1
|
Punde A, Rayrikar A, Maity S, Patra C. Extracellular matrix in cardiac morphogenesis, fibrosis, and regeneration. Cells Dev 2025:204023. [PMID: 40154789 DOI: 10.1016/j.cdev.2025.204023] [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: 10/17/2024] [Revised: 03/14/2025] [Accepted: 03/22/2025] [Indexed: 04/01/2025]
Abstract
The extracellular matrix (ECM) plays a crucial role in providing structural integrity and regulating cell communication essential for organ development, homeostasis, and regeneration, including hearts. Evidence indicates that disruptions in the spatiotemporal expression or alterations in ECM components lead to cardiac malformations, including a wide range of congenital heart diseases (CHDs). Furthermore, research on injured hearts across various vertebrate species, some of which show effective regeneration while others experience irreversible fibrosis, underscores the significance of ECM molecules in cardiac regeneration. This review presents an overview of heart development and the dynamics of ECM during cardiac morphogenesis, beginning with the formation of the contractile heart tube and advancing to the development of distinct chambers separated by valves to facilitate unidirectional blood flow. Furthermore, we discuss research emphasizing the multifaceted roles of secreted molecules in mediating fibrosis and regeneration following myocardial injury.
Collapse
Affiliation(s)
- Ashwini Punde
- Department of Developmental Biology, Agharkar Research Institute, Pune, Maharashtra, 411004, India
| | - Amey Rayrikar
- Department of Developmental Biology, Agharkar Research Institute, Pune, Maharashtra, 411004, India
| | - Shreya Maity
- Department of Developmental Biology, Agharkar Research Institute, Pune, Maharashtra, 411004, India
| | - Chinmoy Patra
- Department of Developmental Biology, Agharkar Research Institute, Pune, Maharashtra, 411004, India.
| |
Collapse
|
2
|
Hamze J, Broadwin M, Stone C, Muir KC, Sellke FW, Abid MR. Developments in Extracellular Matrix-Based Angiogenesis Therapy for Ischemic Heart Disease: A Review of Current Strategies, Methodologies and Future Directions. BIOTECH 2025; 14:23. [PMID: 40227326 PMCID: PMC11940646 DOI: 10.3390/biotech14010023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 04/15/2025] Open
Abstract
Ischemic heart disease (IHD) is the leading cause of mortality worldwide, underscoring the urgent need for innovative therapeutic strategies. The cardiac extracellular matrix (ECM) undergoes extreme transformations during IHD, adversely influencing the heart's structure, mechanics, and cellular signaling. Researchers investigating the regenerative capacity of the diseased heart have turned their attention to exploring the modulation of ECM to improve therapeutic outcomes. In this review, we thoroughly examine the current state of knowledge regarding the cardiac ECM and its therapeutic potential in the ischemic myocardium. We begin by providing an overview of the fundamentals of cardiac ECM, focusing on the structural, functional, and regulatory mechanisms that drive its modulation. Subsequently, we examine the ECM's interactions within both chronically ischemic and acutely infarcted myocardium, emphasizing key ECM components and their roles in modulating angiogenesis. Finally, we discuss recent ECM-based approaches in biomedical engineering, focusing on different types of scaffolds as delivery tools and their compositions, and conclude with future directions for therapeutic research. By harnessing the potential of these emerging ECM-based therapies, we aim to contribute to the development of novel therapeutic modalities for IHD.
Collapse
Affiliation(s)
| | | | | | | | | | - M. Ruhul Abid
- Division of Cardiothoracic Surgery, Cardiovascular Research Center, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI 02903, USA
| |
Collapse
|
3
|
Holme S, Richardson SM, Bella J, Pinali C. Hydrogels for Cardiac Tissue Regeneration: Current and Future Developments. Int J Mol Sci 2025; 26:2309. [PMID: 40076929 PMCID: PMC11900105 DOI: 10.3390/ijms26052309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/23/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Myocardial infarction remains a leading cause of death worldwide due to the heart's limited regenerative capability and the current lack of viable therapeutic solutions. Therefore, there is an urgent need to develop effective treatment options to restore cardiac function after a heart attack. Stem cell-derived cardiac cells have been extensively utilised in cardiac tissue regeneration studies. However, the use of Matrigel as a substrate for the culture and maturation of these cells has been a major limitation for the translation of this research into clinical application. Hydrogels are emerging as a promising system to overcome this problem. They are biocompatible and can provide stem cells with a supportive scaffold that mimics the extracellular matrix, which is essential for repairing damaged tissue in the myocardium after an infarction. Thus, hydrogels provide an alternative and reproducible option in addressing myocardial infarction due to their unique potential therapeutic benefits. This review explores the different types of natural and synthetic polymers used to create hydrogels and their various delivery methods, the most common being via injection and cardiac patches and other applications such as bioprinting. Many challenges remain before hydrogels can be used in a clinical setting, but they hold great promise for the future of cardiac tissue regeneration.
Collapse
Affiliation(s)
- Sonja Holme
- Division of Cell Matrix Biology & Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK; (S.H.); (S.M.R.)
| | - Stephen M. Richardson
- Division of Cell Matrix Biology & Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK; (S.H.); (S.M.R.)
| | - Jordi Bella
- Division of Cell Matrix Biology & Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK; (S.H.); (S.M.R.)
| | - Christian Pinali
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9NT, UK
| |
Collapse
|
4
|
Boodagh P, De Mohac LM, Hayashi Y, Vella D, Ye SH, Cosentino F, Fujii T, Gorge E, Coyan G, Soto JDL, Burriesci G, Wagner WR, D'Amore A. Photooxidation Cross-Linked, Glutaraldehyde Cross-Linked, or Enzyme and Hydrostatic Pressure Processed Decellularized Biomaterials for Cardiovascular Repair Do Not Affect Host Response in a Rat Right Ventricular Outflow Flow Tract Reconstruction (RVOT) Model. J Biomed Mater Res B Appl Biomater 2025; 113:e35529. [PMID: 39853946 DOI: 10.1002/jbm.b.35529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/06/2024] [Accepted: 12/26/2024] [Indexed: 01/26/2025]
Abstract
Cardiovascular diseases (CVDs) were responsible for approximately 19 million deaths in 2020, marking an increase of 18.7% since 2010. Biological decellularized patches are common therapeutic solutions for CVD such as cardiac and valve defects. The preparation of biomaterials for cardiac patches involves two main processing methods: glutaraldehyde or photooxidation cross-linking (fixation) and noncross-linked (nonfixation) processing. Despite the variety of products available in the market, cardiac patches still suffer from significant limitations, failing to adequately mimic the properties of biological tissue and restore its function. This study assesses the impact of different processing methodologies on the biological and biomechanical outcomes of three commercially available cardiac patches (CorPatch, CardioCel, PhotoFix) and one newly developed decellularized cardiac patch (Adeka) when implanted as right ventricular outflow tract (RVOT) repair material on a rat model. Four different patches for cardiovascular repair were selected based on their processing approaches and included: photooxidation crosslinked (PhotoFix), glutaraldehyde crosslinked (CardioCel), noncross-linked small intestine submucosa (CorPatch) or enzyme, and hydrostatic pressure (Adeka) processed decellularized biomaterials. Structure and function were characterized prior to implantation via thickness mapping, cross-section morphology, 2D surface topography, 3D volume microstructure, biaxial testing, uniaxial tensile testing, ball burst, and suture retention. Their host-biomaterials response was assessed in vivo using a relevant model for cardiovascular repair: a rat (RVOT) reconstruction with 8 and 16-week timepoints. Topological analysis showed that the crosslinked cardiac patches had a more homogeneous thickness distribution when compared to the noncrosslinked patches. This agreed with histological evaluation, where cross-linking processed materials better preserved collagen content than noncrosslinked patches who were also more delaminated. Biaxial data demonstrated that all patches, except CorPatch, recapitulated the anisotropic behavior of healthy left ventricle tissue. The Adeka patch in-plane mechanics at 16 weeks was the one who better resembled the mechanics of healthy cardiac tissue. All patches showed appropriate biocompatibility and function at 8- and 16-week timepoints for RVOT patching. This included echocardiographic assessment, biomechanics, macrophage infiltration and polarization, and angiogenesis. Consistently with a more porous laminae structure, explants histology showed higher cell infiltration in non-crosslinked Adeka when compared to the crosslinked PhotoFix. Overall, both in vitro and in vivo tests indicate that the material processing does not impact the function, biomechanical performance, and the host response of the patches that can be considered as equally effective as materials based cardiac repair solutions.
Collapse
Affiliation(s)
- Parnaz Boodagh
- McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania, USA
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | - Yasurani Hayashi
- McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania, USA
| | | | - Sang-Ho Ye
- McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania, USA
| | | | - Taro Fujii
- McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania, USA
| | - Emily Gorge
- McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania, USA
| | - Garrett Coyan
- McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania, USA
| | | | - Gaetano Burriesci
- Ri.MED Foundation, Palermo, Italy
- UCL Mechanical Engineering, University College London, London, UK
- University of Palermo, Department of Engineering, Viale delle Scienze, Palermo, Italy
| | - William R Wagner
- McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania, USA
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Chemical and Petroleum Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Antonio D'Amore
- McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania, USA
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Ri.MED Foundation, Palermo, Italy
- Clinical Translational Science Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Precision Medicine Me.Pre.C.C, University of Palermo, Palermo, Italy
- Department of Medicina di Precisione in Area Medica, Chirurgica e Critica, University of Palermo, Via Liborio Giuffrè, Palermo, Italy
| |
Collapse
|
5
|
Arıcı Ş, Kamali AR, Ege D. CMC/Gel/GO 3D-printed cardiac patches: GO and CMC improve flexibility and promote H9C2 cell proliferation, while EDC/NHS enhances stability. Biofabrication 2024; 17:015025. [PMID: 39496201 DOI: 10.1088/1758-5090/ad8e87] [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: 05/31/2024] [Accepted: 11/04/2024] [Indexed: 11/06/2024]
Abstract
In this research, carboxymethyl cellulose (CMC)/gelatin (Gel)/graphene oxide (GO)-based scaffolds were produced by using extrusion-based 3D printing for cardiac tissue regeneration. Rheological studies were conducted to evaluate the printability of CMC/Gel/GO inks, which revealed that CMC increased viscosity and enhanced printability. The 3D-printed cardiac patches were crosslinked with N-(3-dimethylaminopropyl)-n'-ethylcarbodiimide hydrochloride (EDC)/N-hydroxysuccinimide (NHS) (100:20 mM, 50:10 mM, 25:5 mM) and then characterized by mechanical analysis, electrical conductivity testing, contact angle measurements and degradation studies. Subsequently, cell culture studies were conducted to evaluate the viability of H9C2 cardiomyoblast cells by using the Alamar Blue assay and fluorescence imaging. A high concentration of EDC/NHS (100:20 mM) led to the stability of the patches; however, it drastically reduced the flexibility of the scaffolds. Conversely, a concentration of 25:5 mM resulted in flexible but unstable scaffolds in phosphate buffer saline solution. The suitable EDC/NHS concentration was found to be 50:10 mM, as it produced flexible, stable, and stiff cardiac scaffolds with high ultimate tensile strength. Mechanical characterization revealed that % strain at break of C15/G7.5/GO1 exhibited a remarkable increase of 61.03% compared to C15/G7.5 samples. The improvement of flexibility was attributed to the hydrogen bonding between CMC, Gel and GO. The electrical conductivity of 3D printed CMC/Gel/GO cardiac patches was 7.0 × 10-3S cm-1, demonstrating suitability for mimicking the desired electrical conductivity of human myocardium. The incorporation of 1 wt% of GO and addition of CMC concentration from 7.5 wt% to 15 wt% significantly enhanced relative % cell viability. Overall, although this research is at its infancy, CMC/Gel/GO cardiac patches have potential to improve the physiological function of cardiac tissue.
Collapse
Affiliation(s)
- Şule Arıcı
- Institute of Biomedical Engineering, Boğaziçi University, Rasathane Cd. Kandilli Campus, Kandilli Mah., 34684 Istanbul, Turkey
| | - Ali Reza Kamali
- Energy and Environmental Materials Research Centre (E2MC), School of Metallurgy, Northeastern University, 110819 Shenyang, People's Republic of China
| | - Duygu Ege
- Institute of Biomedical Engineering, Boğaziçi University, Rasathane Cd. Kandilli Campus, Kandilli Mah., 34684 Istanbul, Turkey
| |
Collapse
|
6
|
Carton F, Rizzi M, Canciani E, Sieve G, Di Francesco D, Casarella S, Di Nunno L, Boccafoschi F. Use of Hydrogels in Regenerative Medicine: Focus on Mechanical Properties. Int J Mol Sci 2024; 25:11426. [PMID: 39518979 PMCID: PMC11545898 DOI: 10.3390/ijms252111426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 10/16/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Bioengineered materials represent an innovative option to support the regenerative processes of damaged tissues, with the final objective of creating a functional environment closely mimicking the native tissue. Among the different available biomaterials, hydrogels represent the solution of choice for tissue regeneration, thanks to the easy synthesis process and the highly tunable physical and mechanical properties. Moreover, hydrogels are biocompatible and biodegradable, able to integrate in biological environments and to support cellular interactions in order to restore damaged tissues' functionality. This review offers an overview of the current knowledge concerning hydrogel synthesis and characterization and of the recent achievements in their experimental use in supporting skin, bone, cartilage, and muscle regeneration. The currently available in vitro and in vivo results are of great interest, highlighting the need for carefully designed and controlled preclinical studies and clinical trials to support the transition of these innovative biomaterials from the bench to the bedside.
Collapse
Affiliation(s)
- Flavia Carton
- Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, Italy (E.C.); (S.C.)
| | - Manuela Rizzi
- Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, Italy (E.C.); (S.C.)
| | - Elena Canciani
- Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, Italy (E.C.); (S.C.)
| | - Gianluca Sieve
- Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, Italy (E.C.); (S.C.)
| | - Dalila Di Francesco
- Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, Italy (E.C.); (S.C.)
- Laboratory for Biomaterials and Bioengineering, CRC-I, Department of Min-Met-Materials Engineering, University Hospital Research Center, Regenerative Medicine, Laval University, Quebec City, QC G1V 0A6, Canada
| | - Simona Casarella
- Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, Italy (E.C.); (S.C.)
| | - Luca Di Nunno
- Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, Italy (E.C.); (S.C.)
