1
|
Azam HMH, Rößling RI, Geithe C, Khan MM, Dinter F, Hanack K, Prüß H, Husse B, Roggenbuck D, Schierack P, Rödiger S. MicroRNA biomarkers as next-generation diagnostic tools for neurodegenerative diseases: a comprehensive review. Front Mol Neurosci 2024; 17:1386735. [PMID: 38883980 PMCID: PMC11177777 DOI: 10.3389/fnmol.2024.1386735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 04/12/2024] [Indexed: 06/18/2024] Open
Abstract
Neurodegenerative diseases (NDs) are characterized by abnormalities within neurons of the brain or spinal cord that gradually lose function, eventually leading to cell death. Upon examination of affected tissue, pathological changes reveal a loss of synapses, misfolded proteins, and activation of immune cells-all indicative of disease progression-before severe clinical symptoms become apparent. Early detection of NDs is crucial for potentially administering targeted medications that may delay disease advancement. Given their complex pathophysiological features and diverse clinical symptoms, there is a pressing need for sensitive and effective diagnostic methods for NDs. Biomarkers such as microRNAs (miRNAs) have been identified as potential tools for detecting these diseases. We explore the pivotal role of miRNAs in the context of NDs, focusing on Alzheimer's disease, Parkinson's disease, Multiple sclerosis, Huntington's disease, and Amyotrophic Lateral Sclerosis. The review delves into the intricate relationship between aging and NDs, highlighting structural and functional alterations in the aging brain and their implications for disease development. It elucidates how miRNAs and RNA-binding proteins are implicated in the pathogenesis of NDs and underscores the importance of investigating their expression and function in aging. Significantly, miRNAs exert substantial influence on post-translational modifications (PTMs), impacting not just the nervous system but a wide array of tissues and cell types as well. Specific miRNAs have been found to target proteins involved in ubiquitination or de-ubiquitination processes, which play a significant role in regulating protein function and stability. We discuss the link between miRNA, PTM, and NDs. Additionally, the review discusses the significance of miRNAs as biomarkers for early disease detection, offering insights into diagnostic strategies.
Collapse
Affiliation(s)
- Hafiz Muhammad Husnain Azam
- Institute of Biotechnology, Faculty of Environment and Natural Sciences, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany
| | - Rosa Ilse Rößling
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Department of Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Christiane Geithe
- Institute of Biotechnology, Faculty of Environment and Natural Sciences, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany
- Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology Cottbus - Senftenberg, The Brandenburg Medical School Theodor Fontane and the University of Potsdam, Berlin, Germany
| | - Muhammad Moman Khan
- Institute of Biotechnology, Faculty of Environment and Natural Sciences, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany
| | - Franziska Dinter
- Institute of Biotechnology, Faculty of Environment and Natural Sciences, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany
- PolyAn GmbH, Berlin, Germany
| | - Katja Hanack
- Institute of Biochemistry and Biology, University of Potsdam, Potsdam, Germany
| | - Harald Prüß
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Department of Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Britta Husse
- Institute of Biotechnology, Faculty of Environment and Natural Sciences, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany
| | - Dirk Roggenbuck
- Institute of Biotechnology, Faculty of Environment and Natural Sciences, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany
| | - Peter Schierack
- Institute of Biotechnology, Faculty of Environment and Natural Sciences, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany
| | - Stefan Rödiger
- Institute of Biotechnology, Faculty of Environment and Natural Sciences, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany
- Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology Cottbus - Senftenberg, The Brandenburg Medical School Theodor Fontane and the University of Potsdam, Berlin, Germany
| |
Collapse
|
2
|
Karakan MÇ, Ewoldt JK, Segarra AJ, Sundaram S, Wang MC, White AE, Chen CS, Ekinci KL. Geometry and length control of 3D engineered heart tissues using direct laser writing. LAB ON A CHIP 2024; 24:1685-1701. [PMID: 38317604 PMCID: PMC10929702 DOI: 10.1039/d3lc00752a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 01/23/2024] [Indexed: 02/07/2024]
Abstract
Geometry and mechanical characteristics of the environment surrounding the Engineered Heart Tissues (EHT) affect their structure and function. Here, we employed a 3D tissue culture platform fabricated using two-photon direct laser writing with a high degree of accuracy to control parameters that are relevant to EHT maturation. Using this platform, we first explore the effects of geometry based on two distinct shapes: a rectangular seeding well with two attachment sites, and a stadium-like seeding well with six attachment sites that are placed symmetrically along hemicylindrical membranes. The former geometry promotes uniaxial contraction of the tissues; the latter additionally induces diagonal fiber alignment. We systematically increase the length of the seeding wells for both configurations and observe a positive correlation between fiber alignment at the center of the EHTs and tissue length. With increasing length, an undesirable thinning and "necking" also emerge, leading to the failure of longer tissues over time. In the second step, we optimize the stiffness of the seeding wells and modify some of the attachment sites of the platform and the seeding parameters to achieve tissue stability for each length and geometry. Furthermore, we use the platform for electrical pacing and calcium imaging to evaluate the functional dynamics of EHTs as a function of frequency.
Collapse
Affiliation(s)
- M Çağatay Karakan
- Department of Mechanical Engineering, Boston University, Boston, MA 02215, USA.
- Photonics Center, Boston University, Boston, MA 02215, USA
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Jourdan K Ewoldt
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Addianette J Segarra
- Photonics Center, Boston University, Boston, MA 02215, USA
- Department of Biomedical Engineering, Polytechnic University of Puerto Rico, San Juan 00918, Puerto Rico
| | - Subramanian Sundaram
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Miranda C Wang
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
- Harvard-MIT Program in Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Alice E White
- Department of Mechanical Engineering, Boston University, Boston, MA 02215, USA.
- Photonics Center, Boston University, Boston, MA 02215, USA
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Division of Materials Science and Engineering, Boston University, Boston, Massachusetts 02215, USA
- Department of Physics, Boston University, Boston, MA 02215, USA
| | - Christopher S Chen
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Kamil L Ekinci
- Department of Mechanical Engineering, Boston University, Boston, MA 02215, USA.
- Photonics Center, Boston University, Boston, MA 02215, USA
- Division of Materials Science and Engineering, Boston University, Boston, Massachusetts 02215, USA
| |
Collapse
|
3
|
Ren Z, Harriot AD, Mair DB, Chung MK, Lee PHU, Kim DH. Biomanufacturing of 3D Tissue Constructs in Microgravity and their Applications in Human Pathophysiological Studies. Adv Healthc Mater 2023; 12:e2300157. [PMID: 37483106 DOI: 10.1002/adhm.202300157] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 06/27/2023] [Indexed: 07/25/2023]
Abstract
The growing interest in bioengineering in-vivo-like 3D functional tissues has led to novel approaches to the biomanufacturing process as well as expanded applications for these unique tissue constructs. Microgravity, as seen in spaceflight, is a unique environment that may be beneficial to the tissue-engineering process but cannot be completely replicated on Earth. Additionally, the expense and practical challenges of conducting human and animal research in space make bioengineered microphysiological systems an attractive research model. In this review, published research that exploits real and simulated microgravity to improve the biomanufacturing of a wide range of tissue types as well as those studies that use microphysiological systems, such as organ/tissue chips and multicellular organoids, for modeling human diseases in space are summarized. This review discusses real and simulated microgravity platforms and applications in tissue-engineered microphysiological systems across three topics: 1) application of microgravity to improve the biomanufacturing of tissue constructs, 2) use of tissue constructs fabricated in microgravity as models for human diseases on Earth, and 3) investigating the effects of microgravity on human tissues using biofabricated in vitro models. These current achievements represent important progress in understanding the physiological effects of microgravity and exploiting their advantages for tissue biomanufacturing.
Collapse
Affiliation(s)
- Zhanping Ren
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Anicca D Harriot
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Devin B Mair
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
| | | | - Peter H U Lee
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, 02912, USA
- Department of Cardiothoracic Surgery, Southcoast Health, Fall River, MA, 02720, USA
| | - Deok-Ho Kim
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Center for Microphysiological Systems, Johns Hopkins University, Baltimore, MD, 21205, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, 21218, USA
| |
Collapse
|
4
|
Singh BN, Yucel D, Garay BI, Tolkacheva EG, Kyba M, Perlingeiro RCR, van Berlo JH, Ogle BM. Proliferation and Maturation: Janus and the Art of Cardiac Tissue Engineering. Circ Res 2023; 132:519-540. [PMID: 36795845 PMCID: PMC9943541 DOI: 10.1161/circresaha.122.321770] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
During cardiac development and morphogenesis, cardiac progenitor cells differentiate into cardiomyocytes that expand in number and size to generate the fully formed heart. Much is known about the factors that regulate initial differentiation of cardiomyocytes, and there is ongoing research to identify how these fetal and immature cardiomyocytes develop into fully functioning, mature cells. Accumulating evidence indicates that maturation limits proliferation and conversely proliferation occurs rarely in cardiomyocytes of the adult myocardium. We term this oppositional interplay the proliferation-maturation dichotomy. Here we review the factors that are involved in this interplay and discuss how a better understanding of the proliferation-maturation dichotomy could advance the utility of human induced pluripotent stem cell-derived cardiomyocytes for modeling in 3-dimensional engineered cardiac tissues to obtain truly adult-level function.
Collapse
Affiliation(s)
- Bhairab N. Singh
- Department of Pediatrics, University of Minnesota, MN, USA
- Department of Biomedical Engineering, University of Minnesota, MN, USA
- Stem Cell Institute, University of Minnesota, MN, USA
| | - Dogacan Yucel
- Stem Cell Institute, University of Minnesota, MN, USA
- Department of Medicine, Cardiovascular Division, University of Minnesota, MN, USA
- Lillehei Heart Institute, University of Minnesota, MN, USA
| | - Bayardo I. Garay
- Stem Cell Institute, University of Minnesota, MN, USA
- Department of Medicine, Cardiovascular Division, University of Minnesota, MN, USA
- Lillehei Heart Institute, University of Minnesota, MN, USA
- Medical Scientist Training Program, University of Minnesota Medical School, MN, USA
| | - Elena G. Tolkacheva
- Department of Biomedical Engineering, University of Minnesota, MN, USA
- Lillehei Heart Institute, University of Minnesota, MN, USA
- Institute for Engineering in Medicine, University of Minnesota, MN, USA
| | - Michael Kyba
- Department of Pediatrics, University of Minnesota, MN, USA
- Stem Cell Institute, University of Minnesota, MN, USA
- Lillehei Heart Institute, University of Minnesota, MN, USA
| | - Rita C. R. Perlingeiro
- Stem Cell Institute, University of Minnesota, MN, USA
- Department of Medicine, Cardiovascular Division, University of Minnesota, MN, USA
- Lillehei Heart Institute, University of Minnesota, MN, USA
| | - Jop H. van Berlo
- Stem Cell Institute, University of Minnesota, MN, USA
- Department of Medicine, Cardiovascular Division, University of Minnesota, MN, USA
- Lillehei Heart Institute, University of Minnesota, MN, USA
| | - Brenda M. Ogle
- Department of Pediatrics, University of Minnesota, MN, USA
- Department of Biomedical Engineering, University of Minnesota, MN, USA
- Stem Cell Institute, University of Minnesota, MN, USA
- Lillehei Heart Institute, University of Minnesota, MN, USA
- Institute for Engineering in Medicine, University of Minnesota, MN, USA
- Masonic Cancer Center, University of Minnesota, MN, USA
| |
Collapse
|
5
|
Zhu L, Liu K, Feng Q, Liao Y. Cardiac Organoids: A 3D Technology for Modeling Heart Development and Disease. Stem Cell Rev Rep 2022; 18:2593-2605. [PMID: 35525908 DOI: 10.1007/s12015-022-10385-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2022] [Indexed: 12/14/2022]
Abstract
Cardiac organoids (COs) are miniaturized and simplified organ structures that can be used in heart development biology, drug screening, disease modeling, and regenerative medicine. This cardiac organoid (CO) model is revolutionizing our perspective on answering major cardiac physiology and pathology issues. Recently, many research groups have reported various methods for modeling the heart in vitro. However, there are differences in methodologies and concepts. In this review, we discuss the recent advances in cardiac organoid technologies derived from human embryonic stem cells (hESCs) and human-induced pluripotent stem cells (hiPSCs), with a focus on the summary of methods for organoid generation. In addition, we introduce CO applications in modeling heart development and cardiovascular diseases and discuss the prospects for and common challenges of CO that still need to be addressed. A detailed understanding of the development of CO will help us design better methods, explore and expand its application in the cardiovascular field.
