1
|
Ireland J, Kilian KA. The importance of matrix in cardiomyogenesis: Defined substrates for maturation and chamber specificity. Matrix Biol Plus 2024; 24:100160. [PMID: 39291079 PMCID: PMC11403269 DOI: 10.1016/j.mbplus.2024.100160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 09/19/2024] Open
Abstract
Human embryonic stem cell-derived cardiomyocytes (hESC-CM) are a promising source of cardiac cells for disease modelling and regenerative medicine. However, current protocols invariably lead to mixed population of cardiac cell types and often generate cells that resemble embryonic phenotypes. Here we developed a combinatorial approach to assess the importance of extracellular matrix proteins (ECMP) in directing the differentiation of cardiomyocytes from human embryonic stem cells (hESC). We did this by focusing on combinations of ECMP commonly found in the developing heart with a broad goal of identifying combinations that promote maturation and influence chamber specific differentiation. We formulated 63 unique ECMP combinations fabricated from collagen 1, collagen 3, collagen 4, fibronectin, laminin, and vitronectin, presented alone and in combinations, leading to the identification of specific ECMP combinations that promote hESC proliferation, pluripotency, and germ layer specification. When hESC were subjected to a differentiation protocol on the ECMP combinations, it revealed precise protein combinations that enhance differentiation as determined by the expression of cardiac progenitor markers kinase insert domain receptor (KDR) and mesoderm posterior transcription factor 1 (MESP1). High expression of cardiac troponin (cTnT) and the relative expression of myosin light chain isoforms (MLC2a and MLC2v) led to the identification of three surfaces that promote a mature cardiomyocyte phenotype. Action potential morphology was used to assess chamber specificity, which led to the identification of matrices that promote chamber-specific cardiomyocytes. This study provides a matrix-based approach to improve control over cardiomyocyte phenotypes during differentiation, with the scope for translation to cardiac laboratory models and for the generation of functional chamber specific cardiomyocytes for regenerative therapies.
Collapse
Affiliation(s)
- Jake Ireland
- School of Chemistry, UNSW Sydney, Sydney, New South Wales, Australia
| | - Kristopher A Kilian
- School of Chemistry, UNSW Sydney, Sydney, New South Wales, Australia
- School of Materials Science and Engineering, UNSW Sydney, Sydney, New South Wales, Australia
- Australian Centre for NanoMedicine, UNSW Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
2
|
Gao ML, Wang TY, Lin X, Tang C, Li M, Bai ZP, Liu ZC, Chen LJ, Kong QR, Pan SH, Zeng SS, Guo Y, Cai JQ, Huang XF, Zhang J. Retinal Organoid Microenvironment Enhanced Bioactivities of Microglia-Like Cells Derived From HiPSCs. Invest Ophthalmol Vis Sci 2024; 65:19. [PMID: 39392440 PMCID: PMC11472886 DOI: 10.1167/iovs.65.12.19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 07/12/2024] [Indexed: 10/12/2024] Open
Abstract
Purpose Microglia-like cells derived from stem cells (iMG) provide a plentiful cell source for studying the functions of microglia in both normal and pathological conditions. Our goal is to establish a simplified and effective method for generating iMG in a precisely defined system. Additionally, we aim to achieve functional maturation of iMG through coculture with retinal organoids. Methods In this study, iMG were produced under precisely defined conditions. They were subjected to LPS and poly IC stimulation. Additionally, we examined distinct phenotypic and functional variances between iMG and HMC3, a commonly used human microglia cell line. To investigate how the retinal cell interaction enhances microglial properties, iMG were cocultured with retinal organoids, producing CC-iMG. We performed RNA sequencing, electrophysiological analysis, and transmission electron microscope (TEM) to examine the maturation of CC-iMG compared to iMG. Results Our results demonstrated that iMG performed immune-responsive profiles closely resembling those of primary human microglia. Compared to HMC3, iMG expressed a higher level of typical microglial markers and exhibited enhanced phagocytic activity. The transcriptomic analysis uncovered notable alterations in the ion channel profile of CC-iMG compared to iMG. Electrophysiological examination demonstrated a heightened intensity of inward- and outward-rectifying K+ currents in CC-iMG. Furthermore, CC-iMG displayed elevated numbers of lysosomes and mitochondria, coupled with increased phagocytic activity. Conclusions These findings contribute to advancing our understanding of human microglial biology, specifically in characterizing and elucidating the functions of CC-iMG, thereby offering an in vitro microglial model for future scientific research and potential clinical applications in cell therapy.
Collapse
Affiliation(s)
- Mei-Ling Gao
- The State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Wenzhou, China
- Laboratory of Retinal Physiology and Disease, Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, China
| | - Tong-Yu Wang
- The State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Wenzhou, China
- Laboratory of Retinal Physiology and Disease, Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, China
| | - Xin Lin
- The State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Wenzhou, China
- Laboratory of Retinal Physiology and Disease, Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, China
| | - Chun Tang
- The State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Wenzhou, China
- Laboratory of Retinal Physiology and Disease, Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, China
| | - Mengyao Li
- Department of Critical Care Medicine, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China
| | - Zhan-Pei Bai
- Zhejiang Provincial Clinical Research Center for Pediatric Disease, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhi-Cong Liu
- The State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Wenzhou, China
- Laboratory of Retinal Physiology and Disease, Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, China
| | - Li-Jun Chen
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Qing-Ran Kong
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Shao-Hui Pan
- The State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Wenzhou, China
| | - Shan-Shan Zeng
- China National Institute of Standardization, Beijing, China
| | - Ya Guo
- China National Institute of Standardization, Beijing, China
| | - Jian-Qi Cai
- China National Institute of Standardization, Beijing, China
| | - Xiu-Feng Huang
- Zhejiang Provincial Clinical Research Center for Pediatric Disease, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jun Zhang
- The State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Wenzhou, China
- Laboratory of Retinal Physiology and Disease, Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
3
|
Lu X, Perr E, Naqvi T, Galitz D, Andersen M, Grabowski D, Person A, Kalyuzhny A, Flynn KC. A Novel Recombinant Vitronectin Variant Supports the Expansion and Differentiation of Pluripotent Stem Cells in Defined Animal-Free Workflows. Cells 2024; 13:1566. [PMID: 39329750 PMCID: PMC11429963 DOI: 10.3390/cells13181566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 09/04/2024] [Accepted: 09/09/2024] [Indexed: 09/28/2024] Open
Abstract
An essential aspect of harnessing the potential of pluripotent stem cells (PSCs) and their derivatives for regenerative medicine is the development of animal-free and chemically defined conditions for ex vivo cultivation. PSCs, including embryonic and induced PSCs (iPSCs), are in the early stages of clinical trials for various indications, including degenerative diseases and traumatic injury. A key step in the workflows generating these cells for more widespread clinical use is their safe and robust ex vivo cultivation. This entails optimization of cell culture media and substrates that are safe and consistent while maintaining robust functionality. Here, we describe the design of a human vitronectin (hVTN) variant with improved manufacturability in a bacterial expression system along with improved function in comparison to wild-type VTN and other previously characterized polypeptide fragments. In conjunction with an animal component-free media formulation, our hVTN fragment provides animal-free conditions for the enhanced expansion of iPSCs. This hVTN variant also supports the reprogramming of PBMCs into iPSCs. Furthermore, we show that these iPSCs can be efficiently differentiated into the three major germ layers and cortical neurons, thereby closing the loop on a completely defined animal-free workflow for cell types relevant for regenerative medicine.
Collapse
Affiliation(s)
- Xi Lu
- Stem Cell & Gene Therapy, Bio-Techne, Minneapolis, MN 55413, USA
| | - Eli Perr
- Stem Cell & Gene Therapy, Bio-Techne, Minneapolis, MN 55413, USA
| | - Tahmina Naqvi
- Stem Cell & Gene Therapy, Bio-Techne, Minneapolis, MN 55413, USA
| | - David Galitz
- Stem Cell & Gene Therapy, Bio-Techne, Minneapolis, MN 55413, USA
| | | | | | - Anthony Person
- Protein Development, Bio-Techne, Minneapolis, MN 55413, USA
| | - Alex Kalyuzhny
- Antibody Applications, Bio-Techne, Minneapolis, MN 55413, USA
| | - Kevin C Flynn
- Stem Cell & Gene Therapy, Bio-Techne, Minneapolis, MN 55413, USA
- Department of Biochemistry & Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
4
|
Lee NK, Jang WB, Seo DS, Goo HG, Lim HJ, Lee EJ, Kwon SM. Development of advanced cardiac progenitor cell culture system through fibronectin and vitronectin derived peptide coated plate. Stem Cell Res 2024; 79:103476. [PMID: 38941882 DOI: 10.1016/j.scr.2024.103476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 05/23/2024] [Accepted: 06/17/2024] [Indexed: 06/30/2024] Open
Abstract
Cardiovascular disease remains a global health concern. Stem cell therapy utilizing human cardiac progenitor cells (hCPCs) shows promise in treating cardiac vascular disease. However, limited availability and senescence of hCPCs hinder their widespread use. To address these challenges, researchers are exploring innovative approaches. In this study, a bioengineered cell culture plate was developed to mimic the natural cardiac tissue microenvironment. It was coated with a combination of extracellular matrix (ECM) peptide motifs and mussel adhesive protein (MAP). The selected ECM peptide motifs, derived from fibronectin and vitronectin, play crucial roles in hCPCs. Results revealed that the Fibro-P and Vitro-P coated plates significantly improved hCPC adhesion, proliferation, migration, and differentiation compared to uncoated plates. Additionally, long-term culture on the coated plates delayed cellular senescence and maintained hCPC stemness. These enhancements were attributed to the activation of integrin downstream signaling pathways. The findings suggest that the engineered ECM peptide motif-MAP-coated plates hold potential for enhancing the therapeutic efficacy of stem cell-based therapies in cardiac tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Na Kyung Lee
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Korea; Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Korea
| | - Woong Bi Jang
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Korea; Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Korea
| | - Dong Sik Seo
- AMO Lifescience Co., Ltd., Seoul, Seocho-gu, Republic of Korea
| | - Hui-Gwan Goo
- AMO Lifescience Co., Ltd., Seoul, Seocho-gu, Republic of Korea
| | - Hye Ji Lim
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Korea; Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Korea
| | - Eun Ji Lee
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Korea; Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Korea
| | - Sang-Mo Kwon
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Korea; Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Korea.
| |
Collapse
|
5
|
Cho Y, Lee H, Jeong W, Jung KB, Lee SY, Park S, Yeun J, Kwon O, Son JG, Lee TG, Son MY, Im SG. Long-Term Culture of Human Pluripotent Stem Cells in Xeno-Free Condition Using Functional Polymer Films. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2403952. [PMID: 39015054 DOI: 10.1002/adma.202403952] [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: 03/18/2024] [Revised: 06/13/2024] [Indexed: 07/18/2024]
Abstract
Human pluripotent stem cells (hPSCs), encompassing human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs), hold immense potential in regenerative medicine, offering new opportunities for personalized cell therapies. However, their clinical translation is hindered by the inevitable reliance on xenogeneic components in culture environments. This study addresses this challenge by engineering a fully synthetic, xeno-free culture substrate, whose surface composition is tailored systematically for xeno-free culture of hPSCs. A functional polymer surface, pGC2 (poly(glycidyl methacrylate-grafting-guanidine-co-carboxylic acrylate)), offers excellent cell-adhesive properties as well as non-cytotoxicity, enabling robust hESCs and hiPSCs growth while presenting cost-competitiveness and scalability over Matrigel. This investigation includes comprehensive evaluations of pGC2 across diverse experimental conditions, demonstrating its wide adaptability with various pluripotent stem cell lines, culture media, and substrates. Crucially, pGC2 supports long-term hESCs and hiPSCs expansion, up to ten passages without compromising their stemness and pluripotency. Notably, this study is the first to confirm an identical proteomic profile after ten passages of xeno-free cultivation of hiPSCs on a polymeric substrate compared to Matrigel. The innovative substrate bridges the gap between laboratory research and clinical translation, offering a new promising avenue for advancing stem cell-based therapies.
Collapse
Affiliation(s)
- Younghak Cho
- Brain Science Institute, Korea Institute of Science and Technology (KIST), 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
- Functional Thin Film Laboratory, Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Hana Lee
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Wonji Jeong
- Functional Thin Film Laboratory, Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Kwang Bo Jung
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Sun Young Lee
- Korea Research Institute of Standards and Science (KRISS), Daejeon, 34141, Republic of Korea
| | - Seonghyeon Park
- Functional Thin Film Laboratory, Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Jemin Yeun
- Functional Thin Film Laboratory, Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Ohman Kwon
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Jin Gyeong Son
- Korea Research Institute of Standards and Science (KRISS), Daejeon, 34141, Republic of Korea
| | - Tae Geol Lee
- Korea Research Institute of Standards and Science (KRISS), Daejeon, 34141, Republic of Korea
| | - Mi-Young Son
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
- Department of Biological Science, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Sung Gap Im
- Functional Thin Film Laboratory, Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| |
Collapse
|
6
|
López-Carrasco A, Vieco-Martí I, Granados-Aparici S, Acevedo-León D, Estañ-Capell N, Portugal R, Huerta-Aragonés J, Cañete A, Navarro S, Noguera R. Vitronectin Levels in the Plasma of Neuroblastoma Patients and Culture Media of 3D Models: A Prognostic Circulating Biomarker? Int J Mol Sci 2024; 25:8733. [PMID: 39201421 PMCID: PMC11354570 DOI: 10.3390/ijms25168733] [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: 07/09/2024] [Revised: 08/02/2024] [Accepted: 08/09/2024] [Indexed: 09/02/2024] Open
Abstract
Vitronectin is a glycoprotein present in plasma and the extracellular matrix that is implicated in cell migration. The high amount of vitronectin found in neuroblastoma biopsies has been associated with poor prognosis. Moreover, increased vitronectin levels have been described in the plasma of patients with different cancers. Our aim was to assess vitronectin as a potential circulating biomarker of neuroblastoma prognosis. Vitronectin concentration was quantified using ELISA in culture media of four neuroblastoma cell lines grown in a monolayer and in 3D models, and in the plasma of 114 neuroblastoma patients. Three of the neuroblastoma cell lines secreted vitronectin to culture media when cultured in a monolayer and 3D models. Vitronectin release was higher by neuroblastoma cells cultured in 3D models than in the monolayer and was still elevated when cells were grown in 3D scaffolds with cross-linked vitronectin. Vitronectin secretion occurred independently of cell numbers in cultures. Its concentration in the plasma of neuroblastoma patients ranged between 52.4 and 870 µg/mL (median, 218 µg/mL). A ROC curve was used to establish a cutoff of 361 µg/mL, above which patients over 18 months old had worse prognosis (p = 0.0018). Vitronectin could be considered a new plasma prognostic biomarker in neuroblastoma and warrants confirmation in collaborative studies. Drugs inhibiting vitronectin interactions with cells and/or the extracellular matrix could represent a significant improvement in survival for neuroblastoma patients.
Collapse
Affiliation(s)
- Amparo López-Carrasco
- Incliva Biomedical Health Research Institute, 46010 Valencia, Spain; (A.L.-C.); (S.G.-A.); (S.N.)
- CIBER of Cancer (CIBERONC), 28029 Madrid, Spain
| | - Isaac Vieco-Martí
- Incliva Biomedical Health Research Institute, 46010 Valencia, Spain; (A.L.-C.); (S.G.-A.); (S.N.)
- CIBER of Cancer (CIBERONC), 28029 Madrid, Spain
| | - Sofía Granados-Aparici
- Incliva Biomedical Health Research Institute, 46010 Valencia, Spain; (A.L.-C.); (S.G.-A.); (S.N.)
