1
|
Dumbali SP, Horton PD, Moore TI, Wenzel PL. Mitochondrial permeability transition dictates mitochondrial maturation upon switch in cellular identity of hematopoietic precursors. Commun Biol 2024; 7:967. [PMID: 39122870 PMCID: PMC11316084 DOI: 10.1038/s42003-024-06671-y] [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: 10/22/2023] [Accepted: 08/02/2024] [Indexed: 08/12/2024] Open
Abstract
The mitochondrial permeability transition pore (mPTP) is a supramolecular channel that regulates exchange of solutes across cristae membranes, with executive roles in mitochondrial function and cell death. The contribution of the mPTP to normal physiology remains debated, although evidence implicates the mPTP in mitochondrial inner membrane remodeling in differentiating progenitor cells. Here, we demonstrate that strict control over mPTP conductance shapes metabolic machinery as cells transit toward hematopoietic identity. Cells undergoing the endothelial-to-hematopoietic transition (EHT) tightly control chief regulatory elements of the mPTP. During EHT, maturing arterial endothelium restricts mPTP activity just prior to hematopoietic commitment. After transition in cellular identity, mPTP conductance is restored. In utero treatment with NIM811, a molecule that blocks sensitization of the mPTP to opening by Cyclophilin D (CypD), amplifies oxidative phosphorylation (OXPHOS) in hematopoietic precursors and increases hematopoiesis in the embryo. Additionally, differentiating pluripotent stem cells (PSCs) acquire greater organization of mitochondrial cristae and hematopoietic activity following knockdown of the CypD gene, Ppif. Conversely, knockdown of Opa1, a GTPase critical for proper cristae architecture, induces cristae irregularity and impairs hematopoiesis. These data elucidate a mechanism that regulates mitochondrial maturation in hematopoietic precursors and underscore a role for the mPTP in the acquisition of hematopoietic fate.
Collapse
Affiliation(s)
- Sandeep P Dumbali
- Department of Integrative Biology & Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Paulina D Horton
- Department of Integrative Biology & Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Center for Stem Cell and Regenerative Medicine, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Immunology Program, The University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Travis I Moore
- Department of Integrative Biology & Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Molecular & Translational Biology Program, The University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Pamela L Wenzel
- Department of Integrative Biology & Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA.
- Center for Stem Cell and Regenerative Medicine, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA.
- Immunology Program, The University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA.
- Molecular & Translational Biology Program, The University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA.
| |
Collapse
|
2
|
Son B, Park S, Cho S, Kim JA, Baek SH, Yoo KH, Han D, Joo J, Park HH, Park TH. Improved Neural Inductivity of Size-Controlled 3D Human Embryonic Stem Cells Using Magnetic Nanoparticles. Biomater Res 2024; 28:0011. [PMID: 38500782 PMCID: PMC10944702 DOI: 10.34133/bmr.0011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 02/12/2024] [Indexed: 03/20/2024] Open
Abstract
Background: To improve the efficiency of neural development from human embryonic stem cells, human embryoid body (hEB) generation is vital through 3-dimensional formation. However, conventional approaches still have limitations: long-term cultivation and laborious steps for lineage determination. Methods: In this study, we controlled the size of hEBs for ectodermal lineage specification using cell-penetrating magnetic nanoparticles (MNPs), which resulted in reduced time required for initial neural induction. The magnetized cells were applied to concentrated magnetic force for magnet-derived multicellular organization. The uniformly sized hEBs were differentiated in neural induction medium (NIM) and suspended condition. This neurally induced MNP-hEBs were compared with other groups. Results: As a result, the uniformly sized MNP-hEBs in NIM showed significantly improved neural inductivity through morphological analysis and expression of neural markers. Signaling pathways of the accelerated neural induction were detected via expression of representative proteins; Wnt signaling, dopaminergic neuronal pathway, intercellular communications, and mechanotransduction. Consequently, we could shorten the time necessary for early neurogenesis, thereby enhancing the neural induction efficiency. Conclusion: Overall, this study suggests not only the importance of size regulation of hEBs at initial differentiation stage but also the efficacy of MNP-based neural induction method and stimulations for enhanced neural tissue regeneration.
Collapse
Affiliation(s)
- Boram Son
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
- Department of Bioengineering, Hanyang University, 222 Wangsimri-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Sora Park
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Sungwoo Cho
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Jeong Ah Kim
- Center for Scientific Instrumentation, Korea Basic Science Institute, Cheongju, Chungbuk 28119, Republic of Korea
| | - Seung-Ho Baek
- Center for Bio-based Chemistry, Korea Research Institute of Chemical Technology (KRICT), Ulsan 44429, Korea
| | - Ki Hyun Yoo
- SIMPLE Planet Inc., 48 Achasan-ro 17-gil, Seongdong-gu, Seoul 04799, Korea
| | - Dongoh Han
- SIMPLE Planet Inc., 48 Achasan-ro 17-gil, Seongdong-gu, Seoul 04799, Korea
| | - Jinmyoung Joo
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Hee Ho Park
- Department of Bioengineering, Hanyang University, 222 Wangsimri-ro, Seongdong-gu, Seoul 04763, Republic of Korea
- Research Institute for Convergence of Basic Science, Hanyang University, Seoul 04763, Republic of Korea
| | - Tai Hyun Park
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
- Department of Nutritional Science and Food Management, Ewha Womans University, Seodaemun-gu, Seoul 03760, Republic of Korea
| |
Collapse
|
3
|
Lee SH, Choi J. The Need for Research on the Comparison of Sensory Characteristics between Cultured Meat Produced Using Scaffolds and Meat. Food Sci Anim Resour 2024; 44:269-283. [PMID: 38764515 PMCID: PMC11097029 DOI: 10.5851/kosfa.2023.e81] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/13/2023] [Accepted: 12/17/2023] [Indexed: 05/21/2024] Open
Abstract
Cultured meat is one of the research areas currently in the spotlight in the agricultural and livestock industry, and refers to cells obtained from livestock that are proliferated and differentiated and processed into edible meat. These cell-cultured meats are mainly studied at the lab-scale by culturing them in flasks, and for commercial use, they are produced using scaffolds that mimic cell supports. Scaffolds are broadly divided into fiber scaffolds, hydrogels, and micro-carrier beads, and these are classified according to processing methods and materials. In particular, a scaffold is essential for mass production, which allows it to have appearance, texture, and flavor characteristics similar to meat. Because cultured meat is cultured in a state where oxygen is blocked, it may be lighter in color or produce less flavor substances than edible meat, but these can be compensated for by adding natural substances to the scaffolds or improving fat adhesion. In addition, it has the advantage of being able to express the texture characteristics of the scaffolds that make up the meat in various ways depending on the materials and manufacturing methods of the scaffolds. As a result, to increase consumers' preference for cultured meat and its similarity to edible meat, it is believed that manufacturing scaffolds taking into account the characteristics of edible meat will serve as an important factor. Therefore, continued research and interest in scaffolds is believed to be necessary.
Collapse
Affiliation(s)
- Sol-Hee Lee
- Department of Animal Science, Chungbuk
National University, Cheongju 28644, Korea
| | - Jungseok Choi
- Department of Animal Science, Chungbuk
National University, Cheongju 28644, Korea
| |
Collapse
|
4
|
Dijkhuis L, Johns A, Ragusa D, van den Brink SC, Pina C. Haematopoietic development and HSC formation in vitro: promise and limitations of gastruloid models. Emerg Top Life Sci 2023; 7:439-454. [PMID: 38095554 PMCID: PMC10754337 DOI: 10.1042/etls20230091] [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: 08/22/2023] [Revised: 11/23/2023] [Accepted: 11/27/2023] [Indexed: 12/19/2023]
Abstract
Haematopoietic stem cells (HSCs) are the most extensively studied adult stem cells. Yet, six decades after their first description, reproducible and translatable generation of HSC in vitro remains an unmet challenge. HSC production in vitro is confounded by the multi-stage nature of blood production during development. Specification of HSC is a late event in embryonic blood production and depends on physical and chemical cues which remain incompletely characterised. The precise molecular composition of the HSC themselves is incompletely understood, limiting approaches to track their origin in situ in the appropriate cellular, chemical and mechanical context. Embryonic material at the point of HSC emergence is limiting, highlighting the need for an in vitro model of embryonic haematopoietic development in which current knowledge gaps can be addressed and exploited to enable HSC production. Gastruloids are pluripotent stem cell-derived 3-dimensional (3D) cellular aggregates which recapitulate developmental events in gastrulation and early organogenesis with spatial and temporal precision. Gastruloids self-organise multi-tissue structures upon minimal and controlled external cues, and are amenable to live imaging, screening, scaling and physicochemical manipulation to understand and translate tissue formation. In this review, we consider the haematopoietic potential of gastruloids and review early strategies to enhance blood progenitor and HSC production. We highlight possible strategies to achieve HSC production from gastruloids, and discuss the potential of gastruloid systems in illuminating current knowledge gaps in HSC specification.
Collapse
Affiliation(s)
- Liza Dijkhuis
- Department of Hematopoiesis, Sanquin Research, 1066 CX Amsterdam, The Netherlands
| | - Ayona Johns
- College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, U.K
- Centre for Genome Engineering and Maintenance, Brunel University London, Uxbridge UB8 3PH, U.K
| | - Denise Ragusa
- College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, U.K
- Centre for Genome Engineering and Maintenance, Brunel University London, Uxbridge UB8 3PH, U.K
| | | | - Cristina Pina
- College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, U.K
- Centre for Genome Engineering and Maintenance, Brunel University London, Uxbridge UB8 3PH, U.K
| |
Collapse
|
5
|
Ho DLL, Lee S, Du J, Weiss JD, Tam T, Sinha S, Klinger D, Devine S, Hamfeldt A, Leng HT, Herrmann JE, He M, Fradkin LG, Tan TK, Standish D, Tomasello P, Traul D, Dianat N, Ladi R, Vicard Q, Katikireddy K, Skylar‐Scott MA. Large-Scale Production of Wholly Cellular Bioinks via the Optimization of Human Induced Pluripotent Stem Cell Aggregate Culture in Automated Bioreactors. Adv Healthc Mater 2022; 11:e2201138. [PMID: 36314397 PMCID: PMC10234214 DOI: 10.1002/adhm.202201138] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 10/10/2022] [Indexed: 01/28/2023]
Abstract
Combining the sustainable culture of billions of human cells and the bioprinting of wholly cellular bioinks offers a pathway toward organ-scale tissue engineering. Traditional 2D culture methods are not inherently scalable due to cost, space, and handling constraints. Here, the suspension culture of human induced pluripotent stem cell-derived aggregates (hAs) is optimized using an automated 250 mL stirred tank bioreactor system. Cell yield, aggregate morphology, and pluripotency marker expression are maintained over three serial passages in two distinct cell lines. Furthermore, it is demonstrated that the same optimized parameters can be scaled to an automated 1 L stirred tank bioreactor system. This 4-day culture results in a 16.6- to 20.4-fold expansion of cells, generating approximately 4 billion cells per vessel, while maintaining >94% expression of pluripotency markers. The pluripotent aggregates can be subsequently differentiated into derivatives of the three germ layers, including cardiac aggregates, and vascular, cortical and intestinal organoids. Finally, the aggregates are compacted into a wholly cellular bioink for rheological characterization and 3D bioprinting. The printed hAs are subsequently differentiated into neuronal and vascular tissue. This work demonstrates an optimized suspension culture-to-3D bioprinting pipeline that enables a sustainable approach to billion cell-scale organ engineering.
