1
|
Hashemi M, Finklea FB, Hammons H, Tian Y, Young N, Kim E, Halloin C, Triebert W, Zweigerdt R, Mitra AK, Lipke EA. Hydrogel microsphere stem cell encapsulation enhances cardiomyocyte differentiation and functionality in scalable suspension system. Bioact Mater 2025; 43:423-440. [PMID: 39399838 PMCID: PMC11471139 DOI: 10.1016/j.bioactmat.2024.08.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 08/30/2024] [Accepted: 08/31/2024] [Indexed: 10/15/2024] Open
Abstract
A reliable suspension-based platform for scaling engineered cardiac tissue (ECT) production from human induced pluripotent stem cells (hiPSCs) is crucial for regenerative therapies. Here, we compared the production and functionality of ECTs formed using our scaffold-based, engineered tissue microsphere differentiation approach with those formed using the prevalent scaffold-free aggregate platform. We utilized a microfluidic system for the rapid (1 million cells/min), high density (30, 40, 60 million cells/ml) encapsulation of hiPSCs within PEG-fibrinogen hydrogel microspheres. HiPSC-laden microspheres and aggregates underwent suspension-based cardiac differentiation in chemically defined media. In comparison to aggregates, microspheres maintained consistent size and shape initially, over time, and within and between batches. Initial size and shape coefficients of variation for microspheres were eight and three times lower, respectively, compared to aggregates. On day 10, microsphere cardiomyocyte (CM) content was 27 % higher and the number of CMs per initial hiPSC was 250 % higher than in aggregates. Contraction and relaxation velocities of microspheres were four and nine times higher than those of aggregates, respectively. Microsphere contractile functionality also improved with culture time, whereas aggregate functionality remained unchanged. Additionally, microspheres displayed improved β-adrenergic signaling responsiveness and uniform calcium transient propagation. Transcriptomic analysis revealed that while both microspheres and aggregates demonstrated similar gene regulation patterns associated with cardiomyocyte differentiation, heart development, cardiac muscle contraction, and sarcomere organization, the microspheres exhibited more pronounced transcriptional changes over time. Taken together, these results highlight the capability of the microsphere platform for scaling up biomanufacturing of ECTs in a suspension-based culture platform.
Collapse
Affiliation(s)
| | - Ferdous B. Finklea
- Department of Chemical Engineering, Auburn University, Auburn, AL, United States
| | - Hanna Hammons
- Department of Chemical Engineering, Auburn University, Auburn, AL, United States
| | - Yuan Tian
- Department of Chemical Engineering, Auburn University, Auburn, AL, United States
| | - Nathan Young
- Department of Chemical Engineering, Auburn University, Auburn, AL, United States
| | - Emma Kim
- Department of Chemical Engineering, Auburn University, Auburn, AL, United States
| | - Caroline Halloin
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hanover, Germany
| | - Wiebke Triebert
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hanover, Germany
| | - Robert Zweigerdt
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hanover, Germany
| | - Amit Kumar Mitra
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL, United States
| | - Elizabeth A. Lipke
- Department of Chemical Engineering, Auburn University, Auburn, AL, United States
| |
Collapse
|
2
|
Septiana WL, Pawitan JA. Potential Use of Organoids in Regenerative Medicine. Tissue Eng Regen Med 2024; 21:1125-1139. [PMID: 39412646 PMCID: PMC11589048 DOI: 10.1007/s13770-024-00672-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 08/30/2024] [Accepted: 09/05/2024] [Indexed: 11/26/2024] Open
Abstract
BACKGROUND In vitro cell culture is crucial for studying human diseases and development. Compared to traditional monolayer cultures, 3D culturing with organoids offers significant advantages by more accurately replicating natural tissues' structural and functional features. This advancement enhances disease modeling, drug testing, and regenerative medicine applications. Organoids, derived from stem cells, mimic tissue physiology in a more relevant manner. Despite their promise, the clinical use of regenerative medicine currently needs to be improved by reproducibility, scalability, and maturation issues. METHODS This article overviews recent organoid research, focusing on their types, sources, 3D culturing methods, and applications in regenerative medicine. A literature review of "organoid" and "regenerative medicine" in PubMed/MEDLINE highlighted relevant studies published over the past decade, emphasizing human-sourced organoids and their regenerative benefits, as well as the availability of free full-text articles. The review uses descriptive data, including tables and text, to illustrate the challenges and potential of organoids in regenerative medicine. RESULTS The transition from 2D to 3D models, particularly organoids, has significantly advanced in vitro research. This review covers a decade of progress in various organoid types-such as liver, cholangiocyte, intestinal, pancreatic, cardiac, brain, thymus, and mammary organoids-and their 3D culture methods and applications. It addresses critical issues of maturity, scalability, and reproducibility and underscores the need for standardization and improved production techniques to facilitate broader clinical applications in regenerative medicine. CONCLUSIONS Successful therapy requires increased scalability and standardization. Organoids have enormous potential in biological research, notwithstanding obstacles.
Collapse
Affiliation(s)
- Wahyunia L Septiana
- Department of Histology Faculty of Medicine, Gunadarma University, Depok, Indonesia.
| | - Jeanne A Pawitan
- Department of Histology Faculty of Medicine,, Universitas Indonesia, Jakarta, Indonesia
- Stem Cell and Tissue Engineering Research Center (SCTE) IMERI, Jakarta, Indonesia
| |
Collapse
|
3
|
Heinzelmann E, Piraino F, Costa M, Roch A, Norkin M, Garnier V, Homicsko K, Brandenberg N. iPSC-derived and Patient-Derived Organoids: Applications and challenges in scalability and reproducibility as pre-clinical models. Curr Res Toxicol 2024; 7:100197. [PMID: 40276485 PMCID: PMC12020925 DOI: 10.1016/j.crtox.2024.100197] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/20/2024] [Accepted: 09/26/2024] [Indexed: 04/26/2025] Open
Abstract
Recent advancements in stem cell technology have led to the development of organoids - three-dimensional (3D) cell cultures that closely mimic the structural and functional characteristics of human organs. These organoids represent a significant improvement over traditional two-dimensional (2D) cell cultures by preserving native tissue architecture and cellular interactions critical for physiological relevance. This review provides a comprehensive comparison between two main types of organoids: induced Pluripotent Stem Cell (iPSC)-derived and Adult Stem Cell (ASC)-derived (also known as Patient-Derived Organoids, PDOs). iPSC-derived organoids, derived from reprogrammed cells, exhibit remarkable plasticity, and can model a wide range of tissues and developmental stages. They are particularly valuable for studying early human development, genetic disorders, and complex diseases. However, challenges such as prolonged differentiation protocols and variability in maturation levels remain significant hurdles. In contrast, ASC-derived organoids, generated directly from patient tissues, faithfully recapitulate tissue-specific characteristics and disease phenotypes. This fidelity makes them indispensable for personalized medicine applications, including drug screening, disease modeling, and understanding individualized treatment responses. The review highlights the unique advantages and limitations of each organoid type, emphasizing their roles in advancing biomedical research and drug discovery. It addresses key challenges in organoid technology, such as scalability, reproducibility, and the need for standardized culture protocols. Furthermore, it explores recent innovations in scaffold-guided organoid engineering and the integration of organoids with advanced technologies like artificial intelligence and high-throughput screening. The integration of organoids with cutting-edge technologies holds promise for enhancing their utility in modeling complex human diseases and accelerating drug discovery and development. By providing more physiologically relevant models of human organs, organoid technology is poised to revolutionize biomedical research, offering new insights into disease mechanisms and personalized therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | | | - Maxim Norkin
- Department of Oncology, CHUV, Lausanne, Switzerland
| | | | | | | |
Collapse
|
4
|
Zheng H, Harcum SW, Pei J, Xie W. Stochastic biological system-of-systems modelling for iPSC culture. Commun Biol 2024; 7:39. [PMID: 38191636 PMCID: PMC10774284 DOI: 10.1038/s42003-023-05653-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 11/30/2023] [Indexed: 01/10/2024] Open
Abstract
Large-scale manufacturing of induced pluripotent stem cells (iPSCs) is essential for cell therapies and regenerative medicines. Yet, iPSCs form large cell aggregates in suspension bioreactors, resulting in insufficient nutrient supply and extra metabolic waste build-up for the cells located at the core. Since subtle changes in micro-environment can lead to a heterogeneous cell population, a novel Biological System-of-Systems (Bio-SoS) framework is proposed to model cell-to-cell interactions, spatial and metabolic heterogeneity, and cell response to micro-environmental variation. Building on stochastic metabolic reaction network, aggregation kinetics, and reaction-diffusion mechanisms, the Bio-SoS model characterizes causal interdependencies at individual cell, aggregate, and cell population levels. It has a modular design that enables data integration and improves predictions for different monolayer and aggregate culture processes. In addition, a variance decomposition analysis is derived to quantify the impact of factors (i.e., aggregate size) on cell product health and quality heterogeneity.
