1
|
Chen Z, Li XJ. Targeting cholesterol trafficking to mitigate axonal degeneration in hereditary spastic paraplegia. Neural Regen Res 2025; 20:1397-1398. [PMID: 39075901 PMCID: PMC11624880 DOI: 10.4103/nrr.nrr-d-24-00138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/18/2024] [Accepted: 05/07/2024] [Indexed: 07/31/2024] Open
Affiliation(s)
- Zhenyu Chen
- Department of Biomedical Sciences, University of Illinois College of Medicine Rockford, Rockford, IL, USA; Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, USA
| | - Xue-Jun Li
- Department of Biomedical Sciences, University of Illinois College of Medicine Rockford, Rockford, IL, USA; Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
2
|
Verma I, Seshagiri PB. Current Applications of Human Pluripotent Stem Cells in Neuroscience Research and Cell Transplantation Therapy for Neurological Disorders. Stem Cell Rev Rep 2025:10.1007/s12015-025-10851-6. [PMID: 40186708 DOI: 10.1007/s12015-025-10851-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/05/2025] [Indexed: 04/07/2025]
Abstract
Many neurological diseases involving tissue damage cannot be treated with drug-based approaches, and the inaccessibility of human brain samples further hampers the study of these diseases. Human pluripotent stem cells (PSCs), including embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), provide an excellent model for studying neural development and function. PSCs can be differentiated into various neural cell types, providing a renewal source of functional human brain cells. Therefore, PSC-derived neural cells are increasingly used for multiple applications, including neurodevelopmental and neurotoxicological studies, neurological disease modeling, drug screening, and regenerative medicine. In addition, the neural cells generated from patient iPSCs can be used to study patient-specific disease signatures and progression. With the recent advances in genome editing technologies, it is possible to remove the disease-related mutations in the patient iPSCs to generate corrected iPSCs. The corrected iPSCs can differentiate into neural cells with normal physiological functions, which can be used for autologous transplantation. This review highlights the current progress in using PSCs to understand the fundamental principles of human neurodevelopment and dissect the molecular mechanisms of neurological diseases. This knowledge can be applied to develop better drugs and explore cell therapy options. We also discuss the basic requirements for developing cell transplantation therapies for neurological disorders and the current status of the ongoing clinical trials.
Collapse
Affiliation(s)
- Isha Verma
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, 560012, India.
- Department of Neurology, University of Michigan, Ann Arbor, 48109, USA.
| | - Polani B Seshagiri
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, 560012, India
| |
Collapse
|
3
|
Ramakrishnan S, Mohan N, Dong Z, Liu M, Qiang L. Unraveling Isoform Complexity: The Roles of M1- and M87-Spastin in Spastic Paraplegia 4 (SPG4). Mov Disord 2025; 40:420-430. [PMID: 39614608 PMCID: PMC11928279 DOI: 10.1002/mds.30072] [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: 08/26/2024] [Revised: 10/30/2024] [Accepted: 11/12/2024] [Indexed: 12/01/2024] Open
Abstract
Spastic Paraplegia 4 (SPG4) is a debilitating neurodegenerative disorder characterized by progressive muscle weakness and spasticity in the lower limbs, often leading to gait impairment. Central to SPG4 pathology is the die-back degeneration of corticospinal tracts, primarily driven by mutations in the spastin protein encoded by the SPAST gene. SPAST gives rise to two major spastin isoforms, M1- and M87-spastin, which are generated from distinct translation initiation sites. Although spastin is implicated in various cellular functions, the specific roles of each isoform in the pathogenesis of SPG4 remain poorly understood. This review offers an overview of the genetic and structural organization of the M1- and M87-spastin isoforms, highlighting their distinct and overlapping functions, and exploring their potential roles in the haploinsufficiency and gain-of-toxicity mechanisms underlying SPG4. We also present a novel perspective on the evolutionary emergence of M1-spastin and its potential unique involvement in the pathogenesis of SPG4. Drawing upon the latest research, we propose an intriguing hypothesis regarding the hetero-oligomerization of M1- and M87-spastin, exploring how their interaction may drive disease progression and open new avenues for therapeutic intervention. By integrating the current research with these fresh insights, we seek to illuminate the complex molecular mechanisms driving SPG4 and foster the development of innovative therapeutic strategies. This review not only incorporates existing knowledge but also lays the groundwork for future studies aimed at uncovering the isoform-specific roles of spastin in SPG4, with the ultimate goal of advancing targeted treatments for this challenging neurodegenerative disorder. © 2024 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Skandha Ramakrishnan
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129
| | - Neha Mohan
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129
| | - Zhangji Dong
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, China
| | - Mei Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, China
| | - Liang Qiang
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129
| |
Collapse
|
4
|
Deng K, Ruan H, Yu F, Pei Z, Xu C, Zhang S. Dominant negative effect as a novel mechanism of SPAST gene mutation in a large family with hereditary spastic paraplegia. Genes Dis 2024; 11:101152. [PMID: 38882014 PMCID: PMC11176630 DOI: 10.1016/j.gendis.2023.101152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 09/16/2023] [Indexed: 06/18/2024] Open
Affiliation(s)
- Ke Deng
- Shanghai Ji Ai Genetics & IVF Institute, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200000, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200000, China
| | - Haibo Ruan
- The First People's Hospital of Wenling, Wenling, Zhejiang 317500, China
| | - Feifei Yu
- Rushan Hospital of Traditional Chinese Medicine, Rushan, Shandong 264599, China
| | - Zhenle Pei
- Shanghai Ji Ai Genetics & IVF Institute, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200000, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200000, China
| | - Congjian Xu
- Shanghai Ji Ai Genetics & IVF Institute, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200000, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200000, China
- Department of Obstetrics and Gynecology, Fudan University, Shanghai 200000, China
| | - Shuo Zhang
- Shanghai Ji Ai Genetics & IVF Institute, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200000, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200000, China
| |
Collapse
|
5
|
Lin X, Jiang JY, Hong DJ, Lin KJ, Li JJ, Chen YJ, Qiu YS, Wang Z, Liao YC, Yang K, Shi Y, Wang MW, Hsu SL, Hong S, Zeng YH, Chen XC, Wang N, Lee YC, Chen WJ. Biallelic COQ4 Variants in Hereditary Spastic Paraplegia: Clinical and Molecular Characterization. Mov Disord 2024; 39:152-163. [PMID: 38014483 DOI: 10.1002/mds.29664] [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: 01/06/2023] [Revised: 10/31/2023] [Accepted: 11/07/2023] [Indexed: 11/29/2023] Open
Abstract
BACKGROUND Hereditary spastic paraplegias (HSP) are neurologic disorders characterized by progressive lower-extremity spasticity. Despite the identification of several HSP-related genes, many patients lack a genetic diagnosis. OBJECTIVES The aims were to confirm the pathogenic role of biallelic COQ4 mutations in HSP and elucidate the clinical, genetic, and functional molecular features of COQ4-associated HSP. METHODS Whole exome sequences of 310 index patients with HSP of unknown cause from three distinct populations were analyzed to identify potential HSP causal genes. Clinical data obtained from patients harboring candidate causal mutations were examined. Functional characterization of COQ4 variants was performed using bioinformatic tools, single-cell RNA sequencing, biochemical assays in cell lines, primary fibroblasts, induced pluripotent stem cell-derived pyramidal neurons, and zebrafish. RESULTS Compound heterozygous variants in COQ4, which cosegregated with HSP in pedigrees, were identified in 7 patients from six unrelated families. Patients from four of the six families presented with pure HSP, whereas probands of the other two families exhibited complicated HSP with epilepsy or with cerebellar ataxia. In patient-derived fibroblasts and COQ4 knockout complementation lines, stable expression of these missense variants exerted loss-of-function effects, including mitochondrial reactive oxygen species accumulation, decreased mitochondrial membrane potential, and lower ubiquinone biosynthesis. Whereas differentiated pyramidal neurons expressed high COQ4 levels, coq4 knockdown zebrafish displayed severe motor dysfunction, reflecting motor neuron dysregulation. CONCLUSIONS Our study confirms that loss-of-function, compound heterozygous, pathogenic COQ4 variants are causal for autosomal recessive pure and complicated HSP. Moreover, reduced COQ4 levels attributable to variants correspond with decreased ubiquinone biosynthesis, impaired mitochondrial function, and higher phenotypic disease severity. © 2023 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Xiang Lin
- Department of Neurology, Department of Rare Diseases, Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
| | - Jun-Yi Jiang
- Department of Neurology, Department of Rare Diseases, Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
| | - Dao-Jun Hong
- Department of Neurology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Kai-Jun Lin
- Department of Neurology, Department of Rare Diseases, Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
| | - Jin-Jing Li
- Department of Neurology, Department of Rare Diseases, Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
| | - Yi-Jun Chen
- Department of Neurology, Department of Rare Diseases, Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
| | - Yu-Sen Qiu
- Department of Neurology, Department of Rare Diseases, Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
| | - Zishuai Wang
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Yi-Chu Liao
- Department of Neurology, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Neurology, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Kang Yang
- Department of Neurology, Department of Rare Diseases, Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
| | - Yan Shi
- Department of Neurology, Department of Rare Diseases, Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
| | - Meng-Wen Wang
- Department of Neurology, Department of Rare Diseases, Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
| | - Shao-Lun Hsu
- Department of Neurology, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Neurology, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Shunyan Hong
- Department of Neurology, Department of Rare Diseases, Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
| | - Yi-Heng Zeng
- Department of Neurology, Department of Rare Diseases, Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
| | - Xiao-Chun Chen
- Fujian Key Laboratory of Molecular Neurology, Institute of Neuroscience, Fujian Medical University, Fuzhou, China
| | - Ning Wang
- Department of Neurology, Department of Rare Diseases, Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
| | - Yi-Chung Lee
- Department of Neurology, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Neurology, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Wan-Jin Chen
- Department of Neurology, Department of Rare Diseases, Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
| |
Collapse
|
6
|
Wang J, Wu Y, Dong H, Ji Y, Zhang L, Liu Y, Liu Y, Gao X, Jia Y, Wang X. A novel truncated variant in SPAST results in spastin accumulation and defects in microtubule dynamics. BMC Med Genomics 2023; 16:321. [PMID: 38066582 PMCID: PMC10704811 DOI: 10.1186/s12920-023-01759-6] [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: 06/05/2023] [Accepted: 12/02/2023] [Indexed: 12/18/2023] Open
Abstract
OBJECTIVE Haploinsufficiency is widely accepted as the pathogenic mechanism of hereditary spastic paraplegias type 4 (SPG4). However, there are some cases that cannot be explained by reduced function of the spastin protein encoded by SPAST. The aim of this study was to identify the causative variant of SPG4 in a large Chinese family and explore its pathological mechanism. MATERIALS AND METHODS A five-generation family with 49 members including nine affected (4 males and 5 females) and 40 unaffected individuals in Mongolian nationality was recruited. Whole exome sequencing was employed to investigate the genetic etiology. Western blotting and immunofluorescence were used to analyze the effects of the mutant proteins in vitro. RESULTS A novel frameshift variant NM_014946.4: c.483_484delinsC (p.Val162Leufs*2) was identified in SPAST from a pedigree with SPG4. The variant segregated with the disease in the family and thus determined as the disease-causing variant. The c.483_484delinsC variant produced two truncated mutants (mutant M1 and M87 isoforms). They accumulated to a higher level and presented increased stability than their wild-type counterparts and may lost the microtubule severing activity. CONCLUSION SPAST mutations leading to premature stop codons do not always act through haploinsufficiency. The potential toxicity to the corticospinal tract caused by the intracellular accumulation of truncated spastin should be considered as the pathological mechanism of SPG4.
