1
|
Riaz Z, Richardson GS, Jin H, Zenitsky G, Anantharam V, Kanthasamy A, Kanthasamy AG. Nuclear pore and nucleocytoplasmic transport impairment in oxidative stress-induced neurodegeneration: relevance to molecular mechanisms in Pathogenesis of Parkinson's and other related neurodegenerative diseases. Mol Neurodegener 2024; 19:87. [PMID: 39578912 PMCID: PMC11585115 DOI: 10.1186/s13024-024-00774-0] [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: 08/23/2024] [Accepted: 10/28/2024] [Indexed: 11/24/2024] Open
Abstract
Nuclear pore complexes (NPCs) are embedded in the nuclear envelope and facilitate the exchange of macromolecules between the nucleus and cytoplasm in eukaryotic cells. The dysfunction of the NPC and nuclear transport plays a significant role in aging and the pathogenesis of various neurodegenerative diseases. Common features among these neurodegenerative diseases, including Parkinson's disease (PD), encompass mitochondrial dysfunction, oxidative stress and the accumulation of insoluble protein aggregates in specific brain regions. The susceptibility of dopaminergic neurons to mitochondrial stress underscores the pivotal role of mitochondria in PD progression. Disruptions in mitochondrial-nuclear communication are exacerbated by aging and α-synuclein-induced oxidative stress in PD. The precise mechanisms underlying mitochondrial impairment-induced neurodegeneration in PD are still unclear. Evidence suggests that perturbations in dopaminergic neuronal nuclei are linked to PD-related neurodegeneration. These perturbations involve structural damage to the nuclear envelope and mislocalization of pivotal transcription factors, potentially driven by oxidative stress or α-synuclein pathology. The presence of protein aggregates, pathogenic mutations, and ongoing oxidative stress can exacerbate the dysfunction of NPCs, yet this mechanism remains understudied in the context of oxidative stress-induced PD. This review summarizes the link between mitochondrial dysfunction and dopaminergic neurodegeneration and outlines the current evidence for nuclear envelope and nuclear transport abnormalities in PD, particularly in oxidative stress. We highlight the potential role of nuclear pore and nucleocytoplasmic transport dysfunction in PD and stress the importance of systematically investigating NPC components in PD.
Collapse
Affiliation(s)
- Zainab Riaz
- Isakson Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, Athens, GA, USA
| | - Gabriel S Richardson
- Isakson Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, Athens, GA, USA
| | - Huajun Jin
- Isakson Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, Athens, GA, USA
| | - Gary Zenitsky
- Isakson Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, Athens, GA, USA
| | - Vellareddy Anantharam
- Isakson Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, Athens, GA, USA
| | - Arthi Kanthasamy
- Isakson Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, Athens, GA, USA
| | - Anumantha G Kanthasamy
- Isakson Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, Athens, GA, USA.
| |
Collapse
|
2
|
Mahmoodi M, Ayatollahi Mehrgardi A, Momen M, Serpell JA, Esmailizadeh A. Deciphering the genetic basis of behavioral traits in dogs: Observed-trait GWAS and latent-trait GWAS analysis reveal key genes and variants. Vet J 2024; 308:106251. [PMID: 39368730 DOI: 10.1016/j.tvjl.2024.106251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/21/2024] [Accepted: 09/26/2024] [Indexed: 10/07/2024]
Abstract
Dogs exhibit remarkable phenotypic diversity, particularly in behavioral traits, making them an excellent model for studying the genetic basis of complex behaviors. Behavioral traits such as aggression and fear are highly heritable among different dog breeds, but their genetic basis is largely unknown. We used the genome-wide association study (GWAS) to identify candidate genes associated with nine behavioral traits including; stranger-directed aggression (SDA), owner-directed aggression (ODA), dog-directed aggression (DDA), stranger-directed fear (SDF), nonsocial fear (NF), dog-directed fear (DDF), touch sensitivity (TS), separation-related behavior (SRB) and attachment attention-seeking (AAS). The observed behavioral traits were collected from 38,714 to 40,460 individuals across 108 modern dog breeds. We performed a GWAS based on a latent trait extracted using the confirmatory factor analysis (CFA) method with nine observable behavioral traits and compared the results with those from the GWAS of the observed traits. Using both observed-trait and latent-trait GWAS, we identified 41 significant SNPs that were common between both GWAS methods, of which 26 were pleiotropic, as well as 10 SNPs unique to the latent-trait GWAS, and 5 SNPs unique to the observed-trait GWAS discovered. These SNPs were associated with 21 genes in latent-trait GWAS and 22 genes in the observed-trait GWAS, with 19 genes shared by both. According to previous studies, some of the genes from this study have been reported to be related to behavioral and neurological functions in dogs. In the human population, these identified genes play a role in either the formation of the nervous system or are linked to various mental health conditions. Taken together, our findings suggest that latent-trait GWAS for behavioral traits in dogs identifies significant latent genes that are neurologically prioritized.