Collapse
Affiliation(s)
- Liyuan Zhu
- Xiamen Key Laboratory of Cardiovascular Disease, Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Kui Liu
- Xiamen Key Laboratory of Cardiovascular Disease, Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Qi Feng
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yingnan Liao
- Xiamen Key Laboratory of Cardiovascular Disease, Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.
| |
Collapse
|
6
|
Pisanu A, Reid G, Fusco D, Sileo A, Robles Diaz D, Tarhini H, Putame G, Massai D, Isu G, Marsano A. Bizonal cardiac engineered tissues with differential maturation features in a mid-throughput multimodal bioreactor. iScience 2022; 25:104297. [PMID: 35586070 PMCID: PMC9108516 DOI: 10.1016/j.isci.2022.104297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 11/26/2021] [Accepted: 04/21/2022] [Indexed: 12/03/2022] Open
Abstract
Functional three-dimensional (3D) engineered cardiac tissue (ECT) models are essential for effective drug screening and biological studies. Application of physiological cues mimicking those typical of the native myocardium is known to promote the cardiac maturation and functionality in vitro. Commercially available bioreactors can apply one physical force type at a time and often in a restricted loading range. To overcome these limitations, a millimetric-scale microscope-integrated bioreactor was developed to deliver multiple biophysical stimuli to ECTs. In this study, we showed that the single application of auxotonic loading (passive) generated a bizonal ECT with a unique cardiac maturation pattern. Throughout the statically cultured constructs and in the ECT region exposed to high passive loading, cardiomyocytes predominantly displayed a round morphology and poor contractility ability. The ECT region with a low passive mechanical stimulation instead showed both rat- and human-origin cardiac cell maturation and organization, as well as increased ECT functionality. Mid-throughput culture platform to engineer reproducible 3D cardiac in vitro models 3D culture under multiphysical stimuli mimicking the in vivo heart environment Passive loading leads to bizonal constructs with different cardiac maturation stages
Collapse
|
7
|
Abstract
Cardiac congenital disabilities are the most common organ malformations, but we still do not understand how they arise in the human embryo. Moreover, although cardiovascular disease is the most common cause of death globally, the development of new therapies is lagging compared with other fields. One major bottleneck hindering progress is the lack of self-organizing human cardiac models that recapitulate key aspects of human heart development, physiology and disease. Current in vitro cardiac three-dimensional systems are either engineered constructs or spherical aggregates of cardiomyocytes and other cell types. Although tissue engineering enables the modeling of some electro-mechanical properties, it falls short of mimicking heart development, morphogenetic defects and many clinically relevant aspects of cardiomyopathies. Here, we review different approaches and recent efforts to overcome these challenges in the field using a new generation of self-organizing embryonic and cardiac organoids.
Collapse
Affiliation(s)
- Pablo Hofbauer
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter, Dr. Bohr Gasse 3, 1030 Vienna, Austria
| | - Stefan M Jahnel
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter, Dr. Bohr Gasse 3, 1030 Vienna, Austria
| | - Sasha Mendjan
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter, Dr. Bohr Gasse 3, 1030 Vienna, Austria
| |
Collapse
|
8
|
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: 7] [Impact Index Per Article: 1.8] [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
|
9
|
Song Y, Wang H, Yue F, Lv Q, Cai B, Dong N, Wang Z, Wang L. Silk-Based Biomaterials for Cardiac Tissue Engineering. Adv Healthc Mater 2020; 9:e2000735. [PMID: 32939999 DOI: 10.1002/adhm.202000735] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 08/29/2020] [Indexed: 12/18/2022]
Abstract
Cardiovascular diseases are one of the leading causes of death globally. Among various cardiovascular diseases, myocardial infarction is an important one. Compared with conventional treatments, cardiac tissue engineering provides an alternative to repair and regenerate the injured tissue. Among various types of materials used for tissue engineering applications, silk biomaterials have been increasingly utilized due to their biocompatibility, biological functions, and many favorable physical/chemical properties. Silk biomaterials are often used alone or in combination with other materials in the forms of patches or hydrogels, and serve as promising delivery systems for bioactive compounds in tissue engineering repair scenarios. This review focuses primarily on the promising characteristics of silk biomaterials and their recent advances in cardiac tissue engineering.
Collapse
Affiliation(s)
- Yu Song
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Huifang Wang
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Feifei Yue
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qiying Lv
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Bo Cai
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Nianguo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zheng Wang
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Lin Wang
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| |
Collapse
|
10
|
Majid QA, Fricker ATR, Gregory DA, Davidenko N, Hernandez Cruz O, Jabbour RJ, Owen TJ, Basnett P, Lukasiewicz B, Stevens M, Best S, Cameron R, Sinha S, Harding SE, Roy I. Natural Biomaterials for Cardiac Tissue Engineering: A Highly Biocompatible Solution. Front Cardiovasc Med 2020; 7:554597. [PMID: 33195451 PMCID: PMC7644890 DOI: 10.3389/fcvm.2020.554597] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 09/10/2020] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular diseases (CVD) constitute a major fraction of the current major global diseases and lead to about 30% of the deaths, i.e., 17.9 million deaths per year. CVD include coronary artery disease (CAD), myocardial infarction (MI), arrhythmias, heart failure, heart valve diseases, congenital heart disease, and cardiomyopathy. Cardiac Tissue Engineering (CTE) aims to address these conditions, the overall goal being the efficient regeneration of diseased cardiac tissue using an ideal combination of biomaterials and cells. Various cells have thus far been utilized in pre-clinical studies for CTE. These include adult stem cell populations (mesenchymal stem cells) and pluripotent stem cells (including autologous human induced pluripotent stem cells or allogenic human embryonic stem cells) with the latter undergoing differentiation to form functional cardiac cells. The ideal biomaterial for cardiac tissue engineering needs to have suitable material properties with the ability to support efficient attachment, growth, and differentiation of the cardiac cells, leading to the formation of functional cardiac tissue. In this review, we have focused on the use of biomaterials of natural origin for CTE. Natural biomaterials are generally known to be highly biocompatible and in addition are sustainable in nature. We have focused on those that have been widely explored in CTE and describe the original work and the current state of art. These include fibrinogen (in the context of Engineered Heart Tissue, EHT), collagen, alginate, silk, and Polyhydroxyalkanoates (PHAs). Amongst these, fibrinogen, collagen, alginate, and silk are isolated from natural sources whereas PHAs are produced via bacterial fermentation. Overall, these biomaterials have proven to be highly promising, displaying robust biocompatibility and, when combined with cells, an ability to enhance post-MI cardiac function in pre-clinical models. As such, CTE has great potential for future clinical solutions and hence can lead to a considerable reduction in mortality rates due to CVD.
Collapse
Affiliation(s)
- Qasim A. Majid
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Annabelle T. R. Fricker
- Department of Material Science and Engineering, Faculty of Engineering, University of Sheffield, Sheffield, United Kingdom
| | - David A. Gregory
- Department of Material Science and Engineering, Faculty of Engineering, University of Sheffield, Sheffield, United Kingdom
| | - Natalia Davidenko
- Department of Materials Science and Metallurgy, Cambridge Centre for Medical Materials, University of Cambridge, Cambridge, United Kingdom
| | - Olivia Hernandez Cruz
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Department of Bioengineering, Department of Materials, IBME, Faculty of Engineering, Imperial College London, United Kingdom
| | - Richard J. Jabbour
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Thomas J. Owen
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Pooja Basnett
- Applied Biotechnology Research Group, School of Life Sciences, College of Liberal Arts and Sciences, University of Westminster, London, United Kingdom
| | - Barbara Lukasiewicz
- Applied Biotechnology Research Group, School of Life Sciences, College of Liberal Arts and Sciences, University of Westminster, London, United Kingdom
| | - Molly Stevens
- Department of Bioengineering, Department of Materials, IBME, Faculty of Engineering, Imperial College London, United Kingdom
| | - Serena Best
- Department of Materials Science and Metallurgy, Cambridge Centre for Medical Materials, University of Cambridge, Cambridge, United Kingdom
| | - Ruth Cameron
- Department of Materials Science and Metallurgy, Cambridge Centre for Medical Materials, University of Cambridge, Cambridge, United Kingdom
| | - Sanjay Sinha
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Sian E. Harding
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Ipsita Roy
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Department of Material Science and Engineering, Faculty of Engineering, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
11
|
Intensive Care Unit-Acquired Weakness: Not just Another Muscle Atrophying Condition. Int J Mol Sci 2020; 21:ijms21217840. [PMID: 33105809 PMCID: PMC7660068 DOI: 10.3390/ijms21217840] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/18/2020] [Accepted: 10/19/2020] [Indexed: 02/07/2023] Open
Abstract
Intensive care unit-acquired weakness (ICUAW) occurs in critically ill patients stemming from the critical illness itself, and results in sustained disability long after the ICU stay. Weakness can be attributed to muscle wasting, impaired contractility, neuropathy, and major pathways associated with muscle protein degradation such as the ubiquitin proteasome system and dysregulated autophagy. Furthermore, it is characterized by the preferential loss of myosin, a distinct feature of the condition. While many risk factors for ICUAW have been identified, effective interventions to offset these changes remain elusive. In addition, our understanding of the mechanisms underlying the long-term, sustained weakness observed in a subset of patients after discharge is minimal. Herein, we discuss the various proposed pathways involved in the pathophysiology of ICUAW, with a focus on the mechanisms underpinning skeletal muscle wasting and impaired contractility, and the animal models used to study them. Furthermore, we will explore the contributions of inflammation, steroid use, and paralysis to the development of ICUAW and how it pertains to those with the corona virus disease of 2019 (COVID-19). We then elaborate on interventions tested as a means to offset these decrements in muscle function that occur as a result of critical illness, and we propose new strategies to explore the molecular mechanisms of ICUAW, including serum-related biomarkers and 3D human skeletal muscle culture models.
