51
|
Abstract
Regenerative therapies, including both gene and cellular therapies, aim to induce regeneration of cells, tissues and organs and restore their functions. In this short Spotlight, we summarize the latest advances in cellular therapies using pluripotent stem cells (PSCs), highlighting the current status of clinical trials using induced (i)PSC-derived cells. We also discuss the different cellular products that might be used in clinical studies, and consider safety issues and other challenges in iPSC-based cell therapy.
Collapse
Affiliation(s)
- Hideyuki Okano
- Keio University School of Medicine, Department of Physiology, 35 Shinano-machi, Shinjuku-ku, Tokyo 160-8582, Japan .,Keio University Global Research Institute, 2-15-45 Mita, Minato-ku, Tokyo 108-8345, Japan
| | - Doug Sipp
- Keio University School of Medicine, Department of Physiology, 35 Shinano-machi, Shinjuku-ku, Tokyo 160-8582, Japan.,Keio University Global Research Institute, 2-15-45 Mita, Minato-ku, Tokyo 108-8345, Japan.,RIKEN Center for Developmental Biology, 2-2-3 Minatojima Minamimachi, Chuo-ku, Kobe 650-0047, Japan.,RIKEN Center for Advanced Intelligence Project, Nihonbashi 1-chome Mitsui Building, 15th floor, 1-4-1 Nihonbashi, Chuo-ku, Tokyo 103-0027, Japan
| |
Collapse
|
52
|
Cherkashova EA, Salikhova DI, Namestnikova DD, Leonov GE, Gubskiy IL, Solovieva AA, Akopyan GB, Kurilo VV, Nikitina MP, Fatkhudinov TK, Chekhonin VP, Gubskiy LV, Yarygin KN, Bukharova TB, Goldshtein DV. Therapeutic effects of intra-arterial administration of glial progenitor cells-conditioned medium in acute experimental ishemic stroke in rats. GENES & CELLS 2020; 15:80-84. [DOI: 10.23868/202011013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Transplantation of various types of stem cells as a possible therapy for stroke has been tested for years and the results are promising. Recently, most researchers are inclined to assume that the therapeutic effect of stem cell therapy is based on the mechanism of paracrine action associated with the secretion wide set of regulatory proteins. The aim of this study was to evaluate therapeutic effects of iPSC-derived glial progenitor cells conditioned medium in the rat middle cerebral artery occlusion model of the ischemic stroke. We showed that intra-arterial administration of glial progenitor cells conditioned medium promoted faster decrease of neurological deficit compared to the control group. Moreover, expression of gap43, bax, and tnfa genes involved in neuritogenesis, apoptosis and neuroinflammation was altered. However, no significant enhanced reduction of the infarct volume was registered. Our results demonstrated that administration of glial progenitor cells conditioned medium induced functional recovery after experimental stroke and may affect brain plasticity.
Collapse
|
53
|
Induced Pluripotent Stem Cells: Hope in the Treatment of Diseases, including Muscular Dystrophies. Int J Mol Sci 2020; 21:ijms21155467. [PMID: 32751747 PMCID: PMC7432218 DOI: 10.3390/ijms21155467] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 04/22/2020] [Accepted: 04/27/2020] [Indexed: 02/07/2023] Open
Abstract
Induced pluripotent stem (iPS) cells are laboratory-produced cells that combine the biological advantages of somatic adult and stem cells for cell-based therapy. The reprogramming of cells, such as fibroblasts, to an embryonic stem cell-like state is done by the ectopic expression of transcription factors responsible for generating embryonic stem cell properties. These primary factors are octamer-binding transcription factor 4 (Oct3/4), sex-determining region Y-box 2 (Sox2), Krüppel-like factor 4 (Klf4), and the proto-oncogene protein homolog of avian myelocytomatosis (c-Myc). The somatic cells can be easily obtained from the patient who will be subjected to cellular therapy and be reprogrammed to acquire the necessary high plasticity of embryonic stem cells. These cells have no ethical limitations involved, as in the case of embryonic stem cells, and display minimal immunological rejection risks after transplant. Currently, several clinical trials are in progress, most of them in phase I or II. Still, some inherent risks, such as chromosomal instability, insertional tumors, and teratoma formation, must be overcome to reach full clinical translation. However, with the clinical trials and extensive basic research studying the biology of these cells, a promising future for human cell-based therapies using iPS cells seems to be increasingly clear and close.
Collapse
|
54
|
Hamamura K, Hotta H, Murakumo Y, Shibuya H, Kondo Y, Furukawa K. SSEA-3 and 4 are not essential for the induction or properties of mouse iPS cells. J Oral Sci 2020; 62:393-396. [PMID: 32684574 DOI: 10.2334/josnusd.19-0513] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
Stage-specific embryonic antigens (SSEA-1, 3, and 4) are carbohydrate antigens that have been used as markers of embryonic stem (ES) cells. However, the roles of these antigens in the establishment and maintenance of stemness of ES and induced pluripotent stem (iPS) cells are still poorly understood. This study investigated the biological and functional significance of globo-series glycolipids such as SSEA-3 and 4 in mouse iPS cells induced from tail-tip fibroblasts (TTFs) of α1,4Gal-T-knockout mice (lacking SSEA-3 and 4). These iPS cells were induced by retroviral transduction of four factors (Oct3/4, Sox2, Klf4, and c-Myc) into TTFs, and colonies were picked up. Morphologically, the colonies resembled ES cells and were positive for alkaline phosphatase and ES cell markers. Furthermore, in vitro-differentiated induction experiments after embryoid body formation revealed that some colonies derived from α1, 4Gal-T knockout mice were able to differentiate into three germ layers. Three germ layers were also observed in teratomas from iPS cells derived from α1,4Gal-T-knockout mice. These results suggest that SSEA-3 and 4 are not essential, at least for the establishment and maintenance of stemness of mouse iPS cells.
Collapse
Affiliation(s)
- Kazunori Hamamura
- Department of Pharmacology, School of Dentistry, Aichi Gakuin University
| | - Hiroshi Hotta
- Department of Biochemistry II, Nagoya University Graduate School of Medicine
| | | | - Hidenobu Shibuya
- Department of Biochemistry II, Nagoya University Graduate School of Medicine
| | - Yuji Kondo
- Department of Biochemistry II, Nagoya University Graduate School of Medicine
| | - Koichi Furukawa
- Department of Biochemistry II, Nagoya University Graduate School of Medicine.,Department of Biomedical Sciences, Chubu University College of Life and Health Sciences
| |
Collapse
|
55
|
Suzuki N, Akiyama T, Warita H, Aoki M. Omics Approach to Axonal Dysfunction of Motor Neurons in Amyotrophic Lateral Sclerosis (ALS). Front Neurosci 2020; 14:194. [PMID: 32269505 PMCID: PMC7109447 DOI: 10.3389/fnins.2020.00194] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 02/24/2020] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is an intractable adult-onset neurodegenerative disease that leads to the loss of upper and lower motor neurons (MNs). The long axons of MNs become damaged during the early stages of ALS. Genetic and pathological analyses of ALS patients have revealed dysfunction in the MN axon homeostasis. However, the molecular pathomechanism for the degeneration of axons in ALS has not been fully elucidated. This review provides an overview of the proposed axonal pathomechanisms in ALS, including those involving the neuronal cytoskeleton, cargo transport within axons, axonal energy supply, clearance of junk protein, neuromuscular junctions (NMJs), and aberrant axonal branching. To improve understanding of the global changes in axons, the review summarizes omics analyses of the axonal compartments of neurons in vitro and in vivo, including a motor nerve organoid approach that utilizes microfluidic devices developed by this research group. The review also discusses the relevance of intra-axonal transcription factors frequently identified in these omics analyses. Local axonal translation and the relationship among these pathomechanisms should be pursued further. The development of novel strategies to analyze axon fractions provides a new approach to establishing a detailed understanding of resilience of long MN and MN pathology in ALS.
Collapse
Affiliation(s)
- Naoki Suzuki
- Department of Neurology, Tohoku University School of Medicine, Sendai, Japan.,Department of Neurology, Shodo-kai Southern Tohoku General Hospital, Miyagi, Japan
| | - Tetsuya Akiyama
- Department of Neurology, Tohoku University School of Medicine, Sendai, Japan
| | - Hitoshi Warita
- Department of Neurology, Tohoku University School of Medicine, Sendai, Japan
| | - Masashi Aoki
- Department of Neurology, Tohoku University School of Medicine, Sendai, Japan
| |
Collapse
|
56
|
Moayeri A, Darvishi M, Amraei M. Homing of Super Paramagnetic Iron Oxide Nanoparticles (SPIONs) Labeled Adipose-Derived Stem Cells by Magnetic Attraction in a Rat Model of Parkinson's Disease. Int J Nanomedicine 2020; 15:1297-1308. [PMID: 32161459 PMCID: PMC7049746 DOI: 10.2147/ijn.s238266] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 02/07/2020] [Indexed: 12/11/2022] Open
Abstract
Introduction Stem cell therapies for neurodegenerative diseases such as Parkinson’s disease (PD) are intended to replace lost dopaminergic neurons. The basis of this treatment is to guide the migration of transplanted cells into the target tissue or injury site. The aim of this study is an evaluation of the homing of superparamagnetic iron oxide nanoparticles (SPIONs) labeled adipose-derived stem cells (ADSC) by an external magnetic field in a rat model of PD. Methods ADSCs were obtained from perinephric regions of male adult rats and cultured in a DMEM medium. ADSC markers were assessed by immunostaining with CD90, CD105, CD49d, and CD45. The SPION was coated using poly-L-lysine hydrobromide and transfection was determined in rat ADSC using the GFP reporter gene. For this in vivo study, rats with PD were divided into five groups: a positive control group, a control group with PD (lesion with 6-HD injection), and three treatment groups: the PD/ADSC group (PD transplant with ADSCs transfected by BrdU), PD/ADSC/SPION group (PD transplant with ADSCs labeled with SPION and transfected by GFP), and the PD/ADSC/SPION/EM group (PD transplant with ADSCs labeled with SPION and transfected by GFP induced with external magnet). Results ADSCs were immunoreactive to fat markers CD90 (90.73±1.7), CD105 (87.4±2.9) and CD49d (79.6±2.6), with negative immunostaining at the hematopoietic stem cell marker (CD45: 1.4±0.4). The efficiency of cells with SPION/PLL was about 96% of ADSC. The highest number of GFP-positive cells was in the ADSC/SPION/EM group (54.5±1.3), which was significantly different from that in ADSC/SPION group (30.83±3 and P<0.01). Conclusion Transfection of ADSC by SPION/PLL is an appropriate protocol for cell therapy. External magnets can be used for the delivery and homing of transplanted stem cells in the target tissue.
Collapse
Affiliation(s)
- Ardeshir Moayeri
- Department of Anatomy, Faculty of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Marzieh Darvishi
- Department of Anatomy, Faculty of Medicine, Ilam University of Medical Sciences, Ilam, Iran.,Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| | - Mansour Amraei
- Department of Physiology, Faculty of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| |
Collapse
|
57
|
Ishikawa M, Aoyama T, Shibata S, Sone T, Miyoshi H, Watanabe H, Nakamura M, Morota S, Uchino H, Yoo AS, Okano H. miRNA-Based Rapid Differentiation of Purified Neurons from hPSCs Advancestowards Quick Screening for Neuronal Disease Phenotypes In Vitro. Cells 2020; 9:E532. [PMID: 32106535 PMCID: PMC7140514 DOI: 10.3390/cells9030532] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/18/2020] [Accepted: 02/20/2020] [Indexed: 12/12/2022] Open
Abstract
Obtaining differentiated cells with high physiological functions by an efficient, but simple and rapid differentiation method is crucial for modeling neuronal diseases in vitro using human pluripotent stem cells (hPSCs). Currently, methods involving the transient expression of one or a couple of transcription factors have been established as techniques for inducing neuronal differentiation in a rapid, single step. It has also been reported that microRNAs can function as reprogramming effectors for directly reprogramming human dermal fibroblasts to neurons. In this study, we tested the effect of adding neuronal microRNAs, miRNA-9/9*, and miR-124 (miR-9/9*-124), for the neuronal induction method of hPSCs using Tet-On-driven expression of the Neurogenin2 gene (Ngn2), a proneural factor. While it has been established that Ngn2 can facilitate differentiation from pluripotent stem cells into neurons with high purity due to its neurogenic effect, a long or indefinite time is required for neuronal maturation with Ngn2 misexpression alone. With the present method, the cells maintained a high neuronal differentiation rate while exhibiting increased gene expression of neuronal maturation markers, spontaneous calcium oscillation, and high electrical activity with network bursts as assessed by a multipoint electrode system. Moreover, when applying this method to iPSCs from Alzheimer's disease (AD) patients with presenilin-1 (PS1) or presenilin-2 (PS2) mutations, cellular phenotypes such as increased amount of extracellular secretion of amyloid β42, abnormal oxygen consumption, and increased reactive oxygen species in the cells were observed in a shorter culture period than those previously reported. Therefore, it is strongly anticipated that the induction method combining Ngn2 and miR-9/9*-124 will enable more rapid and simple screening for various types of neuronal disease phenotypes and promote drug discovery.
