1
|
Siddique AB, Williams KA, Swami NS. Nanogrooved Elastomeric Diaphragm Arrays for Assessment of Cardiomyocytes under Synergistic Effects of Circular Mechanical Stimuli and Electrical Conductivity to Enhance Intercellular Communication. ACS Biomater Sci Eng 2024. [PMID: 39679605 DOI: 10.1021/acsbiomaterials.4c01298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Cardiovascular diseases remain the leading cause of mortality, necessitating advancements in in vitro cardiac tissue engineering platforms for improved disease modeling, drug screening, and regenerative therapies. The chief challenge to recapitulating the beating behavior of cardiomyocytes is creation of the circular stress profile experienced by hollow organs in the natural heart due to filling pressure and integrated strategies for intercellular communication to promote cell-to-cell connections. We present a platform featuring addressable arrays of nanogrooved polydimethylsiloxane (PDMS) diaphragms for cell alignment and circular mechanical stimulation, with embedded silver nanowires (AgNWs) for electrical cues, so that cardiomyocyte functionality can be assessed under these synergistic influences. Central to our innovation is a two-layer PDMS diaphragm design that electrically isolates the liquid metal (EGaIn) strain sensor in the bottom layer to enable detection and control of mechanical stimulation from conductive portions of embedded AgNWs in the top layer that supports cardiomyocyte culture and communication. In this manner, through localized detection and control of the circular mechanical stimulation, the essential role of multiaxial stretching on cardiomyocyte function is elucidated based on their contractility, sarcomere length, and connexin-43 expression. This in vitro platform can potentially transform cardiac tissue engineering, drug screening, and precision medicine approaches.
Collapse
Affiliation(s)
- Abdullah-Bin Siddique
- Electrical and Computer Engineering, University of Virginia, Charlottesville, Virginia 22904, United States
| | - Keith A Williams
- Electrical and Computer Engineering, University of Virginia, Charlottesville, Virginia 22904, United States
| | - Nathan S Swami
- Electrical and Computer Engineering, University of Virginia, Charlottesville, Virginia 22904, United States
- Chemistry, University of Virginia, Charlottesville, Virginia 22904, United States
| |
Collapse
|
2
|
Hill MC, Simonson B, Roselli C, Xiao L, Herndon CN, Chaffin M, Mantineo H, Atwa O, Bhasin H, Guedira Y, Bedi KC, Margulies KB, Klattenhoff CA, Tucker NR, Ellinor PT. Large-scale single-nuclei profiling identifies role for ATRNL1 in atrial fibrillation. Nat Commun 2024; 15:10002. [PMID: 39562555 PMCID: PMC11576987 DOI: 10.1038/s41467-024-54296-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 11/06/2024] [Indexed: 11/21/2024] Open
Abstract
Atrial fibrillation (AF) is the most common sustained arrhythmia in humans, yet the molecular basis of AF remains incompletely understood. To determine the cell type-specific transcriptional changes underlying AF, we perform single-nucleus RNA-seq (snRNA-seq) on left atrial (LA) samples from patients with AF and controls. From more than 175,000 nuclei we find that only cardiomyocytes (CMs) and macrophages (MΦs) have a significant number of differentially expressed genes in patients with AF. Attractin Like 1 (ATRNL1) was overexpressed in CMs among patients with AF and localized to the intercalated disks. Further, in both knockdown and overexpression experiments we identify a potent role for ATRNL1 in cell stress response, and in the modulation of the cardiac action potential. Finally, we detect an unexpected expression pattern for a leading AF candidate gene, KCNN3. In sum, we uncover a role for ATRNL1 which may serve as potential therapeutic target for this common arrhythmia.
Collapse
Affiliation(s)
- Matthew C Hill
- Cardiovascular Disease Initiative & Precision Cardiology Laboratory, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Bridget Simonson
- Cardiovascular Disease Initiative & Precision Cardiology Laboratory, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Carolina Roselli
- Cardiovascular Disease Initiative & Precision Cardiology Laboratory, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ling Xiao
- Cardiovascular Disease Initiative & Precision Cardiology Laboratory, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Caroline N Herndon
- Cardiovascular Disease Initiative & Precision Cardiology Laboratory, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Mark Chaffin
- Cardiovascular Disease Initiative & Precision Cardiology Laboratory, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Helene Mantineo
- Cardiovascular Disease Initiative & Precision Cardiology Laboratory, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ondine Atwa
- Cardiovascular Disease Initiative & Precision Cardiology Laboratory, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Harshit Bhasin
- Cardiovascular Disease Initiative & Precision Cardiology Laboratory, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Yasmine Guedira
- Cardiovascular Disease Initiative & Precision Cardiology Laboratory, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kenneth C Bedi
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kenneth B Margulies
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Nathan R Tucker
- Departments of Pharmacology and Medicine, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Patrick T Ellinor
- Cardiovascular Disease Initiative & Precision Cardiology Laboratory, The Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
3
|
Guo F, Jooken S, Ahmad A, Yu W, Deschaume O, Thielemans W, Bartic C. Optically Active, Paper-Based Scaffolds for 3D Cardiac Pacing. ACS APPLIED MATERIALS & INTERFACES 2024; 16:53449-53459. [PMID: 39332816 PMCID: PMC11472259 DOI: 10.1021/acsami.4c10183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 09/29/2024]
Abstract
In this work, we report the design and fabrication of a light-addressable, paper-based nanocomposite scaffold for optical pacing and read-out of in vitro grown cardiac tissue. The scaffold consists of paper cellulose microfibers functionalized with gold nanorods (GNRs) and semiconductor quantum dots (QDs), embedded in a cell-permissive collagen matrix. The GNRs enable cardiomyocyte activity modulation through local temperature gradients induced by modulated near-infrared (NIR) laser illumination, with the local temperature changes reported by temperature-dependent QD photoluminescence (PL). The micrometer-sized paper fibers promote the tubular organization of HL-1 cardiac muscle cells, while the NIR plasmonic stimulation modulates reversibly their activity. Given the nanoscale spatial resolution and facile fabrication, paper-based nanocomposite scaffolds with NIR modulation offer excellent alternatives to electrode-based or optogenetic methods for cell activity modulation, at the single cell level, and are compatible with 3D tissue constructs. Such paper-based optical platforms can provide new possibilities for the development of in vitro drug screening assays and heart disease modeling.
Collapse
Affiliation(s)
- Fanglei Guo
- Laboratory
for Soft Matter and Biophysics, Department of Physics and Astronomy, KU Leuven, 3001 Leuven, Belgium
| | - Stijn Jooken
- Laboratory
for Soft Matter and Biophysics, Department of Physics and Astronomy, KU Leuven, 3001 Leuven, Belgium
| | - Amin Ahmad
- Laboratory
for Soft Matter and Biophysics, Department of Physics and Astronomy, KU Leuven, 3001 Leuven, Belgium
| | - Wei Yu
- Laboratory
for Soft Matter and Biophysics, Department of Physics and Astronomy, KU Leuven, 3001 Leuven, Belgium
| | - Olivier Deschaume
- Laboratory
for Soft Matter and Biophysics, Department of Physics and Astronomy, KU Leuven, 3001 Leuven, Belgium
| | - Wim Thielemans
- Sustainable
Materials Lab, Department of Chemical Engineering, KU Leuven, Campus Kulak Kortrijk, Etienne Sabbelaan 53, Kortrijk 8500, Belgium
| | - Carmen Bartic
- Laboratory
for Soft Matter and Biophysics, Department of Physics and Astronomy, KU Leuven, 3001 Leuven, Belgium
| |
Collapse
|
4
|
Mair DB, Tsui JH, Higashi T, Koenig P, Dong Z, Chen JF, Meir JU, Smith AST, Lee PHU, Ahn EH, Countryman S, Sniadecki NJ, Kim DH. Spaceflight-induced contractile and mitochondrial dysfunction in an automated heart-on-a-chip platform. Proc Natl Acad Sci U S A 2024; 121:e2404644121. [PMID: 39312653 PMCID: PMC11459163 DOI: 10.1073/pnas.2404644121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 07/17/2024] [Indexed: 09/25/2024] Open
Abstract
With current plans for manned missions to Mars and beyond, the need to better understand, prevent, and counteract the harmful effects of long-duration spaceflight on the body is becoming increasingly important. In this study, an automated heart-on-a-chip platform was flown to the International Space Station on a 1-mo mission during which contractile cardiac function was monitored in real-time. Upon return to Earth, engineered human heart tissues (EHTs) were further analyzed with ultrastructural imaging and RNA sequencing to investigate the impact of prolonged microgravity on cardiomyocyte function and health. Spaceflight EHTs exhibited significantly reduced twitch forces, increased incidences of arrhythmias, and increased signs of sarcomere disruption and mitochondrial damage. Transcriptomic analyses showed an up-regulation of genes and pathways associated with metabolic disorders, heart failure, oxidative stress, and inflammation, while genes related to contractility and calcium signaling showed significant down-regulation. Finally, in silico modeling revealed a potential link between oxidative stress and mitochondrial dysfunction that corresponded with RNA sequencing results. This represents an in vitro model to faithfully reproduce the adverse effects of spaceflight on three-dimensional (3D)-engineered heart tissue.