- CIBER of Cancer (CIBERONC), 28029 Madrid, Spain
| | | | | | | | | | - Adela Cañete
- Politechnic and University Hospital La Fe, 46026 Valencia, Spain
| | - Samuel Navarro
- Incliva Biomedical Health Research Institute, 46010 Valencia, Spain; (A.L.-C.); (S.G.-A.); (S.N.)
- CIBER of Cancer (CIBERONC), 28029 Madrid, Spain
- Pathology Department, Medical School, University of Valencia, 46010 Valencia, Spain
| | - Rosa Noguera
- Incliva Biomedical Health Research Institute, 46010 Valencia, Spain; (A.L.-C.); (S.G.-A.); (S.N.)
- CIBER of Cancer (CIBERONC), 28029 Madrid, Spain
- Pathology Department, Medical School, University of Valencia, 46010 Valencia, Spain
| |
Collapse
|
7
|
Yehya H, Raudins S, Padmanabhan R, Jensen J, Bukys MA. Addressing bioreactor hiPSC aggregate stability, maintenance and scaleup challenges using a design of experiment approach. Stem Cell Res Ther 2024; 15:191. [PMID: 38956608 PMCID: PMC11218057 DOI: 10.1186/s13287-024-03802-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 06/16/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Stem cell-derived therapies hold the potential for treatment of regenerative clinical indications. Static culture has a limited ability to scale up thus restricting its use. Suspension culturing can be used to produce target cells in large quantities, but also presents challenges related to stress and aggregation stability. METHODS Utilizing a design of experiments (DoE) approach in vertical wheel bioreactors, we evaluated media additives that have versatile properties. The additives evaluated are Heparin sodium salt (HS), polyethylene glycol (PEG), poly (vinyl alcohol) (PVA), Pluronic F68 and dextran sulfate (DS). Multiple response variables were chosen to assess cell growth, pluripotency maintenance and aggregate stability in response to the additive inputs, and mathematical models were generated and tuned for maximal predictive power. RESULTS Expansion of iPSCs using 100 ml vertical wheel bioreactor assay for 4 days on 19 different media combinations resulted in models that can optimize pluripotency, stability, and expansion. The expansion optimization resulted in the combination of PA, PVA and PEG with E8. This mixture resulted in an expansion doubling time that was 40% shorter than that of E8 alone. Pluripotency optimizer highlighted the importance of adding 1% PEG to the E8 medium. Aggregate stability optimization that minimizes aggregate fusion in 3D culture indicated that the interaction of both Heparin and PEG can limit aggregation as well as increase the maintenance capacity and expansion of hiPSCs, suggesting that controlling fusion is a critical parameter for expansion and maintenance. Validation of optimized solution on two cell lines in bioreactors with decreased speed of 40 RPM, showed consistency and prolonged control over aggregates that have high frequency of pluripotency markers of OCT4 and SOX2 (> 90%). A doubling time of around 1-1.4 days was maintained after passaging as clumps in the optimized medium. Controlling aggregate fusion allowed for a decrease in bioreactor speed and therefore shear stress exerted on the cells in a large-scale expansion. CONCLUSION This study resulted in a control of aggregate size within suspension cultures, while informing about concomitant state control of the iPSC state. Wider application of this approach can address media optimization complexity and bioreactor scale-up challenges.
Collapse
Affiliation(s)
- Haneen Yehya
- Trailhead Biosystems, 23215 Commerce Park, Beachwood, OH, 44122, USA
- Cleveland State University, 2121 Euclid Ave, Cleveland, OH, 44115, USA
| | - Sofija Raudins
- Trailhead Biosystems, 23215 Commerce Park, Beachwood, OH, 44122, USA
| | | | - Jan Jensen
- Trailhead Biosystems, 23215 Commerce Park, Beachwood, OH, 44122, USA
| | - Michael A Bukys
- Trailhead Biosystems, 23215 Commerce Park, Beachwood, OH, 44122, USA.
| |
Collapse
|
8
|
KIMURA K, NAGAKURA H, TSUKAMOTO M, YOSHIDA T, SUGISAKI H, SHISHIDA K, TACHI Y, SHIMASAKI S, SUGIURA K, HATOYA S. Canine induced pluripotent stem cells can be successfully maintained in weekend-free culture systems. J Vet Med Sci 2024; 86:247-257. [PMID: 38171744 PMCID: PMC10963097 DOI: 10.1292/jvms.23-0422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 12/27/2023] [Indexed: 01/05/2024] Open
Abstract
Canine induced pluripotent stem cells (ciPSCs) can provide useful insights into novel therapies in both veterinary and medical fields. However, limited accessibility to the present culture medium and requirement of considerable time, effort, and cost for routine ciPSC maintenance restrict advancement in ciPSC research. In addition, it is unknown whether ciPSC culture conditions influence differentiation propensity. We investigated the availability of the common human pluripotent stem cells (hPSCs) culture systems for ciPSC maintenance and the differentiation propensities of the ciPSCs maintained in these culture systems. StemFlex and mTeSR Plus supported PSC-like colony formation and pluripotency markers expression in ciPSCs even after five passages. Additionally, ciPSCs were maintained under weekend-free culture conditions with a stable growth rate, pluripotency marker expression, and differentiation abilities using vitronectin (VTN-N) and Geltrex. Following maintenance of spontaneously differentiated ciPSCs under various conditions by embryoid body formation, there were few differences in the differentiation propensities of ciPSCs among the tested culture conditions. Thus, ciPSCs were successfully cultured under weekend-free conditions for ciPSC maintenance using StemFlex or mTeSR Plus with VTN-N or Geltrex. The present study offers simpler and more effort-, time-, and cost-saving options for ciPSC culture systems, which may lead to further development in research using ciPSCs.
Collapse
Affiliation(s)
- Kazuto KIMURA
- Department of Advanced Pathobiology, Graduate School of Veterinary Science, Osaka Metropolitan University, Osaka, Japan
| | - Hiroya NAGAKURA
- Department of Advanced Pathobiology, Graduate School of Veterinary Science, Osaka Metropolitan University, Osaka, Japan
| | - Masaya TSUKAMOTO
- Department of Advanced Pathobiology, Graduate School of Veterinary Science, Osaka Metropolitan University, Osaka, Japan
| | - Takumi YOSHIDA
- Department of Advanced Pathobiology, Graduate School of Veterinary Science, Osaka Metropolitan University, Osaka, Japan
| | - Hiroko SUGISAKI
- Department of Advanced Pathobiology, Graduate School of Veterinary Science, Osaka Metropolitan University, Osaka, Japan
| | - Kohei SHISHIDA
- Department of Advanced Pathobiology, Graduate School of Veterinary Science, Osaka Metropolitan University, Osaka, Japan
| | - Yuta TACHI
- Department of Advanced Pathobiology, Graduate School of Veterinary Science, Osaka Metropolitan University, Osaka, Japan
| | - Shoko SHIMASAKI
- Department of Advanced Pathobiology, Graduate School of Veterinary Science, Osaka Metropolitan University, Osaka, Japan
| | - Kikuya SUGIURA
- Department of Advanced Pathobiology, Graduate School of Veterinary Science, Osaka Metropolitan University, Osaka, Japan
| | - Shingo HATOYA
- Department of Advanced Pathobiology, Graduate School of Veterinary Science, Osaka Metropolitan University, Osaka, Japan
| |
Collapse
|
9
|
Cheng YS, Taniguchi Y, Yunoki Y, Masai S, Nogi M, Doi H, Sekiguchi K, Nakagawa M. Simultaneous binding of bFGF to both FGFR and integrin maintains properties of primed human induced pluripotent stem cells. Regen Ther 2024; 25:113-127. [PMID: 38226057 PMCID: PMC10788407 DOI: 10.1016/j.reth.2023.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 12/07/2023] [Accepted: 12/17/2023] [Indexed: 01/17/2024] Open
Abstract
Introduction Basic fibroblast growth factor (bFGF, FGF2) and integrin α6β1 are important for maintaining the pluripotency of human pluripotent stem cells (hPSCs). Although bFGF-integrin binding contributes to biofunctions in cancer cells, the relationship in hPSCs remains unclear. Methods To investigate the relationship between bFGF and integrin in human induced pluripotent stem cells (hiPSCs), we generated recombinant human bFGF wild-type and mutant proteins, that do not bind to integrin, FGFR, or both. We then cultured hiPSCs with these recombinant bFGF proteins. To evaluate the abilities of recombinant bFGF proteins in maintaining hPSC properties, pluripotent markers, ERK activity, and focal adhesion structure were analyzed through flow cytometry, immunofluorescence (IF), and immunoblotting (IB). Result We identified an interaction between bFGF and integrin α6β1 in vitro and in hiPSCs. The integrin non-binding mutant was incapable of inducing the hPSC properties, such as proliferation, ERK activity, and large focal adhesions at the edges of hiPSC colonies. Signaling induced by bFGF-FGFR binding was essential during the first 24 h after cell seeding for maintaining the properties of hPSCs, followed by a shift towards intracellular signaling via the bFGF-integrin interaction. The mixture of the two bFGF mutants also failed to maintain hPSC properties, indicating that bFGF binds to both FGFR and integrin. Conclusion Our study demonstrates that the integrin-bFGF-FGFR ternary complex maintains the properties of hPSCs via intracellular signaling, providing insights into the functional crosstalk between bFGF and integrins in hiPSCs.
Collapse
Affiliation(s)
- Yu-Shen Cheng
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507, Japan
| | - Yukimasa Taniguchi
- Division of Matrixome Research and Application, Institute for Protein Research, Osaka University, Osaka, 565-0871, Japan
| | - Yasuhiro Yunoki
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507, Japan
| | - Satomi Masai
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507, Japan
| | - Mizuho Nogi
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507, Japan
| | - Hatsuki Doi
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507, Japan
| | - Kiyotoshi Sekiguchi
- Division of Matrixome Research and Application, Institute for Protein Research, Osaka University, Osaka, 565-0871, Japan
| | - Masato Nakagawa
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507, Japan
| |
Collapse
|
10
|
Yang Y, Yang H, Kiskin FN, Zhang JZ. The new era of cardiovascular research: revolutionizing cardiovascular research with 3D models in a dish. MEDICAL REVIEW (2021) 2024; 4:68-85. [PMID: 38515776 PMCID: PMC10954298 DOI: 10.1515/mr-2023-0059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 01/18/2024] [Indexed: 03/23/2024]
Abstract
Cardiovascular research has heavily relied on studies using patient samples and animal models. However, patient studies often miss the data from the crucial early stage of cardiovascular diseases, as obtaining primary tissues at this stage is impracticable. Transgenic animal models can offer some insights into disease mechanisms, although they usually do not fully recapitulate the phenotype of cardiovascular diseases and their progression. In recent years, a promising breakthrough has emerged in the form of in vitro three-dimensional (3D) cardiovascular models utilizing human pluripotent stem cells. These innovative models recreate the intricate 3D structure of the human heart and vessels within a controlled environment. This advancement is pivotal as it addresses the existing gaps in cardiovascular research, allowing scientists to study different stages of cardiovascular diseases and specific drug responses using human-origin models. In this review, we first outline various approaches employed to generate these models. We then comprehensively discuss their applications in studying cardiovascular diseases by providing insights into molecular and cellular changes associated with cardiovascular conditions. Moreover, we highlight the potential of these 3D models serving as a platform for drug testing to assess drug efficacy and safety. Despite their immense potential, challenges persist, particularly in maintaining the complex structure of 3D heart and vessel models and ensuring their function is comparable to real organs. However, overcoming these challenges could revolutionize cardiovascular research. It has the potential to offer comprehensive mechanistic insights into human-specific disease processes, ultimately expediting the development of personalized therapies.
Collapse
Affiliation(s)
- Yuan Yang
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen, Guangdong Province, China
| | - Hao Yang
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen, Guangdong Province, China
| | - Fedir N. Kiskin
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen, Guangdong Province, China
| | - Joe Z. Zhang
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen, Guangdong Province, China
| |
Collapse
|
11
|
Falk J, Donadeu FX. Equine Induced Pluripotent Stem Cell Culture. Methods Mol Biol 2024; 2749:175-184. [PMID: 38133784 DOI: 10.1007/978-1-0716-3609-1_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
Groundbreaking work by Takahashi and Yamanaka in 2006 demonstrated that non-embryonic cells can be reprogrammed into pluripotent stem cells (PSCs) by forcing the expression of a defined set of transcription factors in culture, thus overcoming ethical concerns linked to embryonic stem cells. Induced PSCs have since revolutionized biomedical research, holding tremendous potential also in other areas such as livestock production and wildlife conservation. iPSCs exhibit broad accessibility, having been derived from a multitude of cell types and species. Apart from humans, iPSCs hold particular medical promise in the horse. The potential of iPSCs has been shown in a variety of biomedical contexts in the horse. However, progress in generating therapeutically useful equine iPSCs has lagged behind that reported in humans, with the generation of footprint-free iPSCs using non-integrative reprogramming approaches having proven particularly challenging. A greater understanding of the underlying molecular pathways and essential factors required for the generation and maintenance of equine iPSCs and their differentiation into relevant lineages will be critical for realizing their significant potential in veterinary regenerative medicine. This article outlines up-to-date protocols for the successful culture of equine iPSC, including colony selection, expansion, and adaptation to feeder-free conditions.
Collapse
Affiliation(s)
- Julia Falk
- Division of Translational Bioscience, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK.
| | - F Xavier Donadeu
- Division of Translational Bioscience, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
12
|
Iworima DG, Baker RK, Piret JM, Kieffer TJ. Analysis of the effects of bench-scale cell culture platforms and inoculum cell concentrations on PSC aggregate formation and culture. Front Bioeng Biotechnol 2023; 11:1267007. [PMID: 38107616 PMCID: PMC10722899 DOI: 10.3389/fbioe.2023.1267007] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 10/18/2023] [Indexed: 12/19/2023] Open
Abstract
Introduction: Human pluripotent stem cells (hPSCs) provide many opportunities for application in regenerative medicine due to their ability to differentiate into cells from all three germ layers, proliferate indefinitely, and replace damaged or dysfunctional cells. However, such cell replacement therapies require the economical generation of clinically relevant cell numbers. Whereas culturing hPSCs as a two-dimensional monolayer is widely used and relatively simple to perform, their culture as suspended three-dimensional aggregates may enable more economical production in large-scale stirred tank bioreactors. To be more relevant to this biomanufacturing, bench-scale differentiation studies should be initiated from aggregated hPSC cultures. Methods: We compared five available bench-scale platforms for generating undifferentiated cell aggregates of human embryonic stem cells (hESCs) using AggreWell™ plates, low attachment plates on an orbital shaker, roller bottles, spinner flasks, and vertical-wheel bioreactors (PBS-Minis). Thereafter, we demonstrated the incorporation of an hPSC aggregation step prior to directed differentiation to pancreatic progenitors and endocrine cells. Results and discussion: The AggreWell™ system had the highest aggregation yield. The initial cell concentrations had an impact on the size of aggregates generated when using AggreWell™ plates as well as in roller bottles. However, aggregates made with low attachment plates, spinner flasks and PBS-Minis were similar regardless of the initial cell number. Aggregate morphology was compact and relatively homogenously distributed in all platforms except for the roller bottles. The size of aggregates formed in PBS-Minis was modulated by the agitation rate during the aggregation. In all cell culture platforms, the net growth rate of cells in 3D aggregates was lower (range: -0.01-0.022 h-1) than cells growing as a monolayer (range: 0.039-0.045 h-1). Overall, this study describes operating ranges that yield high-quality undifferentiated hESC aggregates using several of the most commonly used bench-scale cell culture platforms. In all of these systems, methods were identified to obtain PSC aggregates with greater than 70% viability, and mean diameters between 60 and 260 mm. Finally, we showed the capacity of hPSC aggregates formed with PBS-Minis to differentiate into viable pancreatic progenitors and endocrine cell types.
