1
|
Grant-Bier J, Ruppert K, Hayward B, Usdin K, Kumari D. MSH2 is not required for either maintenance of DNA methylation or repeat contraction at the FMR1 locus in fragile X syndrome or the FXN locus in Friedreich's ataxia. Epigenetics Chromatin 2025; 18:24. [PMID: 40296143 PMCID: PMC12036138 DOI: 10.1186/s13072-025-00588-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 04/11/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND Repeat-induced epigenetic changes are observed in many repeat expansion disorders (REDs). These changes result in transcriptional deficits and/or silencing of the associated gene. MSH2, a mismatch repair protein that is required for repeat expansion in the REDs, has been implicated in the maintenance of DNA methylation seen in the region upstream of the expanded CTG repeats at the DMPK locus in myotonic dystrophy type 1 (DM1). Here, we investigated the role of MSH2 in aberrant DNA methylation in two additional REDs, fragile X syndrome (FXS) that is caused by a CGG repeat expansion in the 5' untranslated region (UTR) of the Fragile X Messenger Ribonucleoprotein 1 (FMR1) gene, and Friedreich's ataxia (FRDA) that is caused by a GAA repeat expansion in intron 1 of the frataxin (FXN) gene. RESULTS In contrast to what is seen at the DMPK locus in DM1, loss of MSH2 did not decrease DNA methylation at the FMR1 promoter in FXS embryonic stem cells (ESCs) or increase FMR1 transcription. This difference was not due to the differences in the CpG density of the two loci as a decrease in DNA methylation was also not observed in a less CpG dense region upstream of the expanded GAA repeats in the FXN gene in MSH2 null induced pluripotent stem cells (iPSCs) derived from FRDA patient fibroblasts. Surprisingly, given previous reports, we found that FMR1 reactivation was associated with a high frequency of MSH2-independent CGG-repeat contractions that resulted a permanent loss of DNA methylation. MSH2-independent GAA-repeat contractions were also seen in FRDA cells. CONCLUSIONS Our results suggest that there are mechanistic differences in the way that DNA methylation is maintained in the region upstream of expanded repeats among different REDs even though they share a similar mechanism of repeat expansion. The high frequency of transcription-induced MSH2-dependent and MSH2-independent contractions we have observed may contribute to the mosaicism that is frequently seen in carriers of FMR1 alleles with expanded CGG-repeat tracts. These contractions may reflect the underlying problems associated with transcription through the repeat. Given the recent interest in the therapeutic use of transcription-driven repeat contractions, our data may have interesting mechanistic, prognostic, and therapeutic implications.
Collapse
Affiliation(s)
- Jessalyn Grant-Bier
- Section on Gene Structure and Disease, Laboratory of Cell and Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
- Present address: Cellular and Molecular Biology Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Kathryn Ruppert
- Section on Gene Structure and Disease, Laboratory of Cell and Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Bruce Hayward
- Section on Gene Structure and Disease, Laboratory of Cell and Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Karen Usdin
- Section on Gene Structure and Disease, Laboratory of Cell and Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Daman Kumari
- Section on Gene Structure and Disease, Laboratory of Cell and Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
2
|
Guo X, Wang X, Wang J, Ma M, Ren Q. Current Development of iPSC-Based Modeling in Neurodegenerative Diseases. Int J Mol Sci 2025; 26:3774. [PMID: 40332425 PMCID: PMC12027653 DOI: 10.3390/ijms26083774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 04/08/2025] [Accepted: 04/09/2025] [Indexed: 05/08/2025] Open
Abstract
Over the past two decades, significant advancements have been made in the induced pluripotent stem cell (iPSC) technology. These developments have enabled the broader application of iPSCs in neuroscience, improved our understanding of disease pathogenesis, and advanced the investigation of therapeutic targets and methods. Specifically, optimizations in reprogramming protocols, coupled with improved neuronal differentiation and maturation techniques, have greatly facilitated the generation of iPSC-derived neural cells. The integration of the cerebral organoid technology and CRISPR/Cas9 genome editing has further propelled the application of iPSCs in neurodegenerative diseases to a new stage. Patient-derived or CRISPR-edited cerebral neurons and organoids now serve as ideal disease models, contributing to our understanding of disease pathophysiology and identifying novel therapeutic targets and candidates. In this review, we examine the development of iPSC-based models in neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, and Huntington's disease.
Collapse
Affiliation(s)
- Xiangge Guo
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang 050017, China; (X.G.); (X.W.); (J.W.)
| | - Xumeng Wang
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang 050017, China; (X.G.); (X.W.); (J.W.)
| | - Jiaxuan Wang
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang 050017, China; (X.G.); (X.W.); (J.W.)
| | - Min Ma
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang 050017, China; (X.G.); (X.W.); (J.W.)
- Human Brain Bank, Hebei Medical University, Shijiazhuang 050017, China
| | - Qian Ren
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang 050017, China; (X.G.); (X.W.); (J.W.)
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang 050017, China
- Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Hebei Medical University, Shijiazhuang 050017, China
| |
Collapse
|
3
|
Ashrafi A, Runyon W, Hu S, Kumar R, Catchpole T, Singh A, Ratnapriya R, Csaky KG, Sripathi SR. Generation of human induced pluripotent stem cell lines from an age-related macular degeneration patient with hyperreflective foci overlying drusen (RFSCi002-A) and an unaffected sibling (RFSCi001-A). Stem Cell Res 2025; 86:103715. [PMID: 40273531 DOI: 10.1016/j.scr.2025.103715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 04/04/2025] [Accepted: 04/13/2025] [Indexed: 04/26/2025] Open
Abstract
Age-related macular degeneration (AMD) is a leading cause of vision loss, driven by retinal pigment epithelium (RPE) and photoreceptor degeneration. A key feature is drusen accumulation between the RPE and Bruch's membrane. In intermediate AMD, hyperreflective foci (HRF)-bright intraretinal lesions visible on optical coherence tomography (OCT) imaging-serve as biomarkers of disease progression. To study HRF mechanisms, we generated induced pluripotent stem cell (iPSC) lines from an AMD patient with HRF overlying drusen (RFSC4) and their unaffected sibling (RFSC3). These iPSC models offer a platform to explore disease mechanisms and develop therapies for AMD.
Collapse
Affiliation(s)
- Adnin Ashrafi
- Henderson Ocular Stem Cell Laboratory, Retina Foundation of the Southwest, Dallas, TX 75231, USA
| | - Wendy Runyon
- Stem Cell Core, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Sam Hu
- Stem Cell Core, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Ritu Kumar
- Stem Cell Core, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Timothy Catchpole
- Molecular Ophthalmology Laboratory, Clinical Center of Innovation for AMD, Retina Foundation of the Southwest, Dallas, TX 75231, USA
| | - Ajeet Singh
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Rinki Ratnapriya
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Karl G Csaky
- Molecular Ophthalmology Laboratory, Clinical Center of Innovation for AMD, Retina Foundation of the Southwest, Dallas, TX 75231, USA; Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Srinivasa R Sripathi
- Henderson Ocular Stem Cell Laboratory, Retina Foundation of the Southwest, Dallas, TX 75231, USA; Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
4
|
Awaya T, Ajiro M, Kobayashi H, Sawada T, Gotanda K, Noji T, Takemoto N, Iida K, Saito MK, Niu DM, Hagiwara M. Invention of an oral medication for cardiac Fabry disease caused by RNA mis-splicing. SCIENCE ADVANCES 2025; 11:eadt9695. [PMID: 40203112 PMCID: PMC11980850 DOI: 10.1126/sciadv.adt9695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 03/03/2025] [Indexed: 04/11/2025]
Abstract
Pathogenic RNA splicing variants have emerged as promising therapeutic targets due to their role in disease while preserving coding sequences. In this study, we developed RECTAS-2.0, a small molecule designed to correct RNA mis-splicing caused by the GLA c.639+919G>A mutation, which leads to the inclusion of a 57-nucleotide poison exon, resulting in later-onset Fabry disease, particularly prevalent in East Asia. RECTAS-2.0 restored normal GLA mRNA splicing and α-galactosidase activity in patient-derived B-lymphoblastoid cell lines and induced pluripotent stem cell-derived cardiomyocytes. Furthermore, oral administration of RECTAS-2.0 effectively corrected splicing in a transgenic mouse model, demonstrating its substantial splice-switching activity and safety for clinical application. RECTAS-2.0 demonstrated potential applicability to other genetic disorders that involve similar exon competition. These findings underscore the therapeutic potential of RECTAS-2.0 for Fabry disease and highlight its broader implications for RNA splicing-targeted therapies in genetic disorders.
Collapse
Affiliation(s)
- Tomonari Awaya
- Department of Anatomy and Developmental Biology, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan
| | - Masahiko Ajiro
- Department of Anatomy and Developmental Biology, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan
- Department of Drug Discovery Medicine, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan
| | - Hiroko Kobayashi
- Department of Anatomy and Developmental Biology, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan
- Department of Drug Discovery Medicine, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan
| | - Teruo Sawada
- Department of Anatomy and Developmental Biology, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan
- Department of Drug Discovery Medicine, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan
| | - Kentoku Gotanda
- Modality Research Laboratories III, Shinagawa R&D Center, Daiichi Sankyo Co. Ltd., Tokyo 140-8710, Japan
| | - Toshiharu Noji
- Modality Research Laboratories I, Shinagawa R&D Center, Daiichi Sankyo Co. Ltd., Tokyo 140-8710, Japan
| | - Naohiro Takemoto
- Modality Research Laboratories I, Shinagawa R&D Center, Daiichi Sankyo Co. Ltd., Tokyo 140-8710, Japan
| | - Kei Iida
- Medical Support Center, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan
| | - Megumu K. Saito
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Dau-Ming Niu
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei 11217, Taiwan
- Institute of Clinical Medicine, National Yang-Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Masatoshi Hagiwara
- Department of Anatomy and Developmental Biology, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan
- Department of Drug Discovery Medicine, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan
| |
Collapse
|
5
|
Zueva AS, Shevchenko AI, Medvedev SP, Elisaphenko EA, Sleptcov AA, Nazarenko MS, Tmoyan NA, Zakian SM, Zakharova IS. Isogenic induced pluripotent stem cell line ICGi036-A-1 from a patient with familial hypercholesterolaemia, derived by correcting a pathogenic variant of the gene LDLR c.530C>T. Vavilovskii Zhurnal Genet Selektsii 2025; 29:189-199. [PMID: 40264801 PMCID: PMC12011625 DOI: 10.18699/vjgb-25-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 12/04/2024] [Accepted: 12/09/2024] [Indexed: 04/24/2025] Open
Abstract
Familial hypercholesterolaemia is a common monogenic disorder characterized by high plasma cholesterol levels leading to chronic cardiovascular disease with high risk and often early manifestation due to atherosclerotic lesions of the blood vessels. The atherosclerotic lesions in familial hypercholesterolaemia are mainly caused by pathogenic variants of the low-density lipoprotein receptor (LDLR) gene, which plays an important role in cholesterol metabolism. Normally, cholesterol-laden low-density lipoproteins bind to the LDLR receptor on the surface of liver cells to be removed from the bloodstream by internalisation with hepatocytes. In familial hypercholesterolaemia, the function of the receptor is impaired and the uptake of low-density lipoproteins is significantly reduced. As a result, cholesterol accumulates in the subendothelial space on the inner wall of blood vessels, triggering atherogenesis, the formation of atherosclerotic plaques. At present, there are no effective and universal approaches to the diagnosis and treatment of familial hypercholesterolaemia. A relevant approach to study the molecular genetic mechanisms of the disease and to obtain systems for screening chemical compounds as potential drugs is the generation of cellular models based on patient-specific induced pluripotent stem cells. The aim of our work was to derive an isogenic genetically modified induced pluripotent stem cell line by correcting the pathogenic allelic variant c.530C of the LDLR gene in the original iPSC previously obtained from a compound heterozygote patient with familial hypercholesterolaemia. The resulting isogenic iPSC line differs from the original by only one corrected nucleotide substitution, allowing us to study the direct effect of this pathogenic genetic variant on physiological changes in relevant differentiated cells. CRISPR/Cas-mediated base editing was used to correct the single nucleotide substitution. The resulting genetically modified iPSC line has pluripotency traits, a normal karyotype, a set of short tandem repeats identical to that in the original line and can be used to obtain differentiated derivatives necessary for the elaboration of relevant cell models.
Collapse
Affiliation(s)
- A S Zueva
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia Novosibirsk State University, Novosibirsk, Russia
| | - A I Shevchenko
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - S P Medvedev
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - E A Elisaphenko
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - A A Sleptcov
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - M S Nazarenko
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - N A Tmoyan
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia National Medical Research Center of Cardiology named after academician E.I. Chazov, Moscow, Russia
| | - S M Zakian
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - I S Zakharova
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
6
|
Raina K, Modak K, Premkumar C, Joshi G, Palani D, Nandy K, Sivamani Y, Velayudhan SR, Thummer RP. UTF1 Expression is Important for the Generation and Maintenance of Human iPSCs. Stem Cell Rev Rep 2025; 21:859-871. [PMID: 39754619 DOI: 10.1007/s12015-024-10836-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2024] [Indexed: 01/06/2025]
Abstract
BACKGROUND Undifferentiated embryonic cell transcription factor 1 (UTF1) is predominantly expressed in pluripotent stem cells and plays a vital role in embryonic development and pluripotency maintenance. Despite its established importance in murine models, the role of UTF1 on human induced pluripotent stem cells (iPSCs) has not been comprehensively studied. METHODS This study utilized CRISPR/Cas9 gene editing to create UTF1 knockout in human fibroblasts and iPSCs. We employed episomal vectors for reprogramming UTF1 knockout fibroblasts into iPSCs and analyzed the effects of UTF1 depletion on cellular morphology, pluripotency, and viability through Western blotting, PCR, and flow cytometry. In addition, we integrated an shRNA that downregulated the expression of UTF1 for mechanistic studies to understand the impact of UTF1 depletion in iPSC pluripotency and differentiation. RESULTS UTF1 knockout resulted in significantly reduced reprogramming efficiency and increased spontaneous differentiation, indicating its crucial role in maintaining human iPSC identity and stability. In knockdown experiments, gradual loss of UTF1 led to change in cellular morphologies and decreased expression of core pluripotency markers OCT4 and SOX2. Interestingly, unlike complete UTF1 knockout, the gradual downregulation of UTF1 in iPSCs did not result in apoptosis, suggesting that the loss of pluripotency can occur independently of the apoptotic pathways. CONCLUSIONS UTF1 is essential for maintaining the pluripotency and viability of human iPSCs. Its depletion affects the fundamental properties of stem cells, underscoring the potential challenges in using UTF1-deficient cells for therapeutic applications. Future studies should explore the mechanistic pathways through which UTF1 controls pluripotency and differentiation, which could provide insights into improving iPSC stability for clinical applications.
Collapse
Affiliation(s)
- Khyati Raina
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Kirti Modak
- Department of Hematology, Christian Medical College, Vellore, India
| | - Chitra Premkumar
- Center for Stem Cell Research, Christian Medical College, Vellore, India
| | - Gaurav Joshi
- Department of Hematology, Christian Medical College, Vellore, India
| | - Dhavapriya Palani
- Center for Stem Cell Research, Christian Medical College, Vellore, India
| | - Krittika Nandy
- Center for Stem Cell Research, Christian Medical College, Vellore, India
| | - Yazhini Sivamani
- Center for Stem Cell Research, Christian Medical College, Vellore, India
| | - Shaji R Velayudhan
- Department of Hematology, Christian Medical College, Vellore, India
- Center for Stem Cell Research, Christian Medical College, Vellore, India
| | - Rajkumar P Thummer
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India.
| |
Collapse
|
7
|
Kuang YL, Locatelli CA, Qin Y, Zhang Y, Theusch E, Muñoz-Howell A, Sanchez G, Lu M, Nguyen MA, Yalamanchili T, Wang X, Nalula G, Mattis AN, Oni-Orisan A, Iribarren C, Krauss RM, Mulvihill EE, Medina MW. MIR192 Upregulates GLP-1 Receptor and Improves Statin-Induced Impairment of Insulin Secretion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.18.643960. [PMID: 40166140 PMCID: PMC11956930 DOI: 10.1101/2025.03.18.643960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Statins are a commonly prescribed cholesterol lowering drug class that can increase the risk of new-onset diabetes (NOD). To investigate the molecular mechanisms underlying this effect, we generated human induced pluripotent stem cells (iPSCs) from individuals identified from electronic health records of Kaiser Permanente of Northern California who were susceptible to developing NOD after statin initiation or controls who maintained stable fasting glucose on statin treatment. RNA-seq analysis of iPSCs incubated with atorvastatin, simvastatin or mock buffer for 24 hours identified the long non-coding RNA MIR194-2HG as a top candidate gene. Statin-induced increases in its expression were observed in NOD resistant controls, while statin-induced reductions occurred in NOD susceptible cases. MIR194-2HG encompasses two microRNA genes: MIR192 and MIR194-2. The mature microRNA miR-192-5p, derived from the 5' arm of MIR192, was predicted to bind the 3'UTR of the glucagon like peptide 1 (GLP-1) receptor (GLP1R) transcript. Transfection of a rat insulinoma cell line INS-1 with a miR-192-5p mimic increased Glp1r transcript (1.41-fold) and protein (1.51-fold) levels compared to a scrambled control. Using a luciferase reporter containing the human GLP1R 3'UTR, miR-192-5p overexpression similarly increased luciferase signal (1.44-fold). The miR-192-5p mimic enhanced glucose stimulated insulin secretion (GSIS) in response to GLP1R agonists (1.64-1.81-fold) and rescued simvastatin-induced GSIS impairment in INS-1 cells. Wildtype mice treated with miR-192 AAV8 had improved glucose sensitivity. Islets isolated from these mice exhibited enhanced GLP-1 potentiated GSIS during perifusion ex vivo. These effects were absent in the DIRKO (Glp1r/Gipr double knockout) mouse islets, consistent with the idea that miR-192 promotes GLP-1 mediated GSIS through GLP1R. These findings implicate MIR192 in statin-induced impairment of GSIS by modulating GLP1R, potentially contributing to the susceptibility to NOD in statin users.