Collapse
Affiliation(s)
- Devin B. Mair
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD21218
| | - Jonathan H. Tsui
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD21218
| | - Ty Higashi
- Department of Mechanical Engineering, University of Washington, Seattle, WA98195
| | - Paul Koenig
- BioServe Space Technologies, Ann and HJ Smead Department of Aerospace Engineering Sciences, University of Colorado, Boulder, CO80303
| | - Zhipeng Dong
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD21218
| | - Jeffrey F. Chen
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD21218
| | - Jessica U. Meir
- The National Aeronautics and Space Administration, NASA Johnson Space Center, Houston, TX77058
| | - Alec S. T. Smith
- Department of Physiology and Biophysics, University of Washington, Seattle, WA98195
| | - Peter H. U. Lee
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI02912
| | - Eun Hyun Ahn
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD21218
- Center for Microphysiological Systems, Johns Hopkins University, Baltimore, MD21205
| | - Stefanie Countryman
- BioServe Space Technologies, Ann and HJ Smead Department of Aerospace Engineering Sciences, University of Colorado, Boulder, CO80303
| | - Nathan J. Sniadecki
- Department of Mechanical Engineering, University of Washington, Seattle, WA98195
- Department of Bioengineering, University of Washington, Seattle, WA98195
- Center for Cardiovascular Biology, University of Washington, Seattle, WA98109
- Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA98109
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD21218
- Department of Bioengineering, University of Washington, Seattle, WA98195
- Center for Cardiovascular Biology, University of Washington, Seattle, WA98109
- Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA98109
- Department of Medicine, Johns Hopkins University, Baltimore, MD21205
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD21218
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD21218
| |
Collapse
|
5
|
Kistamás K, Lamberto F, Vaiciuleviciute R, Leal F, Muenthaisong S, Marte L, Subías-Beltrán P, Alaburda A, Arvanitis DN, Zana M, Costa PF, Bernotiene E, Bergaud C, Dinnyés A. The Current State of Realistic Heart Models for Disease Modelling and Cardiotoxicity. Int J Mol Sci 2024; 25:9186. [PMID: 39273136 PMCID: PMC11394806 DOI: 10.3390/ijms25179186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/18/2024] [Accepted: 08/22/2024] [Indexed: 09/15/2024] Open
Abstract
One of the many unresolved obstacles in the field of cardiovascular research is an uncompromising in vitro cardiac model. While primary cell sources from animal models offer both advantages and disadvantages, efforts over the past half-century have aimed to reduce their use. Additionally, obtaining a sufficient quantity of human primary cardiomyocytes faces ethical and legal challenges. As the practically unlimited source of human cardiomyocytes from induced pluripotent stem cells (hiPSC-CM) is now mostly resolved, there are great efforts to improve their quality and applicability by overcoming their intrinsic limitations. The greatest bottleneck in the field is the in vitro ageing of hiPSC-CMs to reach a maturity status that closely resembles that of the adult heart, thereby allowing for more appropriate drug developmental procedures as there is a clear correlation between ageing and developing cardiovascular diseases. Here, we review the current state-of-the-art techniques in the most realistic heart models used in disease modelling and toxicity evaluations from hiPSC-CM maturation through heart-on-a-chip platforms and in silico models to the in vitro models of certain cardiovascular diseases.
Collapse
Affiliation(s)
- Kornél Kistamás
- BioTalentum Ltd., Aulich Lajos Str 26, H-2100 Gödöllő, Hungary
| | - Federica Lamberto
- BioTalentum Ltd., Aulich Lajos Str 26, H-2100 Gödöllő, Hungary
- Department of Physiology and Animal Health, Institute of Physiology and Animal Nutrition, Hungarian University of Agriculture and Life Sciences, Páter Károly Str 1, H-2100 Gödöllő, Hungary
| | - Raminta Vaiciuleviciute
- Department of Regenerative Medicine, State Research Institute Innovative Medicine Centre, Santariskiu g. 5, LT-08406 Vilnius, Lithuania
| | - Filipa Leal
- Biofabics Lda, Rua Alfredo Allen 455, 4200-135 Porto, Portugal
| | | | - Luis Marte
- Digital Health Unit, Eurecat-Centre Tecnològic de Catalunya, 08005 Barcelona, Spain
| | - Paula Subías-Beltrán
- Digital Health Unit, Eurecat-Centre Tecnològic de Catalunya, 08005 Barcelona, Spain
| | - Aidas Alaburda
- Department of Regenerative Medicine, State Research Institute Innovative Medicine Centre, Santariskiu g. 5, LT-08406 Vilnius, Lithuania
- Institute of Biosciences, Life Sciences Center, Vilnius University, Sauletekio al. 7, LT-10257 Vilnius, Lithuania
| | - Dina N Arvanitis
- Laboratory for Analysis and Architecture of Systems-French National Centre for Scientific Research (LAAS-CNRS), 7 Avenue du Colonel Roche, F-31400 Toulouse, France
| | - Melinda Zana
- BioTalentum Ltd., Aulich Lajos Str 26, H-2100 Gödöllő, Hungary
| | - Pedro F Costa
- Biofabics Lda, Rua Alfredo Allen 455, 4200-135 Porto, Portugal
| | - Eiva Bernotiene
- Department of Regenerative Medicine, State Research Institute Innovative Medicine Centre, Santariskiu g. 5, LT-08406 Vilnius, Lithuania
- Faculty of Fundamental Sciences, Vilnius Tech, Sauletekio al. 11, LT-10223 Vilnius, Lithuania
| | - Christian Bergaud
- Laboratory for Analysis and Architecture of Systems-French National Centre for Scientific Research (LAAS-CNRS), 7 Avenue du Colonel Roche, F-31400 Toulouse, France
| | - András Dinnyés
- BioTalentum Ltd., Aulich Lajos Str 26, H-2100 Gödöllő, Hungary
- Department of Physiology and Animal Health, Institute of Physiology and Animal Nutrition, Hungarian University of Agriculture and Life Sciences, Páter Károly Str 1, H-2100 Gödöllő, Hungary
| |
Collapse
|
6
|
Farboud SP, Fathi E, Valipour B, Farahzadi R. Toward the latest advancements in cardiac regeneration using induced pluripotent stem cells (iPSCs) technology: approaches and challenges. J Transl Med 2024; 22:783. [PMID: 39175068 PMCID: PMC11342568 DOI: 10.1186/s12967-024-05499-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 07/10/2024] [Indexed: 08/24/2024] Open
Abstract
A novel approach to treating heart failures was developed with the introduction of iPSC technology. Knowledge in regenerative medicine, developmental biology, and the identification of illnesses at the cellular level has exploded since the discovery of iPSCs. One of the most frequent causes of mortality associated with cardiovascular disease is the loss of cardiomyocytes (CMs), followed by heart failure. A possible treatment for heart failure involves restoring cardiac function and replacing damaged tissue with healthy, regenerated CMs. Significant strides in stem cell biology during the last ten years have transformed the in vitro study of human illness and enhanced our knowledge of the molecular pathways underlying human disease, regenerative medicine, and drug development. We seek to examine iPSC advancements in disease modeling, drug discovery, iPSC-Based cell treatments, and purification methods in this article.
Collapse
Affiliation(s)
- Seyedeh Parya Farboud
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Ezzatollah Fathi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran.
| | - Behnaz Valipour
- Department of Anatomical Sciences, Sarab Faculty of Medical Sciences, Sarab, Iran
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Raheleh Farahzadi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
7
|
Cooke JP, Youker KA, Lai L. Myocardial Recovery versus Myocardial Regeneration: Mechanisms and Therapeutic Modulation. Methodist Debakey Cardiovasc J 2024; 20:31-41. [PMID: 39184159 PMCID: PMC11342844 DOI: 10.14797/mdcvj.1400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 06/12/2024] [Indexed: 08/27/2024] Open
Abstract
Myocardial recovery is characterized by a return toward normal structure and function of the heart after an injury. Mechanisms of myocardial recovery include restoration and/or adaptation of myocyte structure and function, mitochondrial activity and number, metabolic homeostasis, electrophysiological stability, extracellular matrix remodeling, and myocardial perfusion. Myocardial regeneration is an element of myocardial recovery that involves the generation of new myocardial tissue, a process which is limited in adult humans but may be therapeutically augmented. Understanding the mechanisms of myocardial recovery and myocardial regeneration will lead to novel therapies for heart failure.