- Laboratory for Biomaterials and Bioengineering, CRC-I, Department of Min-Met-Materials Engineering, University Hospital Research Center, Regenerative Medicine, Laval University, Quebec City, QC G1V 0A6, Canada
| | - Francesca Boccafoschi
- Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, Italy (E.C.); (S.C.)
| |
Collapse
|
7
|
Qiao A, Wei Y, Liu Y, Kahn-Krell A, Ye L, Nguyen T, Zhang J. Doxycycline-Mediated Control of Cyclin D2 Overexpression in Human-Induced Pluripotent Stem Cells. Int J Mol Sci 2024; 25:8714. [PMID: 39201401 PMCID: PMC11354523 DOI: 10.3390/ijms25168714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 07/24/2024] [Accepted: 07/29/2024] [Indexed: 09/02/2024] Open
Abstract
Previous studies have demonstrated that when the cyclin D2 (CCND2), a cell-cycle regulatory protein, is overexpressed in human-induced pluripotent stem cells (hiPSCs), cardiomyocytes (CMs) differentiated from these CCND2-overexpressing hiPSCs can proliferate after transplantation into infarcted hearts, which significantly improves the cells' potency for myocardial regeneration. However, persistent CM proliferation could lead to tumor growth or the development of arrhythmogenic complications; thus, the goal of the current study was to generate a line of hiPSCs in which CCND2 overexpression could be tightly controlled. First, we transfected hiPSCs with vectors coding for a doxycycline-inducible Tet-On transactivator and S. pyogenes dCas9 fused to the VPR activation domain; then, the same hiPSCs were engineered to express guide RNAs targeting the CCND2 promotor. Thus, treatment with doxycycline (dox) activated dCas9-VPR expression, and the guide RNAs directed dCas9-VPR to the CCND2 promoter, which activated CCND2 expression. Subsequent experiments confirmed that CCND2 expression was dox-dependent in this newly engineered line of hiPSCs (doxCCND2-hiPSCs): CCND2 protein was abundantly expressed after 48 h of treatment with dox and declined to near baseline level ~96 h after dox treatment was discontinued.
Collapse
Affiliation(s)
- Aijun Qiao
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (A.Q.); (Y.W.); (Y.L.); (A.K.-K.); (L.Y.); (T.N.)
| | - Yuhua Wei
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (A.Q.); (Y.W.); (Y.L.); (A.K.-K.); (L.Y.); (T.N.)
| | - Yanwen Liu
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (A.Q.); (Y.W.); (Y.L.); (A.K.-K.); (L.Y.); (T.N.)
| | - Asher Kahn-Krell
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (A.Q.); (Y.W.); (Y.L.); (A.K.-K.); (L.Y.); (T.N.)
| | - Lei Ye
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (A.Q.); (Y.W.); (Y.L.); (A.K.-K.); (L.Y.); (T.N.)
| | - Thanh Nguyen
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (A.Q.); (Y.W.); (Y.L.); (A.K.-K.); (L.Y.); (T.N.)
| | - Jianyi Zhang
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (A.Q.); (Y.W.); (Y.L.); (A.K.-K.); (L.Y.); (T.N.)
- Department of Medicine/Cardiovascular Diseases, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
8
|
Gao H, Huang X, Chen W, Feng Z, Zhao Z, Li P, Tan C, Wang J, Zhuang Q, Gao Y, Min S, Yao Q, Qian M, Ma X, Wu F, Yan W, Sheng W, Huang G. Association of copy number variation in X chromosome-linked PNPLA4 with heterotaxy and congenital heart disease. Chin Med J (Engl) 2024; 137:1823-1834. [PMID: 38973237 PMCID: PMC12077557 DOI: 10.1097/cm9.0000000000003192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Indexed: 07/09/2024] Open
Abstract
BACKGROUND Heterotaxy (HTX) is a thoracoabdominal organ anomaly syndrome and commonly accompanied by congenital heart disease (CHD). The aim of this study was to analyze rare copy number variations (CNVs) in a HTX/CHD cohort and to examine the potential mechanisms contributing to HTX/CHD. METHODS Chromosome microarray analysis was used to identify rare CNVs in a cohort of 120 unrelated HTX/CHD patients, and available samples from parents were used to confirm the inheritance pattern. Potential candidate genes in CNVs region were prioritized via the DECIPHER database, and PNPLA4 was identified as the leading candidate gene. To validate, we generated PNPLA4 -overexpressing human induced pluripotent stem cell lines as well as pnpla4 -overexpressing zebrafish model, followed by a series of transcriptomic, biochemical and cellular analyses. RESULTS Seventeen rare CNVs were identified in 15 of the 120 HTX/CHD patients (12.5%). Xp22.31 duplication was one of the inherited CNVs identified in this HTX/CHD cohort, and PNPLA4 in the Xp22.31 was a candidate gene associated with HTX/CHD. PNPLA4 is expressed in the lateral plate mesoderm, which is known to be critical for left/right embryonic patterning as well as cardiomyocyte differentiation, and in the neural crest cell lineage. Through a series of in vivo and in vitro analyses at the molecular and cellular levels, we revealed that the biological function of PNPLA4 is importantly involved in the primary cilia formation and function via its regulation of energy metabolism and mitochondria-mediated ATP production. CONCLUSIONS Our findings demonstrated a significant association between CNVs and HTX/CHD. Our data strongly suggested that an increased genetic dose of PNPLA4 due to Xp22.31 duplication is a disease-causing risk factor for HTX/CHD.
Collapse
Affiliation(s)
- Han Gao
- Children’s Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
| | - Xianghui Huang
- Fujian Key Laboratory of Neonatal Diseases, Xiamen Children’s Hospital, Xiamen, Fujian 361006, China
| | - Weicheng Chen
- Children’s Hospital of Fudan University, Shanghai 201102, China
| | - Zhiyu Feng
- Children’s Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
| | - Zhengshan Zhao
- Children’s Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
| | - Ping Li
- Children’s Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
| | - Chaozhong Tan
- Children’s Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
| | - Jinxin Wang
- Children’s Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
| | - Quannan Zhuang
- Children’s Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
| | - Yuan Gao
- Children’s Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
| | - Shaojie Min
- Children’s Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
| | - Qinyu Yao
- Children’s Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
| | - Maoxiang Qian
- Children’s Hospital of Fudan University, Shanghai 201102, China
| | - Xiaojing Ma
- Children’s Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
| | - Feizhen Wu
- Children’s Hospital of Fudan University, Shanghai 201102, China
| | - Weili Yan
- Children’s Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
- Research Unit of Early Intervention of Genetically Related Childhood Cardiovascular Diseases, Chinese Academy of Medical Sciences, Shanghai 201102, China
| | - Wei Sheng
- Children’s Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
- Fujian Key Laboratory of Neonatal Diseases, Xiamen Children’s Hospital, Xiamen, Fujian 361006, China
- Research Unit of Early Intervention of Genetically Related Childhood Cardiovascular Diseases, Chinese Academy of Medical Sciences, Shanghai 201102, China
| | - Guoying Huang
- Children’s Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
- Fujian Key Laboratory of Neonatal Diseases, Xiamen Children’s Hospital, Xiamen, Fujian 361006, China
- Research Unit of Early Intervention of Genetically Related Childhood Cardiovascular Diseases, Chinese Academy of Medical Sciences, Shanghai 201102, China
| |
Collapse
|
9
|
Cyr JA, Burdett C, Pürstl JT, Thompson RP, Troughton SC, Sinha S, Best SM, Cameron RE. Characterizing collagen scaffold compliance with native myocardial strains using an ex-vivo cardiac model: The physio-mechanical influence of scaffold architecture and attachment method. Acta Biomater 2024; 184:239-253. [PMID: 38942187 DOI: 10.1016/j.actbio.2024.06.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 05/30/2024] [Accepted: 06/21/2024] [Indexed: 06/30/2024]
Abstract
Applied to the epicardium in-vivo, regenerative cardiac patches support the ventricular wall, reduce wall stresses, encourage ventricular wall thickening, and improve ventricular function. Scaffold engraftment, however, remains a challenge. After implantation, scaffolds are subject to the complex, time-varying, biomechanical environment of the myocardium. The mechanical capacity of engineered tissue to biomimetically deform and simultaneously support the damaged native tissue is crucial for its efficacy. To date, however, the biomechanical response of engineered tissue applied directly to live myocardium has not been characterized. In this paper, we utilize optical imaging of a Langendorff ex-vivo cardiac model to characterize the native deformation of the epicardium as well as that of attached engineered scaffolds. We utilize digital image correlation, linear strain, and 2D principal strain analysis to assess the mechanical compliance of acellular ice templated collagen scaffolds. Scaffolds had either aligned or isotropic porous architecture and were adhered directly to the live epicardial surface with either sutures or cyanoacrylate glue. We demonstrate that the biomechanical characteristics of native myocardial deformation on the epicardial surface can be reproduced by an ex-vivo cardiac model. Furthermore, we identified that scaffolds with unidirectionally aligned pores adhered with suture fixation most accurately recapitulated the deformation of the native epicardium. Our study contributes a translational characterization methodology to assess the physio-mechanical performance of engineered cardiac tissue and adds to the growing body of evidence showing that anisotropic scaffold architecture improves the functional biomimetic capacity of engineered cardiac tissue. STATEMENT OF SIGNIFICANCE: Engineered cardiac tissue offers potential for myocardial repair, but engraftment remains a challenge. In-vivo, engineered scaffolds are subject to complex biomechanical stresses and the mechanical capacity of scaffolds to biomimetically deform is critical. To date, the biomechanical response of engineered scaffolds applied to live myocardium has not been characterized. In this paper, we utilize optical imaging of an ex-vivo cardiac model to characterize the deformation of the native epicardium and scaffolds attached directly to the heart. Comparing scaffold architecture and fixation method, we demonstrate that sutured scaffolds with anisotropic pores aligned with the native alignment of the superficial myocardium best recapitulate native deformation. Our study contributes a physio-mechanical characterization methodology for cardiac tissue engineering scaffolds.
Collapse
Affiliation(s)
- Jamie A Cyr
- Department of Materials Science & Metallurgy Cambridge University 27 Charles Babbage Road, Cambridge CB3 0FS, UK.
| | - Clare Burdett
- Department of Materials Science & Metallurgy Cambridge University 27 Charles Babbage Road, Cambridge CB3 0FS, UK
| | - Julia T Pürstl
- Department of Materials Science & Metallurgy Cambridge University 27 Charles Babbage Road, Cambridge CB3 0FS, UK
| | - Robert P Thompson
- Department of Materials Science & Metallurgy Cambridge University 27 Charles Babbage Road, Cambridge CB3 0FS, UK
| | - Samuel C Troughton
- Department of Materials Science & Metallurgy Cambridge University 27 Charles Babbage Road, Cambridge CB3 0FS, UK
| | - Sanjay Sinha
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge University, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Serena M Best
- Department of Materials Science & Metallurgy Cambridge University 27 Charles Babbage Road, Cambridge CB3 0FS, UK.
| | - Ruth E Cameron
- Department of Materials Science & Metallurgy Cambridge University 27 Charles Babbage Road, Cambridge CB3 0FS, UK.
| |
Collapse
|
10
|
Min S, Kim S, Sim WS, Choi YS, Joo H, Park JH, Lee SJ, Kim H, Lee MJ, Jeong I, Cui B, Jo SH, Kim JJ, Hong SB, Choi YJ, Ban K, Kim YG, Park JU, Lee HA, Park HJ, Cho SW. Versatile human cardiac tissues engineered with perfusable heart extracellular microenvironment for biomedical applications. Nat Commun 2024; 15:2564. [PMID: 38519491 PMCID: PMC10960018 DOI: 10.1038/s41467-024-46928-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 03/13/2024] [Indexed: 03/25/2024] Open
Abstract
Engineered human cardiac tissues have been utilized for various biomedical applications, including drug testing, disease modeling, and regenerative medicine. However, the applications of cardiac tissues derived from human pluripotent stem cells are often limited due to their immaturity and lack of functionality. Therefore, in this study, we establish a perfusable culture system based on in vivo-like heart microenvironments to improve human cardiac tissue fabrication. The integrated culture platform of a microfluidic chip and a three-dimensional heart extracellular matrix enhances human cardiac tissue development and their structural and functional maturation. These tissues are comprised of cardiovascular lineage cells, including cardiomyocytes and cardiac fibroblasts derived from human induced pluripotent stem cells, as well as vascular endothelial cells. The resultant macroscale human cardiac tissues exhibit improved efficacy in drug testing (small molecules with various levels of arrhythmia risk), disease modeling (Long QT Syndrome and cardiac fibrosis), and regenerative therapy (myocardial infarction treatment). Therefore, our culture system can serve as a highly effective tissue-engineering platform to provide human cardiac tissues for versatile biomedical applications.
Collapse
Affiliation(s)
- Sungjin Min
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Suran Kim
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
- Cellartgen, Seoul, 03722, Republic of Korea
| | - Woo-Sup Sim
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Yi Sun Choi
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Hyebin Joo
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Jae-Hyun Park
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Su-Jin Lee
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea
| | - Hyeok Kim
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Mi Jeong Lee
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Inhea Jeong
- Department of Materials Science and Engineering, Yonsei University, Seoul, 03722, Republic of Korea
| | - Baofang Cui
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Sung-Hyun Jo
- Department of Chemical Engineering, Soongsil University, Seoul, 06978, Republic of Korea
| | - Jin-Ju Kim
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Seok Beom Hong
- Department of Thoracic and Cardiovascular Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Yeon-Jik Choi
- Division of Cardiology, Department of Internal Medicine, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 03312, Republic of Korea
| | - Kiwon Ban
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, 999077, Hong Kong
| | - Yun-Gon Kim
- Department of Chemical Engineering, Soongsil University, Seoul, 06978, Republic of Korea
| | - Jang-Ung Park
- Department of Materials Science and Engineering, Yonsei University, Seoul, 03722, Republic of Korea
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, 03722, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul, 03722, Republic of Korea
| | - Hyang-Ae Lee
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea
| | - Hun-Jun Park
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
- Cell Death Disease Research Center, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea.
- Cellartgen, Seoul, 03722, Republic of Korea.
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, 03722, Republic of Korea.
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul, 03722, Republic of Korea.
| |
Collapse
|
11
|
Katili PA, Karima AP, Azwani W, Antarianto RD, Djer MM. Application of Human Induced Pluripotent Stem Cells for Tissue Engineered Cardiomyocyte Modelling. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2023; 9:431-446. [DOI: 10.1007/s40883-023-00294-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 01/02/2023] [Accepted: 01/06/2023] [Indexed: 01/06/2025]
Abstract
Abstract
Purpose
Cardiac
tissue engineering opens up opportunities for regenerative therapy in heart diseases. Current technologies improve engineered cardiac tissue characteristics by combining human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) with non-cardiomyocytes, selective biomaterials, and additional growth factors. Animal models are still required to determine cardiac patches’ overall in vivo effect before initiating human trials. Here, we review the current in vivo studies of cardiac patches using hiPSC-CMs.