Collapse
Affiliation(s)
- Mitsuru Ishikawa
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Takeshi Aoyama
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Shoichiro Shibata
- Department of Anesthesiology, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| | - Takefumi Sone
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Hiroyuki Miyoshi
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Hirotaka Watanabe
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Mari Nakamura
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Saori Morota
- Department of Anesthesiology, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| | - Hiroyuki Uchino
- Department of Anesthesiology, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| | - Andrew S Yoo
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| |
Collapse
|
58
|
Watanabe H, Ito T, Tsuda I. A mathematical model for neuronal differentiation in terms of an evolved dynamical system. Neurosci Res 2020; 156:206-216. [PMID: 32084446 DOI: 10.1016/j.neures.2020.02.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 01/08/2020] [Accepted: 02/05/2020] [Indexed: 11/30/2022]
Abstract
We attempted to create a mathematical model for neuronal differentiation. The present study was performed within the framework of self-organization with constraints by looking for an optimized informational unit. We treated networks of individual dynamical system units with an external input, which was provided by coupled one-dimensional maps with possible forms of unidirectionally feed-forward network, random network, small-world network, and fully-connected network. We used a genetic algorithm to maximize the information transmission for each type of network. Optimized maps were obtained depending on the coupling strength and network structure. These maps can be classified into three types: passive, excitable, and oscillatory. In particular, the excitable and oscillatory types of dynamical systems possess characteristics that are quite similar to those of neurons, whereas the passive and oscillatory types of dynamical system may represent glial cells.
Collapse
Affiliation(s)
- Hiroshi Watanabe
- Chubu University Academy of Emerging Sciences, Kasugai, Aichi, 487-8501, Japan.
| | - Takao Ito
- Hokkaido Prefectural Police, Sapporo, Hokkaido, 060-8520, Japan
| | - Ichiro Tsuda
- Chubu University Academy of Emerging Sciences, Kasugai, Aichi, 487-8501, Japan
| |
Collapse
|
59
|
Onodera K, Shimojo D, Ishihara Y, Yano M, Miya F, Banno H, Kuzumaki N, Ito T, Okada R, de Araújo Herculano B, Ohyama M, Yoshida M, Tsunoda T, Katsuno M, Doyu M, Sobue G, Okano H, Okada Y. Unveiling synapse pathology in spinal bulbar muscular atrophy by genome-wide transcriptome analysis of purified motor neurons derived from disease specific iPSCs. Mol Brain 2020; 13:18. [PMID: 32070397 PMCID: PMC7029484 DOI: 10.1186/s13041-020-0561-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Accepted: 01/29/2020] [Indexed: 02/09/2023] Open
Abstract
Spinal bulbar muscular atrophy (SBMA) is an adult-onset, slowly progressive motor neuron disease caused by abnormal CAG repeat expansion in the androgen receptor (AR) gene. Although ligand (testosterone)-dependent mutant AR aggregation has been shown to play important roles in motor neuronal degeneration by the analyses of transgenic mice models and in vitro cell culture models, the underlying disease mechanisms remain to be fully elucidated because of the discrepancy between model mice and SBMA patients. Thus, novel human disease models that recapitulate SBMA patients’ pathology more accurately are required for more precise pathophysiological analysis and the development of novel therapeutics. Here, we established disease specific iPSCs from four SBMA patients, and differentiated them into spinal motor neurons. To investigate motor neuron specific pathology, we purified iPSC-derived motor neurons using flow cytometry and cell sorting based on the motor neuron specific reporter, HB9e438::Venus, and proceeded to the genome-wide transcriptome analysis by RNA sequences. The results revealed the involvement of the pathology associated with synapses, epigenetics, and endoplasmic reticulum (ER) in SBMA. Notably, we demonstrated the involvement of the neuromuscular synapse via significant upregulation of Synaptotagmin, R-Spondin2 (RSPO2), and WNT ligands in motor neurons derived from SBMA patients, which are known to be associated with neuromuscular junction (NMJ) formation and acetylcholine receptor (AChR) clustering. These aberrant gene expression in neuromuscular synapses might represent a novel therapeutic target for SBMA.
Collapse
Affiliation(s)
- Kazunari Onodera
- Department of Neurology, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan.,Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Daisuke Shimojo
- Department of Neurology, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan.,Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Yasuharu Ishihara
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Masato Yano
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 951-8510, Japan
| | - Fuyuki Miya
- Department of Medical Science Mathematics, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, 113-8510, Japan.,Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, 113-0033, Japan.,Laboratory for Medical Science Mathematics, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
| | - Haruhiko Banno
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Naoko Kuzumaki
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan.,Department of Pharmacology, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Tokyo, 142-8501, Japan
| | - Takuji Ito
- Department of Neurology, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan
| | - Rina Okada
- Department of Neurology, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan
| | - Bruno de Araújo Herculano
- Department of Neurology, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan
| | - Manabu Ohyama
- Department of Dermatology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Mari Yoshida
- Department of Neuropathology, Institute for Medical Science of Aging, Aichi Medical University, Nagakute, Aichi, 480-1195, Japan
| | - Tatsuhiko Tsunoda
- Department of Medical Science Mathematics, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, 113-8510, Japan.,Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, 113-0033, Japan.,Laboratory for Medical Science Mathematics, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
| | - Masahisa Katsuno
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Manabu Doyu
- Department of Neurology, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan
| | - Gen Sobue
- Research Division of Dementia and Neurodegenerative Disease, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Yohei Okada
- Department of Neurology, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan.
| |
Collapse
|
60
|
Okano H, Yasuda D, Fujimori K, Morimoto S, Takahashi S. Ropinirole, a New ALS Drug Candidate Developed Using iPSCs. Trends Pharmacol Sci 2020; 41:99-109. [PMID: 31926602 DOI: 10.1016/j.tips.2019.12.002] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 11/28/2019] [Accepted: 12/04/2019] [Indexed: 12/12/2022]
Abstract
Induced pluripotent stem cells (iPSCs) are increasingly used in the study of disease mechanisms and the development of effective disease-modifying therapies for neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS). Recently, three candidate anti-ALS drugs - ropinirole (ROPI), retigabine, and bosutinib - have been identified in iPSC-based drug screens and are now being evaluated in clinical trials for safety and effectiveness. We review the preclinical data, clinical research design, and rationale for ROPI as an anti-ALS drug candidate compared with those of the other two drugs. We also discuss the use of iPSCs for understanding and monitoring treatment response as well as for new insights into the development of new drugs and therapeutic interventions for major neurodegenerative diseases.
Collapse
Affiliation(s)
- Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan.
| | - Daisuke Yasuda
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo, Japan
| | - Koki Fujimori
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan
| | - Satoru Morimoto
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan
| | - Shinichi Takahashi
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan; Department of Neurology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan; Department of Neurology and Stroke, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka-shi, Saitama, Japan
| |
Collapse
|
61
|
Haldar S, Ghosh S, Kumar V, Roy P, Lahiri D. The Evolving Neural Tissue Engineering Landscape of India. ACS APPLIED BIO MATERIALS 2019; 2:5446-5459. [PMID: 35021543 DOI: 10.1021/acsabm.9b00567] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The healthcare sector in India is witnessing unprecedented advancement. Tissue engineering has become an integral part of healthcare and medicine, particularly where treatments involve functional restoration of any injured or deceased part of the body. Not falling behind much with the progressing medical and healthcare sector of India, tissue engineering is also gaining momentum in the country. Out of several arenas of tissue engineering, India has made its mark in orthopedic and bone regeneration, cosmetic and skin regeneration, and very importantly neural regeneration. There are several articles reviewing the progress and prospects of orthopedic and skin regeneration research in India. However, there is no systematic review on progress, prospects, and pitfalls associated with neural tissue engineering in Indian context. The existing ones mainly focus on the technical advancements in the field from a global perspective. Therefore, it is worthwhile to have an organized look at the evolving neural tissue engineering landscape of India. This review will walk the readers systematically through different aspects of the topic. The review starts with an introduction to the nervous system to help readers appreciate the complexity that must be dealt with while engineering neural tissue. This is followed with a global picture of the neural tissue engineering, prominent research groups working on neural tissue engineering in India, factors that have and are currently molding the prospects of this field, and concluding with an overall perspective on present and future of neural tissue engineering in India.
Collapse
|
62
|
Kathuria A, Lopez-Lengowski K, Watmuff B, McPhie D, Cohen BM, Karmacharya R. Synaptic deficits in iPSC-derived cortical interneurons in schizophrenia are mediated by NLGN2 and rescued by N-acetylcysteine. Transl Psychiatry 2019; 9:321. [PMID: 31780643 PMCID: PMC6882825 DOI: 10.1038/s41398-019-0660-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 09/06/2019] [Accepted: 10/20/2019] [Indexed: 02/08/2023] Open
Abstract
Human postmortem studies suggest a major role for abnormalities in GABAergic interneurons in the prefrontal cortex in schizophrenia. Cortical interneurons differentiated from induced pluripotent stem cells (iPSCs) of schizophrenia subjects showed significantly lower levels of glutamate decarboxylase 67 (GAD67), replicating findings from multiple postmortem studies, as well as reduced levels of synaptic proteins gehpyrin and NLGN2. Co-cultures of the interneurons with excitatory cortical pyramidal neurons from schizophrenia iPSCs showed reduced synaptic puncta density and lower action potential frequency. NLGN2 overexpression in schizophrenia neurons rescued synaptic puncta deficits while NLGN2 knockdown in healthy neurons resulted in reduced synaptic puncta density. Schizophrenia interneurons also had significantly smaller nuclear area, suggesting an innate oxidative stressed state. The antioxidant N-acetylcysteine increased the nuclear area in schizophrenia interneurons, increased NLGN2 expression and rescued synaptic deficits. These results implicate specific deficiencies in the synaptic machinery in cortical interneurons as critical regulators of synaptic connections in schizophrenia and point to a nexus between oxidative stress and NLGN2 expression in mediating synaptic deficits in schizophrenia.
Collapse
Affiliation(s)
- Annie Kathuria
- 0000 0004 0386 9924grid.32224.35Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA USA ,grid.66859.34Chemical Biology and Therapeutic Science Program, Broad Institute of MIT & Harvard, Cambridge, MA USA ,000000041936754Xgrid.38142.3cDepartment of Psychiatry, Harvard Medical School, Boston, MA USA
| | - Kara Lopez-Lengowski
- 0000 0004 0386 9924grid.32224.35Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA USA ,grid.66859.34Chemical Biology and Therapeutic Science Program, Broad Institute of MIT & Harvard, Cambridge, MA USA
| | - Bradley Watmuff
- 0000 0004 0386 9924grid.32224.35Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA USA ,grid.66859.34Chemical Biology and Therapeutic Science Program, Broad Institute of MIT & Harvard, Cambridge, MA USA ,000000041936754Xgrid.38142.3cDepartment of Psychiatry, Harvard Medical School, Boston, MA USA
| | - Donna McPhie
- 000000041936754Xgrid.38142.3cDepartment of Psychiatry, Harvard Medical School, Boston, MA USA ,0000 0000 8795 072Xgrid.240206.2Schizophrenia and Bipolar Disorder Program, McLean Hospital, Belmont, MA USA
| | - Bruce M. Cohen
- 000000041936754Xgrid.38142.3cDepartment of Psychiatry, Harvard Medical School, Boston, MA USA ,0000 0000 8795 072Xgrid.240206.2Schizophrenia and Bipolar Disorder Program, McLean Hospital, Belmont, MA USA
| | - Rakesh Karmacharya
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA. .,Chemical Biology and Therapeutic Science Program, Broad Institute of MIT & Harvard, Cambridge, MA, USA. .,Department of Psychiatry, Harvard Medical School, Boston, MA, USA. .,Schizophrenia and Bipolar Disorder Program, McLean Hospital, Belmont, MA, USA. .,Graduate Program in Chemical Biology, Harvard University, Cambridge, MA, USA. .,Program in Neuroscience, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
63
|
Lam M, Sanosaka T, Lundin A, Imaizumi K, Etal D, Karlsson FH, Clausen M, Cairns J, Hicks R, Kohyama J, Kele M, Okano H, Falk A. Single-cell study of neural stem cells derived from human iPSCs reveals distinct progenitor populations with neurogenic and gliogenic potential. Genes Cells 2019; 24:836-847. [PMID: 31651061 PMCID: PMC6916357 DOI: 10.1111/gtc.12731] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 10/04/2019] [Accepted: 10/22/2019] [Indexed: 12/30/2022]
Abstract
We used single‐cell RNA sequencing (seq) on several human induced pluripotent stem (iPS) cell‐derived neural stem cell (NSC) lines and one fetal brain‐derived NSC line to study inherent cell type heterogeneity at proliferating neural stem cell stage and uncovered predisposed presence of neurogenic and gliogenic progenitors. We observed heterogeneity in neurogenic progenitors that differed between the iPS cell‐derived NSC lines and the fetal‐derived NSC line, and we also observed differences in spontaneous differentiation potential for inhibitory and excitatory neurons between the iPS cell‐derived NSC lines and the fetal‐derived NSC line. In addition, using a recently published glia patterning protocol we enriched for gliogenic progenitors and generated glial cells from an iPS cell‐derived NSC line.
