflux in vitro mimic, improved cell viability, reduced cell aggregation, and had an inhibitory effect on hypoxic reactions due to cell deposition with hiPSC-RPE. Additionally, gelatin hydrolysate did not affect cell adhesion and morphology, and decreased the expression of major histocompatibility complex class II molecules, which suggests reduced immunogenicity of hiPSC-RPE. Conclusion Gelatin hydrolysate is considered a valuable and useful candidate for future regenerative therapies in hiPSC-RPE suspension transplantation.
Collapse
Affiliation(s)
- Shohei Kitahata
- Department of Ophthalmology and Micro-technology, Yokohama City University, 4-57 Urafunecho, Minami-ku, Yokohama, Japan
- Kobe City Eye Hospital, 2-1-8 Minatojima-minamimachi, Chuo-ku, Kobe, Japan
| | - Michiko Mandai
- Kobe City Eye Hospital, 2-1-8 Minatojima-minamimachi, Chuo-ku, Kobe, Japan
| | - Hinako Ichikawa
- Department of Ophthalmology and Micro-technology, Yokohama City University, 4-57 Urafunecho, Minami-ku, Yokohama, Japan
| | - Yuji Tanaka
- Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, Japan
| | - Toshika Senba
- Vision Care Inc., Kobe Eye Center 5F, 2-1-8 Minatojima-minamimachi, Chuo-ku, Kobe, Japan
| | - Keisuke Kajita
- Vision Care Inc., Kobe Eye Center 5F, 2-1-8 Minatojima-minamimachi, Chuo-ku, Kobe, Japan
- Department of Ophthalmology, Tokushima University, 2-24 Shinkura-cho, Tokushima, Japan
| | - Sunao Sugita
- Kobe City Eye Hospital, 2-1-8 Minatojima-minamimachi, Chuo-ku, Kobe, Japan
- Vision Care Inc., Kobe Eye Center 5F, 2-1-8 Minatojima-minamimachi, Chuo-ku, Kobe, Japan
| | - Kazuaki Kadonosono
- Department of Ophthalmology and Micro-technology, Yokohama City University, 4-57 Urafunecho, Minami-ku, Yokohama, Japan
| | - Masayo Takahashi
- Kobe City Eye Hospital, 2-1-8 Minatojima-minamimachi, Chuo-ku, Kobe, Japan
- Vision Care Inc., Kobe Eye Center 5F, 2-1-8 Minatojima-minamimachi, Chuo-ku, Kobe, Japan
| |
Collapse
|
7
|
Hashida A, Nakazato T, Uemura T, Liu L, Miyagawa S, Sawa Y, Kino-oka M. Effect of morphological change on the maturation of human induced pluripotent stem cell-derived cardiac tissue in rotating flow culture. Regen Ther 2023; 24:479-488. [PMID: 37767182 PMCID: PMC10520276 DOI: 10.1016/j.reth.2023.09.002] [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: 06/17/2023] [Revised: 08/06/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Introduction Understanding the critical factors for the maturation of human induced pluripotent stem cell (hiPSC)-derived cardiac tissue is important for further development of culture techniques. Rotating flow culture, where the tissues float in the culture medium by balancing its gravitational settling and the medium flow generated in rotating disk-shaped culture vessels, is one of culture systems used for tissue engineering. It has previously been demonstrated that rotating flow culture leads to the formation of matured cardiac tissue with higher levels of function and structure than the other culture systems. However, the detailed mechanisms underlying the maturation of cardiac tissue remain unclear. This study investigated the maturation process of hiPSC-derived cardiac tissue in rotating flow culture with a focus on morphological changes in the tissue, which is a trigger for maturation. Methods The cardiac tissue, which consisted of cardiomyocytes derived from hiPSCs, was cultured on the 3D scaffold of poly (lactic-co-glycolic) acid (PLGA)-aligned nanofibers, in rotating flow culture for 5 days. During the culture, the time profile of projected area of tissue and formation of maturation marker proteins (β-myosin heavy chain and Connexin-43), tissue structure, and formation of nuclear lamina proteins (Lamin A/C) were compared with that in static suspension culture. Results The ratio of the projected area of tissue significantly decreased from Day 0 to Day 3 due to tissue shrinkage. In contrast, Western blot analysis revealed that maturation protein markers of cardiomyocytes significantly increased after Day 3. In addition, in rotating flow culture, flat-shaped nuclei and fiber-like cytoskeletal structures were distributed in the surface region of tissue where medium flow was continuously applied. Moreover, Lamin A/C, which are generally formed in differentiated cells owing to mechanical force across the cytoskeleton and critically affect the maturation of cardiomyocytes, were significantly formed in the tissue of rotating flow culture. Conclusions In this study, we found that spatial heterogeneity of tissue structure and tissue shrinkage occurred in rotating flow culture, which was not observed in static suspension culture. Moreover, from the quantitative analysis, it was also suggested that tissue shrinkage in rotating flow culture contributed its following tissue maturation. These findings showed one of the important characteristics of rotating flow culture which was not revealed in previous studies.
Collapse
Affiliation(s)
- Akihiro Hashida
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Taro Nakazato
- Department of Surgery, Division of Cardiovascular Surgery, Graduate School of Medicine, Osaka University, 2-15, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Toshimasa Uemura
- Department of Precise and Science Technology, Graduate School of Engineering, Osaka University, 2-1, Yamadaoka, Suita, Osaka, 565-0871, Japan
- Cell Culture Marketing & Research Center, JTEC Corporation, 2-1, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Li Liu
- Department of Surgery, Division of Cardiovascular Surgery, Graduate School of Medicine, Osaka University, 2-15, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Shigeru Miyagawa
- Department of Surgery, Division of Cardiovascular Surgery, Graduate School of Medicine, Osaka University, 2-15, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yoshiki Sawa
- Division of Health and Sciences, Graduate School of Medicine, Osaka University, 2-15, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Masahiro Kino-oka
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1, Yamadaoka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
8
|
Bernava G, Iop L. Advances in the design, generation, and application of tissue-engineered myocardial equivalents. Front Bioeng Biotechnol 2023; 11:1247572. [PMID: 37811368 PMCID: PMC10559975 DOI: 10.3389/fbioe.2023.1247572] [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: 06/26/2023] [Accepted: 08/29/2023] [Indexed: 10/10/2023] Open
Abstract
Due to the limited regenerative ability of cardiomyocytes, the disabling irreversible condition of myocardial failure can only be treated with conservative and temporary therapeutic approaches, not able to repair the damage directly, or with organ transplantation. Among the regenerative strategies, intramyocardial cell injection or intravascular cell infusion should attenuate damage to the myocardium and reduce the risk of heart failure. However, these cell delivery-based therapies suffer from significant drawbacks and have a low success rate. Indeed, cardiac tissue engineering efforts are directed to repair, replace, and regenerate native myocardial tissue function. In a regenerative strategy, biomaterials and biomimetic stimuli play a key role in promoting cell adhesion, proliferation, differentiation, and neo-tissue formation. Thus, appropriate biochemical and biophysical cues should be combined with scaffolds emulating extracellular matrix in order to support cell growth and prompt favorable cardiac microenvironment and tissue regeneration. In this review, we provide an overview of recent developments that occurred in the biomimetic design and fabrication of cardiac scaffolds and patches. Furthermore, we sift in vitro and in situ strategies in several preclinical and clinical applications. Finally, we evaluate the possible use of bioengineered cardiac tissue equivalents as in vitro models for disease studies and drug tests.
Collapse
Affiliation(s)
| | - Laura Iop
- Department of Cardiac Thoracic Vascular Sciences and Public Health, Padua Medical School, University of Padua, Padua, Italy
| |
Collapse
|
9
|
Bui TQ, Binh NT, Pham TLB, Le Van T, Truong NH, Nguyen DPH, Luu TTT, Nguyen-Xuan Pham T, Cam Tran T, Nguyen HTT, Thuy-Trinh N, Tran PA. The Efficacy of Transplanting Human Umbilical Cord Mesenchymal Stem Cell Sheets in the Treatment of Myocardial Infarction in Mice. Biomedicines 2023; 11:2187. [PMID: 37626684 PMCID: PMC10452263 DOI: 10.3390/biomedicines11082187] [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: 06/15/2023] [Revised: 07/17/2023] [Accepted: 07/21/2023] [Indexed: 08/27/2023] Open
Abstract
The transplantation of mesenchymal stem cell (MSC) sheets derived from human umbilical cords (hUCs) was investigated in this study as a potential application in treating myocardial infarction (MI). Two groups of hUC-MSC sheets were formed by populating LunaGelTM, which are 3D scaffolds of photo-crosslinkable gelatin-based hydrogel with two different cell densities. An MI model was created by ligating the left anterior descending coronary artery of healthy BALB/c mice. After two weeks, the cell sheets were applied directly to the MI area and the efficacy of the treatment was evaluated over the next two weeks by monitoring the mice's weight, evaluating the left ventricle ejection fraction, and assessing the histology of the heart tissue at the end of the experiment. Higher cell density showed significantly greater efficiency in MI mice treatment in terms of weight gain and the recovery of ejection fraction. The heart tissue of the groups receiving cell sheets showed human-CD44-positive staining and reduced fibrosis and apoptosis. In conclusion, the hUC-MSC sheets ameliorated heart MI injury in mice and the efficacy of the cell sheets improved as the number of cells increased.
Collapse
Affiliation(s)
| | - Nguyen Trong Binh
- Biotechnology Center of Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam; (T.L.-B.P.); (D.P.-H.N.); (T.N.-X.P.)
| | - Truc Le-Buu Pham
- Biotechnology Center of Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam; (T.L.-B.P.); (D.P.-H.N.); (T.N.-X.P.)
- Faculty of Biotechnology, Ho Chi Minh City Open University, Ho Chi Minh City 700000, Vietnam
| | - Trinh Le Van
- Laboratory of Stem Cell Research and Application, University of Science, Ho Chi Minh City 700000, Vietnam; (T.L.V.); (N.H.T.)
- Vietnam National University, Ho Chi Minh City 700000, Vietnam;
| | - Nhung Hai Truong
- Laboratory of Stem Cell Research and Application, University of Science, Ho Chi Minh City 700000, Vietnam; (T.L.V.); (N.H.T.)
- Vietnam National University, Ho Chi Minh City 700000, Vietnam;
| | - Dang Phu-Hai Nguyen
- Biotechnology Center of Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam; (T.L.-B.P.); (D.P.-H.N.); (T.N.-X.P.)
| | - Thao Thi-Thu Luu
- Histology-Embryology-Pathology Department, Faculty of Medicine, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam;
| | - Trang Nguyen-Xuan Pham
- Biotechnology Center of Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam; (T.L.-B.P.); (D.P.-H.N.); (T.N.-X.P.)
| | - Tu Cam Tran
- Institute of Tropical Biology, Ho Chi Minh City 700000, Vietnam;
| | - Huyen Thuong-Thi Nguyen
- Divison of Human and Animal Physiology, HCMC University of Education, Ho Chi Minh City 700000, Vietnam;
| | - Nhu Thuy-Trinh
- Vietnam National University, Ho Chi Minh City 700000, Vietnam;
- School of Biomedical Engineering, International University, Ho Chi Minh City 700000, Vietnam
| | - Phong Anh Tran
- Interface Science and Materials Engineering Group, School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology, Brisbane City, QLD 4000, Australia;
| |
Collapse
|
10
|
Tabata Y, Joanna I, Higuchi A. Stem cell culture and differentiation in 3-D scaffolds. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 199:109-127. [PMID: 37678968 DOI: 10.1016/bs.pmbts.2023.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/09/2023]
Abstract
Conventional two-dimensional (2-D) cultivation are easy to utilize for human pluripotent stem (hPS) cell cultivation in standard techniques and are important for analysis or development of the signal pathways to keep pluripotent state of hPS cells cultivated on 2-D cell culture materials. However, the most efficient protocol to prepare hPS cells is the cell culture in a three dimensional (3-D) cultivation unit because huge numbers of hPS cells should be utilized in clinical treatment. Some 3-D cultivation strategies for hPS cells are considered: (a) microencapsulated cell cultivation in suspended hydrogels, (b) cell cultivation on microcarriers (MCs), (c) cell cultivation on self-aggregated spheroid [cell aggregates; embryoid bodies (EBs) and organoids], (d) cell cultivation on microfibers or nanofibers, and (e) cell cultivation in macroporous scaffolds. These cultivation ways are described in this chapter.
