101
|
Khurana A, Sayed N, Singh V, Khurana I, Allawadhi P, Rawat PS, Navik U, Pasumarthi SK, Bharani KK, Weiskirchen R. A comprehensive overview of CRISPR/Cas 9 technology and application thereof in drug discovery. J Cell Biochem 2022; 123:1674-1698. [PMID: 36128934 DOI: 10.1002/jcb.30329] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 08/13/2022] [Accepted: 09/01/2022] [Indexed: 11/07/2022]
Abstract
Clustered Regularly Interspaced Short Palindromic Repeat (CRISPR)-Cas technology possesses revolutionary potential to positively affect various domains of drug discovery. It has initiated a rise in the area of genetic engineering and its advantages range from classical science to translational medicine. These genome editing systems have given a new dimension to our capabilities to alter, detect and annotate specified gene sequences. Moreover, the ease, robustness and adaptability of the CRISPR/Cas9 technology have led to its extensive utilization in research areas in such a short period of time. The applications include the development of model cell lines, understanding disease mechanisms, discovering disease targets, developing transgenic animals and plants, and transcriptional modulation. Further, the technology is rapidly growing; hence, an overlook of progressive success is crucial. This review presents the current status of the CRISPR-Cas technology in a tailor-made format from its discovery to several advancements for drug discovery alongwith future trends associated with possibilities and hurdles including ethical concerns.
Collapse
Affiliation(s)
- Amit Khurana
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH Aachen University Hospital, Aachen, Germany
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science (CVSc), PVNRTVU, Hyderabad, Telangana, India
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science (CVSc), PVNRTVU, Mamnoor, Warangal, Telangana, India
| | - Nilofer Sayed
- Department of Pharmacy, Pravara Rural Education Society's (P.R.E.S.'s) College of Pharmacy, Shreemati Nathibai Damodar Thackersey (SNDT) Women's University, Nashik, Maharashtra, India
| | - Vishakha Singh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India
| | - Isha Khurana
- Department of Pharmaceutical Chemistry, University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Chandigarh, India
| | - Prince Allawadhi
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India
| | - Pushkar Singh Rawat
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, Punjab, India
| | - Umashanker Navik
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, Punjab, India
| | | | - Kala Kumar Bharani
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science (CVSc), PVNRTVU, Mamnoor, Warangal, Telangana, India
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH Aachen University Hospital, Aachen, Germany
| |
Collapse
|
102
|
Basara G, Bahcecioglu G, Ozcebe SG, Ellis BW, Ronan G, Zorlutuna P. Myocardial infarction from a tissue engineering and regenerative medicine point of view: A comprehensive review on models and treatments. BIOPHYSICS REVIEWS 2022; 3:031305. [PMID: 36091931 PMCID: PMC9447372 DOI: 10.1063/5.0093399] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 08/08/2022] [Indexed: 05/12/2023]
Abstract
In the modern world, myocardial infarction is one of the most common cardiovascular diseases, which are responsible for around 18 million deaths every year or almost 32% of all deaths. Due to the detrimental effects of COVID-19 on the cardiovascular system, this rate is expected to increase in the coming years. Although there has been some progress in myocardial infarction treatment, translating pre-clinical findings to the clinic remains a major challenge. One reason for this is the lack of reliable and human representative healthy and fibrotic cardiac tissue models that can be used to understand the fundamentals of ischemic/reperfusion injury caused by myocardial infarction and to test new drugs and therapeutic strategies. In this review, we first present an overview of the anatomy of the heart and the pathophysiology of myocardial infarction, and then discuss the recent developments on pre-clinical infarct models, focusing mainly on the engineered three-dimensional cardiac ischemic/reperfusion injury and fibrosis models developed using different engineering methods such as organoids, microfluidic devices, and bioprinted constructs. We also present the benefits and limitations of emerging and promising regenerative therapy treatments for myocardial infarction such as cell therapies, extracellular vesicles, and cardiac patches. This review aims to overview recent advances in three-dimensional engineered infarct models and current regenerative therapeutic options, which can be used as a guide for developing new models and treatment strategies.
Collapse
Affiliation(s)
- Gozde Basara
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - Gokhan Bahcecioglu
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - S. Gulberk Ozcebe
- Bioengineering Graduate Program, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - Bradley W Ellis
- Bioengineering Graduate Program, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - George Ronan
- Bioengineering Graduate Program, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - Pinar Zorlutuna
- Present address: 143 Multidisciplinary Research Building, University of Notre Dame, Notre Dame, IN 46556. Author to whom correspondence should be addressed:. Tel.: +1 574 631 8543. Fax: +1 574 631 8341
| |
Collapse
|
103
|
Zhang Z, Wang Y, Wang Q, Shang L. Smart Film Actuators for Biomedical Applications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2105116. [PMID: 35038215 DOI: 10.1002/smll.202105116] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 11/23/2021] [Indexed: 06/14/2023]
Abstract
Taking inspiration from the extremely flexible motion abilities in natural organisms, soft actuators have emerged in the past few decades. Particularly, smart film actuators (SFAs) demonstrate unique superiority in easy fabrication, tailorable geometric configurations, and programmable 3D deformations. Thus, they are promising in many biomedical applications, such as soft robotics, tissue engineering, delivery system, and organ-on-a-chip. In this review, the latest achievements of SFAs applied in biomedical fields are summarized. The authors start by introducing the fabrication techniques of SFAs, then shift to the topology design of SFAs, followed by their material selections and distinct actuating mechanisms. After that, their biomedical applications are categorized in practical aspects. The challenges and prospects of this field are finally discussed. The authors believe that this review can boost the development of soft robotics, biomimetics, and human healthcare.
Collapse
Affiliation(s)
- Zhuohao Zhang
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, and the Shanghai Key Laboratory of Medical Epigenetics, the International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Yu Wang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Qiao Wang
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, and the Shanghai Key Laboratory of Medical Epigenetics, the International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Luoran Shang
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, and the Shanghai Key Laboratory of Medical Epigenetics, the International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| |
Collapse
|
104
|
Zhang J, Liu X, Nie J, Shi Y. Restoration of mitophagy ameliorates cardiomyopathy in Barth syndrome. Autophagy 2022; 18:2134-2149. [PMID: 34985382 PMCID: PMC9466615 DOI: 10.1080/15548627.2021.2020979] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Barth syndrome (BTHS) is an X-linked genetic disorder caused by mutations in the TAFAZZIN/Taz gene which encodes a transacylase required for cardiolipin remodeling. Cardiolipin is a mitochondrial signature phospholipid that plays a pivotal role in maintaining mitochondrial membrane structure, respiration, mtDNA biogenesis, and mitophagy. Mutations in the TAFAZZIN gene deplete mature cardiolipin, leading to mitochondrial dysfunction, dilated cardiomyopathy, and premature death in BTHS patients. Currently, there is no effective treatment for this debilitating condition. In this study, we showed that TAFAZZIN deficiency caused hyperactivation of MTORC1 signaling and defective mitophagy, leading to accumulation of autophagic vacuoles and dysfunctional mitochondria in the heart of Tafazzin knockdown mice, a rodent model of BTHS. Consequently, treatment of TAFAZZIN knockdown mice with rapamycin, a potent inhibitor of MTORC1, not only restored mitophagy, but also mitigated mitochondrial dysfunction and dilated cardiomyopathy. Taken together, these findings identify MTORC1 as a novel therapeutic target for BTHS, suggesting that pharmacological restoration of mitophagy may provide a novel treatment for BTHS.Abbreviations: BTHS: Barth syndrome; CCCP: carbonyl cyanide 3-chlorophenylhydrazone; CL: cardiolipin; EIF4EBP1/4E-BP1: eukaryotic translation initiation factor 4E binding protein 1; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; KD: knockdown; KO: knockout; LAMP1: lysosomal-associated membrane protein 1; LV: left ventricle; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; MEFs: mouse embryonic fibroblasts; MTORC1: mechanistic target of rapamycin kinase complex 1; OCR: oxygen consumption rate; PE: phosphatidylethanolamine; PIK3C3/VPS34: phosphatidylinositol 3-kinase catalytic subunit type 3; PINK1: PTEN induced putative kinase 1; PRKN/Parkin: parkin RBR E3 ubiquitin protein ligase; qRT-PCR: quantitative real-time polymerase chain reaction; RPS6KB/S6K: ribosomal protein S6 kinase beta; SQSTM1/p62: sequestosome 1; TLCL: tetralinoleoyl cardiolipin; WT: wild-type.
Collapse
Affiliation(s)
- Jun Zhang
- Sam and Ann Barshop Institute for Longevity and Aging Studies, Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Xueling Liu
- Sam and Ann Barshop Institute for Longevity and Aging Studies, Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA,Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, People’s Republic of China
| | - Jia Nie
- Sam and Ann Barshop Institute for Longevity and Aging Studies, Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Yuguang Shi
- Sam and Ann Barshop Institute for Longevity and Aging Studies, Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA,Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, People’s Republic of China,CONTACT Yuguang Shi Joe R. & Teresa Lozano Long Distinguished Chair in Metabolic Biology, Barshop Institute for Longevity and Aging Studies, Department of Pharmacology, University of Texas Health Science Center, San Antonio 4939 Charles Katz Drive, San Antonio, TX78229, USA
| |
Collapse
|
105
|
Greenwell AA, Tabatabaei Dakhili SA, Ussher JR. Myocardial disturbances of intermediary metabolism in Barth syndrome. Front Cardiovasc Med 2022; 9:981972. [PMID: 36035919 PMCID: PMC9399503 DOI: 10.3389/fcvm.2022.981972] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 07/25/2022] [Indexed: 11/13/2022] Open
Abstract
Barth Syndrome (BTHS) is a rare X-linked mitochondrial disorder due to mutations in the gene TAFAZZIN, which leads to immature cardiolipin (CL) remodeling and is characterized by the development of cardiomyopathy. The immature CL remodeling in BTHS results in electron transport chain respiratory defects and destabilization of supercomplexes, thereby impairing ATP production. Thus, BTHS-related cardiomyopathy appears to share metabolic characteristics of the failing heart being an "engine out of fuel." As CL associates with numerous mitochondrial enzymes involved in ATP production, BTHS is also characterized by several defects in intermediary energy metabolism. Herein we will describe the primary disturbances in intermediary energy metabolism relating to the heart's major fuel sources, fatty acids, carbohydrates, ketones, and amino acids. In addition, we will interrogate whether these disturbances represent potential metabolic targets for alleviating BTHS-related cardiomyopathy.
Collapse
Affiliation(s)
- Amanda A. Greenwell
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
- Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, Canada
| | - Seyed Amirhossein Tabatabaei Dakhili
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
- Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, Canada
| | - John R. Ussher
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
- Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
106
|
Zegallai HM, Abu-El-Rub E, Olayinka-Adefemi F, Cole LK, Sparagna GC, Marshall AJ, Hatch GM. Tafazzin deficiency in mouse mesenchymal stem cells promote reprogramming of activated B lymphocytes toward immunosuppressive phenotypes. FASEB J 2022; 36:e22443. [PMID: 35816277 DOI: 10.1096/fj.202200145r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 05/20/2022] [Accepted: 06/27/2022] [Indexed: 11/11/2022]
Abstract
Barth Syndrome (BTHS) is a rare X-linked genetic disorder caused by mutation in the TAFAZZIN gene. Tafazzin (Taz) deficiency in BTHS patients results in an increased risk of infections. Mesenchymal stem cells (MSCs) are well known for their immune-inhibitory function. We examined how Taz-deficiency in murine MSCs impact their ability to modulate the function of lipopolysaccharide (LPS)-activated wild type (WT) B lymphocytes. MSCs from tafazzin knockdown (TazKD) mice exhibited a reduction in mitochondrial cardiolipin compared to wild type (WT) MSCs. However, mitochondrial bioenergetics and membrane potential were unaltered. In contrast, TazKD MSCs exhibited increased reactive oxygen species generation and increased glycolysis. The increased glycolysis was associated with an elevated proliferation, phosphatidylinositol-3-kinase expression and expression of the immunosuppressive markers indoleamine-2,3-dioxygenase, cytotoxic T-lymphocyte-associated protein 4, interleukin-10, and cluster of differentiation 59 compared to controls. Inhibition of glycolysis with 2-deoxyglucose attenuated the TazKD-mediated increased expression of cytotoxic T-lymphocyte-associated protein 4 and interleukin-10. When co-cultured with LPS-activated WT B cells, TazKD MSCs inhibited B cell proliferation and growth rate and reduced B cell secretion of immunoglobulin M compared to controls. In addition, co-culture of LPS-activated WT B cells with TazKD MSCs promoted B cell differentiation toward interleukin-10 secreting plasma cells and B regulatory cells compared to controls. The results indicate that Taz deficiency in MSCs promote reprogramming of activated B lymphocytes toward immunosuppressive phenotypes.