Collapse
Affiliation(s)
- Yu-Lin Kuang
- Department of Pediatrics, University of California San Francisco, Oakland, CA, USA
| | - Cassandra A.A. Locatelli
- Department of Biochemistry, Microbiology and Immunology, The University of Ottawa, Ottawa, Ontario, Canada
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Yuanyuan Qin
- Department of Pediatrics, University of California San Francisco, Oakland, CA, USA
| | - Yuqing Zhang
- Department of Pediatrics, University of California San Francisco, Oakland, CA, USA
| | - Elizabeth Theusch
- Department of Pediatrics, University of California San Francisco, Oakland, CA, USA
| | - Antonio Muñoz-Howell
- Department of Pediatrics, University of California San Francisco, Oakland, CA, USA
| | - Gabriela Sanchez
- Kaiser Permanente Division of Research, 2000 Broadway, Oakland, CA, USA
| | - Meng Lu
- Kaiser Permanente Division of Research, 2000 Broadway, Oakland, CA, USA
| | - My-Anh Nguyen
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Tanvi Yalamanchili
- Department of Pediatrics, University of California San Francisco, Oakland, CA, USA
| | - Xuanwen Wang
- Department of Pediatrics, University of California San Francisco, Oakland, CA, USA
| | - Gilbert Nalula
- Department of Pediatrics, University of California San Francisco, Oakland, CA, USA
| | - Aras N. Mattis
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
- Board Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- The Liver Center, University of California San Francisco, San Francisco, CA, USA
| | - Akinyemi Oni-Orisan
- Department of Clinical Pharmacy, University of California San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
| | - Carlos Iribarren
- Kaiser Permanente Division of Research, 2000 Broadway, Oakland, CA, USA
| | - Ronald M. Krauss
- Department of Pediatrics, University of California San Francisco, Oakland, CA, USA
- Department of Medicine, University of California San Francisco, Oakland, CA, USA
| | - Erin E. Mulvihill
- Department of Biochemistry, Microbiology and Immunology, The University of Ottawa, Ottawa, Ontario, Canada
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Marisa W. Medina
- Department of Pediatrics, University of California San Francisco, Oakland, CA, USA
- The Liver Center, University of California San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
8
|
Hébert-Milette I, Lévesque C, Paquette J, Rivard MÈ, Villeneuve L, Boucher G, Goyette P, Charron G, Rioux JD. Inflammatory bowel disease risk gene C1ORF106 regulates actin dynamics in intestinal epithelial cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.14.643205. [PMID: 40161582 PMCID: PMC11952551 DOI: 10.1101/2025.03.14.643205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Background and aims C1ORF106 has previously been associated with inflammatory bowel diseases (IBD) via large-scale genetic studies. Increased intestinal permeability is a hallmark of IBD and is observed in at-risk individuals prior to the appearance of clinical symptoms. C1ORF106 was previously shown to regulate intestinal barrier permeability through the regulation of adherens junction stability and through the formation of tight junctions, which impacted actin assembly. However, the downstream impact and molecular mechanisms involved in actin regulation by C1ORF106 haven't been explored. Our study aimed at identifying which pathways involved in intestinal epithelial barrier regulation and F-actin regulation are impacted by C1ORF106 and its IBD-associated variant. Methods We knocked down (KD) the expression of C1ORF106 in human colonic epithelial cells and characterized the function of the 333F variant in intestinal epithelial spheroid cultures obtained from patient-derived human induced pluripotent stem cell (hiPSC). We measured barrier permeability and characterized spheroid formation, actin regulation and cell migration though immunofluorescence, western blots and permeability assays. Results C1ORF106 KD leads to impaired cortical actin belt dynamics and regulation of stress fiber formation, resulting in increased cell constriction, impaired barrier permeability, cell polarity and cell migration. Moreover, we demonstrated that an inhibition of ROCK rescues the actin belt and cell polarity phenotypes in C1ORF106 KD cells, demonstrating that C1ORF106 regulates these phenotypes through a ROCK-dependent mechanism. We also observed an altered nmMYO2-P localization in C1ORF106 KD cells associated with the formation of Vacuolar Apical Compartments (VACs), which are important for 3D epithelial spheroid formation. We observed a similar impact on cell polarity in intestinal epithelial spheroids obtained from hiPSC carrying the 333F variant, providing additional support that this pathway is involved in disease development. Conclusion We provide insights into the molecular mechanisms by which C1ORF106 controls actin dynamics to regulate intestinal epithelial integrity.
Collapse
Affiliation(s)
- Isabelle Hébert-Milette
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, Montreal, Quebec, Canada
- Université de Montréal, Montreal, Quebec, Canada
| | - Chloé Lévesque
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, Montreal, Quebec, Canada
| | - Jean Paquette
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, Montreal, Quebec, Canada
| | - Marie-Ève Rivard
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, Montreal, Quebec, Canada
| | - Louis Villeneuve
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, Montreal, Quebec, Canada
| | - Gabrielle Boucher
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, Montreal, Quebec, Canada
| | - Philippe Goyette
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, Montreal, Quebec, Canada
| | - Guy Charron
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, Montreal, Quebec, Canada
| | - John D. Rioux
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, Montreal, Quebec, Canada
- Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
9
|
Kado Abdalkader R, Konishi S, Fujita T. Development of a flexible 3D printed TPU-PVC microfluidic devices for organ-on-a-chip applications. Sci Rep 2025; 15:6125. [PMID: 39972010 PMCID: PMC11839916 DOI: 10.1038/s41598-025-90470-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 02/13/2025] [Indexed: 02/21/2025] Open
Abstract
The development of cost-effective, flexible, and scalable microfluidic devices is crucial for advancing organ-on-a-chip (OoC) technology for drug discovery and disease modeling applications. In this study, we present a novel 3D-printed flexible microfluidic device (3D-FlexTPU-MFD) fabricated through a one-step fused deposition modeling (FDM) process using thermoplastic polyurethane (TPU) as the printing filament and polyvinyl chloride (PVC) as the bonding substrate. The device's compatibility was evaluated with various cell types, including human primary myoblasts, human primary endothelial cells (HUVEC), and human iPSC-derived optic vesicle (OV) organoids. Myoblasts cultured within the device exhibited high viability, successful differentiation, and the formation of aligned myotube bundles, outperforming conventional well-plate cultures. Additionally, iPSC-derived OV organoids-maintained viability, displayed neurite outgrowth, and sustained expression of the eye marker PAX6. These results demonstrate that the 3D-FlexTPU-MFD effectively supports cell growth, differentiation, and alignment, making it a promising platform for tissue modeling and OoC applications in future.
Collapse
Affiliation(s)
- Rodi Kado Abdalkader
- Ritsumeikan Global-Innovation Research Organization (R-GIRO), Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, Shiga, 525-8577, Japan.
| | - Satoshi Konishi
- Ritsumeikan Global-Innovation Research Organization (R-GIRO), Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, Shiga, 525-8577, Japan
- Department of Mechanical Engineering, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, Shiga, 525-8577, Japan
| | - Takuya Fujita
- Ritsumeikan Global-Innovation Research Organization (R-GIRO), Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, Shiga, 525-8577, Japan
- Department of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, Shiga, 525-8577, Japan
| |
Collapse
|
10
|
Kamon M, Wakatsuki S, Nakamori M, Takahashi MP, Mori-Yoshimura M, Komaki H, Araki T. Identification of ZNF850 as a novel CTG repeat expansion-related gene in myotonic dystrophy type 1 patient-derived iPSCs. Hum Mol Genet 2025; 34:327-337. [PMID: 39679849 DOI: 10.1093/hmg/ddae186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/25/2024] [Accepted: 12/08/2024] [Indexed: 12/17/2024] Open
Abstract
Myotonic dystrophy type 1 (DM1) is a dominantly inherited multi-system disease caused by expanded CTG repeats in the 3' untranslated region of the dystrophia myotonica protein kinase (DMPK) gene. Similar to other repeat disorders, the expanded trinucleotide repeat is unstable and demonstrates a tendency to increase repeat size with age in affected tissues. DNA mismatch repair system is implicated in somatic instability. It has been demonstrated that DM1 patient-derived induced pluripotent stem cells (DM1-iPSCs) show repeat instability, in which involvement of mismatch repair proteins has been suggested. Here we identified ZNF850 as a novel CTG repeat expansion-related molecule in DM1-iPSCs. ZNF850 was downregulated in a DM1-iPSC clone whose CTG repeat is exceptionally stable. We found that RNAi-mediated ZNF850 downregulation in DM1-iPSCs significantly reduced the repeat expansion and resulting instability. In adult skeletal muscle tissue of DM1 patients, ZNF850 expression levels were positively correlated with the repeat size. Furthermore, we found that ZNF850 protein can bind to the expanded CTG repeat sequence, and is located in proximity to MutSβ components. These results suggest that ZNF850 might play a role in repeat instability in DM1 by recruiting MutSβ to the repeat sequence.
Collapse
Affiliation(s)
- Masayoshi Kamon
- Department of Peripheral Nervous System Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1, Ogawa-Higashi, Kodaira, Tokyo 187-8502, Japan
- Department of Developmental Biology and Functional Genomics, Ehime University School of Medicine, 454 Shitsukawa, Toon, Ehime 791-0295, Japan
| | - Shuji Wakatsuki
- Department of Peripheral Nervous System Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1, Ogawa-Higashi, Kodaira, Tokyo 187-8502, Japan
| | - Masayuki Nakamori
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Masanori P Takahashi
- Clinical Neurophysiology, Department of Clinical Laboratory and Biomedical Sciences, Osaka University Graduate School of Medicine, 1-7 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Madoka Mori-Yoshimura
- Department of Neurology, National Center Hospital, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo 187-8551, Japan
| | - Hirofumi Komaki
- Translational Medical Center, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo 187-8551, Japan
| | - Toshiyuki Araki
- Department of Peripheral Nervous System Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1, Ogawa-Higashi, Kodaira, Tokyo 187-8502, Japan
| |
Collapse
|
11
|
Mima A, Kimura A, Ito R, Hatano Y, Tsujimoto H, Mae SI, Yamane J, Fujibuchi W, Uza N, Toyoda T, Seno H, Osafune K. Mechanistic elucidation of human pancreatic acinar development using single-cell transcriptome analysis on a human iPSC differentiation model. Sci Rep 2025; 15:4668. [PMID: 39920294 PMCID: PMC11806057 DOI: 10.1038/s41598-025-88690-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 01/30/2025] [Indexed: 02/09/2025] Open
Abstract
Few effective treatments have been developed for intractable pancreatic exocrine disorders due to the lack of suitable disease models using human cells. Pancreatic acinar cells differentiated from human induced pluripotent stem cells (hiPSCs) have the potential to solve this issue. In this study, we aimed to elucidate the developmental mechanisms of pancreatic exocrine acinar lineages to establish a directed differentiation method for pancreatic acinar cells from hiPSCs. hiPSC-derived pancreatic endoderm cells were spontaneously differentiated into both pancreatic exocrine and endocrine tissues by implantation into the renal subcapsular space of NOD/SCID mice. Single-cell RNA-seq analysis of the retrieved grafts confirmed the differentiation of pancreatic acinar lineage cells and identified REG4 as a candidate marker for pancreatic acinar progenitor cells. Furthermore, differential gene expression analysis revealed upregulated pathways, including cAMP-related signals, involved in the differentiation of hiPSC-derived pancreatic acinar lineage cells in vivo, and we found that a cAMP activator, forskolin, facilitates the differentiation from hiPSC-derived pancreatic endoderm into pancreatic acinar progenitor cells in our in vitro differentiation culture. Therefore, this platform contributes to our understanding of the developmental mechanisms of pancreatic acinar lineage cells and the establishment of differentiation methods for acinar cells from hiPSCs.
Collapse
Affiliation(s)
- Atsushi Mima
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
- Department of Gastroenterology and Hepatology, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Azuma Kimura
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
- Rege Nephro Co., Ltd., Med-Pharm Collaboration Building, Kyoto University, 46-29 Yoshidashimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Ryo Ito
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yu Hatano
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Hiraku Tsujimoto
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
- Rege Nephro Co., Ltd., Med-Pharm Collaboration Building, Kyoto University, 46-29 Yoshidashimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Shin-Ichi Mae
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Junko Yamane
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Wataru Fujibuchi
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Norimitsu Uza
- Department of Gastroenterology and Hepatology, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Taro Toyoda
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.
| | - Hiroshi Seno
- Department of Gastroenterology and Hepatology, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Kenji Osafune
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.
| |
Collapse
|
12
|
Podvysotskaya VS, Grigor'eva EV, Malakhova AA, Minina JM, Vyatkin YV, Khabarova EA, Rzaev JA, Medvedev SP, Kovalenko LV, Zakian SM. Generation and characterisation of seven induced pluripotent stem cell lines from two patients with Parkinson's disease carrying the pathological variant c.1087G>T of the LGR4 gene. Vavilovskii Zhurnal Genet Selektsii 2025; 29:15-25. [PMID: 40144372 PMCID: PMC11933898 DOI: 10.18699/vjgb-25-03] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 11/20/2024] [Accepted: 11/21/2024] [Indexed: 03/28/2025] Open
Abstract
Parkinson's disease is a neurodegenerative disorder affecting dopaminergic neurons of the substantia nigra pars compacta. The known pathological genetic variants may explain the cause of only 5 % of cases of the disease. In our study, we found two patients with a clinical diagnosis of Parkinson's disease with the genetic variant c.1087G>T (p.Gly363Cys) of the LGR4 gene. The LGR4 gene encodes the membrane receptor LGR4 (leucine rich repeat containing G protein-coupled receptor 4) associated with the G protein. We hypothesize that the LGR4 gene may be either a direct cause or a risk factor for this disease, since it is one of the main participants of the WNT/β-catenin signalling pathway. This signalling pathway is necessary for the proliferation of neurons during their differentiation, which may lead to Parkinson's disease. To study the relationship between this genetic variant and Parkinson's disease, an ideal tool is a cellular model based on induced pluripotent stem cells (iPSCs) and their differentiated derivatives, dopaminergic neurons. We reprogrammed the peripheral blood mononuclear cells of the two patients with the c.1087G>T variant of the LGR4 gene with non-integrating episomal vectors expressing OCT4, SOX2, KLF4, LIN28, L-MYC and mp53DD proteins. The obtained seven lines of induced pluripotent stem cells were characterised in detail. The iPSCs lines obtained meet all the requirements of pluripotent cells, namely, they stably proliferate, form colonies with a morphology characteristic of human pluripotent cells, have a normal diploid karyotype, express endogenous alkaline phosphatase and pluripotency markers (OCT4, NANOG, SSEA-4 and SOX2) and are capable to differentiate into derivatives of the three germ layers. The iPSC lines obtained in this work can be used as a tool to generate a relevant model to study the effect of the pathological variant c.1087G>T of the LGR4 gene on dopaminergic neuron differentiation.
Collapse
Affiliation(s)
- V S Podvysotskaya
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia Novosibirsk State University, Novosibirsk, Russia
| | - E V Grigor'eva
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia Institute of Chemical Biology and Fundamental Medicine of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - A A Malakhova
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia Institute of Chemical Biology and Fundamental Medicine of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - J M Minina
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | | | - E A Khabarova
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia Federal Neurosurgical Center of the Ministry of Health of the Russian Federation, Novosibirsk, Russia
| | - J A Rzaev
- Federal Neurosurgical Center of the Ministry of Health of the Russian Federation, Novosibirsk, Russia
| | - S P Medvedev
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia Institute of Chemical Biology and Fundamental Medicine of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - L V Kovalenko
- Surgut State University, Surgut, Khanty-Mansiysk Autonomous Okrug - Ugra, Russia
| | - S M Zakian
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia Institute of Chemical Biology and Fundamental Medicine of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
13
|
Tanabe M, Saito Y, Takasaki A, Nakano K, Yamamoto S, Suzuki C, Kawamura N, Hattori A, Oikawa M, Nagashima S, Yanagi S, Yamaguchi T, Fukuda T. Role of immature choroid plexus in the pathology of model mice and human iPSC-derived organoids with autism spectrum disorder. Cell Rep 2025; 44:115133. [PMID: 39731733 DOI: 10.1016/j.celrep.2024.115133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 10/22/2024] [Accepted: 12/11/2024] [Indexed: 12/30/2024] Open
Abstract
During gestation, the choroid plexus (ChP) produces protein-rich cerebrospinal fluid and matures prior to brain development. It is assumed that ChP dysfunction has a profound effect on developmental neuropsychiatric disorders, such as autism spectrum disorder (ASD). However, the mechanisms linking immature ChP to the onset of ASD remain unclear. Here, we find that ChP-specific CAMDI-knockout mice develop an immature ChP alongside decreased multiciliogenesis and expression of differentiation marker genes following disruption of the cerebrospinal fluid barrier. These mice exhibit ASD-like behaviors, including anxiety and impaired socialization. Additionally, the administration of metformin, an FDA-approved drug, before the social critical period achieves ChP maturation and restores social behaviors. Furthermore, both the ASD model mice and organoids derived from patients with ASD developed an immature ChP. These results propose the involvement of an immature ChP in the pathogenesis of ASD and suggest the targeting of functional maturation of the ChP as a therapeutic strategy for ASD.
Collapse
Affiliation(s)
- Motoi Tanabe
- Laboratory of Regenerative Medicine, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Yuga Saito
- Laboratory of Regenerative Medicine, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Ayaka Takasaki
- Laboratory of Regenerative Medicine, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Keita Nakano
- Laboratory of Regenerative Medicine, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Shunta Yamamoto
- Laboratory of Regenerative Medicine, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Chikako Suzuki
- Laboratory of Regenerative Medicine, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Nao Kawamura
- Laboratory of Regenerative Medicine, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Aki Hattori
- Laboratory of Regenerative Medicine, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Mami Oikawa
- Laboratory of Regenerative Medicine, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Shun Nagashima
- Laboratory of Regenerative Medicine, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Shigeru Yanagi
- Department of Life Science, Faculty of Science, Gakushuin University, Toshima-ku, Tokyo, Japan
| | - Tomoyuki Yamaguchi
- Laboratory of Regenerative Medicine, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Toshifumi Fukuda
- Laboratory of Regenerative Medicine, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan.
| |
Collapse
|
14
|
Pozner T, Grandizio C, Mitchell MW, Turan N, Scheinfeldt L. Human iPSC Reprogramming Success: The Impact of Approaches and Source Materials. Stem Cells Int 2025; 2025:2223645. [PMID: 39850337 PMCID: PMC11756937 DOI: 10.1155/sci/2223645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 12/06/2024] [Indexed: 01/25/2025] Open
Abstract
Since their discovery, human induced pluripotent stem cells (hiPSCs) have been instrumental in biomedical research, particularly in the fields of disease modelling, drug screening and regenerative therapies. Their use has significantly increased over recent years driven by the ability of hiPSCs to provide differentiated cell models without requiring embryonic stem cells. Furthermore, the transition from integrating to non-integrating reprogramming methodologies has contributed to the increase in utilisation. This shift minimises the risk of genomic alterations, enhancing the safety and reliability of hiPSCs. However, the factors that contribute to reprogramming success are still not well understood. In this study, we conducted a comparative analysis of the most prevalent non-integrating reprogramming methods across a range of starting source materials to assess their impact on reprogramming success rates. We found that while source material does not significantly impact success rates, the Sendai virus reprogramming method yields significantly higher success rates relative to the episomal reprogramming method. Our findings offer important insights from a biobanking perspective, for which long-term reliability, integrity and reproducibility of hiPSCs are crucial.
