1
|
Chen Y, Xu Y, Cai J, Lauwers M, Xiang L, Zheng Y, Chu H, Chen X, Ker DFE, Zhang C, Wang DM, Zhang Z. Automated and Enclosed Three-Dimensional Biofabrication System for Mesenchymal Stem Cell Culture to Enhance Diabetic Wound Healing. Biomater Res 2025; 29:0205. [PMID: 40421058 PMCID: PMC12104559 DOI: 10.34133/bmr.0205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 04/01/2025] [Accepted: 04/19/2025] [Indexed: 05/28/2025] Open
Abstract
The industrialization of mesenchymal stem cells for regenerative medicine faces substantial challenges, particularly in large-scale production. Conventional 2-dimensional (2D) culture systems demonstrate limitations in meeting clinical requirements, such as inadequate cell yield, and poor cell-cell and cell-matrix interactions. These challenges can potentially be addressed by employing a 3D culture platform, which offers higher cell yields and enhanced efficacy. Moreover, it is essential to conduct a systematic and rigorous evaluation of cells produced in 3D culture systems to ensure their successful clinical translation. In this study, we cultured human umbilical cord mesenchymal stem cells (hUCMSCs) using an automated, scalable, and enclosed 3D microcarrier-bioreactor system, and comprehensively investigated their biological characteristics and potential therapeutic effects for diabetic wound repair. Our findings revealed that hUCMSCs harvested from this 3D microcarrier-bioreactor system are genetically stable and maintain the trilineage differentiation potential. Compared to hUCMSCs expanded under 2D conditions, those cultured in 3D exhibited reduced senescence and enhanced capabilities in migration, angiogenesis, and anti-inflammatory responses across different passages in vitro. RNA-sequencing analysis showed higher expression levels of genes related to angiogenesis and anti-inflammatory pathways in hUCMSCs cultured in 3D compared to those in 2D, which was further validated using quantitative real-time polymerase chain reaction and Western blot analysis. Additionally, 3D-cultured hUCMSCs demonstrated superior therapeutic effects for diabetic wound repair in mice, potentially due to their enhanced angiogenetic and anti-inflammatory effects. Collectively, our finding showcases the high quality of hUCMSCs cultured using an automated and enclosed 3D microcarrier-bioreactor system and their promising potential for diabetic wound repair.
Collapse
Affiliation(s)
- Yanmei Chen
- Translational Research Centre of Regenerative Medicine and 3D Printing, Department of Orthopaedic Surgery, Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, Guangdong Province Engineering Research Center for Biomedical Engineering, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital,
Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yang Xu
- School of Biomedical Sciences, Faculty of Medicine,
The Chinese University of Hong Kong, Hong Kong SAR, China
- Institute for Tissue Engineering and Regenerative Medicine,
The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jiawei Cai
- Translational Research Centre of Regenerative Medicine and 3D Printing, Department of Orthopaedic Surgery, Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, Guangdong Province Engineering Research Center for Biomedical Engineering, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital,
Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Marianne Lauwers
- Centre for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Hong Kong SAR, China
| | - Liwei Xiang
- Translational Research Centre of Regenerative Medicine and 3D Printing, Department of Orthopaedic Surgery, Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, Guangdong Province Engineering Research Center for Biomedical Engineering, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital,
Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yali Zheng
- Translational Research Centre of Regenerative Medicine and 3D Printing, Department of Orthopaedic Surgery, Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, Guangdong Province Engineering Research Center for Biomedical Engineering, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital,
Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Hua Chu
- Translational Research Centre of Regenerative Medicine and 3D Printing, Department of Orthopaedic Surgery, Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, Guangdong Province Engineering Research Center for Biomedical Engineering, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital,
Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xianglong Chen
- Translational Research Centre of Regenerative Medicine and 3D Printing, Department of Orthopaedic Surgery, Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, Guangdong Province Engineering Research Center for Biomedical Engineering, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital,
Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Dai Fei Elmer Ker
- Department of Biomedical Engineering, Faculty of Engineering, The
Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Cheng Zhang
- Translational Research Centre of Regenerative Medicine and 3D Printing, Department of Orthopaedic Surgery, Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, Guangdong Province Engineering Research Center for Biomedical Engineering, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital,
Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Dan Michelle Wang
- School of Biomedical Sciences, Faculty of Medicine,
The Chinese University of Hong Kong, Hong Kong SAR, China
- Institute for Tissue Engineering and Regenerative Medicine,
The Chinese University of Hong Kong, Hong Kong SAR, China
- Centre for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Hong Kong SAR, China
- Department of Orthopaedics and Traumatology,
The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Zhiyong Zhang
- Translational Research Centre of Regenerative Medicine and 3D Printing, Department of Orthopaedic Surgery, Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, Guangdong Province Engineering Research Center for Biomedical Engineering, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital,
Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
2
|
Hamann C, Kjar A, Kim H, Simmons AJ, Brien HJ, Quartey CI, Walton BL, Lau KS, Lippmann ES, Brunger JM. Induced Neural Progenitor Specification from Human Pluripotent Stem Cells by a Refined Synthetic Notch Platform. ACS Synth Biol 2025; 14:1482-1495. [PMID: 40327355 PMCID: PMC12090341 DOI: 10.1021/acssynbio.4c00742] [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: 10/25/2024] [Revised: 04/22/2025] [Accepted: 04/23/2025] [Indexed: 05/07/2025]
Abstract
Historically, studying the development of brain and central nervous system (CNS) tissues has been challenging. Human pluripotent stem cell (hPSC) technology has allowed for the in vitro reconstitution of relevant, early cell trajectories by using small molecules and recombinant proteins to guide differentiation of cells toward relevant brain and CNS phenotypes. However, many of these protocols fail to recapitulate the cell-guided differentiation programs intrinsic to embryonic development, particularly the signaling centers that emerge within the neural tube during brain formation. Located on the ventral end of the neural tube, the floor plate acts as one such signaling center to pattern the dorsal/ventral axis by secreting the morphogen Sonic Hedgehog (SHH). Here, we present a method for cell-guided differentiation using the synthetic Notch (synNotch) receptor platform to regulate SHH production and subsequent cell fate specification. We show that the widely used configuration of the orthogonal synNotch ligand green fluorescent protein (GFP) mounted on a platelet-derived growth factor receptor-β transmembrane chassis does not allow for robust artificial signaling in synNotch-hPSCs ("receivers") cocultured with ligand-presenting hPSCs ("senders"). We discovered that refined designs of membrane-bound GFP-ligand allow for efficient receptor activation in hPSC receivers. A variant of this enhanced synNotch system drives the production of SHH in hPSC sender:hPSC receiver cocultures and gives rise to floor plate-like cell types seen during neural tube development. This revised synNotch platform has the potential to pattern hPSC differentiation programs in synthetic morphogenesis studies designed to uncover key paradigms of human CNS development.
Collapse
Affiliation(s)
- Catherine
A. Hamann
- Department
of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Andrew Kjar
- Department
of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Hyosung Kim
- Department
of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Alan J. Simmons
- Department
of Cell and Developmental Biology, Vanderbilt
University, Nashville, Tennessee 37235, United States
| | - Hannah J. Brien
- Department
of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Cheryl I. Quartey
- Department
of Biological Sciences, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Bonnie L. Walton
- Department
of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Ken S. Lau
- Department
of Cell and Developmental Biology, Vanderbilt
University, Nashville, Tennessee 37235, United States
- Center
for Stem Cell Biology, Vanderbilt University, Nashville, Tennessee 37235, United States
- Center
for
Computational Systems Biology, Vanderbilt
University, Nashville, Tennessee 37235, United States
| | - Ethan S. Lippmann
- Department
of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
- Center
for Stem Cell Biology, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Jonathan M. Brunger
- Department
of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
- Center
for Stem Cell Biology, Vanderbilt University, Nashville, Tennessee 37235, United States
- Center
for
Computational Systems Biology, Vanderbilt
University, Nashville, Tennessee 37235, United States
| |
Collapse
|
3
|
Liu X, Wu J, Peng Y, Qian H, Lv X, Li F, Jin K, Niu Y, Song J, Han W, Chen G, Li B, Zuo Q. Chicken Primordial Germ Cells Do Not Proliferate in Insulin-Lacking Media. Int J Mol Sci 2025; 26:3122. [PMID: 40243906 PMCID: PMC11988930 DOI: 10.3390/ijms26073122] [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: 03/02/2025] [Revised: 03/25/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Abstract
Insulin is an important component of stem cell cultures; however, its role in the proliferation of avian primordial germ cells (PGCs) is unknown. The proliferation of PGCs in cultures varies and the growth factors and signaling pathways necessary to induce the proliferation of PGCs in chickens are unknown. Therefore, we conducted the present study to investigate the effect of insulin on the survival and proliferation of PGCs. In this study, we observed that under this culture system, PGCs proliferate in the presence of insulin, but do not proliferate in the absence of insulin. Furthermore, in insulin-lacking media, the expression of pluripotency genes, including LIN28, NANOG, POUV, and SOX2, was markedly decreased. Similarly, the expression of cell adhesion proteins ZO-1, Occludin, and JAM-A was significantly reduced. Elevated levels of ROS, GSSG, and MDA reduced the redox capacity of the cells and induced apoptosis. Subsequent transcriptome analyses revealed that insulin is one of the key factors in the proliferation of chicken PGCs through the regulation of downstream genes by PI3K/AKT, ECM-receptor interaction, Wnt, and P53 signaling, and that these downstream genes may be important for PGCs' proliferation and survival.
Collapse
Affiliation(s)
- Xin Liu
- Key Laboratory of Animal Genetics, Breeding and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Jun Wu
- Key Laboratory of Animal Genetics, Breeding and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Yixiu Peng
- Key Laboratory of Animal Genetics, Breeding and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Hongwu Qian
- Key Laboratory of Animal Genetics, Breeding and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Xiaoqian Lv
- Key Laboratory of Animal Genetics, Breeding and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Fan Li
- Key Laboratory of Animal Genetics, Breeding and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Kai Jin
- Key Laboratory of Animal Genetics, Breeding and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Yingjie Niu
- Key Laboratory of Animal Genetics, Breeding and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Jiuzhou Song
- Animal & Avian Sciences, University of Maryland, College Park, MA 20742, USA
| | - Wei Han
- Poultry Institute, Chinese Academy of Agricultural Sciences Poultry Institute of Jiangsu, Yangzhou 225003, China
| | - Guohong Chen
- Key Laboratory of Animal Genetics, Breeding and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Bichun Li
- Key Laboratory of Animal Genetics, Breeding and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
- College of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China
| | - Qisheng Zuo
- Key Laboratory of Animal Genetics, Breeding and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
4
|
Krishnamoorthy VK, Hamdani F, Shukla P, Rao RA, Anaitullah S, Biligiri KK, Kadumuri RV, Pothula PR, Chavali S, Rampalli S. NSD3 protein methylation and stabilization transforms human ES cells into variant state. Life Sci Alliance 2025; 8:e202402871. [PMID: 39741006 PMCID: PMC11707394 DOI: 10.26508/lsa.202402871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 11/29/2024] [Accepted: 11/29/2024] [Indexed: 01/02/2025] Open
Abstract
Cultured human embryonic stem cells (hESCs) can develop genetic anomalies that increase their susceptibility to transformation. In this study, we characterized a variant hESC (vhESC) line and investigated the molecular mechanisms leading to the drift towards a transformed state. Our findings revealed that vhESCs up-regulate EMT-specific markers, accelerate wound healing, exhibit compromised lineage differentiation, and retain pluripotency gene expression in teratomas. Furthermore, we discovered an altered epigenomic landscape and overexpression of the lysine methyltransferases EHMT1, EHMT2, and NSD group of proteins in vhESCs. Remarkably, depleting NSD3 oncogene reversed the molecular and phenotypic changes in vhESCs. We identified a detailed mechanism where EHMT2 interacts and methylates NSD3 at lysine 477, stabilizing its protein levels in vhESCs. In addition, we showed that NSD3 levels are regulated by protein degradation in hESCs, and its stabilization leads to the emergence of the variant state. Overall, our study identify that misregulation of NSD3 in pluripotent stem cells, through methylation-mediated abrogation of its protein degradation, drives hESCs towards oncogenic transformation.
Collapse
Affiliation(s)
- Vignesh K Krishnamoorthy
- https://ror.org/05ef28661 Council of Scientific and Industrial Research (CSIR) - Institute of Genomics and Integrative Biology (IGIB), New Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Fariha Hamdani
- https://ror.org/05ef28661 Council of Scientific and Industrial Research (CSIR) - Institute of Genomics and Integrative Biology (IGIB), New Delhi, India
| | - Pooja Shukla
- https://ror.org/05ef28661 Council of Scientific and Industrial Research (CSIR) - Institute of Genomics and Integrative Biology (IGIB), New Delhi, India
| | - Radhika Arasala Rao
- Institute for Stem Cell Science and Regenerative Medicine (DBT-inStem), GKVK Campus, Bangalore, India
| | - Shaikh Anaitullah
- Institute for Stem Cell Science and Regenerative Medicine (DBT-inStem), GKVK Campus, Bangalore, India
| | - Kriti Kestur Biligiri
- https://ror.org/05ef28661 Council of Scientific and Industrial Research (CSIR) - Institute of Genomics and Integrative Biology (IGIB), New Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Rajashekar Varma Kadumuri
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati, India
| | | | - Sreenivas Chavali
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati, India
| | - Shravanti Rampalli
- https://ror.org/05ef28661 Council of Scientific and Industrial Research (CSIR) - Institute of Genomics and Integrative Biology (IGIB), New Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
5
|
Jiang L, Wang J, Liu Z, Zhang Q, Yang XL. Seryl-tRNA synthetase inhibits Wnt signaling and breast cancer progression and metastasis. FASEB J 2025; 39:e70294. [PMID: 39760229 PMCID: PMC11817322 DOI: 10.1096/fj.202401720r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 12/06/2024] [Accepted: 12/23/2024] [Indexed: 01/07/2025]
Abstract
Tumors require ample protein synthesis to grow, and aminoacyl-tRNA synthetases, as critical translation factors, are expected to support cancer progression. Unexpectedly, overexpression of seryl-tRNA synthetase (SerRS) suppresses primary tumor growth of breast cancer. However, the effects of SerRS on metastasis have not been studied. We observe a decrease in SerRS expression in breast cancer patient metastases compared with matched primary tumors, suggesting an inhibitory role of SerRS in metastasis. Through mouse metastasis models using breast cancer cell lines overexpressing SerRS, we show that SerRS impedes not only primary tumor growth but also establishment of metastases, and the effect of SerRS on metastasis can be independent of its impact on the primary tumor. SerRS also inhibits tumor growth with induced, post-tumor-onset overexpression, demonstrating its potential as an anticancer therapeutic. Tumor RNA-seq analysis identified Wnt signaling among the top SerRS-regulated pathways. Using cell-based studies, we confirm SerRS suppresses Wnt signaling and metastatic processes in breast cancer cells. To the best of our knowledge, this is the first study to show a component of the translation machinery can act as both a tumor and metastasis suppressor.
