351
|
Huebsch N, Loskill P, Deveshwar N, Spencer CI, Judge LM, Mandegar MA, Fox CB, Mohamed TMA, Ma Z, Mathur A, Sheehan AM, Truong A, Saxton M, Yoo J, Srivastava D, Desai TA, So PL, Healy KE, Conklin BR. Miniaturized iPS-Cell-Derived Cardiac Muscles for Physiologically Relevant Drug Response Analyses. Sci Rep 2016; 6:24726. [PMID: 27095412 PMCID: PMC4837370 DOI: 10.1038/srep24726] [Citation(s) in RCA: 166] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 04/05/2016] [Indexed: 01/16/2023] Open
Abstract
Tissue engineering approaches have the potential to increase the physiologic relevance of human iPS-derived cells, such as cardiomyocytes (iPS-CM). However, forming Engineered Heart Muscle (EHM) typically requires >1 million cells per tissue. Existing miniaturization strategies involve complex approaches not amenable to mass production, limiting the ability to use EHM for iPS-based disease modeling and drug screening. Micro-scale cardiospheres are easily produced, but do not facilitate assembly of elongated muscle or direct force measurements. Here we describe an approach that combines features of EHM and cardiospheres: Micro-Heart Muscle (μHM) arrays, in which elongated muscle fibers are formed in an easily fabricated template, with as few as 2,000 iPS-CM per individual tissue. Within μHM, iPS-CM exhibit uniaxial contractility and alignment, robust sarcomere assembly, and reduced variability and hypersensitivity in drug responsiveness, compared to monolayers with the same cellular composition. μHM mounted onto standard force measurement apparatus exhibited a robust Frank-Starling response to external stretch, and a dose-dependent inotropic response to the β-adrenergic agonist isoproterenol. Based on the ease of fabrication, the potential for mass production and the small number of cells required to form μHM, this system provides a potentially powerful tool to study cardiomyocyte maturation, disease and cardiotoxicology in vitro.
Collapse
Affiliation(s)
- Nathaniel Huebsch
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158.,Department of Pediatrics, University of California, San Francisco, CA 94143
| | - Peter Loskill
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, California 94720, USA.,Department of Materials Science and Engineering, University of California at Berkeley, Berkeley, California 94720, USA
| | - Nikhil Deveshwar
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, California 94720, USA
| | - C Ian Spencer
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158
| | - Luke M Judge
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158.,Department of Pediatrics, University of California, San Francisco, CA 94143
| | | | - Cade B Fox
- University of California, San Francisco, Schools of Pharmacy and Medicine, Department of Bioengineering and Therapeutic Sciences, San Francisco, CA 94158
| | - Tamer M A Mohamed
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158.,Institute of Cardiovascular Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, United Kingdom.,Faculty of Pharmacy, Zagazig University, EL-Sharkiak, Egypt
| | - Zhen Ma
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, California 94720, USA.,Department of Materials Science and Engineering, University of California at Berkeley, Berkeley, California 94720, USA
| | - Anurag Mathur
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, California 94720, USA.,Department of Materials Science and Engineering, University of California at Berkeley, Berkeley, California 94720, USA
| | - Alice M Sheehan
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158
| | - Annie Truong
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158
| | - Mike Saxton
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158
| | - Jennie Yoo
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158
| | - Deepak Srivastava
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158.,Department of Pediatrics, University of California, San Francisco, CA 94143
| | - Tejal A Desai
- University of California, San Francisco, Schools of Pharmacy and Medicine, Department of Bioengineering and Therapeutic Sciences, San Francisco, CA 94158
| | - Po-Lin So
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158
| | - Kevin E Healy
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, California 94720, USA.,Department of Materials Science and Engineering, University of California at Berkeley, Berkeley, California 94720, USA
| | - Bruce R Conklin
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158.,Departments of Medicine, and Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158
| |
Collapse
|
352
|
Human induced pluripotent stem cell-derived cardiomyocytes recapitulate the predilection of breast cancer patients to doxorubicin-induced cardiotoxicity. Nat Med 2016; 22:547-56. [PMID: 27089514 DOI: 10.1038/nm.4087] [Citation(s) in RCA: 549] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 03/18/2016] [Indexed: 12/14/2022]
Abstract
Doxorubicin is an anthracycline chemotherapy agent effective in treating a wide range of malignancies, but it causes a dose-related cardiotoxicity that can lead to heart failure in a subset of patients. At present, it is not possible to predict which patients will be affected by doxorubicin-induced cardiotoxicity (DIC). Here we demonstrate that patient-specific human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) can recapitulate the predilection to DIC of individual patients at the cellular level. hiPSC-CMs derived from individuals with breast cancer who experienced DIC were consistently more sensitive to doxorubicin toxicity than hiPSC-CMs from patients who did not experience DIC, with decreased cell viability, impaired mitochondrial and metabolic function, impaired calcium handling, decreased antioxidant pathway activity, and increased reactive oxygen species production. Taken together, our data indicate that hiPSC-CMs are a suitable platform to identify and characterize the genetic basis and molecular mechanisms of DIC.
