1
|
Kang B, Jeong E, Han SY, Heo JH, Lee Y, Choi S, Choi Y, Kang D, Hwang YH, Lee J, Seo JH, Kim J, Jeong I, Kim E, Lee J, Kim DE, Park JU, Cho SR, Jin Y, Cho SW, Lee H. Acoustofluidic bioassembly induced morphogenesis for therapeutic tissue fabrication. Nat Commun 2025; 16:4174. [PMID: 40324975 PMCID: PMC12053659 DOI: 10.1038/s41467-025-59026-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 04/07/2025] [Indexed: 05/07/2025] Open
Abstract
To build in vitro tissues for therapeutic applications, it is essential to replicate the spatial distribution of cells that occurs during morphogenesis in vivo. However, it remains technically challenging to simultaneously regulate the geometric alignment and aggregation of cells during tissue fabrication. Here, we introduce the acoustofluidic bioassembly induced morphogenesis, which is the combination of precise arrangement of cells by the mechanical forces produced by acoustofluidic cues, and the morphological and functional changes of cells in the following in vitro and in vivo cultures. The acoustofluidic bioassembly can be used to create tissues with regulated nano-, micro-, and macro-structures. We demonstrate that the neuromuscular tissue fabricated with the acoustofluidic bioassembly exhibits enhanced contraction dynamics, electrophysiology, and therapeutic efficacy. The potential of the acoustofluidic bioassembly as an in situ application is demonstrated by fabricating artificial tissues at the defect sites of living tissues. The acoustofluidic bioassembly induced morphogenesis can provide a pioneering platform to fabricate tissues for biomedical applications.
Collapse
Affiliation(s)
- Byungjun Kang
- School of Mechanical Engineering, Yonsei University, Seoul, 03722, Republic of Korea
| | - Eunseon Jeong
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Seung Yeop Han
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
- Department of Biomaterials Science and Engineering, Yonsei University, Seoul, 03722, Republic of Korea
| | - Jeong Hyun Heo
- Department of Physiology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Yunam Lee
- School of Mechanical Engineering, Yonsei University, Seoul, 03722, Republic of Korea
| | - Suah Choi
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Yunjung Choi
- School of Mechanical Engineering, Yonsei University, Seoul, 03722, Republic of Korea
| | - Donyoung Kang
- School of Mechanical Engineering, Yonsei University, Seoul, 03722, Republic of Korea
| | - Youn-Hoo Hwang
- School of Mechanical Engineering, Yonsei University, Seoul, 03722, Republic of Korea
| | - Jiin Lee
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Jung Hwa Seo
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Jinyoung Kim
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Graduate Program of Biomedical Engineering, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Inhea Jeong
- Department of Materials Science and Engineering, Yonsei University, Seoul, 03722, Republic of Korea
| | - Enji Kim
- Department of Materials Science and Engineering, Yonsei University, Seoul, 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, 03722, Republic of Korea
| | - Juyoung Lee
- School of Mechanical Engineering, Yonsei University, Seoul, 03722, Republic of Korea
| | - Dae-Eun Kim
- School of Mechanical Engineering, Yonsei University, Seoul, 03722, Republic of Korea
| | - Jang-Ung Park
- Department of Materials Science and Engineering, Yonsei University, Seoul, 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, 03722, Republic of Korea
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul, 03722, Republic of Korea
| | - Sung-Rae Cho
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Graduate Program of Biomedical Engineering, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Rehabilitation Institute of Neuromuscular Disease, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Department of Rehabilitation Medicine, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Brain research institute, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Yoonhee Jin
- Department of Physiology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea.
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, 03722, Republic of Korea.
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul, 03722, Republic of Korea.
| | - Hyungsuk Lee
- School of Mechanical Engineering, Yonsei University, Seoul, 03722, Republic of Korea.
| |
Collapse
|
2
|
Hossain MK, Kim HR, Chae HJ. Aging phenotype in AD brain organoids: Track to success and challenges. Ageing Res Rev 2024; 96:102256. [PMID: 38460555 DOI: 10.1016/j.arr.2024.102256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 02/19/2024] [Accepted: 02/27/2024] [Indexed: 03/11/2024]
Abstract
Alzheimer's disease (AD) poses a complex challenge, with abnormal protein accumulation in the brain causing memory loss and cognitive decline. Traditional models fall short in AD research, prompting interest in 3D brain organoids (BOs) from human stem cells. These findings hold promise for unveiling the mechanisms of AD, especially in relation to aging. However, an understanding of the aging impact of AD remains elusive. BOs offer insight but face challenges. This review delves into the role of BOs in deciphering aging-related AD and acknowledges limitations. Strategies to enhance BOs for accurate aging modeling in AD brains are suggested. Strengthened by molecular advancements, BOs have the potential to uncover the aging phenotype, advancing AD research.
Collapse
Affiliation(s)
| | - Hyung-Ryong Kim
- Department of Pharmacology, College of Dentistry, Jeonbuk National University, Jeonju 54896, Republic of Korea.
| | - Han Jung Chae
- School of Pharmacy, Jeonbuk National University, Jeonju 54896, Republic of Korea.
| |
Collapse
|
3
|
Pinton L, Khedr M, Lionello VM, Sarcar S, Maffioletti SM, Dastidar S, Negroni E, Choi S, Khokhar N, Bigot A, Counsell JR, Bernardo AS, Zammit PS, Tedesco FS. 3D human induced pluripotent stem cell-derived bioengineered skeletal muscles for tissue, disease and therapy modeling. Nat Protoc 2023; 18:1337-1376. [PMID: 36792780 DOI: 10.1038/s41596-022-00790-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 11/02/2022] [Indexed: 02/17/2023]
Abstract
Skeletal muscle is a complex tissue composed of multinucleated myofibers responsible for force generation that are supported by multiple cell types. Many severe and lethal disorders affect skeletal muscle; therefore, engineering models to reproduce such cellular complexity and function are instrumental for investigating muscle pathophysiology and developing therapies. Here, we detail the modular 3D bioengineering of multilineage skeletal muscles from human induced pluripotent stem cells, which are first differentiated into myogenic, neural and vascular progenitor cells and then combined within 3D hydrogels under tension to generate an aligned myofiber scaffold containing vascular networks and motor neurons. 3D bioengineered muscles recapitulate morphological and functional features of human skeletal muscle, including establishment of a pool of cells expressing muscle stem cell markers. Importantly, bioengineered muscles provide a high-fidelity platform to study muscle pathology, such as emergence of dysmorphic nuclei in muscular dystrophies caused by mutant lamins. The protocol is easy to follow for operators with cell culture experience and takes between 9 and 30 d, depending on the number of cell lineages in the construct. We also provide examples of applications of this advanced platform for testing gene and cell therapies in vitro, as well as for in vivo studies, providing proof of principle of its potential as a tool to develop next-generation neuromuscular or musculoskeletal therapies.
Collapse
Affiliation(s)
- Luca Pinton
- Department of Cell and Developmental Biology, University College London, London, UK
- The Francis Crick Institute, London, UK
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, UK
| | - Moustafa Khedr
- Department of Cell and Developmental Biology, University College London, London, UK
- The Francis Crick Institute, London, UK
| | - Valentina M Lionello
- Department of Cell and Developmental Biology, University College London, London, UK
- The Francis Crick Institute, London, UK
| | - Shilpita Sarcar
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Sara M Maffioletti
- Department of Cell and Developmental Biology, University College London, London, UK
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), Milan, Italy
| | - Sumitava Dastidar
- Department of Cell and Developmental Biology, University College London, London, UK
- The Francis Crick Institute, London, UK
| | - Elisa Negroni
- Department of Cell and Developmental Biology, University College London, London, UK
- Center for Research in Myology UMRS974, Sorbonne Université, INSERM, Myology Institute AIM, Paris, France
| | - SungWoo Choi
- Department of Cell and Developmental Biology, University College London, London, UK
- The Francis Crick Institute, London, UK
| | - Noreen Khokhar
- Department of Cell and Developmental Biology, University College London, London, UK
- The Francis Crick Institute, London, UK
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, UK
| | - Anne Bigot
- Center for Research in Myology UMRS974, Sorbonne Université, INSERM, Myology Institute AIM, Paris, France
| | - John R Counsell
- UCL Division of Surgery and Interventional Science, Royal Free Hospital, London, UK
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health and Great Ormond Street Hospital for Children, London, UK
| | - Andreia Sofia Bernardo
- The Francis Crick Institute, London, UK
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Peter S Zammit
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, UK
| | - Francesco Saverio Tedesco
- Department of Cell and Developmental Biology, University College London, London, UK.
- The Francis Crick Institute, London, UK.
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health and Great Ormond Street Hospital for Children, London, UK.
| |
Collapse
|
4
|
Dwivedi S, Choudhary P, Gupta A, Singh S. Therapeutical growth in oligodendroglial fate induction via transdifferentiation of stem cells for neuroregenerative therapy. Biochimie 2023; 211:35-56. [PMID: 36842627 DOI: 10.1016/j.biochi.2023.02.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 12/20/2022] [Accepted: 02/21/2023] [Indexed: 02/27/2023]
Abstract
The merits of stem cell therapy and research are undisputed due to their widespread usage in the treatment of neurodegenerative diseases and demyelinating disorders. Cell replacement therapy especially revolves around stem cells and their induction into different cell lineages both adult and progenitor - belonging to each germ layer, prior to transplantation or disease modeling studies. The nervous system is abundant in glial cells and among these are oligodendrocytes capable of myelinating new-born neurons and remyelination of axons with lost or damaged myelin sheath. But demyelinating diseases generate tremendous deficit between myelin loss and recovery. To compensate for this loss, analyze the defects in remyelination mechanisms as well as to trigger full recovery in such patients mesenchymal stem cells (MSCs) have been induced to transdifferentiate into oligodendrocytes. But such experiments are riddled with problems like prolonged, tenuous and complicated protocols that stretch longer than the time taken for the spread of demyelination-associated after-effects. This review delves into such protocols and the combinations of different molecules and factors that have been recruited to derive bona fide oligodendrocytes from in vitro differentiation of embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs) and MSCs with special focus on MSC-derived oligodendrocytes.
Collapse
Affiliation(s)
- Shrey Dwivedi
- Department of Applied Sciences, Indian Institute of Information Technology, Allahabad, U.P., India
| | - Princy Choudhary
- Department of Applied Sciences, Indian Institute of Information Technology, Allahabad, U.P., India
| | - Ayushi Gupta
- Department of Applied Sciences, Indian Institute of Information Technology, Allahabad, U.P., India
| | - Sangeeta Singh
- Department of Applied Sciences, Indian Institute of Information Technology, Allahabad, U.P., India.
| |
Collapse
|
5
|
Yan L, Rodríguez-delaRosa A, Pourquié O. Human muscle production in vitro from pluripotent stem cells: Basic and clinical applications. Semin Cell Dev Biol 2021; 119:39-48. [PMID: 33941447 PMCID: PMC8530835 DOI: 10.1016/j.semcdb.2021.04.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 04/19/2021] [Indexed: 10/21/2022]
Abstract
Human pluripotent stem cells (PSCs), which have the capacity to self-renew and differentiate into multiple cell types, offer tremendous therapeutic potential and invaluable flexibility as research tools. Recently, remarkable progress has been made in directing myogenic differentiation of human PSCs. The differentiation strategies, which were inspired by our knowledge of myogenesis in vivo, have provided an important platform for the study of human muscle development and modeling of muscular diseases, as well as a promising source of cells for cell therapy to treat muscular dystrophies. In this review, we summarize the current state of skeletal muscle generation from human PSCs, including transgene-based and transgene-free differentiation protocols, and 3D muscle tissue production through bioengineering approaches. We also highlight their basic and clinical applications, which facilitate the study of human muscle biology and deliver new hope for muscular disease treatment.
