51
|
Faruqu FN, Zhou S, Sami N, Gheidari F, Lu H, Al‐Jamal KT. Three-dimensional culture of dental pulp pluripotent-like stem cells (DPPSCs) enhances Nanog expression and provides a serum-free condition for exosome isolation. FASEB Bioadv 2020; 2:419-433. [PMID: 32676582 PMCID: PMC7354694 DOI: 10.1096/fba.2020-00025] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/18/2020] [Accepted: 05/26/2020] [Indexed: 12/18/2022] Open
Abstract
Stem cell-derived exosomes have been identified as novel cell-free therapeutics for regenerative medicine. Three-dimensional (3D) culture of stem cells were reported to improve their "stemness" and therapeutic efficacy. This work focused on establishing serum-free 3D culture of dental pulp pluripotent-like stem cells (DPPSCs)-a newly characterized pluripotent-like stem cell for exosome production. DPPSCs were expanded in regular 2D culture in human serum-supplemented (HS)-medium and transferred to a micropatterned culture plate for 3D culture in HS-medium (default) and medium supplemented with KnockOut™ serum replacement (KO-medium). Bright-field microscopy observation throughout the culture period (24 days) revealed that DPPSCs in KO-medium formed spheroids of similar morphology and size to that in HS-medium. qRT-PCR analysis showed similar Oct4A gene expression in DPPSC spheroids in both HS-medium and KO-medium, but Nanog expression significantly increased in the latter. Vesicles isolated from DPPSC spheroids in KO-medium in the first 12 days of culture showed sizes that fall within the exosomal size range by nanoparticle tracking analysis (NTA) and express the canonical exosomal markers. It is concluded that 3D culture of DPPSCs in KO-medium provided an optimal serum-free condition for successful isolation of DPPSC-derived exosomes for subsequent applications in regenerative medicine.
Collapse
Affiliation(s)
- Farid N. Faruqu
- Institute of Pharmaceutical ScienceKing’s College LondonLondonUK
| | - Shuai Zhou
- Institute of Pharmaceutical ScienceKing’s College LondonLondonUK
| | - Noor Sami
- Institute of Pharmaceutical ScienceKing’s College LondonLondonUK
| | - Fatemeh Gheidari
- Institute of Pharmaceutical ScienceKing’s College LondonLondonUK
| | - Han Lu
- Genomics CentreKing’s College LondonLondonUK
| | | |
Collapse
|
52
|
Aronsson C, Jury M, Naeimipour S, Boroojeni FR, Christoffersson J, Lifwergren P, Mandenius CF, Selegård R, Aili D. Dynamic peptide-folding mediated biofunctionalization and modulation of hydrogels for 4D bioprinting. Biofabrication 2020; 12:035031. [DOI: 10.1088/1758-5090/ab9490] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
53
|
Requirements for Proper Immunosuppressive Regimens to Limit Translational Failure of Cardiac Cell Therapy in Preclinical Large Animal Models. J Cardiovasc Transl Res 2020; 14:88-99. [PMID: 32476086 PMCID: PMC7892682 DOI: 10.1007/s12265-020-10035-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 05/19/2020] [Indexed: 12/16/2022]
Abstract
Various cell-based therapies are currently investigated in an attempt to tackle the high morbidity and mortality associated with heart failure. The need for these therapies to move towards the clinic is pressing. Therefore, preclinical large animal studies that use non-autologous cells are needed to evaluate their potential. However, non-autologous cells are highly immunogenic and trigger immune rejection responses resulting in potential loss of efficacy. To overcome this issue, adequate immunosuppressive regimens are of imminent importance but clear guidelines are currently lacking. In this review, we assess the immunological barriers regarding non-autologous cell transplantation and immune modulation with immunosuppressive drugs. In addition, we provide recommendations with respect to immunosuppressive regimens in preclinical cardiac cell-replacement studies.
Collapse
|
54
|
Miwa T, Idiris A, Kumagai H. A novel cardiac differentiation method of a large number and uniformly-sized spheroids using microfabricated culture vessels. Regen Ther 2020; 15:18-26. [PMID: 32490063 PMCID: PMC7256449 DOI: 10.1016/j.reth.2020.04.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 03/30/2020] [Accepted: 04/14/2020] [Indexed: 12/30/2022] Open
Abstract
Cardiomyocytes differentiated from human induced pluripotent stem cells (hiPSCs) have great potential for regenerative medicine and drug discovery. In this study, we developed a novel protocol to more reproducibly and efficiently induce cardiomyocytes. A large quantity of uniformly sized spheroids were generated from hiPSCs using microfabricated vessels and induced into cardiac differentiation. In the middle of the cardiac differentiation process, spheroids were then dissociated into single cells and reaggregated into smaller spheroids using the microfabricated vessels. This reaggregation process raised WNT5A and WNT11 expression levels and improved the quality of cardiomyocyte population compared to that in a control group in which dissociation and reaggregation were not performed. Microfabricated culture vessels were used to form large number of hiPSC spheroids. High purity cardiomyocytes were obtained by reaggregation of hiPSC-derived spheroids. Maturation of cardiomyocyte was promoted by the reaggregation process. WNTs were also increased the reaggregation process during cardiac differentiation.
Collapse
Affiliation(s)
- Tatsuaki Miwa
- Developing & Planning Division, Technology Development General Division, Electronics Company, AGC Inc., (reside in AGC Techno Glass Co.), 3583-5 Kawashiri Yoshida-cho, Haibara-gun, Shizuoka 421-0302, Japan
| | - Alimjan Idiris
- Biochemistry Team, Bio Science Division, Material Integration Laboratories, Technology General Division, AGC Inc., 1150 Hazawa-cho, Kanagawa-ku, Yokohama-shi, Kanagawa 221-8755, Japan
| | - Hiromichi Kumagai
- AGC Research Institute, Inc., Shin-Marunouchi Bldg. 1-5-1, Marunouchi Chiyoda-ku, Tokyo 100-6530 Japan
| |
Collapse
|
55
|
Ulmer BM, Eschenhagen T. Human pluripotent stem cell-derived cardiomyocytes for studying energy metabolism. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2020; 1867:118471. [PMID: 30954570 PMCID: PMC7042711 DOI: 10.1016/j.bbamcr.2019.04.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 03/26/2019] [Accepted: 04/01/2019] [Indexed: 12/25/2022]
Abstract
Cardiomyocyte energy metabolism is altered in heart failure, and primary defects of metabolic pathways can cause heart failure. Studying cardiac energetics in rodent models has principal shortcomings, raising the question to which extent human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CM) can provide an alternative. As metabolic maturation of CM occurs mostly after birth during developmental hypertrophy, the immaturity of hiPSC-CM is an important limitation. Here we shortly review the physiological drivers of metabolic maturation and concentrate on methods to mature hiPSC-CM with the goal to benchmark the metabolic state of hiPSC-CM against in vivo data and to see how far known abnormalities in inherited metabolic disorders can be modeled in hiPSC-CM. The current data indicate that hiPSC-CM, despite their immature, approximately mid-fetal state of energy metabolism, faithfully recapitulate some basic metabolic disease mechanisms. Efforts to improve their metabolic maturity are underway and shall improve the validity of this model.
Collapse
Affiliation(s)
- Bärbel M Ulmer
- University Medical Center Hamburg-Eppendorf, Institute of Experimental Pharmacology and Toxicology, 20246 Hamburg, Germany; German Centre for Heart Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany.
| | - Thomas Eschenhagen
- University Medical Center Hamburg-Eppendorf, Institute of Experimental Pharmacology and Toxicology, 20246 Hamburg, Germany; German Centre for Heart Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany.
| |
Collapse
|
56
|
Jiang B, Yan L, Shamul JG, Hakun M, He X. Stem cell therapy of myocardial infarction: a promising opportunity in bioengineering. ADVANCED THERAPEUTICS 2020; 3:1900182. [PMID: 33665356 PMCID: PMC7928435 DOI: 10.1002/adtp.201900182] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Indexed: 02/06/2023]
Abstract
Myocardial infarction (MI) is a life-threatening disease resulting from irreversible death of cardiomyocytes (CMs) and weakening of the heart blood-pumping function. Stem cell-based therapies have been studied for MI treatment over the last two decades with promising outcome. In this review, we critically summarize the past work in this field to elucidate the advantages and disadvantages of treating MI using pluripotent stem cells (PSCs) including both embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), adult stem cells, and cardiac progenitor cells. The main advantage of the latter is their cytokine production capability to modulate immune responses and control the progression of healing. However, human adult stem cells have very limited (if not 'no') capacity to differentiate into functional CMs in vitro or in vivo. In contrast, PSCs can be differentiated into functional CMs although the protocols for the cardiac differentiation of PSCs are mainly for adherent cells under 2D culture. Derivation of PSC-CMs in 3D, allowing for large-scale production of CMs via modulation of the Wnt/β-catenin signal pathway with defined chemicals and medium, may be desired for clinical translation. Furthermore, the technology of purification and maturation of the PSC-CMs may need further improvements to eliminate teratoma formation after in vivo implantation of the PSC-CMs for treating MI. In addition, in vitro derived PSC-CMs may have mechanical and electrical mismatch with the patient's cardiac tissue, which causes arrhythmia. This supports the use of PSC-derived cells committed to cardiac lineage without beating for implantation to treat MI. In this case, the PSC derived cells may utilize the mechanical, electrical, and chemical cues in the heart to further differentiate into mature/functional CMs in situ. Another major challenge facing stem cell therapy of MI is the low retention/survival of stem cells or their derivatives (e.g., PSC-CMs) in the heart for MI treatment after injection in vivo. This may be resolved by using biomaterials to engineer stem cells for reduced immunogenicity, immobilization of the cells in the heart, and increased integration with the host cardiac tissue. Biomaterials have also been applied in the derivation of CMs in vitro to increase the efficiency and maturation of differentiation. Collectively, a lot has been learned from the past failure of simply injecting intact stem cells or their derivatives in vivo for treating MI, and bioengineering stem cells with biomaterials is expected to be a valuable strategy for advancing stem cell therapy towards its widespread application for treating MI in the clinic.
Collapse
Affiliation(s)
- Bin Jiang
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| | - Li Yan
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| | - James G Shamul
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| | - Maxwell Hakun
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| | - Xiaoming He
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| |
Collapse
|
57
|
Chemically-Defined, Xeno-Free, Scalable Production of hPSC-Derived Definitive Endoderm Aggregates with Multi-Lineage Differentiation Potential. Cells 2019; 8:cells8121571. [PMID: 31817235 PMCID: PMC6953099 DOI: 10.3390/cells8121571] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 11/29/2019] [Accepted: 12/03/2019] [Indexed: 12/30/2022] Open
Abstract
For the production and bio-banking of differentiated derivatives from human pluripotent stem cells (hPSCs) in large quantities for drug screening and cellular therapies, well-defined and robust procedures for differentiation and cryopreservation are required. Definitive endoderm (DE) gives rise to respiratory and digestive epithelium, as well as thyroid, thymus, liver, and pancreas. Here, we present a scalable, universal process for the generation of DE from human-induced pluripotent stem cells (hiPSCs) and embryonic stem cells (hESCs). Optimal control during the differentiation process was attained in chemically-defined and xeno-free suspension culture, and high flexibility of the workflow was achieved by the introduction of an efficient cryopreservation step at the end of DE differentiation. DE aggregates were capable of differentiating into hepatic-like, pancreatic, intestinal, and lung progenitor cells. Scale-up of the differentiation process using stirred-tank bioreactors enabled production of large quantities of DE aggregates. This process provides a useful advance for versatile applications of DE lineages, in particular for cell therapies and drug screening.
Collapse
|
58
|
Sung TC, Liu CH, Huang WL, Lee YC, Kumar SS, Chang Y, Ling QD, Hsu ST, Higuchi A. Efficient differentiation of human ES and iPS cells into cardiomyocytes on biomaterials under xeno-free conditions. Biomater Sci 2019; 7:5467-5481. [PMID: 31656967 DOI: 10.1039/c9bm00817a] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Current xeno-free and chemically defined methods for the differentiation of hPSCs (human pluripotent stem cells) into cardiomyocytes are not efficient and are sometimes not reproducible. Therefore, it is necessary to develop reliable and efficient methods for the differentiation of hPSCs into cardiomyocytes for future use in cardiovascular research related to drug discovery, cardiotoxicity screening, and disease modeling. We evaluated two representative differentiation methods that were reported previously, and we further developed original, more efficient methods for the differentiation of hPSCs into cardiomyocytes under xeno-free, chemically defined conditions. The developed protocol successively differentiated hPSCs into cardiomyocytes, approximately 90-97% of which expressed the cardiac marker cTnT, with beating speeds and sarcomere lengths that were similar to those of a healthy adult human heart. The optimal cell culture biomaterials for the cardiac differentiation of hPSCs were also evaluated using extracellular matrix-mimetic material-coated dishes. Synthemax II-coated and Laminin-521-coated dishes were found to be the most effective and efficient biomaterials for the cardiac differentiation of hPSCs according to the observation of hPSC-derived cardiomyocytes with high survival ratios, high beating colony numbers, a similar beating frequency to that of a healthy adult human heart, high purity levels (high cTnT expression) and longer sarcomere lengths similar to those of a healthy adult human heart.