Collapse
Affiliation(s)
- John P. Cooke
- Houston Methodist Academic Institute, Houston, Texas, US
| | | | - Li Lai
- Houston Methodist Academic Institute, Houston, Texas, US
| |
Collapse
|
8
|
Soroudi S, Jaafari MR, Arabi L. Lipid nanoparticle (LNP) mediated mRNA delivery in cardiovascular diseases: Advances in genome editing and CAR T cell therapy. J Control Release 2024; 372:113-140. [PMID: 38876358 DOI: 10.1016/j.jconrel.2024.06.023] [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: 01/09/2024] [Revised: 06/05/2024] [Accepted: 06/09/2024] [Indexed: 06/16/2024]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of global mortality among non-communicable diseases. Current cardiac regeneration treatments have limitations and may lead to adverse reactions. Hence, innovative technologies are needed to address these shortcomings. Messenger RNA (mRNA) emerges as a promising therapeutic agent due to its versatility in encoding therapeutic proteins and targeting "undruggable" conditions. It offers low toxicity, high transfection efficiency, and controlled protein production without genome insertion or mutagenesis risk. However, mRNA faces challenges such as immunogenicity, instability, and difficulty in cellular entry and endosomal escape, hindering its clinical application. To overcome these hurdles, lipid nanoparticles (LNPs), notably used in COVID-19 vaccines, have a great potential to deliver mRNA therapeutics for CVDs. This review highlights recent progress in mRNA-LNP therapies for CVDs, including Myocardial Infarction (MI), Heart Failure (HF), and hypercholesterolemia. In addition, LNP-mediated mRNA delivery for CAR T-cell therapy and CRISPR/Cas genome editing in CVDs and the related clinical trials are explored. To enhance the efficiency, safety, and clinical translation of mRNA-LNPs, advanced technologies like artificial intelligence (AGILE platform) in RNA structure design, and optimization of LNP formulation could be integrated. We conclude that the strategies to facilitate the extra-hepatic delivery and targeted organ tropism of mRNA-LNPs (SORT, ASSET, SMRT, and barcoded LNPs) hold great prospects to accelerate the development and translation of mRNA-LNPs in CVD treatment.
Collapse
Affiliation(s)
- Setareh Soroudi
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Leila Arabi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
9
|
Ghasemzaie N, Jeyhani M, Joshi K, Lee WL, Tsai SSH. ATPSpin: A Single Microfluidic Platform that Produces Diversified ATPS-Alginate Microfibers. ACS Biomater Sci Eng 2024; 10:3896-3908. [PMID: 38748191 DOI: 10.1021/acsbiomaterials.4c00110] [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] [Indexed: 06/11/2024]
Abstract
Microfluidic spinning is emerging as a useful technique in the fabrication of alginate fibers, enabling applications in drug screening, disease modeling, and disease diagnostics. In this paper, by capitalizing on the benefits of aqueous two-phase systems (ATPS) to produce diverse alginate fiber forms, we introduce an ATPS-Spinning platform (ATPSpin). This ATPS-enabled method efficiently circumvents the rapid clogging challenges inherent to traditional fiber production techniques by regulating the interaction between alginate and cross-linking agents like Ba2+ ions. By varying system parameters under the guidance of a regime map, our system produces several fiber forms─solid, hollow, and droplet-filled─consistently and reproducibly from a single device. We demonstrate that the resulting alginate fibers possess distinct features, including biocompatibility. We also encapsulate HEK293 cells in the microfibers as a proof-of-concept that this versatile microfluidic fiber generation platform may have utility in tissue engineering and regenerative medicine applications.
Collapse
Affiliation(s)
- Niloofar Ghasemzaie
- Biomedical Engineering Graduate Program, Toronto Metropolitan University, Toronto, Ontario, Canada M5B 2K3
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada M5B 1T8
- Institute for Biomedical Engineering, Science and Technology (iBEST), Toronto, Ontario, Canada M5B 1T8
| | - Morteza Jeyhani
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada M5B 1T8
- Department of Mechanical, Industrial, and Mechatronics Engineering, Toronto Metropolitan University, Toronto, Ontario, Canada M5B 2K3
- Institute for Biomedical Engineering, Science and Technology (iBEST), Toronto, Ontario, Canada M5B 1T8
| | - Kushal Joshi
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada M5B 1T8
- Department of Mechanical, Industrial, and Mechatronics Engineering, Toronto Metropolitan University, Toronto, Ontario, Canada M5B 2K3
- Institute for Biomedical Engineering, Science and Technology (iBEST), Toronto, Ontario, Canada M5B 1T8
| | - Warren L Lee
- Biomedical Engineering Graduate Program, Toronto Metropolitan University, Toronto, Ontario, Canada M5B 2K3
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada M5B 1T8
- Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Ontario, Canada M5S 1A1
- Institute for Biomedical Engineering, Science and Technology (iBEST), Toronto, Ontario, Canada M5B 1T8
| | - Scott S H Tsai
- Biomedical Engineering Graduate Program, Toronto Metropolitan University, Toronto, Ontario, Canada M5B 2K3
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada M5B 1T8
- Department of Mechanical, Industrial, and Mechatronics Engineering, Toronto Metropolitan University, Toronto, Ontario, Canada M5B 2K3
- Institute for Biomedical Engineering, Science and Technology (iBEST), Toronto, Ontario, Canada M5B 1T8
| |
Collapse
|
10
|
Ma J, Ross L, Grube C, Wang HS. Toxicity of low dose bisphenols in human iPSC-derived cardiomyocytes and human cardiac organoids - Impact on contractile function and hypertrophy. CHEMOSPHERE 2024; 353:141567. [PMID: 38417488 DOI: 10.1016/j.chemosphere.2024.141567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 02/23/2024] [Accepted: 02/25/2024] [Indexed: 03/01/2024]
Abstract
Bisphenol A (BPA) and its analogs are common environmental chemicals with various adverse health impacts, including cardiac toxicity. In this study, we examined the long term effect of low dose BPA and three common BPA analogs, bisphenol S (BPS), bisphenol F (BPF), and bisphenol AF (BPAF), in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) based models. HiPSC-CMs and human cardiac organoids were exposed to these chemicals for 4-5 or 20 days. 1 nM BPA, BPS, and BPAF, but not BPF, resulted in suppressed myocyte contractility, retarded contraction kinetics, and aberrant Ca2+ transients in hiPSC-CMs. In cardiac organoids, BPAF and BPA, but not the other bisphenols, resulted in suppressed contraction and Ca2+ transients, and aberrant contraction kinetics. The order of toxicities was BPAF > BPA>∼BPS > BPF and the toxicities of BPAF and BPA were more pronounced under longer exposure. The impact of BPAF on myocyte contraction and Ca2+ handling was mediated by reduction of sarcoplasmic reticulum Ca2+ load and inhibition of L-type Ca2+ channel involving alternation of Ca2+ handling proteins. Impaired myocyte Ca2+ handling plays a key role in cardiac pathophysiology and is a characteristic of cardiac hypertrophy; therefore we examined the potential pro-hypertrophic cardiotoxicity of these bisphenols. Four to five day exposure to BPAF did not cause hypertrophy in normal hiPSC-CMs, but significantly exacerbated the hypertrophic phenotype in myocytes with existing hypertrophy induced by endothelin-1, characterized by increased cell size and elevated expression of the hypertrophic marker proBNP. This pro-hypertrophic cardiotoxicity was also occurred in cardiac organoids, with BPAF having the strongest toxicity, followed by BPA. Our findings demonstrate that long term exposures to BPA and some of its analogs cause contractile dysfunction and abnormal Ca2+ handling, and have potential pro-hypertrophic cardiotoxicity in human heart cells/tissues, and suggest that some bisphenol chemicals may be a risk factor for cardiac hypertrophy in human hearts.
Collapse
Affiliation(s)
- Jianyong Ma
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267 USA.
| | - Leah Ross
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267 USA
| | - Christian Grube
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267 USA
| | - Hong-Sheng Wang
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267 USA
| |
Collapse
|
11
|
Raniga K, Nasir A, Vo NTN, Vaidyanathan R, Dickerson S, Hilcove S, Mosqueira D, Mirams GR, Clements P, Hicks R, Pointon A, Stebbeds W, Francis J, Denning C. Strengthening cardiac therapy pipelines using human pluripotent stem cell-derived cardiomyocytes. Cell Stem Cell 2024; 31:292-311. [PMID: 38366587 DOI: 10.1016/j.stem.2024.01.007] [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: 09/14/2023] [Revised: 11/27/2023] [Accepted: 01/19/2024] [Indexed: 02/18/2024]
Abstract
Advances in hiPSC isolation and reprogramming and hPSC-CM differentiation have prompted their therapeutic application and utilization for evaluating potential cardiovascular safety liabilities. In this perspective, we showcase key efforts toward the large-scale production of hiPSC-CMs, implementation of hiPSC-CMs in industry settings, and recent clinical applications of this technology. The key observations are a need for traceable gender and ethnically diverse hiPSC lines, approaches to reduce cost of scale-up, accessible clinical trial datasets, and transparent guidelines surrounding the safety and efficacy of hiPSC-based therapies.