Collapse
Affiliation(s)
- Debbie L. L. Ho
- Department of BioengineeringStanford UniversityStanfordCA94305USA
| | - Stacey Lee
- Department of BioengineeringStanford UniversityStanfordCA94305USA
| | - Jianyi Du
- Department of BioengineeringStanford UniversityStanfordCA94305USA
| | | | - Tony Tam
- Department of BioengineeringStanford UniversityStanfordCA94305USA
| | - Soham Sinha
- Department of BioengineeringStanford UniversityStanfordCA94305USA
| | - Danielle Klinger
- Department of BioengineeringStanford UniversityStanfordCA94305USA
| | - Sean Devine
- Sartorius Stedim North America Inc565 Johnson AvenueBohemiaNY11716USA
| | - Art Hamfeldt
- Sartorius Stedim North America Inc565 Johnson AvenueBohemiaNY11716USA
| | - Hope T. Leng
- Department of BioengineeringStanford UniversityStanfordCA94305USA
| | - Jessica E. Herrmann
- Department of BioengineeringStanford UniversityStanfordCA94305USA
- School of MedicineStanford UniversityStanfordCA94305USA
| | - Mengdi He
- Materials Science and EngineeringStanford UniversityStanfordCA94305USA
| | - Lee G. Fradkin
- Department of BioengineeringStanford UniversityStanfordCA94305USA
| | - Tze Kai Tan
- Institute of Stem Cell Biology and Regenerative MedicineStanford University School of MedicineStanfordCA94305USA
- Department of GeneticsStanford University School of MedicineStanfordCA94305USA
- Department of PathologyStanford University School of MedicineStanfordCA94305USA
| | - David Standish
- Sartorius Stedim North America Inc565 Johnson AvenueBohemiaNY11716USA
| | - Peter Tomasello
- Sartorius Stedim North America Inc565 Johnson AvenueBohemiaNY11716USA
| | - Donald Traul
- Sartorius Stedim North America Inc565 Johnson AvenueBohemiaNY11716USA
| | - Noushin Dianat
- Sartorius Stedim France S.A.SZone Industrielle les PaludsAvenue de Jouques CS 71058Aubagne Cedex13781France
| | - Rukmini Ladi
- Sartorius Stedim North America Inc565 Johnson AvenueBohemiaNY11716USA
| | - Quentin Vicard
- Sartorius Stedim France S.A.SZone Industrielle les PaludsAvenue de Jouques CS 71058Aubagne Cedex13781France
| | | | - Mark A. Skylar‐Scott
- Department of BioengineeringStanford UniversityStanfordCA94305USA
- Basic Science and Engineering InitiativeChildren's Heart CenterStanford UniversityStanfordCA94305USA
- Chan Zuckerberg BiohubSan FranciscoCA94158USA
| |
Collapse
|
6
|
Wang S, Tao Y. Construction of graphene oxide-modified peptide-coated nanofibrous enhances the osteogenic conversion of induced pluripotent stem cells. INT J POLYM MATER PO 2022. [DOI: 10.1080/00914037.2022.2100374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Affiliation(s)
- Shu Wang
- Chongqing Emergency Medical Center, Chongqing, China
- Chongqing Key Laboratory of Emergency Medicine, Chongqing, China
| | - Yang Tao
- Chongqing Emergency Medical Center, Chongqing, China
- Chongqing Key Laboratory of Emergency Medicine, Chongqing, China
| |
Collapse
|
7
|
Piersma AH, Baker NC, Daston GP, Flick B, Fujiwara M, Knudsen TB, Spielmann H, Suzuki N, Tsaioun K, Kojima H. Pluripotent stem cell assays: Modalities and applications for predictive developmental toxicity. Curr Res Toxicol 2022; 3:100074. [PMID: 35633891 PMCID: PMC9130094 DOI: 10.1016/j.crtox.2022.100074] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 04/21/2022] [Accepted: 05/09/2022] [Indexed: 12/02/2022] Open
Abstract
This manuscript provides a review focused on embryonic stem cell-based models and their place within the landscape of alternative developmental toxicity assays. Against the background of the principles of developmental toxicology, the wide diversity of alternative methods using pluripotent stem cells developed in this area over the past half century is reviewed. In order to provide an overview of available models, a systematic scoping review was conducted following a published protocol with inclusion criteria, which were applied to select the assays. Critical aspects including biological domain, readout endpoint, availability of standardized protocols, chemical domain, reproducibility and predictive power of each assay are described in detail, in order to review the applicability and limitations of the platform in general and progress moving forward to implementation. The horizon of innovative routes of promoting regulatory implementation of alternative methods is scanned, and recommendations for further work are given.
Collapse
Affiliation(s)
- Aldert H. Piersma
- Center for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | | | - George P. Daston
- Global Product Stewardship, The Procter & Gamble Company, Cincinnati, OH, USA
| | - Burkhard Flick
- Experimental Toxicology and Ecology, BASF SE, Ludwigshafen am Rhein, Germany
| | - Michio Fujiwara
- Drug Safety Research Labs, Astellas Pharma Inc., Tsukuba-shi, Japan
| | - Thomas B. Knudsen
- Center for Computational Toxicology and Exposure, U.S. Environmental Protection Agency, Research Triangle Park, USA
| | - Horst Spielmann
- Institute for Pharmacy, Faculty of Biology, Chemistry, and Pharmacy, Freie Universität, Berlin, Germany
| | - Noriyuki Suzuki
- Cell Science Group Environmental Health Science Laboratory, Sumitomo Chemical Co., Ltd., Osaka, Japan
| | - Katya Tsaioun
- Evidence-Based Toxicology Collaboration at Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Hajime Kojima
- National Institute of Health Sciences, Kawasaki, Japan
| |
Collapse
|
8
|
Cha JM, Hwang YS, Kang DK, Lee J, Cooper ES, Mantalaris A. Development of a Novel Perfusion Rotating Wall Vessel Bioreactor with Ultrasound Stimulation for Mass-Production of Mineralized Tissue Constructs. Tissue Eng Regen Med 2022; 19:739-754. [PMID: 35532736 PMCID: PMC9294093 DOI: 10.1007/s13770-022-00447-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 02/06/2022] [Accepted: 02/20/2022] [Indexed: 10/18/2022] Open
Abstract
BACKGROUND As stem cells are considered a promising cell source for tissue engineering, many culture strategies have been extensively studied to generate in vitro stem cell-based tissue constructs. However, most approaches using conventional tissue culture plates are limited by the lack of biological relevance in stem cell microenvironments required for neotissue formation. In this study, a novel perfusion rotating wall vessel (RWV) bioreactor was developed for mass-production of stem cell-based 3D tissue constructs. METHODS An automated RWV bioreactor was fabricated, which is capable of controlling continuous medium perfusion, highly efficient gas exchange with surrounding air, as well as low-intensity pulsed ultrasound (LIPUS) stimulation. Embryonic stem cells encapsulated in alginate/gelatin hydrogel were cultured in the osteogenic medium by using our bioreactor system. Cellular viability, growth kinetics, and osteogenesis/mineralization were thoroughly evaluated, and culture media were profiled at real time. The in vivo efficacy was examined by a rabbit cranial defect model. RESULTS Our bioreactor successfully maintained the optimal culture environments for stem cell proliferation, osteogenic differentiation, and mineralized tissue formation during the culture period. The mineralized tissue constructs produced by our bioreactor demonstrated higher void filling efficacy in the large bone defects compared to the group implanted with hydrogel beads only. In addition, the LIPUS modules mounted on our bioreactor successfully reached higher mineralization of the tissue constructs compared to the groups without LIPUS stimulation. CONCLUSION This study suggests an effective biomanufacturing strategy for mass-production of implantable mineralized tissue constructs from stem cells that could be applicable to future clinical practice.
Collapse
|
9
|
Gordeeva O, Gordeev A, Erokhov P. Archetypal Architecture Construction, Patterning, and Scaling Invariance in a 3D Embryoid Body Differentiation Model. Front Cell Dev Biol 2022; 10:852071. [PMID: 35573693 PMCID: PMC9091174 DOI: 10.3389/fcell.2022.852071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 04/11/2022] [Indexed: 11/13/2022] Open
Abstract
Self-organized patterning and architecture construction studying is a priority goal for fundamental developmental and stem cell biology. To study the spatiotemporal patterning of pluripotent stem cells of different origins, we developed a three-dimensional embryoid body (EB) differentiation model quantifying volumetric parameters and investigated how the EB architecture formation, patterning, and scaling depend on the proliferation, cavitation, and differentiation dynamics, external environmental factors, and cell numbers. We identified three similar spatiotemporal patterns in the EB architectures, regardless of cell origin, which constitute the EB archetype and mimick the pre-gastrulation embryonic patterns. We found that the EB patterning depends strongly on cellular positional information, culture media factor/morphogen content, and free diffusion from the external environment and between EB cell layers. However, the EB archetype formation is independent of the EB size and initial cell numbers forming EBs; therefore, it is capable of scaling invariance and patterning regulation. Our findings indicate that the underlying principles of reaction-diffusion and positional information concepts can serve as the basis for EB architecture construction, patterning, and scaling. Thus, the 3D EB differentiation model represents a highly reproducible and reliable platform for experimental and theoretical research on developmental and stem cell biology issues.
Collapse
Affiliation(s)
- Olga Gordeeva
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia
| | - Andrey Gordeev
- National Institutes of Health’s National Library of Medicine, Bethesda, MD, United States
| | - Pavel Erokhov
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
10
|
Varzideh F, Mone P, Santulli G. Bioengineering Strategies to Create 3D Cardiac Constructs from Human Induced Pluripotent Stem Cells. Bioengineering (Basel) 2022; 9:168. [PMID: 35447728 PMCID: PMC9028595 DOI: 10.3390/bioengineering9040168] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 04/06/2022] [Accepted: 04/08/2022] [Indexed: 12/12/2022] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) can be used to generate various cell types in the human body. Hence, hiPSC-derived cardiomyocytes (hiPSC-CMs) represent a significant cell source for disease modeling, drug testing, and regenerative medicine. The immaturity of hiPSC-CMs in two-dimensional (2D) culture limit their applications. Cardiac tissue engineering provides a new promise for both basic and clinical research. Advanced bioengineered cardiac in vitro models can create contractile structures that serve as exquisite in vitro heart microtissues for drug testing and disease modeling, thereby promoting the identification of better treatments for cardiovascular disorders. In this review, we will introduce recent advances of bioengineering technologies to produce in vitro cardiac tissues derived from hiPSCs.
Collapse
Affiliation(s)
- Fahimeh Varzideh
- Department of Medicine, Wilf Family Cardiovascular Research Institute, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA; (F.V.); (P.M.)
- Department of Molecular Pharmacology, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Pasquale Mone
- Department of Medicine, Wilf Family Cardiovascular Research Institute, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA; (F.V.); (P.M.)
| | - Gaetano Santulli
- Department of Medicine, Wilf Family Cardiovascular Research Institute, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA; (F.V.); (P.M.)
- Department of Molecular Pharmacology, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, New York, NY 10461, USA
| |
Collapse
|
11
|
Francis HS, Harold CL, Beagrie RA, King AJ, Gosden ME, Blayney JW, Jeziorska DM, Babbs C, Higgs DR, Kassouf MT. Scalable in vitro production of defined mouse erythroblasts. PLoS One 2022; 17:e0261950. [PMID: 34995303 PMCID: PMC8741028 DOI: 10.1371/journal.pone.0261950] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 12/14/2021] [Indexed: 01/23/2023] Open
Abstract
Mouse embryonic stem cells (mESCs) can be manipulated in vitro to recapitulate the process of erythropoiesis, during which multipotent cells undergo lineage specification, differentiation and maturation to produce erythroid cells. Although useful for identifying specific progenitors and precursors, this system has not been fully exploited as a source of cells to analyse erythropoiesis. Here, we establish a protocol in which characterised erythroblasts can be isolated in a scalable manner from differentiated embryoid bodies (EBs). Using transcriptional and epigenetic analysis, we demonstrate that this system faithfully recapitulates normal primitive erythropoiesis and fully reproduces the effects of natural and engineered mutations seen in primary cells obtained from mouse models. We anticipate this system to be of great value in reducing the time and costs of generating and maintaining mouse lines in a number of research scenarios.
Collapse
Affiliation(s)
- Helena S. Francis
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Caroline L. Harold
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Robert A. Beagrie
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Andrew J. King
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Matthew E. Gosden
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Joseph W. Blayney
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Danuta M. Jeziorska
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Christian Babbs
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Douglas R. Higgs
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Mira T. Kassouf
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
12
|
Effects of Simulated Microgravity on Wild Type and Marfan hiPSCs-Derived Embryoid Bodies. Cell Mol Bioeng 2021; 14:613-626. [PMID: 34900014 PMCID: PMC8630351 DOI: 10.1007/s12195-021-00680-1] [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: 12/23/2020] [Accepted: 05/20/2021] [Indexed: 11/03/2022] Open
Abstract
Background Mechanical unloading in microgravity is thought to induce tissue degeneration by various mechanisms, including the inhibition of regenerative stem cell differentiation. In this work, we investigate the effects of microgravity simulation on early lineage commitment of hiPSCs from healthy and Marfan Syndrome (MFS; OMIM #154700) donors, using the embryoid bodies model of tissue differentiation and evaluating their ultra-structural conformation. MFS model involves an anomalous organization of the extracellular matrix for a deficit of fibrillin-1, an essential protein of connective tissue. Methods In vitro models require the use of embryoid bodies derived from hiPSCs. A DRPM was used to simulate microgravity conditions. Results Our data suggest an increase of the stemness of those EBs maintained in SMG condition. EBs are still capable of external migration, but are less likely to distinguish, providing a measure of the remaining progenitor or stem cell populations in the earlier stage. The microgravity response appears to vary between WT and Marfan EBs, presumably as a result of a cell structural component deficiency due to fibrillin-1 protein lack. In fact, MFS EBs show a reduced adaptive capacity to the environment of microgravity that prevented them from reacting and making rapid adjustments, while healthy EBs show stem retention, without any structural changes due to microgravity conditions. Conclusion EBs formation specifically mimics stem cell differentiation into embryonic tissues, this process has also significant similarities with adult stem cell-based tissue regeneration. The use of SMG devices for the maintenance of stem cells on regenerative medicine applications is becoming increasingly more feasible. Supplementary Information The online version contains supplementary material available at 10.1007/s12195-021-00680-1.