Collapse
Affiliation(s)
- Hana M Zegallai
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme, Children's Hospital Research Institute of Manitoba, Department of Pharmacology & Therapeutics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Ejlal Abu-El-Rub
- Physiology and Pathophysiology, Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid, Jordan
- Physiology and Pathophysiology, Regenerative Medicine, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Folayemi Olayinka-Adefemi
- Department of Immunology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Laura K Cole
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme, Children's Hospital Research Institute of Manitoba, Department of Pharmacology & Therapeutics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Genevieve C Sparagna
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Center, Aurora, Colorado, USA
| | - Aaron J Marshall
- Department of Immunology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Grant M Hatch
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme, Children's Hospital Research Institute of Manitoba, Department of Pharmacology & Therapeutics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
107
|
Li H, Henty-Ridilla JL, Bernstein AM, Ganapathy PS, Herberg S. TGFβ2 Regulates Human Trabecular Meshwork Cell Contractility via ERK and ROCK Pathways with Distinct Signaling Crosstalk Dependent on the Culture Substrate. Curr Eye Res 2022; 47:1165-1178. [PMID: 35481448 PMCID: PMC9782738 DOI: 10.1080/02713683.2022.2071943] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
PURPOSE Transforming growth factor-beta 2 (TGFβ2) is a major contributor to the pathologic changes occurring in human trabecular meshwork (HTM) cells in primary open-angle glaucoma (POAG). TGFβ2 activates extracellular-signal-regulated kinase (ERK) and Rho-associated kinase (ROCK) signaling pathways, both affecting HTM cell behavior. However, exactly how these signaling pathways converge to regulate HTM cell contractility is unclear. Here, we investigated the molecular mechanism underlying TGFβ2-induced pathologic HTM cell contractility, and the crosstalk between ERK and ROCK signaling pathways with different culture substrates. METHODS Hydrogels were engineered by mixing collagen type I, elastin-like polypeptide, and hyaluronic acid, each containing photoactive functional groups, followed by UV crosslinking. Primary HTM cells were seeded atop pre-formed hydrogels for comparisons with glass, or encapsulated within the hydrogels. Changes in actin cytoskeleton, extracellular matrix (ECM) production, phospho-myosin light chain (p-MLC) levels, and hydrogel contraction were assessed. RESULTS HTM cell morphology and filamentous (F)-actin organization were affected by the underlying culture substrates. TGFβ2 increased HTM cell contractility via ERK and ROCK signaling pathways by differentially regulating F-actin, α-smooth muscle actin (αSMA), fibronectin (FN), and p-MLC in HTM cells. ERK inhibition, even as short as 4 h, further increased TGFβ2-induced p-MLC in HTM cells on hydrogels, but not on glass. This translated into hypercontractility of HTM cell-laden hydrogels. ROCK inhibition had precisely the opposite effects and potently relaxed the TGFβ2-induced hydrogels. CONCLUSIONS Our data suggest that ERK signaling negatively regulates ROCK-mediated HTM cell contractility. These findings emphasize the critical importance of using tissue-mimetic ECM substrates for investigating HTM cell physiology and glaucomatous pathophysiology in vitro.
Collapse
Affiliation(s)
- Haiyan Li
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, NY 13210, USA,Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA,BioInspired Institute, Syracuse University, Syracuse, NY 13244, USA
| | - Jessica L. Henty-Ridilla
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA,Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY 13210, USA,BioInspired Institute, Syracuse University, Syracuse, NY 13244, USA
| | - Audrey M. Bernstein
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, NY 13210, USA,Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA,Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA,BioInspired Institute, Syracuse University, Syracuse, NY 13244, USA,Syracuse VA Medical Center, New York VA Health Care, Syracuse, NY 13210, USA
| | - Preethi S. Ganapathy
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, NY 13210, USA,Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY 13210, USA,BioInspired Institute, Syracuse University, Syracuse, NY 13244, USA
| | - Samuel Herberg
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, NY 13210, USA,Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA,Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA,BioInspired Institute, Syracuse University, Syracuse, NY 13244, USA,Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY 13244, USA,To whom correspondence should be addressed: Samuel Herberg, PhD, Assistant Professor; Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, 505 Irving Avenue, Neuroscience Research Building Room 4609, Syracuse, NY 13210, USA,
| |
Collapse
|
108
|
Dou W, Malhi M, Cui T, Wang M, Wang T, Shan G, Law J, Gong Z, Plakhotnik J, Filleter T, Li R, Simmons CA, Maynes JT, Sun Y. A Carbon-Based Biosensing Platform for Simultaneously Measuring the Contraction and Electrophysiology of iPSC-Cardiomyocyte Monolayers. ACS NANO 2022; 16:11278-11290. [PMID: 35715006 DOI: 10.1021/acsnano.2c04676] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Heart beating is triggered by the generation and propagation of action potentials through the myocardium, resulting in the synchronous contraction of cardiomyocytes. This process highlights the importance of electrical and mechanical coordination in organ function. Investigating the pathogenesis of heart diseases and potential therapeutic actions in vitro requires biosensing technologies which allow for long-term and simultaneous measurement of the contractility and electrophysiology of cardiomyocytes. However, the adoption of current biosensing approaches for functional measurement of in vitro cardiac models is hampered by low sensitivity, difficulties in achieving multifunctional detection, and costly manufacturing processes. Leveraging carbon-based nanomaterials, we developed a biosensing platform that is capable of performing on-chip and simultaneous measurement of contractility and electrophysiology of human induced pluripotent stem-cell-derived cardiomyocyte (iPSC-CM) monolayers. This platform integrates with a flexible thin-film cantilever embedded with a carbon black (CB)-PDMS strain sensor for high-sensitivity contraction measurement and four pure carbon nanotube (CNT) electrodes for the detection of extracellular field potentials with low electrode impedance. Cardiac functional properties including contractile stress, beating rate, beating rhythm, and extracellular field potential were evaluated to quantify iPSC-CM responses to common cardiotropic agents. In addition, an in vitro model of drug-induced cardiac arrhythmia was established to further validate the platform for disease modeling and drug testing.
Collapse
Affiliation(s)
- Wenkun Dou
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario M5S 3G8, Canada
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | - Manpreet Malhi
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
- Department of Biochemistry, University of Toronto, Toronto, M5S 1A8, Canada
| | - Teng Cui
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario M5S 3G8, Canada
| | - Minyao Wang
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, M5S 3G9, Canada
- Division of Cardiovascular Surgery, Department of Surgery, University Health Network and University of Toronto, Toronto, M5G 1L7, Canada
| | - Tiancong Wang
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario M5S 3G8, Canada
| | - Guanqiao Shan
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario M5S 3G8, Canada
| | - Junhui Law
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario M5S 3G8, Canada
| | - Zheyuan Gong
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario M5S 3G8, Canada
| | - Julia Plakhotnik
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
- Department of Biochemistry, University of Toronto, Toronto, M5S 1A8, Canada
| | - Tobin Filleter
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario M5S 3G8, Canada
| | - Renke Li
- Division of Cardiovascular Surgery, Department of Surgery, University Health Network and University of Toronto, Toronto, M5G 1L7, Canada
| | - Craig A Simmons
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario M5S 3G8, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, M5S 3G9, Canada
- Translational Biology & Engineering Program, Ted Rogers Centre for Heart Research, Toronto, M5G 1M1, Canada
| | - Jason T Maynes
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
- Department of Biochemistry, University of Toronto, Toronto, M5S 1A8, Canada
- Department of Anesthesia and Pain Medicine, The Hospital for Sick Children, Toronto, M5G 1X8, Canada
| | - Yu Sun
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario M5S 3G8, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, M5S 3G9, Canada
- Department of Electrical and Computer Engineering, University of Toronto, Toronto, M5S 3G4, Canada
- Department of Computer Science, University of Toronto, Toronto, M5T 3A1, Canada
| |
Collapse
|
109
|
Caudal A, Ren L, Tu C, Wu JC. Human Induced Pluripotent Stem Cells for Studying Mitochondrial Diseases in the Heart. FEBS Lett 2022; 596:1735-1745. [PMID: 35788991 DOI: 10.1002/1873-3468.14444] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 05/11/2022] [Accepted: 06/28/2022] [Indexed: 11/09/2022]
Abstract
Mitochondrial dysfunction is known to contribute to a range of diseases, and primary mitochondrial defects strongly impact high-energy organs such as the heart. Platforms for high-throughput and human-relevant assessment of mitochondrial diseases are currently lacking, hindering the development of targeted therapies. In the past decade, human induced pluripotent stem cells (iPSCs) have become a promising technology for drug discovery in basic and clinical research. In particular, human iPSC-derived cardiomyocytes (iPSC-CMs) offer a unique tool to study a wide range of mitochondrial functions and possess the potential to become a key translational asset for mitochondrial drug development. This review summarizes mitochondrial functions and recent therapeutic discoveries, advancements, and limitations of using iPSC-CMs to study mitochondrial diseases of the heart with an emphasis on cardiac applications.
Collapse
Affiliation(s)
- Arianne Caudal
- Stanford Cardiovascular Institute.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford.,Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Lu Ren
- Stanford Cardiovascular Institute.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford.,Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Chengyi Tu
- Stanford Cardiovascular Institute.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford.,Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford.,Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| |
Collapse
|
110
|
Mousavi A, Stefanek E, Jafari A, Ajji Z, Naghieh S, Akbari M, Savoji H. Tissue-engineered heart chambers as a platform technology for drug discovery and disease modeling. BIOMATERIALS ADVANCES 2022; 138:212916. [PMID: 35913255 DOI: 10.1016/j.bioadv.2022.212916] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 04/29/2022] [Accepted: 05/20/2022] [Indexed: 06/15/2023]
Abstract
Current drug screening approaches are incapable of fully detecting and characterizing drug effectiveness and toxicity of human cardiomyocytes. The pharmaceutical industry uses mathematical models, cell lines, and in vivo models. Many promising drugs are abandoned early in development, and some cardiotoxic drugs reach humans leading to drug recalls. Therefore, there is an unmet need to have more reliable and predictive tools for drug discovery and screening applications. Biofabrication of functional cardiac tissues holds great promise for developing a faithful 3D in vitro disease model, optimizing drug screening efficiencies enabling precision medicine. Different fabrication techniques including molding, pull spinning and 3D bioprinting were used to develop tissue-engineered heart chambers. The big challenge is to effectively organize cells into tissue with structural and physiological features resembling native tissues. Some advancements have been made in engineering miniaturized heart chambers that resemble a living pump for drug screening and disease modeling applications. Here, we review the currently developed tissue-engineered heart chambers and discuss challenges and prospects.
Collapse
Affiliation(s)
- Ali Mousavi
- Institute of Biomedical Engineering, Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada; Research Center, Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, H3T 1C5 Canada; Montreal TransMedTech Institute (iTMT), Montreal, QC H3T 1C5, Canada
| | - Evan Stefanek
- Laboratory for Innovation in Microengineering (LiME), Department of Mechanical Engineering, Center for Biomedical Research, University of Victoria, Victoria, BC V8P 2C5, Canada; Centre for Advanced Materials and Related Technologies (CAMTEC), University of Victoria, Victoria, BC V8P 5C2, Canada
| | - Arman Jafari
- Institute of Biomedical Engineering, Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada; Research Center, Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, H3T 1C5 Canada; Montreal TransMedTech Institute (iTMT), Montreal, QC H3T 1C5, Canada
| | - Zineb Ajji
- Institute of Biomedical Engineering, Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada; Research Center, Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, H3T 1C5 Canada; Montreal TransMedTech Institute (iTMT), Montreal, QC H3T 1C5, Canada
| | - Saman Naghieh
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK S7N 5A9, Canada
| | - Mohsen Akbari
- Laboratory for Innovation in Microengineering (LiME), Department of Mechanical Engineering, Center for Biomedical Research, University of Victoria, Victoria, BC V8P 2C5, Canada; Centre for Advanced Materials and Related Technologies (CAMTEC), University of Victoria, Victoria, BC V8P 5C2, Canada; Biotechnology Center, Silesian University of Technology, Akademicka 2A, 44-100 Gliwice, Poland
| | - Houman Savoji
- Institute of Biomedical Engineering, Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada; Research Center, Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, H3T 1C5 Canada; Montreal TransMedTech Institute (iTMT), Montreal, QC H3T 1C5, Canada.
| |
Collapse
|
111
|
Okhovatian S, Mohammadi MH, Rafatian N, Radisic M. Engineering Models of the Heart Left Ventricle. ACS Biomater Sci Eng 2022; 8:2144-2160. [PMID: 35523206 DOI: 10.1021/acsbiomaterials.1c00636] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Despite capturing the imagination of scientists for decades, the goal of creating an artificial heart for transplantation proved to be significantly more challenging than initially anticipated. Toward this goal, recent ground-breaking studies demonstrate the development of functional left ventricular (LV) models. LV models are artificially constructed 3D chambers that are capable of containing liquid within the engineered cavity and exhibit the functionality of native LV including contraction, ejection of fluid, and electrical impulse propagation. Various hydrogels and polymers have been used in manufacturing of LV models, relying on techniques such as electrospinning, bioprinting, casting, and molding. Most studies scaled down the models based on the dimensions of the human or rat ventricle. Initially, neonatal rat cardiomyocytes were the cell type of choice for construction the LV models. Yet, as the stem cell biology field advanced, recent studies focused on the use of cardiomyocytes derived from human induced pluripotent stem cells. In this review, we first describe the physiological characteristics of the human heart, to establish the parameter space for modeling. We then elaborate on current advances in the field and compare recently developed LV models among themselves and with the native human left ventricle. Fabrication methods, cell types, biomaterials, functional properties, and disease modeling capability are some of the major parameters that have distinguished these models. We also highlight some of the current challenges in this field, such as vascularization, cell composition and fidelity, and discuss potential solutions to overcome them.
