1
|
Yang M, You D, Liu G, Lu Y, Yang G, O'Brien T, Henshall DC, Hardiman O, Cai L, Liu M, Shen S. Polyethyleneimine facilitates the growth and electrophysiological characterization of iPSC-derived motor neurons. Sci Rep 2024; 14:26106. [PMID: 39478194 PMCID: PMC11525838 DOI: 10.1038/s41598-024-77710-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 10/24/2024] [Indexed: 11/02/2024] Open
Abstract
Induced pluripotent stem cell (iPSC) technology, in combination with electrophysiological characterization via multielectrode array (MEA), has facilitated the utilization of iPSC-derived motor neurons (iPSC-MNs) as highly valuable models for underpinning pathogenic mechanisms and developing novel therapeutic interventions for motor neuron diseases (MNDs). However, the challenge of MN adherence to the MEA plate and the heterogeneity presented in iPSC-derived cultures raise concerns about the reproducibility of the findings obtained from these cellular models. We discovered that one novel factor modulating the electrophysiological activity of iPSC-MNs is the extracellular matrix (ECM) used in the coating to support in vitro growth, differentiation and maturation of iPSC-MNs. The current study showed that two coating conditions, namely, Poly-L-ornithine/Matrigel (POM) and Polyethyleneimine (PEI) strongly promoted attachment of iPSC-MNs on MEA culture dishes compared to three other coating conditions, and both facilitated the maturation of iPSC-MNs as characterized by the detection of extensive electrophysiological activities from the MEA plates. POM coating accelerated the maturation of the iPSC-MNs for up to 5 weeks, which suits modeling of neurodevelopmental disorders. However, the application of PEI resulted in more even distribution of the MNs on the culture dish and reduced variability of electrophysiological signals from the iPSC-MNs in 7-week cultures, which permitted the detection of enhanced excitability in iPSC-MNs from patients with amyotrophic lateral sclerosis (ALS). This study provides a comprehensive comparison of five coating conditions and offers POM and PEI as favorable coatings for in vitro modeling of neurodevelopmental and neurodegenerative disorders, respectively.
Collapse
Affiliation(s)
- Meimei Yang
- Key Laboratory of Measurement and Evaluation in Exercise Bioinformation of Hebei Province, School of Physical Education, Hebei Normal University, Shijiazhuang, 050024, China.
- Regenerative Medicine Institute, School of Medicine, University of Galway, Galway, H91 W2TY, Ireland.
- FutureNeuro SFI Research Centre for Chronic and Rare Neurological Diseases and Department of Physiology and Medical Physics, RCSI University of Medicine and Health Sciences, Dublin, D02 YN77, Ireland.
| | - Daofeng You
- Emergency Department, The First Hospital of Hebei Medical University, No. 89, Donggang Road, Shijiazhuang, China
| | - Gang Liu
- Department of Cardiology, Hebei Key Laboratory of Cardiac Injury Repair Mechanism Study; Hebei Key Laboratory of Heart and Metabolism, Hebei Engineering Research Center of Intelligent Medical Clinical Application, Hebei International Joint Research Center for Structural Heart Disease, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yin Lu
- College of Pharmacy, Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Guangming Yang
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
- Confucius Institute of Chinese and Regenerative Medicine, University of Galway, Galway, H91 W2TY, Ireland
| | - Timothy O'Brien
- Regenerative Medicine Institute, School of Medicine, University of Galway, Galway, H91 W2TY, Ireland
- Confucius Institute of Chinese and Regenerative Medicine, University of Galway, Galway, H91 W2TY, Ireland
| | - David C Henshall
- FutureNeuro SFI Research Centre for Chronic and Rare Neurological Diseases and Department of Physiology and Medical Physics, RCSI University of Medicine and Health Sciences, Dublin, D02 YN77, Ireland
- Department of Physiology and Medical Physics, RCSI University of Medicine & Health Sciences, Dublin, D02 YN77, Ireland
| | - Orla Hardiman
- FutureNeuro SFI Research Centre for Chronic and Rare Neurological Diseases and Department of Physiology and Medical Physics, RCSI University of Medicine and Health Sciences, Dublin, D02 YN77, Ireland
- Academic Unit of Neurology, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland
| | - Li Cai
- Department of Ophthalmology, Shenzhen University General Hospital, Xueyuan Road 1098, Shenzhen, 518000, China.
| | - Min Liu
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Key Laboratory of Molecular and Cellular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China.
| | - Sanbing Shen
- Regenerative Medicine Institute, School of Medicine, University of Galway, Galway, H91 W2TY, Ireland.
- FutureNeuro SFI Research Centre for Chronic and Rare Neurological Diseases and Department of Physiology and Medical Physics, RCSI University of Medicine and Health Sciences, Dublin, D02 YN77, Ireland.
- Confucius Institute of Chinese and Regenerative Medicine, University of Galway, Galway, H91 W2TY, Ireland.
| |
Collapse
|
2
|
Chang FC, James MM, Zhou Y, Ando Y, Zareie HM, Yang J, Zhang M. Human Neural Stem Cell Expansion in Natural Polymer Scaffolds Under Chemically Defined Condition. Adv Biol (Weinh) 2024; 8:e2400224. [PMID: 38963310 DOI: 10.1002/adbi.202400224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 06/06/2024] [Indexed: 07/05/2024]
Abstract
The maintenance and expansion of human neural stem cells (hNSCs) in 3D tissue scaffolds is a promising strategy in producing cost-effective hNSCs with quality and quantity applicable for clinical applications. A few biopolymers have been extensively used to fabricate 3D scaffolds, including hyaluronic acid, collagen, alginate, and chitosan, due to their bioactive nature and availability. However, these polymers are usually applied in combination with other biomolecules, leading to their responses difficult to ascribe to. Here, scaffolds made of chitosan, alginate, hyaluronic acid, or collagen, are explored for hNSC expansion under xeno-free and chemically defined conditions and compared for hNSC multipotency maintenance. This study shows that the scaffolds made of pure chitosan support the highest adhesion and growth of hNSCs, yielding the most viable cells with NSC marker protein expression. In contrast, the presence of alginate, hyaluronic acid, or collagen induces differentiation toward immature neurons and astrocytes even in the maintenance medium and absence of differentiation factors. The cells in pure chitosan scaffolds preserve the level of transmembrane protein profile similar to that of standard culture. These findings point to the potential of using pure chitosan scaffolds as a base scaffolding material for hNSC expansion in 3D.
Collapse
Affiliation(s)
- Fei-Chien Chang
- Department of Materials Science and Engineering, University of Washington, Seattle, WA, 98195, USA
| | - Matthew Michael James
- Department of Materials Science and Engineering, University of Washington, Seattle, WA, 98195, USA
| | - Yang Zhou
- Department of Materials Science and Engineering, University of Washington, Seattle, WA, 98195, USA
| | - Yoshiki Ando
- Materials Department, Medical R&D Center, Corporate R&D Group, KYOCERA Corporation, Yasu, Shiga, 520-2362, Japan
| | - Hadi M Zareie
- Department of Materials Science and Engineering, University of Washington, Seattle, WA, 98195, USA
| | - Jihui Yang
- Department of Materials Science and Engineering, University of Washington, Seattle, WA, 98195, USA
| | - Miqin Zhang
- Department of Materials Science and Engineering, University of Washington, Seattle, WA, 98195, USA
| |
Collapse
|
3
|
Ortega JA, Soares de Aguiar GP, Chandravanshi P, Levy N, Engel E, Álvarez Z. Exploring the properties and potential of the neural extracellular matrix for next-generation regenerative therapies. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1962. [PMID: 38723788 DOI: 10.1002/wnan.1962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 04/05/2024] [Accepted: 04/09/2024] [Indexed: 05/24/2024]
Abstract
The extracellular matrix (ECM) is a dynamic and complex network of proteins and molecules that surrounds cells and tissues in the nervous system and orchestrates a myriad of biological functions. This review carefully examines the diverse interactions between cells and the ECM, as well as the transformative chemical and physical changes that the ECM undergoes during neural development, aging, and disease. These transformations play a pivotal role in shaping tissue morphogenesis and neural activity, thereby influencing the functionality of the central nervous system (CNS). In our comprehensive review, we describe the diverse behaviors of the CNS ECM in different physiological and pathological scenarios and explore the unique properties that make ECM-based strategies attractive for CNS repair and regeneration. Addressing the challenges of scalability, variability, and integration with host tissues, we review how advanced natural, synthetic, and combinatorial matrix approaches enhance biocompatibility, mechanical properties, and functional recovery. Overall, this review highlights the potential of decellularized ECM as a powerful tool for CNS modeling and regenerative purposes and sets the stage for future research in this exciting field. This article is categorized under: Implantable Materials and Surgical Technologies > Nanotechnology in Tissue Repair and Replacement Therapeutic Approaches and Drug Discovery > Nanomedicine for Neurological Disease Implantable Materials and Surgical Technologies > Nanomaterials and Implants.
Collapse
Affiliation(s)
- J Alberto Ortega
- Department of Pathology and Experimental Therapeutics, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
- Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet del Llobregat, Spain
| | - Gisele P Soares de Aguiar
- Department of Pathology and Experimental Therapeutics, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
- Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet del Llobregat, Spain
| | - Palash Chandravanshi
- Biomaterials for Neural Regeneration Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Natacha Levy
- Biomaterials for Neural Regeneration Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Elisabeth Engel
- IMEM-BRT Group, Department of Materials Science and Engineering, EEBE, Technical University of Catalonia (UPC), Barcelona, Spain
- Biomaterials for Regenerative Therapies Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, Madrid, Spain
| | - Zaida Álvarez
- Biomaterials for Neural Regeneration Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, Madrid, Spain
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
4
|
Chen X, Liu C, Wadsworth M, Zeng EZ, Driscoll T, Zeng C, Li Y. Surface Engineering of Auxetic Scaffolds for Neural and Vascular Differentiation from Human Pluripotent Stem Cells. Adv Healthc Mater 2023; 12:e2202511. [PMID: 36403987 PMCID: PMC9992167 DOI: 10.1002/adhm.202202511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/10/2022] [Indexed: 11/22/2022]
Abstract
Auxetic materials are the materials that can display negative Poisson's ratio that describes the degree to which a material contracts (or expands) transversally when axially strained. Human stem cells sense the mechanical properties of the microenvironment, including material surface properties, stiffness, and Poisson's ratio. In this study, six different auxetic polyurethane (PU) foams with different elastic modulus (0.7-1.8 kPa) and Poisson's ratio (-0.1 to -0.5) are used to investigate lineage specification of human induced pluripotent stem cells (hiPSCs). The surfaces of the foams are modified with chitosan or heparin to enhance the adhesion and proliferation of hiPSCs. Then, the vascular and neural differentiation of hiPSCs are investigated on different foams with distinct elastic modulus and Poisson's ratio. With different auxetic foams, cells show differential adherent density and differentiation capacity. Chitosan and heparin surface functionalization promote the hindbrain and hippocampal markers, but not forebrain markers during neural patterning of hiPSCs. Properly surface engineered auxetic scaffolds can also promote vascular differentiation of hiPSCs. This study represents a versatile and multifunctional scaffold fabrication approach and can lead to a suitable system for establishing hiPSC culture models in applications of neurovascular disease modeling and drug screening.