Collapse
Affiliation(s)
- Matti Lam
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Tsukasa Sanosaka
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | | | - Kent Imaizumi
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | | | | | | | | | | | - Jun Kohyama
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Malin Kele
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Anna Falk
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
64
|
In Vitro Modeling of the Bipolar Disorder and Schizophrenia Using Patient-Derived Induced Pluripotent Stem Cells with Copy Number Variations of PCDH15 and RELN. eNeuro 2019; 6:ENEURO.0403-18.2019. [PMID: 31540999 PMCID: PMC6800292 DOI: 10.1523/eneuro.0403-18.2019] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 08/30/2019] [Accepted: 09/03/2019] [Indexed: 12/12/2022] Open
Abstract
Bipolar disorder (BP) and schizophrenia (SCZ) are major psychiatric disorders, but the molecular mechanisms underlying the complicated pathologies of these disorders remain unclear. It is difficult to establish adequate in vitro models for pathological analysis because of the heterogeneity of these disorders. In the present study, to recapitulate the pathologies of these disorders in vitro, we established in vitro models by differentiating mature neurons from human induced pluripotent stem cells (hiPSCs) derived from BP and SCZ patient with contributive copy number variations, as follows: two BP patients with PCDH15 deletion and one SCZ patient with RELN deletion. Glutamatergic neurons and GABAergic neurons were induced from hiPSCs under optimized conditions. Both types of induced neurons from both hiPSCs exhibited similar phenotypes of MAP2 (microtubule-associated protein 2)-positive dendrite shortening and decreasing synapse numbers. Additionally, we analyzed isogenic PCDH15- or RELN-deleted cells. The dendrite and synapse phenotypes of isogenic neurons were partially similar to those of patient-derived neurons. These results suggest that the observed phenotypes are general phenotypes of psychiatric disorders, and our in vitro models using hiPSC-based technology may be suitable for analysis of the pathologies of psychiatric disorders.
Collapse
|
65
|
Evaluating the efficacy of small molecules for neural differentiation of common marmoset ESCs and iPSCs. Neurosci Res 2019; 155:1-11. [PMID: 31586586 DOI: 10.1016/j.neures.2019.09.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Revised: 09/12/2019] [Accepted: 09/26/2019] [Indexed: 12/15/2022]
Abstract
The common marmoset (marmoset; Callithrix jacchus) harbors various desired features as a non-human primate (NHP) model for neuroscience research. Recently, efforts have been made to induce neural cells in vitro from marmoset pluripotent stem cells (PSCs), including embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), which are characterized by their capacity to differentiate into all cell types from the three germ layers. Successful generation of marmoset neural cells is not only invaluable for understanding neural development and for modeling neurodegenerative and psychiatric disorders, but is also necessary for the phenotypic screening of genetically-modified marmosets. However, differences in the differentiation propensity among PSC lines hamper the applicability and the reproducibility of differentiation methods. To overcome this limitation, we evaluated the efficacy of small molecules for neural differentiation of marmoset ESCs (cjESCs) and iPSCs using multiple differentiation methods. By immunochemical and transcriptomic analyses, we confirmed that our methods using the small molecules are efficient for various differentiation protocols by either enhancing the yield of a mixture of neural cells including both neurons and glial cells, or a pure population of neurons. Collectively, our findings optimized in vitro neural differentiation methods for marmoset PSCs, which would ultimately help enhance the utility of the animal model in neuroscience.
Collapse
|
66
|
Antill-O'Brien N, Bourke J, O'Connell CD. Layer-By-Layer: The Case for 3D Bioprinting Neurons to Create Patient-Specific Epilepsy Models. MATERIALS (BASEL, SWITZERLAND) 2019; 12:E3218. [PMID: 31581436 PMCID: PMC6804258 DOI: 10.3390/ma12193218] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 09/26/2019] [Accepted: 09/26/2019] [Indexed: 02/06/2023]
Abstract
The ability to create three-dimensional (3D) models of brain tissue from patient-derived cells, would open new possibilities in studying the neuropathology of disorders such as epilepsy and schizophrenia. While organoid culture has provided impressive examples of patient-specific models, the generation of organised 3D structures remains a challenge. 3D bioprinting is a rapidly developing technology where living cells, encapsulated in suitable bioink matrices, are printed to form 3D structures. 3D bioprinting may provide the capability to organise neuronal populations in 3D, through layer-by-layer deposition, and thereby recapitulate the complexity of neural tissue. However, printing neuron cells raises particular challenges since the biomaterial environment must be of appropriate softness to allow for the neurite extension, properties which are anathema to building self-supporting 3D structures. Here, we review the topic of 3D bioprinting of neurons, including critical discussions of hardware and bio-ink formulation requirements.
Collapse
Affiliation(s)
- Natasha Antill-O'Brien
- BioFab3D, Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Fitzroy, VIC 3065, Australia.
| | - Justin Bourke
- BioFab3D, Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Fitzroy, VIC 3065, Australia.
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, Innovation Campus, University of Wollongong, NSW 2522, Australia.
- Department of Medicine, St Vincent's Hospital Melbourne, University of Melbourne, Fitzroy, VIC 3065, Australia.
| | - Cathal D O'Connell
- BioFab3D, Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Fitzroy, VIC 3065, Australia.
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, Innovation Campus, University of Wollongong, NSW 2522, Australia.
| |
Collapse
|
67
|
Csobonyeiova M, Polak S, Zamborsky R, Danisovic L. Recent Progress in the Regeneration of Spinal Cord Injuries by Induced Pluripotent Stem Cells. Int J Mol Sci 2019; 20:3838. [PMID: 31390782 PMCID: PMC6695701 DOI: 10.3390/ijms20153838] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 07/27/2019] [Accepted: 08/02/2019] [Indexed: 12/13/2022] Open
Abstract
Regeneration of injuries occurring in the central nervous system, particularly spinal cord injuries (SCIs), is extremely difficult. The complex pathological events following a SCI often restrict regeneration of nervous tissue at the injury site and frequently lead to irreversible loss of motor and sensory function. Neural stem/progenitor cells (NSCs/NPCs) possess neuroregenerative and neuroprotective features, and transplantation of such cells into the site of damaged tissue is a promising stem cell-based therapy for SCI. However, NSC/NPCs have mostly been induced from embryonic stem cells or fetal tissue, leading to ethical concerns. The pioneering work of Yamanaka and colleagues gave rise to the technology to induce pluripotent stem cells (iPSCs) from somatic cells, overcoming these ethical issues. The advent of iPSCs technology has meant significant progress in the therapy of neurodegenerative disease and nerve tissue damage. A number of published studies have described the successful differentiation of NSCs/NPCs from iPSCs and their subsequent engraftment into SCI animal models, followed by functional recovery of injury. The aim of this present review is to summarize various iPSC- NPCs differentiation methods, SCI modelling, and the current status of possible iPSC- NPCs- based therapy of SCI.
Collapse
Affiliation(s)
- Maria Csobonyeiova
- Institute of Histology and Embryology, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Stefan Polak
- Institute of Histology and Embryology, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Radoslav Zamborsky
- Department of Orthopaedics, Faculty of Medicine, Comenius University and National Institute of Children's Diseases, Limbova 1, 833 40 Bratislava, Slovakia
| | - Lubos Danisovic
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia.
- Regenmed Ltd., Medena 29, 811 01 Bratislava, Slovakia.
| |
Collapse
|
68
|
Shao A, Tu S, Lu J, Zhang J. Crosstalk between stem cell and spinal cord injury: pathophysiology and treatment strategies. Stem Cell Res Ther 2019; 10:238. [PMID: 31387621 PMCID: PMC6683526 DOI: 10.1186/s13287-019-1357-z] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The injured spinal cord is difficult to repair and regenerate. Traditional treatments are not effective. Stem cells are a type of cells that have the potential to differentiate into various cells, including neurons. They exert a therapeutic effect by safely and effectively differentiating into neurons or replacing damaged cells, secreting neurotrophic factors, and inhibiting the inflammatory response. Many types of stem cells have been used for transplantation, and each has its own advantages and disadvantages. This review discusses the possible mechanisms of stem cell therapy for spinal cord injury, and the types of stem cells commonly used in experiments, to provide a reference for basic and clinical research on stem cell therapy for spinal cord injury.
Collapse
Affiliation(s)
- Anwen Shao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China.
| | - Sheng Tu
- Department of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Jianan Lu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Jianmin Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China.,Brain Research Institute, Zhejiang University, Hangzhou, 310003, China.,Collaborative Innovation Center for Brain Science, Zhejiang University, Hangzhou, 310003, China
| |
Collapse
|
69
|
Nakazawa T, Hashimoto R, Takuma K, Hashimoto H. Modeling of psychiatric disorders using induced pluripotent stem cell-related technologies. J Pharmacol Sci 2019; 140:321-324. [DOI: 10.1016/j.jphs.2019.06.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 05/29/2019] [Accepted: 06/03/2019] [Indexed: 02/06/2023] Open
|
70
|
Nakajima M, Yoshimatsu S, Sato T, Nakamura M, Okahara J, Sasaki E, Shiozawa S, Okano H. Establishment of induced pluripotent stem cells from common marmoset fibroblasts by RNA-based reprogramming. Biochem Biophys Res Commun 2019; 515:593-599. [DOI: 10.1016/j.bbrc.2019.05.175] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 05/30/2019] [Indexed: 10/26/2022]
|
71
|
Yamashita T, Shang J, Nakano Y, Morihara R, Sato K, Takemoto M, Hishikawa N, Ohta Y, Abe K. In vivo direct reprogramming of glial linage to mature neurons after cerebral ischemia. Sci Rep 2019; 9:10956. [PMID: 31358888 PMCID: PMC6662847 DOI: 10.1038/s41598-019-47482-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 07/17/2019] [Indexed: 01/23/2023] Open
Abstract
The therapeutic effect of in vivo direct reprogramming on ischemic stroke has not been evaluated. In the present study, a retroviral solution (1.5–2.0 × 107 /ul) of mock pMX-GFP (n = 13) or pMX-Ascl1/Sox2/NeuroD1 (ASN) (n = 14) was directly injected into the ipsilateral striatum and cortex 3 days after 30 min of transient cerebral ischemia. The reprogrammed cells first expressed neuronal progenitor marker Dcx 7 and 21 days after viral injection, then expressed mature neuronal marker NeuN. This was accompanied by morphological changes, including long processes and synapse-like structures, 49 days after viral injection. Meanwhile, therapeutic improvement was not detected both in clinical scores or infarct volume. The present study provides a future novel self-repair strategy for ischemic stroke with beneficial modifications of the inducer-suppressor balance.
Collapse
Affiliation(s)
- Toru Yamashita
- Department of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Jingwei Shang
- Department of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yumiko Nakano
- Department of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Ryuta Morihara
- Department of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kota Sato
- Department of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Mami Takemoto
- Department of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Nozomi Hishikawa
- Department of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yasuyuki Ohta
- Department of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Koji Abe
- Department of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.
| |
Collapse
|
72
|
Popa-Wagner A, Hermann D, Gresita A. Genetic conversion of proliferative astroglia into neurons after cerebral ischemia: a new therapeutic tool for the aged brain? GeroScience 2019; 41:363-368. [PMID: 31300928 DOI: 10.1007/s11357-019-00084-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 07/02/2019] [Indexed: 12/24/2022] Open
Abstract
Ischemic stroke represents the 2nd leading cause of death worldwide and the leading cause for long-term disabilities, for which no cure exists. After stroke, neurons are frequently lost in the infarct core. On the other hand, other cells such as astrocytes become reactive and proliferative, disrupting the neurovascular unit in the lesioned area, especially in the aged brain. Therefore, restoring the balance between neurons and nonneuronal cells within the perilesional area is crucial for post stroke recovery. In addition, the aged post stroke brain mounts a fulminant proliferative astroglial response leading to the buildup of gliotic scars that prevent neural regeneration. Therefore, "melting" glial scars has been attempted for decades, albeit with little success. Alternative strategies include transforming inhibitory gliotic tissue into an environment conducive to neuronal regeneration and axonal growth by genetic conversion of astrocytes into neurons. The latter idea has gained momentum following the discovery that in vivo direct lineage reprogramming in the adult mammalian brain is a feasible strategy for reprogramming nonneuronal cells into neurons. This exciting new technology emerged as a new approach to circumvent cell transplantation for stroke therapy. However, the potential of this new methodology has not been yet tested to improve restoration of structure and function in the hostile environment caused by the fulminant inflammatory reaction in the brains of aged animals.