Collapse
Affiliation(s)
- Jie Wang
- Department of Genetics, Inner Mongolia Maternity and Child Health Care Hospital, Hohhot, 010020, China
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock (RRBGL), Inner Mongolia University, Hohhot, 010070, China
| | - Yihan Wu
- Department of Family Medicine, Inner Mongolia People's Hospital, Hohhot, 010057, China
| | - Hong Dong
- Department of Genetics, Inner Mongolia Maternity and Child Health Care Hospital, Hohhot, 010020, China
| | - Yunpeng Ji
- Department of Genetics, Inner Mongolia Maternity and Child Health Care Hospital, Hohhot, 010020, China
| | - Lichun Zhang
- Department of Genetics, Inner Mongolia Maternity and Child Health Care Hospital, Hohhot, 010020, China
| | - Yaxian Liu
- Department of Genetics, Inner Mongolia Maternity and Child Health Care Hospital, Hohhot, 010020, China
| | - Yueshi Liu
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock (RRBGL), Inner Mongolia University, Hohhot, 010070, China
| | - Xin Gao
- Department of Pediatrics, Inner Mongolia Maternity and Child Health Care Hospital, Hohhot, 010020, China
| | - Yueqi Jia
- Department of Genetics, Inner Mongolia Maternity and Child Health Care Hospital, Hohhot, 010020, China.
| | - Xiaohua Wang
- Department of Genetics, Inner Mongolia Maternity and Child Health Care Hospital, Hohhot, 010020, China.
| |
Collapse
|
7
|
Lv S, He E, Luo J, Liu Y, Liang W, Xu S, Zhang K, Yang Y, Wang M, Song Y, Wu Y, Cai X. Using Human-Induced Pluripotent Stem Cell Derived Neurons on Microelectrode Arrays to Model Neurological Disease: A Review. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301828. [PMID: 37863819 PMCID: PMC10667858 DOI: 10.1002/advs.202301828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 09/04/2023] [Indexed: 10/22/2023]
Abstract
In situ physiological signals of in vitro neural disease models are essential for studying pathogenesis and drug screening. Currently, an increasing number of in vitro neural disease models are established using human-induced pluripotent stem cell (hiPSC) derived neurons (hiPSC-DNs) to overcome interspecific gene expression differences. Microelectrode arrays (MEAs) can be readily interfaced with two-dimensional (2D), and more recently, three-dimensional (3D) neural stem cell-derived in vitro models of the human brain to monitor their physiological activity in real time. Therefore, MEAs are emerging and useful tools to model neurological disorders and disease in vitro using human iPSCs. This is enabling a real-time window into neuronal signaling at the network scale from patient derived. This paper provides a comprehensive review of MEA's role in analyzing neural disease models established by hiPSC-DNs. It covers the significance of MEA fabrication, surface structure and modification schemes for hiPSC-DNs culturing and signal detection. Additionally, this review discusses advances in the development and use of MEA technology to study in vitro neural disease models, including epilepsy, autism spectrum developmental disorder (ASD), and others established using hiPSC-DNs. The paper also highlights the application of MEAs combined with hiPSC-DNs in detecting in vitro neurotoxic substances. Finally, the future development and outlook of multifunctional and integrated devices for in vitro medical diagnostics and treatment are discussed.
Collapse
Affiliation(s)
- Shiya Lv
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Enhui He
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
- The State Key Lab of Brain‐Machine IntelligenceZhejiang UniversityHangzhou321100China
| | - Jinping Luo
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yaoyao Liu
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Wei Liang
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Shihong Xu
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Kui Zhang
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yan Yang
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Mixia Wang
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yilin Song
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yirong Wu
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Xinxia Cai
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| |
Collapse
|
8
|
Menduti G, Boido M. Recent Advances in High-Content Imaging and Analysis in iPSC-Based Modelling of Neurodegenerative Diseases. Int J Mol Sci 2023; 24:14689. [PMID: 37834135 PMCID: PMC10572296 DOI: 10.3390/ijms241914689] [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: 08/13/2023] [Revised: 09/24/2023] [Accepted: 09/25/2023] [Indexed: 10/15/2023] Open
Abstract
In the field of neurodegenerative pathologies, the platforms for disease modelling based on patient-derived induced pluripotent stem cells (iPSCs) represent a valuable molecular diagnostic/prognostic tool. Indeed, they paved the way for the in vitro recapitulation of the pathological mechanisms underlying neurodegeneration and for characterizing the molecular heterogeneity of disease manifestations, also enabling drug screening approaches for new therapeutic candidates. A major challenge is related to the choice and optimization of the morpho-functional study designs in human iPSC-derived neurons to deeply detail the cell phenotypes as markers of neurodegeneration. In recent years, the specific combination of high-throughput screening with subcellular resolution microscopy for cell-based high-content imaging (HCI) screening allowed in-depth analyses of cell morphology and neurite trafficking in iPSC-derived neuronal cells by using specific cutting-edge microscopes and automated computational assays. The present work aims to describe the main recent protocols and advances achieved with the HCI analysis in iPSC-based modelling of neurodegenerative diseases, highlighting technical and bioinformatics tips and tricks for further uses and research. To this end, microscopy requirements and the latest computational pipelines to analyze imaging data will be explored, while also providing an overview of the available open-source high-throughput automated platforms.
Collapse
Affiliation(s)
- Giovanna Menduti
- Department of Neuroscience “Rita Levi Montalcini”, Neuroscience Institute Cavalieri Ottolenghi, University of Turin, Regione Gonzole 10, Orbassano, 10043 Turin, TO, Italy;
| | | |
Collapse
|
9
|
Wali G, Li Y, Liyanage E, Kumar KR, Day ML, Sue CM. Pharmacological rescue of mitochondrial and neuronal defects in SPG7 hereditary spastic paraplegia patient neurons using high throughput assays. Front Neurosci 2023; 17:1231584. [PMID: 37766787 PMCID: PMC10520970 DOI: 10.3389/fnins.2023.1231584] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023] Open
Abstract
SPG7 is the most common form of autosomal recessive hereditary spastic paraplegia (HSP). There is a lack of HSP-SPG7 human neuronal models to understand the disease mechanism and identify new drug treatments. We generated a human neuronal model of HSP-SPG7 using induced pluripotent stem (iPS) cell technology. We first generated iPS cells from three HSP-SPG7 patients carrying different disease-causing variants and three healthy controls. The iPS cells were differentiated to form neural progenitor cells (NPCs) and then from NPCs to mature cortical neurons. Mitochondrial and neuronal defects were measured using a high throughout imaging and analysis-based assay in live cells. Our results show that compared to control NPCs, patient NPCs had aberrant mitochondrial morphology with increased mitochondrial size and reduced membrane potential. Patient NPCs develop to form mature cortical neurons with amplified mitochondrial morphology and functional defects along with defects in neuron morphology - reduced neurite complexity and length, reduced synaptic gene, protein expression and activity, reduced viability and increased axonal degeneration. Treatment of patient neurons with Bz-423, a mitochondria permeability pore regulator, restored the mitochondrial and neurite morphological defects and mitochondrial membrane potential back to control neuron levels and rescued the low viability and increased degeneration in patient neurons. This study establishes a direct link between mitochondrial and neuronal defects in HSP-SPG7 patient neurons. We present a strategy for testing mitochondrial targeting drugs to rescue neuronal defects in HSP-SPG7 patient neurons.