Collapse
|
12
|
Massai D, Pisani G, Isu G, Rodriguez Ruiz A, Cerino G, Galluzzi R, Pisanu A, Tonoli A, Bignardi C, Audenino AL, Marsano A, Morbiducci U. Bioreactor Platform for Biomimetic Culture and in situ Monitoring of the Mechanical Response of in vitro Engineered Models of Cardiac Tissue. Front Bioeng Biotechnol 2020; 8:733. [PMID: 32766218 PMCID: PMC7381147 DOI: 10.3389/fbioe.2020.00733] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 06/10/2020] [Indexed: 12/17/2022] Open
Abstract
In the past two decades, relevant advances have been made in the generation of engineered cardiac constructs to be used as functional in vitro models for cardiac research or drug testing, and with the ultimate but still challenging goal of repairing the damaged myocardium. To support cardiac tissue generation and maturation in vitro, the application of biomimetic physical stimuli within dedicated bioreactors is crucial. In particular, cardiac-like mechanical stimulation has been demonstrated to promote development and maturation of cardiac tissue models. Here, we developed an automated bioreactor platform for tunable cyclic stretch and in situ monitoring of the mechanical response of in vitro engineered cardiac tissues. To demonstrate the bioreactor platform performance and to investigate the effects of cyclic stretch on construct maturation and contractility, we developed 3D annular cardiac tissue models based on neonatal rat cardiac cells embedded in fibrin hydrogel. The constructs were statically pre-cultured for 5 days and then exposed to 4 days of uniaxial cyclic stretch (sinusoidal waveform, 10% strain, 1 Hz) within the bioreactor. Explanatory biological tests showed that cyclic stretch promoted cardiomyocyte alignment, maintenance, and maturation, with enhanced expression of typical mature cardiac markers compared to static controls. Moreover, in situ monitoring showed increasing passive force of the constructs along the dynamic culture. Finally, only the stretched constructs were responsive to external electrical pacing with synchronous and regular contractile activity, further confirming that cyclic stretching was instrumental for their functional maturation. This study shows that the proposed bioreactor platform is a reliable device for cyclic stretch culture and in situ monitoring of the passive mechanical response of the cultured constructs. The innovative feature of acquiring passive force measurements in situ and along the culture allows monitoring the construct maturation trend without interrupting the culture, making the proposed device a powerful tool for in vitro investigation and ultimately production of functional engineered cardiac constructs.
Collapse
Affiliation(s)
- Diana Massai
- PolitoBIOMed Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy.,Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, Turin, Italy
| | - Giuseppe Pisani
- PolitoBIOMed Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy.,Department of Surgery, University Hospital of Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Giuseppe Isu
- PolitoBIOMed Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy.,Department of Surgery, University Hospital of Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Andres Rodriguez Ruiz
- PolitoBIOMed Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - Giulia Cerino
- Department of Surgery, University Hospital of Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Renato Galluzzi
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - Alessia Pisanu
- Department of Surgery, University Hospital of Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Andrea Tonoli
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - Cristina Bignardi
- PolitoBIOMed Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy.,Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, Turin, Italy
| | - Alberto L Audenino
- PolitoBIOMed Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy.,Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, Turin, Italy
| | - Anna Marsano
- Department of Surgery, University Hospital of Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Umberto Morbiducci
- PolitoBIOMed Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy.,Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, Turin, Italy
| |
Collapse
|
13
|
Abstract
Stretchable conductive fabric (SCF) is a durable nontoxic textile material coated or blended with conductive metals. Unlike solid metal, SCF effectively conducts electricity with low resistance and maintains conductance when stretched. Thus, we hypothesized that SCF electrodes are more suitable for cardiac electrophysiology applications in beating hearts than traditional solid metal electrodes. Accordingly, we developed a straightforward protocol for fabricating customized SCF electrodes and then assessed their ability to electrically stimulate and record electrical signals from beating hearts. Compared to flexible copper electrodes, SCF electrodes had similar electrical resistance (112.50 ± 25.81 vs 157.85 ± 17.06 Ω, p = 0.09), activated cardiac tissue with lower stimulus strength (27.25 ± 3.52 vs 15.35 ± 2.15 mA, p = 0.0001), recorded stable electrograms with a higher signal-to-noise ratio (20.54 ± 1.09 vs 13.35 ± 1.46 dB, p = 0.04), and were noncorrosive and harmless to cardiac tissue or vasculature. These results support the use of SCF over metal electrodes for a wide range of cardiac electrophysiology applications in the beating heart.
Collapse
|
14
|
Popular three-dimensional models: Advantages for cancer, Alzheimer's and cardiovascular diseases. Tissue Cell 2020; 65:101367. [PMID: 32746982 DOI: 10.1016/j.tice.2020.101367] [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] [Received: 02/27/2020] [Revised: 04/08/2020] [Accepted: 04/09/2020] [Indexed: 11/21/2022]
Abstract
The increasing prevalence of non-communicable diseases, namely cancer, Alzheimer's (AD) and cardiovascular diseases (CVDs), worldwide continues to be a major health burden. Research attempts have been made to understand the pathophysiology and develop effective therapeutic agents for these diseases using conventional in vitro and ex vivo models. Due to the complexity of human disease mechanisms, often these models fail to recapitulate clinically relevant pathologies. As such, interests are arising in the exploration of three-dimensional (3D) in-vitro models, which create an artificial environment to closely mimic in vivo human conditions. Several studies have developed 3D models for cancer, AD and CVD research which can greatly improve the understanding of biological mechanisms and mirror clinical drug activities. Thus, 3D cultures may provide new in-vitro models that recapitulate the architecture and biological mechanisms of human diseases prior to the need for the use of sentient animals.
Collapse
|
15
|
|
16
|
Fusto A, Moyle LA, Gilbert PM, Pegoraro E. Cored in the act: the use of models to understand core myopathies. Dis Model Mech 2019; 12:dmm041368. [PMID: 31874912 PMCID: PMC6955215 DOI: 10.1242/dmm.041368] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The core myopathies are a group of congenital myopathies with variable clinical expression - ranging from early-onset skeletal-muscle weakness to later-onset disease of variable severity - that are identified by characteristic 'core-like' lesions in myofibers and the presence of hypothonia and slowly or rather non-progressive muscle weakness. The genetic causes are diverse; central core disease is most often caused by mutations in ryanodine receptor 1 (RYR1), whereas multi-minicore disease is linked to pathogenic variants of several genes, including selenoprotein N (SELENON), RYR1 and titin (TTN). Understanding the mechanisms that drive core development and muscle weakness remains challenging due to the diversity of the excitation-contraction coupling (ECC) proteins involved and the differential effects of mutations across proteins. Because of this, the use of representative models expressing a mature ECC apparatus is crucial. Animal models have facilitated the identification of disease progression mechanisms for some mutations and have provided evidence to help explain genotype-phenotype correlations. However, many unanswered questions remain about the common and divergent pathological mechanisms that drive disease progression, and these mechanisms need to be understood in order to identify therapeutic targets. Several new transgenic animals have been described recently, expanding the spectrum of core myopathy models, including mice with patient-specific mutations. Furthermore, recent developments in 3D tissue engineering are expected to enable the study of core myopathy disease progression and the effects of potential therapeutic interventions in the context of human cells. In this Review, we summarize the current landscape of core myopathy models, and assess the hurdles and opportunities of future modeling strategies.
Collapse
Affiliation(s)
- Aurora Fusto
- Department of Neuroscience, University of Padua, Padua 35128, Italy
| | - Louise A Moyle
- Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada
- Institute of Biomaterials and Biochemical Engineering, University of Toronto, Toronto, ON M5S3G9, Canada
| | - Penney M Gilbert
- Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada
- Institute of Biomaterials and Biochemical Engineering, University of Toronto, Toronto, ON M5S3G9, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S3G5, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON M5S1A8, Canada
| | - Elena Pegoraro
- Department of Neuroscience, University of Padua, Padua 35128, Italy
| |
Collapse
|
17
|
Dissection of heterocellular cross-talk in vascularized cardiac tissue mimetics. J Mol Cell Cardiol 2019; 138:269-282. [PMID: 31866374 DOI: 10.1016/j.yjmcc.2019.12.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 12/07/2019] [Accepted: 12/10/2019] [Indexed: 02/07/2023]
Abstract
Cellular specialization and interaction with other cell types in cardiac tissue is essential for the coordinated function of cell populations in the heart. The complex interplay between cardiomyocytes, endothelial cells and fibroblasts is necessary for adaptation but can also lead to pathophysiological remodeling. To understand this complex interplay, we developed 3D vascularized cardiac tissue mimetics (CTM) to study heterocellular cross-talk in hypertrophic, hypoxic and fibrogenic environments. This 3D platform responds to physiologic and pathologic stressors and mimics the microenvironment of diseased tissue. In combination with endothelial cell fluorescence reporters, these cardiac tissue mimetics can be used to precisely visualize and quantify cellular and functional responses upon stress stimulation. Utilizing this platform, we demonstrate that stimulation of α/β-adrenergic receptors with phenylephrine (PE) promotes cardiomyocyte hypertrophy, metabolic maturation and vascularization of CTMs. Increased vascularization was promoted by conditioned medium of PE-stimulated cardiomyocytes and blocked by inhibiting VEGF or upon β-adrenergic receptor antagonist treatment, demonstrating cardiomyocyte-endothelial cross-talk. Pathophysiological stressors such as severe hypoxia reduced angiogenic sprouting and increased cell death, while TGF β2 stimulation increased collagen deposition concomitant to endothelial-to-mesenchymal transition. In sum, we have developed a cardiac 3D culture system that reflects native cardiac tissue function, metabolism and morphology - and for the first time enables the tracking and analysis of cardiac vascularization dynamics in physiology and pathology.
Collapse
|
18
|
Wragg NM, Mosqueira D, Blokpeol-Ferreras L, Capel A, Player DJ, Martin NRW, Liu Y, Lewis MP. Development of a 3D Tissue-Engineered Skeletal Muscle and Bone Co-culture System. Biotechnol J 2019; 15:e1900106. [PMID: 31468704 DOI: 10.1002/biot.201900106] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 07/05/2019] [Indexed: 12/26/2022]
Abstract
In vitro 3D tissue-engineered (TE) structures have been shown to better represent in vivo tissue morphology and biochemical pathways than monolayer culture, and are less ethically questionable than animal models. However, to create systems with even greater relevance, multiple integrated tissue systems should be recreated in vitro. In the present study, the effects and conditions most suitable for the co-culture of TE skeletal muscle and bone are investigated. High-glucose Dulbecco's modified Eagle medium (HG-DMEM) supplemented with 20% fetal bovine serum followed by HG-DMEM with 2% horse serum is found to enable proliferation of both C2C12 muscle precursor cells and TE85 human osteosarcoma cells, fusion of C2C12s into myotubes, as well as an upregulation of RUNX2/CBFa1 in TE85s. Myotube formation is also evident within indirect contact monolayer cultures. Finally, in 3D co-cultures, TE85 collagen/hydroxyapatite constructs have significantly greater expression of RUNX2/CBFa1 and osteocalcin/BGLAP in the presence of collagen-based C2C12 skeletal muscle constructs; however, fusion within these constructs appears reduced. This work demonstrates the first report of the simultaneous co-culture and differentiation of 3D TE skeletal muscle and bone, and represents a significant step toward a full in vitro 3D musculoskeletal junction model.