Collapse
Affiliation(s)
- Sargol Okhovatian
- Institute of Biomaterials Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Mohammad Hossein Mohammadi
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario M5S 3E5, Canada
| | - Naimeh Rafatian
- Institute of Biomaterials Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Milica Radisic
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario M5S 3E5, Canada.,Institute of Biomaterials Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada.,Toronto General Research Institute, Toronto, Ontario M5G 2C4, Canada
| |
Collapse
|
112
|
Abstract
Cardiovascular disease remains the leading cause of morbidity and mortality in the developed world. In recent decades, extraordinary effort has been devoted to defining the molecular and pathophysiological characteristics of the diseased heart and vasculature. Mouse models have been especially powerful in illuminating the complex signaling pathways, genetic and epigenetic regulatory circuits, and multicellular interactions that underlie cardiovascular disease. The advent of CRISPR genome editing has ushered in a new era of cardiovascular research and possibilities for genetic correction of disease. Next-generation sequencing technologies have greatly accelerated the identification of disease-causing mutations, and advances in gene editing have enabled the rapid modeling of these mutations in mice and patient-derived induced pluripotent stem cells. The ability to correct the genetic drivers of cardiovascular disease through delivery of gene editing components in vivo, while still facing challenges, represents an exciting therapeutic frontier. In this review, we provide an overview of cardiovascular disease mechanisms and the potential applications of CRISPR genome editing for disease modeling and correction. We also discuss the extent to which mice can faithfully model cardiovascular disease and the opportunities and challenges that lie ahead.
Collapse
Affiliation(s)
- Ning Liu
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas
| | - Eric N Olson
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas
| |
Collapse
|
113
|
Abstract
An ensemble of in vitro cardiac tissue models has been developed over the past several decades to aid our understanding of complex cardiovascular disorders using a reductionist approach. These approaches often rely on recapitulating single or multiple clinically relevant end points in a dish indicative of the cardiac pathophysiology. The possibility to generate disease-relevant and patient-specific human induced pluripotent stem cells has further leveraged the utility of the cardiac models as screening tools at a large scale. To elucidate biological mechanisms in the cardiac models, it is critical to integrate physiological cues in form of biochemical, biophysical, and electromechanical stimuli to achieve desired tissue-like maturity for a robust phenotyping. Here, we review the latest advances in the directed stem cell differentiation approaches to derive a wide gamut of cardiovascular cell types, to allow customization in cardiac model systems, and to study diseased states in multiple cell types. We also highlight the recent progress in the development of several cardiovascular models, such as cardiac organoids, microtissues, engineered heart tissues, and microphysiological systems. We further expand our discussion on defining the context of use for the selection of currently available cardiac tissue models. Last, we discuss the limitations and challenges with the current state-of-the-art cardiac models and highlight future directions.
Collapse
Affiliation(s)
- Dilip Thomas
- Stanford Cardiovascular Institute
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Suji Choi
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston MA 02134
- Department of Cardiology, Boston Children’s Hospital, Boston, MA 02115
| | - Christina Alamana
- Stanford Cardiovascular Institute
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Kevin K. Parker
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston MA 02134
- Department of Cardiology, Boston Children’s Hospital, Boston, MA 02115
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138
| | - Joseph C. Wu
- Stanford Cardiovascular Institute
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
114
|
Koyilot MC, Natarajan P, Hunt CR, Sivarajkumar S, Roy R, Joglekar S, Pandita S, Tong CW, Marakkar S, Subramanian L, Yadav SS, Cherian AV, Pandita TK, Shameer K, Yadav KK. Breakthroughs and Applications of Organ-on-a-Chip Technology. Cells 2022; 11:cells11111828. [PMID: 35681523 PMCID: PMC9180073 DOI: 10.3390/cells11111828] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/28/2022] [Accepted: 05/30/2022] [Indexed: 12/10/2022] Open
Abstract
Organ-on-a-chip (OOAC) is an emerging technology based on microfluid platforms and in vitro cell culture that has a promising future in the healthcare industry. The numerous advantages of OOAC over conventional systems make it highly popular. The chip is an innovative combination of novel technologies, including lab-on-a-chip, microfluidics, biomaterials, and tissue engineering. This paper begins by analyzing the need for the development of OOAC followed by a brief introduction to the technology. Later sections discuss and review the various types of OOACs and the fabrication materials used. The implementation of artificial intelligence in the system makes it more advanced, thereby helping to provide a more accurate diagnosis as well as convenient data management. We introduce selected OOAC projects, including applications to organ/disease modelling, pharmacology, personalized medicine, and dentistry. Finally, we point out certain challenges that need to be surmounted in order to further develop and upgrade the current systems.
Collapse
Affiliation(s)
- Mufeeda C. Koyilot
- Molecular Robotics, Cochin 682033, India; (M.C.K.); (P.N.); (S.S.); (R.R.); (S.J.); (S.M.); (A.V.C.)
| | - Priyadarshini Natarajan
- Molecular Robotics, Cochin 682033, India; (M.C.K.); (P.N.); (S.S.); (R.R.); (S.J.); (S.M.); (A.V.C.)
| | - Clayton R. Hunt
- Houston Methodist Research Institute, Houston, TX 77030, USA;
| | - Sonish Sivarajkumar
- Molecular Robotics, Cochin 682033, India; (M.C.K.); (P.N.); (S.S.); (R.R.); (S.J.); (S.M.); (A.V.C.)
| | - Romy Roy
- Molecular Robotics, Cochin 682033, India; (M.C.K.); (P.N.); (S.S.); (R.R.); (S.J.); (S.M.); (A.V.C.)
| | - Shreeram Joglekar
- Molecular Robotics, Cochin 682033, India; (M.C.K.); (P.N.); (S.S.); (R.R.); (S.J.); (S.M.); (A.V.C.)
| | - Shruti Pandita
- Mays Cancer Center, University of Texas Health Sciences Center at San Antonio, San Antonio, TX 78229, USA;
| | - Carl W. Tong
- School of Engineering Medicine, Texas A&M University, Houston, TX 77030, USA;
| | - Shamsudheen Marakkar
- Molecular Robotics, Cochin 682033, India; (M.C.K.); (P.N.); (S.S.); (R.R.); (S.J.); (S.M.); (A.V.C.)
| | | | - Shalini S. Yadav
- Department of Immunology, UT MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Anoop V. Cherian
- Molecular Robotics, Cochin 682033, India; (M.C.K.); (P.N.); (S.S.); (R.R.); (S.J.); (S.M.); (A.V.C.)
| | - Tej K. Pandita
- Houston Methodist Research Institute, Houston, TX 77030, USA;
- Center for Genomic and Precision Medicine, Institute of Biosciences and Technology, Department of Translational Medical Sciences, Texas A&M University, Houston, TX 77030, USA
- Correspondence: (T.K.P.); (K.S.); (K.K.Y.)
| | - Khader Shameer
- School of Public Health, Faculty of Medicine, Imperial College London, South Kensington, London SW7 2AZ, UK
- Correspondence: (T.K.P.); (K.S.); (K.K.Y.)
| | - Kamlesh K. Yadav
- School of Engineering Medicine, Texas A&M University, Houston, TX 77030, USA;
- Center for Genomic and Precision Medicine, Institute of Biosciences and Technology, Department of Translational Medical Sciences, Texas A&M University, Houston, TX 77030, USA
- Correspondence: (T.K.P.); (K.S.); (K.K.Y.)
| |
Collapse
|
115
|
Filippi M, Buchner T, Yasa O, Weirich S, Katzschmann RK. Microfluidic Tissue Engineering and Bio-Actuation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2108427. [PMID: 35194852 DOI: 10.1002/adma.202108427] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 02/07/2022] [Indexed: 06/14/2023]
Abstract
Bio-hybrid technologies aim to replicate the unique capabilities of biological systems that could surpass advanced artificial technologies. Soft bio-hybrid robots consist of synthetic and living materials and have the potential to self-assemble, regenerate, work autonomously, and interact safely with other species and the environment. Cells require a sufficient exchange of nutrients and gases, which is guaranteed by convection and diffusive transport through liquid media. The functional development and long-term survival of biological tissues in vitro can be improved by dynamic flow culture, but only microfluidic flow control can develop tissue with fine structuring and regulation at the microscale. Full control of tissue growth at the microscale will eventually lead to functional macroscale constructs, which are needed as the biological component of soft bio-hybrid technologies. This review summarizes recent progress in microfluidic techniques to engineer biological tissues, focusing on the use of muscle cells for robotic bio-actuation. Moreover, the instances in which bio-actuation technologies greatly benefit from fusion with microfluidics are highlighted, which include: the microfabrication of matrices, biomimicry of cell microenvironments, tissue maturation, perfusion, and vascularization.
Collapse
Affiliation(s)
- Miriam Filippi
- Soft Robotics Laboratory, ETH Zurich, Tannenstrasse 3, Zurich, 8092, Switzerland
| | - Thomas Buchner
- Soft Robotics Laboratory, ETH Zurich, Tannenstrasse 3, Zurich, 8092, Switzerland
| | - Oncay Yasa
- Soft Robotics Laboratory, ETH Zurich, Tannenstrasse 3, Zurich, 8092, Switzerland
| | - Stefan Weirich
- Soft Robotics Laboratory, ETH Zurich, Tannenstrasse 3, Zurich, 8092, Switzerland
| | - Robert K Katzschmann
- Soft Robotics Laboratory, ETH Zurich, Tannenstrasse 3, Zurich, 8092, Switzerland
| |
Collapse
|
116
|
Rao KS, Kameswaran V, Bruneau BG. Modeling congenital heart disease: lessons from mice, hPSC-based models, and organoids. Genes Dev 2022; 36:652-663. [PMID: 35835508 PMCID: PMC9296004 DOI: 10.1101/gad.349678.122] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Congenital heart defects (CHDs) are among the most common birth defects, but their etiology has long been mysterious. In recent decades, the development of a variety of experimental models has led to a greater understanding of the molecular basis of CHDs. In this review, we contrast mouse models of CHD, which maintain the anatomical arrangement of the heart, and human cellular models of CHD, which are more likely to capture human-specific biology but lack anatomical structure. We also discuss the recent development of cardiac organoids, which are a promising step toward more anatomically informative human models of CHD.