Collapse
Affiliation(s)
- Xingchi Chen
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University
- High-Performance Materials Institute, FAMU-FSU College of Engineering, Florida State University
| | - Chang Liu
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University
| | - Matthew Wadsworth
- High-Performance Materials Institute, FAMU-FSU College of Engineering, Florida State University
| | - Eric Z. Zeng
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University
| | - Tristan Driscoll
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University
| | - Changchun Zeng
- High-Performance Materials Institute, FAMU-FSU College of Engineering, Florida State University
- Department of Industrial and Manufacturing Engineering, FAMU-FSU College of Engineering, Florida State University
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University
| |
Collapse
|
5
|
Peressotti S, Koehl GE, Goding JA, Green RA. Self-Assembling Hydrogel Structures for Neural Tissue Repair. ACS Biomater Sci Eng 2021; 7:4136-4163. [PMID: 33780230 PMCID: PMC8441975 DOI: 10.1021/acsbiomaterials.1c00030] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 03/10/2021] [Indexed: 12/12/2022]
Abstract
Hydrogel materials have been employed as biological scaffolds for tissue regeneration across a wide range of applications. Their versatility and biomimetic properties make them an optimal choice for treating the complex and delicate milieu of neural tissue damage. Aside from finely tailored hydrogel properties, which aim to mimic healthy physiological tissue, a minimally invasive delivery method is essential to prevent off-target and surgery-related complications. The specific class of injectable hydrogels termed self-assembling peptides (SAPs), provide an ideal combination of in situ polymerization combined with versatility for biofunctionlization, tunable physicochemical properties, and high cytocompatibility. This review identifies design criteria for neural scaffolds based upon key cellular interactions with the neural extracellular matrix (ECM), with emphasis on aspects that are reproducible in a biomaterial environment. Examples of the most recent SAPs and modification methods are presented, with a focus on biological, mechanical, and topographical cues. Furthermore, SAP electrical properties and methods to provide appropriate electrical and electrochemical cues are widely discussed, in light of the endogenous electrical activity of neural tissue as well as the clinical effectiveness of stimulation treatments. Recent applications of SAP materials in neural repair and electrical stimulation therapies are highlighted, identifying research gaps in the field of hydrogels for neural regeneration.
Collapse
Affiliation(s)
- Sofia Peressotti
- Department
of Bioengineering and Centre for Neurotechnology, Imperial College London, London SW72AS, United Kingdom
| | - Gillian E. Koehl
- Department
of Bioengineering and Centre for Neurotechnology, Imperial College London, London SW72AS, United Kingdom
| | - Josef A. Goding
- Department
of Bioengineering and Centre for Neurotechnology, Imperial College London, London SW72AS, United Kingdom
| | - Rylie A. Green
- Department
of Bioengineering and Centre for Neurotechnology, Imperial College London, London SW72AS, United Kingdom
| |
Collapse
|
6
|
Induced Pluripotent Stem Cells to Model Juvenile Myelomonocytic Leukemia: New Perspectives for Preclinical Research. Cells 2021; 10:cells10092335. [PMID: 34571984 PMCID: PMC8465353 DOI: 10.3390/cells10092335] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/24/2021] [Accepted: 08/31/2021] [Indexed: 11/16/2022] Open
Abstract
Juvenile myelomonocytic leukemia (JMML) is a malignant myeloproliferative disorder arising in infants and young children. The origin of this neoplasm is attributed to an early deregulation of the Ras signaling pathway in multipotent hematopoietic stem/progenitor cells. Since JMML is notoriously refractory to conventional cytostatic therapy, allogeneic hematopoietic stem cell transplantation remains the mainstay of curative therapy for most cases. However, alternative therapeutic approaches with small epigenetic molecules have recently entered the stage and show surprising efficacy at least in specific subsets of patients. Hence, the establishment of preclinical models to test novel agents is a priority. Induced pluripotent stem cells (IPSCs) offer an opportunity to imitate JMML ex vivo, after attempts to generate immortalized cell lines from primary JMML material have largely failed in the past. Several research groups have previously generated patient-derived JMML IPSCs and successfully differentiated these into myeloid cells with extensive phenotypic similarities to primary JMML cells. With infinite self-renewal and the capability to differentiate into multiple cell types, JMML IPSCs are a promising resource to advance the development of treatment modalities targeting specific vulnerabilities. This review discusses current reprogramming techniques for JMML stem/progenitor cells, related clinical applications, and the challenges involved.
Collapse
|
7
|
Sachana M, Willett C, Pistollato F, Bal-Price A. The potential of mechanistic information organised within the AOP framework to increase regulatory uptake of the developmental neurotoxicity (DNT) in vitro battery of assays. Reprod Toxicol 2021; 103:159-170. [PMID: 34147625 PMCID: PMC8279093 DOI: 10.1016/j.reprotox.2021.06.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 05/19/2021] [Accepted: 06/04/2021] [Indexed: 12/24/2022]
Abstract
Current in vivo DNT testing for regulatory purposes is not effective. In vitro assays anchored to key neurodevelopmental processes are available. Development of Adverse Outcome Pathways is required to increase mechanistic understanding of DNT effects. DNT Integrated Approaches to Testing and Assessment for various regulatory purposes should be developed. The OECD Guidance Document on use of in vitro DNT battery of assays is currently under development.
A major challenge in regulatory developmental neurotoxicity (DNT) assessment is lack of toxicological information for many compounds. Therefore, the Test Guidelines programme of the Organisation for Economic Cooperation and Development (OECD) took the initiative to coordinate an international collaboration between diverse stakeholders to consider integration of alternative approaches towards improving the current chemical DNT testing. During the past few years, a series of workshops was organized during which a consensus was reached that incorporation of a DNT testing battery that relies on in vitro assays anchored to key neurodevelopmental processes should be developed. These key developmental processes include neural progenitor cell proliferation, neuronal and oligodendrocyte differentiation, neural cell migration, neurite outgrowth, synaptogenesis and neuronal network formation, as well key events identified in the existing Adverse Outcome Pathways (AOPs). AOPs deliver mechanistic information on the causal links between molecular initiating event, intermediate key events and an adverse outcome of regulatory concern, providing the biological context to facilitate development of Integrated Approaches to Testing and Assessment (IATA) for various regulatory purposes. Developing IATA case studies, using mechanistic information derived from AOPs, is expected to increase scientific confidence for the use of in vitro methods within an IATA, thereby facilitating regulatory uptake. This manuscript summarizes the current state of international efforts to enhance DNT testing by using an in vitro battery of assays focusing on the role of AOPs in informing the development of IATA for different regulatory purposes, aiming to deliver an OECD guidance document on use of in vitro DNT battery of assays that include in vitro data interpretation.
Collapse
Affiliation(s)
- Magdalini Sachana
- Environment Health and Safety Division, Environment Directorate, Organisation for Economic Co-Operation and Development (OECD), 75775, Paris Cedex 16, France
| | - Catherine Willett
- Humane Society International, 1255 23rd Street NW, Washington, DC, 20037, USA
| | | | - Anna Bal-Price
- European Commission Joint Research Centre (JRC), Ispra, Italy.
| |
Collapse
|
8
|
Zhang Y, Wiesholler LM, Rabie H, Jiang P, Lai J, Hirsch T, Lee KB. Remote Control of Neural Stem Cell Fate Using NIR-Responsive Photoswitching Upconversion Nanoparticle Constructs. ACS APPLIED MATERIALS & INTERFACES 2020; 12:40031-40041. [PMID: 32805826 DOI: 10.1021/acsami.0c10145] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Light-mediated remote control of stem cell fate, such as proliferation, differentiation, and migration, can bring a significant impact on stem cell biology and regenerative medicine. Current UV/vis-mediated control approaches are limited in terms of nonspecific absorption, poor tissue penetration, and phototoxicity. Upconversion nanoparticle (UCNP)-based near-infrared (NIR)-mediated control systems have gained increasing attention for vast applications with minimal nonspecific absorption, good penetration depth, and minimal phototoxicity from NIR excitations. Specifically, 808 nm NIR-responsive upconversion nanomaterials have shown clear advantages for biomedical applications owing to diminished heating effects and better tissue penetration. Herein, a novel 808 nm NIR-mediated control method for stem cell differentiation has been developed using multishell UCNPs, which are optimized for upconverting 808 nm NIR light to UV emission. The locally generated UV emissions further toggle photoswitching polymer capping ligands to achieve spatiotemporally controlled small-molecule release. More specifically, with 808 nm NIR excitation, stem cell differentiation factors can be released to guide neural stem cell (NSC) differentiation in a highly controlled manner. Given the challenges in stem cell behavior control, the developed 808 nm NIR-responsive UCNP-based approach to control stem cell differentiation can represent a new tool for studying single-molecule roles in stem cell and developmental biology.
Collapse
Affiliation(s)
- Yixiao Zhang
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - Lisa M Wiesholler
- Institute of Analytical Chemistry, Chemo- and Biosensors, University of Regensburg, 93040 Regensburg, Germany
| | - Hudifah Rabie
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - Pengfei Jiang
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - Jinping Lai
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - Thomas Hirsch
- Institute of Analytical Chemistry, Chemo- and Biosensors, University of Regensburg, 93040 Regensburg, Germany
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| |
Collapse
|
9
|
Jeon BM, Yeon GB, Goo HG, Lee KE, Kim DS. PVDF Nanofiber Scaffold Coated with a Vitronectin Peptide Facilitates the Neural Differentiation of Human Embryonic Stem Cells. Dev Reprod 2020; 24:135-147. [PMID: 32734130 PMCID: PMC7375977 DOI: 10.12717/dr.2020.24.2.135] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/08/2020] [Accepted: 05/16/2020] [Indexed: 12/24/2022]
Abstract
Polyvinylidene fluoride (PVDF) is a stable and biocompatible material that has been broadly used in biomedical applications. Due to its piezoelectric property, the electrospun nanofiber of PVDF has been used to culture electroactive cells, such as osteocytes and cardiomyocytes. Here, taking advantage of the piezoelectric property of PVDF, we have fabricated a PVDF nanofiber scaffolds using an electrospinning technique for differentiating human embryonic stem cells (hESCs) into neural precursors (NPs). Surface coating with a peptide derived from vitronectin enables hESCs to firmly adhere onto the nanofiber scaffolds and differentiate into NPs under dual-SMAD inhibition. Our nanofiber scaffolds supported the differentiation of hESCs into SOX1-positive NPs more significantly than Matrigel. The NPs generated on the nanofiber scaffolds could give rise to neurons, astrocytes, and oligodendrocyte precursors. Furthermore, comparative transcriptome analysis revealed the variable expressions of 27 genes in the nanofiber scaffold groups, several of which are highly related to the biological processes required for neural differentiation. These results suggest that a PVDF nanofiber scaffold coated with a vitronectin peptide can serve as a highly efficient and defined culture platform for the neural differentiation of hESCs.