Collapse
Affiliation(s)
- Nicholas M Wragg
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK.,Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Loughborough, UK
| | - Diogo Mosqueira
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Lia Blokpeol-Ferreras
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Andrew Capel
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Darren J Player
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK.,Institute of Orthopaedics and Musculoskeletal Sciences, RNOH University College London, Stanmore, UK
| | - Neil R W Martin
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Yang Liu
- Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Loughborough, UK
| | - Mark P Lewis
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| |
Collapse
|
19
|
Gude NA, Sussman MA. Cardiac regenerative therapy: Many paths to repair. Trends Cardiovasc Med 2019; 30:338-343. [PMID: 31515053 DOI: 10.1016/j.tcm.2019.08.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 08/14/2019] [Accepted: 08/29/2019] [Indexed: 12/17/2022]
Abstract
Cardiovascular disease remains the primary cause of death in the United States and in most nations worldwide, despite ongoing intensive efforts to promote cardiac health and treat heart failure. Replacing damaged myocardium represents perhaps the most promising treatment strategy, but also the most challenging given that the adult mammalian heart is notoriously resistant to endogenous repair. Cardiac regeneration following pathologic challenge would require proliferation of surviving tissue, expansion and differentiation of resident progenitors, or transdifferentiation of exogenously applied progenitor cells into functioning myocardium. Adult cardiomyocyte proliferation has been the focus of investigation for decades, recently enjoying a renaissance of interest as a therapeutic strategy for reversing cardiomyocyte loss due in large part to ongoing controversies and frustrations with myocardial cell therapy outcomes. The promise of cardiac cell therapy originated with reports of resident adult cardiac stem cells that could be isolated, expanded and reintroduced into damaged myocardium, producing beneficial effects in preclinical animal models. Despite modest functional improvements, Phase I clinical trials using autologous cardiac derived cells have proven safe and effective, setting the stage for an ongoing multi-center Phase II trial combining autologous cardiac stem cell types to enhance beneficial effects. This overview will examine the history of these two approaches for promoting cardiac repair and attempt to provide context for current and future directions in cardiac regenerative research.
Collapse
Affiliation(s)
- Natalie A Gude
- SDSU Heart Institute and Biology Department, San Diego State University, 5500 Campanile Drive, San Diego, CA 92182, USA
| | - Mark A Sussman
- SDSU Heart Institute and Biology Department, San Diego State University, 5500 Campanile Drive, San Diego, CA 92182, USA.
| |
Collapse
|
20
|
Owen TJ, Harding SE. Multi-cellularity in cardiac tissue engineering, how close are we to native heart tissue? J Muscle Res Cell Motil 2019; 40:151-157. [PMID: 31222588 PMCID: PMC6726707 DOI: 10.1007/s10974-019-09528-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 06/15/2019] [Indexed: 12/25/2022]
Abstract
Tissue engineering is a complex field where the elements of biology and engineering are combined in an attempt to recapitulate the native environment of the body. Tissue engineering has shown one thing categorically; that the human body is extremely complex and it is truly a difficult task to generate this in the lab. There have been varied attempts at trying to generate a model for the heart with numerous cell types and different scaffolds or materials. The common underlying theme in these approaches is to combine together matrix material and different cell types to make something similar to heart tissue. Multi-cellularity is an essential aspect of the heart and therefore critical to any approach which would try to mimic such a complex tissue. The heart is made up of many cell types that combine to form complex structures like: deformable chambers, a tri-layered heart muscle, and vessels. Thus, in this review we will summarise how tissue engineering has progressed in modelling the heart and what gaps still exist in this dynamic field.
Collapse
Affiliation(s)
- Thomas J Owen
- National Heart and Lung Institute, Imperial College London Hammersmith Campus, Imperial Centre for Translational and Experimental Medicine, Du Cane Road, London, W12 0NN, UK.
| | - Sian E Harding
- National Heart and Lung Institute, Imperial College London Hammersmith Campus, Imperial Centre for Translational and Experimental Medicine, Du Cane Road, London, W12 0NN, UK
| |
Collapse
|
21
|
Nakao M, Imashiro C, Kuribara T, Kurashina Y, Totani K, Takemura K. Formation of Large Scaffold-Free 3-D Aggregates in a Cell Culture Dish by Ultrasound Standing Wave Trapping. ULTRASOUND IN MEDICINE & BIOLOGY 2019; 45:1306-1315. [PMID: 30799124 DOI: 10.1016/j.ultrasmedbio.2019.01.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Revised: 12/25/2018] [Accepted: 01/15/2019] [Indexed: 05/06/2023]
Abstract
Cellular aggregates that mimic cell-cell interactions in vitro are essential for biological research. This study introduces a method to form large scaffold-free 3-D aggregates in a clinically ubiquitous cell culture dish using kilohertz-order ultrasound standing wave trapping (USWT). We fabricated an aggregate formation system in which a 60-mm dish was set above a Langevin transducer via water. The transducer was excited at 110.8 kHz, and then C2C12 myoblasts were injected into the dish and trapped at the node position of the standing wave. The diameter and thickness of the formed aggregate were 8 and 2.7 mm, respectively, which are larger than those of aggregates formed previously by USWT. Moreover, we confirmed that >94% of cells constituting the aggregates survived 9 h, and the protein expression of cells was not altered significantly. This method can be applied to form aggregates with high functionality, which contributes to the development of biological research methodology.
Collapse
Affiliation(s)
- Misa Nakao
- School of Science for Open and Environmental Systems, Graduate School of Science and Technology, Keio University, Yokohama, Kanagawa, Japan
| | - Chikahiro Imashiro
- School of Science for Open and Environmental Systems, Graduate School of Science and Technology, Keio University, Yokohama, Kanagawa, Japan
| | - Taiki Kuribara
- Department of Materials and Life Science, Faculty of Science and Technology, Seikei University, Musashino, Tokyo, Japan
| | - Yuta Kurashina
- Department of Materials Science and Engineering, School of Materials and Chemical Technology, Tokyo Institute of Technology, Yokohama, Kanagawa, Japan 226-8503; Department of Mechanical Engineering, Faculty of Science and Technology, Keio University, Yokohama, Kanagawa, Japan
| | - Kiichiro Totani
- Department of Materials and Life Science, Faculty of Science and Technology, Seikei University, Musashino, Tokyo, Japan
| | - Kenjiro Takemura
- Department of Mechanical Engineering, Faculty of Science and Technology, Keio University, Yokohama, Kanagawa, Japan.
| |
Collapse
|
22
|
Hu JB, Tomov ML, Buikema JW, Chen C, Mahmoudi M, Wu SM, Serpooshan V. Cardiovascular tissue bioprinting: Physical and chemical processes. APPLIED PHYSICS REVIEWS 2018; 5:041106. [PMID: 32550960 PMCID: PMC7187889 DOI: 10.1063/1.5048807] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 09/24/2018] [Indexed: 05/08/2023]
Abstract
Three-dimensional (3D) cardiac tissue bioprinting occupies a critical crossroads position between the fields of materials engineering, cardiovascular biology, 3D printing, and rational organ replacement design. This complex area of research therefore requires expertise from all those disciplines and it poses some unique considerations that must be accounted for. One of the chief hurdles is that there is a relatively limited systematic organization of the physical and chemical characteristics of bioinks that would make them applicable to cardiac bioprinting. This is of great significance, as heart tissue is functionally complex and the in vivo extracellular niche is under stringent controls with little room for variability before a cardiomyopathy manifests. This review explores the critical parameters that are necessary for biologically relevant bioinks to successfully be leveraged for functional cardiac tissue engineering, which can have applications in in vitro heart tissue models, cardiotoxicity studies, and implantable constructs that can be used to treat a range of cardiomyopathies, or in regenerative medicine.
Collapse
Affiliation(s)
- James B. Hu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, USA
| | | | | | - Caressa Chen
- Department of General Surgery, Loyola University Medical Center, Maywood, Illinois 60153, USA
| | | | | | - Vahid Serpooshan
- Author to whom correspondence should be addressed: . Present address: 1760 Haygood Dr. NE, HSRB Bldg., Suite E480, Atlanta, Georgia 30322, USA. Telephone: 404-712-9717. Fax: 404-727-9873
| |
Collapse
|
23
|
Simultaneous AFM Investigation of the Single Cardiomyocyte Electro-Chemo-Mechanics During Excitation-Contraction Coupling. Methods Mol Biol 2018. [PMID: 30374879 DOI: 10.1007/978-1-4939-8894-5_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
The cardiac excitation-contraction coupling is the cellular process through which the heart absolves its blood pumping function, and it is directly affected when cardiac pathologies occur. Cardiomyocytes are the functional units in which this complex biomolecular process takes place: they can be represented as a two-stage electro-chemo and chemo-mechanical transducer, along which each stage can be probed and monitored via appropriate micro/nanotechnology-based tools. Atomic force microscopy (AFM), with its unique nanoresolved force sensitivity and versatile modes of extracting sample properties, can represent a key instrument to study time-dependent heart mechanics and topography at the single cell level. In this work, we show how the integrative possibilities of AFM allowed us to implement an in vitro system which can monitor cardiac electrophysiology, intracellular calcium dynamics, and single cell mechanics. We believe this single cell-sensitive and integrated system will unlock improved, fast, and reliable cardiac in vitro tests in the future.
Collapse
|
24
|
Masumoto H, Yamashita JK. Human iPS cell-engineered three-dimensional cardiac tissues perfused by capillary networks between host and graft. Inflamm Regen 2018; 38:26. [PMID: 30338009 PMCID: PMC6178271 DOI: 10.1186/s41232-018-0084-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Accepted: 08/07/2018] [Indexed: 12/30/2022] Open
Abstract
Stem cell-based cardiac regenerative therapy is expected to be a promising strategy for the treatment of severe heart diseases. Pluripotent stem cells enabled us to reconstruct regenerated myocardium in injured hearts as an engineered tissue aiming for cardiac regeneration. To establish a long-term survival of transplanted three-dimensional (3D) engineered heart tissues in vivo, it is indispensable to induce microcapillaries into the engineered tissues after transplantation. Using temperature-responsive culture surface, we have developed pluripotent stem cell-derived cardiac tissue sheets including multiple cardiac cell lineages. The application of gelatin hydrogel microsphere between the cell sheet stacks enabled us to generate thick stacked cell sheets with functional vascular network in vivo. Another technology to generate 3D engineered cardiac tissues using cardiac cells and biomaterials also validated successful induction of vascular network originated from both host and graft-derived vascular cells.
Collapse
Affiliation(s)
- Hidetoshi Masumoto
- Clinical Translational Research Program, RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047 Japan.,2Clinical Translational Research Program, RIKEN Center for Developmental Biology, Kobe, Japan.,3Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507 Japan.,4Department of Cardiovascular Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Jun K Yamashita
- 3Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507 Japan
| |
Collapse
|
25
|
Caluori G, Pribyl J, Cmiel V, Pesl M, Potocnak T, Provaznik I, Skladal P, Rotrekl V. Simultaneous study of mechanobiology and calcium dynamics on hESC-derived cardiomyocytes clusters. J Mol Recognit 2018; 32:e2760. [PMID: 30084213 DOI: 10.1002/jmr.2760] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 06/11/2018] [Accepted: 07/07/2018] [Indexed: 12/19/2022]
Abstract
Calcium ions act like ubiquitous second messengers in a wide amount of cellular processes. In cardiac myocytes, Ca2+ handling regulates the mechanical contraction necessary to the heart pump function. The field of intracellular and intercellular Ca2+ handling, employing in vitro models of cardiomyocytes, has become a cornerstone to understand the role and adaptation of calcium signalling in healthy and diseased hearts. Comprehensive in vitro systems and cell-based biosensors are powerful tools to enrich and speed up cardiac phenotypic and drug response evaluation. We have implemented a combined setup to measure contractility and calcium waves in human embryonic stem cells-derived cardiomyocyte 3D clusters, obtained from embryoid body differentiation. A combination of atomic force microscopy to monitor cardiac contractility, and sensitive fast scientific complementary metal-oxide-semiconductor camera for epifluorescence video recording, provided correlated signals in real time. To speed up the integrated data processing, we tested several post-processing algorithms, to improve the automatic detection of relevant functional parameters. The validation of our proposed method was assessed by caffeine stimulation (10mM) and detection/characterization of the induced cardiac response. We successfully report the first simultaneous recording of cardiac contractility and calcium waves on the described cardiac 3D models. The drug stimulation confirmed the automatic detection capabilities of the used algorithms, measuring expected physiological response, such as elongation of contraction time and Ca2+ cytosolic persistence, increased calcium basal fluorescence, and transient peaks. These results contribute to the implementation of novel, integrated, high-information, and reliable experimental systems for cardiac models and drug evaluation.