Collapse
Affiliation(s)
- Hua Zheng
- Mechanical and Industrial Engineering, Northeastern University, Boston, MA, 02115, USA
| | | | - Jinxiang Pei
- Mechanical and Industrial Engineering, Northeastern University, Boston, MA, 02115, USA
| | - Wei Xie
- Mechanical and Industrial Engineering, Northeastern University, Boston, MA, 02115, USA.
| |
Collapse
|
5
|
Wauford N, Patel A, Tordoff J, Enghuus C, Jin A, Toppen J, Kemp ML, Weiss R. Synthetic symmetry breaking and programmable multicellular structure formation. Cell Syst 2023; 14:806-818.e5. [PMID: 37689062 PMCID: PMC10919224 DOI: 10.1016/j.cels.2023.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 04/14/2023] [Accepted: 08/02/2023] [Indexed: 09/11/2023]
Abstract
During development, cells undergo symmetry breaking into differentiated subpopulations that self-organize into complex structures.1,2,3,4,5 However, few tools exist to recapitulate these behaviors in a controllable and coupled manner.6,7,8,9 Here, we engineer a stochastic recombinase genetic switch tunable by small molecules to induce programmable symmetry breaking, commitment to downstream cell fates, and morphological self-organization. Inducers determine commitment probabilities, generating tunable subpopulations as a function of inducer dosage. We use this switch to control the cell-cell adhesion properties of cells committed to each fate.10,11 We generate a wide variety of 3D morphologies from a monoclonal population and develop a computational model showing high concordance with experimental results, yielding new quantitative insights into the relationship between cell-cell adhesion strengths and downstream morphologies. We expect that programmable symmetry breaking, generating precise and tunable subpopulation ratios and coupled to structure formation, will serve as an integral component of the toolbox for complex tissue and organoid engineering.
Collapse
Affiliation(s)
- Noreen Wauford
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Akshay Patel
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jesse Tordoff
- Program in Computational and Systems Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Casper Enghuus
- Department of Microbiology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Andrew Jin
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Jack Toppen
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Melissa L Kemp
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Ron Weiss
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Electrical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
6
|
Abdal Dayem A, Lee SB, Lim KM, Kim A, Shin HJ, Vellingiri B, Kim YB, Cho SG. Bioactive peptides for boosting stem cell culture platform: Methods and applications. Biomed Pharmacother 2023; 160:114376. [PMID: 36764131 DOI: 10.1016/j.biopha.2023.114376] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 02/10/2023] Open
Abstract
Peptides, short protein fragments, can emulate the functions of their full-length native counterparts. Peptides are considered potent recombinant protein alternatives due to their specificity, high stability, low production cost, and ability to be easily tailored and immobilized. Stem cell proliferation and differentiation processes are orchestrated by an intricate interaction between numerous growth factors and proteins and their target receptors and ligands. Various growth factors, functional proteins, and cellular matrix-derived peptides efficiently enhance stem cell adhesion, proliferation, and directed differentiation. For that, peptides can be immobilized on a culture plate or conjugated to scaffolds, such as hydrogels or synthetic matrices. In this review, we assess the applications of a variety of peptides in stem cell adhesion, culture, organoid assembly, proliferation, and differentiation, describing the shortcomings of recombinant proteins and their full-length counterparts. Furthermore, we discuss the challenges of peptide applications in stem cell culture and materials design, as well as provide a brief outlook on future directions to advance peptide applications in boosting stem cell quality and scalability for clinical applications in tissue regeneration.
Collapse
Affiliation(s)
- Ahmed Abdal Dayem
- Department of Stem Cell and Regenerative Biotechnology, KU Convergence Science and Technology Institute, Konkuk University, Seoul 05029, Republic of Korea
| | - Soo Bin Lee
- Department of Stem Cell and Regenerative Biotechnology, KU Convergence Science and Technology Institute, Konkuk University, Seoul 05029, Republic of Korea
| | - Kyung Min Lim
- Department of Stem Cell and Regenerative Biotechnology, KU Convergence Science and Technology Institute, Konkuk University, Seoul 05029, Republic of Korea; R&D Team, StemExOne co., ltd. 303, Life Science Bldg, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Aram Kim
- Department of Urology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul 05029, Republic of Korea; R&D Team, StemExOne co., ltd. 303, Life Science Bldg, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Hyun Jin Shin
- Department of Ophthalmology, Research Institute of Medical Science, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul 05029, Republic of Korea; R&D Team, StemExOne co., ltd. 303, Life Science Bldg, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Balachandar Vellingiri
- Stem cell and Regenerative Medicine/Translational Research, Department of Zoology, School of Basic Sciences, Central University of Punjab (CUPB), Bathinda 151401, Punjab, India
| | - Young Bong Kim
- Department of Biomedical Science & Engineering, KU Convergence Science and Technology Institute, Konkuk University, Seoul 05029, Republic of Korea
| | - Ssang-Goo Cho
- Department of Stem Cell and Regenerative Biotechnology, KU Convergence Science and Technology Institute, Konkuk University, Seoul 05029, Republic of Korea; R&D Team, StemExOne co., ltd. 303, Life Science Bldg, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea.
| |
Collapse
|
7
|
Thanuthanakhun N, Kim MH, Kino-oka M. Cell Behavioral Dynamics as a Cue in Optimizing Culture Stabilization in the Bioprocessing of Pluripotent Stem Cells. Bioengineering (Basel) 2022; 9:669. [PMID: 36354580 PMCID: PMC9687444 DOI: 10.3390/bioengineering9110669] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 10/28/2022] [Accepted: 11/05/2022] [Indexed: 04/23/2024] Open
Abstract
Pluripotent stem cells (PSCs) are important for future regenerative medicine therapies. However, in the production of PSCs and derivatives, the control of culture-induced fluctuations in the outcome of cell quality remains challenging. A detailed mechanistic understanding of how PSC behaviors are altered in response to biomechanical microenvironments within a culture is necessary for rational bioprocessing optimization. In this review, we discuss recent insights into the role of cell behavioral and mechanical homeostasis in modulating the states and functions of PSCs during culture processes. We delineate promising ways to manipulate the culture variability through regulating cell behaviors using currently developed tools. Furthermore, we anticipate their potential implementation for designing a culture strategy based on the concept of Waddington's epigenetic landscape that may provide a feasible solution for tuning the culture quality and stability in the bioprocessing space.