Collapse
Affiliation(s)
- Kavita Raniga
- The Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK; Pathology, Non-Clinical Safety, GlaxoSmithKline R&D, Stevenage SG1 2NY, UK.
| | - Aishah Nasir
- The Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | - Nguyen T N Vo
- The Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | | | | | | | - Diogo Mosqueira
- The Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | - Gary R Mirams
- Centre for Mathematical Medicine & Biology, School of Mathematical Sciences, University of Nottingham, Nottingham NG7 2RD, UK
| | - Peter Clements
- Pathology, Non-Clinical Safety, GlaxoSmithKline R&D, Stevenage SG1 2NY, UK
| | - Ryan Hicks
- BioPharmaceuticals R&D Cell Therapy Department, Research and Early Development, Cardiovascular, Renal, and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden; School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, London WC2R 2LS, UK
| | - Amy Pointon
- Safety Sciences, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge CB2 0AA, UK
| | | | - Jo Francis
- Mechanstic Biology and Profiling, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge CB2 0AA, UK
| | - Chris Denning
- The Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK.
| |
Collapse
|
12
|
Garg A, Lavine KJ, Greenberg MJ. Assessing Cardiac Contractility From Single Molecules to Whole Hearts. JACC Basic Transl Sci 2024; 9:414-439. [PMID: 38559627 PMCID: PMC10978360 DOI: 10.1016/j.jacbts.2023.07.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 07/14/2023] [Accepted: 07/14/2023] [Indexed: 04/04/2024]
Abstract
Fundamentally, the heart needs to generate sufficient force and power output to dynamically meet the needs of the body. Cardiomyocytes contain specialized structures referred to as sarcomeres that power and regulate contraction. Disruption of sarcomeric function or regulation impairs contractility and leads to cardiomyopathies and heart failure. Basic, translational, and clinical studies have adapted numerous methods to assess cardiac contraction in a variety of pathophysiological contexts. These tools measure aspects of cardiac contraction at different scales ranging from single molecules to whole organisms. Moreover, these studies have revealed new pathogenic mechanisms of heart disease leading to the development of novel therapies targeting contractility. In this review, the authors explore the breadth of tools available for studying cardiac contractile function across scales, discuss their strengths and limitations, highlight new insights into cardiac physiology and pathophysiology, and describe how these insights can be harnessed for therapeutic candidate development and translational.
Collapse
Affiliation(s)
- Ankit Garg
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri, USA
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Kory J. Lavine
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Michael J. Greenberg
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
13
|
Sakamoto T, Kelly DP. Cardiac maturation. J Mol Cell Cardiol 2024; 187:38-50. [PMID: 38160640 PMCID: PMC10923079 DOI: 10.1016/j.yjmcc.2023.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/12/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024]
Abstract
The heart undergoes a dynamic maturation process following birth, in response to a wide range of stimuli, including both physiological and pathological cues. This process entails substantial re-programming of mitochondrial energy metabolism coincident with the emergence of specialized structural and contractile machinery to meet the demands of the adult heart. Many components of this program revert to a more "fetal" format during development of pathological cardiac hypertrophy and heart failure. In this review, emphasis is placed on recent progress in our understanding of the transcriptional control of cardiac maturation, encompassing the results of studies spanning from in vivo models to cardiomyocytes derived from human stem cells. The potential applications of this current state of knowledge to new translational avenues aimed at the treatment of heart failure is also addressed.
Collapse
Affiliation(s)
- Tomoya Sakamoto
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daniel P Kelly
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
14
|
Babini H, Jiménez-Sábado V, Stogova E, Arslanova A, Butt M, Dababneh S, Asghari P, Moore EDW, Claydon TW, Chiamvimonvat N, Hove-Madsen L, Tibbits GF. hiPSC-derived cardiomyocytes as a model to study the role of small-conductance Ca 2+-activated K + (SK) ion channel variants associated with atrial fibrillation. Front Cell Dev Biol 2024; 12:1298007. [PMID: 38304423 PMCID: PMC10830749 DOI: 10.3389/fcell.2024.1298007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 01/05/2024] [Indexed: 02/03/2024] Open
Abstract
Atrial fibrillation (AF), the most common arrhythmia, has been associated with different electrophysiological, molecular, and structural alterations in atrial cardiomyocytes. Therefore, more studies are required to elucidate the genetic and molecular basis of AF. Various genome-wide association studies (GWAS) have strongly associated different single nucleotide polymorphisms (SNPs) with AF. One of these GWAS identified the rs13376333 risk SNP as the most significant one from the 1q21 chromosomal region. The rs13376333 risk SNP is intronic to the KCNN3 gene that encodes for small conductance calcium-activated potassium channels type 3 (SK3). However, the functional electrophysiological effects of this variant are not known. SK channels represent a unique family of K+ channels, primarily regulated by cytosolic Ca2+ concentration, and different studies support their critical role in the regulation of atrial excitability and consequently in the development of arrhythmias like AF. Since different studies have shown that both upregulation and downregulation of SK3 channels can lead to arrhythmias by different mechanisms, an important goal is to elucidate whether the rs13376333 risk SNP is a gain-of-function (GoF) or a loss-of-function (LoF) variant. A better understanding of the functional consequences associated with these SNPs could influence clinical practice guidelines by improving genotype-based risk stratification and personalized treatment. Although research using native human atrial cardiomyocytes and animal models has provided useful insights, each model has its limitations. Therefore, there is a critical need to develop a human-derived model that represents human physiology more accurately than existing animal models. In this context, research with human induced pluripotent stem cells (hiPSC) and subsequent generation of cardiomyocytes derived from hiPSC (hiPSC-CMs) has revealed the underlying causes of various cardiovascular diseases and identified treatment opportunities that were not possible using in vitro or in vivo studies with animal models. Thus, the ability to generate atrial cardiomyocytes and atrial tissue derived from hiPSCs from human/patients with specific genetic diseases, incorporating novel genetic editing tools to generate isogenic controls and organelle-specific reporters, and 3D bioprinting of atrial tissue could be essential to study AF pathophysiological mechanisms. In this review, we will first give an overview of SK-channel function, its role in atrial fibrillation and outline pathophysiological mechanisms of KCNN3 risk SNPs. We will then highlight the advantages of using the hiPSC-CM model to investigate SNPs associated with AF, while addressing limitations and best practices for rigorous hiPSC studies.
Collapse
Affiliation(s)
- Hosna Babini
- Cellular and Regenerative Medicine Centre, BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Verónica Jiménez-Sábado
- Cellular and Regenerative Medicine Centre, BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
- IIB SANT PAU, and CIBERCV, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Ekaterina Stogova
- Cellular and Regenerative Medicine Centre, BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Alia Arslanova
- Cellular and Regenerative Medicine Centre, BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Mariam Butt
- Cellular and Regenerative Medicine Centre, BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Saif Dababneh
- Cellular and Regenerative Medicine Centre, BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Department of Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Parisa Asghari
- Department of Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Edwin D. W. Moore
- Department of Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Thomas W. Claydon
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | | | - Leif Hove-Madsen
- IIB SANT PAU, and CIBERCV, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Instituto de Investigaciones Biomédicas de Barcelona (IIBB-CSIC), Barcelona, Spain
| | - Glen F. Tibbits
- Cellular and Regenerative Medicine Centre, BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
- Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
15
|
Shar A, Shar A, Joung D. Carbon nanotube nanocomposite scaffolds: advances in fabrication and applications for tissue regeneration and cancer therapy. Front Bioeng Biotechnol 2023; 11:1299166. [PMID: 38179128 PMCID: PMC10764633 DOI: 10.3389/fbioe.2023.1299166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 12/08/2023] [Indexed: 01/06/2024] Open
Abstract
Carbon nanotube (CNT) nanocomposite scaffolds have emerged as highly promising frameworks for tissue engineering research. By leveraging their intrinsic electrical conductivity and valuable mechanical properties, CNTs are commonly dispersed into polymers to create robust, electrically conductive scaffolds that facilitate tissue regeneration and remodeling. This article explores the latest progress and challenges related to CNT dispersion, functionalization, and scaffold printing techniques, including electrospinning and 3D printing. Notably, these CNT scaffolds have demonstrated remarkable positive effects across various cell culture systems, stimulating neuronal growth, promoting cardiomyocyte maturation, and facilitating osteocyte differentiation. These encouraging results have sparked significant interest within the regenerative medicine field, including neural, cardiac, muscle, and bone regenerations. However, addressing the concern of CNT cytotoxicity in these scaffolds remains critical. Consequently, substantial efforts are focused on exploring strategies to minimize cytotoxicity associated with CNT-based scaffolds. Moreover, researchers have also explored the intriguing possibility of utilizing the natural cytotoxic properties of CNTs to selectively target cancer cells, opening up promising avenues for cancer therapy. More research should be conducted on cutting-edge applications of CNT-based scaffolds through phototherapy and electrothermal ablation. Unlike drug delivery systems, these novel methodologies can combine 3D additive manufacturing with the innate physical properties of CNT in response to electromagnetic stimuli to efficiently target localized tumors. Taken together, the unique properties of CNT-based nanocomposite scaffolds position them as promising candidates for revolutionary breakthroughs in both regenerative medicine and cancer treatment. Continued research and innovation in this area hold significant promise for improving healthcare outcomes.