Collapse
Affiliation(s)
- Lu Yan
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA; Department of Genetics, Harvard Medical School, Boston, MA, USA; Harvard Stem Cell Institute, Boston, MA, USA
| | - Alejandra Rodríguez-delaRosa
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA; Department of Genetics, Harvard Medical School, Boston, MA, USA; Harvard Stem Cell Institute, Boston, MA, USA
| | - Olivier Pourquié
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA; Department of Genetics, Harvard Medical School, Boston, MA, USA; Harvard Stem Cell Institute, Boston, MA, USA.
| |
Collapse
|
6
|
Liang KX, Vatne GH, Kristiansen CK, Ievglevskyi O, Kondratskaya E, Glover JC, Chen A, Sullivan GJ, Bindoff LA. N-acetylcysteine amide ameliorates mitochondrial dysfunction and reduces oxidative stress in hiPSC-derived dopaminergic neurons with POLG mutation. Exp Neurol 2020; 337:113536. [PMID: 33264635 DOI: 10.1016/j.expneurol.2020.113536] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 11/22/2020] [Accepted: 11/22/2020] [Indexed: 01/03/2023]
Abstract
The inability to reliably replicate mitochondrial DNA (mtDNA) by mitochondrial DNA polymerase gamma (POLG) leads to a subset of common mitochondrial diseases associated with neuronal death and depletion of neuronal mtDNA. Defining disease mechanisms in neurons remains difficult due to the limited access to human tissue. Using human induced pluripotent stem cells (hiPSCs), we generated functional dopaminergic (DA) neurons showing positive expression of dopaminergic markers TH and DAT, mature neuronal marker MAP2 and functional synaptic markers synaptophysin and PSD-95. These DA neurons were electrophysiologically characterized, and exhibited inward Na + currents, overshooting action potentials and spontaneous postsynaptic currents (sPSCs). POLG patient-specific DA neurons (POLG-DA neurons) manifested a phenotype that replicated the molecular and biochemical changes found in patient post-mortem brain samples namely loss of complex I and depletion of mtDNA. Compared to disease-free hiPSC-derived DA neurons, POLG-DA neurons exhibited loss of mitochondrial membrane potential, loss of complex I and loss of mtDNA and TFAM expression. POLG driven mitochondrial dysfunction also led to neuronal ROS overproduction and increased cellular senescence. This deficit was selectively rescued by treatment with N-acetylcysteine amide (NACA). In conclusion, our study illustrates the promise of hiPSC technology for assessing pathogenetic mechanisms associated with POLG disease, and that NACA can be a promising potential therapy for mitochondrial diseases such as those caused by POLG mutation.
Collapse
Affiliation(s)
- Kristina Xiao Liang
- Neuro-SysMed, Center of Excellence for Clinical Research in Neurological Diseases, Department of Neurology, Haukeland University Hospital, Jonas Lies vei 87, P. O. Box 7804, 5021 Bergen, Norway; Department of Clinical Medicine (K1), University of Bergen, Jonas Lies vei 87, P. O. Box 7804, 5021 Bergen, Norway.
| | - Guro Helén Vatne
- Department of Clinical Medicine (K1), University of Bergen, Jonas Lies vei 87, P. O. Box 7804, 5021 Bergen, Norway
| | - Cecilie Katrin Kristiansen
- Department of Clinical Medicine (K1), University of Bergen, Jonas Lies vei 87, P. O. Box 7804, 5021 Bergen, Norway
| | - Oleksandr Ievglevskyi
- The Intervention Centre, Oslo University Hospital, P. O. Box 4950, Nydalen, 0424 Oslo, Norway; Laboratory of Neural Development and Optical Recording (NDEVOR), Division of Physiology, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, P. O. Box 1103, Blindern, 0317 Oslo, Norway
| | - Elena Kondratskaya
- Laboratory of Neural Development and Optical Recording (NDEVOR), Division of Physiology, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, P. O. Box 1103, Blindern, 0317 Oslo, Norway
| | - Joel C Glover
- Laboratory of Neural Development and Optical Recording (NDEVOR), Division of Physiology, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, P. O. Box 1103, Blindern, 0317 Oslo, Norway; Norwegian Center for Stem Cell Research, Department of Immunology and Transfusion Medicine, Oslo University Hospital, P. O. Box 4950, Nydalen, 0424 Oslo, Norway
| | - Anbin Chen
- Department of Clinical Medicine (K1), University of Bergen, Jonas Lies vei 87, P. O. Box 7804, 5021 Bergen, Norway; Department of Neurosurgery, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Jinan 250012, Shandong Province, China; Shandong Key Laboratory of Brain Function Remodeling, Shandong University, 107 Wenhua Xi Road, Jinan 250012, Shandong Province, China
| | - Gareth John Sullivan
- Norwegian Center for Stem Cell Research, Department of Immunology and Transfusion Medicine, Oslo University Hospital, P. O. Box 4950, Nydalen, 0424 Oslo, Norway; Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, P. O. Box 1105, Blindern, 0317 Oslo, Norway; Institute of Immunology, Oslo University Hospital, PO Box 4950, 0424 Oslo, Norway; Hybrid Technology Hub - Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, P. O. Box 1110, Blindern, 0317 Oslo, Norway; Department of Pediatric Research, Oslo University Hospital, P. O. Box 4950, Nydalen, 0424 Oslo, Norway
| | - Laurence A Bindoff
- Neuro-SysMed, Center of Excellence for Clinical Research in Neurological Diseases, Department of Neurology, Haukeland University Hospital, Jonas Lies vei 87, P. O. Box 7804, 5021 Bergen, Norway; Department of Clinical Medicine (K1), University of Bergen, Jonas Lies vei 87, P. O. Box 7804, 5021 Bergen, Norway.
| |
Collapse
|
7
|
Todorova D, Zhang Y, Chen Q, Liu J, He J, Fu X, Xu Y. hESC-derived immune suppressive dendritic cells induce immune tolerance of parental hESC-derived allografts. EBioMedicine 2020; 62:103120. [PMID: 33242828 PMCID: PMC7695963 DOI: 10.1016/j.ebiom.2020.103120] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 09/28/2020] [Accepted: 10/26/2020] [Indexed: 12/21/2022] Open
Abstract
Background With their inherent capability of unlimited self-renewal and unique potential to differentiate into functional cells of the three germ layers, human embryonic stem cells (hESCs) hold great potential in regenerative medicine. A major challenge in the application of hESC-based cell therapy is the allogeneic immune rejection of hESC-derived allografts. Methods We derived dendritic cell-like cells (DCLs) from wild type and CTLA4-Ig/PD-L1 knock-in hESCs, denoted WT DCLs and CP DCLs. The expression of DC-related genes and surface molecules was evaluated, as well as their DCL capacity to stimulate allogeneic T cells and induce regulatory T (Treg) cells in vitro. Using an immune system humanized mouse model, we investigated whether the adoptive transfer of CP DCLs can induce long-term immune tolerance of parental hESC-derived smooth muscle and cardiomyocyte allografts. Findings CP DCLs can maintain immune suppressive properties after robust inflammatory stimulation and induce Treg cells. While CP DCLs survive transiently in vivo, they induce long-term immune tolerance of parental hESC-derived allografts. Interpretation This strategy does not cause systemic immune suppression but induces immune tolerance specific for DCL-specific HLAs, and thus it presents a safe and effective approach to induce immune tolerance of allografts derived from any clinically approved hESC line. Funding NSFC, leading talents of Guangdong Province Program (No. 00201516), Key R&D Program of Guangdong Province (2019B020235003), Science and Technology Innovation Committee of Shenzhen Municipality (JCYJ20180504170301309), National High-tech R&D Program (863 Program No. 2015AA020310), Shenzhen “Sanming” Project of Medicine (SZSM201602102), Development and Reform Commission of Shenzhen Municipality (S2016004730009), CIRM (DISC2–10559).
Collapse
Affiliation(s)
- Dilyana Todorova
- Division of Biological Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Yue Zhang
- Guangzhou University of Chinese Medicine, Second Clinical Medical College, 232 Waihuan Road E, Guangzhou, Guangdong 510006, China
| | - Qu Chen
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong 518033, China
| | - Jingfeng Liu
- Division of Biological Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Jingjin He
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong 518033, China
| | - Xuemei Fu
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong 518033, China; Shenzhen Children's Hospital, Shenzhen 518026, China..
| | - Yang Xu
- Division of Biological Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Guangzhou University of Chinese Medicine, Second Clinical Medical College, 232 Waihuan Road E, Guangzhou, Guangdong 510006, China; The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong 518033, China.
| |
Collapse
|
8
|
Liang KX, Kristiansen CK, Mostafavi S, Vatne GH, Zantingh GA, Kianian A, Tzoulis C, Høyland LE, Ziegler M, Perez RM, Furriol J, Zhang Z, Balafkan N, Hong Y, Siller R, Sullivan GJ, Bindoff LA. Disease-specific phenotypes in iPSC-derived neural stem cells with POLG mutations. EMBO Mol Med 2020; 12:e12146. [PMID: 32840960 PMCID: PMC7539330 DOI: 10.15252/emmm.202012146] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 07/30/2020] [Accepted: 07/31/2020] [Indexed: 12/12/2022] Open
Abstract
Mutations in POLG disrupt mtDNA replication and cause devastating diseases often with neurological phenotypes. Defining disease mechanisms has been hampered by limited access to human tissues, particularly neurons. Using patient cells carrying POLG mutations, we generated iPSCs and then neural stem cells. These neural precursors manifested a phenotype that faithfully replicated the molecular and biochemical changes found in patient post‐mortem brain tissue. We confirmed the same loss of mtDNA and complex I in dopaminergic neurons generated from the same stem cells. POLG‐driven mitochondrial dysfunction led to neuronal ROS overproduction and increased cellular senescence. Loss of complex I was associated with disturbed NAD+ metabolism with increased UCP2 expression and reduced phosphorylated SirT1. In cells with compound heterozygous POLG mutations, we also found activated mitophagy via the BNIP3 pathway. Our studies are the first that show it is possible to recapitulate the neuronal molecular and biochemical defects associated with POLG mutation in a human stem cell model. Further, our data provide insight into how mitochondrial dysfunction and mtDNA alterations influence cellular fate determining processes.
Collapse
Affiliation(s)
- Kristina Xiao Liang
- Neuro-SysMed, Center of Excellence for Clinical Research in Neurological Diseases, Haukeland University Hospital, Bergen, Norway.,Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | | | - Sepideh Mostafavi
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Guro Helén Vatne
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Gina Alien Zantingh
- Leiden University Medical Centre, Leiden University, Leiden, The Netherlands
| | - Atefeh Kianian
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Charalampos Tzoulis
- Neuro-SysMed, Center of Excellence for Clinical Research in Neurological Diseases, Haukeland University Hospital, Bergen, Norway.,Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | | | - Mathias Ziegler
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | | | - Jessica Furriol
- Department of Clinical Medicine, University of Bergen, Bergen, Norway.,Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Zhuoyuan Zhang
- State Key Laboratory of Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, China.,Department of Head and Neck Cancer Surgery, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Novin Balafkan
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Yu Hong
- Neuro-SysMed, Center of Excellence for Clinical Research in Neurological Diseases, Haukeland University Hospital, Bergen, Norway.,Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Richard Siller
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.,Norwegian Center for Stem Cell Research, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Gareth John Sullivan
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.,Norwegian Center for Stem Cell Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,Institute of Immunology, Oslo University Hospital, Oslo, Norway.,Hybrid Technology Hub - Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.,Department of Pediatric Research, Oslo University Hospital, Oslo, Norway
| | - Laurence A Bindoff
- Neuro-SysMed, Center of Excellence for Clinical Research in Neurological Diseases, Haukeland University Hospital, Bergen, Norway.,Department of Clinical Medicine, University of Bergen, Bergen, Norway
| |
Collapse
|
9
|
Sunny DE, Hammer E, Strempel S, Joseph C, Manchanda H, Ittermann T, Hübner S, Weiss FU, Völker U, Heckmann M. Nup133 and ERα mediate the differential effects of hyperoxia-induced damage in male and female OPCs. Mol Cell Pediatr 2020; 7:10. [PMID: 32844334 PMCID: PMC7447710 DOI: 10.1186/s40348-020-00102-8] [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: 04/16/2020] [Accepted: 08/02/2020] [Indexed: 02/07/2023] Open
Abstract
Background Hyperoxia is a well-known cause of cerebral white matter injury in preterm infants with male sex being an independent and critical risk factor for poor neurodevelopmental outcome. Sex is therefore being widely considered as one of the major decisive factors for prognosis and treatment of these infants. But unfortunately, we still lack a clear view of the molecular mechanisms that lead to such a profound difference. Hence, using mouse-derived primary oligodendrocyte progenitor cells (OPCs), we investigated the molecular factors and underlying mechanisms behind the differential response of male and female cells towards oxidative stress. Results We demonstrate that oxidative stress severely affects cellular functions related to energy metabolism, stress response, and maturation in the male-derived OPCs, whereas the female cells remain largely unaffected. CNPase protein level was found to decline following hyperoxia in male but not in female cells. This impairment of maturation was accompanied by the downregulation of nucleoporin and nuclear lamina proteins in the male cells. We identify Nup133 as a novel target protein affected by hyperoxia, whose inverse regulation may mediate this differential response in the male and female cells. Nup133 protein level declined following hyperoxia in male but not in female cells. We show that nuclear respiratory factor 1 (Nrf1) is a direct downstream target of Nup133 and that Nrf1 mRNA declines following hyperoxia in male but not in female cells. The female cells may be rendered resistant due to synergistic protection via the estrogen receptor alpha (ERα) which was upregulated following hyperoxia in female but not in male cells. Both Nup133 and ERα regulate mitochondrial function and oxidative stress response by transcriptional regulation of Nrf1. Conclusions These findings from a basic cell culture model establish prominent sex-based differences and suggest a novel mechanism involved in the differential response of OPCs towards oxidative stress. It conveys a strong message supporting the need to study how complex cellular processes are regulated differently in male and female brains during development and for a better understanding of how the brain copes up with different forms of stress after preterm birth.