Collapse
Affiliation(s)
- Naruchit Thanuthanakhun
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita 565-0871, Osaka, Japan
| | - Mee-Hae Kim
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita 565-0871, Osaka, Japan
| | - Masahiro Kino-oka
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita 565-0871, Osaka, Japan
- Research Base for Cell Manufacturability, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita 565-0871, Osaka, Japan
| |
Collapse
|
8
|
Lee B, Jung S, Hashimura Y, Lee M, Borys BS, Dang T, Kallos MS, Rodrigues CAV, Silva TP, Cabral JMS. Cell Culture Process Scale-Up Challenges for Commercial-Scale Manufacturing of Allogeneic Pluripotent Stem Cell Products. Bioengineering (Basel) 2022; 9:bioengineering9030092. [PMID: 35324781 PMCID: PMC8945133 DOI: 10.3390/bioengineering9030092] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/10/2022] [Accepted: 02/14/2022] [Indexed: 01/01/2023] Open
Abstract
Allogeneic cell therapy products, such as therapeutic cells derived from pluripotent stem cells (PSCs), have amazing potential to treat a wide variety of diseases and vast numbers of patients globally. However, there are various challenges related to manufacturing PSCs in single-use bioreactors, particularly at larger volumetric scales. This manuscript addresses these challenges and presents potential solutions to alleviate the anticipated bottlenecks for commercial-scale manufacturing of high-quality therapeutic cells derived from PSCs.
Collapse
Affiliation(s)
- Brian Lee
- PBS Biotech, Inc., Camarillo, CA 93012, USA; (S.J.); (Y.H.); (M.L.); (B.S.B.)
- Correspondence:
| | - Sunghoon Jung
- PBS Biotech, Inc., Camarillo, CA 93012, USA; (S.J.); (Y.H.); (M.L.); (B.S.B.)
| | - Yas Hashimura
- PBS Biotech, Inc., Camarillo, CA 93012, USA; (S.J.); (Y.H.); (M.L.); (B.S.B.)
| | - Maximilian Lee
- PBS Biotech, Inc., Camarillo, CA 93012, USA; (S.J.); (Y.H.); (M.L.); (B.S.B.)
| | - Breanna S. Borys
- PBS Biotech, Inc., Camarillo, CA 93012, USA; (S.J.); (Y.H.); (M.L.); (B.S.B.)
- Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, 2500 University Dr. NW, Calgary, AB T2N 1N4, Canada; (T.D.); (M.S.K.)
| | - Tiffany Dang
- Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, 2500 University Dr. NW, Calgary, AB T2N 1N4, Canada; (T.D.); (M.S.K.)
| | - Michael S. Kallos
- Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, 2500 University Dr. NW, Calgary, AB T2N 1N4, Canada; (T.D.); (M.S.K.)
| | - Carlos A. V. Rodrigues
- iBB–Institute for Bioengineering and Biosciences and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal; (C.A.V.R.); (T.P.S.); (J.M.S.C.)
| | - Teresa P. Silva
- iBB–Institute for Bioengineering and Biosciences and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal; (C.A.V.R.); (T.P.S.); (J.M.S.C.)
| | - Joaquim M. S. Cabral
- iBB–Institute for Bioengineering and Biosciences and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal; (C.A.V.R.); (T.P.S.); (J.M.S.C.)
| |
Collapse
|
9
|
Tao T, Deng P, Wang Y, Zhang X, Guo Y, Chen W, Qin J. Microengineered Multi-Organoid System from hiPSCs to Recapitulate Human Liver-Islet Axis in Normal and Type 2 Diabetes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103495. [PMID: 34951149 PMCID: PMC8844474 DOI: 10.1002/advs.202103495] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 11/21/2021] [Indexed: 05/07/2023]
Abstract
Type 2 diabetes mellitus (T2DM) is a systematic multi-organ metabolic disease, which is characterized by the dynamic interplay among different organs. The increasing incidence of T2DM reflects an urgent need for the development of in vitro human-relevant models for disease study and drug therapy. Here, a new microfluidic multi-organoid system is developed that recapitulates the human liver-pancreatic islet axis in normal and disease states. The system contains two compartmentalized regions connected by a microchannel network, enabling 3D co-culture of human induced pluripotent stem cells (hiPSC)-derived liver and islet organoids for up to 30 days under circulatory perfusion conditions. The co-cultured liver and islet organoids exhibit favorable growth and improved tissue-specific functions. Transcriptional analyses reveal the activation of metabolically relevant signaling pathways in the co-cultured organoids. Notably, the co-culture system facilitates sensitive glucose-stimulated insulin secretion from islet organoids and increased glucose utilization in liver organoids by glucose tolerance tests. Both liver and islet organoids display mitochondrial dysfunction and decreased glucose transport capacity under high glucose conditions, which can be alleviated by metformin treatment. This novel multi-organoid system can recapitulate human-relevant liver-islet axis under both physiological and pathological conditions, providing a unique platform for future T2DM research and drug development.
Collapse
Affiliation(s)
- Tingting Tao
- Division of BiotechnologyDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
- University of Chinese Academy of SciencesBeijing100049China
| | - Pengwei Deng
- Division of BiotechnologyDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yaqing Wang
- Division of BiotechnologyDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
| | - Xu Zhang
- Division of BiotechnologyDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
| | - Yaqiong Guo
- Division of BiotechnologyDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
- University of Chinese Academy of SciencesBeijing100049China
| | - Wenwen Chen
- Division of BiotechnologyDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
- University of Chinese Academy of SciencesBeijing100049China
| | - Jianhua Qin
- Division of BiotechnologyDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijing100101China
- CAS Center for Excellence in Brain Science and Intelligence TechnologyChinese Academy of SciencesShanghai200031China
- University of Chinese Academy of SciencesBeijing100049China
| |
Collapse
|
10
|
Clément JP, Al-Alwan L, Glasgow SD, Stolow A, Ding Y, Quevedo Melo T, Khayachi A, Liu Y, Hellmund M, Haag R, Milnerwood AJ, Grütter P, Kennedy TE. Dendritic Polyglycerol Amine: An Enhanced Substrate to Support Long-Term Neural Cell Culture. ASN Neuro 2022; 14:17590914211073276. [PMID: 35023760 PMCID: PMC8784910 DOI: 10.1177/17590914211073276] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Long-term stable cell culture is a critical tool to better understand cell function. Most adherent cell culture models require a polymer substrate coating of poly-lysine or poly-ornithine for the cells to adhere and survive. However, polypeptide-based substrates are degraded by proteolysis and it remains a challenge to maintain healthy cell cultures for extended periods of time. Here, we report the development of an enhanced cell culture substrate based on a coating of dendritic polyglycerol amine (dPGA), a non-protein macromolecular biomimetic of poly-lysine, to promote the adhesion and survival of neurons in cell culture. We show that this new polymer coating provides enhanced survival, differentiation and long-term stability for cultures of primary neurons or neurons derived from human induced pluripotent stem cells (hiPSCs). Atomic force microscopy analysis provides evidence that greater nanoscale roughness contributes to the enhanced capacity of dPGA-coated surfaces to support cells in culture. We conclude that dPGA is a cytocompatible, functionally superior, easy to use, low cost and highly stable alternative to poly-cationic polymer cell culture substrate coatings such as poly-lysine and poly-ornithine.