Collapse
Affiliation(s)
- Yasuhiko Tabata
- Department of Regeneration Science and Engineering, Institute for Life and Medical Sciences, Kyoto University, Kawara-cho, Shogoin, Sakyo-ku, Kyoto, Japan.
| | - Idaszek Joanna
- Division of Materials Design, Faculty of Materials Science and Engineering, Warsaw University of Technology, Woloska Street, Warsaw, Poland
| | - Akon Higuchi
- Department of Chemical and Materials Engineering, National Central University, Jhongli, Taoyuan, Taiwan; State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China.
| |
Collapse
|
11
|
Chepeleva EV, Pavlova SV, Bgatova NP, Volkov AM, Kazanskaya GM, Sergeevichev DS. Functional Activity of Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes on a Mouse Renal Subcapsular Xenograft Model. Int J Mol Sci 2023; 24:9792. [PMID: 37372940 DOI: 10.3390/ijms24129792] [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: 04/28/2023] [Revised: 06/02/2023] [Accepted: 06/04/2023] [Indexed: 06/29/2023] Open
Abstract
In the treatment of coronary heart disease, the most promising approach for replacing lost contractile elements involves obtaining cardiomyocytes through cardiac differentiation of pluripotent cells. The objective of this study is to develop a technology for creating a functional layer of cardiomyocytes derived from iPSCs, capable of generating rhythmic activity and synchronous contractions. To expedite the maturation of cardiomyocytes, a renal subcapsular transplantation model was employed in SCID mice. Following explantation, the formation of the cardiomyocyte contractile apparatus was assessed using fluorescence and electron microscopy, while the cytoplasmic oscillation of calcium ions was evaluated through visualization using the fluorescent calcium binding dye Fluo-8. The results demonstrate that transplanted human iPSC-derived cardiomyocyte cell layers, placed under the fibrous capsules of SCID mouse kidneys (for up to 6 weeks), initiate the development of an organized contractile apparatus and retain functional activity along with the ability to generate calcium ion oscillations even after removal from the body.
Collapse
Affiliation(s)
- Elena V Chepeleva
- Federal State Budgetary Institution National Medical Research Center Named after Academician E.N. Meshalkin of the Ministry of Health of the Russian Federation, 15, Rechkunovskaya Str., 630055 Novosibirsk, Russia
- Research Institute of Clinical and Experimental Lymphology-Branch of the Institute of Cytology and Genetics Siberian Branch of Russian Academy of Sciences, 2, Timakova Str., 630060 Novosibirsk, Russia
| | - Sophia V Pavlova
- Federal State Budgetary Institution National Medical Research Center Named after Academician E.N. Meshalkin of the Ministry of Health of the Russian Federation, 15, Rechkunovskaya Str., 630055 Novosibirsk, Russia
- Federal Research Center Institute of Cytology and Genetics Siberian Branch of Russian Academy of Sciences, 10, Ac. Lavrentiev Ave., 630090 Novosibirsk, Russia
| | - Nataliya P Bgatova
- Research Institute of Clinical and Experimental Lymphology-Branch of the Institute of Cytology and Genetics Siberian Branch of Russian Academy of Sciences, 2, Timakova Str., 630060 Novosibirsk, Russia
| | - Alexander M Volkov
- Federal State Budgetary Institution National Medical Research Center Named after Academician E.N. Meshalkin of the Ministry of Health of the Russian Federation, 15, Rechkunovskaya Str., 630055 Novosibirsk, Russia
| | - Galina M Kazanskaya
- Institute of Molecular Biology and Biophysics-Subdivision of FRC FTM, 2/12, Timakova Str., 630060 Novosibirsk, Russia
| | - David S Sergeevichev
- Federal State Budgetary Institution National Medical Research Center Named after Academician E.N. Meshalkin of the Ministry of Health of the Russian Federation, 15, Rechkunovskaya Str., 630055 Novosibirsk, Russia
| |
Collapse
|
12
|
Abdolahzadeh H, Rad NK, Shpichka A, Golroo R, Rahi K, Timashev P, Hassan M, Vosough M. Progress and promise of cell sheet assisted cardiac tissue engineering in regenerative medicine. Biomed Mater 2023; 18. [PMID: 36758240 DOI: 10.1088/1748-605x/acbad4] [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: 09/26/2022] [Accepted: 02/09/2023] [Indexed: 02/11/2023]
Abstract
Cardiovascular diseases (CVDs) are the most common leading causes of premature deaths in all countries. To control the harmful side effects of CVDs on public health, it is necessary to understand the current and prospective strategies in prevention, management, and monitoring CVDs.In vitro,recapitulating of cardiac complex structure with its various cell types is a challenging topic in tissue engineering. Cardiac tissue engineering (CTE) is a multi-disciplinary strategy that has been considered as a novel alternative approach for cardiac regenerative medicine and replacement therapies. In this review, we overview various cell types and approaches in cardiac regenerative medicine. Then, the applications of cell-sheet-assisted CTE in cardiac diseases were discussed. Finally, we described how this technology can improve cardiac regeneration and function in preclinical and clinical models.
Collapse
Affiliation(s)
- Hadis Abdolahzadeh
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Niloofar Khoshdel Rad
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Anastasia Shpichka
- World-Class Research Center 'Digital Biodesign and Personalized Healthcare', Sechenov University, Moscow, Russia.,Institute for Regenerative Medicine, Sechenov University, Moscow, Russia.,Chemistry Department, Lomonosov Moscow State University, Moscow, Russia
| | - Reihaneh Golroo
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Kosar Rahi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Peter Timashev
- World-Class Research Center 'Digital Biodesign and Personalized Healthcare', Sechenov University, Moscow, Russia.,Institute for Regenerative Medicine, Sechenov University, Moscow, Russia.,Chemistry Department, Lomonosov Moscow State University, Moscow, Russia
| | - Moustapha Hassan
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Chemistry Department, Lomonosov Moscow State University, Moscow, Russia.,Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
13
|
Qu X, Li J, Liu L, Zhang J, Hua Y, Suzuki K, Harada A, Ishida M, Yoshida N, Okuzaki D, Sakai Y, Sawa Y, Miyagawa S. ONO-1301 enhances post-transplantation survival of human induced pluripotent stem cell-derived cardiac tissue sheet by promoting angiogenesis. J Heart Lung Transplant 2023; 42:716-729. [PMID: 36964085 DOI: 10.1016/j.healun.2023.01.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 01/11/2023] [Accepted: 01/30/2023] [Indexed: 02/11/2023] Open
Abstract
BACKGROUND Transplanting human induced pluripotent stem cell-derived cardiomyocyte (hiPSC-CM) tissue sheets effectively treat ischemic cardiomyopathy. Cardiac functional recovery relies on graft survival in which angiogenesis played an important part. ONO-1301 is a synthetic prostacyclin analog with proangiogenic effects. We hypothesized that transplantation of hiPSC-CM tissue sheets with slow-release ONO-1301 scaffold could promote hostgraft angiogenesis, enhance tissue survival and therapeutic effect. METHODS We developed hiPSC-CM tissue sheets with ONO-1301 slow-release scaffold and evaluated their morphology, gene expression, and effects on angiogenesis. Three tissue sheet layers were transplanted into a rat myocardial infarction (MI) model. Left ventricular ejection fraction, gene expression in the MI border zone, and angiogenesis effects were investigated 4 weeks after transplantation. RESULTS In vitro assessment confirmed the slow-release of ONO-1301, and its pro-angiogenesis effects. In addition, in vivo data demonstrated that ONO-1301 administration positively correlated with graft survival. Cardiac tissue as thick as ∼900 μm was retained in the ONO (+) treated group. Additionally, left ventricular ejection fraction of the ONO (+) group was significantly enhanced, compared to ONO (-) group. The ONO (+) group also showed significantly improved interstitial fibrosis, higher capillary density, increased number of mature blood vessels, along with an enhanced supply of oxygen, and nutrients. CONCLUSIONS Slow-release ONO-1301 scaffold provided an efficient delivery method for thick hiPSC-CM tissue. ONO-1301 promotes angiogenesis between the host and graft and improves nutritional and oxygen supply, thereby enhancing the survival of transplanted cells, effectively improving ejection fraction, and therapeutic effects.
Collapse
Affiliation(s)
- Xiang Qu
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Junjun Li
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Li Liu
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Jingbo Zhang
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Ying Hua
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Kota Suzuki
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Akima Harada
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Masako Ishida
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Noriko Yoshida
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Daisuke Okuzaki
- Laboratory of Human Immunology (Single Cell Genomics), WPI Immunology Research Center, Osaka University, Osaka, Japan; Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Yoshiki Sakai
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yoshiki Sawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Shigeru Miyagawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.
| |
Collapse
|
14
|
Identification of marker genes to monitor residual iPSCs in iPSC-derived products. Cytotherapy 2023; 25:59-67. [PMID: 36319564 DOI: 10.1016/j.jcyt.2022.09.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 09/08/2022] [Accepted: 09/27/2022] [Indexed: 12/27/2022]
Abstract
BACKGROUND Engineered tissues and cell therapies based on human induced pluripotent stem cells (iPSCs) represent a promising approach for novel medicines. However, iPSC-derived cells and tissues may contain residual undifferentiated iPSCs that could lead to teratoma formation after implantation into patients. As a consequence, highly sensitive and specific methods for detecting residual undifferentiated iPSCs are indispensable for safety evaluations of iPSC-based therapies. The present study provides an approach for identifying potential marker genes for iPSC impurities in iPSC-derived cells using RNA sequencing data from iPSCs and various differentiated cell types. METHODS Identifying iPSC marker genes for each cell type individually provided a larger and more specific set of potential marker genes than considering all cell types in the analysis. Thus, the authors focused on identifying markers for iPSC impurities in iPSC-derived cardiomyocytes (iCMs) and validated the selected genes by reverse transcription quantitative polymerase chain reaction. The sensitivity of the candidate genes was determined by spiking different amounts of iPSCs into iCMs and their performance was compared with the previously suggested marker lin-28 homolog A (LIN28A). RESULTS Embryonic stem cell-related gene (ESRG), long intergenic non-protein coding RNA 678 (LINC00678), CaM kinase-like vesicle-associated (CAMKV), indoleamine 2,3-dioxygenase 1 (IDO1), chondromodulin (CNMD), LINE1-type transposase domain containing 1 (L1DT1), LIN28A, lymphocyte-specific protein tyrosine kinase (LCK), vertebrae development-associated (VRTN) and zinc finger and SCAN domain containing 10 (ZSCAN10) detected contaminant iPSCs among iCMs with a limit of detection that ranged from 0.001% to 0.1% depending on the gene and iCM batch used. CONCLUSIONS Using the example of iCMs, the authors provide a strategy for identifying a set of highly specific and sensitive markers that can be used for quality assessment of iPSC-derived products.
Collapse
|
15
|
Tsujimoto H, Osafune K. Current status and future directions of clinical applications using iPS cells-focus on Japan. FEBS J 2022; 289:7274-7291. [PMID: 34407307 DOI: 10.1111/febs.16162] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 08/04/2021] [Accepted: 08/17/2021] [Indexed: 01/13/2023]
Abstract
Regenerative medicine using iPS cell technologies has progressed remarkably in recent years. In this review, we summarize these technologies and their clinical application. First, we discuss progress in the establishment of iPS cells, including the HLA-homo iPS cell stock project in Japan and the advancement of low antigenic iPS cells using genome-editing technology. Then, we describe iPS cell-based therapies in or approaching clinical application, including those for ophthalmological, neurological, cardiac, hematological, cartilage, and metabolic diseases. Next, we introduce disease models generated from patient iPS cells and successfully used to identify therapeutic agents for intractable diseases. Clinical medicine using iPS cells has advanced safely and effectively by making full use of current scientific standards, but tests on cell safety need to be further developed and validated. The next decades will see the further spread of iPS cell technology-based regenerative medicine.