Collapse
Affiliation(s)
- Byeong-Min Jeon
- Dept. of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea.,Institute of Animal Molecular Biotechnology, Korea University, Seoul 02841, Korea
| | - Gyu-Bum Yeon
- Dept. of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea.,Institute of Animal Molecular Biotechnology, Korea University, Seoul 02841, Korea
| | | | - Kyung Eun Lee
- Advance Analysis Center, Korean Institute of Science and Technology, Seoul 02792, Korea
| | - Dae-Sung Kim
- Dept. of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea.,Institute of Animal Molecular Biotechnology, Korea University, Seoul 02841, Korea.,Dept. of Pediatrics, Korea University College of Medicine, Seoul 08308, Korea
| |
Collapse
|
10
|
Åstrand C, Chotteau V, Falk A, Hedhammar M. Assembly of FN-silk with laminin-521 to integrate hPSCs into a three-dimensional culture for neural differentiation. Biomater Sci 2020; 8:2514-2525. [PMID: 32215392 DOI: 10.1039/c9bm01624d] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Three-dimensional (3D) neural tissue cultures recapitulate the basic concepts during development and disease better than what can be obtained using conventional two-dimensional cultures. Here, we use a recombinant spider silk protein functionalized with a cell binding motif from fibronectin (FN-silk) in combination with a human recombinant laminin 521 (LN-521) to create a fully defined stem cell niche in 3D. A novel method to assemble silk blended with LN-521 together with human pluripotent stem cells (hPSC) is used to create centimeter-sized foams, which upon cultivation develop into 3D cell constructs supported by a microfibrillar network. After initial cell expansion, neural differentiation was induced to form a homogenous layer of continuous neuroectodermal tissue that allows further differentiation into neuronal subtypes. The silk-supported 3D cell constructs could then be detached from the bottom of the well and cultured as floating entities, where cells appeared in distinctive radial organization resembling early neural tube. This shows that the neural progenitors retain their cellular self-organization ability in the FN-silk/LN-521-supported 3D culture. Calcium imaging demonstrated spontaneous activity, which is important for the formation of neuronal networks. Together, the results show that hPSCs integrated into FN-silk/LN-521 foam develop into neural progenitors and that these stay viable during long-term differentiations. FN-silk/LN-521 also supports morphogenesis mimicking the human brain development and can serve as base for engineering of hPSC-derived neural tissue.
Collapse
Affiliation(s)
- Carolina Åstrand
- Dept. of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH-Royal Institute of Technology, SE-10691, Stockholm, Sweden.
| | | | | | | |
Collapse
|
11
|
Kruger TM, Bell KJ, Lansakara TI, Tivanski AV, Doorn JA, Stevens LL. A Soft Mechanical Phenotype of SH-SY5Y Neuroblastoma and Primary Human Neurons Is Resilient to Oligomeric Aβ(1-42) Injury. ACS Chem Neurosci 2020; 11:840-850. [PMID: 32058688 DOI: 10.1021/acschemneuro.9b00401] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Aggregated amyloid beta (Aβ) is widely reported to cause neuronal dystrophy and toxicity through multiple pathways: oxidative stress, disrupting calcium homeostasis, and cytoskeletal dysregulation. The neuro-cytoskeleton is a dynamic structure that reorganizes to maintain cell homeostasis in response to varying soluble and physical cues presented from the extracellular matrix (ECM). Due this relationship between cell health and the ECM, we hypothesize that amyloid toxicity may be directly influenced by physical changes to the ECM (stiffness and dimensionality) through mechanosensitive pathways, and while previous studies demonstrated that Aβ can distort focal adhesion signaling with pathological consequences, these studies do not address the physical contribution from a physiologically relevant matrix. To test our hypothesis that physical cues can adjust Aβ toxicity, SH-SY5Y human neuroblastoma and primary human cortical neurons were plated on soft and stiff, 2D polyacrylamide matrices or suspended in 3D collagen gels. Each cell culture was exposed to escalating concentrations of oligomeric or fibrillated Aβ(1-42) with MTS viability and lactate dehydrogenase toxicity assessed. Actin restructuring was further monitored in live cells by atomic force microscopy nanoindentation, and our results demonstrate that increasing either matrix stiffness or exposure to oligomeric Aβ promotes F-actin polymerization and cell stiffening, while mature Aβ fibrils yielded no apparent cell stiffening and minor toxicity. Moreover, the rounded, softer mechanical phenotype displayed by cells plated onto a compliant matrix also demonstrated a resilience to oligomeric Aβ as noted by a significant recovery of viability when compared to same-dosed cells plated on traditional tissue culture plastic. This recovery was reproduced pharmacologically through inhibiting actin polymerization with cytochalasin D prior to Aβ exposure. These studies indicate that the cell-ECM interface can modify amyloid toxicity in neurons and the matrix-mediated pathways that promote this protection may offer unique targets in amyloid pathologies like Alzheimer's disease.
Collapse
Affiliation(s)
- Terra M. Kruger
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, The University of Iowa, Iowa City, Iowa 52242, United States
| | - Kendra J. Bell
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, The University of Iowa, Iowa City, Iowa 52242, United States
| | | | - Alexei V. Tivanski
- Department of Chemistry, The University of Iowa, Iowa City, Iowa 52242, United States
| | - Jonathan A. Doorn
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, The University of Iowa, Iowa City, Iowa 52242, United States
| | - Lewis L. Stevens
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, The University of Iowa, Iowa City, Iowa 52242, United States
| |
Collapse
|
12
|
Kośla K, Płuciennik E, Styczeń-Binkowska E, Nowakowska M, Orzechowska M, Bednarek AK. The WWOX Gene Influences Cellular Pathways in the Neuronal Differentiation of Human Neural Progenitor Cells. Front Cell Neurosci 2019; 13:391. [PMID: 31543760 PMCID: PMC6730490 DOI: 10.3389/fncel.2019.00391] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 08/08/2019] [Indexed: 11/13/2022] Open
Abstract
The brain is the most functionally organized structure of all organs. It manages behavior, perception and higher cognitive functions. The WWOX gene is non-classical tumor suppressor gene, which has been shown to have an impact on proliferation, apoptosis and migration processes. Moreover, genetic aberrations in WWOX induce severe neuropathological phenotypes in humans and rodents. The aim of the present study was to investigate in detail the impact of WWOX on human neural progenitor cell (hNPC) maintenance and how depletion of WWOX disturbs signaling pathways playing a pivotal role in neuronal differentiation and central nervous system (CNS) organogenesis. hNPC with a silenced WWOX gene exhibited lowered mitochondrial redox potential, enhanced adhesion to fibronectin and extracellular matrix protein mixture, downregulation of MMP2/9 expression and impaired 3D growth. Global transcriptome analysis using cap analysis of gene expression (CAGE) found that WWOX downregulation significantly changes the expression of multiple genes engaged in cytoskeleton organization, adhesion, cell signaling and chromatin remodeling. The massive changes in gene expression caused by WWOX silencing may strongly affect the differentiation and migration of neurons in organogenesis, brain injury, cancerogenesis or neurodifferentiation. WWOX gene appears to be an important regulator of neural tissue architecture and function.
Collapse
Affiliation(s)
- Katarzyna Kośla
- Department of Molecular Carcinogenesis, Medical University of Łódź, Łódź, Poland
| | - Elżbieta Płuciennik
- Department of Molecular Carcinogenesis, Medical University of Łódź, Łódź, Poland
| | | | - Magdalena Nowakowska
- Department of Molecular Carcinogenesis, Medical University of Łódź, Łódź, Poland
| | | | - Andrzej K Bednarek
- Department of Molecular Carcinogenesis, Medical University of Łódź, Łódź, Poland
| |
Collapse
|
13
|
Insolia V, Priori EC, Gasperini C, Coppa F, Cocchia M, Iervasi E, Ferrari B, Besio R, Maruelli S, Bernocchi G, Forlino A, Bottone MG. Prolidase enzyme is required for extracellular matrix integrity and impacts on postnatal cerebellar cortex development. J Comp Neurol 2019; 528:61-80. [PMID: 31246278 DOI: 10.1002/cne.24735] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 06/11/2019] [Accepted: 06/20/2019] [Indexed: 12/12/2022]
Abstract
The extracellular matrix is essential for brain development, lamination, and synaptogenesis. In particular, the basement membrane below the pial meninx (pBM) is required for correct cortical development. The last step in the catabolism of the most abundant protein in pBM, collagen Type IV, requires prolidase, an exopeptidase cleaving the imidodipeptides containing pro or hyp at the C-terminal end. Mutations impairing prolidase activity lead in humans to the rare disease prolidase deficiency characterized by severe skin ulcers and mental impairment. Thus, the dark-like (dal) mouse, in which the prolidase is knocked-out, was used to investigate whether the deficiency of prolidase affects the neuronal maturation during development of a brain cortex area. Focusing on the cerebellar cortex, thinner collagen fibers and disorganized pBM were found. Aberrant cortical granule cell proliferation and migration occurred, associated to defects in brain lamination, and in particular in maturation of Purkinje neurons and formation of synaptic contacts. This study deeply elucidates a link between prolidase activity and neuronal maturation shedding new light on the molecular basis of functional aspects in the prolidase deficiency.
Collapse
Affiliation(s)
- Violetta Insolia
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Erica C Priori
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Caterina Gasperini
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Federica Coppa
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Marco Cocchia
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Erika Iervasi
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Beatrice Ferrari
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Roberta Besio
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Pavia, Italy
| | - Silvia Maruelli
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Pavia, Italy
| | | | - Antonella Forlino
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Pavia, Italy
| | - Maria G Bottone
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| |
Collapse
|
14
|
Silva TP, Cotovio JP, Bekman E, Carmo-Fonseca M, Cabral JMS, Fernandes TG. Design Principles for Pluripotent Stem Cell-Derived Organoid Engineering. Stem Cells Int 2019; 2019:4508470. [PMID: 31149014 PMCID: PMC6501244 DOI: 10.1155/2019/4508470] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 02/12/2019] [Accepted: 02/24/2019] [Indexed: 12/17/2022] Open
Abstract
Human morphogenesis is a complex process involving distinct microenvironmental and physical signals that are manipulated in space and time to give rise to complex tissues and organs. Advances in pluripotent stem cell (PSC) technology have promoted the in vitro recreation of processes involved in human morphogenesis. The development of organoids from human PSCs represents one reliable source for modeling a large spectrum of human disorders, as well as a promising approach for drug screening and toxicological tests. Based on the "self-organization" capacity of stem cells, different PSC-derived organoids have been created; however, considerable differences between in vitro-generated PSC-derived organoids and their in vivo counterparts have been reported. Advances in the bioengineering field have allowed the manipulation of different components, including cellular and noncellular factors, to better mimic the in vivo microenvironment. In this review, we focus on different examples of bioengineering approaches used to promote the self-organization of stem cells, including assembly, patterning, and morphogenesis in vitro, contributing to tissue-like structure formation.
