1
|
Li M, Suzuki K, Wang M, Benner C, Ku M, Ma L, Kobari L, Kim NY, Montserrat N, Chang CJ, Liu G, Qu J, Xu J, Zhang Y, Aizawa E, Wu J, Douay L, Esteban CR, Belmonte JCI. Dynamic WNT signaling controls differentiation of hematopoietic progenitor cells from human pluripotent stem cells. SCIENCE CHINA. LIFE SCIENCES 2025:10.1007/s11427-024-2816-0. [PMID: 40080269 DOI: 10.1007/s11427-024-2816-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 12/11/2024] [Indexed: 03/15/2025]
Abstract
Human pluripotent stem cells (hPSCs) can in theory give rise to any hematopoietic lineages, thereby offering opportunities for disease modeling, drug screening and cell therapies. However, gaps in our knowledge of the signaling requirements for the specification of human hematopoietic stem/progenitor cells (HSPCs), which lie at the apex of all hematopoietic lineages, greatly limit the potential of hPSC in hematological research and application. Transcriptomic analysis reveals aberrant regulation of WNT signaling during maturation of hPSC-derived hematopoietic progenitor cells (hPSC-HPCs), which results in higher mitochondria activity, misregulation of HOX genes, loss of self-renewal and precocious differentiation. These defects are partly due to the activation of the WNT target gene CDX2. Late-stage WNT inhibition improves the yield, self-renewal, multilineage differentiation, and transcriptional and metabolic profiles of hPSC-HPCs. Genome-wide mapping of transcription factor (TF) accessible chromatin reveals a significant overrepresentation of myeloid TF binding motifs in hPSC-HPCs, which could underlie their myeloid-biased lineage potential. Together our findings uncover a previously unappreciated dynamic requirement of the WNT signaling pathway during the specification of human HSPCs. Modulating the WNT pathway with small molecules normalizes the molecular differences between hPSC-HPCs and endogenous hematopoietic stem cells (HSCs), thereby representing a promising approach to improve the differentiation and function of hPSC-HPCs.
Collapse
Affiliation(s)
- Mo Li
- Bioscience Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia.
- KAUST Center of Excellence for Smart Health (KCSH), Thuwal, 23955-6900, Kingdom of Saudi Arabia.
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, 92037, USA.
| | - Keiichiro Suzuki
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, 92037, USA
- Institute for Advanced Co-Creation Studies, Osaka University, Osaka, 560-8531, Japan
| | - Mengge Wang
- Bioscience Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
- KAUST Center of Excellence for Smart Health (KCSH), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Christopher Benner
- Integrative Genomics and Bioinformatics Core, Salk Institute for Biological Studies, La Jolla, 92037, USA
| | - Manching Ku
- Next Generation Sequencing Core, Salk Institute for Biological Studies, La Jolla, 92037, USA
| | - Li Ma
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, 92037, USA
| | - Ladan Kobari
- Prolifération et Différentiation des Cellules Souches, UPMC Univ Paris 06, UMR_S938 CDR Saint-Antoine, Paris, F-75012, France
- Prolifération et Différentiation des Cellules Souches, INSERM, UMR_S938, Paris, F-75012, France
| | - Na Young Kim
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, 92037, USA
| | - Nuria Montserrat
- Center for Regenerative Medicine in Barcelona, Barcelona, 08003, Spain
| | - Chan-Jung Chang
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, 92037, USA
| | - Guanghui Liu
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, 92037, USA
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jing Qu
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, 92037, USA
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jinna Xu
- Bioscience Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
- KAUST Center of Excellence for Smart Health (KCSH), Thuwal, 23955-6900, Kingdom of Saudi Arabia
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, 92037, USA
| | - Yingzi Zhang
- Bioscience Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Emi Aizawa
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, 92037, USA
| | - Jun Wu
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, 92037, USA
| | - Luc Douay
- Prolifération et Différentiation des Cellules Souches, UPMC Univ Paris 06, UMR_S938 CDR Saint-Antoine, Paris, F-75012, France
- Unité d'Ingénierie et de Thérapie Cellulaire, EFS Ile de France, Créteil, F-94017, France
- Service d'Hématologie et immunologie biologique, AP-HP Hôpital Saint Antoine/Armand Trousseau, Paris, F-75012, France
| | - Concepcion Rodriguez Esteban
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, 92037, USA
- Altos Labs, San Diego, 92122, USA
| | - Juan Carlos Izpisua Belmonte
- Bioscience Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia.
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, 92037, USA.
- Altos Labs, San Diego, 92122, USA.
| |
Collapse
|
2
|
Benati D, Leung A, Perdigao P, Toulis V, van der Spuy J, Recchia A. Induced Pluripotent Stem Cells and Genome-Editing Tools in Determining Gene Function and Therapy for Inherited Retinal Disorders. Int J Mol Sci 2022; 23:ijms232315276. [PMID: 36499601 PMCID: PMC9735568 DOI: 10.3390/ijms232315276] [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] [Received: 10/04/2022] [Revised: 11/25/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022] Open
Abstract
Inherited retinal disorders (IRDs) affect millions of people worldwide and are a major cause of irreversible blindness. Therapies based on drugs, gene augmentation or transplantation approaches have been widely investigated and proposed. Among gene therapies for retinal degenerative diseases, the fast-evolving genome-editing CRISPR/Cas technology has emerged as a new potential treatment. The CRISPR/Cas system has been developed as a powerful genome-editing tool in ophthalmic studies and has been applied not only to gain proof of principle for gene therapies in vivo, but has also been extensively used in basic research to model diseases-in-a-dish. Indeed, the CRISPR/Cas technology has been exploited to genetically modify human induced pluripotent stem cells (iPSCs) to model retinal disorders in vitro, to test in vitro drugs and therapies and to provide a cell source for autologous transplantation. In this review, we will focus on the technological advances in iPSC-based cellular reprogramming and gene editing technologies to create human in vitro models that accurately recapitulate IRD mechanisms towards the development of treatments for retinal degenerative diseases.
Collapse
Affiliation(s)
- Daniela Benati
- Centre for Regenerative Medicine, Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Amy Leung
- UCL Institute of Ophthalmology, London EC1V 9EL, UK
| | - Pedro Perdigao
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | | | | | - Alessandra Recchia
- Centre for Regenerative Medicine, Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
- Correspondence: (J.v.d.S.); (A.R.)
| |
Collapse
|
3
|
Yuan B, Zhou X, Suzuki K, Ramos-Mandujano G, Wang M, Tehseen M, Cortés-Medina LV, Moresco JJ, Dunn S, Hernandez-Benitez R, Hishida T, Kim NY, Andijani MM, Bi C, Ku M, Takahashi Y, Xu J, Qiu J, Huang L, Benner C, Aizawa E, Qu J, Liu GH, Li Z, Yi F, Ghosheh Y, Shao C, Shokhirev M, Comoli P, Frassoni F, Yates JR, Fu XD, Esteban CR, Hamdan S, Li M, Izpisua Belmonte JC. Wiskott-Aldrich syndrome protein forms nuclear condensates and regulates alternative splicing. Nat Commun 2022; 13:3646. [PMID: 35752626 PMCID: PMC9233711 DOI: 10.1038/s41467-022-31220-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 06/06/2022] [Indexed: 11/09/2022] Open
Abstract
The diverse functions of WASP, the deficiency of which causes Wiskott-Aldrich syndrome (WAS), remain poorly defined. We generated three isogenic WAS models using patient induced pluripotent stem cells and genome editing. These models recapitulated WAS phenotypes and revealed that WASP deficiency causes an upregulation of numerous RNA splicing factors and widespread altered splicing. Loss of WASP binding to splicing factor gene promoters frequently leads to aberrant epigenetic activation. WASP interacts with dozens of nuclear speckle constituents and constrains SRSF2 mobility. Using an optogenetic system, we showed that WASP forms phase-separated condensates that encompasses SRSF2, nascent RNA and active Pol II. The role of WASP in gene body condensates is corroborated by ChIPseq and RIPseq. Together our data reveal that WASP is a nexus regulator of RNA splicing that controls the transcription of splicing factors epigenetically and the dynamics of the splicing machinery through liquid-liquid phase separation.