Collapse
Affiliation(s)
- Aurel Popa-Wagner
- Center of Clinical and Experimental Medicine, University of Medicine and Pharmacy, Craiova, Romania. .,Vascular Neurology, Dementia and Ageing Research, Department of Neurology, University of Duisburg-Essen, University Hospital Essen, 45122, Essen, Germany.
| | - Dirk Hermann
- Vascular Neurology, Dementia and Ageing Research, Department of Neurology, University of Duisburg-Essen, University Hospital Essen, 45122, Essen, Germany
| | - Andrei Gresita
- Center of Clinical and Experimental Medicine, University of Medicine and Pharmacy, Craiova, Romania
| |
Collapse
|
73
|
Aberrant axon branching via Fos-B dysregulation in FUS-ALS motor neurons. EBioMedicine 2019; 45:362-378. [PMID: 31262712 PMCID: PMC6642224 DOI: 10.1016/j.ebiom.2019.06.013] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 05/20/2019] [Accepted: 06/09/2019] [Indexed: 12/18/2022] Open
Abstract
Background The characteristic structure of motor neurons (MNs), particularly of the long axons, becomes damaged in the early stages of amyotrophic lateral sclerosis (ALS). However, the molecular pathophysiology of axonal degeneration remains to be fully elucidated. Method Two sets of isogenic human-induced pluripotent stem cell (hiPSCs)-derived MNs possessing the single amino acid difference (p.H517D) in the fused in sarcoma (FUS) were constructed. By combining MN reporter lentivirus, MN specific phenotype was analyzed. Moreover, RNA profiling of isolated axons were conducted by applying the microfluidic devices that enable axon bundles to be produced for omics analysis. The relationship between the target gene, which was identified as a pathological candidate in ALS with RNA-sequencing, and the MN phenotype was confirmed by intervention with si-RNA or overexpression to hiPSCs-derived MNs and even in vivo. The commonality was further confirmed with other ALS-causative mutant hiPSCs-derived MNs and human pathology. Findings We identified aberrant increasing of axon branchings in FUS-mutant hiPSCs-derived MN axons compared with isogenic controls as a novel phenotype. We identified increased level of Fos-B mRNA, the binding target of FUS, in FUS-mutant MNs. While Fos-B reduction using si-RNA or an inhibitor ameliorated the observed aberrant axon branching, Fos-B overexpression resulted in aberrant axon branching even in vivo. The commonality of those phenotypes was further confirmed with other ALS causative mutation than FUS. Interpretation Analyzing the axonal fraction of hiPSC-derived MNs using microfluidic devices revealed that Fos-B is a key regulator of FUS-mutant axon branching. Fund Japan Agency for Medical Research and development; Japanese Ministry of Education, Culture, Sports, Science and Technology Clinical Research, Innovation and Education Center, Tohoku University Hospital; Japan Intractable Diseases (Nanbyo) Research Foundation; the Kanae Foundation for the Promotion of Medical Science; and “Inochi-no-Iro” ALS research grant.
Collapse
|
74
|
Ahlfors JE, Azimi A, El-Ayoubi R, Velumian A, Vonderwalde I, Boscher C, Mihai O, Mani S, Samoilova M, Khazaei M, Fehlings MG, Morshead CM. Examining the fundamental biology of a novel population of directly reprogrammed human neural precursor cells. Stem Cell Res Ther 2019; 10:166. [PMID: 31196173 PMCID: PMC6567617 DOI: 10.1186/s13287-019-1255-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 04/29/2019] [Accepted: 05/06/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Cell reprogramming is a promising avenue for cell-based therapies as it allows for the generation of multipotent, unipotent, or mature somatic cells without going through a pluripotent state. While the use of autologous cells is considered ideal, key challenges for their clinical translation include the ability to reproducibly generate sufficient quantities of cells within a therapeutically relevant time window. METHODS We performed transfection of three distinct human somatic starting populations of cells with a non-integrating synthetic plasmid expressing Musashi 1 (MSI1), Neurogenin 2 (NGN2), and Methyl-CpG-Binding Domain 2 (MBD2). The resulting directly reprogrammed neural precursor cells (drNPCs) were examined in vitro using RT-qPCR, karyotype analysis, immunohistochemistry, and FACS at early and late time post-transfection. Electrophysiology (patch clamp) was performed on drNPC-derived neurons to determine their capacity to generate action potentials. In vivo characterization was performed following transplantation of drNPCs into two animal models (Shiverer and SCID/Beige mice), and the numbers, location, and differentiation profile of the transplanted cells were examined using immunohistochemistry. RESULTS Human somatic cells can be directly reprogrammed within two weeks to neural precursor cells (drNPCs) by transient exposure to Msi1, Ngn2, and MBD2 using non-viral constructs. The drNPCs generate all three neural cell types (astrocytes, oligodendrocytes, and neurons) and can be passaged in vitro to generate large numbers of cells within four weeks. drNPCs can respond to in vivo differentiation and migration cues as demonstrated by their migration to the olfactory bulb and contribution to neurogenesis in vivo. Differentiation profiles of transplanted cells onto the corpus callosum of myelin-deficient mice reveal the production of oligodendrocytes and astrocytes. CONCLUSIONS Human drNPCs can be efficiently and rapidly produced from donor somatic cells and possess all the important characteristics of native neural multipotent cells including differentiation into neurons, astrocytes, and oligodendrocytes, and in vivo neurogenesis and myelination.
Collapse
Affiliation(s)
| | - Ashkan Azimi
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario M5S 1A8 Canada
- Division of Anatomy, Department of Surgery, University of Toronto, Ontario, M5S 1A8 Canada
| | | | - Alexander Velumian
- Division of Neurosurgery, Department of Surgery, University of Toronto, Ontario, M5T 1P5 Canada
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario M5T 2S8 Canada
| | - Ilan Vonderwalde
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9 Canada
| | | | - Oana Mihai
- New World Laboratories, Laval, Quebec, H7V 5B7 Canada
| | - Sarathi Mani
- New World Laboratories, Laval, Quebec, H7V 5B7 Canada
| | - Marina Samoilova
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario M5T 2S8 Canada
| | - Mohamad Khazaei
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario M5T 2S8 Canada
| | - Michael G. Fehlings
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario M5S 1A8 Canada
- Division of Neurosurgery, Department of Surgery, University of Toronto, Ontario, M5T 1P5 Canada
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario M5T 2S8 Canada
| | - Cindi M Morshead
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario M5S 1A8 Canada
- Division of Anatomy, Department of Surgery, University of Toronto, Ontario, M5S 1A8 Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9 Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario M5S3E1 Canada
| |
Collapse
|
75
|
Blair JD, Bateup HS. New frontiers in modeling tuberous sclerosis with human stem cell-derived neurons and brain organoids. Dev Dyn 2019; 249:46-55. [PMID: 31070828 DOI: 10.1002/dvdy.60] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 05/01/2019] [Accepted: 05/02/2019] [Indexed: 12/16/2022] Open
Abstract
Recent advances in human stem cell and genome engineering have enabled the generation of genetically defined human cellular models for brain disorders. These models can be established from a patient's own cells and can be genetically engineered to generate isogenic, controlled systems for mechanistic studies. Given the challenges of obtaining and working with primary human brain tissue, these models fill a critical gap in our understanding of normal and abnormal human brain development and provide an important complement to animal models. Recently, there has been major progress in modeling the neuropathophysiology of the canonical "mTORopathy" tuberous sclerosis complex (TSC) with such approaches. Studies using two- and three-dimensional cultures of human neurons and glia have provided new insights into how mutations in the TSC1 and TSC2 genes impact human neural development and function. Here we discuss recent progress in human stem cell-based modeling of TSC and highlight challenges and opportunities for further efforts in this area.
Collapse
Affiliation(s)
- John D Blair
- Department of Molecular and Cell Biology, University of California, Berkeley, California
| | - Helen S Bateup
- Department of Molecular and Cell Biology, University of California, Berkeley, California.,Helen Wills Neuroscience Institute, University of California, Berkeley, California.,Chan Zuckerberg Biohub, San Francisco, California
| |
Collapse
|
76
|
Nie Y, Wang W, Xu X, Zou J, Bhuvanesh T, Schulz B, Ma N, Lendlein A. Enhancement of human induced pluripotent stem cells adhesion through multilayer laminin coating. Clin Hemorheol Microcirc 2019; 70:531-542. [PMID: 30347612 DOI: 10.3233/ch-189318] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Bioengineered cell substrates are a highly promising tool to govern the differentiation of stem cells in vitro and to modulate the cellular behavior in vivo. While this technology works fine for adult stem cells, the cultivation of human induced pluripotent stem cells (hiPSCs) is challenging as these cells typically show poor attachment on the bioengineered substrates, which among other effects causes substantial cell death. Thus, very limited types of surfaces have been demonstrated suitable for hiPSC cultures. The multilayer coating approach that renders the surface with diverse chemical compositions, architectures, and functions can be used to improve the adhesion of hiPSCs on the bioengineered substrates. We hypothesized that a multilayer formation based on the attraction of molecules with opposite charges could functionalize the polystyrene (PS) substrates to improve the adhesion of hiPSCs. Polymeric substrates were stepwise coated, first with dopamine to form a polydopamine (PDA) layer, second with polylysine and last with Laminin-521. The multilayer formation resulted in the variation of hydrophilicity and chemical functionality of the surfaces. Hydrophilicity was detected using captive bubble method and the amount of primary and secondary amines on the surface was quantified by fluorescent staining. The PDA layer effectively immobilized the upper layers and thereby improved the attachment of hiPSCs. Cell adhesion was enhanced on the surfaces coated with multilayers, as compared to those without PDA and/or polylysine. Moreover, hiPSCs spread well over this multilayer laminin substrate. These cells maintained their proliferation capacity and differentiation potential. The multilayer coating strategy is a promising attempt for engineering polymer-based substrates for the cultivation of hiPSCs and of interest for expanding the application scope of hiPSCs.
Collapse
Affiliation(s)
- Yan Nie
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
| | - Weiwei Wang
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
| | - Xun Xu
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany.,Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Jie Zou
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany.,Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Thanga Bhuvanesh
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany.,Institute of Chemistry, University of Potsdam, Potsdam, Germany
| | - Burkhard Schulz
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany.,Institute of Chemistry, University of Potsdam, Potsdam, Germany
| | - Nan Ma
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany.,Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany.,Helmholtz Virtual Institute - Multifunctional Biomaterials for Medicine, Teltow, Germany
| | - Andreas Lendlein
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany.,Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany.,Institute of Chemistry, University of Potsdam, Potsdam, Germany.,Helmholtz Virtual Institute - Multifunctional Biomaterials for Medicine, Teltow, Germany
| |
Collapse
|
77
|
Kojima K, Miyoshi H, Nagoshi N, Kohyama J, Itakura G, Kawabata S, Ozaki M, Iida T, Sugai K, Ito S, Fukuzawa R, Yasutake K, Renault‐Mihara F, Shibata S, Matsumoto M, Nakamura M, Okano H. Selective Ablation of Tumorigenic Cells Following Human Induced Pluripotent Stem Cell-Derived Neural Stem/Progenitor Cell Transplantation in Spinal Cord Injury. Stem Cells Transl Med 2019; 8:260-270. [PMID: 30485733 PMCID: PMC6392358 DOI: 10.1002/sctm.18-0096] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 10/12/2018] [Indexed: 12/29/2022] Open
Abstract
Tumorigenesis is an important problem that needs to be addressed in the field of human stem/progenitor cell transplantation for the treatment of subacute spinal cord injury (SCI). When certain "tumorigenic" cell lines are transplanted into the spinal cord of SCI mice model, there is initial improvement of motor function, followed by abrupt deterioration secondary to the effect of tumor growth. A significant proportion of the transplanted cells remains undifferentiated after transplantation and is thought to increase the risk of tumorigenesis. In this study, using lentiviral vectors, we introduced the herpes simplex virus type 1 thymidine kinase (HSVtk) gene into a human induced pluripotent stem cell-derived neural stem/progenitor cell (hiPSC-NS/PC) line that is known to undergo tumorigenic transformation. Such approach enables selective ablation of the immature proliferating cells and thereby prevents subsequent tumor formation. In vitro, the HSVtk system successfully ablated the immature proliferative neural cells while preserving mature postmitotic neuronal cells. Similar results were observed in vivo following transplantation into the injured spinal cords of immune-deficient (nonobese diabetic-severe combined immune-deficient) mice. Ablation of the proliferating cells exerted a protective effect on the motor function which was regained after transplantation, simultaneously defending the spinal cord from the harmful tumor growth. These results suggest a potentially promising role of suicide genes in opposing tumorigenesis during stem cell therapy. This system allows both preventing and treating tumorigenesis following hiPSC-NS/PC transplantation without sacrificing the improved motor function. Stem Cells Translational Medicine 2019;8:260&270.