Collapse
Affiliation(s)
- Hiraku Tsujimoto
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Japan.,RegeNephro Co., Ltd., MIC bldg. Graduate School of Medicine, Kyoto University, Japan
| | - Kenji Osafune
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Japan.,Meiji University International Institute for Bio-Resource Research, Meiji University, Kanagawa, Japan
| |
Collapse
|
16
|
Fang J, Li JJ, Zhong X, Zhou Y, Lee RJ, Cheng K, Li S. Engineering stem cell therapeutics for cardiac repair. J Mol Cell Cardiol 2022; 171:56-68. [PMID: 35863282 DOI: 10.1016/j.yjmcc.2022.06.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 05/18/2022] [Accepted: 06/25/2022] [Indexed: 10/17/2022]
Abstract
Cardiovascular disease is the leading cause of death in the world. Stem cell-based therapies have been widely investigated for cardiac regeneration in patients with heart failure or myocardial infarction (MI) and surged ahead on multiple fronts over the past two decades. To enhance cellular therapy for cardiac regeneration, numerous engineering techniques have been explored to engineer cells, develop novel scaffolds, make constructs, and deliver cells or their derivatives. This review summarizes the state-of-art stem cell-based therapeutics for cardiac regeneration and discusses the emerged bioengineering approaches toward the enhancement of therapeutic efficacy of stem cell therapies in cardiac repair. We cover the topics in stem cell source and engineering, followed by stem cell-based therapies such as cell aggregates and cell sheets, and biomaterial-mediated stem cell therapies such as stem cell delivery with injectable hydrogel, three-dimensional scaffolds, and microneedle patches. Finally, we discuss future directions and challenges of engineering stem cell therapies for clinical translation.
Collapse
Affiliation(s)
- Jun Fang
- Department of Bioengineering, Department of Medicine, University of California, Los Angeles, Los Angeles, California 90095, USA; School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Jennifer J Li
- Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA; Department of Medicine, Cardiovascular Research Institute and Institute for Regeneration Medicine, University of California, San Francisco, CA 94143, USA
| | - Xintong Zhong
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yue Zhou
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Randall J Lee
- Department of Medicine, Cardiovascular Research Institute and Institute for Regeneration Medicine, University of California, San Francisco, CA 94143, USA
| | - Ke Cheng
- Department of Biomedical Engineering, North Carolina State University, NC, USA
| | - Song Li
- Department of Bioengineering, Department of Medicine, University of California, Los Angeles, Los Angeles, California 90095, USA; Eli and Edythe Broad Stem Cell Research Center, University of California, Los Angeles, California 90095, USA.
| |
Collapse
|
17
|
Nii T, Tabata Y. Immunosuppressive mesenchymal stem cells aggregates incorporating hydrogel microspheres promote an in vitro invasion of cancer cells. Regen Ther 2022; 18:516-522. [PMID: 34977285 PMCID: PMC8668441 DOI: 10.1016/j.reth.2021.11.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/16/2021] [Accepted: 11/24/2021] [Indexed: 12/20/2022] Open
Abstract
Introduction The objective of this study is to design a co-culture system of cancer cells and three-dimensional (3D) mesenchymal stem cells (MSC) aggregates for the in vitro evaluation of cancer invasion. Methods First, the MSC of an immunosuppressive phenotype (MSC2) were prepared by the MSC stimulation of polyriboinosinic polyribocytidylic acid. By simple mixing MSC2 and gelatin hydrogel microspheres (GM) in a U-bottomed well of 96 well plates which had been pre-coated with poly (vinyl alcohol), 3D MSC2 aggregates incorporating GM were obtained. The amount of chemokine (C–C motif) ligand 5 (CCL5) secreted from the MSC2 aggregates incorporating GM. Finally, an invasion assay was performed to evaluate the cancer invasion rate by co-cultured cancer cells and the 3D MSC2 incorporating GM. Results The amount of CCL5 secreted for the 3D MSC2 aggregates incorporating GM was significantly higher than that of two-dimensional (2D) MSC, 2D MSC2, and 3D MSC aggregates incorporating GM. When MDA-MB-231 human breast cancer cells were co-cultured with the 3D MSC2 aggregates incorporating GM, the invasion rate of cancer cells was significantly high compared with that of 2D MSC or 2D MSC2 and 3D MSC aggregates incorporating GM. In addition, high secretion of matrix metalloproteinase-2 was observed for the 3D MSC2 aggregates/cancer cells system. Conclusions It is concluded that the co-culture system of 3D MSC2 aggregates incorporating GM and cancer cells is promising to evaluate the invasion of cancer cells in vitro. This invasion model is an important tool for anti-cancer drug screening. Mesenchymal stem cells of an immunosuppressive phenotype (MSC2) were obtained. 3D MSC2 aggregates incorporating gelatin hydrogel microspheres were prepared. 3D MSC2 aggregates promoted the invasion rate of cancer cells.
Collapse
Key Words
- (CCL)5, chemokine (C–C motif) ligand
- 2D, two-dimensional
- 3D, three-dimensional
- Anti-cancer drug screening
- CAF, cancer-associated fibroblasts
- Cancer invasion model
- DDW, double-distilled water
- DMEM, Dulbecco's modified Eagle's medium
- ELISA, enzyme-linked immunosolvent assay
- FCS, fetal calf serum
- GM, gelatin hydrogel microspheres
- Gelatin hydrogel microspheres
- MEM, minimum essential medium
- MMP, matrix metalloproteinase
- MSC, mesenchymal stem cells
- MSC2, MSC of an immunosuppressive phenotype
- Mesenchymal stem cells
- PBS, phosphate buffered-saline
- PVA, poly (vinyl alcohol)
- TAM, tumor-associated macrophages
- Three-dimensional cell culture
Collapse
Affiliation(s)
- Teruki Nii
- Laboratory of Biomaterials, Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Kawara-cho Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yasuhiko Tabata
- Laboratory of Biomaterials, Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Kawara-cho Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| |
Collapse
|
18
|
Osada H, Kawatou M, Fujita D, Tabata Y, Minatoya K, Yamashita JK, Masumoto H. Therapeutic potential of clinical-grade human induced pluripotent stem cell-derived cardiac tissues. JTCVS OPEN 2021; 8:359-374. [PMID: 36004071 PMCID: PMC9390608 DOI: 10.1016/j.xjon.2021.09.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 09/24/2021] [Indexed: 11/12/2022]
Abstract
Objectives To establish a protocol to prepare and transplant clinical-grade human induced pluripotent stem cell (hiPSC)-derived cardiac tissues (HiCTs) and to evaluate the therapeutic potential in an animal myocardial infarction (MI) model. Methods We simultaneously differentiated clinical-grade hiPSCs into cardiovascular cell lineages with or without the administration of canonical Wnt inhibitors, generated 5- layer cell sheets with insertion of gelatin hydrogel microspheres (GHMs) (HiCTs), and transplanted them onto an athymic rat MI model. Cardiac function was evaluated by echocardiography and cardiac magnetic resonance imaging and compared with that in animals with sham and transplantation of 5-layer cell sheets without GHMs. Graft survival, ventricular remodeling, and neovascularization were evaluated histopathologically. Results The administration of Wnt inhibitors significantly promoted cardiomyocyte (CM) (P < .0001) and vascular endothelial cell (EC) (P = .006) induction, which resulted in cellular components of 52.0 ± 6.1% CMs and 9.9 ± 3.0% ECs. Functional analyses revealed the significantly lowest left ventricular end-diastolic volume and highest ejection fraction in the HiCT group. Histopathologic evaluation revealed that the HiCT group had a significantly larger median engrafted area (4 weeks, GHM(-) vs HiCT: 0.4 [range, 0.2-0.7] mm2 vs 2.2 [range, 1.8-3.1] mm2; P = .005; 12 weeks, 0 [range, 0-0.2] mm2 vs 1.9 [range, 0.1-3.2] mm2; P = .026), accompanied by the smallest scar area and highest vascular density at the MI border zone. Conclusions Transplantation of HiCTs generated from clinical-grade hiPSCs exhibited a prominent therapeutic potential in a rat MI model and may provide a promising therapeutic strategy in cardiac regenerative medicine.
Collapse
|
19
|
Kilmarx SE, Balsam LB. Commentary: The far-out prospect of cardiac regeneration after myocardial infarction. JTCVS OPEN 2021; 8:375-376. [PMID: 36004111 PMCID: PMC9390668 DOI: 10.1016/j.xjon.2021.10.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 10/15/2021] [Accepted: 10/22/2021] [Indexed: 10/26/2022]
Affiliation(s)
| | - Leora B. Balsam
- Division of Cardiac Surgery, UMass Memorial Medical Center, Worcester, Mass
| |
Collapse
|
20
|
Possible Treatment of Myocardial Infarct Based on Tissue Engineering Using a Cellularized Solid Collagen Scaffold Functionalized with Arg-Glyc-Asp (RGD) Peptide. Int J Mol Sci 2021; 22:ijms222212563. [PMID: 34830447 PMCID: PMC8620820 DOI: 10.3390/ijms222212563] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/23/2021] [Accepted: 11/15/2021] [Indexed: 12/13/2022] Open
Abstract
Currently, the clinical impact of cell therapy after a myocardial infarction (MI) is limited by low cell engraftment due to low cell retention, cell death in inflammatory and poor angiogenic infarcted areas, secondary migration. Cells interact with their microenvironment through integrin mechanoreceptors that control their survival/apoptosis/differentiation/migration and proliferation. The association of cells with a three-dimensional material may be a way to improve interactions with their integrins, and thus outcomes, especially if preparations are epicardially applied. In this review, we will focus on the rationale for using collagen as a polymer backbone for tissue engineering of a contractile tissue. Contractilities are reported for natural but not synthetic polymers and for naturals only for: collagen/gelatin/decellularized-tissue/fibrin/Matrigel™ and for different material states: hydrogels/gels/solids. To achieve a thick/long-term contractile tissue and for cell transfer, solid porous compliant scaffolds are superior to hydrogels or gels. Classical methods to produce solid scaffolds: electrospinning/freeze-drying/3D-printing/solvent-casting and methods to reinforce and/or maintain scaffold properties by reticulations are reported. We also highlight the possibility of improving integrin interaction between cells and their associated collagen by its functionalizing with the RGD-peptide. Using a contractile patch that can be applied epicardially may be a way of improving ventricular remodeling and limiting secondary cell migration.
Collapse
|
21
|
Müllenbroich MC, Kelly A, Acker C, Bub G, Bruegmann T, Di Bona A, Entcheva E, Ferrantini C, Kohl P, Lehnart SE, Mongillo M, Parmeggiani C, Richter C, Sasse P, Zaglia T, Sacconi L, Smith GL. Novel Optics-Based Approaches for Cardiac Electrophysiology: A Review. Front Physiol 2021; 12:769586. [PMID: 34867476 PMCID: PMC8637189 DOI: 10.3389/fphys.2021.769586] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 10/18/2021] [Indexed: 12/31/2022] Open
Abstract
Optical techniques for recording and manipulating cellular electrophysiology have advanced rapidly in just a few decades. These developments allow for the analysis of cardiac cellular dynamics at multiple scales while largely overcoming the drawbacks associated with the use of electrodes. The recent advent of optogenetics opens up new possibilities for regional and tissue-level electrophysiological control and hold promise for future novel clinical applications. This article, which emerged from the international NOTICE workshop in 2018, reviews the state-of-the-art optical techniques used for cardiac electrophysiological research and the underlying biophysics. The design and performance of optical reporters and optogenetic actuators are reviewed along with limitations of current probes. The physics of light interaction with cardiac tissue is detailed and associated challenges with the use of optical sensors and actuators are presented. Case studies include the use of fluorescence recovery after photobleaching and super-resolution microscopy to explore the micro-structure of cardiac cells and a review of two photon and light sheet technologies applied to cardiac tissue. The emergence of cardiac optogenetics is reviewed and the current work exploring the potential clinical use of optogenetics is also described. Approaches which combine optogenetic manipulation and optical voltage measurement are discussed, in terms of platforms that allow real-time manipulation of whole heart electrophysiology in open and closed-loop systems to study optimal ways to terminate spiral arrhythmias. The design and operation of optics-based approaches that allow high-throughput cardiac electrophysiological assays is presented. Finally, emerging techniques of photo-acoustic imaging and stress sensors are described along with strategies for future development and establishment of these techniques in mainstream electrophysiological research.