Collapse
Affiliation(s)
- Lei Jiang
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Justin Wang
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Ze Liu
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Qian Zhang
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Xiang-Lei Yang
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, CA, 92037, USA
| |
Collapse
|
6
|
Matsuo-Takasaki M, Kambayashi S, Hemmi Y, Wakabayashi T, Shimizu T, An Y, Ito H, Takeuchi K, Ibuki M, Kawashima T, Masayasu R, Suzuki M, Kawai Y, Umekage M, Kato TM, Noguchi M, Nakade K, Nakamura Y, Nakaishi T, Nishishita N, Tsukahara M, Hayashi Y. Complete suspension culture of human induced pluripotent stem cells supplemented with suppressors of spontaneous differentiation. eLife 2024; 12:RP89724. [PMID: 39529479 PMCID: PMC11556790 DOI: 10.7554/elife.89724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) are promising resources for producing various types of tissues in regenerative medicine; however, the improvement in a scalable culture system that can precisely control the cellular status of hiPSCs is needed. Utilizing suspension culture without microcarriers or special materials allows for massive production, automation, cost-effectiveness, and safety assurance in industrialized regenerative medicine. Here, we found that hiPSCs cultured in suspension conditions with continuous agitation without microcarriers or extracellular matrix components were more prone to spontaneous differentiation than those cultured in conventional adherent conditions. Adding PKCβ and Wnt signaling pathway inhibitors in the suspension conditions suppressed the spontaneous differentiation of hiPSCs into ectoderm and mesendoderm, respectively. In these conditions, we successfully completed the culture processes of hiPSCs, including the generation of hiPSCs from peripheral blood mononuclear cells with the expansion of bulk population and single-cell sorted clones, long-term culture with robust self-renewal characteristics, single-cell cloning, direct cryopreservation from suspension culture and their successful recovery, and efficient mass production of a clinical-grade hiPSC line. Our results demonstrate that precise control of the cellular status in suspension culture conditions paves the way for their stable and automated clinical application.
Collapse
Affiliation(s)
- Mami Matsuo-Takasaki
- iPS Cell Advanced Characterization and Development Team, RIKEN BioResource Research CenterIbarakiJapan
| | - Sho Kambayashi
- Regenerative Medicine and Cell Therapy Laboratories, KANEKA CORPORATIONKobeJapan
| | - Yasuko Hemmi
- iPS Cell Advanced Characterization and Development Team, RIKEN BioResource Research CenterIbarakiJapan
| | - Tamami Wakabayashi
- iPS Cell Advanced Characterization and Development Team, RIKEN BioResource Research CenterIbarakiJapan
| | - Tomoya Shimizu
- iPS Cell Advanced Characterization and Development Team, RIKEN BioResource Research CenterIbarakiJapan
| | - Yuri An
- iPS Cell Advanced Characterization and Development Team, RIKEN BioResource Research CenterIbarakiJapan
| | - Hidenori Ito
- iPS Cell Advanced Characterization and Development Team, RIKEN BioResource Research CenterIbarakiJapan
| | - Kazuhiro Takeuchi
- Regenerative Medicine and Cell Therapy Laboratories, KANEKA CORPORATIONKobeJapan
| | - Masato Ibuki
- Regenerative Medicine and Cell Therapy Laboratories, KANEKA CORPORATIONKobeJapan
| | - Terasu Kawashima
- Regenerative Medicine and Cell Therapy Laboratories, KANEKA CORPORATIONKobeJapan
| | - Rio Masayasu
- Regenerative Medicine and Cell Therapy Laboratories, KANEKA CORPORATIONKobeJapan
| | - Manami Suzuki
- Regenerative Medicine and Cell Therapy Laboratories, KANEKA CORPORATIONKobeJapan
| | - Yoshikazu Kawai
- Regenerative Medicine and Cell Therapy Laboratories, KANEKA CORPORATIONKobeJapan
| | | | - Tomoaki M Kato
- Research and Development Center, CiRA FoundationKyotoJapan
| | - Michiya Noguchi
- Cell Engineering Division, RIKEN BioResource Research CenterIbarakiJapan
| | - Koji Nakade
- Gene Engineering Division, RIKEN BioResource Research CenterIbarakiJapan
| | - Yukio Nakamura
- Cell Engineering Division, RIKEN BioResource Research CenterIbarakiJapan
| | - Tomoyuki Nakaishi
- Regenerative Medicine and Cell Therapy Laboratories, KANEKA CORPORATIONKobeJapan
| | - Naoki Nishishita
- Regenerative Medicine and Cell Therapy Laboratories, KANEKA CORPORATIONKobeJapan
| | | | - Yohei Hayashi
- iPS Cell Advanced Characterization and Development Team, RIKEN BioResource Research CenterIbarakiJapan
- Faculty of Medicine and School of Integrative and Global Majors, University of TsukubaIbarakiJapan
| |
Collapse
|
7
|
Ma H, Xu L, Wu S, Wang S, Li J, Ai S, Yang Z, Mo R, Lin L, Li Y, Wang S, Gao J, Li C, Kong D. Supragel-mediated efficient generation of pancreatic progenitor clusters and functional glucose-responsive islet-like clusters. Bioact Mater 2024; 41:1-14. [PMID: 39101030 PMCID: PMC11292262 DOI: 10.1016/j.bioactmat.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/19/2024] [Accepted: 07/04/2024] [Indexed: 08/06/2024] Open
Abstract
Although several synthetic hydrogels with defined stiffness have been developed to facilitate the proliferation and maintenance of human pluripotent stem cells (hPSCs), the influence of biochemical cues in lineage-specific differentiation and functional cluster formation has been rarely reported. Here, we present the application of Supragel, a supramolecular hydrogel formed by synthesized biotinylated peptides, for islet-like cluster differentiation. We observed that Supragel, with a peptide concentration of 5 mg/mL promoted spontaneous hPSCs formation into uniform clusters, which is mainly attributable to a supporting stiffness of ∼1.5 kPa as provided by the Supragel matrix. Supragel was also found to interact with the hPSCs and facilitate endodermal and subsequent insulin-secreting cell differentiation, partially through its components: the sequences of RGD and YIGSR that interacts with cell membrane molecules of integrin receptor. Compared to Matrigel and suspension culturing conditions, more efficient differentiation of the hPSCs was also observed at the stages 3 and 4, as well as the final stage toward generation of insulin-secreting cells. This could be explained by 1) suitable average size of the hPSCs clusters cultured on Supragel; 2) appropriate level of cell adhesive sites provided by Supragel during differentiation. It is worth noting that the Supragel culture system was more tolerance in terms of the initial seeding densities and less demanding, since a standard static cell culture condition was sufficient for the entire differentiation process. Our observations demonstrate a positive role of Supragel for hPSCs differentiation into islet-like cells, with additional potential in facilitating germ layer differentiation.
Collapse
Affiliation(s)
- Hongmeng Ma
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Lilin Xu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Shengjie Wu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Songdi Wang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Jie Li
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Sifan Ai
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Zhuangzhuang Yang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Rigen Mo
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Lei Lin
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Yan Li
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Shusen Wang
- Research Institute of Transplant Medicine, Organ Transplant Center, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Jie Gao
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Chen Li
- Tianjin Key Laboratory of Biomedical Materials, Biomedical Barriers Research Center, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
| | - Deling Kong
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin, 300071, China
- College of Life Science, Key Laboratory of Bioactive Materials (Ministry of Education), State Key Laboratory of Medicinal Chemical Biology, Xu Rongxiang Regeneration Life Science Center, Nankai University, 300071, Tianjin, China
| |
Collapse
|
8
|
Liu X, Ye L, Ding Y, Gong W, Qian H, Jin K, Niu Y, Zuo Q, Song J, Han W, Chen G, Li B. Role of PI3K/AKT signaling pathway involved in self-renewing and maintaining biological properties of chicken primordial germ cells. Poult Sci 2024; 103:104140. [PMID: 39173217 PMCID: PMC11379996 DOI: 10.1016/j.psj.2024.104140] [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: 01/17/2024] [Revised: 06/29/2024] [Accepted: 07/25/2024] [Indexed: 08/24/2024] Open
Abstract
Avian primordial germ cells (PGCs) are important culture cells for the production of transgenic chickens and preservation of the genetic resources of endangered species; however, culturing these cells in vitro proves challenging. Although the proliferation of chicken PGCs is dependent on insulin, the underlying molecular mechanisms remain unclear. In the present study, we explored the expression of the PI3K/AKT signaling pathway in PGCs, investigated its effects on PGC self-renewal and biological properties, and identified the underlying mechanisms. Our findings indicated that although supplementation with the PI3K/AKT activator IGF-1 failed to promote proliferation under the assessed culture conditions, the PI3K/AKT inhibitor LY294002 resulted in retarded cell proliferation and reduced expression of germ cell-related markers. We further demonstrated that inhibition of PI3K/AKT regulates the cell cycle and promotes apoptosis in PGCs by activating the expression of BAX and inhibiting that of Bcl-2. These findings indicated that the PI3K/AKT pathway is required for cell renewal, apoptosis, and maintenance of the reproductive potential in chicken PGCs. This study aimed to provide a theoretical basis for the optimization and improvement of a culture system for chicken PGCs and provide insights into the self-renewal of vertebrate PGCs as well as potential evolutionary changes in this unique cell population.
Collapse
Affiliation(s)
- Xin Liu
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Liu Ye
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Ying Ding
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Wei Gong
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Hongwu Qian
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Kai Jin
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Yingjie Niu
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Qisheng Zuo
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Jiuzhou Song
- Animal & Avian Sciences, University of Maryland, College Park, MA 20742, USA
| | - Wei Han
- Poultry Institute, Chinese Academy of Agricultural Sciences Poultry Institute of Jiangsu, Yangzhou 225003, China
| | - Guohong Chen
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Bichun Li
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; College of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China.
| |
Collapse
|
9
|
Seitz M, Song Y, Lian XL, Ma Z, Jain E. Soft Polyethylene Glycol Hydrogels Support Human PSC Pluripotency and Morphogenesis. ACS Biomater Sci Eng 2024; 10:4525-4540. [PMID: 38973308 PMCID: PMC11234337 DOI: 10.1021/acsbiomaterials.4c00923] [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: 05/20/2024] [Revised: 06/05/2024] [Accepted: 06/05/2024] [Indexed: 07/09/2024]
Abstract
Lumenogenesis within the epiblast represents a critical step in early human development, priming the embryo for future specification and patterning events. However, little is known about the specific mechanisms that drive this process due to the inability to study the early embryo in vivo. While human pluripotent stem cell (hPSC)-based models recapitulate many aspects of the human epiblast, most approaches for generating these 3D structures rely on ill-defined, reconstituted basement membrane matrices. Here, we designed synthetic, nonadhesive polyethylene glycol (PEG) hydrogel matrices to better understand the role of matrix mechanical cues in iPSC morphogenesis, specifically elastic modulus. First, we identified a narrow range of hydrogel moduli that were conducive to the hPSC viability, pluripotency, and differentiation. We then used this platform to investigate the effects of the hydrogel modulus on lumenogenesis, finding that matrices of intermediate stiffness yielded the most epiblast-like aggregates. Conversely, stiffer matrices impeded lumen formation and apico-basal polarization, while the softest matrices yielded polarized but aberrant structures. Our approach offers a simple, modular platform for modeling the human epiblast and investigating the role of matrix cues in its morphogenesis.
Collapse
Affiliation(s)
- Michael
P. Seitz
- Department
of Biomedical and Chemical Engineering, Syracuse University, Syracuse, New York 13244, United States
- Bioinspired
Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, New York 13244, United States
| | - Yuanhui Song
- Department
of Biomedical and Chemical Engineering, Syracuse University, Syracuse, New York 13244, United States
- Bioinspired
Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, New York 13244, United States
| | - Xiaojun Lance Lian
- Department
of Biomedical Engineering, The Huck Institutes of the Life Sciences,
Department of Biology, Pennsylvania State
University, University
Park, Pennsylvania 16802, United States
| | - Zhen Ma
- Department
of Biomedical and Chemical Engineering, Syracuse University, Syracuse, New York 13244, United States
- Bioinspired
Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, New York 13244, United States
| | - Era Jain
- Department
of Biomedical and Chemical Engineering, Syracuse University, Syracuse, New York 13244, United States
- Bioinspired
Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, New York 13244, United States
| |
Collapse
|
10
|
Kim MH, Thanuthanakhun N, Kino-oka M. Stable and efficient generation of functional iPSC-derived neural progenitor cell rosettes through regulation of collective cell-cell behavior. Front Bioeng Biotechnol 2024; 11:1269108. [PMID: 38268936 PMCID: PMC10806250 DOI: 10.3389/fbioe.2023.1269108] [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: 07/31/2023] [Accepted: 12/21/2023] [Indexed: 01/26/2024] Open
Abstract
Although the potential of stem cells to differentiate into several cell types has shown promise in regenerative medicine, low differentiation efficiency and poor reproducibility significantly limit their practical application. We developed an effective and robust differentiation strategy for the efficient and robust generation of neural progenitor cell rosettes from induced pluripotent stem cells (iPSCs) incorporating botulinum hemagglutinin (HA). Treatment with HA suppressed the spontaneous differentiation of iPSCs cultured under undirected differentiation conditions, resulting in the preservation of their pluripotency. Moreover, treatment with HA during neural progenitor differentiation combined with dual SMAD inhibition generated a highly homogeneous population of PAX6-and SOX1-expressing neural progenitor cells with 8.4-fold higher yields of neural progenitor cells than untreated control cultures. These neural progenitor cells formed radially organized rosettes surrounding the central lumen. This differentiation method enhanced the generation of functional iPSC-derived neural progenitor cell rosettes throughout the culture vessel, suggesting that the regulation of collective cell-cell behavior using HA plays a morphogenetically important role in rosette formation and maturation. These findings show the significance of HA in the suppression of spontaneous differentiation through spatial homogeneity. The study proposes a novel methodology for the efficient derivation of functional iPSC-derived neural progenitor cell rosettes.