Collapse
Affiliation(s)
- Kota Kojima
- Department of PhysiologyKeio University School of MedicineTokyoJapan
- Department of Orthopaedic SurgeryKeio University School of MedicineTokyoJapan
| | - Hiroyuki Miyoshi
- Department of PhysiologyKeio University School of MedicineTokyoJapan
| | - Narihito Nagoshi
- Department of Orthopaedic SurgeryKeio University School of MedicineTokyoJapan
| | - Jun Kohyama
- Department of PhysiologyKeio University School of MedicineTokyoJapan
| | - Go Itakura
- Department of PhysiologyKeio University School of MedicineTokyoJapan
- Department of Orthopaedic SurgeryKeio University School of MedicineTokyoJapan
| | - Soya Kawabata
- Department of PhysiologyKeio University School of MedicineTokyoJapan
- Department of Orthopaedic SurgeryKeio University School of MedicineTokyoJapan
| | - Masahiro Ozaki
- Department of PhysiologyKeio University School of MedicineTokyoJapan
- Department of Orthopaedic SurgeryKeio University School of MedicineTokyoJapan
| | - Tsuyoshi Iida
- Department of PhysiologyKeio University School of MedicineTokyoJapan
- Department of Orthopaedic SurgeryKeio University School of MedicineTokyoJapan
| | - Keiko Sugai
- Department of PhysiologyKeio University School of MedicineTokyoJapan
- Department of Orthopaedic SurgeryKeio University School of MedicineTokyoJapan
| | - Shuhei Ito
- Department of PhysiologyKeio University School of MedicineTokyoJapan
- Department of Orthopaedic SurgeryKeio University School of MedicineTokyoJapan
| | - Ryuji Fukuzawa
- Department of PathologyInternational University of Health and WelfareChibaJapan
| | - Kaori Yasutake
- Department of Orthopaedic SurgeryKeio University School of MedicineTokyoJapan
| | | | - Shinsuke Shibata
- Department of PhysiologyKeio University School of MedicineTokyoJapan
| | - Morio Matsumoto
- Department of Orthopaedic SurgeryKeio University School of MedicineTokyoJapan
| | - Masaya Nakamura
- Department of Orthopaedic SurgeryKeio University School of MedicineTokyoJapan
| | - Hideyuki Okano
- Department of PhysiologyKeio University School of MedicineTokyoJapan
| |
Collapse
|
78
|
Liu Q, Du J, Fan J, Li W, Guo W, Feng H, Lin J. Generation and Characterization of Induced Pluripotent Stem Cells from Mononuclear Cells in Schizophrenic Patients. CELL JOURNAL 2019; 21:161-168. [PMID: 30825289 PMCID: PMC6397609 DOI: 10.22074/cellj.2019.5871] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 07/01/2018] [Indexed: 12/17/2022]
Abstract
Objective Schizophrenia (SZ) is a mental disorder in which psychotic symptoms are the main problem. The
pathogenesis of SZ is not fully understood, partly because of limitations in current disease models and technology. The
development of induced pluripotent stem cell (iPSC) technology has opened up the possibility of elucidating disease
mechanisms in neurodegenerative diseases. Here, we aimed to obtain iPSCs from peripheral blood mononuclear cells
(PBMCs) of normal and schizophrenic individuals and analyze the inflammatory response in these iPSCs.
Materials and Methods In this experimental study, we isolated PBMCs from whole blood of healthy individuals and
SZ patients and reprogrammed them into iPSCs by transfection of recombinant lentiviruses that contained Yamanaka
factors (Oct4, Sox2, Klf4 and c-Myc). We calculated the numbers of iPSC clones and stained them with alkaline
phosphatase (ALP), Nanog, SSEA4, Nestin, Vimentin, and AFP to confirm their efficiency and pluripotency. The iPSCs
were analyzed by real-time quantitative polymerase chain reaction (qRT-PCR) for the expressions of inflammatory
factors.
Results iPSCs from schizophrenic patients (SZ-iPSCs) exhibited typical morphology and highly expressed pluripotent
markers. These iPSCs retained their normal karyotype and differentiated in vitro to form embryoid bodies (EBs) that
expressed markers of all 3 germ layers. However, iPSCs from the SZ-iPSCs group had a weak capacity to differentiate
into ectoderm compared to the normal iPSCs (Con-iPSC). An elevated, stronger inflammatory response existed in
iPSCs from schizophrenic individuals.
Conclusion We successfully obtained iPSCs from PBMCs of schizophrenic patients without genetic operation and analyzed
the expressions of pluripotent markers and inflammatory factors between the Con-iPSC and SZ-iPSC groups. Taken together,
our results may assist to explain the pathogenesis of SZ and develop new strategies for clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Qing Liu
- Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, Xinxiang, China.,College of Life Science and Technology, Xinxiang Medical University, Xinxiang, China.,Henan Key Lab of Biological Psychiatry, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Jiang Du
- Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, Xinxiang, China.,College of Biomedical Engineering, Xinxiang Medical University, Xinxiang, China
| | - Jinyu Fan
- Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, Xinxiang, China.,College of Life Science and Technology, Xinxiang Medical University, Xinxiang, China
| | - Wenqiang Li
- Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, Xinxiang, China.,Henan Key Lab of Biological Psychiatry, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Weiyun Guo
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, China
| | - Huigen Feng
- Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, Xinxiang, China.,College of Life Science and Technology, Xinxiang Medical University, Xinxiang, China.Electronic Address:
| | - Juntang Lin
- Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, Xinxiang, China.,College of Biomedical Engineering, Xinxiang Medical University, Xinxiang, China.Electronic Address:
| |
Collapse
|
79
|
Increased Cytotoxicity of Herpes Simplex Virus Thymidine Kinase Expression in Human Induced Pluripotent Stem Cells. Int J Mol Sci 2019; 20:ijms20040810. [PMID: 30769780 PMCID: PMC6413063 DOI: 10.3390/ijms20040810] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 02/07/2019] [Accepted: 02/11/2019] [Indexed: 02/07/2023] Open
Abstract
Human induced pluripotent stem cells (iPSCs) hold enormous promise for regenerative medicine. The major safety concern is the tumorigenicity of transplanted cells derived from iPSCs. A potential solution would be to introduce a suicide gene into iPSCs as a safety switch. The herpes simplex virus type 1 thymidine kinase (HSV-TK) gene, in combination with ganciclovir, is the most widely used enzyme/prodrug suicide system from basic research to clinical applications. In the present study, we attempted to establish human iPSCs that stably expressed HSV-TK with either lentiviral vectors or CRISPR/Cas9-mediated genome editing. However, this task was difficult to achieve, because high-level and/or constitutive expression of HSV-TK resulted in the induction of cell death or silencing of HSV-TK expression. A nucleotide metabolism analysis suggested that excessive accumulation of thymidine triphosphate, caused by HSV-TK expression, resulted in an imbalance in the dNTP pools. This unbalanced state led to DNA synthesis inhibition and cell death in a process similar to a “thymidine block”, but more severe. We also demonstrated that the Tet-inducible system was a feasible solution for overcoming the cytotoxicity of HSV-TK expression. Our results provided a warning against using the HSV-TK gene in human iPSCs, particularly in clinical applications.
Collapse
|
80
|
Ishikawa KI, Yamaguchi A, Okano H, Akamatsu W. Assessment of Mitophagy in iPS Cell-Derived Neurons. Methods Mol Biol 2019; 1759:59-67. [PMID: 28324490 DOI: 10.1007/7651_2017_10] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2023]
Abstract
Aberrant mitochondrial function is associated with many neurological diseases. Mitophagy is a key mechanism for the elimination of damaged mitochondria and maintenance of mitochondrial homeostasis. Induced pluripotent stem (iPS) cell technologies developed over the last decade have allowed us to analyze functions of the human neuron. Here we describe an efficient induction method from human iPS cells to neurons, followed by an image-based mitophagy assay.
Collapse
Affiliation(s)
- Kei-Ichi Ishikawa
- Department of Neurology, Juntendo University School of Medicine, Bunkyo, Tokyo, 113-8421, Japan
| | - Akihiro Yamaguchi
- Center for Genomic and Regenerative Medicine, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo, Tokyo, 113-8421, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Shinjuku, Tokyo, 160-8582, Japan
| | - Wado Akamatsu
- Center for Genomic and Regenerative Medicine, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo, Tokyo, 113-8421, Japan.
| |
Collapse
|
81
|
Hosoya M, Saeki T, Saegusa C, Matsunaga T, Okano H, Fujioka M, Ogawa K. Estimating the concentration of therapeutic range using disease-specific iPS cells: Low-dose rapamycin therapy for Pendred syndrome. Regen Ther 2018; 10:54-63. [PMID: 30581897 PMCID: PMC6299162 DOI: 10.1016/j.reth.2018.11.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 10/28/2018] [Accepted: 11/12/2018] [Indexed: 01/31/2023] Open
Abstract
Introduction Pendred syndrome is an autosomal-recessive disease characterized by congenital hearing loss and thyroid goiter. Previously, cell stress susceptibilities were shown to increase in patient-derived cells with intracellular aggregation using an in vitro acute cochlear cell model derived from patient-specific pluripotent stem (iPS) cells. Moreover, we showed that rapamycin can relieve cell death. However, studies regarding long-term cell survival without cell stressors that mimic the natural course of disease or the rational minimum concentration of rapamycin that prevents cell death are missing. Methods In this report, we first investigated the rational minimum concentration of rapamycin using patient-specific iPS cells derived-cochlear cells with three different conditions of acute stress. We next confirmed the effects of rapamycin in long-term cell survival and phenotypes by using cochlear cells derived from three different patient-derived iPS cells. Results We found that inner ear cells derived from Pendred syndrome patients are more vulnerable than those from healthy individuals during long-term culturing; however, this susceptibility was relieved via treatment with low-dose rapamycin. The slow progression of hearing loss in patients may be explained, in part, by the vulnerability observed in patient cells during long-term culturing. We successfully evaluated the rational minimum concentration of rapamycin for treatment of Pendred syndrome. Conclusion Our results suggest that low-dose rapamycin not only decreases acute symptoms but may prevent progression of hearing loss in Pendred syndrome patients. In vitro chronic disorder model of Pendred syndrome is established. The vulnerability observed during long-term culturing explains progression of PDS. Low-dose rapamycin relief the cell vulnerability observed in PDS patients. PDS iPSCs reveal a rational treatment strategy for chronic progressive hearing loss.
Collapse
Affiliation(s)
- Makoto Hosoya
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, 35 Shinanomachi Shinjuku-ku, Tokyo 160-8582, Japan
| | - Tsubasa Saeki
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi Shinjuku-ku, Tokyo 160-8582, Japan
| | - Chika Saegusa
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, 35 Shinanomachi Shinjuku-ku, Tokyo 160-8582, Japan
| | - Tatsuo Matsunaga
- The Laboratory of Auditory Disorders and Division of Hearing and Balance Research, National Institute of Sensory Organs, National Tokyo Medical Center, 2-5-1 Higashigaoka, Meguro-ku, Tokyo 152-8902, Japan.,Medical Genetics Center, National Tokyo Medical Center, 2-5-1 Higashigaoka, Meguro-ku, Tokyo 152-8902, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi Shinjuku-ku, Tokyo 160-8582, Japan
| | - Masato Fujioka
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, 35 Shinanomachi Shinjuku-ku, Tokyo 160-8582, Japan
| | - Kaoru Ogawa
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, 35 Shinanomachi Shinjuku-ku, Tokyo 160-8582, Japan
| |
Collapse
|
82
|
Tsuji O, Sugai K, Yamaguchi R, Tashiro S, Nagoshi N, Kohyama J, Iida T, Ohkubo T, Itakura G, Isoda M, Shinozaki M, Fujiyoshi K, Kanemura Y, Yamanaka S, Nakamura M, Okano H. Concise Review: Laying the Groundwork for a First-In-Human Study of an Induced Pluripotent Stem Cell-Based Intervention for Spinal Cord Injury. Stem Cells 2018; 37:6-13. [PMID: 30371964 PMCID: PMC7379555 DOI: 10.1002/stem.2926] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 08/16/2018] [Accepted: 09/22/2018] [Indexed: 12/21/2022]
Abstract
There have been numerous attempts to develop stem cell transplantation approaches to promote the regeneration of spinal cord injury (SCI). Our multicenter team is currently planning to launch a first-in-human clinical study of an induced pluripotent stem cell (iPSC)-based cell transplant intervention for subacute SCI. This trial was conducted as class I regenerative medicine protocol as provided for under Japan's Act on the Safety of Regenerative Medicine, using neural stem/progenitor cells derived from a clinical-grade, integration-free human "iPSC stock" generated by the Kyoto University Center for iPS Cell Research and Application. In the present article, we describe how we are preparing to initiate this clinical study, including addressing the issues of safety and tumorigenesis as well as practical problems that must be overcome to enable the development of therapeutic interventions for patients with chronic SCI. Stem Cells 2019;37:6-13.
Collapse
Affiliation(s)
- Osahiko Tsuji
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Keiko Sugai
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Ryo Yamaguchi
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan.,Department of Physiology, Keio University School of Medicine, Tokyo, Japan.,Regenerative & Cellular Medicine Office, Sumitomo Dainippon Pharma Co., Ltd., Kobe, Japan
| | - Syoichi Tashiro
- Department of Rehabilitation Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Narihito Nagoshi
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Jun Kohyama
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Tsuyoshi Iida
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan.,Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Toshiki Ohkubo
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan.,Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Go Itakura
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan.,Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Miho Isoda
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan.,Regenerative & Cellular Medicine Office, Sumitomo Dainippon Pharma Co., Ltd., Kobe, Japan
| | - Munehisa Shinozaki
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Kanehiro Fujiyoshi
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan.,Department of Orthopaedic Surgery, National Hospital Organization Murayama Medical Center, Tokyo, Japan
| | - Yonehiro Kanemura
- Department of Biomedical Research and Innovation, Institute for Clinical Research and Department of Neurosurgery, Osaka National Hospital, National Hospital Organization, Osaka, Japan
| | - Shinya Yamanaka
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Masaya Nakamura
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
83
|
Hatami M, Conrad S, Naghsh P, Alvarez-Bolado G, Skutella T. Cell-Biological Requirements for the Generation of Dentate Gyrus Granule Neurons. Front Cell Neurosci 2018; 12:402. [PMID: 30483057 PMCID: PMC6240695 DOI: 10.3389/fncel.2018.00402] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 10/18/2018] [Indexed: 12/22/2022] Open
Abstract
The dentate gyrus (DG) receives highly processed information from the associative cortices functionally integrated in the trisynaptic hippocampal circuit, which contributes to the formation of new episodic memories and the spontaneous exploration of novel environments. Remarkably, the DG is the only brain region currently known to have high rates of neurogenesis in adults (Andersen et al., 1966, 1971). The DG is involved in several neurodegenerative disorders, including clinical dementia, schizophrenia, depression, bipolar disorder and temporal lobe epilepsy. The principal neurons of the DG are the granule cells. DG granule cells generated in culture would be an ideal model to investigate their normal development and the causes of the pathologies in which they are involved and as well as possible therapies. Essential to establish such in vitro models is the precise definition of the most important cell-biological requirements for the differentiation of DG granule cells. This requires a deeper understanding of the precise molecular and functional attributes of the DG granule cells in vivo as well as the DG cells derived in vitro. In this review we outline the neuroanatomical, molecular and cell-biological components of the granule cell differentiation pathway, including some growth- and transcription factors essential for their development. We summarize the functional characteristics of DG granule neurons, including the electrophysiological features of immature and mature granule cells and the axonal pathfinding characteristics of DG neurons. Additionally, we discuss landmark studies on the generation of dorsal telencephalic precursors from pluripotent stem cells (PSCs) as well as DG neuron differentiation in culture. Finally, we provide an outlook and comment critical aspects.