Collapse
Affiliation(s)
- Andy Shar
- Department of Physics, Virginia Commonwealth University, Richmond, VA, United States
| | - Angela Shar
- College of Medicine, University of Florida, Gainesville, FL, United States
| | - Daeha Joung
- Department of Physics, Virginia Commonwealth University, Richmond, VA, United States
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
16
|
Moriwaki T, Tani H, Haga K, Morita-Umei Y, Soma Y, Umei TC, Sekine O, Takatsuna K, Kishino Y, Kanazawa H, Fujita J, Fukuda K, Tohyama S, Ieda M. Scalable production of homogeneous cardiac organoids derived from human pluripotent stem cells. CELL REPORTS METHODS 2023; 3:100666. [PMID: 38113855 PMCID: PMC10753388 DOI: 10.1016/j.crmeth.2023.100666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 08/24/2023] [Accepted: 11/16/2023] [Indexed: 12/21/2023]
Abstract
Three-dimensional (3D) cultures are known to more closely mimic in vivo conditions compared with 2D cultures. Cardiac spheroids (CSs) and organoids (COs) are useful for 3D tissue engineering and are advantageous for their simplicity and mass production for regenerative therapy and drug discovery. Herein, we describe a large-scale method for producing homogeneous human induced pluripotent stem cell (hiPSC)-derived CSs (hiPSC-CSs) and COs without scaffolds using a porous 3D microwell substratum with a suction system. Our method has many advantages, such as increased efficiency and improved functionality, homogeneity, and sphericity of hiPSC-CSs. Moreover, we have developed a substratum on a clinically relevant large scale for regenerative therapy and have succeeded in producing approximately 40,000 hiPSC-CSs with high sphericity at once. Furthermore, we efficiently produced a fused CO model consisting of hiPSC-derived atrial and ventricular cardiomyocytes localized on opposite sides of one organoid. This method will facilitate progress toward hiPSC-based clinical applications.
Collapse
Affiliation(s)
- Taijun Moriwaki
- Department of Cardiology, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Hidenori Tani
- Department of Cardiology, Keio University School of Medicine, Shinjuku, Tokyo, Japan; Joint Research Laboratory for Medical Innovation in Heart Disease, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Kotaro Haga
- Department of Cardiology, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Yuika Morita-Umei
- Department of Cardiology, Keio University School of Medicine, Shinjuku, Tokyo, Japan; Kanagawa Institute of Industrial Science and Technology (KISTEC), Kawasaki, Kanagawa, Japan
| | - Yusuke Soma
- Department of Cardiology, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Tomohiko C Umei
- Department of Cardiology, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Otoya Sekine
- Department of Cardiology, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Kaworu Takatsuna
- Department of Cardiology, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Yoshikazu Kishino
- Department of Cardiology, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Hideaki Kanazawa
- Department of Cardiology, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Jun Fujita
- Department of Cardiology, Keio University School of Medicine, Shinjuku, Tokyo, Japan; Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Keiichi Fukuda
- Department of Cardiology, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Shugo Tohyama
- Department of Cardiology, Keio University School of Medicine, Shinjuku, Tokyo, Japan.
| | - Masaki Ieda
- Department of Cardiology, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| |
Collapse
|
17
|
Finkel S, Sweet S, Locke T, Smith S, Wang Z, Sandini C, Imredy J, He Y, Durante M, Lagrutta A, Feinberg A, Lee A. FRESH™ 3D bioprinted cardiac tissue, a bioengineered platform for in vitro pharmacology. APL Bioeng 2023; 7:046113. [PMID: 38046544 PMCID: PMC10693443 DOI: 10.1063/5.0163363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 10/30/2023] [Indexed: 12/05/2023] Open
Abstract
There is critical need for a predictive model of human cardiac physiology in drug development to assess compound effects on human tissues. In vitro two-dimensional monolayer cultures of cardiomyocytes provide biochemical and cellular readouts, and in vivo animal models provide information on systemic cardiovascular response. However, there remains a significant gap in these models due to their incomplete recapitulation of adult human cardiovascular physiology. Recent efforts in developing in vitro models from engineered heart tissues have demonstrated potential for bridging this gap using human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) in three-dimensional tissue structure. Here, we advance this paradigm by implementing FRESH™ 3D bioprinting to build human cardiac tissues in a medium throughput, well-plate format with controlled tissue architecture, tailored cellular composition, and native-like physiological function, specifically in its drug response. We combined hiPSC-CMs, endothelial cells, and fibroblasts in a cellular bioink and FRESH™ 3D bioprinted this mixture in the format of a thin tissue strip stabilized on a tissue fixture. We show that cardiac tissues could be fabricated directly in a 24-well plate format were composed of dense and highly aligned hiPSC-CMs at >600 million cells/mL and, within 14 days, demonstrated reproducible calcium transients and a fast conduction velocity of ∼16 cm/s. Interrogation of these cardiac tissues with the β-adrenergic receptor agonist isoproterenol showed responses consistent with positive chronotropy and inotropy. Treatment with calcium channel blocker verapamil demonstrated responses expected of hiPSC-CM derived cardiac tissues. These results confirm that FRESH™ 3D bioprinted cardiac tissues represent an in vitro platform that provides data on human physiological response.
Collapse
Affiliation(s)
| | | | - Tyler Locke
- FluidForm, Inc., Waltham, Massachusetts 02451, USA
| | - Sydney Smith
- FluidForm, Inc., Waltham, Massachusetts 02451, USA
| | - Zhefan Wang
- FluidForm, Inc., Waltham, Massachusetts 02451, USA
| | | | - John Imredy
- In Vitro Safety Pharmacology, Genetic and Cellular Toxicology, Merck & Co. Inc., Rahway, New Jersey 07065, USA
| | - Yufang He
- Division of Technology, Infrastructure, Operations and Experience, Merck & Co. Inc., Rahway, New Jersey 07065, USA
| | - Marc Durante
- Division of Technology, Infrastructure, Operations and Experience, Merck & Co. Inc., Rahway, New Jersey 07065, USA
| | - Armando Lagrutta
- In Vitro Safety Pharmacology, Genetic and Cellular Toxicology, Merck & Co. Inc., Rahway, New Jersey 07065, USA
| | | | - Andrew Lee
- FluidForm, Inc., Waltham, Massachusetts 02451, USA
| |
Collapse
|
18
|
Aitova A, Berezhnoy A, Tsvelaya V, Gusev O, Lyundup A, Efimov AE, Agapov I, Agladze K. Biomimetic Cardiac Tissue Models for In Vitro Arrhythmia Studies. Biomimetics (Basel) 2023; 8:487. [PMID: 37887618 PMCID: PMC10604593 DOI: 10.3390/biomimetics8060487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/26/2023] [Accepted: 10/03/2023] [Indexed: 10/28/2023] Open
Abstract
Cardiac arrhythmias are a major cause of cardiovascular mortality worldwide. Many arrhythmias are caused by reentry, a phenomenon where excitation waves circulate in the heart. Optical mapping techniques have revealed the role of reentry in arrhythmia initiation and fibrillation transition, but the underlying biophysical mechanisms are still difficult to investigate in intact hearts. Tissue engineering models of cardiac tissue can mimic the structure and function of native cardiac tissue and enable interactive observation of reentry formation and wave propagation. This review will present various approaches to constructing cardiac tissue models for reentry studies, using the authors' work as examples. The review will highlight the evolution of tissue engineering designs based on different substrates, cell types, and structural parameters. A new approach using polymer materials and cellular reprogramming to create biomimetic cardiac tissues will be introduced. The review will also show how computational modeling of cardiac tissue can complement experimental data and how such models can be applied in the biomimetics of cardiac tissue.