Methods
We performed a literature search for studies on cardiac patch in vivo application and compared outcomes based on cell engraftment, functional changes, and safety profiles.
Results
Present studies confirm the beneficial results of combining hiPSC-CMs with other cardiac cell lineages and biomaterials. They improved the functional capacity of the heart, showed a reduction in infarct size, and initiated an adaptive inflammatory process through neovascularisation.
Conclusion
The cardiac patch is currently the most effective delivery system, proving safety and improvements in animal models, which are suggested to be the role of the paracrine mechanism. Further studies should focus on honing in vitro patch characteristics to achieve ideal results.
Lay Summary
Cardiac tissue engineering answers the demand for regenerative therapy in heart diseases. Combining human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) with biomaterials and growth factors in cardiac patches improves the heart’s structural and functional characteristics. This delivery system is safe and efficient for delivering many cells and minimising cellular loss in vivo. Rat and porcine models of ischemic and non-ischemic heart diseases demonstrated the benefits of this therapy, which include cell engraftment, reduced infarct size, and increased left ventricular (LV) systolic function, with no reported critical adverse events. These reports sufficiently provide evidence of feasible improvements to proceed towards further trials.
Collapse
|
12
|
Cyr JA, Colzani M, Bayraktar S, Köhne M, Bax DV, Graup V, Farndale R, Sinha S, Best SM, Cameron RE. Extracellular macrostructure anisotropy improves cardiac tissue-like construct function and phenotypic cellular maturation. BIOMATERIALS ADVANCES 2023; 155:213680. [PMID: 37944449 DOI: 10.1016/j.bioadv.2023.213680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 10/02/2023] [Accepted: 10/30/2023] [Indexed: 11/12/2023]
Abstract
Regenerative cardiac tissue is a promising field of study with translational potential as a therapeutic option for myocardial repair after injury, however, poor electrical and contractile function has limited translational utility. Emerging research suggests scaffolds that recapitulate the structure of the native myocardium improve physiological function. Engineered cardiac constructs with anisotropic extracellular architecture demonstrate improved tissue contractility, signaling synchronicity, and cellular organization when compared to constructs with reduced architectural order. The complexity of scaffold fabrication, however, limits isolated variation of individual structural and mechanical characteristics. Thus, the isolated impact of scaffold macroarchitecture on tissue function is poorly understood. Here, we produce isotropic and aligned collagen scaffolds seeded with embryonic stem cell derived cardiomyocytes (hESC-CM) while conserving all confounding physio-mechanical features to independently assess the effects of macroarchitecture on tissue function. We quantified spatiotemporal tissue function through calcium signaling and contractile strain. We further examined intercellular organization and intracellular development. Aligned tissue constructs facilitated improved signaling synchronicity and directional contractility as well as dictated uniform cellular alignment. Cells on aligned constructs also displayed phenotypic and genetic markers of increased maturity. Our results isolate the influence of scaffold macrostructure on tissue function and inform the design of optimized cardiac tissue for regenerative and model medical systems.
Collapse
Affiliation(s)
- Jamie A Cyr
- Department of Materials Science & Metallurgy, Cambridge University, 27 Charles Babbage Road, Cambridge CB3 0FS, UK
| | - Maria Colzani
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge University, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Semih Bayraktar
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge University, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Maria Köhne
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge University, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Daniel V Bax
- Department of Materials Science & Metallurgy, Cambridge University, 27 Charles Babbage Road, Cambridge CB3 0FS, UK
| | - Vera Graup
- Department of Materials Science & Metallurgy, Cambridge University, 27 Charles Babbage Road, Cambridge CB3 0FS, UK
| | - Richard Farndale
- Department of Biochemistry, Cambridge University, Hopkins Building Tennis Court Road, Cambridge CB2 1QW, UK
| | - Sanjay Sinha
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge University, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge CB2 0AW, UK.
| | - Serena M Best
- Department of Materials Science & Metallurgy, Cambridge University, 27 Charles Babbage Road, Cambridge CB3 0FS, UK.
| | - Ruth E Cameron
- Department of Materials Science & Metallurgy, Cambridge University, 27 Charles Babbage Road, Cambridge CB3 0FS, UK.
| |
Collapse
|
13
|
El-Husseiny HM, Mady EA, Usui T, Ishihara Y, Yoshida T, Kobayashi M, Sasaki K, Ma D, Yairo A, Mandour AS, Hendawy H, Doghish AS, Mohammed OA, Takahashi K, Tanaka R. Adipose Stem Cell-Seeded Decellularized Porcine Pericardium: A Promising Functional Biomaterial to Synergistically Restore the Cardiac Functions Post-Myocardial Infarction. Vet Sci 2023; 10:660. [PMID: 37999483 PMCID: PMC10675230 DOI: 10.3390/vetsci10110660] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/19/2023] [Accepted: 11/15/2023] [Indexed: 11/25/2023] Open
Abstract
Myocardial infarction (MI) is a serious cardiovascular disease as the leading cause of death globally. Hence, reconstruction of the cardiac tissue comes at the forefront of strategies adopted to restore heart functions following MI. In this investigation, we studied the capacity of rat adipose-derived mesenchymal stem cells (r-AdMSCs) and decellularized porcine pericardium (DPP) to restore heart functions in MI animals. MI was induced in four different groups, three of which were treated either using DPP (MI-DPP group), stem cells (MI-SC group), or both (MI-SC/DPP group). Cardiac functions of these groups and the Sham group were evaluated using echocardiography, the intraventricular pressure gradient (IVPG) on weeks 2 and 4, and intraventricular hemodynamics on week 4. On day 31, the animals were euthanized for histological analysis. Echocardiographic, IVPG and hemodynamic findings indicated that the three treatment strategies shared effectively in the regeneration process. However, the MI-SC/DPP group had a unique synergistic ability to restore heart functions superior to the other treatment protocols. Histology showed that the MI-SC/DPP group presented the lowest (p < 0.05) degeneration score and fibrosis % compared to the other groups. Conclusively, stem cell-seeded DPP is a promising platform for the delivery of stem cells and restoration of heart functions post-MI.
Collapse
Affiliation(s)
- Hussein M. El-Husseiny
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai Cho, Fuchu-shi 183-8509, Tokyo, Japan; (K.S.); (D.M.); (A.Y.); (A.S.M.); (H.H.)
- Department of Surgery, Anesthesiology, and Radiology, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh 13736, Elqaliobiya, Egypt
| | - Eman A. Mady
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai Cho, Fuchu-shi 183-8509, Tokyo, Japan;
- Department of Animal Hygiene, Behavior and Management, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh 13736, Elqaliobiya, Egypt
| | - Tatsuya Usui
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai Cho, Fuchu-shi 183-8509, Tokyo, Japan; (T.U.); (Y.I.)
| | - Yusuke Ishihara
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai Cho, Fuchu-shi 183-8509, Tokyo, Japan; (T.U.); (Y.I.)
| | - Toshinori Yoshida
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi 183-8509, Tokyo, Japan; (T.Y.); (M.K.)
| | - Mio Kobayashi
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi 183-8509, Tokyo, Japan; (T.Y.); (M.K.)
| | - Kenta Sasaki
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai Cho, Fuchu-shi 183-8509, Tokyo, Japan; (K.S.); (D.M.); (A.Y.); (A.S.M.); (H.H.)
| | - Danfu Ma
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai Cho, Fuchu-shi 183-8509, Tokyo, Japan; (K.S.); (D.M.); (A.Y.); (A.S.M.); (H.H.)
- College of Veterinary Medicine, Nanjing Agricultural University, No. 1 Wei-Gang, Xuanwu District, Nanjing 210095, China
| | - Akira Yairo
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai Cho, Fuchu-shi 183-8509, Tokyo, Japan; (K.S.); (D.M.); (A.Y.); (A.S.M.); (H.H.)
| | - Ahmed S. Mandour
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai Cho, Fuchu-shi 183-8509, Tokyo, Japan; (K.S.); (D.M.); (A.Y.); (A.S.M.); (H.H.)
- Department of Animal Medicine (Internal Medicine), Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Ismailia, Egypt
| | - Hanan Hendawy
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai Cho, Fuchu-shi 183-8509, Tokyo, Japan; (K.S.); (D.M.); (A.Y.); (A.S.M.); (H.H.)
- Department of Veterinary Surgery, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Ismailia, Egypt
| | - Ahmed S. Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City 11829, Cairo, Egypt;
- Department of Biochemistry, and Molecular Biology Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11651, Cairo, Egypt
| | - Osama A. Mohammed
- Department of Clinical Pharmacology, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia;
| | - Ken Takahashi
- Department of Pediatrics and Adolescent Medicine, Juntendo University Graduate School of Medicine, Bunkyo 113-8421, Tokyo, Japan;
| | - Ryou Tanaka
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai Cho, Fuchu-shi 183-8509, Tokyo, Japan; (K.S.); (D.M.); (A.Y.); (A.S.M.); (H.H.)
| |
Collapse
|
14
|
Chen TA, Sharma D, Jia W, Ha D, Man K, Zhang J, Yang Y, Zhou Y, Kamp TJ, Zhao F. Detergent-Based Decellularization for Anisotropic Cardiac-Specific Extracellular Matrix Scaffold Generation. Biomimetics (Basel) 2023; 8:551. [PMID: 37999192 PMCID: PMC10669368 DOI: 10.3390/biomimetics8070551] [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: 08/09/2023] [Revised: 09/19/2023] [Accepted: 11/09/2023] [Indexed: 11/25/2023] Open
Abstract
Cell-derived extracellular matrix (ECM) has become increasingly popular in tissue engineering applications due to its ability to provide tailored signals for desirable cellular responses. Anisotropic cardiac-specific ECM scaffold decellularized from human induced pluripotent stem cell (hiPSC)-derived cardiac fibroblasts (hiPSC-CFs) mimics the native cardiac microenvironment and provides essential biochemical and signaling cues to hiPSC-derived cardiomyocytes (hiPSC-CMs). The objective of this study was to assess the efficacy of two detergent-based decellularization methods: (1) a combination of ethylenediaminetetraacetic acid and sodium dodecyl sulfate (EDTA + SDS) and (2) a combination of sodium deoxycholate and deoxyribonuclease (SD + DNase), in preserving the composition and bioactive substances within the aligned ECM scaffold while maximumly removing cellular components. The decellularization effects were evaluated by characterizing the ECM morphology, quantifying key structural biomacromolecules, and measuring preserved growth factors. Results showed that both treatments met the standard of cell removal (less than 50 ng/mg ECM dry weight) and substantially preserved major ECM biomacromolecules and growth factors. The EDTA + SDS treatment was more time-efficient and has been determined to be a more efficient method for generating an anisotropic ECM scaffold from aligned hiPSC-CFs. Moreover, this cardiac-specific ECM has demonstrated effectiveness in supporting the alignment of hiPSC-CMs and their expression of mature structural and functional proteins in in vitro cultures, which is crucial for cardiac tissue engineering.
Collapse
Affiliation(s)
- Te-An Chen
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Dhavan Sharma
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Wenkai Jia
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Donggi Ha
- Department of Mechanical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Kun Man
- Department of Biomedical Engineering, University of North Texas, Denton, TX 76203, USA
| | - Jianhua Zhang
- Stem Cell and Regenerative Medicine Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Yong Yang
- Department of Biomedical Engineering, University of North Texas, Denton, TX 76203, USA
| | - Yuxiao Zhou
- Department of Mechanical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Timothy J. Kamp
- Stem Cell and Regenerative Medicine Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Feng Zhao
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
15
|
Kafili G, Kabir H, Jalali Kandeloos A, Golafshan E, Ghasemi S, Mashayekhan S, Taebnia N. Recent advances in soluble decellularized extracellular matrix for heart tissue engineering and organ modeling. J Biomater Appl 2023; 38:577-604. [PMID: 38006224 PMCID: PMC10676626 DOI: 10.1177/08853282231207216] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2023]
Abstract
Despite the advent of tissue engineering (TE) for the remodeling, restoring, and replacing damaged cardiovascular tissues, the progress is hindered by the optimal mechanical and chemical properties required to induce cardiac tissue-specific cellular behaviors including migration, adhesion, proliferation, and differentiation. Cardiac extracellular matrix (ECM) consists of numerous structural and functional molecules and tissue-specific cells, therefore it plays an important role in stimulating cell proliferation and differentiation, guiding cell migration, and activating regulatory signaling pathways. With the improvement and modification of cell removal methods, decellularized ECM (dECM) preserves biochemical complexity, and bio-inductive properties of the native matrix and improves the process of generating functional tissue. In this review, we first provide an overview of the latest advancements in the utilization of dECM in in vitro model systems for disease and tissue modeling, as well as drug screening. Then, we explore the role of dECM-based biomaterials in cardiovascular regenerative medicine (RM), including both invasive and non-invasive methods. In the next step, we elucidate the engineering and material considerations in the preparation of dECM-based biomaterials, namely various decellularization techniques, dECM sources, modulation, characterizations, and fabrication approaches. Finally, we discuss the limitations and future directions in fabrication of dECM-based biomaterials for cardiovascular modeling, RM, and clinical translation.