Collapse
|
13
|
Hulme AJ, Maksour S, St-Clair Glover M, Miellet S, Dottori M. Making neurons, made easy: The use of Neurogenin-2 in neuronal differentiation. Stem Cell Reports 2021; 17:14-34. [PMID: 34971564 PMCID: PMC8758946 DOI: 10.1016/j.stemcr.2021.11.015] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 11/27/2021] [Accepted: 11/29/2021] [Indexed: 01/01/2023] Open
Abstract
Directed neuronal differentiation of human pluripotent stem cells (hPSCs), neural progenitors, or fibroblasts using transcription factors has allowed for the rapid and highly reproducible differentiation of mature and functional neurons. Exogenous expression of the transcription factor Neurogenin-2 (NGN2) has been widely used to generate different populations of neurons, which have been used in neurodevelopment studies, disease modeling, drug screening, and neuronal replacement therapies. Could NGN2 be a “one-glove-fits-all” approach for neuronal differentiations? This review summarizes the cellular roles of NGN2 and describes the applications and limitations of using NGN2 for the rapid and directed differentiation of neurons.
Collapse
Affiliation(s)
- Amy J Hulme
- Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia; School of Medicine, University of Wollongong, Wollongong, NSW, Australia; Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia
| | - Simon Maksour
- Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia; School of Medicine, University of Wollongong, Wollongong, NSW, Australia; Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia
| | - Mitchell St-Clair Glover
- Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia; School of Medicine, University of Wollongong, Wollongong, NSW, Australia; Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia
| | - Sara Miellet
- Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia; School of Medicine, University of Wollongong, Wollongong, NSW, Australia; Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia
| | - Mirella Dottori
- Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia; School of Medicine, University of Wollongong, Wollongong, NSW, Australia; Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia.
| |
Collapse
|
14
|
Song MH, Choi SC, Noh JM, Joo HJ, Park CY, Cha JJ, Ahn TH, Ko TH, Choi JI, Na JE, Rhyu IJ, Jang Y, Park Y, Gim JA, Kim JH, Lim DS. LEFTY-PITX2 signaling pathway is critical for generation of mature and ventricular cardiac organoids in human pluripotent stem cell-derived cardiac mesoderm cells. Biomaterials 2021; 278:121133. [PMID: 34571434 DOI: 10.1016/j.biomaterials.2021.121133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 09/10/2021] [Accepted: 09/15/2021] [Indexed: 02/07/2023]
Abstract
The generation of mature ventricular cardiomyocytes (CMs) resembling adult CMs from human pluripotent stem cells (hPSCs) is necessary for disease modeling and drug discovery. To investigate the effect of self-organizing capacity on the generation of mature cardiac organoids (COs), we generated cardiac mesoderm cell-derived COs (CMC-COs) and CM-derived COs (CM-COs) and evaluated COs. CMC-COs exhibited more organized sarcomere structures and mitochondria, well-arranged t-tubule structures, and evenly distributed intercalated discs. Increased expressions of ventricular CM, cardiac metabolic, t-tubule formation, K+ ion channel, and junctional markers were confirmed in CMC-COs. Mature ventricular-like function such as faster motion vector speed, decreased beats per min, increased peak-to-peak duration, and prolonged APD50 and APD90 were observed in CMC-COs. Transcriptional profiling revealed that extracellular matrix-integrin, focal adhesion, and LEFTY-PITX2 signaling pathways are upregulated in CMC-COs. LEFTY knockdown affected ECM-integrin-FA signaling pathways in CMC-COs. Here, we found that high self-organizing capacity of CMCs is critical for the generation of mature and ventricular COs. We also demonstrated that LEFTY-PITX2 signaling plays key roles for CM maturation and specification into ventricular-like CM subtype in CMC-COs. CMC-COs are an attractive resource for disease modeling and drug discovery.
Collapse
Affiliation(s)
- Myeong-Hwa Song
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, South Korea
| | - Seung-Cheol Choi
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, South Korea; R&D Center for Companion Diagnostic, SOL Bio Corporation, Suite 510, 27, Seongsui-ro7-gil, Seongdong-gu, Seoul, 04780, South Korea
| | - Ji-Min Noh
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, South Korea
| | - Hyung Joon Joo
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, South Korea
| | - Chi-Yeon Park
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, South Korea
| | - Jung-Joon Cha
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, South Korea
| | - Tae Hoon Ahn
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, South Korea
| | - Tae Hee Ko
- Division of Cardiology, Department of Internal Medicine, Korea University College of Medicine and Korea University Anam Hospital, Seoul, 02841, South Korea
| | - Jong-Il Choi
- Division of Cardiology, Department of Internal Medicine, Korea University College of Medicine and Korea University Anam Hospital, Seoul, 02841, South Korea
| | - Ji Eun Na
- Department of Anatomy, College of Medicine, Korea University, Seoul, 02841, South Korea
| | - Im Joo Rhyu
- Department of Anatomy, College of Medicine, Korea University, Seoul, 02841, South Korea
| | - Yongjun Jang
- Department of Biomedical Sciences, College of Medicine, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, South Korea
| | - Yongdoo Park
- Department of Biomedical Sciences, College of Medicine, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, South Korea
| | - Jeong-An Gim
- Medical Science Research Center, College of Medicine, Korea University Guro Hospital, Seoul,08308, South Korea
| | - Jong-Hoon Kim
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, South Korea
| | - Do-Sun Lim
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, South Korea.
| |
Collapse
|
15
|
Samiei M, Fathi M, Barar J, Fathi N, Amiryaghoubi N, Omidi Y. Bioactive hydrogel-based scaffolds for the regeneration of dental pulp tissue. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
16
|
Zeng H, Peng F, Wang J, Meng R, Zhang J. Effects of Fruquintinib on the Pluripotency Maintenance and Differentiation Potential of Mouse Embryonic Stem Cells. Cell Reprogram 2021; 23:180-190. [PMID: 34077681 DOI: 10.1089/cell.2020.0100] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Mouse embryonic stem cells (mESCs) can maintain self-renewal and differentiate into any cell type of the three primary germ layers. The vascular endothelial growth factor (VEGF) is involved in the regulation of mESC differentiation and induces the activation of a series of kinase responses and several cell signaling pathways by binding to its respective transmembrane receptors, vascular endothelial growth factor receptor VEGFR1, and VEGFR2. Fruquintinib is a selective inhibitor of VEGFRs, and we used it to investigate the effects on the maintenance of pluripotency and differentiation potential of mESCs in this study. Our results showed that fruquintinib-treated cells expressed higher levels of pluripotent markers, including Oct4, Nanog, Sox2, and Esrrb under serum and leukemia inhibitory factor (LIF) condition, whereas the expression of phosphorylated Erk1/2 was restricted. Mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (MEK) signaling inhibitor (PD0325901) and glycogen synthase kinase 3 (GSK3) signaling inhibitor (CHIR99021) (also known as 2i) enable cells to maintain naive pluripotency with LIF, and fruquintinib can also promote cells to maintain naive pluripotent state even under serum/LIF condition, whereas VEGF addition limits the pluripotency characteristics in serum/LIF mESCs. Furthermore, fruquintinib could inhibit the three-germ layer establishment in embryoid body formation and maintain the undifferentiated characteristics of mESCs, indicating that fruquintinib could promote the maintenance of naive pluripotency and inhibit early differentiation programs.
Collapse
Affiliation(s)
- Hanyi Zeng
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Fanke Peng
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Jiachen Wang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Ru Meng
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Jun Zhang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| |
Collapse
|
17
|
Smith LA, Hidalgo Aguilar A, Owens DDG, Quelch RH, Knight E, Przyborski SA. Using Advanced Cell Culture Techniques to Differentiate Pluripotent Stem Cells and Recreate Tissue Structures Representative of Teratoma Xenografts. Front Cell Dev Biol 2021; 9:667246. [PMID: 34026759 PMCID: PMC8134696 DOI: 10.3389/fcell.2021.667246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 04/12/2021] [Indexed: 11/24/2022] Open
Abstract
Various methods are currently used to investigate human tissue differentiation, including human embryo culture and studies utilising pluripotent stem cells (PSCs) such as in vitro embryoid body formation and in vivo teratoma assays. Each method has its own distinct advantages, yet many are limited due to being unable to achieve the complexity and maturity of tissue structures observed in the developed human. The teratoma xenograft assay allows maturation of more complex tissue derivatives, but this method has ethical issues surrounding animal usage and significant protocol variation. In this study, we have combined three-dimensional (3D) in vitro cell technologies including the common technique of embryoid body (EB) formation with a novel porous scaffold membrane, in order to prolong cell viability and extend the differentiation of PSC derived EBs. This approach enables the formation of more complex morphologically identifiable 3D tissue structures representative of all three primary germ layers. Preliminary in vitro work with the human embryonal carcinoma line TERA2.SP12 demonstrated improved EB viability and enhanced tissue structure formation, comparable to teratocarcinoma xenografts derived in vivo from the same cell line. This is thought to be due to reduced diffusion distances as the shape of the spherical EB transforms and flattens, allowing for improved nutritional/oxygen support to the developing structures over extended periods. Further work with EBs derived from murine embryonic stem cells demonstrated that the formation of a wide range of complex, recognisable tissue structures could be achieved within 2–3 weeks of culture. Rudimentary tissue structures from all three germ layers were present, including epidermal, cartilage and epithelial tissues, again, strongly resembling tissue structure of teratoma xenografts of the same cell line. Proof of concept work with EBs derived from the human embryonic stem cell line H9 also showed the ability to form complex tissue structures within this system. This novel yet simple model offers a controllable, reproducible method to achieve complex tissue formation in vitro. It has the potential to be used to study human developmental processes, as well as offering an animal free alternative method to the teratoma assay to assess the developmental potential of novel stem cell lines.
Collapse
Affiliation(s)
- L A Smith
- Department of Biosciences, Durham University, Durham, United Kingdom
| | - A Hidalgo Aguilar
- Department of Biosciences, Durham University, Durham, United Kingdom
| | - D D G Owens
- Department of Biosciences, Durham University, Durham, United Kingdom
| | - R H Quelch
- Department of Biosciences, Durham University, Durham, United Kingdom
| | - E Knight
- Department of Biosciences, Durham University, Durham, United Kingdom
| | - S A Przyborski
- Department of Biosciences, Durham University, Durham, United Kingdom.,Reprocell Europe, NETPark, Sedgefield, United Kingdom
| |
Collapse
|
18
|
Miller SA, Damle M, Kim J, Kingston RE. Full methylation of H3K27 by PRC2 is dispensable for initial embryoid body formation but required to maintain differentiated cell identity. Development 2021; 148:dev196329. [PMID: 33688077 PMCID: PMC8077505 DOI: 10.1242/dev.196329] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 03/02/2021] [Indexed: 12/13/2022]
Abstract
Polycomb repressive complex 2 (PRC2) catalyzes methylation of histone H3 on lysine 27 and is required for normal development of complex eukaryotes. The nature of that requirement is not clear. H3K27me3 is associated with repressed genes, but the modification is not sufficient to induce repression and, in some instances, is not required. We blocked full methylation of H3K27 with both a small molecule inhibitor, GSK343, and by introducing a point mutation into EZH2, the catalytic subunit of PRC2, in the mouse CJ7 cell line. Cells with substantively decreased H3K27 methylation differentiate into embryoid bodies, which contrasts with EZH2 null cells. PRC2 targets had varied requirements for H3K27me3, with a subset that maintained normal levels of repression in the absence of methylation. The primary cellular phenotype of blocked H3K27 methylation was an inability of altered cells to maintain a differentiated state when challenged. This phenotype was determined by H3K27 methylation in embryonic stem cells through the first 4 days of differentiation. Full H3K27 methylation therefore was not necessary for formation of differentiated cell states during embryoid body formation but was required to maintain a stable differentiated state.