Collapse
Affiliation(s)
- Mee-Hae Kim
- Department of Biotechnology, Graduate School of Engineering, Osaka University, Osaka, Japan
| | | | - Masahiro Kino-oka
- Department of Biotechnology, Graduate School of Engineering, Osaka University, Osaka, Japan
- Research Base for Cell Manufacturability, Osaka University, Osaka, Japan
| |
Collapse
|
11
|
Kim MH, Thanuthanakhun N, Kino-Oka M. A simple tool for the synchronous differentiation of human induced pluripotent stem cells into pancreatic progenitors. Biotechnol J 2024; 19:e2300364. [PMID: 37955342 DOI: 10.1002/biot.202300364] [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: 07/28/2023] [Revised: 10/01/2023] [Accepted: 11/09/2023] [Indexed: 11/14/2023]
Abstract
Efficient differentiation of human induced pluripotent stem cells (hiPSCs) into functional pancreatic cells holds great promise for diabetes research and treatment. However, a robust culture strategy for producing pancreatic progenitors with high homogeneity is lacking. Here, we established a simple differentiation strategy for generating synchronous iPSC-derived pancreatic progenitors via a two-step method of sequential cell synchronization using botulinum hemagglutinin (HA), an E-cadherin function-blocking agent. Of the various methods tested, the first-step synchronization method with HA exposure induces a synchronous switch from E- to N-cadherin and N- to E-cadherin expression by spatially controlling heterogeneous cell distribution, subsequently improving their competency for directed differentiation into definitive endodermal cells from iPSCs. The iPSC-derived definitive endodermal cells can efficiently generate PDX1+ and NKX6.1+ pancreatic progenitor cells in high yields. The PDX1+ and PDX1+ /NKX6.1+ cell densities showed 1.6- and 2.2-fold increases, respectively, compared with those from unsynchronized cultures. The intra-run and inter-run coefficient of variation were below 10%, indicating stable and robust differentiation across different cultures and runs. Our approach is a simple and efficient strategy to produce large quantities of differentiated cells with the highest homogeneity during multistage pancreatic progenitor differentiation, providing a potential tool for guided differentiation of iPSCs to functional insulin-producing cells.
Collapse
Affiliation(s)
- Mee-Hae Kim
- Department of Biotechnology, Graduate School of Engineering, Osaka University, Suita, Osaka, Japan
| | - Naruchit Thanuthanakhun
- Department of Biotechnology, Graduate School of Engineering, Osaka University, Suita, Osaka, Japan
| | - Masahiro Kino-Oka
- Department of Biotechnology, Graduate School of Engineering, Osaka University, Suita, Osaka, Japan
- Research Base for Cell Manufacturability, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
12
|
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
|
13
|
Abdal Dayem A, Lee SB, Lim KM, Kim A, Shin HJ, Vellingiri B, Kim YB, Cho SG. Bioactive peptides for boosting stem cell culture platform: Methods and applications. Biomed Pharmacother 2023; 160:114376. [PMID: 36764131 DOI: 10.1016/j.biopha.2023.114376] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 02/10/2023] Open
Abstract
Peptides, short protein fragments, can emulate the functions of their full-length native counterparts. Peptides are considered potent recombinant protein alternatives due to their specificity, high stability, low production cost, and ability to be easily tailored and immobilized. Stem cell proliferation and differentiation processes are orchestrated by an intricate interaction between numerous growth factors and proteins and their target receptors and ligands. Various growth factors, functional proteins, and cellular matrix-derived peptides efficiently enhance stem cell adhesion, proliferation, and directed differentiation. For that, peptides can be immobilized on a culture plate or conjugated to scaffolds, such as hydrogels or synthetic matrices. In this review, we assess the applications of a variety of peptides in stem cell adhesion, culture, organoid assembly, proliferation, and differentiation, describing the shortcomings of recombinant proteins and their full-length counterparts. Furthermore, we discuss the challenges of peptide applications in stem cell culture and materials design, as well as provide a brief outlook on future directions to advance peptide applications in boosting stem cell quality and scalability for clinical applications in tissue regeneration.
Collapse
Affiliation(s)
- Ahmed Abdal Dayem
- Department of Stem Cell and Regenerative Biotechnology, KU Convergence Science and Technology Institute, Konkuk University, Seoul 05029, Republic of Korea
| | - Soo Bin Lee
- Department of Stem Cell and Regenerative Biotechnology, KU Convergence Science and Technology Institute, Konkuk University, Seoul 05029, Republic of Korea
| | - Kyung Min Lim
- Department of Stem Cell and Regenerative Biotechnology, KU Convergence Science and Technology Institute, Konkuk University, Seoul 05029, Republic of Korea; R&D Team, StemExOne co., ltd. 303, Life Science Bldg, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Aram Kim
- Department of Urology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul 05029, Republic of Korea; R&D Team, StemExOne co., ltd. 303, Life Science Bldg, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Hyun Jin Shin
- Department of Ophthalmology, Research Institute of Medical Science, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul 05029, Republic of Korea; R&D Team, StemExOne co., ltd. 303, Life Science Bldg, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Balachandar Vellingiri
- Stem cell and Regenerative Medicine/Translational Research, Department of Zoology, School of Basic Sciences, Central University of Punjab (CUPB), Bathinda 151401, Punjab, India
| | - Young Bong Kim
- Department of Biomedical Science & Engineering, KU Convergence Science and Technology Institute, Konkuk University, Seoul 05029, Republic of Korea
| | - Ssang-Goo Cho
- Department of Stem Cell and Regenerative Biotechnology, KU Convergence Science and Technology Institute, Konkuk University, Seoul 05029, Republic of Korea; R&D Team, StemExOne co., ltd. 303, Life Science Bldg, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea.
| |
Collapse
|
14
|
Ragusa D, Dijkhuis L, Pina C, Tosi S. Mechanisms associated with t(7;12) acute myeloid leukaemia: from genetics to potential treatment targets. Biosci Rep 2023; 43:BSR20220489. [PMID: 36622782 PMCID: PMC9894016 DOI: 10.1042/bsr20220489] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 01/04/2023] [Accepted: 01/09/2023] [Indexed: 01/10/2023] Open
Abstract
Acute myeloid leukaemia (AML), typically a disease of elderly adults, affects 8 children per million each year, with the highest paediatric incidence in infants aged 0-2 of 18 per million. Recurrent cytogenetic abnormalities contribute to leukaemia pathogenesis and are an important determinant of leukaemia classification. The t(7;12)(q36;p13) translocation is a high-risk AML subtype exclusively associated with infants and represents the second most common abnormality in this age group. Mechanisms of t(7;12) leukaemogenesis remain poorly understood. The translocation relocates the entire MNX1 gene within the ETV6 locus, but a fusion transcript is present in only half of the patients and its significance is unclear. Instead, research has focused on ectopic MNX1 expression, a defining feature of t(7;12) leukaemia, which has nevertheless failed to produce transformation in conventional disease models. Recently, advances in genome editing technologies have made it possible to recreate the t(7;12) rearrangement at the chromosomal level. Together with recent studies of MNX1 involvement using murine in vivo, in vitro, and organoid-based leukaemia models, specific investigation on the biology of t(7;12) can provide new insights into this AML subtype. In this review, we provide a comprehensive up-to-date analysis of the biological features of t(7;12), and discuss recent advances in mechanistic understanding of the disease which may deliver much-needed therapeutic opportunities to a leukaemia of notoriously poor prognosis.
Collapse
Affiliation(s)
- Denise Ragusa
- College of Health, Medicine and Life Sciences, Division of Biosciences, Brunel University London, Uxbridge, UB8 3PH, U.K
- Centre for Genome Engineering and Maintenance (CenGEM), Brunel University London, Kingston Lane, UB8 3PH, U.K
| | - Liza Dijkhuis
- College of Health, Medicine and Life Sciences, Division of Biosciences, Brunel University London, Uxbridge, UB8 3PH, U.K
| | - Cristina Pina
- College of Health, Medicine and Life Sciences, Division of Biosciences, Brunel University London, Uxbridge, UB8 3PH, U.K
- Centre for Genome Engineering and Maintenance (CenGEM), Brunel University London, Kingston Lane, UB8 3PH, U.K
| | - Sabrina Tosi
- College of Health, Medicine and Life Sciences, Division of Biosciences, Brunel University London, Uxbridge, UB8 3PH, U.K
- Centre for Genome Engineering and Maintenance (CenGEM), Brunel University London, Kingston Lane, UB8 3PH, U.K
| |
Collapse
|
15
|
Arkenberg MR, Koehler K, Lin CC. Heparinized Gelatin-Based Hydrogels for Differentiation of Induced Pluripotent Stem Cells. Biomacromolecules 2022; 23:4141-4152. [PMID: 36074748 PMCID: PMC9554908 DOI: 10.1021/acs.biomac.2c00585] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
![]()
Chemically defined
hydrogels are increasingly utilized to define
the effects of extracellular matrix (ECM) components on cellular fate
determination of human embryonic and induced pluripotent stem cell
(hESC and hiPSCs). In particular, hydrogels cross-linked by orthogonal
click chemistry, including thiol-norbornene photopolymerization and
inverse electron demand Diels–Alder (iEDDA) reactions, are
explored for 3D culture of hESC/hiPSCs owing to the specificity, efficiency,
cytocompatibility, and modularity of the cross-linking reactions.
In this work, we exploited the modularity of thiol-norbornene photopolymerization
to create a biomimetic hydrogel platform for 3D culture and differentiation
of hiPSCs. A cell-adhesive, protease-labile, and cross-linkable gelatin
derivative, gelatin-norbornene (GelNB), was used as the backbone polymer
for constructing hiPSC-laden biomimetic hydrogels. GelNB was further
heparinized via the iEDDA click reaction using tetrazine-modified
heparin (HepTz), creating GelNB-Hep. GelNB or GelNB-Hep was modularly
cross-linked with either inert macromer poly(ethylene glycol)-tetra-thiol
(PEG4SH) or another bioactive macromer-thiolated hyaluronic acid (THA).
The formulations of these hydrogels were modularly tuned to afford
biomimetic matrices with similar elastic moduli but varying bioactive
components, enabling the understanding of each bioactive component
on supporting hiPSC growth and ectodermal, mesodermal, and endodermal
fate commitment under identical soluble differentiation cues.
Collapse
Affiliation(s)
- Matthew R Arkenberg
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Karl Koehler
- Departments of Otolaryngology and Plastic and Oral Surgery, F.M. Kirby Neurobiology Center, Boston Children's Hospital/Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Chien-Chi Lin
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana 47907, United States.,Department of Biomedical Engineering, Purdue School of Engineering & Technology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana 46202, United States
| |
Collapse
|
16
|
Liu Y, Bertels S, Reischl M, Peravali R, Bastmeyer M, Popova AA, Levkin PA. Droplet Microarray Based Screening Identifies Proteins for Maintaining Pluripotency of hiPSCs. Adv Healthc Mater 2022; 11:e2200718. [PMID: 35799451 PMCID: PMC11468593 DOI: 10.1002/adhm.202200718] [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: 03/31/2022] [Revised: 06/10/2022] [Indexed: 01/27/2023]
Abstract
Human induced pluripotent stem cells (hiPSCs) are crucial for disease modeling, drug discovery, and personalized medicine. Animal-derived materials hinderapplications of hiPSCs in medical fields. Thus, novel and well-defined substrate coatings capable of maintaining hiPSC pluripotency are important for advancing biomedical applications of hiPSCs. Here a miniaturized droplet microarray (DMA) platform to investigate 11 well-defined proteins, their 55 binary and 165 ternary combinations for their ability to maintainpluripotency of hiPSCs when applied as a surface coating, is used. Using this screening approach, ten protein group coatings are identified, which promote significantly higher NANOG expression of hiPSCs in comparison with Matrigel coating. With two of the identified coatings, long-term pluripotency maintenance of hiPSCs and subsequent differentiation into three germ layers are achieved. Compared with conventional high-throughput screening (HTS) in 96-well plates, the DMA platform uses only 83 µL of protein solution (0.83 µg total protein) and only ≈2.8 × 105 cells, decreasing the amount of proteins and cells ≈860 and 25-fold, respectively. The identified proteins will be essential for research and applications using hiPSCs, while the DMA platform demonstrates great potential for miniaturized HTS of scarce cells or expensive materials such as recombinant proteins.
Collapse
Affiliation(s)
- Yanxi Liu
- Institute of Biological and Chemical Systems – Functional Molecular SystemsKarlsruhe Institute of TechnologyHermann‐von‐Helmholtz‐Platz 176344Eggenstein‐LeopoldshafenGermany
| | - Sarah Bertels
- Zoological InstituteCell‐ and NeurobiologyKarlsruhe Institute of TechnologyFritz‐Haber‐Weg 476131KarlsruheGermany
| | - Markus Reischl
- Institute for Automation and Applied InformaticsKarlsruhe Institute of TechnologyHermann‐von Helmholtz‐Platz 176344Eggenstein‐LeopoldshafenGermany
| | - Ravindra Peravali
- Institute of Biological and Chemical Systems – Biological Information ProcessingKarlsruhe Institute of TechnologyHermann‐von‐Helmholtz‐Platz 176344Eggenstein‐LeopoldshafenGermany
| | - Martin Bastmeyer
- Zoological InstituteCell‐ and NeurobiologyKarlsruhe Institute of TechnologyFritz‐Haber‐Weg 476131KarlsruheGermany
- Institute of Biological and Chemical Systems – Biological Information ProcessingKarlsruhe Institute of TechnologyHermann‐von‐Helmholtz‐Platz 176344Eggenstein‐LeopoldshafenGermany
| | - Anna A. Popova
- Institute of Biological and Chemical Systems – Functional Molecular SystemsKarlsruhe Institute of TechnologyHermann‐von‐Helmholtz‐Platz 176344Eggenstein‐LeopoldshafenGermany
| | - Pavel A. Levkin
- Institute of Biological and Chemical Systems – Functional Molecular SystemsKarlsruhe Institute of TechnologyHermann‐von‐Helmholtz‐Platz 176344Eggenstein‐LeopoldshafenGermany
- Institute of Organic ChemistryKarlsruhe Institute of TechnologyKaiserstraße 1276131KarlsruheGermany
| |
Collapse
|
17
|
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: 3.7] [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
|
18
|
Wu H, Tang X, Wang Y, Wang N, Chen Q, Xie J, Liu S, Zhong Z, Qiu Y, Situ P, Zern MA, Wang J, Chen H, Duan Y. Dextran sulfate prevents excess aggregation of human pluripotent stem cells in 3D culture by inhibiting ICAM1 expression coupled with down-regulating E-cadherin through activating the Wnt signaling pathway. Stem Cell Res Ther 2022; 13:218. [PMID: 35619172 PMCID: PMC9137216 DOI: 10.1186/s13287-022-02890-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 04/25/2022] [Indexed: 11/29/2022] Open
Abstract
Background Human pluripotent stem cells (hPSCs) have great potential in applications for regenerative medicine and drug development. However, 3D suspension culture systems for clinical-grade hPSC large-scale production have been a major challenge. Accumulating evidence has demonstrated that the addition of dextran sulfate (DS) could prevent excessive adhesion of hPSCs from forming larger aggregates in 3D suspension culture. However, the signaling and molecular mechanisms underlying this phenomenon remain elusive. Methods By using a cell aggregate culture assay and separating big and small aggregates in suspension culture systems, the potential mechanism and downstream target genes of DS were investigated by mRNA sequence analysis, qRT-PCR validation, colony formation assay, and interference assay. Results Since cellular adhesion molecules (CAMs) play important roles in hPSC adhesion and aggregation, we assumed that DS might prevent excess adhesion through affecting the expression of CAMs in hPSCs. As expected, after DS treatment, we found that the expression of CAMs was significantly down-regulated, especially E-cadherin (E-cad) and intercellular adhesion molecule 1 (ICAM1), two highly expressed CAMs in hPSCs. The role of E-cad in the adhesion of hPSCs has been widely investigated, but the function of ICAM1 in hPSCs is hardly understood. In the present study, we demonstrated that ICAM1 exhibited the capacity to promote the adhesion in hPSCs, and this adhesion was suppressed by the treatment with DS. Furthermore, transcriptomic analysis of RNA-seq revealed that DS treatment up-regulated genes related to Wnt signaling resulting in the activation of Wnt signaling in which SLUG, TWIST, and MMP3/7 were highly expressed, and further inhibited the expression of E-cad. Conclusion Our results demonstrated that DS played an important role in controlling the size of hPSC aggregates in 3D suspension culture by inhibiting the expression of ICAM1 coupled with the down-regulation of E-cad through the activation of the Wnt signaling pathway. These results represent a significant step toward developing the expansion of hPSCs under 3D suspension condition in large-scale cultures. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-02890-4.