Collapse
|
353
|
Chen IY, Matsa E, Wu JC. Induced pluripotent stem cells: at the heart of cardiovascular precision medicine. Nat Rev Cardiol 2016; 13:333-49. [PMID: 27009425 DOI: 10.1038/nrcardio.2016.36] [Citation(s) in RCA: 133] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The advent of human induced pluripotent stem cell (hiPSC) technology has revitalized the efforts in the past decade to realize more fully the potential of human embryonic stem cells for scientific research. Adding to the possibility of generating an unlimited amount of any cell type of interest, hiPSC technology now enables the derivation of cells with patient-specific phenotypes. Given the introduction and implementation of the large-scale Precision Medicine Initiative, hiPSC technology will undoubtedly have a vital role in the advancement of cardiovascular research and medicine. In this Review, we summarize the progress that has been made in the field of hiPSC technology, with particular emphasis on cardiovascular disease modelling and drug development. The growing roles of hiPSC technology in the practice of precision medicine will also be discussed.
Collapse
Affiliation(s)
- Ian Y Chen
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California 94305, USA.,Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Elena Matsa
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Joseph C Wu
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California 94305, USA.,Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, USA.,Department of Radiology, Stanford University School of Medicine, Stanford, California 94305, USA
| |
Collapse
|
354
|
Gowran A, Rasponi M, Visone R, Nigro P, Perrucci GL, Righetti S, Zanobini M, Pompilio G. Young at Heart: Pioneering Approaches to Model Nonischaemic Cardiomyopathy with Induced Pluripotent Stem Cells. Stem Cells Int 2016; 2016:4287158. [PMID: 27110250 PMCID: PMC4823509 DOI: 10.1155/2016/4287158] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 02/09/2016] [Indexed: 01/01/2023] Open
Abstract
A mere 9 years have passed since the revolutionary report describing the derivation of induced pluripotent stem cells from human fibroblasts and the first in-patient translational use of cells obtained from these stem cells has already been achieved. From the perspectives of clinicians and researchers alike, the promise of induced pluripotent stem cells is alluring if somewhat beguiling. It is now evident that this technology is nascent and many areas for refinement have been identified and need to be considered before induced pluripotent stem cells can be routinely used to stratify, treat and cure patients, and to faithfully model diseases for drug screening purposes. This review specifically addresses the pioneering approaches to improve induced pluripotent stem cell based models of nonischaemic cardiomyopathy.
Collapse
Affiliation(s)
- Aoife Gowran
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino-IRCCS, Via Parea 4, 20138 Milan, Italy
| | - Marco Rasponi
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Piazza Leonardo da Vinci 32, Building No. 21, 20133 Milan, Italy
| | - Roberta Visone
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Piazza Leonardo da Vinci 32, Building No. 21, 20133 Milan, Italy
| | - Patrizia Nigro
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino-IRCCS, Via Parea 4, 20138 Milan, Italy
| | - Gianluca L. Perrucci
- Department of Clinical Sciences and Community Health, University of Milan, Via Festa del Perdono 7, 20122 Milan, Italy
| | - Stefano Righetti
- Cardiology Unit, San Gerardo Hospital, Via Giambattista Pergolesi 33, 20052 Monza, Italy
| | - Marco Zanobini
- Department of Cardiac Surgery, Centro Cardiologico Monzino-IRCCS, Via Parea 4, 20138 Milan, Italy
| | - Giulio Pompilio
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino-IRCCS, Via Parea 4, 20138 Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Via Festa del Perdono 7, 20122 Milan, Italy
- Department of Cardiac Surgery, Centro Cardiologico Monzino-IRCCS, Via Parea 4, 20138 Milan, Italy
| |
Collapse
|
355
|
Del Álamo JC, Lemons D, Serrano R, Savchenko A, Cerignoli F, Bodmer R, Mercola M. High throughput physiological screening of iPSC-derived cardiomyocytes for drug development. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:1717-27. [PMID: 26952934 DOI: 10.1016/j.bbamcr.2016.03.003] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 03/01/2016] [Accepted: 03/03/2016] [Indexed: 12/25/2022]
Abstract
Cardiac drug discovery is hampered by the reliance on non-human animal and cellular models with inadequate throughput and physiological fidelity to accurately identify new targets and test novel therapeutic strategies. Similarly, adverse drug effects on the heart are challenging to model, contributing to costly failure of drugs during development and even after market launch. Human induced pluripotent stem cell derived cardiac tissue represents a potentially powerful means to model aspects of heart physiology relevant to disease and adverse drug effects, providing both the human context and throughput needed to improve the efficiency of drug development. Here we review emerging technologies for high throughput measurements of cardiomyocyte physiology, and comment on the promises and challenges of using iPSC-derived cardiomyocytes to model disease and introduce the human context into early stages of drug discovery. This article is part of a Special Issue entitled: Cardiomyocyte biology: Integration of Developmental and Environmental Cues in the Heart edited by Marcus Schaub and Hughes Abriel.