Summary statement
Here, we describe a novel dendritic polyglycerol amine-based substrate coating, demonstrating superior performance compared to current polymer coatings for long-term culture of primary neurons and neurons derived from induced pluripotent stem cells.
Collapse
Affiliation(s)
- Jean-Pierre Clément
- Program in Neuroengineering, Montreal Neurological Institute, McGill University, Montreal, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute and Hospital, Montreal, Canada
| | - Laila Al-Alwan
- Program in Neuroengineering, Montreal Neurological Institute, McGill University, Montreal, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute and Hospital, Montreal, Canada
| | - Stephen D. Glasgow
- Program in Neuroengineering, Montreal Neurological Institute, McGill University, Montreal, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute and Hospital, Montreal, Canada
| | - Avya Stolow
- Department of Physics, McGill University, Montreal, Canada
| | - Yi Ding
- Department of Physics, McGill University, Montreal, Canada
| | - Thaiany Quevedo Melo
- Department of Neurology and Neurosurgery, Montreal Neurological Institute and Hospital, Montreal, Canada
| | - Anouar Khayachi
- Department of Neurology and Neurosurgery, Montreal Neurological Institute and Hospital, Montreal, Canada
| | - Yumin Liu
- Department of Neurology and Neurosurgery, Montreal Neurological Institute and Hospital, Montreal, Canada
| | - Markus Hellmund
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Rainer Haag
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Austen J Milnerwood
- Department of Neurology and Neurosurgery, Montreal Neurological Institute and Hospital, Montreal, Canada
| | - Peter Grütter
- Department of Physics, McGill University, Montreal, Canada
| | - Timothy E. Kennedy
- Program in Neuroengineering, Montreal Neurological Institute, McGill University, Montreal, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute and Hospital, Montreal, Canada
| |
Collapse
|
11
|
Niari SA, Rahbarghazi R, Geranmayeh MH, Karimipour M. Biomaterials patterning regulates neural stem cells fate and behavior: The interface of biology and material science. J Biomed Mater Res A 2021; 110:725-737. [PMID: 34751503 DOI: 10.1002/jbm.a.37321] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 09/19/2021] [Accepted: 10/06/2021] [Indexed: 11/12/2022]
Abstract
The combination of nanotechnology and stem cell biology is one of the most promising advances in the field of regenerative medicine. This novel combination has widely been utilized in vitro settings in an attempt to develop efficient therapeutic strategies to overcome the limited capacity of the central nervous system (CNS) in replacing degenerating neural cells with functionally normal cells after the onset of acute and chronic neurological disorders. Importantly, biomaterials, not only, enhance the endogenous CNS neurogenesis and plasticity, but also, could provide a desirable supportive microenvironment to harness the full potential of the in vitro expanded neural stem cells (NSCs) for regenerative purposes. Here, first, we discuss how the physical and biochemical properties of biomaterials, such as their stiffness and elasticity, could influence the behavior of NSCs. Then, since the NSCs niche or microenvironment is of fundamental importance in controlling the dynamic destiny of NSCs such as their quiescent and proliferative states, topographical effects of surface diversity in biomaterials, that is, the micro-and nano-patterned surfaces will be discussed in detail. Finally, the influence of biomaterials as artificial microenvironments on the behavior of NSCs through the specific mechanotransduction signaling pathway mediated by focal adhesion formation will be reviewed.
Collapse
Affiliation(s)
- Shabnam Asghari Niari
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Hossein Geranmayeh
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Neurosciences Research Center (NSRC), Imam Reza Medical Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Karimipour
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
12
|
Sen D, Voulgaropoulos A, Keung AJ. Effects of early geometric confinement on the transcriptomic profile of human cerebral organoids. BMC Biotechnol 2021; 21:59. [PMID: 34641840 PMCID: PMC8507123 DOI: 10.1186/s12896-021-00718-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 09/28/2021] [Indexed: 12/21/2022] Open
Abstract
Background Human cerebral organoids (hCO) are attractive systems due to their ability to model important brain regions and transcriptomics of early in vivo brain development. To date, they have been used to understand the effects of genetics and soluble factors on neurodevelopment. Interestingly, one of the main advantages of hCOs are that they provide three dimensionality that better mimics the in vivo environment; yet, despite this central feature it remains unclear how spatial and mechanical properties regulate hCO and neurodevelopment. While biophysical factors such as shape and mechanical forces are known to play crucial roles in stem cell differentiation, embryogenesis and neurodevelopment, much of this work investigated two dimensional systems or relied on correlative observations of native developing tissues in three dimensions. Using hCOs to establish links between spatial factors and neurodevelopment will require the use of new approaches and could reveal fundamental principles of brain organogenesis as well as improve hCOs as an experimental model. Results Here, we investigated the effects of early geometric confinements on transcriptomic changes during hCO differentiation. Using a custom and tunable agarose microwell platform we generated embryoid bodies (EB) of diverse shapes mimicking several structures from embryogenesis and neurodevelopment and then further differentiated those EBs to whole brain hCOs. Our results showed that the microwells did not have negative gross impacts on the ability of the hCOs to differentiate towards neural fates, and there were clear shape dependent effects on neural lineage specification. In particular we observed that non-spherical shapes showed signs of altered neurodevelopmental kinetics and favored the development of medial ganglionic eminence-associated brain regions and cell types over cortical regions. Transcriptomic analysis suggests these mechanotransducive effects may be mediated by integrin and Wnt signaling. Conclusions The findings presented here suggest a role for spatial factors in brain region specification during hCO development. Understanding these spatial patterning factors will not only improve understanding of in vivo development and differentiation, but also provide important handles with which to advance and improve control over human model systems for in vitro applications. Supplementary Information The online version contains supplementary material available at 10.1186/s12896-021-00718-2.
Collapse
Affiliation(s)
- Dilara Sen
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Campus Box 7905, Raleigh, NC, 27695-7905, USA
| | - Alexis Voulgaropoulos
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Campus Box 7905, Raleigh, NC, 27695-7905, USA
| | - Albert J Keung
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Campus Box 7905, Raleigh, NC, 27695-7905, USA.
| |
Collapse
|
13
|
Salem T, Frankman Z, Churko J. Tissue engineering techniques for iPSC derived three-dimensional cardiac constructs. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:891-911. [PMID: 34476988 PMCID: PMC9419978 DOI: 10.1089/ten.teb.2021.0088] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Recent developments in applied developmental physiology have provided well-defined methodologies for producing human stem cell derived cardiomyocytes. Cardiomyocytes produced in this way have become commonplace as cardiac physiology research models. This accessibility has also allowed for the development of tissue engineered human heart constructs for drug screening, surgical intervention, and investigating cardiac pathogenesis. However, cardiac tissue engineering is an interdisciplinary field that involves complex engineering and physiological concepts, which limits its accessibility. This review provides a readable, broad reaching, and thorough discussion of major factors to consider for the development of cardiovascular tissues from stem cell derived cardiomyocytes. This review will examine important considerations in undertaking a cardiovascular tissue engineering project, and will present, interpret, and summarize some of the recent advancements in this field. This includes reviewing different forms of tissue engineered constructs, a discussion on cardiomyocyte sources, and an in-depth discussion of the fabrication and maturation procedures for tissue engineered heart constructs.