Collapse
Affiliation(s)
- Maryam Mahmoodi
- Department of Animal Science, Faculty of Agriculture, Shahid Bahonar University of Kerman, Kerman, Iran.
| | - Ahmad Ayatollahi Mehrgardi
- Department of Animal Science, Faculty of Agriculture, Shahid Bahonar University of Kerman, Kerman, Iran.
| | - Mehdi Momen
- Department of Surgical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - James A Serpell
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ali Esmailizadeh
- Department of Animal Science, Faculty of Agriculture, Shahid Bahonar University of Kerman, Kerman, Iran
| |
Collapse
|
3
|
Colussi C, Bertozzi A, Leone L, Rinaudo M, Sollazzo R, Conte F, Paccosi E, Nardella L, Aceto G, Li Puma DD, Ripoli C, Vita MG, Marra C, D'Ascenzo M, Grassi C. Nucleoporin 153 deficiency in adult neural stem cells defines a pathological protein-network signature and defective neurogenesis in a mouse model of AD. Stem Cell Res Ther 2024; 15:275. [PMID: 39227892 PMCID: PMC11373261 DOI: 10.1186/s13287-024-03805-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 06/17/2024] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND Reduction of adult hippocampal neurogenesis is an early critical event in Alzheimer's disease (AD), contributing to progressive memory loss and cognitive decline. Reduced levels of the nucleoporin 153 (Nup153), a key epigenetic regulator of NSC stemness, characterize the neural stem cells isolated from a mouse model of AD (3×Tg) (AD-NSCs) and determine their altered plasticity and gene expression. METHODS Nup153-regulated mechanisms contributing to NSC function were investigated: (1) in cultured NSCs isolated from AD and wild type (WT) mice by proteomics; (2) in vivo by lentiviral-mediated delivery of Nup153 or GFP in the hippocampus of AD and control mice analyzing neurogenesis and cognitive function; (3) in human iPSC-derived brain organoids obtained from AD patients and control subjects as a model of neurodevelopment. RESULTS Proteomic approach identified Nup153 interactors in WT- and AD-NSCs potentially implicated in neurogenesis regulation. Gene ontology (GO) analysis showed that Nup153-bound proteins in WT-NSCs were involved in RNA metabolism, nuclear import and epigenetic mechanisms. Nup153-bound proteins in AD-NSCs were involved in pathways of neurodegeneration, mitochondrial dysfunction, proteasomal processing and RNA degradation. Furthermore, recovery of Nup153 levels in AD-NSCs reduced the levels of oxidative stress markers and recovered proteasomal activity. Lentiviral-mediated delivery of Nup153 in the hippocampal niche of AD mice increased the proliferation of early progenitors, marked by BrdU/DCX and BrdU/PSANCAM positivity and, later, the integration of differentiating neurons in the cell granule layer (BrdU/NeuN+ cells) compared with GFP-injected AD mice. Consistently, Nup153-injected AD mice showed an improvement of cognitive performance in comparison to AD-GFP mice at 1 month after virus delivery assessed by Morris Water Maze. To validate the role of Nup153 in neurogenesis we took advantage of brain organoids derived from AD-iPSCs characterized by fewer neuroepithelial progenitor loops and reduced differentiation areas. The upregulation of Nup153 in AD organoids recovered the formation of neural-like tubes and differentiation. CONCLUSIONS Our data suggest that the positive effect of Nup153 on neurogenesis is based on a complex regulatory network orchestrated by Nup153 and that this protein is a valuable disease target.
Collapse
Affiliation(s)
- Claudia Colussi
- Istituto di Analisi dei Sistemi ed Informatica "Antonio Ruberti" (IASI) - CNR , National Research Council, Via dei Taurini 19, Rome, 00185, Italy.