Collapse
Affiliation(s)
- Baolei Yuan
- Bioscience Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Xuan Zhou
- Bioscience Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Keiichiro Suzuki
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037, USA.,Institute for Advanced Co-Creation Studies, Graduate School of Engineering Science, Osaka University, Osaka, Japan
| | - Gerardo Ramos-Mandujano
- Bioscience Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Mengge Wang
- Bioscience Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Muhammad Tehseen
- Bioscience Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Lorena V Cortés-Medina
- Bioscience Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - James J Moresco
- Department of Cell Biology, Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Sarah Dunn
- The Waitt Advanced Biophotonics Core Facility, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Reyna Hernandez-Benitez
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037, USA.,Altos Labs, Inc. 5510 Morehouse Drive, Suite 300, San Diego, CA, 92121, USA
| | - Tomoaki Hishida
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037, USA.,Laboratory of Biological Chemistry, School of Pharmaceutical Sciences, Wakayama Medical University, 25-1 Shitibancho, Wakayama, Wakayama, 640-8156, Japan
| | - Na Young Kim
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Manal M Andijani
- Bioscience Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Chongwei Bi
- Bioscience Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Manching Ku
- Next-generation sequencing core, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Yuta Takahashi
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037, USA.,Life Science Center, Tsukuba Advanced Research Alliance, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki, 305-8577, Japan
| | - Jinna Xu
- Bioscience Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Jinsong Qiu
- Department of Cellular & Molecular Medicine, University of California at San Diego, La Jolla, CA, 92093, USA
| | - Ling Huang
- Integrative Genomics and Bioinformatics Core, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Christopher Benner
- Integrative Genomics and Bioinformatics Core, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Emi Aizawa
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037, USA.,Institute for Advanced Co-Creation Studies, Graduate School of Engineering Science, Osaka University, Osaka, Japan
| | - Jing Qu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Guang-Hui Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Zhongwei Li
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037, USA.,University of Southern California, 1333 San Pablo Street, MMR 618, Los Angeles, CA, 90033, USA
| | - Fei Yi
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037, USA.,Ambys Medicines, 131 Oyster Point Blvd. Suite 200, South San Francisco, CA, 94080, USA
| | - Yanal Ghosheh
- Bioscience Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Changwei Shao
- Department of Cellular & Molecular Medicine, University of California at San Diego, La Jolla, CA, 92093, USA
| | - Maxim Shokhirev
- Integrative Genomics and Bioinformatics Core, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Patrizia Comoli
- Pediatric Hematology/Oncology and Cell Factory, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Francesco Frassoni
- Department of Research Laboratories and Director of Center for Stem Cell and Cell Therapy, Instituto G. Gaslini Children Hospital Scientific Institute, 16147, Genova, Italy
| | - John R Yates
- Department of Cell Biology, Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Xiang-Dong Fu
- Department of Cellular & Molecular Medicine, University of California at San Diego, La Jolla, CA, 92093, USA
| | - Concepcion Rodriguez Esteban
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037, USA.,Altos Labs, Inc. 5510 Morehouse Drive, Suite 300, San Diego, CA, 92121, USA
| | - Samir Hamdan
- Bioscience Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Mo Li
- Bioscience Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia.
| | - Juan Carlos Izpisua Belmonte
- Bioscience Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia. .,Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037, USA. .,Altos Labs, Inc. 5510 Morehouse Drive, Suite 300, San Diego, CA, 92121, USA.
| |
Collapse
|
4
|
Wang J, Liu C, Chen Y, Wang W. Taiji-reprogram: a framework to uncover cell-type specific regulators and predict cellular reprogramming cocktails. NAR Genom Bioinform 2021; 3:lqab100. [PMID: 34761218 PMCID: PMC8573821 DOI: 10.1093/nargab/lqab100] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 09/29/2021] [Accepted: 10/05/2021] [Indexed: 12/21/2022] Open
Abstract
Cellular reprogramming is a promising technology to develop disease models and cell-based therapies. Identification of the key regulators defining the cell type specificity is pivotal to devising reprogramming cocktails for successful cell conversion but remains a great challenge. Here, we present a systems biology approach called Taiji-reprogram to efficiently uncover transcription factor (TF) combinations for conversion between 154 diverse cell types or tissues. This method integrates the transcriptomic and epigenomic data to construct cell-type specific genetic networks and assess the global importance of TFs in the network. Comparative analysis across cell types revealed TFs that are specifically important in a particular cell type and often tightly associated with cell-type specific functions. A systematic search of TFs with differential importance in the source and target cell types uncovered TF combinations for desired cell conversion. We have shown that Taiji-reprogram outperformed the existing methods to better recover the TFs in the experimentally validated reprogramming cocktails. This work not only provides a comprehensive catalog of TFs defining cell specialization but also suggests TF combinations for direct cell conversion.
Collapse
Affiliation(s)
- Jun Wang
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093-0359, USA
| | - Cong Liu
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093-0359, USA
| | - Yue Chen
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093-0359, USA
| | - Wei Wang
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093-0359, USA
| |
Collapse
|
5
|
Goldenberg D, McLaughlin C, Koduru SV, Ravnic DJ. Regenerative Engineering: Current Applications and Future Perspectives. Front Surg 2021; 8:731031. [PMID: 34805257 PMCID: PMC8595140 DOI: 10.3389/fsurg.2021.731031] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 10/13/2021] [Indexed: 12/12/2022] Open
Abstract
Many pathologies, congenital defects, and traumatic injuries are untreatable by conventional pharmacologic or surgical interventions. Regenerative engineering represents an ever-growing interdisciplinary field aimed at creating biological replacements for injured tissues and dysfunctional organs. The need for bioengineered replacement parts is ubiquitous among all surgical disciplines. However, to date, clinical translation has been limited to thin, small, and/or acellular structures. Development of thicker tissues continues to be limited by vascularization and other impediments. Nevertheless, currently available materials, methods, and technologies serve as robust platforms for more complex tissue fabrication in the future. This review article highlights the current methodologies, clinical achievements, tenacious barriers, and future perspectives of regenerative engineering.
Collapse
Affiliation(s)
- Dana Goldenberg
- Irvin S. Zubar Plastic Surgery Research Laboratory, Penn State College of Medicine, Hershey, PA, United States
- Department of Surgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Caroline McLaughlin
- Irvin S. Zubar Plastic Surgery Research Laboratory, Penn State College of Medicine, Hershey, PA, United States
- Department of Surgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Srinivas V. Koduru
- Irvin S. Zubar Plastic Surgery Research Laboratory, Penn State College of Medicine, Hershey, PA, United States
- Department of Surgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Dino J. Ravnic
- Irvin S. Zubar Plastic Surgery Research Laboratory, Penn State College of Medicine, Hershey, PA, United States
- Department of Surgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, United States
| |
Collapse
|
6
|
Mutukula N, Man Z, Takahashi Y, Iniesta Martinez F, Morales M, Carreon-Guarnizo E, Hernandez Clares R, Garcia-Bernal D, Martinez Martinez L, Lajara J, Nuñez Delicado E, Meca Lallana JE, Izpisua Belmonte JC. Generation of RRMS and PPMS specific iPSCs as a platform for modeling Multiple Sclerosis. Stem Cell Res 2021; 53:102319. [PMID: 33894548 DOI: 10.1016/j.scr.2021.102319] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 03/05/2021] [Accepted: 03/26/2021] [Indexed: 11/25/2022] Open
Abstract
The advent of cellular reprogramming technology converting somatic cells into induced pluripotent stem cells (iPSCs) has revolutionized our understandings of neurodegenerative diseases that are otherwise hard to access and model. Multiple Sclerosis (MS) is a chronic demyelinating, inflammatory disease of central nervous system eventually causing neuronal death and accompanied disabilities. Here, we report the generation of several relapsing-remitting MS (RRMS) and primary progressive MS (PPMS) iPSC lines from MS patients along with their age matched healthy controls from peripheral blood mononuclear cells (PBMC). These patient specific iPSC lines displayed characteristic embryonic stem cell (ESC) morphology and exhibited pluripotency marker expression. Moreover, these MS iPSC lines were successfully differentiated into neural progenitor cells (NPC) after subjecting to neural induction. Furthermore, we identified the elevated expression of cellular senescence hallmarks in RRMS and PPMS neural progenitors unveiling a novel drug target avenue of MS pathophysiology. Thus, our study altogether offers both RRMS and PPMS iPSC cellular models as a good tool for better understanding of MS pathologies and drug testing.