Collapse
Affiliation(s)
- Sara A. Miller
- Department of Molecular Biology, Massachusetts General Hospital Research Institute, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Manashree Damle
- Department of Molecular Biology, Massachusetts General Hospital Research Institute, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Jongmin Kim
- Department of Molecular Biology, Massachusetts General Hospital Research Institute, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Robert E. Kingston
- Department of Molecular Biology, Massachusetts General Hospital Research Institute, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
19
|
Roshandel M, Dorkoosh F. Cardiac tissue engineering, biomaterial scaffolds, and their fabrication techniques. POLYM ADVAN TECHNOL 2021. [DOI: 10.1002/pat.5273] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Marjan Roshandel
- School of Chemical Engineering, College of Engineering University of Tehran Tehran Iran
| | - Farid Dorkoosh
- Department of Pharmaceutics, Faculty of Pharmacy Tehran University of Medical Sciences Tehran Iran
- Medical Biomaterial Research Centre (MBRC) Tehran University of Medical Sciences Tehran Iran
| |
Collapse
|
20
|
Jansch C, Ziegler GC, Forero A, Gredy S, Wäldchen S, Vitale MR, Svirin E, Zöller JEM, Waider J, Günther K, Edenhofer F, Sauer M, Wischmeyer E, Lesch KP. Serotonin-specific neurons differentiated from human iPSCs form distinct subtypes with synaptic protein assembly. J Neural Transm (Vienna) 2021; 128:225-241. [PMID: 33560471 PMCID: PMC7914246 DOI: 10.1007/s00702-021-02303-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 01/10/2021] [Indexed: 02/06/2023]
Abstract
Human induced pluripotent stem cells (hiPSCs) have revolutionized the generation of experimental disease models, but the development of protocols for the differentiation of functionally active neuronal subtypes with defined specification is still in its infancy. While dysfunction of the brain serotonin (5-HT) system has been implicated in the etiology of various neuropsychiatric disorders, investigation of functional human 5-HT specific neurons in vitro has been restricted by technical limitations. We describe an efficient generation of functionally active neurons from hiPSCs displaying 5-HT specification by modification of a previously reported protocol. Furthermore, 5-HT specific neurons were characterized using high-end fluorescence imaging including super-resolution microscopy in combination with electrophysiological techniques. Differentiated hiPSCs synthesize 5-HT, express specific markers, such as tryptophan hydroxylase 2 and 5-HT transporter, and exhibit an electrophysiological signature characteristic of serotonergic neurons, with spontaneous rhythmic activities, broad action potentials and large afterhyperpolarization potentials. 5-HT specific neurons form synapses reflected by the expression of pre- and postsynaptic proteins, such as Bassoon and Homer. The distribution pattern of Bassoon, a marker of the active zone along the soma and extensions of neurons, indicates functionality via volume transmission. Among the high percentage of 5-HT specific neurons (~ 42%), a subpopulation of CDH13 + cells presumably designates dorsal raphe neurons. hiPSC-derived 5-HT specific neuronal cell cultures reflect the heterogeneous nature of dorsal and median raphe nuclei and may facilitate examining the association of serotonergic neuron subpopulations with neuropsychiatric disorders.
Collapse
Affiliation(s)
- Charline Jansch
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Margarete-Höppel-Platz 1, 97080, Würzburg, Germany
| | - Georg C Ziegler
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Margarete-Höppel-Platz 1, 97080, Würzburg, Germany.
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University of Würzburg, Würzburg, Germany.
| | - Andrea Forero
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Margarete-Höppel-Platz 1, 97080, Würzburg, Germany
| | - Sina Gredy
- Institute of Physiology, Molecular Electrophysiology, University of Würzburg, Würzburg, Germany
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Sina Wäldchen
- Department of Biotechnology and Biophysics, Biocenter, University of Würzburg, Würzburg, Germany
| | - Maria Rosaria Vitale
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Margarete-Höppel-Platz 1, 97080, Würzburg, Germany
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Evgeniy Svirin
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Margarete-Höppel-Platz 1, 97080, Würzburg, Germany
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Johanna E M Zöller
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Margarete-Höppel-Platz 1, 97080, Würzburg, Germany
- Department of Translational Neuroscience, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Maastricht, The Netherlands
| | - Jonas Waider
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Margarete-Höppel-Platz 1, 97080, Würzburg, Germany
| | - Katharina Günther
- Department of Genomics, Stem Cell Biology and Regenerative Medicine, Institute of Molecular Biology and CMBI, Leopold-Franzens-University Innsbruck, Innsbruck, Austria
- Institute of Molecular Regenerative Medicine, SCI-TReCS, Paracelsus Medical University, Salzburg, Austria
| | - Frank Edenhofer
- Department of Genomics, Stem Cell Biology and Regenerative Medicine, Institute of Molecular Biology and CMBI, Leopold-Franzens-University Innsbruck, Innsbruck, Austria
| | - Markus Sauer
- Department of Biotechnology and Biophysics, Biocenter, University of Würzburg, Würzburg, Germany
| | - Erhard Wischmeyer
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Margarete-Höppel-Platz 1, 97080, Würzburg, Germany
- Institute of Physiology, Molecular Electrophysiology, University of Würzburg, Würzburg, Germany
| | - Klaus-Peter Lesch
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Margarete-Höppel-Platz 1, 97080, Würzburg, Germany.
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russia.
- Department of Translational Neuroscience, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
21
|
Oss-Ronen L, Redden RA, Lelkes PI. Enhanced Induction of Definitive Endoderm Differentiation of Mouse Embryonic Stem Cells in Simulated Microgravity. Stem Cells Dev 2020; 29:1275-1284. [PMID: 32731794 DOI: 10.1089/scd.2020.0097] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Directed in vitro differentiation of pluripotent stem cells toward definitive endoderm (DE) offers great research and therapeutic potential since these cells can further differentiate into cells of the respiratory and gastrointestinal tracts, as well as associated organs such as pancreas, liver, and thyroid. We hypothesized that culturing mouse embryonic stem cells (mESCs) under simulated microgravity (SMG) conditions in rotary bioreactors (BRs) will enhance the induction of directed DE differentiation. To test our hypothesis, we cultured the cells for 6 days in two-dimensional monolayer colony cultures or as embryoid bodies (EBs) in either static conditions or, dynamically, in the rotary BRs. We used flow cytometry and quantitative polymerase chain reaction to analyze the expression of marker proteins and genes, respectively, for pluripotency (Oct3/4) and mesendodermal (Brachyury T), endodermal (FoxA2, Sox17, CxCr4), and mesodermal (Vimentin, Meox1) lineages. Culture in the form of EBs in maintenance media in the presence of leukemia inhibitory factor, in static or SMG conditions, induced expression of some of the differentiation markers, suggesting heterogeneity of the cells. This is in line with previous studies showing that differentiation is initiated as cells are aggregated into EBs even without supplementing differentiation factors to the media. Culturing EBs in static conditions in differentiation media (DM) in the presence of activin A reduced Oct3/4 expression and significantly increased Brachyury T and CxCr4 expression, but downregulated FoxA2 and Sox17. However, culturing in SMG BRs in DM upregulated Brachyury T and all of the DE markers and reduced Oct3/4 expression, indicating the advantage of dynamic cultures in BRs to specifically enhance directed DE differentiation. Given the potential discrepancies between the SMG conditions on earth and actual microgravity conditions, as observed in other studies, future experiments in space flight are required to validate the effects of reduced gravity on mESC differentiation.
Collapse
Affiliation(s)
- Liat Oss-Ronen
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, Pennsylvania, USA
| | - Robert A Redden
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, Pennsylvania, USA
| | - Peter I Lelkes
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
22
|
Ibrahim M, Xie B, Richardson MK. The growth of endothelial-like cells in zebrafish embryoid body culture. Exp Cell Res 2020; 392:112032. [PMID: 32353375 DOI: 10.1016/j.yexcr.2020.112032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 04/21/2020] [Indexed: 11/25/2022]
Abstract
There is increasing interest in the possibility of culturing organ-like tissues (organoids) in vitro for biomedical applications. The ability to culture organoids would be greatly enhanced by having a functional circulation in vitro. The endothelial cell is the most important cell type in this context. Endothelial cells can be derived from pluripotent embryonic blastocyst cells in aggregates called embryoid bodies. Here, we examine the yield of endothelial-like cells in embryoid bodies (EBs) developed from transgenic zebrafish fli:GFP and kdrl:GFP blastocyst embryos. The isolated blastocyst cells developed into EBs within the first 24 h of culture and contained fli:GFP+ (putative endothelial, hematopoietic and other cell types); or kdrl:GFP+ (endothelial) cells. The addition of endothelial growth supplements to the media and culture on collagen type-I substratum increased the percentages of fli:GFP+ and kdrl:GFP+ cells in culture. We found that EBs developed in hanging-drop cultures possessed a higher percentage of fli:GFP+ (45.0 ± 3.1%) and kdrl:GFP+ cells (8.7 ± 0.7%) than those developed on conventional substrata (34.5 ± 1.4% or 5.2 ± 0.4%, respectively). The transcriptome analysis showed a higher expression of VEGF and TGFβ genes in EB cultures compared to the adherent cultures. When transferred to conventional culture, the percentage of fli:GFP+ or kdrl:GFP+ cells declined significantly over subsequent days in the EBs. The fli:GFP+ cells formed a monolayer around the embryoid bodies, while the kdrl:GFP+ cells formed vascular network-like structures in the embryoid bodies. Differences were observed in the spreading of fli:GFP+ cells, and network formation of kdrl:GFP+ cells on different substrates. The fli:GFP+ cells could be maintained in primary culture and sub-cultures. By contrast, kdrl:GFP+ cells were almost completely absent at 8d of primary culture. Our culture model allows real-time observation of fli:GFP+ and kdrl:GFP+ cells in culture. The results obtained from this study will be important for the development of vascular and endothelial cell culture using embryonic cells.
Collapse
Affiliation(s)
- Muhammad Ibrahim
- Institute of Biology Leiden, Leiden University, The Netherlands; Animal Biotechnology Division, Institute of Biotechnology and Genetic Engineering, The University of Agriculture Peshawar, Pakistan
| | - Bing Xie
- Institute of Biology Leiden, Leiden University, The Netherlands
| | | |
Collapse
|
23
|
Hanafiah A, Geng Z, Wang Q, Gao Z. Differentiation and Characterization of Neural Progenitors and Neurons from Mouse Embryonic Stem Cells. J Vis Exp 2020. [PMID: 32478755 DOI: 10.3791/61446] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
We describe the step-by-step procedure for culturing and differentiating mouse embryonic stem cells into neuronal lineages, followed by a series of assays to characterize the differentiated cells. The E14 mouse embryonic stem cells were used to form embryoid bodies through the hanging drop method, and then induced to differentiate into neural progenitor cells by retinoic acid, and finally differentiated into neurons. Quantitative reverse transcription polymerase chain reaction (RT-qPCR) and immunofluorescence experiments revealed that the neural progenitors and neurons exhibit corresponding markers (nestin for neural progenitors and neurofilament for neurons) at day 8 and 12 post-differentiation, respectively. Flow cytometry experiments on an E14 line expressing a Sox1 promoter-driven GFP reporter showed that about 60% of cells at day 8 are GFP positive, indicating the successful differentiation of neural progenitor cells at this stage. Finally, RNA-seq analysis was used to profile the global transcriptomic changes. These methods are useful for analyzing the involvement of specific genes and pathways in regulating the cell identity transition during neuronal differentiation.
Collapse
Affiliation(s)
- Aflah Hanafiah
- Departments of Biochemistry and Molecular Biology, Penn State College of Medicine, Penn State Hershey Cancer Institute
| | - Zhuangzhuang Geng
- Departments of Biochemistry and Molecular Biology, Penn State College of Medicine, Penn State Hershey Cancer Institute
| | - Qiang Wang
- Departments of Biochemistry and Molecular Biology, Penn State College of Medicine, Penn State Hershey Cancer Institute
| | - Zhonghua Gao
- Departments of Biochemistry and Molecular Biology, Penn State College of Medicine, Penn State Hershey Cancer Institute;
| |
Collapse
|
24
|
Bedell ML, Melchiorri AJ, Aleman J, Skardal A, Mikos AG. A high-throughput approach to compare the biocompatibility of candidate bioink formulations. ACTA ACUST UNITED AC 2020. [DOI: 10.1016/j.bprint.2019.e00068] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
25
|
Kim YY, Kim H, Suh CS, Liu HC, Rosenwaks Z, Ku SY. Effects of Natural Progesterone and Synthetic Progestin on Germ Layer Gene Expression in a Human Embryoid Body Model. Int J Mol Sci 2020; 21:769. [PMID: 31991577 PMCID: PMC7036864 DOI: 10.3390/ijms21030769] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 01/16/2020] [Accepted: 01/17/2020] [Indexed: 11/17/2022] Open
Abstract
Natural progesterone and synthetic progestin are widely used for the treatment of threatened abortion or in in vitro fertilization (IVF) cycles. This in vitro study aimed to assess whether the treatment with natural progesterone or synthetic progestin influences the germ layer gene expression on the early human embryonic development using human embryonic stem cells (hESCs)-derived embryoid bodies (hEBs) as a surrogate of early stage human embryonic development. Human EBs derived from hESCs were cultured for nine days, and were treated with natural progesterone (P4) or synthetic progestin, medroxyprogesterone acetate (MPA) at 10-7 M for five days. To reverse the effects of treatment, mifepristone (RU486) as progesterone antagonist was added to the hEBs for four days starting one day after the initiation of treatment. Mouse blastocysts (mBLs) were cultured in vitro for 24 h, and P4 or MPA at 10-7 M was treated for an additional 24 h. The treated embryos were further transferred onto in vitro cultured endometrial cells to evaluate chorionic gonadotropin (CG) expression. To analyze the effects of P4 or MPA, the expression of differentiation genes representing the three germ layers was investigated, GATA-binding factor 4 (GATA4), α-fetoprotein (AFP), hepatocyte nuclear factor (HNF)-3β, hepatocyte nuclear factor (HNF)-4α (endoderm), Brachyury, cardiac actin (cACT) (mesoderm), and Nestin (ectoderm), using quantitative reverse transcription PCR (qRT-PCR) and immunostaining. Significantly lower expressions of HNF-3β, HNF-4α, Brachyury, and Nestin were observed in MPA-treated hEBs (all p < 0.05), which was negated by RU486 treatment. This inhibitory effect of MPA was also observed in mouse embryos. Conclusively, the effects of natural progesterone and synthetic progestin may differ in the germ layer gene expression in the hEB model, which suggests that caution is necessary in the use of progestogen.