Collapse
Affiliation(s)
- Juan C Del Álamo
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, 9500 Gilman Drive MC 0411, La Jolla, CA 92093-0411, USA
| | - Derek Lemons
- Department of Bioengineering, University of California, San Diego, 9500 Gilman Drive MC 0412, La Jolla, CA 92093-0412, USA; Sanford-Burnham-Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, CA 92037, USA
| | - Ricardo Serrano
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, 9500 Gilman Drive MC 0411, La Jolla, CA 92093-0411, USA
| | - Alex Savchenko
- Department of Bioengineering, University of California, San Diego, 9500 Gilman Drive MC 0412, La Jolla, CA 92093-0412, USA; Sanford-Burnham-Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, CA 92037, USA; Stanford Cardiovascular Institute, 265 Campus Dr., Stanford, CA 94305-5454, USA
| | - Fabio Cerignoli
- ACEA Biosciences, Inc., 6779 Mesa Ridge Road, San Diego, CA 92121, USA
| | - Rolf Bodmer
- Sanford-Burnham-Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, CA 92037, USA
| | - Mark Mercola
- Department of Bioengineering, University of California, San Diego, 9500 Gilman Drive MC 0412, La Jolla, CA 92093-0412, USA; Sanford-Burnham-Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, CA 92037, USA; Stanford Cardiovascular Institute, 265 Campus Dr., Stanford, CA 94305-5454, USA.
| |
Collapse
|
356
|
Marsano A, Conficconi C, Lemme M, Occhetta P, Gaudiello E, Votta E, Cerino G, Redaelli A, Rasponi M. Beating heart on a chip: a novel microfluidic platform to generate functional 3D cardiac microtissues. LAB ON A CHIP 2016; 16:599-610. [PMID: 26758922 DOI: 10.1039/c5lc01356a] [Citation(s) in RCA: 254] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
In the past few years, microfluidic-based technology has developed microscale models recapitulating key physical and biological cues typical of the native myocardium. However, the application of controlled physiological uniaxial cyclic strains on a defined three-dimension cellular environment is not yet possible. Two-dimension mechanical stimulation was particularly investigated, neglecting the complex three-dimensional cell-cell and cell-matrix interactions. For this purpose, we developed a heart-on-a-chip platform, which recapitulates the physiologic mechanical environment experienced by cells in the native myocardium. The device includes an array of hanging posts to confine cell-laden gels, and a pneumatic actuation system to induce homogeneous uniaxial cyclic strains to the 3D cell constructs during culture. The device was used to generate mature and highly functional micro-engineered cardiac tissues (μECTs), from both neonatal rat and human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM), strongly suggesting the robustness of our engineered cardiac micro-niche. Our results demonstrated that the cyclic strain was effectively highly uniaxial and uniformly transferred to cells in culture. As compared to control, stimulated μECTs showed superior cardiac differentiation, as well as electrical and mechanical coupling, owing to a remarkable increase in junction complexes. Mechanical stimulation also promoted early spontaneous synchronous beating and better contractile capability in response to electric pacing. Pacing analyses of hiPSC-CM constructs upon controlled administration of isoprenaline showed further promising applications of our platform in drug discovery, delivery and toxicology fields. The proposed heart-on-a-chip device represents a relevant step forward in the field, providing a standard functional three-dimensional cardiac model to possibly predict signs of hypertrophic changes in cardiac phenotype by mechanical and biochemical co-stimulation.
Collapse
Affiliation(s)
- Anna Marsano
- Departments of Surgery and Biomedicine, University Basel, University Hospital Basel, Hebelstrasse 20, 4031 Basel, Switzerland.
| | - Chiara Conficconi
- Departments of Surgery and Biomedicine, University Basel, University Hospital Basel, Hebelstrasse 20, 4031 Basel, Switzerland. and Department of Electronics, Information and Bioengineering, Politecnico di Milano, Piazza Leonardo da Vinci 32, Building #21, 20133 Milano, Italy.
| | - Marta Lemme
- Departments of Surgery and Biomedicine, University Basel, University Hospital Basel, Hebelstrasse 20, 4031 Basel, Switzerland. and Department of Electronics, Information and Bioengineering, Politecnico di Milano, Piazza Leonardo da Vinci 32, Building #21, 20133 Milano, Italy.
| | - Paola Occhetta
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Piazza Leonardo da Vinci 32, Building #21, 20133 Milano, Italy.