Collapse
Affiliation(s)
- Tzu-Cheng Sung
- The Eye Hospital of Wenzhou Medical University, No. 270, Xueyuan Road, Wenzhou, Zhejiang 325027, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
59
|
Hamad S, Derichsweiler D, Papadopoulos S, Nguemo F, Šarić T, Sachinidis A, Brockmeier K, Hescheler J, Boukens BJ, Pfannkuche K. Generation of human induced pluripotent stem cell-derived cardiomyocytes in 2D monolayer and scalable 3D suspension bioreactor cultures with reduced batch-to-batch variations. Theranostics 2019; 9:7222-7238. [PMID: 31695764 PMCID: PMC6831300 DOI: 10.7150/thno.32058] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 07/16/2019] [Indexed: 02/06/2023] Open
Abstract
Human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CMs) are promising candidates to treat myocardial infarction and other cardiac diseases. Such treatments require pure cardiomyocytes (CMs) in large quantities. Methods: In the present study we describe an improved protocol for production of hiPSC-CMs in which hiPSCs are first converted into mesodermal cells by stimulation of wingless (Wnt) signaling using CHIR99021, which are then further differentiated into CM progenitors by simultaneous inhibition of porcupine and tankyrase pathways using IWP2 and XAV939 under continuous supplementation of ascorbate during the entire differentiation procedure. Results: The protocol resulted in reproducible generation of >90% cardiac troponin T (TNNT2)-positive cells containing highly organized sarcomeres. In 2D monolayer cultures CM yields amounted to 0.5 million cells per cm2 growth area, and on average 72 million cells per 100 mL bioreactor suspension culture without continuous perfusion. The differentiation efficiency was hardly affected by the initial seeding density of undifferentiated hiPSCs. Furthermore, batch-to-batch variations were reduced by combinatorial use of ascorbate, IWP2, and XAV939. Conclusion: Combined inhibition of porcupine and tankyrase sub-pathways of Wnt signaling and continuous ascorbate supplementation, enable robust and efficient production of hiPSC-CMs.
Collapse
Affiliation(s)
- Sarkawt Hamad
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, University of Cologne, Medical Faculty, Cologne, Germany
- Biology Department, Faculty of Science, Soran University, Soran, Kurdistan region-Iraq
| | - Daniel Derichsweiler
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, University of Cologne, Medical Faculty, Cologne, Germany
| | - Symeon Papadopoulos
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, University of Cologne, Medical Faculty, Cologne, Germany
- Center for Physiology and Pathophysiology, Institute for Vegetative Physiology, University of Cologne, Medical Faculty, Cologne, Germany
| | - Filomain Nguemo
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, University of Cologne, Medical Faculty, Cologne, Germany
| | - Tomo Šarić
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, University of Cologne, Medical Faculty, Cologne, Germany
| | - Agapios Sachinidis
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, University of Cologne, Medical Faculty, Cologne, Germany
| | - Konrad Brockmeier
- Department of Pediatric Cardiology, University Clinics of Cologne, Cologne, Germany
| | - Jürgen Hescheler
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, University of Cologne, Medical Faculty, Cologne, Germany
| | - Bastiaan J Boukens
- Department of Medical Biology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Kurt Pfannkuche
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, University of Cologne, Medical Faculty, Cologne, Germany
- Department of Pediatric Cardiology, University Clinics of Cologne, Cologne, Germany
| |
Collapse
|
60
|
Skylar-Scott MA, Uzel SGM, Nam LL, Ahrens JH, Truby RL, Damaraju S, Lewis JA. Biomanufacturing of organ-specific tissues with high cellular density and embedded vascular channels. SCIENCE ADVANCES 2019; 5:eaaw2459. [PMID: 31523707 PMCID: PMC6731072 DOI: 10.1126/sciadv.aaw2459] [Citation(s) in RCA: 503] [Impact Index Per Article: 83.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Accepted: 08/02/2019] [Indexed: 05/17/2023]
Abstract
Engineering organ-specific tissues for therapeutic applications is a grand challenge, requiring the fabrication and maintenance of densely cellular constructs composed of ~108 cells/ml. Organ building blocks (OBBs) composed of patient-specific-induced pluripotent stem cell-derived organoids offer a pathway to achieving tissues with the requisite cellular density, microarchitecture, and function. However, to date, scant attention has been devoted to their assembly into 3D tissue constructs. Here, we report a biomanufacturing method for assembling hundreds of thousands of these OBBs into living matrices with high cellular density into which perfusable vascular channels are introduced via embedded three-dimensional bioprinting. The OBB matrices exhibit the desired self-healing, viscoplastic behavior required for sacrificial writing into functional tissue (SWIFT). As an exemplar, we created a perfusable cardiac tissue that fuses and beats synchronously over a 7-day period. Our SWIFT biomanufacturing method enables the rapid assembly of perfusable patient- and organ-specific tissues at therapeutic scales.
Collapse
Affiliation(s)
- Mark A. Skylar-Scott
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02138, USA
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
| | - Sebastien G. M. Uzel
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02138, USA
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
| | - Lucy L. Nam
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02138, USA
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
| | - John H. Ahrens
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02138, USA
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
| | - Ryan L. Truby
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02138, USA
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
| | - Sarita Damaraju
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02138, USA
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
| | - Jennifer A. Lewis
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02138, USA
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
- Corresponding author.
| |
Collapse
|
61
|
Schwedhelm I, Zdzieblo D, Appelt-Menzel A, Berger C, Schmitz T, Schuldt B, Franke A, Müller FJ, Pless O, Schwarz T, Wiedemann P, Walles H, Hansmann J. Automated real-time monitoring of human pluripotent stem cell aggregation in stirred tank reactors. Sci Rep 2019; 9:12297. [PMID: 31444389 PMCID: PMC6707254 DOI: 10.1038/s41598-019-48814-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 08/13/2019] [Indexed: 12/21/2022] Open
Abstract
The culture of human induced pluripotent stem cells (hiPSCs) at large scale becomes feasible with the aid of scalable suspension setups in continuously stirred tank reactors (CSTRs). Innovative monitoring options and emerging automated process control strategies allow for the necessary highly defined culture conditions. Next to standard process characteristics such as oxygen consumption, pH, and metabolite turnover, a reproducible and steady formation of hiPSC aggregates is vital for process scalability. In this regard, we developed a hiPSC-specific suspension culture unit consisting of a fully monitored CSTR system integrated into a custom-designed and fully automated incubator. As a step towards cost-effective hiPSC suspension culture and to pave the way for flexibility at a large scale, we constructed and utilized tailored miniature CSTRs that are largely made from three-dimensional (3D) printed polylactic acid (PLA) filament, which is a low-cost material used in fused deposition modelling. Further, the monitoring tool for hiPSC suspension cultures utilizes in situ microscopic imaging to visualize hiPSC aggregation in real-time to a statistically significant degree while omitting the need for time-intensive sampling. Suitability of our culture unit, especially concerning the developed hiPSC-specific CSTR system, was proven by demonstrating pluripotency of CSTR-cultured hiPSCs at RNA (including PluriTest) and protein level.
Collapse
Affiliation(s)
- Ivo Schwedhelm
- University Hospital Würzburg, Department Tissue Engineering and Regenerative Medicine (TERM), 97070, Würzburg, Germany
| | - Daniela Zdzieblo
- University Hospital Würzburg, Department Tissue Engineering and Regenerative Medicine (TERM), 97070, Würzburg, Germany
| | - Antje Appelt-Menzel
- University Hospital Würzburg, Department Tissue Engineering and Regenerative Medicine (TERM), 97070, Würzburg, Germany
- Translational Center for Regenerative Therapies, Fraunhofer Institute for Silicate Research ISC, 97070, Würzburg, Germany
| | - Constantin Berger
- University Hospital Würzburg, Department Tissue Engineering and Regenerative Medicine (TERM), 97070, Würzburg, Germany
| | - Tobias Schmitz
- University Hospital Würzburg, Department Tissue Engineering and Regenerative Medicine (TERM), 97070, Würzburg, Germany
| | - Bernhard Schuldt
- University Hospital Schleswig-Holstein, Department of Psychiatry and Psychotherapy, 24105, Kiel, Germany
| | - Andre Franke
- University Hospital Schleswig-Holstein, Institute of Clinical Molecular Biology, 24105, Kiel, Germany
| | - Franz-Josef Müller
- University Hospital Schleswig-Holstein, Department of Psychiatry and Psychotherapy, 24105, Kiel, Germany
| | - Ole Pless
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, 22525, Hamburg, Germany
| | - Thomas Schwarz
- Translational Center for Regenerative Therapies, Fraunhofer Institute for Silicate Research ISC, 97070, Würzburg, Germany
| | - Philipp Wiedemann
- Mannheim University of Applied Sciences, Institute of Molecular and Cell Biology, 68163, Mannheim, Germany
| | - Heike Walles
- University Hospital Würzburg, Department Tissue Engineering and Regenerative Medicine (TERM), 97070, Würzburg, Germany
- Translational Center for Regenerative Therapies, Fraunhofer Institute for Silicate Research ISC, 97070, Würzburg, Germany
| | - Jan Hansmann
- University Hospital Würzburg, Department Tissue Engineering and Regenerative Medicine (TERM), 97070, Würzburg, Germany.
- Translational Center for Regenerative Therapies, Fraunhofer Institute for Silicate Research ISC, 97070, Würzburg, Germany.
| |
Collapse
|
62
|
Wiegand C, Banerjee I. Recent advances in the applications of iPSC technology. Curr Opin Biotechnol 2019; 60:250-258. [PMID: 31386977 DOI: 10.1016/j.copbio.2019.05.011] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 05/03/2019] [Accepted: 05/11/2019] [Indexed: 12/18/2022]
Abstract
Pluripotent stems cells (PSCs) can be expanded indefinitely and differentiated into almost any organ-specific cell type. This has enabled the generation of disease relevant tissues from patients in scalable quantities. iPSC-derived organs and organoids are currently being evaluated both in regenerative therapy which are proceeding towards clinical trials, and for disease modeling, which are facilitating drug screening efforts for discovery of novel therapeutics. Here we will review the current efforts and advances in iPSC technology and its subsequent applications and provide a brief commentary on future outlook of this promising technology.
Collapse
Affiliation(s)
- Connor Wiegand
- Department of Chemical and Petroleum Engineering, University of Pittsburgh, United States
| | - Ipsita Banerjee
- Department of Chemical and Petroleum Engineering, University of Pittsburgh, United States; Department of Bioengineering, University of Pittsburgh, United States; McGowan Institute for Regenerative Medicine, United States.
| |
Collapse
|
63
|
de la Roche J, Angsutararux P, Kempf H, Janan M, Bolesani E, Thiemann S, Wojciechowski D, Coffee M, Franke A, Schwanke K, Leffler A, Luanpitpong S, Issaragrisil S, Fischer M, Zweigerdt R. Comparing human iPSC-cardiomyocytes versus HEK293T cells unveils disease-causing effects of Brugada mutation A735V of Na V1.5 sodium channels. Sci Rep 2019; 9:11173. [PMID: 31371804 PMCID: PMC6673693 DOI: 10.1038/s41598-019-47632-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 07/22/2019] [Indexed: 11/23/2022] Open
Abstract
Loss-of-function mutations of the SCN5A gene encoding for the sodium channel α-subunit NaV1.5 result in the autosomal dominant hereditary disease Brugada Syndrome (BrS) with a high risk of sudden cardiac death in the adult. We here engineered human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) carrying the CRISPR/Cas9 introduced BrS-mutation p.A735V-NaV1.5 (g.2204C > T in exon 14 of SCN5A) as a novel model independent of patient´s genetic background. Recent studies raised concern regarding the use of hiPSC-CMs for studying adult-onset hereditary diseases due to cells' immature phenotype. To tackle this concern, long-term cultivation of hiPSC-CMs on a stiff matrix (27-42 days) was applied to promote maturation. Patch clamp recordings of A735V mutated hiPSC-CMs revealed a substantially reduced upstroke velocity and sodium current density, a prominent rightward shift of the steady state activation curve and decelerated recovery from inactivation as compared to isogenic hiPSC-CMs controls. These observations were substantiated by a comparative study on mutant A735V-NaV1.5 channels heterologously expressed in HEK293T cells. In contrast to mutated hiPSC-CMs, a leftward shift of sodium channel inactivation was not observed in HEK293T, emphasizing the importance of investigating mechanisms of BrS in independent systems. Overall, our approach supports hiPSC-CMs' relevance for investigating channelopathies in a dish.