Collapse
Affiliation(s)
| | - Allen Kelly
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Corey Acker
- Center for Cell Analysis and Modeling, UConn Health, Farmington, CT, United States
| | - Gil Bub
- Department of Physiology, McGill University, Montréal, QC, Canada
| | - Tobias Bruegmann
- Institute for Cardiovascular Physiology, University Medical Center Goettingen, Goettingen, Germany
| | - Anna Di Bona
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Emilia Entcheva
- Department of Biomedical Engineering, The George Washington University, Washington, DC, United States
| | | | - Peter Kohl
- Institute for Experimental Cardiovascular Medicine, University Heart Center and Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Stephan E. Lehnart
- Heart Research Center Göttingen, University Medical Center Göttingen, Göttingen, Germany
- Department of Cardiology and Pneumology, Georg-August University Göttingen, Göttingen, Germany
- Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells” (MBExC), University of Göttingen, Göttingen, Germany
| | - Marco Mongillo
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, Padova, Italy
| | | | - Claudia Richter
- German Primate Center - Leibniz Institute for Primate Research, Göttingen, Germany
| | - Philipp Sasse
- Institute of Physiology I, Medical Faculty, University of Bonn, Bonn, Germany
| | - Tania Zaglia
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, Padova, Italy
| | - Leonardo Sacconi
- European Laboratory for Nonlinear Spectroscopy, Sesto Fiorentino, Italy
- Institute for Experimental Cardiovascular Medicine, University Heart Center and Medical Faculty, University of Freiburg, Freiburg, Germany
- National Institute of Optics, National Research Council, Florence, Italy
| | - Godfrey L. Smith
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
22
|
Nii T. Strategies Using Gelatin Microparticles for Regenerative Therapy and Drug Screening Applications. Molecules 2021; 26:molecules26226795. [PMID: 34833885 PMCID: PMC8617939 DOI: 10.3390/molecules26226795] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 11/17/2022] Open
Abstract
Gelatin, a denatured form of collagen, is an attractive biomaterial for biotechnology. In particular, gelatin particles have been noted due to their attractive properties as drug carriers. The drug release from gelatin particles can be easily controlled by the crosslinking degree of gelatin molecule, responding to the purpose of the research. The gelatin particles capable of drug release are effective in wound healing, drug screening models. For example, a sustained release of growth factors for tissue regeneration at the injured sites can heal a wound. In the case of the drug screening model, a tissue-like model composed of cells with high activity by the sustained release of drug or growth factor provides reliable results of drug effects. Gelatin particles are effective in drug delivery and the culture of spheroids or cell sheets because the particles prevent hypoxia-derived cell death. This review introduces recent research on gelatin microparticles-based strategies for regenerative therapy and drug screening models.
Collapse
Affiliation(s)
- Teruki Nii
- Graduate School of Systems Life Sciences, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan;
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| |
Collapse
|
23
|
Gelatin hydrogel nonwoven fabrics of a cell culture scaffold to formulate 3-dimensional cell constructs. Regen Ther 2021; 18:418-429. [PMID: 34722838 PMCID: PMC8524245 DOI: 10.1016/j.reth.2021.09.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 09/09/2021] [Accepted: 09/29/2021] [Indexed: 12/12/2022] Open
Abstract
The objective of this study is to evaluate the possibility of gelatin hydrogel nonwoven fabrics (GHNF) of a cell culture scaffold to formulate 3-dimensional (3D) cell construct. The thickness of cell construct is about 1 mm and the cells inside are live and bio-active, irrespective of their internal distribution. The GHNF were prepared by the solution blow method of gelatin, following by dehydrothermal crosslinking. The GHNF showed a mechanical strength strong enough not to allow the shape to deform even in a wet state. The wet GHNF also showed resistance against repeated compression. After human mesenchymal stromal cells (hMSC) were seeded and cultured, the inner distribution in GHNF, the apoptosis, hypoxia inducible factor (HIF)-1α, Ki67, collagen or sulfated glycosaminoglycan (sGAG) secretion of cells were evaluated. The hMSC proliferated inside the GHNF with time while a homogeneous distribution in the number of cells proliferated from the surface to the 1000 μm depth of GHNF was observed. The number of apoptosis and HIF-1α positive cells was significantly low compared with that of polypropylene nonwoven fabrics with the similar fiber diameters and intra-structure. The GHNF were degraded during cell culture, and completely replaced by collagen and sGAG secreted. It is concluded that the GHNF is a promising cell culture scaffold for 3D cell constructs.
Collapse
|
24
|
Vaka R, Davis DR. State-of-play for cellular therapies in cardiac repair and regeneration. Stem Cells 2021; 39:1579-1588. [PMID: 34448513 PMCID: PMC9290630 DOI: 10.1002/stem.3446] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 08/10/2021] [Indexed: 12/24/2022]
Abstract
Cardiovascular disease is the primary cause of death around the world. For almost two decades, cell therapy has been proposed as a solution for heart disease. In this article, we report on the “state‐of‐play” of cellular therapies for cardiac repair and regeneration. We outline the progression of new ideas from the preclinical literature to ongoing clinical trials. Recent data supporting the mechanics and mechanisms of myogenic and paracrine therapies are evaluated in the context of long‐term cardiac engraftment. This discussion informs on promising new approaches to indicate future avenues for the field.
Collapse
Affiliation(s)
- Ramana Vaka
- Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, Canada
| | - Darryl R Davis
- Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, Canada
| |
Collapse
|
25
|
Biomaterial-Assisted Regenerative Medicine. Int J Mol Sci 2021; 22:ijms22168657. [PMID: 34445363 PMCID: PMC8395440 DOI: 10.3390/ijms22168657] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/06/2021] [Accepted: 08/10/2021] [Indexed: 12/11/2022] Open
Abstract
This review aims to show case recent regenerative medicine based on biomaterial technologies. Regenerative medicine has arousing substantial interest throughout the world, with “The enhancement of cell activity” one of the essential concepts for the development of regenerative medicine. For example, drug research on drug screening is an important field of regenerative medicine, with the purpose of efficient evaluation of drug effects. It is crucial to enhance cell activity in the body for drug research because the difference in cell condition between in vitro and in vivo leads to a gap in drug evaluation. Biomaterial technology is essential for the further development of regenerative medicine because biomaterials effectively support cell culture or cell transplantation with high cell viability or activity. For example, biomaterial-based cell culture and drug screening could obtain information similar to preclinical or clinical studies. In the case of in vivo studies, biomaterials can assist cell activity, such as natural healing potential, leading to efficient tissue repair of damaged tissue. Therefore, regenerative medicine combined with biomaterials has been noted. For the research of biomaterial-based regenerative medicine, the research objective of regenerative medicine should link to the properties of the biomaterial used in the study. This review introduces regenerative medicine with biomaterial.
Collapse
|
26
|
Yin S, Cao Y. Hydrogels for Large-Scale Expansion of Stem Cells. Acta Biomater 2021; 128:1-20. [PMID: 33746032 DOI: 10.1016/j.actbio.2021.03.026] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/25/2021] [Accepted: 03/10/2021] [Indexed: 12/18/2022]
Abstract
Stem cells demonstrate considerable promise for various preclinical and clinical applications, including drug screening, disease treatments, and regenerative medicine. Producing high-quality and large amounts of stem cells is in demand for these applications. Despite challenges, as hydrogel-based cell culture technology has developed, tremendous progress has been made in stem cell expansion and directed differentiation. Hydrogels are soft materials with abundant water. Many hydrogel properties, including biodegradability, mechanical strength, and porosity, have been shown to play essential roles in regulating stem cell proliferation and differentiation. The biochemical and physical properties of hydrogels can be specifically tailored to mimic the native microenvironment that various stem cells reside in vivo. A few hydrogel-based systems have been developed for successful stem cell cultures and expansion in vitro. In this review, we summarize various types of hydrogels that have been designed to effectively enhance the proliferation of hematopoietic stem cells (HSCs), mesenchymal stem/stromal cells (MSCs), and pluripotent stem cells (PSCs), respectively. According to each stem cell type's preference, we also discuss strategies for fabricating hydrogels with biochemical and mechanical cues and other characteristics representing microenvironments of stem cells in vivo. STATEMENT OF SIGNIFICANCE: In this review article we summarize current progress on the construction of hydrogel systems for the culture and expansion of various stem cells, including hematopoietic stem cells (HSCs), mesenchymal stem/stromal cells (MSCs), and pluripotent stem cells (PSCs). The Significance includes: (1) Provide detailed discussion on the stem cell niches that should be considered for stem cell in vitro expansion. (2) Summarize various strategies to construct hydrogels that can largely recapture the microenvironment of native stem cells. (3) Suggest a few future directions that can be implemented to improve current in vitro stem cell expansion systems.
Collapse
Affiliation(s)
- Sheng Yin
- National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, 210093, China; Chemistry and Biomedicine innovation center, Nanjing University, Nanjing, 210093, China; Institute for Brain Sciences, Nanjing University, Nanjing, 210093, China; Shenzhen Research Institute of Nanjing University, Shenzhen, China, 518057
| | - Yi Cao
- National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, 210093, China; Chemistry and Biomedicine innovation center, Nanjing University, Nanjing, 210093, China; Institute for Brain Sciences, Nanjing University, Nanjing, 210093, China; Shenzhen Research Institute of Nanjing University, Shenzhen, China, 518057.
| |
Collapse
|
27
|
Gähwiler EKN, Motta SE, Martin M, Nugraha B, Hoerstrup SP, Emmert MY. Human iPSCs and Genome Editing Technologies for Precision Cardiovascular Tissue Engineering. Front Cell Dev Biol 2021; 9:639699. [PMID: 34262897 PMCID: PMC8273765 DOI: 10.3389/fcell.2021.639699] [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: 12/09/2020] [Accepted: 03/31/2021] [Indexed: 12/12/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) originate from the reprogramming of adult somatic cells using four Yamanaka transcription factors. Since their discovery, the stem cell (SC) field achieved significant milestones and opened several gateways in the area of disease modeling, drug discovery, and regenerative medicine. In parallel, the emergence of clustered regularly interspaced short palindromic repeats (CRISPR)-associated protein 9 (CRISPR-Cas9) revolutionized the field of genome engineering, allowing the generation of genetically modified cell lines and achieving a precise genome recombination or random insertions/deletions, usefully translated for wider applications. Cardiovascular diseases represent a constantly increasing societal concern, with limited understanding of the underlying cellular and molecular mechanisms. The ability of iPSCs to differentiate into multiple cell types combined with CRISPR-Cas9 technology could enable the systematic investigation of pathophysiological mechanisms or drug screening for potential therapeutics. Furthermore, these technologies can provide a cellular platform for cardiovascular tissue engineering (TE) approaches by modulating the expression or inhibition of targeted proteins, thereby creating the possibility to engineer new cell lines and/or fine-tune biomimetic scaffolds. This review will focus on the application of iPSCs, CRISPR-Cas9, and a combination thereof to the field of cardiovascular TE. In particular, the clinical translatability of such technologies will be discussed ranging from disease modeling to drug screening and TE applications.