Collapse
Affiliation(s)
- Haibin Wu
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, No. 382 Waihuan East Road, Suite 406, Higher Education Mega Center, Guangzhou, 510006, People's Republic of China
| | - Xianglian Tang
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, No. 382 Waihuan East Road, Suite 406, Higher Education Mega Center, Guangzhou, 510006, People's Republic of China.,School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 510180, People's Republic of China.,Guangxi Key Laboratory of Reproductive Health and Birth Defects Prevention, Guangxi Health Commission Key Laboratory of Precise Diagnosis and Treatment of Genetic Diseases, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530003, Guangxi, People's Republic of China.,Genetic and Metabolic Central Laboratory, Guangxi Birth Defects Research and Prevention Institute, Nanning, 530003, Guangxi, People's Republic of China
| | - Yiyu Wang
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, No. 382 Waihuan East Road, Suite 406, Higher Education Mega Center, Guangzhou, 510006, People's Republic of China
| | - Ning Wang
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, No. 382 Waihuan East Road, Suite 406, Higher Education Mega Center, Guangzhou, 510006, People's Republic of China.,School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 510180, People's Republic of China
| | - Qicong Chen
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, No. 382 Waihuan East Road, Suite 406, Higher Education Mega Center, Guangzhou, 510006, People's Republic of China.,School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 510180, People's Republic of China
| | - Jinghe Xie
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, No. 382 Waihuan East Road, Suite 406, Higher Education Mega Center, Guangzhou, 510006, People's Republic of China.,School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 510180, People's Republic of China
| | - Shoupei Liu
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, No. 382 Waihuan East Road, Suite 406, Higher Education Mega Center, Guangzhou, 510006, People's Republic of China
| | - Zhiyong Zhong
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, No. 382 Waihuan East Road, Suite 406, Higher Education Mega Center, Guangzhou, 510006, People's Republic of China.,School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 510180, People's Republic of China
| | - Yaqi Qiu
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, No. 382 Waihuan East Road, Suite 406, Higher Education Mega Center, Guangzhou, 510006, People's Republic of China
| | - Ping Situ
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, No. 382 Waihuan East Road, Suite 406, Higher Education Mega Center, Guangzhou, 510006, People's Republic of China.,School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 510180, People's Republic of China
| | - Mark A Zern
- Department of Internal Medicine, University of California Davis Medical Center, Sacramento, CA, 95817, USA
| | - Jue Wang
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, No. 382 Waihuan East Road, Suite 406, Higher Education Mega Center, Guangzhou, 510006, People's Republic of China.
| | - Honglin Chen
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, No. 382 Waihuan East Road, Suite 406, Higher Education Mega Center, Guangzhou, 510006, People's Republic of China. .,National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510180, People's Republic of China. .,Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510180, People's Republic of China. .,Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510180, People's Republic of China.
| | - Yuyou Duan
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, No. 382 Waihuan East Road, Suite 406, Higher Education Mega Center, Guangzhou, 510006, People's Republic of China. .,National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510180, People's Republic of China. .,Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510180, People's Republic of China. .,Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510180, People's Republic of China.
| |
Collapse
|
19
|
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: 2.3] [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
|
20
|
Barhouse PS, Andrade MJ, Smith Q. Home Away From Home: Bioengineering Advancements to Mimic the Developmental and Adult Stem Cell Niche. FRONTIERS IN CHEMICAL ENGINEERING 2022. [DOI: 10.3389/fceng.2022.832754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The inherent self-organizing capacity of pluripotent and adult stem cell populations has advanced our fundamental understanding of processes that drive human development, homeostasis, regeneration, and disease progression. Translating these principles into in vitro model systems has been achieved with the advent of organoid technology, driving innovation to harness patient-specific, cell-laden regenerative constructs that can be engineered to augment or replace diseased tissue. While developmental organization and regenerative adult stem cell niches are tightly regulated in vivo, in vitro analogs lack defined architecture and presentation of physicochemical cues, leading to the unhindered arrangement of mini-tissues that lack complete physiological mimicry. This review aims to highlight the recent integrative engineering approaches that elicit spatio-temporal control of the extracellular niche to direct the structural and functional maturation of pluripotent and adult stem cell derivatives. While the advances presented here leverage multi-pronged strategies ranging from synthetic biology to microfabrication technologies, the methods converge on recreating the biochemical and biophysical milieu of the native tissue to be modeled or regenerated.
Collapse
|
21
|
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: 9] [Impact Index Per Article: 3.0] [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
|
22
|
Chen C, Zhang X, Wang Y, Chen X, Chen W, Dan S, She S, Hu W, Dai J, Hu J, Cao Q, Liu Q, Huang Y, Qin B, Kang B, Wang YJ. Translational and post-translational control of human naïve versus primed pluripotency. iScience 2022; 25:103645. [PMID: 35005567 PMCID: PMC8718978 DOI: 10.1016/j.isci.2021.103645] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 10/22/2021] [Accepted: 12/10/2021] [Indexed: 01/05/2023] Open
Abstract
Deciphering the regulatory network for human naive and primed pluripotency is of fundamental theoretical and applicable significance. Here, by combining quantitative proteomics, phosphoproteomics, and acetylproteomics analyses, we revealed RNA processing and translation as the most differentially regulated processes between naive and primed human embryonic stem cells (hESCs). Although glycolytic primed hESCs rely predominantly on the eukaryotic initiation factor 4E (eIF4E)-mediated cap-dependent pathway for protein translation, naive hESCs with reduced mammalian target of rapamycin complex (mTORC1) activity are more tolerant to eIF4E inhibition, and their bivalent metabolism allows for translating selective mRNAs via both eIF4E-dependent and eIF4E-independent/eIF4A2-dependent pathways to form a more compact naive proteome. Globally up-regulated proteostasis and down-regulated post-translational modifications help to further refine the naive proteome that is compatible with the more rapid cycling of naive hESCs, where CDK1 plays an indispensable coordinative role. These findings may assist in better understanding the unrestricted lineage potential of naive hESCs and in further optimizing conditions for future clinical applications RNA processing and translation are most different between naive and primed hESCs Glycolytic primed hESCs mainly rely on eIF4E-dependent translation Bivalent metabolism in naive hESCs promotes eIF4E-independent translation CDK1 is required for naive pluripotency partially by activating E-cadherin signaling
Collapse
Affiliation(s)
- Cheng Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China.,Shaoxing People's Hospital, Shaoxing Hospital, Zhejiang University School of Medicine, Shaoxing, Zhejiang 312000, China
| | - Xiaobing Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Yisha Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Xinyu Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Wenjie Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Songsong Dan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Shiqi She
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China.,Zhejiang Museum of Natural History, Hangzhou, Zhejiang 310014, China
| | - Weiwei Hu
- Shanghai Bioprofile Technology Co., Ltd., Shanghai 200241, China
| | - Jie Dai
- Shanghai Bioprofile Technology Co., Ltd., Shanghai 200241, China
| | - Jianwen Hu
- Shanghai Bioprofile Technology Co., Ltd., Shanghai 200241, China
| | - Qingyi Cao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Qianyu Liu
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Yinghua Huang
- Laboratory of Metabolism and Cell Fate, Center for Cell Lineage and Development, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Baoming Qin
- Laboratory of Metabolism and Cell Fate, Center for Cell Lineage and Development, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Bo Kang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Ying-Jie Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China.,Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310058, China
| |
Collapse
|
23
|
Romero JJ, De Rossi MC, Oses C, Echegaray CV, Verneri P, Francia M, Guberman A, Levi V. Nucleus-cytoskeleton communication impacts on OCT4-chromatin interactions in embryonic stem cells. BMC Biol 2022; 20:6. [PMID: 34996451 PMCID: PMC8742348 DOI: 10.1186/s12915-021-01207-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 12/06/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The cytoskeleton is a key component of the system responsible for transmitting mechanical cues from the cellular environment to the nucleus, where they trigger downstream responses. This communication is particularly relevant in embryonic stem (ES) cells since forces can regulate cell fate and guide developmental processes. However, little is known regarding cytoskeleton organization in ES cells, and thus, relevant aspects of nuclear-cytoskeletal interactions remain elusive. RESULTS We explored the three-dimensional distribution of the cytoskeleton in live ES cells and show that these filaments affect the shape of the nucleus. Next, we evaluated if cytoskeletal components indirectly modulate the binding of the pluripotency transcription factor OCT4 to chromatin targets. We show that actin depolymerization triggers OCT4 binding to chromatin sites whereas vimentin disruption produces the opposite effect. In contrast to actin, vimentin contributes to the preservation of OCT4-chromatin interactions and, consequently, may have a pro-stemness role. CONCLUSIONS Our results suggest roles of components of the cytoskeleton in shaping the nucleus of ES cells, influencing the interactions of the transcription factor OCT4 with the chromatin and potentially affecting pluripotency and cell fate.
Collapse
Affiliation(s)
- Juan José Romero
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, C1428EGA, Buenos Aires, Argentina
| | - María Cecilia De Rossi
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, C1428EGA, Buenos Aires, Argentina
| | - Camila Oses
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, C1428EGA, Buenos Aires, Argentina
| | - Camila Vázquez Echegaray
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, C1428EGA, Buenos Aires, Argentina
| | - Paula Verneri
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, C1428EGA, Buenos Aires, Argentina
| | - Marcos Francia
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, C1428EGA, Buenos Aires, Argentina
| | - Alejandra Guberman
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, C1428EGA, Buenos Aires, Argentina.
- Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, C1428EGA, Buenos Aires, Argentina.
| | - Valeria Levi
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, C1428EGA, Buenos Aires, Argentina.
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, C1428EGA, Buenos Aires, Argentina.
| |
Collapse
|
24
|
Gao X, Sprando RL, Yourick JJ. Rapid and Highly Efficient Isolation and Purification of Human Induced Pluripotent Stem Cells. Methods Mol Biol 2022; 2429:3-14. [PMID: 35507151 DOI: 10.1007/978-1-0716-1979-7_1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Human induced pluripotent stem cells (iPSCs) hold great promise for biomedical applications. However, establishment of new iPSC lines still presents many challenges. Here we describe a simple yet highly efficient two-step protocol for the isolation and purification of human iPSC lines. The first step adapts iPSCs to single cell culture and passaging, promoting survival and self-renewal; the second step enables the isolation and purification of bona fide iPSCs from a mixed population using column-based positive selection of cells expressing pluripotency markers such as TRA-1-60. Both steps utilize commercially available reagents. Using this protocol, iPSCs can be purified from cell preparations containing differentiated or unreprogrammed cells, or even be isolated directly from reprogramming vessels. The protocol could be adopted for high throughput isolation and expansion of iPSC lines and facilitate the widespread use of iPSCs in future applications.
Collapse
Affiliation(s)
- Xiugong Gao
- Division of Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, MD, USA.
| | - Robert L Sprando
- Division of Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, MD, USA
| | - Jeffrey J Yourick
- Division of Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, MD, USA
| |
Collapse
|
25
|
Gallerani G, Rossi T, Valgiusti M, Angeli D, Fici P, De Fanti S, Bandini E, Cocchi C, Frassineti GL, Bonafè M, Fabbri F. CNA Profiling of Single CTCs in Locally Advanced Esophageal Cancer Patients during Therapy Highlights Unexplored Molecular Pathways. Cancers (Basel) 2021; 13:6369. [PMID: 34944989 PMCID: PMC8699413 DOI: 10.3390/cancers13246369] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/15/2021] [Accepted: 12/16/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Here, we monitored the evolution of CTCs spread in 11 patients affected by locally advanced EC who were undergoing therapy. METHODS In this perspective study, we designed multiple blood biopsies from individual patients: before and after neoadjuvant chemo-radio therapy and after surgery. We developed a multi-target array, named Grab-all assay, to estimate CTCs for their epithelial (EpCAM/E-Cadherin/Cytokeratins) and mesenchymal/stem (N-Cadherin/CD44v6/ABCG2) phenotypes. Identified CTCs were isolated as single cells by DEPArray, subjected to whole genome amplification, and copy number aberration (CNA) profiles were determined. Through bioinformatic analysis, we assessed the genomic imbalance of single CTCs, investigated specific focal copy number changes previously reported in EC and aberrant pathways using enrichment analysis. RESULTS Longitudinal monitoring allowed the identification of CTCs in at least one time-point per patient. Through single cell CNA analysis, we revealed that CTCs showed significantly dynamic genomic imbalance during treatment. Individual CTCs from relapsed patients displayed a higher degree of genomic imbalance relative to disease-free patients' groups. Genomic aberrations previously reported in EC occurred mostly in post-neoadjuvant therapy CTCs. In-depth analysis showed that networks enrichment in all time-point CTCs were inherent to innate immune system. Transcription/gene regulation, post-transcriptional and epigenetic modifications were uniquely affected in CTCs of relapsed patients. CONCLUSIONS Our data add clues to the comprehension of the role of CTCs in EC aggressiveness: chromosomal aberrations on genes related to innate immune system behave as relevant to the onset of CTC-status, whilst pathways of transcription/gene regulation, post-transcriptional and epigenetic modifications seem linked to patients' outcome.