Collapse
Affiliation(s)
- Naresh Mutukula
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Zhiqiu Man
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Yuta Takahashi
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Francisca Iniesta Martinez
- Clinical Neuroimmunology Unit and Multiple Sclerosis CSUR, Department of Neurology. "Virgen de la Arrixaca" Clinical University Hospital, IMIB-Arrixaca, Murcia, Spain; Universidad Católica San Antonio de Murcia (UCAM), Murcia, Spain
| | - Mariana Morales
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Ester Carreon-Guarnizo
- Clinical Neuroimmunology Unit and Multiple Sclerosis CSUR, Department of Neurology. "Virgen de la Arrixaca" Clinical University Hospital, IMIB-Arrixaca, Murcia, Spain; Universidad Católica San Antonio de Murcia (UCAM), Murcia, Spain
| | - Rocio Hernandez Clares
- Clinical Neuroimmunology Unit and Multiple Sclerosis CSUR, Department of Neurology. "Virgen de la Arrixaca" Clinical University Hospital, IMIB-Arrixaca, Murcia, Spain; Universidad Católica San Antonio de Murcia (UCAM), Murcia, Spain
| | - David Garcia-Bernal
- Hematopoietic Transplant and Cellular Therapy Unit, Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca, Medicine Department, University of Murcia, Murcia, Spain
| | | | - Jeronimo Lajara
- Universidad Católica San Antonio de Murcia (UCAM), Murcia, Spain
| | | | - Jose E Meca Lallana
- Clinical Neuroimmunology Unit and Multiple Sclerosis CSUR, Department of Neurology. "Virgen de la Arrixaca" Clinical University Hospital, IMIB-Arrixaca, Murcia, Spain; Universidad Católica San Antonio de Murcia (UCAM), Murcia, Spain.
| | | |
Collapse
|
7
|
Sriram S, Kang NY, Subramanian S, Nandi T, Sudhagar S, Xing Q, Tong GJL, Chen AKL, Srijaya TC, Tan P, Loh YH, Chang YT, Sugii S. Novel live cell fluorescent probe for human-induced pluripotent stem cells highlights early reprogramming population. Stem Cell Res Ther 2021; 12:113. [PMID: 33546754 PMCID: PMC7866770 DOI: 10.1186/s13287-021-02171-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 01/15/2021] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Despite recent rapid progress in method development and biological understanding of induced pluripotent stem (iPS) cells, there has been a relative shortage of tools that monitor the early reprogramming process into human iPS cells. METHODS We screened the in-house built fluorescent library compounds that specifically bind human iPS cells. After tertiary screening, the selected probe was analyzed for its ability to detect reprogramming cells in the time-dependent manner using high-content imaging analysis. The probe was compared with conventional dyes in different reprogramming methods, cell types, and cell culture conditions. Cell sorting was performed with the fluorescent probe to analyze the early reprogramming cells for their pluripotent characteristics and genome-wide gene expression signatures by RNA-seq. Finally, the candidate reprogramming factor identified was investigated for its ability to modulate reprogramming efficiency. RESULTS We identified a novel BODIPY-derived fluorescent probe, BDL-E5, which detects live human iPS cells at the early reprogramming stage. BDL-E5 can recognize authentic reprogramming cells around 7 days before iPS colonies are formed and stained positive with conventional pluripotent markers. Cell sorting of reprogrammed cells with BDL-E5 allowed generation of an increased number and higher quality of iPS cells. RNA sequencing analysis of BDL-E5-positive versus negative cells revealed early reprogramming patterns of gene expression, which notably included CREB1. Reprogramming efficiency was significantly increased by overexpression of CREB1 and decreased by knockdown of CREB1. CONCLUSION Collectively, BDL-E5 offers a valuable tool for delineating the early reprogramming pathway and clinically applicable commercial production of human iPS cells.
Collapse
Affiliation(s)
- Sandhya Sriram
- Fat Metabolism and Stem Cell Group, Singapore Bioimaging Consortium, A*STAR, 11 Biopolis Way, Singapore, 138667, Singapore
| | - Nam-Young Kang
- Laboratory of Bioimaging Probe Development, Singapore Bioimaging Consortium, A*STAR, 11 Biopolis Way, Singapore, 138667, Singapore.,Department of Creative IT Engineering, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea
| | - Subha Subramanian
- Fat Metabolism and Stem Cell Group, Singapore Bioimaging Consortium, A*STAR, 11 Biopolis Way, Singapore, 138667, Singapore
| | - Tannistha Nandi
- Cancer Therapeutics and Stratified Oncology, Genome Institute of Singapore, 60 Biopolis Street, Genome #02-01, Singapore, 138672, Singapore
| | - Samydurai Sudhagar
- Genome Institute of Singapore, 60 Biopolis Street, Genome, #02-01, Singapore, 138672, Singapore
| | - Qiaorui Xing
- Epigenetics and Cell Fates Laboratory, Institute of Molecular and Cell Biology, 61 Biopolis Drive, Singapore, 138673, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Gerine Jin-Ling Tong
- Bioprocessing Technology Institute, A*STAR, 20 Biopolis Way, #06-01 Centros, Singapore, 138668, Singapore
| | - Allen Kuan-Liang Chen
- Bioprocessing Technology Institute, A*STAR, 20 Biopolis Way, #06-01 Centros, Singapore, 138668, Singapore
| | | | - Patrick Tan
- Genome Institute of Singapore, 60 Biopolis Street, Genome, #02-01, Singapore, 138672, Singapore.,Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore.,SingHealth/Duke-NUS Institute of Precision Medicine, Singapore, 168752, Singapore
| | - Yuin-Han Loh
- Epigenetics and Cell Fates Laboratory, Institute of Molecular and Cell Biology, 61 Biopolis Drive, Singapore, 138673, Singapore.,Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore, 117543, Singapore
| | - Young-Tae Chang
- Laboratory of Bioimaging Probe Development, Singapore Bioimaging Consortium, A*STAR, 11 Biopolis Way, Singapore, 138667, Singapore.,Department of Chemistry, National University of Singapore, 3 Science Drive 3, Singapore, 117543, Singapore.,Department of Chemistry, POSTECH, Pohang, Gyeongbuk, 37673, Republic of Korea.,Center for Self-assembly and Complexity, Institute for Basic Science (IBS), Pohang, 37673, Republic of Korea
| | - Shigeki Sugii
- Fat Metabolism and Stem Cell Group, Singapore Bioimaging Consortium, A*STAR, 11 Biopolis Way, Singapore, 138667, Singapore. .,Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore. .,Institute of Bioengineering and Nanotechnology, A*STAR, 31 Biopolis Way, Singapore, 138669, Singapore.
| |
Collapse
|
8
|
"Betwixt Mine Eye and Heart a League Is Took": The Progress of Induced Pluripotent Stem-Cell-Based Models of Dystrophin-Associated Cardiomyopathy. Int J Mol Sci 2020; 21:ijms21196997. [PMID: 32977524 PMCID: PMC7582534 DOI: 10.3390/ijms21196997] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/17/2020] [Accepted: 09/21/2020] [Indexed: 12/19/2022] Open
Abstract
The ultimate goal of precision disease modeling is to artificially recreate the disease of affected people in a highly controllable and adaptable external environment. This field has rapidly advanced which is evident from the application of patient-specific pluripotent stem-cell-derived precision therapies in numerous clinical trials aimed at a diverse set of diseases such as macular degeneration, heart disease, spinal cord injury, graft-versus-host disease, and muscular dystrophy. Despite the existence of semi-adequate treatments for tempering skeletal muscle degeneration in dystrophic patients, nonischemic cardiomyopathy remains one of the primary causes of death. Therefore, cardiovascular cells derived from muscular dystrophy patients' induced pluripotent stem cells are well suited to mimic dystrophin-associated cardiomyopathy and hold great promise for the development of future fully effective therapies. The purpose of this article is to convey the realities of employing precision disease models of dystrophin-associated cardiomyopathy. This is achieved by discussing, as suggested in the title echoing William Shakespeare's words, the settlements (or "leagues") made by researchers to manage the constraints ("betwixt mine eye and heart") distancing them from achieving a perfect precision disease model.