Collapse
Affiliation(s)
- Tori Salem
- University of Arizona Medical Center - University Campus, 22165, Cellular and Molecular Medicine, Tucson, Arizona, United States;
| | - Zachary Frankman
- University of Arizona Medical Center - University Campus, 22165, Biomedical Engineering, Tucson, Arizona, United States;
| | - Jared Churko
- University of Arizona Medical Center - University Campus, 22165, 1501 N Campbell RD, SHC 6143, Tucson, Arizona, United States, 85724-5128;
| |
Collapse
|
14
|
Dang T, Borys BS, Kanwar S, Colter J, Worden H, Blatchford A, Croughan MS, Hossan T, Rancourt DE, Lee B, Kallos MS, Jung S. Computational fluid dynamic characterization of vertical‐wheel bioreactors used for effective scale‐up of human induced pluripotent stem cell aggregate culture. CAN J CHEM ENG 2021. [DOI: 10.1002/cjce.24253] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Tiffany Dang
- Pharmaceutical Production Research Facility, Schulich School of Engineering University of Calgary Calgary Alberta Canada
- Biomedical Engineering Graduate Program University of Calgary Calgary Alberta Canada
- Department of Chemical and Petroleum Engineering, Schulich School of Engineering University of Calgary Calgary Alberta Canada
| | - Breanna S. Borys
- Pharmaceutical Production Research Facility, Schulich School of Engineering University of Calgary Calgary Alberta Canada
- Biomedical Engineering Graduate Program University of Calgary Calgary Alberta Canada
- Department of Chemical and Petroleum Engineering, Schulich School of Engineering University of Calgary Calgary Alberta Canada
- PBS Biotech Inc. Camarillo California USA
| | - Shivek Kanwar
- Pharmaceutical Production Research Facility, Schulich School of Engineering University of Calgary Calgary Alberta Canada
- Department of Mechanical and Manufacturing Engineering, Schulich School of Engineering University of Calgary Calgary Alberta Canada
| | - James Colter
- Pharmaceutical Production Research Facility, Schulich School of Engineering University of Calgary Calgary Alberta Canada
- Biomedical Engineering Graduate Program University of Calgary Calgary Alberta Canada
- Department of Chemical and Petroleum Engineering, Schulich School of Engineering University of Calgary Calgary Alberta Canada
| | | | | | | | - Tareq Hossan
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine University of Calgary Calgary Alberta Canada
| | - Derrick E. Rancourt
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine University of Calgary Calgary Alberta Canada
| | - Brian Lee
- PBS Biotech Inc. Camarillo California USA
| | - Michael S. Kallos
- Pharmaceutical Production Research Facility, Schulich School of Engineering University of Calgary Calgary Alberta Canada
- Biomedical Engineering Graduate Program University of Calgary Calgary Alberta Canada
- Department of Chemical and Petroleum Engineering, Schulich School of Engineering University of Calgary Calgary Alberta Canada
| | | |
Collapse
|
15
|
Organoids in modelling infectious diseases. Drug Discov Today 2021; 27:223-233. [PMID: 34418577 DOI: 10.1016/j.drudis.2021.08.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 05/14/2021] [Accepted: 08/13/2021] [Indexed: 12/12/2022]
Abstract
Approaches based on animal and two-dimensional (2D) cell culture models cannot ensure reliable results in modeling novel pathogens or in drug testing in the short term; therefore, there is rising interest in platforms such as organoids. To develop a toolbox that can be used successfully to overcome current issues in modeling various infections, it is essential to provide a framework of recent achievements in applying organoids. Organoids have been used to study viruses, bacteria, and protists that cause, for example, respiratory, gastrointestinal, and liver diseases. Their future as models of infection will be associated with improvements in system complexity, including abilities to model tissue structure, a dynamic microenvironment, and coinfection. Teaser. Organoids are a flexible tool for modelling viral, bacterial and protist infections. They can provide fast and reliable information on the biology of pathogens and in drug screening, and thus have become essential in combatting emerging infectious diseases.
Collapse
|
16
|
Xie AW, Zacharias NA, Binder BYK, Murphy WL. Controlled aggregation enhances immunomodulatory potential of mesenchymal stromal cell aggregates. Stem Cells Transl Med 2021; 10:1184-1201. [PMID: 33818906 PMCID: PMC8284773 DOI: 10.1002/sctm.19-0414] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 02/04/2021] [Accepted: 03/08/2021] [Indexed: 02/06/2023] Open
Abstract
Human mesenchymal stromal cells (MSCs) are promising candidates for cell therapy due to their ease of isolation and expansion and their ability to secrete antiapoptotic, pro-angiogenic, and immunomodulatory factors. Three-dimensional (3D) aggregation "self-activates" MSCs to augment their pro-angiogenic and immunomodulatory potential, but the microenvironmental features and culture parameters that promote optimal MSC immunomodulatory function in 3D aggregates are poorly understood. Here, we generated MSC aggregates via three distinct methods and compared them with regard to their (a) aggregate structure and (b) immunomodulatory phenotype under resting conditions and in response to inflammatory stimulus. Methods associated with fast aggregation kinetics formed aggregates with higher cell packing density and reduced extracellular matrix (ECM) synthesis compared to those with slow aggregation kinetics. While all three methods of 3D aggregation enhanced MSC expression of immunomodulatory factors compared to two-dimensional culture, different aggregation methods modulated cells' temporal expression of these factors. A Design of Experiments approach, in which aggregate size and aggregation kinetics were systematically covaried, identified a significant effect of both parameters on MSCs' ability to regulate immune cells. Compared to small aggregates formed with fast kinetics, large aggregates with slow assembly kinetics were more effective at T-cell suppression and macrophage polarization toward anti-inflammatory phenotypes. Thus, culture parameters including aggregation method, kinetics, and aggregate size influence both the structural properties of aggregates and their paracrine immunomodulatory function. These findings underscore the utility of engineering strategies to control properties of 3D MSC aggregates, which may identify new avenues for optimizing the immunomodulatory function of MSC-based cell therapies.
Collapse
Affiliation(s)
- Angela W. Xie
- Department of Biomedical EngineeringUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Nicholas A. Zacharias
- Department of Biomedical EngineeringUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Bernard Y. K. Binder
- Department of Orthopedics and RehabilitationUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - William L. Murphy
- Department of Biomedical EngineeringUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Department of Orthopedics and RehabilitationUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Department of Materials Science and EngineeringUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| |
Collapse
|
17
|
Silva TP, Sousa-Luís R, Fernandes TG, Bekman EP, Rodrigues CAV, Vaz SH, Moreira LM, Hashimura Y, Jung S, Lee B, Carmo-Fonseca M, Cabral JMS. Transcriptome profiling of human pluripotent stem cell-derived cerebellar organoids reveals faster commitment under dynamic conditions. Biotechnol Bioeng 2021; 118:2781-2803. [PMID: 33871054 DOI: 10.1002/bit.27797] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 03/30/2021] [Accepted: 04/14/2021] [Indexed: 12/14/2022]
Abstract
Human-induced pluripotent stem cells (iPSCs) have great potential for disease modeling. However, generating iPSC-derived models to study brain diseases remains a challenge. In particular, the ability to recapitulate cerebellar development in vitro is still limited. We presented a reproducible and scalable production of cerebellar organoids by using the novel single-use Vertical-Wheel bioreactors, in which functional cerebellar neurons were obtained. Here, we evaluate the global gene expression profiles by RNA sequencing (RNA-seq) across cerebellar differentiation, demonstrating a faster cerebellar commitment in this novel dynamic differentiation protocol. Furthermore, transcriptomic profiles suggest a significant enrichment of extracellular matrix (ECM) in dynamic-derived cerebellar organoids, which can better mimic the neural microenvironment and support a consistent neuronal network. Thus, an efficient generation of organoids with cerebellar identity was achieved for the first time in a continuous process using a dynamic system without the need of organoids encapsulation in ECM-based hydrogels, allowing the possibility of large-scale production and application in high-throughput processes. The presence of factors that favors angiogenesis onset was also detected in dynamic conditions, which can enhance functional maturation of cerebellar organoids. We anticipate that large-scale production of cerebellar organoids may help developing models for drug screening, toxicological tests, and studying pathological pathways involved in cerebellar degeneration.