| | - Emanuele Gaudiello
- Departments of Surgery and Biomedicine, University Basel, University Hospital Basel, Hebelstrasse 20, 4031 Basel, Switzerland.
| | - Emiliano Votta
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Piazza Leonardo da Vinci 32, Building #21, 20133 Milano, Italy.
| | - Giulia Cerino
- Departments of Surgery and Biomedicine, University Basel, University Hospital Basel, Hebelstrasse 20, 4031 Basel, Switzerland.
| | - Alberto Redaelli
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Piazza Leonardo da Vinci 32, Building #21, 20133 Milano, Italy.
| | - Marco Rasponi
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Piazza Leonardo da Vinci 32, Building #21, 20133 Milano, Italy.
| |
Collapse
|
357
|
Csöbönyeiová M, Polák Š, Danišovič L. Toxicity testing and drug screening using iPSC-derived hepatocytes, cardiomyocytes, and neural cells. Can J Physiol Pharmacol 2016; 94:687-94. [PMID: 27128322 DOI: 10.1139/cjpp-2015-0459] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Unexpected toxicity in areas such as cardiotoxicity, hepatotoxicity, and neurotoxicity is a serious complication of clinical therapy and one of the key causes for failure of promising drug candidates in development. Animal studies have been widely used for toxicology research to provide preclinical security evaluation of various therapeutic agents under development. Species differences in drug penetration of the blood-brain barrier, drug metabolism, and related toxicity contribute to failure of drug trials from animal models to human. The existing system for drug discovery has relied on immortalized cell lines, animal models of human disease, and clinical trials in humans. Moreover, drug candidates that are passed as being safe in the preclinical stage often show toxic effects during the clinical stage. Only around 16% drugs are approved for human use. Research on induced pluripotent stem cells (iPSCs) promises to enhance drug discovery and development by providing simple, reproducible, and economically effective tools for drug toxicity screening under development and, on the other hand, for studying the disease mechanism and pathways. In this review, we provide an overview of basic information about iPSCs, and discuss efforts aimed at the use of iPSC-derived hepatocytes, cardiomyocytes, and neural cells in drug discovery and toxicity testing.
Collapse
Affiliation(s)
- Mária Csöbönyeiová
- a Institute of Histology and Embryology, Faculty of Medicine, Comenius University in Bratislava, Sasinkova 4, 811 08 Bratislava, Slovak Republic
| | - Štefan Polák
- a Institute of Histology and Embryology, Faculty of Medicine, Comenius University in Bratislava, Sasinkova 4, 811 08 Bratislava, Slovak Republic
| | - L'uboš Danišovič
- b Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University in Bratislava, Sasinkova 4, 811 08 Bratislava, Slovak Republic
| |
Collapse
|
358
|
Rodgers K, Papinska A, Mordwinkin N. Regulatory aspects of small molecule drugs for heart regeneration. Adv Drug Deliv Rev 2016; 96:245-52. [PMID: 26150343 DOI: 10.1016/j.addr.2015.06.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 06/05/2015] [Accepted: 06/30/2015] [Indexed: 01/14/2023]
Abstract
Even though recent discoveries prove the existence of cardiac progenitor cells, internal regenerative capacity of the heart is minimal. As cardiovascular disease is the leading cause of deaths in the United States, a number of approaches are being used to develop treatments for heart repair and regeneration. Small molecule drugs are of particular interest as they are suited for oral administration and can be chemically synthesized. However, the regulatory process for the development of new treatment modalities is protracted, complex and expensive. One of the hurdles to development of appropriate therapies is the need for predictive preclinical models. The use of patient-derived cardiomyocytes from iPSC cells represents a novel tool for this purpose. Among other concepts for induction of heart regeneration, the most advanced is the combination of DPP-IV inhibitors with stem cell mobilizers. This review will focus on regulatory aspects as well as preclinical hurdles of development of new treatments for heart regeneration.
Collapse
Affiliation(s)
- Kathleen Rodgers
- Titus Family Department of Clinical Pharmacy and Pharmacoeconomics and Policy, School of Pharmacy, University of Southern California, 1985 Zonal Avenue, Los Angeles, CA 90089, United States.