Collapse
Affiliation(s)
- Yoon Young Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul 03080, Korea; (Y.Y.K.); (H.K.); (C.S.S.)
- Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Hoon Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul 03080, Korea; (Y.Y.K.); (H.K.); (C.S.S.)
- Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Chang Suk Suh
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul 03080, Korea; (Y.Y.K.); (H.K.); (C.S.S.)
- Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Hung-Ching Liu
- Center for Reproductive Medicine and Infertility, Weill Medical College of Cornell University, New York, NY 10065, USA; (H.-C.L.); (Z.R.)
| | - Zev Rosenwaks
- Center for Reproductive Medicine and Infertility, Weill Medical College of Cornell University, New York, NY 10065, USA; (H.-C.L.); (Z.R.)
| | - Seung-Yup Ku
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul 03080, Korea; (Y.Y.K.); (H.K.); (C.S.S.)
- Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University College of Medicine, Seoul 03080, Korea
| |
Collapse
|
26
|
Rassouli H, Khalaj M, Hassani SN, Nemati SH, Hosseini Salekdeh GH, Baharvand H. Gene Expression Patterns of Royan Human Embryonic Stem Cells Correlate with Their Propensity and Culture Systems. CELL JOURNAL 2019; 21:290-299. [PMID: 31210435 PMCID: PMC6582416 DOI: 10.22074/cellj.2019.6128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 11/20/2018] [Indexed: 11/09/2022]
Abstract
OBJECTIVE Human embryonic stem cells (hESCs) have the potential to give rise to all types of cells in the human body when appropriately induced to differentiate. Stem cells can differentiate spontaneously into the three-germ layer derivatives by embryoid bodies (EBs) formation. However, the two-dimensional (2D) adherent culture of hESCs under defined conditions is commonly used for directed differentiation toward a specific type of mature cells. In this study, we aimed to determine the propensity of the Royan hESC lines based on comparison of expression levels of 46 lineage specific markers. MATERIALS AND METHODS In this experimental study, we have compared the expression of lineage-specific markers in hESC lines during EB versus adherent-based spontaneous differentiation. We used quantitative real-time polymerase chain reaction (qRT-PCR) to assess expressions of 46 lineage-specific markers in 4 hESC lines, Royan H1 (RH1), RH2, RH5, and RH6, during spontaneous differentiation in both EB and adherent cultures at 0, 10, and 30 days after initiation of differentiation. RESULTS Based on qRT-PCR data analysis, the liver and neuronal markers had higher expression levels in EBs, whereas skin-specific markers expressed at higher levels in the adherent culture. The results showed differential expression patterns of some lineage-specific markers in EBs compared with the adherent cultures. CONCLUSION According to these results, possibly the spontaneous differentiation technique could be a useful method for optimization of culture conditions to differentiate stem cells into specific cell types such ectoderm, neuron, endoderm and hepatocyte. This approach might prove beneficial for further work on maximizing the efficiency of directed differentiation and development of novel differentiation protocols.
Collapse
Affiliation(s)
- Hassan Rassouli
- Department of Molecular Systems Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mona Khalaj
- Department of Molecular Systems Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Seyedeh-Nafiseh Hassani
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - S Hiva Nemati
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - G Hasem Hosseini Salekdeh
- Department of Molecular Systems Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Systems Biology, Agricultural Biotechnology Research Institute of Iran, Agricultural Research, Education, and Extension Organization, Karaj, Iran. Electronic Address:
- Department of Molecular Sciences, Macquarie University, Sydney, NSW, Australia
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran. Electronic Address:
- Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| |
Collapse
|
27
|
Kim HJ, Sung IY, Cho YC, Kang MS, Rho GJ, Byun JH, Park WU, Son MG, Park BW, Lee HJ, Kang YH. Three-Dimensional Spheroid Formation of Cryopreserved Human Dental Follicle-Derived Stem Cells Enhances Pluripotency and Osteogenic Induction Properties. Tissue Eng Regen Med 2019; 16:513-523. [PMID: 31624706 DOI: 10.1007/s13770-019-00203-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 07/03/2019] [Accepted: 07/05/2019] [Indexed: 02/07/2023] Open
Abstract
Background Enhancement and maintenance of the stemness of mesenchymal stem cells (MSCs) is one of the most important factors contributing to the successful in vivo therapeutic application of these cells. In this regard, three-dimensional (3D) spheroid formation has been developed as reliable method for increasing the pluripotency of MSCs. Moreover, using a new protocol, we have previously shown that dental tissues of extracted wisdom teeth can be effectively cryopreserved for subsequent use as a source of autologous stem cells. The main purpose of this study is to analyze the stemness and in vitro osteogenic differentiation potential of 3D spheroid dental MSCs compared with conventional mono-layer cultured MSCs. Methods In this study, MSC-characterized stem cells were isolated and cultured from long-term cryopreserved dental follicles (hDFSCs), and then 2D hDFSCs were cultured under 3D spheroid-forming conditions using a newly designed microchip dish. The spheroids (3D hDFSCs) thus produced were investigated and characterized with respect to stemness, MSC marker expression, apoptosis, cell cycle analysis, extracellular matrix (ECM) production, and osteogenic and adipogenic differentiation properties. Results In terms of MSC and senescence markers, spheroid cells showed no difference when compared with 2D hDFSCs; however, 3D hDFSCs were observed to have a higher proportion of cell cycle arrest and a larger number of apoptotic cells. Moreover, spheroids showed substantially increased levels of pluripotency marker (early transcription factors) and ECM protein expression. Compared with 2D hDFSCs, there was also a notable enhancement in the osteogenic induction potential of spheroids, although no differences were observed with respect to in vitro adipogenesis. Conclusion To the best of our knowledge, this is the first study to demonstrate the application of a spheroid culture system for dental follicle-derived stem cells using a microchip dish. Although further studies are needed, including in vivo transplantation, the results obtained in this study indicate that spheroid hDFSCs derived from cryopreserved dental follicle tissues could be used as a valuable source of autologous stem cells for bone tissue regeneration.
Collapse
Affiliation(s)
- Hyo-Jung Kim
- 1Department of Oral and Maxillofacial Surgery, College of Medicine, University of Ulsan, 877 Bangeojinsunhwando-ro, Dong-gu, Ulsan, 44033 Republic of Korea
| | - Iel-Yong Sung
- 1Department of Oral and Maxillofacial Surgery, College of Medicine, University of Ulsan, 877 Bangeojinsunhwando-ro, Dong-gu, Ulsan, 44033 Republic of Korea
| | - Yeong-Cheol Cho
- 1Department of Oral and Maxillofacial Surgery, College of Medicine, University of Ulsan, 877 Bangeojinsunhwando-ro, Dong-gu, Ulsan, 44033 Republic of Korea
| | - Min-Su Kang
- 2Department of Oral and Maxillofacial Surgery, Changwon Gyeongsang National University Hospital, 11 Samjeongja-ro, Seongsan-gu, Changwon, 51472 Republic of Korea
| | - Gyu-Jin Rho
- 3Department of Theriogenology and Biotechnology, College of Veterinary Medicine and Research Institute of Life Science, Gyeongsang National University, 15 Jinju-daero 816 beon-gil, Jinju-si, Gyeongsangnam-do 52727 Republic of Korea
| | - June-Ho Byun
- 4Department of Dentistry, Institute of Health Science, Gyeongsang National University School of Medicine, Jinju, 816-15 Republic of Korea
| | - Won-Uk Park
- Department of Dental Technology, Jinju Health College, Jinju, Republic of Korea
| | - Myeong-Gyun Son
- Department of Dentistry, Hanil Hospital, Jinju, Republic of Korea
| | - Bong-Wook Park
- Department of Dentistry, Hanil Hospital, Jinju, Republic of Korea
| | - Hyeon-Jeong Lee
- 3Department of Theriogenology and Biotechnology, College of Veterinary Medicine and Research Institute of Life Science, Gyeongsang National University, 15 Jinju-daero 816 beon-gil, Jinju-si, Gyeongsangnam-do 52727 Republic of Korea
| | - Young-Hoon Kang
- 2Department of Oral and Maxillofacial Surgery, Changwon Gyeongsang National University Hospital, 11 Samjeongja-ro, Seongsan-gu, Changwon, 51472 Republic of Korea.,4Department of Dentistry, Institute of Health Science, Gyeongsang National University School of Medicine, Jinju, 816-15 Republic of Korea
| |
Collapse
|
28
|
Abstract
Several techniques have been established over the last decades to produce three-dimensional (3D) cellular spheroids and each method has its advantages and limitations. The unique self-assembly properties of surface layer (S-layer) proteins have already been applied to a broad range of life science applications. The bacterial S-layer protein SbpA displays a strong antifouling behavior when recrystallized on planar surfaces and offers the opportunity to induce 3D cell aggregation. In this chapter, an S-layer nanointerface is presented as novel ultralow attachment material for the formation of functional spheroids of reproducible sizes. The system is compatible with standard microwell plates and enables long-term 3D cell culture and in situ monitoring of cellular viability. Moreover, this facile and stable biointerface has potential for use in toxicity screening assays and represents an alternative to conventional materials like polyethylene glycol (PEG), agarose, or hydrogel surfaces.
Collapse
|
29
|
Diomede F, Zini N, Pizzicannella J, Merciaro I, Pizzicannella G, D’Orazio M, Piattelli A, Trubiani O. 5-Aza Exposure Improves Reprogramming Process Through Embryoid Body Formation in Human Gingival Stem Cells. Front Genet 2018; 9:419. [PMID: 30349553 PMCID: PMC6186780 DOI: 10.3389/fgene.2018.00419] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 09/06/2018] [Indexed: 12/18/2022] Open
Abstract
Embryoid bodies (EBs) are three-dimensional aggregates formed by pluripotent stem cells, including embryonic stem cells and induced pluripotent stem cells. They are used as an in vitro model to evaluate early extraembryonic tissue formation and differentiation process. In the adult organisms, cell differentiation is controlled and realized through the epigenetic regulation of gene expression, which consists of various mechanisms including DNA methylation. One demethylating agent is represented by 5-Azacytidine (5-Aza), considered able to induce epigenetic changes through gene derepression. Human gingival mesenchymal stem cells (hGMSCs), an easily accessible stem cells population, migrated from neural crest. They are particularly apt as an in vitro study model in regenerative medicine and in systemic diseases. The ability of 5-Aza treatment to induce hGMSCs toward a dedifferentiation stage and in particular versus EBs formation was investigated. For this purpose hGMSCs were treated for 48 h with 5-Aza (5 μM). After treatment, hGMSCs are organized as round 3D structures (EBs-hGMSCs). At light and transmission electron microscopy, the cells at the periphery of EBs-hGMSCs appear elongated, while ribbon-shaped cells and smaller cells with irregular shape surrounded by extracellular matrix were present in the center. By RT-PCR, EBs-hGMSCs expressed specific transcription markers related to the three germ layers as MAP-2, PAX-6 (ectoderm), MSX-1, Flk-1 (mesoderm), GATA-4, and GATA-6 (endoderm). Moreover, in EB-hGMSCs the overexpression of DNMT1 and ACH3 other than the down regulation of p21 was detectable. Immunofluorescence staining also showed a positivity for specific etodermal and mesodermal markers. In conclusion, 5-Aza was able to induce the direct conversion of adult hGMSCs into cells of three embryonic lineages: endoderm, ectoderm, and mesoderm, suggesting their possible application in autologous cell therapy for clinical organ repair.