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, 00168, Italy.
| | - Alessia Bertozzi
- Istituto di Analisi dei Sistemi ed Informatica "Antonio Ruberti" (IASI) - CNR , National Research Council, Via dei Taurini 19, Rome, 00185, Italy
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, 00168, Italy
| | - Lucia Leone
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, 00168, Italy
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, 00168, Italy
| | - Marco Rinaudo
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, 00168, Italy
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, 00168, Italy
| | - Raimondo Sollazzo
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, 00168, Italy
| | - Federica Conte
- Istituto di Analisi dei Sistemi ed Informatica "Antonio Ruberti" (IASI) - CNR , National Research Council, Via dei Taurini 19, Rome, 00185, Italy
| | - Elena Paccosi
- Istituto di Analisi dei Sistemi ed Informatica "Antonio Ruberti" (IASI) - CNR , National Research Council, Via dei Taurini 19, Rome, 00185, Italy
| | - Luca Nardella
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, 00168, Italy
| | - Giuseppe Aceto
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, 00168, Italy
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, 00168, Italy
| | - Domenica Donatella Li Puma
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, 00168, Italy
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, 00168, Italy
| | - Cristian Ripoli
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, 00168, Italy
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, 00168, Italy
| | | | - Camillo Marra
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, 00168, Italy
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, 00168, Italy
| | - Marcello D'Ascenzo
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, 00168, Italy
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, 00168, Italy
| | - Claudio Grassi
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, 00168, Italy
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, 00168, Italy
| |
Collapse
|
4
|
Wang Z, Zhang J, Luo L, Zhang C, Huang X, Liu S, Chen H, Miao W. Nucleoporin 93 Regulates Cancer Cell Growth and Stemness in Bladder Cancer via Wnt/β-Catenin Signaling. Mol Biotechnol 2024:10.1007/s12033-024-01184-9. [PMID: 38744786 DOI: 10.1007/s12033-024-01184-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 04/16/2024] [Indexed: 05/16/2024]
Abstract
Bladder cancer (BLCA) is a prevalent cancer type with an unmet need for new therapeutic strategies. Nucleoporin 93 (Nup93) is implicated in the pathophysiology of several cancers, but its relationship with bladder cancer remains unclear. Nup93 expression was analyzed in TCGA datasets and 88 BLCA patient samples. Survival analysis and Cox regression models evaluated the association between Nup93 levels and patient prognosis. BLCA cells were used to investigate the effects of Nup93 overexpression or knockdown on cell growth, invasion, stemness (sphere formation and ALDH2 + cancer stem cell marker), and Wnt/β-catenin signaling in vitro. The Wnt activator BML-284 was used to confirm the involvement of Wnt/β-catenin signaling pathway. A xenograft mouse model validated the in vitro findings. Nup93 was highly expressed in BLCA tissues and cell lines, and high Nup93 expression correlated with poor prognosis in BLCA patients. Nup93 silencing inhibited BLCA cell proliferation, Wnt/β-catenin activation, and cancer cell stemness. Conversely, Nup93 overexpression promoted these effects. BML-284 partially rescued the reduction in cell growth and stemness markers caused by Nup93 knockdown. Nup93 knockdown also suppressed the tumor formation of BLCA cells in vivo. Nup93 regulates BLCA cell growth and stemness via the Wnt/β-catenin pathway, suggesting its potential as a prognostic marker and therapeutic target in BLCA.
Collapse
Affiliation(s)
- Zhe Wang
- Urology Department, The First Affiliated Hospital of Hebei North University, No. 12, Changqing Road, Zhangjiakou, 050051, Hebei Province, China
| | - Jing Zhang
- Department of Medical Oncology, The First Affiliated Hospital of Hebei North University, No. 12, Changqing Road, Zhangjiakou, 050051, Hebei Province, China
| | - Lina Luo
- Urology Department, The First Affiliated Hospital of Hebei North University, No. 12, Changqing Road, Zhangjiakou, 050051, Hebei Province, China
| | - Chao Zhang
- Urology Department, The First Affiliated Hospital of Hebei North University, No. 12, Changqing Road, Zhangjiakou, 050051, Hebei Province, China
| | - Xiaomeng Huang
- Medical Department, The First Affiliated Hospital of Hebei North University, No. 12, Changqing Road, Zhangjiakou, 050051, Hebei Province, China
| | - Shuo Liu
- Urology Department, The First Affiliated Hospital of Hebei North University, No. 12, Changqing Road, Zhangjiakou, 050051, Hebei Province, China
| | - Huaian Chen
- Urology Department, The First Affiliated Hospital of Hebei North University, No. 12, Changqing Road, Zhangjiakou, 050051, Hebei Province, China
| | - Wenlong Miao
- Urology Department, The First Affiliated Hospital of Hebei North University, No. 12, Changqing Road, Zhangjiakou, 050051, Hebei Province, China.