Collapse
Affiliation(s)
- Gautam Wali
- Neuroscience Research Australia, Sydney, NSW, Australia
- Kolling Institute for Medical Research, University of Sydney, NSW, Australia
| | - Yan Li
- Neuroscience Research Australia, Sydney, NSW, Australia
- Kolling Institute for Medical Research, University of Sydney, NSW, Australia
- University of New South Wales, Sydney, NSW, Australia
| | - Erandhi Liyanage
- Neuroscience Research Australia, Sydney, NSW, Australia
- Kolling Institute for Medical Research, University of Sydney, NSW, Australia
- University of New South Wales, Sydney, NSW, Australia
| | - Kishore R. Kumar
- University of New South Wales, Sydney, NSW, Australia
- Translational Neurogenomics Group, Molecular Medicine Laboratory and Department of Neurology, Concord Repatriation General Hospital, Concord Clinical School, University of Sydney, Concord, NSW, Australia
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Margot L. Day
- School of Medical Science, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Carolyn M. Sue
- Neuroscience Research Australia, Sydney, NSW, Australia
- Kolling Institute for Medical Research, University of Sydney, NSW, Australia
- University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
10
|
Wang J, Bu WT, Zhu MJ, Tang JY, Liu XM. Novel mutation of SPG4 gene in a Chinese family with hereditary spastic paraplegia: A case report. World J Clin Cases 2023; 11:3288-3294. [PMID: 37274038 PMCID: PMC10237142 DOI: 10.12998/wjcc.v11.i14.3288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 03/15/2023] [Accepted: 04/12/2023] [Indexed: 05/16/2023] Open
Abstract
BACKGROUND Hereditary spastic paraplegia (HSP) is a group of neurogenetic diseases of the corticospinal tract, accompanied by distinct spasticity and weakness of the lower extremities. Mutations in the spastic paraplegia type 4 (SPG4) gene, encoding the spastin protein, are the major cause of the disease. This study reported a Chinese family with HSP caused by a novel mutation of the SPG4 gene.
CASE SUMMARY A 44-year-old male was admitted to our hospital for long-term right lower limb weakness, leg stiffness, and unstable walking. His symptoms gradually worsened, while no obvious muscle atrophy in the lower limbs was found. Neurological examinations revealed that the muscle strength of the lower limbs was normal, and knee reflex hyperreflexia and bilateral positive Babinski signs were detected. Members of his family also had the same symptoms. Using mutation analysis, a novel heterozygous duplication mutation, c.1053dupA, p. (Gln352Thrfs*15), was identified in the SPG4 gene in this family.
CONCLUSION A Chinese family with HSP had a novel mutation of the SPG4 gene, which is autosomal dominant and inherited as pure HSP. The age of onset, sex distribution, and clinical manifestations of all existing living patients in this family were analyzed. The findings may extend the current knowledge on the existing mutations in the SPG4 gene.
Collapse
Affiliation(s)
- Jie Wang
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University of Traditional Chinese Medicine, Jinan 250014, Shandong Province, China
| | - Wei-Ting Bu
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Weifang Medical University, Jinan 250014, Shandong Province, China
| | - Mei-Jia Zhu
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, Shandong Province, China
| | - Ji-You Tang
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, Shandong Province, China
| | - Xiao-Min Liu
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, Shandong Province, China
| |
Collapse
|
11
|
Mou Y, Nandi G, Mukte S, Chai E, Chen Z, Nielsen JE, Nielsen TT, Criscuolo C, Blackstone C, Fraidakis MJ, Li XJ. Chenodeoxycholic acid rescues axonal degeneration in induced pluripotent stem cell-derived neurons from spastic paraplegia type 5 and cerebrotendinous xanthomatosis patients. Orphanet J Rare Dis 2023; 18:72. [PMID: 37024986 PMCID: PMC10080795 DOI: 10.1186/s13023-023-02666-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 03/11/2023] [Indexed: 04/08/2023] Open
Abstract
BACKGROUND Biallelic mutations in CYP27A1 and CYP7B1, two critical genes regulating cholesterol and bile acid metabolism, cause cerebrotendinous xanthomatosis (CTX) and hereditary spastic paraplegia type 5 (SPG5), respectively. These rare diseases are characterized by progressive degeneration of corticospinal motor neuron axons, yet the underlying pathogenic mechanisms and strategies to mitigate axonal degeneration remain elusive. METHODS To generate induced pluripotent stem cell (iPSC)-based models for CTX and SPG5, we reprogrammed patient skin fibroblasts into iPSCs by transducing fibroblast cells with episomal vectors containing pluripotency factors. These patient-specific iPSCs, as well as control iPSCs, were differentiated into cortical projection neurons (PNs) and examined for biochemical alterations and disease-related phenotypes. RESULTS CTX and SPG5 patient iPSC-derived cortical PNs recapitulated several disease-specific biochemical changes and axonal defects of both diseases. Notably, the bile acid chenodeoxycholic acid (CDCA) effectively mitigated the biochemical alterations and rescued axonal degeneration in patient iPSC-derived neurons. To further examine underlying disease mechanisms, we developed CYP7B1 knockout human embryonic stem cell (hESC) lines using CRISPR-cas9-mediated gene editing and, following differentiation, examined hESC-derived cortical PNs. Knockout of CYP7B1 resulted in similar axonal vesiculation and degeneration in human cortical PN axons, confirming a cause-effect relationship between gene deficiency and axonal degeneration. Interestingly, CYP7B1 deficiency led to impaired neurofilament expression and organization as well as axonal degeneration, which could be rescued with CDCA, establishing a new disease mechanism and therapeutic target to mitigate axonal degeneration. CONCLUSIONS Our data demonstrate disease-specific lipid disturbances and axonopathy mechanisms in human pluripotent stem cell-based neuronal models of CTX and SPG5 and identify CDCA, an established treatment of CTX, as a potential pharmacotherapy for SPG5. We propose this novel treatment strategy to rescue axonal degeneration in SPG5, a currently incurable condition.
Collapse
Affiliation(s)
- Yongchao Mou
- Department of Biomedical Sciences, University of Illinois College of Medicine Rockford, Rockford, IL, 61107, USA
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Ghata Nandi
- Department of Biomedical Sciences, University of Illinois College of Medicine Rockford, Rockford, IL, 61107, USA
| | - Sukhada Mukte
- Department of Biomedical Sciences, University of Illinois College of Medicine Rockford, Rockford, IL, 61107, USA
| | - Eric Chai
- Department of Biomedical Sciences, University of Illinois College of Medicine Rockford, Rockford, IL, 61107, USA
| | - Zhenyu Chen
- Department of Biomedical Sciences, University of Illinois College of Medicine Rockford, Rockford, IL, 61107, USA
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Jorgen E Nielsen
- Neurogenetics Clinic & Research Laboratory, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Troels T Nielsen
- Neurogenetics Clinic & Research Laboratory, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Chiara Criscuolo
- Department of Neuroscience, Reproductive Sciences and Odontostomatology, Federico II University, Naples, Italy
| | - Craig Blackstone
- Movement Disorders Division, Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Charlestown, Boston, MA, 02129, USA
| | - Matthew J Fraidakis
- Rare Neurological Diseases Unit, Department of Neurology, Attikon University Hospital, Medical School of the University of Athens, Athens, Greece
| | - Xue-Jun Li
- Department of Biomedical Sciences, University of Illinois College of Medicine Rockford, Rockford, IL, 61107, USA.
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, 60607, USA.
| |
Collapse
|
12
|
The interconnection of endoplasmic reticulum and microtubule and its implication in Hereditary Spastic Paraplegia. Comput Struct Biotechnol J 2023; 21:1670-1677. [PMID: 36860342 PMCID: PMC9968982 DOI: 10.1016/j.csbj.2023.02.025] [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: 08/15/2022] [Revised: 02/14/2023] [Accepted: 02/14/2023] [Indexed: 02/22/2023] Open
Abstract
The endoplasmic reticulum (ER) and microtubule (MT) network form extensive contact with each other and their interconnection plays a pivotal role in ER maintenance and distribution as well as MT stability. The ER participates in a variety of biological processes including protein folding and processing, lipid biosynthesis, and Ca2+ storage. MTs specifically regulate cellular architecture, provide routes for transport of molecules or organelles, and mediate signaling events. The ER morphology and dynamics are regulated by a class of ER shaping proteins, which also provide the physical contact structure for linking of ER and MT. In addition to these ER-localized and MT-binding proteins, specific motor proteins and adaptor-linking proteins also mediate bidirectional communication between the two structures. In this review, we summarize the current understanding of the structure and function of ER-MT interconnection. We further highlight the morphologic factors which coordinate the ER-MT network and maintain the normal physiological function of neurons, with their defect causing neurodegenerative diseases such as Hereditary Spastic Paraplegia (HSP). These findings promote our understanding of the pathogenesis of HSP and provide important therapeutic targets for treatment of these diseases.