Collapse
Affiliation(s)
- Teresa P Silva
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Rui Sousa-Luís
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Tiago G Fernandes
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Evguenia P Bekman
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Carlos A V Rodrigues
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Sandra H Vaz
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal, Portugal
| | - Leonilde M Moreira
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | | | | | - Brian Lee
- PBS Biotech, Camarillo, California, USA
| | - Maria Carmo-Fonseca
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Joaquim M S Cabral
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
18
|
Ratri MC, Brilian AI, Setiawati A, Nguyen HT, Soum V, Shin K. Recent Advances in Regenerative Tissue Fabrication: Tools, Materials, and Microenvironment in Hierarchical Aspects. ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202000088] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
- Monica Cahyaning Ratri
- Department of Chemistry and Institute of Biological Interfaces Sogang University Seoul 04107 Republic of Korea
- Department of Chemistry Education Sanata Dharma University Yogyakarta 55281 Indonesia
| | - Albertus Ivan Brilian
- Department of Chemistry and Institute of Biological Interfaces Sogang University Seoul 04107 Republic of Korea
| | - Agustina Setiawati
- Department of Chemistry and Institute of Biological Interfaces Sogang University Seoul 04107 Republic of Korea
- Department of Life Science Sogang University Seoul 04107 Republic of Korea
- Faculty of Pharmacy Sanata Dharma University Yogyakarta 55281 Indonesia
| | - Huong Thanh Nguyen
- Department of Chemistry and Institute of Biological Interfaces Sogang University Seoul 04107 Republic of Korea
| | - Veasna Soum
- Department of Chemistry and Institute of Biological Interfaces Sogang University Seoul 04107 Republic of Korea
| | - Kwanwoo Shin
- Department of Chemistry and Institute of Biological Interfaces Sogang University Seoul 04107 Republic of Korea
| |
Collapse
|
19
|
Ando Y, Okeyo KO, Sunaga J, Adachi T. Edge-localized alteration in pluripotency state of mouse ES cells forming topography-confined layers on designed mesh substrates. Stem Cell Res 2021; 53:102352. [PMID: 33901814 DOI: 10.1016/j.scr.2021.102352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 03/15/2021] [Accepted: 04/09/2021] [Indexed: 10/21/2022] Open
Abstract
Self-organization of pluripotent stem cells during tissue formation is directed by the adhesion microenvironment, which defines the resulting tissue topography. Although the influence of tissue topography on pluripotency state has been inferred, this aspect of self-organization remains largely unexplored. In this study, to determine the effect of self-organized tissue topography on pluripotency loss, we designed novel island mesh substrates to confine the self-organization process of mouse embryonic stem cells, enabling us to generate isolated cell layers with an island-like topography and overhanging edges. Using immunofluorescence microscopy, we determined that cells at the tissue edge exhibited deformed nuclei associated with low OCT3/4, in contrast with cells nested in the tissue interior which had round-shaped nuclei and exhibited sustained OCT3/4 expression. Interestingly, F-actin and phospho-myosin light chain were visibly enriched at the tissue edge where ERK activation and elevated AP-2γ expression were also found to be localized, as determined using both immunofluorescence microscopy and RT-qPCR analysis. Since actomyosin contractility is known to cause ERK activation, these results suggest that mechanical condition at the tissue edge can contribute to loss of pluripotency leading to differentiation. Thus, our study draws attention to the influence of self-organized tissue topography in stem cell culture and differentiation.
Collapse
Affiliation(s)
- Yuta Ando
- Department of Micro Engineering, Graduate School of Engineering, Kyoto University, Kyoto daigaku-katsura, Nishikyo-ku, Kyoto 615-8530, Japan; Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Kennedy Omondi Okeyo
- Department of Micro Engineering, Graduate School of Engineering, Kyoto University, Kyoto daigaku-katsura, Nishikyo-ku, Kyoto 615-8530, Japan; Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan; Division of Systemic Life Science, Graduate School of Biostudies, Kyoto University, Yoshida-Konoecho, Sakyo-ku, Kyoto 606-8501, Japan.
| | - Junko Sunaga
- Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Taiji Adachi
- Department of Micro Engineering, Graduate School of Engineering, Kyoto University, Kyoto daigaku-katsura, Nishikyo-ku, Kyoto 615-8530, Japan; Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan; Division of Systemic Life Science, Graduate School of Biostudies, Kyoto University, Yoshida-Konoecho, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
20
|
Khalil AS, Jaenisch R, Mooney DJ. Engineered tissues and strategies to overcome challenges in drug development. Adv Drug Deliv Rev 2020; 158:116-139. [PMID: 32987094 PMCID: PMC7518978 DOI: 10.1016/j.addr.2020.09.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/29/2020] [Accepted: 09/23/2020] [Indexed: 12/16/2022]
Abstract
Current preclinical studies in drug development utilize high-throughput in vitro screens to identify drug leads, followed by both in vitro and in vivo models to predict lead candidates' pharmacokinetic and pharmacodynamic properties. The goal of these studies is to reduce the number of lead drug candidates down to the most likely to succeed in later human clinical trials. However, only 1 in 10 drug candidates that emerge from preclinical studies will succeed and become an approved therapeutic. Lack of efficacy or undetected toxicity represents roughly 75% of the causes for these failures, despite these parameters being the primary exclusion criteria in preclinical studies. Recently, advances in both biology and engineering have created new tools for constructing new preclinical models. These models can complement those used in current preclinical studies by helping to create more realistic representations of human tissues in vitro and in vivo. In this review, we describe current preclinical models to identify their value and limitations and then discuss select areas of research where improvements in preclinical models are particularly needed to advance drug development. Following this, we discuss design considerations for constructing preclinical models and then highlight recent advances in these efforts. Taken together, we aim to review the advances as of 2020 surrounding the prospect of biological and engineering tools for adding enhanced biological relevance to preclinical studies to aid in the challenges of failed drug candidates and the burden this poses on the drug development enterprise and thus healthcare.
Collapse
Affiliation(s)
- Andrew S Khalil
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02115, USA; Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Rudolf Jaenisch
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.
| | - David J Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02115, USA.
| |
Collapse
|
21
|
Aisenbrey EA, Murphy WL. Synthetic alternatives to Matrigel. NATURE REVIEWS. MATERIALS 2020; 5:539-551. [PMID: 32953138 PMCID: PMC7500703 DOI: 10.1038/s41578-020-0199-8] [Citation(s) in RCA: 536] [Impact Index Per Article: 107.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/31/2020] [Indexed: 05/19/2023]
Abstract
Matrigel, a basement-membrane matrix extracted from Engelbreth-Holm-Swarm mouse sarcomas, has been used for more than four decades for a myriad of cell culture applications. However, Matrigel is limited in its applicability to cellular biology, therapeutic cell manufacturing and drug discovery owing to its complex, ill-defined and variable composition. Variations in the mechanical and biochemical properties within a single batch of Matrigel - and between batches - have led to uncertainty in cell culture experiments and a lack of reproducibility. Moreover, Matrigel is not conducive to physical or biochemical manipulation, making it difficult to fine-tune the matrix to promote intended cell behaviours and achieve specific biological outcomes. Recent advances in synthetic scaffolds have led to the development of xenogenic-free, chemically defined, highly tunable and reproducible alternatives. In this Review, we assess the applications of Matrigel in cell culture, regenerative medicine and organoid assembly, detailing the limitations of Matrigel and highlighting synthetic scaffold alternatives that have shown equivalent or superior results. Additionally, we discuss the hurdles that are limiting a full transition from Matrigel to synthetic scaffolds and provide a brief perspective on the future directions of synthetic scaffolds for cell culture applications.