Collapse
Affiliation(s)
- Golara Kafili
- Institute for Nanoscience and Nanotechnology, Sharif University of Technology, Tehran, Iran
| | - Hannaneh Kabir
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California, Berkeley, CA, USA
| | | | - Elham Golafshan
- Institute for Nanoscience and Nanotechnology, Sharif University of Technology, Tehran, Iran
| | - Sara Ghasemi
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| | - Shohreh Mashayekhan
- Institute for Nanoscience and Nanotechnology, Sharif University of Technology, Tehran, Iran
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| | - Nayere Taebnia
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
16
|
Tani H, Kobayashi E, Yagi S, Tanaka K, Kameda-Haga K, Shibata S, Moritoki N, Takatsuna K, Moriwaki T, Sekine O, Umei TC, Morita Y, Soma Y, Kishino Y, Kanazawa H, Fujita J, Hattori S, Fukuda K, Tohyama S. Heart-derived collagen promotes maturation of engineered heart tissue. Biomaterials 2023; 299:122174. [PMID: 37285642 DOI: 10.1016/j.biomaterials.2023.122174] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 05/17/2023] [Accepted: 05/19/2023] [Indexed: 06/09/2023]
Abstract
Although the extracellular matrix (ECM) plays essential roles in heart tissue engineering, the optimal ECM components for heart tissue organization have not previously been elucidated. Here, we focused on the main ECM component, fibrillar collagen, and analyzed the effects of collagens on heart tissue engineering, by comparing the use of porcine heart-derived collagen and other organ-derived collagens in generating engineered heart tissue (EHT). We demonstrate that heart-derived collagen induces better contraction and relaxation of human induced pluripotent stem cell-derived EHT (hiPSC-EHT) and that hiPSC-EHT with heart-derived collagen exhibit more mature profiles than those with collagens from other organs. Further, we found that collagen fibril formation and gel stiffness influence the contraction, relaxation, and maturation of hiPSC-EHT, suggesting the importance of collagen types III and type V, which are relatively abundant in the heart. Thus, we demonstrate the effectiveness of organ-specific collagens in tissue engineering and drug discovery.
Collapse
Affiliation(s)
- Hidenori Tani
- Department of Cardiology, Japan; Joint Research Laboratory for Medical Innovation in Heart Disease, Japan
| | - Eiji Kobayashi
- Department of Organ Fabrication, Japan; Department of Kidney Regenerative Medicine, The Jikei University School of Medicine, Tokyo, Japan.
| | - Shinomi Yagi
- Nippi Research Institute of Biomatrix, Toride, Ibaraki, Japan
| | - Keisuke Tanaka
- Nippi Research Institute of Biomatrix, Toride, Ibaraki, Japan
| | | | - Shinsuke Shibata
- Electron Microscope Laboratory, Keio University School of Medicine, Shinjuku, Tokyo, Japan; Division of Microscopic Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Nobuko Moritoki
- Electron Microscope Laboratory, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | | | | | | | | | - Yuika Morita
- Department of Cardiology, Japan; Kanagawa Institute of Industrial Science and Technology (KISTEC), Kawasaki, Kanagawa, Japan
| | | | | | | | - Jun Fujita
- Department of Cardiology, Japan; Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Shunji Hattori
- Department of Kidney Regenerative Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | | | | |
Collapse
|
17
|
Sigaroodi F, Rahmani M, Parandakh A, Boroumand S, Rabbani S, Khani MM. Designing cardiac patches for myocardial regeneration–a review. INT J POLYM MATER PO 2023. [DOI: 10.1080/00914037.2023.2180510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Affiliation(s)
- Faraz Sigaroodi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahya Rahmani
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Azim Parandakh
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Safieh Boroumand
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shahram Rabbani
- Research Center for Advanced Technologies in Cardiovascular Medicine, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad-Mehdi Khani
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
18
|
Whole-Heart Tissue Engineering and Cardiac Patches: Challenges and Promises. BIOENGINEERING (BASEL, SWITZERLAND) 2023; 10:bioengineering10010106. [PMID: 36671678 PMCID: PMC9855348 DOI: 10.3390/bioengineering10010106] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/02/2023] [Accepted: 01/05/2023] [Indexed: 01/15/2023]
Abstract
Despite all the advances in preventing, diagnosing, and treating cardiovascular disorders, they still account for a significant part of mortality and morbidity worldwide. The advent of tissue engineering and regenerative medicine has provided novel therapeutic approaches for the treatment of various diseases. Tissue engineering relies on three pillars: scaffolds, stem cells, and growth factors. Gene and cell therapy methods have been introduced as primary approaches to cardiac tissue engineering. Although the application of gene and cell therapy has resulted in improved regeneration of damaged cardiac tissue, further studies are needed to resolve their limitations, enhance their effectiveness, and translate them into the clinical setting. Scaffolds from synthetic, natural, or decellularized sources have provided desirable characteristics for the repair of cardiac tissue. Decellularized scaffolds are widely studied in heart regeneration, either as cell-free constructs or cell-seeded platforms. The application of human- or animal-derived decellularized heart patches has promoted the regeneration of heart tissue through in vivo and in vitro studies. Due to the complexity of cardiac tissue engineering, there is still a long way to go before cardiac patches or decellularized whole-heart scaffolds can be routinely used in clinical practice. This paper aims to review the decellularized whole-heart scaffolds and cardiac patches utilized in the regeneration of damaged cardiac tissue. Moreover, various decellularization methods related to these scaffolds will be discussed.
Collapse
|
19
|
Joddar B, Natividad-Diaz SL, Padilla AE, Esparza AA, Ramirez SP, Chambers DR, Ibaroudene H. Engineering approaches for cardiac organoid formation and their characterization. Transl Res 2022; 250:46-67. [PMID: 35995380 PMCID: PMC10370285 DOI: 10.1016/j.trsl.2022.08.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/05/2022] [Accepted: 08/15/2022] [Indexed: 11/29/2022]
Abstract
Cardiac organoids are 3-dimensional (3D) structures composed of tissue or niche-specific cells, obtained from diverse sources, encapsulated in either a naturally derived or synthetic, extracellular matrix scaffold, and include exogenous biochemical signals such as essential growth factors. The overarching goal of developing cardiac organoid models is to establish a functional integration of cardiomyocytes with physiologically relevant cells, tissues, and structures like capillary-like networks composed of endothelial cells. These organoids used to model human heart anatomy, physiology, and disease pathologies in vitro have the potential to solve many issues related to cardiovascular drug discovery and fundamental research. The advent of patient-specific human-induced pluripotent stem cell-derived cardiovascular cells provide a unique, single-source approach to study the complex process of cardiovascular disease progression through organoid formation and incorporation into relevant, controlled microenvironments such as microfluidic devices. Strategies that aim to accomplish such a feat include microfluidic technology-based approaches, microphysiological systems, microwells, microarray-based platforms, 3D bioprinted models, and electrospun fiber mat-based scaffolds. This article discusses the engineering or technology-driven practices for making cardiac organoid models in comparison with self-assembled or scaffold-free methods to generate organoids. We further discuss emerging strategies for characterization of the bio-assembled cardiac organoids including electrophysiology and machine-learning and conclude with prospective points of interest for engineering cardiac tissues in vitro.
Collapse
Affiliation(s)
- Binata Joddar
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL); Department of Metallurgical, Materials and Biomedical Engineering, University of Texas at El Paso, El Paso, Texas; Border Biomedical Research Center, University of Texas at El Paso, El Paso, Texas.
| | - Sylvia L Natividad-Diaz
- Department of Metallurgical, Materials and Biomedical Engineering, University of Texas at El Paso, El Paso, Texas; Border Biomedical Research Center, University of Texas at El Paso, El Paso, Texas
| | - Andie E Padilla
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL); Department of Metallurgical, Materials and Biomedical Engineering, University of Texas at El Paso, El Paso, Texas
| | - Aibhlin A Esparza
- Department of Metallurgical, Materials and Biomedical Engineering, University of Texas at El Paso, El Paso, Texas
| | - Salma P Ramirez
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL); Department of Metallurgical, Materials and Biomedical Engineering, University of Texas at El Paso, El Paso, Texas
| | | | | |
Collapse
|
20
|
McInnes AD, Moser MAJ, Chen X. Preparation and Use of Decellularized Extracellular Matrix for Tissue Engineering. J Funct Biomater 2022; 13:jfb13040240. [PMID: 36412881 PMCID: PMC9680265 DOI: 10.3390/jfb13040240] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/22/2022] [Accepted: 11/05/2022] [Indexed: 11/16/2022] Open
Abstract
The multidisciplinary fields of tissue engineering and regenerative medicine have the potential to revolutionize the practise of medicine through the abilities to repair, regenerate, or replace tissues and organs with functional engineered constructs. To this end, tissue engineering combines scaffolding materials with cells and biologically active molecules into constructs with the appropriate structures and properties for tissue/organ regeneration, where scaffolding materials and biomolecules are the keys to mimic the native extracellular matrix (ECM). For this, one emerging way is to decellularize the native ECM into the materials suitable for, directly or in combination with other materials, creating functional constructs. Over the past decade, decellularized ECM (or dECM) has greatly facilitated the advance of tissue engineering and regenerative medicine, while being challenged in many ways. This article reviews the recent development of dECM for tissue engineering and regenerative medicine, with a focus on the preparation of dECM along with its influence on cell culture, the modification of dECM for use as a scaffolding material, and the novel techniques and emerging trends in processing dECM into functional constructs. We highlight the success of dECM and constructs in the in vitro, in vivo, and clinical applications and further identify the key issues and challenges involved, along with a discussion of future research directions.
Collapse
Affiliation(s)
- Adam D. McInnes
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK S7N 5A9, Canada
- Correspondence: ; Tel.: +1-306-966-5435
| | - Michael A. J. Moser
- Department of Surgery, Health Sciences Building, University of Saskatchewan, Saskatoon, SK S7N 0W8, Canada
| | - Xiongbiao Chen
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK S7N 5A9, Canada
- Department of Mechanical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK S7N 5A9, Canada
| |
Collapse
|
21
|
Barbulescu GI, Bojin FM, Ordodi VL, Goje ID, Barbulescu AS, Paunescu V. Decellularized Extracellular Matrix Scaffolds for Cardiovascular Tissue Engineering: Current Techniques and Challenges. Int J Mol Sci 2022; 23:13040. [PMID: 36361824 PMCID: PMC9658138 DOI: 10.3390/ijms232113040] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 10/18/2022] [Accepted: 10/26/2022] [Indexed: 08/13/2023] Open
Abstract
Cardiovascular diseases are the leading cause of global mortality. Over the past two decades, researchers have tried to provide novel solutions for end-stage heart failure to address cardiac transplantation hurdles such as donor organ shortage, chronic rejection, and life-long immunosuppression. Cardiac decellularized extracellular matrix (dECM) has been widely explored as a promising approach in tissue-regenerative medicine because of its remarkable similarity to the original tissue. Optimized decellularization protocols combining physical, chemical, and enzymatic agents have been developed to obtain the perfect balance between cell removal, ECM composition, and function maintenance. However, proper assessment of decellularized tissue composition is still needed before clinical translation. Recellularizing the acellular scaffold with organ-specific cells and evaluating the extent of cardiomyocyte repopulation is also challenging. This review aims to discuss the existing literature on decellularized cardiac scaffolds, especially on the advantages and methods of preparation, pointing out areas for improvement. Finally, an overview of the state of research regarding the application of cardiac dECM and future challenges in bioengineering a human heart suitable for transplantation is provided.
Collapse
Affiliation(s)
- Greta Ionela Barbulescu
- Immuno-Physiology and Biotechnologies Center (CIFBIOTEH), Department of Functional Sciences, “Victor Babes” University of Medicine and Pharmacy, No 2 Eftimie Murgu Square, 300041 Timisoara, Romania
- Department of Clinical Practical Skills, “Victor Babes” University of Medicine and Pharmacy, No 2 Eftimie Murgu Square, 300041 Timisoara, Romania
| | - Florina Maria Bojin
- Immuno-Physiology and Biotechnologies Center (CIFBIOTEH), Department of Functional Sciences, “Victor Babes” University of Medicine and Pharmacy, No 2 Eftimie Murgu Square, 300041 Timisoara, Romania
- Clinical Emergency County Hospital “Pius Brinzeu” Timisoara, Center for Gene and Cellular Therapies in the Treatment of Cancer Timisoara-OncoGen, No 156 Liviu Rebreanu, 300723 Timisoara, Romania
| | - Valentin Laurentiu Ordodi
- Clinical Emergency County Hospital “Pius Brinzeu” Timisoara, Center for Gene and Cellular Therapies in the Treatment of Cancer Timisoara-OncoGen, No 156 Liviu Rebreanu, 300723 Timisoara, Romania
- Faculty of Industrial Chemistry and Environmental Engineering, “Politehnica” University Timisoara, No 2 Victoriei Square, 300006 Timisoara, Romania
| | - Iacob Daniel Goje
- Department of Medical Semiology I, “Victor Babes” University of Medicine and Pharmacy, No 2 Eftimie Murgu Square, 300041 Timisoara, Romania
- Advanced Cardiology and Hemostaseology Research Center, “Victor Babes” University of Medicine and Pharmacy, No 2 Eftimie Murgu Square, 300041 Timisoara, Romania
| | - Andreea Severina Barbulescu
- Center for Advanced Research in Gastroenterology and Hepatology, Department of Internal Medicine II, Division of Gastroenterology and Hepatology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Virgil Paunescu
- Immuno-Physiology and Biotechnologies Center (CIFBIOTEH), Department of Functional Sciences, “Victor Babes” University of Medicine and Pharmacy, No 2 Eftimie Murgu Square, 300041 Timisoara, Romania
- Clinical Emergency County Hospital “Pius Brinzeu” Timisoara, Center for Gene and Cellular Therapies in the Treatment of Cancer Timisoara-OncoGen, No 156 Liviu Rebreanu, 300723 Timisoara, Romania
| |
Collapse
|
22
|
Li J, Liu L, Zhang J, Qu X, Kawamura T, Miyagawa S, Sawa Y. Engineered Tissue for Cardiac Regeneration: Current Status and Future Perspectives. Bioengineering (Basel) 2022; 9:605. [PMID: 36354516 PMCID: PMC9688015 DOI: 10.3390/bioengineering9110605] [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: 09/20/2022] [Revised: 10/12/2022] [Accepted: 10/20/2022] [Indexed: 11/12/2023] Open
Abstract
Heart failure (HF) is the leading cause of death worldwide. The most effective HF treatment is heart transplantation, the use of which is restricted by the limited supply of donor hearts. The human pluripotent stem cell (hPSC), including human embryonic stem cell (hESC) and the induced pluripotent stem cells (hiPSC), could be produced in an infinite manner and differentiated into cardiomyocytes (CMs) with high efficiency. The hPSC-CMs have, thus, offered a promising alternative for heart transplant. In this review, we introduce the tissue-engineering technologies for hPSC-CM, including the materials for cell culture and tissue formation, and the delivery means into the heart. The most recent progress in clinical application of hPSC-CMs is also introduced. In addition, the bottleneck limitations and future perspectives for clinical translation are further discussed.