Collapse
Affiliation(s)
- Jeanne de la Roche
- Institute for Neurophysiology, Hannover Medical School, Carl-Neuberg-Str.1, 30625, Hannover, Germany.
| | - Paweorn Angsutararux
- Siriraj Center of Excellence for Stem Cell Research (SiSCR), Faculty of Medicine, Siriraj Hospital, 2, Bangkoknoi, Bangkok, 10700, Thailand
| | - Henning Kempf
- Department of Cardiac, Thoracic, Transplantation and Vascular Surgery (HTTG), Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO) and Hans Borst Zentrum (HBZ), Hannover Medical School, Carl-Neuberg-Str.1, 30625, Hannover, Germany
- Department of Stem Cell Discovery, Novo Nordisk A/S, 2760, Maaloev, Denmark
| | - Montira Janan
- Siriraj Center of Excellence for Stem Cell Research (SiSCR), Faculty of Medicine, Siriraj Hospital, 2, Bangkoknoi, Bangkok, 10700, Thailand
| | - Emiliano Bolesani
- Department of Cardiac, Thoracic, Transplantation and Vascular Surgery (HTTG), Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO) and Hans Borst Zentrum (HBZ), Hannover Medical School, Carl-Neuberg-Str.1, 30625, Hannover, Germany
| | - Stefan Thiemann
- Institute for Neurophysiology, Hannover Medical School, Carl-Neuberg-Str.1, 30625, Hannover, Germany
| | - Daniel Wojciechowski
- Institute for Neurophysiology, Hannover Medical School, Carl-Neuberg-Str.1, 30625, Hannover, Germany
| | - Michelle Coffee
- Department of Cardiac, Thoracic, Transplantation and Vascular Surgery (HTTG), Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO) and Hans Borst Zentrum (HBZ), Hannover Medical School, Carl-Neuberg-Str.1, 30625, Hannover, Germany
| | - Annika Franke
- Department of Cardiac, Thoracic, Transplantation and Vascular Surgery (HTTG), Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO) and Hans Borst Zentrum (HBZ), Hannover Medical School, Carl-Neuberg-Str.1, 30625, Hannover, Germany
| | - Kristin Schwanke
- Department of Cardiac, Thoracic, Transplantation and Vascular Surgery (HTTG), Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO) and Hans Borst Zentrum (HBZ), Hannover Medical School, Carl-Neuberg-Str.1, 30625, Hannover, Germany
| | - Andreas Leffler
- Department of Anaesthesiology and Intensive Care, Hannover Medical School, Carl-Neuberg-Str.1, 30625, Hannover, Germany
| | - Sudjit Luanpitpong
- Siriraj Center of Excellence for Stem Cell Research (SiSCR), Faculty of Medicine, Siriraj Hospital, 2, Bangkoknoi, Bangkok, 10700, Thailand
| | - Surapol Issaragrisil
- Siriraj Center of Excellence for Stem Cell Research (SiSCR), Faculty of Medicine, Siriraj Hospital, 2, Bangkoknoi, Bangkok, 10700, Thailand.
| | - Martin Fischer
- Institute for Neurophysiology, Hannover Medical School, Carl-Neuberg-Str.1, 30625, Hannover, Germany
| | - Robert Zweigerdt
- Department of Cardiac, Thoracic, Transplantation and Vascular Surgery (HTTG), Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO) and Hans Borst Zentrum (HBZ), Hannover Medical School, Carl-Neuberg-Str.1, 30625, Hannover, Germany.
| |
Collapse
|
64
|
Halloin C, Schwanke K, Löbel W, Franke A, Szepes M, Biswanath S, Wunderlich S, Merkert S, Weber N, Osten F, de la Roche J, Polten F, Christoph Wollert K, Kraft T, Fischer M, Martin U, Gruh I, Kempf H, Zweigerdt R. Continuous WNT Control Enables Advanced hPSC Cardiac Processing and Prognostic Surface Marker Identification in Chemically Defined Suspension Culture. Stem Cell Reports 2019; 13:366-379. [PMID: 31353227 PMCID: PMC6700605 DOI: 10.1016/j.stemcr.2019.06.004] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 06/21/2019] [Accepted: 06/24/2019] [Indexed: 02/06/2023] Open
Abstract
Aiming at clinical translation, robust directed differentiation of human pluripotent stem cells (hPSCs), preferentially in chemically defined conditions, is a key requirement. Here, feasibility of suspension culture based hPSC-cardiomyocyte (hPSC-CM) production in low-cost, xeno-free media compatible with good manufacturing practice standards is shown. Applying stirred tank bioreactor systems at increasing dimensions, our advanced protocol enables routine production of about 1 million hPSC-CMs/mL, yielding ∼1.3 × 108 CM in 150 mL and ∼4.0 × 108 CMs in 350–500 mL process scale at >90% lineage purity. Process robustness and efficiency is ensured by uninterrupted chemical WNT pathway control at early stages of differentiation and results in the formation of almost exclusively ventricular-like CMs. Modulated WNT pathway regulation also revealed the previously unappreciated role of ROR1/CD13 as superior surrogate markers for predicting cardiac differentiation efficiency as soon as 72 h of differentiation. This monitoring strategy facilitates process upscaling and controlled mass production of hPSC derivatives. Chemically defined hPSC cardiac differentiation applicable to stirred tank reactors Protocol generates >90% purity of ventricular-like cardiomyocytes Uninterrupted WNT pathway control enables superior cardiac mesoderm formation Novel ROR1/CD13 combination as superior, predictive marker of cardiomyogenesis
Collapse
Affiliation(s)
- Caroline Halloin
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic-, Transplantation and Vascular Surgery, REBIRTH-Cluster of Excellence, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Kristin Schwanke
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic-, Transplantation and Vascular Surgery, REBIRTH-Cluster of Excellence, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Wiebke Löbel
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic-, Transplantation and Vascular Surgery, REBIRTH-Cluster of Excellence, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Annika Franke
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic-, Transplantation and Vascular Surgery, REBIRTH-Cluster of Excellence, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Monika Szepes
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic-, Transplantation and Vascular Surgery, REBIRTH-Cluster of Excellence, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Santoshi Biswanath
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic-, Transplantation and Vascular Surgery, REBIRTH-Cluster of Excellence, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Stephanie Wunderlich
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic-, Transplantation and Vascular Surgery, REBIRTH-Cluster of Excellence, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Sylvia Merkert
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic-, Transplantation and Vascular Surgery, REBIRTH-Cluster of Excellence, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Natalie Weber
- Institute of Molecular and Cell Physiology, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625, Hannover, Germany
| | - Felix Osten
- Institute of Molecular and Cell Physiology, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625, Hannover, Germany
| | - Jeanne de la Roche
- Institute for Neurophysiology, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Felix Polten
- Division of Molecular and Translational Cardiology and Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Kai Christoph Wollert
- Division of Molecular and Translational Cardiology and Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Theresia Kraft
- Institute of Molecular and Cell Physiology, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625, Hannover, Germany
| | - Martin Fischer
- Institute for Neurophysiology, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Ulrich Martin
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic-, Transplantation and Vascular Surgery, REBIRTH-Cluster of Excellence, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Ina Gruh
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic-, Transplantation and Vascular Surgery, REBIRTH-Cluster of Excellence, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Henning Kempf
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic-, Transplantation and Vascular Surgery, REBIRTH-Cluster of Excellence, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany.
| | - Robert Zweigerdt
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic-, Transplantation and Vascular Surgery, REBIRTH-Cluster of Excellence, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany.
| |
Collapse
|
65
|
Abstract
PURPOSE OF REVIEW Cardiovascular disease is the leading cause of mortality worldwide. Pluripotent stem cell-derived cardiomyocytes (PSC-CMs) have great potential to treat heart disease, owing to their capacity of engraftment and remuscularization in the host heart after transplantation. In the current review, we provide an overview of PSC-CMs for clinical transplantation. RECENT FINDINGS Studies have shown that PSC-CMs can survive, engraft, and form gap junctions after transplantation, with functional benefit. Engrafted PSC-CMs matured gradually in host hearts. Only in a large animal model, transient ventricular arrhythmias were detected, mainly because of the ectopic pacing from the grafted PSC-CMs. Although intense immunosuppression is unavoidable in xenotransplantation, immunosuppression remains necessary for MHC-matched allogenic non-human primate PSC-CMs transplantation. This review offers insights on how PSC-CMs contribute to functional benefit after transplantation to injured non-human primate hearts. We believe that PSC-CM transplantation represents a potentially novel treatment for ischemic heart diseases, provided that several technological and biological limitations can be overcome.
Collapse
Affiliation(s)
- Shin Kadota
- Department of Regenerative Science and Medicine, Institute for Biomedical Sciences, Shinshu University, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
| | - Yuji Shiba
- Department of Regenerative Science and Medicine, Institute for Biomedical Sciences, Shinshu University, 3-1-1 Asahi, Matsumoto, 390-8621, Japan.
| |
Collapse
|
66
|
Le MNT, Hasegawa K. Expansion Culture of Human Pluripotent Stem Cells and Production of Cardiomyocytes. Bioengineering (Basel) 2019; 6:E48. [PMID: 31137703 PMCID: PMC6632060 DOI: 10.3390/bioengineering6020048] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 05/15/2019] [Accepted: 05/18/2019] [Indexed: 12/25/2022] Open
Abstract
Transplantation of human pluripotent stem cell (hPSCs)-derived cardiomyocytes for the treatment of heart failure is a promising therapy. In order to implement this therapy requiring numerous cardiomyocytes, substantial production of hPSCs followed by cardiac differentiation seems practical. Conventional methods of culturing hPSCs involve using a 2D culture monolayer that hinders the expansion of hPSCs, thereby limiting their productivity. Advanced culture of hPSCs in 3D aggregates in the suspension overcomes the limitations of 2D culture and attracts immense attention. Although the hPSC production needs to be suitable for subsequent cardiac differentiation, many studies have independently focused on either expansion of hPSCs or cardiac differentiation protocols. In this review, we summarize the recent approaches to expand hPSCs in combination with cardiomyocyte differentiation. A comparison of various suspension culture methods and future prospects for dynamic culture of hPSCs are discussed in this study. Understanding hPSC characteristics in different models of dynamic culture helps to produce numerous cells that are useful for further clinical applications.
Collapse
Affiliation(s)
- Minh Nguyen Tuyet Le
- Institute for Integrated Cell-Material Sciences (iCeMS), Institute for Advanced Study, Kyoto University, Kyoto 606-8501, Japan.
| | - Kouichi Hasegawa
- Institute for Integrated Cell-Material Sciences (iCeMS), Institute for Advanced Study, Kyoto University, Kyoto 606-8501, Japan.
| |
Collapse
|
67
|
Abstract
Myocardial infarction leads to an irreversible loss of vital myocardial cells. The transplantation of new cardiomyocytes into the heart was first described over 20 years ago and represents a straightforward approach to remuscularize a damaged heart. Due to the lack of human cells a clinical application seemed ambitious; however, dramatic progress in stem cell biology over the last two decades has paved the way towards a clinical application. This is especially important as the prognosis for patients with terminal heart failure is still poor. The transplantation of either cardiomyocytes or engineered heart tissue derived from pluripotent stem cells (either embryonic stem cells or induced pluripotent stem cells) might represent a new regenerative approach. Transplantation of either cells or tissue constructs has now been evaluated in several preclinical models, which have demonstrated that an injured heart can be (partially) remuscularized; however, major hurdles towards a clinical application are the transplantation-related occurrence of arrhythmia, the potential tumorigenicity of pluripotent cells and the required immunosuppression. Several groups are working hard to solve these problems and we are optimistic that the first clinical studies will take place within the next few years.
Collapse
|
68
|
Yoshida S, Sumomozawa K, Nagamine K, Nishizawa M. Hydrogel Microchambers Integrated with Organic Electrodes for Efficient Electrical Stimulation of Human iPSC-Derived Cardiomyocytes. Macromol Biosci 2019; 19:e1900060. [PMID: 31038841 DOI: 10.1002/mabi.201900060] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 03/30/2019] [Indexed: 12/20/2022]
Abstract
A hydrogel-based microchamber with organic electrodes for efficient electrical stimulations of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) is described. The microchamber is made from molecularly permeable, optically transparent, and electrically conductive polyvinyl alcohol (PVA) hydrogel and highly capacitive carbon electrode modified with poly(3,4-ethylenedioxythiophene) (PEDOT). Spheroids of hiPSC-CMs are cultured in microchambers, and electrically stimulated by the electrode for maturation. The large interfacial capacitance of the electrodes enables several days of electrical stimulation without generation of cytotoxic bubbles even when the electrodes are placed near the spheroids. The spheroids can be cultivated in the closed microchambers because of the permeated nutrients through the hydrogel, thus the spheroids are stably addressable and the culture medium around the sealed microchambers can be simply exchanged. Synchronized beating of the spheroids can be optically analyzed in situ, which makes it possible to selectively collect electrically responsive cells for further use. As the hydrogel is electrically conductive, the amount of electrical charge needed for maturing the spheroids can be reduced by configuring electrodes on the top and the bottom of the microchamber. The bioreactor will be useful for efficient production of matured hiPSC-CMs for regenerative medicine and drug screening.