Collapse
Affiliation(s)
- Donna Elizabeth Sunny
- Department of Neonatology and Pediatric Intensive Care, University of Medicine Greifswald, Ferdinand-Sauerbruchstrasse, 17475, Greifswald, Germany.
| | - Elke Hammer
- Department of Functional Genomics, University of Medicine Greifswald, Greifswald, Germany
| | | | - Christy Joseph
- Department of Pharmacology, Center of Drug Absorption and Transport (C_DAT), University of Medicine Greifswald, Greifswald, Germany
| | - Himanshu Manchanda
- Department of Bioinformatics, University of Medicine Greifswald, Greifswald, Germany
| | - Till Ittermann
- Institute for Community Medicine, University of Medicine Greifswald, Greifswald, Germany
| | - Stephanie Hübner
- Department of Neonatology and Pediatric Intensive Care, University of Medicine Greifswald, Ferdinand-Sauerbruchstrasse, 17475, Greifswald, Germany
| | - Frank Ulrich Weiss
- Department of Internal Medicine A, University of Medicine Greifswald, Greifswald, Germany
| | - Uwe Völker
- Department of Functional Genomics, University of Medicine Greifswald, Greifswald, Germany
| | - Matthias Heckmann
- Department of Neonatology and Pediatric Intensive Care, University of Medicine Greifswald, Ferdinand-Sauerbruchstrasse, 17475, Greifswald, Germany
| |
Collapse
|
10
|
Human Embryonic Stem Cell-Derived Neural Lineages as In Vitro Models for Screening the Neuroprotective Properties of Lignosus rhinocerus (Cooke) Ryvarden. BIOMED RESEARCH INTERNATIONAL 2019; 2019:3126376. [PMID: 33204680 PMCID: PMC7658738 DOI: 10.1155/2019/3126376] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Revised: 07/02/2019] [Accepted: 07/11/2019] [Indexed: 11/17/2022]
Abstract
In the biomedical field, there is growing interest in using human stem cell-derived neurons as in vitro models for pharmacological and toxicological screening of bioactive compounds extracted from natural products. Lignosus rhinocerus (Tiger Milk Mushroom) is used by indigenous communities in Malaysia as a traditional medicine to treat various diseases. The sclerotium of L. rhinocerus has been reported to have medicinal properties, including various bioactivities such as neuritogenic, anti-inflammatory, and anticancer effects. This study aims to investigate the neuroprotective activities of L. rhinocerus sclerotial extracts. Human embryonic stem cell (hESC)-derived neural lineages exposed to the synthetic glucocorticoid, dexamethasone (DEX), were used as the in vitro models. Excess glucocorticoids have been shown to adversely affect fetal brain development and impair differentiation of neural progenitor cells. Screening of different L. rhinocerus sclerotial extracts and DEX on the hESC-derived neural lineages was conducted using cell viability and neurite outgrowth assays. The neuroprotective effects of L. rhinocerus sclerotial extracts against DEX were further evaluated using apoptosis assays and Western blot analysis. Hot aqueous and methanol extracts of L. rhinocerus sclerotium promoted neurite outgrowth of hESC-derived neural stem cells (NSCs) with negligible cytotoxicity. Treatment with DEX decreased viability of NSCs by inducing apoptosis. Coincubation of L. rhinocerus methanol extract with DEX attenuated the DEX-induced apoptosis and reduction in phospho-Akt (pAkt) level in NSCs. These results suggest the involvement of Akt signaling in the neuroprotection of L. rhinocerus methanol extract against DEX-induced apoptosis in NSCs. Methanol extract of L. rhinocerus sclerotium exhibited potential neuroprotective activities against DEX-induced toxicity in hESC-derived NSCs. This study thus validates the use of human stem cell-derived neural lineages as potential in vitro models for screening of natural products with neuroprotective properties.
Collapse
|
11
|
Zunke F, Mazzulli JR. Modeling neuronopathic storage diseases with patient-derived culture systems. Neurobiol Dis 2019; 127:147-162. [PMID: 30790616 PMCID: PMC6588474 DOI: 10.1016/j.nbd.2019.01.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 01/23/2019] [Accepted: 01/25/2019] [Indexed: 02/08/2023] Open
Abstract
Lysosomes are organelles involved in the degradation and recycling of macromolecules, and play a critical role in sensing metabolic information in the cell. A class of rare metabolic diseases called lysosomal storage disorders (LSD) are characterized by lysosomal dysfunction and the accumulation of macromolecular substrates. The central nervous system appears to be particularly vulnerable to lysosomal dysfunction, since many LSDs are characterized by severe, widespread neurodegeneration with pediatric onset. Furthermore, variants in lysosomal genes are strongly associated with some common neurodegenerative disorders such as Parkinson's disease (PD). To better understand disease pathology and develop novel treatment strategies, it is critical to study the fundamental molecular disease mechanisms in the affected cell types that harbor endogenously expressed mutations. The discovery of methods for reprogramming of patient-derived somatic cells into induced pluripotent stem cells (iPSCs), and their differentiation into distinct neuronal and glial cell types, have provided novel opportunities to study mechanisms of lysosomal dysfunction within the relevant, vulnerable cell types. These models also expand our ability to develop and test novel therapeutic targets. We discuss recently developed methods for iPSC differentiation into distinct neuronal and glial cell types, while addressing the need for meticulous experimental techniques and parameters that are essential to accurately identify inherent cellular pathologies. iPSC models for neuronopathic LSDs and their relationship to sporadic age-related neurodegeneration are also discussed. These models should facilitate the discovery and development of personalized therapies in the future.
Collapse
Affiliation(s)
- Friederike Zunke
- Institute of Biochemistry, Christian-Albrechts-Universität zu Kiel, Kiel 24118, Germany.
| | - Joseph R Mazzulli
- Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
12
|
Wen H, Xiao W, Biswas S, Cong ZQ, Liu XM, Lam KS, Liao YH, Deng W. Alginate Hydrogel Modified with a Ligand Interacting with α3β1 Integrin Receptor Promotes the Differentiation of 3D Neural Spheroids toward Oligodendrocytes in Vitro. ACS APPLIED MATERIALS & INTERFACES 2019; 11:5821-5833. [PMID: 30645095 DOI: 10.1021/acsami.8b19438] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
In this study, we established a long-term three-dimensional (3D) culture system by using integrin ligand modified alginate hydrogels to encapsulate and differentiate neural progenitor cells (NPCs) toward oligodendrocyte (OL) lineage cells. The porosity of the hydrogel was optimized by varying the alginate concentrations and then characterized by scanning electronic microscopy (SEM). The surface plasmon resonance (SPR) test was used to confirm the ligand-integrin interactions indicating adherence between the NPC surfaces and the hydrogels. Following encapsulation in the hydrogels, both mouse and human NPC sphere cultures could be maintained up to 90 days. Mouse NPC spheres were differentiated into viable neurons, astrocytes and mature OLs by day 60 in all groups whereas human NPC spheres were differentiated into neurons and later into GFAP positive astrocytes and O4 positive pre-OL within 90 days. The species difference in the timeline of OL development between mouse and human was reflected in this system. The ligand LXY30 interacting with the α3β1 integrin receptor was more effective in promoting the differentiation of hNPCs to OL lineage cells compared with the ligand LXW64 interacting with the αvβ3 integrin receptor, hyaluronic acid interacting with CD44 receptor or without any ligand. This study is the first to differentiate O4+ pre-OLs from hNPCs in a LXY30-α3β1 (integrin-ligand) modified alginate 3D hydrogel culture. This 3D platform could serve as a valuable tool in disease modeling, drug discovery, and NPC transplantation.
Collapse
Affiliation(s)
- Han Wen
- Institute of Medicinal Plant Development , Chinese Academy of Medical Sciences and Peking Union Medical College , 151 Malianwa North Road , Haidian District, Beijing 100193 , China
- Department of Biochemistry and Molecular Medicine, School of Medicine , University of California , 2700 Stockton Blvd , Davis , California 95817 , United States
- Institute for Pediatric Regenerative Medicine , Shriners Hospitals for Children-Northern California , 2425 Stockton Blvd , Sacramento , California 95817 , United States
| | - Wenwu Xiao
- Department of Biochemistry and Molecular Medicine, School of Medicine , University of California , 2700 Stockton Blvd , Davis , California 95817 , United States
| | - Sangita Biswas
- Department of Biochemistry and Molecular Medicine, School of Medicine , University of California , 2700 Stockton Blvd , Davis , California 95817 , United States
- Institute for Pediatric Regenerative Medicine , Shriners Hospitals for Children-Northern California , 2425 Stockton Blvd , Sacramento , California 95817 , United States
- School of Pharmaceutical Sciences, Shenzhen Campus , Sun Yat-Sen University , 135 Xingang Xi Road , Guangzhou 510275 , China
| | - Zhao-Qing Cong
- Institute of Medicinal Plant Development , Chinese Academy of Medical Sciences and Peking Union Medical College , 151 Malianwa North Road , Haidian District, Beijing 100193 , China
| | - Xin-Min Liu
- Institute of Medicinal Plant Development , Chinese Academy of Medical Sciences and Peking Union Medical College , 151 Malianwa North Road , Haidian District, Beijing 100193 , China
| | - Kit S Lam
- Department of Biochemistry and Molecular Medicine, School of Medicine , University of California , 2700 Stockton Blvd , Davis , California 95817 , United States
| | - Yong-Hong Liao
- Institute of Medicinal Plant Development , Chinese Academy of Medical Sciences and Peking Union Medical College , 151 Malianwa North Road , Haidian District, Beijing 100193 , China
| | - Wenbin Deng
- Department of Biochemistry and Molecular Medicine, School of Medicine , University of California , 2700 Stockton Blvd , Davis , California 95817 , United States
- Institute for Pediatric Regenerative Medicine , Shriners Hospitals for Children-Northern California , 2425 Stockton Blvd , Sacramento , California 95817 , United States
- School of Pharmaceutical Sciences, Shenzhen Campus , Sun Yat-Sen University , 135 Xingang Xi Road , Guangzhou 510275 , China
| |
Collapse
|
13
|
Rikhtegar R, Yousefi M, Dolati S, Kasmaei HD, Charsouei S, Nouri M, Shakouri SK. Stem cell-based cell therapy for neuroprotection in stroke: A review. J Cell Biochem 2018; 120:8849-8862. [PMID: 30506720 DOI: 10.1002/jcb.28207] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 11/15/2018] [Indexed: 12/15/2022]
Abstract
Neurological disorders, such as stroke, are triggered by a loss of neurons and glial cells. Ischemic stroke remains a substantial problem for industrialized countries. Over the previous few decades our understanding about the pathophysiology of stroke has enhanced, nevertheless, more awareness is required to advance the field of stroke recovery. Existing therapies are incapable to adequately relief the disease outcome and are not appropriate to all patients. Meanwhile, the majority of patients continue to show neurological deficits even subsequent effective thrombolysis, recuperative therapies are immediately required that stimulate brain remodeling and repair once stroke damage has happened. Cell therapy is emergent as a hopeful new modality for increasing neurological recovery in ischemic stroke. Numerous types of stem cells from various sources have been identified and their possibility and efficiency for the treatment of stroke have been investigated. Stem cell therapy in patients with stroke using adult stem cells have been first practiced in clinical trials since 15 years ago. Even though stem cells have revealed a hopeful role in ischemic stroke in investigational studies besides early clinical pilot studies, cellular therapy in human is still at a primary stage. In this review, we summarize the types of stem cells, various delivery routes, and clinical application of stem cell-based therapy for stroke treatment.