Collapse
Affiliation(s)
- Giulia Gallerani
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (T.R.); (P.F.); (E.B.); (C.C.); (F.F.)
| | - Tania Rossi
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (T.R.); (P.F.); (E.B.); (C.C.); (F.F.)
| | - Martina Valgiusti
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (M.V.); (G.L.F.)
| | - Davide Angeli
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy;
| | - Pietro Fici
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (T.R.); (P.F.); (E.B.); (C.C.); (F.F.)
| | - Sara De Fanti
- Interdepartmental Centre “Alma Mater Research Institute on Global Challenges and Climate Change (Alma Climate)”, University of Bologna, 40126 Bologna, Italy;
| | - Erika Bandini
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (T.R.); (P.F.); (E.B.); (C.C.); (F.F.)
| | - Claudia Cocchi
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (T.R.); (P.F.); (E.B.); (C.C.); (F.F.)
| | - Giovanni Luca Frassineti
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (M.V.); (G.L.F.)
| | - Massimiliano Bonafè
- Department of Experimental and Diagnostic Medicine, University of Bologna, 40126 Bologna, Italy;
| | - Francesco Fabbri
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (T.R.); (P.F.); (E.B.); (C.C.); (F.F.)
| |
Collapse
|
26
|
Muscolino E, Di Stefano AB, Trapani M, Sabatino MA, Giacomazza D, Moschella F, Cordova A, Toia F, Dispenza C. Injectable xyloglucan hydrogels incorporating spheroids of adipose stem cells for bone and cartilage regeneration. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 131:112545. [PMID: 34857257 DOI: 10.1016/j.msec.2021.112545] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 10/31/2021] [Accepted: 11/07/2021] [Indexed: 12/12/2022]
Abstract
Cartilage or bone regeneration approaches based on the direct injection of mesenchymal stem cells (MSCs) at the lesion site encounter several challenges, related to uncontrolled cell spreading and differentiation, reduced cell viability and poor engrafting. This work presents a simple and versatile strategy based on the synergic combination of in-situ forming hydrogels and spheroids of adipose stem cells (SASCs) with great potential for minimally invasive regenerative interventions aimed to threat bone and cartilage defects. Aqueous dispersions of partially degalactosylated xyloglucan (dXG) are mixed with SASCs derived from liposuction and either a chondroinductive or an osteoinductive medium. The dispersions rapidly set into hydrogels when temperature is brought to 37 °C. The physico-chemical and mechanical properties of the hydrogels are controlled by polymer concentration. The hydrogels, during 21 day incubation at 37 °C, undergo significant structural rearrangements that support cell proliferation and spreading. In formulations containing 1%w dXG cell viability increases up to 300% for SASCs-derived osteoblasts and up to 1000% for SASCs-derived chondrocytes if compared with control 2D cultures. The successful differentiation into the target cells is supported by the expression of lineage-specific genes. Cell-cell and cell-matrix interactions are also investigated. All formulations resulted injectable, and the incorporated cells are fully viable after injection.
Collapse
Affiliation(s)
- Emanuela Muscolino
- Dipartimento di Ingegneria, Università degli Studi di Palermo, Viale delle Scienze 6, 90128 Palermo, Italy
| | - Anna Barbara Di Stefano
- BIOPLAST-Laboratory of BIOlogy and Regenerative Medicine-PLASTic Surgery, Dipartimento di Discipline Chirurgiche, Oncologiche e Stomatologiche, Università degli Studi di Palermo, via del Vespro 129, 90127 Palermo, Italy
| | - Marco Trapani
- BIOPLAST-Laboratory of BIOlogy and Regenerative Medicine-PLASTic Surgery, Dipartimento di Discipline Chirurgiche, Oncologiche e Stomatologiche, Università degli Studi di Palermo, via del Vespro 129, 90127 Palermo, Italy
| | - Maria Antonietta Sabatino
- Dipartimento di Ingegneria, Università degli Studi di Palermo, Viale delle Scienze 6, 90128 Palermo, Italy
| | - Daniela Giacomazza
- Istituto di Biofisica, Consiglio Nazionale delle Ricerche, Via U. La Malfa 153, 90146, Palermo, Italy
| | - Francesco Moschella
- BIOPLAST-Laboratory of BIOlogy and Regenerative Medicine-PLASTic Surgery, Dipartimento di Discipline Chirurgiche, Oncologiche e Stomatologiche, Università degli Studi di Palermo, via del Vespro 129, 90127 Palermo, Italy; Dipartimento di Discipline Chirurgiche, Oncologiche e Stomatologiche, Università degli Studi di Palermo, via del Vespro 129, 90127 Palermo, Italy
| | - Adriana Cordova
- BIOPLAST-Laboratory of BIOlogy and Regenerative Medicine-PLASTic Surgery, Dipartimento di Discipline Chirurgiche, Oncologiche e Stomatologiche, Università degli Studi di Palermo, via del Vespro 129, 90127 Palermo, Italy; Dipartimento di Discipline Chirurgiche, Oncologiche e Stomatologiche, Università degli Studi di Palermo, via del Vespro 129, 90127 Palermo, Italy
| | - Francesca Toia
- Dipartimento di Discipline Chirurgiche, Oncologiche e Stomatologiche, Università degli Studi di Palermo, via del Vespro 129, 90127 Palermo, Italy
| | - Clelia Dispenza
- Dipartimento di Ingegneria, Università degli Studi di Palermo, Viale delle Scienze 6, 90128 Palermo, Italy; Istituto di Biofisica, Consiglio Nazionale delle Ricerche, Via U. La Malfa 153, 90146, Palermo, Italy.
| |
Collapse
|
27
|
Tadokoro S, Tokuyama-Toda R, Tatehara S, Ide S, Umeki H, Miyoshi K, Noma T, Satomura K. A New Induction Method for the Controlled Differentiation of Human-Induced Pluripotent Stem Cells Using Frozen Sections. Cells 2021; 10:2827. [PMID: 34831050 PMCID: PMC8616214 DOI: 10.3390/cells10112827] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/15/2021] [Accepted: 10/19/2021] [Indexed: 02/04/2023] Open
Abstract
Considering that every tissue/organ has the most suitable microenvironment for its functional cells, controlling induced pluripotent stem cell (iPSC) differentiation by culture on frozen sections having a suitable microenvironment is possible. Induced PSCs were cultured on frozen sections of the liver, the brain, the spinal cord, and cover glasses (control) for 9 days. The iPSCs cultured on the sections of the liver resembled hepatocytes, whereas those on sections of the brain and the spinal cord resembled neuronal cells. The percentage of hepatocytic marker-positive cells in the iPSCs cultured on the sections of the liver was statistically higher than that of those in the iPSCs cultured on the sections of the brain and the spinal cord or on cover glasses. In contrast, the iPSCs cultured on the sections of the brain and the spinal cord revealed a high percentage of neural marker-positive cells. Thus, iPSCs can be differentiated into a specific cell lineage in response to specific factors within frozen sections of tissues/organs. Differentiation efficacy of the frozen sections markedly differed between the iPSC clones. Therefore, our induction method could be simple and effective for evaluating the iPSC quality.
Collapse
Affiliation(s)
- Susumu Tadokoro
- Department of Oral Medicine and Stomatology, Tsurumi University School of Dental Medicine, 2-1-3, Tsurumi, Tsurumi-ku, Yokohama 230-8501, Japan; (S.T.); (R.T.-T.); (S.T.); (S.I.); (H.U.)
| | - Reiko Tokuyama-Toda
- Department of Oral Medicine and Stomatology, Tsurumi University School of Dental Medicine, 2-1-3, Tsurumi, Tsurumi-ku, Yokohama 230-8501, Japan; (S.T.); (R.T.-T.); (S.T.); (S.I.); (H.U.)
| | - Seiko Tatehara
- Department of Oral Medicine and Stomatology, Tsurumi University School of Dental Medicine, 2-1-3, Tsurumi, Tsurumi-ku, Yokohama 230-8501, Japan; (S.T.); (R.T.-T.); (S.T.); (S.I.); (H.U.)
| | - Shinji Ide
- Department of Oral Medicine and Stomatology, Tsurumi University School of Dental Medicine, 2-1-3, Tsurumi, Tsurumi-ku, Yokohama 230-8501, Japan; (S.T.); (R.T.-T.); (S.T.); (S.I.); (H.U.)
| | - Hirochika Umeki
- Department of Oral Medicine and Stomatology, Tsurumi University School of Dental Medicine, 2-1-3, Tsurumi, Tsurumi-ku, Yokohama 230-8501, Japan; (S.T.); (R.T.-T.); (S.T.); (S.I.); (H.U.)
| | - Keiko Miyoshi
- Department of Oral Bioscience, Tokushima University Graduate School of Biomedical Sciences, 3-18-15, Kuramoto, Tokushima 770-8504, Japan;
| | - Takafumi Noma
- Department of Nutrition and Health Promotion, Hiroshima Jogakuin University, 4-13-1, Ushitahigashi, Higashiku, Hiroshima 732-0063, Japan;
| | - Kazuhito Satomura
- Department of Oral Medicine and Stomatology, Tsurumi University School of Dental Medicine, 2-1-3, Tsurumi, Tsurumi-ku, Yokohama 230-8501, Japan; (S.T.); (R.T.-T.); (S.T.); (S.I.); (H.U.)
| |
Collapse
|
28
|
Allen TA, Cullen MM, Hawkey N, Mochizuki H, Nguyen L, Schechter E, Borst L, Yoder JA, Freedman JA, Patierno SR, Cheng K, Eward WC, Somarelli JA. A Zebrafish Model of Metastatic Colonization Pinpoints Cellular Mechanisms of Circulating Tumor Cell Extravasation. Front Oncol 2021; 11:641187. [PMID: 34631514 PMCID: PMC8495265 DOI: 10.3389/fonc.2021.641187] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 08/31/2021] [Indexed: 01/18/2023] Open
Abstract
Metastasis is a multistep process in which cells must detach, migrate/invade local structures, intravasate, circulate, extravasate, and colonize. A full understanding of the complexity of this process has been limited by the lack of ability to study these steps in isolation with detailed molecular analyses. Leveraging a comparative oncology approach, we injected canine osteosarcoma cells into the circulation of transgenic zebrafish with fluorescent blood vessels in a biologically dynamic metastasis extravasation model. Circulating tumor cell clusters that successfully extravasated the vasculature as multicellular units were isolated under intravital imaging (n = 6). These extravasation-positive tumor cell clusters sublines were then molecularly profiled by RNA-Seq. Using a systems-level analysis, we pinpointed the downregulation of KRAS signaling, immune pathways, and extracellular matrix (ECM) organization as enriched in extravasated cells (p < 0.05). Within the extracellular matrix remodeling pathway, we identified versican (VCAN) as consistently upregulated and central to the ECM gene regulatory network (p < 0.05). Versican expression is prognostic for a poorer metastasis-free and overall survival in patients with osteosarcoma. Together, our results provide a novel experimental framework to study discrete steps in the metastatic process. Using this system, we identify the versican/ECM network dysregulation as a potential contributor to osteosarcoma circulating tumor cell metastasis.
Collapse
Affiliation(s)
- Tyler A Allen
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, United States
| | - Mark M Cullen
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, United States
| | - Nathan Hawkey
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, United States
| | - Hiroyuki Mochizuki
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States
| | - Lan Nguyen
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, United States
| | - Elyse Schechter
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, United States
| | - Luke Borst
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Jeffrey A Yoder
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States
| | - Jennifer A Freedman
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, United States.,Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, NC, United States
| | - Steven R Patierno
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, United States.,Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, NC, United States
| | - Ke Cheng
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States.,Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC, United States
| | - William C Eward
- Department of Orthopedics, Duke University Medical Center, Durham, NC, United States
| | - Jason A Somarelli
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, United States.,Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
29
|
Kim MH, Thanuthanakhun N, Fujimoto S, Kino-Oka M. Effect of initial seeding density on cell behavior-driven epigenetic memory and preferential lineage differentiation of human iPSCs. Stem Cell Res 2021; 56:102534. [PMID: 34530397 DOI: 10.1016/j.scr.2021.102534] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 08/22/2021] [Accepted: 09/03/2021] [Indexed: 10/20/2022] Open
Abstract
Understanding the cellular behavioral mechanisms underlying memory formation and maintenance in human induced pluripotent stem cell (hiPSC) culture provides key strategies for achieving stability and robustness of cell differentiation. Here, we show that changes in cell behavior-driven epigenetic memory of hiPSC cultures alter their pluripotent state and subsequent differentiation. Interestingly, pluripotency-associated genes were activated during the entire cell growth phases along with increased active modifications and decreased repressive modifications. This memory effect can last several days in the long-term stationary phase and was sustained in the aspect of cell behavioral changes after subculture. Further, changes in growth-related cell behavior were found to induce nucleoskeletal reorganization and active versus repressive modifications, thereby enabling hiPSCs to change their differentiation potential. Overall, we discuss the cell behavior-driven epigenetic memory induced by the culture environment, and the effect of previous memory on cell lineage specification in the process of hiPSC differentiation.
Collapse
Affiliation(s)
- Mee-Hae Kim
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | - Naruchit Thanuthanakhun
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Shun Fujimoto
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Masahiro Kino-Oka
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
30
|
Chang PC, Lin ZJ, Luo HT, Tu CC, Tai WC, Chang CH, Chang YC. Degradable RGD-Functionalized 3D-Printed Scaffold Promotes Osteogenesis. J Dent Res 2021; 100:1109-1117. [PMID: 34334009 DOI: 10.1177/00220345211024634] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
To establish an ideal microenvironment for regenerating maxillofacial defects, recent research interests have concentrated on developing scaffolds with intricate configurations and manipulating the stiffness of extracellular matrix toward osteogenesis. Herein, we propose to infuse a degradable RGD-functionalized alginate matrix (RAM) with osteoid-like stiffness, as an artificial extracellular matrix, to a rigid 3D-printed hydroxyapatite scaffold for maxillofacial regeneration. The 3D-printed hydroxyapatite scaffold was produced by microextrusion technology and showed good dimensional stability with consistent microporous detail. RAM was crosslinked by calcium sulfate to manipulate the stiffness, and its degradation was accelerated by partial oxidation using sodium periodate. The results revealed that viability of bone marrow stem cells was significantly improved on the RAM and was promoted on the oxidized RAM. In addition, the migration and osteogenic differentiation of bone marrow stem cells were promoted on the RAM with osteoid-like stiffness, specifically on the oxidized RAM. The in vivo evidence revealed that nonoxidized RAM with osteoid-like stiffness upregulated osteogenic genes but prevented ingrowth of newly formed bone, leading to limited regeneration. Oxidized RAM with osteoid-like stiffness facilitated collagen synthesis, angiogenesis, and osteogenesis and induced robust bone formation, thereby significantly promoting maxillofacial regeneration. Overall, this study supported that in the stabilized microenvironment, oxidized RAM with osteoid-like stiffness offered requisite mechanical cues for osteogenesis and an appropriate degradation profile to facilitate bone formation. Combining the 3D-printed hydroxyapatite scaffold and oxidized RAM with osteoid-like stiffness may be an advantageous approach for maxillofacial regeneration.