Collapse
Affiliation(s)
- Junjun Li
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Li Liu
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Jingbo Zhang
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Xiang Qu
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Takuji Kawamura
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Shigeru Miyagawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Yoshiki Sawa
- Cardiovascular Division, Osaka Police Hospital, Tennoji, Osaka 543-0035, Japan
| |
Collapse
|
23
|
Kałużna E, Nadel A, Zimna A, Rozwadowska N, Kolanowski T. Modeling the human heart ex vivo-current possibilities and strive for future applications. J Tissue Eng Regen Med 2022; 16:853-874. [PMID: 35748158 PMCID: PMC9796015 DOI: 10.1002/term.3335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 04/20/2022] [Accepted: 06/03/2022] [Indexed: 12/30/2022]
Abstract
The high organ specification of the human heart is inversely proportional to its functional recovery after damage. The discovery of induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) has accelerated research in human heart regeneration and physiology. Nevertheless, due to the immaturity of iPSC-CMs, they are far from being an representative model of the adult heart physiology. Therefore, number of laboratories strive to obtain a heart tissues by engineering methods by structuring iPSC-CMs into complex and advanced platforms. By using the iPSC-CMs and arranging them in 3D cultures it is possible to obtain a human heart muscle with physiological capabilities potentially similar to the adult heart, while remaining in vitro. Here, we attempt to describe existing examples of heart muscle either in vitro or ex vivo models and discuss potential options for the further development of such structures. This will be a crucial step for ultimate derivation of complete heart tissue-mimicking organs and their future use in drug development, therapeutic approaches testing, pre-clinical studies, and clinical applications. This review particularly aims to compile available models of advanced human heart tissue for scientists considering which model would best fit their research needs.
Collapse
Affiliation(s)
- Ewelina Kałużna
- Institute of Human GeneticsPolish Academy of SciencesPoznanPoland
| | - Agnieszka Nadel
- Institute of Human GeneticsPolish Academy of SciencesPoznanPoland
| | - Agnieszka Zimna
- Institute of Human GeneticsPolish Academy of SciencesPoznanPoland
| | | | | |
Collapse
|
24
|
Sharma V, Manhas A, Gupta S, Dikshit M, Jagavelu K, Verma RS. Fabrication, characterization and in vivo assessment of cardiogel loaded chitosan patch for myocardial regeneration. Int J Biol Macromol 2022; 222:3045-3056. [DOI: 10.1016/j.ijbiomac.2022.10.079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 10/06/2022] [Accepted: 10/09/2022] [Indexed: 11/05/2022]
|
25
|
Csöbönyeiová M, Beerová N, Klein M, Debreová-Čeháková M, Danišovič Ľ. Cell-Based and Selected Cell-Free Therapies for Myocardial Infarction: How Do They Compare to the Current Treatment Options? Int J Mol Sci 2022; 23:10314. [PMID: 36142245 PMCID: PMC9499607 DOI: 10.3390/ijms231810314] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/03/2022] [Accepted: 09/05/2022] [Indexed: 11/17/2022] Open
Abstract
Because of cardiomyocyte death or dysfunction frequently caused by myocardial infarction (MI), heart failure is a leading cause of morbidity and mortality in modern society. Paradoxically, only limited and non-curative therapies for heart failure or MI are currently available. As a result, over the past two decades research has focused on developing cell-based approaches promoting the regeneration of infarcted tissue. Cell-based therapies for myocardial regeneration include powerful candidates, such as multipotent stem cells (mesenchymal stem cells (MSCs), bone-marrow-derived stem cells, endothelial progenitor cells, and hematopoietic stem cells) and induced pluripotent stem cells (iPSCs). These possess unique properties, such as potency to differentiate into desired cell types, proliferation capacity, and patient specificity. Preclinical and clinical studies have demonstrated modest improvement in the myocardial regeneration and reduced infarcted areas upon transplantation of pluripotent or multipotent stem cells. Another cell population that need to be considered as a potential source for cardiac regeneration are telocytes found in different organs, including the heart. Their therapeutic effect has been studied in various heart pathologies, such as MI, arrhythmias, or atrial amyloidosis. The most recent cell-free therapeutic tool relies on the cardioprotective effect of complex cargo carried by small membrane-bound vesicles-exosomes-released from stem cells via exocytosis. The MSC/iPSC-derived exosomes could be considered a novel exosome-based therapy for cardiovascular diseases thanks to their unique content. There are also other cell-free approaches, e.g., gene therapy, or acellular cardiac patches. Therefore, our review provides the most recent insights into the novel strategies for myocardial repair based on the regenerative potential of different cell types and cell-free approaches.
Collapse
Affiliation(s)
- Mária Csöbönyeiová
- National Institute of Rheumatic Diseases, Nábrežie I. Krasku 4, 921 12 Piešťany, Slovakia
- Institute of Histology and Embryology, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Nikoleta Beerová
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Martin Klein
- National Institute of Rheumatic Diseases, Nábrežie I. Krasku 4, 921 12 Piešťany, Slovakia
- Institute of Histology and Embryology, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Michaela Debreová-Čeháková
- National Institute of Rheumatic Diseases, Nábrežie I. Krasku 4, 921 12 Piešťany, Slovakia
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Ľuboš Danišovič
- National Institute of Rheumatic Diseases, Nábrežie I. Krasku 4, 921 12 Piešťany, Slovakia
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| |
Collapse
|
26
|
Wu Y, Guo X, Han T, Feng K, Zhang P, Xu Y, Yang Y, Xia Y, Chen Y, Xi J, Yang H, Wan X, Kang J. Cmarr/miR-540-3p axis promotes cardiomyocyte maturation transition by orchestrating Dtna expression. MOLECULAR THERAPY - NUCLEIC ACIDS 2022; 29:481-497. [PMID: 36035750 PMCID: PMC9382425 DOI: 10.1016/j.omtn.2022.07.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 07/20/2022] [Indexed: 10/31/2022]
Abstract
The immature phenotype of embryonic stem cell-derived cardiomyocytes (ESC-CMs) limits their application. However, the molecular mechanisms of cardiomyocyte maturation remain largely unexplored. This study found that overexpression of long noncoding RNA (lncRNA)-Cmarr, which was highly expressed in cardiomyocytes, promoted the maturation change and physiological maturation of mouse ESC-CMs (mESC-CMs). Moreover, transplantation of cardiac patch overexpressing Cmarr exhibited better retention of mESC-CMs, reduced infarct area by enhancing vascular density in the host heart, and improved cardiac function in mice after myocardial infarction. Mechanism studies identified that Cmarr acted as a competitive endogenous RNA to impede the repression of miR-540-3p on Dtna expression and promoted the binding of the dystrophin-glycoprotein complex (DGC) and yes-associated protein (YAP), which in turn reduced the proportion of nuclear YAP and the expression of YAP target genes. Therefore, this study revealed the function and mechanism of Cmarr in promoting cardiomyocyte maturation and provided a lncRNA that can be used as a functional factor in the construction of cardiac patches for the treatment of myocardial infarction.
Collapse
|
27
|
Assali M, Kittana N, Alhaj-Qasem S, Hajjyahya M, Abu-Rass H, Alshaer W, Al-Buqain R. Noncovalent functionalization of carbon nanotubes as a scaffold for tissue engineering. Sci Rep 2022; 12:12062. [PMID: 35835926 PMCID: PMC9283586 DOI: 10.1038/s41598-022-16247-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 07/07/2022] [Indexed: 11/10/2022] Open
Abstract
Tissue engineering is one of the hot topics in recent research that needs special requirements. It depends on the development of scaffolds that allow tissue formation with certain characteristics, carbon nanotubes (CNTs)-collagen composite attracted the attention of the researchers with this respect. However, CNTs suffer from low water dispersibility, which hampered their utilization. Therefore, we aim to functionalize CNTs non-covalently with pyrene moiety using an appropriate hydrophilic linker derivatized from polyethylene glycol (PEG) terminated with hydroxyl or carboxyl group to disperse them in water. The functionalization of the CNTs is successfully confirmed by TEM, absorption spectroscopy, TGA, and zeta potential analysis. 3T3 cells-based engineered connective tissues (ECTs) are generated with different concentrations of the functionalized CNTs (f-CNTs). These tissues show a significant enhancement in electrical conductivity at a concentration of 0.025%, however, the cell viability is reduced by about 10 to 20%. All ECTs containing f-CNTs show a significant reduction in tissue fibrosis and matrix porosity relative to the control tissues. Taken together, the developed constructs show great potential for further in vivo studies as engineered tissue.
Collapse
Affiliation(s)
- Mohyeddin Assali
- Department of Pharmacy, Faculty of Medicine & Health Sciences, An-Najah National University, Nablus, Palestine.
| | - Naim Kittana
- Department of Biomedical Sciences, Faculty of Medicine & Health Sciences, An-Najah National University, Nablus, Palestine.
| | - Sahar Alhaj-Qasem
- Department of Pharmacy, Faculty of Medicine & Health Sciences, An-Najah National University, Nablus, Palestine
| | - Muna Hajjyahya
- Department of Physics, Faculty of Sciences, An-Najah National University, Nablus, Palestine
| | - Hanood Abu-Rass
- Department of Biomedical Sciences, Faculty of Medicine & Health Sciences, An-Najah National University, Nablus, Palestine
| | - Walhan Alshaer
- Cell Therapy Center, The University of Jordan, Amman, 11942, Jordan
| | - Rula Al-Buqain
- Cell Therapy Center, The University of Jordan, Amman, 11942, Jordan
| |
Collapse
|
28
|
Varzideh F, Mone P, Santulli G. Bioengineering Strategies to Create 3D Cardiac Constructs from Human Induced Pluripotent Stem Cells. Bioengineering (Basel) 2022; 9:168. [PMID: 35447728 PMCID: PMC9028595 DOI: 10.3390/bioengineering9040168] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 04/06/2022] [Accepted: 04/08/2022] [Indexed: 12/12/2022] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) can be used to generate various cell types in the human body. Hence, hiPSC-derived cardiomyocytes (hiPSC-CMs) represent a significant cell source for disease modeling, drug testing, and regenerative medicine. The immaturity of hiPSC-CMs in two-dimensional (2D) culture limit their applications. Cardiac tissue engineering provides a new promise for both basic and clinical research. Advanced bioengineered cardiac in vitro models can create contractile structures that serve as exquisite in vitro heart microtissues for drug testing and disease modeling, thereby promoting the identification of better treatments for cardiovascular disorders. In this review, we will introduce recent advances of bioengineering technologies to produce in vitro cardiac tissues derived from hiPSCs.
Collapse
Affiliation(s)
- Fahimeh Varzideh
- Department of Medicine, Wilf Family Cardiovascular Research Institute, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA; (F.V.); (P.M.)
- Department of Molecular Pharmacology, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Pasquale Mone
- Department of Medicine, Wilf Family Cardiovascular Research Institute, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA; (F.V.); (P.M.)
| | - Gaetano Santulli
- Department of Medicine, Wilf Family Cardiovascular Research Institute, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA; (F.V.); (P.M.)
- Department of Molecular Pharmacology, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, New York, NY 10461, USA
| |
Collapse
|
29
|
Yang J, Dang H, Xu Y. Recent advancement of decellularization extracellular matrix for tissue engineering and biomedical application. Artif Organs 2022; 46:549-567. [PMID: 34855994 DOI: 10.1111/aor.14126] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/18/2021] [Accepted: 11/15/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Decellularized extracellular matrixs (dECMs) derived from organs and tissues have emerged as a promising tool, as they encompass the characteristics of an ideal tissue scaffold: complex composition, vascular networks and unique tissue-specific architecture. Consequently, their use has propagated throughout tissue engineering and regenerative medicine. dECM can be easily obtained from various tissues/organs by appropriate decellularization protocolsand is entitled to provide necessary cues to cells homing. METHODS In this review, we describe the decellularization and sterilization methods that are commonly used in recent research, the effects of these methods upon biologic scaffold material are discussed. Also, we summarize the recent developments of recellularization and vascularization techniques in regeneration medicine. Additionally, dECM preservation methods is mentioned, which provides the basis for the establishment of organ bank. RESULTS Biomedical applications and the status of current research developments relating to dECM biomaterials are outlined, including transplantation in vivo, disease models and drug screening, organoid, 3D bioprinting, tissue reconstruction and rehabilitation and cell transplantation and culture. Finally, critical challenges and future developing technologies are discussed. CONCLUSIONS With the development of tissue engineering and regenerative medicine, dECM will have broader applications in the field of biomedicine in the near future.
Collapse
Affiliation(s)
- Jiamin Yang
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Hangyu Dang
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Yi Xu
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, China
| |
Collapse
|
30
|
Li M, Wu H, Yuan Y, Hu B, Gu N. Recent fabrications and applications of cardiac patch in myocardial infarction treatment. VIEW 2022. [DOI: 10.1002/viw.20200153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Mei Li
- School of Biomedical Engineering and Informatics Nanjing Medical University Nanjing China
- The Laboratory Center for Basic Medical Sciences Nanjing Medical University Nanjing China
| | - Hao Wu
- School of Biomedical Engineering and Informatics Nanjing Medical University Nanjing China
| | - Yuehui Yuan
- School of Biomedical Engineering and Informatics Nanjing Medical University Nanjing China
| | - Benhui Hu
- School of Biomedical Engineering and Informatics Nanjing Medical University Nanjing China
| | - Ning Gu
- School of Biomedical Engineering and Informatics Nanjing Medical University Nanjing China
- State Key Laboratory of Bioelectronics Jiangsu Key Laboratory for Biomaterials and Devices School of Biological Sciences and Medical Engineering Southeast University Nanjing China
| |
Collapse
|
31
|
Tan YH, Helms HR, Nakayama KH. Decellularization Strategies for Regenerating Cardiac and Skeletal Muscle Tissues. Front Bioeng Biotechnol 2022; 10:831300. [PMID: 35295645 PMCID: PMC8918733 DOI: 10.3389/fbioe.2022.831300] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 01/28/2022] [Indexed: 12/24/2022] Open
Abstract
Cardiovascular disease is the leading cause of death worldwide and is associated with approximately 17.9 million deaths each year. Musculoskeletal conditions affect more than 1.71 billion people globally and are the leading cause of disability. These two areas represent a massive global health burden that is perpetuated by a lack of functionally restorative treatment options. The fields of regenerative medicine and tissue engineering offer great promise for the development of therapies to repair damaged or diseased tissues. Decellularized tissues and extracellular matrices are cornerstones of regenerative biomaterials and have been used clinically for decades and many have received FDA approval. In this review, we first discuss and compare methods used to produce decellularized tissues and ECMs from cardiac and skeletal muscle. We take a focused look at how different biophysical properties such as spatial topography, extracellular matrix composition, and mechanical characteristics influence cell behavior and function in the context of regenerative medicine. Lastly, we describe emerging research and forecast the future high impact applications of decellularized cardiac and skeletal muscle that will drive novel and effective regenerative therapies.