Collapse
Affiliation(s)
- Diepiriye G. Iworima
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, BC, Canada
- School of Biomedical Engineering, The University of British Columbia, Vancouver, BC, Canada
| | - Robert K. Baker
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, BC, Canada
| | - James M. Piret
- School of Biomedical Engineering, The University of British Columbia, Vancouver, BC, Canada
- Michael Smith Laboratories, The University of British Columbia, Vancouver, BC, Canada
- Department of Chemical and Biological Engineering, The University of British Columbia, Vancouver, BC, Canada
| | - Timothy J. Kieffer
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, BC, Canada
- School of Biomedical Engineering, The University of British Columbia, Vancouver, BC, Canada
- Department of Surgery, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
13
|
Souralova T, Hulinova D, Jeseta M, Ventruba P, Hampl A, Koutna I. Truncated vitronectin with E-cadherin enables the xeno-free derivation of human embryonic stem cells. Sci Rep 2023; 13:15062. [PMID: 37700192 PMCID: PMC10497536 DOI: 10.1038/s41598-023-42236-5] [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: 06/27/2023] [Accepted: 09/07/2023] [Indexed: 09/14/2023] Open
Abstract
Human embryonic stem cells (hESCs) have unique abilities that enable their use in cell therapy, disease modeling, and drug development. Their derivation is usually performed using a feeder layer, which is undefined and can potentially cause a contamination by xeno components, therefore there is a tendency to replace feeders with xeno-free defined substrates in recent years. Three hESC lines were successfully derived on the vitronectin with a truncated N-terminus (VTN-N) in combination with E-cadherin in xeno-free conditions for the first time, and their undifferentiated state, hESC morphology, and standard karyotypes together with their potential to differentiate into three germ layers were confirmed. These results support the conclusion that the VTN-N/E-cadherin is a suitable substrate for the xeno-free derivation of hESCs and can be used for the derivation of hESCs according to good manufacturing practices.
Collapse
Affiliation(s)
- Tereza Souralova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
- International Clinical Research Center, Cell and Tissue Engineering Facility, St. Anne's University Hospital, Pekarska 53, 602 00, Brno, Czech Republic
| | - Daniela Hulinova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
- International Clinical Research Center, Cell and Tissue Engineering Facility, St. Anne's University Hospital, Pekarska 53, 602 00, Brno, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - Michal Jeseta
- Department of Gynecology and Obstetrics, Faculty of Medicine, Center of Assisted Reproduction, Masaryk University Brno and University Hospital, Obilni Trh 11, 602 00, Brno, Czech Republic
| | - Pavel Ventruba
- Department of Gynecology and Obstetrics, Faculty of Medicine, Center of Assisted Reproduction, Masaryk University Brno and University Hospital, Obilni Trh 11, 602 00, Brno, Czech Republic
| | - Ales Hampl
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
- International Clinical Research Center, Cell and Tissue Regeneration, St. Anne's University Hospital, Pekarska 53, 602 00, Brno, Czech Republic
| | - Irena Koutna
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic.
- International Clinical Research Center, Cell and Tissue Engineering Facility, St. Anne's University Hospital, Pekarska 53, 602 00, Brno, Czech Republic.
| |
Collapse
|
14
|
Timilsina S, McCandliss KF, Trivedi E, Villa-Diaz LG. Enhanced Expansion of Human Pluripotent Stem Cells and Somatic Cell Reprogramming Using Defined and Xeno-Free Culture Conditions. Bioengineering (Basel) 2023; 10:999. [PMID: 37760101 PMCID: PMC10525589 DOI: 10.3390/bioengineering10090999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/07/2023] [Accepted: 08/14/2023] [Indexed: 09/29/2023] Open
Abstract
Human embryonic stem cells and induced pluripotent stem cells (hPSC) have an unprecedented opportunity to revolutionize the fields of developmental biology as well as tissue engineering and regenerative medicine. However, their applications have been significantly limited by the lack of chemically defined and xeno-free culture conditions. The demand for the high-quality and scaled-up production of cells for use in both research and clinical studies underscores the need to develop tools that will simplify the in vitro culture process while reducing the variables. Here, we describe a systematic study to identify the optimal conditions for the initial cell attachment of hPSC to tissue culture dishes grafted with polymers of N-(3-Sulfopropyl)-N-Methacryloxyethyl-N, N-Dimethylammoniun Betaine (PMEDSAH) in combination with chemically defined and xeno-free culture media. After testing multiple supplements and chemicals, we identified that pre-conditioning of PMEDSAH grafted plates with 10% human serum (HS) supported the initial cell attachment, which allowed for the long-term culture and maintenance of hPSC compared to cells cultured on Matrigel-coated plates. Using this culture condition, a 2.1-fold increase in the expansion of hPSC was observed without chromosomal abnormalities. Furthermore, this culture condition supported a higher reprogramming efficiency (0.37% vs. 0.22%; p < 0.0068) of somatic cells into induced pluripotent stem cells compared to the non-defined culture conditions. This defined and xeno-free hPSC culture condition may be used in obtaining the large populations of hPSC and patient-derived iPSC required for many applications in regenerative and translational medicine.
Collapse
Affiliation(s)
- Suraj Timilsina
- Department of Biomarkers and Investigative Pathology Unit (BIPU), Charles River Laboratories, Mattawan, MI 49071, USA;
| | | | - Evan Trivedi
- Department of Chemistry, Oakland University, Rochester, MI 48309, USA;
| | - Luis G. Villa-Diaz
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, USA;
- Department of Bioengineering, Oakland University, Rochester, MI 48309, USA
| |
Collapse
|
15
|
Barrachina L, Arshaghi TE, O'Brien A, Ivanovska A, Barry F. Induced pluripotent stem cells in companion animals: how can we move the field forward? Front Vet Sci 2023; 10:1176772. [PMID: 37180067 PMCID: PMC10168294 DOI: 10.3389/fvets.2023.1176772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 04/04/2023] [Indexed: 05/15/2023] Open
Abstract
Following a one medicine approach, the development of regenerative therapies for human patients leads to innovative treatments for animals, while pre-clinical studies on animals provide knowledge to advance human medicine. Among many different biological products under investigation, stem cells are among the most prominent. Mesenchymal stromal cells (MSCs) are extensively investigated, but they present challenges such as senescence and limited differentiation ability. Embryonic stem cells (ESCs) are pluripotent cells with a virtually unlimited capacity for self-renewal and differentiation, but the use of embryos carries ethical concerns. Induced pluripotent stem cells (iPSCs) can overcome all of these limitations, as they closely resemble ESCs but are derived from adult cells by reprogramming in the laboratory using pluripotency-associated transcription factors. iPSCs hold great potential for applications in therapy, disease modeling, drug screening, and even species preservation strategies. However, iPSC technology is less developed in veterinary species compared to human. This review attempts to address the specific challenges associated with generating and applying iPSCs from companion animals. Firstly, we discuss strategies for the preparation of iPSCs in veterinary species and secondly, we address the potential for different applications of iPSCs in companion animals. Our aim is to provide an overview on the state of the art of iPSCs in companion animals, focusing on equine, canine, and feline species, as well as to identify which aspects need further optimization and, where possible, to provide guidance on future advancements. Following a "step-by-step" approach, we cover the generation of iPSCs in companion animals from the selection of somatic cells and the reprogramming strategies, to the expansion and characterization of iPSCs. Subsequently, we revise the current applications of iPSCs in companion animals, identify the main hurdles, and propose future paths to move the field forward. Transferring the knowledge gained from human iPSCs can increase our understanding in the biology of pluripotent cells in animals, but it is critical to further investigate the differences among species to develop specific approaches for animal iPSCs. This is key for significantly advancing iPSC application in veterinary medicine, which at the same time will also allow gaining pre-clinical knowledge transferable to human medicine.
Collapse
Affiliation(s)
| | | | | | | | - Frank Barry
- Regenerative Medicine Institute (REMEDI), Biosciences, University of Galway, Galway, Ireland
| |
Collapse
|
16
|
Abdal Dayem A, Lee SB, Lim KM, Kim A, Shin HJ, Vellingiri B, Kim YB, Cho SG. Bioactive peptides for boosting stem cell culture platform: Methods and applications. Biomed Pharmacother 2023; 160:114376. [PMID: 36764131 DOI: 10.1016/j.biopha.2023.114376] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 02/10/2023] Open
Abstract
Peptides, short protein fragments, can emulate the functions of their full-length native counterparts. Peptides are considered potent recombinant protein alternatives due to their specificity, high stability, low production cost, and ability to be easily tailored and immobilized. Stem cell proliferation and differentiation processes are orchestrated by an intricate interaction between numerous growth factors and proteins and their target receptors and ligands. Various growth factors, functional proteins, and cellular matrix-derived peptides efficiently enhance stem cell adhesion, proliferation, and directed differentiation. For that, peptides can be immobilized on a culture plate or conjugated to scaffolds, such as hydrogels or synthetic matrices. In this review, we assess the applications of a variety of peptides in stem cell adhesion, culture, organoid assembly, proliferation, and differentiation, describing the shortcomings of recombinant proteins and their full-length counterparts. Furthermore, we discuss the challenges of peptide applications in stem cell culture and materials design, as well as provide a brief outlook on future directions to advance peptide applications in boosting stem cell quality and scalability for clinical applications in tissue regeneration.
Collapse
Affiliation(s)
- Ahmed Abdal Dayem
- Department of Stem Cell and Regenerative Biotechnology, KU Convergence Science and Technology Institute, Konkuk University, Seoul 05029, Republic of Korea
| | - Soo Bin Lee
- Department of Stem Cell and Regenerative Biotechnology, KU Convergence Science and Technology Institute, Konkuk University, Seoul 05029, Republic of Korea
| | - Kyung Min Lim
- Department of Stem Cell and Regenerative Biotechnology, KU Convergence Science and Technology Institute, Konkuk University, Seoul 05029, Republic of Korea; R&D Team, StemExOne co., ltd. 303, Life Science Bldg, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Aram Kim
- Department of Urology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul 05029, Republic of Korea; R&D Team, StemExOne co., ltd. 303, Life Science Bldg, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Hyun Jin Shin
- Department of Ophthalmology, Research Institute of Medical Science, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul 05029, Republic of Korea; R&D Team, StemExOne co., ltd. 303, Life Science Bldg, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Balachandar Vellingiri
- Stem cell and Regenerative Medicine/Translational Research, Department of Zoology, School of Basic Sciences, Central University of Punjab (CUPB), Bathinda 151401, Punjab, India
| | - Young Bong Kim
- Department of Biomedical Science & Engineering, KU Convergence Science and Technology Institute, Konkuk University, Seoul 05029, Republic of Korea
| | - Ssang-Goo Cho
- Department of Stem Cell and Regenerative Biotechnology, KU Convergence Science and Technology Institute, Konkuk University, Seoul 05029, Republic of Korea; R&D Team, StemExOne co., ltd. 303, Life Science Bldg, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea.
| |
Collapse
|
17
|
Wang T, Yu T, Tsai CY, Hong ZY, Chao WH, Su YS, Subbiah SK, Renuka RR, Hsu ST, Wu GJ, Higuchi A. Xeno-free culture and proliferation of hPSCs on 2D biomaterials. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 199:63-107. [PMID: 37678982 DOI: 10.1016/bs.pmbts.2023.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
Human pluripotent stem cells (human embryonic stem cells (hESCs) and induced pluripotent stem cells (hiPSCs)) have unlimited proliferative potential, whereas adult stem cells such as bone marrow-derived stem cells and adipose-derived stem cells have problems with aging. When hPSCs are intended to be cultured on feeder-free or xeno-free conditions without utilizing mouse embryonic fibroblasts or human fibroblasts, they cannot be cultured on conventional tissue culture polystyrene dishes, as adult stem cells can be cultured but should be cultivated on material surfaces grafted or coated with (a) natural or recombinant extracellular matrix (ECM) proteins, (b) ECM protein-derived peptides and specific synthetic polymer surfaces in xeno-free and/or chemically defined conditions. This review describes current developing cell culture biomaterials for the proliferation of hPSCs while maintaining the pluripotency and differentiation potential of the cells into 3 germ layers. Biomaterials for the cultivation of hPSCs without utilizing a feeder layer are essential to decrease the risk of xenogenic molecules, which contributes to the potential clinical usage of hPSCs. ECM proteins such as human recombinant vitronectin, laminin-511 and laminin-521 have been utilized instead of Matrigel for the feeder-free cultivation of hPSCs. The following biomaterials are also discussed for hPSC cultivation: (a) decellularized ECM, (b) peptide-grafted biomaterials derived from ECM proteins, (c) recombinant E-cadherin-coated surface, (d) polysaccharide-immobilized surface, (e) synthetic polymer surfaces with and without bioactive sites, (f) thermoresponsive polymer surfaces with and without bioactive sites, and (g) synthetic microfibrous scaffolds.
Collapse
Affiliation(s)
- Ting Wang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China
| | - Tao Yu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China
| | - Chang-Yen Tsai
- Department of Chemical and Materials Engineering, National Central University, Jhongli, Taoyuan, Taiwan
| | - Zhao-Yu Hong
- Department of Chemical and Materials Engineering, National Central University, Jhongli, Taoyuan, Taiwan
| | - Wen-Hui Chao
- Department of Chemical and Materials Engineering, National Central University, Jhongli, Taoyuan, Taiwan
| | - Yi-Shuo Su
- Department of Chemical and Materials Engineering, National Central University, Jhongli, Taoyuan, Taiwan
| | - Suresh Kumar Subbiah
- Centre for Materials Engineering and Regenerative Medicine, Bharath Institute of Higher Education and Research, Chennai, India
| | - Remya Rajan Renuka
- Centre for Materials Engineering and Regenerative Medicine, Bharath Institute of Higher Education and Research, Chennai, India
| | - Shih-Tien Hsu
- Department of Internal Medicine, Landseed International Hospital, Pingjen City, Taoyuan, Taiwan
| | - Gwo-Jang Wu
- Graduate Institute of Medical Sciences and Department of Obstetrics & Gynecology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.
| | - Akon Higuchi
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China; Graduate Institute of Medical Sciences and Department of Obstetrics & Gynecology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.
| |
Collapse
|
18
|
Wang T, Liu Q, Chang YT, Liu J, Yu T, Maitiruze K, Ban LK, Sung TC, Subbiah SK, Renuka RR, Jen SH, Lee HHC, Higuchi A. Designed peptide-grafted hydrogels for human pluripotent stem cell culture and differentiation. J Mater Chem B 2023; 11:1434-1444. [PMID: 36541288 DOI: 10.1039/d2tb02521c] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Human pluripotent stem cells (hPSCs) have the ability to differentiate into cells derived from three germ layers and are an attractive cell source for cell therapy in regenerative medicine. However, hPSCs cannot be cultured on conventional tissue culture flasks but can be cultured on biomaterials with specific hPSC integrin interaction sites. We designed hydrogels conjugated with several designed peptides that had laminin-β4 active sites, optimal elasticities and different zeta potentials. A higher expansion fold of hPSCs cultured on the hydrogels was found with the increasing zeta potential of the hydrogels conjugated with designed peptides, where positive amino acid (lysine) insertion into the peptides promoted higher zeta potentials of the hydrogels and higher expansion folds of hPSCs when cultured on the hydrogels using xeno-free protocols. The hPSCs cultured on hydrogels conjugated with the optimal peptides showed a higher expansion fold than those on recombinant vitronectin-coated plates, which are the gold standard of hPSC cultivation dishes. The hPSCs could differentiate into specific cell lineages, such as mesenchymal stem cells (MSCs) and MSC-derived osteoblasts, even after being cultivated on hydrogels conjugated with optimal peptides for long periods of time, such as 10 passages.