Collapse
Affiliation(s)
- Guido Caluori
- International Clinical Research Centre of Saint Anne Hospital of Brno (FNUSA-ICRC), Interventional Cardiac Electrophysiology Group, Brno, Czech Republic.,Nanobiotechnology Group, Central European Institute of Technology of Masaryk University (CEITEC-MU), Brno, Czech Republic
| | - Jan Pribyl
- Nanobiotechnology Group, Central European Institute of Technology of Masaryk University (CEITEC-MU), Brno, Czech Republic
| | - Vratislav Cmiel
- Department of Biomedical Engineering, Brno University of Technology, Faculty of Electrical Engineering and Communication, Brno, Czech Republic
| | - Martin Pesl
- International Clinical Research Centre of Saint Anne Hospital of Brno (FNUSA-ICRC), Interventional Cardiac Electrophysiology Group, Brno, Czech Republic.,Department of Biology, Masaryk University, Faculty of Medicine, Brno, Czech Republic
| | - Tomas Potocnak
- Department of Biomedical Engineering, Brno University of Technology, Faculty of Electrical Engineering and Communication, Brno, Czech Republic
| | - Ivo Provaznik
- Department of Biomedical Engineering, Brno University of Technology, Faculty of Electrical Engineering and Communication, Brno, Czech Republic
| | - Petr Skladal
- Nanobiotechnology Group, Central European Institute of Technology of Masaryk University (CEITEC-MU), Brno, Czech Republic
| | - Vladimir Rotrekl
- Department of Biology, Masaryk University, Faculty of Medicine, Brno, Czech Republic
| |
Collapse
|
26
|
Oliveira ALDA, Scheffer JP, Markoski M, Koche A, Balbinot A, Antunes F, Kalil R. Vascular endothelial growth factor association with angiopoietin 1 promotes improvement in ventricular function after ischemic cardiomyopathy induced in mini pigs. Acta Cir Bras 2018; 33:386-395. [PMID: 29768541 DOI: 10.1590/s0102-865020180040000010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 03/23/2018] [Indexed: 01/14/2023] Open
Abstract
PURPOSE To investigate the safety and clinical, hemodynamic and tissue improvement ability in mini pigs undergoing cell and gene therapy for the treatment of acute myocardial infarction. METHODS Thirty-two mini pigs Br1 lineage, 12 months old, undergoing induction of acute myocardial infarction by occlusion of the diagonal branch of the paraconal coronary. They were divided into 4 groups: one control group and 3 treatment groups (cell therapy and gene cell therapy). Echocardiography reviews were performed on three occasions and histopathological analysis was performed after 4 weeks. Analysis of variance (ANOVA), Tukey and Wilcoxon tests, were performed. RESULTS Association of vascular endothelial growth factor (VEGF) with angiopoietin1 (Ang1) presented satisfactory results in the improvement of ventricular function following ischemic cardiomyopathy in mini pigs when compared to the results of the other treated groups. CONCLUSION The therapy with VEGF and the combination of VEGF with Ang1, promoted recovered function of the myocardium, characterized by reduced akinetic area and induction of neovascularization.
Collapse
Affiliation(s)
- André Lacerda de Abreu Oliveira
- PhD, Associate Professor, Laboratory of Animal Health, Center for Agricultural Sciences and Technologies, Animal Experimentation Unit, Universidade Estadual do Norte Fluminense Darcy Ribeiro (UENF), Campos dos Goytacazes-RJ, Brazil. Technical procedures, critical revision, final approval the manuscript
| | - Jussara Peters Scheffer
- Fellow PhD degree, Laboratory of Animal Health, Center for Agricultural Sciences and Technologies, Animal Experimentation Unit, UENF Darcy Ribeiro, Campos dos Goytacazes-RJ, Brazil. Conception and design of the study, technical procedures, acquisition and analysis of data, manuscript preparation
| | - Melissa Markoski
- PhD, Associate Professor, Institute of Cardiology of Rio Grande do Sul, University Foundation of Cardiology, Porto Alegre-RS, Brazil. Critical revision, final approval the manuscript
| | - Andreia Koche
- PhD, Associate Professor, Institute of Cardiology of Rio Grande do Sul, University Foundation of Cardiology, Porto Alegre-RS, Brazil. Technical procedures
| | - Alexsandra Balbinot
- PhD, Associate Professor, Institute of Cardiology of Rio Grande do Sul, University Foundation of Cardiology, Porto Alegre-RS, Brazil. Technical procedures
| | - Fernanda Antunes
- PhD, Associate Professor, Laboratory of Animal Health, Center for Agricultural Sciences and Technologies, Animal Experimentation Unit, UENF Darcy Ribeiro, Campos dos Goytacazes-RJ, Brazil. Technical procedures
| | - Renato Kalil
- PhD, Associate Professor, Institute of Cardiology of Rio Grande do Sul, University Foundation of Cardiology, Porto Alegre-RS, Brazil. Critical revision, final approval the manuscript
| |
Collapse
|
27
|
Abstract
Despite considerable advances in medicine, cardiovascular disease is still rising, with ischemic heart disease being the leading cause of death and disability worldwide. Thus extensive efforts are continuing to establish effective therapeutic modalities that would improve both quality of life and survival in this patient population. Novel therapies are being investigated not only to protect the myocardium against ischemia-reperfusion injury but also to regenerate the heart. Stem cell therapy, such as potential use of human mesenchymal stem cells and induced pluripotent stem cells and their exosomes, will make it possible not only to address molecular mechanisms of cardiac conditioning, but also to develop new therapies for ischemic heart disease. Despite the studies and progress made over the last 15 years on the use of stem cell therapy for cardiovascular disease, the efforts are still in their infancy. Even though the expectations have been high, the findings indicate that most of the clinical trials generally have been small and the results inconclusive. Because of many negative findings, there is certain pessimism that cardiac cell therapy is likely to yield any meaningful results over the next decade or so. Similar to other new technologies, early failures are not unusual and they may be followed by impressive success. Nevertheless, there has been considerable attention to safety by the clinical investigators because the adverse events of stem cell therapy have been impressively rare. In summary, although regenerative biology might not help the cardiovascular patient in the near term, it is destined to do so over the next several decades.
Collapse
Affiliation(s)
- Maia Terashvili
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI
| | - Zeljko J Bosnjak
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI.
| |
Collapse
|
28
|
Tee R, Morrison WA, Dilley RJ. A novel microsurgical rodent model for the transplantation of engineered cardiac muscle flap. Microsurgery 2018; 38:544-552. [DOI: 10.1002/micr.30325] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 01/30/2018] [Accepted: 03/16/2018] [Indexed: 11/12/2022]
Affiliation(s)
- Richard Tee
- O'Brien Institute; 42, Fitzroy Street, Fitzroy Victoria, 3065 Australia
- Department of Surgery; University of Melbourne; Victoria, 3065 Australia
| | - Wayne Allan Morrison
- O'Brien Institute; 42, Fitzroy Street, Fitzroy Victoria, 3065 Australia
- Department of Surgery; University of Melbourne; Victoria, 3065 Australia
| | - Rodney J. Dilley
- O'Brien Institute; 42, Fitzroy Street, Fitzroy Victoria, 3065 Australia
- Department of Surgery; University of Melbourne; Victoria, 3065 Australia
- School of Surgery; University of Western Australia; Nedlands Western Australia, 6009 Australia
| |
Collapse
|
29
|
Rotenberg MY, Tian B. Talking to cells: semiconductor nanomaterials at the cellular interface. ADVANCED BIOSYSTEMS 2018; 2:1700242. [PMID: 30906852 PMCID: PMC6430216 DOI: 10.1002/adbi.201700242] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The interface of biological components with semiconductors is a growing field with numerous applications. For example, the interfaces can be used to sense and modulate the electrical activity of single cells and tissues. From the materials point of view, silicon is the ideal option for such studies due to its controlled chemical synthesis, scalable lithography for functional devices, excellent electronic and optical properties, biocompatibility and biodegradability. Recent advances in this area are pushing the bio-interfaces from the tissue and organ level to the single cell and sub-cellular regimes. In this progress report, we will describe some fundamental studies focusing on miniaturizing the bioelectric and biomechanical interfaces. Additionally, many of our highlighted examples involve freestanding silicon-based nanoscale systems, in addition to substrate-bound structures or devices; the former offers new promise for basic research and clinical application. In this report, we will describe recent developments in the interfacing of neuronal and cardiac cells and their networks. Moreover, we will briefly discuss the incorporation of semiconductor nanostructures for interfacing non-excitable cells in applications such as probing intracellular force dynamics and drug delivery. Finally, we will suggest several directions for future exploration.
Collapse
Affiliation(s)
| | - Bozhi Tian
- The James Franck Institute, the University of Chicago, Chicago, IL 60637
- Department of Chemistry, the University of Chicago, Chicago, IL 60637
- The Institute for Biophysical Dynamics, Chicago, IL 60637
| |
Collapse
|
30
|
Onwuka E, King N, Heuer E, Breuer C. The Heart and Great Vessels. Cold Spring Harb Perspect Med 2018; 8:cshperspect.a031922. [PMID: 28289246 DOI: 10.1101/cshperspect.a031922] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Cardiovascular disease is the leading cause of mortality worldwide. We have made large strides over the past few decades in management, but definitive therapeutic options to address this health-care burden are still limited. Given the ever-increasing need, much effort has been spent creating engineered tissue to replaced diseased tissue. This article gives a general overview of this work as it pertains to the development of great vessels, myocardium, and heart valves. In each area, we focus on currently studied methods, limitations, and areas for future study.
Collapse
Affiliation(s)
- Ekene Onwuka
- Tissue Engineering and Surgical Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio 43205.,College of Medicine, The Ohio State University, Columbus, Ohio 43210
| | - Nakesha King
- Tissue Engineering and Surgical Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio 43205.,College of Medicine, The Ohio State University, Columbus, Ohio 43210
| | - Eric Heuer
- Tissue Engineering and Surgical Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio 43205
| | - Christopher Breuer
- Tissue Engineering and Surgical Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio 43205.,College of Medicine, The Ohio State University, Columbus, Ohio 43210.,Department of Pediatric Surgery, Nationwide Children's Hospital, Columbus, Ohio 43205
| |
Collapse
|
31
|
Fabrication of Hydrogel Materials for Biomedical Applications. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1077:197-224. [PMID: 30357691 DOI: 10.1007/978-981-13-0947-2_12] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Hydrogels are three-dimensional hydrophilic polymeric networks that can be made from a wide range of natural and synthetic polymers. This review discusses recent advanced engineering methods to fabricate hydrogels for biomedical applications with emphasis in cardiac constructs and wound healing. Layer-by-Layer (LbL) assembly offers a tissue-engineered construct with robust and highly ordered structures for cell proliferation and differentiation. Three-dimensional printings, including inkjet printing, fused deposition modeling, and stereolithographic apparatus, have been widely employed to fabricate complex structures (e.g., heart valves). Moreover, the state-of-the-art design of intelligent/stimulus-responsive hydrogels can be used for a wide range of biomedical applications, including drug delivery, glucose delivery, shape memory, wound dressings, and so on. In the future, an increasing number of hydrogels with tunable mechanical properties and versatile functions will be developed for biomedical applications by employing advanced engineering techniques with novel material design.