Collapse
Affiliation(s)
| | | | - Karina H. Nakayama
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
32
|
Vasu S, Zhou J, Chen J, Johnston PV, Kim DH. Biomaterials-based Approaches for Cardiac Regeneration. Korean Circ J 2021; 51:943-960. [PMID: 34854577 PMCID: PMC8636758 DOI: 10.4070/kcj.2021.0291] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 09/08/2021] [Indexed: 12/29/2022] Open
Abstract
Cardiovascular disease is a prevalent cause of mortality and morbidity, largely due to the limited ability of cardiomyocytes to proliferate. Existing therapies for cardiac regeneration include cell-based therapies and bioactive molecules. However, delivery remains one of the major challenges impeding such therapies from having significant clinical impact. Recent advancements in biomaterials-based approaches for cardiac regeneration have shown promise in improving cardiac function, promoting angiogenesis, and reducing adverse immune response in both human clinical trials and animal studies. These advances in therapeutic delivery via extracellular vesicles, cardiac patches, and hydrogels have the potential to enable clinical impact of cardiac regeneration therapies. The limited ability of cardiomyocytes to proliferate is a major cause of mortality and morbidity in cardiovascular diseases. There exist therapies for cardiac regeneration that are cell-based as well as that involve bioactive molecules. However, delivery remains one of the major challenges impeding such therapies from having clinical impact. Recent advancements in biomaterials-based approaches for cardiac regeneration have shown promise in clinical trials and animal studies in improving cardiac function, promoting angiogenesis, and reducing adverse immune response. This review will focus on current clinical studies of three contemporary biomaterials-based approaches for cardiac regeneration (extracellular vesicles, injectable hydrogels, and cardiac patches), remaining challenges and shortcomings to be overcome, and future directions for the use of biomaterials to promote cardiac regeneration.
Collapse
Affiliation(s)
- Samhita Vasu
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Justin Zhou
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Jeffrey Chen
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Peter V Johnston
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA.,Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
33
|
Jin Y, Cho SW. Bioengineering platforms for cell therapeutics derived from pluripotent and direct reprogramming. APL Bioeng 2021; 5:031501. [PMID: 34258498 PMCID: PMC8263070 DOI: 10.1063/5.0040621] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 06/01/2021] [Indexed: 12/13/2022] Open
Abstract
Pluripotent and direct reprogramming technologies hold great potential for tissue repair and restoration of tissue and organ function. The implementation of induced pluripotent stem cells and directly reprogrammed cells in biomedical research has resulted in a significant leap forward in the highly promising area of regenerative medicine. While these therapeutic strategies are promising, there are several obstacles to overcome prior to the introduction of these therapies into clinical settings. Bioengineering technologies, such as biomaterials, bioprinting, microfluidic devices, and biostimulatory systems, can enhance cell viability, differentiation, and function, in turn the efficacy of cell therapeutics generated via pluripotent and direct reprogramming. Therefore, cellular reprogramming technologies, in combination with tissue-engineering platforms, are poised to overcome current bottlenecks associated with cell-based therapies and create new ways of producing engineered tissue substitutes.
Collapse
Affiliation(s)
- Yoonhee Jin
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | | |
Collapse
|
34
|
Cho J, Kim S, Lee H, Rah W, Cho HC, Kim NK, Bae S, Shin DH, Lee MG, Park IH, Tanaka Y, Shin E, Yi H, Han JW, Hwang PTJ, Jun HW, Park HJ, Cho K, Lee SW, Jung JK, Levit RD, Sussman MA, Harvey RP, Yoon YS. Regeneration of infarcted mouse hearts by cardiovascular tissue formed via the direct reprogramming of mouse fibroblasts. Nat Biomed Eng 2021; 5:880-896. [PMID: 34426676 PMCID: PMC8809198 DOI: 10.1038/s41551-021-00783-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 07/13/2021] [Indexed: 02/07/2023]
Abstract
Fibroblasts can be directly reprogrammed into cardiomyocytes, endothelial cells or smooth muscle cells. Here we report the reprogramming of mouse tail-tip fibroblasts simultaneously into cells resembling these three cell types using the microRNA mimic miR-208b-3p, ascorbic acid and bone morphogenetic protein 4, as well as the formation of tissue-like structures formed by the directly reprogrammed cells. Implantation of the formed cardiovascular tissue into the infarcted hearts of mice led to the migration of reprogrammed cells to the injured tissue, reducing regional cardiac strain and improving cardiac function. The migrated endothelial cells and smooth muscle cells contributed to vessel formation, and the migrated cardiomyocytes, which initially displayed immature characteristics, became mature over time and formed gap junctions with host cardiomyocytes. Direct reprogramming of somatic cells to make cardiac tissue may aid the development of applications in cell therapy, disease modelling and drug discovery for cardiovascular diseases.
Collapse
Affiliation(s)
- Jaeyeaon Cho
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sangsung Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyein Lee
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Woongchan Rah
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hee Cheol Cho
- Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Nam Kyun Kim
- Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Seongho Bae
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Dong Hoon Shin
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Min Goo Lee
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - In-Hyun Park
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT, USA
| | - Yoshiaki Tanaka
- Department of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Eric Shin
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Hong Yi
- Robert P. Apkarian Integrated Electron Microscopy Core, Emory University, Atlanta, GA, USA
| | - Ji Woong Han
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Patrick Tae Joon Hwang
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ho-Wook Jun
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hun-Jun Park
- Division of Cardiology, Department of Internal Medicine, Seoul St Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Kyuwon Cho
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Sang Wook Lee
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Jae Kyung Jung
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Rebecca D Levit
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Mark A Sussman
- San Diego State University Heart Institute, San Diego State University, San Diego, CA, USA
- Department of Biology, San Diego State University, San Diego, CA, USA
| | - Richard P Harvey
- Victor Chang Cardiac Research Institute, St Vincent's Hospital, Darlinghurst, New South Wales, Australia
| | - Young-Sup Yoon
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA.
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
35
|
Litowczenko J, Woźniak-Budych MJ, Staszak K, Wieszczycka K, Jurga S, Tylkowski B. Milestones and current achievements in development of multifunctional bioscaffolds for medical application. Bioact Mater 2021; 6:2412-2438. [PMID: 33553825 PMCID: PMC7847813 DOI: 10.1016/j.bioactmat.2021.01.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/23/2020] [Accepted: 01/07/2021] [Indexed: 12/13/2022] Open
Abstract
Tissue engineering (TE) is a rapidly growing interdisciplinary field, which aims to restore or improve lost tissue function. Despite that TE was introduced more than 20 years ago, innovative and more sophisticated trends and technologies point to new challenges and development. Current challenges involve the demand for multifunctional bioscaffolds which can stimulate tissue regrowth by biochemical curves, biomimetic patterns, active agents and proper cell types. For those purposes especially promising are carefully chosen primary cells or stem cells due to its high proliferative and differentiation potential. This review summarized a variety of recently reported advanced bioscaffolds which present new functions by combining polymers, nanomaterials, bioactive agents and cells depending on its desired application. In particular necessity of study biomaterial-cell interactions with in vitro cell culture models, and studies using animals with in vivo systems were discuss to permit the analysis of full material biocompatibility. Although these bioscaffolds have shown a significant therapeutic effect in nervous, cardiovascular and muscle, tissue engineering, there are still many remaining unsolved challenges for scaffolds improvement.
Collapse
Affiliation(s)
- Jagoda Litowczenko
- NanoBioMedical Centre, Adam Mickiewicz University in Poznan, Wszechnicy Piastowskiej 3, Poznan, Poland
| | - Marta J. Woźniak-Budych
- NanoBioMedical Centre, Adam Mickiewicz University in Poznan, Wszechnicy Piastowskiej 3, Poznan, Poland
| | - Katarzyna Staszak
- Institute of Technology and Chemical Engineering, Poznan University of Technology, ul. Berdychowo 4, Poznan, Poland
| | - Karolina Wieszczycka
- Institute of Technology and Chemical Engineering, Poznan University of Technology, ul. Berdychowo 4, Poznan, Poland
| | - Stefan Jurga
- NanoBioMedical Centre, Adam Mickiewicz University in Poznan, Wszechnicy Piastowskiej 3, Poznan, Poland
| | - Bartosz Tylkowski
- Eurecat, Centre Tecnològic de Catalunya, Chemical Technologies Unit, Marcel·lí Domingo s/n, Tarragona, 43007, Spain
| |
Collapse
|
36
|
Adams E, McCloy R, Jordan A, Falconer K, Dykes IM. Direct Reprogramming of Cardiac Fibroblasts to Repair the Injured Heart. J Cardiovasc Dev Dis 2021; 8:72. [PMID: 34206355 PMCID: PMC8306371 DOI: 10.3390/jcdd8070072] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/14/2021] [Accepted: 06/18/2021] [Indexed: 02/06/2023] Open
Abstract
Coronary heart disease is a leading cause of mortality and morbidity. Those that survive acute myocardial infarction are at significant risk of subsequent heart failure due to fibrotic remodelling of the infarcted myocardium. By applying knowledge from the study of embryonic cardiovascular development, modern medicine offers hope for treatment of this condition through regeneration of the myocardium by direct reprogramming of fibrotic scar tissue. Here, we will review mechanisms of cell fate specification leading to the generation of cardiovascular cell types in the embryo and use this as a framework in which to understand direct reprogramming. Driving expression of a network of transcription factors, micro RNA or small molecule epigenetic modifiers can reverse epigenetic silencing, reverting differentiated cells to a state of induced pluripotency. The pluripotent state can be bypassed by direct reprogramming in which one differentiated cell type can be transdifferentiated into another. Transdifferentiating cardiac fibroblasts to cardiomyocytes requires a network of transcription factors similar to that observed in embryonic multipotent cardiac progenitors. There is some flexibility in the composition of this network. These studies raise the possibility that the failing heart could one day be regenerated by directly reprogramming cardiac fibroblasts within post-infarct scar tissue.
Collapse
Affiliation(s)
- Emma Adams
- Pharmacy and Biomolecular Science, Liverpool John Moores University, Liverpool L3 3AF, UK; (E.A.); (R.M.); (A.J.); (K.F.)
| | - Rachel McCloy
- Pharmacy and Biomolecular Science, Liverpool John Moores University, Liverpool L3 3AF, UK; (E.A.); (R.M.); (A.J.); (K.F.)
| | - Ashley Jordan
- Pharmacy and Biomolecular Science, Liverpool John Moores University, Liverpool L3 3AF, UK; (E.A.); (R.M.); (A.J.); (K.F.)
| | - Kaitlin Falconer
- Pharmacy and Biomolecular Science, Liverpool John Moores University, Liverpool L3 3AF, UK; (E.A.); (R.M.); (A.J.); (K.F.)
| | - Iain M. Dykes
- Pharmacy and Biomolecular Science, Liverpool John Moores University, Liverpool L3 3AF, UK; (E.A.); (R.M.); (A.J.); (K.F.)
- Liverpool Centre for Cardiovascular Science, Liverpool John Moores University, Liverpool L3 3AF, UK
| |
Collapse
|
37
|
Seguret M, Vermersch E, Jouve C, Hulot JS. Cardiac Organoids to Model and Heal Heart Failure and Cardiomyopathies. Biomedicines 2021; 9:563. [PMID: 34069816 PMCID: PMC8157277 DOI: 10.3390/biomedicines9050563] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/04/2021] [Accepted: 05/10/2021] [Indexed: 12/18/2022] Open
Abstract
Cardiac tissue engineering aims at creating contractile structures that can optimally reproduce the features of human cardiac tissue. These constructs are becoming valuable tools to model some of the cardiac functions, to set preclinical platforms for drug testing, or to alternatively be used as therapies for cardiac repair approaches. Most of the recent developments in cardiac tissue engineering have been made possible by important advances regarding the efficient generation of cardiac cells from pluripotent stem cells and the use of novel biomaterials and microfabrication methods. Different combinations of cells, biomaterials, scaffolds, and geometries are however possible, which results in different types of structures with gradual complexities and abilities to mimic the native cardiac tissue. Here, we intend to cover key aspects of tissue engineering applied to cardiology and the consequent development of cardiac organoids. This review presents various facets of the construction of human cardiac 3D constructs, from the choice of the components to their patterning, the final geometry of generated tissues, and the subsequent readouts and applications to model and treat cardiac diseases.
Collapse
Affiliation(s)
- Magali Seguret
- INSERM, PARCC, Université de Paris, F-75006 Paris, France; (M.S.); (E.V.); (C.J.)
| | - Eva Vermersch
- INSERM, PARCC, Université de Paris, F-75006 Paris, France; (M.S.); (E.V.); (C.J.)
| | - Charlène Jouve
- INSERM, PARCC, Université de Paris, F-75006 Paris, France; (M.S.); (E.V.); (C.J.)
| | - Jean-Sébastien Hulot
- INSERM, PARCC, Université de Paris, F-75006 Paris, France; (M.S.); (E.V.); (C.J.)