Collapse
Affiliation(s)
- Kavitha S Rao
- Gladstone Institutes, San Francisco, California 94158, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone, San Francisco, California 94158, USA
| | - Vasumathi Kameswaran
- Gladstone Institutes, San Francisco, California 94158, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone, San Francisco, California 94158, USA
| | - Benoit G Bruneau
- Gladstone Institutes, San Francisco, California 94158, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone, San Francisco, California 94158, USA
- Department of Pediatrics and Cardiovascular Research Institute, University of California at San Francisco, San Francisco, California 94158, USA
| |
Collapse
|
117
|
Cardio PyMEA: A user-friendly, open-source Python application for cardiomyocyte microelectrode array analysis. PLoS One 2022; 17:e0266647. [PMID: 35617323 PMCID: PMC9135279 DOI: 10.1371/journal.pone.0266647] [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: 03/23/2022] [Accepted: 05/16/2022] [Indexed: 11/19/2022] Open
Abstract
Open source analytical software for the analysis of electrophysiological cardiomyocyte data offers a variety of new functionalities to complement closed-source, proprietary tools. Here, we present the Cardio PyMEA application, a free, modifiable, and open source program for the analysis of microelectrode array (MEA) data obtained from cardiomyocyte cultures. Major software capabilities include: beat detection; pacemaker origin estimation; beat amplitude and interval; local activation time, upstroke velocity, and conduction velocity; analysis of cardiomyocyte property-distance relationships; and robust power law analysis of pacemaker spatiotemporal instability. Cardio PyMEA was written entirely in Python 3 to provide an accessible, integrated workflow that possesses a user-friendly graphical user interface (GUI) written in PyQt5 to allow for performant, cross-platform utilization. This application makes use of object-oriented programming (OOP) principles to facilitate the relatively straightforward incorporation of custom functionalities, e.g. power law analysis, that suit the needs of the user. Cardio PyMEA is available as an open source application under the terms of the GNU General Public License (GPL). The source code for Cardio PyMEA can be downloaded from Github at the following repository: https://github.com/csdunhamUC/cardio_pymea.
Collapse
|
118
|
Vásquez-Limeta A, Lukasik K, Kong D, Sullenberger C, Luvsanjav D, Sahabandu N, Chari R, Loncarek J. CPAP insufficiency leads to incomplete centrioles that duplicate but fragment. J Cell Biol 2022; 221:213119. [PMID: 35404385 PMCID: PMC9007748 DOI: 10.1083/jcb.202108018] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 01/13/2022] [Accepted: 02/28/2022] [Indexed: 11/22/2022] Open
Abstract
Centrioles are structures that assemble centrosomes. CPAP is critical for centrosome assembly, and its mutations are found in patients with diseases such as primary microcephaly. CPAP’s centrosomal localization, its dynamics, and the consequences of its insufficiency in human cells are poorly understood. Here we use human cells genetically engineered for fast degradation of CPAP, in combination with superresolution microscopy, to address these uncertainties. We show that three independent centrosomal CPAP populations are dynamically regulated during the cell cycle. We confirm that CPAP is critical for assembly of human centrioles, but not for recruitment of pericentriolar material on already assembled centrioles. Further, we reveal that CPAP insufficiency leads to centrioles with incomplete microtubule triplets that can convert to centrosomes, duplicate, and form mitotic spindle poles, but fragment owing to loss of cohesion between microtubule blades. These findings further our basic understanding of the role of CPAP in centrosome biogenesis and help understand how CPAP aberrations can lead to human diseases.
Collapse
Affiliation(s)
- Alejandra Vásquez-Limeta
- Laboratory of Protein Dynamics and Signaling, National Institutes of Health, National Cancer Institute, Center for Cancer Research, Frederick, MD
| | - Kimberly Lukasik
- Laboratory of Protein Dynamics and Signaling, National Institutes of Health, National Cancer Institute, Center for Cancer Research, Frederick, MD
| | - Dong Kong
- Laboratory of Protein Dynamics and Signaling, National Institutes of Health, National Cancer Institute, Center for Cancer Research, Frederick, MD
| | - Catherine Sullenberger
- Laboratory of Protein Dynamics and Signaling, National Institutes of Health, National Cancer Institute, Center for Cancer Research, Frederick, MD
| | - Delgermaa Luvsanjav
- Laboratory of Protein Dynamics and Signaling, National Institutes of Health, National Cancer Institute, Center for Cancer Research, Frederick, MD
| | - Natalie Sahabandu
- Laboratory of Protein Dynamics and Signaling, National Institutes of Health, National Cancer Institute, Center for Cancer Research, Frederick, MD
| | - Raj Chari
- Genome Modification Core, Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Jadranka Loncarek
- Laboratory of Protein Dynamics and Signaling, National Institutes of Health, National Cancer Institute, Center for Cancer Research, Frederick, MD
| |
Collapse
|
119
|
Opportunities and challenges in cardiac tissue engineering from an analysis of two decades of advances. Nat Biomed Eng 2022; 6:327-338. [PMID: 35478227 DOI: 10.1038/s41551-022-00885-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 03/08/2022] [Indexed: 12/20/2022]
Abstract
Engineered human cardiac tissues facilitate progress in regenerative medicine, disease modelling and drug development. In this Perspective, we reflect on the most notable advances in cardiac tissue engineering from the past two decades by analysing pivotal studies and critically examining the most consequential developments. This retrospective analysis led us to identify key milestones and to outline a set of opportunities, along with their associated challenges, for the further advancement of engineered human cardiac tissues.
Collapse
|
120
|
Wei L, Xia S, Li Y, Qi Y, Wang Y, Zhang D, Hua Y, Luo S. Application of hiPSC as a Drug Tester Via Mimicking a Personalized Mini Heart. Front Genet 2022; 13:891159. [PMID: 35495144 PMCID: PMC9046785 DOI: 10.3389/fgene.2022.891159] [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: 03/07/2022] [Accepted: 03/30/2022] [Indexed: 12/02/2022] Open
Abstract
Human induced pluripotent stem cells (hIPSC) have been used to produce almost all types of human cells currently, which makes them into several potential applications with replicated patient-specific genotype. Furthermore, hIPSC derived cardiomyocytes assembled engineering heart tissue can be established to achieve multiple functional evaluations by tissue engineering technology. This short review summarized the current advanced applications based on the hIPSC derived heart tissue in molecular mechanisms elucidating and high throughput drug screening.
Collapse
Affiliation(s)
- Li Wei
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Shutao Xia
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, China
| | - Yifei Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yan Qi
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, China
| | - Yue Wang
- Department of Cardiovascular Surgery, Pediatric Heart Center, West China Hospital, Sichuan University, Chengdu, China
| | - Donghui Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, China
- *Correspondence: Donghui Zhang, ; Yimin Hua, ; Shuhua Luo,
| | - Yimin Hua
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- *Correspondence: Donghui Zhang, ; Yimin Hua, ; Shuhua Luo,
| | - Shuhua Luo
- Department of Cardiovascular Surgery, Pediatric Heart Center, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Donghui Zhang, ; Yimin Hua, ; Shuhua Luo,
| |
Collapse
|
121
|
Pang J, Bao Y, Mitchell-Silbaugh K, Veevers J, Fang X. Barth Syndrome Cardiomyopathy: An Update. Genes (Basel) 2022; 13:genes13040656. [PMID: 35456462 PMCID: PMC9030331 DOI: 10.3390/genes13040656] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 03/23/2022] [Accepted: 04/02/2022] [Indexed: 12/28/2022] Open
Abstract
Barth syndrome (BTHS) is an X-linked mitochondrial lipid disorder caused by mutations in the TAFAZZIN (TAZ) gene, which encodes a mitochondrial acyltransferase/transacylase required for cardiolipin (CL) biosynthesis. Cardiomyopathy is a major clinical feature of BTHS. During the past four decades, we have witnessed many landmark discoveries that have led to a greater understanding of clinical features of BTHS cardiomyopathy and their molecular basis, as well as the therapeutic targets for this disease. Recently published Taz knockout mouse models provide useful experimental models for studying BTHS cardiomyopathy and testing potential therapeutic approaches. This review aims to summarize key findings of the clinical features, molecular mechanisms, and potential therapeutic approaches for BTHS cardiomyopathy, with particular emphasis on the most recent studies.
Collapse
Affiliation(s)
- Jing Pang
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; (J.P.); (Y.B.); (K.M.-S.); (J.V.)
- Department of Biological Science, University of California San Diego, La Jolla, CA 92093, USA
| | - Yutong Bao
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; (J.P.); (Y.B.); (K.M.-S.); (J.V.)
- Department of Biological Science, University of California San Diego, La Jolla, CA 92093, USA
| | - Kalia Mitchell-Silbaugh
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; (J.P.); (Y.B.); (K.M.-S.); (J.V.)
| | - Jennifer Veevers
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; (J.P.); (Y.B.); (K.M.-S.); (J.V.)
| | - Xi Fang
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; (J.P.); (Y.B.); (K.M.-S.); (J.V.)
- Correspondence: ; Tel.: +1-858-246-4637
| |
Collapse
|
122
|
Magdy T, Burridge PW. Prime time for doxorubicin-induced cardiotoxicity genetic testing. Pharmacogenomics 2022; 23:335-338. [PMID: 35380470 PMCID: PMC9006338 DOI: 10.2217/pgs-2022-0032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 03/14/2022] [Indexed: 11/21/2022] Open
Affiliation(s)
- Tarek Magdy
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Paul W Burridge
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
123
|
Wang Y, Wang P, Qin J. Human Organoids and Organs-on-Chips for Addressing COVID-19 Challenges. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2105187. [PMID: 35107217 PMCID: PMC8981475 DOI: 10.1002/advs.202105187] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 01/15/2022] [Indexed: 05/07/2023]
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), poses an imminent threat to our lives. Although animal models and monolayer cell cultures are utilized for pathogenesis studies and the development of COVID-19 therapeutics, models that can more accurately reflect human-relevant responses to this novel virus are still lacking. Stem cell organoids and bioengineered organs-on-chips have emerged as two cutting-edge technologies used to construct biomimetic in vitro three-dimensional (3D) tissue or organ models. In this review, the key features of these two model systems that allow them to recapitulate organ physiology and function are introduced. The recent progress of these technologies for virology research is summarized and their utility in meeting the COVID-19 pandemic is highlighted. Future opportunities and challenges in the development of advanced human organ models and their potential to accelerate translational applications to provide vaccines and therapies for COVID-19 and other emerging epidemics are also discussed.
Collapse
Affiliation(s)
- Yaqing Wang
- Division of BiotechnologyDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
| | - Peng Wang
- Division of BiotechnologyDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
| | - Jianhua Qin
- Division of BiotechnologyDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
- Beijing Institute For Stem Cell and Regeneration MedicineBeijing100101China
- CAS Center for Excellence in Brain Science and Intelligence TechnologyChinese Academy of SciencesShanghai200031China
- University of Chinese Academy of SciencesBeijing100049China
| |
Collapse
|
124
|
Huebsch N, Charrez B, Neiman G, Siemons B, Boggess SC, Wall S, Charwat V, Jæger KH, Cleres D, Telle Å, Lee-Montiel FT, Jeffreys NC, Deveshwar N, Edwards AG, Serrano J, Snuderl M, Stahl A, Tveito A, Miller EW, Healy KE. Metabolically driven maturation of human-induced-pluripotent-stem-cell-derived cardiac microtissues on microfluidic chips. Nat Biomed Eng 2022; 6:372-388. [PMID: 35478228 PMCID: PMC10344596 DOI: 10.1038/s41551-022-00884-4] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 03/14/2022] [Indexed: 12/29/2022]
Abstract
The immature physiology of cardiomyocytes derived from human induced pluripotent stem cells (hiPSCs) limits their utility for drug screening and disease modelling. Here we show that suitable combinations of mechanical stimuli and metabolic cues can enhance the maturation of hiPSC-derived cardiomyocytes, and that the maturation-inducing cues have phenotype-dependent effects on the cells' action-potential morphology and calcium handling. By using microfluidic chips that enhanced the alignment and extracellular-matrix production of cardiac microtissues derived from genetically distinct sources of hiPSC-derived cardiomyocytes, we identified fatty-acid-enriched maturation media that improved the cells' mitochondrial structure and calcium handling, and observed divergent cell-source-dependent effects on action-potential duration (APD). Specifically, in the presence of maturation media, tissues with abnormally prolonged APDs exhibited shorter APDs, and tissues with aberrantly short APDs displayed prolonged APDs. Regardless of cell source, tissue maturation reduced variabilities in spontaneous beat rate and in APD, and led to converging cell phenotypes (with APDs within the 300-450 ms range characteristic of human left ventricular cardiomyocytes) that improved the modelling of the effects of pro-arrhythmic drugs on cardiac tissue.