Collapse
Affiliation(s)
- Tatyana Pozner
- Biobanking Department, Coriell Institute for Medical Research, Camden 08003, New Jersey, USA
| | - Christine Grandizio
- Biobanking Department, Coriell Institute for Medical Research, Camden 08003, New Jersey, USA
| | - Matthew W. Mitchell
- Biobanking Department, Coriell Institute for Medical Research, Camden 08003, New Jersey, USA
| | - Nahid Turan
- Biobanking Department, Coriell Institute for Medical Research, Camden 08003, New Jersey, USA
| | - Laura Scheinfeldt
- Biobanking Department, Coriell Institute for Medical Research, Camden 08003, New Jersey, USA
| |
Collapse
|
15
|
Kitajima K, Hara T. Generation of chimeric antigen receptor-macrophages by using human induced pluripotent stem cells. Biochem Biophys Res Commun 2025; 743:151158. [PMID: 39673975 DOI: 10.1016/j.bbrc.2024.151158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 12/04/2024] [Accepted: 12/09/2024] [Indexed: 12/16/2024]
Abstract
Cancer immunotherapy using chimeric antigen receptor (CAR) cells shows high therapeutic efficacy against several types of leukemia. Among acute lymphoblastic leukemias (ALLs), B cell-derived ALL can be cured by CAR-expressing T cells (CAR-Ts); however, CAR-T cells cannot be simply applied for T cell-derived ALL (T-ALL) because antigens expressed by T-ALL cells, but not by CAR-T cells, have not yet been identified. To apply CAR-T therapy for T-ALL, gene editing of CAR-T cells is required to avoid attacking CAR-T cells themselves. Alternatively, CAR-expressing macrophages (CAR-Ms) have proven to be effective against various cancers, suggesting that CAR-Ms may also be effective against T-ALL. Recently, we developed an efficient differentiation induction system to generate a large number of macrophages from human induced pluripotent stem cells (iPSCs). Here, we asked whether these human iPSC-derived macrophages (iPS-MACs) can be used to develop and evaluate CAR-based immunotherapy against T-ALLs. When non-transduced iPS-MACs were co-cultured with human T-ALL-derived cells, the iPS-MACs appeared to phagocytose parts of T-ALL cells; this method of phagocytosis operated mainly through incorporation of small, "bite-sized" vesicles derived from the T-ALL cells into iPS-MACs (similar to trogocytosis). By contrast, when CAR-expressing iPS-MACs were co-cultured with T-ALL cells, iPS-MACs engulfed the whole T-ALL cell. Thus, our differentiation induction system may be a promising tool for building up CAR-M therapy for T-ALLs.
Collapse
Affiliation(s)
- Kenji Kitajima
- Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.
| | - Takahiko Hara
- Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan; Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Graduate School of Science, Department of Biological Science, Tokyo Metropolitan University, Tokyo, Japan.
| |
Collapse
|
16
|
Li R, Tsuboi H, Ito H, Takagi D, Chang YH, Shimizu T, Arai Y, Matsuo-Takasaki M, Noguchi M, Nakamura Y, Ohnuma K, Takahashi S, Hayashi Y. Generation of human induced pluripotent stem cell lines derived from two glucose transporter 1 deficiency syndrome patients. Stem Cell Res 2024; 81:103584. [PMID: 39490212 DOI: 10.1016/j.scr.2024.103584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 10/12/2024] [Accepted: 10/15/2024] [Indexed: 11/05/2024] Open
Abstract
Glucose transporter 1 deficiency syndrome (GLUT1DS), caused by impaired glucose transport at the blood-brain barriers, leads to various central nervous system dysfunctions. A comprehensive understanding of the underlying disease pathogenesis is still lacking. In this study, we have generated GLUT1DS-specific human induced pluripotent stem cells (hiPSCs) derived from two patients. These established GLUT1DS-specific hiPSC lines showed self-renewal and pluripotency and carried heterozygous frameshift or missense mutations in the responsible SLC2A1 gene. These novel cell resources provide new avenues for understanding disease mechanisms and developing new therapies for GLUT1DS.
Collapse
Affiliation(s)
- Rui Li
- iPS Cell Advanced Characterization and Development Team, BioResource Research Center, RIKEN, 3-1-1 Koyadai, Tsukuba, Ibaraki 305-0074, Japan; School of Integrative and Global Majors, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
| | - Hazuki Tsuboi
- Department of Materials Science and Bioengineering, Nagaoka University of Technology, 1603-1 Kami-Tomioka, Nagaoka, Niigata 940-2188, Japan
| | - Hidenori Ito
- iPS Cell Advanced Characterization and Development Team, BioResource Research Center, RIKEN, 3-1-1 Koyadai, Tsukuba, Ibaraki 305-0074, Japan
| | - Daigo Takagi
- iPS Cell Advanced Characterization and Development Team, BioResource Research Center, RIKEN, 3-1-1 Koyadai, Tsukuba, Ibaraki 305-0074, Japan
| | - Yun-Hsuan Chang
- iPS Cell Advanced Characterization and Development Team, BioResource Research Center, RIKEN, 3-1-1 Koyadai, Tsukuba, Ibaraki 305-0074, Japan
| | - Tomoya Shimizu
- iPS Cell Advanced Characterization and Development Team, BioResource Research Center, RIKEN, 3-1-1 Koyadai, Tsukuba, Ibaraki 305-0074, Japan
| | - Yutaka Arai
- iPS Cell Advanced Characterization and Development Team, BioResource Research Center, RIKEN, 3-1-1 Koyadai, Tsukuba, Ibaraki 305-0074, Japan
| | - Mami Matsuo-Takasaki
- iPS Cell Advanced Characterization and Development Team, BioResource Research Center, RIKEN, 3-1-1 Koyadai, Tsukuba, Ibaraki 305-0074, Japan
| | - Michiya Noguchi
- Cell Engineering Division, BioResource Research Center, RIKEN, 3-1-1 Koyadai, Tsukuba, Ibaraki 305-0074, Japan
| | - Yukio Nakamura
- Cell Engineering Division, BioResource Research Center, RIKEN, 3-1-1 Koyadai, Tsukuba, Ibaraki 305-0074, Japan
| | - Kiyoshi Ohnuma
- Department of Materials Science and Bioengineering, Nagaoka University of Technology, 1603-1 Kami-Tomioka, Nagaoka, Niigata 940-2188, Japan; Department of Science of Technology Innovation, Nagaoka University of Technology, 1603-1 Kami-Tomioka, Nagaoka, Niigata 940-2188, Japan
| | - Satoru Takahashi
- Department of Pediatrics, Asahikawa Medical University, 2-1-1-1 Midorigaoka Higashi, Asahikawa, Hokkaido 078-8510, Japan.
| | - Yohei Hayashi
- iPS Cell Advanced Characterization and Development Team, BioResource Research Center, RIKEN, 3-1-1 Koyadai, Tsukuba, Ibaraki 305-0074, Japan; School of Integrative and Global Majors, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan.
| |
Collapse
|
17
|
Li T, Ma Y, Cheng Y, Zhao Y, Qiu Z, Liu H, Zhang D, Wu J, Li J, Zhang S, Wu J. Single-Cell Transcriptomic Dataset of RPGR-associated Retinitis Pigmentosa Patient-Derived Retinal Organoids. Sci Data 2024; 11:1285. [PMID: 39592612 PMCID: PMC11599861 DOI: 10.1038/s41597-024-04124-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
X-linked retinitis pigmentosa (XLRP) is a severe hereditary retinal disorder marked by progressive vision loss due to photoreceptor dysfunction. The retinitis pigmentosa GTPase regulator (RPGR) gene, responsible for most XLRP cases, encodes a protein crucial for the transport of visual signal proteins between the photoreceptor inner and outer segments. However, the mechanism of RPGR mutation causing photoreceptor disorder is not clear and effective treatments remain elusive. This study utilized retinal organoids (ROs) derived from normal and RPGR-mutant human induced pluripotent stem cells (hiPSC) at four developmental stages (40, 90, 150, and 200 days). Single-cell RNA sequencing (scRNA-seq) was conducted on 71,096 cells, including 33,839 cells from the control group and 37,257 cells from the RPGR group. Key retinal cell types were identified and the obtained scRNAseq dataset was validated reliable and high -quality. This study has provided data resources and references for exploring the mechanism of RPGR-related retinal degeneration and support the development of targeted therapies.
Collapse
Affiliation(s)
- Ting Li
- Qingdao Institute, College of Medicine, Fudan University, Qingdao, 266500, China
- Department of Ophthalmology, Eye and ENT Hospital, College of Medicine, Fudan University, Shanghai, 200000, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality, Shanghai, 200000, China
- Key Laboratory of Myopia (Fudan University), Chinese Academy of Medical Sciences, National Health Commission, Shanghai, 200000, China
| | - Yuting Ma
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
- BGI Genomics, Shenzhen, 518083, China
| | - Yun Cheng
- Department of Ophthalmology, Eye and ENT Hospital, College of Medicine, Fudan University, Shanghai, 200000, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality, Shanghai, 200000, China
- Key Laboratory of Myopia (Fudan University), Chinese Academy of Medical Sciences, National Health Commission, Shanghai, 200000, China
| | - Yingke Zhao
- Department of Ophthalmology, Eye and ENT Hospital, College of Medicine, Fudan University, Shanghai, 200000, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality, Shanghai, 200000, China
- Key Laboratory of Myopia (Fudan University), Chinese Academy of Medical Sciences, National Health Commission, Shanghai, 200000, China
| | - Zhixu Qiu
- BGI Genomics, Shenzhen, 518083, China
| | - Hongli Liu
- Department of Ophthalmology, Eye and ENT Hospital, College of Medicine, Fudan University, Shanghai, 200000, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality, Shanghai, 200000, China
- Key Laboratory of Myopia (Fudan University), Chinese Academy of Medical Sciences, National Health Commission, Shanghai, 200000, China
| | - Daowei Zhang
- Department of Ophthalmology, Eye and ENT Hospital, College of Medicine, Fudan University, Shanghai, 200000, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality, Shanghai, 200000, China
- Key Laboratory of Myopia (Fudan University), Chinese Academy of Medical Sciences, National Health Commission, Shanghai, 200000, China
| | - Jiawen Wu
- Department of Ophthalmology, Eye and ENT Hospital, College of Medicine, Fudan University, Shanghai, 200000, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality, Shanghai, 200000, China
- Key Laboratory of Myopia (Fudan University), Chinese Academy of Medical Sciences, National Health Commission, Shanghai, 200000, China
| | - Junfeng Li
- Department of Ophthalmology, Eye and ENT Hospital, College of Medicine, Fudan University, Shanghai, 200000, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality, Shanghai, 200000, China
- Key Laboratory of Myopia (Fudan University), Chinese Academy of Medical Sciences, National Health Commission, Shanghai, 200000, China
| | - Shenghai Zhang
- Department of Ophthalmology, Eye and ENT Hospital, College of Medicine, Fudan University, Shanghai, 200000, China.
- Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality, Shanghai, 200000, China.
- Key Laboratory of Myopia (Fudan University), Chinese Academy of Medical Sciences, National Health Commission, Shanghai, 200000, China.
| | - Jihong Wu
- Qingdao Institute, College of Medicine, Fudan University, Qingdao, 266500, China.
- Department of Ophthalmology, Eye and ENT Hospital, College of Medicine, Fudan University, Shanghai, 200000, China.
- Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality, Shanghai, 200000, China.
- Key Laboratory of Myopia (Fudan University), Chinese Academy of Medical Sciences, National Health Commission, Shanghai, 200000, China.
| |
Collapse
|
18
|
Ge N, Suzuki K, Sato I, Noguchi M, Nakamura Y, Matsuo-Takasaki M, Fujishiro J, Hayashi Y. Generation of human induced pluripotent stem cell lines derived from patients of cystic biliary atresia. Hum Cell 2024; 38:18. [PMID: 39532815 PMCID: PMC11557646 DOI: 10.1007/s13577-024-01147-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 11/03/2024] [Indexed: 11/16/2024]
Abstract
Biliary atresia (BA), resulting from abnormal development of the liver's internal or external bile ducts, can lead to liver damage and potentially fatal cirrhosis. Type I cystic biliary atresia is a relatively uncommon, but clinically significant variant of BA. It is critical to develop experimental models of BA to examine the etiology and pathogenesis, which remain elusive, and to develop future therapeutics. Here, we have successfully generated a panel of human induced pluripotent stem cells (hiPSCs) from five Japanese patients carrying type I cystic BA. These hiPSC lines exhibited characteristics of self-renewal and pluripotency. These cells held normal karyotypes mostly, but one of them carried hemizygous deletions, the clinical significance of which is unknown yet. Whole genome sequence analysis indicated that some of the mutations or single nucleotide polymorphisms (SNPs) commonly found in these patients are related to hepatobiliary abnormality. Given the limited understanding of the molecular pathogenesis of cystic BA, attributed to unknown factors of genetic and environmental causes, these cellular resources will be instrumental in replicating disease phenotypes and in advancing novel therapies for this disease.
Collapse
Affiliation(s)
- Ningxin Ge
- iPS Cell Advanced Characterization and Development Team, BioResource Research Center, RIKEN, 3-1-1 Koyadai, Tsukuba, Ibaraki, 305-0074, Japan
- School of Integrative and Global Majors, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan
| | - Kan Suzuki
- Division of Pediatric Surgery, Surgical Oncology Graduate School of Medicine, Dokkyo Medical University, Tochigi, Japan.
- Department of Pediatric Surgery, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Iori Sato
- iPS Cell Advanced Characterization and Development Team, BioResource Research Center, RIKEN, 3-1-1 Koyadai, Tsukuba, Ibaraki, 305-0074, Japan
| | - Michiya Noguchi
- Cell Engineering Division, BioResource Research Center, RIKEN, 3-1-1 Koyadai, Tsukuba, Ibaraki, 305-0074, Japan
| | - Yukio Nakamura
- Cell Engineering Division, BioResource Research Center, RIKEN, 3-1-1 Koyadai, Tsukuba, Ibaraki, 305-0074, Japan
| | - Mami Matsuo-Takasaki
- iPS Cell Advanced Characterization and Development Team, BioResource Research Center, RIKEN, 3-1-1 Koyadai, Tsukuba, Ibaraki, 305-0074, Japan
| | - Jun Fujishiro
- Department of Pediatric Surgery, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Yohei Hayashi
- iPS Cell Advanced Characterization and Development Team, BioResource Research Center, RIKEN, 3-1-1 Koyadai, Tsukuba, Ibaraki, 305-0074, Japan.
- School of Integrative and Global Majors, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan.
| |
Collapse
|
19
|
Sebastian R, Song Y, Pak C. Probing the molecular and cellular pathological mechanisms of schizophrenia using human induced pluripotent stem cell models. Schizophr Res 2024; 273:4-23. [PMID: 35835709 PMCID: PMC9832179 DOI: 10.1016/j.schres.2022.06.028] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 06/21/2022] [Accepted: 06/23/2022] [Indexed: 01/13/2023]
Abstract
With recent advancements in psychiatric genomics, as a field, "stem cell-based disease modelers" were given the exciting yet daunting task of translating the extensive list of disease-associated risks into biologically and clinically relevant information in order to deliver therapeutically meaningful leads and insights. Despite their limitations, human induced pluripotent stem cell (iPSCs) based models have greatly aided our understanding of the molecular and cellular mechanisms underlying the complex etiology of brain disorders including schizophrenia (SCZ). In this review, we summarize the major findings from studies in the past decade which utilized iPSC models to investigate cell type-specific phenotypes relevant to idiopathic SCZ and disease penetrant alleles. Across cell type differences, several biological themes emerged, serving as potential neurodevelopmental mechanisms of SCZ, including oxidative stress and mitochondrial dysfunction, depletion of progenitor pools and insufficient differentiation potential of these progenitors, and structural and functional deficits of neurons and other supporting cells. Here, we discuss both the recent progress as well as challenges and improvements needed for future studies utilizing iPSCs as a model for SCZ and other neuropsychiatric disorders.
Collapse
Affiliation(s)
- Rebecca Sebastian
- Department of Biochemistry and Molecular Biology, University of Massachusetts, Amherst, MA 01003, USA; Neuroscience and Behavior Graduate Program, University of Massachusetts, Amherst, MA 01003, USA
| | - Yoonjae Song
- Department of Biochemistry and Molecular Biology, University of Massachusetts, Amherst, MA 01003, USA
| | - ChangHui Pak
- Department of Biochemistry and Molecular Biology, University of Massachusetts, Amherst, MA 01003, USA.
| |
Collapse
|
20
|
Grigor'eva EV, Malakhova AA, Yarkova ES, Minina JM, Vyatkin YV, Nadtochy JA, Khabarova EA, Rzaev JA, Medvedev SP, Zakian SM. Generation and characterization of two induced pluripotent stem cell lines (ICGi052-A and ICGi052-B) from a patient with frontotemporal dementia with parkinsonism-17 associated with the pathological variant c.2013T>G in the MAPT gene. Vavilovskii Zhurnal Genet Selektsii 2024; 28:679-687. [PMID: 39722675 PMCID: PMC11668817 DOI: 10.18699/vjgb-24-76] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/28/2024] [Accepted: 09/02/2024] [Indexed: 12/28/2024] Open
Abstract
Frontotemporal dementia with parkinsonism-17 is a neurodegenerative disease characterised by pathological aggregation of the tau protein with the formation of neurofibrillary tangles and subsequent neuronal death. The inherited form of frontotemporal dementia can be caused by mutations in several genes, including the MAPT gene on chromosome 17, which encodes the tau protein. As there are currently no medically approved treatments for frontotemporal dementia, there is an urgent need for research using in vitro cell models to understand the molecular genetic mechanisms that lead to the development of the disease, to identify targets for therapeutic intervention and to test potential drugs to prevent neuronal death. Analysis of exome sequencing data from a 46-year-old patient with a clinical diagnosis of Parkinson's disease revealed the presence of the pathological variant c.2013T>G (rs63750756) in the MAPT gene, which is associated with frontotemporal dementia with parkinsonism-17. By reprogramming the patient's peripheral blood mononuclear cells, we obtained induced pluripotent stem cells (iPSCs). Two iPSC lines were characterised in detail. Reprogramming was performed by transfection with non-integrating episomal vectors expressing the OCT4, SOX2, KLF4, LIN28, L-MYC and mp53DD proteins. The iPSC lines ICGi052-A and ICGi052-B proliferate stably, form colonies with a morphology characteristic of human pluripotent cells, have a normal diploid karyotype (46,XX), express endogenous alkaline phosphatase and pluripotency markers (OCT4, NANOG, SSEA-4 and TRA-1-60) and are able to differentiate into derivatives of three germ layers: ento-, ecto- and mesoderm. The iPSC lines obtained and characterised in detail in this work represent a unique tool for studying the molecular genetic mechanisms of the pathogenesis of frontotemporal dementia with parkinsonism-17, as well as for testing potential drugs in vitro.