Collapse
Affiliation(s)
- Reza Rikhtegar
- Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Yousefi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sanam Dolati
- Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Student's Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hosein Delavar Kasmaei
- Department of Neurology, Shohada-e-Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeid Charsouei
- Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Nouri
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyed Kazem Shakouri
- Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Physical Medicine and Rehabilitation Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
14
|
Andreopoulou E, Arampatzis A, Patsoni M, Kazanis I. Being a Neural Stem Cell: A Matter of Character But Defined by the Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1041:81-118. [PMID: 29204830 DOI: 10.1007/978-3-319-69194-7_6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The cells that build the nervous system, either this is a small network of ganglia or a complicated primate brain, are called neural stem and progenitor cells. Even though the very primitive and the very recent neural stem cells (NSCs) share common basic characteristics that are hard-wired within their character, such as the expression of transcription factors of the SoxB family, their capacity to give rise to extremely different neural tissues depends significantly on instructions from the microenvironment. In this chapter we explore the nature of the NSC microenvironment, looking through evolution, embryonic development, maturity and even disease. Experimental work undertaken over the last 20 years has revealed exciting insight into the NSC microcosmos. NSCs are very capable in producing their own extracellular matrix and in regulating their behaviour in an autocrine and paracrine manner. Nevertheless, accumulating evidence indicates an important role for the vasculature, especially within the NSC niches of the postnatal brain; while novel results reveal direct links between the metabolic state of the organism and the function of NSCs.
Collapse
Affiliation(s)
- Evangelia Andreopoulou
- Lab of Developmental Biology, Department of Biology, University of Patras, Patras, Greece
| | - Asterios Arampatzis
- Wellcome Trust- MRC Cambridge Stem Cell Biology Institute, University of Cambridge, Cambridge, UK
- School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Melina Patsoni
- Lab of Developmental Biology, Department of Biology, University of Patras, Patras, Greece
| | - Ilias Kazanis
- Lab of Developmental Biology, Department of Biology, University of Patras, Patras, Greece.
- Wellcome Trust- MRC Cambridge Stem Cell Biology Institute, University of Cambridge, Cambridge, UK.
| |
Collapse
|
15
|
Kokaia Z, Llorente IL, Carmichael ST. Customized Brain Cells for Stroke Patients Using Pluripotent Stem Cells. Stroke 2018; 49:1091-1098. [PMID: 29669871 DOI: 10.1161/strokeaha.117.018291] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 10/30/2017] [Accepted: 11/06/2017] [Indexed: 12/13/2022]
Affiliation(s)
- Zaal Kokaia
- From the Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, University Hospital, Sweden (Z.K.)
| | - Irene L Llorente
- Department of Neurology, David Geffen School of Medicine, Los Angeles, CA (I.L.L., S.T.C.)
| | - S Thomas Carmichael
- Department of Neurology, David Geffen School of Medicine, Los Angeles, CA (I.L.L., S.T.C.).
| |
Collapse
|
16
|
Maffioletti SM, Sarcar S, Henderson ABH, Mannhardt I, Pinton L, Moyle LA, Steele-Stallard H, Cappellari O, Wells KE, Ferrari G, Mitchell JS, Tyzack GE, Kotiadis VN, Khedr M, Ragazzi M, Wang W, Duchen MR, Patani R, Zammit PS, Wells DJ, Eschenhagen T, Tedesco FS. Three-Dimensional Human iPSC-Derived Artificial Skeletal Muscles Model Muscular Dystrophies and Enable Multilineage Tissue Engineering. Cell Rep 2018; 23:899-908. [PMID: 29669293 PMCID: PMC5917451 DOI: 10.1016/j.celrep.2018.03.091] [Citation(s) in RCA: 210] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 02/21/2018] [Accepted: 03/20/2018] [Indexed: 01/07/2023] Open
Abstract
Generating human skeletal muscle models is instrumental for investigating muscle pathology and therapy. Here, we report the generation of three-dimensional (3D) artificial skeletal muscle tissue from human pluripotent stem cells, including induced pluripotent stem cells (iPSCs) from patients with Duchenne, limb-girdle, and congenital muscular dystrophies. 3D skeletal myogenic differentiation of pluripotent cells was induced within hydrogels under tension to provide myofiber alignment. Artificial muscles recapitulated characteristics of human skeletal muscle tissue and could be implanted into immunodeficient mice. Pathological cellular hallmarks of incurable forms of severe muscular dystrophy could be modeled with high fidelity using this 3D platform. Finally, we show generation of fully human iPSC-derived, complex, multilineage muscle models containing key isogenic cellular constituents of skeletal muscle, including vascular endothelial cells, pericytes, and motor neurons. These results lay the foundation for a human skeletal muscle organoid-like platform for disease modeling, regenerative medicine, and therapy development.
Collapse
Affiliation(s)
| | - Shilpita Sarcar
- Department of Cell and Developmental Biology, University College London, London WC1E 6DE, UK
| | - Alexander B H Henderson
- Department of Cell and Developmental Biology, University College London, London WC1E 6DE, UK
| | - Ingra Mannhardt
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf (UKE), 20246 Hamburg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany
| | - Luca Pinton
- Department of Cell and Developmental Biology, University College London, London WC1E 6DE, UK; Randall Centre for Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK
| | - Louise Anne Moyle
- Department of Cell and Developmental Biology, University College London, London WC1E 6DE, UK
| | - Heather Steele-Stallard
- Department of Cell and Developmental Biology, University College London, London WC1E 6DE, UK; Randall Centre for Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK
| | - Ornella Cappellari
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London NW1 0TU, UK
| | - Kim E Wells
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London NW1 0TU, UK
| | - Giulia Ferrari
- Department of Cell and Developmental Biology, University College London, London WC1E 6DE, UK
| | - Jamie S Mitchell
- Institute of Neurology, University College London, London WC1N 3BG, UK; The Francis Crick Institute, London NW1 1AT, UK
| | - Giulia E Tyzack
- Institute of Neurology, University College London, London WC1N 3BG, UK; The Francis Crick Institute, London NW1 1AT, UK
| | - Vassilios N Kotiadis
- Department of Cell and Developmental Biology, University College London, London WC1E 6DE, UK
| | - Moustafa Khedr
- Department of Cell and Developmental Biology, University College London, London WC1E 6DE, UK
| | - Martina Ragazzi
- Department of Cell and Developmental Biology, University College London, London WC1E 6DE, UK
| | - Weixin Wang
- Department of Cell and Developmental Biology, University College London, London WC1E 6DE, UK
| | - Michael R Duchen
- Department of Cell and Developmental Biology, University College London, London WC1E 6DE, UK
| | - Rickie Patani
- Institute of Neurology, University College London, London WC1N 3BG, UK; The Francis Crick Institute, London NW1 1AT, UK
| | - Peter S Zammit
- Randall Centre for Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK
| | - Dominic J Wells
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London NW1 0TU, UK
| | - Thomas Eschenhagen
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf (UKE), 20246 Hamburg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany
| | | |
Collapse
|
17
|
Rybová J, Ledvinová J, Sikora J, Kuchař L, Dobrovolný R. Neural cells generated from human induced pluripotent stem cells as a model of CNS involvement in mucopolysaccharidosis type II. J Inherit Metab Dis 2018; 41:221-229. [PMID: 29168031 DOI: 10.1007/s10545-017-0108-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 10/19/2017] [Accepted: 10/22/2017] [Indexed: 01/24/2023]
Abstract
Mucopolysaccharidosis type II (MPSII) is a rare X-linked lysosomal storage disorder caused by mutations in the iduronate-2-sulfatase (IDS) gene (IDS, Xq28). MPSII is characterized by skeletal deformities, hearing loss, airway obstruction, hepatosplenomegaly, cardiac valvular disease, and progressive neurological impairment. At the cellular level, IDS deficiency leads to lysosomal storage of glycosaminoglycans (GAGs), dominated by accumulation of dermatan and heparan sulfates. Human induced pluripotent stem cells (iPSC) represent an alternative system that complements the available MPSII murine model. Herein we report on the reprogramming of peripheral white blood cells from male and female MPSII patients into iPSC using a non-integrating protocol based on the Sendai virus vector system. We differentiated the iPSC lines into IDS deficient and GAG accumulating β-Tubulin III+ neurons, GFAP+ astrocytes, and CNPase+ oligodendrocytes. The lysosomal system in these cells displayed structural abnormalities reminiscent of those previously found in patient tissues and murine IDS deficient neuronal stem cells. Furthermore, quantitative determination of GAGs revealed a moderate increase in GAG levels in IDS deficient neurons and glia. We also tested the effects of recombinant IDS and found that the exogenous enzyme was internalized from the culture media and partially decreased the intracellular GAG levels in iPSC-derived neural cells; however, it failed to completely prevent accumulation of GAGs. In summary, we demonstrate that this human iPSC based model expresses the cellular and biochemical features of MPSII, and thus represents a useful experimental tool for further pathogenesis studies as well as therapy development and testing.
Collapse
Affiliation(s)
- Jitka Rybová
- Rare Diseases Research Unit, Department of Pediatrics and Adolescent Medicine, Charles University, Ke Karlovu 2, Prague, 12808, Czech Republic
| | - Jana Ledvinová
- Rare Diseases Research Unit, Department of Pediatrics and Adolescent Medicine, Charles University, Ke Karlovu 2, Prague, 12808, Czech Republic
| | - Jakub Sikora
- Rare Diseases Research Unit, Department of Pediatrics and Adolescent Medicine, Charles University, Ke Karlovu 2, Prague, 12808, Czech Republic
- Institute of Pathology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Ladislav Kuchař
- Rare Diseases Research Unit, Department of Pediatrics and Adolescent Medicine, Charles University, Ke Karlovu 2, Prague, 12808, Czech Republic
| | - Robert Dobrovolný
- Rare Diseases Research Unit, Department of Pediatrics and Adolescent Medicine, Charles University, Ke Karlovu 2, Prague, 12808, Czech Republic.
| |
Collapse
|
18
|
Yasui T, Uezono N, Nakashima H, Noguchi H, Matsuda T, Noda-Andoh T, Okano H, Nakashima K. Hypoxia Epigenetically Confers Astrocytic Differentiation Potential on Human Pluripotent Cell-Derived Neural Precursor Cells. Stem Cell Reports 2018; 8:1743-1756. [PMID: 28591654 PMCID: PMC5470174 DOI: 10.1016/j.stemcr.2017.05.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 05/01/2017] [Accepted: 05/01/2017] [Indexed: 12/22/2022] Open
Abstract
Human neural precursor cells (hNPCs) derived from pluripotent stem cells display a high propensity for neuronal differentiation, but they require long-term culturing to differentiate efficiently into astrocytes. The mechanisms underlying this biased fate specification of hNPCs remain elusive. Here, we show that hypoxia confers astrocytic differentiation potential on hNPCs through epigenetic gene regulation, and that this was achieved by cooperation between hypoxia-inducible factor 1α and Notch signaling, accompanied by a reduction of DNA methylation level in the promoter region of a typical astrocyte-specific gene, Glial fibrillary acidic protein. Furthermore, we found that this hypoxic culture condition could be applied to rapid generation of astrocytes from Rett syndrome patient-derived hNPCs, and that these astrocytes impaired neuronal development. Thus, our findings shed further light on the molecular mechanisms regulating hNPC differentiation and provide attractive tools for the development of therapeutic strategies for treating astrocyte-mediated neurological disorders.
Collapse
Affiliation(s)
- Tetsuro Yasui
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka 812-8582, Japan; Department of Otorhinolaryngology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka 812-8582, Japan
| | - Naohiro Uezono
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka 812-8582, Japan
| | - Hideyuki Nakashima
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka 812-8582, Japan
| | - Hirofumi Noguchi
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka 812-8582, Japan
| | - Taito Matsuda
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka 812-8582, Japan
| | - Tomoko Noda-Andoh
- Department of Physiology, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Hideyuki Okano
- Department of Physiology, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Kinichi Nakashima
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka 812-8582, Japan; Laboratory of Molecular Neuroscience, Graduate School of Biological Sciences, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan.
| |
Collapse
|
19
|
Zhuo Y, Wang L, Ge L, Li X, Duan D, Teng X, Jiang M, Liu K, Yuan T, Wu P, Wang H, Deng Y, Xie H, Chen P, Xia Y, Lu M. Hypoxic Culture Promotes Dopaminergic-Neuronal Differentiation of Nasal Olfactory Mucosa Mesenchymal Stem Cells via Upregulation of Hypoxia-Inducible Factor-1α. Cell Transplant 2017; 26:1452-1461. [PMID: 28901191 PMCID: PMC5680974 DOI: 10.1177/0963689717720291] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 02/14/2017] [Accepted: 02/24/2017] [Indexed: 01/09/2023] Open
Abstract
Olfactory mucosa mesenchymal stem cells (OM-MSCs) display significant clonogenic activity and may be easily propagated for Parkinson's disease therapies. Methods of inducing OM-MSCs to differentiate into dopaminergic (DAergic) neurons using olfactory ensheathing cells (OECs) are thus an attractive topic of research. We designed a hypoxic induction protocol to generate DAergic neurons from OM-MSCs using a physiological oxygen (O2) level of 3% and OEC-conditioned medium (OCM; HI group). The normal induction (NI) group was cultured in O2 at ambient air level (21%). The role of hypoxia-inducible factor-1α (HIF-1α) in the differentiation of OM-MSCs under hypoxia was investigated by treating cells with an HIF-1α inhibitor before induction (HIR group). The proportions of β-tubulin- and tyrosine hydroxylase (TH)-positive cells were significantly increased in the HI group compared with the NI and HIR groups, as shown by immunocytochemistry and Western blotting. Furthermore, the level of dopamine was significantly increased in the HI group. A slow outward potassium current was recorded in differentiated cells after 21 d of induction using whole-cell voltage-clamp tests. A hypoxic environment thus promotes OM-MSCs to differentiate into DAergic neurons by increasing the expression of HIF-1α and by activating downstream target gene TH. This study indicated that OCM under hypoxic conditions could significantly upregulate key transcriptional factors involved in the development of DAergic neurons from OM-MSCs, mediated by HIF-1α. Hypoxia promotes DAergic neuronal differentiation of OM-MSCs, and HIF-1α may play an important role in hypoxia-inducible pathways during DAergic lineage specification and differentiation in vitro.