Collapse
Affiliation(s)
- Francesca Diomede
- Department of Medical, Oral and Biotechnological Sciences, D’Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Nicoletta Zini
- CNR-National Research Council of Italy, IGM, Bologna, Italy
- IRCCS, Rizzoli Orthopaedic Institute, Bologna, Italy
| | | | - Ilaria Merciaro
- Department of Medical, Oral and Biotechnological Sciences, D’Annunzio University of Chieti-Pescara, Chieti, Italy
| | | | - Monica D’Orazio
- Division of Rheumatology and Immunology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Adriano Piattelli
- Department of Medical, Oral and Biotechnological Sciences, D’Annunzio University of Chieti-Pescara, Chieti, Italy
- Chair of Biomaterials Engineering, Catholic University of San Antonio of Murcia (UCAM), Murcia, Spain
| | - Oriana Trubiani
- Department of Medical, Oral and Biotechnological Sciences, D’Annunzio University of Chieti-Pescara, Chieti, Italy
| |
Collapse
|
30
|
Luz AL, Tokar EJ. Pluripotent Stem Cells in Developmental Toxicity Testing: A Review of Methodological Advances. Toxicol Sci 2018; 165:31-39. [PMID: 30169765 PMCID: PMC6111785 DOI: 10.1093/toxsci/kfy174] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Millions of children are born each year with a birth defect. Many of these defects are caused by environmental factors, although the underlying etiology is often unknown. In vivo mammalian models are frequently used to determine if a chemical poses a risk to the developing fetus. However, there are over 80 000 chemicals registered for use in the United States, many of which have undergone little safety testing, necessitating the need for higher-throughput methods to assess developmental toxicity. Pluripotent stem cells (PSCs) are an ideal in vitro model to investigate developmental toxicity as they possess the capacity to differentiate into nearly any cell type in the human body. Indeed, a burst of research has occurred in the field of stem cell toxicology over the past decade, which has resulted in numerous methodological advances that utilize both mouse and human PSCs, as well as cutting-edge technology in the fields of metabolomics, transcriptomics, transgenics, and high-throughput imaging. Here, we review the wide array of approaches used to detect developmental toxicants, suggest areas for further research, and highlight critical aspects of stem cell biology that should be considered when utilizing PSCs in developmental toxicity testing.
Collapse
Affiliation(s)
- Anthony L Luz
- Stem Cell Toxicology Group, National Toxicology Program Laboratory, Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709
| | - Erik J Tokar
- Stem Cell Toxicology Group, National Toxicology Program Laboratory, Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709
| |
Collapse
|
31
|
Moritani Y, Usui M, Sano K, Nakazawa K, Hanatani T, Nakatomi M, Iwata T, Sato T, Ariyoshi W, Nishihara T, Nakashima K. Spheroid culture enhances osteogenic potential of periodontal ligament mesenchymal stem cells. J Periodontal Res 2018; 53:870-882. [PMID: 29900548 DOI: 10.1111/jre.12577] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2018] [Indexed: 12/13/2022]
Abstract
OBJECTIVE AND BACKGROUND Human periodontal ligament mesenchymal stem cells (hPDLMSCs) are reported to be responsible for homeostasis and regeneration of periodontal tissue. Although hPDLMSCs are commonly cultured in monolayers, monolayer cultures have been reported as inferior to 3-dimensional cultures such as spheroids, which are spherical clusters of cells formed by self-assembly. The aim of this study was to examine the osteogenic phenotype of spheroids of hPDLMSCs, compared with monolayer cultures of hPDLMSC, in vitro and in vivo. MATERIAL AND METHODS Spheroids were formed using microwell chips that were tagged with polyethylene glycol. Mesenchymal stem cell (MSC) markers in hPDLMSC spheroids were examined by flow cytometer. Real-time polymerase chain reaction analysis was examined to measure the expressions of stemness markers and osteogenesis-related genes in monolayer and spheroid-cultured hPDLMSCs. Immunofluorescence analysis was performed to confirm protein expressions of stemness markers in PDLMSC spheroids. Nodule formation assay, alkaline phosphatase (ALP) activity assay and transplantation assay in a mouse calvarial defect model were performed to confirm the osteogenic potential of hPDLMSC spheroids. To elucidate the mechanism of spheroid culture enhanced osteogenesis in hPDLMSCs with osteoinductive medium (OIM), a small interfering RNA (siRNA) assay targeted with secreted frizzled-related protein 3 (SFRP3) was examined. The levels of SFRP3 expression in monolayer and spheroid-cultured hPDLMSCs with OIM were measured by real-time polymerase chain reaction and western blotting analysis. ALP gene expression and ALP activity were examined in SFRP3-deficient hPDLMSC spheroids. RESULTS The hPDLMSC spheroids expressed MSC markers, which were similar to hPDLMSCs grown in monolayer cultures. Intriguingly, the protein and mRNA expressions of transcription factors that regulate "stemness" were significantly increased in hPDLMSC spheroids, compared with hPDLMSCs in monolayer cultures. Nodule formation by hPDLMSCs was significantly increased in spheroid cultures grown with OIM, compared with monolayer-cultured hPDLMSCs. ALP activity and expression of osteogenesis-related genes were also significantly enhanced in hPDLMSC spheroids, compared with monolayer cultures. Treatment with hPDLMSC spheroids significantly enhanced new bone formation in a murine calvarial defect model, compared with hPDLMSCs in monolayer culture. Finally, to elucidate mechanisms by which spheroid culture enhances ALP activation in hPDLMSCs grown with OIM, an siRNA assay was used to manipulate expression of SFRP3, a Wnt signaling antagonist. Knockdown of SFRP3 suppressed ALP gene expression in hPDLMSCs grown in OIM; further, it suppressed ALP activity in spheroid culture. These data suggest that the enhancement of osteogenic potential in hPDLMSC spheroids is regulated through SFRP3-mediated ALP activation. CONCLUSION Spheroid cultures of hPDLMSCs may be a novel and useful tool in regenerative medicine.
Collapse
Affiliation(s)
- Y Moritani
- Division of Periodontology, Department of Oral Function, Kyushu Dental University, Kitakyushu, Japan
| | - M Usui
- Division of Periodontology, Department of Oral Function, Kyushu Dental University, Kitakyushu, Japan
| | - K Sano
- Division of Periodontology, Department of Oral Function, Kyushu Dental University, Kitakyushu, Japan
| | - K Nakazawa
- Department of Life and Environment Engineering, The University of Kitakyushu, Kitakyushu, Japan
| | - T Hanatani
- Division of Periodontology, Department of Oral Function, Kyushu Dental University, Kitakyushu, Japan
| | - M Nakatomi
- Division of Anatomy, Department of Health Promotion, Kyushu Dental University, Kitakyushu, Japan
| | - T Iwata
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University (TWIns), Tokyo, Japan
| | - T Sato
- Department of Oral and Maxillofacial Surgery, Saitama Medical University, Saitama, Japan
| | - W Ariyoshi
- Division of Infections and Molecular Biology, Department of Health Promotion, Kyushu Dental University, Kitakyushu, Japan
| | - T Nishihara
- Division of Infections and Molecular Biology, Department of Health Promotion, Kyushu Dental University, Kitakyushu, Japan
| | - K Nakashima
- Division of Periodontology, Department of Oral Function, Kyushu Dental University, Kitakyushu, Japan
| |
Collapse
|
32
|
Lempereur A, Canto PY, Richard C, Martin S, Thalgott J, Raymond K, Lebrin F, Drevon C, Jaffredo T. The TGFβ pathway is a key player for the endothelial-to-hematopoietic transition in the embryonic aorta. Dev Biol 2017; 434:292-303. [PMID: 29253505 DOI: 10.1016/j.ydbio.2017.12.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 12/07/2017] [Accepted: 12/08/2017] [Indexed: 12/30/2022]
Abstract
The embryonic aorta produces hematopoietic stem and progenitor cells from a hemogenic endothelium localized in the aortic floor through an endothelial to hematopoietic transition. It has been long proposed that the Bone Morphogenetic Protein (BMP)/Transforming Growth Factor ß (TGFß) signaling pathway was implicated in aortic hematopoiesis but the very nature of the signal was unknown. Here, using thorough expression analysis of the BMP/TGFß signaling pathway members in the endothelial and hematopoietic compartments of the aorta at pre-hematopoietic and hematopoietic stages, we show that the TGFß pathway is preferentially balanced with a prominent role of Alk1/TgfßR2/Smad1 and 5 on both chicken and mouse species. Functional analysis using embryonic stem cells mutated for Acvrl1 revealed an enhanced propensity to produce hematopoietic cells. Collectively, we reveal that TGFß through the Alk1/TgfßR2 receptor axis is acting on endothelial cells to produce hematopoiesis.
Collapse
Affiliation(s)
- A Lempereur
- Sorbonne Universités, UPMC Univ Paris 06, IBPS, CNRS UMR7622, Inserm U 1156, Laboratoire de Biologie du Développement, 75005 Paris, France
| | - P Y Canto
- Sorbonne Universités, UPMC Univ Paris 06, IBPS, CNRS UMR7622, Inserm U 1156, Laboratoire de Biologie du Développement, 75005 Paris, France
| | - C Richard
- Sorbonne Universités, UPMC Univ Paris 06, IBPS, CNRS UMR7622, Inserm U 1156, Laboratoire de Biologie du Développement, 75005 Paris, France
| | - S Martin
- CNRS UMR 7241/INSERM U1050, Center for Interdisciplinary Research in Biology, Collège de France, 11 Place Marcelin Berthelot, 75231 Paris CEDEX 05, France; MEMOLIFE Laboratory of Excellence and Paris Sciences et Lettres Research University, France
| | - J Thalgott
- Einthoven Laboratory for Experimental Vascular Medicine, Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, The Netherlands
| | - K Raymond
- Einthoven Laboratory for Experimental Vascular Medicine, Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, The Netherlands
| | - F Lebrin
- CNRS UMR 7241/INSERM U1050, Center for Interdisciplinary Research in Biology, Collège de France, 11 Place Marcelin Berthelot, 75231 Paris CEDEX 05, France; Einthoven Laboratory for Experimental Vascular Medicine, Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, The Netherlands; MEMOLIFE Laboratory of Excellence and Paris Sciences et Lettres Research University, France
| | - C Drevon
- Sorbonne Universités, UPMC Univ Paris 06, IBPS, CNRS UMR7622, Inserm U 1156, Laboratoire de Biologie du Développement, 75005 Paris, France
| | - T Jaffredo
- Sorbonne Universités, UPMC Univ Paris 06, IBPS, CNRS UMR7622, Inserm U 1156, Laboratoire de Biologie du Développement, 75005 Paris, France.
| |
Collapse
|
33
|
Son B, Kim JA, Cho S, Jeong GJ, Kim BS, Hwang NS, Park TH. Lineage Specific Differentiation of Magnetic Nanoparticle-Based Size Controlled Human Embryoid Body. ACS Biomater Sci Eng 2017; 3:1719-1729. [DOI: 10.1021/acsbiomaterials.7b00141] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Boram Son
- School
of Chemical and Biological Engineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 151-742, Republic of Korea
| | - Jeong Ah Kim
- Biomedical
Omics Group, Korea Basic Science Institute, Cheongju, Chungbuk 28119, Republic of Korea
| | - Sungwoo Cho
- School
of Chemical and Biological Engineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 151-742, Republic of Korea
| | - Gun-Jae Jeong
- School
of Chemical and Biological Engineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 151-742, Republic of Korea
| | - Byung Soo Kim
- School
of Chemical and Biological Engineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 151-742, Republic of Korea
| | - Nathaniel S. Hwang
- School
of Chemical and Biological Engineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 151-742, Republic of Korea
| | - Tai Hyun Park
- School
of Chemical and Biological Engineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 151-742, Republic of Korea
| |
Collapse
|
34
|
Yang J, Wu C, Stefanescu I, Horowitz A. Analysis of Retinoic Acid-induced Neural Differentiation of Mouse Embryonic Stem Cells in Two and Three-dimensional Embryoid Bodies. J Vis Exp 2017. [PMID: 28518115 DOI: 10.3791/55621] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Mouse embryonic stem cells (ESCs) isolated from the inner mass of the blastocyst (typically at day E3.5), can be used as in vitro model system for studying early embryonic development. In the absence of leukemia inhibitory factor (LIF), ESCs differentiate by default into neural precursor cells. They can be amassed into a three dimensional (3D) spherical aggregate termed embryoid body (EB) due to its similarity to the early stage embryo. EBs can be seeded on fibronectin-coated coverslips, where they expand by growing two dimensional (2D) extensions, or implanted in 3D collagen matrices where they continue growing as spheroids, and differentiate into the three germ layers: endodermal, mesodermal, and ectodermal. The 3D collagen culture mimics the in vivo environment more closely than the 2D EBs. The 2D EB culture facilitates analysis by immunofluorescence and immunoblotting to track differentiation. We have developed a two-step neural differentiation protocol. In the first step, EBs are generated by the hanging-drop technique, and, simultaneously, are induced to differentiate by exposure to retinoic acid (RA). In the second step, neural differentiation proceeds in a 2D or 3D format in the absence of RA.