Collapse
|
13
|
Rizo T, Gebhardt L, Riedlberger J, Eberhardt E, Fester L, Alansary D, Winkler J, Turan S, Arnold P, Niemeyer BA, Fischer MJM, Winner B. Store-operated calcium entry is reduced in spastin-linked hereditary spastic paraplegia. Brain 2022; 145:3131-3146. [PMID: 36103408 PMCID: PMC9473359 DOI: 10.1093/brain/awac122] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 03/17/2022] [Accepted: 03/22/2022] [Indexed: 01/04/2023] Open
Abstract
Pathogenic variants in SPAST, the gene coding for spastin, are the single most common cause of hereditary spastic paraplegia, a progressive motor neuron disease. Spastin regulates key cellular functions, including microtubule-severing and endoplasmic reticulum-morphogenesis. However, it remains unclear how alterations in these cellular functions due to SPAST pathogenic variants result in motor neuron dysfunction. Since spastin influences both microtubule network and endoplasmic reticulum structure, we hypothesized that spastin is necessary for the regulation of Ca2+ homeostasis via store-operated calcium entry. Here, we show that the lack of spastin enlarges the endoplasmic reticulum and reduces store-operated calcium entry. In addition, elevated levels of different spastin variants induced clustering of STIM1 within the endoplasmic reticulum, altered the transport of STIM1 to the plasma membrane and reduced store-operated calcium entry, which could be rescued by exogenous expression of STIM1. Importantly, store-operated calcium entry was strongly reduced in induced pluripotent stem cell-derived neurons from hereditary spastic paraplegia patients with pathogenic variants in SPAST resulting in spastin haploinsufficiency. These neurons developed axonal swellings in response to lack of spastin. We were able to rescue both store-operated calcium entry and axonal swellings in SPAST patient neurons by restoring spastin levels, using CRISPR/Cas9 to correct the pathogenic variants in SPAST. These findings demonstrate that proper amounts of spastin are a key regulatory component for store-operated calcium entry mediated Ca2+ homeostasis and suggest store-operated calcium entry as a disease relevant mechanism of spastin-linked motor neuron disease.
Collapse
Affiliation(s)
- Tania Rizo
- Department of Stem Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Lisa Gebhardt
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Julia Riedlberger
- Department of Stem Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Esther Eberhardt
- Present address: Department of Anesthesiology, RWTH Aachen University, 52074 Aachen, Germany
| | - Lars Fester
- Institute of Anatomy and Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Dalia Alansary
- Molecular Biophysics, University of Saarland, Center for Integrative Physiology and Molecular Medicine, 66421 Homburg/Saar, Germany
| | - Jürgen Winkler
- Department of Molecular Neurology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany,Center of Rare Diseases Erlangen (ZSEER), Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Soeren Turan
- Institute of Biochemistry (Emil-Fischer-Center), Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Philipp Arnold
- Institute of Anatomy, Functional and Clinical Anatomy, Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | | | | | - Beate Winner
- Correspondence to: Beate Winner Department of Stem Cell Biology Friedrich-Alexander University Erlangen-Nürnberg Glückstraße 6 91054 Erlangen, Germany E-mail:
| |
Collapse
|
14
|
Gather F, Ihrig-Biedert I, Kohlhas P, Krutenko T, Peitz M, Brüstle O, Pautz A, Kleinert H. A specific, non-immune system-related isoform of the human inducible nitric oxide synthase is expressed during differentiation of human stem cells into various cell types. Cell Commun Signal 2022; 20:47. [PMID: 35392923 PMCID: PMC8991583 DOI: 10.1186/s12964-022-00855-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 03/03/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND NOS2 expression is mostly found in bacteria-exposed or cytokine-treated tissues and is mostly connected to innate immune reactions. There are three isoforms of NOS2 (NOS2-1 to -3). In RNA-seq data sets, analyzing inflammatory gene expression, only expression of the NOS2-1 mRNA isoform is detected. However, the expression of NOS2 in differentiating human pluripotent stems (hPSCs) has not been analyzed yet. METHODS Public available RNA-seq databases were screened for data of hPSCs during differentiation to different target cells. An isoform specific algorithm was used to analyze NOS2 mRNA isoform expression. In addition, we differentiated four different human iPSC cell lines toward cortical neurons and analyzed NOS2 mRNA expression by qRT-PCR and 5'-RACE. The functionality of the NOS2-2 protein was analyzed by transient transfection of expression clones in human DLD1 cells and nitrate measurement in the supernatant of these cells. RESULTS In RNA-seq databases we detected a transient expression of the NOS2 mRNA during the differentiation of hPSCs to cardiomyocytes, chondrocytes, mesenchymal stromal cells, neurons, syncytiotrophoblast cells, and trophoblasts. NOS2 mRNA isoform specific analyses showed, that the transiently expressed NOS2 mRNA in differentiating hPSC (NOS2-2; "diff-iNOS") differ remarkably from the already described NOS2 transcript found in colon or induced islets (NOS2-1; "immuno-iNOS"). Also, analysis of the NOS2 mRNA- and protein expression during the differentiation of four different hiPSC lines towards cortical neurons showed a transient expression of the NOS2 mRNA and NOS2 protein on day 18 of the differentiation course. 5'-RACE experiments and isoform specific qRT-PCR analyses revealed that only the NOS2-2 mRNA isoform was expressed in these experiments. To analyze the functionality of the NOS2-2 protein, we transfected human DLD-1 cells with tetracycline inducible expression clones encoding the NOS2-1- or -2 coding sequence. After induction of the NOS2-1 or -2 mRNA expression by tetracycline a similar nitrate production was measured proofing the functionality of the NOS2-2 protein isoform. CONCLUSIONS Our data show that a differentiation specific NOS2 isoform (NOS2-2) is transiently expressed during differentiation of hPSC. Video Abstract.
Collapse
Affiliation(s)
- Fabian Gather
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany.,Department of Molecular Embryology, Institute for Anatomy and Cell Biology, Freiburg, Germany
| | - Irmgard Ihrig-Biedert
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Paul Kohlhas
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Tamara Krutenko
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty & University Hospital Bonn, Bonn, Germany
| | - Michael Peitz
- Cell Programming Core Facility, Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty & University Hospital Bonn, Bonn, Germany.,Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty & University Hospital Bonn, Bonn, Germany
| | - Oliver Brüstle
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty & University Hospital Bonn, Bonn, Germany
| | - Andrea Pautz
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany.
| | - Hartmut Kleinert
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany.
| |
Collapse
|
15
|
Hauser S, Helm J, Kraft M, Korneck M, Hübener-Schmid J, Schöls L. Allele-specific targeting of mutant ataxin-3 by antisense oligonucleotides in SCA3-iPSC-derived neurons. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 27:99-108. [PMID: 34938609 PMCID: PMC8649108 DOI: 10.1016/j.omtn.2021.11.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 11/16/2021] [Indexed: 12/01/2022]
Abstract
Spinocerebellar ataxia type 3 (SCA3) is caused by an expanded polyglutamine stretch in ataxin-3. While wild-type ataxin-3 has important functions, e.g., as a deubiquitinase, downregulation of mutant ataxin-3 is likely to slow down the course of this fatal disease. We established a screening platform with human neurons of patients and controls derived from induced pluripotent stem cells to test antisense oligonucleotides (ASOs) for their effects on ataxin-3 expression. We identified an ASO that suppressed mutant and wild-type ataxin-3 levels by >90% after a singular treatment. Next, we screened pairs of ASOs designed to selectively target the mutant or the wild-type allele by taking advantage of a SNP (c.987G > C) in ATXN3 that is present in most SCA3 patients. We found ASOmut4 to reduce levels of mutant ataxin-3 by 80% after 10 days while leaving expression of wild-type ataxin-3 largely unaffected. In a long-term study we proved this effect to last for about 4 weeks after a single treatment without signs of neurotoxicity. This study provides proof of principle that allele-specific lowering of poly(Q)-expanded ataxin-3 by selective ASOs is feasible and long lasting, with sparing of wild-type ataxin-3 expression in a human cell culture model that is genetically identical to SCA3 patients.