Collapse
Affiliation(s)
- Yi Zhuo
- Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Lei Wang
- Department of Neurosurgery, Affiliated Haikou Hospital of Xiangya School of Central South University, Haikou, China
| | - Lite Ge
- Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Xuan Li
- Cardiopulmonary Function Test Center, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Da Duan
- Department of Neurosurgery, The Second Affiliated Hospital of Hunan Normal University (PLA 163 Hospital), Changsha, China
| | - Xiaohua Teng
- Department of Neurosurgery, The Second Affiliated Hospital of Hunan Normal University (PLA 163 Hospital), Changsha, China
| | - Miao Jiang
- Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Kai Liu
- Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Ting Yuan
- Department of Neurosurgery, The Second Affiliated Hospital of Hunan Normal University (PLA 163 Hospital), Changsha, China
| | - Pei Wu
- Department of Neurosurgery, The Second Affiliated Hospital of Hunan Normal University (PLA 163 Hospital), Changsha, China
| | - Hao Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Hunan Normal University (PLA 163 Hospital), Changsha, China
| | - Yujia Deng
- Department of Neurosurgery, The Second Affiliated Hospital of Hunan Normal University (PLA 163 Hospital), Changsha, China
| | - Huali Xie
- Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Ping Chen
- Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Ying Xia
- Department of Neurosurgery, Affiliated Haikou Hospital of Xiangya School of Central South University, Haikou, China
| | - Ming Lu
- Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, China
| |
Collapse
|
20
|
Qiu J, McQueen J, Bilican B, Dando O, Magnani D, Punovuori K, Selvaraj BT, Livesey M, Haghi G, Heron S, Burr K, Patani R, Rajan R, Sheppard O, Kind PC, Simpson TI, Tybulewicz VLJ, Wyllie DJA, Fisher EMC, Lowell S, Chandran S, Hardingham GE. Evidence for evolutionary divergence of activity-dependent gene expression in developing neurons. eLife 2016; 5:e20337. [PMID: 27692071 PMCID: PMC5092045 DOI: 10.7554/elife.20337] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 09/30/2016] [Indexed: 12/12/2022] Open
Abstract
Evolutionary differences in gene regulation between humans and lower mammalian experimental systems are incompletely understood, a potential translational obstacle that is challenging to surmount in neurons, where primary tissue availability is poor. Rodent-based studies show that activity-dependent transcriptional programs mediate myriad functions in neuronal development, but the extent of their conservation in human neurons is unknown. We compared activity-dependent transcriptional responses in developing human stem cell-derived cortical neurons with those induced in developing primary- or stem cell-derived mouse cortical neurons. While activity-dependent gene-responsiveness showed little dependence on developmental stage or origin (primary tissue vs. stem cell), notable species-dependent differences were observed. Moreover, differential species-specific gene ortholog regulation was recapitulated in aneuploid mouse neurons carrying human chromosome-21, implicating promoter/enhancer sequence divergence as a factor, including human-specific activity-responsive AP-1 sites. These findings support the use of human neuronal systems for probing transcriptional responses to physiological stimuli or indeed pharmaceutical agents.
Collapse
Affiliation(s)
- Jing Qiu
- School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Jamie McQueen
- School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Bilada Bilican
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Owen Dando
- School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative Medicine, National Centre for Biological Sciences, Bangalore, India
| | - Dario Magnani
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Karolina Punovuori
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Bhuvaneish T Selvaraj
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Matthew Livesey
- School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Ghazal Haghi
- School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Samuel Heron
- School of Informatics, University of Edinburgh, Edinburgh, United Kingdom
| | - Karen Burr
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Rickie Patani
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Rinku Rajan
- School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Olivia Sheppard
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | - Peter C Kind
- School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative Medicine, National Centre for Biological Sciences, Bangalore, India
| | - T Ian Simpson
- School of Informatics, University of Edinburgh, Edinburgh, United Kingdom
| | - Victor LJ Tybulewicz
- The Francis Crick Institute, London, United Kingdom
- Imperial College, London, United Kingdom
| | - David JA Wyllie
- School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Elizabeth MC Fisher
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | - Sally Lowell
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Siddharthan Chandran
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative Medicine, National Centre for Biological Sciences, Bangalore, India
| | - Giles E Hardingham
- School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
21
|
Establishment of Human Neural Progenitor Cells from Human Induced Pluripotent Stem Cells with Diverse Tissue Origins. Stem Cells Int 2016; 2016:7235757. [PMID: 27212953 PMCID: PMC4861799 DOI: 10.1155/2016/7235757] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Accepted: 03/28/2016] [Indexed: 11/17/2022] Open
Abstract
Human neural progenitor cells (hNPCs) have previously been generated from limited numbers of human induced pluripotent stem cell (hiPSC) clones. Here, 21 hiPSC clones derived from human dermal fibroblasts, cord blood cells, and peripheral blood mononuclear cells were differentiated using two neural induction methods, an embryoid body (EB) formation-based method and an EB formation method using dual SMAD inhibitors (dSMADi). Our results showed that expandable hNPCs could be generated from hiPSC clones with diverse somatic tissue origins. The established hNPCs exhibited a mid/hindbrain-type neural identity and uniform expression of neural progenitor genes.
Collapse
|
22
|
Yap MS, Tang YQ, Yeo Y, Lim WL, Lim LW, Tan KO, Richards M, Othman I, Poh CL, Heng BC. Pluripotent Human embryonic stem cell derived neural lineages for in vitro modelling of enterovirus 71 infection and therapy. Virol J 2016; 13:5. [PMID: 26738773 PMCID: PMC4704260 DOI: 10.1186/s12985-015-0454-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 12/14/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The incidence of neurological complications and fatalities associated with Hand, Foot & Mouth disease has increased over recent years, due to emergence of newly-evolved strains of Enterovirus 71 (EV71). In the search for new antiviral therapeutics against EV71, accurate and sensitive in vitro cellular models for preliminary studies of EV71 pathogenesis is an essential prerequisite, before progressing to expensive and time-consuming live animal studies and clinical trials. METHODS This study thus investigated whether neural lineages derived from pluripotent human embryonic stem cells (hESC) can fulfil this purpose. EV71 infection of hESC-derived neural stem cells (NSC) and mature neurons (MN) was carried out in vitro, in comparison with RD and SH-SY5Y cell lines. RESULTS Upon assessment of post-infection survivability and EV71 production by the various types, it was observed that NSC were significantly more susceptible to EV71 infection compared to MN, RD (rhabdomyosarcoma) and SH-SY5Y cells, which was consistent with previous studies on mice. The SP81 peptide had significantly greater inhibitory effect on EV71 production by NSC and MN compared to the cancer-derived RD and SH-SY5Y cell lines. CONCLUSIONS Hence, this study demonstrates that hESC-derived neural lineages can be utilized as in vitro models for studying EV71 pathogenesis and for screening of antiviral therapeutics.
Collapse
Affiliation(s)
- May Shin Yap
- Department of Biological Sciences, Faculty of Science & Technology, Sunway University, No. 5 Jalan Universiti, Bandar Sunway, 47500, , Selangor Darul Ehsan, Malaysia.
| | - Yin Quan Tang
- Department of Biological Sciences, Faculty of Science & Technology, Sunway University, No. 5 Jalan Universiti, Bandar Sunway, 47500, , Selangor Darul Ehsan, Malaysia.
| | - Yin Yeo
- Department of Biological Sciences, Faculty of Science & Technology, Sunway University, No. 5 Jalan Universiti, Bandar Sunway, 47500, , Selangor Darul Ehsan, Malaysia.
| | - Wei Ling Lim
- Department of Biological Sciences, Faculty of Science & Technology, Sunway University, No. 5 Jalan Universiti, Bandar Sunway, 47500, , Selangor Darul Ehsan, Malaysia.
| | - Lee Wei Lim
- Department of Biological Sciences, Faculty of Science & Technology, Sunway University, No. 5 Jalan Universiti, Bandar Sunway, 47500, , Selangor Darul Ehsan, Malaysia. .,The University of Hong Kong, Pokfulam, Hong Kong.
| | - Kuan Onn Tan
- Department of Biological Sciences, Faculty of Science & Technology, Sunway University, No. 5 Jalan Universiti, Bandar Sunway, 47500, , Selangor Darul Ehsan, Malaysia.
| | - Mark Richards
- School of Chemical & Life Sciences, Nanyang Polytechnic, 180 Ang Mo Kio Avenue 8, Singapore, 569830, Singapore.
| | - Iekhsan Othman
- Jeffrey Cheah School of Medicine, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor Darul Ehsan, Malaysia.
| | - Chit Laa Poh
- Department of Biological Sciences, Faculty of Science & Technology, Sunway University, No. 5 Jalan Universiti, Bandar Sunway, 47500, , Selangor Darul Ehsan, Malaysia.
| | - Boon Chin Heng
- Department of Biological Sciences, Faculty of Science & Technology, Sunway University, No. 5 Jalan Universiti, Bandar Sunway, 47500, , Selangor Darul Ehsan, Malaysia. .,The University of Hong Kong, Pokfulam, Hong Kong.
| |
Collapse
|
23
|
Řeboun M, Rybová J, Dobrovolný R, Včelák J, Veselková T, Štorkánová G, Mušálková D, Hřebíček M, Ledvinová J, Magner M, Zeman J, Pešková K, Dvořáková L. X-Chromosome Inactivation Analysis in Different Cell Types and Induced Pluripotent Stem Cells Elucidates the Disease Mechanism in a Rare Case of Mucopolysaccharidosis Type II in a Female. Folia Biol (Praha) 2016; 62:82-9. [PMID: 27187040 DOI: 10.14712/fb2016062020082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Mucopolysaccharidosis type II (MPS II) is an X-linked lysosomal storage disorder resulting from deficiency of iduronate-2-sulphatase activity. The disease manifests almost exclusively in males; only 16 symptomatic heterozygote girls have been reported so far. We describe the results of X-chromosome inactivation analysis in a 5-year-old girl with clinically severe disease and heterozygous mutation p.Arg468Gln in the IDS gene. X inactivation analysed at three X-chromosome loci showed extreme skewing (96/4 to 99/1) in two patient's cell types. This finding correlated with exclusive expression of the mutated allele. Induced pluripotent stem cells (iPSC) generated from the patient's peripheral blood demonstrated characteristic pluripotency markers, deficiency of enzyme activity, and mutation in the IDS gene. These cells were capable of differentiation into other cell types (cardiomyocytes, neurons). In MPS II iPSC clones, the X inactivation ratio remained highly skewed in culture conditions that led to partial X inactivation reset in Fabry disease iPSC clones. Our data, in accordance with the literature, suggest that extremely skewed X inactivation favouring the mutated allele is a crucial condition for manifestation of MPS II in females. This suggests that the X inactivation status and enzyme activity have a prognostic value and should be used to evaluate MPS II in females. For the first time, we show generation of iPSC from a symptomatic MPS II female patient that can serve as a cellular model for further research of the pathogenesis and treatment of this disease.