Collapse
Affiliation(s)
| | - William L. Murphy
- Department of Biomedical Engineering, University of Wisconsin–Madison, WI, USA
- Department of Orthopedics and Rehabilitation, University of Wisconsin–Madison, WI, USA
| |
Collapse
|
22
|
Khalil AS, Xie AW, Johnson HJ, Murphy WL. Sustained release and protein stabilization reduce the growth factor dosage required for human pluripotent stem cell expansion. Biomaterials 2020; 248:120007. [PMID: 32302801 PMCID: PMC8445021 DOI: 10.1016/j.biomaterials.2020.120007] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Revised: 02/24/2020] [Accepted: 03/24/2020] [Indexed: 12/17/2022]
Abstract
Translation of human pluripotent stem cell (hPSC)-derived therapies to the clinic demands scalable, cost-effective methods for cell expansion. Culture media currently used for hPSC expansion rely on high concentrations and frequent supplementation of recombinant growth factors due to their short half-life at physiological temperatures. Here, we developed a biomaterial strategy using mineral-coated microparticles (MCMs) to sustain delivery of basic fibroblast growth factor (bFGF), a thermolabile protein critical for hPSC pluripotency and proliferation. We show that the MCMs stabilize bFGF against thermally induced activity loss and provide more efficient sustained release of active growth factor compared to polymeric carriers commonly used for growth factor delivery. Using a statistically driven optimization approach called Design of Experiments, we generated a bFGF-loaded MCM formulation that supported hPSC expansion over 25 passages without the need for additional bFGF supplementation to the media, resulting in greater than 80% reduction in bFGF usage compared to standard approaches. This materials-based strategy to stabilize and sustain delivery of a thermolabile growth factor has broad potential to reduce costs associated with recombinant protein supplements in scalable biomanufacturing of emerging cell therapies.
Collapse
Affiliation(s)
- Andrew S Khalil
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Angela W Xie
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Hunter J Johnson
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - William L Murphy
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53705, USA; Department of Materials Science and Engineering, University of Wisconsin-Madison, Madison, WI, 53705, USA; Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI, 53705, USA.
| |
Collapse
|
23
|
Hedegaard CL, Mata A. Integrating self-assembly and biofabrication for the development of structures with enhanced complexity and hierarchical control. Biofabrication 2020; 12:032002. [DOI: 10.1088/1758-5090/ab84cb] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
24
|
Size-Optimized Microspace Culture Facilitates Differentiation of Mouse Induced Pluripotent Stem Cells into Osteoid-Rich Bone Constructs. Stem Cells Int 2020; 2020:7082679. [PMID: 32508932 PMCID: PMC7244985 DOI: 10.1155/2020/7082679] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 01/29/2020] [Accepted: 02/15/2020] [Indexed: 01/15/2023] Open
Abstract
Microspace culture is promising for self-organization of induced pluripotent stem cells (iPSCs). However, the optimal size of microspaces for osteogenic differentiation is unclear. We hypothesized that a specific microspace size could facilitate self-organizing iPSC differentiation to form bone-like tissue in vitro. The objectives of this study were to investigate such effects of microspace size and to evaluate bone regeneration upon transplantation of the resulting osteogenic constructs. Dissociated mouse gingival fibroblast-derived iPSCs were plated in ultra-low-attachment microspace culture wells containing hundreds of U-bottom-shaped microwell spots per well to form cell aggregates in growth medium. The microwells had different aperture diameters/depths (400/560 μm (Elp400), 500/700 μm (Elp500), and 900/700 μm (Elp900)) (Kuraray; Elplasia). After 5 days of aggregation, cells were maintained in osteogenic induction medium for 35 days. Only cells in the Elp500 condition tightly aggregated and maintained high viability during osteogenic induction. After 10 days of induction, Elp500 cell constructs showed significantly higher gene expression of Runx2, Osterix, Collagen 1a1, Osteocalcin, Bone sialoprotein, and Osteopontin compared to constructs in Elp400 and Elp900. In methylene blue-counterstained von Kossa staining and Movat's pentachrome staining, only Elp500 constructs showed robust osteoid formation on day 35, with high expression of type I collagen (a major osteoid component) and osteocalcin proteins. Cell constructs were transplanted into rat calvarial bone defects, and micro-CT analysis after 3 weeks showed better bone repair with significantly higher bone mineral density in the Elp500 group compared to the Elp900 group. These results suggest that microspace size affects self-organized osteogenic differentiation of iPSCs. Elp500 microspace culture specifically induces mouse iPSCs into osteoid-rich bone-like tissue possessing high bone regeneration capacity.
Collapse
|
25
|
Lee B, Borys BS, Kallos MS, Rodrigues CAV, Silva TP, Cabral JMS. Challenges and Solutions for Commercial Scale Manufacturing of Allogeneic Pluripotent Stem Cell Products. Bioengineering (Basel) 2020; 7:E31. [PMID: 32231012 PMCID: PMC7355837 DOI: 10.3390/bioengineering7020031] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 03/25/2020] [Accepted: 03/26/2020] [Indexed: 01/09/2023] Open
Abstract
Allogeneic cell therapy products, such as therapeutic cells derived from pluripotent stem cells (PSCs), have amazing potential to treat a wide variety of diseases and vast numbers of patients globally. However, there are various challenges related to the manufacturing of PSCs in large enough quantities to meet commercial needs. This manuscript addresses the challenges for the process development of PSCs production in a bioreactor, and also presents a scalable bioreactor technology that can be a possible solution to remove the bottleneck for the large-scale manufacturing of high-quality therapeutic cells derived from PSCs.
Collapse
Affiliation(s)
- Brian Lee
- PBS Biotech, Inc., Camarillo, CA 93012, USA
| | - Breanna S. Borys
- Department of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, 2500 University Dr. NW, Calgary, AB T2N 1N4, Canada; (B.S.B.); (M.S.K.)
| | - Michael S. Kallos
- Department of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, 2500 University Dr. NW, Calgary, AB T2N 1N4, Canada; (B.S.B.); (M.S.K.)
| | - Carlos A. V. Rodrigues
- Ibb—Institute for Bioengineering and Biosciences and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisboa 1049-001, Portugal; (C.A.V.R.); (T.P.S.); (J.M.S.C.)
| | - Teresa P. Silva
- Ibb—Institute for Bioengineering and Biosciences and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisboa 1049-001, Portugal; (C.A.V.R.); (T.P.S.); (J.M.S.C.)
| | - Joaquim M. S. Cabral
- Ibb—Institute for Bioengineering and Biosciences and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisboa 1049-001, Portugal; (C.A.V.R.); (T.P.S.); (J.M.S.C.)