Collapse
Affiliation(s)
- Nathaniel Huebsch
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA, USA
- Department of Biomedical Engineering, The Washington University in Saint Louis, Saint Louis, MO, USA
| | - Berenice Charrez
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA, USA
| | - Gabriel Neiman
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA, USA
| | - Brian Siemons
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA, USA
| | - Steven C Boggess
- Department of Chemistry, University of California at Berkeley, Berkeley, CA, USA
| | | | - Verena Charwat
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA, USA
| | | | - David Cleres
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA, USA
| | | | - Felipe T Lee-Montiel
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA, USA
| | - Nicholas C Jeffreys
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA, USA
| | - Nikhil Deveshwar
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA, USA
| | - Andrew G Edwards
- Department of Pharmacology, School of Medicine, University of California, Davis, CA, USA
| | - Jonathan Serrano
- Department of Pathology, New York University Langone Health and Medical School, New York, NY, USA
| | - Matija Snuderl
- Department of Pathology, New York University Langone Health and Medical School, New York, NY, USA
| | - Andreas Stahl
- Department of Nutritional Sciences and Toxicology, University of California at Berkeley, Berkeley, CA, USA
| | | | - Evan W Miller
- Department of Chemistry, University of California at Berkeley, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA, USA
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Kevin E Healy
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA, USA.
- Department of Materials Science and Engineering, University of California at Berkeley, Berkeley, CA, USA.
| |
Collapse
|
125
|
Qi Y, Ye Y, Wang R, Yu S, Zhang Y, Lv J, Jin W, Xia S, Jiang W, Li Y, Zhang D. Mitochondrial dysfunction by TFAM depletion disrupts self-renewal and lineage differentiation of human PSCs by affecting cell proliferation and YAP response. Redox Biol 2022; 50:102248. [PMID: 35091324 PMCID: PMC8802056 DOI: 10.1016/j.redox.2022.102248] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 01/10/2022] [Accepted: 01/20/2022] [Indexed: 02/08/2023] Open
Abstract
Genetic mitochondrial dysfunction is frequently associated with various embryonic developmental defects. However, how mitochondria contribute to early development and cell fate determination is poorly studied, especially in humans. Using human pluripotent stem cells (hPSCs), we established a Dox-induced knockout model with mitochondrial dysfunction and evaluated the effect of mitochondrial dysfunction on human pluripotency maintenance and lineage differentiation. The nucleus-encoded gene TFAM (transcription factor A, mitochondrial), essential for mitochondrial gene transcription and mitochondrial DNA replication, is targeted to construct the mitochondrial dysfunction model. The hPSCs with TFAM depletion exhibit the decrease of mtDNA level and oxidative respiration efficiency, representing a typical mitochondrial dysfunction phenotype. Mitochondrial dysfunction leads to impaired self-renewal in hPSCs due to proliferation arrest. Although the mitochondrial dysfunction does not affect pluripotent gene expression, it results in a severe defect in lineage differentiation. Further study in mesoderm differentiation reveals that mitochondrial dysfunction causes proliferation disability and YAP nuclear translocalization and thus together blocks mesoderm lineage differentiation. These findings provide new insights into understanding the mitochondrial function in human pluripotency maintenance and mesoderm differentiation.
Collapse
Affiliation(s)
- Yan Qi
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, 430062, China
| | - Yida Ye
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, 430062, China
| | - Ruxiang Wang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, 430062, China
| | - Senlin Yu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, 430062, China
| | - Yue Zhang
- Department of Pediatrics and Ministry of Education Key Laboratory of Women and Children's Diseases and Birth Defects, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Jing Lv
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, 430071, China
| | - Wenwen Jin
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, 430071, China
| | - Shutao Xia
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, 430062, China
| | - Wei Jiang
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, 430071, China.
| | - Yifei Li
- Department of Pediatrics and Ministry of Education Key Laboratory of Women and Children's Diseases and Birth Defects, West China Second University Hospital, Sichuan University, Chengdu, 610041, China.
| | - Donghui Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, 430062, China.
| |
Collapse
|
126
|
Progress in Bioengineering Strategies for Heart Regenerative Medicine. Int J Mol Sci 2022; 23:ijms23073482. [PMID: 35408844 PMCID: PMC8998628 DOI: 10.3390/ijms23073482] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 03/20/2022] [Accepted: 03/21/2022] [Indexed: 02/05/2023] Open
Abstract
The human heart has the least regenerative capabilities among tissues and organs, and heart disease continues to be a leading cause of mortality in the industrialized world with insufficient therapeutic options and poor prognosis. Therefore, developing new therapeutic strategies for heart regeneration is a major goal in modern cardiac biology and medicine. Recent advances in stem cell biology and biotechnologies such as human pluripotent stem cells (hPSCs) and cardiac tissue engineering hold great promise for opening novel paths to heart regeneration and repair for heart disease, although these areas are still in their infancy. In this review, we summarize and discuss the recent progress in cardiac tissue engineering strategies, highlighting stem cell engineering and cardiomyocyte maturation, development of novel functional biomaterials and biofabrication tools, and their therapeutic applications involving drug discovery, disease modeling, and regenerative medicine for heart disease.
Collapse
|
127
|
Cho J, Lee H, Rah W, Chang HJ, Yoon YS. From engineered heart tissue to cardiac organoid. Theranostics 2022; 12:2758-2772. [PMID: 35401829 PMCID: PMC8965483 DOI: 10.7150/thno.67661] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 03/01/2022] [Indexed: 12/03/2022] Open
Abstract
The advent of human pluripotent stem cells (hPSCs) presented a new paradigm to employ hPSC-derived cardiomyocytes (hPSC-CMs) in drug screening and disease modeling. However, hPSC-CMs differentiated in conventional two-dimensional systems are structurally and functionally immature. Moreover, these differentiation systems generate predominantly one type of cell. Since the heart includes not only CMs but other cell types, such monolayer cultures have limitations in simulating the native heart. Accordingly, three-dimensional (3D) cardiac tissues have been developed as a better platform by including various cardiac cell types and extracellular matrices. Two advances were made for 3D cardiac tissue generation. One type is engineered heart tissues (EHTs), which are constructed by 3D cell culture of cardiac cells using an engineering technology. This system provides a convenient real-time analysis of cardiac function, as well as a precise control of the input/output flow and mechanical/electrical stimulation. The other type is cardiac organoids, which are formed through self-organization of differentiating cardiac lineage cells from hPSCs. While mature cardiac organoids are more desirable, at present only primitive forms of organoids are available. In this review, we discuss various models of hEHTs and cardiac organoids emulating the human heart, focusing on their unique features, utility, and limitations.
Collapse
Affiliation(s)
- Jaeyeaon Cho
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyein Lee
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Woongchan Rah
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyuk Jae Chang
- Division of Cardiology, Department of Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Young-sup Yoon
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
- Karis Bio Inc., Seoul, Republic of Korea
| |
Collapse
|
128
|
Cardiac Cell Therapy with Pluripotent Stem Cell-Derived Cardiomyocytes: What Has Been Done and What Remains to Do? Curr Cardiol Rep 2022; 24:445-461. [PMID: 35275365 PMCID: PMC9068652 DOI: 10.1007/s11886-022-01666-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/05/2022] [Indexed: 01/14/2023]
Abstract
PURPOSE OF REVIEW Exciting pre-clinical data presents pluripotent stem cell-derived cardiomyocytes (PSC-CM) as a novel therapeutic prospect following myocardial infarction, and worldwide clinical trials are imminent. However, despite notable advances, several challenges remain. Here, we review PSC-CM pre-clinical studies, identifying key translational hurdles. We further discuss cell production and characterization strategies, identifying markers that may help generate cells which overcome these barriers. RECENT FINDINGS PSC-CMs can robustly repopulate infarcted myocardium with functional, force generating cardiomyocytes. However, current differentiation protocols produce immature and heterogenous cardiomyocytes, creating related issues such as arrhythmogenicity, immunogenicity and poor engraftment. Recent efforts have enhanced our understanding of cardiovascular developmental biology. This knowledge may help implement novel differentiation or gene editing strategies that could overcome these limitations. PSC-CMs are an exciting therapeutic prospect. Despite substantial recent advances, limitations of the technology remain. However, with our continued and increasing biological understanding, these issues are addressable, with several worldwide clinical trials anticipated in the coming years.
Collapse
|
129
|
Arslan U, Moruzzi A, Nowacka J, Mummery CL, Eckardt D, Loskill P, Orlova VV. Microphysiological stem cell models of the human heart. Mater Today Bio 2022; 14:100259. [PMID: 35514437 PMCID: PMC9062349 DOI: 10.1016/j.mtbio.2022.100259] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 04/08/2022] [Accepted: 04/10/2022] [Indexed: 11/10/2022] Open
Abstract
Models of heart disease and drug responses are increasingly based on human pluripotent stem cells (hPSCs) since their ability to capture human heart (dys-)function is often better than animal models. Simple monolayer cultures of hPSC-derived cardiomyocytes, however, have shortcomings. Some of these can be overcome using more complex, multi cell-type models in 3D. Here we review modalities that address this, describe efforts to tailor readouts and sensors for monitoring tissue- and cell physiology (exogenously and in situ) and discuss perspectives for implementation in industry and academia.
Collapse
Affiliation(s)
- Ulgu Arslan
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Alessia Moruzzi
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
- Institute for Biomedical Engineering, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Joanna Nowacka
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Christine L. Mummery
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, the Netherlands
| | | | - Peter Loskill
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
- Institute for Biomedical Engineering, Eberhard Karls University Tübingen, Tübingen, Germany
- 3R-Center for in Vitro Models and Alternatives to Animal Testing, Tübingen, Germany
| | - Valeria V. Orlova
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, the Netherlands
| |
Collapse
|
130
|
Yester J, Feingold B. Extended recovery of cardiac function after severe infantile cardiomyopathy presentation of Barth syndrome. JIMD Rep 2022; 63:114-122. [PMID: 35281665 PMCID: PMC8898717 DOI: 10.1002/jmd2.12264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 11/05/2021] [Accepted: 11/24/2021] [Indexed: 11/20/2022] Open
Abstract
Cardiomyopathy is the most common presenting feature of Barth syndrome, often presenting in infancy with severe heart failure and cardiac dysfunction. Historically, affected infants commonly died early after presentation, sometimes before a diagnosis of Barth syndrome was made. With increases in awareness of Barth syndrome and in the care of infants with severe heart failure, survival of children with Barth syndrome and severe heart failure has improved. We describe our experience caring for five unrelated boys with Barth syndrome who presented with severe cardiomyopathy and heart failure prior to age 2 who have had marked improvement with long-term response to medical heart failure therapy.
Collapse
Affiliation(s)
- Jessie Yester
- Heart InstituteUPMC Children's Hospital of PittsburghPittsburghPennsylvaniaUSA
| | - Brian Feingold
- Heart InstituteUPMC Children's Hospital of PittsburghPittsburghPennsylvaniaUSA
- Department of PediatricsUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Clinical and Translational ScienceUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| |
Collapse
|
131
|
Dou W, Malhi M, Zhao Q, Wang L, Huang Z, Law J, Liu N, Simmons CA, Maynes JT, Sun Y. Microengineered platforms for characterizing the contractile function of in vitro cardiac models. MICROSYSTEMS & NANOENGINEERING 2022; 8:26. [PMID: 35299653 PMCID: PMC8882466 DOI: 10.1038/s41378-021-00344-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 11/12/2021] [Accepted: 12/03/2021] [Indexed: 05/08/2023]
Abstract
Emerging heart-on-a-chip platforms are promising approaches to establish cardiac cell/tissue models in vitro for research on cardiac physiology, disease modeling and drug cardiotoxicity as well as for therapeutic discovery. Challenges still exist in obtaining the complete capability of in situ sensing to fully evaluate the complex functional properties of cardiac cell/tissue models. Changes to contractile strength (contractility) and beating regularity (rhythm) are particularly important to generate accurate, predictive models. Developing new platforms and technologies to assess the contractile functions of in vitro cardiac models is essential to provide information on cell/tissue physiologies, drug-induced inotropic responses, and the mechanisms of cardiac diseases. In this review, we discuss recent advances in biosensing platforms for the measurement of contractile functions of in vitro cardiac models, including single cardiomyocytes, 2D monolayers of cardiomyocytes, and 3D cardiac tissues. The characteristics and performance of current platforms are reviewed in terms of sensing principles, measured parameters, performance, cell sources, cell/tissue model configurations, advantages, and limitations. In addition, we highlight applications of these platforms and relevant discoveries in fundamental investigations, drug testing, and disease modeling. Furthermore, challenges and future outlooks of heart-on-a-chip platforms for in vitro measurement of cardiac functional properties are discussed.