Collapse
Affiliation(s)
- M Řeboun
- Institute of Inherited Metabolic Disorders, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Czech Republic
| | - J Rybová
- Institute of Inherited Metabolic Disorders, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Czech Republic
| | - R Dobrovolný
- Institute of Inherited Metabolic Disorders, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Czech Republic
| | - J Včelák
- Institute of Inherited Metabolic Disorders, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Czech Republic
| | - T Veselková
- Institute of Inherited Metabolic Disorders, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Czech Republic
| | - G Štorkánová
- Institute of Inherited Metabolic Disorders, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Czech Republic
| | - D Mušálková
- Institute of Inherited Metabolic Disorders, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Czech Republic
| | - M Hřebíček
- Institute of Inherited Metabolic Disorders, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Czech Republic
| | - J Ledvinová
- Institute of Inherited Metabolic Disorders, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Czech Republic
| | - M Magner
- Department of Paediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Czech Republic
| | - J Zeman
- Department of Paediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Czech Republic
| | - K Pešková
- Institute of Inherited Metabolic Disorders, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Czech Republic
| | - L Dvořáková
- Institute of Inherited Metabolic Disorders, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Czech Republic
| |
Collapse
|
24
|
Hypoxia-inducible factor-1α upregulates tyrosine hydroxylase and dopamine transporter by nuclear receptor ERRγ in SH-SY5Y cells. Neuroreport 2015; 26:380-6. [PMID: 25807177 DOI: 10.1097/wnr.0000000000000356] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Hypoxia-inducible factor-1α (HIF-1α) is a transcription factor relevant to the development of many mammalian organs including the brain. However, the molecular mechanisms by which signaling events mediate neuronal differentiation have not been fully elucidated. In the present study, we show for the first time that the orphan nuclear receptor estrogen-related receptor γ (ERRγ) is upregulated by HIF-1α and plays essential roles in HIF-1α-induced upregulation of dopaminergic marker molecules such as tyrosine hydroxylase and dopamine transporter. We found that deferoxamine upregulated HIF-1α and enhanced the dopaminergic phenotype and neurite outgrowth of SH-SY5Y cells. Deferoxamine activated transcription and protein expression of ERRγ, and deferoxamine-induced upregulation of tyrosine hydroxylase and dopamine transporter was attenuated by using the ERRγ inverse agonist or silencing ERRγ. Altogether, these results suggest that HIF-1α can positively regulate the dopaminergic phenotype through ERRγ. This study could provide new perspectives for understanding the mechanisms underlying the promotion of dopaminergic neuronal differentiation by hypoxia.
Collapse
|
25
|
Jiang H, Xu Z, Zhong P, Ren Y, Liang G, Schilling HA, Hu Z, Zhang Y, Wang X, Chen S, Yan Z, Feng J. Cell cycle and p53 gate the direct conversion of human fibroblasts to dopaminergic neurons. Nat Commun 2015; 6:10100. [PMID: 26639555 PMCID: PMC4672381 DOI: 10.1038/ncomms10100] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 11/03/2015] [Indexed: 12/22/2022] Open
Abstract
The direct conversion of fibroblasts to induced dopaminergic (iDA) neurons and other cell types demonstrates the plasticity of cell fate. The low efficiency of these relatively fast conversions suggests that kinetic barriers exist to safeguard cell-type identity. Here we show that suppression of p53, in conjunction with cell cycle arrest at G1 and appropriate extracellular environment, markedly increase the efficiency in the transdifferentiation of human fibroblasts to iDA neurons by Ascl1, Nurr1, Lmx1a and miR124. The conversion is dependent on Tet1, as G1 arrest, p53 knockdown or expression of the reprogramming factors induces Tet1 synergistically. Tet1 knockdown abolishes the transdifferentiation while its overexpression enhances the conversion. The iDA neurons express markers for midbrain DA neurons and have active dopaminergic transmission. Our results suggest that overcoming these kinetic barriers may enable highly efficient epigenetic reprogramming in general and will generate patient-specific midbrain DA neurons for Parkinson's disease research and therapy. The efficiency of reprogramming of fibroblasts into induced dopaminergic neurons is often low. Here, Jiang et al. demonstrate increased transdifferentiation rates by inducing cell cycle arrest, suppressing p53, Tet 1 and by adding neurotrophic factors to the media.
Collapse
Affiliation(s)
- Houbo Jiang
- Veterans Affairs Western New York Healthcare System, Buffalo, New York 14215, USA.,Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, New York 14214, USA
| | - Zhimin Xu
- Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, New York 14214, USA.,Department of Neurology and Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ping Zhong
- Veterans Affairs Western New York Healthcare System, Buffalo, New York 14215, USA.,Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, New York 14214, USA
| | - Yong Ren
- Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, New York 14214, USA
| | - Gaoyang Liang
- Howard Hughes Medical Institute, Departments of Genetics &Pediatrics, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Haley A Schilling
- Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, New York 14214, USA
| | - Zihua Hu
- Center for Computational Research, New York State Center of Excellence in Bioinformatics &Life Sciences, State University of New York at Buffalo, Buffalo, New York 14260, USA
| | - Yi Zhang
- Howard Hughes Medical Institute, Departments of Genetics &Pediatrics, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Xiaomin Wang
- Department of Neurobiology, Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Shengdi Chen
- Department of Neurology and Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhen Yan
- Veterans Affairs Western New York Healthcare System, Buffalo, New York 14215, USA.,Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, New York 14214, USA
| | - Jian Feng
- Veterans Affairs Western New York Healthcare System, Buffalo, New York 14215, USA.,Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, New York 14214, USA.,Department of Neurobiology, Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, China
| |
Collapse
|
26
|
O'Brien LC, Keeney PM, Bennett JP. Differentiation of Human Neural Stem Cells into Motor Neurons Stimulates Mitochondrial Biogenesis and Decreases Glycolytic Flux. Stem Cells Dev 2015; 24:1984-94. [PMID: 25892363 DOI: 10.1089/scd.2015.0076] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Differentiation of human pluripotent stem cells (hPSCs) in vitro offers a way to study cell types that are not accessible in living patients. Previous research suggests that hPSCs generate ATP through anaerobic glycolysis, in contrast to mitochondrial oxidative phosphorylation (OXPHOS) in somatic cells; however, specialized cell types have not been assessed. To test if mitobiogenesis is increased during motor neuron differentiation, we differentiated human embryonic stem cell (hESC)- and induced pluripotent stem cell-derived human neural stem cells (hNSCs) into motor neurons. After 21 days of motor neuron differentiation, cells increased mRNA and protein levels of genes expressed by postmitotic spinal motor neurons. Electrophysiological analysis revealed voltage-gated currents characteristic of excitable cells and action potential formation. Quantitative PCR revealed an increase in peroxisome proliferator-activated receptor gamma coactivator 1-α (PGC-1α), an upstream regulator of transcription factors involved in mitobiogenesis, and several of its downstream targets in hESC-derived cultures. This correlated with an increase in protein expression of respiratory subunits, but no increase in protein reflecting mitochondrial mass in either cell type. Respiration analysis revealed a decrease in glycolytic flux in both cell types on day 21 (D21), suggesting a switch from glycolysis to OXPHOS. Collectively, our findings suggest that mitochondrial biogenesis, but not mitochondrial mass, is increased during differentiation of hNSCs into motor neurons. These findings help us to understand human motor neuron mitobiogenesis, a process impaired in amyotrophic lateral sclerosis, a neurodegenerative disease characterized by death of motor neurons in the brain and spinal cord.
Collapse
Affiliation(s)
- Laura C O'Brien
- 1 Department of Physiology and Biophysics, Virginia Commonwealth University , Richmond, Virginia
| | - Paula M Keeney
- 2 VCU Parkinson's and Movement Disorders Center, Virginia Commonwealth University , Richmond, Virginia
| | - James P Bennett
- 1 Department of Physiology and Biophysics, Virginia Commonwealth University , Richmond, Virginia.,2 VCU Parkinson's and Movement Disorders Center, Virginia Commonwealth University , Richmond, Virginia.,3 Department of Neurology, Virginia Commonwealth University , Richmond, Virginia.,4 Department of Psychiatry, Virginia Commonwealth University , Richmond, Virginia
| |
Collapse
|
27
|
Li J, Yan Q, Ma Y, Feng Z, Wang T. Directional induction of dopaminergic neurons from neural stem cells using substantia nigra homogenates and basic fibroblast growth factor. Neural Regen Res 2015; 7:511-6. [PMID: 25745437 PMCID: PMC4348997 DOI: 10.3969/j.issn.1673-5374.2012.07.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2011] [Accepted: 01/06/2012] [Indexed: 11/18/2022] Open
Abstract
To date, complex components of available reagents have been used for directional induction of neural stem cells into dopaminergic neurons, resulting in a poor ability to repeat experiments. This study sought to investigate whether a homogenate of the substantia nigra of adult rats and/or basic fibroblast growth factor could directionally induce neural stem cells derived from the subventricular zone of embryonic rats to differentiate into dopaminergic neurons. Tyrosine hydroxylase-positive cells were observed exclusively after induction with the homogenate supernatant of the substantia nigra from adult rats and basic fibroblast growth factor for 48 hours in vitro. However, in the groups treated with homogenate supernatant or basic fibroblast growth factor alone, tyrosine hydroxylase expression was not observed. Moreover, the content of dopamine in the culture medium of subventricular zone neurons was significantly increased at 48 hours after induction with the homogenate supernatant of the substantia nigra from adult rats and basic fibroblast growth factor. Experimental findings indicate that the homogenate supernatant of the substantia nigra from adult rats and basic fibroblast growth factor could directionally induce neural stem cells derived from the subventricular zone of embryonic rats to differentiate into dopaminergic neurons in the substantia nigra with the ability to secrete dopamine.
Collapse
Affiliation(s)
- Jintao Li
- Institute of Neuroscience, Kunming Medical College, Kunming 650031, Yunnan Province, China
| | - Qi Yan
- Department of Minimally Invasive Neurosurgery, First Hospital of Kunming Medical College, Kunming 650032, Yunnan Province, China
| | - Yiliu Ma
- Department of Minimally Invasive Neurosurgery, First Hospital of Kunming Medical College, Kunming 650032, Yunnan Province, China
| | - Zhongtang Feng
- Institute of Neuroscience, Kunming Medical College, Kunming 650031, Yunnan Province, China
| | - Tinghua Wang
- Institute of Neuroscience, Kunming Medical College, Kunming 650031, Yunnan Province, China
| |
Collapse
|
28
|
Montgomery A, Wong A, Gabers N, Willerth SM. Engineering personalized neural tissue by combining induced pluripotent stem cells with fibrin scaffolds. Biomater Sci 2014. [PMID: 26218131 DOI: 10.1039/c4bm00299g] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Induced pluripotent stem cells (iPSCs) are generated from adult somatic cells through the induction of key transcription factors that restore the ability to become any cell type found in the body. These cells are of interest for tissue engineering due to their potential for developing patient-specific therapies. As the technology for generating iPSCs advances, it is important to concurrently investigate protocols for the efficient differentiation of these cells to desired downstream phenotypes in combination with biomaterial scaffolds as a way of engineering neural tissue. For such applications, the generation of neurons within three dimensional fibrin scaffolds has been well characterized as a cell-delivery platform for murine embryonic stem cells (ESCs) but has not yet been applied to murine iPSCs. Given that iPSCs have been reported to differentiate less effectively than ESCs, a key objective of this investigation is to maximize the proportion of iPSC-derived neurons in fibrin through the choice of differentiation protocol. To this end, this study compares two EB-mediated protocols for generating neurons from murine iPSCs and ESCs: an 8 day 4-/4+ protocol using soluble retinoic acid in the last 4 days and a 6 day 2-/4+ protocol using soluble retinoic acid and the small molecule sonic hedgehog agonist purmorphamine in the last 4 days. EBs were then seeded in fibrin scaffolds for 14 days to allow further differentiation into neurons. EBs generated by the 2-/4+ protocol yielded a higher percentage of neurons compared to those from the 4-/4+ protocol for both iPSCs and ESCs. The results demonstrate the successful translation of the fibrin-based cell-delivery platform for use with murine iPSCs and furthermore that the proportion of neurons generated from murine iPSC-derived EBs seeded in fibrin can be maximized using the 2-/4+ differentiation protocol. Together, these findings validate the further exploration of 3D fibrin-based scaffolds as a method of delivering neuronal cells derived from iPSCs - an important step toward the development of iPSC-based tissue engineering strategies for spinal cord injury repair.
