1
|
Yahata N, Goto YI, Hata R. Optimization of mtDNA-targeted platinum TALENs for bi-directionally modifying heteroplasmy levels in patient-derived m.3243A>G-iPSCs. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102521. [PMID: 40242044 PMCID: PMC12002989 DOI: 10.1016/j.omtn.2025.102521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 03/16/2025] [Indexed: 04/18/2025]
Abstract
Patient-derived induced pluripotent stem cells (iPSCs) are a useful pathological model for debilitating diseases caused by mitochondrial DNA (mtDNA) mutations. We established iPSCs derived from mitochondrial disease patients, heteroplasmic for the m.3243A>G mutation. The proportion of a selected mtDNA can be reduced by delivering a programmable nuclease into the mitochondria, and we developed various mtDNA-targeted Platinum TALENs (mpTALENs) to modify m.3243A>G-iPSC heteroplasmy levels in either wild-type or mutant direction. For TALEN optimization, the use of non-conventional repeat-variable di-residues (ncRVD)-LK/WK or NM-enhanced cleavage activity and specificity, and the replacement of conventional with obligate heterodimeric FokI nuclease domains increased target specificity and protected mtDNA from copy number depletion. In vitro, depending on whether wild-type or mutant mtDNA was targeted, we could obtain m.3243A>G-iPSCs with a higher or lower mutation load, while the cells retained their ability to differentiate into three germ layers. These results demonstrate that our mpTALEN optimization created a useful tool for altering heteroplasmy levels in m.3243A>G-iPSCs, improving the potential for studying mutation pathology. The enhanced efficiency also holds promise for using m.3243G(MUT)-mpTALEN as a therapeutic strategy for treating patients suffering from m.3243A>G mitochondrial diseases.
Collapse
Affiliation(s)
- Naoki Yahata
- Department of Anatomy I, Fujita Health University School of Medicine, Toyoake, Aichi 470-1192, Japan
- Department of Developmental Biology, Fujita Health University School of Medicine, Toyoake, Aichi 470-1192, Japan
- Division of Developmental Neurobiology, International Center for Brain Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Yu-ichi Goto
- Medical Genome Center, National Center of Neurology and Psychiatry, Kodaira, Tokyo 187-8551, Japan
| | - Ryuji Hata
- Department of Anatomy I, Fujita Health University School of Medicine, Toyoake, Aichi 470-1192, Japan
- Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka Prefectural Hospital Organization, Hirakata, Osaka 573-0022, Japan
| |
Collapse
|
2
|
Innachai P, Pornratananont G, Satirapod C, Anurathapan U, Songdej D, Tangprasittipap A, Hongeng S. Generation of an integration-free induced pluripotent stem cell line, MURAi006-A, from a hemoglobin E/β-thalassemia patient harboring the β E/β 0 (Codon 17, A > T) compound heterozygous mutation. Stem Cell Res 2025; 85:103702. [PMID: 40179812 DOI: 10.1016/j.scr.2025.103702] [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: 01/08/2025] [Revised: 03/11/2025] [Accepted: 03/21/2025] [Indexed: 04/05/2025] Open
Abstract
The HBB gene encodes the β-globin protein, one of the two main components of adult hemoglobin A (HbA) responsible for oxygen transport. β-thalassemia is a genetic disorder caused by mutations affecting β-globin chain synthesis, leading to reduced or absent β-globin production, impaired erythropoiesis, and generally results in anemia. In this study, the human-induced pluripotent stem cell line (hiPSC) MURAi006-A was generated from male fetal skin fibroblasts carrying both a β⁰-thalassemia mutation at codon 17 (A > T) and a codon 26 (G > A) HbE mutation using non-integrative reprogramming episomes.
Collapse
Affiliation(s)
- Pawarit Innachai
- Offices of Health Science Research, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Gunn Pornratananont
- Program in Translational Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Chonthicha Satirapod
- Department of Obstetrics and Gynecology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Usanarat Anurathapan
- Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Duantida Songdej
- Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Amornrat Tangprasittipap
- Offices of Health Science Research, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand.
| | - Suradej Hongeng
- Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| |
Collapse
|
3
|
Kiyuna LA, Horcas‐Nieto JM, Odendaal C, Langelaar‐Makkinje M, Gerding A, Broekhuis MJC, Bonanini F, Singh M, Kurek D, Harms AC, Hankemeier T, Foijer F, Derks TGJ, Bakker BM. iPSC-Derived Liver Organoids as a Tool to Study Medium Chain Acyl-CoA Dehydrogenase Deficiency. J Inherit Metab Dis 2025; 48:e70028. [PMID: 40199742 PMCID: PMC11978564 DOI: 10.1002/jimd.70028] [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: 03/06/2024] [Revised: 02/28/2025] [Accepted: 03/19/2025] [Indexed: 04/10/2025]
Abstract
Medium chain acyl-CoA dehydrogenase deficiency (MCADD) is an inherited metabolic disease, characterized by biallelic variants in the ACADM gene. Interestingly, even with the same genotype, patients often present with very heterogeneous symptoms, ranging from fully asymptomatic to life-threatening hypoketotic hypoglycemia. The mechanisms underlying this heterogeneity remain unclear. Therefore, there is a need for in vitro models of MCADD that recapitulate the clinical phenotype as a tool to study the pathophysiology of the disease. Fibroblasts of control and symptomatic MCADD patients with the c.985A>G (p.K329E) were reprogrammed into induced pluripotent stem cells (iPSCs). iPSCs were then differentiated into hepatic expandable organoids (EHOs), further matured to Mat-EHOs, and functionally characterized. EHOs and Mat-EHOs performed typical hepatic metabolic functions, such as albumin and urea production. The organoids metabolized fatty acids, as confirmed by acyl-carnitine profiling and high-resolution respirometry. MCAD protein was fully ablated in MCADD organoids, in agreement with the instability of the mutated MCAD protein. MCADD organoids accumulated medium-chain acyl-carnitines, with a strongly elevated C8/C10 ratio, characteristic of the biochemical phenotype of the disease. Notably, C2 and C14 acyl-carnitines were found decreased in MCADD Mat-EHOs. Finally, MCADD organoids exhibited differential expression of genes involved in ω-oxidation, mitochondrial β-oxidation, TCA cycle, and peroxisomal coenzyme A metabolism, particularly upregulation of NUDT7. iPSC-derived organoids of MCADD patients recapitulated the major biochemical phenotype of the disease. Mat-EHOs expressed relevant pathways involved in putative compensatory mechanisms, notably CoA metabolism and the TCA cycle. The upregulation of NUDT7 expression may play a role in preventing excessive accumulation of dicarboxylic acids in MCADD. This patient-specific hepatic organoid system is a promising platform to study the phenotypic heterogeneity between MCADD patients.
Collapse
Affiliation(s)
- Ligia A. Kiyuna
- Laboratory of PediatricsUniversity Medical Center Groningen, University of GroningenGroningenthe Netherlands
| | - José M. Horcas‐Nieto
- Laboratory of PediatricsUniversity Medical Center Groningen, University of GroningenGroningenthe Netherlands
| | - Christoff Odendaal
- Laboratory of PediatricsUniversity Medical Center Groningen, University of GroningenGroningenthe Netherlands
| | - Miriam Langelaar‐Makkinje
- Laboratory of PediatricsUniversity Medical Center Groningen, University of GroningenGroningenthe Netherlands
| | - Albert Gerding
- Laboratory of PediatricsUniversity Medical Center Groningen, University of GroningenGroningenthe Netherlands
- Department of Laboratory MedicineUniversity Medical Center Groningen, University of GroningenGroningenthe Netherlands
| | - Mathilde J. C. Broekhuis
- European Research Institute for the Biology of AgeingUniversity Medical Center Groningen, University of GroningenGroningenthe Netherlands
| | | | - Madhulika Singh
- Metabolomics and Analytics CentreLeiden Academic Centre for Drug Research, Leiden UniversityLeidenthe Netherlands
| | | | - Amy C. Harms
- Metabolomics and Analytics CentreLeiden Academic Centre for Drug Research, Leiden UniversityLeidenthe Netherlands
| | - Thomas Hankemeier
- Metabolomics and Analytics CentreLeiden Academic Centre for Drug Research, Leiden UniversityLeidenthe Netherlands
| | - Floris Foijer
- European Research Institute for the Biology of AgeingUniversity Medical Center Groningen, University of GroningenGroningenthe Netherlands
| | - Terry G. J. Derks
- Section of Metabolic Diseases, Beatrix Children's HospitalUniversity Medical Centre Groningen, University of GroningenGroningenthe Netherlands
| | - Barbara M. Bakker
- Laboratory of PediatricsUniversity Medical Center Groningen, University of GroningenGroningenthe Netherlands
| |
Collapse
|
4
|
Zhou N, Zhang S, Wang C, Zheng B, Zhang A, Zhou W. Generation of human induced pluripotent stem cell lines derived from a patient carrying an intragenic deletion in the NFIA gene. Hum Cell 2025; 38:95. [PMID: 40266456 DOI: 10.1007/s13577-025-01222-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: 01/24/2025] [Accepted: 04/09/2025] [Indexed: 04/24/2025]
Abstract
Brain malformations with or without urinary tract defects (BRMUTD) are caused by heterozygous variants in the NFIA gene. BRMUTD is a neurodevelopmental disorder characterized by hypoplasia or absence of the corpus callosum, hydrocephalus or ventriculomegaly, and developmental delay, which may or may not be accompanied by urinary tract defects. Here, we report the successful generation of induced pluripotent stem cells (hiPSCs) from a 3-year-old male BRMUTD patient using Sendai virus-based non-integrating reprogramming technology. This patient-derived cell line harbors an intragenic deletion within the NFIA gene (NC_000001.10: g.61650967_61842967del [GRCh37]), which is associated with a significant reduction in NFIA expression. This cell line maintains a normal karyotype, expresses pluripotency markers, and can differentiate into three germ layers. The established hiPSCs line will provide an in vitro model for studying pathological mechanisms and potential therapies of NFIA-related neurodevelopmental disorder.
Collapse
Affiliation(s)
- Ning Zhou
- School of Medicine, Southeast University, Nanjing, 210009, China
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China
| | - Shengnan Zhang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China
| | - Chunli Wang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China
| | - Bixia Zheng
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China
| | - Aihua Zhang
- School of Medicine, Southeast University, Nanjing, 210009, China.
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China.
| | - Wei Zhou
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China.
| |
Collapse
|
5
|
Filippi K, Riße I, Judge LM, Conklin BR, Fleischmann BK, Hesse M. Generation and characterization of a human induced pluripotent stem cell (iPSC) line from a patient with BAG3 P209L myofibrillar myopathy-6. Stem Cell Res 2025; 86:103718. [PMID: 40318522 DOI: 10.1016/j.scr.2025.103718] [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: 10/22/2024] [Revised: 04/11/2025] [Accepted: 04/14/2025] [Indexed: 05/07/2025] Open
Abstract
Bag3 is important for protein homeostasis in mechanically stressed muscle proteins as member of the chaperone-assisted selective autophagy (CASA) complex. Patients with BAG3P209L myofibrillar myopathy-6 (MFM6) carry a point mutation (p.P209L; c.626C>T) in the BAG3 gene and display clinical features such as restrictive cardiomyopathy, skeletal muscle dystrophy and polyneuropathy. To obtain a representative MFM6-model, biopsies from a female BAG3P209L-patient were used to generate a human induced pluripotent stem cell (iPSC) line. For quality control, germ layer differentiation and pluripotency analyses were conducted. This iPSC allows us to characterize the pathophysiology of MFM6 and develop innovative experimental therapeutic strategies.
Collapse
Affiliation(s)
- Kerstin Filippi
- Institute of Physiology I, Medical Faculty, University of Bonn, Germany
| | - Isabelle Riße
- Institute of Physiology I, Medical Faculty, University of Bonn, Germany
| | - Luke M Judge
- Gladstone Institutes, San Francisco, USA; University of California, San Francisco, USA
| | - Bruce R Conklin
- Gladstone Institutes, San Francisco, USA; University of California, San Francisco, USA
| | | | - Michael Hesse
- Institute of Physiology I, Medical Faculty, University of Bonn, Germany.
| |
Collapse
|
6
|
Sawamoto N, Doi D, Nakanishi E, Sawamura M, Kikuchi T, Yamakado H, Taruno Y, Shima A, Fushimi Y, Okada T, Kikuchi T, Morizane A, Hiramatsu S, Anazawa T, Shindo T, Ueno K, Morita S, Arakawa Y, Nakamoto Y, Miyamoto S, Takahashi R, Takahashi J. Phase I/II trial of iPS-cell-derived dopaminergic cells for Parkinson's disease. Nature 2025:10.1038/s41586-025-08700-0. [PMID: 40240591 DOI: 10.1038/s41586-025-08700-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 01/24/2025] [Indexed: 04/18/2025]
Abstract
Parkinson's disease is caused by the loss of dopamine neurons, causing motor symptoms. Initial cell therapies using fetal tissues showed promise but had complications and ethical concerns1-5. Pluripotent stem (PS) cells emerged as a promising alternative for developing safe and effective treatments6. In this phase I/II trial at Kyoto University Hospital, seven patients (ages 50-69) received bilateral transplantation of dopaminergic progenitors derived from induced PS (iPS) cells. Primary outcomes focused on safety and adverse events, while secondary outcomes assessed motor symptom changes and dopamine production for 24 months. There were no serious adverse events, with 73 mild to moderate events. Patients' anti-parkinsonian medication doses were maintained unless therapeutic adjustments were required, resulting in increased dyskinesia. Magnetic resonance imaging showed no graft overgrowth. Among six patients subjected to efficacy evaluation, four showed improvements in the Movement Disorder Society Unified Parkinson's Disease Rating Scale part III OFF score, and five showed improvements in the ON scores. The average changes of all six patients were 9.5 (20.4%) and 4.3 points (35.7%) for the OFF and ON scores, respectively. Hoehn-Yahr stages improved in four patients. Fluorine-18-L-dihydroxyphenylalanine (18F-DOPA) influx rate constant (Ki) values in the putamen increased by 44.7%, with higher increases in the high-dose group. Other measures showed minimal changes. This trial (jRCT2090220384) demonstrated that allogeneic iPS-cell-derived dopaminergic progenitors survived, produced dopamine and did not form tumours, therefore suggesting safety and potential clinical benefits for Parkinson's disease.
Collapse
Affiliation(s)
- Nobukatsu Sawamoto
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Daisuke Doi
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Etsuro Nakanishi
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masanori Sawamura
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takayuki Kikuchi
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hodaka Yamakado
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yosuke Taruno
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Atsushi Shima
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yasutaka Fushimi
- Department of Diagnostic Imaging and Nuclear Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tomohisa Okada
- Department of Diagnostic Imaging and Nuclear Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tetsuhiro Kikuchi
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Asuka Morizane
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Satoe Hiramatsu
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Takayuki Anazawa
- Department of Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takero Shindo
- Department of Hematology/Oncology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kentaro Ueno
- Department of Biomedical Statistics and Bioinformatics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Satoshi Morita
- Department of Biomedical Statistics and Bioinformatics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yoshiki Arakawa
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yuji Nakamoto
- Department of Diagnostic Imaging and Nuclear Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Susumu Miyamoto
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ryosuke Takahashi
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Jun Takahashi
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan.
| |
Collapse
|
7
|
Peng T, Ma X, Hua W, Wang C, Chu Y, Sun M, Fermi V, Hamelmann S, Lindner K, Shao C, Zaman J, Tian W, Zhuo Y, Harim Y, Stöffler N, Hammann L, Xiao Q, Jin X, Warta R, Lotsch C, Zhuang X, Feng Y, Fu M, Zhang X, Zhang J, Xu H, Qiu F, Xie L, Zhang Y, Zhu W, Du Z, Salgueiro L, Schneider M, Eichhorn F, Lefevre A, Pusch S, Grinevich V, Ratliff M, Loges S, Bunse L, Sahm F, Xiang Y, Unterberg A, von Deimling A, Platten M, Herold-Mende C, Wu Y, Liu HK, Mao Y. Individualized patient tumor organoids faithfully preserve human brain tumor ecosystems and predict patient response to therapy. Cell Stem Cell 2025; 32:652-669.e11. [PMID: 39938519 DOI: 10.1016/j.stem.2025.01.002] [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: 12/22/2023] [Revised: 09/27/2024] [Accepted: 01/03/2025] [Indexed: 02/14/2025]
Abstract
Tumor organoids are important tools for cancer research, but current models have drawbacks that limit their applications for predicting response to therapy. Here, we developed a fast, efficient, and complex culture system (IPTO, individualized patient tumor organoid) that accurately recapitulates the cellular and molecular pathology of human brain tumors. Patient-derived tumor explants were cultured in induced pluripotent stem cell (iPSC)-derived cerebral organoids, thus enabling culture of a wide range of human tumors in the central nervous system (CNS), including adult, pediatric, and metastatic brain cancers. Histopathological, genomic, epigenomic, and single-cell RNA sequencing (scRNA-seq) analyses demonstrated that the IPTO model recapitulates cellular heterogeneity and molecular features of original tumors. Crucially, we showed that the IPTO model predicts patient-specific drug responses, including resistance mechanisms, in a prospective patient cohort. Collectively, the IPTO model represents a major breakthrough in preclinical modeling of human cancers, which provides a path toward personalized cancer therapy.