Collapse
Affiliation(s)
- Shotaro Yoshida
- Department of Finemechanics, Tohoku University, 6-6-1 Aramaki Aoba, Sendai, 980-8579, Japan
| | - Kensuke Sumomozawa
- Department of Finemechanics, Tohoku University, 6-6-1 Aramaki Aoba, Sendai, 980-8579, Japan
| | - Kuniaki Nagamine
- Department of Finemechanics, Tohoku University, 6-6-1 Aramaki Aoba, Sendai, 980-8579, Japan
| | - Matsuhiko Nishizawa
- Department of Finemechanics, Tohoku University, 6-6-1 Aramaki Aoba, Sendai, 980-8579, Japan
| |
Collapse
|
69
|
Müller D, Hagenah D, Biswanath S, Coffee M, Kampmann A, Zweigerdt R, Heisterkamp A, Kalies SMK. Femtosecond laser-based nanosurgery reveals the endogenous regeneration of single Z-discs including physiological consequences for cardiomyocytes. Sci Rep 2019; 9:3625. [PMID: 30842507 PMCID: PMC6403391 DOI: 10.1038/s41598-019-40308-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 02/13/2019] [Indexed: 11/24/2022] Open
Abstract
A highly organized cytoskeleton architecture is the basis for continuous and controlled contraction in cardiomyocytes (CMs). Abnormalities in cytoskeletal elements, like the Z-disc, are linked to several diseases. It is challenging to reveal the mechanisms of CM failure, endogenous repair, or mechanical homeostasis on the scale of single cytoskeletal elements. Here, we used a femtosecond (fs) laser to ablate single Z-discs in human pluripotent stem cells (hPSC) -derived CMs (hPSC-CM) and neonatal rat CMs. We show, that CM viability was unaffected by the loss of a single Z-disc. Furthermore, more than 40% of neonatal rat and 68% of hPSC-CMs recovered the Z-disc loss within 24 h. Significant differences to control cells, after the Z-disc loss, in terms of cell perimeter, x- and y-expansion and calcium homeostasis were not found. Only 14 days in vitro old hPSC-CMs reacted with a significant decrease in cell area, x- and y-expansion 24 h past nanosurgery. This demonstrates that CMs can compensate the loss of a single Z-disc and recover a regular sarcomeric pattern during spontaneous contraction. It also highlights the significant potential of fs laser-based nanosurgery to physically micro manipulate CMs to investigate cytoskeletal functions and organization of single elements.
Collapse
Affiliation(s)
- Dominik Müller
- Institute of Quantum Optics, Leibniz University Hannover, Hannover, Germany. .,REBIRTH-Cluster of Excellence, Hannover Medical School, Hannover, Germany. .,Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany.
| | - Dorian Hagenah
- Institute of Quantum Optics, Leibniz University Hannover, Hannover, Germany.,REBIRTH-Cluster of Excellence, Hannover Medical School, Hannover, Germany.,Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany
| | - Santoshi Biswanath
- REBIRTH-Cluster of Excellence, Hannover Medical School, Hannover, Germany.,Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic, Transplantation and Vascular Surgery (HTTG), Hannover Medical, School, Hannover, Germany
| | - Michelle Coffee
- REBIRTH-Cluster of Excellence, Hannover Medical School, Hannover, Germany.,Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic, Transplantation and Vascular Surgery (HTTG), Hannover Medical, School, Hannover, Germany
| | - Andreas Kampmann
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany.,Clinic for Cranio-Maxillo-Facial Surgery, Hannover Medical School, Hannover, Germany
| | - Robert Zweigerdt
- REBIRTH-Cluster of Excellence, Hannover Medical School, Hannover, Germany.,Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic, Transplantation and Vascular Surgery (HTTG), Hannover Medical, School, Hannover, Germany
| | - Alexander Heisterkamp
- Institute of Quantum Optics, Leibniz University Hannover, Hannover, Germany.,REBIRTH-Cluster of Excellence, Hannover Medical School, Hannover, Germany.,Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany
| | - Stefan M K Kalies
- Institute of Quantum Optics, Leibniz University Hannover, Hannover, Germany.,REBIRTH-Cluster of Excellence, Hannover Medical School, Hannover, Germany.,Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany
| |
Collapse
|
70
|
Madonna R, Van Laake LW, Botker HE, Davidson SM, De Caterina R, Engel FB, Eschenhagen T, Fernandez-Aviles F, Hausenloy DJ, Hulot JS, Lecour S, Leor J, Menasché P, Pesce M, Perrino C, Prunier F, Van Linthout S, Ytrehus K, Zimmermann WH, Ferdinandy P, Sluijter JPG. ESC Working Group on Cellular Biology of the Heart: position paper for Cardiovascular Research: tissue engineering strategies combined with cell therapies for cardiac repair in ischaemic heart disease and heart failure. Cardiovasc Res 2019; 115:488-500. [PMID: 30657875 PMCID: PMC6383054 DOI: 10.1093/cvr/cvz010] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 12/21/2018] [Accepted: 01/10/2019] [Indexed: 12/15/2022] Open
Abstract
Morbidity and mortality from ischaemic heart disease (IHD) and heart failure (HF) remain significant in Europe and are increasing worldwide. Patients with IHD or HF might benefit from novel therapeutic strategies, such as cell-based therapies. We recently discussed the therapeutic potential of cell-based therapies and provided recommendations on how to improve the therapeutic translation of these novel strategies for effective cardiac regeneration and repair. Despite major advances in optimizing these strategies with respect to cell source and delivery method, the clinical outcome of cell-based therapy remains unsatisfactory. Major obstacles are the low engraftment and survival rate of transplanted cells in the harmful microenvironment of the host tissue, and the paucity or even lack of endogenous cells with repair capacity. Therefore, new ways of delivering cells and their derivatives are required in order to empower cell-based cardiac repair and regeneration in patients with IHD or HF. Strategies using tissue engineering (TE) combine cells with matrix materials to enhance cell retention or cell delivery in the transplanted area, and have recently received much attention for this purpose. Here, we summarize knowledge on novel approaches emerging from the TE scenario. In particular, we will discuss how combinations of cell/bio-materials (e.g. hydrogels, cell sheets, prefabricated matrices, microspheres, and injectable matrices) combinations might enhance cell retention or cell delivery in the transplantation areas, thereby increase the success rate of cell therapies for IHD and HF. We will not focus on the use of classical engineering approaches, employing fully synthetic materials, because of their unsatisfactory material properties which render them not clinically applicable. The overall aim of this Position Paper from the ESC Working Group Cellular Biology of the Heart is to provide recommendations on how to proceed in research with these novel TE strategies combined with cell-based therapies to boost cardiac repair in the clinical settings of IHD and HF.
Collapse
Affiliation(s)
- Rosalinda Madonna
- Institute of Cardiology and Center of Excellence on Aging, “G. d’Annunzio” University—Chieti, Italy
- University of Texas Medical School in Houston, USA
| | - Linda W Van Laake
- Cardiology and UMC Utrecht Regenerative Medicine Center, University Medical Center Utrecht, The Netherlands
| | - Hans Erik Botker
- Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, London, UK
| | - Raffaele De Caterina
- Institute of Cardiology and Center of Excellence on Aging, “G. d’Annunzio” University—Chieti, Italy
- University of Texas Medical School in Houston, USA
- University of Pisa, Pisa University Hospital, Pisa, Italy
| | - Felix B Engel
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany; Muscle Research Center Erlangen, MURCE
| | - Thomas Eschenhagen
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Francesco Fernandez-Aviles
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain
- CIBERCV, ISCIII, Madrid, Spain
| | - Derek J Hausenloy
- The Hatter Cardiovascular Institute, University College London, London, UK
- Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore
- National Heart Research Institute Singapore, National Heart Centre, Singapore
- Yong Loo Lin School of Medicine, National University Singapore, Singapore
- The National Institute of Health Research University College London Hospitals Biomedical Research Centre, Research & Development, London, UK
- Tecnologico de Monterrey, Centro de Biotecnologia-FEMSA, Nuevo Leon, Mexico
| | - Jean-Sebastien Hulot
- Université Paris-Descartes, Sorbonne Paris Cité, Paris, France
- Paris Cardiovascular Research Center (PARCC), INSERM UMRS 970, Paris, France
- Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Sandrine Lecour
- Hatter Cardiovascular Research Institute, University of Cape Town, South Africa
| | - Jonathan Leor
- Tamman and Neufeld Cardiovascular Research Institutes, Sackler Faculty of Medicine, Tel-Aviv University and Sheba Medical Center, Tel-Hashomer, Israel
| | - Philippe Menasché
- Department of Cardiovascular Surgery, Hôpital Européen Georges Pompidou, Paris, France
- Université Paris-Descartes, Sorbonne Paris Cité, Paris, France
- INSERM UMRS 970, Paris, France
| | - Maurizio Pesce
- Unità di Ingegneria Tissutale Cardiovascolare, Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | - Cinzia Perrino
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Fabrice Prunier
- Institut Mitovasc, INSERM, CNRS, Université d’Angers, Service de Cardiologie, CHU Angers, Angers, France
| | - Sophie Van Linthout
- Berlin-Brandenburg Center for Regenerative Therapies, Charité, University Medicine Berlin, Campus Virchow Klinikum, Berlin, Germany
- Department of Cardiology, Charité, University Medicine Berlin, Campus Virchow Klinikum, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Kirsti Ytrehus
- Department of Medical Biology, UiT, The Arctic University of Norway, Norway
| | - Wolfram-Hubertus Zimmermann
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Peter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvárad tér 4, III-V Floor, H-1089 Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Joost P G Sluijter
- Department of Cardiology, Experimental Cardiology Laboratory, Regenerative Medicine Center, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, CX Utrecht, the Netherlands
| |
Collapse
|
71
|
Biomaterializing the promise of cardiac tissue engineering. Biotechnol Adv 2019; 42:107353. [PMID: 30794878 DOI: 10.1016/j.biotechadv.2019.02.009] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 02/18/2019] [Accepted: 02/19/2019] [Indexed: 12/14/2022]
Abstract
During an average individual's lifespan, the human heart pumps nearly 200 million liters of blood delivered by approximately 3 billion heartbeats. Therefore, it is not surprising that native myocardium under this incredible demand is extraordinarily complex, both structurally and functionally. As a result, successful engineering of adult-mimetic functional cardiac tissues is likely to require utilization of highly specialized biomaterials representative of the native extracellular microenvironment. There is currently no single biomaterial that fully recapitulates the architecture or the biochemical and biomechanical properties of adult myocardium. However, significant effort has gone toward designing highly functional materials and tissue constructs that may one day provide a ready source of cardiac tissue grafts to address the overwhelming burden of cardiomyopathic disease. In the near term, biomaterial-based scaffolds are helping to generate in vitro systems for querying the mechanisms underlying human heart homeostasis and disease and discovering new, patient-specific therapeutics. When combined with advances in minimally-invasive cardiac delivery, ongoing efforts will likely lead to scalable cell and biomaterial technologies for use in clinical practice. In this review, we describe recent progress in the field of cardiac tissue engineering with particular emphasis on use of biomaterials for therapeutic tissue design and delivery.
Collapse
|
72
|
Wnorowski A, Yang H, Wu JC. Progress, obstacles, and limitations in the use of stem cells in organ-on-a-chip models. Adv Drug Deliv Rev 2019; 140:3-11. [PMID: 29885330 DOI: 10.1016/j.addr.2018.06.001] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 05/16/2018] [Accepted: 06/01/2018] [Indexed: 12/18/2022]
Abstract
In recent years, drug development costs have soared, primarily due to the failure of preclinical animal and cell culture models, which do not directly translate to human physiology. Organ-on-a-chip (OOC) is a burgeoning technology with the potential to revolutionize disease modeling, drug discovery, and toxicology research by strengthening the relevance of culture-based models while reducing costly animal studies. Although OOC models can incorporate a variety of tissue sources, the most robust and relevant OOC models going forward will include stem cells. In this review, we will highlight the benefits of stem cells as a tissue source while considering current limitations to their complete and effective implementation into OOC models.