| |
Collapse
|
5
|
Wang ZQ, Wu ZX, Wang ZP, Bao JX, Wu HD, Xu DY, Li HF, Xu YY, Wu RX, Dai XX. Pan-cancer analysis of NUP155 and validation of its role in breast cancer cell proliferation, migration, and apoptosis. BMC Cancer 2024; 24:353. [PMID: 38504158 PMCID: PMC10953186 DOI: 10.1186/s12885-024-12039-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 02/21/2024] [Indexed: 03/21/2024] Open
Abstract
NUP155 is reported to be correlated with tumor development. However, the role of NUP155 in tumor physiology and the tumor immune microenvironment (TIME) has not been previously examined. This study comprehensively investigated the expression, immunological function, and prognostic significance of NUP155 in different cancer types. Bioinformatics analysis revealed that NUP155 was upregulated in 26 types of cancer. Additionally, NUP155 upregulation was strongly correlated with advanced pathological or clinical stages and poor prognosis in several cancers. Furthermore, NUP155 was significantly and positively correlated with DNA methylation, tumor mutational burden, microsatellite instability, and stemness score in most cancers. Additionally, NUP155 was also found to be involved in TIME and closely associated with tumor infiltrating immune cells and immunoregulation-related genes. Functional enrichment analysis revealed a strong correlation between NUP155 and immunomodulatory pathways, especially antigen processing and presentation. The role of NUP155 in breast cancer has not been examined. This study, for the first time, demonstrated that NUP155 was upregulated in breast invasive carcinoma (BRCA) cells and revealed its oncogenic role in BRCA using molecular biology experiments. Thus, our study highlights the potential value of NUP155 as a biomarker in the assessment of prognostic prediction, tumor microenvironment and immunotherapeutic response in pan-cancer.
Collapse
Affiliation(s)
- Zi-Qiong Wang
- Quzhou People's Hospital, The Quzhou Affiliated Hospital of Wenzhou Medical University, 100 Minjiang Avenue, Quzhou, Zhejiang, 324000, Zhejiang, China
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Zhi-Xuan Wu
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Zong-Pan Wang
- Quzhou People's Hospital, The Quzhou Affiliated Hospital of Wenzhou Medical University, 100 Minjiang Avenue, Quzhou, Zhejiang, 324000, Zhejiang, China
| | - Jing-Xia Bao
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Hao-Dong Wu
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Di-Yan Xu
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Hong-Feng Li
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Yi-Yin Xu
- Quzhou People's Hospital, The Quzhou Affiliated Hospital of Wenzhou Medical University, 100 Minjiang Avenue, Quzhou, Zhejiang, 324000, Zhejiang, China
| | - Rong-Xing Wu
- Quzhou People's Hospital, The Quzhou Affiliated Hospital of Wenzhou Medical University, 100 Minjiang Avenue, Quzhou, Zhejiang, 324000, Zhejiang, China.
| | - Xuan-Xuan Dai
- Quzhou People's Hospital, The Quzhou Affiliated Hospital of Wenzhou Medical University, 100 Minjiang Avenue, Quzhou, Zhejiang, 324000, Zhejiang, China.
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
6
|
Ravindran E, Lesca G, Januel L, Goldgruber L, Dickmanns A, Margot H, Kaindl AM. Case report: Compound heterozygous NUP85 variants cause autosomal recessive primary microcephaly. Front Neurol 2023; 14:1124886. [PMID: 36846113 PMCID: PMC9947397 DOI: 10.3389/fneur.2023.1124886] [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: 12/15/2022] [Accepted: 01/13/2023] [Indexed: 02/11/2023] Open
Abstract
Nucleoporin (NUP) 85 is a member of the Y-complex of nuclear pore complex (NPC) that is key for nucleocytoplasmic transport function, regulation of mitosis, transcription, and chromatin organization. Mutations in various nucleoporin genes have been linked to several human diseases. Among them, NUP85 was linked to childhood-onset steroid-resistant nephrotic syndrome (SRNS) in four affected individuals with intellectual disability but no microcephaly. Recently, we broaden the phenotype spectrum of NUP85-associated disease by reporting NUP85 variants in two unrelated individuals with primary autosomal recessive microcephaly (MCPH) and Seckel syndrome (SCKS) spectrum disorders (MCPH-SCKS) without SRNS. In this study, we report compound heterozygous NUP85 variants in an index patient with only MCPH phenotype, but neither Seckel syndrome nor SRNS was reported. We showed that the identified missense variants cause reduced cell viability of patient-derived fibroblasts. Structural simulation analysis of double variants is predicted to alter the structure of NUP85 and its interactions with neighboring NUPs. Our study thereby further expands the phenotypic spectrum of NUP85-associated human disorder and emphasizes the crucial role of NUP85 in the brain development and function.