Collapse
Affiliation(s)
- Tianping Peng
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University; Shanghai Clinical Research and Trial Center, Shanghai 201210, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xiujian Ma
- Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Im Neuenheimer Feld 581, Heidelberg 69120, Germany
| | - Wei Hua
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University; National Center for Neurological Disorders, Shanghai 200040, China
| | - Changwen Wang
- Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Im Neuenheimer Feld 581, Heidelberg 69120, Germany
| | - Youjun Chu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University; Shanghai Clinical Research and Trial Center, Shanghai 201210, China
| | - Meng Sun
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University; Shanghai Clinical Research and Trial Center, Shanghai 201210, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Valentina Fermi
- Division of Experimental Neurosurgery, Department of Neurosurgery, University Hospital Heidelberg, INF400, Heidelberg 69120, Germany
| | - Stefan Hamelmann
- Deptment of Neuropathology, University Hospital Heidelberg, CCU Neuropathology, German Cancer Research Center (DKFZ), University Heidelberg, Heidelberg 69120, Germany
| | - Katharina Lindner
- DKTK Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany; Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Tanslational Neuroscience (MCTN), Heidelberg University, Heidelberg 69120, Germany; Immune Monitoring Unit, National Center for Tumor Diseases (NCT), Heidelberg 69120, Germany
| | - Chunxuan Shao
- Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Im Neuenheimer Feld 581, Heidelberg 69120, Germany
| | - Julia Zaman
- Deptment of Neuropathology, University Hospital Heidelberg, CCU Neuropathology, German Cancer Research Center (DKFZ), University Heidelberg, Heidelberg 69120, Germany
| | - Weili Tian
- Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Im Neuenheimer Feld 581, Heidelberg 69120, Germany
| | - Yue Zhuo
- Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Im Neuenheimer Feld 581, Heidelberg 69120, Germany
| | - Yassin Harim
- Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Im Neuenheimer Feld 581, Heidelberg 69120, Germany
| | - Nadja Stöffler
- Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Im Neuenheimer Feld 581, Heidelberg 69120, Germany
| | - Linda Hammann
- Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Im Neuenheimer Feld 581, Heidelberg 69120, Germany
| | - Qungen Xiao
- Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Im Neuenheimer Feld 581, Heidelberg 69120, Germany
| | - Xiaoliang Jin
- Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Im Neuenheimer Feld 581, Heidelberg 69120, Germany
| | - Rolf Warta
- Division of Experimental Neurosurgery, Department of Neurosurgery, University Hospital Heidelberg, INF400, Heidelberg 69120, Germany
| | - Catharina Lotsch
- Division of Experimental Neurosurgery, Department of Neurosurgery, University Hospital Heidelberg, INF400, Heidelberg 69120, Germany
| | - Xuran Zhuang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yuan Feng
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University; National Center for Neurological Disorders, Shanghai 200040, China
| | - Minjie Fu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University; National Center for Neurological Disorders, Shanghai 200040, China
| | - Xin Zhang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University; National Center for Neurological Disorders, Shanghai 200040, China
| | - Jinsen Zhang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University; National Center for Neurological Disorders, Shanghai 200040, China
| | - Hao Xu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University; National Center for Neurological Disorders, Shanghai 200040, China
| | - Fufang Qiu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University; National Center for Neurological Disorders, Shanghai 200040, China
| | - Liqian Xie
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University; National Center for Neurological Disorders, Shanghai 200040, China
| | - Yi Zhang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University; National Center for Neurological Disorders, Shanghai 200040, China
| | - Wei Zhu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University; National Center for Neurological Disorders, Shanghai 200040, China
| | - Zunguo Du
- Department of Pathology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Lorena Salgueiro
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim 68167, Germany; Division of Personalized Medical Oncology (A420), German Cancer Research Center (DKFZ), Heidelberg 69120, Germany; Department of Personalized Oncology, University Hospital Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim 68167, Germany
| | - Mark Schneider
- Translational Research Unit, Thoraxklinik at Heidelberg University, Heidelberg 69120, Germany; Translational Lung Research Center Heidelberg (TRLC), German Center for Lung Research (DZL), Heidelberg 69120, Germany
| | - Florian Eichhorn
- Department of Thoracic Surgery, Thoraxklinik, University Hospital Heidelberg, Roentgenstrasse 1, Heidelberg 69126, Germany; Translational Lung Research Center Heidelberg (TRLC), German Center for Lung Research (DZL), Heidelberg 69120, Germany
| | - Arthur Lefevre
- Department of Neuropeptide Research in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim 68167, Germany
| | - Stefan Pusch
- Deptment of Neuropathology, University Hospital Heidelberg, CCU Neuropathology, German Cancer Research Center (DKFZ), University Heidelberg, Heidelberg 69120, Germany
| | - Valery Grinevich
- Department of Neuropeptide Research in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim 68167, Germany
| | - Miriam Ratliff
- DKTK Clinical Cooperation Unit (CCU) Neurooncology, German Cancer Research Center (DKFZ), Department of Neurosurgery, University Hospital Mannheim, University of Heidelberg, Mannheim 68167, Germany
| | - Sonja Loges
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim 68167, Germany; Division of Personalized Medical Oncology (A420), German Cancer Research Center (DKFZ), Heidelberg 69120, Germany; Department of Personalized Oncology, University Hospital Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim 68167, Germany; Translational Lung Research Center Heidelberg (TRLC), German Center for Lung Research (DZL), Heidelberg 69120, Germany
| | - Lukas Bunse
- DKTK Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany; Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Tanslational Neuroscience (MCTN), Heidelberg University, Heidelberg 69120, Germany; Immune Monitoring Unit, National Center for Tumor Diseases (NCT), Heidelberg 69120, Germany
| | - Felix Sahm
- Deptment of Neuropathology, University Hospital Heidelberg, CCU Neuropathology, German Cancer Research Center (DKFZ), University Heidelberg, Heidelberg 69120, Germany
| | - Yangfei Xiang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China; Shanghai Clinical Research and Trial Center, Shanghai 201210, China
| | - Andreas Unterberg
- Division of Experimental Neurosurgery, Department of Neurosurgery, University Hospital Heidelberg, INF400, Heidelberg 69120, Germany
| | - Andreas von Deimling
- Deptment of Neuropathology, University Hospital Heidelberg, CCU Neuropathology, German Cancer Research Center (DKFZ), University Heidelberg, Heidelberg 69120, Germany
| | - Michael Platten
- DKTK Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany; DKFZ Hector Cancer Institute at the University Medical Center Mannheim, Helmholtz Institute of Translational Oncology Mainz (HI-TRON Mainz) - a Helmholtz Institute of the DKFZ, Mainz 55131, Germany; Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Tanslational Neuroscience (MCTN), Heidelberg University, Heidelberg 69120, Germany; Immune Monitoring Unit, National Center for Tumor Diseases (NCT), Heidelberg 69120, Germany; German Cancer Consortium (DKTK), DKFZ, Core Center, Heidelberg 69120, Germany
| | - Christel Herold-Mende
- Division of Experimental Neurosurgery, Department of Neurosurgery, University Hospital Heidelberg, INF400, Heidelberg 69120, Germany
| | - Yonghe Wu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University; Shanghai Clinical Research and Trial Center, Shanghai 201210, China.
| | - Hai-Kun Liu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University; Shanghai Clinical Research and Trial Center, Shanghai 201210, China; Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Im Neuenheimer Feld 581, Heidelberg 69120, Germany.
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University; National Center for Neurological Disorders, Shanghai 200040, China.
| |
Collapse
|
8
|
Zhao X, Ni L, Kubo M, Matsuto M, Sakurai H, Shimizu M, Takahashi Y, Sato R, Yamauchi Y. Modeling statin-induced myopathy with hiPSC-derived myocytes reveals that impaired proteostasis underlies the myotoxicity and is targetable for the prevention. Am J Physiol Cell Physiol 2025; 328:C1247-C1259. [PMID: 40055879 DOI: 10.1152/ajpcell.00714.2024] [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: 09/24/2024] [Revised: 10/16/2024] [Accepted: 02/18/2025] [Indexed: 04/01/2025]
Abstract
Statins, 3-hydroxy-3-methylglutaryl (HMG)-CoA reductase inhibitors, have been widely prescribed to lower circulating low-density lipoprotein cholesterol levels and reduce the risk of cardiovascular disease. Although statins are well tolerated, statin-associated muscle symptoms (SAMS) are the major adverse effect and cause statin intolerance. Therefore, understanding the molecular mechanisms of SAMS and developing effective strategies for its prevention are of significant clinical importance; however, both remain unclear. Here, we establish a model of statin-induced myopathy (SIM) with human induced pluripotent stem cell (hiPSC)-derived myocytes (iPSC-MCs) and investigate the effect of statins on protein homeostasis (proteostasis) that affects skeletal muscle wasting and myotoxicity. We show that treating hiPSC-MCs with statins induces atrophic phenotype and myotoxicity, establishing an hiPSC-based SIM model. We then examine whether statins impair the balance between protein synthesis and degradation. The results show that statins not only suppress protein synthesis but also promote protein degradation by upregulating the expression of the muscle-specific E3 ubiquitin ligase Atrogin-1 in a mevalonate pathway-dependent manner. Mechanistically, blocking the mevalonate pathway inactivates the protein kinase Akt, leading to the inhibition of mTOR complex 1 (mTORC1) but the activation of GSK3β and FOXO1. These changes explain the statin-induced impairment in proteostasis. Finally, we show that pharmacological blockage of FOXO1 prevents SIM in hiPSC-MCs, implicating FOXO1 as a key mediator of SIM. Taken together, this study suggests that the mevalonate pathway is critical for maintaining skeletal muscle proteostasis and identifies FOXO1 as a potential target for preventing SIM.NEW & NOTEWORTHY This work established a human induced pluripotent stem (iPS) cell-based model for statin-induced myopathy (SIM) and demonstrated that blocking the mevalonate pathway disrupts the balance between protein synthesis and degradation, leading to myopathy. Furthermore, the present study showed that pharmacological inhibition of the transcription factor FOXO1 prevents SIM in human iPS cell-derived myocytes, suggesting that FOXO1 is a key mediator of SIM and a potential target for its prevention.
Collapse
Affiliation(s)
- Xiaolin Zhao
- Laboratory of Food Biochemistry, Department of Applied Biological Chemistry, Graduate School of Life and Agricultural Sciences, The University of Tokyo, Tokyo, Japan
| | - Liyang Ni
- Laboratory of Food Biochemistry, Department of Applied Biological Chemistry, Graduate School of Life and Agricultural Sciences, The University of Tokyo, Tokyo, Japan
| | - Miharu Kubo
- Nutri-Life Science Laboratory, Department of Applied Biological Chemistry, Graduate School of Life and Agricultural Sciences, The University of Tokyo, Tokyo, Japan
- Laboratory of Genome and Biosignals, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Mariko Matsuto
- Nutri-Life Science Laboratory, Department of Applied Biological Chemistry, Graduate School of Life and Agricultural Sciences, The University of Tokyo, Tokyo, Japan
| | - Hidetoshi Sakurai
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Makoto Shimizu
- Nutri-Life Science Laboratory, Department of Applied Biological Chemistry, Graduate School of Life and Agricultural Sciences, The University of Tokyo, Tokyo, Japan
| | - Yu Takahashi
- Laboratory of Food Biochemistry, Department of Applied Biological Chemistry, Graduate School of Life and Agricultural Sciences, The University of Tokyo, Tokyo, Japan
| | - Ryuichiro Sato
- Laboratory of Food Biochemistry, Department of Applied Biological Chemistry, Graduate School of Life and Agricultural Sciences, The University of Tokyo, Tokyo, Japan
- Nutri-Life Science Laboratory, Department of Applied Biological Chemistry, Graduate School of Life and Agricultural Sciences, The University of Tokyo, Tokyo, Japan
| | - Yoshio Yamauchi
- Laboratory of Food Biochemistry, Department of Applied Biological Chemistry, Graduate School of Life and Agricultural Sciences, The University of Tokyo, Tokyo, Japan
- Nutri-Life Science Laboratory, Department of Applied Biological Chemistry, Graduate School of Life and Agricultural Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
9
|
Trudler D, Ghatak S, Bula M, Parker J, Talantova M, Luevanos M, Labra S, Grabauskas T, Noveral SM, Teranaka M, Schahrer E, Dolatabadi N, Bakker C, Lopez K, Sultan A, Patel P, Chan A, Choi Y, Kawaguchi R, Stankiewicz P, Garcia-Bassets I, Kozbial P, Rosenfeld MG, Nakanishi N, Geschwind DH, Chan SF, Lin W, Schork NJ, Ambasudhan R, Lipton SA. Dysregulation of miRNA expression and excitation in MEF2C autism patient hiPSC-neurons and cerebral organoids. Mol Psychiatry 2025; 30:1479-1496. [PMID: 39349966 PMCID: PMC11919750 DOI: 10.1038/s41380-024-02761-9] [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: 01/11/2024] [Revised: 09/13/2024] [Accepted: 09/20/2024] [Indexed: 03/20/2025]
Abstract
MEF2C is a critical transcription factor in neurodevelopment, whose loss-of-function mutation in humans results in MEF2C haploinsufficiency syndrome (MHS), a severe form of autism spectrum disorder (ASD)/intellectual disability (ID). Despite prior animal studies of MEF2C heterozygosity to mimic MHS, MHS-specific mutations have not been investigated previously, particularly in a human context as hiPSCs afford. Here, for the first time, we use patient hiPSC-derived cerebrocortical neurons and cerebral organoids to characterize MHS deficits. Unexpectedly, we found that decreased neurogenesis was accompanied by activation of a micro-(mi)RNA-mediated gliogenesis pathway. We also demonstrate network-level hyperexcitability in MHS neurons, as evidenced by excessive synaptic and extrasynaptic activity contributing to excitatory/inhibitory (E/I) imbalance. Notably, the predominantly extrasynaptic (e)NMDA receptor antagonist, NitroSynapsin, corrects this aberrant electrical activity associated with abnormal phenotypes. During neurodevelopment, MEF2C regulates many ASD-associated gene networks, suggesting that treatment of MHS deficits may possibly help other forms of ASD as well.
Collapse
Affiliation(s)
- Dorit Trudler
- Neurodegeneration New Medicines Center and Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
- Neurodegenerative Disease Center, Scintillon Institute, San Diego, CA, USA
| | - Swagata Ghatak
- Neurodegeneration New Medicines Center and Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
- Neurodegenerative Disease Center, Scintillon Institute, San Diego, CA, USA
- School of Biological Sciences, National Institute of Science Education and Research (NISER)-Bhubaneswar, an Off Campus Center of Homi Bhabha National Institute, Jatani, Odisha, India
| | - Michael Bula
- Neurodegeneration New Medicines Center and Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - James Parker
- Neurodegenerative Disease Center, Scintillon Institute, San Diego, CA, USA
| | - Maria Talantova
- Neurodegeneration New Medicines Center and Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
- Neurodegenerative Disease Center, Scintillon Institute, San Diego, CA, USA
| | - Melissa Luevanos
- Neurodegeneration New Medicines Center and Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Sergio Labra
- Neurodegeneration New Medicines Center and Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Titas Grabauskas
- Neurodegeneration New Medicines Center and Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Sarah Moore Noveral
- Neurodegenerative Disease Center, Scintillon Institute, San Diego, CA, USA
- Department of Neurosciences, University of California, San Diego, School of Medicine, La Jolla, CA, USA
| | - Mayu Teranaka
- Neurodegenerative Disease Center, Scintillon Institute, San Diego, CA, USA
| | - Emily Schahrer
- Neurodegeneration New Medicines Center and Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Nima Dolatabadi
- Neurodegeneration New Medicines Center and Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
- Neurodegenerative Disease Center, Scintillon Institute, San Diego, CA, USA
| | - Clare Bakker
- Neurodegeneration New Medicines Center and Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Kevin Lopez
- Neurodegenerative Disease Center, Scintillon Institute, San Diego, CA, USA
| | - Abdullah Sultan
- Neurodegenerative Disease Center, Scintillon Institute, San Diego, CA, USA
| | - Parth Patel
- Neurodegeneration New Medicines Center and Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Agnes Chan
- Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Yongwook Choi
- Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Riki Kawaguchi
- Departments of Psychiatry and Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Pawel Stankiewicz
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Ivan Garcia-Bassets
- Howard Hughes Medical Institute, School and Department of Medicine, University of California, San Diego School of Medicine, La Jolla, CA, USA
| | - Piotr Kozbial
- Howard Hughes Medical Institute, School and Department of Medicine, University of California, San Diego School of Medicine, La Jolla, CA, USA
| | - Michael G Rosenfeld
- Howard Hughes Medical Institute, School and Department of Medicine, University of California, San Diego School of Medicine, La Jolla, CA, USA
| | - Nobuki Nakanishi
- Neurodegenerative Disease Center, Scintillon Institute, San Diego, CA, USA
| | - Daniel H Geschwind
- Department of Neurology, Center for Autism Research and Treatment, Program in Neurobehavioral Genetics, Department of Human Genetics, Department of Psychiatry, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Shing Fai Chan
- Center for Neuroscience, Aging, and Stem Cell Research, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
- Department of Medicine, Indiana University-Purdue University, Indianapolis, IN, USA
| | - Wei Lin
- Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Nicholas J Schork
- Translational Genomics Research Institute, Phoenix, AZ, USA
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Rajesh Ambasudhan
- Neurodegeneration New Medicines Center and Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
- Neurodegenerative Disease Center, Scintillon Institute, San Diego, CA, USA
| | - Stuart A Lipton
- Neurodegeneration New Medicines Center and Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA.
- Neurodegenerative Disease Center, Scintillon Institute, San Diego, CA, USA.