Collapse
|
32
|
Cabiati M, Vozzi F, Gemma F, Montemurro F, De Maria C, Vozzi G, Domenici C, Del Ry S. Cardiac tissue regeneration: A preliminary study on carbon-based nanotubes gelatin scaffold. J Biomed Mater Res B Appl Biomater 2017; 106:2750-2762. [PMID: 29206329 DOI: 10.1002/jbm.b.34056] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 11/06/2017] [Accepted: 11/18/2017] [Indexed: 01/15/2023]
Abstract
The aim of this study was set-up and test of gelatin and carbon nanotubes scaffolds. Gelatin-based (5%) genipin cross-linked (0.2%) scaffolds embedding single-walled carbon nanotubes (SWCNTs, 0.3, 0.5, 0.7, 0.9, and 1.3% w/w) were prepared and mechanically/electrically characterized. For biological evaluation, H9c2 cell line was cultured for 10 days. Cytotoxicity, cell growth and differentiation, immunohistochemistry, and real-time PCR analysis were performed. Myoblast and cardiac differentiation were obtained by serum reduction to 1% (C1% ) and stimulation with 50 nM all trans-retinoic acid (CRA ), respectively. Immunohistochemistry showed elongated myotubes in C1% while round and multinucleated cells in CRA with also a significantly increased expression of natriuretic peptides (NP) and ET-1 receptors in parallel with a decreased ET-1. On scaffolds, cell viability was similar for Gel-SWCNT0.3%/0.9% ; NP and ET systems expression decreased in both concentrations with respect to control and CX-43, mainly due to a lacking of complete differentiation in cardiac phenotype during that time. Although further analyses on novel biomaterials are necessary, these results represent a useful starting point to develop new biomaterial-based scaffolds. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 106B: 2750-2762, 2018.
Collapse
Affiliation(s)
| | | | | | - Francesca Montemurro
- Research Centre "E. Piaggio" and Department of Information Engineering, University of Pisa, Pisa, Italy
| | - Carmelo De Maria
- Research Centre "E. Piaggio" and Department of Information Engineering, University of Pisa, Pisa, Italy
| | - Giovanni Vozzi
- Research Centre "E. Piaggio" and Department of Information Engineering, University of Pisa, Pisa, Italy
| | | | | |
Collapse
|
33
|
Abstract
To curb the high cost of drug development, there is an urgent need to develop more predictive tissue models using human cells to determine drug efficacy and safety in advance of clinical testing. Recent insights gained through fundamental biological studies have validated the importance of dynamic cell environments and cellular communication to the expression of high fidelity organ function. Building on this knowledge, emerging organ-on-a-chip technology is poised to fill the gaps in drug screening by offering predictive human tissue models with methods of sophisticated tissue assembly. Organ-on-a-chip start-ups have begun to spawn from academic research to fill this commercial space and are attracting investment to transform the drug discovery industry. This review traces the history, examines the scientific foundation and envisages the prospect of these renowned organ-on-a-chip technologies. It serves as a guide for new members of this dynamic field to navigate the existing scientific and market space.
Collapse
Affiliation(s)
- Boyang Zhang
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada.
| | | |
Collapse
|
34
|
Gouveia PJ, Rosa S, Ricotti L, Abecasis B, Almeida HV, Monteiro L, Nunes J, Carvalho FS, Serra M, Luchkin S, Kholkin AL, Alves PM, Oliveira PJ, Carvalho R, Menciassi A, das Neves RP, Ferreira LS. Flexible nanofilms coated with aligned piezoelectric microfibers preserve the contractility of cardiomyocytes. Biomaterials 2017. [PMID: 28622605 DOI: 10.1016/j.biomaterials.2017.05.048] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The use of engineered cardiac tissue for high-throughput drug screening/toxicology assessment remains largely unexplored. Here we propose a scaffold that mimics aspects of cardiac extracellular matrix while preserving the contractility of cardiomyocytes. The scaffold is based on a poly(caprolactone) (PCL) nanofilm with magnetic properties (MNF, standing for magnetic nanofilm) coated with a layer of piezoelectric (PIEZO) microfibers of poly(vinylidene fluoride-trifluoroethylene) (MNF+PIEZO). The nanofilm creates a flexible support for cell contraction and the aligned PIEZO microfibers deposited on top of the nanofilm creates conditions for cell alignment and electrical stimulation of the seeded cells. Our results indicate that MNF+PIEZO scaffold promotes rat and human cardiac cell attachment and alignment, maintains the ratio of cell populations overtime, promotes cell-cell communication and metabolic maturation, and preserves cardiomyocyte (CM) contractility for at least 12 days. The engineered cardiac construct showed high toxicity against doxorubicin, a cardiotoxic molecule, and responded to compounds that modulate CM contraction such as epinephrine, propranolol and heptanol.
Collapse
Affiliation(s)
- P José Gouveia
- CNC-Center of Neurosciences and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal; Instituto de Investigação Interdisciplinar, University of Coimbra, Casa Costa Alemão - Pólo II, Rua Dom Francisco de Lemos, 3030-789 Coimbra, Portugal
| | - S Rosa
- CNC-Center of Neurosciences and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
| | - L Ricotti
- The BioRobotics Institute, Scuola Superiore Sant' Anna, Viale Rinaldo Piaggio 34, 56025 Pontedera (PI), Italy
| | - B Abecasis
- Instituto de Tecnologia Química e Biologica António Xavier, New University of Lisbon, Av. da Republica, 2780-157 Oeiras, Portugal
| | - H V Almeida
- CNC-Center of Neurosciences and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
| | - L Monteiro
- CNC-Center of Neurosciences and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
| | - J Nunes
- Center for Mechanical Engineering, University of Coimbra, 3030-788 Coimbra, Portugal
| | - F Sofia Carvalho
- CNC-Center of Neurosciences and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal; Instituto de Investigação Interdisciplinar, University of Coimbra, Casa Costa Alemão - Pólo II, Rua Dom Francisco de Lemos, 3030-789 Coimbra, Portugal
| | - M Serra
- Instituto de Tecnologia Química e Biologica António Xavier, New University of Lisbon, Av. da Republica, 2780-157 Oeiras, Portugal
| | - S Luchkin
- CICECO - Materials Institute of Aveiro & Physics Department, University of Aveiro, Campus de Santiago, 3810-193 Aveiro, Portugal
| | - A Leonidovitch Kholkin
- CICECO - Materials Institute of Aveiro & Physics Department, University of Aveiro, Campus de Santiago, 3810-193 Aveiro, Portugal; School of Natural Sciences and Mathematics, Ural Federal University, 620000 Ekaterinburg, Russia
| | - P Marques Alves
- Instituto de Tecnologia Química e Biologica António Xavier, New University of Lisbon, Av. da Republica, 2780-157 Oeiras, Portugal
| | - P Jorge Oliveira
- Instituto de Investigação Interdisciplinar, University of Coimbra, Casa Costa Alemão - Pólo II, Rua Dom Francisco de Lemos, 3030-789 Coimbra, Portugal
| | - R Carvalho
- Instituto de Investigação Interdisciplinar, University of Coimbra, Casa Costa Alemão - Pólo II, Rua Dom Francisco de Lemos, 3030-789 Coimbra, Portugal; Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, 3000-456 Coimbra, Portugal
| | - A Menciassi
- The BioRobotics Institute, Scuola Superiore Sant' Anna, Viale Rinaldo Piaggio 34, 56025 Pontedera (PI), Italy
| | - R Pires das Neves
- CNC-Center of Neurosciences and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal; Instituto de Investigação Interdisciplinar, University of Coimbra, Casa Costa Alemão - Pólo II, Rua Dom Francisco de Lemos, 3030-789 Coimbra, Portugal
| | - L Silva Ferreira
- CNC-Center of Neurosciences and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal.
| |
Collapse
|
35
|
Pedde RD, Mirani B, Navaei A, Styan T, Wong S, Mehrali M, Thakur A, Mohtaram NK, Bayati A, Dolatshahi-Pirouz A, Nikkhah M, Willerth SM, Akbari M. Emerging Biofabrication Strategies for Engineering Complex Tissue Constructs. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2017; 29:1606061. [PMID: 28370405 DOI: 10.1002/adma.201606061] [Citation(s) in RCA: 227] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 01/16/2017] [Indexed: 05/24/2023]
Abstract
The demand for organ transplantation and repair, coupled with a shortage of available donors, poses an urgent clinical need for the development of innovative treatment strategies for long-term repair and regeneration of injured or diseased tissues and organs. Bioengineering organs, by growing patient-derived cells in biomaterial scaffolds in the presence of pertinent physicochemical signals, provides a promising solution to meet this demand. However, recapitulating the structural and cytoarchitectural complexities of native tissues in vitro remains a significant challenge to be addressed. Through tremendous efforts over the past decade, several innovative biofabrication strategies have been developed to overcome these challenges. This review highlights recent work on emerging three-dimensional bioprinting and textile techniques, compares the advantages and shortcomings of these approaches, outlines the use of common biomaterials and advanced hybrid scaffolds, and describes several design considerations including the structural, physical, biological, and economical parameters that are crucial for the fabrication of functional, complex, engineered tissues. Finally, the applications of these biofabrication strategies in neural, skin, connective, and muscle tissue engineering are explored.
Collapse
Affiliation(s)
- R Daniel Pedde
- Laboratory for Innovations in Microengineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC, V8P 5C2, Canada
| | - Bahram Mirani
- Laboratory for Innovations in Microengineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC, V8P 5C2, Canada
| | - Ali Navaei
- School of Biological and Health Systems Engineering (SBHSE), Arizona State University, Tempe, AZ, 85281, USA
| | - Tara Styan
- Willerth Laboratory, Department of Mechanical Engineering and Division of Medical Sciences, University of Victoria, Victoria, V8P 5C2, Canada
| | - Sarah Wong
- Willerth Laboratory, Department of Mechanical Engineering and Division of Medical Sciences, University of Victoria, Victoria, V8P 5C2, Canada
| | - Mehdi Mehrali
- Department of Micro- and Nanotechnology, Center for Nanomedicine and Theranostics, Technical University of Denmark, Kgs. Lyngby, 2800, Denmark
| | - Ashish Thakur
- Department of Micro- and Nanotechnology, Center for Nanomedicine and Theranostics, Technical University of Denmark, Kgs. Lyngby, 2800, Denmark
| | - Nima Khadem Mohtaram
- Laboratory for Innovations in Microengineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC, V8P 5C2, Canada
| | - Armin Bayati
- Willerth Laboratory, Department of Mechanical Engineering and Division of Medical Sciences, University of Victoria, Victoria, V8P 5C2, Canada
| | - Alireza Dolatshahi-Pirouz
- Department of Micro- and Nanotechnology, Center for Nanomedicine and Theranostics, Technical University of Denmark, Kgs. Lyngby, 2800, Denmark
| | - Mehdi Nikkhah
- School of Biological and Health Systems Engineering (SBHSE), Arizona State University, Tempe, AZ, 85281, USA
| | - Stephanie M Willerth
- Willerth Laboratory, Department of Mechanical Engineering and Division of Medical Sciences, University of Victoria, Victoria, V8P 5C2, Canada
| | - Mohsen Akbari
- Laboratory for Innovations in Microengineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC, V8P 5C2, Canada
- Center for Advanced Materials and Related Technologies (CAMTEC), University of Victoria, Victoria, V8P 5C2, Canada
- Center for Biomedical Research, University of Victoria, Victoria, V8P 5C2, Canada
| |
Collapse
|
36
|
Novel approaches toward the generation of bioscaffolds as a potential therapy in cardiovascular tissue engineering. Int J Cardiol 2017; 228:319-326. [DOI: 10.1016/j.ijcard.2016.11.210] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 11/06/2016] [Indexed: 12/18/2022]
|
37
|
Smith AST, Macadangdang J, Leung W, Laflamme MA, Kim DH. Human iPSC-derived cardiomyocytes and tissue engineering strategies for disease modeling and drug screening. Biotechnol Adv 2017; 35:77-94. [PMID: 28007615 PMCID: PMC5237393 DOI: 10.1016/j.biotechadv.2016.12.002] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 12/16/2016] [Accepted: 12/17/2016] [Indexed: 01/13/2023]
Abstract
Improved methodologies for modeling cardiac disease phenotypes and accurately screening the efficacy and toxicity of potential therapeutic compounds are actively being sought to advance drug development and improve disease modeling capabilities. To that end, much recent effort has been devoted to the development of novel engineered biomimetic cardiac tissue platforms that accurately recapitulate the structure and function of the human myocardium. Within the field of cardiac engineering, induced pluripotent stem cells (iPSCs) are an exciting tool that offer the potential to advance the current state of the art, as they are derived from somatic cells, enabling the development of personalized medical strategies and patient specific disease models. Here we review different aspects of iPSC-based cardiac engineering technologies. We highlight methods for producing iPSC-derived cardiomyocytes (iPSC-CMs) and discuss their application to compound efficacy/toxicity screening and in vitro modeling of prevalent cardiac diseases. Special attention is paid to the application of micro- and nano-engineering techniques for the development of novel iPSC-CM based platforms and their potential to advance current preclinical screening modalities.