| | - Anna Papinska
- Titus Family Department of Clinical Pharmacy and Pharmacoeconomics and Policy, School of Pharmacy, University of Southern California, 1985 Zonal Avenue, Los Angeles, CA 90089, United States
| | - Nicholas Mordwinkin
- Miltenyi Biotec, Inc., 2303 Lindbergh Street, Auburn, CA 95602, United States
| |
Collapse
|
359
|
Stoppel WL, Kaplan DL, Black LD. Electrical and mechanical stimulation of cardiac cells and tissue constructs. Adv Drug Deliv Rev 2016; 96:135-55. [PMID: 26232525 DOI: 10.1016/j.addr.2015.07.009] [Citation(s) in RCA: 169] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 07/16/2015] [Accepted: 07/25/2015] [Indexed: 12/19/2022]
Abstract
The field of cardiac tissue engineering has made significant strides over the last few decades, highlighted by the development of human cell derived constructs that have shown increasing functional maturity over time, particularly using bioreactor systems to stimulate the constructs. However, the functionality of these tissues is still unable to match that of native cardiac tissue and many of the stem-cell derived cardiomyocytes display an immature, fetal like phenotype. In this review, we seek to elucidate the biological underpinnings of both mechanical and electrical signaling, as identified via studies related to cardiac development and those related to an evaluation of cardiac disease progression. Next, we review the different types of bioreactors developed to individually deliver electrical and mechanical stimulation to cardiomyocytes in vitro in both two and three-dimensional tissue platforms. Reactors and culture conditions that promote functional cardiomyogenesis in vitro are also highlighted. We then cover the more recent work in the development of bioreactors that combine electrical and mechanical stimulation in order to mimic the complex signaling environment present in vivo. We conclude by offering our impressions on the important next steps for physiologically relevant mechanical and electrical stimulation of cardiac cells and engineered tissue in vitro.
Collapse
|
360
|
Tzatzalos E, Abilez OJ, Shukla P, Wu JC. Engineered heart tissues and induced pluripotent stem cells: Macro- and microstructures for disease modeling, drug screening, and translational studies. Adv Drug Deliv Rev 2016; 96:234-244. [PMID: 26428619 DOI: 10.1016/j.addr.2015.09.010] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 09/16/2015] [Accepted: 09/23/2015] [Indexed: 01/01/2023]
Abstract
Engineered heart tissue has emerged as a personalized platform for drug screening. With the advent of induced pluripotent stem cell (iPSC) technology, patient-specific stem cells can be developed and expanded into an indefinite source of cells. Subsequent developments in cardiovascular biology have led to efficient differentiation of cardiomyocytes, the force-producing cells of the heart. iPSC-derived cardiomyocytes (iPSC-CMs) have provided potentially limitless quantities of well-characterized, healthy, and disease-specific CMs, which in turn has enabled and driven the generation and scale-up of human physiological and disease-relevant engineered heart tissues. The combined technologies of engineered heart tissue and iPSC-CMs are being used to study diseases and to test drugs, and in the process, have advanced the field of cardiovascular tissue engineering into the field of precision medicine. In this review, we will discuss current developments in engineered heart tissue, including iPSC-CMs as a novel cell source. We examine new research directions that have improved the function of engineered heart tissue by using mechanical or electrical conditioning or the incorporation of non-cardiomyocyte stromal cells. Finally, we discuss how engineered heart tissue can evolve into a powerful tool for therapeutic drug testing.
Collapse
Affiliation(s)
- Evangeline Tzatzalos
- Stanford Cardiovascular Institute
- Institute for Stem Cell Biology and Regenerative Medicine
| | - Oscar J Abilez
- Stanford Cardiovascular Institute
- Institute for Stem Cell Biology and Regenerative Medicine
- Bio-X Program
- Department of Medicine, Division of Cardiovascular Medicine
| | - Praveen Shukla
- Stanford Cardiovascular Institute
- Institute for Stem Cell Biology and Regenerative Medicine
| | - Joseph C Wu
- Stanford Cardiovascular Institute
- Institute for Stem Cell Biology and Regenerative Medicine
- Bio-X Program
- Department of Medicine, Division of Cardiovascular Medicine
| |
Collapse
|
361
|
Kurokawa YK, George SC. Tissue engineering the cardiac microenvironment: Multicellular microphysiological systems for drug screening. Adv Drug Deliv Rev 2016; 96:225-33. [PMID: 26212156 PMCID: PMC4869857 DOI: 10.1016/j.addr.2015.07.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 07/07/2015] [Accepted: 07/17/2015] [Indexed: 12/29/2022]
Abstract
The ability to accurately detect cardiotoxicity has become increasingly important in the development of new drugs. Since the advent of human pluripotent stem cell-derived cardiomyocytes, researchers have explored their use in creating an in vitro drug screening platform. Recently, there has been increasing interest in creating 3D microphysiological models of the heart as a tool to detect cardiotoxic compounds. By recapitulating the complex microenvironment that exists in the native heart, cardiac microphysiological systems have the potential to provide a more accurate pharmacological response compared to current standards in preclinical drug screening. This review aims to provide an overview on the progress made in creating advanced models of the human heart, including the significance and contributions of the various cellular and extracellular components to cardiac function.