Collapse
Affiliation(s)
- Aleria Aitova
- Laboratory of Experimental and Cellular Medicine, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Russia
- M.F. Vladimirsky Moscow Regional Clinical Research Institute, 129110 Moscow, Russia
- Almetyevsk State Oil Institute, 423450 Almetyevsk, Russia
| | - Andrey Berezhnoy
- Laboratory of Experimental and Cellular Medicine, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Russia
- M.F. Vladimirsky Moscow Regional Clinical Research Institute, 129110 Moscow, Russia
- Almetyevsk State Oil Institute, 423450 Almetyevsk, Russia
| | - Valeriya Tsvelaya
- Laboratory of Experimental and Cellular Medicine, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Russia
- M.F. Vladimirsky Moscow Regional Clinical Research Institute, 129110 Moscow, Russia
- Almetyevsk State Oil Institute, 423450 Almetyevsk, Russia
| | - Oleg Gusev
- Regulatory Genomics Research Center, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420018 Kazan, Russia
- Life Improvement by Future Technologies (LIFT) Center, 143025 Moscow, Russia
- Intractable Disease Research Center, Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
| | | | - Anton E. Efimov
- Academician V.I. Shumakov National Medical Research Center of Transplantology and Artificial Organs, Ministry of Health of the Russian Federation, 123182 Moscow, Russia
| | - Igor Agapov
- Academician V.I. Shumakov National Medical Research Center of Transplantology and Artificial Organs, Ministry of Health of the Russian Federation, 123182 Moscow, Russia
| | - Konstantin Agladze
- Laboratory of Experimental and Cellular Medicine, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Russia
- M.F. Vladimirsky Moscow Regional Clinical Research Institute, 129110 Moscow, Russia
| |
Collapse
|
19
|
Sekine O, Kanaami S, Masumoto K, Aihara Y, Morita-Umei Y, Tani H, Soma Y, Umei TC, Haga K, Moriwaki T, Kawai Y, Ohno M, Kishino Y, Kanazawa H, Fukuda K, Ieda M, Tohyama S. Seamless and non-destructive monitoring of extracellular microRNAs during cardiac differentiation from human pluripotent stem cells. Stem Cell Reports 2023; 18:1925-1939. [PMID: 37738969 PMCID: PMC10656301 DOI: 10.1016/j.stemcr.2023.08.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 08/20/2023] [Accepted: 08/21/2023] [Indexed: 09/24/2023] Open
Abstract
Monitoring cardiac differentiation and maturation from human pluripotent stem cells (hPSCs) and detecting residual undifferentiated hPSCs are indispensable for the development of cardiac regenerative therapy. MicroRNA (miRNA) is secreted from cells into the extracellular space, and its role as a biomarker is attracting attention. Here, we performed an miRNA array analysis of supernatants during the process of cardiac differentiation and maturation from hPSCs. We demonstrated that the quantification of extracellular miR-489-3p and miR-1/133a-3p levels enabled the monitoring of mesoderm and cardiac differentiation, respectively, even in clinical-grade mass culture systems. Moreover, extracellular let-7c-5p levels showed the greatest increase with cardiac maturation during long-term culture. We also verified that residual undifferentiated hPSCs in hPSC-derived cardiomyocytes (hPSC-CMs) were detectable by measuring miR-302b-3p expression, with a detection sensitivity of 0.01%. Collectively, we demonstrate that our method of seamlessly monitoring specific miRNAs secreted into the supernatant is non-destructive and effective for the quality evaluation of hPSC-CMs.
Collapse
Affiliation(s)
- Otoya Sekine
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Sayaka Kanaami
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; Heartseed Inc, The Artcomplex Center of Tokyo, #302, 12-9, Daikyo-cho, Shinjuku-ku, Tokyo 160-0015, Japan
| | - Kanako Masumoto
- Sysmex Corporation, Central Research Laboratories, 4-4-4 Takatsukadai, Nishi-ku, Kobe 651-2271, Japan
| | - Yuki Aihara
- Sysmex Corporation, Central Research Laboratories, 4-4-4 Takatsukadai, Nishi-ku, Kobe 651-2271, Japan
| | - Yuika Morita-Umei
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; Kanagawa Institute of Industrial Science and Technology (KISTEC), Kawasaki, Kanagawa, Japan
| | - Hidenori Tani
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; Joint Research Laboratory for Medical Innovation in Heart Disease, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Yusuke Soma
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Tomohiko C Umei
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Kotaro Haga
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Taijun Moriwaki
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Yujiro Kawai
- Department of Cardiovascular Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Masatoshi Ohno
- Department of Cardiovascular Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Yoshikazu Kishino
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Hideaki Kanazawa
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Keiichi Fukuda
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; Heartseed Inc, The Artcomplex Center of Tokyo, #302, 12-9, Daikyo-cho, Shinjuku-ku, Tokyo 160-0015, Japan
| | - Masaki Ieda
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Shugo Tohyama
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan.
| |
Collapse
|
20
|
Thakar RG, Fenton KN. Bioethical implications of organ-on-a-chip on modernizing drug development. Artif Organs 2023; 47:1553-1558. [PMID: 37578206 PMCID: PMC10615722 DOI: 10.1111/aor.14620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Organ-on-chips are three-dimensional microdevices that emulate the structure, functionality, and behavior of specific tissues or organs using human cells. Combining organoids with microfabricated fluidic channels and microelectronics, these systems offer a promising platform for studying disease mechanisms, drug responses, and tissue performance. By replicating the in vivo microenvironment, these devices can recreate complex cell interactions in controlled conditions and facilitate research in various fields, including drug toxicity and efficacy studies, biochemical analysis, and disease pathogenesis. Integrating human induced pluripotent stem cells further enhances their applicability, thereby enabling patient-specific disease modeling for precision medicine. Although challenges like economy-of-scale, multichip integration, and regulatory compliance exist, advances in this modular technology show promise for lowering drug development costs, improving reproducibility, and reducing the reliance on animal testing. The ethical landscape surrounding organ-on-chip usage presents both benefits and concerns. While these chips offer an alternative to animal testing and potential cost savings, they raise ethical considerations related to community engagement, informed consent, and the need for standardized guidelines. Ensuring public acceptance and involvement in decision-making is vital to address misinformation and mistrust. Furthermore, personalized medicine models using patient-derived cells demand careful consideration of potential ethical dilemmas, such as modeling physiological functions of fetuses or brains and determining the extent of protection for these models. To achieve the full potential of organ-on-a-chip models, collaboration between scientists, ethicists, and regulators is essential to fulfil the promise of transforming drug development, advancing personalized medicine, and contributing to a more ethical and efficient biomedical research landscape.
Collapse
Affiliation(s)
- Rahul G. Thakar
- Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Kathleen N. Fenton
- Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
- Department of Bioethics, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
21
|
Ainsworth MJ, Chirico N, de Ruijter M, Hrynevich A, Dokter I, Sluijter JPG, Malda J, van Mil A, Castilho M. Convergence of melt electrowriting and extrusion-based bioprinting for vascular patterning of a myocardial construct. Biofabrication 2023; 15:035025. [PMID: 37343567 DOI: 10.1088/1758-5090/ace07f] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 06/21/2023] [Indexed: 06/23/2023]
Abstract
To progress cardiac tissue engineering strategies closer to the clinic, thicker constructs are required to meet the functional need following a cardiac event. Consequently, pre-vascularization of these constructs needs to be investigated to ensure survival and optimal performance of implantable engineered heart tissue. The aim of this research is to investigate the potential of combining extrusion-based bioprinting (EBB) and melt electrowriting for the fabrication of a myocardial construct with a precisely patterned pre-vascular pathway. Gelatin methacryloyl (GelMA) was investigated as a base hydrogel for the respective myocardial and vascular bioinks with collagen, Matrigel and fibrinogen as interpenetrating polymers to support myocardial functionality. Subsequently, extrusion-based printability and viability were investigated to determine the optimal processing parameters for printing into melt electrowritten meshes. Finally, an anatomically inspired vascular pathway was implemented in a dual EBB set-up into melt electrowritten meshes, creating a patterned pre-vascularized myocardial construct. It was determined that a blend of 5% GelMA and 0.8 mg·ml-1collagen with a low crosslinked density was optimal for myocardial cellular arrangement and alignment within the constructs. For the vascular fraction, the optimized formulation consisted of 5% GelMA, 0.8 mg·ml-1collagen and 1 mg·ml-1fibrinogen with a higher crosslinked density, which led to enhanced vascular cell connectivity. Printability assessment confirmed that the optimized bioinks could effectively fill the microfiber mesh while supporting cell viability (∼70%). Finally, the two bioinks were applied using a dual EBB system for the fabrication of a pre-vascular pathway with the shape of a left anterior descending artery within a myocardial construct, whereby the distinct cell populations could be visualized in their respective patterns up to D14. This research investigated the first step towards developing a thick engineered cardiac tissue construct in which a pre-vascularization pathway is fabricated within a myocardial construct.