Collapse
Affiliation(s)
- Maryam Hatami
- Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | | | - Pooyan Naghsh
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
| | | | - Thomas Skutella
- Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
84
|
Hirayama M. Advances in Functional Restoration of the Lacrimal Glands. Invest Ophthalmol Vis Sci 2018; 59:DES174-DES182. [PMID: 30481824 DOI: 10.1167/iovs.17-23528] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The lacrimal glands produce tears to support a healthy homeostatic environment on the ocular surface. The lacrimal gland dysfunction characteristic of dry eye disease causes ocular discomfort and visual disturbances and in severe cases can result in a loss of vision. The demand for adequate restoration of lacrimal gland function has been intensified due to advances in stem cell biology, developmental biology, and bioengineering technologies. In addition to conventional therapies, including artificial tears, tear alternatives (such as autologous serum eye drops) and salivary gland transplantation, a regenerative medicine approach has been identified as a novel strategy to restore the function of the lacrimal gland. Recent studies have demonstrated the potential of progenitor cell injection therapy to repair the tissue of the lacrimal glands. A current three-dimensional (3D) tissue engineering technique has been shown to regenerate a secretory gland structure by reproducing reciprocal epithelial-mesenchymal interactions during ontogenesis in vitro and in vivo. A novel direct reprogramming method has suggested a possibility to induce markers in the lacrimal gland developmental process from human pluripotent stem cells. The development of this method is supported by advances in our understanding of gene expression and regulatory networks involved in the development and differentiation of the lacrimal glands. Engineering science has proposed a medical device to stimulate tearing and a bio-hybrid scaffold to reconstruct the 3D lacrimal gland structure. In this review, we will summarize recent bioengineering advances in lacrimal gland regeneration toward the functional restoration of the lacrimal glands as a future dry eye therapy.
Collapse
Affiliation(s)
- Masatoshi Hirayama
- Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan.,Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, California, United States
| |
Collapse
|
85
|
Tabata Y, Imaizumi Y, Sugawara M, Andoh-Noda T, Banno S, Chai M, Sone T, Yamazaki K, Ito M, Tsukahara K, Saya H, Hattori N, Kohyama J, Okano H. T-type Calcium Channels Determine the Vulnerability of Dopaminergic Neurons to Mitochondrial Stress in Familial Parkinson Disease. Stem Cell Reports 2018; 11:1171-1184. [PMID: 30344006 PMCID: PMC6234903 DOI: 10.1016/j.stemcr.2018.09.006] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Revised: 09/17/2018] [Accepted: 09/18/2018] [Indexed: 12/11/2022] Open
Abstract
Parkinson disease (PD) is a progressive neurological disease caused by selective degeneration of dopaminergic (DA) neurons in the substantia nigra. Although most cases of PD are sporadic cases, familial PD provides a versatile research model for basic mechanistic insights into the pathogenesis of PD. In this study, we generated DA neurons from PARK2 patient-specific, isogenic PARK2 null and PARK6 patient-specific induced pluripotent stem cells and found that these neurons exhibited more apoptosis and greater susceptibility to rotenone-induced mitochondrial stress. From phenotypic screening with an FDA-approved drug library, one voltage-gated calcium channel antagonist, benidipine, was found to suppress rotenone-induced apoptosis. Furthermore, we demonstrated the dysregulation of calcium homeostasis and increased susceptibility to rotenone-induced stress in PD, which is prevented by T-type calcium channel knockdown or antagonists. These findings suggest that calcium homeostasis in DA neurons might be a useful target for developing new drugs for PD patients. Patient-derived DA neurons recapitulate several PD-related disease phenotypes Establishment of a system for drug screening against PD using patient-derived cells Calcium channel antagonists suppress rotenone-induced apoptosis in PARK2 DA neurons The involvement of dysregulated T-type calcium channels in the progression of PD
Collapse
Affiliation(s)
- Yoshikuni Tabata
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; Tsukuba Research Laboratories, Eisai Co., Ltd, 5-1-3 Tokodai, Tsukuba-shi, Ibaraki 300-2635, Japan
| | - Yoichi Imaizumi
- Tsukuba Research Laboratories, Eisai Co., Ltd, 5-1-3 Tokodai, Tsukuba-shi, Ibaraki 300-2635, Japan
| | - Michiko Sugawara
- Tsukuba Research Laboratories, Eisai Co., Ltd, 5-1-3 Tokodai, Tsukuba-shi, Ibaraki 300-2635, Japan
| | - Tomoko Andoh-Noda
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Satoe Banno
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - MuhChyi Chai
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Takefumi Sone
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Kazuto Yamazaki
- Tsukuba Research Laboratories, Eisai Co., Ltd, 5-1-3 Tokodai, Tsukuba-shi, Ibaraki 300-2635, Japan
| | - Masashi Ito
- Tsukuba Research Laboratories, Eisai Co., Ltd, 5-1-3 Tokodai, Tsukuba-shi, Ibaraki 300-2635, Japan
| | - Kappei Tsukahara
- Tsukuba Research Laboratories, Eisai Co., Ltd, 5-1-3 Tokodai, Tsukuba-shi, Ibaraki 300-2635, Japan
| | - Hideyuki Saya
- Division of Gene Regulation, Institute for Advanced Medical Research, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Jun Kohyama
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan.
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan.
| |
Collapse
|
86
|
Lin H, Du Q, Li Q, Wang O, Wang Z, Liu K, Elowsky C, Zhang C, Lei Y. Hydrogel-Based Bioprocess for Scalable Manufacturing of Human Pluripotent Stem Cell-Derived Neural Stem Cells. ACS APPLIED MATERIALS & INTERFACES 2018; 10:29238-29250. [PMID: 30091584 DOI: 10.1021/acsami.8b05780] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Neural stem cells derived from human pluripotent stem cells (hPSC-NSCs) are of great value for modeling diseases, developing drugs, and treating neurological disorders. However, manufacturing high-quantity and -quality hPSC-NSCs, especially for clinical applications, remains a challenge. Here, we report a chemically defined, high-yield, and scalable bioprocess for manufacturing hPSC-NSCs. hPSCs are expanded and differentiated into NSCs in microscale tubes made with alginate hydrogels. The tubes are used to isolate cells from the hydrodynamic stresses in the culture vessel and limit the radial diameter of the cell mass to less than 400 μm to ensure efficient mass transport during the culture. The hydrogel tubes provide uniform, reproducible, and cell-friendly microspaces and microenvironments for cells. With this new technology, we showed that hPSC-NSCs could be produced in 12 days with high viability (∼95%), high purity (>90%), and high yield (∼5 × 108 cells/mL of microspace). The volumetric yield is about 250 times more than the current state-of-the-art. Whole transcriptome analysis and quantitative real-time polymerase chain reaction showed that hPSC-NSCs made by this process had a similar gene expression to hPSC-NSCs made by the conventional culture technology. The produced hPSC-NSCs could mature into both neurons and glial cells in vitro and in vivo. The process developed in this paper can be used to produce large numbers of hPSC-NSCs for various biomedical applications in the future.
Collapse
Affiliation(s)
| | | | | | | | - Zhanqi Wang
- Department of Vascular Surgery, Beijing Anzhen Hospital of Capital Medical University , Beijing Institute of Heart Lung and Blood Vessel Diseases , Beijing 100029 , China
| | | | | | | | | |
Collapse
|
87
|
Modeling sporadic ALS in iPSC-derived motor neurons identifies a potential therapeutic agent. Nat Med 2018; 24:1579-1589. [PMID: 30127392 DOI: 10.1038/s41591-018-0140-5] [Citation(s) in RCA: 244] [Impact Index Per Article: 34.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 07/11/2018] [Indexed: 12/21/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a heterogeneous motor neuron disease for which no effective treatment is available, despite decades of research into SOD1-mutant familial ALS (FALS). The majority of ALS patients have no familial history, making the modeling of sporadic ALS (SALS) essential to the development of ALS therapeutics. However, as mutations underlying ALS pathogenesis have not yet been identified, it remains difficult to establish useful models of SALS. Using induced pluripotent stem cell (iPSC) technology to generate stem and differentiated cells retaining the patients' full genetic information, we have established a large number of in vitro cellular models of SALS. These models showed phenotypic differences in their pattern of neuronal degeneration, types of abnormal protein aggregates, cell death mechanisms, and onset and progression of these phenotypes in vitro among cases. We therefore developed a system for case clustering capable of subdividing these heterogeneous SALS models by their in vitro characteristics. We further evaluated multiple-phenotype rescue of these subclassified SALS models using agents selected from non-SOD1 FALS models, and identified ropinirole as a potential therapeutic candidate. Integration of the datasets acquired in this study permitted the visualization of molecular pathologies shared across a wide range of SALS models.
Collapse
|
88
|
Stem cells purified from human induced pluripotent stem cell-derived neural crest-like cells promote peripheral nerve regeneration. Sci Rep 2018; 8:10071. [PMID: 29968745 PMCID: PMC6030210 DOI: 10.1038/s41598-018-27952-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 06/13/2018] [Indexed: 12/12/2022] Open
Abstract
Strategies for therapeutic cell transplantation have been assessed for use in the treatment of massive peripheral nerve defects. To support safe and efficient cell transplantation, we have focused on the purification of cells using cell surface markers. Our group previously reported low-affinity nerve growth factor receptor (LNGFR)- and thymocyte antigen-1 (THY-1)-positive neural crest-like cells (LT-NCLCs), generated from human induced pluripotent stem cells (hiPSCs). In the present study, we investigated the efficacy of transplantation of hiPSC-derived LT-NCLCs in a murine massive peripheral nerve defect model. Animals with a sciatic nerve defect were treated with a bridging silicone tube prefilled with LT-NCLCs or medium in the transplantation (TP) and negative control (NC) groups, respectively. The grafted LT-NCLCs survived and enhanced myelination and angiogenesis, as compared to the NC group. Behavioral analysis indicated that motor functional recovery in the TP group was superior to that in the NC group, and similar to that in the autograft (Auto) group. LT-NCLCs promoted axonal regrowth and remyelination by Schwann cells. Transplantation of LT-NCLCs is a promising approach for nerve regeneration treatment of massive peripheral nerve defects.
Collapse
|
89
|
O'Day E, Hosta-Rigau L, Oyarzún DA, Okano H, de Lorenzo V, von Kameke C, Alsafar H, Cao C, Chen GQ, Ji W, Roberts RJ, Ronaghi M, Yeung K, Zhang F, Lee SY. Are We There Yet? How and When Specific Biotechnologies Will Improve Human Health. Biotechnol J 2018; 14:e1800195. [PMID: 29799175 DOI: 10.1002/biot.201800195] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 05/11/2018] [Indexed: 12/11/2022]
Abstract
Patient X: A 67-year-old Caucasian man slips on a patch of ice. He has abrasions to his hands and has sustained significant damage to his hip. At the emergency room, he informs clinicians he takes atorvastatin, metformin, and glimepiride to treat hypertension and Type 2 Diabetes Mellitus (T2DM). X-rays reveal a fractured hip, which will require total hip replacement surgery.