Collapse
Affiliation(s)
- Yosuke K Kurokawa
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA.
| | - Steven C George
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA; Department of Energy, Environment, and Chemical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA.
| |
Collapse
|
362
|
In vitro cardiac tissue models: Current status and future prospects. Adv Drug Deliv Rev 2016; 96:203-13. [PMID: 26428618 DOI: 10.1016/j.addr.2015.09.011] [Citation(s) in RCA: 123] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 09/14/2015] [Accepted: 09/21/2015] [Indexed: 01/15/2023]
Abstract
Cardiovascular disease is the leading cause of death worldwide. Achieving the next phase of potential treatment strategies and better prognostic tools will require a concerted effort from interdisciplinary fields. Biomaterials-based cardiac tissue models are revolutionizing the area of preclinical research and translational applications. The goal of in vitro cardiac tissue modeling is to create physiological functional models of the human myocardium, which is a difficult task due to the complex structure and function of the human heart. This review describes the advances made in area of in vitro cardiac models using biomaterials and bioinspired platforms. The field has progressed extensively in the past decade, and we envision its applications in the areas of drug screening, disease modeling, and precision medicine.
Collapse
|
363
|
Aung A, Bhullar IS, Theprungsirikul J, Davey SK, Lim HL, Chiu YJ, Ma X, Dewan S, Lo YH, McCulloch A, Varghese S. 3D cardiac μtissues within a microfluidic device with real-time contractile stress readout. LAB ON A CHIP 2016; 16:153-62. [PMID: 26588203 PMCID: PMC4681661 DOI: 10.1039/c5lc00820d] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
We present the development of three-dimensional (3D) cardiac microtissues within a microfluidic device with the ability to quantify real-time contractile stress measurements in situ. Using a 3D patterning technology that allows for the precise spatial distribution of cells within the device, we created an array of 3D cardiac microtissues from neonatal mouse cardiomyocytes. We integrated the 3D micropatterning technology with microfluidics to achieve perfused cell-laden structures. The cells were encapsulated within a degradable gelatin methacrylate hydrogel, which was sandwiched between two polyacrylamide hydrogels. The polyacrylamide hydrogels were used as "stress sensors" to acquire the contractile stresses generated by the beating cardiac cells. The cardiac-specific response of the engineered 3D system was examined by exposing it to epinephrine, an adrenergic neurotransmitter known to increase the magnitude and frequency of cardiac contractions. In response to exogenous epinephrine the engineered cardiac tissues exhibited an increased beating frequency and stress magnitude. Such cost-effective and easy-to-adapt 3D cardiac systems with real-time functional readout could be an attractive technological platform for drug discovery and development.
Collapse
Affiliation(s)
- Aereas Aung
- Department of Bioengineering, University of California-San Diego, La Jolla, CA, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
364
|
Wang L, Zhang X, Xu C, Liu H, Qin J. Human induced pluripotent stem cell-derived cardiac tissue on a thin collagen membrane with natural microstructures. Biomater Sci 2016; 4:1655-1662. [DOI: 10.1039/c6bm00522e] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
We present a new strategy to produce a thin collagen membrane from porcine tendons and engineered cardiac tissues using hiPSC-derived cardiomyocytes.
Collapse
Affiliation(s)
- Li Wang
- Division of Biotechnology
- Dalian Institute of Chemical Physics
- Chinese Academy of Sciences
- Dalian 116023
- PR China
| | - Xiaoqing Zhang
- Division of Biotechnology
- Dalian Institute of Chemical Physics
- Chinese Academy of Sciences
- Dalian 116023
- PR China
| | - Cong Xu
- Division of Biotechnology
- Dalian Institute of Chemical Physics
- Chinese Academy of Sciences
- Dalian 116023
- PR China
| | - Hui Liu
- Division of Biotechnology
- Dalian Institute of Chemical Physics
- Chinese Academy of Sciences
- Dalian 116023
- PR China
| | - Jianhua Qin
- Division of Biotechnology
- Dalian Institute of Chemical Physics
- Chinese Academy of Sciences
- Dalian 116023
- PR China
| |
Collapse
|
365
|
Laurila E, Ahola A, Hyttinen J, Aalto-Setälä K. Methods for in vitro functional analysis of iPSC derived cardiomyocytes - Special focus on analyzing the mechanical beating behavior. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1863:1864-72. [PMID: 26707468 DOI: 10.1016/j.bbamcr.2015.12.013] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2015] [Revised: 12/09/2015] [Accepted: 12/16/2015] [Indexed: 02/06/2023]
Abstract
A rapidly increasing number of papers describing novel iPSC models for cardiac diseases are being published. To be able to understand the disease mechanisms in more detail, we should also take the full advantage of the various methods for analyzing these cell models. The traditionally and commonly used electrophysiological analysis methods have been recently accompanied by novel approaches for analyzing the mechanical beatingbehavior of the cardiomyocytes. In this review, we provide first a concise overview on the methodology for cardiomyocyte functional analysis and then concentrate on the video microscopy, which provides a promise for a new faster yet reliable method for cardiomyocyte functional analysis. We also show how analysis conditions may affect the results. Development of the methodology not only serves the basic research on the disease models, but could also provide the much needed efficient early phase screening method for cardiac safety toxicology. This article is part of a Special Issue entitled: Cardiomyocyte Biology: Integration of Developmental and Environmental Cues in the Heart edited by Marcus Schaub and Hughes Abriel.