Collapse
Affiliation(s)
- Eric K. N. Gähwiler
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
| | - Sarah E. Motta
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
- Wyss Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Marcy Martin
- Division of Pediatric Cardiology, Department of Pediatrics, Stanford School of Medicine, Stanford, CA, United States
- Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford School of Medicine, Stanford, CA, United States
- Stanford Cardiovascular Institute, Stanford School of Medicine, Stanford, CA, United States
| | - Bramasta Nugraha
- Molecular Parasitology Lab, Institute of Parasitology, University of Zurich, Zurich, Switzerland
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism, R&D BioPharmaceuticals, AstraZeneca, Gothenburg, Sweden
| | - Simon P. Hoerstrup
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
- Wyss Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Maximilian Y. Emmert
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
- Wyss Zurich, University and ETH Zurich, Zurich, Switzerland
- Department of Cardiovascular Surgery, Charité Universitätsmedizin Berlin, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany
| |
Collapse
|
28
|
Wongin S, Wangdee C, Nantavisai S, Banlunara W, Nakbunnum R, Waikakul S, Chotiyarnwong P, Roytrakul S, Viravaidya-Pasuwat K. Evaluation of osteochondral-like tissues using human freeze-dried cancellous bone and chondrocyte sheets to treat osteochondral defects in rabbits. Biomater Sci 2021; 9:4701-4716. [PMID: 34019604 DOI: 10.1039/d1bm00239b] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Human freeze-dried cancellous bone combined with human chondrocyte sheets have recently been used to construct an osteochondral-like tissue, which resembled a cartilage layer on a subchondral bone layer. Nevertheless, the efficacy of these human tissues in a xenogeneic model has been rarely reported. Therefore, this study aimed to evaluate the potential of human freeze-dried cancellous bones combined with human chondrocyte sheets for the treatment of osteochondral defects in rabbits. The key roles of the extracellular matrix (ECM) and released cytokines in these tissues in osteochondral repair were also assessed. Triple-layered chondrocyte sheets were constructed using a temperature-responsive culture surface. Then, they were placed onto cancellous bone to form chondrocyte sheet-cancellous bone tissues. The immunostaining of collagen type II (COL2) and the proteomic analysis of the human tissues were carried out before the transplantation. In our in vitro study, the triple-layered chondrocyte sheets adhered well on the cancellous bone, and the COL2 expression was apparent throughout the tissue structures. From the proteomic analysis results, it was found that the major function of the secreted proteins found in these tissues was protein binding. The distinct pathways were focal adhesion and the ECM-receptor interaction pathways. Among the highly expressed proteins, laminin-alpha 5 (LAMA5) and fibronectin (FN) not only played roles in the protein binding and ECM-receptor interaction, but also were involved in the cytokine-mediated signaling pathway. At 12 weeks after xenogeneic transplantation, compared to the control group, the defects treated with the chondrocyte sheets showed more hyaline-like cartilage tissue, as indicated by the abundance of safranin-O and COL2 with a partial collagen type I (COL1) expression. At 4, 8, and 12 weeks, compared to the defects treated with the cancellous bone, the staining of safranin-O and COL2 was more apparent in the defects treated with the chondrocyte sheet-cancellous bone tissues. Therefore, the human chondrocyte sheets and chondrocyte sheet-cancellous bone tissues provide a potential treatment for rabbit femoral condyle defect. LAMA5 and FN found in these human xenografts and their culture media might play key roles in the ECM-receptor interaction and might be involved in the cytokine-mediated signaling pathway during tissue repair.
Collapse
Affiliation(s)
- Sopita Wongin
- Biological Engineering Program, Faculty of Engineering, King Mongkut's University of Technology Thonburi, Bangkok 10140, Thailand.
| | - Chalika Wangdee
- Department of Veterinary Surgery, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand.
| | - Sirirat Nantavisai
- Special Task Force for Activating Research (STAR) in Biology of Embryo and Stem Cell Research in Veterinary Science, Veterinary Stem Cells and Bioengineering Innovation Center (VSCBIC), Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand.
| | - Wijit Banlunara
- Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand.
| | - Rapeepat Nakbunnum
- Department of Orthopedic Surgery, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand.
| | - Saranatra Waikakul
- Department of Orthopedic Surgery, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand.
| | - Pojchong Chotiyarnwong
- Department of Orthopedic Surgery, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand.
| | - Sittiruk Roytrakul
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Thailand Science Park, Pathum Thani, 12120, Thailand.
| | - Kwanchanok Viravaidya-Pasuwat
- Biological Engineering Program, Faculty of Engineering, King Mongkut's University of Technology Thonburi, Bangkok 10140, Thailand. and Department of Chemical Engineering, Faculty of Engineering, King Mongkut's University of Technology Thonburi, Bangkok 10140, Thailand.
| |
Collapse
|
29
|
Guo R, Wan F, Morimatsu M, Xu Q, Feng T, Yang H, Gong Y, Ma S, Chang Y, Zhang S, Jiang Y, Wang H, Chang D, Zhang H, Ling Y, Lan F. Cell sheet formation enhances the therapeutic effects of human umbilical cord mesenchymal stem cells on myocardial infarction as a bioactive material. Bioact Mater 2021; 6:2999-3012. [PMID: 33732969 PMCID: PMC7941025 DOI: 10.1016/j.bioactmat.2021.01.036] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 01/21/2021] [Accepted: 01/28/2021] [Indexed: 12/13/2022] Open
Abstract
Stem cell-based therapy has been used to treat ischaemic heart diseases for two decades. However, optimal cell types and transplantation methods remain unclear. This study evaluated the therapeutic effects of human umbilical cord mesenchymal stem cell (hUCMSC) sheet on myocardial infarction (MI). Methods hUCMSCs expressing luciferase were generated by lentiviral transduction for in vivo bio-luminescent imaging tracking of cells. We applied a temperature-responsive cell culture surface-based method to form the hUCMSC sheet. Cell retention was evaluated using an in vivo bio-luminescent imaging tracking system. Unbiased transcriptional profiling of infarcted hearts and further immunohistochemical assessment of monocyte and macrophage subtypes were used to determine the mechanisms underlying the therapeutic effects of the hUCMSC sheet. Echocardiography and pathological analyses of heart sections were performed to evaluate cardiac function, angiogenesis and left ventricular remodelling. Results When transplanted to the infarcted mouse hearts, hUCMSC sheet significantly improved the retention and survival compared with cell suspension. At the early stage of MI, hUCMSC sheet modulated inflammation by decreasing Mcp1-positive monocytes and CD68-positive macrophages and increasing Cx3cr1-positive non-classical macrophages, preserving the cardiomyocytes from acute injury. Moreover, the extracellular matrix produced by hUCMSC sheet then served as bioactive scaffold for the host cells to graft and generate new epicardial tissue, providing mechanical support and routes for revascularsation. These effects of hUCMSC sheet treatment significantly improved the cardiac function at days 7 and 28 post-MI. Conclusions hUCMSC sheet formation dramatically improved the biological functions of hUCMSCs, mitigating adverse post-MI remodelling by modulating the inflammatory response and providing bioactive scaffold upon transplantation into the heart. Translational perspective Due to its excellent availability as well as superior local cellular retention and survival, allogenic transplantation of hUCMSC sheets can more effectively acquire the biological functions of hUCMSCs, such as modulating inflammation and enhancing angiogenesis. Moreover, the hUCMSC sheet method allows the transfer of an intact extracellular matrix without introducing exogenous or synthetic biomaterial, further improving its clinical applicability. Cell sheet formation of hUCMSCs dramatically improves post transplantation cell survival in the infarcted heart. hUCMSC sheet protects cardiomyocytes from infarction by alleviating acute inflammation. The ECM of cell sheet serves as bioactive scaffold to allow the host cells to integrate and form new epicardial tissue. The new epicardial tissue can provide mechanical support and new routes for revascularization.
Collapse
Affiliation(s)
- Rui Guo
- Department of Cardiac Surgery, Peking University Third Hospital, Beijing, 100191, China.,Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, 700-8558, Japan
| | - Feng Wan
- Department of Cardiac Surgery, Peking University Third Hospital, Beijing, 100191, China.,Department of Cardiovascular Surgery, Tongji University East Hospital, Shanghai, 200120, China
| | - Masatoshi Morimatsu
- Department of Cardiac Surgery, Peking University Third Hospital, Beijing, 100191, China.,Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, 700-8558, Japan
| | - Qing Xu
- Department of Cardiac Surgery, Peking University Third Hospital, Beijing, 100191, China
| | - Tian Feng
- Department of Cardiac Surgery, Peking University Third Hospital, Beijing, 100191, China.,Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, 700-8558, Japan
| | - Hang Yang
- Department of Cardiac Surgery, Peking University Third Hospital, Beijing, 100191, China
| | - Yichen Gong
- Department of Cardiac Surgery, Peking University Third Hospital, Beijing, 100191, China
| | - Shuhong Ma
- Department of Cardiac Surgery, Peking University Third Hospital, Beijing, 100191, China.,State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Yun Chang
- Department of Cardiac Surgery, Peking University Third Hospital, Beijing, 100191, China.,State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Siyao Zhang
- Department of Cardiac Surgery, Peking University Third Hospital, Beijing, 100191, China.,State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Youxu Jiang
- Department of Cardiac Surgery, Peking University Third Hospital, Beijing, 100191, China.,State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Heqing Wang
- Department of Cardiac Surgery, Peking University Third Hospital, Beijing, 100191, China.,Department of Cardiovascular Surgery, Tongji University East Hospital, Shanghai, 200120, China
| | - Dehua Chang
- Department of Cardiac Surgery, Peking University Third Hospital, Beijing, 100191, China.,Department of Cardiac Surgery, The University of Tokyo Hospital, Tokyo, 113-8655, Japan
| | - Hongjia Zhang
- Department of Cardiac Surgery, Peking University Third Hospital, Beijing, 100191, China.,Beijing Laboratory for Cardiovascular Precision Medicine, MOE Key Laboratory of Medical Engineering for Cardiovascular Diseases, Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Yunpeng Ling
- Department of Cardiac Surgery, Peking University Third Hospital, Beijing, 100191, China
| | - Feng Lan
- Department of Cardiac Surgery, Peking University Third Hospital, Beijing, 100191, China.,State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.,Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, Shenzhen, China
| |
Collapse
|
30
|
Recent progress in induced pluripotent stem cell-derived 3D cultures for cardiac regeneration. Cell Tissue Res 2021; 384:231-240. [PMID: 33544212 DOI: 10.1007/s00441-021-03414-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 01/10/2021] [Indexed: 12/12/2022]
Abstract
Cardiovascular diseases are the leading cause of death in the world due to the high incidence of the diseases coupled with the limited therapeutic options. In recent years, advances in regenerative medicine have emerged as a promising treatment. Differentiation of induced pluripotent stem cells (iPSCs) into cardiac cells and emerging technologies allowing arrangement of cells into complex 3D tissue-like structures open new frontiers for transplantation and engraftment of these tissue patches onto the damaged heart. Despite the cells integrating and presenting initial neovascularization, the functional and electric properties of these patches are still not comparable with those of the host cardiac tissue. Future research optimizing maturation and integration of the iPSC-derived cardiomyocytes is paramount for cardiac cell therapy to attain clinical use. Herein, we will review the state of the art and the different approaches to constructing these 3D transplantable structures.
Collapse
|
31
|
Lee J, Park D, Seo Y, Chung JJ, Jung Y, Kim SH. Organ-Level Functional 3D Tissue Constructs with Complex Compartments and their Preclinical Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e2002096. [PMID: 33103834 DOI: 10.1002/adma.202002096] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 06/16/2020] [Indexed: 06/11/2023]
Abstract
There is an increasing interest in organ-level 3D tissue constructs, owing to their mirroring of in vivo-like features. This has resulted in a wide range of preclinical applications to obtain cell- or tissue-specific responses. Additionally, the development and improvement of sophisticated technologies, such as organoid generation, microfluidics, hydrogel engineering, and 3D printing, have enhanced 3D tissue constructs to become more elaborate. In particular, recent studies have focused on including complex compartments, i.e., vascular and innervation structured 3D tissue constructs, which mimic the nature of the human body in that all tissues/organs are interconnected and physiological phenomena are mediated through vascular and neural systems. Here, the strategies are categorized according to the number of dimensions (0D, 1D, 2D, and 3D) of the starting materials for scaling up, and novel approaches to introduce increased complexity in 3D tissue constructs are highlighted. Recent advances in preclinical applications are also investigated to gain insight into the future direction of 3D tissue construct research. Overcoming the challenges in improving organ-level functional 3D tissue constructs both in vitro and in vivo will ultimately become a life-saving tool in the biomedical field.