Collapse
Affiliation(s)
- Junning Yang
- Department of Medicine, Cardeza Vascular Research Center, Sidney Kimmel Medical College, Thomas Jefferson University
| | - Chuanshen Wu
- Department of Molecular Cardiology, Cleveland Clinic Foundation
| | - Ioana Stefanescu
- Department of Medicine, Cardeza Vascular Research Center, Sidney Kimmel Medical College, Thomas Jefferson University
| | - Arie Horowitz
- Department of Medicine, Cardeza Vascular Research Center, Sidney Kimmel Medical College, Thomas Jefferson University; Department of Cancer Biology, Cardeza Vascular Research Center, Sidney Kimmel Medical College, Thomas Jefferson University;
| |
Collapse
|
35
|
Kandasamy M, Roll L, Langenstroth D, Brüstle O, Faissner A. Glycoconjugates reveal diversity of human neural stem cells (hNSCs) derived from human induced pluripotent stem cells (hiPSCs). Cell Tissue Res 2017; 368:531-549. [DOI: 10.1007/s00441-017-2594-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 02/23/2017] [Indexed: 12/20/2022]
|
36
|
Deng Y, Yang Y, Wei S. Peptide-Decorated Nanofibrous Niche Augments In Vitro Directed Osteogenic Conversion of Human Pluripotent Stem Cells. Biomacromolecules 2017; 18:587-598. [DOI: 10.1021/acs.biomac.6b01748] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Yi Deng
- School
of Chemical Engineering, Sichuan University, Chengdu 610065, China
| | - Yuanyi Yang
- Department
of Materials Engineering, Sichuan College of Architectural Technology, Deyang 618000, China
| | | |
Collapse
|
37
|
Rashidi H, Hay D. Generation and Application of 3DCulture Systems in Human Drug Discovery and Medicine. STEM CELLS IN TOXICOLOGY AND MEDICINE 2016:265-287. [DOI: 10.1002/9781119135449.ch14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
38
|
Sharma VS, Khalife R, Tostoes R, Leung L, Kinsella R, Ruban L, Veraitch FS. Early retinal differentiation of human pluripotent stem cells in microwell suspension cultures. Biotechnol Lett 2016; 39:339-350. [PMID: 27812821 PMCID: PMC5247545 DOI: 10.1007/s10529-016-2244-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 10/27/2016] [Indexed: 11/17/2022]
Abstract
Objective To develop a microwell suspension platform for the adaption of attached stem cell differentiation protocols into mixed suspension culture. Results We adapted an adherent protocol for the retinal differentiation of human induced pluripotent stem cells (hiPSCs) using a two-step protocol. Establishing the optimum embryoid body (EB) starting size and shaking speed resulted in the translation of the original adherent process into suspension culture. Embryoid bodies expanded in size as the culture progressed resulting in the expression of characteristic markers of early (Rx, Six and Otx2) and late (Crx, Nrl and Rhodopsin) retinal differentiation. The new process also eliminated the use of matrigel, an animal-derived extracellular matrix coating. Conclusions Shaking microwells offer a fast and cost-effective method for proof-of-concept studies to establish whether pluripotent stem cell differentiation processes can be translated into mixed suspension culture. Electronic supplementary material The online version of this article (doi:10.1007/s10529-016-2244-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Vishal S Sharma
- Department of Biochemical Engineering, University College London, Gower Street, London, WC1E 6BT, UK
| | - Rana Khalife
- Department of Biochemical Engineering, University College London, Gower Street, London, WC1E 6BT, UK
| | - Rui Tostoes
- Department of Biochemical Engineering, University College London, Gower Street, London, WC1E 6BT, UK
| | - Leonard Leung
- Department of Biochemical Engineering, University College London, Gower Street, London, WC1E 6BT, UK
| | - Rose Kinsella
- Department of Biochemical Engineering, University College London, Gower Street, London, WC1E 6BT, UK
| | - Ludmilla Ruban
- Department of Biochemical Engineering, University College London, Gower Street, London, WC1E 6BT, UK
| | - Farlan S Veraitch
- Department of Biochemical Engineering, University College London, Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
39
|
Wu HW, Hsiao YH, Chen CC, Yet SF, Hsu CH. A PDMS-Based Microfluidic Hanging Drop Chip for Embryoid Body Formation. Molecules 2016; 21:molecules21070882. [PMID: 27399655 PMCID: PMC6272923 DOI: 10.3390/molecules21070882] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Revised: 06/27/2016] [Accepted: 06/29/2016] [Indexed: 01/08/2023] Open
Abstract
The conventional hanging drop technique is the most widely used method for embryoid body (EB) formation. However, this method is labor intensive and limited by the difficulty in exchanging the medium. Here, we report a microfluidic chip-based approach for high-throughput formation of EBs. The device consists of microfluidic channels with 6 × 12 opening wells in PDMS supported by a glass substrate. The PDMS channels were fabricated by replicating polydimethyl-siloxane (PDMS) from SU-8 mold. The droplet formation in the chip was tested with different hydrostatic pressures to obtain optimal operation pressures for the wells with 1000 μm diameter openings. The droplets formed at the opening wells were used to culture mouse embryonic stem cells which could subsequently developed into EBs in the hanging droplets. This device also allows for medium exchange of the hanging droplets making it possible to perform immunochemistry staining and characterize EBs on chip.
Collapse
Affiliation(s)
- Huei-Wen Wu
- Institutes of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Zhunan 35053, Taiwan.
| | - Yi-Hsing Hsiao
- Institute of Nano Engineering and MicroSystems, National Tsing Hua University, Hsinchu 30013, Taiwan.
| | - Chih-Chen Chen
- Institute of Nano Engineering and MicroSystems, National Tsing Hua University, Hsinchu 30013, Taiwan.
- Department of Power Mechanical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan.
| | - Shaw-Fang Yet
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan 35053, Taiwan.
| | - Chia-Hsien Hsu
- Institutes of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Zhunan 35053, Taiwan.
- Institute of Nano Engineering and MicroSystems, National Tsing Hua University, Hsinchu 30013, Taiwan.
| |
Collapse
|
40
|
Abstract
Soluble morphogen gradients have long been studied in the context of heart specification and patterning. However, recent data have begun to challenge the notion that long-standing in vivo observations are driven solely by these gradients alone. Evidence from multiple biological models, from stem cells to ex vivo biophysical assays, now supports a role for mechanical forces in not only modulating cell behavior but also inducing it de novo in a process termed mechanotransduction. Structural proteins that connect the cell to its niche, for example, integrins and cadherins, and that couple to other growth factor receptors, either directly or indirectly, seem to mediate these changes, although specific mechanistic details are still being elucidated. In this review, we summarize how the wingless (Wnt), transforming growth factor-β, and bone morphogenetic protein signaling pathways affect cardiomyogenesis and then highlight the interplay between each pathway and mechanical forces. In addition, we will outline the role of integrins and cadherins during cardiac development. For each, we will describe how the interplay could change multiple processes during cardiomyogenesis, including the specification of undifferentiated cells, the establishment of heart patterns to accomplish tube and chamber formation, or the maturation of myocytes in the fully formed heart.
Collapse
Affiliation(s)
- Cassandra L Happe
- From the Department of Bioengineering, University of California, San Diego, La Jolla; and Sanford Consortium for Regenerative Medicine, La Jolla, CA
| | - Adam J Engler
- From the Department of Bioengineering, University of California, San Diego, La Jolla; and Sanford Consortium for Regenerative Medicine, La Jolla, CA.
| |
Collapse
|
41
|
Lee ES, Park J, Wang J, Lee H, Hwang NS. Osteogenic commitment of human induced pluripotent stem cell-derived mesenchymal progenitor-like cells on biomimetic scaffolds. J IND ENG CHEM 2016. [DOI: 10.1016/j.jiec.2016.03.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
42
|
Vrij EJ, Espinoza S, Heilig M, Kolew A, Schneider M, van Blitterswijk CA, Truckenmüller RK, Rivron NC. 3D high throughput screening and profiling of embryoid bodies in thermoformed microwell plates. LAB ON A CHIP 2016; 16:734-742. [PMID: 26775648 DOI: 10.1039/c5lc01499a] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
3D organoids using stem cells to study development and disease are now widespread. These models are powerful to mimic in vivo situations but are currently associated with high variability and low throughput. For biomedical research, platforms are thus necessary to increase reproducibility and allow high-throughput screens (HTS). Here, we introduce a microwell platform, integrated in standard culture plates, for functional HTS. Using micro-thermoforming, we form round-bottom microwell arrays from optically clear cyclic olefin polymer films, and assemble them with bottom-less 96-well plates. We show that embryonic stem cells aggregate faster and more reproducibly (centricity, circularity) as compared to a state-of-the-art microwell array. We then run a screen of a chemical library to direct differentiation into primitive endoderm (PrE) and, using on-chip high content imaging (HCI), we identify molecules, including regulators of the cAMP pathway, regulating tissue size, morphology and PrE gene activity. We propose that this platform will benefit to the systematic study of organogenesis in vitro.
Collapse
Affiliation(s)
- E J Vrij
- Merln Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Dehdilani N, Shamsasenjan K, Movassaghpour A, Akbarzadehlaleh P, Amoughli Tabrizi B, Parsa H, Sabagi F. Improved Survival and Hematopoietic Differentiation of Murine Embryonic Stem Cells on Electrospun Polycaprolactone Nanofiber. CELL JOURNAL 2016; 17:629-38. [PMID: 26862522 PMCID: PMC4746413 DOI: 10.22074/cellj.2016.3835] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2014] [Accepted: 11/13/2014] [Indexed: 11/04/2022]
Abstract
OBJECTIVE Three-dimensional (3D) biomimetic nanofiber scaffolds have widespread ap- plications in biomedical tissue engineering. They provide a suitable environment for cel- lular adhesion, survival, proliferation and differentiation, guide new tissue formation and development, and are one of the outstanding goals of tissue engineering. Electrospinning has recently emerged as a leading technique for producing biomimetic scaffolds with mi- cro to nanoscale topography and a high porosity similar to the natural extracellular matrix (ECM). These scaffolds are comprised of synthetic and natural polymers for tissue engi- neering applications. Several kinds of cells such as human embryonic stem cells (hESCs) and mouse ESCs (mESCs) have been cultured and differentiated on nanofiber scaffolds. mESCs can be induced to differentiate into a particular cell lineage when cultured as em- bryoid bodies (EBs) on nano-sized scaffolds. MATERIALS AND METHODS We cultured mESCs (2500 cells/100 µl) in 96-well plates with knockout Dulbecco's modified eagle medium (DMEM-KO) and Roswell Park Memorial Institute-1640 (RPMI-1640), both supplemented with 20% ESC grade fetal bovine serum (FBS) and essential factors in the presence of leukemia inhibitory factor (LIF). mESCs were seeded at a density of 2500 cells/100 µl onto electrospun polycaprolactone (PCL) nanofibers in 96-well plates. The control group comprised mESCs grown on tissue cul- ture plates (TCP) at a density of 2500 cells/100 µl. Differentiation of mESCs into mouse hematopoietic stem cells (mHSCs) was performed by stem cell factor (SCF), interleukin-3 (IL-3), IL-6 and Fms-related tyrosine kinase ligand (Flt3-L) cytokines for both the PCL and TCP groups. We performed an experimental study of mESCs differentiation. RESULTS PCL was compared to conventional TCP for survival and differentiation of mESCs to mHSCs. There were significantly more mESCs in the PCL group. Flowcyto- metric analysis revealed differences in hematopoietic differentiation between the PCL and TCP culture systems. There were more CD34+(Sca1+) and CD133+cells subpopulations in the PCL group compared to the conventional TCP culture system. CONCLUSION The nanofiber scaffold, as an effective surface, improves survival and differentiation of mESCs into mHSCs compared to gelatin coated TCP. More studies are necessary to understand how the topographical features of electrospun fibers af- fect cell growth and behavior. This can be achieved by designing biomimetic scaffolds for tissue engineering.
Collapse
Affiliation(s)
- Nima Dehdilani
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Karim Shamsasenjan
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Iran Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Aliakbar Movassaghpour
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parvin Akbarzadehlaleh
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bahram Amoughli Tabrizi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Hamed Parsa
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Sabagi
- Iran Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| |
Collapse
|
44
|
Uniform Embryoid Body Production and Enhanced Mesendoderm Differentiation with Murine Embryonic Stem Cells in a Rotary Suspension Bioreactor. Methods Mol Biol 2016; 1502:63-75. [PMID: 27115505 DOI: 10.1007/7651_2016_354] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Embryonic stem cells (ESCs) are capable of differentiating into almost all cell types in vitro and hold great promise for drug screening, developmental studies and have a huge potential in many therapeutic areas. ESCs can aggregate to form embryoid body (EB) in static suspension culture by spontaneous differentiation, which resembles an intact embryo; while static suspension culture cannot prevent agglomeration of cells and offers little control over the size and shape of EBs, it results in aggregation of EBs into large, irregular masses, which prejudice the efficiency of differentiation of cells. Recently, bioreactor-based platforms have been shown to not only offer a beneficial effect on increasing diffusion of nutrients and oxygen which promotes cell viability and proliferation but also display local biomechanical properties (e.g., low fluid shear stresses and hydrodynamic force) in tissue development and organogenesis. This chapter describes a protocol for using a rotary suspension bioreactor to produce embryoid bodies and process the differentiation of mouse embryonic stem cells (mESCs), and to assess the efficiency of EB differentiation in the bioreactor by real-time PCR and immunostaining.