Collapse
|
9
|
Salazar‐Roa M, Trakala M, Álvarez‐Fernández M, Valdés‐Mora F, Zhong C, Muñoz J, Yu Y, Peters TJ, Graña‐Castro O, Serrano R, Zapatero‐Solana E, Abad M, Bueno MJ, de Cedrón MG, Fernández‐Piqueras J, Serrano M, Blasco MA, Wang D, Clark SJ, Izpisua‐Belmonte JC, Ortega S, Malumbres M. Transient exposure to miR-203 enhances the differentiation capacity of established pluripotent stem cells. EMBO J 2020; 39:e104324. [PMID: 32614092 PMCID: PMC7429746 DOI: 10.15252/embj.2019104324] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 05/27/2020] [Accepted: 05/28/2020] [Indexed: 11/09/2022] Open
Abstract
Full differentiation potential along with self-renewal capacity is a major property of pluripotent stem cells (PSCs). However, the differentiation capacity frequently decreases during expansion of PSCs in vitro. We show here that transient exposure to a single microRNA, expressed at early stages during normal development, improves the differentiation capacity of already-established murine and human PSCs. Short exposure to miR-203 in PSCs (miPSCs) induces a transient expression of 2C markers that later results in expanded differentiation potency to multiple lineages, as well as improved efficiency in tetraploid complementation and human-mouse interspecies chimerism assays. Mechanistically, these effects are at least partially mediated by direct repression of de novo DNA methyltransferases Dnmt3a and Dnmt3b, leading to transient and reversible erasure of DNA methylation. These data support the use of transient exposure to miR-203 as a versatile method to reset the epigenetic memory in PSCs, and improve their effectiveness in regenerative medicine.
Collapse
Affiliation(s)
- María Salazar‐Roa
- Cell Division and Cancer GroupSpanish National Cancer Research Centre (CNIO)MadridSpain
| | - Marianna Trakala
- Cell Division and Cancer GroupSpanish National Cancer Research Centre (CNIO)MadridSpain
| | | | - Fátima Valdés‐Mora
- Epigenetics Research Program, Genomics and Epigenetics DivisionGarvan Institute of Medical ResearchSydneyNSWAustralia
- St. Vincent's Clinical SchoolUNSW, SydneySydneyNSWAustralia
| | - Cuiqing Zhong
- Gene Expression LaboratoryThe Salk Institute for Biological StudiesLa JollaCAUSA
| | | | - Yang Yu
- Gene Expression LaboratoryThe Salk Institute for Biological StudiesLa JollaCAUSA
| | - Timothy J Peters
- Epigenetics Research Program, Genomics and Epigenetics DivisionGarvan Institute of Medical ResearchSydneyNSWAustralia
| | | | | | | | | | - María José Bueno
- Cell Division and Cancer GroupSpanish National Cancer Research Centre (CNIO)MadridSpain
| | - Marta Gómez de Cedrón
- Cell Division and Cancer GroupSpanish National Cancer Research Centre (CNIO)MadridSpain
| | - José Fernández‐Piqueras
- Centro de Biología Molecular Severo Ochoa (CBMSO)Consejo Superior de Investigaciones Científicas‐Universidad Autónoma de Madrid (CSIC‐UAM)MadridSpain
- Centro de Investigación Biomédica en Red para Enfermedades Raras (CIBERER)Instituto de Salud Carlos IIIMadridSpain
- Instituto de Investigación BiosanitariaFundación Jimenez DíazMadridSpain
| | - Manuel Serrano
- Tumor Suppression GroupCNIOMadridSpain
- Institute for Research in Biomedicine (IRB Barcelona)The Barcelona Institute of Science and Technology (BIST)BarcelonaSpain
- Catalan Institution for Research and Advanced Studies (ICREA)BarcelonaSpain
| | | | - Da‐Zhi Wang
- Cardiovascular Research DivisionBoston Children′s HospitalHarvard Medical SchoolBostonMAUSA
| | - Susan J Clark
- Epigenetics Research Program, Genomics and Epigenetics DivisionGarvan Institute of Medical ResearchSydneyNSWAustralia
- St. Vincent's Clinical SchoolUNSW, SydneySydneyNSWAustralia
| | | | | | - Marcos Malumbres
- Cell Division and Cancer GroupSpanish National Cancer Research Centre (CNIO)MadridSpain
| |
Collapse
|
10
|
Stathakos P, Jiménez-Moreno N, Crompton LA, Nistor PA, Badger JL, Barbuti PA, Kerrigan TL, Randall AD, Caldwell MA, Lane JD. A monolayer hiPSC culture system for autophagy/mitophagy studies in human dopaminergic neurons. Autophagy 2020; 17:855-871. [PMID: 32286126 PMCID: PMC8078667 DOI: 10.1080/15548627.2020.1739441] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Macroautophagy/autophagy cytoplasmic quality control pathways are required during neural development and are critical for the maintenance of functional neuronal populations in the adult brain. Robust evidence now exists that declining neuronal autophagy pathways contribute to human neurodegenerative diseases, including Parkinson disease (PD). Reliable and relevant human neuronal model systems are therefore needed to understand the biology of disease-vulnerable neural populations, to decipher the underlying causes of neurodegenerative disease, and to develop assays to test therapeutic interventions in vitro. Human induced pluripotent stem cell (hiPSC) neural model systems can meet this demand: they provide a renewable source of material for differentiation into regional neuronal sub-types for functional assays; they can be expanded to provide a platform for screening, and they can potentially be optimized for transplantation/neurorestorative therapy. So far, however, hiPSC differentiation protocols for the generation of ventral midbrain dopaminergic neurons (mDANs) – the predominant neuronal sub-type afflicted in PD – have been somewhat restricted by poor efficiency and/or suitability for functional and/or imaging-based in vitro assays. Here, we describe a reliable, monolayer differentiation protocol for the rapid and reproducible production of high numbers of mDANs from hiPSC in a format that is amenable for autophagy/mitophagy research. We characterize these cells with respect to neuronal differentiation and macroautophagy capability and describe qualitative and quantitative assays for the study of autophagy and mitophagy in these important cells. Abbreviations: AA: ascorbic acid; ATG: autophagy-related; BDNF: brain derived neurotrophic factor; CCCP: carbonyl cyanide m-chlorophenylhydrazone; dbcAMP: dibutyryl cAMP; DAN: dopaminergic neuron; DAPI: 4ʹ,6-diamidino-2-phenylindole; DAPT: N-[N-(3,5-difluorophenacetyl)-L-alanyl]-sphenylglycine; DLG4/PSD95: discs large MAGUK scaffold protein 4; DMEM: Dulbecco’s modified eagle’s medium; EB: embryoid body; ECAR: extracellular acidification rate; EGF: epidermal growth factor; FACS: fluorescence-activated cell sorting; FCCP: arbonyl cyanide p-triflouromethoxyphenylhydrazone; FGF: fibroblast growth factor; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; GDNF: glia cell derived neurotrophic factor; hiPSC: human induced pluripotent stem cell; LAMP2A: lysosomal associated membrane protein 2A; LT-R: LysoTracker Red; MAP1LC3: microtubule associated protein 1 light chain 3; mDAN: midbrain dopaminergic neuron; MEF: mouse embryonic fibroblast; MT-GR: MitoTracker Green; MT-R: MitoTracker Red; NAS2: normal SNCA2; NEM: neuroprogenitor expansion media; NR4A2/NURR1: nuclear receptor subfamily group A member 2; OA: oligomycin and antimycin A; OCR: oxygen consumption rate; PD: Parkinson disease; SHH: sonic hedgehog signaling molecule; SNCA/α-synuclein: synuclein alpha; TH: tyrosine hydroxylase; VTN: vitronectin.