Collapse
Affiliation(s)
- Jaeseo Lee
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - DoYeun Park
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
- Biomaterials Research Center, Korea Institute of Science and Technology (KIST), 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Yoojin Seo
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
- Center for BioMicrosystems, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Justin J Chung
- Biomaterials Research Center, Korea Institute of Science and Technology (KIST), 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Youngmee Jung
- Biomaterials Research Center, Korea Institute of Science and Technology (KIST), 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Soo Hyun Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
- Biomaterials Research Center, Korea Institute of Science and Technology (KIST), 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| |
Collapse
|
32
|
Osada H, Ho WJ, Yamashita H, Yamazaki K, Ikeda T, Minatoya K, Masumoto H. Novel device prototyping for endoscopic cell sheet transplantation using a three-dimensional printed simulator. Regen Ther 2020; 15:258-264. [PMID: 33426227 PMCID: PMC7770426 DOI: 10.1016/j.reth.2020.10.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 10/15/2020] [Accepted: 10/28/2020] [Indexed: 12/28/2022] Open
Abstract
Introduction Considering higher risks of candidates for cardiac regenerative therapy with compromised cardiac function, it is anticipated to develop less invasive surgical procedures. In the present study, we aimed to develop a prototype of totally endoscopic cell sheet delivery device and evaluate the surgical technique for epicardial cell sheet placement using three-dimensional (3D) printed simulators based on human computed tomography data. Methods We designed an endoscopic cell sheet delivery device with outer and inner frame with self-expandable applicator which can be opened in thoracic cavity. We launched spout line to provide liquids on the applicator surface and tension line to gently bend the applicator dorsally. We prepared human mesenchymal stem cell (MSC) sheets and compared wet/dry conditions of 3D printed heart/porcine heart and applicator to identify suitable conditions for cell sheet transplantation. Finally we validated the feasibility of endoscopic transplantation to anterior and lateral wall of left ventricle using 3D printed simulators. Results Moist condition of both 3D printed heart/porcine heart surface and applicator at transplantation yielded highest successful rate (100%, p = 0.0197). For both endoscopic transplantation sites, MSC sheets were successfully deployed. The procedure duration was 157 ± 23 s for anterior wall and 123 ± 13 s for the lateral wall in average, respectively. Conclusions We developed a novel prototype of endoscopic cell sheet delivery device for minimally-invasive cardiac regenerative therapy utilizing a 3D printed simulator. The commercialization of the prototype may provide a safe minimally-invasive method to deliver potential cardiac regenerative therapy in the future. We invented a novel device for endoscopic cell sheet transplantation. We optimized the transplantation procedure using three-dimensional printed simulator. Commercialization of the device may provide standardized cardiac regenerative therapy.
Collapse
Affiliation(s)
- Hiroaki Osada
- Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawaharacho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Wen-Jin Ho
- Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawaharacho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Hideki Yamashita
- Industrial Materials Technology Development Department, Ashimori Industry Co., Ltd., 7-11-61, Senrioka, Settsu, Osaka, 566-0001, Japan
| | - Kazuhiro Yamazaki
- Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawaharacho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Tadashi Ikeda
- Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawaharacho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Kenji Minatoya
- Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawaharacho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Hidetoshi Masumoto
- Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawaharacho, Sakyo-ku, Kyoto, 606-8507, Japan.,Clinical Translational Research Program, RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojimaminami-cho, Chuo-ku, Kobe, 650-0047, Japan
| |
Collapse
|
33
|
He L, Chen X. Cardiomyocyte Induction and Regeneration for Myocardial Infarction Treatment: Cell Sources and Administration Strategies. Adv Healthc Mater 2020; 9:e2001175. [PMID: 33000909 DOI: 10.1002/adhm.202001175] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/15/2020] [Indexed: 02/06/2023]
Abstract
Occlusion of coronary artery and subsequent damage or death of myocardium can lead to myocardial infarction (MI) and even heart failure-one of the leading causes of deaths world wide. Notably, myocardium has extremely limited regeneration potential due to the loss or death of cardiomyocytes (i.e., the cells of which the myocardium is comprised) upon MI. A variety of stem cells and stem cell-derived cardiovascular cells, in situ cardiac fibroblasts and endogenous proliferative epicardium, have been exploited to provide renewable cellular sources to treat injured myocardium. Also, different strategies, including direct injection of cell suspensions, bioactive molecules, or cell-incorporated biomaterials, and implantation of artificial cardiac scaffolds (e.g., cell sheets and cardiac patches), have been developed to deliver renewable cells and/or bioactive molecules to the MI site for the myocardium regeneration. This article briefly surveys cell sources and delivery strategies, along with biomaterials and their processing techniques, developed for MI treatment. Key issues and challenges, as well as recommendations for future research, are also discussed.
Collapse
Affiliation(s)
- Lihong He
- Department of Cell Biology Medical College of Soochow University Suzhou 215123 China
| | - Xiongbiao Chen
- Department of Mechanical Engineering Division of Biomedical Engineering University of Saskatchewan Saskatoon S7N5A9 Canada
| |
Collapse
|
34
|
Abstract
The regenerative capacity of the heart has long fascinated scientists. In contrast to other organs such as liver, skin, and skeletal muscle, the heart possesses only a minimal regenerative capacity. It lacks a progenitor cell population, and cardiomyocytes exit the cell cycle shortly after birth and do not re-enter after injury. Thus, any loss of cardiomyocytes is essentially irreversible and can lead to or exaggerate heart failure, which represents a major public health problem. New therapeutic options are urgently needed, but regenerative therapies have remained an unfulfilled promise in cardiovascular medicine until today. Yet, through a clearer comprehension of signaling pathways that regulate the cardiomyocyte cell cycle and advances in stem cell technology, strategies have evolved that demonstrate the potential to generate new myocytes and thereby fulfill an essential central criterion for heart repair.
Collapse
Affiliation(s)
- Florian Weinberger
- Institute for Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; , .,German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Thomas Eschenhagen
- Institute for Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; , .,German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| |
Collapse
|
35
|
Murata K, Ikegawa M, Minatoya K, Masumoto H. Strategies for immune regulation in iPS cell-based cardiac regenerative medicine. Inflamm Regen 2020; 40:36. [PMID: 33005258 PMCID: PMC7523082 DOI: 10.1186/s41232-020-00145-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 09/09/2020] [Indexed: 01/14/2023] Open
Abstract
Cardiac regenerative therapy is expected to be a promising therapeutic option for the treatment of severe cardiovascular diseases. Artificial tissues or organoids made from cardiovascular cell lineages differentiated from human induced pluripotent stem cells (iPSCs) are expected to regenerate the damaged heart. Even though immune rejection rarely occurs when iPSC-derived graft and the recipient have the same HLA type, in some cases, such as tissue transplantation onto hearts, the HLA matching would not be sufficient to fully control immune rejection. The present review introduces recent immunomodulatory strategies in iPSC-based transplantation therapies other than MHC matching including the induction of immune tolerance through iPSC-derived antigen-presenting cells, simultaneous transplantation of syngeneic mesenchymal stem cells, and using the universal donor cells such as gene editing-based HLA modulation in iPSCs to regulate T cell compatibility. In addition, we present future perspectives for proper adjustment of immunosuppression therapy after iPSC-derived tissue/organoid-based cardiac regenerative therapies by identifying biomarkers monitoring immune rejection.
Collapse
Affiliation(s)
- Kozue Murata
- Clinical Translational Research Program, RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047 Japan.,Institute for Advancement of Clinical and Translational Science, Kyoto University Hospital, Kyoto, Japan
| | - Masaya Ikegawa
- Department of Life and Medical Systems, Faculty of Life and Medical Sciences, Doshisha University, Kyoto, Japan
| | - Kenji Minatoya
- Institute for Advancement of Clinical and Translational Science, Kyoto University Hospital, Kyoto, Japan.,Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hidetoshi Masumoto
- Clinical Translational Research Program, RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047 Japan.,Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
36
|
Nakamura K, Saotome T, Shimada N, Matsuno K, Tabata Y. A Gelatin Hydrogel Nonwoven Fabric Facilitates Metabolic Activity of Multilayered Cell Sheets. Tissue Eng Part C Methods 2020; 25:344-352. [PMID: 31062648 DOI: 10.1089/ten.tec.2019.0061] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
IMPACT STATEMENT This study introduces the utility of gelatin hydrogel nonwoven fabrics (GHNFs) for cell sheet engineering. The GHNF had the mechanical property strong enough to hold by forceps even in the swollen condition. The cell sheet harvest and transfer processes were performed simpler and faster than those without using the GHNF. The GHNF facilitates the metabolic activity of three-layered cell sheets, and the cell migration from cell sheets into the GHNF was observed. The GHNF is a promising material used to support cell sheets during the process of assemble formulation and contributes to the improved biological functions of tissue-like cell constructs.
Collapse
Affiliation(s)
- Koichiro Nakamura
- 1 Research and Development Center, The Japan Wool Textile Co., Ltd., Hyogo, Japan.,2 Laboratory of Biomaterials, Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Toshiki Saotome
- 1 Research and Development Center, The Japan Wool Textile Co., Ltd., Hyogo, Japan.,2 Laboratory of Biomaterials, Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Naoki Shimada
- 1 Research and Development Center, The Japan Wool Textile Co., Ltd., Hyogo, Japan
| | - Kumiko Matsuno
- 1 Research and Development Center, The Japan Wool Textile Co., Ltd., Hyogo, Japan.,2 Laboratory of Biomaterials, Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Yasuhiko Tabata
- 2 Laboratory of Biomaterials, Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| |
Collapse
|
37
|
Osada H, Kawatou M, Takeda M, Jo JI, Murakami T, Tabata Y, Minatoya K, Yamashita JK, Masumoto H. Accuracy of spiked cell counting methods for designing a pre-clinical tumorigenicity study model. Heliyon 2020; 6:e04423. [PMID: 32685738 PMCID: PMC7358391 DOI: 10.1016/j.heliyon.2020.e04423] [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: 07/25/2019] [Revised: 01/28/2020] [Accepted: 07/08/2020] [Indexed: 11/09/2022] Open
Abstract
Background Evaluations for the tumorigenicity of transplantation of stem cell products is mandatory for clinical application. It is of importance to establish a system to accurately quantify contaminated tumorigenic cells regardless of the format of stem cell product. In the present report, we aimed to examine the accuracy of the quantification of tumorigenic cell numbers with commonly used 2 methods, quantitative polymerase chain reaction (qPCR) and flow cytometry (FCM) using experimental models of stem cell products spiked with tumorigenic cells. Methods Human mesenchymal stem cells (hMSCs) and melanoma Mewo-Luc cells constitutively expressing luciferase were used. We stained Mewo-Luc cells with a cell linker then spiked onto hMSC suspensions and hMSC sheets. We validated the accuracy of 10-fold serial dilution technique for Mewo-Luc cell suspension using a Coulter counter. The samples spiked with Mewo-Luc cells were subjected to qPCR and FCM analyses, respectively for the quantification of Mewo-Luc cells. Results Ten-fold serial dilutions of Mewo-Luc cells were performed accurately with small deviation. In samples spiked with or less than 100 cells in hMSC suspensions, and samples spiked with or less than 1,000 cells in hMSC sheets showed significantly higher cell numbers in calculations by FCM, respectively (suspensions; qPCR vs FCM: 100 cells: 59 ± 25 vs 232 ± 35 cells, p = 0.022/10 cells: 21 ± 7 vs 114 ± 27 cells, p = 0.030, sheets; qPCR vs FCM: 1,000 cells: 1723 ± 258 vs 5810 ± 878 cells, p = 0.012/100 cells: 110 ± 18 vs 973 ± 232 cells, p = 0.012/10 cells: 20 ± 6 vs 141 ± 36 cells, p = 0.030). Conclusion Differences in accuracy between quantification methods should be considered in designing a tumorigenicity study model.