Collapse
Affiliation(s)
- Ting Wang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, No. 270, Xueyuan Road, Wenzhou, Zhejiang, 325027, China.
| | - Qian Liu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, No. 270, Xueyuan Road, Wenzhou, Zhejiang, 325027, China.
| | - Yu-Tang Chang
- Department of Chemical and Materials Engineering, National Central University, No. 300, Jhongda RD., Jhongli, Taoyuan, 32001, Taiwan.
| | - Jun Liu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, No. 270, Xueyuan Road, Wenzhou, Zhejiang, 325027, China.
| | - Tao Yu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, No. 270, Xueyuan Road, Wenzhou, Zhejiang, 325027, China.
| | - Kailibinuer Maitiruze
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, No. 270, Xueyuan Road, Wenzhou, Zhejiang, 325027, China.
| | - Lee-Kiat Ban
- Department of Surgery, Hsinchu Cathay General Hospital, No. 678, Sec 2, Zhonghua Rd., Hsinchu, 30060, Taiwan
| | - Tzu-Cheng Sung
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, No. 270, Xueyuan Road, Wenzhou, Zhejiang, 325027, China.
| | - Suresh Kumar Subbiah
- Centre for Materials Engineering and Regenerative Medicine, Bharath Institute of Higher Education and Research, 173, Agaram Road, Tambaram East, Chennai-73, 600078, India
| | - Remya Rajan Renuka
- Centre for Materials Engineering and Regenerative Medicine, Bharath Institute of Higher Education and Research, 173, Agaram Road, Tambaram East, Chennai-73, 600078, India
| | - Shih Hsi Jen
- Department of Obstetrics and Gynecology, Taiwan Landseed Hospital, 77, Kuangtai Road, Pingjen City, Taoyuan 32405, Taiwan
| | - Henry Hsin-Chung Lee
- Department of Surgery, Hsinchu Cathay General Hospital, No. 678, Sec 2, Zhonghua Rd., Hsinchu, 30060, Taiwan.,Department of Surgery, Cathay General Hospital, Taipei, 10630, Taiwan. .,Graduate Institute of Translational and Interdisciplinary Medicine, National Central University, No. 300, Jhongda Rd., Jhongli, Taoyuan, 32001, Taiwan
| | - Akon Higuchi
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, No. 270, Xueyuan Road, Wenzhou, Zhejiang, 325027, China. .,Department of Chemical and Materials Engineering, National Central University, No. 300, Jhongda RD., Jhongli, Taoyuan, 32001, Taiwan. .,R&D Center for Membrane Technology, Chung Yuan Christian University, Chungli, Taoyuan 320, Taiwan
| |
Collapse
|
19
|
Ren Y, Zhang S, Liang Y, Gong Z, Cui Y, Song W. Feeder cells treated with ethanol can be used to maintain self-renewal and pluripotency of human pluripotent stem cells. FEBS Open Bio 2023; 13:279-292. [PMID: 36537760 PMCID: PMC9900095 DOI: 10.1002/2211-5463.13538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 11/24/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Feeder cells play an important role in the culture of human pluripotent stem cells (hPSCs) in vitro. Previously, we used methanol as a fixative to prepare feeder cells for the cultivation of pluripotent stem cells (PSCs), and this method could maintain the self-renewal and pluripotency of PSCs. However, methanol is toxic, and so here we examined whether ethanol could be used to prepare feeder cells as a fixative for hPSC culturing. Primed, naïve, and extended human embryonic stem cells and induced pluripotent stem cells can maintain self-renewal and undifferentiated potential on feeder cells treated with ethanol for an extended period. RNA sequencing analysis showed that the expression of collagen-related genes in hPSCs cultured on feeder cells treated with ethanol was significantly lower as compared with hPSCs cultured on feeder cells treated with mitomycin C. Therefore, we speculate that the signaling pathway mediated by collagen-related genes may, at least in part, contribute to the maintenance of self-renewal and pluripotency of PSCs induced by feeder cells treated with chemicals.
Collapse
Affiliation(s)
- Yahui Ren
- School of Life Science and EngineeringHenan University of Urban ConstructionPingdingshanChina
| | - Sijin Zhang
- School of Life Science and EngineeringHenan University of Urban ConstructionPingdingshanChina
| | - Yun Liang
- School of Life Science and EngineeringHenan University of Urban ConstructionPingdingshanChina
| | - Zichao Gong
- School of Life Science and EngineeringHenan University of Urban ConstructionPingdingshanChina
| | - Yongyi Cui
- School of Life Science and EngineeringHenan University of Urban ConstructionPingdingshanChina
| | - Wei Song
- School of Life Science and EngineeringHenan University of Urban ConstructionPingdingshanChina
| |
Collapse
|
20
|
Lin CY, Ching YY, Wu SF, Lee YK, Fan HC, Su LY, Tsai SY, Chen YC, Shen CI, Su HL. Coating-Free Culture Medium for Establishing and Maintaining Human Induced Pluripotent Stem Cells. Cell Transplant 2023; 32:9636897231198172. [PMID: 37698258 PMCID: PMC10498698 DOI: 10.1177/09636897231198172] [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: 05/01/2023] [Revised: 08/11/2023] [Accepted: 08/15/2023] [Indexed: 09/13/2023] Open
Abstract
Cell expansion of human pluripotent stem cells (hPSCs) commonly depends on Matrigel as a coating matrix on two-dimensional (2D) culture plates and 3D microcarriers. However, the xenogenic Matrigel requires sophisticated quality-assurance processes to meet clinical requirements. In this study, we develop an innovative coating-free medium for expanding hPSCs. The xenofree medium supports the weekend-free culture and competitive growth of hPSCs on several cell culture plastics without an additional pre-coating process. The pluripotent stemness of the expanded cells is stably sustained for more than 10 passages, featured with high pluripotent marker expressions, normal karyotyping, and differentiating capacity for three germ layers. The expression levels of some integrins are reduced, compared with those of the hPSCs on Matrigel. This medium also successfully supports the clonal expansion and induced pluripotent stem cell establishment from mitochondrial-defective MELAS (mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes) patient's peripheral blood mononuclear cells. This innovative hPSC medium provides a straightforward scale-up process for producing clinical-orientated hPSCs by excluding the conventional coating procedure.
Collapse
Affiliation(s)
- Chih-Yao Lin
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Yu-Yun Ching
- Duogenic StemCells Corporation, Taichung, Taiwan
| | - Shih-Fang Wu
- The Joint Program of Tissue Engineering and Regenerative Medicine, National Chung Hsing University and National Health Research Institutes, Taichung, Taiwan
| | - Yi-Ko Lee
- Duogenic StemCells Corporation, Taichung, Taiwan
| | - Hueng-Chuen Fan
- Department of Pediatrics, Tungs’ Taichung MetroHarbor Hospital, Wuchi, Taichung, Taiwan
- Department of Nursing, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli, Taiwan
| | - Liang-Yu Su
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - Su-Yi Tsai
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - Yu-Ching Chen
- The Joint Program of Tissue Engineering and Regenerative Medicine, National Chung Hsing University and National Health Research Institutes, Taichung, Taiwan
- Department of Obstetrics and Gynecology, Changhua Christian Hospital, Changhua City, Taiwan
| | - Ching-I Shen
- Duogenic StemCells Corporation, Taichung, Taiwan
| | - Hong-Lin Su
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
21
|
Cesare E, Urciuolo A, Stuart HT, Torchio E, Gesualdo A, Laterza C, Gagliano O, Martewicz S, Cui M, Manfredi A, Di Filippo L, Sabatelli P, Squarzoni S, Zorzan I, Betto RM, Martello G, Cacchiarelli D, Luni C, Elvassore N. 3D ECM-rich environment sustains the identity of naive human iPSCs. Cell Stem Cell 2022; 29:1703-1717.e7. [PMID: 36459970 DOI: 10.1016/j.stem.2022.11.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/07/2022] [Accepted: 11/09/2022] [Indexed: 12/03/2022]
Abstract
The establishment of in vitro naive human pluripotent stem cell cultures opened new perspectives for the study of early events in human development. The role of several transcription factors and signaling pathways have been characterized during maintenance of human naive pluripotency. However, little is known about the role exerted by the extracellular matrix (ECM) and its three-dimensional (3D) organization. Here, using an unbiased and integrated approach combining microfluidic cultures with transcriptional, proteomic, and secretome analyses, we found that naive, but not primed, hiPSC colonies are characterized by a self-organized ECM-rich microenvironment. Based on this, we developed a 3D culture system that supports robust long-term feeder-free self-renewal of naive hiPSCs and also allows direct and timely developmental morphogenesis simply by modulating the signaling environment. Our study opens new perspectives for future applications of naive hiPSCs to study critical stages of human development in 3D starting from a single cell.
Collapse
Affiliation(s)
- Elisa Cesare
- Department of Industrial Engineering, University of Padova, 6/a Via Gradenigo, Padova 35131, Italy; Veneto Institute of Molecular Medicine, 2 Via Orus, Padova 35131, Italy
| | - Anna Urciuolo
- University College London Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK; Institute of Pediatric Research IRP, Corso Stati Uniti, Padova 35127, Italy; Department of Molecular Medicine, University of Padova, Via G. Colombo 3, 35131 Padova, Italy
| | - Hannah T Stuart
- Department of Industrial Engineering, University of Padova, 6/a Via Gradenigo, Padova 35131, Italy; Veneto Institute of Molecular Medicine, 2 Via Orus, Padova 35131, Italy; The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK; Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Campus-Vienna-BioCenter 1, 1030 Vienna, Austria
| | - Erika Torchio
- Veneto Institute of Molecular Medicine, 2 Via Orus, Padova 35131, Italy
| | - Alessia Gesualdo
- Department of Industrial Engineering, University of Padova, 6/a Via Gradenigo, Padova 35131, Italy
| | - Cecilia Laterza
- Department of Industrial Engineering, University of Padova, 6/a Via Gradenigo, Padova 35131, Italy; Veneto Institute of Molecular Medicine, 2 Via Orus, Padova 35131, Italy
| | - Onelia Gagliano
- Department of Industrial Engineering, University of Padova, 6/a Via Gradenigo, Padova 35131, Italy; Veneto Institute of Molecular Medicine, 2 Via Orus, Padova 35131, Italy
| | - Sebastian Martewicz
- Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, 393 Middle Huaxia Road, Pudong, Shanghai 201210, China
| | - Meihua Cui
- Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, 393 Middle Huaxia Road, Pudong, Shanghai 201210, China
| | - Anna Manfredi
- Telethon Institute of Genetics and Medicine (TIGEM), Armenise/Harvard Laboratory of Integrative Genomics, Pozzuoli, Italy; Next Generation Diagnostic srl, Pozzuoli, Italy
| | - Lucio Di Filippo
- Telethon Institute of Genetics and Medicine (TIGEM), Armenise/Harvard Laboratory of Integrative Genomics, Pozzuoli, Italy; Next Generation Diagnostic srl, Pozzuoli, Italy
| | - Patrizia Sabatelli
- CNR - Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza" - Unit of Bologna, Bologna, Italy; IRCCS-Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Stefano Squarzoni
- CNR - Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza" - Unit of Bologna, Bologna, Italy; IRCCS-Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Irene Zorzan
- Epigenetics Programme, Babraham Institute, CB22 3AT Cambridge, UK
| | - Riccardo M Betto
- Department of Molecular Medicine, University of Padova, Via G. Colombo 3, 35131 Padova, Italy
| | - Graziano Martello
- Department of Biology, University of Padova, Via G. Colombo 3, Padova 35131, Italy
| | - Davide Cacchiarelli
- Telethon Institute of Genetics and Medicine (TIGEM), Armenise/Harvard Laboratory of Integrative Genomics, Pozzuoli, Italy; Department of Translational Medicine, University of Naples "Federico II", Naples, Italy; School for Advanced Studies, Genomics and Experimental Medicine Program, University of Naples "Federico II", Naples, Italy
| | - Camilla Luni
- Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, 393 Middle Huaxia Road, Pudong, Shanghai 201210, China; Department of Civil, Chemical, Environmental, and Materials Engineering (DICAM), University of Bologna, Via Terracini 28, Bologna 40131, Italy
| | - Nicola Elvassore
- Department of Industrial Engineering, University of Padova, 6/a Via Gradenigo, Padova 35131, Italy; Veneto Institute of Molecular Medicine, 2 Via Orus, Padova 35131, Italy; University College London Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK.
| |
Collapse
|
22
|
Dias TP, Baltazar T, Pinto SN, Fernandes TG, Fernandes F, Diogo MM, Prieto M, Cabral JMS. Xeno-Free Integrated Platform for Robust Production of Cardiomyocyte Sheets from hiPSCs. Stem Cells Int 2022; 2022:4542719. [PMID: 36467280 PMCID: PMC9712013 DOI: 10.1155/2022/4542719] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 10/17/2022] [Accepted: 11/02/2022] [Indexed: 08/05/2024] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) can be efficiently differentiated into cardiomyocytes (CMs), which can be used for cardiac disease modeling, for drug screening, and to regenerate damaged myocardium. Implementation of xeno-free culture systems is essential to fully explore the potential of these cells. However, differentiation using xeno-free adhesion matrices often results in low CM yields and lack of functional CM sheets, capable of enduring additional maturation stages. Here, we established a xeno-free CM differentiation platform using TeSR/Synthemax, including a replating step and integrated with two versatile purification/enrichment metabolic approaches. Results showed that the replating step was essential to reestablish a fully integrated, closely-knit CM sheet. In addition, replating contributed to increase the cTnT expression from 65% to 75% and the output from 2.2 to 3.1 CM per hiPSC, comparable with the efficiency observed when using TeSR/Matrigel. In addition, supplementation with PluriSin1 and Glu-Lac+ medium allowed increasing the CM content over 80% without compromising CM sheet integrity or functionality. Thus, this xeno-free differentiation platform is a reliable and robust method to produce hiPSC-derived CMs, increasing the possibility of using these cells safely for a wide range of applications.
Collapse
Affiliation(s)
- Tiago P. Dias
- iBB—Institute for Bioengineering and Biosciences and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Tânia Baltazar
- iBB—Institute for Bioengineering and Biosciences and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Sandra N. Pinto
- iBB—Institute for Bioengineering and Biosciences and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Tiago G. Fernandes
- iBB—Institute for Bioengineering and Biosciences and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Fábio Fernandes
- iBB—Institute for Bioengineering and Biosciences and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Maria Margarida Diogo
- iBB—Institute for Bioengineering and Biosciences and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Manuel Prieto
- iBB—Institute for Bioengineering and Biosciences and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Joaquim M. S. Cabral
- iBB—Institute for Bioengineering and Biosciences and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| |
Collapse
|
23
|
Jurj A, Ionescu C, Berindan-Neagoe I, Braicu C. The extracellular matrix alteration, implication in modulation of drug resistance mechanism: friends or foes? J Exp Clin Cancer Res 2022; 41:276. [PMID: 36114508 PMCID: PMC9479349 DOI: 10.1186/s13046-022-02484-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 09/01/2022] [Indexed: 11/10/2022] Open
Abstract
The extracellular matrix (ECM) is an important component of the tumor microenvironment (TME), having several important roles related to the hallmarks of cancer. In cancer, multiple components of the ECM have been shown to be altered. Although most of these alterations are represented by the increased or decreased quantity of the ECM components, changes regarding the functional alteration of a particular ECM component or of the ECM as a whole have been described. These alterations can be induced by the cancer cells directly or by the TME cells, with cancer-associated fibroblasts being of particular interest in this regard. Because the ECM has this wide array of functions in the tumor, preclinical and clinical studies have assessed the possibility of targeting the ECM, with some of them showing encouraging results. In the present review, we will highlight the most relevant ECM components presenting a comprehensive description of their physical, cellular and molecular properties which can alter the therapy response of the tumor cells. Lastly, some evidences regarding important biological processes were discussed, offering a more detailed understanding of how to modulate altered signalling pathways and to counteract drug resistance mechanisms in tumor cells.