Collapse
Affiliation(s)
- Eeva Laurila
- University of Tampere, BioMediTech and School of Medicine, Tampere, Finland.
| | - Antti Ahola
- Tampere University of Technology, Department of Electronics and Communications Engineering, BioMediTech, Tampere, Finland
| | - Jari Hyttinen
- Tampere University of Technology, Department of Electronics and Communications Engineering, BioMediTech, Tampere, Finland
| | - Katriina Aalto-Setälä
- University of Tampere, BioMediTech and School of Medicine, Tampere, Finland; Heart Hospital, Tampere University Hospital, Tampere, Finland
| |
Collapse
|
366
|
Rismani Yazdi S, Shadmani A, Bürgel SC, Misun PM, Hierlemann A, Frey O. Adding the 'heart' to hanging drop networks for microphysiological multi-tissue experiments. LAB ON A CHIP 2015; 15:4138-47. [PMID: 26401602 PMCID: PMC5424877 DOI: 10.1039/c5lc01000d] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Microfluidic hanging-drop networks enable culturing and analysis of 3D microtissue spheroids derived from different cell types under controlled perfusion and investigating inter-tissue communication in multi-tissue formats. In this paper we introduce a compact on-chip pumping approach for flow control in hanging-drop networks. The pump includes one pneumatic chamber located directly above one of the hanging drops and uses the surface tension at the liquid-air-interface for flow actuation. Control of the pneumatic protocol provides a wide range of unidirectional pulsatile and continuous flow profiles. With the proposed concept several independent hanging-drop networks can be operated in parallel with only one single pneumatic actuation line at high fidelity. Closed-loop medium circulation between different organ models for multi-tissue formats and multiple simultaneous assays in parallel are possible. Finally, we implemented a real-time feedback control-loop of the pump actuation based on the beating of a human iPS-derived cardiac microtissue cultured in the same system. This configuration allows for simulating physiological effects on the heart and their impact on flow circulation between the organ models on chip.
Collapse
Affiliation(s)
- Saeed Rismani Yazdi
- ETH Zurich, Department of Biosystems Science and Engineering, Bio Engineering Laboratory, Mattenstrasse 26, CH-4058 Basel, Switzerland.
| | | | | | | | | | | |
Collapse
|
367
|
Loskill P, Marcus SG, Mathur A, Reese WM, Healy KE. μOrgano: A Lego®-Like Plug & Play System for Modular Multi-Organ-Chips. PLoS One 2015; 10:e0139587. [PMID: 26440672 PMCID: PMC4595286 DOI: 10.1371/journal.pone.0139587] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 09/15/2015] [Indexed: 11/19/2022] Open
Abstract
Human organ-on-a-chip systems for drug screening have evolved as feasible alternatives to animal models, which are unreliable, expensive, and at times erroneous. While chips featuring single organs can be of great use for both pharmaceutical testing and basic organ-level studies, the huge potential of the organ-on-a-chip technology is revealed by connecting multiple organs on one chip to create a single integrated system for sophisticated fundamental biological studies and devising therapies for disease. Furthermore, since most organ-on-a-chip systems require special protocols with organ-specific media for the differentiation and maturation of the tissues, multi-organ systems will need to be temporally customizable and flexible in terms of the time point of connection of the individual organ units. We present a customizable Lego®-like plug & play system, μOrgano, which enables initial individual culture of single organ-on-a-chip systems and subsequent connection to create integrated multi-organ microphysiological systems. As a proof of concept, the μOrgano system was used to connect multiple heart chips in series with excellent cell viability and spontaneously physiological beat rates.
Collapse
Affiliation(s)
- Peter Loskill
- Department of Bioengineering, University of California at Berkeley, Berkeley, California, United States of America
- Department of Materials Science and Engineering, University of California at Berkeley, Berkeley, California, United States of America
- California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, California, United States of America
| | - Sivan G. Marcus
- Department of Bioengineering, University of California at Berkeley, Berkeley, California, United States of America
| | - Anurag Mathur
- Department of Bioengineering, University of California at Berkeley, Berkeley, California, United States of America
- Department of Materials Science and Engineering, University of California at Berkeley, Berkeley, California, United States of America
- California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, California, United States of America
| | - Willie Mae Reese
- Department of Materials Science and Engineering, University of California at Berkeley, Berkeley, California, United States of America
| | - Kevin E. Healy
- Department of Bioengineering, University of California at Berkeley, Berkeley, California, United States of America
- Department of Materials Science and Engineering, University of California at Berkeley, Berkeley, California, United States of America
- California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, California, United States of America
| |
Collapse
|
368
|
Tang S, Xie M, Cao N, Ding S. Patient-Specific Induced Pluripotent Stem Cells for Disease Modeling and Phenotypic Drug Discovery. J Med Chem 2015; 59:2-15. [PMID: 26322868 DOI: 10.1021/acs.jmedchem.5b00789] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
In vitro cell models are invaluable tools for studying diseases and discovering drugs. Human induced pluripotent stem cells, particularly derived from patients, are an advantageous resource for generating ample supplies of cells to create unique platforms that model disease. This manuscript will review recent developments in modeling a variety of diseases (including their cellular phenotypes) with induced pluripotent stem cells derived from patients. It will also describe how researchers have exploited these models to validate drugs as potential therapeutics for these devastating diseases.