Collapse
Affiliation(s)
- Stefan Hauser
- German Center for Neurodegenerative Diseases (DZNE), 72076 Tübingen, Germany.,Hertie Institute for Clinical Brain Research, University of Tübingen, 72076 Tübingen, Germany
| | - Jacob Helm
- German Center for Neurodegenerative Diseases (DZNE), 72076 Tübingen, Germany.,Hertie Institute for Clinical Brain Research, University of Tübingen, 72076 Tübingen, Germany.,Graduate School of Cellular and Molecular Neuroscience, University of Tübingen, 72076 Tübingen, Germany
| | - Melanie Kraft
- German Center for Neurodegenerative Diseases (DZNE), 72076 Tübingen, Germany.,Hertie Institute for Clinical Brain Research, University of Tübingen, 72076 Tübingen, Germany
| | - Milena Korneck
- German Center for Neurodegenerative Diseases (DZNE), 72076 Tübingen, Germany.,Hertie Institute for Clinical Brain Research, University of Tübingen, 72076 Tübingen, Germany.,Graduate School of Cellular and Molecular Neuroscience, University of Tübingen, 72076 Tübingen, Germany
| | - Jeannette Hübener-Schmid
- Institute of Medical Genetics and Applied Genomics and Center of Rare Diseases, University of Tübingen, 72076 Tübingen, Germany
| | - Ludger Schöls
- German Center for Neurodegenerative Diseases (DZNE), 72076 Tübingen, Germany.,Hertie Institute for Clinical Brain Research, University of Tübingen, 72076 Tübingen, Germany
| |
Collapse
|
16
|
Johns AE, Maragakis NJ. Exploring Motor Neuron Diseases Using iPSC Platforms. Stem Cells 2022; 40:2-13. [PMID: 35511862 PMCID: PMC9199844 DOI: 10.1093/stmcls/sxab006] [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: 06/18/2021] [Accepted: 09/17/2021] [Indexed: 01/21/2023]
Abstract
The degeneration of motor neurons is a pathological hallmark of motor neuron diseases (MNDs), but emerging evidence suggests that neuronal vulnerability extends well beyond this cell subtype. The ability to assess motor function in the clinic is limited to physical examination, electrophysiological measures, and tissue-based or neuroimaging techniques which lack the resolution to accurately assess neuronal dysfunction as the disease progresses. Spinal muscular atrophy (SMA), spinal and bulbar muscular atrophy (SBMA), hereditary spastic paraplegia (HSP), and amyotrophic lateral sclerosis (ALS) are all MNDs with devastating clinical outcomes that contribute significantly to disease burden as patients are no longer able to carry out normal activities of daily living. The critical need to accurately assess the cause and progression of motor neuron dysfunction, especially in the early stages of those diseases, has motivated the use of human iPSC-derived motor neurons (hiPSC-MN) to study the neurobiological mechanisms underlying disease pathogenesis and to generate platforms for therapeutic discovery and testing. As our understanding of MNDs has grown, so too has our need to develop more complex in vitro models which include hiPSC-MN co-cultured with relevant non-neuronal cells in 2D as well as in 3D organoid and spheroid systems. These more complex hiPSC-derived culture systems have led to the implementation of new technologies, including microfluidics, multielectrode array, and machine learning which offer novel insights into the functional correlates of these emerging model systems.
Collapse
Affiliation(s)
- Alexandra E Johns
- Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | | |
Collapse
|
17
|
Yang JO, Yoon JY, Sung DH, Yun S, Lee JJ, Jun SY, Halder D, Jeon SJ, Woo EJ, Seok JM, Cho JW, Jang JH, Choi JK, Kim BJ, Kim NS. The emerging genetic diversity of hereditary spastic paraplegia in Korean patients. Genomics 2021; 113:4136-4148. [PMID: 34715294 DOI: 10.1016/j.ygeno.2021.10.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 09/09/2021] [Accepted: 10/24/2021] [Indexed: 02/05/2023]
Abstract
Hereditary Spastic Paraplegias (HSP) are a group of rare inherited neurological disorders characterized by progressive loss of corticospinal motor-tract function. Numerous patients with HSP remain undiagnosed despite screening for known genetic causes of HSP. Therefore, identification of novel genetic variations related to HSP is needed. In this study, we identified 88 genetic variants in 54 genes from whole-exome data of 82 clinically well-defined Korean HSP families. Fifty-six percent were known HSP genes, and 44% were composed of putative candidate HSP genes involved in the HSPome and originally reported neuron-related genes, not previously diagnosed in HSP patients. Their inheritance modes were 39, de novo; 33, autosomal dominant; and 10, autosomal recessive. Notably, ALDH18A1 showed the second highest frequency. Fourteen known HSP genes were firstly reported in Koreans, with some of their variants being predictive of HSP-causing protein malfunction. SPAST and REEP1 mutants with unknown function induced neurite abnormality. Further, 54 HSP-related genes were closely linked to the HSP progression-related network. Additionally, the genetic spectrum and variation of known HSP genes differed across ethnic groups. These results expand the genetic spectrum for HSP and may contribute to the accurate diagnosis and treatment for rare HSP.
Collapse
Affiliation(s)
- Jin Ok Yang
- Korea BioInformation Center (KOBIC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea; Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Ji-Yong Yoon
- Rare-disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Duk Hyun Sung
- Department of Physical and Rehabilitation Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sohyun Yun
- Rare-disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Jeong-Ju Lee
- Rare-disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Soo Young Jun
- Rare-disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Debasish Halder
- Rare-disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Su-Jin Jeon
- Rare-disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea; Department of Functional Genomics, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Eui-Jeon Woo
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea; Department of Analytical Bioscience, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Jin Myoung Seok
- Department of Neurology, Soonchunhyang University College of Medicine, Soonchunhyang University Hospital Cheonan, Cheonan, Republic of Korea
| | - Jin Whan Cho
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Ja-Hyun Jang
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jung Kyoon Choi
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.
| | - Byoung Joon Kim
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
| | - Nam-Soon Kim
- Rare-disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea; Department of Functional Genomics, University of Science and Technology (UST), Daejeon, Republic of Korea.
| |
Collapse
|
18
|
ER Morphology in the Pathogenesis of Hereditary Spastic Paraplegia. Cells 2021; 10:cells10112870. [PMID: 34831093 PMCID: PMC8616106 DOI: 10.3390/cells10112870] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/18/2021] [Accepted: 10/20/2021] [Indexed: 12/18/2022] Open
Abstract
The endoplasmic reticulum (ER) is the most abundant and widespread organelle in cells. Its peculiar membrane architecture, formed by an intricate network of tubules and cisternae, is critical to its multifaceted function. Regulation of ER morphology is coordinated by a few ER-specific membrane proteins and is thought to be particularly important in neurons, where organized ER membranes are found even in the most distant neurite terminals. Mutation of ER-shaping proteins has been implicated in the neurodegenerative disease hereditary spastic paraplegia (HSP). In this review we discuss the involvement of these proteins in the pathogenesis of HSP, focusing on the experimental evidence linking their molecular function to disease onset. Although the precise biochemical activity of some ER-related HSP proteins has been elucidated, the pathological mechanism underlying ER-linked HSP is still undetermined and needs to be further investigated.
Collapse
|
19
|
Liu Q, Zhang G, Ji Z, Lin H. Molecular and cellular mechanisms of spastin in neural development and disease (Review). Int J Mol Med 2021; 48:218. [PMID: 34664680 PMCID: PMC8547542 DOI: 10.3892/ijmm.2021.5051] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 09/29/2021] [Indexed: 12/26/2022] Open
Abstract
Spastin is a microtubule (MT)‑severing enzyme identified from mutations of hereditary spastic paraplegia in 1999 and extensive studies indicate its vital role in various cellular activities. In the past two decades, efforts have been made to understand the underlying molecular mechanisms of how spastin is linked to neural development and disease. Recent studies on spastin have unraveled the mechanistic processes of its MT‑severing activity and revealed that spastin acts as an MT amplifier to mediate its remodeling, thus providing valuable insight into the molecular roles of spastin under physiological conditions. In addition, recent research has revealed multiple novel molecular mechanisms of spastin in cellular biological pathways, including endoplasmic reticulum shaping, calcium trafficking, fatty acid trafficking, as well as endosomal fission and trafficking. These processes are closely involved in axonal and dendritic development and maintenance. The current review presents recent biological advances regarding the molecular mechanisms of spastin at the cellular level and provides insight into how it affects neural development and disease.