Collapse
Affiliation(s)
- Ancuta Jurj
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, 400337, Cluj-Napoca, Romania
| | - Calin Ionescu
- 7Th Surgical Department, Iuliu Hațieganu University of Medicine and Pharmacy, 8 Victor Babes Street, 400012, Cluj-Napoca, Romania
- Surgical Department, Municipal Hospital, 400139, Cluj-Napoca, Romania
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, 400337, Cluj-Napoca, Romania.
| | - Cornelia Braicu
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, 400337, Cluj-Napoca, Romania.
- Research Center for Oncopathology and Translational Medicine (CCOMT), George Emil Palade University of Medicine, Pharmacy, Sciences and Technology, 540139, Targu Mures, Romania.
| |
Collapse
|
24
|
Alsobaie S, Alsobaie T, Mantalaris S. Rho-Associated Protein Kinase Inhibitor and Hypoxia Synergistically Enhance the Self-Renewal, Survival Rate, and Proliferation of Human Stem Cells. STEM CELLS AND CLONING: ADVANCES AND APPLICATIONS 2022; 15:43-52. [PMID: 35812359 PMCID: PMC9259205 DOI: 10.2147/sccaa.s365776] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 06/22/2022] [Indexed: 01/16/2023]
Abstract
Introduction High-efficacy single-cell cloning of human-induced pluripotent cells (IPSCs) remains a major challenge. The development of a culture method that supports single-cell passaging while maintaining reproducibility, homogeneity, scalability, and cell expansion to clinically relevant numbers is necessary for clinical application. Methods To address this issue, we combined the use of the rho-associated protein kinase (ROCK) inhibitor Y-27632 and hypoxic conditions in culture to produce a novel, efficient single-cell culture method for human IPSCs and embryonic stem cells. Results Through immunocytochemistry, alkaline phosphatase assays, and flow cytometry, we demonstrated that our method enabled high single-cell proliferation while maintaining self-renewal and pluripotency abilities. Discussion We showed the beneficial effect of the interaction between hypoxia and ROCK inhibition in regulating cell proliferation, pluripotency, and single-cell survival of pluripotent cells.
Collapse
Affiliation(s)
- Sarah Alsobaie
- Department of Clinical Laboratory Science, King Saud University, Riyadh, 11451, Saudi Arabia
- Correspondence: Sarah Alsobaie, Department of Clinical Laboratory Science, King Saud University, Prince Turki Alawal Street, Riyadh, 11451, Saudi Arabia, Tel +966 507191011, Fax +966 114677580, Email
| | - Tamador Alsobaie
- Biological Systems Engineering Laboratory, Department of Chemical Engineering, Imperial College London, London, SW7 2AZ, UK
| | - Sakis Mantalaris
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30322, USA
| |
Collapse
|
25
|
Aly RM, Aglan HA, Eldeen GN, Mahmoud NS, Aboul-Ezz EH, Ahmed HH. Efficient generation of functional pancreatic β cells from dental-derived stem cells via laminin-induced differentiation. J Genet Eng Biotechnol 2022; 20:85. [PMID: 35674918 PMCID: PMC9177930 DOI: 10.1186/s43141-022-00369-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 05/20/2022] [Indexed: 11/15/2022]
Abstract
Background This study was designed to generate functional insulin-producing cells (IPCs) from dental-derived mesenchymal stem cells (MSCs) and further explore their therapeutic potential against diabetes mellitus in vivo. MSCs were isolated from human dental pulp and periodontal ligament and were induced to differentiate into insulin-producing cells (IPCs) using laminin-based differentiation protocol for 14 days. Confirmation of IPCs was performed through real-time PCR analysis and insulin release assay. Then, the generated IPCs were labeled with PKH26 dye prior to transplantation in experimental animals. Twenty-eight days later, blood glucose, serum insulin (INS), c-peptide (CP), and visfatin (VF) levels and pancreatic glucagon (GC) level were estimated. Pancreatic forkhead box protein A2 (Foxa2) and SRY-box transcription factor 17 (Sox17), insulin-like growth factor I (IGF-1), and fibroblast growth factor10 (FGF 10) gene expression levels were analyzed. Results Dental stem cells were successfully differentiated into IPCs that demonstrated increased expression of pancreatic endocrine genes. IPCs released insulin after being subjected to high levels of glucose. In vivo findings uncovered that the implanted IPCs triggered significant decrease in blood glucose, serum VF, and pancreatic GC levels with significant increase in serum INS and CP levels. Furthermore, the implanted IPCs provoked significant upregulation in the expression level of pancreatic genes. Histopathological description of the pancreas tissues revealed that transplantation of IPCs ameliorated the destabilization of pancreas tissue architecture. Conclusion This study demonstrates the significant role of the implantation of IPCs generated from dental-derived stem cells in treatment of diabetes mellitus.
Collapse
Affiliation(s)
- Riham M Aly
- Basic Dental Science Department, Oral Medicine & Dentistry Research Institute, National Research Centre, Dokki, Giza, Egypt. .,Stem Cell Laboratory, Center of Excellence for Advanced Sciences, National Research Centre, 33 El Buhouth St., Dokki, 12622, Giza, Egypt.
| | - Hadeer A Aglan
- Stem Cell Laboratory, Center of Excellence for Advanced Sciences, National Research Centre, 33 El Buhouth St., Dokki, 12622, Giza, Egypt.,Hormones Department, Medicine Research and Clinical Studies Institute, National Research Centre, Dokki, Giza, Egypt
| | - Ghada Nour Eldeen
- Molecular Genetics & Enzymology Department, Human Genetic & Genome Research Institute, National Research Centre, Dokki, Giza, Egypt
| | - Nadia S Mahmoud
- Stem Cell Laboratory, Center of Excellence for Advanced Sciences, National Research Centre, 33 El Buhouth St., Dokki, 12622, Giza, Egypt.,Hormones Department, Medicine Research and Clinical Studies Institute, National Research Centre, Dokki, Giza, Egypt
| | - Eman H Aboul-Ezz
- Basic Dental Science Department, Oral Medicine & Dentistry Research Institute, National Research Centre, Dokki, Giza, Egypt.,Stem Cell Laboratory, Center of Excellence for Advanced Sciences, National Research Centre, 33 El Buhouth St., Dokki, 12622, Giza, Egypt
| | - Hanaa H Ahmed
- Stem Cell Laboratory, Center of Excellence for Advanced Sciences, National Research Centre, 33 El Buhouth St., Dokki, 12622, Giza, Egypt.,Hormones Department, Medicine Research and Clinical Studies Institute, National Research Centre, Dokki, Giza, Egypt
| |
Collapse
|
26
|
Hua Y, Yoshimochi K, Li J, Takekita K, Shimotsuma M, Li L, Qu X, Zhang J, Sawa Y, Liu L, Miyagawa S. Development and evaluation of a novel xeno-free culture medium for human-induced pluripotent stem cells. Stem Cell Res Ther 2022; 13:223. [PMID: 35658933 PMCID: PMC9166585 DOI: 10.1186/s13287-022-02879-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 05/03/2022] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Human-induced pluripotent stem cells (hiPSCs) are considered an ideal resource for regenerative medicine because of their ease of access and infinite expansion ability. To satisfy the sizable requirement for clinical applications of hiPSCs, large-scale, expansion-oriented, xeno-free, and cost-effective media are critical. Although several xeno-free media for hiPSCs have been generated over the past decades, few of them are suitable for scalable expansion of cultured hiPSCs because of their modest potential for proliferation and high cost. METHODS In this study, we developed a xeno-free ON2/AscleStem PSC medium (ON2) and cultured 253G1 hiPSCs on different matrices, including iMatrix-511 and gelatin nanofiber (GNF) in ON2. Over 20 passages, we evaluated cell proliferation by doubling times; pluripotency by flow cytometry, immunofluorescence staining and qRT-PCR; and differentiation ability by three germ layer differentiation in vitro and teratoma formation in severe combined immunodeficiency mice, followed by histological analysis. In addition, we compared the maintenance effect of ON2 on hiPSCs with StemFit® AK02 (AK02N) and Essential 8™ (E8). Besides 253G1 hiPSCs, we cultivated different hiPSC lines, including Ff-l01 hiPSCs, ATCC® ACS-1020™ hiPSCs, and Down's syndrome patient-specific ATCC® ACS-1003™ hiPSCs in ON2. RESULTS We found that 253G1 hiPSCs in ON2 demonstrated normal morphology and karyotype and high self-renewal and differentiation abilities on the tested matrices for over 20 passages. Moreover, 253G1 hiPSCs kept on GNF showed higher growth and stemness, as verified by the shorter doubling time and higher expression levels of pluripotent markers. Compared to AK02N and E8 media, 253G1 hiPSCs grown in ON2 showed higher pluripotency, as demonstrated by the increased expression level of pluripotent factors. In addition, all hiPSC lines cultivated in ON2 were able to grow for at least 10 passages with compact clonal morphology and were positive for all detected pluripotent markers. CONCLUSIONS Our xeno-free ON2 was compatible with various matrices and ideal for long-term expansion and maintenance of not only healthy-derived hiPSCs but also patient-specific hiPSCs. This highly efficient medium enabled the rapid expansion of hiPSCs in a reliable and cost-effective manner and could act as a promising tool for disease modeling and large-scale production for regenerative medicine in the future.
Collapse
Affiliation(s)
- Ying Hua
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, 565-0871, Japan
| | - Kenji Yoshimochi
- NACALAI TESQUE, INC. Research and Development Department, Kyoto, 604-0855, Japan
| | - Junjun Li
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, 565-0871, Japan.,Division of Cardiovascular Surgery, Department of Design for Tissue Regeneration, Graduate School of Medicine, Osaka, 565-0871, Japan
| | - Kazuhiro Takekita
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, 565-0871, Japan
| | - Motoshi Shimotsuma
- NACALAI TESQUE, INC. Research and Development Department, Kyoto, 604-0855, Japan
| | - Lingjun Li
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, 565-0871, Japan
| | - Xiang Qu
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, 565-0871, Japan
| | - Jingbo Zhang
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, 565-0871, Japan
| | | | - Li Liu
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, 565-0871, Japan. .,Division of Cardiovascular Surgery, Department of Design for Tissue Regeneration, Graduate School of Medicine, Osaka, 565-0871, Japan.
| | - Shigeru Miyagawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, 565-0871, Japan.
| |
Collapse
|
27
|
Methanol fixed feeder layers altered the pluripotency and metabolism of bovine pluripotent stem cells. Sci Rep 2022; 12:9177. [PMID: 35654935 PMCID: PMC9163156 DOI: 10.1038/s41598-022-13249-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 05/23/2022] [Indexed: 11/22/2022] Open
Abstract
The pluripotency maintenance of pluripotent stem cells (PSCs) requires the suitable microenvironment, which commonly provided by feeder layers. However, the preparation of feeder layers is time consuming and labor exhaustive, and the feeder cells treated with mitomycin C or γ-ray irradiation bring heterologous contamination. In this study, mouse embryonic fibroblasts (MEFs) were treated by methanol to generate chemical fixed feeder cells, and bovine embryonic stem cells F7 (bESC-F7) cultured on this feeder layer. Then the pluripotency and metabolism of bESC-F7 cultured on methanol-fixed MEFs (MT-MEFs) named MT-F7 was compared with mitomycin C treated MEFs (MC-MEFs). The results showed that bESC-F7 formed alkaline phosphatase positive colonies on MT-MEFs, the relative expression of pluripotent markers of these cells was different from the bESCs cultured on the MC-MEFs (MC-F7). The long-term cultured MT-F7 formed embryoid bodies, showed the ability to differentiate into three germ layers similar to MC-F7. The analyses of RNA-seq data showed that MT-MEFs lead bESCs to novel steady expression patterns of genes regulating pluripotency and metabolism. Furthermore, the bovine expanded pluripotent stem cells (bEPSCs) cultured on MT-MEFs formed classical colonies, maintained pluripotency, and elevated metabolism. In conclusion, MT-MEFs were efficient feeder layer that maintain the distinctive pluripotency and metabolism of PSCs.
Collapse
|
28
|
Yang F, Zhang D, Zhou Q, Li M, Xie C, Li S, Wang X, Wang W, Guo Y, Xiao Q, Wang Y, Gao L. Peptides-modified polystyrene-based polymers as high-performance substrates for the growth and propagation of human embryonic stem cells. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2021.10.028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
29
|
Etezadi F, Le MNT, Shahsavarani H, Alipour A, Moazzezy N, Samani S, Amanzadeh A, Pahlavan S, Bonakdar S, Shokrgozar MA, Hasegawa K. Optimization of a PDMS-Based Cell Culture Substrate for High-Density Human-Induced Pluripotent Stem Cell Adhesion and Long-Term Differentiation into Cardiomyocytes under a Xeno-Free Condition. ACS Biomater Sci Eng 2022; 8:2040-2052. [PMID: 35468288 DOI: 10.1021/acsbiomaterials.2c00162] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Despite the numerous advantages of PDMS-based substrates in various biomedical applications, they are limited by their highly hydrophobic surface that does not optimally interact with cells for attachment and growth. Hence, the lack of lengthy and straightforward procedures for high-density cell production on the PDMS-based substrate is one of the significant challenges in cell production in the cell therapy field. In this study, we found that the PDMS substrate coated with a combination of polydopamine (PDA) and laminin-511 E8 fragments (PDA + LME8-coated PDMS) can support human-induced pluripotent stem cell (hiPSC) attachment and growth for the long term and satisfy their demands of differentiation into cardiomyocytes (iCMs). Compared with prior studies, the density of hiPSCs and their adhesion time on the PDMS surface were increased during iCM production. Although the differentiated iCMs beat and produce mechanical forces, which disturb cellular attachments, the iCMs on the PDA + LME8-coated PDMS substrate showed dramatically better attachment than the control condition. Further, the substrate required less manipulation by enabling one-step seeding throughout the process in iCM formation from hiPSCs under animal-free conditions. In light of the results achieved, the PDA + LME8-coated PDMS substrate will be an up-and-coming tool for cardiomyocyte production for cell therapy and tissue engineering, microfluidics, and organ-on-chip platforms.