Collapse
Affiliation(s)
- Petros Stathakos
- Cell Biology Laboratories, School of Biochemistry, University of Bristol, Bristol, UK.,Regenerative Medicine Laboratory, School of Clinical Sciences, University of Bristol, Bristol, UK
| | | | - Lucy A Crompton
- Cell Biology Laboratories, School of Biochemistry, University of Bristol, Bristol, UK
| | - Paul A Nistor
- Regenerative Medicine Laboratory, School of Clinical Sciences, University of Bristol, Bristol, UK
| | - Jennifer L Badger
- Regenerative Medicine Laboratory, School of Clinical Sciences, University of Bristol, Bristol, UK
| | - Peter A Barbuti
- Regenerative Medicine Laboratory, School of Clinical Sciences, University of Bristol, Bristol, UK
| | - Talitha L Kerrigan
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratory, Exeter, UK.,Dementia Research Group, Institute of Clinical Neurosciences, School of Clinical Sciences, University of Bristol, Bristol, UK
| | - Andrew D Randall
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratory, Exeter, UK
| | - Maeve A Caldwell
- Regenerative Medicine Laboratory, School of Clinical Sciences, University of Bristol, Bristol, UK.,Trinity College Institute for Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Jon D Lane
- Cell Biology Laboratories, School of Biochemistry, University of Bristol, Bristol, UK
| |
Collapse
|
11
|
Amin N, Tan X, Ren Q, Zhu N, Botchway BOA, Hu Z, Fang M. Recent advances of induced pluripotent stem cells application in neurodegenerative diseases. Prog Neuropsychopharmacol Biol Psychiatry 2019; 95:109674. [PMID: 31255650 DOI: 10.1016/j.pnpbp.2019.109674] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 06/03/2019] [Accepted: 06/17/2019] [Indexed: 01/30/2023]
Abstract
Stem cell is defined by its ability to self-renewal and generates differentiated functional cell types, which are derived from the embryo and various sources of postnatal animal. These cells can be divided according to their potential development into totipotent, unipotent, multipotent andpluripotent. Pluripotent is considered as the most important type due to its advantageous capability to create different cell types of the body in a similar behavior as embryonic stem cell. Induced pluripotent stem cells (iPSCs) are adult cells that maintain the characteristics of embryonic stem cells because it can be genetically reprogrammed to an embryonic stem cell-like state via express genes and transcription factors. Such cells provide an efficient pathway to explorehuman diseases and their corresponding therapy, particularly, neurodevelopmental disorders. Consequently, iPSCs can be investigated to check the specific mutations of neurodegenerative disease due to their unique ability to differentiate into neural cell types and/or neural organoids. The current review addresses the different neurodegenerative diseases model by using iPSCs approach such as Alzheimer's diseases (AD), Parkinson diseases (PD),multiplesclerosis(MS) and psychiatric disorders. We also highlight the importance of autophagy in neurodegenerative diseases.
Collapse
Affiliation(s)
- Nashwa Amin
- Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou, China; Department of Zoology, Faculty of Science, Aswan University, Egypt
| | - Xiaoning Tan
- Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiannan Ren
- Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou, China
| | - Ning Zhu
- Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou, China; Hebei North University,Zhangjiakou, China
| | - Benson O A Botchway
- Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhiying Hu
- Obstetrics & Gynecology Department, Zhejiang Integrated Traditional and Western Medicine Hospital, Hangzhou, China.
| | - Marong Fang
- Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
12
|
Ou J, Rosa S, Berchowitz LE, Li W. Induced pluripotent stem cells as a tool for comparative physiology: lessons from the thirteen-lined ground squirrel. J Exp Biol 2019; 222:jeb196493. [PMID: 31585999 PMCID: PMC6806009 DOI: 10.1242/jeb.196493] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Comparative physiologists are often interested in adaptive physiological phenomena found in unconventional model organisms; however, research on these species is frequently constrained by the limited availability of investigative tools. Here, we propose that induced pluripotent stem cells (iPSCs) from unconventional model organisms may retain certain species-specific features that can consequently be investigated in depth in vitro; we use hibernating mammals as an example. Many species (including ground squirrels, bats and bears) can enter a prolonged state of physiological dormancy known as hibernation to survive unfavorable seasonal conditions. Our understanding of the mechanisms underpinning the rapid transition and adaptation to a hypothermic, metabolically suppressed winter torpor state remains limited partially because of the lack of an easily accessible model. To address the fascinating unanswered questions underlying hibernation biology, we have developed a powerful model system: iPSCs from a hibernating species, the thirteen-lined ground squirrel (Ictidomys tridecemlineatus). These stem cells can potentially be differentiated into any cell type, and can be used for the analysis of cell-autonomous mechanisms that facilitate adaptation to hibernation and for comparisons with non-hibernators. Furthermore, we can manipulate candidate molecular and cellular pathways underlying relevant physiological phenomena by pharmacological or RNAi-based methods, and CRISPR/Cas9 gene editing. Moreover, iPSC strategies can be applied to other species (e.g. seals, naked mole rats, humming birds) for in vitro studies on adaptation to extreme physiological conditions. In this Commentary, we discuss factors to consider when attempting to generate iPSCs from unconventional model organisms, based on our experience with the thirteen-lined ground squirrel.
Collapse
Affiliation(s)
- Jingxing Ou
- Retinal Neurophysiology Section, National Eye Institute, US National Institutes of Health, Bethesda, MD 20892, USA
| | - Sarah Rosa
- Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032, USA
- Taub Institute for Research on Alzheimer's and the Aging Brain, New York, NY 10032, USA
| | - Luke E Berchowitz
- Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032, USA
- Taub Institute for Research on Alzheimer's and the Aging Brain, New York, NY 10032, USA
| | - Wei Li
- Retinal Neurophysiology Section, National Eye Institute, US National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
13
|
Ling C, Liu Z, Song M, Zhang W, Wang S, Liu X, Ma S, Sun S, Fu L, Chu Q, Belmonte JCI, Wang Z, Qu J, Yuan Y, Liu GH. Modeling CADASIL vascular pathologies with patient-derived induced pluripotent stem cells. Protein Cell 2019; 10:249-271. [PMID: 30778920 PMCID: PMC6418078 DOI: 10.1007/s13238-019-0608-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Accepted: 12/29/2018] [Indexed: 12/23/2022] Open
Abstract
Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is a rare hereditary cerebrovascular disease caused by a NOTCH3 mutation. However, the underlying cellular and molecular mechanisms remain unidentified. Here, we generated non-integrative induced pluripotent stem cells (iPSCs) from fibroblasts of a CADASIL patient harboring a heterozygous NOTCH3 mutation (c.3226C>T, p.R1076C). Vascular smooth muscle cells (VSMCs) differentiated from CADASIL-specific iPSCs showed gene expression changes associated with disease phenotypes, including activation of the NOTCH and NF-κB signaling pathway, cytoskeleton disorganization, and excessive cell proliferation. In comparison, these abnormalities were not observed in vascular endothelial cells (VECs) derived from the patient's iPSCs. Importantly, the abnormal upregulation of NF-κB target genes in CADASIL VSMCs was diminished by a NOTCH pathway inhibitor, providing a potential therapeutic strategy for CADASIL. Overall, using this iPSC-based disease model, our study identified clues for studying the pathogenic mechanisms of CADASIL and developing treatment strategies for this disease.