Collapse
Affiliation(s)
- Madison Jade Ainsworth
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Nino Chirico
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Cardiology, Experimental Cardiology Laboratory, University Medical Center Utrecht, Utrecht, The Netherlands
- Circulatory Health Research Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Mylène de Ruijter
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Andrei Hrynevich
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Clinical Sciences, Faculty of Veterinary Sciences, Utrecht University, Utrecht, The Netherlands
| | - Inge Dokter
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Cardiology, Experimental Cardiology Laboratory, University Medical Center Utrecht, Utrecht, The Netherlands
- Circulatory Health Research Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Joost P G Sluijter
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Cardiology, Experimental Cardiology Laboratory, University Medical Center Utrecht, Utrecht, The Netherlands
- Circulatory Health Research Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jos Malda
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Clinical Sciences, Faculty of Veterinary Sciences, Utrecht University, Utrecht, The Netherlands
| | - Alain van Mil
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Cardiology, Experimental Cardiology Laboratory, University Medical Center Utrecht, Utrecht, The Netherlands
- Circulatory Health Research Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Miguel Castilho
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| |
Collapse
|
22
|
Li C, Hao J, Zheng Y, Wang C, Yang J, Wang W, Zhang K, Shao C, Hui W, Wang J, Li W, Tang YD. The changing landscape of drug clinical trials on cardiometabolic diseases in China, 2009-2021. Diabetol Metab Syndr 2023; 15:66. [PMID: 37005689 PMCID: PMC10067219 DOI: 10.1186/s13098-023-01043-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 03/25/2023] [Indexed: 04/04/2023] Open
Abstract
BACKGROUND Cardiometabolic disease is a clinical syndrome characterized by multiple metabolic disorders, with atherosclerosis as the core and cardiovascular and cerebrovascular events as the outcome. Drug research and development (R&D) in cardiometabolic diseases has grown rapidly worldwide. However, the development of cardiometabolic drug clinical trials in China remains unclear. This study aims to depict the changing landscape of drug clinical trials for cardiometabolic diseases in China during 2009-2021. METHODS The detailed information of drug trials on cardiometabolic diseases registered in the National Medical Products Administration (NMPA) Registration and Information Disclosure Platform was collected between January 1, 2009, and July 1, 2021. The landscape of cardiometabolic drug clinical trials was analyzed by the characteristics, time trends, indications, pharmacological mechanisms, and geographical distribution. RESULTS A total of 2466 drug clinical trials on cardiometabolic diseases were extracted and analyzed. The annual number of drug trials increased rapidly in the past twelve years. Among all the trials, the bioequivalence trials (1428; 58.3%) accounted for the largest proportion, followed by phase I (555; 22.5%), phase III (278; 11.3%), phase II (169; 6.9%), and phase IV (26; 1.1%). Of 2466 trials, 2133 (86.5%) trials were monomer drugs, only 236 (9.6%) trials were polypills and 97 (3.9%) were traditional Chinese medicine (TCM) compounds. In terms of pharmacological mechanisms, the number of trials in dihydropyridine (DHP) calcium antagonists 321 (11.9%) ranked first, while trials in angiotensin receptor blocker (ARB) 289 (10.7%) and dipeptidyl peptidase-4 (DPP-4) inhibitor 205 (7.6%) ranked second and third place respectively. Of 236 chemical polypills trials, 23 (9.7%) polypills were the combination of DHP calcium antagonists and statins, while others were the combination of two same pharmacological effect agents. As for the geographical distribution of leading units, 36 trials were led by principal investigators (PI) units from Beijing, followed by Jiangsu (n = 29), Shanghai (n = 19), Guangdong (n = 19), and Hunan (n = 19), showing an uneven regional distribution. CONCLUSIONS Great progress has been made in drug clinical trials on cardiometabolic diseases, especially in antihypertensive agents, hypoglycemic agents, and hypolipidemic agents. However, the insufficient innovation of first-in-class drugs and polypills should be carefully considered by all stakeholders in drug trials.
Collapse
Affiliation(s)
- Chen Li
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Jun Hao
- Medical Research and Biometrics Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College, National Clinical Research Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, 100037, China
| | - Yitian Zheng
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Chuangshi Wang
- Medical Research and Biometrics Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College, National Clinical Research Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, 100037, China
| | - Jie Yang
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Wenyao Wang
- Department of Cardiology, Institute of Vascular Medicine, Key Laboratory of Molecular Cardiovascular Science, Peking University Third Hospital, Ministry of Education, Beijing, 100191, China
| | - Kuo Zhang
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Chunli Shao
- Department of Cardiology, Institute of Vascular Medicine, Key Laboratory of Molecular Cardiovascular Science, Peking University Third Hospital, Ministry of Education, Beijing, 100191, China
| | - Wen Hui
- Department of Science and Technology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jiancheng Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Wei Li
- Medical Research and Biometrics Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College, National Clinical Research Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, 100037, China.
| | - Yi-Da Tang
- Department of Cardiology, Institute of Vascular Medicine, Key Laboratory of Molecular Cardiovascular Science, Peking University Third Hospital, Ministry of Education, Beijing, 100191, China.
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
23
|
Kawaguchi N, Nakanishi T. Animal Disease Models and Patient-iPS-Cell-Derived In Vitro Disease Models for Cardiovascular Biology-How Close to Disease? BIOLOGY 2023; 12:468. [PMID: 36979160 PMCID: PMC10045735 DOI: 10.3390/biology12030468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/15/2023] [Accepted: 03/17/2023] [Indexed: 03/22/2023]
Abstract
Currently, zebrafish, rodents, canines, and pigs are the primary disease models used in cardiovascular research. In general, larger animals have more physiological similarities to humans, making better disease models. However, they can have restricted or limited use because they are difficult to handle and maintain. Moreover, animal welfare laws regulate the use of experimental animals. Different species have different mechanisms of disease onset. Organs in each animal species have different characteristics depending on their evolutionary history and living environment. For example, mice have higher heart rates than humans. Nonetheless, preclinical studies have used animals to evaluate the safety and efficacy of human drugs because no other complementary method exists. Hence, we need to evaluate the similarities and differences in disease mechanisms between humans and experimental animals. The translation of animal data to humans contributes to eliminating the gap between these two. In vitro disease models have been used as another alternative for human disease models since the discovery of induced pluripotent stem cells (iPSCs). Human cardiomyocytes have been generated from patient-derived iPSCs, which are genetically identical to the derived patients. Researchers have attempted to develop in vivo mimicking 3D culture systems. In this review, we explore the possible uses of animal disease models, iPSC-derived in vitro disease models, humanized animals, and the recent challenges of machine learning. The combination of these methods will make disease models more similar to human disease.
Collapse
Affiliation(s)
- Nanako Kawaguchi
- Department of Pediatric Cardiology and Adult Congenital Cardiology, Tokyo Women’s Medical University, Tokyo 162-8666, Japan;
| | | |
Collapse
|
24
|
Martin M, Gähwiler EKN, Generali M, Hoerstrup SP, Emmert MY. Advances in 3D Organoid Models for Stem Cell-Based Cardiac Regeneration. Int J Mol Sci 2023; 24:ijms24065188. [PMID: 36982261 PMCID: PMC10049446 DOI: 10.3390/ijms24065188] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/03/2023] [Accepted: 03/06/2023] [Indexed: 03/11/2023] Open
Abstract
The adult human heart cannot regain complete cardiac function following tissue injury, making cardiac regeneration a current clinical unmet need. There are a number of clinical procedures aimed at reducing ischemic damage following injury; however, it has not yet been possible to stimulate adult cardiomyocytes to recover and proliferate. The emergence of pluripotent stem cell technologies and 3D culture systems has revolutionized the field. Specifically, 3D culture systems have enhanced precision medicine through obtaining a more accurate human microenvironmental condition to model disease and/or drug interactions in vitro. In this study, we cover current advances and limitations in stem cell-based cardiac regenerative medicine. Specifically, we discuss the clinical implementation and limitations of stem cell-based technologies and ongoing clinical trials. We then address the advent of 3D culture systems to produce cardiac organoids that may better represent the human heart microenvironment for disease modeling and genetic screening. Finally, we delve into the insights gained from cardiac organoids in relation to cardiac regeneration and further discuss the implications for clinical translation.
Collapse
Affiliation(s)
- Marcy Martin
- Institute for Regenerative Medicine (IREM), University of Zurich, 8952 Schlieren, Switzerland
| | - Eric K. N. Gähwiler
- Institute for Regenerative Medicine (IREM), University of Zurich, 8952 Schlieren, Switzerland
| | - Melanie Generali
- Institute for Regenerative Medicine (IREM), University of Zurich, 8952 Schlieren, Switzerland
| | - Simon P. Hoerstrup
- Institute for Regenerative Medicine (IREM), University of Zurich, 8952 Schlieren, Switzerland
- Wyss Zurich Translational Center, University of Zurich and ETH Zurich, 8092 Zurich, Switzerland
| | - Maximilian Y. Emmert
- Institute for Regenerative Medicine (IREM), University of Zurich, 8952 Schlieren, Switzerland
- Wyss Zurich Translational Center, University of Zurich and ETH Zurich, 8092 Zurich, Switzerland
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), 13353 Berlin, Germany
- Department of Cardiovascular Surgery, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany
- Correspondence: ; Tel.: +41-44-634-5610
| |
Collapse
|
25
|
Motta SE, Martin M, Gähwiler EKN, Visser VL, Zaytseva P, Ehterami A, Hoerstrup SP, Emmert MY. Combining Cell Technologies With Biomimetic Tissue Engineering Applications: A New Paradigm for Translational Cardiovascular Therapies. Stem Cells Transl Med 2023; 12:72-82. [PMID: 36806699 PMCID: PMC9985110 DOI: 10.1093/stcltm/szad002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 12/24/2022] [Indexed: 02/22/2023] Open
Abstract
Cardiovascular disease is a major cause of morbidity and mortality worldwide and, to date, the clinically available prostheses still present several limitations. The design of next-generation regenerative replacements either based on cellular or extracellular matrix technologies can address these shortcomings. Therefore, tissue engineered constructs could potentially become a promising alterative to the current therapeutic options for patients with cardiovascular diseases. In this review, we selectively present an overview of the current tissue engineering tools such as induced pluripotent stem cells, biomimetic materials, computational modeling, and additive manufacturing technologies, with a focus on their application to translational cardiovascular therapies. We discuss how these advanced technologies can help the development of biomimetic tissue engineered constructs and we finally summarize the latest clinical evidence for their use, and their potential therapeutic outcome.