- Department of Neurosciences, University of California, San Diego, School of Medicine, La Jolla, CA, USA.
| |
Collapse
|
10
|
Fang J, Gao Y, Kang Y, Li Y, Chen Q, Chen J, Zhang P. Generation and characterization of a pluripotent stem cell line CIPi005-A derived from a female patient carrying non-canonical splice site c.827 + 1G > A in WDR45. Stem Cell Res 2025; 84:103686. [PMID: 39970510 DOI: 10.1016/j.scr.2025.103686] [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: 08/15/2024] [Revised: 02/09/2025] [Accepted: 02/14/2025] [Indexed: 02/21/2025] Open
Abstract
β-propeller protein-associated neurodegeneration (BPAN) is a rare neurodegenerative disease with developmental retardation, epilepsy and extrapyramidal symptoms, associated with mutations in WDR45. In this article, we generated an iPSC line from peripheral blood mononuclear cells (PBMCs) of a 3-year-old girl who carries a splice site mutation c.827 + 1G > A in WDR45. The generated iPSC exhibited high expression of pluripotency genes, a normal karyotype, good ability of embryoid bodies differentiation.
Collapse
Affiliation(s)
- Jie Fang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, People's Republic of China
| | - Yanyan Gao
- Capital Institute of Pediatrics, People's Republic of China
| | - Yunzhe Kang
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, People's Republic of China
| | - Yin Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, People's Republic of China
| | - Qian Chen
- Capital Institute of Pediatrics, People's Republic of China
| | - Jiayu Chen
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, People's Republic of China
| | - Peipei Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, People's Republic of China.
| |
Collapse
|
11
|
Li B, Wang Y, Yang Y, Shan D, Li J, Wang H, Sun X, Zhan Z, Ji X, Tang Y, Jiao Y, Kong B, Gao B, Sun P, Liu F, Wang Y. Generation of an induced pluripotent stem cell (iPSC) line (INNDSUi008-A) from a patient with Spinocerebellar Ataxia Type 3. Stem Cell Res 2025; 84:103675. [PMID: 39987589 DOI: 10.1016/j.scr.2025.103675] [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: 09/24/2024] [Revised: 01/07/2025] [Accepted: 02/08/2025] [Indexed: 02/25/2025] Open
Abstract
Abnormal trinucleotide CAG repeat expansions in exon 10 of the ataxin-3 (ATXN3) gene has been identified as the cause of Spinocerebellar Ataxia Type 3 (SCA3). We generated and characterized a human induced pluripotent stem cell (iPSC) line from skin fibroblasts of a patient with genetically confirmed SCA3. The pluripotency of these iPSCs was verified by the expression of several undifferentiated hPSCs markers at both RNA and protein levels, as well as their capability to differentiate into all three germ layers.
Collapse
Affiliation(s)
- Bo Li
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Yingxin Wang
- Department of Neurology, Research Institute of Neuromuscular and Neurodegenerative Diseases, Shandong Key Laboratory of Mitochondrial Medicine and Rare Diseases, Jinan, Shandong, China
| | - Yitong Yang
- School of Nursing, Jining Medical University, Jining, Shandong, China
| | - Didi Shan
- Department of Neurology, Research Institute of Neuromuscular and Neurodegenerative Diseases, Shandong Key Laboratory of Mitochondrial Medicine and Rare Diseases, Jinan, Shandong, China
| | - Jianing Li
- Department of Neurology, Research Institute of Neuromuscular and Neurodegenerative Diseases, Shandong Key Laboratory of Mitochondrial Medicine and Rare Diseases, Jinan, Shandong, China
| | - Hongxu Wang
- Department of Neurology, Research Institute of Neuromuscular and Neurodegenerative Diseases, Shandong Key Laboratory of Mitochondrial Medicine and Rare Diseases, Jinan, Shandong, China
| | - Xiaohan Sun
- School of Nursing, Jining Medical University, Jining, Shandong, China
| | - Zexin Zhan
- School of Nursing, Jining Medical University, Jining, Shandong, China
| | - Xinbo Ji
- School of Nursing, Jining Medical University, Jining, Shandong, China
| | - Yao Tang
- School of Nursing, Jining Medical University, Jining, Shandong, China
| | - Yichang Jiao
- School of Nursing, Jining Medical University, Jining, Shandong, China
| | - Bo Kong
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Bo Gao
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Ping Sun
- Prenatal Diagnostic Center of Obstetrics and Department of Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Fuchen Liu
- Department of Neurology, Research Institute of Neuromuscular and Neurodegenerative Diseases, Shandong Key Laboratory of Mitochondrial Medicine and Rare Diseases, Jinan, Shandong, China.
| | - Yu Wang
- Prenatal Diagnostic Center of Obstetrics and Department of Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| |
Collapse
|
12
|
Lee JM, Lee CY, Seol B, Jung CK, Kim Y, Kang D, Yu H, Hong Y, Song CL, Cho YS, Kim M. Tracing genomic instability in induced mesenchymal stromal cell manufacture: an integration-free transfection approach. Exp Mol Med 2025; 57:900-909. [PMID: 40229358 PMCID: PMC12046023 DOI: 10.1038/s12276-025-01439-8] [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: 12/22/2023] [Revised: 12/06/2024] [Accepted: 02/02/2025] [Indexed: 04/16/2025] Open
Abstract
Here we systematically investigated genomic alterations from the initiation of induced pluripotent stem (iPS) cell generation to induced mesenchymal stromal/stem cell differentiation. We observed a total of ten copy number alterations (CNAs) and five single-nucleotide variations (SNVs) during the phases of reprogramming, differentiation and passaging. We identified a higher frequency of CNAs and SNVs in iPS cells generated using the Sendai virus (SV) method compared with those generated with episomal vectors (Epi). Specifically, all SV-iPS cell lines exhibited CNAs during the reprogramming phase, while only 40% of Epi-iPS cells showed such alterations. Additionally, SNVs were observed exclusively in SV-derived cells during passaging and differentiation, with no SNVs detected in Epi-derived lines. Gene expression analysis revealed upregulation of chromosomal instability-related genes in late-passage SV-iPSCs, further indicating increased genomic instability. Notably, TP53 mutations were identified, underscoring the vulnerability of the gene and the critical need for careful genomic scrutiny when preparing iPS cells and derived cell lines.
Collapse
Affiliation(s)
- Jong-Mi Lee
- Catholic Genetic Laboratory Center, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Laboratory Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Chae Yeon Lee
- Catholic Genetic Laboratory Center, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Medical Sciences, Graduate School of The Catholic University of Korea, Seoul, Republic of Korea
| | - Binna Seol
- Stem Cell Research Laboratory, Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Chan Kwon Jung
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yonggoo Kim
- Catholic Genetic Laboratory Center, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Laboratory Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Dain Kang
- Catholic Genetic Laboratory Center, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Haein Yu
- Catholic Genetic Laboratory Center, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yuna Hong
- Catholic Genetic Laboratory Center, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Medical Sciences, Graduate School of The Catholic University of Korea, Seoul, Republic of Korea
| | - Cho Lok Song
- Stem Cell Research Laboratory, Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Yee Sook Cho
- Stem Cell Research Laboratory, Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.
- Department of Bioscience, KRIBB School, University of Science and Technology, Daejeon, Republic of Korea.
| | - Myungshin Kim
- Catholic Genetic Laboratory Center, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
- Department of Medical Sciences, Graduate School of The Catholic University of Korea, Seoul, Republic of Korea.
| |
Collapse
|
13
|
Lee N, Lee D, Lee JH, Lee BS, Kim S, Kim JH, Jeong S. Nanosensor-based imaging of realtime dopamine release in neurons derived from iPSCs of patients with Parkinson's disease. Mater Today Bio 2025; 31:101485. [PMID: 39906200 PMCID: PMC11791356 DOI: 10.1016/j.mtbio.2025.101485] [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: 09/17/2024] [Revised: 01/01/2025] [Accepted: 01/11/2025] [Indexed: 02/06/2025] Open
Abstract
Dopamine (DA) is an essential neuromodulator that underlies critical aspects of cognitive processes, motor function, and reward systems. Disruptions in DA signaling contribute to various neurodegenerative diseases, including Parkinson's disease (PD). Despite its important role in neuronal function, the impact of DA release/uptake on neurochemical imbalances during neuronal development remains unclear. We propose a novel application of near-infrared catecholamine nanosensor (NIRCat) for real-time visualization of DA neurotransmission among neurodegenerative disease cells. The near-infrared fluorescence (900-1400 nm) of NIRCat allows the semi-quantitative measurement of DA release in living neurons and offers insights into cellular dynamics and neuropathological development. In this study, we applied NIRCat to elucidate DA release in human induced pluripotent stem cells (hiPSCs)-derived dopaminergic neurons from both healthy control and PD patient carrying GBA1 mutations. We accurately quantified electrically stimulated DA release events, identifying distinct 'hotspots' of activity across DA neuronal cells. Our findings present a significantly enhanced spatial and temporal resolution of DA signaling, providing a deeper understanding of the role and balance of DA release in the progression of neurodegenerative disease.
Collapse
Affiliation(s)
- Nayeon Lee
- Convergence Stem Cell Research Center, Medical Research Institute, Pusan National University, Yangsan, 50612, Gyeongsangnam-do, Republic of Korea
- Department of Physiology, Pusan National University School of Medicine, Yangsan, 50612, Gyeongsangnam-do, Republic of Korea
| | - Dakyeon Lee
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan, 50612, Gyeongsangnam-do, Republic of Korea
- Department of Chemistry, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Jae Hyeok Lee
- Department of Neurology, School of Medicine, Pusan National University Yangsan Hospital, Yangsan, 50612, Gyeongsangnam-do, Republic of Korea
| | - Bo Seok Lee
- Convergence Stem Cell Research Center, Medical Research Institute, Pusan National University, Yangsan, 50612, Gyeongsangnam-do, Republic of Korea
- Department of Physiology, Pusan National University School of Medicine, Yangsan, 50612, Gyeongsangnam-do, Republic of Korea
| | - Sungjee Kim
- Department of Chemistry, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Jae Ho Kim
- Convergence Stem Cell Research Center, Medical Research Institute, Pusan National University, Yangsan, 50612, Gyeongsangnam-do, Republic of Korea
- Department of Physiology, Pusan National University School of Medicine, Yangsan, 50612, Gyeongsangnam-do, Republic of Korea
| | - Sanghwa Jeong
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan, 50612, Gyeongsangnam-do, Republic of Korea
| |
Collapse
|
14
|
Jiménez A, López-Ornelas A, Gutiérrez-de la Cruz N, Puente-Rivera J, Mayen-Quinto RD, Sánchez-Monciváis A, Ignacio-Mejía I, Albores-Méndez EM, Vargas-Hernández MA, Estudillo E. The Use of Neurons Derived from Pluripotent Stem Cells to Study Nerve-Cancer Cell Interactions. Int J Mol Sci 2025; 26:3057. [PMID: 40243726 PMCID: PMC11988749 DOI: 10.3390/ijms26073057] [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: 03/10/2025] [Revised: 03/24/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Abstract
Tumor innervation is a complex interaction between nerves and cancer cells that consists of axons invading tumors, and its complexity remains largely unknown in humans. Although some retrospective studies have provided important insights into the relationship between nerves and tumors, further knowledge is required about this biological process. Animal experiments have elucidated several molecular and cellular mechanisms of tumor innervation; however, no experimental models currently exist to study interactions between human cancer and nerve cells. Human pluripotent stem cells can differentiate into neurons for research purposes; however, the use of these neurons to study interactions with cancer cells remains largely unexplored. Hence, here we analyze the potential of human pluripotent stem cells to study the interaction of cancer cells and neurons derived from human pluripotent stem cells to unravel the poorly understood mechanisms of human tumor innervation.
Collapse
Affiliation(s)
- Adriana Jiménez
- División de Investigación, Hospital Juárez de México, Mexico City 07760, Mexico; (A.J.); (A.L.-O.); (J.P.-R.)
| | - Adolfo López-Ornelas
- División de Investigación, Hospital Juárez de México, Mexico City 07760, Mexico; (A.J.); (A.L.-O.); (J.P.-R.)
- Hospital Nacional Homeopático, Hospitales Federales de Referencia, Mexico City 06800, Mexico
| | - Neptali Gutiérrez-de la Cruz
- Escuela Militar de Graduados de Sanidad, Secretaría de la Defensa Nacional, Batalla de Celaya 202, Lomas de Sotelo, Miguel Hidalgo, Ciudad de México 11200, Mexico; (N.G.-d.l.C.); (R.D.M.-Q.); (A.S.-M.); (I.I.-M.); (E.M.A.-M.); (M.A.V.-H.)
| | - Jonathan Puente-Rivera
- División de Investigación, Hospital Juárez de México, Mexico City 07760, Mexico; (A.J.); (A.L.-O.); (J.P.-R.)
| | - Rodolfo David Mayen-Quinto
- Escuela Militar de Graduados de Sanidad, Secretaría de la Defensa Nacional, Batalla de Celaya 202, Lomas de Sotelo, Miguel Hidalgo, Ciudad de México 11200, Mexico; (N.G.-d.l.C.); (R.D.M.-Q.); (A.S.-M.); (I.I.-M.); (E.M.A.-M.); (M.A.V.-H.)
| | - Anahí Sánchez-Monciváis
- Escuela Militar de Graduados de Sanidad, Secretaría de la Defensa Nacional, Batalla de Celaya 202, Lomas de Sotelo, Miguel Hidalgo, Ciudad de México 11200, Mexico; (N.G.-d.l.C.); (R.D.M.-Q.); (A.S.-M.); (I.I.-M.); (E.M.A.-M.); (M.A.V.-H.)
| | - Iván Ignacio-Mejía
- Escuela Militar de Graduados de Sanidad, Secretaría de la Defensa Nacional, Batalla de Celaya 202, Lomas de Sotelo, Miguel Hidalgo, Ciudad de México 11200, Mexico; (N.G.-d.l.C.); (R.D.M.-Q.); (A.S.-M.); (I.I.-M.); (E.M.A.-M.); (M.A.V.-H.)
| | - Exsal M. Albores-Méndez
- Escuela Militar de Graduados de Sanidad, Secretaría de la Defensa Nacional, Batalla de Celaya 202, Lomas de Sotelo, Miguel Hidalgo, Ciudad de México 11200, Mexico; (N.G.-d.l.C.); (R.D.M.-Q.); (A.S.-M.); (I.I.-M.); (E.M.A.-M.); (M.A.V.-H.)
| | - Marco Antonio Vargas-Hernández
- Escuela Militar de Graduados de Sanidad, Secretaría de la Defensa Nacional, Batalla de Celaya 202, Lomas de Sotelo, Miguel Hidalgo, Ciudad de México 11200, Mexico; (N.G.-d.l.C.); (R.D.M.-Q.); (A.S.-M.); (I.I.-M.); (E.M.A.-M.); (M.A.V.-H.)
| | - Enrique Estudillo
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City 14269, Mexico
| |
Collapse
|
15
|
Schaefer NK, Pavlovic BJ, Pollen AA. CellBouncer, A Unified Toolkit for Single-Cell Demultiplexing and Ambient RNA Analysis, Reveals Hominid Mitochondrial Incompatibilities. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.23.644821. [PMID: 40166335 PMCID: PMC11957168 DOI: 10.1101/2025.03.23.644821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Pooled processing, in which cells from multiple sources are cultured or captured together, is an increasingly popular strategy for droplet-based single cell sequencing studies. This design allows efficient scaling of experiments, isolation of cell-intrinsic differences, and mitigation of batch effects. We present CellBouncer, a computational toolkit for demultiplexing and analyzing single-cell sequencing data from pooled experiments. We demonstrate that CellBouncer can separate and quantify multi-species and multi-individual cell mixtures, identify unknown mitochondrial haplotypes in cells, assign treatments from lipid-conjugated barcodes or CRISPR sgRNAs, and infer pool composition, outperforming existing methods. We also introduce methods to quantify ambient RNA contamination per cell, infer individual donors' contributions to the ambient RNA pool, and determine a consensus doublet rate harmonized across data types. Applying these tools to tetraploid composite cells, we identify a competitive advantage of human over chimpanzee mitochondria across 10 cell fusion lines and provide evidence for inter-mitochondrial incompatibility and mito-nuclear incompatibility between species.
Collapse
Affiliation(s)
- Nathan K Schaefer
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Bryan J Pavlovic
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Alex A Pollen
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
16
|
Santos JLDS, Paredes BD, Adanho CSA, Nonaka CKV, da Silva KN, Santos IM, Loiola EC, Silva VAO, Rocha CAG, Souza BSDF. Generation and characterization of human-induced pluripotent stem cell lines from patients with autism spectrum disorder and SCN2A variants. Hum Cell 2025; 38:74. [PMID: 40111547 DOI: 10.1007/s13577-025-01199-7] [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: 07/02/2024] [Accepted: 03/02/2025] [Indexed: 03/22/2025]
Abstract
Autism spectrum disorders (ASD) comprise a group of complex neurodevelopmental disorders that affect communication and social interactions. Over a thousand genes have been associated with ASD, with SCN2A standing out due to its critical role in neuronal function and development. Induced pluripotent stem cells (iPSCs) derived from individuals with ASD have become invaluable in vitro models for investigating the cellular and molecular mechanisms underlying the disorder. In this study, we generated and characterized four iPSC clones from peripheral blood mononuclear cells (PBMCs) of two ASD patients carrying loss-of-function variants in the SCN2A gene. These iPSC lines underwent comprehensive characterization through multiple assays. Reverse transcription polymerase chain reaction (RT-PCR), flow cytometry, and immunofluorescence analyses confirmed the presence of pluripotency markers. An embryoid body formation assay demonstrated their potential to differentiate into the three germ layers. Sequencing analysis confirmed the SCN2A variants, while short tandem repeat (STR) analysis authenticated the cell lines, and karyotype analysis ensured chromosomal integrity. The iPSCs exhibited typical morphologic characteristics, including large nuclei with prominent nucleoli, a high nucleus-to-cytoplasm ratio, densely packed cells, and well-defined borders. These cells maintained pluripotency markers, demonstrated the ability to differentiate into the three germ layers, and showed a normal karyotype. Furthermore, we successfully generated cerebral organoids from these cells. Our study establishes a robust platform for further exploration of the pathophysiological mechanisms of ASD, particularly those involving SCN2A.