Collapse
Affiliation(s)
- Chen Ling
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, 100053, China
- Department of Neurology, Peking University First Hospital, Beijing, 100034, China
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Zunpeng Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Moshi Song
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Institute for Stem cell and Regeneration, CAS, Beijing, 100101, China
| | - Weiqi Zhang
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, 100053, China
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Institute for Stem cell and Regeneration, CAS, Beijing, 100101, China
| | - Si Wang
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, 100053, China
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Institute for Stem cell and Regeneration, CAS, Beijing, 100101, China
| | - Xiaoqian Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shuai Ma
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Institute for Stem cell and Regeneration, CAS, Beijing, 100101, China
| | - Shuhui Sun
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lina Fu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qun Chu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Juan Carlos Izpisua Belmonte
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Zhaoxia Wang
- Department of Neurology, Peking University First Hospital, Beijing, 100034, China
| | - Jing Qu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Institute for Stem cell and Regeneration, CAS, Beijing, 100101, China.
| | - Yun Yuan
- Department of Neurology, Peking University First Hospital, Beijing, 100034, China.
| | - Guang-Hui Liu
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Institute for Stem cell and Regeneration, CAS, Beijing, 100101, China.
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou, 510632, China.
- Beijing Institute for Brain Disorders, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
14
|
Kitahata S, Tanaka Y, Hori K, Kime C, Sugita S, Ueda H, Takahashi M. Critical Functionality Effects from Storage Temperature on Human Induced Pluripotent Stem Cell-Derived Retinal Pigment Epithelium Cell Suspensions. Sci Rep 2019; 9:2891. [PMID: 30814559 PMCID: PMC6393435 DOI: 10.1038/s41598-018-38065-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 12/14/2018] [Indexed: 12/22/2022] Open
Abstract
Human induced pluripotent stem cell (hiPSC)-derived retinal pigment epithelium (hiPSC-RPE) cells suspension have the potential for regenerative treatment. However, practical regenerative applications with hiPSC-RPE cells require the development of simple and cost-effective non-freezing preservation methods. We investigated the effect of non-freezing temperatures on suspended hiPSC-RPE cells in various conditions and analysed mechanisms of cell death, anoikis, Rho GTPases, hypoxia, microtubule destruction, and cell metabolism. Cells stored at 37 °C had the lowest viability due to hypoxia from high cell metabolism and cell deposits, and cells preserved at 4 °C were damaged via microtubule fragility. Cell suspensions at 16 °C were optimal with drastically reduced apoptosis and negligible necrosis. Moreover, surviving cells proliferated and secreted key proteins normally, compared to cells without preservation. hiPSC-RPE cell suspensions were optimally preserved at 16 °C. Temperatures above or below the optimal temperature decreased cell viability significantly yet differentially by mechanisms of cell death, cellular metabolism, microtubule destruction, and oxygen tension, all relevant to cell conditions. Surviving cells are expected to function as grafts where high cell death is often reported. This study provides new insight into various non-freezing temperature effects on hiPSC-RPE cells that are highly relevant to clinical applications and may improve cooperation between laboratories and hospitals.
Collapse
Affiliation(s)
- Shohei Kitahata
- Laboratory for Retinal Regeneration, Biosystems Dynamics Research, RIKEN, Kobe, 650-0047, Japan.,Application Biology and Regenerative Medicine, Kyoto University Graduate School of Medicine, Kyoto, 606-8501, Japan
| | - Yuji Tanaka
- Laboratory for Retinal Regeneration, Biosystems Dynamics Research, RIKEN, Kobe, 650-0047, Japan. .,Division of Medicine, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Yamanashi, 409-3898, Japan.
| | - Kanji Hori
- Laboratory for Retinal Regeneration, Biosystems Dynamics Research, RIKEN, Kobe, 650-0047, Japan.,Department of Ophthalmology, Juntendo University School of Medicine, Tokyo, 113-8431, Japan
| | - Cody Kime
- Laboratory for Retinal Regeneration, Biosystems Dynamics Research, RIKEN, Kobe, 650-0047, Japan
| | - Sunao Sugita
- Laboratory for Retinal Regeneration, Biosystems Dynamics Research, RIKEN, Kobe, 650-0047, Japan.,Kobe City Eye Hospital Research Center, Kobe, 650-0047, Japan
| | - Hiroshi Ueda
- Department of Pharmacology and Therapeutic Innovation, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8521, Japan
| | - Masayo Takahashi
- Laboratory for Retinal Regeneration, Biosystems Dynamics Research, RIKEN, Kobe, 650-0047, Japan.,Kobe City Eye Hospital Research Center, Kobe, 650-0047, Japan
| |
Collapse
|
15
|
Hori K, Kuwabara J, Tanaka Y, Nishida M, Koide N, Takahashi M. A simple and static preservation system for shipping retinal pigment epithelium cell sheets. J Tissue Eng Regen Med 2019; 13:459-468. [PMID: 30644171 DOI: 10.1002/term.2805] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 10/25/2018] [Accepted: 12/17/2018] [Indexed: 12/15/2022]
Abstract
The ability to move cells and tissues from bench to bedside is an essential aspect of regenerative medicine. In this study, we propose a simple and static shipping system to deliver tissue-engineered cell sheets. Notably, this system is electronic-device-free and simplified to minimize the number of packing and opening steps involved. Shipping conditions were optimized, and application and verification of the system were performed using human iPS cell-derived or fetal retinal pigment epithelium (RPE) cell sheets. The temperature of the compartments within the insulated container was stable at various conditions, and filling up the cell vessel with medium effectively prevented turbulence-induced mechanical damage to the RPE cell sheets. Furthermore, no abnormal changes were observed in RPE morphology, transepithelial electrical resistance, or mRNA expression after transit by train and car. Taken together, our simple shipping system has the potential to minimize the costs and human error associated with bench to bedside tissue transfer. This specially designed regenerative tissue shipping system, validated for use in this field, can be used without any special training. This study provides a procedure for easily sharing engineered tissues with the goal of promoting collaboration between laboratories and hospitals and enhancing patient care.
Collapse
Affiliation(s)
- Kanji Hori
- Laboratory for Retinal Regeneration, RIKEN Center for Developmental Biology, Kobe, Japan.,Department of Ophthalmology, Juntendo University School of Medicine, Tokyo, Japan
| | | | - Yuji Tanaka
- Laboratory for Retinal Regeneration, RIKEN Center for Developmental Biology, Kobe, Japan
| | - Mitsuhiro Nishida
- Laboratory for Retinal Regeneration, RIKEN Center for Developmental Biology, Kobe, Japan
| | - Naoshi Koide
- Laboratory for Retinal Regeneration, RIKEN Center for Developmental Biology, Kobe, Japan
| | - Masayo Takahashi
- Laboratory for Retinal Regeneration, RIKEN Center for Developmental Biology, Kobe, Japan
| |
Collapse
|
16
|
Sonntag KC, Song B, Lee N, Jung JH, Cha Y, Leblanc P, Neff C, Kong SW, Carter BS, Schweitzer J, Kim KS. Pluripotent stem cell-based therapy for Parkinson's disease: Current status and future prospects. Prog Neurobiol 2018; 168:1-20. [PMID: 29653250 PMCID: PMC6077089 DOI: 10.1016/j.pneurobio.2018.04.005] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 03/13/2018] [Accepted: 04/05/2018] [Indexed: 12/11/2022]
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative disorders, which affects about 0.3% of the general population. As the population in the developed world ages, this creates an escalating burden on society both in economic terms and in quality of life for these patients and for the families that support them. Although currently available pharmacological or surgical treatments may significantly improve the quality of life of many patients with PD, these are symptomatic treatments that do not slow or stop the progressive course of the disease. Because motor impairments in PD largely result from loss of midbrain dopamine neurons in the substantia nigra pars compacta, PD has long been considered to be one of the most promising target diseases for cell-based therapy. Indeed, numerous clinical and preclinical studies using fetal cell transplantation have provided proof of concept that cell replacement therapy may be a viable therapeutic approach for PD. However, the use of human fetal cells as a standardized therapeutic regimen has been fraught with fundamental ethical, practical, and clinical issues, prompting scientists to explore alternative cell sources. Based on groundbreaking establishments of human embryonic stem cells and induced pluripotent stem cells, these human pluripotent stem cells have been the subject of extensive research, leading to tremendous advancement in our understanding of these novel classes of stem cells and promising great potential for regenerative medicine. In this review, we discuss the prospects and challenges of human pluripotent stem cell-based cell therapy for PD.