Collapse
Affiliation(s)
- P-C Chang
- Graduate Institute of Oral Biology, School of Dentistry, National Taiwan University, Taipei.,Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei.,Division of Periodontics, Department of Dentistry, National Taiwan University Hospital, Taipei.,School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung
| | - Z-J Lin
- Graduate Institute of Oral Biology, School of Dentistry, National Taiwan University, Taipei
| | - H-T Luo
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei.,Division of Periodontics, Department of Dentistry, National Taiwan University Hospital, Taipei
| | - C-C Tu
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei.,Division of Periodontics, Department of Dentistry, National Taiwan University Hospital, Taipei
| | - W-C Tai
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei
| | - C-H Chang
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei
| | - Y-C Chang
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei
| |
Collapse
|
31
|
Zhang A, Aslam H, Sharma N, Warmflash A, Fakhouri WD. Conservation of Epithelial-to-Mesenchymal Transition Process in Neural Crest Cells and Metastatic Cancer. Cells Tissues Organs 2021; 210:151-172. [PMID: 34218225 DOI: 10.1159/000516466] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 04/12/2021] [Indexed: 11/19/2022] Open
Abstract
Epithelial to mesenchymal transition (EMT) is a highly conserved cellular process in several species, from worms to humans. EMT plays a fundamental role in early embryogenesis, wound healing, and cancer metastasis. For neural crest cell (NCC) development, EMT typically results in forming a migratory and potent cell population that generates a wide variety of cell and tissue, including cartilage, bone, connective tissue, endocrine cells, neurons, and glia amongst many others. The degree of conservation between the signaling pathways that regulate EMT during development and metastatic cancer (MC) has not been fully established, despite ample studies. This systematic review and meta-analysis dissects the major signaling pathways involved in EMT of NCC development and MC to unravel the similarities and differences. While the FGF, TGFβ/BMP, SHH, and NOTCH pathways have been rigorously investigated in both systems, the EGF, IGF, HIPPO, Factor Receptor Superfamily, and their intracellular signaling cascades need to be the focus of future NCC studies. In general, meta-analyses of the associated signaling pathways show a significant number of overlapping genes (particularly ligands, transcription regulators, and targeted cadherins) involved in each signaling pathway of both systems without stratification by body segments and cancer type. Lack of stratification makes it difficult to meaningfully evaluate the intracellular downstream effectors of each signaling pathway. Finally, pediatric neuroblastoma and melanoma are NCC-derived malignancies, which emphasize the importance of uncovering the EMT events that convert NCC into treatment-resistant malignant cells.
Collapse
Affiliation(s)
- April Zhang
- Center for Craniofacial Research, Department of Diagnostic and Biomedical Sciences, School of Dentistry, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Hira Aslam
- Center for Craniofacial Research, Department of Diagnostic and Biomedical Sciences, School of Dentistry, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Neha Sharma
- Center for Craniofacial Research, Department of Diagnostic and Biomedical Sciences, School of Dentistry, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Aryeh Warmflash
- Department of Biosciences, Rice University, Houston, Texas, USA
| | - Walid D Fakhouri
- Center for Craniofacial Research, Department of Diagnostic and Biomedical Sciences, School of Dentistry, University of Texas Health Science Center at Houston, Houston, Texas, USA.,Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center, Houston, Texas, USA
| |
Collapse
|
32
|
Marca JEL, Somers WG. The Drosophila gonads: models for stem cell proliferation, self-renewal, and differentiation. AIMS GENETICS 2021. [DOI: 10.3934/genet.2014.1.55] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
AbstractThe male and female gonads of Drosophila melanogaster have developed into powerful model systems for both the study of stem cell behaviours, and for understanding how stem cell misregulation can lead to cancers. Using these systems, one is able to observe and manipulate the resident stem cell populations in vivo with a great deal of licence. The tractability of the testis and ovary also allow researchers to explore a range of cellular mechanisms, such as proliferation and polarity, as well as the influence exerted by the local environment through a host of highly-conserved signalling pathways. Importantly, many of the cellular behaviours and processes studied in the Drosophila testis and ovary are known to be disrupted, or otherwise misregulated, in human tumourigenic cells. Here, we review the mechanisms relating to stem cell behaviour, though we acknowledge there are many other fascinating aspects of gametogenesis, including the invasive behaviour of migratory border cells in the Drosophila ovary that, though relevant to the study of tumourigenesis, will unfortunately not be covered.
Collapse
Affiliation(s)
- John E. La Marca
- Department of Genetics, La Trobe University, Melbourne, VIC 3086, Australia
| | | |
Collapse
|
33
|
Xie J, Li X, Zhang Y, Tang T, Chen G, Mao H, Gu Z, Yang J. VE-cadherin-based matrix promoting the self-reconstruction of pro-vascularization microenvironments and endothelial differentiation of human mesenchymal stem cells. J Mater Chem B 2021; 9:3357-3370. [PMID: 33881442 DOI: 10.1039/d1tb00017a] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Regulating the secretion and endothelial differentiation of human mesenchymal stem cells (hMSCs) plays an important role in the vascularization in tissue engineering and regenerative medicine. In this study, a recombinant cadherin fusion protein consisting of a human vascular endothelial-cadherin extracellular domain and immunoglobulin IgG Fc region (hVE-cad-Fc) was developed as a bioartificial matrix for modulating hMSCs. The hVE-cad-Fc matrix significantly enhanced the secretion of angiogenic factors, activated the VE-cadherin-VEGFR2/FAK-AKT/PI3K signaling pathway in hMSCs, and promoted the endothelial differentiation of hMSCs even without extra VEGF. Furthermore, the hVE-cad-Fc matrix was applied for the surface modification of a poly (lactic-co-glycolic acid) (PLGA) porous scaffold, which significantly improved the hemocompatibility and vascularization of the PLGA scaffold in vivo. These results revealed that the hVE-cad-Fc matrix should be a superior bioartificial ECM for remodeling the pro-vascularization extracellular microenvironment by regulating the secretion of hMSCs, and showed great potential for the vascularization in tissue engineering.
Collapse
Affiliation(s)
- Jinghui Xie
- The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin, 300071, China.
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Chowdhury S, Ghosh S. Sialylation of Stem Cells. Stem Cells 2021. [DOI: 10.1007/978-981-16-1638-9_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
35
|
Das R, Fernandez JG. Cellulose Nanofibers for Encapsulation and Pluripotency Preservation in the Early Development of Embryonic Stem Cells. Biomacromolecules 2020; 21:4814-4822. [PMID: 32931265 DOI: 10.1021/acs.biomac.0c01030] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Materials for three-dimensional cultures aim to reproduce the function of the extracellular matrix, enabling cell adhesion and growth by remodeling the environment. However, embryonic stem cells (ESCs) must develop in environments that prevent adhesion and preserve their pluripotency. In this study, we used cellulose nanofiber hydrogels to mimic the developing conditions required for ESCs. These plant-based hydrogels are simultaneously biocompatible and exogenous to mammalian cells, preventing remodeling and attachment. The storage modulus of these hydrogels could be fine-tuned by varying the degree of oxidation to enable selective degradation. The ESCs proliferated in the artificial environment, forming increasingly large embryoid bodies for 15 days. Unlike traditional cultures in which ESCs begin differentiating upon the removal of the chemical inhibition, the expression of pluripotency markers in the ESC population remained high for the entire two weeks. Cellulase from Trichoderma reesei was used to retrieve the ESC cultures selectively. The proposed unique system is a prospective model with which to study the early development of embryonic cells, as well as a nonchemical method of preserving undifferentiated populations of ESCs.
Collapse
Affiliation(s)
- Rupambika Das
- Singapore University of Technology & Design, 8 Somapah Road, Singapore 487372, Singapore
| | - Javier G Fernandez
- Singapore University of Technology & Design, 8 Somapah Road, Singapore 487372, Singapore
| |
Collapse
|
36
|
Park J, Choe G, Oh S, Lee JY. In Situ Formation of Proangiogenic Mesenchymal Stem Cell Spheroids in Hyaluronic Acid/Alginate Core-Shell Microcapsules. ACS Biomater Sci Eng 2020; 6:6938-6948. [PMID: 33320608 DOI: 10.1021/acsbiomaterials.0c01489] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mesenchymal stem-cell (MSC)-based therapies have been recognized as promising strategies for the treatment of various injuries or diseases because of their unique characteristics, such as self-renewal, differentiation potential, and secretion of various bioactive molecules. However, MSC transplantation often results in low efficacy, including a cell viability loss and a low therapeutic activity. Alternatively, MSC spheroids have been studied to improve the viability and therapeutic activity of MSCs. Also, microencapsulation of cells can protect and retain the cells from harsh environments after transplantation. Here, MSC spheroids were formed in hyaluronic acid/alginate (HA@Alg) core-shell microcapsules and employed for neovascularization. A well-defined core-shell structure of HA@Alg microcapsules was produced by optimizing various electrospraying conditions. MSC spheroids could be spontaneously formed in the HA core of the microcapsules after 1 day of incubation. Enhanced secretion of various growth factors was found from MSC spheroids in HA@Alg. In vivo plug assay revealed the significant promotion of angiogenesis by MSC spheroids in HA@Alg compared to that by the controls (i.e., MSCs and MSC spheroids), which is likely because of the better retention of MSC spheroid forms in the microcapsules. Thus, the HA@Alg microcapsules embedding MSC spheroids will be greatly beneficial for various stem cell-based therapies.
Collapse
Affiliation(s)
- Junha Park
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Goeun Choe
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Seulgi Oh
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Jae Young Lee
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| |
Collapse
|
37
|
Richardson T, Wiegand C, Adisa F, Ravikumar K, Candiello J, Kumta P, Banerjee I. Engineered peptide modified hydrogel platform for propagation of human pluripotent stem cells. Acta Biomater 2020; 113:228-239. [PMID: 32603868 DOI: 10.1016/j.actbio.2020.06.034] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 06/19/2020] [Accepted: 06/23/2020] [Indexed: 12/24/2022]
Abstract
Human pluripotent stem cells (hPSCs) have enormous potential to alleviate cell needs for regenerative medicine, however these cells require expansion in cell colonies to maintain cell-cell contact, thus limiting the scalability needed to meet the demands of cell therapy. While the use of a Rho-associated protein kinase (ROCK) inhibitor will allow for culture of single cell hPSCs, typically only 50% of cells are recovered after dissociation. When hPSCs lose cell-cell contact through E-cadherin, dissociation induced apoptosis occurs. In this study, we hypothesized that the extracellular E-cadherin domain of hPSCs will bind to synthetic E-cadherin peptides presented on a hydrogel substrate, mimicking the required cell-cell contact and thereby retaining single-cell viability and clonogenicity. Hence, the objective of this study was to functionalize alginate hydrogels with synthetic peptides mimicking E-cadherin and evaluate peptide performance in promoting cell attachment, viability, maintaining pluripotency, and differentiation potential. We observed that alginate conjugated with synthetic E-cadherin peptides not only supported initial cell attachment with high viability, but also supported hPSC propagation and high fold expansion. hPSCs propagated on the peptide modified substrates maintained the hPSC characteristic pluripotency and differentiation potential, characterized by both spontaneous and directed differentiation. STATEMENT OF SIGNIFICANCE: Human pluripotent stem cells (hPSCs) have enormous potential to alleviate cell needs for regenerative medicine and cell therapy. However, scalable culture of hPSCs is challenged by its need for maintenance of cell-cell contact, dissociation of which triggers the apoptotic pathway. Hence hPSCs are commonly maintained as colonies over Matrigel coated culture plates. Furthermore, use of xenogenic and undefined Matrigel compromises the translational potential of hPSCs. In this work we have developed a completely defined substrate to enable adherent culture of hPSCs as single cells. This substrate prevents apoptosis of the single cells and allows significant fold expansion of hPSCs while maintaining pluripotency and differentiation potential. The developed substrate is expected to be a cost-effective and translatable alternative to Matrigel.
Collapse
Affiliation(s)
- Thomas Richardson
- Department of Chemical and Petroleum Engineering, University of Pittsburgh, United States
| | - Connor Wiegand
- Department of Chemical and Petroleum Engineering, University of Pittsburgh, United States
| | - Fatimah Adisa
- Department of Chemical and Petroleum Engineering, University of Pittsburgh, United States
| | - K Ravikumar
- Department of Chemical and Petroleum Engineering, University of Pittsburgh, United States
| | - Joe Candiello
- Department of Chemical and Petroleum Engineering, University of Pittsburgh, United States
| | - Prashant Kumta
- Department of Chemical and Petroleum Engineering, University of Pittsburgh, United States; Department of Bioengineering, University of Pittsburgh, United States; McGowan Institute for Regenerative Medicine, United States
| | - Ipsita Banerjee
- Department of Chemical and Petroleum Engineering, University of Pittsburgh, United States; Department of Bioengineering, University of Pittsburgh, United States; McGowan Institute for Regenerative Medicine, United States.
| |
Collapse
|
38
|
Zhao X, Wang Y, He J, Deng R, Huang X, Guo Y, Li L, Xie R, Yu J. LncRNA UCA1 maintains the low-tumorigenic and nonmetastatic status by stabilizing E-cadherin in primary prostate cancer cells. Mol Carcinog 2020; 59:1174-1187. [PMID: 32805084 DOI: 10.1002/mc.23247] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 07/28/2020] [Accepted: 08/05/2020] [Indexed: 12/12/2022]
Abstract
Long noncoding RNAs (LncRNAs) have emerged as important players in cancer biology. Increasing evidence suggests that LncRNAs are frequently dysregulated in cancer and may function as oncogenes or tumor suppressors. Urothelial carcinoma associated 1 (UCA1), a LncRNA, firstly identified in bladder transitional cell carcinoma, seems to act as an oncogene in many different types of human cancers by promoting cell proliferation and migration. In this study, we revealed a novel biological function of UCA1, which was different from that reported by previous studies, was responsible for maintaining the low-tumorigenic, nonmetastatic phenotypes in primary prostate epithelial cells. UCA1 could stabilize E-cadherin protein by preventing the interaction between E-cadherin and its E3 ligase MDM2, which suppressed MDM2-mediated ubiquitination and degradation of E-cadherin. In addition, we also found that UCA1 acted as a sponge of miR-296-3p, which targeted E-cadherin gene CDH1 messenger RNA at the posttranscription level. Taken together, these findings demonstrated that UCA1 had a new important role in effectively keeping E-cadherin at a high level through a dual mechanism, which maintained primary prostate cancer cells at the low-tumorigenic and nonmetastatic status.