Collapse
Affiliation(s)
- Teresa P. Silva
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Lisbon Campus, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av Prof Egas Moniz, Edificio Egas Moniz, 1649-028 Lisboa, Portugal
| | - João P. Cotovio
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Lisbon Campus, Universidade de Lisboa, Lisboa, Portugal
| | - Evguenia Bekman
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Lisbon Campus, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av Prof Egas Moniz, Edificio Egas Moniz, 1649-028 Lisboa, Portugal
| | - Maria Carmo-Fonseca
- The Discoveries Centre for Regenerative and Precision Medicine, Lisbon Campus, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av Prof Egas Moniz, Edificio Egas Moniz, 1649-028 Lisboa, Portugal
| | - Joaquim M. S. Cabral
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Lisbon Campus, Universidade de Lisboa, Lisboa, Portugal
| | - Tiago G. Fernandes
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Lisbon Campus, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
15
|
Wen H, Xiao W, Biswas S, Cong ZQ, Liu XM, Lam KS, Liao YH, Deng W. Alginate Hydrogel Modified with a Ligand Interacting with α3β1 Integrin Receptor Promotes the Differentiation of 3D Neural Spheroids toward Oligodendrocytes in Vitro. ACS APPLIED MATERIALS & INTERFACES 2019; 11:5821-5833. [PMID: 30645095 DOI: 10.1021/acsami.8b19438] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
In this study, we established a long-term three-dimensional (3D) culture system by using integrin ligand modified alginate hydrogels to encapsulate and differentiate neural progenitor cells (NPCs) toward oligodendrocyte (OL) lineage cells. The porosity of the hydrogel was optimized by varying the alginate concentrations and then characterized by scanning electronic microscopy (SEM). The surface plasmon resonance (SPR) test was used to confirm the ligand-integrin interactions indicating adherence between the NPC surfaces and the hydrogels. Following encapsulation in the hydrogels, both mouse and human NPC sphere cultures could be maintained up to 90 days. Mouse NPC spheres were differentiated into viable neurons, astrocytes and mature OLs by day 60 in all groups whereas human NPC spheres were differentiated into neurons and later into GFAP positive astrocytes and O4 positive pre-OL within 90 days. The species difference in the timeline of OL development between mouse and human was reflected in this system. The ligand LXY30 interacting with the α3β1 integrin receptor was more effective in promoting the differentiation of hNPCs to OL lineage cells compared with the ligand LXW64 interacting with the αvβ3 integrin receptor, hyaluronic acid interacting with CD44 receptor or without any ligand. This study is the first to differentiate O4+ pre-OLs from hNPCs in a LXY30-α3β1 (integrin-ligand) modified alginate 3D hydrogel culture. This 3D platform could serve as a valuable tool in disease modeling, drug discovery, and NPC transplantation.
Collapse
Affiliation(s)
- Han Wen
- Institute of Medicinal Plant Development , Chinese Academy of Medical Sciences and Peking Union Medical College , 151 Malianwa North Road , Haidian District, Beijing 100193 , China
- Department of Biochemistry and Molecular Medicine, School of Medicine , University of California , 2700 Stockton Blvd , Davis , California 95817 , United States
- Institute for Pediatric Regenerative Medicine , Shriners Hospitals for Children-Northern California , 2425 Stockton Blvd , Sacramento , California 95817 , United States
| | - Wenwu Xiao
- Department of Biochemistry and Molecular Medicine, School of Medicine , University of California , 2700 Stockton Blvd , Davis , California 95817 , United States
| | - Sangita Biswas
- Department of Biochemistry and Molecular Medicine, School of Medicine , University of California , 2700 Stockton Blvd , Davis , California 95817 , United States
- Institute for Pediatric Regenerative Medicine , Shriners Hospitals for Children-Northern California , 2425 Stockton Blvd , Sacramento , California 95817 , United States
- School of Pharmaceutical Sciences, Shenzhen Campus , Sun Yat-Sen University , 135 Xingang Xi Road , Guangzhou 510275 , China
| | - Zhao-Qing Cong
- Institute of Medicinal Plant Development , Chinese Academy of Medical Sciences and Peking Union Medical College , 151 Malianwa North Road , Haidian District, Beijing 100193 , China
| | - Xin-Min Liu
- Institute of Medicinal Plant Development , Chinese Academy of Medical Sciences and Peking Union Medical College , 151 Malianwa North Road , Haidian District, Beijing 100193 , China
| | - Kit S Lam
- Department of Biochemistry and Molecular Medicine, School of Medicine , University of California , 2700 Stockton Blvd , Davis , California 95817 , United States
| | - Yong-Hong Liao
- Institute of Medicinal Plant Development , Chinese Academy of Medical Sciences and Peking Union Medical College , 151 Malianwa North Road , Haidian District, Beijing 100193 , China
| | - Wenbin Deng
- Department of Biochemistry and Molecular Medicine, School of Medicine , University of California , 2700 Stockton Blvd , Davis , California 95817 , United States
- Institute for Pediatric Regenerative Medicine , Shriners Hospitals for Children-Northern California , 2425 Stockton Blvd , Sacramento , California 95817 , United States
- School of Pharmaceutical Sciences, Shenzhen Campus , Sun Yat-Sen University , 135 Xingang Xi Road , Guangzhou 510275 , China
| |
Collapse
|
16
|
Three-dimensional brain-like microenvironments facilitate the direct reprogramming of fibroblasts into therapeutic neurons. Nat Biomed Eng 2018; 2:522-539. [DOI: 10.1038/s41551-018-0260-8] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Accepted: 06/11/2018] [Indexed: 02/07/2023]
|
17
|
Srinivasan G, Morgan D, Varun D, Brookhouser N, Brafman DA. An integrated biomanufacturing platform for the large-scale expansion and neuronal differentiation of human pluripotent stem cell-derived neural progenitor cells. Acta Biomater 2018; 74:168-179. [PMID: 29775730 DOI: 10.1016/j.actbio.2018.05.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 05/03/2018] [Accepted: 05/07/2018] [Indexed: 12/12/2022]
Abstract
Human pluripotent stem cell derived neural progenitor cells (hNPCs) have the unique properties of long-term in vitro expansion as well as differentiation into the various neurons and supporting cell types of the central nervous system (CNS). Because of these characteristics, hNPCs have tremendous potential in the modeling and treatment of various CNS diseases and disorders. However, expansion and neuronal differentiation of hNPCs in quantities necessary for these applications is not possible with current two dimensional (2-D) approaches. Here, we used a fully defined peptide substrate as the basis for a microcarrier (MC)-based suspension culture system. Several independently derived hNPC lines were cultured on MCs for multiple passages as well as efficiently differentiated to neurons. Finally, this MC-based system was used in conjunction with a low shear rotating wall vessel (RWV) bioreactor for the integrated, large-scale expansion and neuronal differentiation of hNPCs. Overall, this fully defined and scalable biomanufacturing system will facilitate the generation of hNPCs and their neuronal derivatives in quantities necessary for basic and translational applications. STATEMENT OF SIGNIFICANCE In this work, we developed a microcarrier (MC)-based culture system that allows for the expansion and neuronal differentiation of human pluripotent stem cell-derived neural progenitor cells (hNPCs) under defined conditions. In turn, this MC approach was implemented in a rotating wall vessel (RWV) bioreactor for the large-scale expansion and neuronal differentiation of hNPCs. This work is of significance as it overcomes current limitations of conventional two dimensional (2-D) culture systems to enable the generation of hNPCs and their neuronal derivatives in quantities required for downstream applications in disease modeling, drug screening, and regenerative medicine.
Collapse
Affiliation(s)
- Gayathri Srinivasan
- School of Biological and Health Systems Engineering, Arizona State University, United States
| | - Daylin Morgan
- School of Biological and Health Systems Engineering, Arizona State University, United States
| | - Divya Varun
- School of Biological and Health Systems Engineering, Arizona State University, United States
| | - Nicholas Brookhouser
- School of Biological and Health Systems Engineering, Arizona State University, United States
| | - David A Brafman
- School of Biological and Health Systems Engineering, Arizona State University, United States.
| |
Collapse
|
18
|
Centeno EGZ, Cimarosti H, Bithell A. 2D versus 3D human induced pluripotent stem cell-derived cultures for neurodegenerative disease modelling. Mol Neurodegener 2018; 13:27. [PMID: 29788997 PMCID: PMC5964712 DOI: 10.1186/s13024-018-0258-4] [Citation(s) in RCA: 157] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 05/08/2018] [Indexed: 12/11/2022] Open
Abstract
Neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD) and amyotrophic lateral sclerosis (ALS), affect millions of people every year and so far, there are no therapeutic cures available. Even though animal and histological models have been of great aid in understanding disease mechanisms and identifying possible therapeutic strategies, in order to find disease-modifying solutions there is still a critical need for systems that can provide more predictive and physiologically relevant results. One possible avenue is the development of patient-derived models, e.g. by reprogramming patient somatic cells into human induced pluripotent stem cells (hiPSCs), which can then be differentiated into any cell type for modelling. These systems contain key genetic information from the donors, and therefore have enormous potential as tools in the investigation of pathological mechanisms underlying disease phenotype, and progression, as well as in drug testing platforms. hiPSCs have been widely cultured in 2D systems, but in order to mimic human brain complexity, 3D models have been proposed as a more advanced alternative. This review will focus on the use of patient-derived hiPSCs to model AD, PD, HD and ALS. In brief, we will cover the available stem cells, types of 2D and 3D culture systems, existing models for neurodegenerative diseases, obstacles to model these diseases in vitro, and current perspectives in the field.
Collapse
Affiliation(s)
- Eduarda G Z Centeno
- Department of Biotechnology, Federal University of Pelotas, Campus Capão do Leão, Pelotas, RS, 96160-000, Brazil.,Department of Pharmacology, Federal University of Santa Catarina, Campus Trindade, Florianópolis, SC, 88040-900, Brazil
| | - Helena Cimarosti
- Department of Pharmacology, Federal University of Santa Catarina, Campus Trindade, Florianópolis, SC, 88040-900, Brazil.
| | - Angela Bithell
- School of Pharmacy, University of Reading, Whiteknights Campus, Reading, RG6 6UB, UK.
| |
Collapse
|
19
|
Gong L, Cao L, Shen Z, Shao L, Gao S, Zhang C, Lu J, Li W. Materials for Neural Differentiation, Trans-Differentiation, and Modeling of Neurological Disease. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1705684. [PMID: 29573284 DOI: 10.1002/adma.201705684] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 12/04/2017] [Indexed: 05/02/2023]
Abstract
Neuron regeneration from pluripotent stem cells (PSCs) differentiation or somatic cells trans-differentiation is a promising approach for cell replacement in neurodegenerative diseases and provides a powerful tool for investigating neural development, modeling neurological diseases, and uncovering the mechanisms that underlie diseases. Advancing the materials that are applied in neural differentiation and trans-differentiation promotes the safety, efficiency, and efficacy of neuron regeneration. In the neural differentiation process, matrix materials, either natural or synthetic, not only provide a structural and biochemical support for the monolayer or three-dimensional (3D) cultured cells but also assist in cell adhesion and cell-to-cell communication. They play important roles in directing the differentiation of PSCs into neural cells and modeling neurological diseases. For the trans-differentiation of neural cells, several materials have been used to make the conversion feasible for future therapy. Here, the most current applications of materials for neural differentiation for PSCs, neuronal trans-differentiation, and neurological disease modeling is summarized and discussed.