Collapse
Affiliation(s)
- Shibing Tang
- Gladstone Institutes , 1650 Owens Street, San Francisco, California 94158, United States
| | - Min Xie
- Gladstone Institutes , 1650 Owens Street, San Francisco, California 94158, United States
| | - Nan Cao
- Gladstone Institutes , 1650 Owens Street, San Francisco, California 94158, United States
| | - Sheng Ding
- Gladstone Institutes , 1650 Owens Street, San Francisco, California 94158, United States
| |
Collapse
|
369
|
Cavero I, Holzgrefe H. CiPA: Ongoing testing, future qualification procedures, and pending issues. J Pharmacol Toxicol Methods 2015; 76:27-37. [PMID: 26159293 DOI: 10.1016/j.vascn.2015.06.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2015] [Revised: 06/04/2015] [Accepted: 06/25/2015] [Indexed: 01/04/2023]
Abstract
INTRODUCTION The comprehensive in vitro proarrhythmia assay (CiPA) is a nonclinical, mechanism-based paradigm for assessing drug proarrhythmic liability. TOPICS COVERED The first CiPA assay determines effects on cloned human cardiac ion channels. The second investigates whether the latter study-generated metrics engender proarrhythmic markers on a computationally reconstructed human ventricular action potential. The third evaluates conclusions from, and searches possibly missed effects by in silico analysis, in human stem cell-derived cardiomyocytes (hSC-CMs). CiPA ad hoc Expert-Working Groups have proposed patch clamp protocols for seven cardiac ion channels, a modified O'Hara-Rudy model for in silico analysis, detailed procedures for field (MEA) and action potential (VSD) measurements in hSC-CMs, and 29 reference drugs for CiPA assay testing and validation. DISCUSSION CiPA adoption as drug development tool for identifying electrophysiological mechanisms conferring proarrhythmic liability to candidate drugs is a complex, multi-functional task requiring significant time, reflection, and efforts to be fully achieved.
Collapse
|
370
|
Monie DD, Bhatia SK. Bioprinting of Dynamic Human Organs-on-Chips: Enabling Technologies for Rapid Drug Development and Personalized Medicine. BIOPRINTING IN REGENERATIVE MEDICINE 2015. [DOI: 10.1007/978-3-319-21386-6_6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
371
|
Warren CR, Cowan CA. [Leukocyte count of puerperal sows]. BERLINER UND MUNCHENER TIERARZTLICHE WOCHENSCHRIFT 1996; 109:330-5. [PMID: 9054332 PMCID: PMC5828525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
147 blood samples of postparturient sows of a secluded housing were taken. The samples were conserved with ACD-solution. The influence of the number and week of the lactation and the health of the sow, determined by puerperal diseases was studied. Hematological values of healthy postparturient sows are: leucocytes 12.6 +/- 2.2 G/l; basophile granulocytes 0.1 +/- 0.1 G/l, eosinophile granulocytes 0.5 +/- 0.4 G/l; banded neutrophile granulocytes 1.3 +/- 0.6 G/l, segmented neutrophile granulocytes 5.2 +/- 1.4 G/l; lymphocytes 5.5 +/- 1.4 G/l, monocytes 0.3 +/- 0.3 G/l. The leucocyte number is lower in the investigated herd compared with quotations in the literature. This is based on the good health conditions in the herd. Changes due to the number and week of the lactation have no clinical relevance. Health status, here described by puerperal diseases is the significant influencing factor of the leucocyte number. The severity of puerperal diseases is significant. Due to puerperal diseases the leucocyte number rises quickly after a short drop about 2 G/l. The number of the neutrophile granulocytes increases, but the lymphocyte number is reduced at the beginning of the illness. The application of ACD-solution for stabilizing of great amounts of blood samples under practical conditions is demonstrated. It is possible to stabilize pigs blood well.
Collapse
Affiliation(s)
- Curtis R. Warren
- Division of Cardiovascular Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
| | - Chad A. Cowan
- Division of Cardiovascular Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
- Broad Institute, Cambridge, Massachusetts 02142, USA
| |
Collapse
|