Collapse
Affiliation(s)
| | - Bruno Diaz Paredes
- Center for Biotechnology and Cell Therapy, São Rafael Hospital, Salvador, Brazil
- D'Or Institute for Research and Education (IDOR), Salvador, Brazil
| | - Corynne Stephanie Ahouefa Adanho
- Center for Biotechnology and Cell Therapy, São Rafael Hospital, Salvador, Brazil
- D'Or Institute for Research and Education (IDOR), Salvador, Brazil
- Pioneer Science Initiative, D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
| | - Carolina Kymie Vasques Nonaka
- Center for Biotechnology and Cell Therapy, São Rafael Hospital, Salvador, Brazil
- D'Or Institute for Research and Education (IDOR), Salvador, Brazil
| | - Katia Nunes da Silva
- Center for Biotechnology and Cell Therapy, São Rafael Hospital, Salvador, Brazil
- D'Or Institute for Research and Education (IDOR), Salvador, Brazil
| | - Ian Marinho Santos
- Center for Biotechnology and Cell Therapy, São Rafael Hospital, Salvador, Brazil
- D'Or Institute for Research and Education (IDOR), Salvador, Brazil
| | - Erick Correia Loiola
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil
- Center for Biotechnology and Cell Therapy, São Rafael Hospital, Salvador, Brazil
- D'Or Institute for Research and Education (IDOR), Salvador, Brazil
| | - Viviane Aline Oliveira Silva
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil
- Center for Biotechnology and Cell Therapy, São Rafael Hospital, Salvador, Brazil
- D'Or Institute for Research and Education (IDOR), Salvador, Brazil
| | - Clarissa Araújo Gurgel Rocha
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil
- Center for Biotechnology and Cell Therapy, São Rafael Hospital, Salvador, Brazil
- D'Or Institute for Research and Education (IDOR), Salvador, Brazil
| | - Bruno Solano de Freitas Souza
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil.
- Center for Biotechnology and Cell Therapy, São Rafael Hospital, Salvador, Brazil.
- D'Or Institute for Research and Education (IDOR), Salvador, Brazil.
- Pioneer Science Initiative, D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil.
| |
Collapse
|
17
|
Zeng CW. Stem Cell-Based Approaches for Spinal Cord Injury: The Promise of iPSCs. BIOLOGY 2025; 14:314. [PMID: 40136570 PMCID: PMC11940451 DOI: 10.3390/biology14030314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/09/2025] [Accepted: 03/19/2025] [Indexed: 03/27/2025]
Abstract
Spinal cord injury (SCI) is a life-altering condition that leads to severe neurological deficits and significantly impacts patients' quality of life. Despite advancements in medical care, current treatment options remain largely palliative, with limited ability to promote meaningful functional recovery. Induced pluripotent stem cells (iPSCs) have emerged as a promising avenue for regenerative medicine, offering patient-specific, cell-based therapeutic potential for SCI repair. This review provides a comprehensive overview of recent advancements in iPSC-based approaches for SCI, detailing the strategies used to generate neural cell types, including neural progenitor cells, oligodendrocytes, astrocytes, and microglia, and their roles in promoting neuroprotection and regeneration. Additionally, we examine key preclinical and clinical studies, highlighting functional recovery assessments and discussing both standardized and debated evaluation metrics. Furthermore, we address critical challenges related to safety, tumorigenicity, immune response, survival, integration, and overcoming the inhibitory microenvironment of the injured spinal cord. We also explore emerging approaches in biomaterial scaffolds, gene editing, and rehabilitation strategies that may enhance the clinical applicability of iPSC-based therapies. By addressing these challenges and refining translational strategies, iPSC-based interventions hold significant potential to revolutionize SCI treatment and improve outcomes for affected individuals.
Collapse
Affiliation(s)
- Chih-Wei Zeng
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
18
|
Mei C, Magliocca V, Chen X, Massey K, Gonzalez-Cordero A, Gray SJ, Tartaglia M, Bertini ES, Corti S, Compagnucci C. Riboflavin transporter deficiency: AAV9-SLC52A2 gene therapy as a new therapeutic strategy. Front Cell Neurosci 2025; 19:1523773. [PMID: 40134705 PMCID: PMC11933037 DOI: 10.3389/fncel.2025.1523773] [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: 11/06/2024] [Accepted: 02/17/2025] [Indexed: 03/27/2025] Open
Abstract
Riboflavin transporter deficiency syndrome (RTD) is a rare childhood-onset neurodegenerative disorder caused by mutations in SLC52A2 and SLC52A3 genes, encoding the riboflavin (RF) transporters hRFVT2 and hRFVT3. In the present study we focused on RTD Type 2, which is due to variants in SLC52A2 gene. There is no cure for RTD patients and, although studies have reported clinical improvements with administration of RF, an effective treatment is still unavailable. Here we tested gene augmentation therapy on RTD type 2 patient-derived motoneurons using an adeno-associated viral vector 2/9 (AAV9) carrying the human codon optimized SLC52A2 cDNA. We optimized the in vitro transduction of motoneurons using sialidase treatment. Treated RTD motoneurons showed a significant increase in neurite's length when compared to untreated samples demonstrating that AAV9-SLC52A2 gene therapy can rescue RTD motoneurons. This leads the path towards in vivo studies offering a potential treatment for RTD patients.
Collapse
Affiliation(s)
- Cecilia Mei
- Department of Pathophysiology and Transplantation (DEPT), Università degli studi di Milano, Milan, Italy
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Valentina Magliocca
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Xin Chen
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | | | - Anai Gonzalez-Cordero
- Stem Cell Medicine Group, Children's Medical Research Institute, University of Sydney, Westmead, NSW, Australia
| | - Steven J. Gray
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Marco Tartaglia
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Enrico Silvio Bertini
- Unit of Neuromuscular and Neurodegenerative Disorders, Translational Pediatrics and Clinical Genetics, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Stefania Corti
- Department of Pathophysiology and Transplantation (DEPT), Università degli studi di Milano, Milan, Italy
| | - Claudia Compagnucci
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| |
Collapse
|
19
|
Santoso J, Do SK, Verma R, Do AV, Hendricks E, Ichida JK, McCain ML. Human iPSC-Derived Motor Neuron Innervation Enhances the Differentiation of Muscle Bundles Engineered with Benchtop Fabrication Techniques. ACS Biomater Sci Eng 2025; 11:1731-1740. [PMID: 39973396 PMCID: PMC11897949 DOI: 10.1021/acsbiomaterials.4c02225] [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: 12/10/2024] [Revised: 02/07/2025] [Accepted: 02/10/2025] [Indexed: 02/21/2025]
Abstract
Engineered skeletal muscle tissues are critical tools for disease modeling, drug screening, and regenerative medicine, but are limited by insufficient maturation. Because innervation is a critical regulator of skeletal muscle development and regeneration in vivo, motor neurons are hypothesized to improve the maturity of engineered skeletal muscle tissues. However, the impact of motor neurons on muscle phenotype when added prior to the onset of muscle differentiation is not clearly established. In this study, benchtop fabrication equipment was used to facilely fabricate chambers for engineering three-dimensional (3D) skeletal muscles bundles and measuring their contractile performance. Primary chick myoblasts were embedded in an extracellular matrix hydrogel solution and differentiated into engineered muscle bundles, with or without the addition of human induced pluripotent stem cell (hiPSC)-derived motor neurons. Muscle bundles differentiated with motor neurons had neurites distributed throughout their volume and a higher myogenic index compared to muscle bundles without motor neurons. Innervated muscle bundles also generated significantly higher twitch and tetanus forces in response to electrical field stimulation after 1 and 2 weeks of differentiation compared to noninnervated muscle bundles cultured with or without neurotrophic factors. Noninnervated muscle bundles also experienced a decline in rise and fall times as the culture progressed, whereas innervated muscle bundles and noninnervated muscle bundles with neurotrophic factors maintained more consistent rise and fall times. Innervated muscle bundles also expressed the highest levels of the genes for slow myosin light chain 3 (MYL3) and myoglobin (MB), which are associated with slow twitch fibers. These data suggest that motor neuron innervation enhances the structural and functional development of engineered skeletal muscle constructs and maintains them in a more oxidative phenotype.
Collapse
Affiliation(s)
- Jeffrey
W. Santoso
- Alfred
E. Mann Department of Biomedical Engineering, USC Viterbi School of
Engineering, University of Southern California, Los Angeles, California 90089, United States
| | - Stephanie K. Do
- Alfred
E. Mann Department of Biomedical Engineering, USC Viterbi School of
Engineering, University of Southern California, Los Angeles, California 90089, United States
| | - Riya Verma
- Department
of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine
of USC, University of Southern California, Los Angeles, California 90033, United States
| | - Alexander V. Do
- Alfred
E. Mann Department of Biomedical Engineering, USC Viterbi School of
Engineering, University of Southern California, Los Angeles, California 90089, United States
- Thomas
Jefferson High School for Science and Technology, Alexandria, Virginia 22312, United States
| | - Eric Hendricks
- Department
of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine
of USC, University of Southern California, Los Angeles, California 90033, United States
| | - Justin K. Ichida
- Department
of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine
of USC, University of Southern California, Los Angeles, California 90033, United States
| | - Megan L. McCain
- Alfred
E. Mann Department of Biomedical Engineering, USC Viterbi School of
Engineering, University of Southern California, Los Angeles, California 90089, United States
- Department
of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine
of USC, University of Southern California, Los Angeles, California 90033, United States
| |
Collapse
|
20
|
Surendran H, Battu R, Gopurappilly R, Vishnuprasad CN, Pal R. Generation of a human induced pluripotent stem cell (iPSC) line ERPLi004-A from an Alpha-1 antitrypsin deficiency (AATD) patient with SERPINA1 mutation. Stem Cell Res 2025; 83:103664. [PMID: 39884160 DOI: 10.1016/j.scr.2025.103664] [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: 09/20/2024] [Revised: 01/18/2025] [Accepted: 01/21/2025] [Indexed: 02/01/2025] Open
Abstract
Alpha-1 antitrypsin deficiency (AATD) is an autosomal disorder that causes liver and lung disease. The risk of developing lung emphysema, chronic obstructive pulmonary disorder and liver cirrhosis is observed in >75 % people affected with a homozygous mutation. Here, we describe the generation of an induced pluripotent stem cell (iPSC) line from peripheral blood mononuclear cells (PBMC) isolated from a AATD patient using non viral and non-integrating episomal vectors. The iPSC line expresses pluripotency markers, generates three germ layers in vitro and retains a normal karyotype (P20) and can provide an ideal tool for disease modelling, drug screening, and personalized medicine.
Collapse
Affiliation(s)
- Harshini Surendran
- The University of Trans-Disciplinary Health Sciences and Technology, Bengaluru 560064, India; Eyestem Research, Centre for Cellular and Molecular Platforms (C-CAMP), Bengaluru 560065, India
| | - Rajani Battu
- Eyestem Research, Centre for Cellular and Molecular Platforms (C-CAMP), Bengaluru 560065, India; Centre for Eye Genetics and Research, Bengaluru 560064, India
| | - Renjitha Gopurappilly
- NKure Therapeutics, Centre for Cellular and Molecular Platforms (C-CAMP), Bengaluru 560065, India
| | - Chethala N Vishnuprasad
- The University of Trans-Disciplinary Health Sciences and Technology, Bengaluru 560064, India
| | - Rajarshi Pal
- The University of Trans-Disciplinary Health Sciences and Technology, Bengaluru 560064, India; Eyestem Research, Centre for Cellular and Molecular Platforms (C-CAMP), Bengaluru 560065, India.
| |
Collapse
|
21
|
Bumroongthai K, Yodtup C, Jantapalaboon D, Phairoh P, Suparak S, Dhepakson P, Tubsuwan A, Wongkidakarn S. Establishing of human induced pluripotent stem cell line DMSCi002-A from the hematopoietic stem cells of a healthy male donor. Stem Cell Res 2025; 83:103655. [PMID: 39778439 DOI: 10.1016/j.scr.2025.103655] [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: 11/14/2024] [Revised: 01/02/2025] [Accepted: 01/04/2025] [Indexed: 01/11/2025] Open
Abstract
Using the integration-free episomal vector containing the reprogramming components OCT3/4/shp53, Sox2/KLF4, L-MYC/LIN28, and EBNA-1, hematopoietic stem cells obtained from a healthy 33-year-old man were effectively reprogrammed and turned into induced pluripotent stem cells (iPSCs). The reprogrammed iPSCs were grown without the use of feeders. They exhibited a normal karyotype, displayed pluripotency markers, and differentiated into cells from the three germ layers. This DMSCi002-A line may serve as a control for investigating disease mechanisms.
Collapse
Affiliation(s)
- Kobkaew Bumroongthai
- Advanced Therapy Medicinal Product Center, Medical Life Sciences Institute, Department of Medical Sciences, Ministry of Public Health, Nonthaburi, Thailand
| | - Chonlada Yodtup
- Advanced Therapy Medicinal Product Center, Medical Life Sciences Institute, Department of Medical Sciences, Ministry of Public Health, Nonthaburi, Thailand
| | - Danai Jantapalaboon
- Advanced Therapy Medicinal Product Center, Medical Life Sciences Institute, Department of Medical Sciences, Ministry of Public Health, Nonthaburi, Thailand
| | - Panapat Phairoh
- Advanced Therapy Medicinal Product Center, Medical Life Sciences Institute, Department of Medical Sciences, Ministry of Public Health, Nonthaburi, Thailand
| | - Supaporn Suparak
- Advanced Therapy Medicinal Product Center, Medical Life Sciences Institute, Department of Medical Sciences, Ministry of Public Health, Nonthaburi, Thailand
| | - Panadda Dhepakson
- Advanced Therapy Medicinal Product Center, Medical Life Sciences Institute, Department of Medical Sciences, Ministry of Public Health, Nonthaburi, Thailand
| | - Alisa Tubsuwan
- Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| | - Sudarat Wongkidakarn
- Advanced Therapy Medicinal Product Center, Medical Life Sciences Institute, Department of Medical Sciences, Ministry of Public Health, Nonthaburi, Thailand.
| |
Collapse
|
22
|
Kim J, Nam Y, Jeon D, Choi Y, Choi S, Hong CP, Kim S, Jung H, Park N, Sohn Y, Rim YA, Ju JH. Generation of hypoimmunogenic universal iPS cells through HLA-type gene knockout. Exp Mol Med 2025; 57:686-699. [PMID: 40087529 PMCID: PMC11958689 DOI: 10.1038/s12276-025-01422-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 10/20/2024] [Accepted: 12/16/2024] [Indexed: 03/17/2025] Open
Abstract
Hypoimmunogenic universal induced pluripotent stemn (iPS) cells were generated through the targeted disruption of key genes, including human leukocyte antigen (HLA)-A, HLA-B and HLA-DR alpha (DRA), using the CRISPR-Cas9 system. This approach aimed to minimize immune recognition and enhance the potential of iPS cells for allogeneic therapy. Heterozygous iPS cells were used for guide RNA design and validation to facilitate the knockout (KO) of the HLA-A, HLA-B and HLA-DRA genes. The electroporation of iPS cells using the selected guide RNAs enabled the generation of triple-KO iPS cells, followed by single-cell cloning for clone selection. Clone A7, an iPS cell with targeted KOs of the HLA-A, HLA-B and HLA-DRA genes, was identified as the final candidate. Messenger RNA analysis revealed robust expression of pluripotency markers, such as octamer-binding transcription factor 4, sex-determining region Y box 2, Krüppel-like factor 4, Lin-28 homolog A and Nanog homeobox, while protein expression assays confirmed the presence of octamer-binding transcription factor 4, stage-specific embryonic antigen 4, Nanog homeobox and tumor rejection antigen 1-60. A karyotype examination revealed no anomalies, and three-germ layer differentiation assays confirmed the differentiation potential. After interferon gamma stimulation, the gene-corrected clone A7 lacked HLA-A, HLA-B and HLA-DR protein expression. Immunogenicity testing further confirmed the hypoimmunogenicity of clone A7, which was evidenced by the absence of proliferation in central memory T cells and effector memory T cells. In conclusion, clone A7, a triple-KO iPS cell clone that demonstrates immune evasion properties, retained its intrinsic iPS cell characteristics and exhibited no immunogenicity.
Collapse
Affiliation(s)
| | - Yoojun Nam
- YiPSCELL Inc., Seoul, Republic of Korea
- Department of Biohealth Regulatory Science, Sungkyunkwan University, Suwon, Republic of Korea
| | | | | | | | | | | | | | | | - Yeowon Sohn
- Department of Biohealth Regulatory Science, Sungkyunkwan University, Suwon, Republic of Korea
| | - Yeri Alice Rim
- CiSTEM Laboratory, Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| | - Ji Hyeon Ju
- YiPSCELL Inc., Seoul, Republic of Korea.