Collapse
Affiliation(s)
- Fatemeh Etezadi
- National Cell Bank of Iran, Pasteur Institute of Iran, No. 69, Pasteur Ave, Tehran 1316943551, Iran.,Institute for Integrated Cell-Material Sciences (iCeMS), Institute for Advanced Study, Kyoto University, Kyoto 606-8501, Japan.,Laboratory of Regenerative Medicine and Biomedical Innovations, Pasteur Institute of Iran, No. 69, Pasteur Ave, Tehran 1316943551, Iran
| | - Minh Nguyen Tuyet Le
- Institute for Integrated Cell-Material Sciences (iCeMS), Institute for Advanced Study, Kyoto University, Kyoto 606-8501, Japan
| | - Hosein Shahsavarani
- Laboratory of Regenerative Medicine and Biomedical Innovations, Pasteur Institute of Iran, No. 69, Pasteur Ave, Tehran 1316943551, Iran.,Department of Cell and Molecular Sciences, Faculty of Life Science and Biotechnology, Shahid Beheshti University, 1983963113 Tehran, Iran
| | - Atefeh Alipour
- Department of Nanobiotechnology, Pasteur Institute of Iran, No. 69, Pasteur Ave, Tehran 1316943551, Iran
| | - Neda Moazzezy
- Molecular Biology Department, Pasteur Institute of Iran, No. 69, Pasteur Ave, Tehran 1316943551, Iran
| | - Saeed Samani
- Department of Tissue Engineering & Applied Cell Sciences, TUMS School of Advanced Technologies in Medicine, No. 88, Italia St, Tehran, 1417755469, Iran
| | - Amir Amanzadeh
- National Cell Bank of Iran, Pasteur Institute of Iran, No. 69, Pasteur Ave, Tehran 1316943551, Iran
| | - Sara Pahlavan
- Department of Stem Cells and Development Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACERCR, Banihashem Ave, Tehran 16635-148, Iran
| | - Shahin Bonakdar
- National Cell Bank of Iran, Pasteur Institute of Iran, No. 69, Pasteur Ave, Tehran 1316943551, Iran
| | - Mohammad Ali Shokrgozar
- National Cell Bank of Iran, Pasteur Institute of Iran, No. 69, Pasteur Ave, Tehran 1316943551, Iran
| | - Kouichi Hasegawa
- Institute for Integrated Cell-Material Sciences (iCeMS), Institute for Advanced Study, Kyoto University, Kyoto 606-8501, Japan
| |
Collapse
|
30
|
Ostrowska-Podhorodecka Z, Ding I, Norouzi M, McCulloch CA. Impact of Vimentin on Regulation of Cell Signaling and Matrix Remodeling. Front Cell Dev Biol 2022; 10:869069. [PMID: 35359446 PMCID: PMC8961691 DOI: 10.3389/fcell.2022.869069] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 02/25/2022] [Indexed: 12/12/2022] Open
Abstract
Vimentin expression contributes to cellular mechanoprotection and is a widely recognized marker of fibroblasts and of epithelial-mesenchymal transition. But it is not understood how vimentin affects signaling that controls cell migration and extracellular matrix (ECM) remodeling. Recent data indicate that vimentin controls collagen deposition and ECM structure by regulating contractile force application to the ECM and through post-transcriptional regulation of ECM related genes. Binding of cells to the ECM promotes the association of vimentin with cytoplasmic domains of adhesion receptors such as integrins. After initial adhesion, cell-generated, myosin-dependent forces and signals that impact vimentin structure can affect cell migration. Post-translational modifications of vimentin determine its adaptor functions, including binding to cell adhesion proteins like paxillin and talin. Accordingly, vimentin regulates the growth, maturation and adhesive strength of integrin-dependent adhesions, which enables cells to tune their attachment to collagen, regulate the formation of cell extensions and control cell migration through connective tissues. Thus, vimentin tunes signaling cascades that regulate cell migration and ECM remodeling. Here we consider how specific properties of vimentin serve to control cell attachment to the underlying ECM and to regulate mesenchymal cell migration and remodeling of the ECM by resident fibroblasts.
Collapse
|
31
|
Zhou P, Qin L, Ge Z, Xie B, Huang H, He F, Ma S, Ren L, Shi J, Pei S, Dong G, Qi Y, Lan F. Design of chemically defined synthetic substrate surfaces for the in vitro maintenance of human pluripotent stem cells: A review. J Biomed Mater Res B Appl Biomater 2022; 110:1968-1990. [PMID: 35226397 DOI: 10.1002/jbm.b.35034] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 01/10/2022] [Accepted: 01/17/2022] [Indexed: 11/11/2022]
Abstract
Human pluripotent stem cells (hPSCs) have the potential of long-term self-renewal and differentiation into nearly all cell types in vitro. Prior to the downstream applications, the design of chemically defined synthetic substrates for the large-scale proliferation of quality-controlled hPSCs is critical. Although great achievements have been made, Matrigel and recombinant proteins are still widely used in the fundamental research and clinical applications. Therefore, much effort is still needed to improve the performance of synthetic substrates in the culture of hPSCs, realizing their commercial applications. In this review, we summarized the design of reported synthetic substrates and especially their limitations in terms of cell culture. Moreover, much attention was paid to the development of promising peptide displaying surfaces. Besides, the biophysical regulation of synthetic substrate surfaces as well as the three-dimensional culture systems were described.
Collapse
Affiliation(s)
- Ping Zhou
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Liying Qin
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Zhangjie Ge
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Biyao Xie
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Hongxin Huang
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Fei He
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Shengqin Ma
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Lina Ren
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Jiamin Shi
- Department of Laboratory Animal Centre, Changzhi Medical College, Changzhi, China
| | - Suying Pei
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Genxi Dong
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Yongmei Qi
- School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Feng Lan
- Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen Key Laboratory of Cardiovascular Disease, State Key Laboratory of Cardiovascular Disease, Shenzhen, China
| |
Collapse
|
32
|
Shimizu E, Iguchi H, Le MNT, Nakamura Y, Kobayashi D, Arai Y, Takakura K, Benno S, Yoshida N, Tsukahara M, Haneda S, Hasegawa K. A chemically-defined plastic scaffold for the xeno-free production of human pluripotent stem cells. Sci Rep 2022; 12:2516. [PMID: 35169157 PMCID: PMC8847402 DOI: 10.1038/s41598-022-06356-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 01/25/2022] [Indexed: 11/09/2022] Open
Abstract
Clinical use of human pluripotent stem cells (hPSCs) is hampered by the technical limitations of their expansion. Here, we developed a chemically synthetic culture substrate for human pluripotent stem cell attachment and maintenance. The substrate comprises a hydrophobic polyvinyl butyral-based polymer (PVB) and a short peptide that enables easy and uniform coating of various types of cell culture ware. The coated ware exhibited thermotolerance, underwater stability and could be stored at room temperature. The substrate supported hPSC expansion in combination with most commercial culture media with an efficiency similar to that of commercial substrates. It supported not only the long-term expansion of examined iPS and ES cell lines with normal karyotypes during their undifferentiated state but also directed differentiation of three germ layers. This substrate resolves major concerns associated with currently used recombinant protein substrates and could be applied in large-scale automated manufacturing; it is suitable for affordable and stable production of clinical-grade hPSCs and hPSC-derived products.
Collapse
Affiliation(s)
- Eiko Shimizu
- Institute for Integrated Cell-Material Sciences (iCeMS), Institute for Advanced Study, Kyoto University, Yoshida-Ushinomiya-cho, Sakyo-ku, Kyoto, 606-8501, Japan
- CiRA Foundation, Kyoto University, 53 Shogoin-Kawara-cho, Sakyo-ku, Kyoto, 606-8397, Japan
| | - Hiroki Iguchi
- Sekisui Chemical Co., Ltd., 2-1 Hyakuyama, Shimamoto-cho, Mishima-gun, Osaka, 618-0021, Japan
| | - Minh Nguyen Tuyet Le
- Institute for Integrated Cell-Material Sciences (iCeMS), Institute for Advanced Study, Kyoto University, Yoshida-Ushinomiya-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Yuta Nakamura
- Sekisui Chemical Co., Ltd., 2-1 Hyakuyama, Shimamoto-cho, Mishima-gun, Osaka, 618-0021, Japan
| | - Daigo Kobayashi
- Sekisui Chemical Co., Ltd., 2-1 Hyakuyama, Shimamoto-cho, Mishima-gun, Osaka, 618-0021, Japan
| | - Yuhei Arai
- Sekisui Chemical Co., Ltd., 2-1 Hyakuyama, Shimamoto-cho, Mishima-gun, Osaka, 618-0021, Japan
| | - Kenta Takakura
- Sekisui Chemical Co., Ltd., 2-1 Hyakuyama, Shimamoto-cho, Mishima-gun, Osaka, 618-0021, Japan
| | - Seiko Benno
- Sekisui Chemical Co., Ltd., 2-1 Hyakuyama, Shimamoto-cho, Mishima-gun, Osaka, 618-0021, Japan
| | - Noriko Yoshida
- Institute for Integrated Cell-Material Sciences (iCeMS), Institute for Advanced Study, Kyoto University, Yoshida-Ushinomiya-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Masayoshi Tsukahara
- CiRA Foundation, Kyoto University, 53 Shogoin-Kawara-cho, Sakyo-ku, Kyoto, 606-8397, Japan
| | - Satoshi Haneda
- Sekisui Chemical Co., Ltd., 2-1 Hyakuyama, Shimamoto-cho, Mishima-gun, Osaka, 618-0021, Japan.
| | - Kouichi Hasegawa
- Institute for Integrated Cell-Material Sciences (iCeMS), Institute for Advanced Study, Kyoto University, Yoshida-Ushinomiya-cho, Sakyo-ku, Kyoto, 606-8501, Japan.
| |
Collapse
|
33
|
Li S, Yoshioka M, Li J, Liu L, Ye S, Kamei KI, Chen Y. Nanocasting of fibrous morphology on a substrate for long-term propagation of human induced pluripotent stem cells. Biomed Mater 2022; 17. [PMID: 35114658 DOI: 10.1088/1748-605x/ac51b8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 02/03/2022] [Indexed: 11/12/2022]
Abstract
Human-induced pluripotent stem cells (hiPSCs) can be self-renewed for many generations on nanofibrous substrates. Herein, a casting method is developed to replicate the nanofibrous morphology into a thin layer of polymethylsiloxane (PDMS). The template is obtained by electrospinning and chemical crosslinking of gelatin nanofibers on a glass slide. The replicas of the template are surface-functionalized by gelatin and used for propagation of hiPSCs over tenth generations. The performance of the propagated hiPSCs is checked by immunofluorescence imaging, flowcytometry, and RT-PCR, confirming the utility of the method. The results are also compared with those obtained using electrospun nanofiber substrates. Inherently, the PDMS replicas is of low stiffness and can be reproduced easily. Compared to other patterning techniques, casting is more flexible and cost-effective, suggesting that this method might find applications in cell-based assays that rely on stringent consideration of both substrate stiffness and surface morphology.
Collapse
Affiliation(s)
- Sisi Li
- Chemistry, Ecole Normale Superieure, 24 rue Lhomond, Paris, Île-de-France, 75230, FRANCE
| | - Momoko Yoshioka
- Kyoto University, Yoshida Ushinomiya-cho, Kyoto, 606-8501, JAPAN
| | - Junjun Li
- Institute for Integrated Cell-Material Sciences, Yoshida Ushinomiya-cho, Kyoto, 606-8501, JAPAN
| | - Li Liu
- Kyoto University, Yoshida Ushinomiya-cho, Kyoto, 606-8501, JAPAN
| | - Sixin Ye
- University of Paris, 94276 Le Kremlin Bicêtre, Paris, 75006, FRANCE
| | - Ken-Ichiro Kamei
- Institute for Integrated Cell-Material Sciences, Yoshida Ushinomiya-cho, Kyoto, 606-8501, JAPAN
| | - Yong Chen
- Chemistry, Ecole Normale Superieure, 24 rue Lhomond, F-75231 Paris Cedex 05, Paris, Île-de-France, 75230, FRANCE
| |
Collapse
|
34
|
Labouesse C, Tan BX, Agley CC, Hofer M, Winkel AK, Stirparo GG, Stuart HT, Verstreken CM, Mulas C, Mansfield W, Bertone P, Franze K, Silva JCR, Chalut KJ. StemBond hydrogels control the mechanical microenvironment for pluripotent stem cells. Nat Commun 2021; 12:6132. [PMID: 34675200 PMCID: PMC8531294 DOI: 10.1038/s41467-021-26236-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 09/22/2021] [Indexed: 12/12/2022] Open
Abstract
Studies of mechanical signalling are typically performed by comparing cells cultured on soft and stiff hydrogel-based substrates. However, it is challenging to independently and robustly control both substrate stiffness and extracellular matrix tethering to substrates, making matrix tethering a potentially confounding variable in mechanical signalling investigations. Moreover, unstable matrix tethering can lead to poor cell attachment and weak engagement of cell adhesions. To address this, we developed StemBond hydrogels, a hydrogel in which matrix tethering is robust and can be varied independently of stiffness. We validate StemBond hydrogels by showing that they provide an optimal system for culturing mouse and human pluripotent stem cells. We further show how soft StemBond hydrogels modulate stem cell function, partly through stiffness-sensitive ERK signalling. Our findings underline how substrate mechanics impact mechanosensitive signalling pathways regulating self-renewal and differentiation, indicating that optimising the complete mechanical microenvironment will offer greater control over stem cell fate specification.
Collapse
Affiliation(s)
- Céline Labouesse
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, UK
| | - Bao Xiu Tan
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, UK
- Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge, CB3 0HE, UK
| | - Chibeza C Agley
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, UK
| | - Moritz Hofer
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, UK
| | - Alexander K Winkel
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3DY, UK
| | - Giuliano G Stirparo
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, UK
| | - Hannah T Stuart
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, UK
| | - Christophe M Verstreken
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, UK
- Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge, CB3 0HE, UK
| | - Carla Mulas
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, UK
| | - William Mansfield
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, UK
| | - Paul Bertone
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, UK
- Department of Medicine, Alpert Medical School, Brown University, Providence, IR, USA
| | - Kristian Franze
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3DY, UK
- Institute of Medical Physics, Friedrich-Alexander University Erlangen-Nuremberg, 91052, Erlangen, Germany
- Max-Planck-Zentrum für Physik und Medizin, 91054, Erlangen, Germany
| | - José C R Silva
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, UK.
- Center for Cell Lineage and Atlas, Guangzhou Laboratory, Guangzhou International Bio Island, 510005, Guangzhou, Guangdong Province, China.
| | - Kevin J Chalut
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, UK.
- Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge, CB3 0HE, UK.
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3DY, UK.
| |
Collapse
|
35
|
Klaas M, Möll K, Mäemets-Allas K, Loog M, Järvekülg M, Jaks V. Long-term maintenance of functional primary human hepatocytes in 3D gelatin matrices produced by solution blow spinning. Sci Rep 2021; 11:20165. [PMID: 34635750 PMCID: PMC8505433 DOI: 10.1038/s41598-021-99659-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 09/07/2021] [Indexed: 12/11/2022] Open
Abstract
Solution blow spinning (SBS) has recently emerged as a novel method that can produce nano- and microfiber structures suitable for tissue engineering. Gelatin is an excellent precursor for SBS as it is derived mainly from collagens that are abundant in natural extracellular matrices. Here we report, for the first time the successful generation of 3D thermally crosslinked preforms by using SBS from porcine gelatin. These SBS mats were shown to have three-dimensional fibrous porous structure similar to that of mammalian tissue extracellular matrix. In pharma industry, there is an urgent need for adequate 3D liver tissue models that could be used in high throughput setting for drug screening and to assess drug induced liver injury. We used SBS mats as culturing substrates for human hepatocytes to create an array of 3D human liver tissue equivalents in 96-well format. The SBS mats were highly cytocompatible, facilitated the induction of hepatocyte specific CYP gene expression in response to common medications, and supported the maintenance of hepatocyte differentiation and polarization status in long term cultures for more than 3 weeks. Together, our results show that SBS-generated gelatin scaffolds are a simple and efficient platform for use in vitro for drug testing applications.