Collapse
Affiliation(s)
- Kai-C Sonntag
- Department of Psychiatry, McLean Hospital, Harvard Medical School, United States; Laboratory for Translational Research on Neurodegeneration, 115 Mill Street, Belmont, MA, 02478, United States; Program for Neuropsychiatric Research, 115 Mill Street, Belmont, MA, 02478, United States
| | - Bin Song
- Department of Psychiatry, McLean Hospital, Harvard Medical School, United States; Molecular Neurobiology Laboratory, Program in Neuroscience and Harvard Stem Cell Institute, McLean Hospital, Harvard Medical School, 115 Mill Street, Belmont, MA, 02478, United States
| | - Nayeon Lee
- Department of Psychiatry, McLean Hospital, Harvard Medical School, United States; Molecular Neurobiology Laboratory, Program in Neuroscience and Harvard Stem Cell Institute, McLean Hospital, Harvard Medical School, 115 Mill Street, Belmont, MA, 02478, United States
| | - Jin Hyuk Jung
- Department of Psychiatry, McLean Hospital, Harvard Medical School, United States; Molecular Neurobiology Laboratory, Program in Neuroscience and Harvard Stem Cell Institute, McLean Hospital, Harvard Medical School, 115 Mill Street, Belmont, MA, 02478, United States
| | - Young Cha
- Department of Psychiatry, McLean Hospital, Harvard Medical School, United States; Molecular Neurobiology Laboratory, Program in Neuroscience and Harvard Stem Cell Institute, McLean Hospital, Harvard Medical School, 115 Mill Street, Belmont, MA, 02478, United States
| | - Pierre Leblanc
- Department of Psychiatry, McLean Hospital, Harvard Medical School, United States; Molecular Neurobiology Laboratory, Program in Neuroscience and Harvard Stem Cell Institute, McLean Hospital, Harvard Medical School, 115 Mill Street, Belmont, MA, 02478, United States
| | - Carolyn Neff
- Kaiser Permanente Medical Group, Irvine, CA, 92618, United States
| | - Sek Won Kong
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, United States; Computational Health Informatics Program, Boston Children's Hospital, Boston, MA, 02115, United States
| | - Bob S Carter
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, 02114, United States
| | - Jeffrey Schweitzer
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, 02114, United States.
| | - Kwang-Soo Kim
- Department of Psychiatry, McLean Hospital, Harvard Medical School, United States; Molecular Neurobiology Laboratory, Program in Neuroscience and Harvard Stem Cell Institute, McLean Hospital, Harvard Medical School, 115 Mill Street, Belmont, MA, 02478, United States.
| |
Collapse
|
17
|
Zheng W, Li Q, Zhao C, Da Y, Zhang HL, Chen Z. Differentiation of Glial Cells From hiPSCs: Potential Applications in Neurological Diseases and Cell Replacement Therapy. Front Cell Neurosci 2018; 12:239. [PMID: 30140204 PMCID: PMC6094089 DOI: 10.3389/fncel.2018.00239] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 07/17/2018] [Indexed: 12/20/2022] Open
Abstract
Glial cells are the most abundant cell type in the central nervous system (CNS) and play essential roles in maintaining brain homeostasis, forming myelin, and providing support and protection for neurons, etc. Over the past decade, significant progress has been made in the reprogramming field. Given the limited accessibility of human glial cells, in vitro differentiation of human induced pluripotent stem cells (hiPSCs) into glia may provide not only a valuable research tool for a better understanding of the functions of glia in the CNS but also a potential cellular source for clinical therapeutic purposes. In this review, we will summarize up-to-date novel strategies for the committed differentiation into the three major glial cell types, i.e., astrocyte, oligodendrocyte, and microglia, from hiPSCs, focusing on the non-neuronal cell effects on the pathology of some representative neurological diseases. Furthermore, the application of hiPSC-derived glial cells in neurological disease modeling will be discussed, so as to gain further insights into the development of new therapeutic targets for treatment of neurological disorders.
Collapse
Affiliation(s)
- Wei Zheng
- Cell Therapy Center, Xuanwu Hospital, Capital Medical University, Beijing, China.,Key Laboratory of Neurodegeneration, Ministry of Education, Beijing, China
| | - Qian Li
- Cell Therapy Center, Xuanwu Hospital, Capital Medical University, Beijing, China.,Key Laboratory of Neurodegeneration, Ministry of Education, Beijing, China
| | - Chao Zhao
- Department of Clinical Neurosciences, Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Yuwei Da
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Hong-Liang Zhang
- Department of Life Sciences, National Natural Science Foundation of China, Beijing, China
| | - Zhiguo Chen
- Cell Therapy Center, Xuanwu Hospital, Capital Medical University, Beijing, China.,Key Laboratory of Neurodegeneration, Ministry of Education, Beijing, China.,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China
| |
Collapse
|
18
|
AAVvector-mediated in vivo reprogramming into pluripotency. Nat Commun 2018; 9:2651. [PMID: 29985406 PMCID: PMC6037684 DOI: 10.1038/s41467-018-05059-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Accepted: 05/14/2018] [Indexed: 12/19/2022] Open
Abstract
In vivo reprogramming of somatic cells into induced pluripotent stem cells (iPSC) holds vast potential for basic research and regenerative medicine. However, it remains hampered by a need for vectors to express reprogramming factors (Oct-3/4, Klf4, Sox2, c-Myc; OKSM) in selected organs. Here, we report OKSM delivery vectors based on pseudotyped Adeno-associated virus (AAV). Using the AAV-DJ capsid, we could robustly reprogram mouse embryonic fibroblasts with low vector doses. Swapping to AAV8 permitted to efficiently reprogram somatic cells in adult mice by intravenous vector delivery, evidenced by hepatic or extra-hepatic teratomas and iPSC in the blood. Notably, we accomplished full in vivo reprogramming without c-Myc. Most iPSC generated in vitro or in vivo showed transcriptionally silent, intronic or intergenic vector integration, likely reflecting the increased host genome accessibility during reprogramming. Our approach crucially advances in vivo reprogramming technology, and concurrently facilitates investigations into the mechanisms and consequences of AAV persistence. In vivo reprogramming of somatic cells is hampered by the need for vectors to express the OKSM factors in selected organs. Here the authors report new AAV-based vectors capable of in vivo reprogramming at low doses.
Collapse
|
19
|
Manipulating cell fate while confronting reproducibility concerns. Biochem Pharmacol 2018; 151:144-156. [DOI: 10.1016/j.bcp.2018.01.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 01/04/2018] [Indexed: 12/13/2022]
|
20
|
Deconstructing the pluripotency gene regulatory network. Nat Cell Biol 2018; 20:382-392. [PMID: 29593328 DOI: 10.1038/s41556-018-0067-6] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 02/16/2018] [Indexed: 12/22/2022]
Abstract
Pluripotent stem cells can be isolated from embryos or derived by reprogramming. Pluripotency is stabilized by an interconnected network of pluripotency genes that cooperatively regulate gene expression. Here we describe the molecular principles of pluripotency gene function and highlight post-transcriptional controls, particularly those induced by RNA-binding proteins and alternative splicing, as an important regulatory layer of pluripotency. We also discuss heterogeneity in pluripotency regulation, alternative pluripotency states and future directions of pluripotent stem cell research.