Collapse
Affiliation(s)
- Hiroaki Osada
- Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Masahide Kawatou
- Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan.,Institute for Advancement of Clinical and Translational Science, Kyoto University Hospital, Kyoto, Japan
| | - Masafumi Takeda
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan.,Institute for Advancement of Clinical and Translational Science, Kyoto University Hospital, Kyoto, Japan
| | - Jun-Ichiro Jo
- Laboratory of Biomaterials, Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Takashi Murakami
- Department of Microbiology, Saitama Medical University, Faculty of Medicine, Saitama, Japan
| | - Yasuhiko Tabata
- Laboratory of Biomaterials, Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Kenji Minatoya
- Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Jun K Yamashita
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Hidetoshi Masumoto
- Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Clinical Translational Research Program, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| |
Collapse
|
38
|
Imashiro C, Hirano M, Morikura T, Fukuma Y, Ohnuma K, Kurashina Y, Miyata S, Takemura K. Detachment of cell sheets from clinically ubiquitous cell culture vessels by ultrasonic vibration. Sci Rep 2020; 10:9468. [PMID: 32528073 PMCID: PMC7289836 DOI: 10.1038/s41598-020-66375-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 05/20/2020] [Indexed: 01/09/2023] Open
Abstract
Proteinases that digest the extracellular matrix are usually used to harvest cells from culture vessels in a general culture process, which lowers the initial adhesion rate in regenerative medicine. Cell sheet engineering is one of the most important technologies in this field, especially for transplantation, because fabricated cell sheets have rich extracellular matrixes providing strong initial adhesion. Current cell sheet fabrication relies on temperature-responsive polymer-coated dishes. Cells are cultured on such specialized dishes and subjected to low temperature. Thus, we developed a simple but versatile cell sheet fabrication method using ubiquitous culture dishes/flasks without any coating or temperature modulation. Confluent mouse myoblasts (C2C12 cell line) were exposed to ultrasonic vibration from underneath and detached as cell sheets from entire culture surfaces. Because of the absence of low temperature, cell metabolism was statically increased compared with the conventional method. Furthermore, viability, morphology, protein expression, and mRNA expression were normal. These analyses indicated no side effects of ultrasonic vibration exposure. Therefore, this novel method may become the standard for cell sheet fabrication. Our method can be easily conducted following a general culture procedure with a typical dish/flask, making cell sheets more accessible to medical experts.
Collapse
Affiliation(s)
- Chikahiro Imashiro
- Department of Mechanical Engineering, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama, 223-8522, Japan.,Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, TWIns, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, Japan
| | - Makoto Hirano
- Department of Pharmacy, Yasuda Women's University, 6-13-1 Yasuhigashi, Asaminami-ku, Hiroshima, Japan
| | - Takashi Morikura
- School of Science for Open and Environmental Systems, Graduate School of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama, 223-8522, Japan
| | - Yuki Fukuma
- School of Science for Open and Environmental Systems, Graduate School of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama, 223-8522, Japan
| | - Kiyoshi Ohnuma
- Department of Bioengineering, Nagaoka University of Technology, 1603-1 Kamitomioka, Nagaoka, Niigata, 940-2188, Japan.,Department of Science of Technology Innovation, Nagaoka University of Technology, 1603-1 Kamitomioka-cho, Nagaoka, Niigata, 940-2188, Japan
| | - Yuta Kurashina
- Department of Mechanical Engineering, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama, 223-8522, Japan.,Department of Materials Science and Engineering, School of Materials and Chemical Technology, Tokyo Institute of Technology, Yokohama, 226-8503, Japan
| | - Shogo Miyata
- Department of Mechanical Engineering, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama, 223-8522, Japan
| | - Kenjiro Takemura
- Department of Mechanical Engineering, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama, 223-8522, Japan.
| |
Collapse
|
39
|
Floy ME, Mateyka TD, Foreman KL, Palecek SP. Human pluripotent stem cell-derived cardiac stromal cells and their applications in regenerative medicine. Stem Cell Res 2020; 45:101831. [PMID: 32446219 PMCID: PMC7931507 DOI: 10.1016/j.scr.2020.101831] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 03/16/2020] [Accepted: 04/15/2020] [Indexed: 02/06/2023] Open
Abstract
Coronary heart disease is one of the leading causes of death in the United States. Recent advances in stem cell biology have led to the development and engineering of human pluripotent stem cell (hPSC)-derived cardiac cells and tissues for application in cellular therapy and cardiotoxicity studies. Initial studies in this area have largely focused on improving differentiation efficiency and maturation states of cardiomyocytes. However, other cell types in the heart, including endothelial and stromal cells, play crucial roles in cardiac development, injury response, and cardiomyocyte function. This review discusses recent advances in differentiation of hPSCs to cardiac stromal cells, identification and classification of cardiac stromal cell types, and application of hPSC-derived cardiac stromal cells and tissues containing these cells in regenerative and drug development applications.
Collapse
Affiliation(s)
- Martha E Floy
- Department of Chemical and Biological Engineering, University of Wisconsin - Madison, Madison, WI, USA
| | - Taylor D Mateyka
- Department of Chemical and Biological Engineering, University of Wisconsin - Madison, Madison, WI, USA
| | - Koji L Foreman
- Department of Chemical and Biological Engineering, University of Wisconsin - Madison, Madison, WI, USA
| | - Sean P Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin - Madison, Madison, WI, USA.
| |
Collapse
|
40
|
Nakamura K, Nobutani K, Shimada N, Tabata Y. Gelatin Hydrogel-Fragmented Fibers Suppress Shrinkage of Cell Sheet. Tissue Eng Part C Methods 2020; 26:216-224. [DOI: 10.1089/ten.tec.2019.0348] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Affiliation(s)
- Koichiro Nakamura
- Research and Development Center, The Japan Wool Textile Co., Ltd., Hyogo, Japan
- Laboratory of Biomaterials, Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Kimiaki Nobutani
- Research and Development Center, The Japan Wool Textile Co., Ltd., Hyogo, Japan
| | - Naoki Shimada
- Research and Development Center, The Japan Wool Textile Co., Ltd., Hyogo, Japan
| | - Yasuhiko Tabata
- Laboratory of Biomaterials, Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| |
Collapse
|
41
|
Stem cell-derived cell sheet transplantation for heart tissue repair in myocardial infarction. Stem Cell Res Ther 2020; 11:19. [PMID: 31915074 PMCID: PMC6950817 DOI: 10.1186/s13287-019-1536-y] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 10/30/2019] [Accepted: 12/19/2019] [Indexed: 12/11/2022] Open
Abstract
Stem cell-derived sheet engineering has been developed as the next-generation treatment for myocardial infarction (MI) and offers attractive advantages in comparison with direct stem cell transplantation and scaffold tissue engineering. Furthermore, induced pluripotent stem cell-derived cell sheets have been indicated to possess higher potential for MI therapy than other stem cell-derived sheets because of their capacity to form vascularized networks for fabricating thickened human cardiac tissue and their long-term therapeutic effects after transplantation in MI. To date, stem cell sheet transplantation has exhibited a dramatic role in attenuating cardiac dysfunction and improving clinical manifestations of heart failure in MI. In this review, we retrospectively summarized the current applications and strategy of stem cell-derived cell sheet technology for heart tissue repair in MI.
Collapse
|
42
|
Engineering of functional cardiac tubes by stepwise transplantation of cardiac cell sheets onto intestinal mesentery. Heart Vessels 2020; 35:859-867. [DOI: 10.1007/s00380-019-01550-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 12/20/2019] [Indexed: 01/01/2023]
|
43
|
Jang Y, Choi SC, Lim DS, Kim JH, Kim J, Park Y. Modulating cardiomyocyte and fibroblast interaction using layer-by-layer deposition facilitates synchronisation of cardiac macro tissues. SOFT MATTER 2020; 16:428-434. [PMID: 31799582 DOI: 10.1039/c9sm01531k] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Maturation and synchronisation of heart cells, including cardiomyocytes and fibroblasts, are essential to develop functional biomimetic cardiac tissues for regenerative medicine and drug discovery. Synchronisation of cells in the biomimetic cardiac tissue requires the structural integrity and functional maturation of cardiomyocytes with other cell types. However, it is challenging to synchronise the beating of macroscale cardiac tissues and induce maturation of cardiomyocytes derived from stem cells. Here, we developed a simple assembly technology to modulate cell-cell interactions by combining layer-by-layer (LBL) deposition and centrifugation of cells with collagen type I to control cell-cell interactions for the preparation of cardiac macro tissues (CMTs). We found that maturation of cardiomyocytes in CMTs was largely enhanced by growth factors FGF-4 and ascorbic acid, but synchronisation of cardiac beating required LBL deposition of cardiomyocytes and cardiac fibroblasts in addition to the growth factors during the maturation process. Our findings have important implications because incorporation of cardiac fibroblasts into the cardiomyocyte layer is a prerequisite for synchronised beating of macroscale cardiac tissues in addition to growth factors to facilitate maturation of stem cell-derived cardiomyocytes.
Collapse
Affiliation(s)
- Yongjun Jang
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, Korea.
| | | | | | | | | | | |
Collapse
|
44
|
Samak M, Hinkel R. Stem Cells in Cardiovascular Medicine: Historical Overview and Future Prospects. Cells 2019; 8:cells8121530. [PMID: 31783680 PMCID: PMC6952821 DOI: 10.3390/cells8121530] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 11/19/2019] [Accepted: 11/23/2019] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular diseases remain the leading cause of death in the developed world, accounting for more than 30% of all deaths. In a large proportion of these patients, acute myocardial infarction is usually the first manifestation, which might further progress to heart failure. In addition, the human heart displays a low regenerative capacity, leading to a loss of cardiomyocytes and persistent tissue scaring, which entails a morbid pathologic sequela. Novel therapeutic approaches are urgently needed. Stem cells, such as induced pluripotent stem cells or embryonic stem cells, exhibit great potential for cell-replacement therapy and an excellent tool for disease modeling, as well as pharmaceutical screening of novel drugs and their cardiac side effects. This review article covers not only the origin of stem cells but tries to summarize their translational potential, as well as potential risks and clinical translation.
Collapse
Affiliation(s)
- Mostafa Samak
- Department of Laboratory Animal Science, Leibnitz-Institut für Primatenforschung, Deutsches Primatenzentrum GmbH, Kellnerweg 4, 37077 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
| | - Rabea Hinkel
- Department of Laboratory Animal Science, Leibnitz-Institut für Primatenforschung, Deutsches Primatenzentrum GmbH, Kellnerweg 4, 37077 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
| |
Collapse
|
45
|
Shear Stress Promotes Arterial Endothelium-Oriented Differentiation of Mouse-Induced Pluripotent Stem Cells. Stem Cells Int 2019; 2019:1847098. [PMID: 31827524 PMCID: PMC6881757 DOI: 10.1155/2019/1847098] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 09/05/2019] [Accepted: 10/17/2019] [Indexed: 12/29/2022] Open
Abstract
Establishment of a functional vascular network, which is required in tissue repair and regeneration, needs large-scale production of specific arterial or venous endothelial cells (ECs) from stem cells. Previous in vitro studies by us and others revealed that shear stress induces EC differentiation of bone marrow-derived mesenchymal stem cells and embryonic stem cells. In this study, we focused on the impact of different magnitudes of shear stress on the differentiation of mouse-induced pluripotent stem cells (iPSCs) towards arterial or venous ECs. When iPSCs were exposed to shear stress (5, 10, and 15 dyne/cm2) with 50 ng/mL vascular endothelial growth factor and 10 ng/mL fibroblast growth factor, the expression levels of the general EC markers and the arterial markers increased, and the stress amplitude of 10 dyne/cm2 could be regarded as a proper promoter, whereas the venous and lymphatic markers had little or no expression. Further, shear stress caused cells to align parallel to the direction of the flow, induced cells forming functional tubes, and increased the secretion of nitric oxide. In addition, Notch1 was significantly upregulated, and the Notch ligand Delta-like 4 was activated in response to shear stress, while inhibition of Notch signaling by DAPT remarkably abolished the shear stress-induced arterial epithelium differentiation. Taken together, our results indicate that exposure to appropriate shear stress facilitated the differentiation of mouse iPSCs towards arterial ECs via Notch signaling pathways, which have potential applications for both disease modeling and regenerative medicine.