- CiSTEM Laboratory, Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
- Department of Biomedicine and Health Sciences, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| |
Collapse
|
23
|
Saware V, Runyon W, Hu S, van Soldt B, Kumar R, Srivastava J. Optimized, Efficient Measurement of the Expression of Undifferentiated Stem Cell Markers in Human Induced Pluripotent Stem Cells (iPSCs) by Flow Cytometry. Curr Protoc 2025; 5:e70105. [PMID: 40028708 DOI: 10.1002/cpz1.70105] [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] [Indexed: 03/05/2025]
Abstract
Induced pluripotent stem cells (iPSCs) have revolutionized the fields of regenerative medicine, disease modeling, and drug discovery. However, the usage of iPSCs for various applications has been hampered by the observed line-to-line variability in their differentiation capacity. Therefore, it is important to verify the pluripotent status of iPSCs. A very effective way to define the pluripotent state of iPSCs is by evaluating the expression of established undifferentiated stem cell markers. A bona fide iPSC must have high, homogeneous expression of these markers. Here, we present a cost-effective platform that can be readily utilized by researchers to define the pluripotency status of iPSCs by measuring the expression of surface and intracellular markers by flow cytometry. © 2025 Wiley Periodicals LLC. Basic Protocol 1: iPSC culture and collection for flow cytometry analysis Basic Protocol 2: Staining of iPSCs for extracellular and intracellular undifferentiated stem cell markers Basic Protocol 3: Flow cytometry acquisition Basic Protocol 4: Flow cytometry data analysis.
Collapse
Affiliation(s)
- Vaishanavi Saware
- Gladstone Flow Cytometry Core, Gladstone Institutes, San Francisco, California
| | - Wendy Runyon
- Gladstone Stem Cell Core, Gladstone Institutes, San Francisco, California
| | - Sam Hu
- Gladstone Stem Cell Core, Gladstone Institutes, San Francisco, California
| | | | - Ritu Kumar
- Gladstone Stem Cell Core, Gladstone Institutes, San Francisco, California
| | - Jane Srivastava
- Gladstone Flow Cytometry Core, Gladstone Institutes, San Francisco, California
| |
Collapse
|
24
|
Mizushina Y, Sun L, Nishio M, Nagata S, Kamakura T, Fukuda M, Tanaka K, Toguchida J, Jin Y. Hydroxycitric acid reconstructs damaged articular cartilages by modifying the metabolic cascade in chondrogenic cells. OSTEOARTHRITIS AND CARTILAGE OPEN 2025; 7:100564. [PMID: 39835169 PMCID: PMC11743121 DOI: 10.1016/j.ocarto.2024.100564] [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: 11/27/2024] [Accepted: 12/19/2024] [Indexed: 01/22/2025] Open
Abstract
Objective Osteoarthritis, a degenerative joint disease, requires innovative therapies due to the limited ability of cartilage to regenerate. Since mesenchymal stem cells (MSCs) provide a cell source for chondrogenic cells, we hypothesize that chemicals capable of enhancing the chondrogenic potential of MSCs with transforming growth factor-beta (TGFβ) in vitro may similarly promote chondrogenesis in articular cartilage in vivo. Design Chemical compounds that enhance the TGFβ signaling for chondrogenesis were investigated utilizing mesenchymal stem cells derived from human induced pluripotent stem cells. The mechanisms of action underlying the identified compound were explored in vitro, and its therapeutic effects were validated in vivo using a mouse model of exercise-induced osteoarthritis. Results Hydroxycitric acid (HCA) emerged as the lead compound. In vitro, HCA effectively enhanced chondrogenesis by inhibiting ATP citrate lyase, inducing citrate and alpha-ketoglutarate (α-KG), while reducing cytosolic acetyl coenzyme A (Ac-CoA). This induction of α-KG promoted collagen prolyl-4-hydroxylase activity, boosting hydroxyproline production and matrix formation. The reduction of Ac-CoA attenuated the inhibitory effect of β-catenin on mitochondrial activity by diminishing its acetylation. In vivo, orally administered HCA accumulated in joint tissues of mice and histological examination demonstrated newly synthesized cartilage tissues in damaged area. Analysis of joint tissue extracts revealed a downregulation of Ac-CoA and an upregulation of citrate and α-KG, accompanied by a systemic increase in an anabolic biomarker. Conclusions HCA demonstrates promise as an osteoarthritis therapy by enhancing chondrogenic differentiation. Its ability to modulate crucial metabolic pathways and facilitate cartilage repair suggests potential for clinical translation, addressing a critical need in the treatment of osteoarthritis.
Collapse
Affiliation(s)
- Yoshiyuki Mizushina
- Department of Regeneration Sciences and Engineering, Institute for Life and Medical Sciences, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-Ku, Kyoto, 606-8507, Japan
- Central R & D Laboratory, Kobayashi Pharmaceutical Co., Ltd., 1-30-3 Toyokawa, Ibaraki, 567-0057, Japan
| | - Liping Sun
- Department of Regeneration Sciences and Engineering, Institute for Life and Medical Sciences, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Megumi Nishio
- Department of Fundamental Cell Technology, Center for iPS Cell Research and Application, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Sanae Nagata
- Department of Fundamental Cell Technology, Center for iPS Cell Research and Application, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Takeshi Kamakura
- Department of Regeneration Sciences and Engineering, Institute for Life and Medical Sciences, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Masayuki Fukuda
- Department of Regeneration Sciences and Engineering, Institute for Life and Medical Sciences, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Kousuke Tanaka
- Central R & D Laboratory, Kobayashi Pharmaceutical Co., Ltd., 1-30-3 Toyokawa, Ibaraki, 567-0057, Japan
| | - Junya Toguchida
- Department of Fundamental Cell Technology, Center for iPS Cell Research and Application, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-Ku, Kyoto, 606-8507, Japan
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Yonghui Jin
- Department of Regeneration Sciences and Engineering, Institute for Life and Medical Sciences, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| |
Collapse
|
25
|
Cui C, Chen J, Shen H, Zhang B. Derivation of induced pluripotent stem cell TUSMi013-A from a 66-year-old Chinese Han Parkinson's disease patient carrying VPS13C and TBP mutations. Stem Cell Res 2025; 83:103631. [PMID: 39914017 DOI: 10.1016/j.scr.2024.103631] [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: 11/06/2024] [Revised: 12/02/2024] [Accepted: 12/09/2024] [Indexed: 02/22/2025] Open
Abstract
Parkinson's Disease (PD), a prevalent neurodegenerative condition, is distinguished by its motor dysfunction. Induced pluripotent stem cells (iPSCs), derived from PD patients, constitute an exquisite investigative tool for elucidating the pathophysiology of the disease, assessing candidate therapeutics, and probing the potential for regenerative medicine approaches. The present study was designed to establish an iPSC line, designated TUSMi013-A, from the dermal fibroblasts of a 66-year-old female afflicted with PD, harboring mutations in VPS13C and TBP. This iPSC line offers a significant resource for the dissection of PD etiology and the innovation of novel therapeutic strategies.
Collapse
Affiliation(s)
- Can Cui
- Department of Neurology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Jian Chen
- Fenggang County People's Hospital, Guizhou Province 564200, China
| | - Hao Shen
- Department of Neurology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Bei Zhang
- Department of Neurology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200092, China.
| |
Collapse
|
26
|
Ingrungruanglert P, Phodang S, Amarinthnukrowh P, Meehart P, Pratedrat P, Suratannon N, Shotelersuk V, Suphapeetiporn K, Israsena N. Gene Correction of Wiskott-Aldrich syndrome iPS Cells Rescues Proplatelet Defects and Improves Platelet Size. Thromb Haemost 2025. [PMID: 39719152 DOI: 10.1055/a-2508-0983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2024]
Abstract
Wiskott-Aldrich syndrome (WAS) is a severe X-linked disorder caused by loss-of-function mutations in the WAS gene, responsible for encoding WAS protein (WASP), a key regulator of the actin cytoskeleton in all hematopoietic cells, except red blood cells. The mechanism underlying microthrombocytopenia, a distinctive feature of WAS and a major contributor to mortality, remains not fully elucidated. In this study, using different gene-editing strategies, we corrected mutations in patient-derived WAS-induced pluripotent stem cell (iPSC) lines, generating isogeneic WAS-iPSC lines. These included lines with direct mutation-specific correction and lines incorporating a WASP transgene cassette regulated by the MND or WAS1.6 kb promoter integrated at the safe harbor AAV1 site. Our results demonstrated that direct mutation correction successfully restored WASP levels to the equivalent of the wild-type in iPSC-derived megakaryocytes (MKs). In contrast, the AAV1-targeted strategy using the MND and WAS1.6 promoters yielded a lower level of WASP. Notably, only the mutation-specific correction lines exhibited improvements in proplatelet structures and generated larger-sized platelets. Our findings underscore the crucial roles of WASP during human thrombopoiesis and suggest that therapeutic approaches, such as direct gene correction, which can achieve physiologic levels of WASP in MKs, hold promise for ameliorating platelet defects in individuals with WAS.
Collapse
Affiliation(s)
- Praewphan Ingrungruanglert
- Center of Excellence for Stem Cell and Cell Therapy, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Excellence Center for Stem Cell and Cell Therapy, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| | - Sarinya Phodang
- Center of Excellence for Stem Cell and Cell Therapy, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Excellence Center for Stem Cell and Cell Therapy, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| | - Pramuk Amarinthnukrowh
- Department of Pediatrics, Faculty of Medicine, Center of Excellence for Medical Genomics, Medical Genomics Cluster, Chulalongkorn University, Bangkok, Thailand
- Excellence Center for Genomics and Precision Medicine, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| | - Phattarawan Meehart
- Center of Excellence for Stem Cell and Cell Therapy, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Excellence Center for Stem Cell and Cell Therapy, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| | - Pornpitra Pratedrat
- Department of Basic Medical Science, Faculty of Medicine Vajira Hospital, Navamindradhiraj University, Bangkok, Thailand
| | - Narissara Suratannon
- Division of Allergy, Immunology, and Rheumatology, Department of Pediatrics, Faculty of Medicine, Center of Excellence for Allergy and Clinical Immunology, Chulalongkorn University, Bangkok, Thailand
- King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| | - Vorasuk Shotelersuk
- Department of Pediatrics, Faculty of Medicine, Center of Excellence for Medical Genomics, Medical Genomics Cluster, Chulalongkorn University, Bangkok, Thailand
- Excellence Center for Genomics and Precision Medicine, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| | - Kanya Suphapeetiporn
- Department of Pediatrics, Faculty of Medicine, Center of Excellence for Medical Genomics, Medical Genomics Cluster, Chulalongkorn University, Bangkok, Thailand
- Excellence Center for Genomics and Precision Medicine, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| | - Nipan Israsena
- Center of Excellence for Stem Cell and Cell Therapy, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Excellence Center for Stem Cell and Cell Therapy, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
- Department of Pharmacology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
27
|
Lee MS, Lin ECY, Sivapatham A, Leiferman EM, Jiao H, Lu Y, Nemke BW, Leiferman M, Markel MD, Li WJ. Autologous iPSC- and MSC-derived chondrocyte implants for cartilage repair in a miniature pig model. Stem Cell Res Ther 2025; 16:86. [PMID: 39988676 PMCID: PMC11849328 DOI: 10.1186/s13287-025-04215-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 02/10/2025] [Indexed: 02/25/2025] Open
Abstract
BACKGROUND Induced pluripotent stem cell (iPSC)-derived mesenchymal stem cells (iMSCs) have greater potential for generating chondrocytes without hypertrophic and fibrotic phenotypes compared to bone marrow-derived mesenchymal stem/stromal cells (BMSCs). However, there is a lack of research demonstrating the use of autologous iMSCs for repairing articular chondral lesions in large animal models. In this study, we aimed to evaluate the effectiveness of autologous miniature pig (minipig) iMSC-chondrocyte (iMSC-Ch)-laden implants in comparison to autologous BMSC-chondrocyte (BMSC-Ch)-laden implants for cartilage repair in porcine femoral condyles. METHODS iMSCs and BMSCs were seeded into fibrin glue/nanofiber constructs and cultured with chondrogenic induction media for 7 days before implantation. To assess the regenerative capacity of the cells, 19 skeletally mature Yucatan minipigs were randomly divided into microfracture control, acellular scaffold, iMSC, and BMSC subgroups. A cylindrical defect measuring 7 mm in diameter and 0.6 mm in depth was created on the articular cartilage surface without violating the subchondral bone. The defects were then left untreated or treated with acellular or cellular implants. RESULTS Both cellular implant-treated groups exhibited enhanced joint repair compared to the microfracture and acellular control groups. Immunofluorescence analysis yielded significant findings, showing that cartilage treated with iMSC-Ch implants exhibited higher expression of COL2A1 and minimal to no expression of COL1A1 and COL10A1, in contrast to the BMSC-Ch-treated group. This indicates that the iMSC-Ch implants generated more hyaline cartilage-like tissue compared to the BMSC-Ch implants. CONCLUSIONS Our findings contribute to filling the knowledge gap regarding the use of autologous iPSC derivatives for cartilage repair in a translational animal model. Moreover, these results highlight their potential as a safe and effective therapeutic strategy.
Collapse
Affiliation(s)
- Ming-Song Lee
- Musculoskeletal Biology and Regenerative Medicine Laboratory, Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
| | - Eric Chang-Yi Lin
- Musculoskeletal Biology and Regenerative Medicine Laboratory, Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
| | - Athillesh Sivapatham
- Musculoskeletal Biology and Regenerative Medicine Laboratory, Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
| | - Ellen M Leiferman
- Musculoskeletal Biology and Regenerative Medicine Laboratory, Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
| | - Hongli Jiao
- Musculoskeletal Biology and Regenerative Medicine Laboratory, Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
| | - Yan Lu
- School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
| | - Brett W Nemke
- School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
| | - Matthew Leiferman
- Musculoskeletal Biology and Regenerative Medicine Laboratory, Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
| | - Mark D Markel
- School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
| | - Wan-Ju Li
- Musculoskeletal Biology and Regenerative Medicine Laboratory, Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA.
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA.
| |
Collapse
|
28
|
Di Matteo F, Bonrath R, Pravata V, Schmidt H, Ayo Martin AC, Di Giaimo R, Menegaz D, Riesenberg S, de Vrij FMS, Maccarrone G, Holzapfel M, Straub T, Kushner SA, Robertson SP, Eder M, Cappello S. Neuronal hyperactivity in neurons derived from individuals with gray matter heterotopia. Nat Commun 2025; 16:1737. [PMID: 39966398 PMCID: PMC11836124 DOI: 10.1038/s41467-025-56998-1] [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: 05/28/2024] [Accepted: 02/05/2025] [Indexed: 02/20/2025] Open
Abstract
Periventricular heterotopia (PH), a common form of gray matter heterotopia associated with developmental delay and drug-resistant seizures, poses a challenge in understanding its neurophysiological basis. Human cerebral organoids (hCOs) derived from patients with causative mutations in FAT4 or DCHS1 mimic PH features. However, neuronal activity in these 3D models has not yet been investigated. Here we show that silicon probe recordings reveal exaggerated spontaneous spike activity in FAT4 and DCHS1 hCOs, suggesting functional changes in neuronal networks. Transcriptome and proteome analyses identify changes in neuronal morphology and synaptic function. Furthermore, patch-clamp recordings reveal a decreased spike threshold specifically in DCHS1 neurons, likely due to increased somatic voltage-gated sodium channels. Additional analyses reveal increased morphological complexity of PH neurons and synaptic alterations contributing to hyperactivity, with rescue observed in DCHS1 neurons by wild-type DCHS1 expression. Overall, we provide new comprehensive insights into the cellular changes underlying symptoms of gray matter heterotopia.
Collapse
Affiliation(s)
- Francesco Di Matteo
- Division of Physiological Genomics, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-University (LMU), Munich, Germany
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), Munich, Germany
- Max Planck Institute of Psychiatry, Munich, Germany
| | - Rebecca Bonrath
- Division of Physiological Genomics, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Veronica Pravata
- Division of Physiological Genomics, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-University (LMU), Munich, Germany
| | | | - Ane Cristina Ayo Martin
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), Munich, Germany
- Max Planck Institute of Psychiatry, Munich, Germany
| | - Rossella Di Giaimo
- Division of Physiological Genomics, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-University (LMU), Munich, Germany
- Max Planck Institute of Psychiatry, Munich, Germany
- Department of Biology, University Federico II, Naples, Italy
| | | | | | - Femke M S de Vrij
- Department of Psychiatry, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | | | | | - Tobias Straub
- Bioinformatics Core, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Steven A Kushner
- Department of Psychiatry, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Department of Psychiatry, Columbia University Medical Center, New York, NY, USA
| | - Stephen P Robertson
- Department of Women's and Children's Health, University of Otago, Dunedin, New Zealand
| | - Matthias Eder
- Max Planck Institute of Psychiatry, Munich, Germany.
| | - Silvia Cappello
- Division of Physiological Genomics, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-University (LMU), Munich, Germany.