Collapse
Affiliation(s)
- Qiuling Liu
- Department of Orthopedics, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510630, P.R. China
| | - Guowei Zhang
- Department of Orthopedics, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510630, P.R. China
| | - Zhisheng Ji
- Department of Orthopedics, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510630, P.R. China
| | - Hongsheng Lin
- Department of Orthopedics, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510630, P.R. China
| |
Collapse
|
20
|
Parittotokkaporn S, Dravid A, Raos BJ, Rosset S, Svirskis D, O'Carroll SJ. Stretchable microchannel-on-a-chip: A simple model for evaluating the effects of uniaxial strain on neuronal injury. J Neurosci Methods 2021; 362:109302. [PMID: 34343573 DOI: 10.1016/j.jneumeth.2021.109302] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 07/14/2021] [Accepted: 07/29/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Axonal injury is a major component of traumatic spinal cord injury (SCI), associated with rapid deformation of spinal tissue and axonal projections. In vitro models enable us to examine these effects and screen potential therapies in a controlled, reproducible manner. NEW METHOD A customized, stretchable microchannel system was developed using polydimethylsiloxane microchannels. Cortical and spinal embryonic rat neurons were cultured within the microchannel structures, allowing a uniaxial strain to be applied to isolated axonal processes. Global strains of up to 52% were applied to the stretchable microchannel-on-a-chip platform leading to local strains of up to 12% being experienced by axons isolated in the microchannels. RESULTS Individual axons exposed to local strains between 3.2% and 8.7% developed beading within 30-minutes of injury. At higher local strains of 9.8% and 12% individual axons ruptured within 30-minutes of injury. Axon bundles, or fascicles, were more resistant to rupture at each strain level, compared to individual axons. At lower local strain of 3.2%, axon bundles inside microchannels and neuronal cells near entrances of them progressively swelled and degenerated over a period of 7 days after injury. COMPARISON WITH EXISTING METHOD(S) This method is simple, reliable and reproducible with good control and measurement of injury tolerance and morphological deformations using standard laboratory equipment. By measuring local strains, we observed that axonal injuries occur at a lower strain magnitude and a lower strain rate than previous methods reporting global strains, which may not accurately reflect the true axonal strain. CONCLUSIONS We describe a novel stretchable microchannel-on-a-chip platform to study the effect of varying local strain on morphological characteristics of neuronal injury.
Collapse
Affiliation(s)
- Sam Parittotokkaporn
- Department of Anatomy and Medical Imaging, School of Medical Sciences, Faculty of Medical and Health Sciences and The Centre for Brain Research, University of Auckland, New Zealand
| | - Anusha Dravid
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Brad J Raos
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Samuel Rosset
- Auckland Bioengineering Institute, University of Auckland, New Zealand
| | - Darren Svirskis
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Simon J O'Carroll
- Department of Anatomy and Medical Imaging, School of Medical Sciences, Faculty of Medical and Health Sciences and The Centre for Brain Research, University of Auckland, New Zealand.
| |
Collapse
|
21
|
Gumeni S, Vantaggiato C, Montopoli M, Orso G. Hereditary Spastic Paraplegia and Future Therapeutic Directions: Beneficial Effects of Small Compounds Acting on Cellular Stress. Front Neurosci 2021; 15:660714. [PMID: 34025345 PMCID: PMC8134669 DOI: 10.3389/fnins.2021.660714] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 03/29/2021] [Indexed: 12/29/2022] Open
Abstract
Hereditary spastic paraplegia (HSP) is a group of inherited neurodegenerative conditions that share a characteristic feature of degeneration of the longest axons within the corticospinal tract, which leads to progressive spasticity and weakness of the lower limbs. Mutations of over 70 genes produce defects in various biological pathways: axonal transport, lipid metabolism, endoplasmic reticulum (ER) shaping, mitochondrial function, and endosomal trafficking. HSPs suffer from an adequate therapeutic plan. Currently the treatments foreseen for patients affected by this pathology are physiotherapy, to maintain the outgoing tone, and muscle relaxant therapies for spasticity. Very few clinical studies have been conducted, and it's urgent to implement preclinical animal studies devoted to pharmacological test and screening, to expand the rose of compounds potentially attractive for clinical trials. Small animal models, such as Drosophila melanogaster and zebrafish, have been generated, analyzed, and used as preclinical model for screening of compounds and their effects. In this work, we briefly described the role of HSP-linked proteins in the organization of ER endomembrane system and in the regulation of ER homeostasis and stress as a common pathological mechanism for these HSP forms. We then focused our attention on the pharmacodynamic and pharmacokinetic features of some recently identified molecules with antioxidant property, such as salubrinal, guanabenz, N-acetyl cysteine, methylene blue, rapamycin, and naringenin, and on their potential use in future clinical studies. Expanding the models and the pharmacological screening for HSP disease is necessary to give an opportunity to patients and clinicians to test new molecules.
Collapse
Affiliation(s)
- Sentiljana Gumeni
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Chiara Vantaggiato
- Laboratory of Molecular Biology, Scientific Institute IRCCS Eugenio Medea, Bosisio Parini, Italy
| | - Monica Montopoli
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padua, Italy
| | - Genny Orso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padua, Italy
| |
Collapse
|
22
|
Mackay-Sim A. Hereditary Spastic Paraplegia: From Genes, Cells and Networks to Novel Pathways for Drug Discovery. Brain Sci 2021; 11:brainsci11030403. [PMID: 33810178 PMCID: PMC8004882 DOI: 10.3390/brainsci11030403] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/14/2021] [Accepted: 03/18/2021] [Indexed: 12/13/2022] Open
Abstract
Hereditary spastic paraplegia (HSP) is a diverse group of Mendelian genetic disorders affecting the upper motor neurons, specifically degeneration of their distal axons in the corticospinal tract. Currently, there are 80 genes or genomic loci (genomic regions for which the causative gene has not been identified) associated with HSP diagnosis. HSP is therefore genetically very heterogeneous. Finding treatments for the HSPs is a daunting task: a rare disease made rarer by so many causative genes and many potential mutations in those genes in individual patients. Personalized medicine through genetic correction may be possible, but impractical as a generalized treatment strategy. The ideal treatments would be small molecules that are effective for people with different causative mutations. This requires identification of disease-associated cell dysfunctions shared across genotypes despite the large number of HSP genes that suggest a wide diversity of molecular and cellular mechanisms. This review highlights the shared dysfunctional phenotypes in patient-derived cells from patients with different causative mutations and uses bioinformatic analyses of the HSP genes to identify novel cell functions as potential targets for future drug treatments for multiple genotypes.
Collapse
Affiliation(s)
- Alan Mackay-Sim
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, QLD 4111, Australia
| |
Collapse
|
23
|
Dobrindt K, Zhang H, Das D, Abdollahi S, Prorok T, Ghosh S, Weintraub S, Genovese G, Powell SK, Lund A, Akbarian S, Eggan K, McCarroll S, Duan J, Avramopoulos D, Brennand KJ. Publicly Available hiPSC Lines with Extreme Polygenic Risk Scores for Modeling Schizophrenia. Complex Psychiatry 2021; 6:68-82. [PMID: 34883504 PMCID: PMC7923934 DOI: 10.1159/000512716] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 10/27/2020] [Indexed: 07/23/2023] Open
Abstract
Schizophrenia (SZ) is a common and debilitating psychiatric disorder with limited effective treatment options. Although highly heritable, risk for this polygenic disorder depends on the complex interplay of hundreds of common and rare variants. Translating the growing list of genetic loci significantly associated with disease into medically actionable information remains an important challenge. Thus, establishing platforms with which to validate the impact of risk variants in cell-type-specific and donor-dependent contexts is critical. Towards this, we selected and characterized a collection of 12 human induced pluripotent stem cell (hiPSC) lines derived from control donors with extremely low and high SZ polygenic risk scores (PRS). These hiPSC lines are publicly available at the California Institute for Regenerative Medicine (CIRM). The suitability of these extreme PRS hiPSCs for CRISPR-based isogenic comparisons of neurons and glia was evaluated across 3 independent laboratories, identifying 9 out of 12 meeting our criteria. We report a standardized resource of publicly available hiPSCs on which we hope to perform genome engineering and generate diverse kinds of functional data, with comparisons across studies facilitated by the use of a common set of genetic backgrounds.
Collapse
Affiliation(s)
- Kristina Dobrindt
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomics, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Hanwen Zhang
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, Illinois, USA
| | - Debamitra Das
- Department of Genetic Medicine and Psychiatry, Johns Hopkins University, Baltimore, Maryland, USA
| | - Sara Abdollahi
- Department of Genetic Medicine and Psychiatry, Johns Hopkins University, Baltimore, Maryland, USA
| | - Tim Prorok
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, Illinois, USA
| | - Sulagna Ghosh
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
| | - Sarah Weintraub
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, Illinois, USA
| | - Giulio Genovese
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
| | - Samuel K. Powell
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomics, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Anina Lund
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomics, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Schahram Akbarian
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kevin Eggan
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA
| | - Steven McCarroll
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
| | - Jubao Duan
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, Illinois, USA
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, Illinois, USA
| | - Dimitrios Avramopoulos
- Department of Genetic Medicine and Psychiatry, Johns Hopkins University, Baltimore, Maryland, USA
| | - Kristen J. Brennand
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomics, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
24
|
Cheffer A, Flitsch LJ, Krutenko T, Röderer P, Sokhranyaeva L, Iefremova V, Hajo M, Peitz M, Schwarz MK, Brüstle O. Human stem cell-based models for studying autism spectrum disorder-related neuronal dysfunction. Mol Autism 2020; 11:99. [PMID: 33308283 PMCID: PMC7733257 DOI: 10.1186/s13229-020-00383-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 09/22/2020] [Indexed: 12/13/2022] Open
Abstract
The controlled differentiation of pluripotent stem cells (PSCs) into neurons and glia offers a unique opportunity to study early stages of human central nervous system development under controlled conditions in vitro. With the advent of cell reprogramming and the possibility to generate induced pluripotent stem cells (iPSCs) from any individual in a scalable manner, these studies can be extended to a disease- and patient-specific level. Autism spectrum disorder (ASD) is considered a neurodevelopmental disorder, with substantial evidence pointing to early alterations in neurogenesis and network formation as key pathogenic drivers. For that reason, ASD represents an ideal candidate for stem cell-based disease modeling. Here, we provide a concise review on recent advances in the field of human iPSC-based modeling of syndromic and non-syndromic forms of ASD, with a particular focus on studies addressing neuronal dysfunction and altered connectivity. We further discuss recent efforts to translate stem cell-based disease modeling to 3D via brain organoid and cell transplantation approaches, which enable the investigation of disease mechanisms in a tissue-like context. Finally, we describe advanced tools facilitating the assessment of altered neuronal function, comment on the relevance of iPSC-based models for the assessment of pharmaceutical therapies and outline potential future routes in stem cell-based ASD research.