Collapse
Affiliation(s)
- Alexa Wnorowski
- Stanford Cardiovascular Institute, Stanford, CA 94305, United States; Department of Bioengineering, Stanford University Schools of Engineering and Medicine, Stanford, CA 943055, United States
| | - Huaxiao Yang
- Stanford Cardiovascular Institute, Stanford, CA 94305, United States
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford, CA 94305, United States; Division of Cardiovascular Medicine, Department of Medicine, Stanford, CA 94305, United States; Department of Radiology, Stanford University School of Medicine, Stanford, CA 94305, United States.
| |
Collapse
|
73
|
Basoli V, Santaniello S, Rinaldi S, Fontani V, Pigliaru G, Wieser M, Strajeriu A, Castagna A, Redl H, Ventura C, Grillari R, Maioli M. Physical stimulation by REAC and BMP4/WNT-1 inhibitor synergistically enhance cardiogenic commitment in iPSCs. PLoS One 2019; 14:e0211188. [PMID: 30673752 PMCID: PMC6343882 DOI: 10.1371/journal.pone.0211188] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 01/08/2019] [Indexed: 12/26/2022] Open
Abstract
It is currently known that pluripotent stem cells can be committed in vitro to the cardiac lineage by the modulation of specific signaling pathways, but it is also well known that, despite the significant increase in cardiomyocyte yield provided by the currently available conditioned media, the resulting cardiogenic commitment remains a highly variable process. Previous studies provided evidence that radio electric fields asymmetrically conveyed through the Radio Electric Asymmetric Conveyer (REAC) technology are able to commit R1 embryonic stem cells and human adipose derived stem cells toward a cardiac phenotype. The present study aimed at investigating whether the effect of physical stimulation by REAC in combination with specific chemical inductors enhance the cardiogenic potential in human induced pluripotent stem cells (iPSCs). The appearance of a cardiac-like phenotype in iPSCs cultured in the presence of a cardiogenic medium, based upon BMP4 and a WNT-inhibitor, was consistently increased by REAC treatment used only during the early fate differentiation for the first 72 hours. REAC-exposed iPSCs exhibited an upregulation in the expression of specific cardiogenic transcripts and morphologically in the number of beating clusters, as compared to cells cultured in the cardiogenic medium alone. Our results indicate that physical modulation of cellular dynamics provided by the REAC offers an affordable strategy to mimic iPSC cardiac-like fates in the presence of a cardiogenic milieu.
Collapse
Affiliation(s)
- Valentina Basoli
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
- Research Department, Rinaldi Fontani Foundation, Florence, Italy
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Sara Santaniello
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
- National Laboratory of Molecular Biology and Stem Cell Engineering - National Institute of Biostructures and Biosystems-Eldor Lab, at Innovation Accelerators, CNR, Bologna, Italy
| | - Salvatore Rinaldi
- Research Department, Rinaldi Fontani Foundation, Florence, Italy
- Department of Regenerative Medicine, Rinaldi Fontani Institute, Florence, Italy
- IRF Shanghai Medical Sciences, Shanghai, China
| | - Vania Fontani
- Research Department, Rinaldi Fontani Foundation, Florence, Italy
- Department of Regenerative Medicine, Rinaldi Fontani Institute, Florence, Italy
- IRF Shanghai Medical Sciences, Shanghai, China
| | - Gianfranco Pigliaru
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
- National Laboratory of Molecular Biology and Stem Cell Engineering - National Institute of Biostructures and Biosystems-Eldor Lab, at Innovation Accelerators, CNR, Bologna, Italy
| | - Matthias Wieser
- Evercyte GmbH, Vienna, Austria
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Agata Strajeriu
- Evercyte GmbH, Vienna, Austria
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Alessandro Castagna
- Department of Regenerative Medicine, Rinaldi Fontani Institute, Florence, Italy
| | - Heinz Redl
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Carlo Ventura
- National Laboratory of Molecular Biology and Stem Cell Engineering - National Institute of Biostructures and Biosystems-Eldor Lab, at Innovation Accelerators, CNR, Bologna, Italy
- Department of Regenerative Medicine, Rinaldi Fontani Institute, Florence, Italy
| | - Regina Grillari
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
- Evercyte GmbH, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Margherita Maioli
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
- IRF Shanghai Medical Sciences, Shanghai, China
- Center for developmental biology and reprogramming - CEDEBIOR, Department of Biomedical Sciences, University of Sassari and National Institute of Biostructures and Biosystems, Sassari, Italy
| |
Collapse
|
74
|
Christoffersson J, Meier F, Kempf H, Schwanke K, Coffee M, Beilmann M, Zweigerdt R, Mandenius CF. Evaluating the Effect of Drug Compounds on Cardiac Spheroids Using the Cardiac Cell Outgrowth Assay. Methods Mol Biol 2019; 1994:185-193. [PMID: 31124116 DOI: 10.1007/978-1-4939-9477-9_17] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The ideal cell culture model should mimic the cell physiology and the mechanical and the chemical cues that are present in specific tissues and organs, within a convenient high-throughput format. A possible key feature for such models is to recapture the cell polarity, the interactions between cells, and the interactions between the cells and the elastic extracellular matrix (ECM) by orienting the cells in a three-dimensional (3D) matrix. A common method to create 3D cell environments is to let the cells aggregate into spheroids with a diameter of around 200 μm. A major challenge for 3D cell cultures is to perform quick and easy imaging of the dense cell population, especially noninvasively. This protocol explains how to take advantage of the number of cells growing out from cell spheroids over time as a readout of the effect of a drug. The assay is compatible with standard imaging techniques and can be performed noninvasively using light microscopy or as a complement to other fluorescent imaging assays.
Collapse
Affiliation(s)
- Jonas Christoffersson
- Division of Biotechnology, Department of Physics, Chemistry and Biology (IFM), Linköping University, Linköping, Sweden
| | - Florian Meier
- Boehringer Ingelheim Pharma GmbH and Co. KG, Nonclinical Drug Safety Germany, Biberach, Germany
| | - Henning Kempf
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Hannover, Germany
| | - Kristin Schwanke
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Hannover, Germany
| | - Michelle Coffee
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Hannover, Germany
| | - Mario Beilmann
- Boehringer Ingelheim Pharma GmbH and Co. KG, Nonclinical Drug Safety Germany, Biberach, Germany
| | - Robert Zweigerdt
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Hannover, Germany
| | - Carl-Fredrik Mandenius
- Division of Biotechnology, Department of Physics, Chemistry and Biology (IFM), Linköping University, Linköping, Sweden.
| |
Collapse
|
75
|
Human Pluripotent Stem Cells: Applications and Challenges for Regenerative Medicine and Disease Modeling. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2019; 171:189-224. [PMID: 31740987 DOI: 10.1007/10_2019_117] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In recent years, human pluripotent stem (hPS) cells have started to emerge as a potential tool with application in fields such as regenerative medicine, disease modeling, and drug screening. In particular, the ability to differentiate human-induced pluripotent stem (hiPS) cells into different cell types and to mimic structures and functions of a specific target organ, resourcing to organoid technology, has introduced novel model systems for disease recapitulation while offering a powerful tool to provide a faster and reproducible approach in the process of drug discovery. All these technologies are expected to improve the overall quality of life of the humankind. Here, we highlight the main applications of hiPS cells and the main challenges associated with the translation of hPS cell derivatives into clinical settings and other biomedical applications, such as the costs of the process and the ability to mimic the complexity of the in vivo systems. Moreover, we focus on the bioprocessing approaches that can be applied towards the production of high numbers of cells as well as their efficient differentiation into the final product and further purification.
Collapse
|
76
|
Ackermann M, Kempf H, Hetzel M, Hesse C, Hashtchin AR, Brinkert K, Schott JW, Haake K, Kühnel MP, Glage S, Figueiredo C, Jonigk D, Sewald K, Schambach A, Wronski S, Moritz T, Martin U, Zweigerdt R, Munder A, Lachmann N. Bioreactor-based mass production of human iPSC-derived macrophages enables immunotherapies against bacterial airway infections. Nat Commun 2018; 9:5088. [PMID: 30504915 PMCID: PMC6269475 DOI: 10.1038/s41467-018-07570-7] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 11/08/2018] [Indexed: 11/18/2022] Open
Abstract
The increasing number of severe infections with multi-drug-resistant pathogens worldwide highlights the need for alternative treatment options. Given the pivotal role of phagocytes and especially alveolar macrophages in pulmonary immunity, we introduce a new, cell-based treatment strategy to target bacterial airway infections. Here we show that the mass production of therapeutic phagocytes from induced pluripotent stem cells (iPSC) in industry-compatible, stirred-tank bioreactors is feasible. Bioreactor-derived iPSC-macrophages (iPSC-Mac) represent a highly pure population of CD45+CD11b+CD14+CD163+ cells, and share important phenotypic, functional and transcriptional hallmarks with professional phagocytes, however with a distinct transcriptome signature similar to primitive macrophages. Most importantly, bioreactor-derived iPSC-Mac rescue mice from Pseudomonas aeruginosa-mediated acute infections of the lower respiratory tract within 4-8 h post intra-pulmonary transplantation and reduce bacterial load. Generation of specific immune-cells from iPSC-sources in scalable stirred-tank bioreactors can extend the field of immunotherapy towards bacterial infections, and may allow for further innovative cell-based treatment strategies. Pulmonary infections constitute a substantial health problem worldwide. Here the authors show that phagocytes similar to primitive macrophages can be generated from human induced pluripotent stem cells, by the use of industry-compatible, stirred-tank bioreactors, and applied as a cell-based therapy to treat acute bacterial infections in mice.
Collapse
Affiliation(s)
- Mania Ackermann
- JRG Translational Hematology of Congenital Diseases, Hannover Medical School, Hannover, Germany.,Institute of Experimental Hematology, REBIRTH Cluster of Excellence, Hannover Medical School, 30625, Hannover, Germany
| | - Henning Kempf
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, REBIRTH Cluster of Excellence, Hannover Medical School, 30625, Hannover, Germany.,Department of Stem Cell Biology, Novo Nordisk A/S, 2760, Maaloev, Denmark
| | - Miriam Hetzel
- Institute of Experimental Hematology, REBIRTH Cluster of Excellence, Hannover Medical School, 30625, Hannover, Germany.,RG Reprogramming and Gene Therapy, Hannover Medical School, 30625, Hannover, Germany
| | - Christina Hesse
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), REBIRTH Cluster-of Excellence, 30625, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), German Center for Lung Research, 30625, Hannover, Germany
| | - Anna Rafiei Hashtchin
- JRG Translational Hematology of Congenital Diseases, Hannover Medical School, Hannover, Germany.,Institute of Experimental Hematology, REBIRTH Cluster of Excellence, Hannover Medical School, 30625, Hannover, Germany
| | - Kerstin Brinkert
- Clinical Research Group 'Cystic Fibrosis', Clinic for Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625, Hannover, Germany
| | - Juliane Wilhelmine Schott
- Institute of Experimental Hematology, REBIRTH Cluster of Excellence, Hannover Medical School, 30625, Hannover, Germany
| | - Kathrin Haake
- JRG Translational Hematology of Congenital Diseases, Hannover Medical School, Hannover, Germany.,Institute of Experimental Hematology, REBIRTH Cluster of Excellence, Hannover Medical School, 30625, Hannover, Germany
| | - Mark Philipp Kühnel
- Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), German Center for Lung Research, 30625, Hannover, Germany.,Institute for Pathology, Hannover Medical School, 30625, Hannover, Germany
| | - Silke Glage
- Institute of Laboratory Animal Science and Central Animal Facility, Hannover Medical School, Hannover, Germany
| | - Constanca Figueiredo
- Institute for Transfusion Medicine, Hannover Medical School, 30625, Hannover, Germany
| | - Danny Jonigk
- Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), German Center for Lung Research, 30625, Hannover, Germany.,Institute for Pathology, Hannover Medical School, 30625, Hannover, Germany
| | - Katherina Sewald
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), REBIRTH Cluster-of Excellence, 30625, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), German Center for Lung Research, 30625, Hannover, Germany
| | - Axel Schambach
- Institute of Experimental Hematology, REBIRTH Cluster of Excellence, Hannover Medical School, 30625, Hannover, Germany.,Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, 02215, USA
| | - Sabine Wronski
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), REBIRTH Cluster-of Excellence, 30625, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), German Center for Lung Research, 30625, Hannover, Germany
| | - Thomas Moritz
- Institute of Experimental Hematology, REBIRTH Cluster of Excellence, Hannover Medical School, 30625, Hannover, Germany.,RG Reprogramming and Gene Therapy, Hannover Medical School, 30625, Hannover, Germany
| | - Ulrich Martin
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, REBIRTH Cluster of Excellence, Hannover Medical School, 30625, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), German Center for Lung Research, 30625, Hannover, Germany
| | - Robert Zweigerdt
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, REBIRTH Cluster of Excellence, Hannover Medical School, 30625, Hannover, Germany
| | - Antje Munder
- Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), German Center for Lung Research, 30625, Hannover, Germany.,Clinical Research Group 'Cystic Fibrosis', Clinic for Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625, Hannover, Germany
| | - Nico Lachmann
- JRG Translational Hematology of Congenital Diseases, Hannover Medical School, Hannover, Germany. .,Institute of Experimental Hematology, REBIRTH Cluster of Excellence, Hannover Medical School, 30625, Hannover, Germany.
| |
Collapse
|
77
|
Dakhore S, Nayer B, Hasegawa K. Human Pluripotent Stem Cell Culture: Current Status, Challenges, and Advancement. Stem Cells Int 2018; 2018:7396905. [PMID: 30595701 PMCID: PMC6282144 DOI: 10.1155/2018/7396905] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 10/23/2018] [Accepted: 10/24/2018] [Indexed: 12/23/2022] Open
Abstract
Over the past two decades, human embryonic stem cells (hESCs) have gained attention due to their pluripotent and proliferative ability which enables production of almost all cell types in the human body in vitro and makes them an excellent tool to study human embryogenesis and disease, as well as for drug discovery and cell transplantation therapies. Discovery of human-induced pluripotent stem cells (hiPSCs) further expanded therapeutic applications of human pluripotent stem cells (PSCs). hPSCs provide a stable and unlimited original cell source for producing suitable cells and tissues for downstream applications. Therefore, engineering the environment in which these cells are grown, for stable and quality-controlled hPSC maintenance and production, is one of the key factors governing the success of these applications. hPSCs are maintained in a particular niche using specific cell culture components. Ideally, the culture should be free of xenobiotic components to render hPSCs suitable for therapeutic applications. Substantial efforts have been put to identify effective components, and develop culture conditions and protocols, for their large-scale expansion without compromising on quality. In this review, we discuss different media, their components and functions, including specific requirements to maintain the pluripotent and proliferative ability of hPSCs. Understanding the role of culture components would enable the development of appropriate conditions to promote large-scale, quality-controlled expansion of hPSCs thereby increasing their potential applications.