Collapse
Affiliation(s)
- Elizabeth O'Day
- Global Future Council on the Future of Biotechnologies, World Economic Forum, Cologny, CH-1223, Geneva, Switzerland.,Olaris Therapeutics, Inc., 45 Moulton St., Cambridge, MA, 02138, USA
| | - Leticia Hosta-Rigau
- Global Future Council on the Future of Biotechnologies, World Economic Forum, Cologny, CH-1223, Geneva, Switzerland.,Department of Micro- and Nanotechnology, Center for Nanomedicine and Theranostics, Technical University of Denmark, 2800, Kongens Lyngby, Denmark
| | - Diego A Oyarzún
- Global Future Council on the Future of Biotechnologies, World Economic Forum, Cologny, CH-1223, Geneva, Switzerland.,Department of Mathematics, Imperial College London, London, SW7 2AZ, UK.,EPSRC Centre for Mathematics of Precision Healthcare, Imperial College London, London, SW7 2AZ, UK
| | - Hideyuki Okano
- Global Future Council on the Future of Biotechnologies, World Economic Forum, Cologny, CH-1223, Geneva, Switzerland.,Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Víctor de Lorenzo
- Global Future Council on the Future of Biotechnologies, World Economic Forum, Cologny, CH-1223, Geneva, Switzerland.,National Center of Biotechnology CSIC, Systems Biology Program, Campus de Cantoblanco, E-28049, Madrid, Spain
| | - Conrad von Kameke
- Global Future Council on the Future of Biotechnologies, World Economic Forum, Cologny, CH-1223, Geneva, Switzerland.,BioInnovators Europe, Berlin, Germany
| | - Habiba Alsafar
- Global Future Council on the Future of Biotechnologies, World Economic Forum, Cologny, CH-1223, Geneva, Switzerland.,Khalifa University Center for Biotechnology, Khalifa University, PO Box 127788, Abu Dhabi, United Arab Emirates
| | - Cong Cao
- Global Future Council on the Future of Biotechnologies, World Economic Forum, Cologny, CH-1223, Geneva, Switzerland.,University of Nottingham, 199 East Taikang Road, Ningbo, 315100, China
| | - Guo-Qiang Chen
- Global Future Council on the Future of Biotechnologies, World Economic Forum, Cologny, CH-1223, Geneva, Switzerland.,Center for Synthetic and Systems Biology, MOE Lab for Industrial Biocatalysis, Tsinghua-Peking University Center of Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Weizhi Ji
- Global Future Council on the Future of Biotechnologies, World Economic Forum, Cologny, CH-1223, Geneva, Switzerland.,Kunming University of Science and Technology, 727 Jingming South Rd. Chenh Gong, Kunming, 650500, Yunnan, China
| | - Richard J Roberts
- Global Future Council on the Future of Biotechnologies, World Economic Forum, Cologny, CH-1223, Geneva, Switzerland.,New England Biolabs, 240 County Road, Ipswich, MA, 01938, USA
| | - Mostafa Ronaghi
- Global Future Council on the Future of Biotechnologies, World Economic Forum, Cologny, CH-1223, Geneva, Switzerland.,Illumina Inc., 5200 Illumina Way, San Diego, CA, 92121, USA
| | - Karen Yeung
- Global Future Council on the Future of Biotechnologies, World Economic Forum, Cologny, CH-1223, Geneva, Switzerland.,Law School and School of Computer Science University of Birmingham, Birmingham, UK, B15 2TT
| | - Feng Zhang
- Global Future Council on the Future of Biotechnologies, World Economic Forum, Cologny, CH-1223, Geneva, Switzerland.,Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA.,McGovern Institute for Brain Research at MIT, Cambridge, MA, 02139, USA.,Department of Brain and Cognitive Sciences and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Sang Yup Lee
- Global Future Council on the Future of Biotechnologies, World Economic Forum, Cologny, CH-1223, Geneva, Switzerland.,Department of Chemical and Biomolecular Engineering (BK21 Plus program), Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-Ro, Daejeon, 34141, Republic of Korea.,The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kemitorvet Bygning 220, 2800, Kongens Lyngby, Denmark
| |
Collapse
|
90
|
Petrella RA, Mollica PA, Zamponi M, Reid JA, Xiao S, Bruno RD, Sachs PC. 3D bioprinter applied picosecond pulsed electric fields for targeted manipulation of proliferation and lineage specific gene expression in neural stem cells. J Neural Eng 2018; 15:056021. [PMID: 29848804 DOI: 10.1088/1741-2552/aac8ec] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Picosecond pulse electric fields (psPEF) have the potential to elicit functional changes in mammalian cells in a non-contact manner. Such electro-manipulation of pluripotent and multipotent cells could be a tool in both neural interface and tissue engineering. Here, we describe the potential of psPEF in directing neural stem cells (NSCs) gene expression, metabolism, and proliferation. As a comparison mesenchymal stem cells (MSCs) were also tested. APPROACH A psPEF electrode was anchored on a customized commercially available 3D printer, which allowed us to deliver pulses with high spatial precision and systematically control the electrode position in three-axes. When the electrodes are continuously energized and their position is shifted by the 3D printer, large numbers of cells on a surface can be exposed to a uniform psPEF. With two electric field strengths (20 and 40 kV cm-1), cell responses, including cell viability, proliferation, and gene expression assays, were quantified and analyzed. MAIN RESULTS Analysis revealed both NSCs and MSCs showed no significant cell death after treatments. Both cell types exhibited an increased metabolic reduction; however, the response rate for MSCs was sensitive to the change of electric field strength, but for NSCs, it appeared independent of electric field strength. The change in proliferation rate was cell-type specific. MSCs underwent no significant change in proliferation whereas NSCs exhibited an electric field dependent response with the higher electric field producing less proliferation. Further, NSCs showed an upregulation of glial fibrillary acidic protein (GFAP) after 24 h to 40 kV cm-1, which is characteristic of astrocyte specific differentiation. SIGNIFICANCE Changes in cell metabolism, proliferation, and gene expression after picosecond pulsed electric field exposure are cell type specific.
Collapse
Affiliation(s)
- Ross A Petrella
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, Virginia, 23529, United States of America. Department of Electrical and Computer Engineering, Old Dominion University, Norfolk, Virginia, 23529, United States of America
| | | | | | | | | | | | | |
Collapse
|
91
|
Rostrocaudal Areal Patterning of Human PSC-Derived Cortical Neurons by FGF8 Signaling. eNeuro 2018; 5:eN-NWR-0368-17. [PMID: 29707616 PMCID: PMC5917473 DOI: 10.1523/eneuro.0368-17.2018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 04/08/2018] [Accepted: 04/09/2018] [Indexed: 12/11/2022] Open
Abstract
The cerebral cortex is subdivided into distinct areas that have particular functions. The rostrocaudal (R-C) gradient of fibroblast growth factor 8 (FGF8) signaling defines this areal identity during neural development. In this study, we recapitulated cortical R-C patterning in human pluripotent stem cell (PSC) cultures. Modulation of FGF8 signaling appropriately regulated the R-C markers, and the patterns of global gene expression resembled those of the corresponding areas of human fetal brains. Furthermore, we demonstrated the utility of this culture system in modeling the area-specific forebrain phenotypes [presumptive upper motor neuron (UMN) phenotypes] of amyotrophic lateral sclerosis (ALS). We anticipate that our culture system will contribute to studies of human neurodevelopment and neurological disease modeling.
Collapse
|
92
|
Nagoshi N, Okano H. iPSC-derived neural precursor cells: potential for cell transplantation therapy in spinal cord injury. Cell Mol Life Sci 2018; 75:989-1000. [PMID: 28993834 PMCID: PMC11105708 DOI: 10.1007/s00018-017-2676-9] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2016] [Revised: 09/03/2017] [Accepted: 10/02/2017] [Indexed: 12/12/2022]
Abstract
A number of studies have demonstrated that transplantation of neural precursor cells (NPCs) promotes functional recovery after spinal cord injury (SCI). However, the NPCs had been mostly harvested from embryonic stem cells or fetal tissue, raising the ethical concern. Yamanaka and his colleagues established induced pluripotent stem cells (iPSCs) which could be generated from somatic cells, and this innovative development has made rapid progression in the field of SCI regeneration. We and other groups succeeded in producing NPCs from iPSCs, and demonstrated beneficial effects after transplantation for animal models of SCI. In particular, efficacy of human iPSC-NPCs in non-human primate SCI models fostered momentum of clinical application for SCI patients. At the same time, however, artificial induction methods in iPSC technology created alternative issues including genetic and epigenetic abnormalities, and tumorigenicity after transplantation. To overcome these problems, it is critically important to select origins of somatic cells, use integration-free system during transfection of reprogramming factors, and thoroughly investigate the characteristics of iPSC-NPCs with respect to quality management. Moreover, since most of the previous studies have focused on subacute phase of SCI, establishment of effective NPC transplantation should be evaluated for chronic phase hereafter. Our group is currently preparing clinical-grade human iPSC-NPCs, and will move forward toward clinical study for subacute SCI patients soon in the near future.
Collapse
Affiliation(s)
- Narihito Nagoshi
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjukuku, Tokyo, 160-8582, Japan.
| |
Collapse
|
93
|
Suda Y, Kuzumaki N, Sone T, Narita M, Tanaka K, Hamada Y, Iwasawa C, Shibasaki M, Maekawa A, Matsuo M, Akamatsu W, Hattori N, Okano H, Narita M. Down-regulation of ghrelin receptors on dopaminergic neurons in the substantia nigra contributes to Parkinson's disease-like motor dysfunction. Mol Brain 2018; 11:6. [PMID: 29458391 PMCID: PMC5819262 DOI: 10.1186/s13041-018-0349-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 01/22/2018] [Indexed: 12/21/2022] Open
Abstract
Ghrelin exerts a wide range of physiological actions throughout the body and appears to be a promising target for disease therapy. Endogenous ghrelin receptors (GHSRs) are present in extrahypothalamic sites including the substantia nigra pars compacta (SNc), which is related to phenotypic dysregulation or frank degeneration in Parkinson’s disease (PD). Here we found a dramatic decrease in the expression of GHSR in PD-specific induced pluripotent stem cell (iPSC)-derived dopaminergic (DAnergic) neurons generated from patients carrying parkin gene (PARK2) mutations compared to those from healthy controls. Consistently, a significant decrease in the expression of GHSR was found in DAnergic neurons of isogenic PARK2-iPSC lines that mimicked loss of function of the PARK2 gene through CRISPR Cas9 technology. Furthermore, either intracerebroventricular injection or microinjection into the SNc of the selective GHSR1a antagonist [D-Lys3]-GHRP6 in normal mice produced cataleptic behaviors related to dysfunction of motor coordination. These findings suggest that the down-regulation of GHSRs in SNc-DA neurons induced the initial dysfunction of DA neurons, leading to extrapyramidal disorder under PD.
Collapse
Affiliation(s)
- Yukari Suda
- Department of Pharmacology, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Ebara, Shinagawa-ku, Tokyo, 142-8501, Japan
| | - Naoko Kuzumaki
- Department of Pharmacology, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Ebara, Shinagawa-ku, Tokyo, 142-8501, Japan. .,Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| | - Takefumi Sone
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Michiko Narita
- Department of Pharmacology, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Ebara, Shinagawa-ku, Tokyo, 142-8501, Japan
| | - Kenichi Tanaka
- Department of Pharmacology, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Ebara, Shinagawa-ku, Tokyo, 142-8501, Japan
| | - Yusuke Hamada
- Department of Pharmacology, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Ebara, Shinagawa-ku, Tokyo, 142-8501, Japan
| | - Chizuru Iwasawa
- Department of Pharmacology, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Ebara, Shinagawa-ku, Tokyo, 142-8501, Japan
| | - Masahiro Shibasaki
- Department of Pharmacology, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Ebara, Shinagawa-ku, Tokyo, 142-8501, Japan
| | - Aya Maekawa
- Laboratory of Molecular Genetics, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| | - Miri Matsuo
- Department of Pharmacology, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Ebara, Shinagawa-ku, Tokyo, 142-8501, Japan
| | - Wado Akamatsu
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.,Center for Genomic and Regenerative Medicine, Juntendo University, School of Medicine, Bunkyo-ku, Tokyo, 113-8431, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University School of Medicine, Bunkyo, Tokyo, 113-8421, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan. .,Life Science Tokyo Advanced Research Center (L-StaR), Hoshi University School of Pharmacy and Pharmaceutical Sciences, Ebara, Shinagawa-ku, Tokyo, 142-8501, Japan.
| | - Minoru Narita
- Department of Pharmacology, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Ebara, Shinagawa-ku, Tokyo, 142-8501, Japan. .,Life Science Tokyo Advanced Research Center (L-StaR), Hoshi University School of Pharmacy and Pharmaceutical Sciences, Ebara, Shinagawa-ku, Tokyo, 142-8501, Japan.
| |
Collapse
|
94
|
Shen H, Yao X, Li H, Li X, Zhang T, Sun Q, Ji C, Chen G. Role of Exosomes Derived from miR-133b Modified MSCs in an Experimental Rat Model of Intracerebral Hemorrhage. J Mol Neurosci 2018; 64:421-430. [PMID: 29455449 DOI: 10.1007/s12031-018-1041-2] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Accepted: 02/06/2018] [Indexed: 12/11/2022]
Abstract
Intracerebral hemorrhage (ICH) has poor outcomes due to high mortality and morbidity, but until now, the effective treatments remain limited. MicroRNAs (miRNAs) are vital regulators of gene expression and demonstrated to be linked to the pathogenesis of various central nervous system (CNS) diseases. Exosomes are considered as cell-to-cell communication vectors and secreted largely by mesenchymal stromal cells (MSCs). The present study investigated the role of miR-133b delivered by exosomes secreted from MSCs to brain tissues in rats after ICH. An autologous arterial blood ICH model in adult male Sprague-Dawley (SD) rats was used in this study. At 72 h after transfection with miR-133b mimics in MSCs, miR-133b-modified MSC-derived exosomes were collected from medium of MSCs and then injected to rats via tail vein. The levels of miR-133b in secreted exosomes and brain tissues of rats in various groups and the levels of RhoA, phosphorylations of extracellular signal regulating kinase (ERK1/2), and cAMP response element-binding protein (CREB) were detected by real-time PCR and western blot analysis, respectively. The effects of miR-133b on neuronal apoptosis and degeneration were respectively evaluated by TUNEL and fluoro-jade B staining. The miR-133b levels were reduced in brain tissues of rats at 24 h and peaked at 72 h after ICH. At 24 h after miR-133b-modified exosome administration, the level of miR-133b was significantly increased, while the apoptotic and neurodegenerative neurons were obviously reduced in brain tissues after ICH. The results of western blot analysis showed that miR-133b modified exosomes treatment remarkably suppressed RhoA expression and activated ERK1/2/CREB in brain tissues after ICH. Collectively, our investigation suggested that exosomes derived from miR-133b modified MSCs exhibited neuroprotective role for anti-apoptotic effect of miR-133b mediating RhoA and ERK1/2/CREB in rats after ICH.