Collapse
|
21
|
The genes involved in asthma with the treatment of human embryonic stem cell-derived mesenchymal stem cells. Mol Immunol 2018; 95:47-55. [PMID: 29407576 DOI: 10.1016/j.molimm.2018.01.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 12/29/2017] [Accepted: 01/24/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND Asthma is affecting more than 300 million people worldwide, which represents the most common chronic disease among children. We previously found that mesenchymal stem cells (MSCs) derived from induced pluripotent stem cells (iPSCs) modulated the immune response on Th2-mediated asthma in vivo and in vitro. This study further evaluated the immunomodulatory effects of MSCs from human embryonic stem cells (hESCs) on asthma. METHODS Multipotent hESC-MSCs were obtained using a feeder-free method. The hESC-MSCs were analysed for the expression of stem cell surface markers by flow cytometry, their differentiation potentials were analysed using in vitro trilineage differentiation methods hESC-MSCs were transplanted into the murine model with ovalbumin (OVA)-induced airway allergic inflammation. The expression levels of allergic related genes were measured by the mRNA PCR arrays. RESULTS The hESC-MSCs expressed classical MSC markers and held the capability of differentiation into multiple mesoderm-type cell lineages. hESC-MSCs were able to suppress allergic inflammation by modulating Th2 cells and eosinophils in the mice, and reversed the reduction of regulatory T cells. By using PCR array, 5 mRNAs- chemokine (C-C motif) ligand 11 (Ccl11), Ccl24, interleukin13 (Il13), Il33 and eosinophil-associated, ribonuclease A family, member 11 (Ear11) were identified the most relevant in murine airway allergic inflammation and hESC-MSCs treatment. CONCLUSIONS The therapeutic effects of hESC-MSCs were identified in the murine model of airway allergic inflammation with key mRNAs involved. This study will provide a better understanding regarding the mechanisms underlying hESC-MSCs therapeutic application in airway allergic inflammation.
Collapse
|
22
|
Modeling the Role of Wnt Signaling in Human and Drosophila Stem Cells. Genes (Basel) 2018; 9:genes9020101. [PMID: 29462894 PMCID: PMC5852597 DOI: 10.3390/genes9020101] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 02/08/2018] [Accepted: 02/09/2018] [Indexed: 12/15/2022] Open
Abstract
The discovery of induced pluripotent stem (iPS) cells, barely more than a decade ago, dramatically transformed the study of stem cells and introduced a completely new way to approach many human health concerns. Although advances have pushed the field forward, human application remains some years away, in part due to the need for an in-depth mechanistic understanding. The role of Wnts in stem cells predates the discovery of iPS cells with Wnts established as major pluripotency promoting factors. Most work to date has been done using mouse and tissue culture models and few attempts have been made in other model organisms, but the recent combination of clustered regularly interspaced short palindromic repeats (CRISPR) gene editing with iPS cell technology provides a perfect avenue for exploring iPS cells in model organisms. Drosophila is an ideal organism for such studies, but fly iPS cells have not yet been made. In this opinion article, we draw parallels between Wnt signaling in human and Drosophila stem cell systems, propose ways to obtain Drosophila iPS cells, and suggest ways to exploit the versatility of the Drosophila system for future stem cell studies.
Collapse
|
23
|
Ribeiro D, Andersson EM, Heath N, Persson-Kry A, Collins R, Hicks R, Dekker N, Forslöw A. Human pancreatic islet-derived extracellular vesicles modulate insulin expression in 3D-differentiating iPSC clusters. PLoS One 2017; 12:e0187665. [PMID: 29117231 PMCID: PMC5678888 DOI: 10.1371/journal.pone.0187665] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 10/24/2017] [Indexed: 01/02/2023] Open
Abstract
It has been suggested that extracellular vesicles (EVs) can mediate crosstalk between hormones and metabolites within pancreatic tissue. However, the possible effect of pancreatic EVs on stem cell differentiation into pancreatic lineages remains unknown. Herein, human islet-derived EVs (h-Islet-EVs) were isolated, characterized and subsequently added to human induced pluripotent stem cell (iPSC) clusters during pancreatic differentiation. The h-islet-EVs had a mean size of 117±7 nm and showed positive expression of CD63 and CD81 EV markers as measured by ELISA. The presence of key pancreatic transcription factor mRNA, such as NGN3, MAFA and PDX1, and pancreatic hormone proteins such as C-peptide and glucagon, were confirmed in h-Islet-EVs. iPSC clusters were differentiated in suspension and at the end stages of the differentiation protocol, the mRNA expression of the main pancreatic transcription factors and pancreatic hormones was increased. H-Islet-EVs were supplemented to the iPSC clusters in the later stages of differentiation. It was observed that h-Islet-EVs were able to up-regulate the intracellular levels of C-peptide in iPSC clusters in a concentration-dependent manner. The effect of h-Islet-EVs on the differentiation of iPSC clusters cultured in 3D-collagen hydrogels was also assessed. Although increased mRNA expression for pancreatic markers was observed when culturing the iPSC clusters in 3D-collagen hydrogels, delivery of EVs did not affect the insulin or C-peptide intracellular content. Our results provide new information on the role of h-Islet-EVs in the regulation of insulin expression in differentiating iPSC clusters, and are highly relevant for pancreatic tissue engineering applications.
Collapse
Affiliation(s)
- Diana Ribeiro
- Discovery Sciences, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden.,Department of Biology and Bioengineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Eva-Marie Andersson
- Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Nikki Heath
- Discovery Sciences, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Alderley Park, Macclesfield, United Kingdom
| | - Anette Persson-Kry
- Discovery Sciences, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Richard Collins
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - Ryan Hicks
- Discovery Sciences, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Niek Dekker
- Discovery Sciences, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Anna Forslöw
- Discovery Sciences, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
24
|
Direct conversion from skin fibroblasts to functional dopaminergic neurons for biomedical application. BIOMEDICAL DERMATOLOGY 2017. [DOI: 10.1186/s41702-017-0004-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
25
|
Induced Pluripotent Stem Cell Neuronal Models for the Study of Autophagy Pathways in Human Neurodegenerative Disease. Cells 2017; 6:cells6030024. [PMID: 28800101 PMCID: PMC5617970 DOI: 10.3390/cells6030024] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 08/08/2017] [Accepted: 08/09/2017] [Indexed: 02/06/2023] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) are invaluable tools for research into the causes of diverse human diseases, and have enormous potential in the emerging field of regenerative medicine. Our ability to reprogramme patient cells to become hiPSCs, and to subsequently direct their differentiation towards those classes of neurons that are vulnerable to stress, is revealing how genetic mutations cause changes at the molecular level that drive the complex pathogeneses of human neurodegenerative diseases. Autophagy dysregulation is considered to be a major contributor in neural decline during the onset and progression of many human neurodegenerative diseases, meaning that a better understanding of the control of non-selective and selective autophagy pathways (including mitophagy) in disease-affected classes of neurons is needed. To achieve this, it is essential that the methodologies commonly used to study autophagy regulation under basal and stressed conditions in standard cell-line models are accurately applied when using hiPSC-derived neuronal cultures. Here, we discuss the roles and control of autophagy in human stem cells, and how autophagy contributes to neural differentiation in vitro. We also describe how autophagy-monitoring tools can be applied to hiPSC-derived neurons for the study of human neurodegenerative disease in vitro.
Collapse
|
26
|
Abstract
Pluripotency is a state that exists transiently in the early embryo and, remarkably, can be recapitulated in vitro by deriving embryonic stem cells or by reprogramming somatic cells to become induced pluripotent stem cells. The state of pluripotency, which is stabilized by an interconnected network of pluripotency-associated genes, integrates external signals and exerts control over the decision between self-renewal and differentiation at the transcriptional, post-transcriptional and epigenetic levels. Recent evidence of alternative pluripotency states indicates the regulatory flexibility of this network. Insights into the underlying principles of the pluripotency network may provide unprecedented opportunities for studying development and for regenerative medicine.
Collapse
|