Collapse
Affiliation(s)
- Sushrut Dakhore
- Institute for Stem Cell Biology and Regenerative Medicine (inStem), National Centre for Biological Sciences (NCBS), Bangalore, India
| | - Bhavana Nayer
- Institute for Stem Cell Biology and Regenerative Medicine (inStem), National Centre for Biological Sciences (NCBS), Bangalore, India
| | - Kouichi Hasegawa
- Institute for Stem Cell Biology and Regenerative Medicine (inStem), National Centre for Biological Sciences (NCBS), Bangalore, India
- Institute for Integrated Cell-Material Sciences (iCeMS), Institute for Advanced Study, Kyoto University, Japan
| |
Collapse
|
78
|
Scalable Cardiac Differentiation of Pluripotent Stem Cells Using Specific Growth Factors and Small Molecules. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2018; 163:39-69. [PMID: 29071404 DOI: 10.1007/10_2017_30] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The envisioned routine application of human pluripotent stem cell (hPSC)-derived cardiomyocytes (CMs) for therapies and industry-compliant screening approaches will require efficient and highly reproducible processes for the mass production of well-characterized CM batches.On their way toward beating CMs, hPSCs initially undergo an epithelial-to-mesenchymal transition into a primitive-streak (PS)-like population that later gives rise to all endodermal and mesodermal lineages, including cardiovascular progenies (CVPs). CVPs are multipotent and possess the capability to give rise to all major cell types of the heart, including CMs, endothelial cells, cardiac fibroblasts, and smooth muscle cells. This article provides an historical overview and describes the stepwise development of protocols that typically result in the appearance of beating CMs within 7-12 days of hPSC differentiation.We describe the development of directed and closely controlled cardiomyogenic differentiation, which now enables the induction of >90% CM purity without further lineage enrichment. Although secreted lineage specifiers (revealed from developmental biology) were initially used, we outline the advantages of chemical pathway modulators, as defined by more recent screening approaches. Subsequently, we discuss the use of defined culture media for upscaling the production of hPSC-CMs in controlled bioreactors and how this, in principle, unlimited source of human CMs can be used to progress heart regeneration and stimulate the drug discovery pipeline. Graphical Abstract.
Collapse
|
79
|
A fully defined static suspension culture system for large-scale human embryonic stem cell production. Cell Death Dis 2018; 9:892. [PMID: 30166524 PMCID: PMC6117302 DOI: 10.1038/s41419-018-0863-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 06/16/2018] [Accepted: 06/21/2018] [Indexed: 12/30/2022]
Abstract
Human embryonic stem cells (hESCs) play an important role in regenerative medicine due to their potential to differentiate into various functional cells. However, the conventional adherent culture system poses challenges to mass production of high-quality hESCs. Though scientists have made many attempts to establish a robust and economical hESC suspension culture system, there are existing limitations, including suboptimal passage methods and shear force caused by dynamic stirring. Here, we report on an efficient large-scale culture system, which enables long-term, GMP grade, single-cell inoculation, and serial expansion of hESCs with a yield of about 1.5 × 109 cells per 1.5-L culture, while maintaining good pluripotency. The suspension culture system was enlarged gradually from a 100-mm dish to a 1.8-L culture bag with methylcellulose involvement to avoid sphere fusion. Under the optimal experimental protocol, this 3D system resolves current problems that limit mass production and clinical application of hESCs, and thus can be used in commercial-level hESC production for cell therapy and pharmaceutics screening in the future.
Collapse
|
80
|
Ting S, Lam A, Tong G, Chen A, Wei H, Wu J, Lam YN, Reuveny S, Oh S. Meticulous optimization of cardiomyocyte yields in a 3-stage continuous integrated agitation bioprocess. Stem Cell Res 2018; 31:161-173. [DOI: 10.1016/j.scr.2018.07.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 07/12/2018] [Accepted: 07/23/2018] [Indexed: 01/15/2023] Open
|
81
|
De Santis MM, Bölükbas DA, Lindstedt S, Wagner DE. How to build a lung: latest advances and emerging themes in lung bioengineering. Eur Respir J 2018; 52:13993003.01355-2016. [PMID: 29903859 DOI: 10.1183/13993003.01355-2016] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Accepted: 05/30/2018] [Indexed: 12/19/2022]
Abstract
Chronic respiratory diseases remain a major cause of morbidity and mortality worldwide. The only option at end-stage disease is lung transplantation, but there are not enough donor lungs to meet clinical demand. Alternative options to increase tissue availability for lung transplantation are urgently required to close the gap on this unmet clinical need. A growing number of tissue engineering approaches are exploring the potential to generate lung tissue ex vivo for transplantation. Both biologically derived and manufactured scaffolds seeded with cells and grown ex vivo have been explored in pre-clinical studies, with the eventual goal of generating functional pulmonary tissue for transplantation. Recently, there have been significant efforts to scale-up cell culture methods to generate adequate cell numbers for human-scale bioengineering approaches. Concomitantly, there have been exciting efforts in designing bioreactors that allow for appropriate cell seeding and development of functional lung tissue over time. This review aims to present the current state-of-the-art progress for each of these areas and to discuss promising new ideas within the field of lung bioengineering.
Collapse
Affiliation(s)
- Martina M De Santis
- Lung Bioengineering and Regeneration, Dept of Experimental Medical Sciences, Lund University, Lund, Sweden.,Lung Repair and Regeneration (LRR), Comprehensive Pneumology Center (CPC), Helmholtz Zentrum Munich, Member of the German Center for Lung Research (DZL), Munich, Germany.,Stem Cell Centre, Lund University, Lund, Sweden.,Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Deniz A Bölükbas
- Lung Bioengineering and Regeneration, Dept of Experimental Medical Sciences, Lund University, Lund, Sweden.,Stem Cell Centre, Lund University, Lund, Sweden.,Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Sandra Lindstedt
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden.,Dept of Cardiothoracic Surgery, Heart and Lung Transplantation, Lund University Hospital, Lund, Sweden
| | - Darcy E Wagner
- Lung Bioengineering and Regeneration, Dept of Experimental Medical Sciences, Lund University, Lund, Sweden .,Lung Repair and Regeneration (LRR), Comprehensive Pneumology Center (CPC), Helmholtz Zentrum Munich, Member of the German Center for Lung Research (DZL), Munich, Germany.,Stem Cell Centre, Lund University, Lund, Sweden.,Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| |
Collapse
|
82
|
Large-scale production of megakaryocytes in microcarrier-supported stirred suspension bioreactors. Sci Rep 2018; 8:10146. [PMID: 29977045 PMCID: PMC6033877 DOI: 10.1038/s41598-018-28459-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 06/21/2018] [Indexed: 01/18/2023] Open
Abstract
Megakaryocytes (MKs) are the precursors of platelets (PLTs) and may be used for PLT production in vivo or in vitro, as well as a source for PLT-derived growth factors. Induced pluripotent stem cells represent an unlimited cell source for the in vitro production of MKs. This study aimed at developing an effective, xeno-free and scalable system to produce high numbers of MKs. In particular, microcarrier beads-assisted stirred bioreactors were evaluated as a means of improving MK yields. This method resulted in the production of 18.7 × 107 MKs per 50 ml medium. Laminin-coated microcarriers increased MK production per iPSC by up to 10-fold. MKs obtained in this system showed typical features of mature MKs and were able to produce PLTs in vitro and in vivo. To increase safety, MKs produced in the bioreactors were irradiated; a procedure that did not affect their capability to form proPLTs and PTLs after transfusion. In vitro generated MKs represent a promising alternative to donor PLTs and open the possibility for the development of innovative MK-based cell therapies.
Collapse
|
83
|
Oikonomopoulos A, Kitani T, Wu JC. Pluripotent Stem Cell-Derived Cardiomyocytes as a Platform for Cell Therapy Applications: Progress and Hurdles for Clinical Translation. Mol Ther 2018; 26:1624-1634. [PMID: 29699941 PMCID: PMC6035734 DOI: 10.1016/j.ymthe.2018.02.026] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 02/26/2018] [Accepted: 02/26/2018] [Indexed: 12/17/2022] Open
Abstract
Cardiovascular diseases are the leading cause of morbidity and mortality worldwide. Regenerative therapy has been applied to restore lost cardiac muscle and cardiac performance. Induced pluripotent stem cells (iPSCs) can provide an unlimited source of cardiomyocytes and therefore play a key role in cardiac regeneration. Despite initial encouraging results from pre-clinical studies, progress toward clinical applications has been hampered by issues such as tumorigenesis, arrhythmogenesis, immune rejection, scalability, low graft-cell survival, and poor engraftment. Here, we review recent developments in iPSC research on regenerating injured heart tissue, including novel advances in cell therapy and potential strategies to overcome current obstacles in the field.
Collapse
Affiliation(s)
- Angelos Oikonomopoulos
- Stanford Cardiovascular Institute, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA 94305, USA; Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Tomoya Kitani
- Stanford Cardiovascular Institute, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA 94305, USA; Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA 94305, USA; Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
84
|
Towards Multi-Organoid Systems for Drug Screening Applications. Bioengineering (Basel) 2018; 5:bioengineering5030049. [PMID: 29933623 PMCID: PMC6163436 DOI: 10.3390/bioengineering5030049] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 06/15/2018] [Accepted: 06/19/2018] [Indexed: 12/13/2022] Open
Abstract
A low percentage of novel drug candidates succeed and reach the end of the drug discovery pipeline, mainly due to poor initial screening and assessment of the effects of the drug and its metabolites over various tissues in the human body. For that, emerging technologies involving the production of organoids from human pluripotent stem cells (hPSCs) and the use of organ-on-a-chip devices are showing great promise for developing a more reliable, rapid and cost-effective drug discovery process when compared with the current use of animal models. In particular, the possibility of virtually obtaining any type of cell within the human body, in combination with the ability to create patient-specific tissues using human induced pluripotent stem cells (hiPSCs), broadens the horizons in the fields of drug discovery and personalized medicine. In this review, we address the current progress and challenges related to the process of obtaining organoids from different cell lineages emerging from hPSCs, as well as how to create devices that will allow a precise examination of the in vitro effects generated by potential drugs in different organ systems.
Collapse
|
85
|
Brandão KO, Tabel VA, Atsma DE, Mummery CL, Davis RP. Human pluripotent stem cell models of cardiac disease: from mechanisms to therapies. Dis Model Mech 2018; 10:1039-1059. [PMID: 28883014 PMCID: PMC5611968 DOI: 10.1242/dmm.030320] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
It is now a decade since human induced pluripotent stem cells (hiPSCs) were first described. The reprogramming of adult somatic cells to a pluripotent state has become a robust technology that has revolutionised our ability to study human diseases. Crucially, these cells capture all the genetic aspects of the patient from which they were derived. Combined with advances in generating the different cell types present in the human heart, this has opened up new avenues to study cardiac disease in humans and investigate novel therapeutic approaches to treat these pathologies. Here, we provide an overview of the current state of the field regarding the generation of cardiomyocytes from human pluripotent stem cells and methods to assess them functionally, an essential requirement when investigating disease and therapeutic outcomes. We critically evaluate whether treatments suggested by these in vitro models could be translated to clinical practice. Finally, we consider current shortcomings of these models and propose methods by which they could be further improved.