Collapse
Affiliation(s)
- Lulu Gong
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Lining Cao
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Zhenmin Shen
- The VIP Department, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Li Shao
- The VIP Department, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Shaorong Gao
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Chao Zhang
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Jianfeng Lu
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Weida Li
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| |
Collapse
|
20
|
Huang Y, Ng TK, Chen CB, Huang B, Liang J, Pang CP, Zhang M. Notch Signaling Activation Enhances Human Adipose-Derived Stem Cell Retinal Differentiation. Stem Cells Int 2018; 2018:9201374. [PMID: 30410544 PMCID: PMC6206515 DOI: 10.1155/2018/9201374] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 08/01/2018] [Accepted: 08/14/2018] [Indexed: 02/05/2023] Open
Abstract
Retinal disease treatment by stem cell-based replacement relies on stem cell differentiation into retinal cells. We previously demonstrated that human periodontal ligament-derived stem cells can be directed into retinal lineage upon induction. Here, we report the transdifferentiation potential of human adipose-derived stem cells (ASCs) into retinal lineage and its enhancement by Notch signaling modulation. Human ASCs, isolated from abdominal fat, expressed mesenchymal but not hematopoietic stem cell markers, and they can differentiate into adipocytes, chondrocytes, and osteoblasts in vitro. Upon noggin/Dkk-1/IGF-1 induction, the treated ASCs showed elevated expression of retinal progenitor, retinal ganglion, and photoreceptor cell markers as well as the glutamate-evoked calcium response, which was not observed in the noninduced cells. Compared to the regular induction treatment, Notch signaling activation by JAG1 enhanced the expression of retinal progenitor and precursor markers without affecting the glutamate-evoked calcium response. In contrast, Notch signaling inhibition by DAPT showed more retinal ganglion cells, but delayed the response to glutamate stimulation. In summary, our results revealed that human ASCs possess a retinal transdifferentiation potential upon noggin/Dkk-1/IGF-1 induction, which can further be enhanced by Notch signaling activation.
Collapse
Affiliation(s)
- Yuqiang Huang
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, Shantou, Guangdong, China
| | - Tsz Kin Ng
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, Shantou, Guangdong, China
- Shantou University Medical College, Shantou, Guandong, China
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Chong-Bo Chen
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, Shantou, Guangdong, China
| | - Bing Huang
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, Shantou, Guangdong, China
| | - Jiajian Liang
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, Shantou, Guangdong, China
| | - Chi Pui Pang
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, Shantou, Guangdong, China
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Mingzhi Zhang
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, Shantou, Guangdong, China
| |
Collapse
|
21
|
Lineage- and developmental stage-specific mechanomodulation of induced pluripotent stem cell differentiation. Stem Cell Res Ther 2017; 8:216. [PMID: 28962663 PMCID: PMC5622562 DOI: 10.1186/s13287-017-0667-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 07/18/2017] [Accepted: 09/12/2017] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND To maximize the translational utility of human induced pluripotent stem cells (iPSCs), the ability to precisely modulate the differentiation of iPSCs to target phenotypes is critical. Although the effects of the physical cell niche on stem cell differentiation are well documented, current approaches to direct step-wise differentiation of iPSCs have been typically limited to the optimization of soluble factors. In this regard, we investigated how temporally varied substrate stiffness affects the step-wise differentiation of iPSCs towards various lineages/phenotypes. METHODS Electrospun nanofibrous substrates with different reduced Young's modulus were utilized to subject cells to different mechanical environments during the differentiation process towards representative phenotypes from each of three germ layer derivatives including motor neuron, pancreatic endoderm, and chondrocyte. Phenotype-specific markers of each lineage/stage were utilized to determine differentiation efficiency by reverse-transcription polymerase chain reaction (RT-PCR) and immunofluorescence imaging for gene and protein expression analysis, respectively. RESULTS The results presented in this proof-of-concept study are the first to systematically demonstrate the significant role of the temporally varied mechanical microenvironment on the differentiation of stem cells. Our results demonstrate that the process of differentiation from pluripotent cells to functional end-phenotypes is mechanoresponsive in a lineage- and differentiation stage-specific manner. CONCLUSIONS Lineage/developmental stage-dependent optimization of electrospun substrate stiffness provides a unique opportunity to enhance differentiation efficiency of iPSCs for their facilitated therapeutic applications.
Collapse
|
22
|
Lee YJ, Seo TH, Lee S, Jang W, Kim MJ, Sung JS. Neuronal differentiation of human mesenchymal stem cells in response to the domain size of graphene substrates. J Biomed Mater Res A 2017; 106:43-51. [DOI: 10.1002/jbm.a.36215] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Revised: 08/10/2017] [Accepted: 08/24/2017] [Indexed: 11/10/2022]
Affiliation(s)
- Yoo-Jung Lee
- Department of Life Science; Dongguk University; Goyang Gyeonggi-do 10326 Republic of Korea
| | - Tae Hoon Seo
- Applied Quantum Composites Research Center; Korea Institute of Science and Technology; Jeonbuk 565-905 Republic of Korea
| | - Seula Lee
- Applied Quantum Composites Research Center; Korea Institute of Science and Technology; Jeonbuk 565-905 Republic of Korea
| | - Wonhee Jang
- Department of Life Science; Dongguk University; Goyang Gyeonggi-do 10326 Republic of Korea
| | - Myung Jong Kim
- Applied Quantum Composites Research Center; Korea Institute of Science and Technology; Jeonbuk 565-905 Republic of Korea
| | - Jung-Suk Sung
- Department of Life Science; Dongguk University; Goyang Gyeonggi-do 10326 Republic of Korea
| |
Collapse
|
23
|
Li Y, Li L, Chen ZN, Gao G, Yao R, Sun W. Engineering-derived approaches for iPSC preparation, expansion, differentiation and applications. Biofabrication 2017; 9:032001. [DOI: 10.1088/1758-5090/aa7e9a] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
24
|
Schutte RJ, Xie Y, Ng NN, Figueroa P, Pham AT, O'Dowd DK. Astrocyte-enriched feeder layers from cryopreserved cells support differentiation of spontaneously active networks of human iPSC-derived neurons. J Neurosci Methods 2017; 294:91-101. [PMID: 28746822 DOI: 10.1016/j.jneumeth.2017.07.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 07/19/2017] [Accepted: 07/19/2017] [Indexed: 01/08/2023]
Abstract
BACKGROUND Human induced pluripotent stem cell (hiPSC)-derived neuronal cultures are a useful tool for studying the mechanisms of neurological disorders and developing novel therapeutics. While plating hiPSC-derived neuronal progenitors onto glial feeder layers prepared from rodent cortex has been reported to promote functional differentiation of neuronal networks, this has not been examined in detail. NEW METHOD Here we describe a method of using cryopreserved cells from primary cultures for generation of mouse astrocyte-enriched, neuron-free feeder layers that grow from 10% to 100% confluence in 1 week. RESULTS Electrophysiological analysis demonstrated that compared to biochemical substrates alone, astrocyte-enriched feeder layers support more rapid differentiation of hiPSC-derived progenitors into excitable neurons that form spontaneously active networks in culture. There was a positive correlation between the degree of astroglial confluence at the time of progenitor plating and the average frequency of postsynaptic currents 3 weeks after plating. One disadvantage to plating on 100% confluent feeder layers was a high incidence of the astroglial layer with the overlying neurons detaching from the coverslips during transfer to the recording chamber. COMPARISON WITH EXISTING METHOD(S) Prevailing methods using primary glial feeder layers can result in possible contamination with rodent neurons and an unpredictable rate of growth. We provide a reliable method of generating mouse astroglial feeder layers from cryopreserved primary cultures to support differentiation of hiPSC-derived neurons. CONCLUSIONS The ability to make astrocyte-enriched feeder layers of defined confluence from cryopreserved primary cultures will facilitate the use of human stem cell derived neuronal cultures for disease modeling.
Collapse
Affiliation(s)
- Ryan J Schutte
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, United States
| | - Yunyao Xie
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, United States
| | - Nathan N Ng
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, United States
| | - Priscilla Figueroa
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, United States
| | - An T Pham
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, United States
| | - Diane K O'Dowd
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, United States.
| |
Collapse
|
25
|
Hsu CH, Huang TY, Chen RD, Liu YX, Chin TY, Chen-Yang YW, Yeh JM. Biomolding Technique to Fabricate the Hierarchical Topographical Scaffold of POMA To Enhance the Differentiation of Neural Stem Cells. ACS Biomater Sci Eng 2017; 3:1527-1534. [PMID: 33429639 DOI: 10.1021/acsbiomaterials.7b00091] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
In this paper, a biomolding technique was first used to fabricate a scaffold of hierarchical topography with biomimetic morphology for tissue engineering. First, poly(ortho-methoxyaniline) (POMA) was synthesized by conventional oxidative polymerization, followed by characterizations with Fourier transform infrared spectroscopy (FTIR) and gel permeation chromatography (GPC). Moreover, the POMA scaffold with 3D biomimetic morphology was fabricated using poly(dimethylsiloxane) (PDMS) as negative soft template from natural leaf surfaces of Xanthosoma sagittifolium, followed by transferring the pattern of PDMS template to POMA. The as-fabricated POMA scaffold with biomimetic morphology was investigated by scanning electron microscopy (SEM). Subsequently, cell-scaffold interactions were carried out by culturing rat neural stem cells (rNSCs) on biomimetic and nonbiomimetic, or flat, POMA scaffolds, as well as on poly(d-lysine) (PDL)-coated substrate, and evaluating the corresponding adhesion, cell viability, and differentiation of rNSCs. Results showed that there was no significant difference in the attachment of rNSCs on the three surface types, however, both the biomimetic and flat POMA scaffolds induced growth arrest relative to the PDL-coated substrate. In addition, the percentage of cells with elongated neurites after 19 days of culture was higher on the biomimetic POMA scaffold relative to flat POMA and PDL. In summary, the POMA scaffold with biomimetic morphology shows promise in promoting rNSCs differentiation and neurite outgrowth for long-term studies on nerve regenerative medicine.