Collapse
Affiliation(s)
- Ethiraj Ravindran
- Institute of Cell Biology and Neurobiology, Charité – Universitätsmedizin Berlin, Berlin, Germany,Department of Pediatric Neurology, Charité – Universitätsmedizin Berlin, Berlin, Germany,Center for Chronically Sick Children (Sozialpädiatrisches Zentrum, SPZ), Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Gaetan Lesca
- Department of Genetics, Hospices Civils de Lyon, Groupe Hospitalier Est, Bron, France,Institut NeuroMyoGene PNMG, CNRS UMR5310, INSERM U1217, Université Claude Bernard Lyon 1, Lyon, France
| | - Louis Januel
- Department of Genetics, Hospices Civils de Lyon, Groupe Hospitalier Est, Bron, France
| | - Linus Goldgruber
- Department of Biomedical Engineering, Veterinärmedizinische Universität (Vetmeduni), Vienna, Austria
| | - Achim Dickmanns
- Department of Molecular Structural Biology, Institute for Microbiology and Genetics (GZMB), Georg-August-University Göttingen, Göttingen, Germany
| | - Henri Margot
- Department of Medical Genetics, University of Bordeaux, MRGM INSERM U1211, CHU de Bordeaux, Bordeaux, France,*Correspondence: Henri Margot ✉
| | - Angela M. Kaindl
- Institute of Cell Biology and Neurobiology, Charité – Universitätsmedizin Berlin, Berlin, Germany,Department of Pediatric Neurology, Charité – Universitätsmedizin Berlin, Berlin, Germany,Center for Chronically Sick Children (Sozialpädiatrisches Zentrum, SPZ), Charité – Universitätsmedizin Berlin, Berlin, Germany,Angela M. Kaindl ✉
| |
Collapse
|
7
|
Rasool D, Soleimani VD, Jahani-Asl A. Isolation of Adult Mouse Neural Stem Cells and Assessment of Self-Renewal by ELDA. Methods Mol Biol 2022; 2515:343-354. [PMID: 35776362 DOI: 10.1007/978-1-0716-2409-8_21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The generation of new neurons in the adult brain throughout life is integral to brain plasticity and repair. Adult neural stem cells (aNSCs), present in the subventricular zone (SVZ) of the lateral ventricle wall and the subgranular zone (SGZ) of the hippocampal dentate gyrus, divide symmetrically or asymmetrically to maintain the stem cell pool or become committed progenitors and differentiate into various cell lineages. Depletion or dysregulation of aNSCs impairs proper brain connectivity and function and can contribute to several brain diseases including cognitive and neurodegenerative disorders and brain cancer. In this chapter, we present our optimized method to obtain and maintain reproducible neurosphere cultures from the adult mouse brain followed by evaluation of self-renewal using the extreme limiting dilution assay (ELDA) software. We use this assay routinely on aNSCs obtained from patient mouse models to generate log fraction plots and provide confidence intervals for all limiting dilution assay (LDA) data. At the same time, given the low number of NSCs required for the completion of the ELDA experiment, it is feasible to employ this approach to conduct high-content compound screening for therapeutic interventions aimed at enhancing the stem cell pool or combating a cohort of genetic and epigenetic disorders.
Collapse
Affiliation(s)
- Dilan Rasool
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
- Lady Davis Institute for Medical Research, Jewish General Hospital, Chemin de la Côte-Sainte-Catherine, Montreal, QC, Canada
- Division of Experimental Medicine, McGill University, Montreal, QC, Canada
- University of Ottawa Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Vahab D Soleimani
- Lady Davis Institute for Medical Research, Jewish General Hospital, Chemin de la Côte-Sainte-Catherine, Montreal, QC, Canada
- Division of Experimental Medicine, McGill University, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Arezu Jahani-Asl
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada.
- Division of Experimental Medicine, McGill University, Montreal, QC, Canada.
- University of Ottawa Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|