- CIC1418 and DMU CARTE, Assistance Publique Hôpitaux de Paris (AP-HP), Hôpital Européen Georges-Pompidou, F-75015 Paris, France
| |
Collapse
|
38
|
Zhao M, Nakada Y, Wei Y, Bian W, Chu Y, Borovjagin AV, Xie M, Zhu W, Nguyen T, Zhou Y, Serpooshan V, Walcott GP, Zhang J. Cyclin D2 Overexpression Enhances the Efficacy of Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes for Myocardial Repair in a Swine Model of Myocardial Infarction. Circulation 2021; 144:210-228. [PMID: 33951921 DOI: 10.1161/circulationaha.120.049497] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
BACKGROUND Human induced pluripotent stem cells with normal (wild-type) or upregulated (overexpressed) levels of CCND2 (cyclin D2) expression were differentiated into cardiomyocytes (CCND2WTCMs or CCND2OECMs, respectively) and injected into infarcted pig hearts. METHODS Acute myocardial infarction was induced by a 60-minute occlusion of the left anterior descending coronary artery. Immediately after reperfusion, CCND2WTCMs or CCND2OECMs (3×107 cells each) or an equivalent volume of the delivery vehicle was injected around the infarct border zone area. RESULTS The number of the engrafted CCND2OECMs exceeded that of the engrafted CCND2WTCMs from 6- to 8-fold, rising from 1 week to 4 weeks after implantation. In contrast to the treatment with the CCND2WTCMs or the delivery vehicle, the administration of CCND2OECM was associated with significantly improved left ventricular function, as revealed by magnetic resonance imaging. This correlated with reduction of infarct size, fibrosis, ventricular hypertrophy, and cardiomyocyte apoptosis, and increase of vascular density and arterial density, as per histologic analysis of the treated hearts. Expression of cell proliferation markers (eg, Ki67, phosphorylated histone 3, and Aurora B kinase) was also significantly upregulated in the recipient cardiomyocytes from the CCND2OECM-treated than from the CCND2WTCM-treated pigs. The cell proliferation rate and the hypoxia tolerance measured in cultured human induced pluripotent stem cell cardiomyocytes were significantly greater after treatment with exosomes isolated from the CCND2OECMs (CCND2OEExos) than from the CCND2WTCMs (CCND2WTExos). As demonstrated by our study, CCND2OEExos can also promote the proliferation activity of postnatal rat and adult mouse cardiomyocytes. A bulk miRNA sequencing analysis of CCND2OEExos versus CCND2WTExos identified 206 and 91 miRNAs that were significantly upregulated and downregulated, respectively. Gene ontology enrichment analysis identified significant differences in the expression profiles of miRNAs from various functional categories and pathways, including miRNAs implicated in cell-cycle checkpoints (G2/M and G1/S transitions), or the mechanism of cytokinesis. CONCLUSIONS We demonstrated that enhanced potency of CCND2OECMs promoted myocyte proliferation in both grafts and recipient tissue in a large mammal acute myocardial infarction model. These results suggest that CCND2OECMs transplantation may be a potential therapeutic strategy for the repair of infarcted hearts.
Collapse
Affiliation(s)
- Meng Zhao
- Department of Biomedical Engineering (M.Z., Y.N., Y.W., W.B., A.V.B., Y.Z., G.P.W., J.Z.), the University of Alabama at Birmingham
| | - Yuji Nakada
- Department of Biomedical Engineering (M.Z., Y.N., Y.W., W.B., A.V.B., Y.Z., G.P.W., J.Z.), the University of Alabama at Birmingham
| | - Yuhua Wei
- Department of Biomedical Engineering (M.Z., Y.N., Y.W., W.B., A.V.B., Y.Z., G.P.W., J.Z.), the University of Alabama at Birmingham
| | - Weihua Bian
- Department of Biomedical Engineering (M.Z., Y.N., Y.W., W.B., A.V.B., Y.Z., G.P.W., J.Z.), the University of Alabama at Birmingham
| | - Yuxin Chu
- Division of Cardiology, Department of Medicine (Y.C., M.X., G.P.W., J.Z.), the University of Alabama at Birmingham
| | - Anton V Borovjagin
- Department of Biomedical Engineering (M.Z., Y.N., Y.W., W.B., A.V.B., Y.Z., G.P.W., J.Z.), the University of Alabama at Birmingham
| | - Min Xie
- Division of Cardiology, Department of Medicine (Y.C., M.X., G.P.W., J.Z.), the University of Alabama at Birmingham
| | - Wuqiang Zhu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Mayo Clinic Arizona, Scottsdale (W.Z.)
| | - Thanh Nguyen
- School of Medicine and School of Engineering, and Informatics Institute (T.N.), the University of Alabama at Birmingham
| | - Yang Zhou
- Department of Biomedical Engineering (M.Z., Y.N., Y.W., W.B., A.V.B., Y.Z., G.P.W., J.Z.), the University of Alabama at Birmingham
| | - Vahid Serpooshan
- Wallace H. Coulter Department of Biomedical Engineering, Department of Pediatrics, Emory University and Georgia Institute of Technology, Atlanta (V.S.)
| | - Gregory P Walcott
- Department of Biomedical Engineering (M.Z., Y.N., Y.W., W.B., A.V.B., Y.Z., G.P.W., J.Z.), the University of Alabama at Birmingham.,Division of Cardiology, Department of Medicine (Y.C., M.X., G.P.W., J.Z.), the University of Alabama at Birmingham
| | - Jianyi Zhang
- Department of Biomedical Engineering (M.Z., Y.N., Y.W., W.B., A.V.B., Y.Z., G.P.W., J.Z.), the University of Alabama at Birmingham.,Division of Cardiology, Department of Medicine (Y.C., M.X., G.P.W., J.Z.), the University of Alabama at Birmingham
| |
Collapse
|
39
|
Chang YC, Mirhaidari G, Kelly J, Breuer C. Current Challenges and Solutions to Tissue Engineering of Large-scale Cardiac Constructs. Curr Cardiol Rep 2021; 23:47. [PMID: 33733317 DOI: 10.1007/s11886-021-01474-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/11/2021] [Indexed: 01/06/2023]
Abstract
PURPOSE OF REVIEW Large-scale tissue engineering of cardiac constructs is a rapidly advancing field; however, there are several barriers still associated with the creation and clinical application of large-scale engineered cardiac tissues. We provide an overview of the current challenges and recently (within the last 5 years) described promising solutions to overcoming said challenges. RECENT FINDINGS The five major criteria yet to be met for clinical application of engineered cardiac tissues are successful electrochemical/mechanical cell coupling, efficient maturation of cardiomyocytes, functional vascularization of large tissues, balancing appropriate immune response, and large-scale generation of constructs. Promising solutions include the use of carbon/graphene in conjunction with existing scaffold designs, utilization of biological hormones, 3D bioprinting, and gene editing. While some of the described barriers to generation of large-scale cardiac tissue have seen encouraging advancements, there is no solution that yet achieves all 5 described criteria. It is vital then to consider a combination of techniques to achieve the optimal construct. Critically, following the demonstration of a viable construct, there remain important considerations to address associated with good manufacturing practices and establishing a standard for clinical trials.
Collapse
Affiliation(s)
- Yu-Chun Chang
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Research Building III, Columbus, OH, 43215, USA.,Biomedical Sciences Graduate Program, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Gabriel Mirhaidari
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Research Building III, Columbus, OH, 43215, USA.,Biomedical Sciences Graduate Program, The Ohio State University College of Medicine, Columbus, OH, USA
| | - John Kelly
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Research Building III, Columbus, OH, 43215, USA.,Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA.,The Heart Center, Nationwide Children's Hospital, Columbus, OH, USA
| | - Christopher Breuer
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Research Building III, Columbus, OH, 43215, USA. .,Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA. .,Department of Surgery, Nationwide Children's Hospital, Columbus, OH, USA.
| |
Collapse
|
40
|
Portillo Esquivel LE, Zhang B. Application of Cell, Tissue, and Biomaterial Delivery in Cardiac Regenerative Therapy. ACS Biomater Sci Eng 2021; 7:1000-1021. [PMID: 33591735 DOI: 10.1021/acsbiomaterials.0c01805] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cardiovascular diseases (CVD) are the leading cause of death around the world, being responsible for 31.8% of all deaths in 2017 (Roth, G. A. et al. The Lancet 2018, 392, 1736-1788). The leading cause of CVD is ischemic heart disease (IHD), which caused 8.1 million deaths in 2013 (Benjamin, E. J. et al. Circulation 2017, 135, e146-e603). IHD occurs when coronary arteries in the heart are narrowed or blocked, preventing the flow of oxygen and blood into the cardiac muscle, which could provoke acute myocardial infarction (AMI) and ultimately lead to heart failure and death. Cardiac regenerative therapy aims to repair and refunctionalize damaged heart tissue through the application of (1) intramyocardial cell delivery, (2) epicardial cardiac patch, and (3) acellular biomaterials. In this review, we aim to examine these current approaches and challenges in the cardiac regenerative therapy field.
Collapse
Affiliation(s)
| | - Boyang Zhang
- Department of Chemical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada.,School of Biomedical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontaria L8S 4L8, Canada
| |
Collapse
|
41
|
He F, Tao T, Liu H, Wang Y, Cui K, Guo Y, Qin J. Controllable Fabrication of Composite Core-Shell Capsules at a Macroscale as Organoid Biocarriers. ACS APPLIED BIO MATERIALS 2021; 4:1584-1596. [PMID: 35014507 DOI: 10.1021/acsabm.0c01441] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The cell encapsulation technology is promising for generation of functional carriers with well-tailored structures for efficient transplantation and immunoprotection of cells/tissues. Stem cell organoids are highly potential for recapitulating the intricate architectures and functionalities of native organs and also providing an unlimited cell source for cellular replacement therapy. However, it remains challenging for loading the organoids with hundreds of micrometers size by current existing cell carriers. Herein, a simple and facile coextrusion strategy is developed for controllable fabrication of Ca-alginate/poly(ethylene imine) (Alg/PEI) macrocapsules for efficient encapsulation and cultivation of organoids. Human-induced pluripotent stem cell (hiPSC)-derived islet organoids are encapsulated in the aqueous compartments of the capsules and immunoisolated by a semipermeable Alg/PEI shell. Via electrostatic interactions, a PEI polyelectrolyte can be incorporated in the shell for restricting its swelling, thus effectively improving the stability of the capsules. The Alg/PEI macrocapsules are featured with desirable selective permeability for immunoisolation of antibodies from reaching the loaded organoids. Meanwhile, they also exhibit excellent permeability for mass transfer due to their well-defined core-shell structure. As such, the encapsulated islet organoids contain islet-specific multicellular components, with high viability and sensitive glucose-stimulated insulin secretion function. The proposed approach provides a versatile encapsulation system for tissue engineering and regenerative medicine applications.
Collapse
Affiliation(s)
- Fan He
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, P. R. China
| | - Tingting Tao
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, P. R. China.,University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Haitao Liu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, P. R. China.,University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Yaqing Wang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, P. R. China.,University of Chinese Academy of Sciences, Beijing 100049, P. R. China.,School of Chemistry, Dalian University of Technology, Dalian 116024, P. R. China
| | - Kangli Cui
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, P. R. China.,University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Yaqiong Guo
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, P. R. China.,University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Jianhua Qin
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, P. R. China.,University of Chinese Academy of Sciences, Beijing 100049, P. R. China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, P. R. China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, P. R. China
| |
Collapse
|
42
|
Yin F, Zhang X, Wang L, Wang Y, Zhu Y, Li Z, Tao T, Chen W, Yu H, Qin J. HiPSC-derived multi-organoids-on-chip system for safety assessment of antidepressant drugs. LAB ON A CHIP 2021; 21:571-581. [PMID: 33319899 DOI: 10.1039/d0lc00921k] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The poor predictive power of existing preclinical models has spurred efforts to develop human-relevant models for accurate assessment of drug safety. In this work, we developed a multi-organoids-on-a-chip system derived from human induced pluripotent stem cells (hiPSCs), which allows for the assessment of the cardiac safety of an antidepressant drug, following liver metabolism in vitro. This liver-heart organoids-on-chip device contains compartmentalized chambers separated by a porous membrane, which permits the co-culture of 3D human liver organoids in the upper multi-well chamber and cardiac organoids in the bottom micropillar array simultaneously. The co-cultured liver and heart organoids on chip maintained good viability and human organ-specific functions respectively, including the synthesis of albumin and urea of liver organoids, and the beating function of cardiac organoids. In particular, the liver organoids displayed proper metabolic capabilities with high expression of CYP450 enzyme genes. Clomipramine, a widely used antidepressant drug, can be metabolized into an active metabolite (desmethylclomipramine) through the hepatic CYP450 enzymes of liver organoids on chip identified by mass spectrometry. After exposure to 1 μM clomipramine in the liver chamber for 24 h and 48 h, the co-cultured heart organoids in the bottom layer showed significantly reduced cell viability, impaired functions of cardiac beating and calcium flux, indicating the hepatic metabolism-dependent cardiotoxicity induced by clomipramine. By combining stem cell biology and microengineered technology, this proposed hiPSC-derived multi-organoids-on-a-chip system can reflect human organ-specific functions, as well as the complex process of drug metabolism and responses at the multi-organ level. It may provide a novel platform for the assessment of drug effectiveness and safety in vitro.
Collapse
Affiliation(s)
- Fangchao Yin
- CAS Key Laboratory of SSAC, Department of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, China. and University of Chinese Academy of Sciences, Beijing, China
| | - Xu Zhang
- CAS Key Laboratory of SSAC, Department of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, China.
| | - Li Wang
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yaqing Wang
- CAS Key Laboratory of SSAC, Department of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, China. and University of Chinese Academy of Sciences, Beijing, China
| | - Yujuan Zhu
- CAS Key Laboratory of SSAC, Department of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, China. and University of Chinese Academy of Sciences, Beijing, China
| | - Zhongyu Li
- CAS Key Laboratory of SSAC, Department of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, China.
| | - Tingting Tao
- CAS Key Laboratory of SSAC, Department of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, China. and University of Chinese Academy of Sciences, Beijing, China
| | - Wenwen Chen
- CAS Key Laboratory of SSAC, Department of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, China. and University of Chinese Academy of Sciences, Beijing, China
| | - Hao Yu
- CAS Key Laboratory of SSAC, Department of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, China.
| | - Jianhua Qin
- CAS Key Laboratory of SSAC, Department of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, China. and University of Chinese Academy of Sciences, Beijing, China and Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China and CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
43
|
Silva AC, Pereira C, Fonseca ACRG, Pinto-do-Ó P, Nascimento DS. Bearing My Heart: The Role of Extracellular Matrix on Cardiac Development, Homeostasis, and Injury Response. Front Cell Dev Biol 2021; 8:621644. [PMID: 33511134 PMCID: PMC7835513 DOI: 10.3389/fcell.2020.621644] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 12/07/2020] [Indexed: 12/12/2022] Open
Abstract
The extracellular matrix (ECM) is an essential component of the heart that imparts fundamental cellular processes during organ development and homeostasis. Most cardiovascular diseases involve severe remodeling of the ECM, culminating in the formation of fibrotic tissue that is deleterious to organ function. Treatment schemes effective at managing fibrosis and promoting physiological ECM repair are not yet in reach. Of note, the composition of the cardiac ECM changes significantly in a short period after birth, concurrent with the loss of the regenerative capacity of the heart. This highlights the importance of understanding ECM composition and function headed for the development of more efficient therapies. In this review, we explore the impact of ECM alterations, throughout heart ontogeny and disease, on cardiac cells and debate available approaches to deeper insights on cell–ECM interactions, toward the design of new regenerative therapies.