Collapse
Affiliation(s)
- Alec S T Smith
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Jesse Macadangdang
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Winnie Leung
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Michael A Laflamme
- Toronto General Research Institute, McEwen Centre for Regenerative Medicine, University Health Network, Toronto, ON, Canada
| | - Deok-Ho Kim
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA; Center for Cardiovascular Biology, University of Washington, Seattle, WA 98109, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA.
| |
Collapse
|
38
|
Dai X, Zhou W, Gao T, Liu J, Lieber CM. Three-dimensional mapping and regulation of action potential propagation in nanoelectronics-innervated tissues. NATURE NANOTECHNOLOGY 2016; 11:776-82. [PMID: 27347837 PMCID: PMC5014560 DOI: 10.1038/nnano.2016.96] [Citation(s) in RCA: 119] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 05/10/2016] [Indexed: 05/19/2023]
Abstract
Real-time mapping and manipulation of electrophysiology in three-dimensional (3D) tissues could have important impacts on fundamental scientific and clinical studies, yet realization is hampered by a lack of effective methods. Here we introduce tissue-scaffold-mimicking 3D nanoelectronic arrays consisting of 64 addressable devices with subcellular dimensions and a submillisecond temporal resolution. Real-time extracellular action potential (AP) recordings reveal quantitative maps of AP propagation in 3D cardiac tissues, enable in situ tracing of the evolving topology of 3D conducting pathways in developing cardiac tissues and probe the dynamics of AP conduction characteristics in a transient arrhythmia disease model and subsequent tissue self-adaptation. We further demonstrate simultaneous multisite stimulation and mapping to actively manipulate the frequency and direction of AP propagation. These results establish new methodologies for 3D spatiotemporal tissue recording and control, and demonstrate the potential to impact regenerative medicine, pharmacology and electronic therapeutics.
Collapse
Affiliation(s)
- Xiaochuan Dai
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, USA
| | - Wei Zhou
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, USA
| | - Teng Gao
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, USA
| | - Jia Liu
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, USA
| | - Charles M Lieber
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, USA
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts 02138, USA
| |
Collapse
|
39
|
Abstract
Soluble morphogen gradients have long been studied in the context of heart specification and patterning. However, recent data have begun to challenge the notion that long-standing in vivo observations are driven solely by these gradients alone. Evidence from multiple biological models, from stem cells to ex vivo biophysical assays, now supports a role for mechanical forces in not only modulating cell behavior but also inducing it de novo in a process termed mechanotransduction. Structural proteins that connect the cell to its niche, for example, integrins and cadherins, and that couple to other growth factor receptors, either directly or indirectly, seem to mediate these changes, although specific mechanistic details are still being elucidated. In this review, we summarize how the wingless (Wnt), transforming growth factor-β, and bone morphogenetic protein signaling pathways affect cardiomyogenesis and then highlight the interplay between each pathway and mechanical forces. In addition, we will outline the role of integrins and cadherins during cardiac development. For each, we will describe how the interplay could change multiple processes during cardiomyogenesis, including the specification of undifferentiated cells, the establishment of heart patterns to accomplish tube and chamber formation, or the maturation of myocytes in the fully formed heart.
Collapse
Affiliation(s)
- Cassandra L Happe
- From the Department of Bioengineering, University of California, San Diego, La Jolla; and Sanford Consortium for Regenerative Medicine, La Jolla, CA
| | - Adam J Engler
- From the Department of Bioengineering, University of California, San Diego, La Jolla; and Sanford Consortium for Regenerative Medicine, La Jolla, CA.
| |
Collapse
|
40
|
In Vitro Differentiation of First Trimester Human Umbilical Cord Perivascular Cells into Contracting Cardiomyocyte-Like Cells. Stem Cells Int 2016; 2016:7513252. [PMID: 27123009 PMCID: PMC4829731 DOI: 10.1155/2016/7513252] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 01/30/2016] [Accepted: 02/11/2016] [Indexed: 12/13/2022] Open
Abstract
Myocardial infarction (MI) causes an extensive loss of heart muscle cells and leads to congestive heart disease (CAD), the leading cause of mortality and morbidity worldwide. Mesenchymal stromal cell- (MSC-) based cell therapy is a promising option to replace invasive interventions. However the optimal cell type providing significant cardiac regeneration after MI is yet to be found. The aim of our study was to investigate the cardiomyogenic differentiation potential of first trimester human umbilical cord perivascular cells (FTM HUCPVCs), a novel, young source of immunoprivileged mesenchymal stromal cells. Based on the expression of cardiomyocyte markers (cTnT, MYH6, SIRPA, and CX43) FTM and term HUCPVCs achieved significantly increased cardiomyogenic differentiation compared to bone marrow MSCs, while their immunogenicity remained significantly lower as indicated by HLA-A and HLA-G expression and susceptibility to T cell mediated cytotoxicity. When applying aggregate-based differentiation, FTM HUCPVCs showed increased aggregate formation potential and generated contracting cells within 1 week of coculture, making them the first MSC type with this ability. Our results indicate that young FTM HUCPVCs have superior cardiomyogenic potential coupled with beneficial immunogenic properties when compared to MSCs of older tissue sources, suggesting that in vitro predifferentiation could be a potential strategy to increase their effectiveness in vivo.
Collapse
|
41
|
Li Y, Huang G, Li M, Wang L, Elson EL, Lu TJ, Genin GM, Xu F. An approach to quantifying 3D responses of cells to extreme strain. Sci Rep 2016; 6:19550. [PMID: 26887698 PMCID: PMC4757889 DOI: 10.1038/srep19550] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 11/18/2015] [Indexed: 12/25/2022] Open
Abstract
The tissues of hollow organs can routinely stretch up to 2.5 times their length. Although significant pathology can arise if relatively large stretches are sustained, the responses of cells are not known at these levels of sustained strain. A key challenge is presenting cells with a realistic and well-defined three-dimensional (3D) culture environment that can sustain such strains. Here, we describe an in vitro system called microscale, magnetically-actuated synthetic tissues (micro-MASTs) to quantify these responses for cells within a 3D hydrogel matrix. Cellular strain-threshold and saturation behaviors were observed in hydrogel matrix, including strain-dependent proliferation, spreading, polarization, and differentiation, and matrix adhesion retained at strains sufficient for apoptosis. More broadly, the system shows promise for defining and controlling the effects of mechanical environment upon a broad range of cells.
Collapse
Affiliation(s)
- Yuhui Li
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China.,Bioinspired Engineering and Biomechanics Center, Xi'an Jiaotong University, Xi'an 710049, China
| | - Guoyou Huang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China.,Bioinspired Engineering and Biomechanics Center, Xi'an Jiaotong University, Xi'an 710049, China
| | - Moxiao Li
- Bioinspired Engineering and Biomechanics Center, Xi'an Jiaotong University, Xi'an 710049, China
| | - Lin Wang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China.,Bioinspired Engineering and Biomechanics Center, Xi'an Jiaotong University, Xi'an 710049, China.,Department of Biochemistry and Molecular Biophysics, Saint Louis, Missouri 63110, USA
| | - Elliot L Elson
- Bioinspired Engineering and Biomechanics Center, Xi'an Jiaotong University, Xi'an 710049, China.,Department of Biochemistry and Molecular Biophysics, Saint Louis, Missouri 63110, USA.,Department of Mechanical Engineering and Materials Science, Washington University, Saint Louis, Missouri 63130, USA
| | - Tian Jian Lu
- Bioinspired Engineering and Biomechanics Center, Xi'an Jiaotong University, Xi'an 710049, China
| | - Guy M Genin
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China.,Bioinspired Engineering and Biomechanics Center, Xi'an Jiaotong University, Xi'an 710049, China.,Department of Neurological Surgery, Washington University School of Medicine, Saint Louis, Missouri 63110, USA.,Department of Mechanical Engineering and Materials Science, Washington University, Saint Louis, Missouri 63130, USA
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China.,Bioinspired Engineering and Biomechanics Center, Xi'an Jiaotong University, Xi'an 710049, China
| |
Collapse
|
42
|
Eder A, Vollert I, Hansen A, Eschenhagen T. Human engineered heart tissue as a model system for drug testing. Adv Drug Deliv Rev 2016; 96:214-24. [PMID: 26026976 DOI: 10.1016/j.addr.2015.05.010] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 05/11/2015] [Accepted: 05/21/2015] [Indexed: 12/29/2022]
Abstract
Drug development is time- and cost-intensive and, despite extensive efforts, still hampered by the limited value of current preclinical test systems to predict side effects, including proarrhythmic and cardiotoxic effects in clinical practice. Part of the problem may be related to species-dependent differences in cardiomyocyte biology. Therefore, the event of readily available human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes (CM) has raised hopes that this human test bed could improve preclinical safety pharmacology as well as drug discovery approaches. However, hiPSC-CM are immature and exhibit peculiarities in terms of ion channel function, gene expression, structural organization and functional responses to drugs that limit their present usefulness. Current efforts are thus directed towards improving hiPSC-CM maturity and high-content readouts. Culturing hiPSC-CM as 3-dimensional engineered heart tissue (EHT) improves CM maturity and anisotropy and, in a 24-well format using silicone racks, enables automated, multiplexed high content readout of contractile function. This review summarizes the principal technology and focuses on advantages and disadvantages of this technology and its potential for preclinical drug screening.
Collapse
|
43
|
van Marion MH, Bax NA, van Turnhout M, Mauretti A, van der Schaft DW, Goumans MJT, Bouten CV. Behavior of CMPCs in unidirectional constrained and stress-free 3D hydrogels. J Mol Cell Cardiol 2015; 87:79-91. [DOI: 10.1016/j.yjmcc.2015.08.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 08/03/2015] [Accepted: 08/10/2015] [Indexed: 11/16/2022]
|
44
|
Chen H, Liu Y, Hu Q. A novel bioactive membrane by cell electrospinning. Exp Cell Res 2015; 338:261-6. [PMID: 26297530 DOI: 10.1016/j.yexcr.2015.08.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 08/17/2015] [Indexed: 12/27/2022]
Abstract
Electrospinning permits fabrication of biodegradable matrices that can resemble the both scale and mechanical behavior of the native extracellular matrix. However, achieving high-cellular density and infiltration of cells within matrices with traditional technique remain challenging and time consuming. The cell electrospinning technique presented in this paper can mitigate the problems associated with these limitations. Cells encapsulated by the material in the cell electrospinning technique survived well and distributed homogenously within the nanofibrous membrane, and their vitality was improved to 133% after being cultured for 28 days. The electrospun nanofibrous membrane has a certain degradation property and favorable cell-membrane interaction that supports the active biocompatibility of the membrane. Its properties are helpful for supporting cell attachment and growth, maintaining phenotypic shape, and secreting an ample amount of extracellular matrix (ECM). This novel membrane may be a potential application within the field of tissue engineering. The ability of cell electrospinning to microintegrate cells into a biodegradable fibrous matrix embodies a novel tissue engineering approach that could be applied to fabricate a high cell density elastic tissue mimetic.