Collapse
Affiliation(s)
- Chien-Hua Hsu
- Department of Chemistry, Center for Nanotechnology and Institute of Biomedical Technology, ‡Master Program in Nanotechnology and Center for Nanotechnology, and §Department of Bioscience Technology, Centre for Nanotechnology and Institute of Biomedical Technology, Chung Yuan Christian University, Chung Li, Taiwan 32023, Republic of China
| | - Ting-Yu Huang
- Department of Chemistry, Center for Nanotechnology and Institute of Biomedical Technology, Master Program in Nanotechnology and Center for Nanotechnology, and §Department of Bioscience Technology, Centre for Nanotechnology and Institute of Biomedical Technology, Chung Yuan Christian University, Chung Li, Taiwan 32023, Republic of China
| | - Rui-Da Chen
- Department of Chemistry, Center for Nanotechnology and Institute of Biomedical Technology, Master Program in Nanotechnology and Center for Nanotechnology, and Department of Bioscience Technology, Centre for Nanotechnology and Institute of Biomedical Technology, Chung Yuan Christian University, Chung Li, Taiwan 32023, Republic of China
| | - Yuan-Xian Liu
- Department of Chemistry, Center for Nanotechnology and Institute of Biomedical Technology, Master Program in Nanotechnology and Center for Nanotechnology, and Department of Bioscience Technology, Centre for Nanotechnology and Institute of Biomedical Technology, Chung Yuan Christian University, Chung Li, Taiwan 32023, Republic of China
| | - Ting-Yu Chin
- Department of Chemistry, Center for Nanotechnology and Institute of Biomedical Technology, Master Program in Nanotechnology and Center for Nanotechnology, and Department of Bioscience Technology, Centre for Nanotechnology and Institute of Biomedical Technology, Chung Yuan Christian University, Chung Li, Taiwan 32023, Republic of China
| | - Yui Whei Chen-Yang
- Department of Chemistry, Center for Nanotechnology and Institute of Biomedical Technology, Master Program in Nanotechnology and Center for Nanotechnology, and Department of Bioscience Technology, Centre for Nanotechnology and Institute of Biomedical Technology, Chung Yuan Christian University, Chung Li, Taiwan 32023, Republic of China
| | - Jui-Ming Yeh
- Department of Chemistry, Center for Nanotechnology and Institute of Biomedical Technology, Master Program in Nanotechnology and Center for Nanotechnology, and Department of Bioscience Technology, Centre for Nanotechnology and Institute of Biomedical Technology, Chung Yuan Christian University, Chung Li, Taiwan 32023, Republic of China
| |
Collapse
|
26
|
Li Z, Wang W, Kratz K, Küchler J, Xu X, Zou J, Deng Z, Sun X, Gossen M, Ma N, Lendlein A. Influence of surface roughness on neural differentiation of human induced pluripotent stem cells. Clin Hemorheol Microcirc 2017; 64:355-366. [DOI: 10.3233/ch-168121] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Zhengdong Li
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Weiwei Wang
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
| | - Karl Kratz
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
- Helmholtz Virtual Institute - Multifunctional Materials in Medicine, Teltow, Germany
| | - Judit Küchler
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
| | - Xun Xu
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Jie Zou
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Zijun Deng
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
| | - Xianlei Sun
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
| | - Manfred Gossen
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
- Helmholtz Virtual Institute - Multifunctional Materials in Medicine, Teltow, Germany
| | - Nan Ma
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
- Helmholtz Virtual Institute - Multifunctional Materials in Medicine, Teltow, Germany
| | - Andreas Lendlein
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
- Helmholtz Virtual Institute - Multifunctional Materials in Medicine, Teltow, Germany
| |
Collapse
|
27
|
Kim BC, Jun SM, Kim SY, Kwon YD, Choe SC, Kim EC, Lee JH, Kim J, Suh JKF, Hwang YS. Engineering three dimensional micro nerve tissue using postnatal stem cells from human dental apical papilla. Biotechnol Bioeng 2016; 114:903-914. [PMID: 27775170 DOI: 10.1002/bit.26205] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 10/16/2016] [Accepted: 10/19/2016] [Indexed: 12/26/2022]
Abstract
The in vitro generation of cell-based three dimensional (3D) nerve tissue is an attractive subject to improve graft survival and integration into host tissue for neural tissue regeneration or to model biological events in stem cell differentiation. Although 3D organotypic culture strategies are well established for 3D nerve tissue formation of pluripotent stem cells to study underlying biology in nerve development, cell-based nerve tissues have not been developed using human postnatal stem cells with therapeutic potential. Here, we established a culture strategy for the generation of in vitro cell-based 3D nerve tissue from postnatal stem cells from apical papilla (SCAPs) of teeth, which originate from neural crest-derived ectomesenchyme cells. A stem cell population capable of differentiating into neural cell lineages was generated during the ex vivo expansion of SCAPs in the presence of EGF and bFGF, and SCAPs differentiated into neural cells, showing neural cell lineage-related molecular and gene expression profiles, morphological changes and electrophysical property under neural-inductive culture conditions. Moreover, we showed the first evidence that 3D cell-based nerve-like tissue with axons and myelin structures could be generated from SCAPs via 3D organotypic culture using an integrated bioprocess composed of polyethylene glycol (PEG) microwell-mediated cell spheroid formation and subsequent dynamic culture in a high aspect ratio vessel (HARV) bioreactor. In conclusion, the culture strategy in our study provides a novel approach to develop in vitro engineered nerve tissue using SCAPs and a foundation to study biological events in the neural differentiation of postnatal stem cells. Biotechnol. Bioeng. 2017;114: 903-914. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Byung-Chul Kim
- Department of Maxillofacial Biomedical Engineering and Institute of Oral Biology, School of Dentistry, Kyung Hee University, 1 Hoegi-dong, Dongdaemun-gu, Seoul 130-701, Seoul, Korea
| | - Sung-Min Jun
- Department of Maxillofacial Biomedical Engineering and Institute of Oral Biology, School of Dentistry, Kyung Hee University, 1 Hoegi-dong, Dongdaemun-gu, Seoul 130-701, Seoul, Korea.,The Center for Bionics, Korea Institute of Science and Technology, Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul 136-791, Seoul, Korea
| | - So Yeon Kim
- Department of Maxillofacial Biomedical Engineering and Institute of Oral Biology, School of Dentistry, Kyung Hee University, 1 Hoegi-dong, Dongdaemun-gu, Seoul 130-701, Seoul, Korea
| | - Yong-Dae Kwon
- Department of Oral Maxillofacial Surgery, Kyung Hee University, Seoul, Korea
| | - Sung Chul Choe
- Department of Pediatric Dentistry, Kyung Hee University, Seoul, Korea
| | - Eun-Chul Kim
- Department of Oral and Maxillofacial Pathology, School of Dentistry, Kyung Hee University, Seoul, Korea
| | - Jae-Hyung Lee
- Department of Maxillofacial Biomedical Engineering and Institute of Oral Biology, School of Dentistry, Kyung Hee University, 1 Hoegi-dong, Dongdaemun-gu, Seoul 130-701, Seoul, Korea.,Department of Life and Nanopharmaceutical Sciences, Kyung Hee University, Seoul, Korea
| | - Jinseok Kim
- The Center for Bionics, Korea Institute of Science and Technology, Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul 136-791, Seoul, Korea
| | - Jun-Kyo Francis Suh
- The Center for Bionics, Korea Institute of Science and Technology, Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul 136-791, Seoul, Korea
| | - Yu-Shik Hwang
- Department of Maxillofacial Biomedical Engineering and Institute of Oral Biology, School of Dentistry, Kyung Hee University, 1 Hoegi-dong, Dongdaemun-gu, Seoul 130-701, Seoul, Korea
| |
Collapse
|
28
|
Wiley LA, Burnight ER, DeLuca AP, Anfinson KR, Cranston CM, Kaalberg EE, Penticoff JA, Affatigato LM, Mullins RF, Stone EM, Tucker BA. cGMP production of patient-specific iPSCs and photoreceptor precursor cells to treat retinal degenerative blindness. Sci Rep 2016; 6:30742. [PMID: 27471043 PMCID: PMC4965859 DOI: 10.1038/srep30742] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 07/07/2016] [Indexed: 12/21/2022] Open
Abstract
Immunologically-matched, induced pluripotent stem cell (iPSC)-derived photoreceptor precursor cells have the potential to restore vision to patients with retinal degenerative diseases like retinitis pigmentosa. The purpose of this study was to develop clinically-compatible methods for manufacturing photoreceptor precursor cells from adult skin in a non-profit cGMP environment. Biopsies were obtained from 35 adult patients with inherited retinal degeneration and fibroblast lines were established under ISO class 5 cGMP conditions. Patient-specific iPSCs were then generated, clonally expanded and validated. Post-mitotic photoreceptor precursor cells were generated using a stepwise cGMP-compliant 3D differentiation protocol. The recapitulation of the enhanced S-cone phenotype in retinal organoids generated from a patient with NR2E3 mutations demonstrated the fidelity of these protocols. Transplantation into immune compromised animals revealed no evidence of abnormal proliferation or tumor formation. These studies will enable clinical trials to test the safety and efficiency of patient-specific photoreceptor cell replacement in humans.
Collapse
Affiliation(s)
- Luke A Wiley
- Stephen A. Wynn Institute for Vision Research, Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Erin R Burnight
- Stephen A. Wynn Institute for Vision Research, Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Adam P DeLuca
- Stephen A. Wynn Institute for Vision Research, Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Kristin R Anfinson
- Stephen A. Wynn Institute for Vision Research, Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Cathryn M Cranston
- Stephen A. Wynn Institute for Vision Research, Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Emily E Kaalberg
- Stephen A. Wynn Institute for Vision Research, Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Jessica A Penticoff
- Stephen A. Wynn Institute for Vision Research, Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Louisa M Affatigato
- Stephen A. Wynn Institute for Vision Research, Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Robert F Mullins
- Stephen A. Wynn Institute for Vision Research, Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Edwin M Stone
- Stephen A. Wynn Institute for Vision Research, Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Budd A Tucker
- Stephen A. Wynn Institute for Vision Research, Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
29
|
Dzobo K, Vogelsang M, Parker MI. Wnt/β-Catenin and MEK-ERK Signaling are Required for Fibroblast-Derived Extracellular Matrix-Mediated Endoderm Differentiation of Embryonic Stem Cells. Stem Cell Rev Rep 2016; 11:761-73. [PMID: 26022506 DOI: 10.1007/s12015-015-9598-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Human embryonic stem cells (hESCs) have the potential to differentiate into all cells of the three germ layers, thus making them an attractive source of cells for use in regenerative medicine. The greatest challenge lies in regulating the differentiation of hESCs into specific cell lineages by both intrinsic and extrinsic factors. In this study we determined the effect of a fibroblast-derived extracellular matrix (fd-ECM) on hESCs differentiation. We demonstrate that growth of hESCs on fd-ECM results in hESCs losing their stemness and proliferation potential. As the stem cells differentiate they attain gene expression profiles similar to the primitive streak of the in vivo embryo. The activation of both the MEK-ERK and Wnt/β-catenin signaling pathways is required for the fd-ECM-mediated differentiation of hESCs towards the endoderm and involves integrins α1, α2, α3 and β1. This study illustrates the importance of the cellular microenvironment in directing stem cell fate and that the nature and composition of the extracellular matrix is a crucial determining factor.