Collapse
Affiliation(s)
- Arquimedes Cheffer
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty & University Hospital Bonn, Venusberg-Campus 1, Building 76, 53127, Bonn, Germany
| | - Lea Jessica Flitsch
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty & University Hospital Bonn, Venusberg-Campus 1, Building 76, 53127, Bonn, Germany
| | - Tamara Krutenko
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty & University Hospital Bonn, Venusberg-Campus 1, Building 76, 53127, Bonn, Germany
| | - Pascal Röderer
- Life & Brain GmbH, Platform Cellomics, Venusberg-Campus 1, Building 76, 53127, Bonn, Germany
| | - Liubov Sokhranyaeva
- Institute of Experimental Epileptology and Cognition Research, University of Bonn Medical Faculty & University Hospital Bonn, Venusberg-Campus 1, Building 76, 53127, Bonn, Germany
| | - Vira Iefremova
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty & University Hospital Bonn, Venusberg-Campus 1, Building 76, 53127, Bonn, Germany
| | - Mohamad Hajo
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty & University Hospital Bonn, Venusberg-Campus 1, Building 76, 53127, Bonn, Germany
| | - Michael Peitz
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty & University Hospital Bonn, Venusberg-Campus 1, Building 76, 53127, Bonn, Germany
- Life & Brain GmbH, Platform Cellomics, Venusberg-Campus 1, Building 76, 53127, Bonn, Germany
- Cell Programming Core Facility, University of Bonn Medical Faculty, Bonn, Germany
| | - Martin Karl Schwarz
- Life & Brain GmbH, Platform Cellomics, Venusberg-Campus 1, Building 76, 53127, Bonn, Germany
- Institute of Experimental Epileptology and Cognition Research, University of Bonn Medical Faculty & University Hospital Bonn, Venusberg-Campus 1, Building 76, 53127, Bonn, Germany
| | - Oliver Brüstle
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty & University Hospital Bonn, Venusberg-Campus 1, Building 76, 53127, Bonn, Germany.
| |
Collapse
|
25
|
Mou Y, Dong Y, Chen Z, Denton KR, Duff MO, Blackstone C, Zhang SC, Li XJ. Impaired lipid metabolism in astrocytes underlies degeneration of cortical projection neurons in hereditary spastic paraplegia. Acta Neuropathol Commun 2020; 8:214. [PMID: 33287888 PMCID: PMC7720406 DOI: 10.1186/s40478-020-01088-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 11/18/2020] [Indexed: 12/11/2022] Open
Abstract
Hereditary spastic paraplegias (HSPs) are caused by a length-dependent axonopathy of long corticospinal neurons, but how axons of these cortical projection neurons (PNs) degenerate remains elusive. We generated isogenic human pluripotent stem cell (hPSC) lines for two ATL1 missense mutations associated with SPG3A, the most common early-onset autosomal dominant HSP. In hPSC-derived cortical PNs, ATL1 mutations resulted in reduced axonal outgrowth, impaired axonal transport, and accumulated axonal swellings, recapitulating disease-specific phenotypes. Importantly, ATL1 mutations dysregulated proteolipid gene expression, reduced lipid droplet size in astrocytes, and unexpectedly disrupted cholesterol transfer from glia to neurons, leading to cholesterol deficiency in SPG3A cortical PNs. Applying cholesterol or conditioned medium from control astrocytes, a major source of cholesterol in the brain, rescued aberrant axonal transport and swellings in SPG3A cortical PNs. Furthermore, treatment with the NR1H2 agonist GW3965 corrected lipid droplet defects in SPG3A astrocytes and promoted cholesterol efflux from astrocytes, leading to restoration of cholesterol levels and rescue of axonal degeneration in SPG3A cortical PNs. These results reveal a non-cell autonomous mechanism underlying axonal degeneration of cortical PNs mediated by impaired cholesterol homeostasis in glia.
Collapse
|
26
|
Tabari D, Scholl C, Steffens M, Weickhardt S, Elgner F, Bender D, Herrlein ML, Sabino C, Semkova V, Peitz M, Till A, Brüstle O, Hildt E, Stingl J. Impact of Zika Virus Infection on Human Neural Stem Cell MicroRNA Signatures. Viruses 2020; 12:E1219. [PMID: 33121145 PMCID: PMC7693339 DOI: 10.3390/v12111219] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/09/2020] [Accepted: 10/23/2020] [Indexed: 12/11/2022] Open
Abstract
Zika virus (ZIKV) is a mosquito-borne virus, which can cause brain abnormalities in newborns, including microcephaly. MicroRNAs (miRNAs) are small non-coding RNAs, which post- transcriptionally regulate gene expression. They are involved in various processes including neurological development and host responses to viral infection, but their potential role in ZIKV pathogenesis remains poorly understood. MiRNAs can be incorporated into extracellular vesicles (EVs) and mediate cell-to-cell communication. While it is well known that in viral infections EVs carrying miRNAs can play a crucial role in disease pathogenesis, ZIKV effects on EV-delivered miRNAs and their contribution to ZIKV pathogenesis have not been elucidated. In the present study, we profiled intracellular and EV-derived miRNAs by next generation sequencing and analyzed the host mRNA transcriptome of neural stem cells during infection with ZIKV Uganda and French Polynesia strains. We identified numerous miRNAs, including miR-4792, which were dysregulated at the intracellular level and had altered levels in EVs during ZIKV infection. Integrated analyses of differentially expressed genes and miRNAs showed that ZIKV infection had an impact on processes associated with neurodevelopment and oxidative stress. Our results provide insights into the roles of intracellular and EV-associated host miRNAs in ZIKV pathogenesis.
Collapse
Affiliation(s)
- Denna Tabari
- Research Division, Federal Institute for Drugs and Medical Devices, 53175 Bonn, Germany; (D.T.); (M.S.); (S.W.)
| | - Catharina Scholl
- Research Division, Federal Institute for Drugs and Medical Devices, 53175 Bonn, Germany; (D.T.); (M.S.); (S.W.)
| | - Michael Steffens
- Research Division, Federal Institute for Drugs and Medical Devices, 53175 Bonn, Germany; (D.T.); (M.S.); (S.W.)
| | - Sandra Weickhardt
- Research Division, Federal Institute for Drugs and Medical Devices, 53175 Bonn, Germany; (D.T.); (M.S.); (S.W.)
| | - Fabian Elgner
- Department of Virology, Paul-Ehrlich-Institut, 63225 Langen, Germany; (F.E.); (D.B.); (M.-L.H.); (C.S.); (E.H.)
| | - Daniela Bender
- Department of Virology, Paul-Ehrlich-Institut, 63225 Langen, Germany; (F.E.); (D.B.); (M.-L.H.); (C.S.); (E.H.)
| | - Marie-Luise Herrlein
- Department of Virology, Paul-Ehrlich-Institut, 63225 Langen, Germany; (F.E.); (D.B.); (M.-L.H.); (C.S.); (E.H.)
| | - Catarina Sabino
- Department of Virology, Paul-Ehrlich-Institut, 63225 Langen, Germany; (F.E.); (D.B.); (M.-L.H.); (C.S.); (E.H.)
| | - Vesselina Semkova
- Institute of Reconstructive Neurobiology, LIFE & BRAIN Center, University of Bonn Medical Faculty & University Hospital Bonn, 53127 Bonn, Germany; (V.S.); (M.P.); (A.T.); (O.B.)
| | - Michael Peitz
- Institute of Reconstructive Neurobiology, LIFE & BRAIN Center, University of Bonn Medical Faculty & University Hospital Bonn, 53127 Bonn, Germany; (V.S.); (M.P.); (A.T.); (O.B.)