Collapse
Affiliation(s)
- Amy Montgomery
- Department of Mechanical Engineering, University of Victoria, Canada.
| | | | | | | |
Collapse
|
29
|
Sart S, Liu Y, Ma T, Li Y. Microenvironment Regulation of Pluripotent Stem Cell-Derived Neural Progenitor Aggregates by Human Mesenchymal Stem Cell Secretome. Tissue Eng Part A 2014; 20:2666-79. [DOI: 10.1089/ten.tea.2013.0437] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Affiliation(s)
- Sébastien Sart
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida
| | - Yijun Liu
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida
| | - Teng Ma
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida
| |
Collapse
|
30
|
Lu D, Chen EYT, Lee P, Wang YC, Ching W, Markey C, Gulstrom C, Chen LC, Nguyen T, Chin WC. Accelerated neuronal differentiation toward motor neuron lineage from human embryonic stem cell line (H9). Tissue Eng Part C Methods 2014; 21:242-52. [PMID: 25036750 DOI: 10.1089/ten.tec.2013.0725] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Motor neurons loss plays a pivotal role in the pathoetiology of various debilitating diseases such as, but not limited to, amyotrophic lateral sclerosis, primary lateral sclerosis, progressive muscular atrophy, progressive bulbar palsy, pseudobulbar palsy, and spinal muscular atrophy. However, advancement in motor neuron replacement therapy has been significantly constrained by the difficulties in large-scale production at a cost-effective manner. Current methods to derive motor neuron heavily rely on biochemical stimulation, chemical biological screening, and complex physical cues. These existing methods are seriously challenged by extensive time requirements and poor yields. An innovative approach that overcomes prior hurdles and enhances the rate of successful motor neuron transplantation in patients is of critical demand. Iron, a trace element, is indispensable for the normal development and function of the central nervous system. Whether ferric ions promote neuronal differentiation and subsequently promote motor neuron lineage has never been considered. Here, we demonstrate that elevated iron concentration can drastically accelerate the differentiation of human embryonic stem cells (hESCs) toward motor neuron lineage potentially via a transferrin mediated pathway. HB9 expression in 500 nM iron-treated hESCs is approximately twofold higher than the control. Moreover, iron treatment generated more matured and functional motor neuron-like cells that are ∼1.5 times more sensitive to depolarization when compared to the control. Our methodology renders an expedited approach to harvest motor neuron-like cells for disease, traumatic injury regeneration, and drug screening.
Collapse
Affiliation(s)
- David Lu
- 1 Bioengineering Program, School of Engineering, University of California , Merced, California
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Influence of oxygen tension on dopaminergic differentiation of human fetal stem cells of midbrain and forebrain origin. PLoS One 2014; 9:e96465. [PMID: 24788190 PMCID: PMC4008610 DOI: 10.1371/journal.pone.0096465] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Accepted: 04/08/2014] [Indexed: 01/09/2023] Open
Abstract
Neural stem cells (NSCs) constitute a promising source of cells for transplantation in Parkinson's disease (PD), but protocols for controlled dopaminergic differentiation are not yet available. Here we investigated the influence of oxygen on dopaminergic differentiation of human fetal NSCs derived from the midbrain and forebrain. Cells were differentiated for 10 days in vitro at low, physiological (3%) versus high, atmospheric (20%) oxygen tension. Low oxygen resulted in upregulation of vascular endothelial growth factor and increased the proportion of tyrosine hydroxylase-immunoreactive (TH-ir) cells in both types of cultures (midbrain: 9.1±0.5 and 17.1±0.4 (P<0.001); forebrain: 1.9±0.4 and 3.9±0.6 (P<0.01) percent of total cells). Regardless of oxygen levels, the content of TH-ir cells with mature neuronal morphologies was higher for midbrain as compared to forebrain cultures. Proliferative Ki67-ir cells were found in both types of cultures, but the relative proportion of these cells was significantly higher for forebrain NSCs cultured at low, as compared to high, oxygen tension. No such difference was detected for midbrain-derived cells. Western blot analysis revealed that low oxygen enhanced β-tubulin III and GFAP expression in both cultures. Up-regulation of β-tubulin III was most pronounced for midbrain cells, whereas GFAP expression was higher in forebrain as compared to midbrain cells. NSCs from both brain regions displayed less cell death when cultured at low oxygen tension. Following mictrotransplantation into mouse striatal slice cultures predifferentiated midbrain NSCs were found to proliferate and differentiate into substantial numbers of TH-ir neurons with mature neuronal morphologies, particularly at low oxygen. In contrast, predifferentiated forebrain NSCs microtransplanted using identical conditions displayed little proliferation and contained few TH-ir cells, all of which had an immature appearance. Our data may reflect differences in dopaminergic differentiation capacity and region-specific requirements of NSCs, with the dopamine-depleted striatum cultured at low oxygen offering an attractive micro-environment for midbrain NSCs.
Collapse
|
32
|
Bilican B, Livesey MR, Haghi G, Qiu J, Burr K, Siller R, Hardingham GE, Wyllie DJA, Chandran S. Physiological normoxia and absence of EGF is required for the long-term propagation of anterior neural precursors from human pluripotent cells. PLoS One 2014; 9:e85932. [PMID: 24465796 PMCID: PMC3895023 DOI: 10.1371/journal.pone.0085932] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Accepted: 12/03/2013] [Indexed: 12/23/2022] Open
Abstract
Widespread use of human pluripotent stem cells (hPSCs) to study neuronal physiology and function is hindered by the ongoing need for specialist expertise in converting hPSCs to neural precursor cells (NPCs). Here, we describe a new methodology to generate cryo-preservable hPSC-derived NPCs that retain an anterior identity and are propagatable long-term prior to terminal differentiation, thus abrogating regular de novo neuralization. Key to achieving passagable NPCs without loss of identity is the combination of both absence of EGF and propagation in physiological levels (3%) of O2. NPCs generated in this way display a stable long-term anterior forebrain identity and importantly retain developmental competence to patterning signals. Moreover, compared to NPCs maintained at ambient O2 (21%), they exhibit enhanced uniformity and speed of functional maturation, yielding both deep and upper layer cortical excitatory neurons. These neurons display multiple attributes including the capability to form functional synapses and undergo activity-dependent gene regulation. The platform described achieves long-term maintenance of anterior neural precursors that can give rise to forebrain neurones in abundance, enabling standardised functional studies of neural stem cell maintenance, lineage choice and neuronal functional maturation for neurodevelopmental research and disease-modelling.
Collapse
Affiliation(s)
- Bilada Bilican
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, United Kingdom
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Neuroregeneration, University of Edinburgh, Edinburgh, United Kingdom
| | - Matthew R. Livesey
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Neuroregeneration, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom
| | - Ghazal Haghi
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Neuroregeneration, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom
| | - Jing Qiu
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom
| | - Karen Burr
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, United Kingdom
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Neuroregeneration, University of Edinburgh, Edinburgh, United Kingdom
| | - Rick Siller
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Neuroregeneration, University of Edinburgh, Edinburgh, United Kingdom
| | - Giles E. Hardingham
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom
| | - David J. A. Wyllie
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom
- * E-mail: (DJAW); (SC)
| | - Siddharthan Chandran
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, United Kingdom
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Neuroregeneration, University of Edinburgh, Edinburgh, United Kingdom
- * E-mail: (DJAW); (SC)
| |
Collapse
|
33
|
Hadden MK. Hedgehog pathway agonism: therapeutic potential and small-molecule development. ChemMedChem 2013; 9:27-37. [PMID: 24203435 DOI: 10.1002/cmdc.201300358] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Indexed: 11/10/2022]
Abstract
The Hedgehog (Hh) pathway is a developmental signaling pathway that plays multiple roles during embryonic development and in adult tissues. Constitutive Hh signaling has been linked to the development and progression of several forms of cancer, and the application of small-molecule pathway inhibitors as anticancer chemotherapeutics is well studied and clearly defined. Activation of the Hh pathway as a therapeutic strategy for a variety of degenerative or ischemic disorders has also been proposed; however, the development of small-molecule Hh agonists has received less attention. The goal of this review is to highlight the recent evidence supporting the therapeutic potential of Hh pathway activators and to provide a comprehensive overview of small-molecule pathway agonists.
Collapse
Affiliation(s)
- M Kyle Hadden
- Department of Pharmaceutical Sciences, University of Connecticut, 69 North Eagleville Road, Unit 3092, Storrs, CT 06269 (USA).
| |
Collapse
|
34
|
Stacpoole S, Spitzer S, Bilican B, Compston A, Karadottir R, Chandran S, Franklin R. High yields of oligodendrocyte lineage cells from human embryonic stem cells at physiological oxygen tensions for evaluation of translational biology. Stem Cell Reports 2013; 1:437-50. [PMID: 24286031 PMCID: PMC3841262 DOI: 10.1016/j.stemcr.2013.09.006] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2013] [Revised: 09/26/2013] [Accepted: 09/27/2013] [Indexed: 02/09/2023] Open
Abstract
We have established and efficient system to specify NG2/PDGF-Rα/OLIG2+ oligodendrocyte precursor cells (OPCs) from human embryonic stem cells (hESCs) at low, physiological (3%) oxygen levels. This was achieved via both forebrain and spinal cord origins, with up to 98% of cells expressing NG2. Developmental insights reveal a critical role for fibroblast growth factor 2 (FGF-2) in OLIG2 induction via ventral forebrain pathways. The OPCs mature in vitro to express O4 (46%) and subsequently become galactocerebroside (GALC), O1, and myelin basic protein-positive (MBP+) multibranching oligodendrocytes. These were cultured alongside hESC-derived neurons. The electrophysiological properties of human OPCs are similar to those of rat OPCs, with large voltage-gated sodium currents and the ability to fire action potentials. Exposure to a selective retinoid X receptor agonist increased the proportion of O4+ oligodendrocytes that express MBP from 5% to 30%. Thus, we have established a developmentally engineered system to investigate the biological properties of human OPCs and test the effects of putative remyelinating agents prior to clinical application.
Collapse
Affiliation(s)
- Sybil R.L. Stacpoole
- Department of Clinical Neurosciences, Cambridge University, Cambridge CB2 0PY, UK
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Veterinary Medicine, Cambridge University, Cambridge CB3 0ES, UK
| | - Sonia Spitzer
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Veterinary Medicine, Cambridge University, Cambridge CB3 0ES, UK
| | - Bilada Bilican
- Centre for Neuroregeneration, Edinburgh University, Edinburgh EH16 4SB, UK
| | - Alastair Compston
- Department of Clinical Neurosciences, Cambridge University, Cambridge CB2 0PY, UK
| | - Ragnhildur Karadottir
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Veterinary Medicine, Cambridge University, Cambridge CB3 0ES, UK
| | | | - Robin J.M. Franklin
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Veterinary Medicine, Cambridge University, Cambridge CB3 0ES, UK
| |
Collapse
|
35
|
Delivery of differentiation factors by mesoporous silica particles assists advanced differentiation of transplanted murine embryonic stem cells. Stem Cells Transl Med 2013. [PMID: 24089415 DOI: 10.5966/sctm.2013-0072] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Stem cell transplantation holds great hope for the replacement of damaged cells in the nervous system. However, poor long-term survival after transplantation and insufficiently robust differentiation of stem cells into specialized cell types in vivo remain major obstacles for clinical application. Here, we report the development of a novel technological approach for the local delivery of exogenous trophic factor mimetics to transplanted cells using specifically designed silica nanoporous particles. We demonstrated that delivering Cintrofin and Gliafin, established peptide mimetics of the ciliary neurotrophic factor and glial cell line-derived neurotrophic factor, respectively, with these particles enabled not only robust functional differentiation of motor neurons from transplanted embryonic stem cells but also their long-term survival in vivo. We propose that the delivery of growth factors by mesoporous nanoparticles is a potentially versatile and widely applicable strategy for efficient differentiation and functional integration of stem cell derivatives upon transplantation.
Collapse
|
36
|
Villeneuve L, Tiede LM, Morsey B, Fox HS. Quantitative proteomics reveals oxygen-dependent changes in neuronal mitochondria affecting function and sensitivity to rotenone. J Proteome Res 2013; 12:4599-606. [PMID: 23971408 DOI: 10.1021/pr400758d] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Mitochondria are implicated in a variety of degenerative disorders and aging. Mitochondria are responsive to the oxygen in their environment, yet tissue culture is performed at atmospheric (21%) oxygen and not at physiological (1-11%) oxygen levels found in tissues. We employed imaging of mitochondrial probes, mass spectrometry, Western blots, and ATP assays of the human neuroblastoma cell-line SH-SY5Y and imaging of mitochondrial probes in human primary neurons under standard nonphysiological oxygen conditions (atmospheric) and under physiological oxygen levels in the nervous system to assess the impact of oxygen on mitochondrial function. SH-SY5Y cells cultured in physiological 5% oxygen exhibited the lowest reactive oxygen species (ROS) production, indicating that culture at 5% oxygen is favored; these results were mimicked in primary human cells. Mass spectrometric analysis revealed extensive mitochondrial proteomic alterations in SH-SY5Y cells based on oxygen culture condition. Among these, the rotenone-sensitive subunit of complex I NDUFV3 was increased in cells cultured at 5% oxygen. Rotenone is a Parkinson's disease-linked toxin, and correspondingly SH-SY5Y cells cultured at 5% oxygen also exhibited over 10 times greater sensitivity to rotenone than those cultured in atmospheric, 21%, oxygen. Our results indicate that neuronal mitochondria are responsive to oxygen levels and produce differential responses under different oxygen levels.