Collapse
Affiliation(s)
- Wenkun Dou
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8 Canada
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, ON M5G 1X8 Canada
| | - Manpreet Malhi
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, ON M5G 1X8 Canada
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8 Canada
| | - Qili Zhao
- Institute of Robotics and Automatic Information System and the Tianjin Key Laboratory of Intelligent Robotics, Nankai University, Tianjin, 300350 China
| | - Li Wang
- School of Mechanical & Automotive Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353 China
| | - Zongjie Huang
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8 Canada
| | - Junhui Law
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8 Canada
| | - Na Liu
- School of Mechatronics Engineering and Automation, Shanghai University, Shanghai, 200444 China
| | - Craig A. Simmons
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8 Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9 Canada
- Translational Biology & Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1 Canada
| | - Jason T. Maynes
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, ON M5G 1X8 Canada
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8 Canada
- Department of Anesthesiology and Pain Medicine, University of Toronto, Toronto, ON M5S 1A8 Canada
- Department of Anesthesia and Pain Medicine, The Hospital for Sick Children, Toronto, ON M5G 1X8 Canada
| | - Yu Sun
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8 Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9 Canada
- Department of Electrical and Computer Engineering, University of Toronto, Toronto, ON M5S 3G4 Canada
- Department of Computer Science, University of Toronto, Toronto, ON M5T 3A1 Canada
| |
Collapse
|
132
|
Bioengineering approaches to treat the failing heart: from cell biology to 3D printing. Nat Rev Cardiol 2022; 19:83-99. [PMID: 34453134 DOI: 10.1038/s41569-021-00603-7] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/12/2021] [Indexed: 02/08/2023]
Abstract
Successfully engineering a functional, human, myocardial pump would represent a therapeutic alternative for the millions of patients with end-stage heart disease and provide an alternative to animal-based preclinical models. Although the field of cardiac tissue engineering has made tremendous advances, major challenges remain, which, if properly resolved, might allow the clinical implementation of engineered, functional, complex 3D structures in the future. In this Review, we provide an overview of state-of-the-art studies, challenges that have not yet been overcome and perspectives on cardiac tissue engineering. We begin with the most clinically relevant cell sources used in this field and discuss the use of topological, biophysical and metabolic stimuli to obtain mature phenotypes of cardiomyocytes, particularly in relation to organized cytoskeletal and contractile intracellular structures. We then move from the cellular level to engineering planar cardiac patches and discuss the need for proper vascularization and the main strategies for obtaining it. Finally, we provide an overview of several different approaches for the engineering of volumetric organs and organ parts - from whole-heart decellularization and recellularization to advanced 3D printing technologies.
Collapse
|
133
|
Liu C, Feng X, Li G, Gokulnath P, Xiao J. Generating 3D human cardiac constructs from pluripotent stem cells. EBioMedicine 2022; 76:103813. [PMID: 35093634 PMCID: PMC8804169 DOI: 10.1016/j.ebiom.2022.103813] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/11/2021] [Accepted: 12/29/2021] [Indexed: 12/14/2022] Open
Abstract
Human pluripotent stem cell (hPSC) technology has offered nearly infinite opportunities to model all kinds of human diseases in vitro. Cardiomyocytes derived from hPSCs have proved to be efficient tools for cardiac disease modeling, drug screening and pathological mechanism studies. In this review, we discuss the advantages and limitations of 2D hPSC-cardiomyocyte (hPSC-CM) system, and introduce the recent development of three-dimensional (3D) culture platforms derived from hPSCs. Although the development of bioengineering technologies has greatly improved 3D platform construction, there are certainly challenges and room for development for further in-depth research.
Collapse
Affiliation(s)
- Chang Liu
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China; Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Xing Feng
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China; Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Guoping Li
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Priyanka Gokulnath
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Junjie Xiao
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China; Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
134
|
Pavez-Giani MG, Cyganek L. Recent Advances in Modeling Mitochondrial Cardiomyopathy Using Human Induced Pluripotent Stem Cells. Front Cell Dev Biol 2022; 9:800529. [PMID: 35083221 PMCID: PMC8784695 DOI: 10.3389/fcell.2021.800529] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 12/20/2021] [Indexed: 12/17/2022] Open
Abstract
Around one third of patients with mitochondrial disorders develop a kind of cardiomyopathy. In these cases, severity is quite variable ranging from asymptomatic status to severe manifestations including heart failure, arrhythmias, and sudden cardiac death. ATP is primarily generated in the mitochondrial respiratory chain via oxidative phosphorylation by utilizing fatty acids and carbohydrates. Genes in both the nuclear and the mitochondrial DNA encode components of this metabolic route and, although mutations in these genes are extremely rare, the risk to develop cardiac symptoms is significantly higher in this patient cohort. Additionally, infants with cardiovascular compromise in mitochondrial deficiency display a worse late survival compared to patients without cardiac symptoms. At this point, the mechanisms behind cardiac disease progression related to mitochondrial gene mutations are poorly understood and current therapies are unable to substantially restore the cardiac performance and to reduce the disease burden. Therefore, new strategies are needed to uncover the pathophysiological mechanisms and to identify new therapeutic options for mitochondrial cardiomyopathies. Here, human induced pluripotent stem cell (iPSC) technology has emerged to provide a suitable patient-specific model system by recapitulating major characteristics of the disease in vitro, as well as to offer a powerful platform for pre-clinical drug development and for the testing of novel therapeutic options. In the present review, we summarize recent advances in iPSC-based disease modeling of mitochondrial cardiomyopathies and explore the patho-mechanistic insights as well as new therapeutic approaches that were uncovered with this experimental platform. Further, we discuss the challenges and limitations of this technology and provide an overview of the latest techniques to promote metabolic and functional maturation of iPSC-derived cardiomyocytes that might be necessary for modeling of mitochondrial disorders.
Collapse
Affiliation(s)
- Mario G Pavez-Giani
- Stem Cell Unit, Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen, Germany
| | - Lukas Cyganek
- Stem Cell Unit, Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen, Germany.,Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells", University of Göttingen, Göttingen, Germany
| |
Collapse
|
135
|
Magdy T, Jouni M, Kuo H, Weddle CJ, Lyra–Leite D, Fonoudi H, Romero–Tejeda M, Gharib M, Javed H, Fajardo G, Ross CJD, Carleton BC, Bernstein D, Burridge PW. Identification of Drug Transporter Genomic Variants and Inhibitors That Protect Against Doxorubicin-Induced Cardiotoxicity. Circulation 2022; 145:279-294. [PMID: 34874743 PMCID: PMC8792344 DOI: 10.1161/circulationaha.121.055801] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
BACKGROUND Multiple pharmacogenomic studies have identified the synonymous genomic variant rs7853758 (G > A, L461L) and the intronic variant rs885004 in SLC28A3 (solute carrier family 28 member 3) as statistically associated with a lower incidence of anthracycline-induced cardiotoxicity. However, the true causal variant(s), the cardioprotective mechanism of this locus, the role of SLC28A3 and other solute carrier (SLC) transporters in anthracycline-induced cardiotoxicity, and the suitability of SLC transporters as targets for cardioprotective drugs has not been investigated. METHODS Six well-phenotyped, doxorubicin-treated pediatric patients from the original association study cohort were recruited again, and human induced pluripotent stem cell-derived cardiomyocytes were generated. Patient-specific doxorubicin-induced cardiotoxicity (DIC) was then characterized using assays of cell viability, activated caspase 3/7, and doxorubicin uptake. The role of SLC28A3 in DIC was then queried using overexpression and knockout of SLC28A3 in isogenic human-induced pluripotent stem cell-derived cardiomyocytes using a CRISPR/Cas9 (Clustered Regularly Interspaced Short Palindromic Repeats/CRISPR-associated protein 9). Fine-mapping of the SLC28A3 locus was then completed after SLC28A3 resequencing and an extended in silico haplotype and functional analysis. Genome editing of the potential causal variant was done using cytosine base editor. SLC28A3-AS1 overexpression was done using a lentiviral plasmid-based transduction and was validated using stranded RNA-sequencing after ribosomal RNA depletion. Drug screening was done using the Prestwick Chemical Library (n = 1200), followed by in vivo validation in mice. The effect of desipramine on doxorubicin cytotoxicity was also investigated in 8 cancer cell lines. RESULTS Here, using the most commonly used anthracycline, doxorubicin, we demonstrate that patient-derived cardiomyocytes recapitulate the cardioprotective effect of the SLC28A3 locus and that SLC28A3 expression influences the severity of DIC. Using Nanopore-based fine-mapping and base editing, we identify a novel cardioprotective single nucleotide polymorphism, rs11140490, in the SLC28A3 locus; its effect is exerted via regulation of an antisense long noncoding RNA (SLC28A3-AS1) that overlaps with SLC28A3. Using high-throughput drug screening in patient-derived cardiomyocytes and whole organism validation in mice, we identify the SLC competitive inhibitor desipramine as protective against DIC. CONCLUSIONS This work demonstrates the power of the human induced pluripotent stem cell model to take a single nucleotide polymorphism from a statistical association through to drug discovery, providing human cell-tested data for clinical trials to attenuate DIC.
Collapse
Affiliation(s)
- Tarek Magdy
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL.,Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Mariam Jouni
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL.,Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Hui–Hsuan Kuo
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL.,Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Carly J. Weddle
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL.,Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Davi Lyra–Leite
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL.,Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Hananeh Fonoudi
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL.,Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Marisol Romero–Tejeda
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL.,Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Mennat Gharib
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL.,Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Hoor Javed
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL.,Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Giovanni Fajardo
- Department of Pediatrics (Division of Cardiology), Stanford University School of Medicine, Stanford, CA
| | - Colin J. D. Ross
- British Columbia Children’s Hospital Research Institute, Vancouver, British Columbia, Canada.,Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Bruce C. Carleton
- British Columbia Children’s Hospital Research Institute, Vancouver, British Columbia, Canada.,Division of Translational Therapeutics Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada.,Pharmaceutical Outcomes Programme, British Columbia Children’s Hospital, Vancouver, British Columbia, Canada
| | - Daniel Bernstein
- Department of Pediatrics (Division of Cardiology), Stanford University School of Medicine, Stanford, CA
| | - Paul W. Burridge
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL.,Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL
| |
Collapse
|
136
|
Xiang Y, Miller K, Guan J, Kiratitanaporn W, Tang M, Chen S. 3D bioprinting of complex tissues in vitro: state-of-the-art and future perspectives. Arch Toxicol 2022; 96:691-710. [PMID: 35006284 PMCID: PMC8850226 DOI: 10.1007/s00204-021-03212-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 12/20/2021] [Indexed: 12/15/2022]
Abstract
The pharmacology and toxicology of a broad variety of therapies and chemicals have significantly improved with the aid of the increasing in vitro models of complex human tissues. Offering versatile and precise control over the cell population, extracellular matrix (ECM) deposition, dynamic microenvironment, and sophisticated microarchitecture, which is desired for the in vitro modeling of complex tissues, 3D bio-printing is a rapidly growing technology to be employed in the field. In this review, we will discuss the recent advancement of printing techniques and bio-ink sources, which have been spurred on by the increasing demand for modeling tactics and have facilitated the development of the refined tissue models as well as the modeling strategies, followed by a state-of-the-art update on the specialized work on cancer, heart, muscle and liver. In the end, the toxicological modeling strategies, substantial challenges, and future perspectives for 3D printed tissue models were explored.
Collapse
Affiliation(s)
- Yi Xiang
- Department of NanoEngineering, University of California San Diego, La Jolla, USA
| | - Kathleen Miller
- Department of NanoEngineering, University of California San Diego, La Jolla, USA
| | - Jiaao Guan
- Department of Electrical and Computer Engineering, University of California San Diego, La Jolla, USA
| | | | - Min Tang
- Department of NanoEngineering, University of California San Diego, La Jolla, USA
| | - Shaochen Chen
- Department of NanoEngineering, University of California San Diego, La Jolla, USA.
- Department of Electrical and Computer Engineering, University of California San Diego, La Jolla, USA.
| |
Collapse
|
137
|
Pozo MR, Meredith GW, Entcheva E. Human iPSC-Cardiomyocytes as an Experimental Model to Study Epigenetic Modifiers of Electrophysiology. Cells 2022; 11:200. [PMID: 35053315 PMCID: PMC8774228 DOI: 10.3390/cells11020200] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/31/2021] [Accepted: 01/01/2022] [Indexed: 02/04/2023] Open
Abstract
The epigenetic landscape and the responses to pharmacological epigenetic regulators in each human are unique. Classes of epigenetic writers and erasers, such as histone acetyltransferases, HATs, and histone deacetylases, HDACs, control DNA acetylation/deacetylation and chromatin accessibility, thus exerting transcriptional control in a tissue- and person-specific manner. Rapid development of novel pharmacological agents in clinical testing-HDAC inhibitors (HDACi)-targets these master regulators as common means of therapeutic intervention in cancer and immune diseases. The action of these epigenetic modulators is much less explored for cardiac tissue, yet all new drugs need to be tested for cardiotoxicity. To advance our understanding of chromatin regulation in the heart, and specifically how modulation of DNA acetylation state may affect functional electrophysiological responses, human-induced pluripotent stem-cell-derived cardiomyocyte (hiPSC-CM) technology can be leveraged as a scalable, high-throughput platform with ability to provide patient-specific insights. This review covers relevant background on the known roles of HATs and HDACs in the heart, the current state of HDACi development, applications, and any adverse cardiac events; it also summarizes relevant differential gene expression data for the adult human heart vs. hiPSC-CMs along with initial transcriptional and functional results from using this new experimental platform to yield insights on epigenetic control of the heart. We focus on the multitude of methodologies and workflows needed to quantify responses to HDACis in hiPSC-CMs. This overview can help highlight the power and the limitations of hiPSC-CMs as a scalable experimental model in capturing epigenetic responses relevant to the human heart.