- Max Planck Institute of Psychiatry, Munich, Germany.
| |
Collapse
|
29
|
Dony L, Krontira AC, Kaspar L, Ahmad R, Demirel IS, Grochowicz M, Schäfer T, Begum F, Sportelli V, Raimundo C, Koedel M, Labeur M, Cappello S, Theis FJ, Cruceanu C, Binder EB. Chronic exposure to glucocorticoids amplifies inhibitory neuron cell fate during human neurodevelopment in organoids. SCIENCE ADVANCES 2025; 11:eadn8631. [PMID: 39951527 PMCID: PMC11827642 DOI: 10.1126/sciadv.adn8631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 01/15/2025] [Indexed: 02/16/2025]
Abstract
Disruptions in the tightly regulated process of human brain development have been linked to increased risk for brain and mental illnesses. While the genetic contribution to these diseases is well established, important environmental factors have been less studied at molecular and cellular levels. Here, we used single-cell and cell type-specific techniques to investigate the effect of glucocorticoid (GC) exposure, a mediator of antenatal environmental risk, on gene regulation and lineage specification in unguided human neural organoids. We characterized the transcriptional response to chronic GC exposure during neural differentiation and studied the underlying gene regulatory networks by integrating single-cell transcriptomics with chromatin accessibility data. We found lasting cell type-specific changes that included autism risk genes and several transcription factors associated with neurodevelopment. Chronic GC exposure influenced lineage specification primarily by priming the inhibitory neuron lineage through transcription factors like PBX3. We provide evidence for convergence of genetic and environmental risk factors through a common mechanism of altering lineage specification.
Collapse
Affiliation(s)
- Leander Dony
- Department Genes and Environment, Max Planck Institute of Psychiatry, 80804 Munich, Germany
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), 80804 Munich, Germany
- Institute of Computational Biology, Computational Health Center, Helmholtz Munich, 85764 Neuherberg, Germany
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, 85354 Freising, Germany
- German Center for Mental Health (DZPG), partner site Munich, Munich, Germany
| | - Anthi C. Krontira
- Department Genes and Environment, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Lea Kaspar
- Department Genes and Environment, Max Planck Institute of Psychiatry, 80804 Munich, Germany
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), 80804 Munich, Germany
| | - Ruhel Ahmad
- Department Genes and Environment, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Ilknur Safak Demirel
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | | | - Tim Schäfer
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Fatema Begum
- Department Genes and Environment, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Vincenza Sportelli
- Department Genes and Environment, Max Planck Institute of Psychiatry, 80804 Munich, Germany
- German Center for Mental Health (DZPG), partner site Munich, Munich, Germany
| | - Catarina Raimundo
- Department Genes and Environment, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Maik Koedel
- Department Genes and Environment, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Marta Labeur
- Department Genes and Environment, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Silvia Cappello
- German Center for Mental Health (DZPG), partner site Munich, Munich, Germany
- Physiological Genomics, Biomedical Center (BMC), LMU Munich Faculty of Medicine, 82152 Planegg-Martinsried, Germany
- Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Fabian J. Theis
- Institute of Computational Biology, Computational Health Center, Helmholtz Munich, 85764 Neuherberg, Germany
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, 85354 Freising, Germany
- German Center for Mental Health (DZPG), partner site Munich, Munich, Germany
- TUM School of Computation, Information and Technology, Technical University of Munich, 85748 Garching bei München, Germany
| | - Cristiana Cruceanu
- Department Genes and Environment, Max Planck Institute of Psychiatry, 80804 Munich, Germany
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Elisabeth B. Binder
- Department Genes and Environment, Max Planck Institute of Psychiatry, 80804 Munich, Germany
- German Center for Mental Health (DZPG), partner site Munich, Munich, Germany
- Max Planck Institute of Psychiatry, 80804 Munich, Germany
| |
Collapse
|
30
|
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
|
31
|
Kim DH, Choi SH, Sung JJ, Kim S, Yi H, Park S, Park CW, Oh YW, Lee J, Kim DS, Kim JH, Park CY, Kim DW. Long-term correction of hemophilia A via integration of a functionally enhanced FVIII gene into the AAVS1 locus by nickase in patient-derived iPSCs. Exp Mol Med 2025; 57:184-192. [PMID: 39762408 PMCID: PMC11799516 DOI: 10.1038/s12276-024-01375-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: 06/12/2024] [Revised: 09/04/2024] [Accepted: 10/08/2024] [Indexed: 02/07/2025] Open
Abstract
Hemophilia A (HA) is caused by mutations in coagulation factor VIII (FVIII). Genome editing in conjunction with patient-derived induced pluripotent stem cells (iPSCs) is a promising cell therapy strategy, as it replaces dysfunctional proteins resulting from genetic mutations with normal proteins. However, the low expression level and short half-life of FVIII still remain significant limiting factors in the efficacy of these approaches in HA. Here, we constructed a functionally enhanced FVIII variant, F309S/E1984V-mutated B domain-deleted (BDD)-FVIII (FE-FVIII), with increased activity and stability. We inserted FE-FVIII with a human elongation factor-1 alpha (EF1α) promoter into the AAVS1 locus of HA patient-derived iPSCs via CRISPR/Cas9 (D10A) nickase to ensure expression in any cell type. FE-FVIII was expressed not only in undifferentiated FE-FVIII-inserted (FE-KI) iPSCs but also in endothelial cells (ECs) differentiated from them in vitro. Compared with mice transplanted with wild-type BDD-FVIII-containing ECs, immunocompetent HA mice intravenously transplanted with FE-KI ECs presented a 2.12-fold increase in FVIII activity in the blood and an approximately 20% greater survival rate after hemorrhagic tail injury. For sustained efficacy, FE-KI ECs were subcutaneously transplanted into immunodeficient HA mice, resulting in amelioration of the hemophilia phenotype for more than 3 months. This strategy can improve FVIII function and may provide a universal therapeutic approach for treating HA.
Collapse
Affiliation(s)
- Do-Hun Kim
- Department of Physiology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea
- S. Biomedics Co., Ltd, 28 Seongsui-ro 26-gil, Seongdong-gu, Seoul, 04797, Korea
| | - Sang-Hwi Choi
- Department of Physiology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea
| | - Jin Jea Sung
- Department of Physiology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea
| | - Sieun Kim
- Department of Physiology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea
- Brain Korea 21 PLUS Program for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea
| | - Hanui Yi
- Department of Physiology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea
- Brain Korea 21 PLUS Program for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea
| | - Sanghyun Park
- Department of Physiology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea
| | - Chan Wook Park
- Department of Physiology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea
| | - Young Woo Oh
- Department of Physiology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea
- Brain Korea 21 PLUS Program for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea
| | - Jungil Lee
- Department of Physiology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea
- Brain Korea 21 PLUS Program for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea
| | - Dae-Sung Kim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Korea
| | - Jong-Hoon Kim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Korea
| | - Chul-Yong Park
- Department of Physiology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea.
- S. Biomedics Co., Ltd, 28 Seongsui-ro 26-gil, Seongdong-gu, Seoul, 04797, Korea.
| | - Dong-Wook Kim
- Department of Physiology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea.
- S. Biomedics Co., Ltd, 28 Seongsui-ro 26-gil, Seongdong-gu, Seoul, 04797, Korea.
- Brain Korea 21 PLUS Program for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea.
| |
Collapse
|
32
|
Byun S, Yoon SH, Hong YJ, Jang HS, Seo BJ, Choi GT, La H, Lee JW, Hong K, Do JT. Expression Patterns of Escape Genes in Turner Syndrome Fibroblasts and Induced Pluripotent Stem Cells. Int J Mol Sci 2025; 26:975. [PMID: 39940742 PMCID: PMC11816654 DOI: 10.3390/ijms26030975] [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: 11/25/2024] [Revised: 01/14/2025] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
Turner syndrome (TS) is an X monosomy-related disorder caused by X chromosome nondisjunction during embryonic development. Patients with TS have only one intact X chromosome, with the other either completely or partially lost. TS affects various tissues, including the liver, kidneys, brain, cardiovascular system, and ovaries. These abnormalities are suggested to involve an altered dosage of escape genes that evade X chromosome inactivation. However, the mechanisms and roles of these escape genes in the TS phenotype remain unclear. We hypothesized that the expression levels of escape genes differ between wild-type (WT) and TS cell lines. In this study, we generated induced pluripotent stem cell (iPSC) lines from WT and TS fibroblasts and examined the expression levels of escape genes in both undifferentiated fibroblasts and reprogrammed iPSCs from WT and TS samples. The reprogrammed WT and TS iPSCs exhibited general characteristics of pluripotency, including the expression of pluripotency markers and the potential to differentiate into all three germ layers. Forty-five escape genes were differentially expressed between the WT and TS cell lines. Among these, five genes (ATP7A, PHKA1, EBP, ZFX, and SMC1A) were suggested to be implicated in the TS phenotype. However, further studies using additional cell lines are necessary to clarify the correlation between TS and escape genes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Jeong-Tae Do
- Department of Stem Cell and Regenerative Biotechnology, Konkuk Institute of Technology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 133-702, Republic of Korea; (S.B.); (S.-H.Y.); (Y.-J.H.); (H.-S.J.); (B.-J.S.); (G.-T.C.); (H.L.); (J.-W.L.); (K.H.)
| |
Collapse
|
33
|
Nunes OBDS, Buranello TW, Farias FDA, Rosero J, Recchia K, Bressan FF. Can cell-cultured meat from stem cells pave the way for sustainable alternative protein? Curr Res Food Sci 2025; 10:100979. [PMID: 40040753 PMCID: PMC11878651 DOI: 10.1016/j.crfs.2025.100979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 01/09/2025] [Accepted: 01/18/2025] [Indexed: 03/06/2025] Open
Abstract
As the global population grows, the demand for food and animal-derived products rises significantly, posing a notable challenge to the progress of society in general. Alternative protein production may adequately address such a challenge, and cell-based meat production emerges as a promising solution. This review investigates methodologies for in vitro myogenesis and adipogenesis from stem cells (adult, embryonic, or induced pluripotent stem cells - iPSCs) across different animal species, as well as the remaining challenges for scalability, the possibility of genetic modification, along with safety concerns regarding the commercialization of cell-cultured meat. Regarding such complexities, interdisciplinary approaches will be vital for assessing the potential of cell-cultured meat as a sustainable protein source, mimicking the sensory and nutritional attributes of conventional livestock meat whilst meeting the demands of a growing global population while mitigating environmental impacts.
Collapse
Affiliation(s)
- Octavio Bignardi da Silva Nunes
- Department of Food Engineering, Faculty of Animal Sciences and Food Engineering, University of São Paulo 13635-000, Pirassununga, SP, Brazil
- Department of Veterinary Medicine, Faculty of Animal Sciences and Food Engineering, University of São Paulo 13635-000, Pirassununga, SP, Brazil
| | - Tiago Willian Buranello
- Department of Veterinary Medicine, Faculty of Animal Sciences and Food Engineering, University of São Paulo 13635-000, Pirassununga, SP, Brazil
- Postgraduate Program in Anatomy of Domestic and Wils Species, Faculty of Veterinary Medicine and Animal Sciences, University of São Paulo, São Paulo 01001-010, SP, Brazil
| | - Fabiana de Andrade Farias
- Department of Food Engineering, Faculty of Animal Sciences and Food Engineering, University of São Paulo 13635-000, Pirassununga, SP, Brazil
- Department of Veterinary Medicine, Faculty of Animal Sciences and Food Engineering, University of São Paulo 13635-000, Pirassununga, SP, Brazil
| | - Jenyffer Rosero
- Department of Veterinary Medicine, Faculty of Animal Sciences and Food Engineering, University of São Paulo 13635-000, Pirassununga, SP, Brazil
- Postgraduate Program in Anatomy of Domestic and Wils Species, Faculty of Veterinary Medicine and Animal Sciences, University of São Paulo, São Paulo 01001-010, SP, Brazil
| | - Kaiana Recchia
- Department of Veterinary Medicine, Faculty of Animal Sciences and Food Engineering, University of São Paulo 13635-000, Pirassununga, SP, Brazil
- Postgraduate Program in Anatomy of Domestic and Wils Species, Faculty of Veterinary Medicine and Animal Sciences, University of São Paulo, São Paulo 01001-010, SP, Brazil
| | - Fabiana Fernandes Bressan
- Department of Veterinary Medicine, Faculty of Animal Sciences and Food Engineering, University of São Paulo 13635-000, Pirassununga, SP, Brazil
- Postgraduate Program in Anatomy of Domestic and Wils Species, Faculty of Veterinary Medicine and Animal Sciences, University of São Paulo, São Paulo 01001-010, SP, Brazil
| |
Collapse
|
34
|
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
|
35
|
Setsu S, Morimoto S, Nakamura S, Ozawa F, Utami KH, Nishiyama A, Suzuki N, Aoki M, Takeshita Y, Tomari Y, Okano H. Swift induction of human spinal lower motor neurons and robust ALS cell screening via single-cell imaging. Stem Cell Reports 2025; 20:102377. [PMID: 39706179 PMCID: PMC11784480 DOI: 10.1016/j.stemcr.2024.11.007] [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: 04/07/2023] [Revised: 11/13/2024] [Accepted: 11/18/2024] [Indexed: 12/23/2024] Open
Abstract
This study introduces a novel method for rapidly and efficiently inducing human spinal lower motor neurons (LMNs) from induced pluripotent stem cells (iPSCs) to eventually elucidate the pathomechanisms of amyotrophic lateral sclerosis (ALS) and facilitate drug screening. Previous methods were limited by low induction efficiency, poor LMN purity, or labor-intensive induction and evaluation processes. Our protocol overcomes these challenges, achieving around 80% induction efficiency within just two weeks by combining a small molecule-based approach with transcription factor transduction. Moreover, to exclude non-LMN cells from the analysis, we utilized time-lapse microscopy and machine learning to analyze the morphology and viability of iPSC-derived LMNs on a single-cell basis, establishing an effective pathophysiological evaluation system. This rapid, efficient, and streamlined protocol, along with our single-cell-based evaluation method, enables large-scale analysis and drug screening using iPSC-derived motor neurons.
Collapse
Affiliation(s)
- Selena Setsu
- Keio University Regenerative Medicine Research Center, Kanagawa 210-0821, Japan; Laboratory of RNA Function, Institute for Quantitative Biosciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0032, Japan; Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0032, Japan
| | - Satoru Morimoto
- Keio University Regenerative Medicine Research Center, Kanagawa 210-0821, Japan; Division of Neurodegenerative Disease Research, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo 173-0015, Japan.
| | - Shiho Nakamura
- Keio University Regenerative Medicine Research Center, Kanagawa 210-0821, Japan; Division of Neurodegenerative Disease Research, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo 173-0015, Japan
| | - Fumiko Ozawa
- Keio University Regenerative Medicine Research Center, Kanagawa 210-0821, Japan; Division of Neurodegenerative Disease Research, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo 173-0015, Japan
| | - Kagistia Hana Utami
- Keio University Regenerative Medicine Research Center, Kanagawa 210-0821, Japan; Division of Neurodegenerative Disease Research, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo 173-0015, Japan
| | - Ayumi Nishiyama
- Department of Neurology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Naoki Suzuki
- Department of Neurology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan; Department of Rehabilitation Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masashi Aoki
- Department of Neurology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Yukio Takeshita
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Yamaguchi 753-8511, Japan; Department of Neurotherapeutics, Yamaguchi University Graduate School of Medicine, Yamaguchi 753-8511, Japan
| | - Yukihide Tomari
- Laboratory of RNA Function, Institute for Quantitative Biosciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0032, Japan; Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0032, Japan
| | - Hideyuki Okano
- Keio University Regenerative Medicine Research Center, Kanagawa 210-0821, Japan; Division of Neurodegenerative Disease Research, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo 173-0015, Japan.
| |
Collapse
|
36
|
Kang X, Ma L, Wen J, Gong W, Liu X, Hu Y, Feng Z, Jing Q, Cai Y, Li S, Cai X, Yuan K, Feng Y. Modeling of auditory neuropathy spectrum disorders associated with the TEME43 variant reveals impaired gap junction function of iPSC-derived glia-like support cells. Front Mol Neurosci 2025; 17:1457874. [PMID: 39834515 PMCID: PMC11743952 DOI: 10.3389/fnmol.2024.1457874] [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: 07/01/2024] [Accepted: 11/22/2024] [Indexed: 01/22/2025] Open
Abstract
Auditory neuropathy spectrum disorder (ANSD) is an auditory dysfunction disorder characterized by impaired speech comprehension. Its etiology is complex and can be broadly categorized into genetic and non-genetic factors. TMEM43 mutation is identified as a causative factor in ANSD. While some studies have been conducted using animal models, its pathogenic mechanisms in humans remain unclear. TMEM43 is predominantly expressed in cochlear glia-like support cells (GLSs) and plays a vital role in gap junction intercellular communication. In this work, we utilized induced pluripotent stem cells from an ANSD patient carrying the TMEM43 gene mutation c.1114C>T (p.Arg372Ter) and directed their differentiation toward GLSs to investigate the effect of TMEM43 mutation on the function of gap junctions in cochlear GLSs in vitro. Reduced expression of genes associated with GLSs characteristics and reduced gap junction intercellular communication in TMEM43 mutant cell lines were observed compared to controls. Transcriptome analysis revealed that differentially expressed genes were significantly enriched in pathways related to cell proliferation, differentiation, extracellular space and adhesion. Furthermore, significant alterations were noted in the PI3K-Akt signaling pathway and the calcium signaling pathway, which could potentially influence gap junction function and contribute to hearing loss. In summary, our study based on patient-derived iPSCs sheds new light on the molecular mechanisms by which TMEM43 mutations may lead to ANSD. These mutations could result in developmental defects in GLSs and a diminished capacity for gap junction function, which may be implicated in the auditory deficits observed in ANSD patients. Our study explored the pathological effects of the TMEM43 mutation and its causal relationship with ANSD using a patient-derived iPSC-based GLSs model, providing a foundation for future mechanistic studies and potential drug screening efforts.