Collapse
Affiliation(s)
- Kevin Dzobo
- Cape Town Component, Wernher and Beit Building (South), UCT Campus, International Centre for Genetic Engineering and Biotechnology (ICGEB), Anzio Road, Observatory, 7925, Cape Town, South Africa
| | | | | |
Collapse
|
30
|
Tatman PD, Muhonen EG, Wickers ST, Gee AO, Kim ES, Kim DH. Self-assembling peptides for stem cell and tissue engineering. Biomater Sci 2016; 4:543-54. [PMID: 26878078 PMCID: PMC4803621 DOI: 10.1039/c5bm00550g] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Regenerative medicine holds great potential to address many shortcomings in current medical therapies. An emerging avenue of regenerative medicine is the use of self-assembling peptides (SAP) in conjunction with stem cells to improve the repair of damaged tissues. The specific peptide sequence, mechanical properties, and nanotopographical cues vary widely between different SAPs, many of which have been used for the regeneration of similar tissues. To evaluate the potential of SAPs to guide stem cell fate, we extensively reviewed the literature for reports of SAPs and stem cell differentiation. To portray the most accurate summary of these studies, we deliberately discuss both the successes and pitfalls, allowing us to make conclusions that span the breadth of this exciting field. We also expand on these conclusions by relating these findings to the fields of nanotopography, mechanotransduction, and the native composition of the extracellular matrix in specific tissues to identify potential directions for future research.
Collapse
Affiliation(s)
- Philip D Tatman
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
- Medical Scientist Training Program, University of Colorado, Aurora, Colorado, USA
| | - Ethan G Muhonen
- School of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Sean T. Wickers
- Department of Chemistry, University of Colorado, Denver, Colorado, USA
| | - Albert O. Gee
- Department of Orthopedics and Sports Medicine, University of Washington, Seattle, WA 98195, USA
| | - Eung-Sam Kim
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
- Department of Biological Sciences, Chonnam National University, Gwangju, Korea
| | - Deok-Ho Kim
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
- Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA 98109, USA
| |
Collapse
|
31
|
Thompson R, Chan C. Signal transduction of the physical environment in the neural differentiation of stem cells. TECHNOLOGY 2016; 4:1-8. [PMID: 27785459 PMCID: PMC5077250 DOI: 10.1142/s2339547816400070] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Neural differentiation is largely dependent on extracellular signals within the cell microenvironment. These extracellular signals are mainly in the form of soluble factors that activate intracellular signaling cascades that drive changes in the cell nucleus. However, it is becoming increasingly apparent that the physical microenvironment provides signals that can also influence lineage commitment and very low modulus surfaces has been repeatedly demonstrated to promote neurogenesis. The molecular mechanisms governing mechano-induced neural differentiation are still largely uncharacterized; however, a growing body of evidence indicates that physical stimuli can regulate known signaling cascades and transcription factors involved in neural differentiation. Understanding how the physical environment affects neural differentiation at the molecular level will enable research and design of materials that will eventually enhance neural stem cell (NSC) differentiation, homogeneity and specificity.
Collapse
Affiliation(s)
- Ryan Thompson
- Cell and Molecular Biology Program, East Lansing, Michigan 48824, USA
| | - Christina Chan
- Cell and Molecular Biology Program, East Lansing, Michigan 48824, USA; Department of Chemical Engineering and Materials Science, East Lansing, Michigan 48824, USA; Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824, USA
| |
Collapse
|
32
|
Dzhoyashvili NA, Shen S, Rochev YA. Natural and Synthetic Materials for Self-Renewal, Long-Term Maintenance, and Differentiation of Induced Pluripotent Stem Cells. Adv Healthc Mater 2015; 4:2342-59. [PMID: 25867178 DOI: 10.1002/adhm.201400798] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 02/02/2015] [Indexed: 12/13/2022]
Abstract
Induced pluripotent stem cells (iPSCs) have attracted considerable attention from the public, clinicians, and scientists since their discovery in 2006, and raised huge expectations for regenerative medicine. One of the distinctive features of iPSCs is their propensity to differentiate into the cells of three germ lines in vitro and in vivo. The human iPSCs can be used to study the mechanisms underlying a disease and to monitor the disease progression, for testing drugs in vitro, and for cell therapy, avoiding many ethical and immunologic concerns. This technology offers the potential to take an individual approach to each patient and allows a more accurate diagnosis and specific treatment. However, there are several obstacles that impede the use of iPSCs. The derivation of fully reprogrammed iPSCs is expensive, time-consuming, and demands meticulous attention to many details. The use of biomaterials could increase the efficacy and safety while decreasing the cost of tissue engineering. The choice of a substrate utilized for iPSC culture is also important because cell-substrate contacts influence cellular behavior such as self-renewal, expansion, and differentiation. This Progress Report aims to summarize the advantages and drawbacks of natural and synthetic biomaterials, and to evaluate their role for maintenance and differentiation of iPSCs.
Collapse
Affiliation(s)
- Nina A. Dzhoyashvili
- School of Chemistry; National University of Ireland; Galway (NUIG), University Road Galway Ireland
- Network of Excellence for Functional Biomaterials (NFB); Biosciences Research Building, Newcastle Road Dangan Galway Ireland
| | - Sanbing Shen
- School of Medicine; National University of Ireland Galway (NUIG); University Road Galway Ireland
- Regenerative Medicine Institute (REMEDI); Biosciences Research Building; Newcastle Road Dangan Galway Ireland
| | - Yury A. Rochev
- School of Chemistry; National University of Ireland; Galway (NUIG), University Road Galway Ireland
- Network of Excellence for Functional Biomaterials (NFB); Biosciences Research Building, Newcastle Road Dangan Galway Ireland
| |
Collapse
|
33
|
Patel S, Chueng STD, Yin PT, Dardir K, Song Z, Pasquale N, Kwan K, Sugiyama H, Lee KB. Induction of stem-cell-derived functional neurons by NanoScript-based gene repression. Angew Chem Int Ed Engl 2015; 54:11983-8. [PMID: 26292201 PMCID: PMC5568028 DOI: 10.1002/anie.201504902] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Indexed: 01/22/2023]
Abstract
Even though gene repression is a powerful approach to exogenously regulate cellular behavior, developing a platform to effectively repress targeted genes, especially for stem-cell applications, remains elusive. Herein, we introduce a nanomaterial-based platform that is capable of mimicking the function of transcription repressor proteins to downregulate gene expression at the transcriptional level for enhancing stem-cell differentiation. We developed the "NanoScript" platform by integrating multiple gene repression molecules with a nanoparticle. First, we show a proof-of-concept demonstration using a GFP-specific NanoScript to knockdown GFP expression in neural stem cells (NSCs-GFP). Then, we show that a Sox9-specific NanoScript can repress Sox9 expression to initiate enhanced differentiation of NSCs into functional neurons. Overall, the tunable properties and gene-knockdown capabilities of NanoScript enables its utilization for gene-repression applications in stem cell biology.
Collapse
Affiliation(s)
- Sahishnu Patel
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854 (USA) http://kblee.rutgers.edu/
| | - Sy-Tsong Dean Chueng
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854 (USA) http://kblee.rutgers.edu/
| | - Perry T Yin
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ 08854 (USA)
| | - Kholud Dardir
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854 (USA) http://kblee.rutgers.edu/
| | - Zhichao Song
- Department of Cell Biology & Neuroscience, W. M. Keck Center for Collaborative Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ 08854 (USA)
| | - Nicholas Pasquale
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854 (USA) http://kblee.rutgers.edu/
| | - Kelvin Kwan
- Department of Cell Biology & Neuroscience, W. M. Keck Center for Collaborative Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ 08854 (USA)
| | - Hiroshi Sugiyama
- Department of Chemistry, Graduate School of Science, Kyoto University, Kyoto, 606-8501 (Japan)
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854 (USA) http://kblee.rutgers.edu/.
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ 08854 (USA).
| |
Collapse
|
34
|
Patel S, Chueng STD, Yin PT, Dardir K, Song Z, Pasquale N, Kwan K, Sugiyama H, Lee KB. Induction of Stem-Cell-Derived Functional Neurons by NanoScript-Based Gene Repression. Angew Chem Int Ed Engl 2015. [DOI: 10.1002/ange.201504902] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
35
|
Wang H, Luo X, Leighton J. Extracellular Matrix and Integrins in Embryonic Stem Cell Differentiation. BIOCHEMISTRY INSIGHTS 2015; 8:15-21. [PMID: 26462244 PMCID: PMC4589090 DOI: 10.4137/bci.s30377] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 09/02/2015] [Accepted: 09/04/2015] [Indexed: 12/17/2022]
Abstract
Embryonic stem cells (ESCs) are pluripotent cells with great therapeutic potentials. The in vitro differentiation of ESC was designed by recapitulating embryogenesis. Significant progress has been made to improve the in vitro differentiation protocols by toning soluble maintenance factors. However, more robust methods for lineage-specific differentiation and maturation are still under development. Considering the complexity of in vivo embryogenesis environment, extracellular matrix (ECM) cues should be considered besides growth factor cues. ECM proteins bind to cells and act as ligands of integrin receptors on cell surfaces. Here, we summarize the role of the ECM and integrins in the formation of three germ layer progenies. Various ECM–integrin interactions were found, facilitating differentiation toward definitive endoderm, hepatocyte-like cells, pancreatic beta cells, early mesodermal progenitors, cardiomyocytes, neuroectoderm lineages, and epidermal cells, such as keratinocytes and melanocytes. In the future, ECM combinations for the optimal ESC differentiation environment will require substantial study.
Collapse
Affiliation(s)
- Han Wang
- Department of Cellular and Structural Biology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Xie Luo
- Department of Cellular and Structural Biology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Jake Leighton
- Department of Cellular and Structural Biology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| |
Collapse
|
36
|
Tsai Y, Cutts J, Kimura A, Varun D, Brafman DA. A chemically defined substrate for the expansion and neuronal differentiation of human pluripotent stem cell-derived neural progenitor cells. Stem Cell Res 2015; 15:75-87. [DOI: 10.1016/j.scr.2015.05.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 05/04/2015] [Accepted: 05/04/2015] [Indexed: 01/27/2023] Open
|
37
|
Neural Differentiation of Human Pluripotent Stem Cells for Nontherapeutic Applications: Toxicology, Pharmacology, and In Vitro Disease Modeling. Stem Cells Int 2015; 2015:105172. [PMID: 26089911 PMCID: PMC4454762 DOI: 10.1155/2015/105172] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 05/06/2015] [Accepted: 05/12/2015] [Indexed: 02/08/2023] Open
Abstract
Human pluripotent stem cells (hPSCs) derived from either blastocyst stage embryos (hESCs) or reprogrammed somatic cells (iPSCs) can provide an abundant source of human neuronal lineages that were previously sourced from human cadavers, abortuses, and discarded surgical waste. In addition to the well-known potential therapeutic application of these cells in regenerative medicine, these are also various promising nontherapeutic applications in toxicological and pharmacological screening of neuroactive compounds, as well as for in vitro modeling of neurodegenerative and neurodevelopmental disorders. Compared to alternative research models based on laboratory animals and immortalized cancer-derived human neural cell lines, neuronal cells differentiated from hPSCs possess the advantages of species specificity together with genetic and physiological normality, which could more closely recapitulate in vivo conditions within the human central nervous system. This review critically examines the various potential nontherapeutic applications of hPSC-derived neuronal lineages and gives a brief overview of differentiation protocols utilized to generate these cells from hESCs and iPSCs.