Collapse
Affiliation(s)
| | | | - Emilia Entcheva
- Department of Biomedical Engineering, George Washington University, Washington, DC 20052, USA; (M.R.P.); (G.W.M.)
| |
Collapse
|
138
|
Youhanna S, Kemas AM, Preiss L, Zhou Y, Shen JX, Cakal SD, Paqualini FS, Goparaju SK, Shafagh RZ, Lind JU, Sellgren CM, Lauschke VM. Organotypic and Microphysiological Human Tissue Models for Drug Discovery and Development-Current State-of-the-Art and Future Perspectives. Pharmacol Rev 2022; 74:141-206. [PMID: 35017176 DOI: 10.1124/pharmrev.120.000238] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 10/12/2021] [Indexed: 12/11/2022] Open
Abstract
The number of successful drug development projects has been stagnant for decades despite major breakthroughs in chemistry, molecular biology, and genetics. Unreliable target identification and poor translatability of preclinical models have been identified as major causes of failure. To improve predictions of clinical efficacy and safety, interest has shifted to three-dimensional culture methods in which human cells can retain many physiologically and functionally relevant phenotypes for extended periods of time. Here, we review the state of the art of available organotypic culture techniques and critically review emerging models of human tissues with key importance for pharmacokinetics, pharmacodynamics, and toxicity. In addition, developments in bioprinting and microfluidic multiorgan cultures to emulate systemic drug disposition are summarized. We close by highlighting important trends regarding the fabrication of organotypic culture platforms and the choice of platform material to limit drug absorption and polymer leaching while supporting the phenotypic maintenance of cultured cells and allowing for scalable device fabrication. We conclude that organotypic and microphysiological human tissue models constitute promising systems to promote drug discovery and development by facilitating drug target identification and improving the preclinical evaluation of drug toxicity and pharmacokinetics. There is, however, a critical need for further validation, benchmarking, and consolidation efforts ideally conducted in intersectoral multicenter settings to accelerate acceptance of these novel models as reliable tools for translational pharmacology and toxicology. SIGNIFICANCE STATEMENT: Organotypic and microphysiological culture of human cells has emerged as a promising tool for preclinical drug discovery and development that might be able to narrow the translation gap. This review discusses recent technological and methodological advancements and the use of these systems for hit discovery and the evaluation of toxicity, clearance, and absorption of lead compounds.
Collapse
Affiliation(s)
- Sonia Youhanna
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Aurino M Kemas
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Lena Preiss
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Yitian Zhou
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Joanne X Shen
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Selgin D Cakal
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Francesco S Paqualini
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Sravan K Goparaju
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Reza Zandi Shafagh
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Johan Ulrik Lind
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Carl M Sellgren
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| |
Collapse
|
139
|
Dudek J, Maack C. Mechano-energetic aspects of Barth syndrome. J Inherit Metab Dis 2022; 45:82-98. [PMID: 34423473 DOI: 10.1002/jimd.12427] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 07/28/2021] [Accepted: 08/19/2021] [Indexed: 12/22/2022]
Abstract
Energy-demanding organs like the heart are strongly dependent on oxidative phosphorylation in mitochondria. Oxidative phosphorylation is governed by the respiratory chain located in the inner mitochondrial membrane. The inner mitochondrial membrane is the only cellular membrane with significant amounts of the phospholipid cardiolipin, and cardiolipin was found to directly interact with a number of essential protein complexes, including respiratory chain complexes I to V. An inherited defect in the biogenesis of cardiolipin causes Barth syndrome, which is associated with cardiomyopathy, skeletal myopathy, neutropenia and growth retardation. Energy conversion is dependent on reducing equivalents, which are replenished by oxidative metabolism in the Krebs cycle. Cardiolipin deficiency in Barth syndrome also affects Krebs cycle activity, metabolite transport and mitochondrial morphology. During excitation-contraction coupling, calcium (Ca2+ ) released from the sarcoplasmic reticulum drives sarcomeric contraction. At the same time, Ca2+ influx into mitochondria drives the activation of Krebs cycle dehydrogenases and the regeneration of reducing equivalents. Reducing equivalents are essential not only for energy conversion, but also for maintaining a redox buffer, which is required to detoxify reactive oxygen species (ROS). Defects in CL may also affect Ca2+ uptake into mitochondria and thereby hamper energy supply and demand matching, but also detoxification of ROS. Here, we review the impact of cardiolipin deficiency on mitochondrial function in Barth syndrome and discuss potential therapeutic strategies.
Collapse
Affiliation(s)
- Jan Dudek
- Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Würzburg, Germany
| | - Christoph Maack
- Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Würzburg, Germany
| |
Collapse
|
140
|
Russo M, Cejas CM, Pitingolo G. Advances in microfluidic 3D cell culture for preclinical drug development. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 187:163-204. [PMID: 35094774 DOI: 10.1016/bs.pmbts.2021.07.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Drug development is often a very long, costly, and risky process due to the lack of reliability in the preclinical studies. Traditional current preclinical models, mostly based on 2D cell culture and animal testing, are not full representatives of the complex in vivo microenvironments and often fail. In order to reduce the enormous costs, both financial and general well-being, a more predictive preclinical model is needed. In this chapter, we review recent advances in microfluidic 3D cell culture showing how its development has allowed the introduction of in vitro microphysiological systems, laying the foundation for organ-on-a-chip technology. These findings provide the basis for numerous preclinical drug discovery assays, which raise the possibility of using micro-engineered systems as emerging alternatives to traditional models, based on 2D cell culture and animals.
Collapse
Affiliation(s)
- Maria Russo
- Microfluidics, MEMS, Nanostructures (MMN), CNRS UMR 8231, Institut Pierre Gilles de Gennes (IPGG) ESPCI Paris, PSL Research University, Paris France.
| | - Cesare M Cejas
- Microfluidics, MEMS, Nanostructures (MMN), CNRS UMR 8231, Institut Pierre Gilles de Gennes (IPGG) ESPCI Paris, PSL Research University, Paris France
| | - Gabriele Pitingolo
- Bioassays, Microsystems and Optical Engineering Unit, BIOASTER, Paris France
| |
Collapse
|
141
|
Camman M, Joanne P, Agbulut O, Hélary C. 3D models of dilated cardiomyopathy: Shaping the chemical, physical and topographical properties of biomaterials to mimic the cardiac extracellular matrix. Bioact Mater 2022; 7:275-291. [PMID: 34466733 PMCID: PMC8379361 DOI: 10.1016/j.bioactmat.2021.05.040] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 05/21/2021] [Accepted: 05/21/2021] [Indexed: 12/12/2022] Open
Abstract
The pathophysiology of dilated cardiomyopathy (DCM), one major cause of heart failure, is characterized by the dilation of the heart but remains poorly understood because of the lack of adequate in vitro models. Current 2D models do not allow for the 3D organotypic organization of cardiomyocytes and do not reproduce the ECM perturbations. In this review, the different strategies to mimic the chemical, physical and topographical properties of the cardiac tissue affected by DCM are presented. The advantages and drawbacks of techniques generating anisotropy required for the cardiomyocytes alignment are discussed. In addition, the different methods creating macroporosity and favoring organotypic organization are compared. Besides, the advances in the induced pluripotent stem cells technology to generate cardiac cells from healthy or DCM patients will be described. Thanks to the biomaterial design, some features of the DCM extracellular matrix such as stiffness, porosity, topography or chemical changes can impact the cardiomyocytes function in vitro and increase their maturation. By mimicking the affected heart, both at the cellular and at the tissue level, 3D models will enable a better understanding of the pathology and favor the discovery of novel therapies.
Collapse
Affiliation(s)
- Marie Camman
- Sorbonne Université, CNRS, UMR 7574, Laboratoire de Chimie de la Matière Condensée de Paris, 4 place Jussieu (case 174), F-75005, Paris, France
- Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), CNRS UMR 8256, Inserm ERL U1164, Biological Adaptation and Ageing, 7 quai St-Bernard (case 256), F-75005, Paris, France
| | - Pierre Joanne
- Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), CNRS UMR 8256, Inserm ERL U1164, Biological Adaptation and Ageing, 7 quai St-Bernard (case 256), F-75005, Paris, France
| | - Onnik Agbulut
- Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), CNRS UMR 8256, Inserm ERL U1164, Biological Adaptation and Ageing, 7 quai St-Bernard (case 256), F-75005, Paris, France
| | - Christophe Hélary
- Sorbonne Université, CNRS, UMR 7574, Laboratoire de Chimie de la Matière Condensée de Paris, 4 place Jussieu (case 174), F-75005, Paris, France
| |
Collapse
|
142
|
Pu WT. Experimental models of Barth syndrome. J Inherit Metab Dis 2022; 45:72-81. [PMID: 34370877 PMCID: PMC8814986 DOI: 10.1002/jimd.12423] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/01/2021] [Accepted: 08/05/2021] [Indexed: 01/03/2023]
Abstract
Mutation of the gene Tafazzin (TAZ) causes Barth syndrome, an X-linked disorder characterized by cardiomyopathy, skeletal muscle weakness, and neutropenia. TAZ is an acyltransferase that catalyzes the remodeling of cardiolipin, the signature phospholipid of the inner mitochondrial membrane. Here, we review the major model systems that have been established to study the role of cardiolipin remodeling in mitochondrial function and the pathogenesis of Barth syndrome. We summarize key features of each model and provide examples of how each has contributed to advance our understanding of TAZ function and Barth syndrome pathophysiology.
Collapse
Affiliation(s)
- William T. Pu
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave., Boston, MA 02115
- Harvard Stem Cell Institute, Harvard University, 7 Divinity Ave, Cambridge, MA 02138
- correspondence:
| |
Collapse
|
143
|
Xu Y, Phoon CKL, Ren M, Schlame M. A simple mechanistic explanation for Barth syndrome and cardiolipin remodeling. J Inherit Metab Dis 2022; 45:51-59. [PMID: 34611930 DOI: 10.1002/jimd.12445] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/24/2021] [Accepted: 10/04/2021] [Indexed: 12/23/2022]
Abstract
Barth syndrome is a multisystem disorder caused by an abnormal metabolism of the mitochondrial lipid cardiolipin. In this review, we discuss physical properties, biosynthesis, membrane assembly, and function of cardiolipin. We hypothesize that cardiolipin reduces packing stress in the inner mitochondrial membrane, which arises as a result of protein crowding. According to this hypothesis, patients with Barth syndrome are unable to meet peak energy demands because they fail to concentrate the proteins of oxidative phosphorylation to a high surface density in the inner mitochondrial membrane.
Collapse
Affiliation(s)
- Yang Xu
- Department of Anesthesiology, New York University School of Medicine, New York, New York, USA
| | - Colin K L Phoon
- Department of Pediatrics, New York University School of Medicine, New York, New York, USA
| | - Mindong Ren
- Department of Anesthesiology, New York University School of Medicine, New York, New York, USA
| | - Michael Schlame
- Department of Anesthesiology, New York University School of Medicine, New York, New York, USA
- Department of Cell Biology, New York University School of Medicine, New York, New York, USA
| |
Collapse
|
144
|
Hargrove-Grimes P, Low LA, Tagle DA. Microphysiological Systems: Stakeholder Challenges to Adoption in Drug Development. Cells Tissues Organs 2022; 211:269-281. [PMID: 34380142 PMCID: PMC8831652 DOI: 10.1159/000517422] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/14/2021] [Indexed: 01/03/2023] Open
Abstract
Microphysiological systems (MPS) or tissue chips/organs-on-chips are novel in vitro models that emulate human physiology at the most basic functional level. In this review, we discuss various hurdles to widespread adoption of MPS technology focusing on issues from multiple stakeholder sectors, e.g., academic MPS developers, commercial suppliers of platforms, the pharmaceutical and biotechnology industries, and regulatory organizations. Broad adoption of MPS technology has thus far been limited by a gap in translation between platform developers, end-users, regulatory agencies, and the pharmaceutical industry. In this brief review, we offer a perspective on the existing barriers and how end-users may help surmount these obstacles to achieve broader adoption of MPS technology.