Collapse
Affiliation(s)
- Xiaoming Kang
- Department of Otorhinolaryngology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Institute of Otorhinolaryngology, Head and Neck Surgery, University of South China, Changsha, China
| | - Lu Ma
- Institute of Otorhinolaryngology, Head and Neck Surgery, University of South China, Changsha, China
- MOE Key Lab of Rare Pediatric Diseases & Institute for Future Sciences, University of South China, Changsha, China
- Institute of Cytology and Genetics, Hengyang Medical School, University of South China, Hengyang, China
| | - Jie Wen
- Department of Otorhinolaryngology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Institute of Otorhinolaryngology, Head and Neck Surgery, University of South China, Changsha, China
| | - Wei Gong
- Department of Otorhinolaryngology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Institute of Otorhinolaryngology, Head and Neck Surgery, University of South China, Changsha, China
| | - Xianlin Liu
- Department of Otorhinolaryngology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Institute of Otorhinolaryngology, Head and Neck Surgery, University of South China, Changsha, China
| | - Yihan Hu
- Department of Otorhinolaryngology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Institute of Otorhinolaryngology, Head and Neck Surgery, University of South China, Changsha, China
| | - Zhili Feng
- Department of Otorhinolaryngology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Institute of Otorhinolaryngology, Head and Neck Surgery, University of South China, Changsha, China
| | - Qiancheng Jing
- Department of Otorhinolaryngology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Institute of Otorhinolaryngology, Head and Neck Surgery, University of South China, Changsha, China
| | - Yuexiang Cai
- Department of Otorhinolaryngology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Institute of Otorhinolaryngology, Head and Neck Surgery, University of South China, Changsha, China
| | - Sijun Li
- Department of Otorhinolaryngology, Xiangya Hospital Central South University, Changsha, China
| | - Xinzhang Cai
- Department of Otorhinolaryngology, Xiangya Hospital Central South University, Changsha, China
| | - Kai Yuan
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Oncology, Xiangya Hospital & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Yong Feng
- Department of Otorhinolaryngology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Institute of Otorhinolaryngology, Head and Neck Surgery, University of South China, Changsha, China
- MOE Key Lab of Rare Pediatric Diseases & Institute for Future Sciences, University of South China, Changsha, China
- Institute of Cytology and Genetics, Hengyang Medical School, University of South China, Hengyang, China
- Department of Otorhinolaryngology, Xiangya Hospital Central South University, Changsha, China
| |
Collapse
|
37
|
Valdes P, Caldwell AB, Liu Q, Fitzgerald MQ, Ramachandran S, Karch CM, Galasko DR, Yuan SH, Wagner SL, Subramaniam S. Integrative multiomics reveals common endotypes across PSEN1, PSEN2, and APP mutations in familial Alzheimer's disease. Alzheimers Res Ther 2025; 17:5. [PMID: 39754192 PMCID: PMC11699654 DOI: 10.1186/s13195-024-01659-6] [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: 05/20/2024] [Accepted: 12/20/2024] [Indexed: 01/06/2025]
Abstract
BACKGROUND PSEN1, PSEN2, and APP mutations cause Alzheimer's disease (AD) with an early age at onset (AAO) and progressive cognitive decline. PSEN1 mutations are more common and generally have an earlier AAO; however, certain PSEN1 mutations cause a later AAO, similar to those observed in PSEN2 and APP. METHODS We examined whether common disease endotypes exist across these mutations with a later AAO (~ 55 years) using hiPSC-derived neurons from familial Alzheimer's disease (FAD) patients harboring mutations in PSEN1A79V, PSEN2N141I, and APPV717I and mechanistically characterized by integrating RNA-seq and ATAC-seq. RESULTS We identified common disease endotypes, such as dedifferentiation, dysregulation of synaptic signaling, repression of mitochondrial function and metabolism, and inflammation. We ascertained the master transcriptional regulators associated with these endotypes, including REST, ASCL1, and ZIC family members (activation), and NRF1 (repression). CONCLUSIONS FAD mutations share common regulatory changes within endotypes with varying severity, resulting in reversion to a less-differentiated state. The regulatory mechanisms described offer potential targets for therapeutic interventions.
Collapse
Affiliation(s)
- Phoebe Valdes
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093, USA
- Bioengineering Graduate Program, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Andrew B Caldwell
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Qing Liu
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, 92093, USA
- Present Address: Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Michael Q Fitzgerald
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093, USA
- Bioengineering Graduate Program, University of California, San Diego, La Jolla, CA, 92093, USA
| | | | - Celeste M Karch
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO, 63110, USA
| | - Douglas R Galasko
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Shauna H Yuan
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, 92093, USA
- Present Address: N. Bud Grossman Center for Memory Research and Care, Department of Neurology, University of Minnesota, GRECC, Minneapolis VA Health Care System, Minneapolis, MN, 55417, USA
| | - Steven L Wagner
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, 92093, USA
- VA San Diego Healthcare System, San Diego, CA, 92161, USA
| | - Shankar Subramaniam
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093, USA.
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, 92093, USA.
- Department of Nanoengineering, University of California, San Diego, La Jolla, CA, 92093, USA.
- Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
38
|
Taei A, Sajadi FS, Salahi S, Enteshari Z, Falah N, Shiri Z, Abasalizadeh S, Hajizadeh-Saffar E, Hassani SN, Baharvand H. The cell replacement therapeutic potential of human pluripotent stem cells. Expert Opin Biol Ther 2025; 25:47-67. [PMID: 39679436 DOI: 10.1080/14712598.2024.2443079] [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: 08/21/2024] [Revised: 11/29/2024] [Accepted: 12/12/2024] [Indexed: 12/17/2024]
Abstract
INTRODUCTION The remarkable ability of human pluripotent stem cells (hPSCs) to differentiate into specialized cells of the human body emphasizes their immense potential in treating various diseases. Advances in hPSC technology are paving the way for personalized and allogeneic cell-based therapies. The first-in-human studies showed improved treatment of diseases with no adverse effects, which encouraged the industrial production of this type of medicine. To ensure the quality, safety and efficacy of hPSC-based products throughout their life cycle, it is important to monitor and control their clinical translation through good practices (GxP) regulations. Understanding these rules in advance will help ensure that the industrial development of hPSC-derived products for widespread clinical implementation is feasible and progresses rapidly. AREAS COVERED In this review, we discuss the key translational obstacles of hPSCs, outline the current hPSC-based clinical trials, and present a workflow for putative clinical hPSC-based products. Finally, we highlight some future therapeutic opportunities for hPSC-derivatives. EXPERT OPINION hPSC-based products continue to show promise for the treatment of a variety of diseases. While clinical trials support the relative safety and efficacy of hPSC-based products, further investigation is required to explore the clinical challenges and achieve exclusive regulations for hPSC-based cell therapies.
Collapse
Affiliation(s)
- Adeleh Taei
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Fatemeh-Sadat Sajadi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Developmental Biology, School of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
| | - Sarvenaz Salahi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Zahra Enteshari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Nasrin Falah
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Zahra Shiri
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Saeed Abasalizadeh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Ensiyeh Hajizadeh-Saffar
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Seyedeh-Nafiseh Hassani
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Developmental Biology, School of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
| |
Collapse
|
39
|
Gavrilov NS, Ignatyeva NV, Medvedeva EV, Timashev PS. Articular cartilage tissue engineering using genetically modified induced pluripotent stem cell lines. GENES & CELLS 2024; 19:404-424. [DOI: 10.17816/gc633492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2025]
Abstract
Mature hyaline cartilage has a low regenerative potential and its repair remains a complex clinical and research issue. Articular cartilage injuries often contribute to the development of osteoarthritis, resulting in loss of joint function and patient disability. Surgical techniques for repairing articular surfaces, such as mosaic chondroplasty and microfracture, which are designed for small defects, cannot be used for degenerative and dystrophic cartilage lesions. Cell therapy using chondrocytes differentiated from induced pluripotent stem cells (iPSCs) is a promising approach to reconstruct articular cartilage tissue. iPSCs have high proliferative activity, which allows the harvesting of autologous cells in quantities necessary to repair a joint defect. CRISPR-Cas genome editing technology, based on the bacterial adaptive immune system, enables the genetic modification of iPSCs to obtain progenitor cells with specific characteristics and properties.
This review describes specific research papers on the combined use of iPSC and CRISPR-Cas technologies for the evaluation of cartilage regenerative medicine. Papers were evaluated for the last twelve years since CRISPR-Cas technology was introduced to the global community. CRISPR-Cas is currently being used to address therapeutic issues in articular cartilage regeneration by increasing the efficiency of chondrogenic differentiation of iPSC lines and harvesting a more homogeneous population of chondroprogenitor cells. Another approach is to remove the pro-inflammatory cytokine receptor sequence to produce inflammation-resistant cartilage. Finally, knocking out genes for components of the major histocompatibility complex allows harvesting chondrocytes that are invisible to the recipient's immune system. This kind of research contributes to personalized healthcare and can improve the quality of life of the world's population in the long term.
Collapse
|
40
|
Yang R, Qi Y, Zhang X, Gao H, Yu Y. Living biobank: Standardization of organoid construction and challenges. Chin Med J (Engl) 2024; 137:3050-3060. [PMID: 39663560 PMCID: PMC11706585 DOI: 10.1097/cm9.0000000000003414] [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: 09/27/2024] [Indexed: 12/13/2024] Open
Abstract
ABSTRACT In multiple areas such as science, technology, and economic activities, it is necessary to unify the management of repetitive tasks or concepts by standardization to obtain the best order and high efficiency. Organoids, as living tissue models, have rapidly developed in the past decade. Organoids can be used repetitively for in vitro culture, cryopreservation, and recovery for further utilization. Because organoids can recapitulate the parental tissues' morphological phenotypes, cell functions, biological behaviors, and genomic profiles, they are known as renewable "living biobanks". Organoids cover two mainstream fields: Adult stem cell-derived organoids (also known as patient-derived organoids) and induced pluripotent stem cell-derived and/or embryonic stem cell-derived organoids. Given the increasing importance of organoids in the development of new drugs, standardized operation, and management in all steps of organoid construction is an important guarantee to ensure the high quality of products. In this review, we systematically introduce the standardization of organoid construction operation procedures, the standardization of laboratory construction, and available standardization documents related to organoid culture that have been published so far. We also proposed the challenges and prospects in this field.
Collapse
Affiliation(s)
- Ruixin Yang
- Department of General Surgery of Ruijin Hospital, Shanghai Institute of Digestive Surgery, and Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yao Qi
- National Engineering Center for Biochip at Shanghai, Shanghai 200120, China
| | - Xiaoyan Zhang
- National Engineering Center for Biochip at Shanghai, Shanghai 200120, China
| | - Hengjun Gao
- National Engineering Center for Biochip at Shanghai, Shanghai 200120, China
| | - Yingyan Yu
- Department of General Surgery of Ruijin Hospital, Shanghai Institute of Digestive Surgery, and Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
41
|
van Ham WB, Meijboom EEM, Ligtermoet ML, Monshouwer-Kloots J, Riele ASJMT, Asselbergs FW, van Rooij E, Bourfiss M, van Veen TAB. An hiPSC-CM approach for electrophysiological phenotyping of a patient-specific case of short-coupled TdP. Stem Cell Res Ther 2024; 15:470. [PMID: 39695883 PMCID: PMC11656816 DOI: 10.1186/s13287-024-04074-8] [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: 09/10/2024] [Accepted: 11/21/2024] [Indexed: 12/20/2024] Open
Abstract
INTRODUCTION A healthy young woman, age 26 without prior cardiac complications, experienced an out-of-hospital cardiac arrest caused by ventricular fibrillation (VF), which coincided with a fever. Comprehensive diagnostics including echo, CMR, exercise testing, and genetic sequencing, did not identify any potential cause. This led to the diagnosis of idiopathic VF and installment of an implantable cardioverter defibrillator, which six months later appropriately intervened another VF episode under conditions comparable to the first event. A second diagnostic opinion concluded short-coupled Torsade de Pointes (scTdP), and the patient was started on a verapamil treatment. METHODS From this patient, human induced pluripotent stem cell cardiomyocyte (hiPSC-CM) lines were generated to study cellular electrophysiology. Without a known genetic pathogenic variation, no isogenic control line could be produced, therefore a healthy age- and sex-matched control hiPSC-CM line was used. Cellular electrophysiology was studied in these cardiomyocytes using calcium- and voltage sensitive fluorescent dyes and measurements were carried out at 37 °C and 39 °C, to mimic the condition of hyperthermia in the patient. mRNA expression of electrophysiologically relevant genes were analyzed to identify a potential underlying mechanism. RESULTS Calcium transients measured in patient lines at a physiological temperature indicated the occurrence of early after transients (EATs). Strikingly, at 39 °C the incidence of EATs further increased. Membrane potential data from the patient also revealed shorter action potentials that, combined with the EATs, indicate the premature release of calcium during diastole, which could be responsible for the extrasystoles in the patient. Gene expression profiles were mainly downregulated in the patient but could not clearly aid in unraveling a mechanism behind the occurrence of EATs. Pharmacological screening was performed to evaluate the treatment regimen and to determine a mechanism of action of the EATs. While verapamil, dantrolene, and flecainide did not decrease the incidence of EATs, calcium handling parameters were affected indicating functionality of the drugs. CONCLUSION This patient-specific case of electrophysiological phenotyping resulted in a hypothesis of the possible mechanism behind the scTdP arrhythmias, but also accentuates the applicability of patient-specific hiPSC-CM disease modeling and phenotyping.
Collapse
Affiliation(s)
- Willem B van Ham
- Department of Medical Physiology, University Medical Center Utrecht, Utrecht, The Netherlands.
| | - Esmeralda E M Meijboom
- Department of Medical Physiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Merel L Ligtermoet
- Department of Medical Physiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jantine Monshouwer-Kloots
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Folkert W Asselbergs
- Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Cardiology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Eva van Rooij
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Mimount Bourfiss
- Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Toon A B van Veen
- Department of Medical Physiology, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
42
|
Das SC, Schulmann A, Callor WB, Jerominski L, Panicker MM, Christensen ED, Bunney WE, Williams ME, Coon H, Vawter MP. Altered transcriptomes, cell type proportions, and dendritic spine morphology in hippocampus of suicide decedents. J Affect Disord 2024; 367:118-128. [PMID: 39191313 DOI: 10.1016/j.jad.2024.08.144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 08/03/2024] [Accepted: 08/23/2024] [Indexed: 08/29/2024]
Abstract
BACKGROUND Suicide is a manner of death resulting from complex environmental and genetic risks that affect millions of people globally. Both structural and functional studies identified the hippocampus as one of the vulnerable brain regions contributing to suicide risk. METHODS We have identified the hippocampal tissue transcriptomes, gene ontology, cell type proportions, and dendritic spine morphology in controls (n = 28) and suicide decedents (n = 22). In addition, the transcriptomic signature in iPSC-derived neuronal precursor cells (NPCs) and neurons were also investigated in controls (n = 2) and suicide decedents (n = 2). RESULTS The hippocampal tissue transcriptomic data revealed that NPAS4 gene expression was downregulated while ALDH1A2, NAAA, and MLXIPL gene expressions were upregulated in hippocampal tissue of suicide decedents. The gene ontology identified 29 significant pathways including NPAS4-associated gene ontology terms "excitatory post-synaptic potential", "regulation of postsynaptic membrane potential" and "long-term memory" indicating alteration of glutamatergic synapses in the hippocampus of suicide decedents. The cell type deconvolution identified decreased excitatory neuron proportion and an increased inhibitory neuron proportion providing evidence of excitation/inhibition imbalance in the hippocampus of suicide decedents. In addition, suicide decedents had increased dendric spine density in the hippocampus, due to an increase of thin (relatively unstable) dendritic spines, compared to controls. The transcriptomes of iPSC-derived hippocampal-like NPCs and neurons revealed 31 and 33 differentially expressed genes in NPC and neurons, respectively, of suicide decedents. CONCLUSIONS Our findings will provide new insights into the hippocampal neuropathology of suicide.
Collapse
Affiliation(s)
- Sujan C Das
- Functional Genomics Laboratory, Department of Psychiatry & Human Behavior, University of California, Irvine, CA, USA
| | | | - William B Callor
- Office of Medical Examiner, Utah Department of Health and Human Services, Salt Lake City, UT, USA
| | - Leslie Jerominski
- Department of Psychiatry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Mitradas M Panicker
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, USA
| | - Erik D Christensen
- Office of Medical Examiner, Utah Department of Health and Human Services, Salt Lake City, UT, USA
| | - William E Bunney
- Department of Psychiatry & Human Behavior, University of California, Irvine, CA, USA
| | - Megan E Williams
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, UT, USA
| | - Hilary Coon
- Department of Psychiatry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Marquis P Vawter
- Functional Genomics Laboratory, Department of Psychiatry & Human Behavior, University of California, Irvine, CA, USA.
| |
Collapse
|
43
|
Liu R, Weng G, Zheng F, Chen J, Wang K, Han J, Huang J, Yan L, Jin J. Generation of an integration-free induced pluripotent stem cell line, FJMAi001-A, from a Marfan syndrome patient with a heterozygous mutation c.2777G > A (p.Cys926Tyr) in FBN1. Stem Cell Res 2024; 81:103591. [PMID: 39515109 DOI: 10.1016/j.scr.2024.103591] [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: 06/07/2024] [Revised: 09/19/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
Marfan syndrome (MFS) is a heritable dominant disorder of fibrous connective tissue, caused by mutations in the gene encoding fibrillin-1 on chromosome 15. Here, we report an induced pluripotent stem cell (iPSC) line generated from a patient with MFS who carries a heterozygous mutation of c.2777G > A(p.Cys926Tyr) in the FBN1 gene using an episomal method. The hiPS-MFS cell line has normal karyotype, expresses pluripotency markers, and has the ability to form three germ layers in vivo.This MFS-specific iPSC line can be used as a cell disease model to study the pathogenesis of Marfan syndrome.