Collapse
Affiliation(s)
- Haiping Chen
- Rapid Manufacturing Engineering Center, Mechatronic Engineering and Automation of Shanghai University, Shanghai 200444, PR China; School of Mechanical and Electrical Engineering, Jinggangshan University, Ji'an 343009, China
| | - Yuanyuan Liu
- Rapid Manufacturing Engineering Center, Mechatronic Engineering and Automation of Shanghai University, Shanghai 200444, PR China; Shanghai Key Laboratory of Intelligent Manufacturing and Roboties, Shanghai 200444, China.
| | - Qingxi Hu
- Rapid Manufacturing Engineering Center, Mechatronic Engineering and Automation of Shanghai University, Shanghai 200444, PR China; Shanghai Key Laboratory of Intelligent Manufacturing and Roboties, Shanghai 200444, China
| |
Collapse
|
45
|
Yu S, Na JY, Lee YJ, Kim KT, Park JT, Kim HS. Forensic application of microRNA-706 as a biomarker for drowning pattern identification. Forensic Sci Int 2015; 255:96-101. [PMID: 26117501 DOI: 10.1016/j.forsciint.2015.06.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 04/23/2015] [Accepted: 06/08/2015] [Indexed: 12/20/2022]
Abstract
Forensic research using microRNA has been used so far only for the identification of body fluids, but its use in understanding biological processes in post-mortem pathology has not been studied before. Therefore, we performed experiments in mice to compare between freshwater and saltwater drowning models, and miRNA expression was analyzed in the brain through a forward bioinformatics screening approach. In this study, we identified eight specific microRNAs whose expression increased in freshwater and decreased in saltwater. Among them, miR-706 - targeting HCN1 - was identified as a potent biomarker for the drowning pattern identification. A higher expression of miR-706 was detected in the freshwater drowning compared to the control and saltwater drowning group (p<0.05, and p<0.01, respectively). HCN1 mRNA expression, a suggested candidate target for miR-706, was lower in the freshwater (p<0.01) than in the saltwater drowning group, and showed statistical difference between freshwater and saltwater drowning (p<0.01). miR-706 was specifically expressed in the hippocampal neurons as detected by in situ hybridization. Our data suggest that a specific microRNA may provide clues to understanding some crime scene investigations and pathobiological processes in the dead body.
Collapse
Affiliation(s)
- SeonYoung Yu
- Department of Forensic Medicine, Chonnam National University Medical School, Gwangju, Republic of Korea; Research Institute of Forensic Science, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Joo-Young Na
- Forensic Medicine Division, National Forensic Service, Jangseong-gun, Jeollanam-do, Republic of Korea
| | - Young-Jik Lee
- Research Institute of Forensic Science, Chonnam National University Medical School, Gwangju, Republic of Korea; Department of Pathology, Saint Carollo Hospital, Sun-Cheon, Chonnam, Republic of Korea
| | - Kyung-Tae Kim
- Department of Anesthesiology and Pain Medicine, Ilsan Paik Hospital, Inje University, Goyang, Republic of Korea
| | - Jong-Tae Park
- Department of Forensic Medicine, Chonnam National University Medical School, Gwangju, Republic of Korea; Research Institute of Forensic Science, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Hyung-Seok Kim
- Department of Forensic Medicine, Chonnam National University Medical School, Gwangju, Republic of Korea; Research Institute of Forensic Science, Chonnam National University Medical School, Gwangju, Republic of Korea.
| |
Collapse
|
46
|
Mu J, Niu H, Zhang J, Hu P, Bo P, Wang Y. Examination of bone marrow mesenchymal stem cells seeded onto poly(3-hydroxybutyrate-co-4-hydroxybutyrate) biological materials for myocardial patch. J Histotechnol 2015. [DOI: 10.1179/2046023615y.0000000006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
47
|
Cao H, Kang BJ, Lee CA, Shung KK, Hsiai TK. Electrical and Mechanical Strategies to Enable Cardiac Repair and Regeneration. IEEE Rev Biomed Eng 2015; 8:114-24. [PMID: 25974948 DOI: 10.1109/rbme.2015.2431681] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Inadequate replacement of lost ventricular myocardium from myocardial infarction leads to heart failure. Investigating the regenerative capacity of mammalian hearts represents an emerging direction for tissue engineering and cell-based therapy. Recent advances in stem cells hold promise to restore cardiac functions. However, embryonic or induced pluripotent stem cell-derived cardiomyocytes lack functional phenotypes of the native myocardium, and transplanted tissues are not fully integrated for synchronized electrical and mechanical coupling with the host. In this context, this review highlights the mechanical and electrical strategies to promote cardiomyocyte maturation and integration, and to assess the functional phenotypes of regenerating myocardium. Simultaneous microelectrocardiogram and high-frequency ultrasound techniques will also be introduced to assess electrical and mechanical coupling for small animal models of heart regeneration.
Collapse
|
48
|
Hastings CL, Roche ET, Ruiz-Hernandez E, Schenke-Layland K, Walsh CJ, Duffy GP. Drug and cell delivery for cardiac regeneration. Adv Drug Deliv Rev 2015; 84:85-106. [PMID: 25172834 DOI: 10.1016/j.addr.2014.08.006] [Citation(s) in RCA: 136] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 07/24/2014] [Accepted: 08/15/2014] [Indexed: 12/12/2022]
Abstract
The spectrum of ischaemic cardiomyopathy, encompassing acute myocardial infarction to congestive heart failure is a significant clinical issue in the modern era. This group of diseases is an enormous source of morbidity and mortality and underlies significant healthcare costs worldwide. Cardiac regenerative therapy, whereby pro-regenerative cells, drugs or growth factors are administered to damaged and ischaemic myocardium has demonstrated significant potential, especially preclinically. While some of these strategies have demonstrated a measure of success in clinical trials, tangible clinical translation has been slow. To date, the majority of clinical studies and a significant number of preclinical studies have utilised relatively simple delivery methods for regenerative therapeutics, such as simple systemic administration or local injection in saline carrier vehicles. Here, we review cardiac regenerative strategies with a particular focus on advanced delivery concepts as a potential means to enhance treatment efficacy and tolerability and ultimately, clinical translation. These include (i) delivery of therapeutic agents in biomaterial carriers, (ii) nanoparticulate encapsulation, (iii) multimodal therapeutic strategies and (iv) localised, minimally invasive delivery via percutaneous transcatheter systems.
Collapse
|
49
|
Stoppel WL, Hu D, Domian IJ, Kaplan DL, Black LD. Anisotropic silk biomaterials containing cardiac extracellular matrix for cardiac tissue engineering. ACTA ACUST UNITED AC 2015; 10:034105. [PMID: 25826196 DOI: 10.1088/1748-6041/10/3/034105] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cardiac malformations and disease are the leading causes of death in the United States in live-born infants and adults, respectively. In both of these cases, a decrease in the number of functional cardiomyocytes often results in improper growth of heart tissue, wound healing complications, and poor tissue repair. The field of cardiac tissue engineering seeks to address these concerns by developing cardiac patches created from a variety of biomaterial scaffolds to be used in surgical repair of the heart. These scaffolds should be fully degradable biomaterial systems with tunable properties such that the materials can be altered to meet the needs of both in vitro culture (e.g. disease modeling) and in vivo application (e.g. cardiac patch). Current platforms do not utilize both structural anisotropy and proper cell-matrix contacts to promote functional cardiac phenotypes and thus there is still a need for critically sized scaffolds that mimic both the structural and adhesive properties of native tissue. To address this need, we have developed a silk-based scaffold platform containing cardiac tissue-derived extracellular matrix (cECM). These silk-cECM composite scaffolds have tunable architectures, degradation rates, and mechanical properties. Subcutaneous implantation in rats demonstrated that addition of the cECM to aligned silk scaffold led to 99% endogenous cell infiltration and promoted vascularization of a critically sized scaffold (10 × 5 × 2.5 mm) after 4 weeks in vivo. In vitro, silk-cECM scaffolds maintained the HL-1 atrial cardiomyocytes and human embryonic stem cell-derived cardiomyocytes and promoted a more functional phenotype in both cell types. This class of hybrid silk-cECM anisotropic scaffolds offers new opportunities for developing more physiologically relevant tissues for cardiac repair and disease modeling.
Collapse
Affiliation(s)
- Whitney L Stoppel
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | | | | | | | | |
Collapse
|
50
|
Gao B, Wang L, Han S, Pingguan-Murphy B, Zhang X, Xu F. Engineering of microscale three-dimensional pancreatic islet models in vitro and their biomedical applications. Crit Rev Biotechnol 2015; 36:619-29. [PMID: 25669871 DOI: 10.3109/07388551.2014.1002381] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Diabetes now is the most common chronic disease in the world inducing heavy burden for the people's health. Based on this, diabetes research such as islet function has become a hot topic in medical institutes of the world. Today, in medical institutes, the conventional experiment platform in vitro is monolayer cell culture. However, with the development of micro- and nano-technologies, several microengineering methods have been developed to fabricate three-dimensional (3D) islet models in vitro which can better mimic the islet of pancreases in vivo. These in vitro islet models have shown better cell function than monolayer cells, indicating their great potential as better experimental platforms to elucidate islet behaviors under both physiological and pathological conditions, such as the molecular mechanisms of diabetes and clinical islet transplantation. In this review, we present the state-of-the-art advances in the microengineering methods for fabricating microscale islet models in vitro. We hope this will help researchers to better understand the progress in the engineering 3D islet models and their biomedical applications such as drug screening and islet transplantation.
Collapse
Affiliation(s)
- Bin Gao
- a The Key Laboratory of Biomedical Information Engineering of Ministry of Education , Xi'an Jiaotong University School of Life Science and Technology , Xi'an , China .,b Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University , Xi'an , China .,c Department of Endocrinology and Metabolism , Xijing Hospital, Fourth Military Medical University , Xi'an , China
| | - Lin Wang
- a The Key Laboratory of Biomedical Information Engineering of Ministry of Education , Xi'an Jiaotong University School of Life Science and Technology , Xi'an , China .,b Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University , Xi'an , China
| | - Shuang Han
- d Institute of Digestive Disease, Xijing Hospital, Fourth Military Medical University , Xi'an , China , and
| | - Belinda Pingguan-Murphy
- e Department of Biomedical Engineering, Faculty of Engineering , University of Malaya , Kuala Lumpur , Malaysia
| | - Xiaohui Zhang
- a The Key Laboratory of Biomedical Information Engineering of Ministry of Education , Xi'an Jiaotong University School of Life Science and Technology , Xi'an , China .,b Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University , Xi'an , China
| | - Feng Xu
- a The Key Laboratory of Biomedical Information Engineering of Ministry of Education , Xi'an Jiaotong University School of Life Science and Technology , Xi'an , China .,b Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University , Xi'an , China
| |
Collapse
|