Collapse
Affiliation(s)
- Passley Hargrove-Grimes
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Lucie A. Low
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Danilo A. Tagle
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
145
|
Budhathoki S, Graham C, Sethu P, Kannappan R. Engineered Aging Cardiac Tissue Chip Model for Studying Cardiovascular Disease. Cells Tissues Organs 2022; 211:348-359. [PMID: 34365455 PMCID: PMC8818062 DOI: 10.1159/000516954] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 04/29/2021] [Indexed: 01/03/2023] Open
Abstract
Due to the rapidly growing number of older people worldwide and the concomitant increase in cardiovascular complications, there is an urgent need for age-related cardiac disease modeling and drug screening platforms. In the present study, we developed a cardiac tissue chip model that incorporates hemodynamic loading and mimics essential aspects of the infarcted aging heart. We induced cellular senescence in H9c2 myoblasts using low-dose doxorubicin treatment. These senescent cells were then used to engineer cardiac tissue fibers, which were subjected to hemodynamic stresses associated with pressure-volume changes in the heart. Myocardial ischemia was modeled in the engineered cardiac tissue via hypoxic treatment. Our results clearly show that acute low-dose doxorubicin treatment-induced senescence, as evidenced by morphological and molecular markers, including enlarged and flattened nuclei, DNA damage response foci, and increased expression of cell cycle inhibitor p16INK4a, p53, and ROS. Under normal hemodynamic load, the engineered cardiac tissues demonstrated cell alignment and retained cardiac cell characteristics. Our senescent cardiac tissue model of hypoxia-induced myocardial infarction recapitulated the pathological disease hallmarks such as increased cell death and upregulated expression of ANP and BNP. In conclusion, the described methodology provides a novel approach to generate stress-induced aging cardiac cell phenotypes and engineer cardiac tissue chip models to study the cardiovascular disease pathologies associated with aging.
Collapse
Affiliation(s)
- Sachin Budhathoki
- Division of Cardiovascular Disease, Departments of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Caleb Graham
- Division of Cardiovascular Disease, Departments of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Palaniappan Sethu
- Division of Cardiovascular Disease, Departments of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ramaswamy Kannappan
- Division of Cardiovascular Disease, Departments of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
146
|
Bozelli JC, Epand RM. Interplay between cardiolipin and plasmalogens in Barth syndrome. J Inherit Metab Dis 2022; 45:99-110. [PMID: 34655242 DOI: 10.1002/jimd.12449] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 10/09/2021] [Accepted: 10/12/2021] [Indexed: 12/28/2022]
Abstract
Barth syndrome (BTHS) is a rare inherited metabolic disease resulting from mutations in the gene of the enzyme tafazzin, which catalyzes the acyl chain remodeling of the mitochondrial-specific lipid cardiolipin (CL). Tissue samples of individuals with BTHS present abnormalities in the level and the molecular species of CL. In addition, in tissues of a tafazzin knockdown mouse as well as in cells derived from BTHS patients it has been shown that plasmalogens, a subclass of glycerophospholipids, also have abnormal levels. Likewise, administration of a plasmalogen precursor to cells derived from BTHS patients led to an increase in plasmalogen and to some extent CL levels. These results indicate an interplay between CL and plasmalogens in BTHS. This interdependence is supported by the concomitant loss in these lipids in different pathological conditions. However, currently the molecular mechanism linking CL and plasmalogens is not fully understood. Here, a review of the evidence showing the linkage between the levels of CL and plasmalogens is presented. In addition, putative mechanisms that might play a role in this interplay are proposed. Finally, the opportunity of therapeutic approaches based on the regulation of plasmalogens as new therapies for the treatment of BTHS is discussed.
Collapse
Affiliation(s)
- José Carlos Bozelli
- Department of Biochemistry and Biomedical Sciences, McMaster University, Health Sciences Centre, Hamilton, Ontario, Canada
| | - Richard M Epand
- Department of Biochemistry and Biomedical Sciences, McMaster University, Health Sciences Centre, Hamilton, Ontario, Canada
| |
Collapse
|
147
|
Zou Z, Luo X, Chen Z, Zhang YS, Wen C. Emerging microfluidics-enabled platforms for osteoarthritis management: from benchtop to bedside. Theranostics 2022; 12:891-909. [PMID: 34976219 PMCID: PMC8692897 DOI: 10.7150/thno.62685] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 11/17/2021] [Indexed: 11/12/2022] Open
Abstract
Osteoarthritis (OA) is a prevalent debilitating age-related joint degenerative disease. It is a leading cause of pain and functional disability in older adults. Unfortunately, there is no cure for OA once the damage is established. Therefore, it promotes an urgent need for early detection and intervention of OA. Theranostics, combining therapy and diagnosis, emerges as a promising approach for OA management. However, OA theranostics is still in its infancy. Three fundamental needs have to be firstly fulfilled: i) a reliable OA model for disease pathogenesis investigation and drug screening, ii) an effective and precise diagnostic platform, and iii) an advanced fabrication approach for drug delivery and therapy. Meanwhile, microfluidics emerges as a versatile technology to address each of the needs and eventually boost the development of OA theranostics. Therefore, this review focuses on the applications of microfluidics, from benchtop to bedside, for OA modelling and drug screening, early diagnosis, and clinical therapy. We first introduce the basic pathophysiology of OA and point out the major unfilled research gaps in current OA management including lack of disease modelling and drug screening platforms, early diagnostic modalities and disease-modifying drugs and delivery approaches. Accordingly, we then summarize the state-of-the-art microfluidics technology for OA management from in vitro modelling and diagnosis to therapy. Given the existing promising results, we further discuss the future development of microfluidic platforms towards clinical translation at the crossroad of engineering and biomedicine.
Collapse
Affiliation(s)
- Zhou Zou
- Department of Biomedical Engineering, Faculty of Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, China
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong, China
| | - Xiaohe Luo
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong, China
| | - Zhengkun Chen
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong, China
- Currently at Department of Chemistry, University of Toronto, Toronto, Ontario M5S 3H6, Canada
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, Massachusetts 02139, United States
| | - Chunyi Wen
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong, China
- Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Hong Kong, China
| |
Collapse
|
148
|
Thompson R, Jefferies J, Wang S, Pu WT, Takemoto C, Hornby B, Heyman A, Chin MT, Vernon HJ. Current and future treatment approaches for Barth syndrome. J Inherit Metab Dis 2022; 45:17-28. [PMID: 34713454 DOI: 10.1002/jimd.12453] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 10/18/2021] [Accepted: 10/26/2021] [Indexed: 12/17/2022]
Abstract
Barth Syndrome is an X-linked disorder of mitochondrial cardiolipin metabolism caused by pathogenic variants in TAFAZZIN with pleiotropic effects including cardiomyopathy, neutropenia, growth delay, and skeletal myopathy. Management requires a multidisciplinary approach to the organ-specific manifestations including specialists from cardiology, hematology, nutrition, physical therapy, genetics, and metabolism. Currently, treatment is centered on management of specific clinical features, and is not targeted toward remediating the underlying biochemical defect. However, two clinical trials have been recently undertaken which target the mitochondrial pathology of this disease: a study to examine the effects of elamipretide, a cardiolipin targeted agent, and a study to examine the effects of bezafibrate, a peroxisome proliferator-activated receptor (PPAR) agonist. Treatments to directly target the defective TAFAZZIN pathway are under development, including enzyme and gene therapies.
Collapse
Affiliation(s)
- Reid Thompson
- Department of Pediatric Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - John Jefferies
- The Cardiovascular Institute, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Suya Wang
- Department of Cardiology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - William T Pu
- Department of Cardiology, Boston Children's Hospital, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| | - Clifford Takemoto
- Division of Clinical Hematology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Brittany Hornby
- Department of Physical Therapy, Kennedy Krieger Institute, Baltimore, Maryland, USA
| | - Andrea Heyman
- Department of Nutrition, Kennedy Krieger Institute, Baltimore, Maryland, USA
| | - Michael T Chin
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, USA
| | - Hilary J Vernon
- Department of Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Neurogenetics, Kennedy Krieger Institute, Baltimore, Maryland, USA
| |
Collapse
|
149
|
Paloschi V, Sabater-Lleal M, Middelkamp H, Vivas A, Johansson S, van der Meer A, Tenje M, Maegdefessel L. Organ-on-a-chip technology: a novel approach to investigate cardiovascular diseases. Cardiovasc Res 2021; 117:2742-2754. [PMID: 33729461 PMCID: PMC8683705 DOI: 10.1093/cvr/cvab088] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 03/13/2021] [Indexed: 02/06/2023] Open
Abstract
The development of organs-on-chip (OoC) has revolutionized in vitro cell-culture experiments by allowing a better mimicry of human physiology and pathophysiology that has consequently led researchers to gain more meaningful insights into disease mechanisms. Several models of hearts-on-chips and vessels-on-chips have been demonstrated to recapitulate fundamental aspects of the human cardiovascular system in the recent past. These 2D and 3D systems include synchronized beating cardiomyocytes in hearts-on-chips and vessels-on-chips with layer-based structures and the inclusion of physiological and pathological shear stress conditions. The opportunities to discover novel targets and to perform drug testing with chip-based platforms have substantially enhanced, thanks to the utilization of patient-derived cells and precise control of their microenvironment. These organ models will provide an important asset for future approaches to personalized cardiovascular medicine and improved patient care. However, certain technical and biological challenges remain, making the global utilization of OoCs to tackle unanswered questions in cardiovascular science still rather challenging. This review article aims to introduce and summarize published work on hearts- and vessels-on chips but also to provide an outlook and perspective on how these advanced in vitro systems can be used to tailor disease models with patient-specific characteristics.
Collapse
Affiliation(s)
- Valentina Paloschi
- Department for Vascular and Endovascular Surgery, Technical University Munich, Klinikum Rechts der Isar, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Berlin, Germany
| | - Maria Sabater-Lleal
- Research Institute of Hospital de la Santa Creu i Sant Pau, IIB Sant Pau, Genomics of Complex Diseases Group, Barcelona, Spain
- Cardiovascular Medicine Unit, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | - Aisen Vivas
- BIOS/Lab on a Chip, University of Twente, Enschede, The Netherlands
- Applied Stem Cell Technologies, University of Twente, Enschede, The Netherlands
| | - Sofia Johansson
- Department of Materials Science and Engineering, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | | | - Maria Tenje
- Department of Materials Science and Engineering, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Lars Maegdefessel
- Department for Vascular and Endovascular Surgery, Technical University Munich, Klinikum Rechts der Isar, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Berlin, Germany
- Molecular Vascular Medicine Unit, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
150
|
From Spheroids to Organoids: The Next Generation of Model Systems of Human Cardiac Regeneration in a Dish. Int J Mol Sci 2021; 22:ijms222413180. [PMID: 34947977 PMCID: PMC8708686 DOI: 10.3390/ijms222413180] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/02/2021] [Accepted: 12/05/2021] [Indexed: 12/12/2022] Open
Abstract
Organoids are tiny, self-organized, three-dimensional tissue cultures that are derived from the differentiation of stem cells. The growing interest in the use of organoids arises from their ability to mimic the biology and physiology of specific tissue structures in vitro. Organoids indeed represent promising systems for the in vitro modeling of tissue morphogenesis and organogenesis, regenerative medicine and tissue engineering, drug therapy testing, toxicology screening, and disease modeling. Although 2D cell cultures have been used for more than 50 years, even for their simplicity and low-cost maintenance, recent years have witnessed a steep rise in the availability of organoid model systems. Exploiting the ability of cells to re-aggregate and reconstruct the original architecture of an organ makes it possible to overcome many limitations of 2D cell culture systems. In vitro replication of the cellular micro-environment of a specific tissue leads to reproducing the molecular, biochemical, and biomechanical mechanisms that directly influence cell behavior and fate within that specific tissue. Lineage-specific self-organizing organoids have now been generated for many organs. Currently, growing cardiac organoid (cardioids) from pluripotent stem cells and cardiac stem/progenitor cells remains an open challenge due to the complexity of the spreading, differentiation, and migration of cardiac muscle and vascular layers. Here, we summarize the evolution of biological model systems from the generation of 2D spheroids to 3D organoids by focusing on the generation of cardioids based on the currently available laboratory technologies and outline their high potential for cardiovascular research.
Collapse
|