Collapse
Affiliation(s)
- E V Grigor'eva
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia Institute of Chemical Biology and Fundamental Medicine of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - A A Malakhova
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia Institute of Chemical Biology and Fundamental Medicine of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - E S Yarkova
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia Novosibirsk State University, Novosibirsk, Russia
| | - J M Minina
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | | | - J A Nadtochy
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia Novosibirsk State University, Novosibirsk, Russia
| | - E A Khabarova
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia Federal Neurosurgical Center of the Ministry of Health of the Russian Federation, Novosibirsk, Russia
| | - J A Rzaev
- Federal Neurosurgical Center of the Ministry of Health of the Russian Federation, Novosibirsk, Russia
| | - S P Medvedev
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia Institute of Chemical Biology and Fundamental Medicine of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - S M Zakian
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia Institute of Chemical Biology and Fundamental Medicine of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
21
|
Xu G, Ge R, Zhang C, Zhao Z, Han L, Zhang W, Yue W, Zhang J, Zhao Y, Hou S, Li L, Wang P. Promotion of nerve regeneration and motor function recovery in SCI rats using LOCAS-iPSCs-NSCs. Stem Cell Res Ther 2024; 15:376. [PMID: 39444002 PMCID: PMC11515548 DOI: 10.1186/s13287-024-03999-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 10/14/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Spinal cord injury (SCI) is a severe traumatic spinal condition with a poor prognosis. In this study, a scaffold called linearly ordered collagen aggregates (LOCAS) was created and loaded with induced pluripotent stem cells (iPSCs)-derived neural stem cells (NSCs) from human umbilical cord blood derived mesenchymal stem cells (hUCB-MSCs) to treat SCI in a rat model. METHODS The rats underwent a complete transection SCI resulting in a 3-mm break at either the T9 or T10 level of the spinal cord. RESULTS Scanning electron microscope analysis revealed a uniform pore structure on the coronal plane of the scaffold. The LOCAS had a porosity of 88.52% and a water absorption of 1161.67%. Its compressive modulus and stress were measured at 4.1 MPa and 205 kPa, respectively, with a degradation time of 10 weeks. After 12 weeks, rats in the LOCAS-iPSCs-NSCs group exhibited significantly higher BBB scores (8.6) compared to the LOCAS-iPSCs-NSCs group (5.6) and the Model group (4.2). The CatWalk analysis showed improved motion trajectory, regularity index (RI), and swing speed in the LOCAS-iPSCs-NSCs group compared to the other groups. Motor evoked potentials latency was lower and amplitude was higher in the LOCAS-iPSCs-NSCs group, indicating better neural function recovery. Histological analysis demonstrated enhanced neuronal differentiation of NSCs and nerve fiber regeneration promoted by LOCAS-iPSCs-NSCs, leading to improved motor function recovery in rats. The LOCAS scaffold facilitated ordered neurofilament extension and guided nerve regeneration. CONCLUSIONS The combination of LOCAS and iPSCs-NSCs demonstrated a positive therapeutic impact on motor function recovery and tissue repair in rats with SCI. This development offers a more resilient bionic microenvironment and presents novel possibilities for clinical SCI repair.
Collapse
Affiliation(s)
- Gang Xu
- Department of Orthopaedics, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning Province, China.
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopaedic Diseases, Liaoning Province, Dalian, 116011, Liaoning Province, China.
| | - Rui Ge
- Department of Orthopaedics, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning Province, China
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopaedic Diseases, Liaoning Province, Dalian, 116011, Liaoning Province, China
| | - Chunli Zhang
- Senior Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, Beijing, 100048, China
- Beijing Engineering Research Center of Orthopedics Implants, Beijing, 100048, China
| | - Ziteng Zhao
- Senior Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, Beijing, 100048, China
- Beijing Engineering Research Center of Orthopedics Implants, Beijing, 100048, China
| | - Liwei Han
- Senior Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, Beijing, 100048, China
- Beijing Engineering Research Center of Orthopedics Implants, Beijing, 100048, China
| | - Wanhao Zhang
- Department of Orthopaedics, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning Province, China
| | - WenJie Yue
- Department of Orthopaedics, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning Province, China
| | - Jing Zhang
- Department of Orthopaedics, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning Province, China
| | - Yantao Zhao
- Senior Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, Beijing, 100048, China
- Beijing Engineering Research Center of Orthopedics Implants, Beijing, 100048, China
| | - Shuxun Hou
- Senior Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, Beijing, 100048, China
- Beijing Engineering Research Center of Orthopedics Implants, Beijing, 100048, China
| | - Li Li
- Senior Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, Beijing, 100048, China.
- Beijing Engineering Research Center of Orthopedics Implants, Beijing, 100048, China.
| | - Peng Wang
- Department of Neurosurgery, The First Medical Center of PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
22
|
Kakizuka T, Natsume T, Nagai T. Compact lens-free imager using a thin-film transistor for long-term quantitative monitoring of stem cell culture and cardiomyocyte production. LAB ON A CHIP 2024. [PMID: 39436381 DOI: 10.1039/d4lc00528g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
With advancements in human induced pluripotent stem cell (hiPSC) technology, there is an increasing demand for quality control techniques to manage the long-term process of target cell production effectively. While monitoring systems designed for use within incubators are promising for assessing culture quality, existing systems still face challenges in terms of compactness, throughput, and available metrics. To address these limitations, we have developed a compact and high-throughput lens-free imaging device named INSPCTOR. The device is as small as a standard culture plate, which allows for the installation of multiple units within an incubator. INSPCTOR utilises a large thin-film transistor image sensor, enabling simultaneous observation of six independent culture environments, each approximately 1 cm2. With this device, we successfully monitored the confluency of hiPSC cultures and identified the onset timing of epithelial-to-mesenchymal transition during mesodermal induction. Additionally, we quantified the beating frequency and conduction of hiPSC-derived cardiomyocytes by using high-speed imaging modes. This enabled us to identify the onset of spontaneous beating during differentiation and assess chronotropic responses in drug evaluations. Moreover, by tracking beating frequency over 10 days of cardiomyocyte maturation, we identified week-scale and daily-scale fluctuations, the latter of which correlated with cellular metabolic activity. The metrics derived from this device would enhance the reproducibility and quality of target cell production.
Collapse
Affiliation(s)
- Taishi Kakizuka
- SANKEN, The University of Osaka, Mihogaoka 8-1, Ibaraki, Osaka 567-0047, Japan.
- Transdimensional Life Imaging Division, Institute for Open and Transdisciplinary Research Initiatives, The University of Osaka, Yamadaoka 2-1, Suita, Osaka 565-0871, Japan
| | - Tohru Natsume
- Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology, 2-3-26 Aoumi, Koto-ku, Tokyo 135-0064, Japan
| | - Takeharu Nagai
- SANKEN, The University of Osaka, Mihogaoka 8-1, Ibaraki, Osaka 567-0047, Japan.
- Transdimensional Life Imaging Division, Institute for Open and Transdisciplinary Research Initiatives, The University of Osaka, Yamadaoka 2-1, Suita, Osaka 565-0871, Japan
| |
Collapse
|
23
|
Beaudreault CP, Wang R, Muh CR, Rosenberg A, Funari A, McGoldrick PE, Wolf SM, Sacknovitz A, Chung S. Overcoming Graft Rejection in Induced Pluripotent Stem Cell-Derived Inhibitory Interneurons for Drug-Resistant Epilepsy. Brain Sci 2024; 14:1027. [PMID: 39452039 PMCID: PMC11506040 DOI: 10.3390/brainsci14101027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/14/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Cell-based therapies for drug-resistant epilepsy using induced pluripotent stem cell-derived inhibitory interneurons are now in early-phase clinical trials, building on findings from trials in Parkinson's disease (PD) and Huntington's disease (HD). Graft rejection and the need for immunosuppressive therapy post-transplantation pose potential barriers to more epilepsy patients becoming potential candidates for inhibitory interneurons transplantation surgery. OBJECTIVES The present literature review weighs the evidence for and against human leukocyte antigen (HLA)-mediated graft rejection in PD and HD and examines the potential advantages and drawbacks to five broad approaches to cell-based therapies, including autologous cell culture and transplantation, in vivo reprogramming of glial cells using viral vectors, allogeneic transplantation using off-the-shelf cell lines, transplantation using inhibitory interneurons cultured from HLA-matched cell lines, and the use of hypoimmunogenic-induced pluripotent stem cell-derived inhibitory interneurons. The impact of surgical technique and associated needle trauma on graft rejection is also discussed. METHODS Non-systematic literature review. RESULTS While cell-based therapies have enjoyed early successes in treating a host of central nervous system disorders, the immunologic reaction against surgical procedures and implanted materials has remained a major obstacle. CONCLUSIONS Adapting cell-based therapies using iPSC-derived inhibitory interneurons for epilepsy surgery will similarly require surmounting the challenge of immunogenicity.
Collapse
Affiliation(s)
- Cameron P. Beaudreault
- School of Medicine, New York Medical College, Valhalla, NY 10595, USA; (C.P.B.); (R.W.); (A.R.); (A.S.)
| | - Richard Wang
- School of Medicine, New York Medical College, Valhalla, NY 10595, USA; (C.P.B.); (R.W.); (A.R.); (A.S.)
| | - Carrie Rebecca Muh
- Department of Neurosurgery, Westchester Medical Center, Valhalla, NY 10595, USA
| | - Ashley Rosenberg
- School of Medicine, New York Medical College, Valhalla, NY 10595, USA; (C.P.B.); (R.W.); (A.R.); (A.S.)
| | - Abigail Funari
- Department of Neurosurgery, SUNY Upstate Medical Center, Syracuse, NY 13210, USA
| | - Patty E. McGoldrick
- School of Medicine, New York Medical College, Valhalla, NY 10595, USA; (C.P.B.); (R.W.); (A.R.); (A.S.)
- Division of Pediatric Neurology, Maria Fareri Children’s Hospital, Valhalla, NY 10595, USA
- Division of Pediatric Neurology, Boston Children’s Health Physicians, Hawthorne, NY 10532, USA
| | - Steven M. Wolf
- School of Medicine, New York Medical College, Valhalla, NY 10595, USA; (C.P.B.); (R.W.); (A.R.); (A.S.)
- Division of Pediatric Neurology, Maria Fareri Children’s Hospital, Valhalla, NY 10595, USA
- Division of Pediatric Neurology, Boston Children’s Health Physicians, Hawthorne, NY 10532, USA
| | - Ariel Sacknovitz
- School of Medicine, New York Medical College, Valhalla, NY 10595, USA; (C.P.B.); (R.W.); (A.R.); (A.S.)
- Department of Neurosurgery, Westchester Medical Center, Valhalla, NY 10595, USA
| | - Sangmi Chung
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY 10595, USA
- Department of Neurosurgery, Brain Health Institute, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
| |
Collapse
|
24
|
Sheeler C, Labrada E, Duvick L, Thompson LM, Zhang Y, Orr HT, Cvetanovic M. Expanded ATXN1 alters transcription and calcium signaling in SCA1 human motor neurons differentiated from induced pluripotent stem cells. Neurobiol Dis 2024; 201:106673. [PMID: 39307401 PMCID: PMC11514977 DOI: 10.1016/j.nbd.2024.106673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/12/2024] [Accepted: 09/16/2024] [Indexed: 10/02/2024] Open
Abstract
Spinocerebellar ataxia type 1 (SCA1) is a dominantly inherited and lethal neurodegenerative disease caused by the abnormal expansion of CAG repeats in the ATAXIN-1 (ATXN1) gene. Pathological studies identified dysfunction and loss of motor neurons (MNs) in the brain stem and spinal cord, which are thought to contribute to premature lethality by affecting the swallowing and breathing of SCA1 patients. However, the molecular and cellular mechanisms of MN pathogenesis remain unknown. To study SCA1 pathogenesis in human MNs, we differentiated induced pluripotent stem cells (iPSCs) derived from SCA1 patients and their unaffected siblings into MNs. We examined proliferation of progenitor cells, neurite outgrowth, spontaneous and glutamate-induced calcium activity of SCA1 MNs to investigate cellular mechanisms of pathogenesis. RNA sequencing was then used to identify transcriptional alterations in iPSC-derived MN progenitors (pMNs) and MNs which could underlie functional changes in SCA1 MNs. We found significantly decreased spontaneous and evoked calcium activity and identified dysregulation of genes regulating calcium signaling in SCA1 MNs. These results indicate that expanded ATXN1 causes dysfunctional calcium signaling in human MNs.
Collapse
Affiliation(s)
- Carrie Sheeler
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States of America; Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, United States of America
| | - Emmanuel Labrada
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States of America
| | - Lisa Duvick
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, United States of America
| | - Leslie M Thompson
- Departments of Psychiatry and Human Behavior and Neurobiology and Behavior, University of California, Irvine, United States of America
| | - Ying Zhang
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States of America
| | - Harry T Orr
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, United States of America; Department of Lab Pathology, University of Minnesota, Minneapolis, MN, United States of America
| | - Marija Cvetanovic
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States of America; Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, United States of America.
| |
Collapse
|
25
|
Baggiani M, Damiani D, Privitera F, Della Vecchia S, Tessa A, Santorelli FM. Generation and Characterization of hiPS Lines from Three Patients Affected by Different Forms of HPDL-Related Neurological Disorders. Int J Mol Sci 2024; 25:10614. [PMID: 39408944 PMCID: PMC11477155 DOI: 10.3390/ijms251910614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/26/2024] [Accepted: 09/29/2024] [Indexed: 10/20/2024] Open
Abstract
Hereditary spastic paraplegias are rare genetic disorders characterized by corticospinal tract impairment. Spastic paraplegia 83 (SPG83) is associated with biallelic mutations in the HPDL gene, leading to varied severities from neonatal to juvenile onset. The function of HPDL is unclear, though it is speculated to play a role in alternative coenzyme Q10 biosynthesis. Here, we report the generation of hiPS lines from primary skin fibroblasts derived from three SPG83 patients with different HPDL mutations, using episomal reprogramming. The patients' clinical characteristics are carefully listed. The hiPS lines were meticulously characterized, demonstrating typical pluripotent characteristics through immunofluorescence assays for stemness markers (OCT4, TRA1-60, NANOG, and SSEA4) and RT-PCR for endogenous gene expression. Genetic integrity and identity were confirmed via Sanger sequencing and short tandem repeat analysis. These hiPS cells displayed typical pluripotent characteristics and were able to differentiate into neocortical neurons via a dual SMAD inhibition protocol. In addition, HPDL mutant neurons assessed via long-term culturing were able to achieve effective maturation, similarly to their wild-type counterparts. The HPDL hiPS lines we generated will provide a valuable model for studying SPG83, offering insights into its molecular mechanisms and potential for developing targeted therapies.
Collapse
Affiliation(s)
- Matteo Baggiani
- Molecular Medicine for Neurodegenerative and Neuromuscular Diseases Unit, IRCCS Fondazione Stella Maris, Via dei Giacinti 2, Calambrone, 56128 Pisa, Italy; (M.B.); (F.P.); (S.D.V.); (A.T.)
| | - Devid Damiani
- Molecular Medicine for Neurodegenerative and Neuromuscular Diseases Unit, IRCCS Fondazione Stella Maris, Via dei Giacinti 2, Calambrone, 56128 Pisa, Italy; (M.B.); (F.P.); (S.D.V.); (A.T.)
| | - Flavia Privitera
- Molecular Medicine for Neurodegenerative and Neuromuscular Diseases Unit, IRCCS Fondazione Stella Maris, Via dei Giacinti 2, Calambrone, 56128 Pisa, Italy; (M.B.); (F.P.); (S.D.V.); (A.T.)
| | - Stefania Della Vecchia
- Molecular Medicine for Neurodegenerative and Neuromuscular Diseases Unit, IRCCS Fondazione Stella Maris, Via dei Giacinti 2, Calambrone, 56128 Pisa, Italy; (M.B.); (F.P.); (S.D.V.); (A.T.)
- Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Viale Pieraccini, 6, 50139 Florence, Italy
| | - Alessandra Tessa
- Molecular Medicine for Neurodegenerative and Neuromuscular Diseases Unit, IRCCS Fondazione Stella Maris, Via dei Giacinti 2, Calambrone, 56128 Pisa, Italy; (M.B.); (F.P.); (S.D.V.); (A.T.)
| | - Filippo Maria Santorelli
- Molecular Medicine for Neurodegenerative and Neuromuscular Diseases Unit, IRCCS Fondazione Stella Maris, Via dei Giacinti 2, Calambrone, 56128 Pisa, Italy; (M.B.); (F.P.); (S.D.V.); (A.T.)
| |
Collapse
|
26
|
Lai W, Zhao Y, Chen Y, Dai Z, Chen R, Niu Y, Chen X, Chen S, Huang G, Shan Z, Zheng J, Hu Y, Chen Q, Gong S, Kang S, Guo H, Ma X, Song Y, Xia K, Wang J, Zhou L, So KF, Wang K, Qiu S, Zhang L, Chen J, Shi L. Autism patient-derived SHANK2B Y29X mutation affects the development of ALDH1A1 negative dopamine neuron. Mol Psychiatry 2024; 29:3180-3194. [PMID: 38704506 PMCID: PMC11449796 DOI: 10.1038/s41380-024-02578-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 04/18/2024] [Accepted: 04/22/2024] [Indexed: 05/06/2024]
Abstract
Autism spectrum disorder (ASD) encompasses a range of neurodevelopmental conditions. Different mutations on a single ASD gene contribute to heterogeneity of disease phenotypes, possibly due to functional diversity of generated isoforms. SHANK2, a causative gene in ASD, demonstrates this phenomenon, but there is a scarcity of tools for studying endogenous SHANK2 proteins in an isoform-specific manner. Here, we report a point mutation on SHANK2, which is found in a patient with autism, located on exon of the SHANK2B transcript variant (NM_133266.5), hereby SHANK2BY29X. This mutation results in an early stop codon and an aberrant splicing event that impacts SHANK2 transcript variants distinctly. Induced pluripotent stem cells (iPSCs) carrying this mutation, from the patient or isogenic editing, fail to differentiate into functional dopamine (DA) neurons, which can be rescued by genetic correction. Available SMART-Seq single-cell data from human midbrain reveals the abundance of SHANK2B transcript in the ALDH1A1 negative DA neurons. We then show that SHANK2BY29X mutation primarily affects SHANK2B expression and ALDH1A1 negative DA neurons in vitro during early neuronal developmental stage. Mice knocked in with the identical mutation exhibit autistic-like behavior, decreased occupancy of ALDH1A1 negative DA neurons and decreased dopamine release in ventral tegmental area (VTA). Our study provides novel insights on a SHANK2 mutation derived from autism patient and highlights SHANK2B significance in ALDH1A1 negative DA neuron.