Collapse
Affiliation(s)
- Xian Zhao
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanli Wang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianfeng He
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rong Deng
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaojun Huang
- Faculty of Health of Sciences, Institute of Translational Medicine, University of Macau, Macau SAR, China
| | - Yanmin Guo
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lian Li
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruiyu Xie
- Faculty of Health of Sciences, Institute of Translational Medicine, University of Macau, Macau SAR, China
| | - Jianxiu Yu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Basic Clinical Research Center, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
39
|
Nowak-Imialek M, Wunderlich S, Herrmann D, Breitschuh-Leibling S, Gohring G, Petersen B, Klein S, Baulain U, Lucas-Hahn A, Martin U, Niemann H. In Vitro and In Vivo Interspecies Chimera Assay Using Early Pig Embryos. Cell Reprogram 2020; 22:118-133. [PMID: 32429746 DOI: 10.1089/cell.2019.0107] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Chimeric pigs harboring organs derived from human stem cells are promising for patient-specific regenerative therapies. Induced pluripotent stem cells (iPSCs) can contribute to all cell types of the fetus, including germline after injection into embryos. However, ethical concerns prohibit testing human iPSCs in chimera assays. Here, we evaluated porcine embryos as hosts for an interspecies chimera assay using iPSCs from either cynomolgus monkeys (cyiPSCs) or mouse (miPSCs). To establish an in vitro culture system compatible for cyiPSCs and porcine embryos, we determined blastocyst development in eight different stem cell media. The highest developmental rates of blastocysts were achieved in Knockout Dulbecco's modified Eagle's medium with 20% knockout serum replacement. We found that cyiPSCs injected into porcine embryos survived in vitro and were mostly located in the trophectoderm (TE). Instead, when miPSCs were injected into porcine embryos, the cells rapidly proliferated. The behavior of chimeras developed in vitro was recapitulated in vivo; cyiPSCs were observed in the TE, but not in the porcine epiblast. However, when miPSCs were injected into in vivo derived porcine embryos, mouse cells were found in both, the epiblast and TE. These results demonstrate that porcine embryos could be useful for evaluating the interspecies chimera-forming ability of iPSCs from different species.
Collapse
Affiliation(s)
- Monika Nowak-Imialek
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, Neustadt, Germany.,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Stephanie Wunderlich
- Leibniz Research Laboratories for Biotechnology and Artificial Organs-LEBAO, Hannover Medical School, Hannover, Germany
| | - Doris Herrmann
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, Neustadt, Germany
| | | | - Gudrun Gohring
- Department of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Björn Petersen
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, Neustadt, Germany
| | - Sabine Klein
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, Neustadt, Germany
| | - Ulrich Baulain
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, Neustadt, Germany
| | - Andrea Lucas-Hahn
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, Neustadt, Germany
| | - Ulrich Martin
- REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany.,Leibniz Research Laboratories for Biotechnology and Artificial Organs-LEBAO, Hannover Medical School, Hannover, Germany
| | - Heiner Niemann
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, Neustadt, Germany.,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| |
Collapse
|
40
|
Bandyopadhayaya S, Ford B, Mandal CC. Cold-hearted: A case for cold stress in cancer risk. J Therm Biol 2020; 91:102608. [PMID: 32716858 DOI: 10.1016/j.jtherbio.2020.102608] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 04/25/2020] [Accepted: 04/25/2020] [Indexed: 02/07/2023]
Abstract
A negative correlation exists between environmental temperature and cancer risk based on both epidemiological and statistical analyses. Previously, cold stress was reported to be an effective cause of tumorigenesis. Several studies have demonstrated that cold temperature serves as a potential risk factor in cancer development. Most recently, a link was demonstrated between the effects of extreme cold climate on cancer incidence, pinpointing its impact on tumour suppressor genes by causing mutation. The underlying mechanism behind cold stress and its association with tumorigenesis is not well understood. Hence, this review intends to shed light on the role of associated factors, genetic and/or non-genetic, which are modulated by cold temperature, and eventually influence tumorigenic potential. While scrutinizing the effect of cold exposure on the body, the expression of certain genes, e.g. uncoupled proteins and heat-shock proteins, were elevated. Biological chemicals such as norepinephrine, thyroxine, and cholesterol were also elevated. Brown adipose tissue, which plays an essential role in thermogenesis, displayed enhanced activity upon cold exposure. Adaptive measures are utilized by the body to tolerate the cold, and in doing so, invites both epigenetic and genetic changes. Unknowingly, these adaptive strategies give rise to a lethal outcome i.e., genesis of cancer. Concisely, this review attempts to draw a link between cold stress, genetic and epigenetic changes, and tumorigenesis and aspires to ascertain the mechanism behind cold temperature-mediated cancer risk.
Collapse
Affiliation(s)
| | - Bridget Ford
- Department of Biology, University of the Incarnate Word, San Antonio, TX, 78209, USA
| | - Chandi C Mandal
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, 305817, India.
| |
Collapse
|
41
|
Scalable Generation of Mesenchymal Stem Cells and Adipocytes from Human Pluripotent Stem Cells. Cells 2020; 9:cells9030710. [PMID: 32183164 PMCID: PMC7140720 DOI: 10.3390/cells9030710] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 03/04/2020] [Accepted: 03/09/2020] [Indexed: 02/06/2023] Open
Abstract
Human pluripotent stem cells (hPSCs) can provide unlimited supply for mesenchymal stem cells (MSCs) and adipocytes that can be used for therapeutic applications. Here we developed a simple and highly efficient all-trans-retinoic acid (RA)-based method for generating an off-the-shelf and scalable number of human pluripotent stem cell (hPSC)-derived MSCs with enhanced adipogenic potential. We showed that short exposure of multiple hPSC lines (hESCs/hiPSCs) to 10 μM RA dramatically enhances embryoid body (EB) formation through regulation of genes activating signaling pathways associated with cell proliferation, survival and adhesion, among others. Disruption of cell adhesion induced the subsequent differentiation of the highly expanded RA-derived EB-forming cells into a pure population of multipotent MSCs (up to 1542-fold increase in comparison to RA-untreated counterparts). Interestingly, the RA-derived MSCs displayed enhanced differentiation potential into adipocytes. Thus, these findings present a novel RA-based approach for providing an unlimited source of MSCs and adipocytes that can be used for regenerative medicine, drug screening and disease modeling applications.
Collapse
|
42
|
Zaky DA, Abouelfadl DM, Nassar NN, Abdallah DM, Al-Shorbagy MY. The paradox of dipeptidyl peptidase IV inhibition in enterocytic differentiation and epithelial-mesenchymal transition in rat cholestatic sepsis. Toxicol Appl Pharmacol 2020; 394:114956. [PMID: 32171571 DOI: 10.1016/j.taap.2020.114956] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 02/19/2020] [Accepted: 03/10/2020] [Indexed: 12/16/2022]
Abstract
Proper enterocytic proliferation/differentiation, besides providing adequate adherens junctions (AJ) integrity, are responsible for strengthening of the gut barrier that acts as a first line defense against endotoxemia. However, the preferential role of the underlying PI3K/Akt (PKB) axis in triggering enterocytic proliferation/differentiation signaling and AJ assembly is still obscure in sepsis. Additionally, the potential involvement of dipeptidyl peptidase (DPP)-IV in cholestatic sepsis has not yet been reported. Common bile duct ligation (CBDL) insult was performed in adult male Sprague-Dawley rats except for sham operated animals; three doses of vildagliptin (VLD3, 10 and 30 mg/kg/d; p.o) were administered for 10 consecutive days post CBDL. VLD3/10/30 dose-dependently decreased DPP-IV and elevated GLP-1, IGF-1, PI3K, pS473-Akt (PKB), pS9-GSK-3β, pS133-CREB and cyclin-D1. VLD3/10 reduced fever, portal/aortic endotoxin and IgG, body weight loss as well as ileal NF-κB, TNF-α, MPO, TBARS, subepithelial/pericryptal and submucosal collagen deposition, vimentin immunoreactivity, N-cadherin, Zeb1 and pY654-β-catenin but increased E-cadherin, NPSH and colon/spleen indices - effects that were quite the opposite of VLD30. Accordingly, maintaining proper enterocytic proliferation/differentiation and phosphorylation inputs consequent to adequate DPP-IV inhibition is integral to AJ assembly in cholestatic sepsis; however, perturbed signals by excessive suppression of the enzyme activity induce toxic effects manifested as AJ disassembly and EMT, hence gut leakage and overt endotoxemia.
Collapse
Affiliation(s)
- Doaa A Zaky
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El-Aini street, Cairo, P.O. Box 11562, Egypt.
| | | | - Noha N Nassar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El-Aini street, Cairo, P.O. Box 11562, Egypt
| | - Dalaal M Abdallah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El-Aini street, Cairo, P.O. Box 11562, Egypt
| | - Muhammad Y Al-Shorbagy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El-Aini street, Cairo, P.O. Box 11562, Egypt; School of Pharmacy, NewGiza University, Giza, Egypt
| |
Collapse
|
43
|
Togo S, Sato K, Kawamura R, Kobayashi N, Noiri M, Nakabayashi S, Teramura Y, Yoshikawa HY. Quantitative evaluation of the impact of artificial cell adhesion via DNA hybridization on E-cadherin-mediated cell adhesion. APL Bioeng 2020; 4:016103. [PMID: 32002498 PMCID: PMC6984976 DOI: 10.1063/1.5123749] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 12/12/2019] [Indexed: 01/10/2023] Open
Abstract
Programmable cell adhesion with DNA hybridization is a promising approach for fabricating various tissue architectures without sophisticated instrumentation. However, little is known about how this artificial interaction influences the binding of cell adhesion proteins, E-cadherin. In this work, we designed a planar and fluid lipid membrane displaying E-cadherin and/or single-strand DNA with well-defined densities. Visualization of cells on membranes by fluorescence and interference microscopy revealed cell adhesion to be a two-step process: artificial adhesion by DNA hybridization within a few minutes followed by biological adhesion via cadherin-cadherin binding within hours. Furthermore, we discovered that DNA hybridization can substantially facilitate E-cadherin-mediated cell adhesion. The promotive effect is probably due to the enforced binding between E-cadherin molecules in geometrical confinement between two membranes. Our in vitro model of cell adhesion can potentially be used to design functional synthetic molecules that can regulate cell adhesion via cell adhesion proteins for tissue engineering.
Collapse
Affiliation(s)
- Shodai Togo
- Department of Chemistry, Saitama University, 338-8570 Saitama, Japan
| | - Ken Sato
- Department of Chemistry, Saitama University, 338-8570 Saitama, Japan
| | | | - Naritaka Kobayashi
- Division of Strategic Research and Development, Saitama University, 338-8570 Saitama, Japan
| | - Makoto Noiri
- Department of Bioengineering, The University of Tokyo, 113-8656 Tokyo, Japan
| | | | | | | |
Collapse
|
44
|
Chan SW, Rizwan M, Yim EKF. Emerging Methods for Enhancing Pluripotent Stem Cell Expansion. Front Cell Dev Biol 2020; 8:70. [PMID: 32117992 PMCID: PMC7033584 DOI: 10.3389/fcell.2020.00070] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 01/27/2020] [Indexed: 12/12/2022] Open
Abstract
Pluripotent stem cells (PSCs) have great potential to revolutionize the fields of tissue engineering and regenerative medicine as well as stem cell therapeutics. However, the end goal of using PSCs for therapeutic use remains distant due to limitations in current PSC production. Conventional methods for PSC expansion have limited potential to be scaled up to produce the number of cells required for the end-goal of therapeutic use due to xenogenic components, high cost or low efficiency. In this mini review, we explore novel methods and emerging technologies of improving PSC expansion: the use of the two-dimensional mechanobiological strategies of topography and stiffness and the use of three-dimensional (3D) expansion methods including encapsulation, microcarrier-based culture, and suspension culture. Additionally, we discuss the limitations of conventional PSC expansion methods as well as the challenges in implementing non-conventional methods.
Collapse
Affiliation(s)
- Sarah W. Chan
- Department of Chemical Engineering, Faculty of Engineering, University of Waterloo, Waterloo, ON, Canada
| | - Muhammad Rizwan
- Department of Chemical Engineering, Faculty of Engineering, University of Waterloo, Waterloo, ON, Canada
| | - Evelyn K. F. Yim
- Department of Chemical Engineering, Faculty of Engineering, University of Waterloo, Waterloo, ON, Canada
- Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, ON, Canada
- Centre for Biotechnology and Bioengineering, University of Waterloo, Waterloo, ON, Canada
| |
Collapse
|
45
|
Aceto N. Bring along your friends: Homotypic and heterotypic circulating tumor cell clustering to accelerate metastasis. Biomed J 2020; 43:18-23. [PMID: 32200952 PMCID: PMC7090281 DOI: 10.1016/j.bj.2019.11.002] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 11/01/2019] [Accepted: 11/07/2019] [Indexed: 11/23/2022] Open
Abstract
Metastasis formation is a hallmark of invasive cancers and it is achieved through the shedding of circulating tumor cells (CTCs) from the primary site into the blood circulation. There, CTCs are found as single cells or as multicellular clusters, with clusters carrying an elevated ability to survive within the bloodstream and initiate new metastatic lesions at distant sites. Clusters of CTCs include homotypic clusters made of cancer cells only, as well as heterotypic clusters that incorporate stromal or immune cells along with cancer cells. Both homotypic and heterotypic CTC clusters are characterized by a high metastasis-forming capability, high proliferation rate and by distinct molecular features compared to single CTCs, and their presence in the peripheral circulation of cancer patients is generally associated with a poor prognosis. In this short review, we summarize the current literature that describes homotypic and heterotypic CTC clusters, both in the context of their molecular characteristics as well as their value in the clinical setting. While CTC clusters have only recently emerged as key players in the metastatic process and many aspects of their biology remain to be investigated, a detailed understanding of their vulnerabilities may pave the way towards the generation of new metastasis-suppressing agents.