- Cell Programming Core Facility, Medical Faculty, University of Bonn, 53172 Bonn, Germany
| | - Andreas Till
- Institute of Reconstructive Neurobiology, LIFE & BRAIN Center, University of Bonn Medical Faculty & University Hospital Bonn, 53127 Bonn, Germany; (V.S.); (M.P.); (A.T.); (O.B.)
| | - Oliver Brüstle
- Institute of Reconstructive Neurobiology, LIFE & BRAIN Center, University of Bonn Medical Faculty & University Hospital Bonn, 53127 Bonn, Germany; (V.S.); (M.P.); (A.T.); (O.B.)
| | - Eberhard Hildt
- Department of Virology, Paul-Ehrlich-Institut, 63225 Langen, Germany; (F.E.); (D.B.); (M.-L.H.); (C.S.); (E.H.)
| | - Julia Stingl
- Department of Clinical Pharmacology, University Hospital, RWTH Aachen University, 52074 Aachen, Germany;
| |
Collapse
|
27
|
Schuster S, Heuten E, Velic A, Admard J, Synofzik M, Ossowski S, Macek B, Hauser S, Schöls L. CHIP mutations affect the heat shock response differently in human fibroblasts and iPSC-derived neurons. Dis Model Mech 2020; 13:13/10/dmm045096. [PMID: 33097556 PMCID: PMC7578354 DOI: 10.1242/dmm.045096] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 08/10/2020] [Indexed: 01/09/2023] Open
Abstract
C-terminus of HSC70-interacting protein (CHIP) encoded by the gene STUB1 is a co-chaperone and E3 ligase that acts as a key regulator of cellular protein homeostasis. Mutations in STUB1 cause autosomal recessive spinocerebellar ataxia type 16 (SCAR16) with widespread neurodegeneration manifesting as spastic-ataxic gait disorder, dementia and epilepsy. CHIP-/- mice display severe cerebellar atrophy, show high perinatal lethality and impaired heat stress tolerance. To decipher the pathomechanism underlying SCAR16, we investigated the heat shock response (HSR) in primary fibroblasts of three SCAR16 patients. We found impaired HSR induction and recovery compared to healthy controls. HSPA1A/B transcript levels (coding for HSP70) were reduced upon heat shock but HSP70 remained higher upon recovery in patient- compared to control-fibroblasts. As SCAR16 primarily affects the central nervous system we next investigated the HSR in cortical neurons (CNs) derived from induced pluripotent stem cells of SCAR16 patients. We found CNs of patients and controls to be surprisingly resistant to heat stress with high basal levels of HSP70 compared to fibroblasts. Although heat stress resulted in strong transcript level increases of many HSPs, this did not translate into higher HSP70 protein levels upon heat shock, independent of STUB1 mutations. Furthermore, STUB1(-/-) neurons generated by CRISPR/Cas9-mediated genome editing from an isogenic healthy control line showed a similar HSR to patients. Proteomic analysis of CNs showed dysfunctional protein (re)folding and higher basal oxidative stress levels in patients. Our results question the role of impaired HSR in SCAR16 neuropathology and highlight the need for careful selection of proper cell types for modeling human diseases.
Collapse
Affiliation(s)
- S Schuster
- Department of Neurology and Hertie Institute for Clinical Brain Research, University of Tübingen, 72076 Tübingen, Germany.,Department of Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), 72076 Tübingen, Germany.,Graduate School of Cellular and Molecular Neuroscience, University of Tübingen, 72076 Tübingen, Germany
| | - E Heuten
- Department of Neurology and Hertie Institute for Clinical Brain Research, University of Tübingen, 72076 Tübingen, Germany
| | - A Velic
- Proteome Center Tübingen, University of Tübingen, 72076 Tübingen, Germany
| | - J Admard
- Institute for Medical Genetics and Applied Genomics, University of Tübingen, 72076 Tübingen, Germany
| | - M Synofzik
- Department of Neurology and Hertie Institute for Clinical Brain Research, University of Tübingen, 72076 Tübingen, Germany
| | - S Ossowski
- Institute for Medical Genetics and Applied Genomics, University of Tübingen, 72076 Tübingen, Germany
| | - B Macek
- Proteome Center Tübingen, University of Tübingen, 72076 Tübingen, Germany
| | - S Hauser
- Department of Neurology and Hertie Institute for Clinical Brain Research, University of Tübingen, 72076 Tübingen, Germany .,Department of Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), 72076 Tübingen, Germany
| | - L Schöls
- Department of Neurology and Hertie Institute for Clinical Brain Research, University of Tübingen, 72076 Tübingen, Germany .,Department of Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), 72076 Tübingen, Germany
| |
Collapse
|
28
|
Hauser S, Schuster S, Heuten E, Höflinger P, Admard J, Schelling Y, Velic A, Macek B, Ossowski S, Schöls L. Comparative Transcriptional Profiling of Motor Neuron Disorder-Associated Genes in Various Human Cell Culture Models. Front Cell Dev Biol 2020; 8:544043. [PMID: 33072739 PMCID: PMC7543453 DOI: 10.3389/fcell.2020.544043] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 09/04/2020] [Indexed: 01/06/2023] Open
Abstract
Disease modeling requires appropriate cellular models that best mimic the underlying pathophysiology. Human origin and an adequate expression of the disease protein are pre-requisites that support information from a model to be meaningful. In this study we investigated expression profiles of (i) PBMCs and (ii) fibroblasts as patient derived cells as well as (iii) lymphoblasts and (iv) induced pluripotent stem cells (iPSC) as immortalized sources, and (v) iPSC-derived cortical neurons to assess their aptitude to model motor neuron diseases (MNDs) including hereditary spastic paraplegia (HSP), amyotrophic lateral sclerosis (ALS) and spinal muscular atrophy (SMA). We generated all five different cell types from two healthy donors and performed RNA sequencing to display expression patterns in MND-related genes. For the ten most common HSP genotypes we validated gene expression by qPCR. To verify the results on protein level, proteome analysis of fibroblasts, iPSCs and cortical neurons was performed. Depending on the specific MND gene we found largely different expression patterns. Out of 168 MND-related genes, 50 had their highest expression in iPSC-derived cortical neurons, 41 were most strongly expressed in fibroblasts, 26 in lymphoblasts, 22 in iPSCs, and 14 in PBMCs. Pathophysiologically related MNDs like HSPs associated with axonal transport deficits shared highest expression in cortical neurons. 15 MND-related genes were not detectable in any of the analyzed cell types. This may reflect the critical dependency of motor neurons on support of other cell types like oligodendrocytes which express myelin proteins like L1CAM (SPG1), PLP1 (SPG2) and MAG (SPG75) which are lacking in neurons but cause MNDs if mutated. This study provides comprehensive information on expression of genes associated with a large spectrum of MNDs. Expression profiles can be used to inform on appropriate cell models for genotype specific motor neuron research.
Collapse
Affiliation(s)
- Stefan Hauser
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany.,Department of Neurology and Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Stefanie Schuster
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany.,Department of Neurology and Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.,Graduate School of Cellular and Molecular Neuroscience, University of Tübingen, Tübingen, Germany
| | - Elena Heuten
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Philip Höflinger
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany.,Department of Neurology and Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.,Graduate School of Cellular and Molecular Neuroscience, University of Tübingen, Tübingen, Germany
| | - Jakob Admard
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Yvonne Schelling
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany.,Department of Neurology and Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Ana Velic
- Proteome Center Tübingen, University of Tübingen, Tübingen, Germany
| | - Boris Macek
- Proteome Center Tübingen, University of Tübingen, Tübingen, Germany
| | - Stephan Ossowski
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany.,Center of Rare Diseases, University of Tübingen, Tübingen, Germany
| | - Ludger Schöls
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany.,Department of Neurology and Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.,Center of Rare Diseases, University of Tübingen, Tübingen, Germany
| |
Collapse
|
29
|
Allison R, Edgar JR, Reid E. Spastin MIT Domain Disease-Associated Mutations Disrupt Lysosomal Function. Front Neurosci 2019; 13:1179. [PMID: 31787869 PMCID: PMC6856053 DOI: 10.3389/fnins.2019.01179] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 10/18/2019] [Indexed: 12/25/2022] Open
Abstract
The hereditary spastic paraplegias (HSPs) are genetic motor neuron diseases characterized by progressive degeneration of corticospinal tract axons. Mutations in SPAST, encoding the microtubule-severing ATPase spastin, are the most common causes of HSP. The broad SPAST mutational spectrum indicates a haploinsufficiency pathogenic mechanism in most cases. Most missense mutations cluster in the ATPase domain, where they disrupt the protein's ability to sever microtubules. However, several putative missense mutations in the protein's microtubule interacting and trafficking (MIT) domain have also been described, but the pathogenicity of these mutations has not been verified with functional studies. Spastin promotes endosomal tubule fission, and defects in this lead to lysosomal enzyme mistrafficking and downstream lysosomal abnormalities. We investigated the function of three disease-associated spastin MIT mutants and found that none was able to promote normal endosomal tubule fission, lysosomal enzyme receptor trafficking, or lysosomal morphology. One of the mutations affected recruitment of spastin to endosomes, a property that requires the canonical function of the MIT domain in binding endosomal sorting complex required for transport (ESCRT)-III proteins. However, the other mutants did not affect spastin's endosomal recruitment, raising the possibility of pathologically important non-canonical roles for the MIT domain. In conclusion, we demonstrate that spastin MIT mutants cause functional abnormalities related to the pathogenesis of HSP. These mutations do not directly affect spastin's microtubule-severing capacity, and so we identify a new molecular pathological mechanism by which spastin mutations may cause disease.
Collapse
Affiliation(s)
- Rachel Allison
- Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - James R Edgar
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Evan Reid
- Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|