Collapse
Affiliation(s)
- Wanjing Lai
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangdong Key Laboratory of Non-human Primate Research, Guangzhou, 510632, China
| | - Yingying Zhao
- Center for Cell Lineage and Development, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Joint School of Life Sciences, Guangzhou Medical University, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, 999077, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yalan Chen
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangdong Key Laboratory of Non-human Primate Research, Guangzhou, 510632, China
| | - Zhenzhu Dai
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangdong Key Laboratory of Non-human Primate Research, Guangzhou, 510632, China
| | - Ruhai Chen
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangdong Key Laboratory of Non-human Primate Research, Guangzhou, 510632, China
| | - Yimei Niu
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangdong Key Laboratory of Non-human Primate Research, Guangzhou, 510632, China
| | - Xiaoxia Chen
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangdong Key Laboratory of Non-human Primate Research, Guangzhou, 510632, China
| | - Shuting Chen
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangdong Key Laboratory of Non-human Primate Research, Guangzhou, 510632, China
| | - Guanqun Huang
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangdong Key Laboratory of Non-human Primate Research, Guangzhou, 510632, China
| | - Ziyun Shan
- Center for Cell Lineage and Development, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Joint School of Life Sciences, Guangzhou Medical University, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jiajun Zheng
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangdong Key Laboratory of Non-human Primate Research, Guangzhou, 510632, China
| | - Yu Hu
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Qingpei Chen
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangdong Key Laboratory of Non-human Primate Research, Guangzhou, 510632, China
| | - Siyi Gong
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangdong Key Laboratory of Non-human Primate Research, Guangzhou, 510632, China
| | - Sai Kang
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangdong Key Laboratory of Non-human Primate Research, Guangzhou, 510632, China
| | - Hui Guo
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, 410008, China
| | - Xiaokuang Ma
- Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, 850004, USA
| | - Youqiang Song
- School of Biomedical Sciences, University of Hong Kong, Hong Kong SAR, China
| | - Kun Xia
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, 410008, China
| | - Jie Wang
- Center for Cell Lineage and Development, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Joint School of Life Sciences, Guangzhou Medical University, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Libing Zhou
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangdong Key Laboratory of Non-human Primate Research, Guangzhou, 510632, China
| | - Kwok-Fai So
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangdong Key Laboratory of Non-human Primate Research, Guangzhou, 510632, China
| | - Kai Wang
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Shenfeng Qiu
- Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, 850004, USA
| | - Li Zhang
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangdong Key Laboratory of Non-human Primate Research, Guangzhou, 510632, China.
| | - Jiekai Chen
- Center for Cell Lineage and Development, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Joint School of Life Sciences, Guangzhou Medical University, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, 999077, China.
| | - Lingling Shi
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangdong Key Laboratory of Non-human Primate Research, Guangzhou, 510632, China.
- Department of Psychiatry, the First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, 510632, China.
- Co-innovation Center of Neuro-regeneration, Nantong University, Nantong, Jiangsu, 226019, China.
- Department of Neurology, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China.
| |
Collapse
|
27
|
Nakamura M, Chonabayashi K, Narita M, Matsumura Y, Nishikawa M, Ochi Y, Nannya Y, Hishizawa M, Inoue D, Delwel R, Ogawa S, Takaori-Kondo A, Yoshida Y. Modelling and drug targeting of a myeloid neoplasm with atypical 3q26/MECOM rearrangement using patient-specific iPSCs. Br J Haematol 2024; 205:1430-1443. [PMID: 39187468 DOI: 10.1111/bjh.19720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 08/11/2024] [Indexed: 08/28/2024]
Abstract
Structural variations involving enhancer hijacking induce aberrant oncogene expression and cause tumorigenesis. A rare translocation, t(3;8)(q26.2;q24), is associated with MECOM and MYC rearrangement, causing myeloid neoplasms with a dismal prognosis. The most recent World Health Organization classification recognises myeloid neoplasms with MECOM rearrangement as acute myeloid leukaemia (AML) with defining genetic abnormalities. Recently, the increasing use of induced pluripotent stem cell (iPSC) technology has helped elucidate the pathogenic processes of haematological malignancies. However, its utility for investigating enhancer hijacking in myeloid neoplasms remains unclear. In this study, we generated iPSC lines from patients with myelodysplastic syndromes (MDS) harbouring t(3;8)(q26.2;q24) and differentiated them into haematopoietic progenitor cells to model the pathophysiology of MDS with t(3;8)(q26.2;q24). Our iPSC model reproduced the primary patient's MECOM expression changes and histone H3 lysine 27 acetylation (H3K27ac) patterns in the MECOM promoter and MYC blood enhancer cluster (BENC). Furthermore, we revealed the apoptotic effects of the bromodomain and extra-terminal motif (BET) inhibitor on iPSC-derived MDS cells by suppressing activated MECOM. Our study demonstrates the usefulness of iPSC models for uncovering the precise mechanism of enhancer hijacking due to chromosomal structural changes and discovering potential therapeutic drug candidates for cancer treatment.
Collapse
MESH Headings
- Humans
- Induced Pluripotent Stem Cells/metabolism
- Induced Pluripotent Stem Cells/drug effects
- Myelodysplastic Syndromes/genetics
- Myelodysplastic Syndromes/pathology
- Myelodysplastic Syndromes/drug therapy
- Myelodysplastic Syndromes/metabolism
- Chromosomes, Human, Pair 3/genetics
- Translocation, Genetic
- Chromosomes, Human, Pair 8/genetics
- Proto-Oncogene Proteins c-myc/genetics
- Proto-Oncogene Proteins c-myc/metabolism
- Gene Rearrangement
- Male
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Azepines/pharmacology
- Female
Collapse
Affiliation(s)
- Momoko Nakamura
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kazuhisa Chonabayashi
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Megumi Narita
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Yasuko Matsumura
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Misato Nishikawa
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Yotaro Ochi
- Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yasuhito Nannya
- Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Division of Hematopoietic Disease Control, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Masakatsu Hishizawa
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Department of Hematology, Kyoto-Katsura Hospital, Kyoto, Japan
| | - Daichi Inoue
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Hyogo, Japan
| | - Ruud Delwel
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Seishi Ogawa
- Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akifumi Takaori-Kondo
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yoshinori Yoshida
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| |
Collapse
|
28
|
Nakashima Y, Tsukahara M. Atelocollagen supports three-dimensional culture of human induced pluripotent stem cells. Mol Ther Methods Clin Dev 2024; 32:101302. [PMID: 39185274 PMCID: PMC11342089 DOI: 10.1016/j.omtm.2024.101302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 07/16/2024] [Indexed: 08/27/2024]
Abstract
As autologous induced pluripotent stem cell (iPSC) therapy requires a custom-made small-lot cell production line, and the cell production method differs significantly from the existing processes for producing allogeneic iPSC stocks for clinical use. Specifically, mass culture to produce stock is no longer necessary; instead, a series of operations from iPSC production to induction of differentiation of therapeutic cells must be performed continuously. A three-dimensional (3D) culture method using small, closed-cell manufacturing devices is suitable for autologous iPSC therapy. The use of such devices avoids the need to handle many patient-derived specimens in a single clean room; handling of cell cultures in an open system in a cell processing facility increases the risk of infection. In this study, atelocollagen beads were evaluated as a 3D biomaterial to assist 3D culture in the establishment, expansion culture, and induction of differentiation of iPSCs. It was found that iPSCs can be handled in a closed-cell device with the same ease as use of a two-dimensional (2D) culture when laminin-511 is added to the medium. In conclusion, atelocollagen beads enable 3D culture of iPSCs, and the quality of the obtained cells is at the same level as those derived from 2D culture.
Collapse
Affiliation(s)
- Yoshiki Nakashima
- CiRA Foundation, Research and Development Center, Nakanoshima Qross, Osaka 530-005, Japan
| | - Masayoshi Tsukahara
- CiRA Foundation, Research and Development Center, Nakanoshima Qross, Osaka 530-005, Japan
| |
Collapse
|
29
|
Terheyden-Keighley D, Hühne M, Berger T, Hiller B, Martins S, Gamerschlag A, Sabour D, Meffert A, Kislat A, Slotta C, Hafezi F, Lichte J, Sudheer S, Tessmer K, Psathaki K, Ader M, Kogler G, Greber B. GMP-compliant iPS cell lines show widespread plasticity in a new set of differentiation workflows for cell replacement and cancer immunotherapy. Stem Cells Transl Med 2024; 13:898-911. [PMID: 39042522 PMCID: PMC11386223 DOI: 10.1093/stcltm/szae047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 06/08/2024] [Indexed: 07/25/2024] Open
Abstract
Cell therapeutic applications based on induced pluripotent stem cells (iPSCs) appear highly promising and challenging at the same time. Good manufacturing practice (GMP) regulations impose necessary yet demanding requirements for quality and consistency when manufacturing iPSCs and their differentiated progeny. Given the scarcity of accessible GMP iPSC lines, we have established a corresponding production workflow to generate the first set of compliant cell banks. Hence, these lines met a comprehensive set of release specifications and, for instance, displayed a low overall mutation load reflecting their neonatal origin, cord blood. Based on these iPSC lines, we have furthermore developed a set of GMP-compatible workflows enabling improved gene targeting at strongly enhanced efficiencies and directed differentiation into critical cell types: A new protocol for the generation of retinal pigment epithelium (RPE) features a high degree of simplicity and efficiency. Mesenchymal stromal cells (MSCs) derived from iPSCs displayed outstanding expansion capacity. A fully optimized cardiomyocyte differentiation protocol was characterized by a particularly high batch-to-batch consistency at purities above 95%. Finally, we introduce a universal immune cell induction platform that converts iPSCs into multipotent precursor cells. These hematopoietic precursors could selectively be stimulated to become macrophages, T cells, or natural killer (NK) cells. A switch in culture conditions upon NK-cell differentiation induced a several thousand-fold expansion, which opens up perspectives for upscaling this key cell type in a feeder cell-independent approach. Taken together, these cell lines and improved manipulation platforms will have broad utility in cell therapy as well as in basic research.
Collapse
Affiliation(s)
| | | | | | - Björn Hiller
- Catalent Düsseldorf GmbH, 40764 Langenfeld, Germany
| | | | | | | | | | | | | | | | - Jens Lichte
- Catalent Düsseldorf GmbH, 40764 Langenfeld, Germany
| | | | - Karen Tessmer
- Center for Regenerative Therapies Dresden (CRTD) and Center for Molecular and Cellular Bioengineering, Dresden University of Technology, 01307 Dresden, Germany
| | - Katherina Psathaki
- Center for Cellular Nanoanalytics (CellNanOs), University of Osnabrück, 49076 Osnabrück, Germany
| | - Marius Ader
- Center for Regenerative Therapies Dresden (CRTD) and Center for Molecular and Cellular Bioengineering, Dresden University of Technology, 01307 Dresden, Germany
| | - Gesine Kogler
- Institute of Transplantation Diagnostics and Cell Therapeutics and Jose Carreras Stem Cell Bank, University Hospital of Düsseldorf, 40225 Düsseldorf, Germany
| | - Boris Greber
- Catalent Düsseldorf GmbH, 40764 Langenfeld, Germany
| |
Collapse
|
30
|
Mazzini L, De Marchi F, Buzanska L, Follenzi A, Glover JC, Gelati M, Lombardi I, Maioli M, Mesa-Herrera F, Mitrečić D, Olgasi C, Pivoriūnas A, Sanchez-Pernaute R, Sgromo C, Zychowicz M, Vescovi A, Ferrari D. Current status and new avenues of stem cell-based preclinical and therapeutic approaches in amyotrophic lateral sclerosis. Expert Opin Biol Ther 2024; 24:933-954. [PMID: 39162129 DOI: 10.1080/14712598.2024.2392307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 08/10/2024] [Indexed: 08/21/2024]
Abstract
INTRODUCTION Cell therapy development represents a critical challenge in amyotrophic lateral sclerosis (ALS) research. Despite more than 20 years of basic and clinical research, no definitive safety and efficacy results of cell-based therapies for ALS have been published. AREAS COVERED This review summarizes advances using stem cells (SCs) in pre-clinical studies to promote clinical translation and in clinical trials to treat ALS. New technologies have been developed and new experimental in vitro and animal models are now available to facilitate pre-clinical research in this field and to determine the most promising approaches to pursue in patients. New clinical trial designs aimed at developing personalized SC-based treatment with biological endpoints are being defined. EXPERT OPINION Knowledge of the basic biology of ALS and on the use of SCs to study and potentially treat ALS continues to grow. However, a consensus has yet to emerge on how best to translate these results into therapeutic applications. The selection and follow-up of patients should be based on clinical, biological, and molecular criteria. Planning of SC-based clinical trials should be coordinated with patient profiling genetically and molecularly to achieve personalized treatment. Much work within basic and clinical research is still needed to successfully transition SC therapy in ALS.
Collapse
Affiliation(s)
- Letizia Mazzini
- ALS Center, Neurology Unit, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Fabiola De Marchi
- ALS Center, Neurology Unit, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Leonora Buzanska
- Department of Stem Cell Bioengineering, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Antonia Follenzi
- Dipartimento di Scienze della Salute, Università del Piemonte Orientale, Novara, Italy
- Dipartimento Attività Integrate Ricerca Innovazione, Azienda Ospedaliero-Universitaria SS. Antonio e Biagio e C. Arrigo, Alessandria, Italy
| | - Joel Clinton Glover
- Norwegian Center for Stem Cell Research, Department of Immunology and Transfusion Medicine, Oslo University Hospital; Laboratory of Neural Development and Optical Recording (NDEVOR), Oslo, Norway
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Maurizio Gelati
- Unità Produttiva per Terapie Avanzate (UPTA), IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Ivan Lombardi
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milano, Italy
| | - Margherita Maioli
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
- Center for Developmental Biology and Reprogramming-CEDEBIOR, University of Sassari, Sassari, Italy
| | - Fatima Mesa-Herrera
- Reprogramming and Neural Regeneration Lab, BioBizkaia Health Research Institute, Barakaldo, Spain
| | - Dinko Mitrečić
- Laboratory for Stem Cells, Croatian Institute for Brain Research and Department of Histology and Embryology, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Cristina Olgasi
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Augustas Pivoriūnas
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| | - Rosario Sanchez-Pernaute
- Reprogramming and Neural Regeneration Lab, BioBizkaia Health Research Institute, Barakaldo, Spain
- Ikerbaske, Basque Foundation for Science, Bilbao, Spain
| | - Chiara Sgromo
- Dipartimento di Scienze della Salute, Università del Piemonte Orientale, Novara, Italy
| | - Marzena Zychowicz
- Department of Stem Cell Bioengineering, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Angelo Vescovi
- Unità Produttiva per Terapie Avanzate (UPTA), IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milano, Italy
| | - Daniela Ferrari
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milano, Italy
| |
Collapse
|
31
|
Munezane H, Imamura K, Fujimoto N, Hotta A, Yukitake H, Inoue H. Elimination of the extra chromosome of Dup15q syndrome iPSCs for cellular and molecular investigation. Eur J Cell Biol 2024; 103:151446. [PMID: 39059105 DOI: 10.1016/j.ejcb.2024.151446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 06/23/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Chromosome 15q11.2-13.1 duplication (Dup15q) syndrome is one of the most common autism spectrum disorders (ASDs) associated with copy number variants (CNVs). For the analysis of CNV-relevant pathological cellular phenotypes, a CNV-corrected isogenic cell line is useful for excluding the influence of genetic background. Here, we devised a strategy to remove the isodicentric chromosome 15 by inserting a puro-ΔTK selection cassette into the extra chromosome using the CRISPR-Cas9 system, followed by a subsequent two-step drug selection. A series of assays, including qPCR-based copy number analysis and karyotype analysis, confirmed the elimination of the extra chromosome. Furthermore, cerebral organoids were generated from the parental Dup15q iPSCs and their isogenic iPSCs. scRNA-seq analysis revealed the alteration of expression levels in ion-channel-related genes and synapse-related genes in glutamatergic and GABAergic neurons in Dup15q organoids, respectively. The established isogenic cell line is a valuable resource for unraveling cellular and molecular alterations associated with Dup15q syndrome.
Collapse
Affiliation(s)
- Haruka Munezane
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan; Takeda-CiRA (T-CiRA) Joint Program, 2-26-1, Muraoka-Higashi, Fujisawa 251-8555, Japan
| | - Keiko Imamura
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan; Takeda-CiRA (T-CiRA) Joint Program, 2-26-1, Muraoka-Higashi, Fujisawa 251-8555, Japan; iPSC-based Drug discovery and Development Team, RIKEN BioResource Research Center, 1-7 Hikaridai, Seika-cho, Soraku-gun, Kyoto 619-0237, Japan; Medical-Risk Avoidance based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP), 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Naoko Fujimoto
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan; Takeda-CiRA (T-CiRA) Joint Program, 2-26-1, Muraoka-Higashi, Fujisawa 251-8555, Japan
| | - Akitsu Hotta
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan; Takeda-CiRA (T-CiRA) Joint Program, 2-26-1, Muraoka-Higashi, Fujisawa 251-8555, Japan
| | - Hiroshi Yukitake
- Takeda-CiRA (T-CiRA) Joint Program, 2-26-1, Muraoka-Higashi, Fujisawa 251-8555, Japan; Global Advanced Platform, Takeda Pharmaceutical Company Limited, 2-26-1, Muraoka-Higashi, Fujisawa 251-8555, Japan
| | - Haruhisa Inoue
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan; Takeda-CiRA (T-CiRA) Joint Program, 2-26-1, Muraoka-Higashi, Fujisawa 251-8555, Japan; iPSC-based Drug discovery and Development Team, RIKEN BioResource Research Center, 1-7 Hikaridai, Seika-cho, Soraku-gun, Kyoto 619-0237, Japan; Medical-Risk Avoidance based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP), 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan.
| |
Collapse
|
32
|
Baggiani M, Santorelli FM, Mero S, Privitera F, Damiani D, Tessa A. Generation of a human induced pluripotent stem cell line (FSMi001-A) from fibroblasts of a patient carrying heterozygous mutation in the REEP1 gene. Stem Cell Res 2024; 79:103472. [PMID: 38889632 PMCID: PMC11307172 DOI: 10.1016/j.scr.2024.103472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/03/2024] [Accepted: 06/10/2024] [Indexed: 06/20/2024] Open
Abstract
Hereditary spastic paraplegias (HSPs) a group of rare, clinically, and genetically heterogeneous disorders characterized by progressive degeneration of the corticospinal tract. Among these HSPs, SPG31 is due to autosomal dominant mutations in the receptor expression-enhancing protein 1 (REEP1) gene. Over 80 genes have been associated with HSPs, and the list is constantly growing as research progresses. This study is aimed to create a patient-derived human induced pluripotent stem cell (hiPSC) line with a specific nonsense mutation to better characterize the etiopathogenesis of the disease.