| |
Collapse
|
26
|
Borys BS, Le A, Roberts EL, Dang T, Rohani L, Hsu CYM, Wyma AA, Rancourt DE, Gates ID, Kallos MS. Using computational fluid dynamics (CFD) modeling to understand murine embryonic stem cell aggregate size and pluripotency distributions in stirred suspension bioreactors. J Biotechnol 2019; 304:16-27. [PMID: 31394111 DOI: 10.1016/j.jbiotec.2019.08.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 07/19/2019] [Accepted: 08/03/2019] [Indexed: 12/15/2022]
Abstract
Computational fluid dynamics (CFD) modeling can be applied to understand hydrodynamics in stirred suspension bioreactors, which can in turn affect cell viability, proliferation, pluripotency and differentiation. In this study, we developed a CFD model to determine the effects of average shear rates and turbulent eddies on the formation and growth of murine embryonic stem cell aggregates. We found a correlation between average eddy size and aggregate size, which depended on bioreactor agitation rates. By relating these computational and biological variables, CFD modeling can predict optimal agitation rates to grow embryonic stem cell aggregates in stirred suspension bioreactors. To examine the effect of hydrodynamics on pluripotency, mESCs cultured in bioreactors under various agitation rates were tested for SSEA-1, Sox-2, and Nanog expression. Cells maintained a minimum of 95% positive expression with no change in the intensity distribution pattern between the different bioreactor conditions. This indicates that the average level of pluripotency marker expression is independent of changes in the hydrodynamic profile and resulting aggregate size distribution. The findings here can be further extended to other cell types that grow as aggregates in stirred suspension bioreactors and offer important insights necessary to realize cell therapies.
Collapse
Affiliation(s)
- Breanna S Borys
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, 2500 University Dr. NW, Calgary, AB, T2N 1N4, Canada; Biomedical Engineering Graduate Program, University of Calgary, 2500 University Dr. NW, Calgary, AB, T2N 1N4, Canada
| | - An Le
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, 2500 University Dr. NW, Calgary, AB, T2N 1N4, Canada; Biomedical Engineering Graduate Program, University of Calgary, 2500 University Dr. NW, Calgary, AB, T2N 1N4, Canada
| | - Erin L Roberts
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, 2500 University Dr. NW, Calgary, AB, T2N 1N4, Canada; Biomedical Engineering Graduate Program, University of Calgary, 2500 University Dr. NW, Calgary, AB, T2N 1N4, Canada
| | - Tiffany Dang
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, 2500 University Dr. NW, Calgary, AB, T2N 1N4, Canada; Department of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, 2500 University Dr. NW, Calgary, AB, T2N 1N4, Canada
| | - Leili Rohani
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr. NW, Calgary, AB, T2N 4N1, Canada
| | - Charlie Yu-Ming Hsu
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr. NW, Calgary, AB, T2N 4N1, Canada
| | - Alexander A Wyma
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, 2500 University Dr. NW, Calgary, AB, T2N 1N4, Canada; Biomedical Engineering Graduate Program, University of Calgary, 2500 University Dr. NW, Calgary, AB, T2N 1N4, Canada
| | - Derrick E Rancourt
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr. NW, Calgary, AB, T2N 4N1, Canada
| | - Ian D Gates
- Department of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, 2500 University Dr. NW, Calgary, AB, T2N 1N4, Canada
| | - Michael S Kallos
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, 2500 University Dr. NW, Calgary, AB, T2N 1N4, Canada; Biomedical Engineering Graduate Program, University of Calgary, 2500 University Dr. NW, Calgary, AB, T2N 1N4, Canada; Department of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, 2500 University Dr. NW, Calgary, AB, T2N 1N4, Canada.
| |
Collapse
|
27
|
Silva TP, Cotovio JP, Bekman E, Carmo-Fonseca M, Cabral JMS, Fernandes TG. Design Principles for Pluripotent Stem Cell-Derived Organoid Engineering. Stem Cells Int 2019; 2019:4508470. [PMID: 31149014 PMCID: PMC6501244 DOI: 10.1155/2019/4508470] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 02/12/2019] [Accepted: 02/24/2019] [Indexed: 12/17/2022] Open
Abstract
Human morphogenesis is a complex process involving distinct microenvironmental and physical signals that are manipulated in space and time to give rise to complex tissues and organs. Advances in pluripotent stem cell (PSC) technology have promoted the in vitro recreation of processes involved in human morphogenesis. The development of organoids from human PSCs represents one reliable source for modeling a large spectrum of human disorders, as well as a promising approach for drug screening and toxicological tests. Based on the "self-organization" capacity of stem cells, different PSC-derived organoids have been created; however, considerable differences between in vitro-generated PSC-derived organoids and their in vivo counterparts have been reported. Advances in the bioengineering field have allowed the manipulation of different components, including cellular and noncellular factors, to better mimic the in vivo microenvironment. In this review, we focus on different examples of bioengineering approaches used to promote the self-organization of stem cells, including assembly, patterning, and morphogenesis in vitro, contributing to tissue-like structure formation.
Collapse
Affiliation(s)
- Teresa P. Silva
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Lisbon Campus, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av Prof Egas Moniz, Edificio Egas Moniz, 1649-028 Lisboa, Portugal
| | - João P. Cotovio
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Lisbon Campus, Universidade de Lisboa, Lisboa, Portugal
| | - Evguenia Bekman
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Lisbon Campus, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av Prof Egas Moniz, Edificio Egas Moniz, 1649-028 Lisboa, Portugal
| | - Maria Carmo-Fonseca
- The Discoveries Centre for Regenerative and Precision Medicine, Lisbon Campus, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av Prof Egas Moniz, Edificio Egas Moniz, 1649-028 Lisboa, Portugal
| | - Joaquim M. S. Cabral
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Lisbon Campus, Universidade de Lisboa, Lisboa, Portugal
| | - Tiago G. Fernandes
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Lisbon Campus, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
28
|
Tao T, Wang Y, Chen W, Li Z, Su W, Guo Y, Deng P, Qin J. Engineering human islet organoids from iPSCs using an organ-on-chip platform. LAB ON A CHIP 2019; 19:948-958. [PMID: 30719525 DOI: 10.1039/c8lc01298a] [Citation(s) in RCA: 141] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Human pluripotent stem cell (hPSC)-derived islet cells provide promising resources for diabetes studies, cell replacement treatment and drug screening. Recently, hPSC-derived organoids have represented a new class of in vitro organ models for disease modeling and regenerative medicine. However, rebuilding biomimetic human islet organoids from hPSCs remains challenging. Here, we present a new strategy to engineer human islet organoids derived from human induced pluripotent stem cells (hiPSCs) using an organ-on-a-chip platform combined with stem cell developmental principles. The microsystem contains a multi-layer microfluidic device that allows controllable aggregation of embryoid bodies (EBs), in situ pancreatic differentiation and generation of heterogeneous islet organoids in parallel under perfused 3D culture in a single device. The generated islet organoids contain heterogeneous islet-specific α and β-like cells that exhibit favorable growth and cell viability. They also show enhanced expression of pancreatic β-cell specific genes and proteins (PDX1 and NKX6.1) and increased β-cell hormone specific INS gene and C-peptide protein expressions under perfused 3D culture conditions compared to static cultures. In addition, the islet organoids exhibit more sensitive glucose-stimulated insulin secretion (GSIS) and higher Ca2+ flux, indicating the role of biomimetic mechanical flow in promoting endocrine cell differentiation and maturation of islet organoids. This islet-on-a-chip system is robust and amenable to real-time imaging and in situ tracking of islet organoid growth, which may provide a promising platform for organoid engineering, disease modeling, drug testing and regenerative medicine.
Collapse
Affiliation(s)
- Tingting Tao
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China.
| | | | | | | | | | | | | | | |
Collapse
|