Collapse
|
45
|
Rapid Fabrication of Hydrogel Microstructures Using UV-Induced Projection Printing. MICROMACHINES 2015. [DOI: 10.3390/mi6121464] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
46
|
Sivakumar M, Dineshshankar J, Sunil PM, Nirmal RM, Sathiyajeeva J, Saravanan B, Senthileagappan AR. Stem cells: An insight into the therapeutic aspects from medical and dental perspectives. J Pharm Bioallied Sci 2015; 7:S361-71. [PMID: 26538878 PMCID: PMC4606620 DOI: 10.4103/0975-7406.163453] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The recent advancements in the field of stem cell (SC) biology have increased the hope of achieving the definitive treatments for the diseases which are now considered incurable such as diabetes, Parkinson's disease and other chronic long standing conditions. To achieve this possibility, it is necessary to understand the basic concepts of SC biology to utilize in various advanced techniques of regenerative medicine including tissue engineering and gene therapy. This article highlights the types of SCs available and their therapeutic capacity in regenerative medical and dental fields.
Collapse
Affiliation(s)
- Muniapillai Sivakumar
- Department of Oral Pathology and Microbiology, Madha Dental College and Hospital, Chennai, Tamil Nadu, India
| | - Janardhanam Dineshshankar
- Department of Oral Pathology and Microbiology, Vivekanandha Dental College for Women, Tiruchengode, Namakkal, Tamil Nadu, India
| | - P M Sunil
- Department of Oral Pathology and Microbiology, Sree Anjaneya Institute of Dental Sciences, Calicut, Kerala, India
| | - R Madhavan Nirmal
- Department of Oral Pathology and Microbiology, Rajah Muthiah Dental College and Hospital, Annamalai University, Chidambaram, Tamil Nadu, India
| | - J Sathiyajeeva
- Department of Oral Pathology and Microbiology, Thai Moogambigai Dental College and Hospital, Chennai, Tamil Nadu, India
| | - Balasubramanian Saravanan
- Department of Oral and Maxillofacial Surgery, Madha Dental College and Hospital, Kundrathur, Chennai, Tamil Nadu, India
| | - A R Senthileagappan
- Department of Pedodontics, Chettinad Dental College and Research Institute, Chettinad Health City, Chennai, Tamil Nadu, India
| |
Collapse
|
47
|
Ouyang L, Yao R, Mao S, Chen X, Na J, Sun W. Three-dimensional bioprinting of embryonic stem cells directs highly uniform embryoid body formation. Biofabrication 2015; 7:044101. [DOI: 10.1088/1758-5090/7/4/044101] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
48
|
Blaber EA, Finkelstein H, Dvorochkin N, Sato KY, Yousuf R, Burns BP, Globus RK, Almeida EAC. Microgravity Reduces the Differentiation and Regenerative Potential of Embryonic Stem Cells. Stem Cells Dev 2015; 24:2605-21. [PMID: 26414276 PMCID: PMC4652210 DOI: 10.1089/scd.2015.0218] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Mechanical unloading in microgravity is thought to induce tissue degeneration by various mechanisms, including inhibition of regenerative stem cell differentiation. To address this hypothesis, we investigated the effects of microgravity on early lineage commitment of mouse embryonic stem cells (mESCs) using the embryoid body (EB) model of tissue differentiation. We found that exposure to microgravity for 15 days inhibits mESC differentiation and expression of terminal germ layer lineage markers in EBs. Additionally, microgravity-unloaded EBs retained stem cell self-renewal markers, suggesting that mechanical loading at Earth's gravity is required for normal differentiation of mESCs. Finally, cells recovered from microgravity-unloaded EBs and then cultured at Earth's gravity showed greater stemness, differentiating more readily into contractile cardiomyocyte colonies. These results indicate that mechanical unloading of stem cells in microgravity inhibits their differentiation and preserves stemness, possibly providing a cellular mechanistic basis for the inhibition of tissue regeneration in space and in disuse conditions on earth.
Collapse
Affiliation(s)
- Elizabeth A Blaber
- 1 Space Biosciences Division, NASA Ames Research Center , Moffett Field, California.,2 School of Biotechnology and Biomolecular Sciences, University of New South Wales , Sydney, Australia
| | - Hayley Finkelstein
- 1 Space Biosciences Division, NASA Ames Research Center , Moffett Field, California
| | - Natalya Dvorochkin
- 1 Space Biosciences Division, NASA Ames Research Center , Moffett Field, California
| | - Kevin Y Sato
- 3 FILMSS Wyle, Space Biology, NASA Ames Research Center , Moffett Field, California
| | - Rukhsana Yousuf
- 1 Space Biosciences Division, NASA Ames Research Center , Moffett Field, California
| | - Brendan P Burns
- 2 School of Biotechnology and Biomolecular Sciences, University of New South Wales , Sydney, Australia .,4 Australian Centre for Astrobiology, University of New South Wales , Sydney, Australia
| | - Ruth K Globus
- 1 Space Biosciences Division, NASA Ames Research Center , Moffett Field, California
| | - Eduardo A C Almeida
- 1 Space Biosciences Division, NASA Ames Research Center , Moffett Field, California
| |
Collapse
|
49
|
Park D, Lim J, Park JY, Lee SH. Concise Review: Stem Cell Microenvironment on a Chip: Current Technologies for Tissue Engineering and Stem Cell Biology. Stem Cells Transl Med 2015; 4:1352-68. [PMID: 26450425 DOI: 10.5966/sctm.2015-0095] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 07/29/2015] [Indexed: 01/09/2023] Open
Abstract
UNLABELLED Stem cells have huge potential in many therapeutic areas. With conventional cell culture methods, however, it is difficult to achieve in vivo-like microenvironments in which a number of well-controlled stimuli are provided for growing highly sensitive stem cells. In contrast, microtechnology-based platforms offer advantages of high precision, controllability, scalability, and reproducibility, enabling imitation of the complex physiological context of in vivo. This capability may fill the gap between the present knowledge about stem cells and that required for clinical stem cell-based therapies. We reviewed the various types of microplatforms on which stem cell microenvironments are mimicked. We have assigned the various microplatforms to four categories based on their practical uses to assist stem cell biologists in using them for research. In particular, many examples are given of microplatforms used for the production of embryoid bodies and aggregates of stem cells in vitro. We also categorized microplatforms based on the types of factors controlling the behaviors of stem cells. Finally, we outline possible future directions for microplatform-based stem cell research, such as research leading to the production of well-defined environments for stem cells to be used in scaled-up systems or organs-on-a-chip, the regulation of induced pluripotent stem cells, and the study of the genetic states of stem cells on microplatforms. SIGNIFICANCE Stem cells are highly sensitive to a variety of physicochemical cues, and their fate can be easily altered by a slight change of environment; therefore, systematic analysis and discrimination of the extracellular signals and intracellular pathways controlling the fate of cells and experimental realization of sensitive and controllable niche environments are critical. This review introduces diverse microplatforms to provide in vitro stem cell niches. Microplatforms could control microenvironments around cells and have recently attracted much attention in biology including stem cell research. These microplatforms and the future directions of stem cell microenvironment are described.
Collapse
Affiliation(s)
- DoYeun Park
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
| | - Jaeho Lim
- School of Biomedical Engineering, College of Health Science, Korea University, Seoul, Republic of Korea
| | - Joong Yull Park
- School of Mechanical Engineering, College of Engineering, Chung-ang University, Seoul, Republic of Korea
| | - Sang-Hoon Lee
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea School of Biomedical Engineering, College of Health Science, Korea University, Seoul, Republic of Korea
| |
Collapse
|
50
|
Chojnowski A, Ong PF, Wong ESM, Lim JSY, Mutalif RA, Navasankari R, Dutta B, Yang H, Liow YY, Sze SK, Boudier T, Wright GD, Colman A, Burke B, Stewart CL, Dreesen O. Progerin reduces LAP2α-telomere association in Hutchinson-Gilford progeria. eLife 2015; 4. [PMID: 26312502 PMCID: PMC4565980 DOI: 10.7554/elife.07759] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 08/23/2015] [Indexed: 12/12/2022] Open
Abstract
Hutchinson-Gilford progeria (HGPS) is a premature ageing syndrome caused by a mutation in LMNA, resulting in a truncated form of lamin A called progerin. Progerin triggers loss of the heterochromatic marker H3K27me3, and premature senescence, which is prevented by telomerase. However, the mechanism how progerin causes disease remains unclear. Here, we describe an inducible cellular system to model HGPS and find that LAP2α (lamina-associated polypeptide-α) interacts with lamin A, while its interaction with progerin is significantly reduced. Super-resolution microscopy revealed that over 50% of telomeres localize to the lamina and that LAP2α association with telomeres is impaired in HGPS. This impaired interaction is central to HGPS since increasing LAP2α levels rescues progerin-induced proliferation defects and loss of H3K27me3, whereas lowering LAP2 levels exacerbates progerin-induced defects. These findings provide novel insights into the pathophysiology underlying HGPS, and how the nuclear lamina regulates proliferation and chromatin organization. DOI:http://dx.doi.org/10.7554/eLife.07759.001 Hutchinson-Gilford Progeria Syndrome (HGPS) is a rare genetic disease in which individuals age prematurely. Newborns appear normal at birth, but start ageing rapidly when they are around a year old. Symptoms of the disease include stunted growth and joint stiffness, and individuals often die of heart failure during their teens. A mutated version of a protein called lamin A causes HGPS; this mutant is known as progerin. In cells that produce progerin, the ‘telomeres’ that protect the ends of chromosomes (the structures that contain most of the cell's DNA) from damage, are unusually short. Every time a cell divides, the telomeres get shorter. If they get too short, the DNA is damaged and the cell stops dividing and enters a state known as senescence. HGPS affects some of the tissues in the body more severely than others, and these tissues tend to produce high levels of progerin. By gradually raising the levels of progerin in human cells, Chojnowski et al. found that DNA damage and cell senescence only occur when the amount of progerin in a cell exceeds a particular threshold. Moreover, the expression of telomerase—a complex that can elongate telomeres—prevented progerin-induced DNA damage and premature senescence. To find out how progerin affects cells, Chojnowski et al. compared how lamin A and progerin interact with other proteins. This revealed that progerin interacts with a protein called LAP2α more weakly than lamin A. LAP2α normally associates with telomeres, but using super-high resolution microscopy, Chojnowski et al. observed that this association is less likely to occur in the cells of people with HGPS. Importantly, increasing the amount of LAP2α in progerin-expressing cells prevented DNA damage and senescence and enabled these cells to continue dividing. Chojnowski et al. propose that in HGPS, the weak interaction between LAP2α and progerin disrupts how LAP2α interacts with telomeres, which prevents cells from dividing. Understanding this process may help to design new ways of treating HGPS, and may also help us to understand other diseases that are caused by mutations in lamin proteins. DOI:http://dx.doi.org/10.7554/eLife.07759.002
Collapse
Affiliation(s)
- Alexandre Chojnowski
- Developmental and Regenerative Biology, Institute of Medical Biology, Singapore, Singapore
| | - Peh Fern Ong
- Cellular Ageing, Institute of Medical Biology, Singapore, Singapore
| | - Esther S M Wong
- Developmental and Regenerative Biology, Institute of Medical Biology, Singapore, Singapore
| | - John S Y Lim
- Microscopy Unit, Institute of Medical Biology, Singapore, Singapore
| | - Rafidah A Mutalif
- Developmental and Regenerative Biology, Institute of Medical Biology, Singapore, Singapore
| | - Raju Navasankari
- Developmental and Regenerative Biology, Institute of Medical Biology, Singapore, Singapore
| | - Bamaprasad Dutta
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Henry Yang
- Bioinformatics Core, Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Yi Y Liow
- Developmental and Regenerative Biology, Institute of Medical Biology, Singapore, Singapore
| | - Siu K Sze
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Thomas Boudier
- Bioinformatics Institute, IPAL UMI 2955, Singapore, Singapore
| | - Graham D Wright
- Microscopy Unit, Institute of Medical Biology, Singapore, Singapore
| | - Alan Colman
- Stem Cell Disease Models, Institute of Medical Biology, Singapore, Singapore
| | - Brian Burke
- Nuclear Dynamics and Architecture, Institute of Medical Biology, Singapore, Singapore
| | - Colin L Stewart
- Developmental and Regenerative Biology, Institute of Medical Biology, Singapore, Singapore
| | - Oliver Dreesen
- Cellular Ageing, Institute of Medical Biology, Singapore, Singapore
| |
Collapse
|