Collapse
|
46
|
Abstract
Myocardial infarction leads to an irreversible loss of vital myocardial cells. The transplantation of new cardiomyocytes into the heart was first described over 20 years ago and represents a straightforward approach to remuscularize a damaged heart. Due to the lack of human cells a clinical application seemed ambitious; however, dramatic progress in stem cell biology over the last two decades has paved the way towards a clinical application. This is especially important as the prognosis for patients with terminal heart failure is still poor. The transplantation of either cardiomyocytes or engineered heart tissue derived from pluripotent stem cells (either embryonic stem cells or induced pluripotent stem cells) might represent a new regenerative approach. Transplantation of either cells or tissue constructs has now been evaluated in several preclinical models, which have demonstrated that an injured heart can be (partially) remuscularized; however, major hurdles towards a clinical application are the transplantation-related occurrence of arrhythmia, the potential tumorigenicity of pluripotent cells and the required immunosuppression. Several groups are working hard to solve these problems and we are optimistic that the first clinical studies will take place within the next few years.
Collapse
|
47
|
Dwenger M, Kowalski WJ, Ye F, Yuan F, Tinney JP, Setozaki S, Nakane T, Masumoto H, Campbell P, Guido W, Keller BB. Chronic optical pacing conditioning of h-iPSC engineered cardiac tissues. J Tissue Eng 2019; 10:2041731419841748. [PMID: 31024681 PMCID: PMC6472158 DOI: 10.1177/2041731419841748] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 03/13/2019] [Indexed: 12/26/2022] Open
Abstract
The immaturity of human induced pluripotent stem cell derived engineered cardiac
tissues limits their ability to regenerate damaged myocardium and to serve as
robust in vitro models for human disease and drug toxicity
studies. Several chronic biomimetic conditioning protocols, including mechanical
stretch, perfusion, and/or electrical stimulation promote engineered cardiac
tissue maturation but have significant technical limitations. Non-contacting
chronic optical stimulation using heterologously expressed channelrhodopsin
light-gated ion channels, termed optogenetics, may be an advantageous
alternative to chronic invasive electrical stimulation for engineered cardiac
tissue conditioning. We designed proof-of-principle experiments to successfully
transfect human induced pluripotent stem cell derived engineered cardiac tissues
with a desensitization resistant, chimeric channelrhodopsin protein, and then
optically paced engineered cardiac tissues to accelerate maturation. We
transfected human induced pluripotent stem cell engineered cardiac tissues using
an adeno-associated virus packaged chimeric channelrhodopsin and then verified
optically paced by whole cell patch clamp. Engineered cardiac tissues were then
chronically optically paced above their intrinsic beat rates in
vitro from day 7 to 14. Chronically optically paced resulted in
improved engineered cardiac tissue electrophysiological properties and subtle
changes in the expression of some cardiac relevant genes, though active force
generation and histology were unchanged. These results validate the feasibility
of a novel chronically optically paced paradigm to explore non-invasive and
scalable optically paced–induced engineered cardiac tissue maturation
strategies.
Collapse
Affiliation(s)
- Marc Dwenger
- Kosair Charities Pediatric Heart Research Program, Cardiovascular Innovation Institute, University of Louisville, Louisville, KY, USA.,Department of Pharmacology & Toxicology, School of Medicine, University of Louisville, Louisville, KY, USA
| | - William J Kowalski
- Kosair Charities Pediatric Heart Research Program, Cardiovascular Innovation Institute, University of Louisville, Louisville, KY, USA.,Department of Pediatrics, School of Medicine, University of Louisville, Louisville, KY, USA.,Laboratory of Stem Cell and Neurovascular Biology, Cell and Developmental Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Fei Ye
- Kosair Charities Pediatric Heart Research Program, Cardiovascular Innovation Institute, University of Louisville, Louisville, KY, USA.,Department of Pediatrics, School of Medicine, University of Louisville, Louisville, KY, USA
| | - Fangping Yuan
- Kosair Charities Pediatric Heart Research Program, Cardiovascular Innovation Institute, University of Louisville, Louisville, KY, USA.,Department of Pediatrics, School of Medicine, University of Louisville, Louisville, KY, USA
| | - Joseph P Tinney
- Kosair Charities Pediatric Heart Research Program, Cardiovascular Innovation Institute, University of Louisville, Louisville, KY, USA.,Department of Pediatrics, School of Medicine, University of Louisville, Louisville, KY, USA
| | - Shuji Setozaki
- Kosair Charities Pediatric Heart Research Program, Cardiovascular Innovation Institute, University of Louisville, Louisville, KY, USA.,Department of Cardiovascular Surgery, Okamura Memorial Hospital, Shimizu, Japan
| | - Takeichiro Nakane
- Kosair Charities Pediatric Heart Research Program, Cardiovascular Innovation Institute, University of Louisville, Louisville, KY, USA.,Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Hidetoshi Masumoto
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan.,RIKEN Center for Biosystems Dynamics Research (BDR), Wako, Japan
| | - Peter Campbell
- Department of Anatomical Sciences and Neurobiology, School of Medicine, University of Louisville, Louisville, KY, USA
| | - William Guido
- Department of Anatomical Sciences and Neurobiology, School of Medicine, University of Louisville, Louisville, KY, USA
| | - Bradley B Keller
- Kosair Charities Pediatric Heart Research Program, Cardiovascular Innovation Institute, University of Louisville, Louisville, KY, USA.,Department of Pharmacology & Toxicology, School of Medicine, University of Louisville, Louisville, KY, USA.,Department of Pediatrics, School of Medicine, University of Louisville, Louisville, KY, USA
| |
Collapse
|
48
|
Jia Z, Guo H, Xie H, Zhou J, Wang Y, Bao X, Huang Y, Chen F. Construction of Pedicled Smooth Muscle Tissues by Combining the Capsule Tissue and Cell Sheet Engineering. Cell Transplant 2019; 28:328-342. [PMID: 30712374 PMCID: PMC6425107 DOI: 10.1177/0963689718821682] [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] [Indexed: 12/16/2022] Open
Abstract
The survival of engineered tissue requires the formation of its own capillary network, which can anastomose with the host vasculature after transplantation. Currently, while many strategies, such as modifying the scaffold material, adding endothelial cells, or angiogenic factors, have been researched, engineered tissue implanted in vivo cannot timely access to sufficient blood supply, leading to ischemic apoptosis or shrinkage. Constructing vascularized engineered tissue with its own axial vessels and subsequent pedicled transfer is promising to solve the problem of vascularization in tissue engineering. In this study, we used the tissue expander capsule as a novel platform for vascularizing autologous smooth muscle cell (SMC) sheets and fabricating vascularized engineered tissue with its own vascular pedicle. First, we verified which time point was the most effective for constructing an axial capsule vascular bed. Second, we compared the outcome of SMC sheet transplantation onto the expander capsule and classical dorsal subcutaneous tissue, which was widely used in other studies for vascularization. Finally, we transplanted multilayered SMC sheets onto the capsule bed twice to verify the feasibility of fabricating thick pedicled engineered smooth muscle tissues. The results indicated that the axial capsule tissue could be successfully induced, and the capsule tissue 1 week after full expansion was the most vascularized. Quantitative comparisons of thickness, vessel density, and apoptosis of cell sheet grafts onto two vascular beds proved that the axial capsule vascular bed was more favorable to the growth and vascularization of transplants than classical subcutaneous tissue. Furthermore, thick vascularized smooth muscle tissues with the vascular pedicle could be constructed by multi-transplanting cell sheets onto the capsule bed. The combination of axial capsule vascular bed and cell sheet engineering may provide an efficient strategy to overcome the problem of slow or insufficient vascularization in tissue engineering.
Collapse
Affiliation(s)
- Zhiming Jia
- 1 Department of Urology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Hailin Guo
- 1 Department of Urology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Hua Xie
- 1 Department of Urology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Junmei Zhou
- 2 Department of Central Laboratory, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yaping Wang
- 1 Department of Urology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xingqi Bao
- 1 Department of Urology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yichen Huang
- 1 Department of Urology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Fang Chen
- 1 Department of Urology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
49
|
Cell sheet technology: a promising strategy in regenerative medicine. Cytotherapy 2019; 21:3-16. [DOI: 10.1016/j.jcyt.2018.10.013] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 09/30/2018] [Accepted: 10/24/2018] [Indexed: 12/31/2022]
|
50
|
Wang BH, Liew D, Huang KW, Huang L, Tang W, Kelly DJ, Reid C, Liu Z. The Challenges of Stem Cell Therapy in Myocardial Infarction and Heart Failure and the Potential Strategies to Improve the Outcomes. ACTA ACUST UNITED AC 2018. [DOI: 10.1142/s1793984418410088] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Cardiovascular disease remains the single highest global cause of death and a significant financial burden on the healthcare system. Despite the advances in medical treatments, the prevalence and mortality for heart failure remain unacceptably high. New approaches are urgently needed to reduce this burden and improve patient outcomes and quality of life. One such promising approach is stem cell therapy, including embryonic stem cells, bone marrow derived stem cells, induced pluripotent stem cells and mesenchymal stem cells. However, the cardiac microenvironment following myocardial infarction poses huge challenges with inflammation, adequate retention, engraftment and functional incorporation all crucial concerns. The lack of cardiac regeneration, cell viability and functional improvement has hindered the success of stem cell therapy in clinical settings. The use of biomaterial scaffolds in conjunction with stem cells has recently been shown to enhance the outcome of stem cell therapy for heart failure and myocardial infarction. This review outlines some of the current challenges in the treatment of heart failure and acute myocardial infarction through improving stem cell therapeutic strategies, as well as the prospect of suitable biomaterial scaffolds to enhance their efficacy and improve patient clinical outcomes.
Collapse
Affiliation(s)
- Bing Hui Wang
- Monash Centre of Cardiovascular Research and Education in Therapeutics, School of Public Health and Preventive Medicine, Monash University, Melbourne 3004, Australia
| | - Danny Liew
- Monash Centre of Cardiovascular Research and Education in Therapeutics, School of Public Health and Preventive Medicine, Monash University, Melbourne 3004, Australia
| | - Kevin W. Huang
- Monash Centre of Cardiovascular Research and Education in Therapeutics, School of Public Health and Preventive Medicine, Monash University, Melbourne 3004, Australia
| | - Li Huang
- Monash Centre of Cardiovascular Research and Education in Therapeutics, School of Public Health and Preventive Medicine, Monash University, Melbourne 3004, Australia
| | - Wenjie Tang
- Department of Cardiovascular and Thoracic Surgery, Research Center for Translational Medicine and Biomedical Multidisciplinary Innovation Research Institute, Shanghai East Hospital, Tongji University, Shanghai 200120, P. R. China
| | - Darren J. Kelly
- Department of Medicine, St Vincent’s Hospital, University of Melbourne, Fitzroy Victoria, Australia
| | - Christopher Reid
- Monash Centre of Cardiovascular Research and Education in Therapeutics, School of Public Health and Preventive Medicine, Monash University, Melbourne 3004, Australia
| | - Zhongmin Liu
- Department of Cardiovascular and Thoracic Surgery, Research Center for Translational Medicine and Biomedical Multidisciplinary Innovation Research Institute, Shanghai East Hospital, Tongji University, Shanghai 200120, P. R. China
| |
Collapse
|