Collapse
Affiliation(s)
- Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China
| | - Xiyang Yao
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China
| | - Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China
| | - Tiejun Zhang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China
| | - Qing Sun
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China.
| | - Chengyuan Ji
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China.
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China
| |
Collapse
|
95
|
Neural Stem Cells Derived from Human-Induced Pluripotent Stem Cells and Their Use in Models of CNS Injury. Results Probl Cell Differ 2018; 66:89-102. [PMID: 30209655 DOI: 10.1007/978-3-319-93485-3_3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Induced pluripotent stem (iPS) cells are derived from differentiated cells by different reprogramming techniques, by introducing specific transcription factors responsible for pluripotency. Induced pluripotent stem cells can serve as an excellent source for differentiated neural stem/progenitor cells (NSCs/NPs). Several methods and protocols are utilized to create a robust number of NSCs/NPs without jeopardizing the safety issues required for in vivo applications. A variety of disease-specific iPS cells have been used to study nervous system diseases. In this chapter, we will focus on some of the derivation and differentiation approaches and the application of iPS-NPs in the treatment of spinal cord injury and stroke.
Collapse
|
96
|
Generation of Induced Pluripotent Stem Cells and Neural Stem/Progenitor Cells from Newborns with Spina Bifida Aperta. Asian Spine J 2017; 11:870-879. [PMID: 29279741 PMCID: PMC5738307 DOI: 10.4184/asj.2017.11.6.870] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 04/18/2017] [Accepted: 04/22/2017] [Indexed: 12/13/2022] Open
Abstract
Study Design We established induced pluripotent stem cells (iPSCs) and neural stem/progenitor cells (NSPCs) from three newborns with spina bifida aperta (SBa) using clinically practical methods. Purpose We aimed to develop stem cell lines derived from newborns with SBa for future therapeutic use. Overview of Literature SBa is a common congenital spinal cord abnormality that causes defects in neurological and urological functions. Stem cell transplantation therapies are predicted to provide beneficial effects for patients with SBa. However, the availability of appropriate cell sources is inadequate for clinical use because of their limited accessibility and expandability, as well as ethical issues. Methods Fibroblast cultures were established from small fragments of skin obtained from newborns with SBa during SBa repair surgery. The cultured cells were transfected with episomal plasmid vectors encoding reprogramming factors necessary for generating iPSCs. These cells were then differentiated into NSPCs by chemical compound treatment, and NSPCs were expanded using neurosphere technology. Results We successfully generated iPSC lines from the neonatal dermal fibroblasts of three newborns with SBa. We confirmed that these lines exhibited the characteristics of human pluripotent stem cells. We successfully generated NSPCs from all SBa newborn-derived iPSCs with a combination of neural induction and neurosphere technology. Conclusions We successfully generated iPSCs and iPSC-NSPCs from surgical samples obtained from newborns with SBa with the goal of future clinical use in patients with SBa.
Collapse
|
97
|
Hoashi Y, Okamoto S, Abe Y, Matsumoto T, Tanaka J, Yoshida Y, Imaizumi K, Mishima K, Akamatsu W, Okano H, Baba K. Generation of neural cells using iPSCs from sleep bruxism patients with 5-HT2A polymorphism. J Prosthodont Res 2017; 61:242-250. [PMID: 27916472 DOI: 10.1016/j.jpor.2016.11.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 10/26/2016] [Accepted: 11/14/2016] [Indexed: 11/28/2022]
Abstract
PURPOSE Sleep bruxism (SB) is classified as a sleep-related movement disorder characterized by grinding and clenching of the teeth during sleep, which is responsible for a variety of clinical problems such as abnormal tooth attrition and fracture of teeth or roots. Little is known about the etiology of SB. Our previous study identified a genomic association of the serotonin 2A receptor (5-HT2A) single nucleotide polymorphism (SNP), rs6313 C>T, with SB, where the C allele carrier is associated with a 4.25-fold increased risk of SB. Based on this finding, the aim of this study was to generate of neural cells using SB patient-specific induced pluripotent stem cells (iPSCs). METHODS Two SB patients with C/C genotype of rs6313 and two controls with T/T genotype were screened by laboratory-based polysomnographic recordings and the TaqMan genotyping assay. Four lines of iPSCs, two from SB patients and two from controls, were established from peripheral blood mononuclear cells by introduction of reprogramming factors. We performed quality control assays on iPSCs using expression of markers for undifferentiated pluripotent cells, immunostaining for pluripotency markers, a three-germ layer assay, and karyotype analysis. The established iPSCs were differentiated into neurons using the neurosphere culture system. 5-HT2A gene expression in these neurons was evaluated by quantitative real-time PCR. RESULTS Patient-specific iPSCs were successfully differentiated into neurons expressing 5-HT2A. CONCLUSIONS This report is the first successful generation of neural cells using iPSCs from sleep bruxism patients with 5-HT2A polymorphism, which has the potential to elucidate the etiology and underlying mechanism of SB.
Collapse
Affiliation(s)
- Yurie Hoashi
- Department of Prosthodontics, Showa University School of Dentistry, Tokyo, Japan
| | - Satoshi Okamoto
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Yuka Abe
- Department of Prosthodontics, Showa University School of Dentistry, Tokyo, Japan
| | - Takashi Matsumoto
- Department of Prosthodontics, Showa University School of Dentistry, Tokyo, Japan
| | - Junichi Tanaka
- Division of Pathology, Department of Oral Diagnostic Sciences, Showa University School of Dentistry, Tokyo, Japan
| | - Yuya Yoshida
- Department of Prosthodontics, Showa University School of Dentistry, Tokyo, Japan
| | - Kent Imaizumi
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Kenji Mishima
- Division of Pathology, Department of Oral Diagnostic Sciences, Showa University School of Dentistry, Tokyo, Japan
| | - Wado Akamatsu
- Center for Genomic and Regenerative Medicine, Juntendo University School of Medicine, Tokyo, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan.
| | - Kazuyoshi Baba
- Department of Prosthodontics, Showa University School of Dentistry, Tokyo, Japan.
| |
Collapse
|
98
|
Engineering new neurons: in vivo reprogramming in mammalian brain and spinal cord. Cell Tissue Res 2017; 371:201-212. [PMID: 29170823 DOI: 10.1007/s00441-017-2729-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 11/02/2017] [Indexed: 12/13/2022]
Abstract
Neurons are postmitotic. Once lost because of injury or degeneration, they do not regenerate in most regions of the mammalian central nervous system. Recent advancements nevertheless clearly reveal that new neurons can be reprogrammed from non-neuronal cells, especially glial cells, in the adult mammalian brain and spinal cord. Here, we give a brief overview concerning cell fate reprogramming in vivo and then focus on the underlying molecular and cellular mechanisms. Specifically, we critically review the cellular sources and the reprogramming factors for in vivo neuronal conversion. Influences of environmental cues and the challenges ahead are also discussed. The ability of inducing new neurons from an abundant and broadly distributed non-neuronal cell source brings new perspectives regarding regeneration-based therapies for traumatic brain and spinal cord injuries and degenerative diseases.
Collapse
|
99
|
Yoshihara M, Hayashizaki Y, Murakawa Y. Genomic Instability of iPSCs: Challenges Towards Their Clinical Applications. Stem Cell Rev Rep 2017; 13:7-16. [PMID: 27592701 PMCID: PMC5346115 DOI: 10.1007/s12015-016-9680-6] [Citation(s) in RCA: 188] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Induced pluripotent stem cells (iPSCs) are a type of pluripotent stem cells generated directly from mature cells through the introduction of key transcription factors. iPSCs can be propagated and differentiated into many cell types in the human body, holding enormous potential in the field of regenerative medicine. However, genomic instability of iPSCs has been reported with the advent of high-throughput technologies such as next-generation sequencing. The presence of genetic variations in iPSCs has raised serious safety concerns, hampering the advancement of iPSC-based novel therapies. Here we summarize our current knowledge on genomic instability of iPSCs, with a particular focus on types of genetic variations and their origins. Importantly, it remains elusive whether genetic variations in iPSCs can be an actual risk factor for adverse effects including malignant outgrowth. Furthermore, we discuss novel approaches to generate iPSCs with fewer genetic variations. Lastly, we outline the safety issues and monitoring strategies of iPSCs in clinical settings.
Collapse
Affiliation(s)
- Masahito Yoshihara
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Kanagawa, Japan.,Department of Ophthalmology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | | | - Yasuhiro Murakawa
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Kanagawa, Japan. .,RIKEN Preventive Medicine and Diagnosis Innovation Program, Wako, Saitama, Japan.
| |
Collapse
|
100
|
Nagashima K, Miwa T, Soumiya H, Ushiro D, Takeda-Kawaguchi T, Tamaoki N, Ishiguro S, Sato Y, Miyamoto K, Ohno T, Osawa M, Kunisada T, Shibata T, Tezuka KI, Furukawa S, Fukumitsu H. Priming with FGF2 stimulates human dental pulp cells to promote axonal regeneration and locomotor function recovery after spinal cord injury. Sci Rep 2017; 7:13500. [PMID: 29044129 PMCID: PMC5647367 DOI: 10.1038/s41598-017-13373-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 09/22/2017] [Indexed: 12/31/2022] Open
Abstract
Human dental pulp cells (DPCs), adherent cells derived from dental pulp tissues, are potential tools for cell transplantation therapy. However, little work has been done to optimize such transplantation. In this study, DPCs were treated with fibroblast growth factor-2 (FGF2) for 5-6 consecutive serial passages and were transplanted into the injury site immediately after complete transection of the rat spinal cord. FGF2 priming facilitated the DPCs to promote axonal regeneration and to improve locomotor function in the rat with spinal cord injury (SCI). Additional analyses revealed that FGF2 priming protected cultured DPCs from hydrogen-peroxide-induced cell death and increased the number of DPCs in the SCI rat spinal cord even 7 weeks after transplantation. The production of major neurotrophic factors was equivalent in FGF2-treated and untreated DPCs. These observations suggest that FGF2 priming might protect DPCs from the post-trauma microenvironment in which DPCs infiltrate and resident immune cells generate cytotoxic reactive oxygen species. Surviving DPCs could increase the availability of neurotrophic factors in the lesion site, thereby promoting axonal regeneration and locomotor function recovery.
Collapse
Affiliation(s)
- Kosuke Nagashima
- Laboratory of Molecular Biology, Department of Biofunctional Analysis, Gifu Pharmaceutical University, 1-25-4 Daigakunishi, Gifu, 501-1196, Japan
| | - Takahiro Miwa
- Laboratory of Molecular Biology, Department of Biofunctional Analysis, Gifu Pharmaceutical University, 1-25-4 Daigakunishi, Gifu, 501-1196, Japan
| | - Hitomi Soumiya
- Laboratory of Molecular Biology, Department of Biofunctional Analysis, Gifu Pharmaceutical University, 1-25-4 Daigakunishi, Gifu, 501-1196, Japan
| | - Daisuke Ushiro
- Laboratory of Molecular Biology, Department of Biofunctional Analysis, Gifu Pharmaceutical University, 1-25-4 Daigakunishi, Gifu, 501-1196, Japan
| | - Tomoko Takeda-Kawaguchi
- Department of Oral and Maxillofacial Science, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Naritaka Tamaoki
- Department of Oral and Maxillofacial Science, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
- National Cancer Institute, National Institutes of Health (NIH), Bethesda, Maryland, 20892, USA
| | - Saho Ishiguro
- Laboratory of Molecular Biology, Department of Biofunctional Analysis, Gifu Pharmaceutical University, 1-25-4 Daigakunishi, Gifu, 501-1196, Japan
| | - Yumi Sato
- Laboratory of Molecular Biology, Department of Biofunctional Analysis, Gifu Pharmaceutical University, 1-25-4 Daigakunishi, Gifu, 501-1196, Japan
| | - Kei Miyamoto
- Department of Orthopaedic Surgery, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
- Department of Orthopaedic Surgery and Spine Center, Gifu Municipal Hospital, 7-1 Kashima, Gifu, 500-8323, Japan
| | - Takatoshi Ohno
- Department of Orthopaedic Surgery, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
- Department of Orthopaedic Surgery, Gifu Red Cross Hospital, 3-36 Iwakura, Gifu, 502-0844, Japan
| | - Masatake Osawa
- Department of Regeneration Technology, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Takahiro Kunisada
- Department of Tissue and Organ Development, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Toshiyuki Shibata
- Department of Oral and Maxillofacial Science, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Ken-Ichi Tezuka
- Department of Tissue and Organ Development, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Shoei Furukawa
- Laboratory of Molecular Biology, Department of Biofunctional Analysis, Gifu Pharmaceutical University, 1-25-4 Daigakunishi, Gifu, 501-1196, Japan
| | - Hidefumi Fukumitsu
- Laboratory of Molecular Biology, Department of Biofunctional Analysis, Gifu Pharmaceutical University, 1-25-4 Daigakunishi, Gifu, 501-1196, Japan.
| |
Collapse
|