Collapse
Affiliation(s)
- Ana Catarina Silva
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,INEB - Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Porto, Portugal.,Gladstone Institutes, San Francisco, CA, United States
| | - Cassilda Pereira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,INEB - Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - Ana Catarina R G Fonseca
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,INEB - Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - Perpétua Pinto-do-Ó
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,INEB - Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Porto, Portugal.,ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Diana S Nascimento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,INEB - Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Porto, Portugal.,ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| |
Collapse
|
44
|
Jiang Y, Sun SJ, Zhen Z, Wei R, Zhang N, Liao SY, Tse HF. Myocardial repair of bioengineered cardiac patches with decellularized placental scaffold and human-induced pluripotent stem cells in a rat model of myocardial infarction. Stem Cell Res Ther 2021; 12:13. [PMID: 33413626 PMCID: PMC7791702 DOI: 10.1186/s13287-020-02066-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 12/02/2020] [Indexed: 12/13/2022] Open
Abstract
Background The creation of a bioengineered cardiac patch (BCP) is a potential novel strategy for myocardial repair. Nevertheless, the ideal scaffold for BCP is unknown. Objective We investigated whether the decellularized placenta (DP) could serve as natural scaffold material to create a BCP for myocardial repair. Methods and results A BCP was created by seeding human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs; 1 × 106/cm2) onto DP. The functional and electrophysiological properties of the BCP were first characterized by in vitro analysis and optical mapping. Next, in vivo therapeutic efficacy of the BCP was evaluated in a rat model of myocardial infarction (MI), created by left descending coronary artery ligation (MI + BCP group), and compared with MI alone (MI group), transplantation of DP (MI + DP group), and hiPSC-CMs (MI + CM group). Cytokine profiling demonstrated that the BCP contained multiple growth and angiogenic factors, including vascular endothelial growth factor, platelet-derived growth factor, insulin-like growth factor-1, basic fibroblast growth factor, angiogenin, and angiopoietin-2. In vitro optical mapping showed that the BCP exhibited organized mechanical contraction and synchronized electrical propagation. RNA sequencing showed that DP enhanced the maturation of hiPSC-CMs compared with the monolayer of cultured hiPSC-CMs. At 4 weeks follow-up, the BCP significantly improved left ventricular (LV) function, as determined by LV ejection fraction, fractional shortening, + dP/dtmax, and end-systolic pressure-volume relationship, compared with the MI, MI + DP, and MI + CM groups. Moreover, histological examination revealed that engraftment of the BCP at the infarct zone decreased infarct size and increased cell retention and neovascularization compared with the MI, MI + DP, and MI + CM groups. Conclusions Our results demonstrate that a DP scaffold contains multiple growth and angiogenic factors that enhance the maturation and survival of seeded hiPSC-CMs. Transplantation of a BCP is superior to DP or hiPSC-CMs alone in reducing infarct size and improving cell retention and neovascularization, thus providing a novel therapy for myocardial repair following MI. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-020-02066-y.
Collapse
Affiliation(s)
- Yu Jiang
- Cardiology Division, Department of Medicine, Queen Mary Hospital, the University of Hong Kong, Hong Kong, SAR, China
| | - Si-Jia Sun
- Cardiology Division, Department of Medicine, Queen Mary Hospital, the University of Hong Kong, Hong Kong, SAR, China
| | - Zhe Zhen
- Cardiology Division, Department of Medicine, Queen Mary Hospital, the University of Hong Kong, Hong Kong, SAR, China
| | - Rui Wei
- Cardiology Division, Department of Medicine, Queen Mary Hospital, the University of Hong Kong, Hong Kong, SAR, China
| | - Nannan Zhang
- Cardiology Division, Department of Medicine, Queen Mary Hospital, the University of Hong Kong, Hong Kong, SAR, China
| | - Song-Yan Liao
- Cardiology Division, Department of Medicine, Queen Mary Hospital, the University of Hong Kong, Hong Kong, SAR, China. .,Shenzhen Institutes of Research and Innovation, the University of Hong Kong, Shenzhen, China.
| | - Hung-Fat Tse
- Cardiology Division, Department of Medicine, Queen Mary Hospital, the University of Hong Kong, Hong Kong, SAR, China. .,Department of Medicine, Shenzhen Hong Kong University Hospital, Shenzhen, China. .,Hong Kong-Guangdong Joint Laboratory on Stem Cell and Regenerative Medicine, the University of Hong Kong, Hong Kong, SAR, China.
| |
Collapse
|
45
|
Pawan KC, Mickey S, Rubia S, Yi H, Ge Z. Preseeding of Mesenchymal Stem Cells Increases Integration of an iPSC-Derived CM Sheet into a Cardiac Matrix. ACS Biomater Sci Eng 2020; 6:6808-6818. [PMID: 33320624 PMCID: PMC9841440 DOI: 10.1021/acsbiomaterials.0c00788] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Cell sheet technology has demonstrated great promise in delivering a large amount of therapeutic cells for tissue repair, including in the myocardium. However, the lack of host integration remains one of the key challenges in using cell sheets for cardiac repair. Paracrine factors secreted by mesenchymal stem cells (MSCs) have been reported to facilitate tissue repair and regeneration in a variety of ways. It has been demonstrated that paracrine factors from MSCs could enhance scaffold recellularization and vascularization. In this study, we used an in vitro cardiac matrix mimic platform to examine the effects of hMSCs preseeding on the interactions between cell sheets and cardiac matrix. The fabricated human induced pluripotent stem cells-derived cardiomyocyte sheets were attached to a decellularized porcine myocardium slice with or without preseeding of hMSCs. The hMSCs preseeding significantly enhanced the interactions between cardiomyocyte sheets and cardiac matrix in terms of cell migration distance, cell distribution, and mature vascular and cardiomyocyte marker expressions in the matrix. Growth factor and matrix metalloproteinases array analysis suggested that hMSCs- induced vascularization and MMPs regulation are the two possible mechanisms that lead to the improved CMs and cardiac matrix interactions. Further examination of these two mechanisms will enable the development of new approaches to facilitate transplanted cells for tissue repair.
Collapse
Affiliation(s)
- KC Pawan
- Department of Biomedical Engineering, The University of Akron, Akron, Ohio 44325, United States
| | - Shah Mickey
- Department of Biomedical Engineering and Department of Integrated Bioscience, The University of Akron, Akron, Ohio 44325, United States
| | - Shaik Rubia
- Department of Biomedical Engineering, The University of Akron, Akron, Ohio 44325, United States
| | - Hong Yi
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Zhang Ge
- Department of Biomedical Engineering, The University of Akron, Akron, Ohio 44325, United States
| |
Collapse
|
46
|
Yang Q, Fang J, Lei Z, Sluijter JPG, Schiffelers R. Repairing the heart: State-of the art delivery strategies for biological therapeutics. Adv Drug Deliv Rev 2020; 160:1-18. [PMID: 33039498 DOI: 10.1016/j.addr.2020.10.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 10/01/2020] [Accepted: 10/03/2020] [Indexed: 12/23/2022]
Abstract
Myocardial infarction (MI) is one of the leading causes of mortality worldwide. It is caused by an acute imbalance between oxygen supply and demand in the myocardium, usually caused by an obstruction in the coronary arteries. The conventional therapy is based on the application of (a combination of) anti-thrombotics, reperfusion strategies to open the occluded artery, stents and bypass surgery. However, numerous patients cannot fully recover after these interventions. In this context, new therapeutic methods are explored. Three decades ago, the first biologicals were tested to improve cardiac regeneration. Angiogenic proteins gained popularity as potential therapeutics. This is not straightforward as proteins are delicate molecules that in order to have a reasonably long time of activity need to be stabilized and released in a controlled fashion requiring advanced delivery systems. To ensure long-term expression, DNA vectors-encoding for therapeutic proteins have been developed. Here, the nuclear membrane proved to be a formidable barrier for efficient expression. Moreover, the development of delivery systems that can ensure entry in the target cell, and also correct intracellular trafficking towards the nucleus are essential. The recent introduction of mRNA as a therapeutic entity has provided an attractive intermediate: prolonged but transient expression from a cytoplasmic site of action. However, protection of the sensitive mRNA and correct delivery within the cell remains a challenge. This review focuses on the application of synthetic delivery systems that target the myocardium to stimulate cardiac repair using proteins, DNA or RNA.
Collapse
Affiliation(s)
- Qiangbing Yang
- Division LAB, CDL Research, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Juntao Fang
- Division Heart & Lungs, Department of Cardiology, Experimental Cardiology Laboratory, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Zhiyong Lei
- Division LAB, CDL Research, University Medical Center Utrecht, Utrecht, the Netherlands; Division Heart & Lungs, Department of Cardiology, Experimental Cardiology Laboratory, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Joost P G Sluijter
- Division Heart & Lungs, Department of Cardiology, Experimental Cardiology Laboratory, University Medical Center Utrecht, Utrecht, the Netherlands; Regenerative Medicine Utrecht, Circulatory Health Laboratory, Utrecht University, Utrecht, the Netherlands
| | - Raymond Schiffelers
- Division LAB, CDL Research, University Medical Center Utrecht, Utrecht, the Netherlands.
| |
Collapse
|
47
|
Sart S, Jeske R, Chen X, Ma T, Li Y. Engineering Stem Cell-Derived Extracellular Matrices: Decellularization, Characterization, and Biological Function. TISSUE ENGINEERING PART B-REVIEWS 2020; 26:402-422. [DOI: 10.1089/ten.teb.2019.0349] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Sébastien Sart
- Hydrodynamics Laboratory, CNRS UMR7646, Ecole Polytechnique, Palaiseau, France
- Laboratory of Physical Microfluidics and Bioengineering, Department of Genome and Genetics, Institut Pasteur, Paris, France
| | - Richard Jeske
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, USA
| | - Xingchi Chen
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, USA
| | - Teng Ma
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, USA
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, USA
| |
Collapse
|
48
|
Wang Y, Wang H, Deng P, Tao T, Liu H, Wu S, Chen W, Qin J. Modeling Human Nonalcoholic Fatty Liver Disease (NAFLD) with an Organoids-on-a-Chip System. ACS Biomater Sci Eng 2020; 6:5734-5743. [DOI: 10.1021/acsbiomaterials.0c00682] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Yaqing Wang
- School of Chemistry, Dalian University of Technology, Dalian 116024, China
- Division of Biotechnology, CAS Key Laboratory of SSAC, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Hui Wang
- Division of Biotechnology, CAS Key Laboratory of SSAC, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Pengwei Deng
- Division of Biotechnology, CAS Key Laboratory of SSAC, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Tingting Tao
- Division of Biotechnology, CAS Key Laboratory of SSAC, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Haitao Liu
- Division of Biotechnology, CAS Key Laboratory of SSAC, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Shuo Wu
- School of Chemistry, Dalian University of Technology, Dalian 116024, China
| | - Wenwen Chen
- Division of Biotechnology, CAS Key Laboratory of SSAC, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jianhua Qin
- Division of Biotechnology, CAS Key Laboratory of SSAC, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100049, China
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
49
|
Wang Y, Liu H, Zhang M, Wang H, Chen W, Qin J. One-step synthesis of composite hydrogel capsules to support liver organoid generation from hiPSCs. Biomater Sci 2020; 8:5476-5488. [PMID: 32914807 DOI: 10.1039/d0bm01085e] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Advances in biomaterials, especially in hydrogels, have offered great opportunities for stem cell organoid engineering with higher controllability and fidelity. Here, we propose a novel strategy for one-step synthesis of composite hydrogel capsules (CHCs) that enable engineering liver organoids from human induced pluripotent stem cells (hiPSCs) in an oil-free droplet microfluidic system. The CHCs composed of a fibrin hydrogel core and an alginate-chitosan composite shell are synthesized by an enzymatic crosslinking reaction and electrostatic complexation within stable aqueous emulsions. The proposed CHCs exhibit high uniformity with biocompatibility, stability and high-throughput properties, as well as defined compositions. Moreover, the established system enables 3D culture, differentiation and self-organized formation of liver organoids in a continuous process by encapsulating hepatocyte-like cells derived from hiPSCs. The encapsulated liver organoids consisting of hepatocyte- and cholangiocyte-like cells show favorable cell viability and growth with consistent size. Furthermore, they maintain proper liver-specific functions including urea synthesis and albumin secretion, replicating the key features of the human liver. By combining stem cell biology, defined hydrogels and the droplet microfluidic technique, the proposed system is easy-to-operate, scalable and stable to engineer stem cell organoids, which may offer a robust platform to advance organoid research and translational applications.
Collapse
Affiliation(s)
- Yaqing Wang
- School of Chemistry, Dalian University of Technology, Dalian 116024, P.R. China.
| | | | | | | | | | | |
Collapse
|
50
|
Yin F, Zhu Y, Wang H, Wang Y, Li D, Qin J. Microengineered hiPSC-Derived 3D Amnion Tissue Model to Probe Amniotic Inflammatory Responses under Bacterial Exposure. ACS Biomater Sci Eng 2020; 6:4644-4652. [PMID: 33455183 DOI: 10.1021/acsbiomaterials.0c00592] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Intra-amniotic infection is a common cause of preterm birth that can lead to adverse neonatal outcomes. Despite the basic and clinical significance, the study in normal and diseased human amnion is highly challenging due to the limited use of human primary tissues and the distinct divergence between animal models and human. Here, we established a microengineered hiPSC-derived amnion tissue model on a chip to investigate the inflammatory responses of amnion tissues to bacterial exposure. The microdevice consisted of two parallel channels with a middle matrix channel, creating a permissive microenvironment for amnion differentiation. Dissociated hiPSCs efficiently self-organized into cell cavity and finally differentiated into a polarized squamous amniotic epithelium on the chip under perfused 3D culture. When exposed to E. coli, amnion tissue exhibited significant functional impairments compared to the control, including induced cell apoptosis, disrupted cell junction integrity, and increased inflammatory factor secretion, recapitulating a series of characteristic clinical signs of intra-amniotic infection at an early stage. Together, this amnion-on-a-chip model provides a promising platform to investigate intrauterine inflammation in early gestation, indicating its potential applications in human embryology and reproductive medicine.
Collapse
Affiliation(s)
- Fangchao Yin
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023 China.,University of Chinese Academy of Sciences, Beijing 100049 China
| | - Yujuan Zhu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023 China.,University of Chinese Academy of Sciences, Beijing 100049 China
| | - Hui Wang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023 China.,University of Chinese Academy of Sciences, Beijing 100049 China
| | - Yaqing Wang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023 China.,University of Chinese Academy of Sciences, Beijing 100049 China
| | - Dong Li
- Dalian Municipal Women and Children's Medical Center, Dalian 116037 China
| | - Jianhua Qin
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023 China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101 China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031 China.,University of Chinese Academy of Sciences, Beijing 100049 China
| |
Collapse
|