Collapse
Affiliation(s)
- Sarah E Motta
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
| | - Marcy Martin
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
| | - Eric K N Gähwiler
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
| | - Valery L Visser
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
| | - Polina Zaytseva
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
| | - Arian Ehterami
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
| | - Simon P Hoerstrup
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
- Wyss Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Maximilian Y Emmert
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
- Wyss Zurich, University and ETH Zurich, Zurich, Switzerland
- Charité Universitätsmedizin Berlin, Berlin, Germany
- Deutsches Herzzentrum der Charité (DHZC), Dept of Cardiothoracic and Vascular Surgery, Berlin, Germany
| |
Collapse
|
26
|
Min S, Cho SW. Engineered human cardiac tissues for modeling heart diseases. BMB Rep 2023; 56:32-42. [PMID: 36443005 PMCID: PMC9887099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Indexed: 01/28/2023] Open
Abstract
Heart disease is one of the major life-threatening diseases with high mortality and incidence worldwide. Several model systems, such as primary cells and animals, have been used to understand heart diseases and establish appropriate treatments. However, they have limitations in accuracy and reproducibility in recapitulating disease pathophysiology and evaluating drug responses. In recent years, three-dimensional (3D) cardiac tissue models produced using tissue engineering technology and human cells have outperformed conventional models. In particular, the integration of cell reprogramming techniques with bioengineering platforms (e.g., microfluidics, scaffolds, bioprinting, and biophysical stimuli) has facilitated the development of heart-ona- chip, cardiac spheroid/organoid, and engineered heart tissue (EHT) to recapitulate the structural and functional features of the native human heart. These cardiac models have improved heart disease modeling and toxicological evaluation. In this review, we summarize the cell types for the fabrication of cardiac tissue models, introduce diverse 3D human cardiac tissue models, and discuss the strategies to enhance their complexity and maturity. Finally, recent studies in the modeling of various heart diseases are reviewed. [BMB Reports 2023; 56(1): 32-42].
Collapse
Affiliation(s)
- Sungjin Min
- Department of Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul 03722, Korea,Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Korea,Corresponding author. Tel: +82-2-2123-5662; Fax: +82-2-362-7265; E-mail:
| |
Collapse
|
27
|
Min S, Cho SW. Engineered human cardiac tissues for modeling heart diseases. BMB Rep 2023; 56:32-42. [PMID: 36443005 PMCID: PMC9887099 DOI: 10.5483/bmbrep.2022-0185] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 11/28/2022] [Accepted: 11/28/2022] [Indexed: 07/30/2023] Open
Abstract
Heart disease is one of the major life-threatening diseases with high mortality and incidence worldwide. Several model systems, such as primary cells and animals, have been used to understand heart diseases and establish appropriate treatments. However, they have limitations in accuracy and reproducibility in recapitulating disease pathophysiology and evaluating drug responses. In recent years, three-dimensional (3D) cardiac tissue models produced using tissue engineering technology and human cells have outperformed conventional models. In particular, the integration of cell reprogramming techniques with bioengineering platforms (e.g., microfluidics, scaffolds, bioprinting, and biophysical stimuli) has facilitated the development of heart-ona- chip, cardiac spheroid/organoid, and engineered heart tissue (EHT) to recapitulate the structural and functional features of the native human heart. These cardiac models have improved heart disease modeling and toxicological evaluation. In this review, we summarize the cell types for the fabrication of cardiac tissue models, introduce diverse 3D human cardiac tissue models, and discuss the strategies to enhance their complexity and maturity. Finally, recent studies in the modeling of various heart diseases are reviewed. [BMB Reports 2023; 56(1): 32-42].
Collapse
Affiliation(s)
- Sungjin Min
- Department of Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul 03722, Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Korea
| |
Collapse
|
28
|
Hwang M, Lee SJ, Lim CH, Shim EB, Lee HA. The three-dimensionality of the hiPSC-CM spheroid contributes to the variability of the field potential. Front Physiol 2023; 14:1123190. [PMID: 37025386 PMCID: PMC10070703 DOI: 10.3389/fphys.2023.1123190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 03/10/2023] [Indexed: 04/08/2023] Open
Abstract
Background: Field potential (FP) signals from human induced pluripotent stem cell-derived cardiomyocyte (hiPSC-CM) spheroid which are used for drug safety tests in the preclinical stage are different from action potential (AP) signals and require working knowledge of the multi-electrode array (MEA) system. In this study, we developed in silico three-dimensional (3-D) models of hiPSC-CM spheroids for the simulation of field potential measurement. We compared our model simulation results against in vitro experimental data under the effect of drugs E-4031 and nifedipine. Methods: In silico 3-D models of hiPSC-CM spheroids were constructed in spherical and discoidal shapes. Tetrahedral meshes were generated inside the models, and the propagation of the action potential in the model was obtained by numerically solving the monodomain reaction-diffusion equation. An electrical model of electrode was constructed and FPs were calculated using the extracellular potentials from the AP propagations. The effects of drugs were simulated by matching the simulation results with in vitro experimental data. Results: The simulated FPs from the 3-D models of hiPSC-CM spheroids exhibited highly variable shapes depending on the stimulation and measurement locations. The values of the IC50 of E-4031 and nifedipine calculated by matching the simulated FP durations with in vitro experimental data were in line with the experimentally measured ones reported in the literature. Conclusion: The 3-D in silico models of hiPSC-CM spheroids generated highly variable FPs similar to those observed in in vitro experiments. The in silico model has the potential to complement the interpretation of the FP signals obtained from in vitro experiments.
Collapse
Affiliation(s)
| | - Su-Jin Lee
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon, Republic of Korea
| | | | - Eun Bo Shim
- AI Medic, Inc., Seoul, Republic of Korea
- Department of Mechanical and Biomedical Engineering, Kangwon National University, Chuncheon, Republic of Korea
- *Correspondence: Eun Bo Shim, ; Hyang-Ae Lee,
| | - Hyang-Ae Lee
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon, Republic of Korea
- *Correspondence: Eun Bo Shim, ; Hyang-Ae Lee,
| |
Collapse
|
29
|
Zhu K, Bao X, Wang Y, Lu T, Zhang L. Human induced pluripotent stem cell (hiPSC)-derived cardiomyocyte modelling of cardiovascular diseases for natural compound discovery. Biomed Pharmacother 2023; 157:113970. [PMID: 36371854 DOI: 10.1016/j.biopha.2022.113970] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/29/2022] [Accepted: 11/01/2022] [Indexed: 11/11/2022] Open
Abstract
Cardiovascular disease (CVD) remains the leading cause of death worldwide. Natural compounds extracted from medicinal plants characterized by diverse biological activities and low toxicity or side effects, are increasingly taking center stage in the search for new drugs. Currently, preclinical evaluation of natural products relies mainly on the use of immortalized cell lines of human origin or animal models. Increasing evidence indicates that cardiomyopathy models based on immortalized cell lines do not recapitulate pathogenic phenotypes accurately and a substantial physiological discrepancy between animals and humans casts doubt on the clinical relevance of animal models for these studies. The newly developed human induced pluripotent stem cell (hiPSC) technology in combination with highly-efficient cardiomyocyte differentiation methods provides an ideal tool for modeling human cardiomyopathies in vitro. Screening of drugs, especially screening of natural products, based on these models has been widely used and has shown that evaluation in such models can recapitulate important aspects of the physiological properties of drugs. The purpose of this review is to provide information on the latest developments in this area of research and to help researchers perform screening of natural products using the hiPSC-CM platform.
Collapse
Affiliation(s)
- Keyang Zhu
- Zhejiang Key Laboratory of Pathophysiology, School of Public Health, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, PR China
| | - Xiaoming Bao
- Department of Cardiology, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang, PR China; Department of Global Health, Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, Zhejiang, PR China
| | - Yingchao Wang
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Ting Lu
- Clinical Research Center of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China.
| | - Ling Zhang
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, PR China.
| |
Collapse
|