Collapse
Affiliation(s)
- Lance Villeneuve
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center , 985800 Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | | | | | | |
Collapse
|
37
|
Drury-Stewart D, Song M, Mohamad O, Guo Y, Gu X, Chen D, Wei L. Highly efficient differentiation of neural precursors from human embryonic stem cells and benefits of transplantation after ischemic stroke in mice. Stem Cell Res Ther 2013; 4:93. [PMID: 23928330 PMCID: PMC3854684 DOI: 10.1186/scrt292] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Accepted: 07/26/2013] [Indexed: 02/07/2023] Open
Abstract
Introduction Ischemic stroke is a leading cause of death and disability, but treatment options are severely limited. Cell therapy offers an attractive strategy for regenerating lost tissues and enhancing the endogenous healing process. In this study, we investigated the use of human embryonic stem cell-derived neural precursors as a cell therapy in a murine stroke model. Methods Neural precursors were derived from human embryonic stem cells by using a fully adherent SMAD inhibition protocol employing small molecules. The efficiency of neural induction and the ability of these cells to further differentiate into neurons were assessed by using immunocytochemistry. Whole-cell patch-clamp recording was used to demonstrate the electrophysiological activity of human embryonic stem cell-derived neurons. Neural precursors were transplanted into the core and penumbra regions of a focal ischemic stroke in the barrel cortex of mice. Animals received injections of bromodeoxyuridine to track regeneration. Neural differentiation of the transplanted cells and regenerative markers were measured by using immunohistochemistry. The adhesive removal test was used to determine functional improvement after stroke and intervention. Results After 11 days of neural induction by using the small-molecule protocol, over 95% of human embryonic stem-derived cells expressed at least one neural marker. Further in vitro differentiation yielded cells that stained for mature neuronal markers and exhibited high-amplitude, repetitive action potentials in response to depolarization. Neuronal differentiation also occurred after transplantation into the ischemic cortex. A greater level of bromodeoxyuridine co-localization with neurons was observed in the penumbra region of animals receiving cell transplantation. Transplantation also improved sensory recovery in transplant animals over that in control animals. Conclusions Human embryonic stem cell-derived neural precursors derived by using a highly efficient small-molecule SMAD inhibition protocol can differentiate into electrophysiologically functional neurons in vitro. These cells also differentiate into neurons in vivo, enhance regenerative activities, and improve sensory recovery after ischemic stroke.
Collapse
|
38
|
Chronic hypoxia leads to a glycolytic phenotype and suppressed HIF-2 signaling in PC12 cells. Biochim Biophys Acta Gen Subj 2013; 1830:3553-69. [DOI: 10.1016/j.bbagen.2013.02.016] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Revised: 01/22/2013] [Accepted: 02/15/2013] [Indexed: 12/12/2022]
|
39
|
Stacpoole SRL, Webber DJ, Bilican B, Compston A, Chandran S, Franklin RJM. Neural precursor cells cultured at physiologically relevant oxygen tensions have a survival advantage following transplantation. Stem Cells Transl Med 2013; 2:464-72. [PMID: 23677643 DOI: 10.5966/sctm.2012-0144] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Traditionally, in vitro stem cell systems have used oxygen tensions that are far removed from the in vivo situation. This is particularly true for the central nervous system, where oxygen (O2) levels range from 8% at the pia to 0.5% in the midbrain, whereas cells are usually cultured in a 20% O2 environment. Cell transplantation strategies therefore typically introduce a stress challenge at the time of transplantation as the cells are switched from 20% to 3% O2 (the average in adult organs). We have modeled the oxygen stress that occurs during transplantation, demonstrating that in vitro transfer of neonatal rat cortical neural precursor cells (NPCs) from a 20% to a 3% O2 environment results in significant cell death, whereas maintenance at 3% O2 is protective. This survival benefit translates to the in vivo environment, where culture of NPCs at 3% rather than 20% O2 approximately doubles survival in the immediate post-transplantation phase. Furthermore, NPC fate is affected by culture at low, physiological O2 tensions (3%), with particularly marked effects on the oligodendrocyte lineage, both in vitro and in vivo. We propose that careful consideration of physiological oxygen environments, and particularly changes in oxygen tension, has relevance for the practical approaches to cellular therapies.
Collapse
Affiliation(s)
- Sybil R L Stacpoole
- Department of Clinical Neurosciences, University Medical Center, the Netherlands.
| | | | | | | | | | | |
Collapse
|
40
|
Wang Y, Yang J, Li H, Wang X, Zhu L, Fan M, Wang X. Hypoxia promotes dopaminergic differentiation of mesenchymal stem cells and shows benefits for transplantation in a rat model of Parkinson's disease. PLoS One 2013; 8:e54296. [PMID: 23342124 PMCID: PMC3546985 DOI: 10.1371/journal.pone.0054296] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2012] [Accepted: 12/10/2012] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells capable of differentiating into dopaminergic (DAergic) neurons, which is one of the major cell types damaged in Parkinson’s disease (PD). For this reason, MSCs are considered a potential cell source for PD therapy. It has been proved that hypoxia is involved in the proliferation and differentiation of stem cells. In this study, we investigated the effect of hypoxia on MSC proliferation and DAergic neuronal differentiation. Our results demonstrate that 3% O2 treatment can enhance rat MSC proliferation by upregulation of phosphorylated p38 MAPK and subsequent nuclear translocation of hypoxia inducible factor (HIF)-1α. During neural differentiation, 3% O2 treatment increases the expression of HIF-1α, phosphorylated ERK and p38 MAPK. These changes are followed by promotion of neurosphere formation and further DAergic neuronal differentiation. Furthermore, we explored the physiological function of hypoxia-induced DAergic neurons from human fetal MSCs by transplanting them into parkinsonian rats. Grafts induced with hypoxia display more survival of DAergic neurons and greater amelioration of behavioral impairments. Altogether, these results suggest that hypoxia can promote MSC proliferation and DAergic neuronal differentiation, and benefit for intrastriatal transplantation. Therefore, this study may provide new perspectives in application of MSCs to clinical PD therapy.
Collapse
Affiliation(s)
- Yue Wang
- Neuroscience Research Institute, Peking University, Key Laboratory of Neuroscience (PKU), Ministry of Education, Peking University Health Science Center, Beijing, China
| | - Jian Yang
- Department of Physiology and Neurobiology, Capital Medical University, Key Laboratory for Neurodegenerative Disease of Education Ministry, Youanmen, Beijing, China
- Beijing An Ding Hospital, Beijing, China
| | - Haisheng Li
- Department of Brain Protection, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Xuan Wang
- Department of Physiology and Neurobiology, Capital Medical University, Key Laboratory for Neurodegenerative Disease of Education Ministry, Youanmen, Beijing, China
| | - Lingling Zhu
- Department of Brain Protection, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Ming Fan
- Department of Brain Protection, Beijing Institute of Basic Medical Sciences, Beijing, China
- * E-mail: (XMW); (MF)
| | - Xiaomin Wang
- Department of Physiology and Neurobiology, Capital Medical University, Key Laboratory for Neurodegenerative Disease of Education Ministry, Youanmen, Beijing, China
- * E-mail: (XMW); (MF)
| |
Collapse
|
41
|
Badger JL, Byrne ML, Veraitch FS, Mason C, Wall IB, Caldwell MA. Hypoxic culture of human pluripotent stem cell lines is permissible using mouse embryonic fibroblasts. Regen Med 2012; 7:675-83. [DOI: 10.2217/rme.12.55] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Aim: Hypoxia is used within in vitro stem cell culture to recreate conditions similar to the in vivo environment surrounding the early blastocyst, from which embryonic stem cells can be isolated. Traditionally, basic research has used a coculture feeder system to culture pluripotent stem cells; however, it is possible that lowered oxygen may restrict cellular metabolic activity of the inactivated mouse embryonic fibroblasts (iMEFs) by disrupting oxygen-dependent pathways, such as ATP production through aerobic respiration. In this work, we examined the potential to continue using routine culture methods, such as iMEFs, to support human pluripotent cell expansion under hypoxia instead of feeder-free methods that can cause cell instability and offer a poor cell attachment rate. Materials & methods: Metabolic activity and viability studies were carried out in normoxic and hypoxic conditions. Pluripotent stem cells were introduced into hypoxia on iMEFs and the rate of colony expansion was compared with normoxic conditions. In addition, pluripotent stem cells were grown in hypoxia for over 6 months to demonstrate maintenance of pluripotency. Immunocytochemistry and western blotting evaluated the activity of the hypoxic transcription factor, HIF1A. Results: Hypoxia does not significantly affect viability or metabolic activity of feeder cells, and there is no detrimental effect on the rate of pluripotent stem cell colony expansion when cells are cultured in hypoxia. In addition, hypoxic pluripotent stem cells maintain their pluripotent nature and ability to differentiate into the three germ layers. Conclusion: The traditional iMEF coculture method is suitable for use in hypoxia and does not need to be replaced with feeder-free systems for hypoxic culture of human pluripotent stem cell lines in basic research.
Collapse
Affiliation(s)
- Jennifer L Badger
- Department of Biochemical Engineering, University College London, Torrington Place, London, WC1E 7JE, UK
- Henry Wellcome Laboratories for Integrative Neuroscience & Endocrinology, Dorothy Hodgkin Building, University of Bristol, Whitson Street, Bristol, BS1 3NY, UK
| | - Meg L Byrne
- Department of Biochemical Engineering, University College London, Torrington Place, London, WC1E 7JE, UK
- Henry Wellcome Laboratories for Integrative Neuroscience & Endocrinology, Dorothy Hodgkin Building, University of Bristol, Whitson Street, Bristol, BS1 3NY, UK
| | - Farlan S Veraitch
- Department of Biochemical Engineering, University College London, Torrington Place, London, WC1E 7JE, UK
| | - Chris Mason
- Department of Biochemical Engineering, University College London, Torrington Place, London, WC1E 7JE, UK
| | - Ivan B Wall
- Department of Biochemical Engineering, University College London, Torrington Place, London, WC1E 7JE, UK
| | - Maeve A Caldwell
- Henry Wellcome Laboratories for Integrative Neuroscience & Endocrinology, Dorothy Hodgkin Building, University of Bristol, Whitson Street, Bristol, BS1 3NY, UK
| |
Collapse
|
42
|
Vukicevic V, Schmid J, Hermann A, Lange S, Qin N, Gebauer L, Chunk KF, Ravens U, Eisenhofer G, Storch A, Ader M, Bornstein SR, Ehrhart-Bornstein M. Differentiation of chromaffin progenitor cells to dopaminergic neurons. Cell Transplant 2012; 21:2471-86. [PMID: 22507143 DOI: 10.3727/096368912x638874] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The differentiation of dopamine-producing neurons from chromaffin progenitors might represent a new valuable source for replacement therapies in Parkinson's disease. However, characterization of their differentiation potential is an important prerequisite for efficient engraftment. Based on our previous studies on isolation and characterization of chromaffin progenitors from adult adrenals, this study investigates their potential to produce dopaminergic neurons and means to enhance their dopaminergic differentiation. Chromaffin progenitors grown in sphere culture showed an increased expression of nestin and Mash1, indicating an increase of the progenitor subset. Proneurogenic culture conditions induced the differentiation into neurons positive for neural markers β-III-tubulin, MAP2, and TH accompanied by a decrease of Mash1 and nestin. Furthermore, Notch2 expression decreased concomitantly with a downregulation of downstream effectors Hes1 and Hes5 responsible for self-renewal and proliferation maintenance of progenitor cells. Chromaffin progenitor-derived neurons secreted dopamine upon stimulation by potassium. Strikingly, treatment of differentiating cells with retinoic and ascorbic acid resulted in a twofold increase of dopamine secretion while norepinephrine and epinephrine were decreased. Initiation of dopamine synthesis and neural maturation is controlled by Pitx3 and Nurr1. Both Pitx3 and Nurr1 were identified in differentiating chromaffin progenitors. Along with the gained dopaminergic function, electrophysiology revealed features of mature neurons, such as sodium channels and the capability to fire multiple action potentials. In summary, this study elucidates the capacity of chromaffin progenitor cells to generate functional dopaminergic neurons, indicating their potential use in cell replacement therapies.
Collapse
Affiliation(s)
- Vladimir Vukicevic
- Molecular Endocrinology, Medical Clinic III, University Clinic Dresden, Dresden University of Technology, Fetscherstrasse 74, Dresden, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
|