Collapse
Affiliation(s)
- Matteo Baggiani
- Department of Neurobiology and Molecular Medicine, IRCCS Fondazione Stella Maris, 56128 Pisa, Italy.
| | - Filippo Maria Santorelli
- Department of Neurobiology and Molecular Medicine, IRCCS Fondazione Stella Maris, 56128 Pisa, Italy.
| | - Serena Mero
- Department of Neurobiology and Molecular Medicine, IRCCS Fondazione Stella Maris, 56128 Pisa, Italy
| | - Flavia Privitera
- Department of Neurobiology and Molecular Medicine, IRCCS Fondazione Stella Maris, 56128 Pisa, Italy
| | - Devid Damiani
- Department of Neurobiology and Molecular Medicine, IRCCS Fondazione Stella Maris, 56128 Pisa, Italy.
| | - Alessandra Tessa
- Department of Neurobiology and Molecular Medicine, IRCCS Fondazione Stella Maris, 56128 Pisa, Italy
| |
Collapse
|
33
|
Saegusa C, Mutai H, Saeki T, Matsuzaki S, Mizukoshi A, Kitajiri SI, Matsunaga T, Hosoya M, Okano H, Fujioka M. Generation of four induced pluripotent stem cell lines (KEIUi004-A, KEIUi005-A, KEIUi006-A, and KEIUi007-A) from patients with sensorineural hearing loss with mutation in EYA4 gene. Stem Cell Res 2024; 79:103489. [PMID: 39002249 DOI: 10.1016/j.scr.2024.103489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 07/06/2024] [Indexed: 07/15/2024] Open
Abstract
Disease-related cells differentiated from patient-derived iPSCs are useful for elucidating the pathophysiological mechanisms underlying these diseases. In this study, four iPSC lines were established from independent patients with sensorineural hearing loss and a mutation in EYA4. These iPSCs showed pluripotency, the capacity to differentiate into three germ layers, and normal karyotypes, suggesting that these lines are useful for the pathological study of sensorineural hearing loss and drug screening for ear disorders.
Collapse
Affiliation(s)
- Chika Saegusa
- Department of Molecular Genetics, Kitasato University School of Medicine, Kanagawa, Japan; Molecular Genetics Unit, Kitasato University Graduate School of Medical Science, Kanagawa, Japan; Department of Otolaryngology, Head and Neck Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Hideki Mutai
- Department of Molecular Genetics, Kitasato University School of Medicine, Kanagawa, Japan; Molecular Genetics Unit, Kitasato University Graduate School of Medical Science, Kanagawa, Japan; Division of Hearing and Balance Research, National Institute of Sensory Organs, NHO Tokyo Medical Center, Tokyo, Japan
| | - Tsubasa Saeki
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Saeko Matsuzaki
- Department of Molecular Genetics, Kitasato University School of Medicine, Kanagawa, Japan; Department of Otolaryngology, Head and Neck Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Akifumi Mizukoshi
- Department of Otolaryngology Head and Neck Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan; Department of Otolaryngology, Kyoto Katsura Hospital, Kyoto, Japan
| | - Shin-Ichiro Kitajiri
- Department of Otolaryngology Head and Neck Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tatsuo Matsunaga
- Division of Hearing and Balance Research, National Institute of Sensory Organs, NHO Tokyo Medical Center, Tokyo, Japan; Medical Genetics Center, NHO Tokyo Medical Center, Tokyo, Japan
| | - Makoto Hosoya
- Department of Otolaryngology, Head and Neck Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan; Keio University Regenerative Medicine Research Center, Kanagawa, Japan; Division of CNS Regeneration and Drug Discovery, International Center for Brain Science, Fujita Health University, Aichi, Japan.
| | - Masato Fujioka
- Department of Molecular Genetics, Kitasato University School of Medicine, Kanagawa, Japan; Molecular Genetics Unit, Kitasato University Graduate School of Medical Science, Kanagawa, Japan; Department of Otolaryngology, Head and Neck Surgery, Keio University School of Medicine, Tokyo, Japan; Clinical and Translational Research Center, Keio University Hospital, Tokyo, Japan.
| |
Collapse
|
34
|
Kishimura U, Soeda S, Ito D, Ueta Y, Harada M, Tanaka M, Taniura H. Pathological analysis of Prader-Willi syndrome using adipocytes. Biochem Biophys Res Commun 2024; 721:150124. [PMID: 38776833 DOI: 10.1016/j.bbrc.2024.150124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 05/08/2024] [Accepted: 05/13/2024] [Indexed: 05/25/2024]
Abstract
Prader-Willi syndrome (PWS) is a complex epigenetic disorder caused by the deficiency of paternally expressed genes in chromosome 15q11-q13. This syndrome also includes endocrine dysfunction, leading to short stature, hypogonadism, and obscure hyperphagia. Although recent progress has been made toward understanding the genetic basis for PWS, the molecular mechanisms underlying its pathology in obesity remain unclear. In this study, we examined the adipocytic characteristics of two PWS-induced pluripotent stem cell (iPSC) lines: those with the 15q11-q13 gene deletion (iPWS cells) and those with 15q11-q13 abnormal methylation (M-iPWS cells). The transcript levels of the lipid-binding protein aP2 were decreased in iPWS and M-iPWS adipocytes. Flow-cytometry analysis showed that PWS adipocytes accumulated more lipid droplets than did normal individual adipocytes. Furthermore, glucose uptake upon insulin stimulation was attenuated compared to that in normal adipocytes. Overall, our results suggest a significantly increased lipid content and defective in glucose metabolism in PWS adipocytes.
Collapse
Affiliation(s)
- Urara Kishimura
- Laboratory of Neurochemistry, College of Pharmaceutical Sciences, Ritsumeikan University, Shiga, 525-8577, Japan
| | - Shuhei Soeda
- Laboratory of Neurochemistry, College of Pharmaceutical Sciences, Ritsumeikan University, Shiga, 525-8577, Japan.
| | - Daiki Ito
- Laboratory of Neurochemistry, College of Pharmaceutical Sciences, Ritsumeikan University, Shiga, 525-8577, Japan
| | - Yoko Ueta
- Laboratory of Neurochemistry, College of Pharmaceutical Sciences, Ritsumeikan University, Shiga, 525-8577, Japan
| | - Maki Harada
- Laboratory of Neurochemistry, College of Pharmaceutical Sciences, Ritsumeikan University, Shiga, 525-8577, Japan
| | - Mai Tanaka
- Laboratory of Neurochemistry, College of Pharmaceutical Sciences, Ritsumeikan University, Shiga, 525-8577, Japan
| | - Hideo Taniura
- Laboratory of Neurochemistry, College of Pharmaceutical Sciences, Ritsumeikan University, Shiga, 525-8577, Japan
| |
Collapse
|
35
|
Masano Y, Saegusa C, Ishikawa M, Matsunaga T, Okano H, Fujioka M. Generation of an induced pluripotent stem cell line (KEIUi008-A) from a hearing loss patient with an A1555G mutation in mitochondrial DNA. Stem Cell Res 2024; 78:103452. [PMID: 38815527 DOI: 10.1016/j.scr.2024.103452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 05/13/2024] [Accepted: 05/24/2024] [Indexed: 06/01/2024] Open
Abstract
We report the establishment of a human induced pluripotent stem cell (iPSC) line from a 54-year-old male patient with an A1555G mutation in the mitochondrial 12S ribosomal RNA gene (MTRNR1), associated with sensorineural hearing loss. The established iPSC line expressed stemness markers or undifferentiated state markers. We also demonstrated the capacity of the cells to differentiate into the three germ layers, suggesting its pluripotency and utility in the pathological study of sensorineural hearing loss and drug screening for ear disorders.
Collapse
Affiliation(s)
- Yusuke Masano
- Department of Molecular Genetics, Kitasato University School of Medicine, Kanagawa, Japan
| | - Chika Saegusa
- Department of Molecular Genetics, Kitasato University School of Medicine, Kanagawa, Japan; Molecular Genetics Unit, Kitasato University Graduate School of Medical Science, Kanagawa, Japan; Department of Otolaryngology, Head and Neck Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Mitsuru Ishikawa
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan; Division of CNS Regeneration and Drug Discovery, International Center for Brain Science, Fujita Health University, Aichi, Japan
| | - Tatsuo Matsunaga
- Division of Hearing and Balance Research, National Institute of Sensory Organs, NHO Tokyo Medical Center, Tokyo, Japan; Medical Genetics Center, NHO Tokyo Medical Center, Tokyo, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan; Division of CNS Regeneration and Drug Discovery, International Center for Brain Science, Fujita Health University, Aichi, Japan; Keio University Regenerative Medicine Research Center, Kanagawa, Japan
| | - Masato Fujioka
- Department of Molecular Genetics, Kitasato University School of Medicine, Kanagawa, Japan; Molecular Genetics Unit, Kitasato University Graduate School of Medical Science, Kanagawa, Japan; Department of Otolaryngology, Head and Neck Surgery, Keio University School of Medicine, Tokyo, Japan; Clinical and Translational Research Center, Keio University Hospital, Tokyo, Japan
| |
Collapse
|
36
|
Oliveira D, Assoni AF, Alves LM, Sakugawa A, Melo US, Teles E Silva AL, Sertie AL, Caires LC, Goulart E, Ghirotto B, Carvalho VM, Ferrari MR, Zatz M. ALS-associated VRK1 R321C mutation causes proteostatic imbalance and mitochondrial defects in iPSC-derived motor neurons. Neurobiol Dis 2024; 198:106540. [PMID: 38806131 DOI: 10.1016/j.nbd.2024.106540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 05/30/2024] Open
Abstract
Vaccinia-related kinase 1 (VRK1) is a gene which has been implicated in the pathological process of a broad range of neurodevelopmental disorders as well as neuropathies, such as Amyotrophic Lateral Sclerosis (ALS). Here we report a family presenting ALS in an autosomal recessive mode of inheritance, segregating with a homozygous missense mutation located in VRK1 gene (p.R321C; Arg321Cys). Proteomic analyses from iPSC-derived motor neurons identified 720 proteins eligible for subsequent investigation, and our exploration of protein profiles revealed significant enrichments in pathways such as mTOR signaling, E2F, MYC targets, DNA repair response, cell proliferation and energetic metabolism. Functional studies further validated such alterations, showing that affected motor neurons presented decreased levels of global protein output, ER stress and downregulation of mTOR signaling. Mitochondrial alterations also pointed to decreased reserve capacity and increased non-mitochondrial oxygen consumption. Taken together, our results present the main pathological alterations associated with VRK1 mutation in ALS.
Collapse
Affiliation(s)
- D Oliveira
- Human Genome and Stem Cell Research Center, Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil; School of Medical Sciences Santa Casa and Pathological Sciences Unit, Irmandade Santa Casa de Misericordia de Sao Paulo, Sao Paulo, Brazil.
| | - A F Assoni
- Human Genome and Stem Cell Research Center, Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil
| | - L M Alves
- Human Genome and Stem Cell Research Center, Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil
| | - A Sakugawa
- Human Genome and Stem Cell Research Center, Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil
| | - U S Melo
- Max Planck Institute for Molecular Genetics, RG Development and Disease, Berlin, Germany
| | | | - A L Sertie
- Hospital Albert Einstein, São Paulo, São Paulo, Brazil
| | - L C Caires
- Human Genome and Stem Cell Research Center, Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil
| | - E Goulart
- Human Genome and Stem Cell Research Center, Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil
| | - B Ghirotto
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - V M Carvalho
- Division of Research and Development, Fleury Group, São Paulo, SP 04344-070, Brazil
| | - M R Ferrari
- Human Genome and Stem Cell Research Center, Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil
| | - M Zatz
- Human Genome and Stem Cell Research Center, Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
37
|
Han JW, Chang HS, Park SC, Yang JY, Kim YJ, Kim JH, Park HS, Jeong H, Lee J, Yoon CK, Yu HG, Woo SJ, Lyu J, Park TK. Early Developmental Characteristics and Features of a Three-Dimensional Retinal Organoid Model of X-Linked Juvenile Retinoschisis. Int J Mol Sci 2024; 25:8203. [PMID: 39125773 PMCID: PMC11311801 DOI: 10.3390/ijms25158203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
X-linked juvenile retinoschisis (XLRS) is a hereditary retinal degeneration affecting young males caused by mutations in the retinoschisin (RS1) gene. We generated human induced pluripotent stem cells (hiPSCs) from XLRS patients and established three-dimensional retinal organoids (ROs) for disease investigation. This disease model recapitulates the characteristics of XLRS, exhibiting defects in RS1 protein production and photoreceptor cell development. XLRS ROs also revealed dysregulation of Na/K-ATPase due to RS1 deficiency and increased ERK signaling pathway activity. Transcriptomic analyses of XLRS ROs showed decreased expression of retinal cells, particularly photoreceptor cells. Furthermore, relevant recovery of the XLRS phenotype was observed when co-cultured with control ROs derived from healthy subject during the early stages of differentiation. In conclusion, our in vitro XLRS RO model presents a valuable tool for elucidating the pathophysiological mechanisms underlying XLRS, offering insights into disease progression. Additionally, this model serves as a robust platform for the development and optimization of targeted therapeutic strategies, potentially improving treatment outcomes for patients with XLRS.
Collapse
Affiliation(s)
- Jung Woo Han
- Department of Ophthalmology, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, 170, Jomaru-ro, Bucheon 14584, Republic of Korea; (J.W.H.); (S.C.P.); (J.H.K.); (H.S.P.)
- Department of Ophthalmology, Soonchunhyang University College of Medicine, Cheonan 31151, Republic of Korea
| | - Hun Soo Chang
- Department of Microbiology, Soonchunhyang University College of Medicine, Cheonan 31151, Republic of Korea;
- Department of Interdisciplinary Program in Biomedical Science, Soonchunhyang Graduate School, Soonchunhyang University Bucheon Hospital, Bucheon 14584, Republic of Korea;
| | - Sung Chul Park
- Department of Ophthalmology, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, 170, Jomaru-ro, Bucheon 14584, Republic of Korea; (J.W.H.); (S.C.P.); (J.H.K.); (H.S.P.)
| | - Jin Young Yang
- Laboratory of Molecular Therapy for Retinal Degeneration, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon 14584, Republic of Korea;
| | - Ye Ji Kim
- Department of Interdisciplinary Program in Biomedical Science, Soonchunhyang Graduate School, Soonchunhyang University Bucheon Hospital, Bucheon 14584, Republic of Korea;
| | - Jin Ha Kim
- Department of Ophthalmology, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, 170, Jomaru-ro, Bucheon 14584, Republic of Korea; (J.W.H.); (S.C.P.); (J.H.K.); (H.S.P.)
- Department of Ophthalmology, Soonchunhyang University College of Medicine, Cheonan 31151, Republic of Korea
| | - Hyo Song Park
- Department of Ophthalmology, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, 170, Jomaru-ro, Bucheon 14584, Republic of Korea; (J.W.H.); (S.C.P.); (J.H.K.); (H.S.P.)
- Department of Ophthalmology, Soonchunhyang University College of Medicine, Cheonan 31151, Republic of Korea
| | - Han Jeong
- Institute of Vision Research, Department of Ophthalmology, Severance Eye Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea;
- Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Junwon Lee
- Institute of Vision Research, Department of Ophthalmology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Republic of Korea;
| | - Chang Ki Yoon
- Department of Ophthalmology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea;
| | - Hyung Gon Yu
- Retina Center, The Sky Eye Institute, Seoul 06536, Republic of Korea;
| | - Se Joon Woo
- Department of Ophthalmology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam 13620, Republic of Korea;
| | - Jungmook Lyu
- Department of Medical Science, Konyang University, Daejun 32992, Republic of Korea;
| | - Tae Kwann Park
- Department of Ophthalmology, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, 170, Jomaru-ro, Bucheon 14584, Republic of Korea; (J.W.H.); (S.C.P.); (J.H.K.); (H.S.P.)
- Department of Ophthalmology, Soonchunhyang University College of Medicine, Cheonan 31151, Republic of Korea
- Department of Interdisciplinary Program in Biomedical Science, Soonchunhyang Graduate School, Soonchunhyang University Bucheon Hospital, Bucheon 14584, Republic of Korea;
- Laboratory of Molecular Therapy for Retinal Degeneration, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon 14584, Republic of Korea;
| |
Collapse
|
38
|
Nakamura Y, Kobayashi H, Fukuda N, Tanaka S, Murata Y, Hatanaka Y, Haketa A, Tsunemi A, Chen L, Abe M. Induced pluripotent stem cells derived renal tubular cells from a patient with pseudohypoparathyroidism and its response to parathyroid hormone stimulation. Mol Biol Rep 2024; 51:790. [PMID: 38990390 DOI: 10.1007/s11033-024-09751-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 06/24/2024] [Indexed: 07/12/2024]
Abstract
INTRODUCTION Creating induced pluripotent stem cells (iPSCs) from somatic cells of patients with genetic diseases offers a pathway to generate disease-specific iPSCs carrying genetic markers. Differentiating these iPSCs into renal tubular cells can aid in understanding the pathophysiology of rare inherited renal tubular diseases through cellular experiments. MATERIALS AND METHODS Two Japanese patients with Pseudohypoparathyroidism (PHP), a 49-year-old woman and a 71-year-old man, were studied. iPSC-derived tubular cells were established from their peripheral blood mononuclear cells (PBMCs). We examined changes in intracellular and extracellular cyclic adenosine monophosphate (cAMP) levels in these cells in response to parathyroid hormone (PTH) stimulation. RESULTS Renal tubular cells, differentiated from iPSCs of a healthy control (648A1), showed a PTH-dependent increase in both intracellular and extracellular cAMP levels. However, the renal tubular cells derived from the PHP patients' iPSCs showed inconsistent changes in cAMP levels upon PTH exposure. CONCLUSION We successfully created disease-specific iPSCs from PHP patients' PBMCs, differentiated them into tubular cells, and replicated the distinctive response of the disease to PTH in vitro. This approach could enhance our understanding of the pathophysiology of inherited renal tubular diseases and contribute to developing effective treatments.