Collapse
|
38
|
Walthers CM, Seidlits SK. Gene delivery strategies to promote spinal cord repair. Biomark Insights 2015; 10:11-29. [PMID: 25922572 PMCID: PMC4395076 DOI: 10.4137/bmi.s20063] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 03/02/2015] [Accepted: 03/04/2015] [Indexed: 12/21/2022] Open
Abstract
Gene therapies hold great promise for the treatment of many neurodegenerative disorders and traumatic injuries in the central nervous system. However, development of effective methods to deliver such therapies in a controlled manner to the spinal cord is a necessity for their translation to the clinic. Although essential progress has been made to improve efficiency of transgene delivery and reduce the immunogenicity of genetic vectors, there is still much work to be done to achieve clinical strategies capable of reversing neurodegeneration and mediating tissue regeneration. In particular, strategies to achieve localized, robust expression of therapeutic transgenes by target cell types, at controlled levels over defined time periods, will be necessary to fully regenerate functional spinal cord tissues. This review summarizes the progress over the last decade toward the development of effective gene therapies in the spinal cord, including identification of appropriate target genes, improvements to design of genetic vectors, advances in delivery methods, and strategies for delivery of multiple transgenes with synergistic actions. The potential of biomaterials to mediate gene delivery while simultaneously providing inductive scaffolding to facilitate tissue regeneration is also discussed.
Collapse
|
39
|
Lam J, Carmichael ST, Lowry WE, Segura T. Hydrogel design of experiments methodology to optimize hydrogel for iPSC-NPC culture. Adv Healthc Mater 2015; 4:534-9. [PMID: 25378176 PMCID: PMC4384641 DOI: 10.1002/adhm.201400410] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 10/02/2014] [Indexed: 12/29/2022]
Abstract
Bioactive signals can be incorporated in hydrogels to direct encapsulated cell behavior. Design of experiments methodology methodically varies the signals systematically to determine the individual and combinatorial effects of each factor on cell activity. Using this approach enables the optimization of three ligands concentrations (RGD, YIGSR, IKVAV) for the survival and differentiation of neural progenitor cells.
Collapse
Affiliation(s)
- Jonathan Lam
- University of California, Los Angeles, Biomedical Engineering Department
| | - S. Thomas Carmichael
- University of California, Los Angeles, Department of Neurology
- University of California, Los Angeles, David Geffen School of Medicine
| | - William E. Lowry
- University of California, Los Angeles, Department of Molecular, Cell and Developmental Biology
- University of California, Los Angeles, Eli and Edythe Broad Center for Regenerative Medicine
| | - Tatiana Segura
- University of California, Los Angeles, Biomedical Engineering Department
- University of California, Los Angeles, Department of Neurology
| |
Collapse
|
40
|
Skalova S, Svadlakova T, Shaikh Qureshi WM, Dev K, Mokry J. Induced pluripotent stem cells and their use in cardiac and neural regenerative medicine. Int J Mol Sci 2015; 16:4043-67. [PMID: 25689424 PMCID: PMC4346943 DOI: 10.3390/ijms16024043] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 01/27/2015] [Accepted: 02/02/2015] [Indexed: 12/20/2022] Open
Abstract
Stem cells are unique pools of cells that are crucial for embryonic development and maintenance of adult tissue homeostasis. The landmark Nobel Prize winning research by Yamanaka and colleagues to induce pluripotency in somatic cells has reshaped the field of stem cell research. The complications related to the usage of pluripotent embryonic stem cells (ESCs) in human medicine, particularly ESC isolation and histoincompatibility were bypassed with induced pluripotent stem cell (iPSC) technology. The human iPSCs can be used for studying embryogenesis, disease modeling, drug testing and regenerative medicine. iPSCs can be diverted to different cell lineages using small molecules and growth factors. In this review we have focused on iPSC differentiation towards cardiac and neuronal lineages. Moreover, we deal with the use of iPSCs in regenerative medicine and modeling diseases like myocardial infarction, Timothy syndrome, dilated cardiomyopathy, Parkinson’s, Alzheimer’s and Huntington’s disease. Despite the promising potential of iPSCs, genome contamination and low efficacy of cell reprogramming remain significant challenges.
Collapse
Affiliation(s)
- Stepanka Skalova
- Department of Histology and Embryology, Medical Faculty in Hradec Kralove, Charles University in Prague, Simkova 870, Hradec Kralove 50038, Czech Republic.
| | - Tereza Svadlakova
- Department of Histology and Embryology, Medical Faculty in Hradec Kralove, Charles University in Prague, Simkova 870, Hradec Kralove 50038, Czech Republic.
| | - Wasay Mohiuddin Shaikh Qureshi
- Department of Histology and Embryology, Medical Faculty in Hradec Kralove, Charles University in Prague, Simkova 870, Hradec Kralove 50038, Czech Republic.
| | - Kapil Dev
- Department of Histology and Embryology, Medical Faculty in Hradec Kralove, Charles University in Prague, Simkova 870, Hradec Kralove 50038, Czech Republic.
| | - Jaroslav Mokry
- Department of Histology and Embryology, Medical Faculty in Hradec Kralove, Charles University in Prague, Simkova 870, Hradec Kralove 50038, Czech Republic.
| |
Collapse
|
41
|
Ebrahimi-Barough S, Hoveizi E, Norouzi Javidan A, Ai J. Investigating the neuroglial differentiation effect of neuroblastoma conditioned medium in human endometrial stem cells cultured on 3D nanofibrous scaffold. J Biomed Mater Res A 2015; 103:2621-7. [DOI: 10.1002/jbm.a.35397] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 12/16/2014] [Accepted: 01/04/2015] [Indexed: 12/27/2022]
Affiliation(s)
- Somayeh Ebrahimi-Barough
- Brain and Spinal Cord Injury Research Center (BASIR), Tehran University of Medical Sciences; Keshavarz Boulevard, Gharib Street Tehran 6114185 Iran
- Department of Tissue Engineering and Applied Cell Sciences; Faculty of Advanced Technologies in Medicine, Tehran University of Medical Sciences; Tehran Iran
| | - Elham Hoveizi
- Department of Biology; Faculty of Sciences, Shahid Chamran University of Ahvaz; Ahvaz Iran
| | - Abbas Norouzi Javidan
- Brain and Spinal Cord Injury Research Center (BASIR), Tehran University of Medical Sciences; Keshavarz Boulevard, Gharib Street Tehran 6114185 Iran
| | - Jafar Ai
- Brain and Spinal Cord Injury Research Center (BASIR), Tehran University of Medical Sciences; Keshavarz Boulevard, Gharib Street Tehran 6114185 Iran
- Department of Tissue Engineering and Applied Cell Sciences; Faculty of Advanced Technologies in Medicine, Tehran University of Medical Sciences; Tehran Iran
| |
Collapse
|
42
|
Hardy JG, Lin P, Schmidt CE. Biodegradable hydrogels composed of oxime crosslinked poly(ethylene glycol), hyaluronic acid and collagen: a tunable platform for soft tissue engineering. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2015; 26:143-61. [DOI: 10.1080/09205063.2014.975393] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
43
|
Laperle A, Masters KS, Palecek SP. Influence of substrate composition on human embryonic stem cell differentiation and extracellular matrix production in embryoid bodies. Biotechnol Prog 2014; 31:212-9. [PMID: 25311359 DOI: 10.1002/btpr.2001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Revised: 09/28/2014] [Indexed: 12/14/2022]
Abstract
Stem cells reside in specialized niches in vivo. Specific factors, including the extracellular matrix (ECM), in these niches are directly responsible for maintaining the stem cell population. During development, components of the stem cell microenvironment also control differentiation with precise spatial and temporal organization. The stem cell microenvironment is dynamically regulated by the cellular component, including stem cells themselves. Thus, a mechanism exists whereby stem cells modify the ECM, which in turn affects the fate of the stem cell. In this study, we investigated whether the type of ECM initially adsorbed to the culture substrate can influence the composition of the ECM deposited by human embryonic stem cells (hESCs) differentiating in embryoid bodies, and whether different ECM composition and deposition profiles elicit distinct differentiation fates. We have shown that the initial ECM environment hESCs are exposed to affects the fate decisions of those cells and that this initial ECM environment is constantly modified during the differentiation process.
Collapse
Affiliation(s)
- Alex Laperle
- Dept. of Biomedical Engineering, University of Wisconsin, Madison, Wisconsin, 53706
| | | | | |
Collapse
|
44
|
Hoveizi E, Tavakol S, Ebrahimi-Barough S. Neuroprotective Effect of Transplanted Neural Precursors Embedded on PLA/CS Scaffold in an Animal Model of Multiple Sclerosis. Mol Neurobiol 2014; 51:1334-42. [DOI: 10.1007/s12035-014-8812-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 07/11/2014] [Indexed: 12/14/2022]
|
45
|
Abstract
The consequence of numerous neurological disorders is the significant loss of neural cells, which further results in multilevel dysfunction or severe functional deficits. The extracellular matrix (ECM) is of tremendous importance for neural regeneration mediating ambivalent functions: ECM serves as a growth-promoting substrate for neurons but, on the other hand, is a major constituent of the inhibitory scar, which results from traumatic injuries of the central nervous system. Therefore, cell and tissue replacement strategies on the basis of ECM mimetics are very promising therapeutic interventions. Numerous synthetic and natural materials have proven effective both in vitro and in vivo. The closer a material's physicochemical and molecular properties are to the original extracellular matrix, the more promising its effectiveness may be. Relevant factors that need to be taken into account when designing such materials for neural repair relate to receptor-mediated cell-matrix interactions, which are dependent on chemical and mechanical sensing. This chapter outlines important characteristics of natural and synthetic ECM materials (scaffolds) and provides an overview of recent advances in design and application of ECM materials for neural regeneration, both in therapeutic applications and in basic biological research.
Collapse
Affiliation(s)
- Veronica Estrada
- Molecular Neurobiology Laboratory, Department of Neurology, Heinrich-Heine-University Medical Center Düsseldorf, Düsseldorf, Germany
| | - Ayse Tekinay
- UNAM-National Nanotechnology Research Center, Institute of Materials Science and Nanotechnology, Bilkent University, Ankara, Turkey
| | - Hans Werner Müller
- Molecular Neurobiology Laboratory, Department of Neurology, Heinrich-Heine-University Medical Center Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|