Collapse
Affiliation(s)
- Nicola Aceto
- Department of Biomedicine, Cancer Metastasis Laboratory, University of Basel and University Hospital Basel, Basel, Switzerland.
| |
Collapse
|
46
|
Ikonomou L, Herriges MJ, Lewandowski SL, Marsland R, Villacorta-Martin C, Caballero IS, Frank DB, Sanghrajka RM, Dame K, Kańduła MM, Hicks-Berthet J, Lawton ML, Christodoulou C, Fabian AJ, Kolaczyk E, Varelas X, Morrisey EE, Shannon JM, Mehta P, Kotton DN. The in vivo genetic program of murine primordial lung epithelial progenitors. Nat Commun 2020; 11:635. [PMID: 32005814 PMCID: PMC6994558 DOI: 10.1038/s41467-020-14348-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 12/23/2019] [Indexed: 12/29/2022] Open
Abstract
Multipotent Nkx2-1-positive lung epithelial primordial progenitors of the foregut endoderm are thought to be the developmental precursors to all adult lung epithelial lineages. However, little is known about the global transcriptomic programs or gene networks that regulate these gateway progenitors in vivo. Here we use bulk RNA-sequencing to describe the unique genetic program of in vivo murine lung primordial progenitors and computationally identify signaling pathways, such as Wnt and Tgf-β superfamily pathways, that are involved in their cell-fate determination from pre-specified embryonic foregut. We integrate this information in computational models to generate in vitro engineered lung primordial progenitors from mouse pluripotent stem cells, improving the fidelity of the resulting cells through unbiased, easy-to-interpret similarity scores and modulation of cell culture conditions, including substratum elastic modulus and extracellular matrix composition. The methodology proposed here can have wide applicability to the in vitro derivation of bona fide tissue progenitors of all germ layers.
Collapse
Affiliation(s)
- Laertis Ikonomou
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA, 02118, USA.
- The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA, 02118, USA.
| | - Michael J Herriges
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA, 02118, USA
- The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Sara L Lewandowski
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA, 02118, USA
- The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Robert Marsland
- Department of Physics, Boston University, Boston, MA, 02215, USA
| | - Carlos Villacorta-Martin
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA, 02118, USA
| | - Ignacio S Caballero
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA, 02118, USA
| | - David B Frank
- Division of Pediatric Cardiology, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Reeti M Sanghrajka
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA, 02118, USA
- The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Keri Dame
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA, 02118, USA
- The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Maciej M Kańduła
- Department of Mathematics & Statistics, Boston University, Boston, MA, 02215, USA
- Chair of Bioinformatics Research Group, Boku University, 1190, Vienna, Austria
| | - Julia Hicks-Berthet
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Matthew L Lawton
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA, 02118, USA
- The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Constantina Christodoulou
- The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA, 02118, USA
| | | | - Eric Kolaczyk
- Department of Mathematics & Statistics, Boston University, Boston, MA, 02215, USA
| | - Xaralabos Varelas
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Edward E Morrisey
- Penn Center for Pulmonary Biology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - John M Shannon
- Division of Pulmonary Biology, Cincinnati Children's Hospital, Cincinnati, OH, 45229, USA
| | - Pankaj Mehta
- Department of Physics, Boston University, Boston, MA, 02215, USA
| | - Darrell N Kotton
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA, 02118, USA.
- The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA, 02118, USA.
| |
Collapse
|
47
|
Yan Z, Xiao Y, Chen Y, Luo G. Screening and identification of epithelial-to-mesenchymal transition-related circRNA and miRNA in prostate cancer. Pathol Res Pract 2019; 216:152784. [PMID: 31882179 DOI: 10.1016/j.prp.2019.152784] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 11/22/2019] [Accepted: 12/10/2019] [Indexed: 01/02/2023]
Abstract
Epithelial-to-mesenchymal transition (EMT) plays a vital role in the progression and metastasis of prostate cancer. However, the molecular mechanisms underlying prostate cancer metastasis are not fully demonstrated. In this study, EMT was induced by interferon-γ (IFN-γ) in PC-3M IE8 cells. High-throughput sequencing was used to screen the differentially expressed circular RNAs (circRNAs) and miRNAs in the cells with or without IFN-γ treatment. EMT-related circRNAs and miRNAs were further identified by quantitative real-time PCR (qPCR). In addition, the relationships among circRNAs, miRNAs, and mRNA were predicted. After cells were treated with IFN-γ, western blot analysis was conducted to detect the expression levels of EMT markers. E-cadherin expression levels were found to be downregulated, and Twist expression levels were found to be upregulated. Our results also found that IFN-γ promoted PC-3M IE8 cell migration and invasion, indicating that IFN-γ could induce EMT in PC-3M IE8 cells. Furthermore, high-throughput sequencing results revealed 827 upregulated and 1279 downregulated circRNAs and 39 upregulated and 2076 downregulated miRNAs in the IFN-γ group compared with the control group. KEGG analysis showed that both differentially expressed circRNAs and differentially expressed miRNAs were enriched in the MAPK signaling pathway related to EMT. Furthermore, the qPCR results revealed that the expression of hsa_circ_0001085, hsa_circ_0004916, hsa_circ_0001165, hsa-miR-196b-5p, and hsa-miR-187-3p in the IFN-γ group was consistent with the sequencing results. hsa_circ_0001165 and hsa_circ_0001085 were used to construct the network of circRNA-miRNA-mRNA. It was found that hsa_circ_0001165 may regulate TNF expression through hsa-miR-187-3p to induce EMT in prostate cancer cells. In addition, hsa_circ_0001085 may indirectly regulate the PI3K-Akt signaling and TGF-β signaling pathways through hsa-miR-196b-5p and the MAPK signaling pathway through has-miR-451a, which played a regulatory role in prostate cancer cells in the EMT induction model. The results obtained in this study lay the foundation for future study.
Collapse
Affiliation(s)
- Zhijian Yan
- Urology Department, Zhongshan Hospital Xiamen University, Xiamen, 361003, China
| | - Yiming Xiao
- Urology Department, Zhongshan Hospital Xiamen University, Xiamen, 361003, China
| | - Yiyan Chen
- Urology Department, Zhongshan Hospital Xiamen University, Xiamen, 361003, China
| | - Guangcheng Luo
- Urology Department, Zhongshan Hospital Xiamen University, Xiamen, 361003, China.
| |
Collapse
|
48
|
Liu W, Deng C, Godoy-Parejo C, Zhang Y, Chen G. Developments in cell culture systems for human pluripotent stem cells. World J Stem Cells 2019; 11:968-981. [PMID: 31768223 PMCID: PMC6851012 DOI: 10.4252/wjsc.v11.i11.968] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 08/21/2019] [Accepted: 08/27/2019] [Indexed: 02/06/2023] Open
Abstract
Human pluripotent stem cells (hPSCs) are important resources for cell-based therapies and pharmaceutical applications. In order to realize the potential of hPSCs, it is critical to develop suitable technologies required for specific applications. Most hPSC technologies depend on cell culture, and are critically influenced by culture medium composition, extracellular matrices, handling methods, and culture platforms. This review summarizes the major technological advances in hPSC culture, and highlights the opportunities and challenges in future therapeutic applications.
Collapse
Affiliation(s)
- Weiwei Liu
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau 999078, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau 999078, China
- Bioimaging and Stem Cell Core Facility, Faculty of Health Sciences, University of Macau, Macau 999078, China
| | - Chunhao Deng
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau 999078, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau 999078, China
| | - Carlos Godoy-Parejo
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau 999078, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau 999078, China
| | - Yumeng Zhang
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau 999078, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau 999078, China
| | - Guokai Chen
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau 999078, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau 999078, China.
| |
Collapse
|
49
|
Gkountela S, Castro-Giner F, Szczerba BM, Vetter M, Landin J, Scherrer R, Krol I, Scheidmann MC, Beisel C, Stirnimann CU, Kurzeder C, Heinzelmann-Schwarz V, Rochlitz C, Weber WP, Aceto N. Circulating Tumor Cell Clustering Shapes DNA Methylation to Enable Metastasis Seeding. Cell 2019; 176:98-112.e14. [PMID: 30633912 PMCID: PMC6363966 DOI: 10.1016/j.cell.2018.11.046] [Citation(s) in RCA: 589] [Impact Index Per Article: 98.2] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 08/31/2018] [Accepted: 11/28/2018] [Indexed: 01/01/2023]
Abstract
The ability of circulating tumor cells (CTCs) to form clusters has been linked to increased metastatic potential. Yet biological features and vulnerabilities of CTC clusters remain largely unknown. Here, we profile the DNA methylation landscape of single CTCs and CTC clusters from breast cancer patients and mouse models on a genome-wide scale. We find that binding sites for stemness- and proliferation-associated transcription factors are specifically hypomethylated in CTC clusters, including binding sites for OCT4, NANOG, SOX2, and SIN3A, paralleling embryonic stem cell biology. Among 2,486 FDA-approved compounds, we identify Na+/K+ ATPase inhibitors that enable the dissociation of CTC clusters into single cells, leading to DNA methylation remodeling at critical sites and metastasis suppression. Thus, our results link CTC clustering to specific changes in DNA methylation that promote stemness and metastasis and point to cluster-targeting compounds to suppress the spread of cancer.
Collapse
Affiliation(s)
- Sofia Gkountela
- Cancer Metastasis Laboratory, Department of Biomedicine, University of Basel and University Hospital Basel, 4058 Basel, Switzerland
| | - Francesc Castro-Giner
- Cancer Metastasis Laboratory, Department of Biomedicine, University of Basel and University Hospital Basel, 4058 Basel, Switzerland; SIB Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Barbara Maria Szczerba
- Cancer Metastasis Laboratory, Department of Biomedicine, University of Basel and University Hospital Basel, 4058 Basel, Switzerland
| | - Marcus Vetter
- Gynecologic Cancer Center, University Hospital Basel, 4056 Basel, Switzerland; Department of Medical Oncology, University Hospital Basel, 4056 Basel, Switzerland
| | - Julia Landin
- Department of Medical Oncology, University Hospital Basel, 4056 Basel, Switzerland; Breast Center, University Hospital Basel and University of Basel, 4031 Basel, Switzerland
| | - Ramona Scherrer
- Cancer Metastasis Laboratory, Department of Biomedicine, University of Basel and University Hospital Basel, 4058 Basel, Switzerland
| | - Ilona Krol
- Cancer Metastasis Laboratory, Department of Biomedicine, University of Basel and University Hospital Basel, 4058 Basel, Switzerland
| | - Manuel C Scheidmann
- Cancer Metastasis Laboratory, Department of Biomedicine, University of Basel and University Hospital Basel, 4058 Basel, Switzerland
| | - Christian Beisel
- Department of Biosystems Science and Engineering, ETH Zurich, 4058 Basel, Switzerland
| | | | - Christian Kurzeder
- Gynecologic Cancer Center, University Hospital Basel, 4056 Basel, Switzerland; Breast Center, University Hospital Basel and University of Basel, 4031 Basel, Switzerland
| | | | - Christoph Rochlitz
- Department of Medical Oncology, University Hospital Basel, 4056 Basel, Switzerland
| | - Walter Paul Weber
- Breast Center, University Hospital Basel and University of Basel, 4031 Basel, Switzerland
| | - Nicola Aceto
- Cancer Metastasis Laboratory, Department of Biomedicine, University of Basel and University Hospital Basel, 4058 Basel, Switzerland.
| |
Collapse
|
50
|
Yang S, Cao Z, Zhu J, Zhang Z, Zhao H, Zhao L, Sun X, Wang X. In Vitro Monolayer Culture of Dispersed Neural Stem Cells on the E-Cadherin-Based Substrate with Long-Term Stemness Maintenance. ACS OMEGA 2019; 4:18136-18146. [PMID: 31720516 PMCID: PMC6843705 DOI: 10.1021/acsomega.9b02053] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 09/13/2019] [Indexed: 05/08/2023]
Abstract
Neural stem cells (NSCs) play an important role in neural tissue engineering because of their capacity of self-renewal and differentiation to multiple cell lineages. The in vitro conventional neurosphere culture protocol has some limitations such as limited nutrition and oxygen penetration and distribution causing the heterogeneity of cells inside, inaccessibility of internal cells, and inhomogeneous cellular morphology and properties. As a result, cultivation as a monolayer is a better way to study NSCs and obtain a homogeneous cell population. The cadherins are a classical family of homophilic cell adhesion molecules mediating cell-cell adhesion. Here, we used a recombinant human E-cadherin mouse IgG Fc chimera protein that self-assembles on a hydrophobic polystyrene surface via hydrophobic interaction to obtain an E-cadherin-coated culture plate (ECP). The rat fetal NSCs were cultured on the ECP and routine tissue culture plate (TCP) from passage 2 to passage 5. NSCs on TCP formed uniform floating neurospheres and grew up over time, while cells on the ECP adhered on the bottom of the plate and exhibited individual cells with scattering morphology, forming intercellular connections between cells. The cell proliferation and differentiation behaviors that were evaluated by Cell Counting Kit-8 assay (CCK-8), immunofluorescence staining, and real-time quantitative polymerase chain reaction showed NSCs could maintain the capacity for self-renewal and ability to differentiate into neurons, oligodendrocytes, and astrocytes after the long-term in vitro cell culture and passaging. Therefore, our study indicated that hE-cad-Fc could provide a homogeneous environment for individual cells in monolayer conditions to maintain the capacity of self-renewal and differentiation by mimicking the cell-cell interaction.
Collapse
Affiliation(s)
- Shuhui Yang
- State
Key Laboratory of New Ceramics and Fine Processing, Key Laboratory
of Advanced Materials of Ministry of Education, School of Materials
Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Zheng Cao
- State
Key Laboratory of New Ceramics and Fine Processing, Key Laboratory
of Advanced Materials of Ministry of Education, School of Materials
Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Jinjin Zhu
- Department
of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College
of Zhejiang University, Sir Run Run Shaw
Institute of Clinical Medicine of Zhejiang University, 3 East Qingchun Road, Hangzhou 310016, Zhejiang Province, China
| | - Zhe Zhang
- State
Key Laboratory of New Ceramics and Fine Processing, Key Laboratory
of Advanced Materials of Ministry of Education, School of Materials
Science and Engineering, Tsinghua University, Beijing 100084, China
| | - He Zhao
- State
Key Laboratory of New Ceramics and Fine Processing, Key Laboratory
of Advanced Materials of Ministry of Education, School of Materials
Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Lingyun Zhao
- State
Key Laboratory of New Ceramics and Fine Processing, Key Laboratory
of Advanced Materials of Ministry of Education, School of Materials
Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Xiaodan Sun
- State
Key Laboratory of New Ceramics and Fine Processing, Key Laboratory
of Advanced Materials of Ministry of Education, School of Materials
Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Xiumei Wang
- State
Key Laboratory of New Ceramics and Fine Processing, Key Laboratory
of Advanced Materials of Ministry of Education, School of Materials
Science and Engineering, Tsinghua University, Beijing 100084, China
| |
Collapse
|