Collapse
Affiliation(s)
- Yoshihiro Nakamura
- Division of Nephrology, Hypertension and Endocrinology, Department of Internal Medicine, Nihon University School of Medicine, 30-1 Oyaguchi Kami-chou, Itabashi-ku, Tokyo, 173-8610, Japan
| | - Hiroki Kobayashi
- Division of Nephrology, Hypertension and Endocrinology, Department of Internal Medicine, Nihon University School of Medicine, 30-1 Oyaguchi Kami-chou, Itabashi-ku, Tokyo, 173-8610, Japan.
| | - Noboru Fukuda
- Division of Nephrology, Hypertension and Endocrinology, Department of Internal Medicine, Nihon University School of Medicine, 30-1 Oyaguchi Kami-chou, Itabashi-ku, Tokyo, 173-8610, Japan
| | - Sho Tanaka
- Division of Nephrology, Hypertension and Endocrinology, Department of Internal Medicine, Nihon University School of Medicine, 30-1 Oyaguchi Kami-chou, Itabashi-ku, Tokyo, 173-8610, Japan
| | - Yusuke Murata
- Division of Nephrology, Hypertension and Endocrinology, Department of Internal Medicine, Nihon University School of Medicine, 30-1 Oyaguchi Kami-chou, Itabashi-ku, Tokyo, 173-8610, Japan
| | - Yoshinari Hatanaka
- Division of Nephrology, Hypertension and Endocrinology, Department of Internal Medicine, Nihon University School of Medicine, 30-1 Oyaguchi Kami-chou, Itabashi-ku, Tokyo, 173-8610, Japan
| | - Akira Haketa
- Division of Nephrology, Hypertension and Endocrinology, Department of Internal Medicine, Nihon University School of Medicine, 30-1 Oyaguchi Kami-chou, Itabashi-ku, Tokyo, 173-8610, Japan
| | - Akiko Tsunemi
- Division of Nephrology, Hypertension and Endocrinology, Department of Internal Medicine, Nihon University School of Medicine, 30-1 Oyaguchi Kami-chou, Itabashi-ku, Tokyo, 173-8610, Japan
| | - Lan Chen
- Division of Nephrology, Hypertension and Endocrinology, Department of Internal Medicine, Nihon University School of Medicine, 30-1 Oyaguchi Kami-chou, Itabashi-ku, Tokyo, 173-8610, Japan
| | - Masanori Abe
- Division of Nephrology, Hypertension and Endocrinology, Department of Internal Medicine, Nihon University School of Medicine, 30-1 Oyaguchi Kami-chou, Itabashi-ku, Tokyo, 173-8610, Japan
| |
Collapse
|
39
|
Murase Y, Yokogawa R, Yabuta Y, Nagano M, Katou Y, Mizuyama M, Kitamura A, Puangsricharoen P, Yamashiro C, Hu B, Mizuta K, Tsujimura T, Yamamoto T, Ogata K, Ishihama Y, Saitou M. In vitro reconstitution of epigenetic reprogramming in the human germ line. Nature 2024; 631:170-178. [PMID: 38768632 PMCID: PMC11222161 DOI: 10.1038/s41586-024-07526-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 05/07/2024] [Indexed: 05/22/2024]
Abstract
Epigenetic reprogramming resets parental epigenetic memories and differentiates primordial germ cells (PGCs) into mitotic pro-spermatogonia or oogonia. This process ensures sexually dimorphic germ cell development for totipotency1. In vitro reconstitution of epigenetic reprogramming in humans remains a fundamental challenge. Here we establish a strategy for inducing epigenetic reprogramming and differentiation of pluripotent stem-cell-derived human PGC-like cells (hPGCLCs) into mitotic pro-spermatogonia or oogonia, coupled with their extensive amplification (about >1010-fold). Bone morphogenetic protein (BMP) signalling is a key driver of these processes. BMP-driven hPGCLC differentiation involves attenuation of the MAPK (ERK) pathway and both de novo and maintenance DNA methyltransferase activities, which probably promote replication-coupled, passive DNA demethylation. hPGCLCs deficient in TET1, an active DNA demethylase abundant in human germ cells2,3, differentiate into extraembryonic cells, including amnion, with de-repression of key genes that bear bivalent promoters. These cells fail to fully activate genes vital for spermatogenesis and oogenesis, and their promoters remain methylated. Our study provides a framework for epigenetic reprogramming in humans and an important advance in human biology. Through the generation of abundant mitotic pro-spermatogonia and oogonia-like cells, our results also represent a milestone for human in vitro gametogenesis research and its potential translation into reproductive medicine.
Collapse
Affiliation(s)
- Yusuke Murase
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ryuta Yokogawa
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yukihiro Yabuta
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masahiro Nagano
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yoshitaka Katou
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Manami Mizuyama
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ayaka Kitamura
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Pimpitcha Puangsricharoen
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Chika Yamashiro
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Bo Hu
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ken Mizuta
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Taro Tsujimura
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan
| | - Takuya Yamamoto
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Medical-Risk Avoidance based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP), Kyoto, Japan
| | - Kosuke Ogata
- Department of Molecular Systems BioAnalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Yasushi Ishihama
- Department of Molecular Systems BioAnalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Mitinori Saitou
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan.
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan.
| |
Collapse
|
40
|
Kagermeier T, Hauser S, Sarieva K, Laugwitz L, Groeschel S, Janzarik WG, Yentür Z, Becker K, Schöls L, Krägeloh-Mann I, Mayer S. Human organoid model of pontocerebellar hypoplasia 2a recapitulates brain region-specific size differences. Dis Model Mech 2024; 17:dmm050740. [PMID: 39034883 PMCID: PMC11552497 DOI: 10.1242/dmm.050740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 05/13/2024] [Indexed: 07/23/2024] Open
Abstract
Pontocerebellar hypoplasia type 2a (PCH2a) is an ultra-rare, autosomal recessive pediatric disorder with limited treatment options. Its anatomical hallmark is hypoplasia of the cerebellum and pons accompanied by progressive microcephaly. A homozygous founder variant in TSEN54, which encodes a tRNA splicing endonuclease (TSEN) complex subunit, is causal. The pathological mechanism of PCH2a remains unknown due to the lack of a model system. Therefore, we developed human models of PCH2a using regionalized neural organoids. We generated induced pluripotent stem cell (iPSC) lines from three males with genetically confirmed PCH2a and subsequently differentiated cerebellar and neocortical organoids. Mirroring clinical neuroimaging findings, PCH2a cerebellar organoids were reduced in size compared to controls starting early in differentiation. Neocortical PCH2a organoids demonstrated milder growth deficits. Although PCH2a cerebellar organoids did not upregulate apoptosis, their stem cell zones showed altered proliferation kinetics, with increased proliferation at day 30 and reduced proliferation at day 50 compared to controls. In summary, we generated a human model of PCH2a, providing the foundation for deciphering brain region-specific disease mechanisms. Our first analyses suggest a neurodevelopmental aspect of PCH2a.
Collapse
Affiliation(s)
- Theresa Kagermeier
- Hertie Institute for Clinical Brain Research, University of Tübingen, 72076Tübingen, Germany
- Graduate Training Centre of Neuroscience, University of Tübingen, 72076Tübingen, Germany
| | - Stefan Hauser
- Hertie Institute for Clinical Brain Research, University of Tübingen, 72076Tübingen, Germany
- German Center for Neurodegenerative Diseases, 72076Tübingen, Germany
| | - Kseniia Sarieva
- Hertie Institute for Clinical Brain Research, University of Tübingen, 72076Tübingen, Germany
- Graduate Training Centre of Neuroscience, University of Tübingen, 72076Tübingen, Germany
- International Max Planck Research School, Graduate Training Centre of Neuroscience, University of Tübingen, 72076Tübingen, Germany
| | - Lucia Laugwitz
- Department of Neuropediatrics, Developmental Neurology and Social Pediatrics, University of Tübingen, 72076 Tübingen, Germany
| | - Samuel Groeschel
- Department of Neuropediatrics, Developmental Neurology and Social Pediatrics, University of Tübingen, 72076 Tübingen, Germany
| | - Wibke G. Janzarik
- Department of Neuropediatrics and Muscle Disorders, Center for Pediatrics and Adolescent Medicine, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Zeynep Yentür
- Hertie Institute for Clinical Brain Research, University of Tübingen, 72076Tübingen, Germany
- Graduate Training Centre of Neuroscience, University of Tübingen, 72076Tübingen, Germany
- International Max Planck Research School, Graduate Training Centre of Neuroscience, University of Tübingen, 72076Tübingen, Germany
- Heidelberger Akademie der Wissenschaften, 69117 Heidelberg, Germany
| | - Katharina Becker
- Hertie Institute for Clinical Brain Research, University of Tübingen, 72076Tübingen, Germany
| | - Ludger Schöls
- Hertie Institute for Clinical Brain Research, University of Tübingen, 72076Tübingen, Germany
- German Center for Neurodegenerative Diseases, 72076Tübingen, Germany
| | - Ingeborg Krägeloh-Mann
- Department of Neuropediatrics, Developmental Neurology and Social Pediatrics, University of Tübingen, 72076 Tübingen, Germany
| | - Simone Mayer
- Hertie Institute for Clinical Brain Research, University of Tübingen, 72076Tübingen, Germany
- Heidelberger Akademie der Wissenschaften, 69117 Heidelberg, Germany
| |
Collapse
|
41
|
Hayward B, Kumari D, Santra S, van Karnebeek CDM, van Kuilenburg ABP, Usdin K. All three MutL complexes are required for repeat expansion in a human stem cell model of CAG-repeat expansion mediated glutaminase deficiency. Sci Rep 2024; 14:13772. [PMID: 38877099 PMCID: PMC11178883 DOI: 10.1038/s41598-024-64480-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 06/10/2024] [Indexed: 06/16/2024] Open
Abstract
The Repeat Expansion Diseases (REDs) arise from the expansion of a disease-specific short tandem repeat (STR). Different REDs differ with respect to the repeat involved, the cells that are most expansion prone and the extent of expansion. Furthermore, whether these diseases share a common expansion mechanism is unclear. To date, expansion has only been studied in a limited number of REDs. Here we report the first studies of the expansion mechanism in induced pluripotent stem cells derived from a patient with a form of the glutaminase deficiency disorder known as Global Developmental Delay, Progressive Ataxia, And Elevated Glutamine (GDPAG; OMIM# 618412) caused by the expansion of a CAG-STR in the 5' UTR of the glutaminase (GLS) gene. We show that alleles with as few as ~ 120 repeats show detectable expansions in culture despite relatively low levels of R-loops formed at this locus. Additionally, using a CRISPR-Cas9 knockout approach we show that PMS2 and MLH3, the constituents of MutLα and MutLγ, the 2 mammalian MutL complexes known to be involved in mismatch repair (MMR), are essential for expansion. Furthermore, PMS1, a component of a less well understood MutL complex, MutLβ, is also important, if not essential, for repeat expansion in these cells. Our results provide insights into the factors important for expansion and lend weight to the idea that, despite some differences, the same mechanism is responsible for expansion in many, if not all, REDs.
Collapse
Affiliation(s)
- Bruce Hayward
- Section On Gene Structure and Disease, Laboratory of Cell and Molecular Biology, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Daman Kumari
- Section On Gene Structure and Disease, Laboratory of Cell and Molecular Biology, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Saikat Santra
- Birmingham Women's and Children's NHS Foundation Trust, Birmingham, B15 2TG, UK
| | - Clara D M van Karnebeek
- Emma Center for Personalized Medicine, Departments of Pediatrics and Human Genetics, Amsterdam Gastro-Enterology Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- United for Metabolic Diseases, Amsterdam, The Netherlands
| | - André B P van Kuilenburg
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
| | - Karen Usdin
- Section On Gene Structure and Disease, Laboratory of Cell and Molecular Biology, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
42
|
Shah Z, Tian L, Li Z, Jin L, Zhang J, Li Z, Barr T, Tang H, Feng M, Caligiuri MA, Yu J. Human anti-PSCA CAR macrophages possess potent antitumor activity against pancreatic cancer. Cell Stem Cell 2024; 31:803-817.e6. [PMID: 38663406 PMCID: PMC11162318 DOI: 10.1016/j.stem.2024.03.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 01/11/2024] [Accepted: 03/28/2024] [Indexed: 05/15/2024]
Abstract
Due to the limitations of autologous chimeric antigen receptor (CAR)-T cells, alternative sources of cellular immunotherapy, including CAR macrophages, are emerging for solid tumors. Human induced pluripotent stem cells (iPSCs) offer an unlimited source for immune cell generation. Here, we develop human iPSC-derived CAR macrophages targeting prostate stem cell antigen (PSCA) (CAR-iMacs), which express membrane-bound interleukin (IL)-15 and truncated epidermal growth factor receptor (EGFR) for immune cell activation and a suicide switch, respectively. These allogeneic CAR-iMacs exhibit strong antitumor activity against human pancreatic solid tumors in vitro and in vivo, leading to reduced tumor burden and improved survival in a pancreatic cancer mouse model. CAR-iMacs appear safe and do not exhibit signs of cytokine release syndrome or other in vivo toxicities. We optimized the cryopreservation of CAR-iMac progenitors that remain functional upon thawing, providing an off-the-shelf, allogeneic cell product that can be developed into CAR-iMacs. Overall, our preclinical data strongly support the potential clinical translation of this human iPSC-derived platform for solid tumors, including pancreatic cancer.
Collapse
Affiliation(s)
- Zahir Shah
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA; Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA 91010, USA
| | - Lei Tian
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA; Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA 91010, USA
| | - Zhixin Li
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA; Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA 91010, USA
| | - Lewei Jin
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA; Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA 91010, USA
| | - Jianying Zhang
- Department of Computational and Quantitative Medicine, City of Hope National Medical Center, Los Angeles, CA 91010, USA
| | - Zhenlong Li
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA; Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA 91010, USA
| | - Tasha Barr
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA; Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA 91010, USA
| | - Hejun Tang
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA; Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA 91010, USA
| | - Mingye Feng
- Department of Immuno-Oncology, City of Hope, Los Angeles, CA 91010, USA
| | - Michael A Caligiuri
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA; Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA 91010, USA; City of Hope Comprehensive Cancer Center, Los Angeles, CA 91010, USA.
| | - Jianhua Yu
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA; Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA 91010, USA; Department of Immuno-Oncology, City of Hope, Los Angeles, CA 91010, USA; City of Hope Comprehensive Cancer Center, Los Angeles, CA 91010, USA.
| |
Collapse
|
43
|
Moratilla A, Martín D, Cadenas-Martín M, Stokking M, Quesada MA, Arnalich F, De Miguel MP. Hypoxia Increases the Efficiencies of Cellular Reprogramming and Oncogenic Transformation in Human Blood Cell Subpopulations In Vitro and In Vivo. Cells 2024; 13:971. [PMID: 38891103 PMCID: PMC11172288 DOI: 10.3390/cells13110971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 05/31/2024] [Accepted: 05/31/2024] [Indexed: 06/21/2024] Open
Abstract
Patients with chronic hypoxia show a higher tumor incidence; however, no primary common cause has been recognized. Given the similarities between cellular reprogramming and oncogenic transformation, we directly compared these processes in human cells subjected to hypoxia. Mouse embryonic fibroblasts were employed as controls to compare transfection and reprogramming efficiency; human adipose-derived mesenchymal stem cells were employed as controls in human cells. Easily obtainable human peripheral blood mononuclear cells (PBMCs) were chosen to establish a standard protocol to compare cell reprogramming (into induced pluripotent stem cells (iPSCs)) and oncogenic focus formation efficiency. Cell reprogramming was achieved for all three cell types, generating actual pluripotent cells capable for differentiating into the three germ layers. The efficiencies of the cell reprogramming and oncogenic transformation were similar. Hypoxia slightly increased the reprogramming efficiency in all the cell types but with no statistical significance for PBMCs. Various PBMC types can respond to hypoxia differently; lymphocytes and monocytes were, therefore, reprogrammed separately, finding a significant difference between normoxia and hypoxia in monocytes in vitro. These differences were then searched for in vivo. The iPSCs and oncogenic foci were generated from healthy volunteers and patients with chronic obstructive pulmonary disease (COPD). Although higher iPSC generation efficiency in the patients with COPD was found for lymphocytes, this increase was not statistically significant for oncogenic foci. Remarkably, a higher statistically significant efficiency in COPD monocytes was demonstrated for both processes, suggesting that physiological hypoxia exerts an effect on cell reprogramming and oncogenic transformation in vivo in at least some cell types.
Collapse
Affiliation(s)
- Adrián Moratilla
- Cell Engineering Laboratory, La Paz University Hospital Health Research Institute, IdiPAZ, 28046 Madrid, Spain; (A.M.); (D.M.); (M.C.-M.); (M.S.)
| | - Diana Martín
- Cell Engineering Laboratory, La Paz University Hospital Health Research Institute, IdiPAZ, 28046 Madrid, Spain; (A.M.); (D.M.); (M.C.-M.); (M.S.)
| | - Marta Cadenas-Martín
- Cell Engineering Laboratory, La Paz University Hospital Health Research Institute, IdiPAZ, 28046 Madrid, Spain; (A.M.); (D.M.); (M.C.-M.); (M.S.)
| | - Martha Stokking
- Cell Engineering Laboratory, La Paz University Hospital Health Research Institute, IdiPAZ, 28046 Madrid, Spain; (A.M.); (D.M.); (M.C.-M.); (M.S.)
| | - Maria Angustias Quesada
- Internal Medicine Service, La Paz University Hospital, IdiPAZ, 28046 Madrid, Spain; (M.A.Q.); (F.A.)
| | - Francisco Arnalich
- Internal Medicine Service, La Paz University Hospital, IdiPAZ, 28046 Madrid, Spain; (M.A.Q.); (F.A.)
| | - Maria P. De Miguel
- Cell Engineering Laboratory, La Paz University Hospital Health Research Institute, IdiPAZ, 28046 Madrid, Spain; (A.M.); (D.M.); (M.C.-M.); (M.S.)
| |
Collapse
|
44
|
Grigor’eva EV, Karapetyan LV, Malakhova AA, Medvedev SP, Minina JM, Hayrapetyan VH, Vardanyan VS, Zakian SM, Arakelyan A, Zakharyan R. Generation of iPSCs from a Patient with the M694V Mutation in the MEFV Gene Associated with Familial Mediterranean Fever and Their Differentiation into Macrophages. Int J Mol Sci 2024; 25:6102. [PMID: 38892289 PMCID: PMC11173119 DOI: 10.3390/ijms25116102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/03/2024] [Accepted: 05/06/2024] [Indexed: 06/21/2024] Open
Abstract
Familial Mediterranean fever (FMF) is a systemic autoinflammatory disorder caused by inherited mutations in the MEFV (Mediterranean FeVer) gene, located on chromosome 16 (16p13.3) and encoding the pyrin protein. Despite the existing data on MEFV mutations, the exact mechanism of their effect on the development of the pathological processes leading to the spontaneous and recurrent autoinflammatory attacks observed in FMF, remains unclear. Induced pluripotent stem cells (iPSCs) are considered an important tool to study the molecular genetic mechanisms of various diseases due to their ability to differentiate into any cell type, including macrophages, which contribute to the development of FMF. In this study, we developed iPSCs from an Armenian patient with FMF carrying the M694V, p.(Met694Val) (c.2080A>G, rs61752717) pathogenic mutation in exon 10 of the MEFV gene. As a result of direct differentiation, macrophages expressing CD14 and CD45 surface markers were obtained. We found that the morphology of macrophages derived from iPSCs of a patient with the MEFV mutation significantly differed from that of macrophages derived from iPSCs of a healthy donor carrying the wild-type MEFV gene.