Collapse
Affiliation(s)
- Mariliis Klaas
- Institute of Molecular and Cell Biology, University of Tartu, Riia 23b, 51010, Tartu, Estonia
| | - Kaidi Möll
- Institute of Technology, University of Tartu, Nooruse 1, 50411, Tartu, Estonia
| | - Kristina Mäemets-Allas
- Institute of Molecular and Cell Biology, University of Tartu, Riia 23b, 51010, Tartu, Estonia
| | - Mart Loog
- Institute of Technology, University of Tartu, Nooruse 1, 50411, Tartu, Estonia
| | - Martin Järvekülg
- Laboratory of Physics of Nanostructures, Institute of Physics, University of Tartu, W. Ostwaldi 1, 50411, Tartu, Estonia
| | - Viljar Jaks
- Institute of Molecular and Cell Biology, University of Tartu, Riia 23b, 51010, Tartu, Estonia.
- Dermatology Clinic, Tartu University Hospital, Raja 31, 50417, Tartu, Estonia.
| |
Collapse
|
36
|
Functional heterogeneity of IFN-γ-licensed mesenchymal stromal cell immunosuppressive capacity on biomaterials. Proc Natl Acad Sci U S A 2021; 118:2105972118. [PMID: 34446555 DOI: 10.1073/pnas.2105972118] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are increasingly combined with biomaterials to enhance their therapeutic properties, including their immunosuppressive function. However, clinical trials utilizing MSCs with or without biomaterials have shown limited success, potentially due to their functional heterogeneity across different donors and among different subpopulations of cells. Here, we evaluated the immunosuppressive capacity, as measured by the ability to reduce T-cell proliferation and activation, of interferon-gamma (IFN-γ)-licensed MSCs from multiple donors on fibrin and collagen hydrogels, the two most commonly utilized biomaterials in combination with MSCs in clinical trials worldwide according to ClinicalTrials.gov Variations in the immunosuppressive capacity between IFN-γ-licensed MSC donors on the biomaterials correlated with the magnitude of indoleamine-2,3-dioxygenase activity. Immunosuppressive capacity of the IFN-γ-licensed MSCs depended on the αV/α5 integrins when cultured on fibrin and on the α2/β1 integrins when cultured on collagen. While all tested MSCs were nearly 100% positive for these integrins, sorted MSCs that expressed higher levels of αV/α5 integrins demonstrated greater immunosuppressive capacity with IFN-γ licensing than MSCs that expressed lower levels of these integrins on fibrin. These findings were equivalent for MSCs sorted based on the α2/β1 integrins on collagen. These results demonstrate the importance of integrin engagement to IFN-γ licensed MSC immunosuppressive capacity and that IFN-γ-licensed MSC subpopulations of varying immunosuppressive capacity can be identified by the magnitude of integrin expression specific to each biomaterial.
Collapse
|
37
|
Zhu XY, Chen YH, Zhang T, Liu SJ, Bai XY, Huang XY, Jiang M, Sun XD. Improvement of human embryonic stem cell-derived retinal pigment epithelium cell adhesion, maturation, and function through coating with truncated recombinant human vitronectin. Int J Ophthalmol 2021; 14:1160-1167. [PMID: 34414078 DOI: 10.18240/ijo.2021.08.04] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 04/21/2021] [Indexed: 11/23/2022] Open
Abstract
AIM To explore an xeno-free and defined coating substrate suitable for the culture of H9 human embryonic stem cell-derived retinal pigment epithelial (hES-RPE) cells in vitro, and compare the behaviors and functions of hES-RPE cells on two culture substrates, laminin521 (LN-521) and truncated recombinant human vitronectin (VTN-N). METHODS hES-RPE cells were used in the experiment. The abilities of LN-521 and VTN-N at different concentrations to adhere to hES-RPE cells were compared with a high-content imaging system. Quantitative real-time polymerase chain reaction was used to evaluate RPE-specific gene expression levels midway (day 10) and at the end (day 20) of the time course. Cell polarity was observed by immunofluorescent staining for apical and basal markers of the RPE. The phagocytic ability of hES-RPE cells was identified by flow cytometry and immunofluorescence. RESULTS The cell adhesion assay showed that the ability of LN-521 to adhere to hES-RPE cells was dose-dependent. With increasing coating concentration, an increasing number of cells attached to the surface of LN-521-coated wells. In contrast, VTN-N presented a strong adhesive ability even at a low concentration. The optimal concentration of LN-521 and VTN-N required to coat and adhesion to hES-RPE cells were 2 and 0.25 µg/cm2, respectively. Furthermore, both LN-521 and VTN-N could facilitate adoption of the desired cobblestone cellular morphology with tight junction and showed polarity by the hES-RPE cells. However, hES-RPE cells cultivated in VTN-N had a greater phagocytic ability, and it took less time for these hES-RPE cells to mature. CONCLUSION VTN-N is a more suitable coating substrate for cultivating hES-RPE cells.
Collapse
Affiliation(s)
- Xin-Yue Zhu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.,National Clinical Research Center for Eye Diseases, Shanghai 200080, China.,Shanghai Key Laboratory of Fundus Diseases, Shanghai 200080, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai 200080, China
| | - Yu-Hong Chen
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.,National Clinical Research Center for Eye Diseases, Shanghai 200080, China.,Shanghai Key Laboratory of Fundus Diseases, Shanghai 200080, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai 200080, China
| | - Ting Zhang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.,National Clinical Research Center for Eye Diseases, Shanghai 200080, China.,Shanghai Key Laboratory of Fundus Diseases, Shanghai 200080, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai 200080, China
| | - Su-Jun Liu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.,National Clinical Research Center for Eye Diseases, Shanghai 200080, China.,Shanghai Key Laboratory of Fundus Diseases, Shanghai 200080, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai 200080, China
| | - Xin-Yue Bai
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.,National Clinical Research Center for Eye Diseases, Shanghai 200080, China.,Shanghai Key Laboratory of Fundus Diseases, Shanghai 200080, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai 200080, China
| | - Xian-Yu Huang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.,National Clinical Research Center for Eye Diseases, Shanghai 200080, China.,Shanghai Key Laboratory of Fundus Diseases, Shanghai 200080, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai 200080, China
| | - Mei Jiang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.,National Clinical Research Center for Eye Diseases, Shanghai 200080, China.,Shanghai Key Laboratory of Fundus Diseases, Shanghai 200080, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai 200080, China
| | - Xiao-Dong Sun
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.,National Clinical Research Center for Eye Diseases, Shanghai 200080, China.,Shanghai Key Laboratory of Fundus Diseases, Shanghai 200080, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai 200080, China
| |
Collapse
|
38
|
Suprunenko EA, Sazonova EA, Vasiliev AV. Extracellular Vesicles of Pluripotent Stem Cells. Russ J Dev Biol 2021. [DOI: 10.1134/s1062360421030073] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
39
|
Rivera T, Zhao Y, Ni Y, Wang J. Human-Induced Pluripotent Stem Cell Culture Methods Under cGMP Conditions. ACTA ACUST UNITED AC 2021; 54:e117. [PMID: 32649060 PMCID: PMC7507179 DOI: 10.1002/cpsc.117] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The discovery of induced pluripotent stem cells (iPSCs) revolutionized the approach to cell therapy in regenerative medicine. Reprogramming of somatic cells into an embryonic-like pluripotent state provides an invaluable resource of patient-specific cells of any lineage. Implementation of procedures and protocols adapted to current good manufacturing practice (cGMP) requirements is critical to ensure robust and consistent high-quality iPSC manufacturing. The technology developed at Allele Biotechnology for iPSC generation under cGMP conditions is a powerful platform for derivation of pluripotent stem cells through a footprint-free, feeder-free, and xeno-free reprogramming method. The cGMP process established by Allele Biotechnology entails fully cGMP compliant iPSC lines where the entire manufacturing process, from tissue collection, cell reprogramming, cell expansion, cell banking and quality control testing are adopted. Previously, we described in this series of publications how to create iPSCs using mRNA only, and how to do so under cGMP conditions. In this article, we describe in detail how to culture, examine and storage cGMP-iPSCs using reagents, materials and equipment compliant with cGMP standards. © 2020 The Authors. Basic Protocol 1: iPSC Dissociation Support Protocol 1: Stem cell media Support Protocol 2: ROCK inhibitor preparation Support Protocol 3: Vitronectin coating Basic Protocol 2: iPSC Cryopreservation Basic Protocol 3: iPSC Thawing.
Collapse
Affiliation(s)
- Teresa Rivera
- Allele Biotechnology and Pharmaceuticals, Inc., San Diego, California
| | - Yuanyuan Zhao
- Allele Biotechnology and Pharmaceuticals, Inc., San Diego, California
| | - Yuhui Ni
- Allele Biotechnology and Pharmaceuticals, Inc., San Diego, California
| | - Jiwu Wang
- Allele Biotechnology and Pharmaceuticals, Inc., San Diego, California
| |
Collapse
|
40
|
Lin SC, Loza A, Antrim L, Talbot P. Video bioinformatics analysis of human pluripotent stem cell morphology, quality, and cellular dynamics. Stem Cells Transl Med 2021; 10:1343-1359. [PMID: 34089307 PMCID: PMC8380446 DOI: 10.1002/sctm.15-0352] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 04/04/2021] [Accepted: 04/20/2021] [Indexed: 12/31/2022] Open
Abstract
StemCellQC is a video bioinformatics software tool for the quantitative analysis of human pluripotent stem cell (hPSC) colonies. Our objective was to use StemCellQC to evaluate and compare various experimental culture conditions, cell lines, and treatments and to demonstrate its applicability to PSC problems. Seven key features were identified that provided useful information on PSC morphology, dynamic behavior, and viability. Colony attachment was better on laminin‐521 than on Matrigel and Geltrex. Growth rates were similar on each matrix when data were normalized. The brightness/area ratio feature showed greater cell death in colonies grown on Matrigel and Geltrex than on laminin‐521 further contributing to an overall greater yield of cells on laminin‐521. Four different PSC culture media performed similarly; however, one medium produced batch‐to‐batch variation in colony morphology and dynamic features. Two embryonic and one induced pluripotent stem cell line showed significant differences in morphology, growth rates, motility, and death rates. Cells from the same vial that became phenotypically different in culture showed measurable differences in morphology, brightness, and motility. Likewise, differentiating and undifferentiated colonies varied in growth rate, intensity, and motility. Three pluripotent cell lines treated with a low concentration of cinnamaldehyde, a chemical used in consumer products, showed adverse effects and differed in their sensitivity to treatment. Our data demonstrate various applications of StemCellQC which could be used in basic and translational research, toxicological and drug testing, and clinical facilities engaged in stem cell therapy.
Collapse
Affiliation(s)
- Sabrina C Lin
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, California, USA
| | - Antonio Loza
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, California, USA
| | - Lauren Antrim
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, California, USA
| | - Prue Talbot
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, California, USA
| |
Collapse
|
41
|
Soto DA, Navarro M, Zheng C, Halstead MM, Zhou C, Guiltinan C, Wu J, Ross PJ. Simplification of culture conditions and feeder-free expansion of bovine embryonic stem cells. Sci Rep 2021; 11:11045. [PMID: 34040070 PMCID: PMC8155104 DOI: 10.1038/s41598-021-90422-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 05/05/2021] [Indexed: 02/04/2023] Open
Abstract
Bovine embryonic stem cells (bESCs) extend the lifespan of the transient pluripotent bovine inner cell mass in vitro. After years of research, derivation of stable bESCs was only recently reported. Although successful, bESC culture relies on complex culture conditions that require a custom-made base medium and mouse embryonic fibroblasts (MEF) feeders, limiting the widespread use of bESCs. We report here simplified bESC culture conditions based on replacing custom base medium with a commercially available alternative and eliminating the need for MEF feeders by using a chemically-defined substrate. bESC lines were cultured and derived using a base medium consisting of N2B27 supplements and 1% BSA (NBFR-bESCs). Newly derived bESC lines were easy to establish, simple to propagate and stable after long-term culture. These cells expressed pluripotency markers and actively proliferated for more than 35 passages while maintaining normal karyotype and the ability to differentiate into derivatives of all three germ lineages in embryoid bodies and teratomas. In addition, NBFR-bESCs grew for multiple passages in a feeder-free culture system based on vitronectin and Activin A medium supplementation while maintaining pluripotency. Simplified conditions will facilitate the use of bESCs for gene editing applications and pluripotency and lineage commitment studies.
Collapse
Affiliation(s)
- Delia Alba Soto
- Department of Animal Science, University of California, 450 Bioletti Way, Davis, CA, 95616, USA
| | - Micaela Navarro
- Department of Animal Science, University of California, 450 Bioletti Way, Davis, CA, 95616, USA
- Instituto de Investigaciones Biotecnológicas 'Dr Rodolfo Ugalde', UNSAM-CONICET, Buenos Aires, Argentina
| | - Canbin Zheng
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390, USA
| | | | - Chuan Zhou
- Department of Animal Science, University of California, 450 Bioletti Way, Davis, CA, 95616, USA
| | - Carly Guiltinan
- Department of Animal Science, University of California, 450 Bioletti Way, Davis, CA, 95616, USA
| | - Jun Wu
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390, USA
| | - Pablo Juan Ross
- Department of Animal Science, University of California, 450 Bioletti Way, Davis, CA, 95616, USA.
| |
Collapse
|
42
|
Defining optimal enzyme and matrix combination for replating of human induced pluripotent stem cell-derived cardiomyocytes at different levels of maturity. Exp Cell Res 2021; 403:112599. [PMID: 33848551 DOI: 10.1016/j.yexcr.2021.112599] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 03/25/2021] [Accepted: 04/04/2021] [Indexed: 11/24/2022]
Abstract
Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM) create an unlimited cell source for basic and translational research. Depending on the maturity of cardiac cultures and the intended applications, obtaining hiPSC-CMs as a single-cell, monolayer or three-dimensional clusters can be challenging. Here, we defined strategies to replate hiPSC-CMs on early days (D15-30) or later more mature (D60-150) differentiation cultures. After generation of hiPSCs and derivation of cardiomyocytes, four dissociation reagents Collagenase A/B, Collagenase II, TrypLE, EDTA and five different extracellular matrix materials Laminin, iMatrix-511, Fibronectin, Matrigel, and Geltrex were comparatively evaluated by imaging, cell viability, and contraction analysis. For early cardiac differentiation cultures mimicking mostly the embryonic stage, the highest adhesion, cell viability, and beating frequencies were achieved by treatment with the TrypLE enzyme. Video-based contraction analysis demonstrated higher beating rates after replating compared to before treatment. For later differentiation days of more mature cardiac cultures, dissociation with EDTA and replating cells on Geltrex or Laminin-derivatives yielded better recovery. Cardiac clusters at various sizes were detected in several groups treated with collagenases. Collectively, our findings revealed the selection criteria of the dissociation approach and coating matrix for replating iPSC-CMs based on the maturity and the requirements of further downstream applications.