Collapse
Affiliation(s)
- Karina O Brandão
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Viola A Tabel
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Douwe E Atsma
- Department of Cardiology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Christine L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Richard P Davis
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| |
Collapse
|
86
|
Christoffersson J, Meier F, Kempf H, Schwanke K, Coffee M, Beilmann M, Zweigerdt R, Mandenius CF. A Cardiac Cell Outgrowth Assay for Evaluating Drug Compounds Using a Cardiac Spheroid-on-a-Chip Device. Bioengineering (Basel) 2018; 5:bioengineering5020036. [PMID: 29734702 PMCID: PMC6027518 DOI: 10.3390/bioengineering5020036] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 04/23/2018] [Accepted: 05/01/2018] [Indexed: 01/09/2023] Open
Abstract
Three-dimensional (3D) models with cells arranged in clusters or spheroids have emerged as valuable tools to improve physiological relevance in drug screening. One of the challenges with cells cultured in 3D, especially for high-throughput applications, is to quickly and non-invasively assess the cellular state in vitro. In this article, we show that the number of cells growing out from human induced pluripotent stem cell (hiPSC)-derived cardiac spheroids can be quantified to serve as an indicator of a drug’s effect on spheroids captured in a microfluidic device. Combining this spheroid-on-a-chip with confocal high content imaging reveals easily accessible, quantitative outgrowth data. We found that effects on outgrowing cell numbers correlate to the concentrations of relevant pharmacological compounds and could thus serve as a practical readout to monitor drug effects. Here, we demonstrate the potential of this semi-high-throughput “cardiac cell outgrowth assay” with six compounds at three concentrations applied to spheroids for 48 h. The image-based readout complements end-point assays or may be used as a non-invasive assay for quality control during long-term culture.
Collapse
Affiliation(s)
- Jonas Christoffersson
- Division of Biotechnology, Department of Physics, Chemistry and Biology (IFM), Linköping University, 58183 Linköping, Sweden.
| | - Florian Meier
- Boehringer Ingelheim Pharma GmbH and Co. KG, Nonclinical Drug Safety Germany, D-88397 Biberach an der Riss, Germany.
| | - Henning Kempf
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany.
| | - Kristin Schwanke
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany.
| | - Michelle Coffee
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany.
| | - Mario Beilmann
- Boehringer Ingelheim Pharma GmbH and Co. KG, Nonclinical Drug Safety Germany, D-88397 Biberach an der Riss, Germany.
| | - Robert Zweigerdt
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany.
| | - Carl-Fredrik Mandenius
- Division of Biotechnology, Department of Physics, Chemistry and Biology (IFM), Linköping University, 58183 Linköping, Sweden.
| |
Collapse
|
87
|
Koch L, Deiwick A, Franke A, Schwanke K, Haverich A, Zweigerdt R, Chichkov B. Laser bioprinting of human induced pluripotent stem cells—the effect of printing and biomaterials on cell survival, pluripotency, and differentiation. Biofabrication 2018; 10:035005. [DOI: 10.1088/1758-5090/aab981] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
88
|
Dunn KK, Palecek SP. Engineering Scalable Manufacturing of High-Quality Stem Cell-Derived Cardiomyocytes for Cardiac Tissue Repair. Front Med (Lausanne) 2018; 5:110. [PMID: 29740580 PMCID: PMC5928319 DOI: 10.3389/fmed.2018.00110] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 04/03/2018] [Indexed: 12/29/2022] Open
Abstract
Recent advances in the differentiation and production of human pluripotent stem cell (hPSC)-derived cardiomyocytes (CMs) have stimulated development of strategies to use these cells in human cardiac regenerative therapies. A prerequisite for clinical trials and translational implementation of hPSC-derived CMs is the ability to manufacture safe and potent cells on the scale needed to replace cells lost during heart disease. Current differentiation protocols generate fetal-like CMs that exhibit proarrhythmogenic potential. Sufficient maturation of these hPSC-derived CMs has yet to be achieved to allow these cells to be used as a regenerative medicine therapy. Insights into the native cardiac environment during heart development may enable engineering of strategies that guide hPSC-derived CMs to mature. Specifically, considerations must be made in regard to developing methods to incorporate the native intercellular interactions and biomechanical cues into hPSC-derived CM production that are conducive to scale-up.
Collapse
Affiliation(s)
- Kaitlin K Dunn
- University of Wisconsin-Madison, Chemical and Biological Engineering, Madison, WI, United States
| | - Sean P Palecek
- University of Wisconsin-Madison, Chemical and Biological Engineering, Madison, WI, United States
| |
Collapse
|
89
|
Olmer R, Engels L, Usman A, Menke S, Malik MNH, Pessler F, Göhring G, Bornhorst D, Bolten S, Abdelilah-Seyfried S, Scheper T, Kempf H, Zweigerdt R, Martin U. Differentiation of Human Pluripotent Stem Cells into Functional Endothelial Cells in Scalable Suspension Culture. Stem Cell Reports 2018; 10:1657-1672. [PMID: 29681541 PMCID: PMC5995343 DOI: 10.1016/j.stemcr.2018.03.017] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 03/20/2018] [Accepted: 03/20/2018] [Indexed: 12/11/2022] Open
Abstract
Endothelial cells (ECs) are involved in a variety of cellular responses. As multifunctional components of vascular structures, endothelial (progenitor) cells have been utilized in cellular therapies and are required as an important cellular component of engineered tissue constructs and in vitro disease models. Although primary ECs from different sources are readily isolated and expanded, cell quantity and quality in terms of functionality and karyotype stability is limited. ECs derived from human induced pluripotent stem cells (hiPSCs) represent an alternative and potentially superior cell source, but traditional culture approaches and 2D differentiation protocols hardly allow for production of large cell numbers. Aiming at the production of ECs, we have developed a robust approach for efficient endothelial differentiation of hiPSCs in scalable suspension culture. The established protocol results in relevant numbers of ECs for regenerative approaches and industrial applications that show in vitro proliferation capacity and a high degree of chromosomal stability. Efficient generation of hiPSC-derived ECs in scalable suspension culture High degree of chromosomal stability of hiPSC-ECs after in vitro expansion Generation of relevant numbers of hiPSC-ECs for regenerative approaches
Collapse
Affiliation(s)
- Ruth Olmer
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG), Hannover Medical School, Carl-Neuberg-Str.1, 30625 Hannover, Germany; REBIRTH-Cluster of Excellence, Hannover Medical School, 30625 Hannover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), 30625 Hannover, Germany
| | - Lena Engels
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG), Hannover Medical School, Carl-Neuberg-Str.1, 30625 Hannover, Germany; REBIRTH-Cluster of Excellence, Hannover Medical School, 30625 Hannover, Germany
| | - Abdulai Usman
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG), Hannover Medical School, Carl-Neuberg-Str.1, 30625 Hannover, Germany; REBIRTH-Cluster of Excellence, Hannover Medical School, 30625 Hannover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), 30625 Hannover, Germany
| | - Sandra Menke
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG), Hannover Medical School, Carl-Neuberg-Str.1, 30625 Hannover, Germany; REBIRTH-Cluster of Excellence, Hannover Medical School, 30625 Hannover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), 30625 Hannover, Germany
| | - Muhammad Nasir Hayat Malik
- TWINCORE Centre for Experimental and Clinical Infection Research, 30625 Hannover, Germany; Helmholtz-Centre for Infection Research Braunschweig, 38124 Braunschweig, Germany; Centre for Individualised Infection Medicine, 30625 Hannover, Germany
| | - Frank Pessler
- TWINCORE Centre for Experimental and Clinical Infection Research, 30625 Hannover, Germany; Helmholtz-Centre for Infection Research Braunschweig, 38124 Braunschweig, Germany; Centre for Individualised Infection Medicine, 30625 Hannover, Germany
| | - Gudrun Göhring
- Institute of Cell and Molecular Pathology, Hannover Medical School, 30625 Hannover, Germany
| | - Dorothee Bornhorst
- REBIRTH-Cluster of Excellence, Hannover Medical School, 30625 Hannover, Germany; Institute of Molecular Biology, Hannover Medical School, 30625 Hannover, Germany
| | - Svenja Bolten
- REBIRTH-Cluster of Excellence, Hannover Medical School, 30625 Hannover, Germany; Institute of Biochemistry and Biology, Potsdam University, Karl-Liebknecht-Straße 24-25, 14476 Potsdam, Germany
| | - Salim Abdelilah-Seyfried
- Institute of Molecular Biology, Hannover Medical School, 30625 Hannover, Germany; Institute of Biochemistry and Biology, Potsdam University, Karl-Liebknecht-Straße 24-25, 14476 Potsdam, Germany
| | - Thomas Scheper
- REBIRTH-Cluster of Excellence, Hannover Medical School, 30625 Hannover, Germany; Institute for Technical Chemistry, Leibniz University Hannover, 30167 Hannover, Germany
| | - Henning Kempf
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG), Hannover Medical School, Carl-Neuberg-Str.1, 30625 Hannover, Germany; REBIRTH-Cluster of Excellence, Hannover Medical School, 30625 Hannover, Germany
| | - Robert Zweigerdt
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG), Hannover Medical School, Carl-Neuberg-Str.1, 30625 Hannover, Germany; REBIRTH-Cluster of Excellence, Hannover Medical School, 30625 Hannover, Germany
| | - Ulrich Martin
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG), Hannover Medical School, Carl-Neuberg-Str.1, 30625 Hannover, Germany; REBIRTH-Cluster of Excellence, Hannover Medical School, 30625 Hannover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), 30625 Hannover, Germany.
| |
Collapse
|
90
|
Richardson TC, Mathew S, Candiello JE, Goh SK, Kumta PN, Banerjee I. Development of an Alginate Array Platform to Decouple the Effect of Multiparametric Perturbations on Human Pluripotent Stem Cells During Pancreatic Differentiation. Biotechnol J 2018; 13. [DOI: 10.1002/biot.201700099] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 12/09/2017] [Indexed: 11/11/2022]
Affiliation(s)
- Thomas C. Richardson
- Department of Chemical and Petroleum Engineering; University of Pittsburgh; Pittsburgh USA
| | - Shibin Mathew
- Department of Chemical and Petroleum Engineering; University of Pittsburgh; Pittsburgh USA
| | | | - Saik K. Goh
- Department of Bioengineering; University of Pittsburgh; Pittsburgh USA
| | - Prashant N. Kumta
- Department of Bioengineering, Chemical and Petroleum Engineering; Mechanical Engineering and Materials Science; University of Pittsburgh; Pittsburgh USA
| | - Ipsita Banerjee
- Department of Chemical and Petroleum Engineering; University of Pittsburgh; Pittsburgh USA
| |
Collapse
|
91
|
Musunuru K, Sheikh F, Gupta RM, Houser SR, Maher KO, Milan DJ, Terzic A, Wu JC. Induced Pluripotent Stem Cells for Cardiovascular Disease Modeling and Precision Medicine: A Scientific Statement From the American Heart Association. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2018; 11:e000043. [PMID: 29874173 PMCID: PMC6708586 DOI: 10.1161/hcg.0000000000000043] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Induced pluripotent stem cells (iPSCs) offer an unprece-dented opportunity to study human physiology and disease at the cellular level. They also have the potential to be leveraged in the practice of precision medicine, for example, personalized drug testing. This statement comprehensively describes the provenance of iPSC lines, their use for cardiovascular disease modeling, their use for precision medicine, and strategies through which to promote their wider use for biomedical applications. Human iPSCs exhibit properties that render them uniquely qualified as model systems for studying human diseases: they are of human origin, which means they carry human genomes; they are pluripotent, which means that in principle, they can be differentiated into any of the human body's somatic cell types; and they are stem cells, which means they can be expanded from a single cell into millions or even billions of cell progeny. iPSCs offer the opportunity to study cells that are genetically matched to individual patients, and genome-editing tools allow introduction or correction of genetic variants. Initial progress has been made in using iPSCs to better understand cardiomyopathies, rhythm disorders, valvular and vascular disorders, and metabolic risk factors for ischemic heart disease. This promising work is still in its infancy. Similarly, iPSCs are only just starting to be used to identify the optimal medications to be used in patients from whom the cells were derived. This statement is intended to (1) summarize the state of the science with respect to the use of iPSCs for modeling of cardiovascular traits and disorders and for therapeutic screening; (2) identify opportunities and challenges in the use of iPSCs for disease modeling and precision medicine; and (3) outline strategies that will facilitate the use of iPSCs for biomedical applications. This statement is not intended to address the use of stem cells as regenerative therapy, such as transplantation into the body to treat ischemic heart disease or heart failure.