Collapse
Affiliation(s)
- Ronghua Liu
- Fujian Key Laboratory of Medical Analysis, Fujian Academy of Medical Sciences, Fuzhou 350001, China
| | - Guoxing Weng
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China.
| | - Fuzhen Zheng
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China
| | - Jinyan Chen
- Fujian Key Laboratory of Medical Analysis, Fujian Academy of Medical Sciences, Fuzhou 350001, China
| | - Kun Wang
- Fujian Key Laboratory of Medical Analysis, Fujian Academy of Medical Sciences, Fuzhou 350001, China
| | - Junyong Han
- Fujian Key Laboratory of Medical Analysis, Fujian Academy of Medical Sciences, Fuzhou 350001, China
| | - Jie Huang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China
| | - Licheng Yan
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China
| | - Jingjun Jin
- Fujian Key Laboratory of Medical Analysis, Fujian Academy of Medical Sciences, Fuzhou 350001, China.
| |
Collapse
|
44
|
Tohari M, Sanjaya R, Yuliana Sari S, Tedjobagaskara B, Ibnu Faisal A, Alvin Prawira M, Oktaviani Dwi Putri A, Faza N, Murti H, Widyastuti HP. Generation of footprint-free human induced pluripotent stem cell line (SCIKFi001-B) from cGMP grade umbilical cord-derived mesenchymal stem cells (UC-MSCs) using episomal-plasmid based reprogramming approach. Stem Cell Res 2024; 81:103566. [PMID: 39332133 DOI: 10.1016/j.scr.2024.103566] [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: 01/10/2024] [Revised: 08/26/2024] [Accepted: 09/19/2024] [Indexed: 09/29/2024] Open
Abstract
UCMSCs were reprogrammed to iPSCs using Yamanaka factor bearing episomal plasmids. SCIKFi001-B exhibited pluripotency, had typical iPSC morphology and didn't retain any residual episomal plasmid. Although karyotyping showed chromosomal translocation, this abnormality seemed to have little impact on the functionality of SCIKFi001-B since it retained its ability to differentiate to three-germ layer. While karyotypic abnormality might negate use in therapeutic and clinical settings, this line remained a valuable educational tool for iPS cell culture techniques. Finally, our study highlighted the importance of routine karyotyping on iPSC lines as abnormal karyotypes oftentimes bear no discernible effect on cell morphology nor functionality.
Collapse
Affiliation(s)
- Marchella Tohari
- Department of Stem Cell Research and Development, Stem Cell and Cancer Institute, PT. Kalbe Farma Tbk., Jakarta, Indonesia
| | - Ricky Sanjaya
- Department of Stem Cell Research and Development, Stem Cell and Cancer Institute, PT. Kalbe Farma Tbk., Jakarta, Indonesia
| | - Siska Yuliana Sari
- Department of Stem Cell Research and Development, Stem Cell and Cancer Institute, PT. Kalbe Farma Tbk., Jakarta, Indonesia
| | - Bintang Tedjobagaskara
- Department of Stem Cell Research and Development, Stem Cell and Cancer Institute, PT. Kalbe Farma Tbk., Jakarta, Indonesia
| | - Andrian Ibnu Faisal
- Department of Stem Cell Research and Development, Stem Cell and Cancer Institute, PT. Kalbe Farma Tbk., Jakarta, Indonesia
| | - Matheus Alvin Prawira
- Department of Stem Cell Research and Development, Stem Cell and Cancer Institute, PT. Kalbe Farma Tbk., Jakarta, Indonesia; ReGeniC Laboratory, PT. Bifarma Adiluhung, Jakarta, Indonesia
| | - Anggia Oktaviani Dwi Putri
- Department of Stem Cell Research and Development, Stem Cell and Cancer Institute, PT. Kalbe Farma Tbk., Jakarta, Indonesia
| | - Naufalia Faza
- Department of Stem Cell Research and Development, Stem Cell and Cancer Institute, PT. Kalbe Farma Tbk., Jakarta, Indonesia
| | - Harry Murti
- Department of Stem Cell Research and Development, Stem Cell and Cancer Institute, PT. Kalbe Farma Tbk., Jakarta, Indonesia; ReGeniC Laboratory, PT. Bifarma Adiluhung, Jakarta, Indonesia
| | - Halida P Widyastuti
- Department of Stem Cell Research and Development, Stem Cell and Cancer Institute, PT. Kalbe Farma Tbk., Jakarta, Indonesia.
| |
Collapse
|
45
|
Wongkidakarn S, Yodtup C, Jantapalaboon D, Phairoh P, Suparak S, Dhepakson P, Tubsuwan A, Bumroongthai K. Generation of human induced pluripotent stem cell (DMSCi001-A) line from hematopoietic stem cells of a healthy female donor. Stem Cell Res 2024; 81:103605. [PMID: 39504763 DOI: 10.1016/j.scr.2024.103605] [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: 07/16/2024] [Revised: 10/22/2024] [Accepted: 10/27/2024] [Indexed: 11/08/2024] Open
Abstract
Hematopoietic stem cell isolated from a healthy 39-year-old woman were successfully reprogrammed and transformed into induced pluripotent stem cell (iPSCs) by using the integration-free episomal vector included OCT3/4/shp53, Sox2/KLF4, L-MYC/LIN28 and EBNA-1 reprogramming factors. The transformed iPSC lines were cultured and expanded under feeder-free condition. They demonstrated the normal karyotype, expressed pluripotency markers and differentiated into cells derived from the three germ layers. These iPSCs are capable of differentiating into numerous cell subtypes for the purposes of drug discovery and mechanism investigation.
Collapse
Affiliation(s)
- Sudarat Wongkidakarn
- Medical Life Sciences Institute, Department of Medical Sciences, Ministry of Public Health, Nonthaburi, Thailand
| | - Chonlada Yodtup
- Medical Life Sciences Institute, Department of Medical Sciences, Ministry of Public Health, Nonthaburi, Thailand
| | - Danai Jantapalaboon
- Medical Life Sciences Institute, Department of Medical Sciences, Ministry of Public Health, Nonthaburi, Thailand
| | - Panapat Phairoh
- Medical Life Sciences Institute, Department of Medical Sciences, Ministry of Public Health, Nonthaburi, Thailand
| | - Supaporn Suparak
- Medical Life Sciences Institute, Department of Medical Sciences, Ministry of Public Health, Nonthaburi, Thailand
| | - Panadda Dhepakson
- Medical Life Sciences Institute, Department of Medical Sciences, Ministry of Public Health, Nonthaburi, Thailand
| | - Alisa Tubsuwan
- Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| | - Kobkaew Bumroongthai
- Medical Life Sciences Institute, Department of Medical Sciences, Ministry of Public Health, Nonthaburi, Thailand.
| |
Collapse
|
46
|
Gao P, Kajiya M, Motoike S, Ikeya M, Yang J. Application of mesenchymal stem/stromal cells in periodontal regeneration: Opportunities and challenges. JAPANESE DENTAL SCIENCE REVIEW 2024; 60:95-108. [PMID: 38314143 PMCID: PMC10837070 DOI: 10.1016/j.jdsr.2024.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 12/06/2023] [Accepted: 01/15/2024] [Indexed: 02/06/2024] Open
Abstract
Guided tissue regeneration (GTR) has been widely used in the periodontal treatment of intrabony and furcation defects for nearly four decades. The treatment outcomes have shown effectiveness in reducing pocket depth, improving attachment gain and bone filling in periodontal tissue. Although applying GTR could reconstruct the periodontal tissue, the surgical indications are relatively narrow, and some complications and race ethic problems bring new challenges. Therefore, it is challenging to achieve a consensus concerning the clinical benefits of GTR. With the appearance of stem cell-based regenerative medicine, mesenchymal stem/stromal cells (MSCs) have been considered a promising cell resource for periodontal regeneration. In this review, we highlight preclinical and clinical periodontal regeneration using MSCs derived from distinct origins, including non-odontogenic and odontogenic tissues and induced pluripotent stem cells, and discuss the transplantation procedures, therapeutic mechanisms, and concerns to evaluate the effectiveness of MSCs.
Collapse
Affiliation(s)
- Pan Gao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of General Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Mikihito Kajiya
- Department of Periodontal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Souta Motoike
- Department of Periodontal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Makoto Ikeya
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Jingmei Yang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| |
Collapse
|
47
|
Kang Y, Liang S, Gao S, Chen J. Generation of a hiPSC line (TONGJIi001-A) from a 46,XX,ins(1;15)(p13.3; q22.31q26.1),inv(2)(p22.1p16.3),t(2;14)(q34;q12) infertility patient. Stem Cell Res 2024; 81:103561. [PMID: 39299133 DOI: 10.1016/j.scr.2024.103561] [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: 05/20/2024] [Revised: 09/08/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024] Open
Abstract
We have successfully derived a hiPSC line from PBMCs obtained from a 41-year-old infertile female. The patient's karyotype, as determined by Bionano OGM, revealed complex chromosomal rearrangements, including 46,XX,ins(1;15)(p13.3;q22.31q26.1),inv(2)(p22.1p16.3),t(2;14)(q34;q12). Specifically, the episomal plasmids encoding key reprogramming factors OCT4, sh-p53, SOX2, KLF4, L-MYC, and LIN28 were applied to generate the integration-free hiPSC line, which was designated as TONGJIi001-A. This line exhibits typical hiPSC morphology, expresses core pluripotency markers and presents the ability to differentiate into all three germ layers in vitro. Collectively, hiPSC TONGJIi001-A provides a valuable resource for investigating the mechanisms underlying chromosomal structural abnormalities associated with infertility.
Collapse
Affiliation(s)
- Yunzhe Kang
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; Frontier Science Center for Stem Cell Research, Tongji University, Shanghai 200092, China
| | - Shanshan Liang
- Department of Assisted Reproductive Medicine, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 201204, China.
| | - Shaorong Gao
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; Frontier Science Center for Stem Cell Research, Tongji University, Shanghai 200092, China.
| | - Jiayu Chen
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; Frontier Science Center for Stem Cell Research, Tongji University, Shanghai 200092, China.
| |
Collapse
|
48
|
Wang Y, Liu Z, Li Y, Nie Z, Xu B, Zhu Y, Duan S, Chen X, Tan H, Dang J, Guan M, Guo Y. A Novel Mutation Located in the N-Terminal Domain of MYO15A Caused Sensorineural Hearing Loss. Mol Genet Genomic Med 2024; 12:e70042. [PMID: 39620501 PMCID: PMC11609997 DOI: 10.1002/mgg3.70042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 11/07/2024] [Accepted: 11/12/2024] [Indexed: 04/06/2025] Open
Abstract
BACKGROUND MYO15A is one of the common genes of severe-to-profound sensorineural deafness. Mutations in this gene can cause both pre- and post-lingual hearing losses. In this study, a novel MYO15A variant (c.2482C>T) was identified to be associated with autosomal recessive non-syndromic hearing loss (ARNSHL) in a Chinese Uighur family. METHODS To examine the effects of the MYO15A mutation on the morphology and function of the derived hair cell-like cells, two iPSCs were generated separately from the proband and a mutation-negative family member and those were then induced to hair cell-like cells. RESULTS Results showed that this homozygous MYO15A mutation (PVS1 + PM2 + PP1 + PP3), which is located in the N-terminal domain, displayed significant differences in the morphology and function of hair cell-like cells between the proband and the normal control, although it had no effect on the totipotency of iPSCs. CONCLUSION Our study demonstrates that the novel variant c.2482C>T in the MYO15A gene may cause inner ear hair cell dysfunction and audiological disorders in this family.
Collapse
Affiliation(s)
- Yanli Wang
- Department of Otolaryngology—Head and Neck SurgeryLanzhou University Second HospitalLanzhouGansuChina
| | - Zengping Liu
- Department of Otolaryngology—Head and Neck SurgeryLanzhou University Second HospitalLanzhouGansuChina
| | - Yong Li
- Department of Otolaryngology—Head and Neck SurgeryLanzhou University Second HospitalLanzhouGansuChina
| | - Zhipeng Nie
- Institute of GeneticsZhejiang University School of MedicineZhejiangHangzhouChina
| | - Baicheng Xu
- Department of Otolaryngology—Head and Neck SurgeryLanzhou University Second HospitalLanzhouGansuChina
| | - Yiming Zhu
- Department of Otolaryngology—Head and Neck SurgeryLanzhou University Second HospitalLanzhouGansuChina
| | - Shihong Duan
- Department of Otolaryngology—Head and Neck SurgeryLanzhou University Second HospitalLanzhouGansuChina
| | - Xingjian Chen
- Department of Otolaryngology—Head and Neck SurgeryLanzhou University Second HospitalLanzhouGansuChina
| | - Huan Tan
- Department of Otolaryngology—Head and Neck SurgeryLanzhou University Second HospitalLanzhouGansuChina
| | - Jiong Dang
- Department of Otolaryngology—Head and Neck SurgeryLanzhou University Second HospitalLanzhouGansuChina
| | - Minxin Guan
- Institute of GeneticsZhejiang University School of MedicineZhejiangHangzhouChina
| | - Yufen Guo
- Department of Otolaryngology—Head and Neck SurgeryLanzhou University Second HospitalLanzhouGansuChina
| |
Collapse
|
49
|
McComish SF, O'Sullivan J, Copas AMM, Imiolek M, Boyle NT, Crompton LA, Lane JD, Caldwell MA. Reactive astrocytes generated from human iPSC are pro-inflammatory and display altered metabolism. Exp Neurol 2024; 382:114979. [PMID: 39357593 DOI: 10.1016/j.expneurol.2024.114979] [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: 06/21/2024] [Revised: 09/21/2024] [Accepted: 09/27/2024] [Indexed: 10/04/2024]
Abstract
Astrocytes are the most abundant type of glial cell in the central nervous system and they play pivotal roles in both normal health and disease. Their dysfunction is detrimental to many brain related pathologies. Under pathological conditions, such as Alzheimer's disease, astrocytes adopt an activated reactive phenotype which can contribute to disease progression. A prominent risk factor for many neurodegenerative diseases is neuroinflammation which is the purview of glial cells, such as astrocytes and microglia. Human in vitro models have the potential to reveal relevant disease specific mechanisms, through the study of individual cell types such as astrocytes or the addition of specific factors, such as those secreted by microglia. The aim of this study was to generate human cortical astrocytes, in order to assess their protein and gene expression, examine their reactivity profile in response to exposure to the microglial secreted factors IL-1α, TNFα and C1q and assess their functionality in terms of calcium signalling and metabolism. The successfully differentiated and stimulated reactive astrocytes display increased IL-6, RANTES and GM-CSF secretion, and increased expression of genes associated with reactivity including, IL-6, ICAM1, LCN2, C3 and SERPINA3. Functional assessment of these reactive astrocytes showed a delayed and sustained calcium response to ATP and a concomitant decrease in the expression of connexin-43. Furthermore, it was demonstrated these astrocytes had an increased glycolytic capacity with no effect on oxidative phosphorylation. These findings not only increase our understanding of astrocyte reactivity but also provides a functional platform for drug discovery.
Collapse
Affiliation(s)
- Sarah F McComish
- Discipline of Physiology & School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Julia O'Sullivan
- Discipline of Physiology & School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Adina Mac Mahon Copas
- Discipline of Physiology & School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Magdalena Imiolek
- Discipline of Physiology & School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Noreen T Boyle
- Discipline of Physiology & School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Lucy A Crompton
- Regenerative Medicine Laboratory, School of Clinical Sciences, University of Bristol, Bristol, UK; Cell Biology Laboratories, School of Biochemistry, University of Bristol, Bristol, UK
| | - Jon D Lane
- Cell Biology Laboratories, School of Biochemistry, University of Bristol, Bristol, UK
| | - Maeve A Caldwell
- Discipline of Physiology & School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
50
|
Mengnan W, Yan C, Qiong X, Man X. Generation of a human induced pluripotent stem cell line (FDIBSi001-A) from a patient with ADNP syndrome carrying ADNP mutation (c. 2059 T>C). Stem Cell Res 2024; 81:103550. [PMID: 39307104 DOI: 10.1016/j.scr.2024.103550] [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: 04/05/2024] [Revised: 08/03/2024] [Accepted: 09/04/2024] [Indexed: 12/15/2024] Open
Abstract
ADNP syndrome is a neurodevelopmental disorder characterized by autism, intellectual disability, and other physical and behavioral health manifestations. Mutations in ADNP gene is responsible for ADNP syndrome. A human iPSC line with a de novo heterozygous ADNP mutation (ADNP c. 2059 T>C) was generated from peripheral blood mononuclear cells of a patient with ADNP syndrome. This iPSC line showed typical human embryonic stem cell-like morphology, normal karyotype, pluripotency, and ability to differentiate into three germ layers. This iPSC line provides a useful resource to study the pathogenesis and drug screening of ADNP syndrome.
Collapse
Affiliation(s)
- Wu Mengnan
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai 200032, China
| | - Cheng Yan
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Xu Qiong
- Department of Child Health Care, Children's Hospital of Fudan University, Shanghai 200032, China.
| | - Xiong Man
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China.
| |
Collapse
|