Collapse
|
43
|
Singh SP, Kharche SD, Pathak M, Ranjan R, Soni YK, Saraswat S, Singh MK, Chauhan MS. Differential effects of extracellular matrix proteins on in vitro culture and growth characteristics of caprine male germ cells. In Vitro Cell Dev Biol Anim 2021; 57:373-380. [PMID: 33826044 DOI: 10.1007/s11626-021-00559-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 03/04/2021] [Indexed: 10/21/2022]
Affiliation(s)
- Shiva Pratap Singh
- Animal Physiology and Reproduction Division, ICAR-Central Institute for Research on Goats, Makhdoom, Farah, Mathura, Uttar Pradesh, 281 122, India.
| | - Suresh Dinkar Kharche
- Animal Physiology and Reproduction Division, ICAR-Central Institute for Research on Goats, Makhdoom, Farah, Mathura, Uttar Pradesh, 281 122, India
| | - Manisha Pathak
- Animal Physiology and Reproduction Division, ICAR-Central Institute for Research on Goats, Makhdoom, Farah, Mathura, Uttar Pradesh, 281 122, India
| | - Ravi Ranjan
- Animal Physiology and Reproduction Division, ICAR-Central Institute for Research on Goats, Makhdoom, Farah, Mathura, Uttar Pradesh, 281 122, India
| | - Yogesh Kumar Soni
- Animal Physiology and Reproduction Division, ICAR-Central Institute for Research on Goats, Makhdoom, Farah, Mathura, Uttar Pradesh, 281 122, India
| | - Sonia Saraswat
- Animal Physiology and Reproduction Division, ICAR-Central Institute for Research on Goats, Makhdoom, Farah, Mathura, Uttar Pradesh, 281 122, India
| | - Manoj Kumar Singh
- Animal Genetics and Breeding Division, ICAR-Central Institute for Research on Goats, Makhdoom, Farah, Mathura, Uttar Pradesh, 281 122, India
| | | |
Collapse
|
44
|
Virdi JK, Pethe P. Biomaterials Regulate Mechanosensors YAP/TAZ in Stem Cell Growth and Differentiation. Tissue Eng Regen Med 2021; 18:199-215. [PMID: 33230800 PMCID: PMC8012461 DOI: 10.1007/s13770-020-00301-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/15/2020] [Accepted: 09/12/2020] [Indexed: 02/07/2023] Open
Abstract
Tissue-resident stem cells are surrounded by a microenvironment known as 'stem cell niche' which is specific for each stem cell type. This niche comprises of cell-intrinsic and -extrinsic factors like biochemical and biophysical signals, which regulate stem cell characteristics and differentiation. Biochemical signals have been thoroughly studied however, the effect of biophysical signals on stem cell regulation is yet to be completely understood. Biomaterials have aided in addressing this issue since they can provide a defined and tuneable microenvironment resembling in vivo conditions. We review various biomaterials used in many studies which have shown a connection between biomaterial-generated mechanical signals and alteration in stem cell behaviour. Researchers probed to understand the mechanism of mechanotransduction and reported that the signals from the extracellular matrix regulate a transcription factor yes-associated protein (YAP)/transcriptional coactivator with PDZ-binding motif (TAZ), which is a downstream-regulator of the Hippo pathway and it transduces the mechanical signals inside the nucleus. We highlight the role of the YAP/TAZ as mechanotransducers in stem cell self-renewal and differentiation in response to substrate stiffness, also the possibility of mechanobiology as the emerging field of regenerative medicines and three-dimensional tissue printing.
Collapse
Affiliation(s)
- Jasmeet Kaur Virdi
- Department of Biological Science, Sunandan Divatia School of Science, SVKM's NMIMS (Deemed to-be) University, Mumbai, India
| | - Prasad Pethe
- Symbiosis Centre for Stem Cell Research (SCSCR), Symbiosis International University, Lavale, Mulshi, Pune, 412115, India.
| |
Collapse
|
45
|
Ornelas-González A, González-González M, Rito-Palomares M. Microcarrier-based stem cell bioprocessing: GMP-grade culture challenges and future trends for regenerative medicine. Crit Rev Biotechnol 2021; 41:1081-1095. [PMID: 33730936 DOI: 10.1080/07388551.2021.1898328] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Recently, stem cell-based therapies have been proposed as an alternative for the treatment of many diseases. Stem cells (SCs) are well known for their capacity to preserve themselves, proliferate, and differentiate into multiple lineages. These characteristics allow stem cells to be a viable option for the treatment of diverse diseases. Traditional methodologies based on 2-dimensional culture techniques (T-flasks and Petri dishes) are simple and well standardized; however, they present disadvantages that limit the production of the cell yield required for regenerative medicine applications. Lately, microcarrier (MC)-based culture techniques have emerged as an attractive platform for expanding stem cells in suspension systems. Although the use of stem cell expansion on MCs has recently shown significant increase, their implementation for medical purposes is been hampered by bottlenecks in upstream and downstream processing. Therefore, there is an urgent need in the development of bioprocesses that simplify stem cell cultures under xeno-free conditions and detachment from MCs without diminishing their pluripotency and viability. A critical analysis of the factors that impact the up and downstream bioprocessing on MC-based stem cell cultures is presented in this review. This analysis aims to raise the awareness of the current drawbacks that limit MC-based stem cell bioprocessing in regenerative medicine and propose alternatives to overcome them.
Collapse
Affiliation(s)
| | | | - Marco Rito-Palomares
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Mexico
| |
Collapse
|
46
|
Suzuki H, Kasai K, Kimura Y, Miyata S. UV/ozone surface modification combined with atmospheric pressure plasma irradiation for cell culture plastics to improve pluripotent stem cell culture. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 123:112012. [PMID: 33812631 DOI: 10.1016/j.msec.2021.112012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 02/24/2021] [Accepted: 02/27/2021] [Indexed: 10/22/2022]
Abstract
Culturing pluripotent stem cells effectively requires substrates coated with feeder cell layers or cell-adhesive matrices. It is difficult to employ pluripotent stem cells as resources for regenerative medicine due to risks of culture system contamination by animal-derived factors, or the large costs associated with the use of adhesive matrices. To enable a coating-free culture system, we focused on UV/ozone surface modification and atmospheric pressure plasma treatment for polystyrene substrates, to improve adhesion and proliferation of pluripotent stem cells. In this study, to develop a feeder- and matrix coating-free culture system for embryonic stem cells (ESCs), mouse ESCs were cultured on polystyrene substrates that were surface-modified using UV/ozone-plasma combined treatment. mESCs could be successfully cultured under feeder-free conditions upon UV/ozone-plasma combined treatment of culture substrates, without any further chemical treatments, and showed similar proliferation rates to those of cells grown on the feeder cell layer or matrix-coated substrates.
Collapse
Affiliation(s)
- Hayato Suzuki
- School of Integrated Design Engineering, Graduate School of Science & Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama, Kanagawa 223-8522, Japan
| | - Kohei Kasai
- School of Integrated Design Engineering, Graduate School of Science & Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama, Kanagawa 223-8522, Japan
| | - Yuka Kimura
- Department of Mechanical Engineering, Faculty of Science & Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama, Kanagawa, 223-8522, Japan
| | - Shogo Miyata
- Department of Mechanical Engineering, Faculty of Science & Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama, Kanagawa, 223-8522, Japan.
| |
Collapse
|
47
|
Nasir A, Thorpe J, Burroughs L, Meurs J, Pijuan‐Galito S, Irvine DJ, Alexander MR, Denning C. Discovery of a Novel Polymer for Xeno-Free, Long-Term Culture of Human Pluripotent Stem Cell Expansion. Adv Healthc Mater 2021; 10:e2001448. [PMID: 33369242 PMCID: PMC11469126 DOI: 10.1002/adhm.202001448] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 12/08/2020] [Indexed: 12/28/2022]
Abstract
Human pluripotent stem cells (hPSCs) can be expanded and differentiated in vitro into almost any adult tissue cell type, and thus have great potential as a source for cell therapies with biomedical application. In this study, a fully-defined polymer synthetic substrate is identified for hPSC culture in completely defined, xenogenic (xeno)-free conditions. This system can overcome the cost, scalability, and reproducibility limitations of current hPSC culture strategies, and facilitate large-scale production. A high-throughput, multi-generational polymer microarray platform approach is used to test over 600 unique polymers and rapidly assess hPSC-polymer interactions in combination with the fully defined xeno-free medium, Essential 8 (E8). This study identifies a novel nanoscale phase separated blend of poly(tricyclodecane-dimethanol diacrylate) and poly(butyl acrylate) (2:1 v/v), which supports long-term expansion of hPSCs and can be readily coated onto standard cultureware. Analysis of cell-polymer interface interactions through mass spectrometry and integrin blocking studies provides novel mechanistic insight into the role of the E8 proteins in promoting integrin-mediated hPSC attachment and maintaining hPSC signaling, including ability to undergo multi-lineage differentiation. This study therefore identifies a novel substrate for long-term serial passaging of hPSCs in serum-free, commercial chemically-defined E8, which provides a promising and economic hPSC expansion platform for clinical-scale application.
Collapse
Affiliation(s)
- Aishah Nasir
- Division of Cancer & Stem CellsBiodiscovery InstituteUniversity of NottinghamNottinghamNG7 2RDUK
| | - Jordan Thorpe
- Division of Cancer & Stem CellsBiodiscovery InstituteUniversity of NottinghamNottinghamNG7 2RDUK
| | | | - Joris Meurs
- School of PharmacyUniversity of NottinghamNottinghamNG7 2RDUK
| | - Sara Pijuan‐Galito
- Division of Cancer & Stem CellsBiodiscovery InstituteUniversity of NottinghamNottinghamNG7 2RDUK
| | - Derek J. Irvine
- Department of Chemical and Environmental EngineeringUniversity of NottinghamNottinghamNG7 2RDUK
| | | | - Chris Denning
- Division of Cancer & Stem CellsBiodiscovery InstituteUniversity of NottinghamNottinghamNG7 2RDUK
| |
Collapse
|
48
|
Human Induced Pluripotent Stem Cell (iPSC) Handling Protocols: Maintenance, Expansion, and Cryopreservation. Methods Mol Biol 2021; 2454:1-15. [PMID: 33837517 DOI: 10.1007/7651_2021_358] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Human induced pluripotent stem cells (iPSCs) have emerged as an invaluable resource for basic research, disease modeling, and drug discovery over recent years. Given the numerous advantages of iPSCs over alternative models-including their human origin, their ability to be differentiated into almost any cell type, and the therapeutic potential of patient-specific iPSCs in personalized medicine-many labs are now considering iPSC models for their studies. As the quality of the starting population of iPSCs is a key determinant in the success of any one of these applications, it is crucial to adhere to best practices in iPSC culture. In the following protocol, we offer a comprehensive guide to the culture, cryopreservation, and quality control methods required for the establishment and maintenance of high-quality iPSC cultures.
Collapse
|
49
|
Zhong C, Tao B, Tang F, Yang X, Peng T, You J, Xia K, Xia X, Chen L, Peng L. Remodeling cancer stemness by collagen/fibronectin via the AKT and CDC42 signaling pathway crosstalk in glioma. Am J Cancer Res 2021; 11:1991-2005. [PMID: 33408794 PMCID: PMC7778591 DOI: 10.7150/thno.50613] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 11/20/2020] [Indexed: 12/13/2022] Open
Abstract
Cancer development is a complex set of proliferative progression, which arises in most cases via multistep pathways associated with various factors, including the tumor microenvironment and extracellular matrix. However, the underlying mechanisms of cancer development remain unclear and this study aimed to explore the role of extracellular matrix in glioma progression. Methods: The expression of type I collagen and fibronectin in tumor tissues from glioma patients was examined by immunofluorescence staining. The correlations between collagen/fibronectin and glioma progression were then analyzed. A 3D collagen/fibronectin cultured system was established for tumor cells culture in vitro. Quantitative, real-time PCR and western blot were used to detect PI3K/ATK and CDC42 signals associated proteins expression in glioma. We used in vitro Cell Counting Kit-8, colony formation, and tumorigenesis assays to investigate the function of PI3K/AKT and CDC42 signals associated proteins. A xenograft glioma mice model was also used to study the anticancer effects of integrin inhibitor in vivo. Results: Our study demonstrated that type I collagen and fibronectin collaborate to regulate glioma cell stemness and tumor growth. In a 3D collagen/fibronectin culture model, glioma cells acquired tumorigenic potential and revealed strengthened proliferative characteristics. More significantly, collagen/fibronectin could facilitate the activation of PI3K/AKT/SOX2 and CDC42/YAP-1/NUPR1/Nestin signaling pathways via integrin αvβ3, eliciting sustained tumor growth and cancer relapse. Combination of the integrin signaling pathway inhibitor and the chemotherapeutic agent efficiently suppressed glioma cell proliferation and tumorigenic ability. Conclusion: We demonstrated that type I collagen and fibronectin could collaborate to promote glioma progression through PI3K/AKT/SOX2 and CDC42/YAP-1/NUPR1/Nestin signaling pathways. Blockade of the upstream molecular integrin αvβ3 revealed improved outcome in glioma therapy, which provide new insights for eradicating tumors and reducing glioma cancer relapse.
Collapse
|
50
|
Fonseca AC, Melchels FPW, Ferreira MJS, Moxon SR, Potjewyd G, Dargaville TR, Kimber SJ, Domingos M. Emulating Human Tissues and Organs: A Bioprinting Perspective Toward Personalized Medicine. Chem Rev 2020; 120:11128-11174. [PMID: 32937071 PMCID: PMC7645917 DOI: 10.1021/acs.chemrev.0c00342] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Indexed: 02/06/2023]
Abstract
The lack of in vitro tissue and organ models capable of mimicking human physiology severely hinders the development and clinical translation of therapies and drugs with higher in vivo efficacy. Bioprinting allow us to fill this gap and generate 3D tissue analogues with complex functional and structural organization through the precise spatial positioning of multiple materials and cells. In this review, we report the latest developments in terms of bioprinting technologies for the manufacturing of cellular constructs with particular emphasis on material extrusion, jetting, and vat photopolymerization. We then describe the different base polymers employed in the formulation of bioinks for bioprinting and examine the strategies used to tailor their properties according to both processability and tissue maturation requirements. By relating function to organization in human development, we examine the potential of pluripotent stem cells in the context of bioprinting toward a new generation of tissue models for personalized medicine. We also highlight the most relevant attempts to engineer artificial models for the study of human organogenesis, disease, and drug screening. Finally, we discuss the most pressing challenges, opportunities, and future prospects in the field of bioprinting for tissue engineering (TE) and regenerative medicine (RM).
Collapse
Affiliation(s)
- Ana Clotilde Fonseca
- Centre
for Mechanical Engineering, Materials and Processes, Department of
Chemical Engineering, University of Coimbra, Rua Sílvio Lima-Polo II, 3030-790 Coimbra, Portugal
| | - Ferry P. W. Melchels
- Institute
of Biological Chemistry, Biophysics and Bioengineering, School of
Engineering and Physical Sciences, Heriot-Watt
University, Edinburgh EH14 4AS, U.K.
| | - Miguel J. S. Ferreira
- Department
of Mechanical, Aerospace and Civil Engineering, School of Engineering,
Faculty of Science and Engineering, The
University of Manchester, Manchester M13 9PL, U.K.
| | - Samuel R. Moxon
- Division
of Neuroscience and Experimental Psychology, School of Biological
Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester M13 9PT, U.K.
| | - Geoffrey Potjewyd
- Division
of Neuroscience and Experimental Psychology, School of Biological
Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester M13 9PT, U.K.
| | - Tim R. Dargaville
- Institute
of Health and Biomedical Innovation, Science and Engineering Faculty, Queensland University of Technology, Queensland 4001, Australia
| | - Susan J. Kimber
- Division
of Cell Matrix Biology and Regenerative Medicine, School of Biological
Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester M13 9PT, U.K.
| | - Marco Domingos
- Department
of Mechanical, Aerospace and Civil Engineering, School of Engineering,
Faculty of Science and Engineering, The
University of Manchester, Manchester M13 9PL, U.K.
| |
Collapse
|