Collapse
Affiliation(s)
- Elena V. Grigor’eva
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (E.V.G.); (A.A.M.); (S.P.M.); (J.M.M.); (S.M.Z.)
- Meshalkin National Medical Research Center, Ministry of Health of the Russian Federation, 630055 Novosibirsk, Russia
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Lana V. Karapetyan
- Department of Bioengineering, Bioinformatics, and Molecular Biology, Institute of Biomedicine and Pharmacy, Russian-Armenian (Slavonic) University, Yerevan 0051, Armenia; (L.V.K.); (V.H.H.); (A.A.)
| | - Anastasia A. Malakhova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (E.V.G.); (A.A.M.); (S.P.M.); (J.M.M.); (S.M.Z.)
- Meshalkin National Medical Research Center, Ministry of Health of the Russian Federation, 630055 Novosibirsk, Russia
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Sergey P. Medvedev
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (E.V.G.); (A.A.M.); (S.P.M.); (J.M.M.); (S.M.Z.)
- Meshalkin National Medical Research Center, Ministry of Health of the Russian Federation, 630055 Novosibirsk, Russia
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Julia M. Minina
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (E.V.G.); (A.A.M.); (S.P.M.); (J.M.M.); (S.M.Z.)
| | - Varduhi H. Hayrapetyan
- Department of Bioengineering, Bioinformatics, and Molecular Biology, Institute of Biomedicine and Pharmacy, Russian-Armenian (Slavonic) University, Yerevan 0051, Armenia; (L.V.K.); (V.H.H.); (A.A.)
- Institute of Molecular Biology NAS RA, Yerevan 0014, Armenia
| | - Valentina S. Vardanyan
- Department of Rheumatology, Yerevan State Medical University after Mkhitar Heratsi (YSMU), Yerevan 0025, Armenia;
- Department of Rheumatology, “Mikaelyan” Institute of Surgery, Yerevan 0052, Armenia
| | - Suren M. Zakian
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (E.V.G.); (A.A.M.); (S.P.M.); (J.M.M.); (S.M.Z.)
- Meshalkin National Medical Research Center, Ministry of Health of the Russian Federation, 630055 Novosibirsk, Russia
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Arsen Arakelyan
- Department of Bioengineering, Bioinformatics, and Molecular Biology, Institute of Biomedicine and Pharmacy, Russian-Armenian (Slavonic) University, Yerevan 0051, Armenia; (L.V.K.); (V.H.H.); (A.A.)
- Institute of Molecular Biology NAS RA, Yerevan 0014, Armenia
| | - Roksana Zakharyan
- Department of Bioengineering, Bioinformatics, and Molecular Biology, Institute of Biomedicine and Pharmacy, Russian-Armenian (Slavonic) University, Yerevan 0051, Armenia; (L.V.K.); (V.H.H.); (A.A.)
- Institute of Molecular Biology NAS RA, Yerevan 0014, Armenia
| |
Collapse
|
45
|
Abujarour R, Dinella J, Pribadi M, Fong LK, Denholtz M, Gutierrez A, Haynes M, Mahmood E, Lee TT, Ding S, Valamehr B. A chemical approach facilitates CRISPRa-only human iPSC generation and minimizes the number of targeted loci required. Future Sci OA 2024; 10:FSO964. [PMID: 38817352 PMCID: PMC11137772 DOI: 10.2144/fsoa-2023-0257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 01/19/2024] [Indexed: 06/01/2024] Open
Abstract
Aim: We explored the generation of human induced pluripotent stem cells (iPSCs) solely through the transcriptional activation of endogenous genes by CRISPR activation (CRISPRa). Methods: Minimal number of human-specific guide RNAs targeting a limited set of loci were used with a unique cocktail of small molecules (CRISPRa-SM). Results: iPSC clones were efficiently generated by CRISPRa-SM, expressed general and naive iPSC markers and clustered with high-quality iPSCs generated using conventional reprogramming methods. iPSCs showed genomic stability and robust pluripotent potential as assessed by in vitro and in vivo. Conclusion: CRISPRa-SM-generated human iPSCs by direct and multiplexed loci activation facilitating a unique and potentially safer cellular reprogramming process to aid potential applications in cellular therapy and regenerative medicine.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Tom T Lee
- Fate Therapeutics, San Diego, CA 92121, USA
| | - Sheng Ding
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | | |
Collapse
|
46
|
Hayward B, Kumari D, Santra S, van Karnebeek CD, van Kuilenburg AB, Usdin K. All three MutL complexes are required for repeat expansion in a human stem cell model of CAG-repeat expansion mediated glutaminase deficiency. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.26.573357. [PMID: 38260514 PMCID: PMC10802475 DOI: 10.1101/2023.12.26.573357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
The Repeat Expansion Diseases (REDs) arise from the expansion of a disease-specific short tandem repeat (STR). Different REDs differ with respect to the repeat involved, the cells that are most expansion prone and the extent of expansion. Furthermore, whether these diseases share a common expansion mechanism is unclear. To date, expansion has only been studied in a limited number of REDs. Here we report the first studies of the expansion mechanism in induced pluripotent stem cells derived from a patient with a form of the glutaminase deficiency disorder known as Global Developmental Delay, Progressive Ataxia, And Elevated Glutamine (GDPAG; OMIM# 618412) caused by the expansion of a CAG-STR in the 5' UTR of the glutaminase (GLS) gene. We show that alleles with as few as ~120 repeats show detectable expansions in culture despite relatively low levels of R-loops formed at this locus. Additionally, using a CRISPR-Cas9 knockout approach we show that PMS2 and MLH3, the constituents of MutLα and MutLγ, the 2 mammalian MutL complexes known to be involved in mismatch repair (MMR), are essential for expansion. Furthermore, PMS1, a component of a less well understood MutL complex, MutLβ, is also important, if not essential, for repeat expansion in these cells. Our results provide insights into the factors important for expansion and lend weight to the idea that, despite some differences, the same mechanism is responsible for expansion in many, if not all, REDs.
Collapse
Affiliation(s)
- Bruce Hayward
- Section on Gene Structure and Disease, Laboratory of Cell and Molecular Biology, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Daman Kumari
- Section on Gene Structure and Disease, Laboratory of Cell and Molecular Biology, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Saikat Santra
- Birmingham Women's and Children's NHS Foundation Trust, Birmingham B15 2TG, United Kingdom
| | - Clara D.M. van Karnebeek
- Amsterdam UMC location University of Amsterdam, Departments of Pediatrics and Human Genetics, Emma Center for Personalized Medicine, Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
- United for Metabolic Diseases, The Netherlands
| | - André B.P. van Kuilenburg
- Amsterdam UMC location University of Amsterdam, Department of Clinical Chemistry, Laboratory Genetic Metabolic Diseases, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
| | - Karen Usdin
- Section on Gene Structure and Disease, Laboratory of Cell and Molecular Biology, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
47
|
Wu L, Lu J, Lan T, Zhang D, Xu H, Kang Z, Peng F, Wang J. Stem cell therapies: a new era in the treatment of multiple sclerosis. Front Neurol 2024; 15:1389697. [PMID: 38784908 PMCID: PMC11111935 DOI: 10.3389/fneur.2024.1389697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 04/22/2024] [Indexed: 05/25/2024] Open
Abstract
Multiple Sclerosis (MS) is an immune-mediated condition that persistently harms the central nervous system. While existing treatments can slow its course, a cure remains elusive. Stem cell therapy has gained attention as a promising approach, offering new perspectives with its regenerative and immunomodulatory properties. This article reviews the application of stem cells in MS, encompassing various stem cell types, therapeutic potential mechanisms, preclinical explorations, clinical research advancements, safety profiles of clinical applications, as well as limitations and challenges, aiming to provide new insights into the treatment research for MS.
Collapse
Affiliation(s)
- Lei Wu
- Changchun University of Chinese Medicine, Changchun, China
| | - Jing Lu
- The Affiliated Hospital to Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Tianye Lan
- The Affiliated Hospital to Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Dongmei Zhang
- The Affiliated Hospital to Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Hanying Xu
- Changchun University of Chinese Medicine, Changchun, China
| | - Zezheng Kang
- Changchun University of Chinese Medicine, Changchun, China
| | - Fang Peng
- Hunan Provincial People's Hospital, Changsha, China
| | - Jian Wang
- The Affiliated Hospital to Changchun University of Traditional Chinese Medicine, Changchun, China
| |
Collapse
|
48
|
Hartley A, Burger L, Wincek CL, Dons L, Li T, Grewenig A, Taşgın T, Urban M, Roig-Merino A, Ghazvini M, Harbottle RP. A Simple Nonviral Method to Generate Human Induced Pluripotent Stem Cells Using SMAR DNA Vectors. Genes (Basel) 2024; 15:575. [PMID: 38790204 PMCID: PMC11121542 DOI: 10.3390/genes15050575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/21/2024] [Accepted: 04/23/2024] [Indexed: 05/26/2024] Open
Abstract
Induced pluripotent stem cells (iPSCs) are a powerful tool for biomedical research, but their production presents challenges and safety concerns. Yamanaka and Takahashi revolutionised the field by demonstrating that somatic cells could be reprogrammed into pluripotent cells by overexpressing four key factors for a sufficient time. iPSCs are typically generated using viruses or virus-based methods, which have drawbacks such as vector persistence, risk of insertional mutagenesis, and oncogenesis. The application of less harmful nonviral vectors is limited as conventional plasmids cannot deliver the levels or duration of the factors necessary from a single transfection. Hence, plasmids that are most often used for reprogramming employ the potentially oncogenic Epstein-Barr nuclear antigen 1 (EBNA-1) system to ensure adequate levels and persistence of expression. In this study, we explored the use of nonviral SMAR DNA vectors to reprogram human fibroblasts into iPSCs. We show for the first time that iPSCs can be generated using nonviral plasmids without the use of EBNA-1 and that these DNA vectors can provide sufficient expression to induce pluripotency. We describe an optimised reprogramming protocol using these vectors that can produce high-quality iPSCs with comparable pluripotency and cellular function to those generated with viruses or EBNA-1 vectors.
Collapse
Affiliation(s)
- Anna Hartley
- DNA Vector Laboratory, German Cancer Research Center, 69120 Heidelberg, Germany; (A.H.); (A.G.); (A.R.-M.)
- Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Luisa Burger
- DNA Vector Laboratory, German Cancer Research Center, 69120 Heidelberg, Germany; (A.H.); (A.G.); (A.R.-M.)
- Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Cornelia L. Wincek
- DNA Vector Laboratory, German Cancer Research Center, 69120 Heidelberg, Germany; (A.H.); (A.G.); (A.R.-M.)
| | - Lieke Dons
- Erasmus MC iPS Core Facility, Erasmus Medical Centre, 3015 GD Rotterdam, The Netherlands (M.G.)
| | - Tracy Li
- Erasmus MC iPS Core Facility, Erasmus Medical Centre, 3015 GD Rotterdam, The Netherlands (M.G.)
| | - Annabel Grewenig
- DNA Vector Laboratory, German Cancer Research Center, 69120 Heidelberg, Germany; (A.H.); (A.G.); (A.R.-M.)
| | - Toros Taşgın
- DNA Vector Laboratory, German Cancer Research Center, 69120 Heidelberg, Germany; (A.H.); (A.G.); (A.R.-M.)
- Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Manuela Urban
- DNA Vector Laboratory, German Cancer Research Center, 69120 Heidelberg, Germany; (A.H.); (A.G.); (A.R.-M.)
- Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Alicia Roig-Merino
- DNA Vector Laboratory, German Cancer Research Center, 69120 Heidelberg, Germany; (A.H.); (A.G.); (A.R.-M.)
- Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Mehrnaz Ghazvini
- Erasmus MC iPS Core Facility, Erasmus Medical Centre, 3015 GD Rotterdam, The Netherlands (M.G.)
| | - Richard P. Harbottle
- DNA Vector Laboratory, German Cancer Research Center, 69120 Heidelberg, Germany; (A.H.); (A.G.); (A.R.-M.)
| |
Collapse
|
49
|
Chen Y, Li M, Wu Y. The occurrence and development of induced pluripotent stem cells. Front Genet 2024; 15:1389558. [PMID: 38699229 PMCID: PMC11063328 DOI: 10.3389/fgene.2024.1389558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 04/08/2024] [Indexed: 05/05/2024] Open
Abstract
The ectopic expression of four transcription factors, Oct3/4, Sox2, Klf4, and c-Myc (OSKM), known as "Yamanaka factors," can reprogram or stimulate the production of induced pluripotent stem cells (iPSCs). Although OSKM is still the gold standard, there are multiple ways to reprogram cells into iPSCs. In recent years, significant progress has been made in improving the efficiency of this technology. Ten years after the first report was published, human pluripotent stem cells have gradually been applied in clinical settings, including disease modeling, cell therapy, new drug development, and cell derivation. Here, we provide a review of the discovery of iPSCs and their applications in disease and development.
Collapse
Affiliation(s)
| | - Meng Li
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yanqing Wu
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
50
|
Kanemura Y, Yamamoto A, Katsuma A, Fukusumi H, Shofuda T, Kanematsu D, Handa Y, Sumida M, Yoshioka E, Mine Y, Yamaguchi R, Okada M, Igarashi M, Sekino Y, Shirao T, Nakamura M, Okano H. Human-Induced Pluripotent Stem Cell-Derived Neural Progenitor Cells Showed Neuronal Differentiation, Neurite Extension, and Formation of Synaptic Structures in Rodent Ischemic Stroke Brains. Cells 2024; 13:671. [PMID: 38667286 PMCID: PMC11048851 DOI: 10.3390/cells13080671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/01/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Ischemic stroke is a major cerebrovascular disease with high morbidity and mortality rates; however, effective treatments for ischemic stroke-related neurological dysfunction have yet to be developed. In this study, we generated neural progenitor cells from human leukocyte antigen major loci gene-homozygous-induced pluripotent stem cells (hiPSC-NPCs) and evaluated their therapeutic effects against ischemic stroke. hiPSC-NPCs were intracerebrally transplanted into rat ischemic brains produced by transient middle cerebral artery occlusion at either the subacute or acute stage, and their in vivo survival, differentiation, and efficacy for functional improvement in neurological dysfunction were evaluated. hiPSC-NPCs were histologically identified in host brain tissues and showed neuronal differentiation into vGLUT-positive glutamatergic neurons, extended neurites into both the ipsilateral infarct and contralateral healthy hemispheres, and synaptic structures formed 12 weeks after both acute and subacute stage transplantation. They also improved neurological function when transplanted at the subacute stage with γ-secretase inhibitor pretreatment. However, their effects were modest and not significant and showed a possible risk of cells remaining in their undifferentiated and immature status in acute-stage transplantation. These results suggest that hiPSC-NPCs show cell replacement effects in ischemic stroke-damaged neural tissues, but their efficacy is insufficient for neurological functional improvement after acute or subacute transplantation. Further optimization of cell preparation methods and the timing of transplantation is required to balance the efficacy and safety of hiPSC-NPC transplantation.
Collapse
Affiliation(s)
- Yonehiro Kanemura
- Department of Biomedical Research and Innovation, Institute for Clinical Research, NHO Osaka National Hospital, Osaka 540-0006, Japan; (A.Y.); (A.K.); (H.F.); (M.S.)
- Department of Neurosurgery, NHO Osaka National Hospital, Osaka 540-0006, Japan
| | - Atsuyo Yamamoto
- Department of Biomedical Research and Innovation, Institute for Clinical Research, NHO Osaka National Hospital, Osaka 540-0006, Japan; (A.Y.); (A.K.); (H.F.); (M.S.)
| | - Asako Katsuma
- Department of Biomedical Research and Innovation, Institute for Clinical Research, NHO Osaka National Hospital, Osaka 540-0006, Japan; (A.Y.); (A.K.); (H.F.); (M.S.)
| | - Hayato Fukusumi
- Department of Biomedical Research and Innovation, Institute for Clinical Research, NHO Osaka National Hospital, Osaka 540-0006, Japan; (A.Y.); (A.K.); (H.F.); (M.S.)
| | - Tomoko Shofuda
- Department of Biomedical Research and Innovation, Institute for Clinical Research, NHO Osaka National Hospital, Osaka 540-0006, Japan; (A.Y.); (A.K.); (H.F.); (M.S.)
| | - Daisuke Kanematsu
- Department of Biomedical Research and Innovation, Institute for Clinical Research, NHO Osaka National Hospital, Osaka 540-0006, Japan; (A.Y.); (A.K.); (H.F.); (M.S.)
| | - Yukako Handa
- Department of Biomedical Research and Innovation, Institute for Clinical Research, NHO Osaka National Hospital, Osaka 540-0006, Japan; (A.Y.); (A.K.); (H.F.); (M.S.)
| | - Miho Sumida
- Department of Biomedical Research and Innovation, Institute for Clinical Research, NHO Osaka National Hospital, Osaka 540-0006, Japan; (A.Y.); (A.K.); (H.F.); (M.S.)
| | - Ema Yoshioka
- Department of Biomedical Research and Innovation, Institute for Clinical Research, NHO Osaka National Hospital, Osaka 540-0006, Japan; (A.Y.); (A.K.); (H.F.); (M.S.)
| | - Yutaka Mine
- Department of Neurosurgery, NHO Tokyo Medical Center, Tokyo 152-8902, Japan;
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan; (R.Y.); (H.O.)
| | - Ryo Yamaguchi
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan; (R.Y.); (H.O.)
- Regenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Kobe 650-0047, Japan
| | - Masayasu Okada
- Department of Brain Tumor Biology, Brain Research Institute, Niigata University, Niigata 951-8585, Japan;
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Michihiro Igarashi
- Department of Neurochemistry and Molecular Cell Biology, School of Medicine, Graduate School of Medical, Dental Sciences Niigata University, Niigata 951-8510, Japan;
| | - Yuko Sekino
- Department of Veterinary Pathophysiology and Animal Health, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan;
| | | | - Masaya Nakamura
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo 160-8582, Japan;
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan; (R.Y.); (H.O.)
- Keio Regenerative Medicine Research Center, Keio University, Kawasaki 210-0821, Japan
| |
Collapse
|