Collapse
|
92
|
Correia C, Koshkin A, Duarte P, Hu D, Carido M, Sebastião MJ, Gomes-Alves P, Elliott DA, Domian IJ, Teixeira AP, Alves PM, Serra M. 3D aggregate culture improves metabolic maturation of human pluripotent stem cell derived cardiomyocytes. Biotechnol Bioeng 2017; 115:630-644. [PMID: 29178315 DOI: 10.1002/bit.26504] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 09/21/2017] [Accepted: 11/24/2017] [Indexed: 12/13/2022]
Abstract
Three-dimensional (3D) cultures of human pluripotent stem cell derived cardiomyocytes (hPSC-CMs) hold great promise for drug discovery, providing a better approximation to the in vivo physiology over standard two-dimensional (2D) monolayer cultures. However, the transition of CM differentiation protocols from 2D to 3D cultures is not straightforward. In this work, we relied on the aggregation of hPSC-derived cardiac progenitors and their culture under agitated conditions to generate highly pure cardiomyocyte aggregates. Whole-transcriptome analysis and 13 C-metabolic flux analysis allowed to demonstrate at both molecular and fluxome levels that such 3D culture environment enhances metabolic maturation of hiPSC-CMs. When compared to 2D, 3D cultures of hiPSC-CMs displayed down-regulation of genes involved in glycolysis and lipid biosynthesis and increased expression of genes involved in OXPHOS. Accordingly, 3D cultures of hiPSC-CMs had lower fluxes through glycolysis and fatty acid synthesis and increased TCA-cycle activity. Importantly, we demonstrated that the 3D culture environment reproducibly improved both CM purity and metabolic maturation across different hPSC lines, thereby providing a robust strategy to derive enriched hPSC-CMs with metabolic features closer to that of adult CMs.
Collapse
Affiliation(s)
- Cláudia Correia
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Alexey Koshkin
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Patrícia Duarte
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Dongjian Hu
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts.,Harvard Stem Cell Institute, Cambridge, Massachusetts
| | - Madalena Carido
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Maria J Sebastião
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Patrícia Gomes-Alves
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - David A Elliott
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Australia
| | - Ibrahim J Domian
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts.,Harvard Stem Cell Institute, Cambridge, Massachusetts
| | - Ana P Teixeira
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Paula M Alves
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Margarida Serra
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| |
Collapse
|
93
|
Yamauchi K, Li J, Morikawa K, Liu L, Shirayoshi Y, Nakatsuji N, Elliott DA, Hisatome I, Suemori H. Isolation and characterization of ventricular-like cells derived from NKX2-5 eGFP/w and MLC2v mCherry/w double knock-in human pluripotent stem cells. Biochem Biophys Res Commun 2017; 495:1278-1284. [PMID: 29175323 DOI: 10.1016/j.bbrc.2017.11.133] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 11/20/2017] [Indexed: 12/11/2022]
Abstract
Human pluripotent stem cell (hPSC)-derived cardiomyocytes (CMs) are a promising source for cell transplantation into the damaged heart, which has limited regenerative ability. Many methods have been developed to obtain large amounts of functional CMs from hPSCs for therapeutic applications. However, during the differentiation process, a mixed population of various cardiac cells, including ventricular, atrial, and pacemaker cells, is generated, which hampers the proper functional analysis and evaluation of cell properties. Here, we established NKX2-5eGFP/w and MLC2vmCherry/w hPSC double knock-ins that allow for labeling, tracing, purification, and analysis of the development of ventricular cells from early to late stages. As with the endogenous transcriptional activities of these genes, MLC2v-mCherry expression following NKX2-5-eGFP expression was observed under previously established culture conditions, which mimic the in vivo cardiac developmental process. Patch-clamp and microelectrode array electrophysiological analyses showed that the NKX2-5 and MLC2v double-positive cells possess ventricular-like properties. The results demonstrate that the NKX2-5eGFP/w and MLC2vmCherry/w hPSCs provide a powerful model system to capture region-specific cardiac differentiation from early to late stages. Our study would facilitate subtype-specific cardiac development and functional analysis using the hPSC-derived sources.
Collapse
Affiliation(s)
- Kaori Yamauchi
- Laboratory of Embryonic Stem Cell Research, Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Junjun Li
- Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Yoshida-ushinomiya-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Kumi Morikawa
- Division of Regenerative Medicine and Therapeutics, Department of Genetic Medicine and Regenerative Therapeutics, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Science, Tottori University, 86 Nishi-machi, Yonago 683-8504, Japan
| | - Li Liu
- Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Yoshida-ushinomiya-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Yasuaki Shirayoshi
- Division of Regenerative Medicine and Therapeutics, Department of Genetic Medicine and Regenerative Therapeutics, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Science, Tottori University, 86 Nishi-machi, Yonago 683-8504, Japan
| | - Norio Nakatsuji
- Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Yoshida-ushinomiya-cho, Sakyo-ku, Kyoto 606-8501, Japan; Laboratory of Developmental Epigenome, Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - David A Elliott
- Murdoch Children's Research Institute, The RoyalChildren's Hospital, Parkville, Victoria 3052, Australia
| | - Ichiro Hisatome
- Division of Regenerative Medicine and Therapeutics, Department of Genetic Medicine and Regenerative Therapeutics, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Science, Tottori University, 86 Nishi-machi, Yonago 683-8504, Japan
| | - Hirofumi Suemori
- Laboratory of Embryonic Stem Cell Research, Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan.
| |
Collapse
|
94
|
Sgodda M, Dai Z, Zweigerdt R, Sharma AD, Ott M, Cantz T. A Scalable Approach for the Generation of Human Pluripotent Stem Cell-Derived Hepatic Organoids with Sensitive Hepatotoxicity Features. Stem Cells Dev 2017; 26:1490-1504. [DOI: 10.1089/scd.2017.0023] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Affiliation(s)
- Malte Sgodda
- Research Group Translational Hepatology and Stem Cell Biology, Cluster of Excellence REBIRTH, Hannover Medical School, Hannover, Germany
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Zhen Dai
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
- Junior Research Group MicroRNA in Liver Regeneration, Cluster of Excellence REBIRTH, Hannover Medical School, Hannover, Germany
| | - Robert Zweigerdt
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department for Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Amar Deep Sharma
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
- Junior Research Group MicroRNA in Liver Regeneration, Cluster of Excellence REBIRTH, Hannover Medical School, Hannover, Germany
| | - Michael Ott
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
- Twincore Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - Tobias Cantz
- Research Group Translational Hepatology and Stem Cell Biology, Cluster of Excellence REBIRTH, Hannover Medical School, Hannover, Germany
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
- Max Planck Institute for Molecular Biomedicine, Cell and Developmental Biology, Münster, Germany
| |
Collapse
|
95
|
Kropp C, Massai D, Zweigerdt R. Progress and challenges in large-scale expansion of human pluripotent stem cells. Process Biochem 2017. [DOI: 10.1016/j.procbio.2016.09.032] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
96
|
Future Challenges in the Generation of Hepatocyte-Like Cells From Human Pluripotent Stem Cells. CURRENT PATHOBIOLOGY REPORTS 2017. [DOI: 10.1007/s40139-017-0150-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
97
|
Massai D, Bolesani E, Diaz DR, Kropp C, Kempf H, Halloin C, Martin U, Braniste T, Isu G, Harms V, Morbiducci U, Dräger G, Zweigerdt R. Sensitivity of human pluripotent stem cells to insulin precipitation induced by peristaltic pump-based medium circulation: considerations on process development. Sci Rep 2017. [PMID: 28638147 PMCID: PMC5479836 DOI: 10.1038/s41598-017-04158-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Controlled large-scale production of human pluripotent stem cells (hPSCs) is indispensable for their envisioned clinical translation. Aiming at advanced process development in suspension culture, the sensitivity of hPSC media to continuous peristaltic pump-based circulation, a well-established technology extensively used in hydraulically-driven bioreactors, was investigated. Unexpectedly, conditioning of low protein media (i.e. E8 and TeSR-E8) in a peristaltic pump circuit induced severe viability loss of hPSCs cultured as aggregates in suspension. Optical, biochemical, and cytological analyses of the media revealed that the applied circulation mode resulted in the reduction of the growth hormone insulin by precipitation of micro-sized particles. Notably, in contrast to insulin depletion, individual withdrawal of other medium protein components (i.e. bFGF, TGFβ1 or transferrin) provoked minor reduction of hPSC viability, if any. Supplementation of the surfactant glycerol or the use of the insulin analogue Aspart did not overcome the issue of insulin precipitation. In contrast, the presence of bovine or human serum albumin (BSA or HSA, respectively) stabilized insulin rescuing its content, possibly by acting as molecular chaperone-like protein, ultimately supporting hPSC maintenance. This study highlights the potential and the requirement of media optimization for automated hPSC processing and has broad implications on media development and bioreactor-based technologies.
Collapse
Affiliation(s)
- Diana Massai
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.,REBIRTH-Cluster of Excellence, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Emiliano Bolesani
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.,REBIRTH-Cluster of Excellence, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Diana Robles Diaz
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.,REBIRTH-Cluster of Excellence, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Christina Kropp
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.,REBIRTH-Cluster of Excellence, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Henning Kempf
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.,REBIRTH-Cluster of Excellence, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Caroline Halloin
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.,REBIRTH-Cluster of Excellence, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Ulrich Martin
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.,REBIRTH-Cluster of Excellence, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Tudor Braniste
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.,National Center for Materials Study and Testing, Technical University of Moldova, Bv. Stefan cel Mare 168, Chisinau, 2004, Republic of Moldova
| | - Giuseppe Isu
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129, Torino, Italy.,Department of Biomedicine, University of Basel and Department of Surgery, University Hospital of Basel, 4031, Basel, Switzerland
| | - Vanessa Harms
- Institute of Organic Chemistry, Leibniz University Hannover, Schneiderberg 1B, 30167, Hannover, Germany
| | - Umberto Morbiducci
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129, Torino, Italy
| | - Gerald Dräger
- REBIRTH-Cluster of Excellence, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.,Institute of Organic Chemistry, Leibniz University Hannover, Schneiderberg 1B, 30167, Hannover, Germany
| | - Robert Zweigerdt
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany. .,REBIRTH-Cluster of Excellence, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| |
Collapse
|
98
|
Martin U, Haverich A. Myokardiales Tissue-Engineering. ZEITSCHRIFT FUR HERZ THORAX UND GEFASSCHIRURGIE 2017. [DOI: 10.1007/s00398-016-0119-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
99
|
Skelton RJP, Kamp TJ, Elliott DA, Ardehali R. Biomarkers of Human Pluripotent Stem Cell-Derived Cardiac Lineages. Trends Mol Med 2017; 23:651-668. [PMID: 28576602 DOI: 10.1016/j.molmed.2017.05.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 04/24/2017] [Accepted: 05/04/2017] [Indexed: 02/07/2023]
Abstract
Human pluripotent stem cells (hPSCs) offer a practical source for the de novo generation of cardiac tissues and a unique opportunity to investigate cardiovascular lineage commitment. Numerous strategies have focused on the in vitro production of cardiomyocytes, smooth muscle, and endothelium from hPSCs. However, these differentiation protocols often yield undesired cell types. Thus, establishing a set of stage-specific markers for pure cardiac subpopulations will assist in defining the hierarchy of cardiac differentiation, aid in the development of cellular therapy, and facilitate drug screening and disease modeling. The recent characterization of many such markers is enabling the isolation of major cardiac lineages and subpopulations from differentiating hPSCs. We provide here a comprehensive review detailing the suite of biomarkers used to differentiate cardiac lineages from mixed hPSC-derived populations.
Collapse
Affiliation(s)
- Rhys J P Skelton
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; Eli and Edythe Broad Stem Cell Research Center, University of California, Los Angeles, CA 90095, USA
| | - Timothy J Kamp
- Department of Medicine, Division of Cardiovascular Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - David A Elliott
- Murdoch Childrens Research Institute, The Royal Children's Hospital, Parkville, Victoria 3052, Australia
| | - Reza Ardehali
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; Eli and Edythe Broad Stem Cell Research Center, University of California, Los Angeles, CA 90095, USA.
| |
Collapse
|
100
|
Abstract
Since the advent of the generation of human induced pluripotent stem cells (hiPSCs), numerous protocols have been developed to differentiate hiPSCs into cardiomyocytes and then subsequently assess their ability to recapitulate the properties of adult human cardiomyocytes. However, hiPSC-derived cardiomyocytes (hiPSC-CMs) are often assessed in single-cell assays. A shortcoming of these assays is the limited ability to characterize the physiological parameters of cardiomyocytes, such as contractile force, due to random orientations. This protocol describes the differentiation of cardiomyocytes from hiPSCs, which occurs within 14 d. After casting, cardiomyocytes undergo 3D assembly. This produces fibrin-based engineered heart tissues (EHTs)-in a strip format-that generate force under auxotonic stretch conditions. 10-15 d after casting, the EHTs can be used for contractility measurements. This protocol describes parallel expansion of hiPSCs; standardized generation of defined embryoid bodies, growth factor and small-molecule-based cardiac differentiation; and standardized generation of EHTs. To carry out the protocol, experience in advanced cell culture techniques is required.
Collapse
|