1
|
Kremer LPM, Cerrizuela S, El-Sammak H, Al Shukairi ME, Ellinger T, Straub J, Korkmaz A, Volk K, Brunken J, Kleber S, Anders S, Martin-Villalba A. DNA methylation controls stemness of astrocytes in health and ischaemia. Nature 2024; 634:415-423. [PMID: 39232166 PMCID: PMC11464379 DOI: 10.1038/s41586-024-07898-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 08/01/2024] [Indexed: 09/06/2024]
Abstract
Astrocytes are the most abundant cell type in the mammalian brain and provide structural and metabolic support to neurons, regulate synapses and become reactive after injury and disease. However, a small subset of astrocytes settles in specialized areas of the adult brain where these astrocytes instead actively generate differentiated neuronal and glial progeny and are therefore referred to as neural stem cells1-3. Common parenchymal astrocytes and quiescent neural stem cells share similar transcriptomes despite their very distinct functions4-6. Thus, how stem cell activity is molecularly encoded remains unknown. Here we examine the transcriptome, chromatin accessibility and methylome of neural stem cells and their progeny, and of astrocytes from the striatum and cortex in the healthy and ischaemic adult mouse brain. We identify distinct methylation profiles associated with either astrocyte or stem cell function. Stem cell function is mediated by methylation of astrocyte genes and demethylation of stem cell genes that are expressed later. Ischaemic injury to the brain induces gain of stemness in striatal astrocytes7. We show that this response involves reprogramming the astrocyte methylome to a stem cell methylome and is absent if the de novo methyltransferase DNMT3A is missing. Overall, we unveil DNA methylation as a promising target for regenerative medicine.
Collapse
Affiliation(s)
- Lukas P M Kremer
- Division of Molecular Neurobiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- BioQuant Centre, University of Heidelberg, Heidelberg, Germany
| | - Santiago Cerrizuela
- Division of Molecular Neurobiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hadil El-Sammak
- Division of Molecular Neurobiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Tobias Ellinger
- Division of Molecular Neurobiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jannes Straub
- Division of Molecular Neurobiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Aylin Korkmaz
- Division of Molecular Neurobiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Katrin Volk
- Division of Molecular Neurobiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jan Brunken
- Division of Molecular Neurobiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Susanne Kleber
- Division of Molecular Neurobiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Simon Anders
- BioQuant Centre, University of Heidelberg, Heidelberg, Germany.
| | - Ana Martin-Villalba
- Division of Molecular Neurobiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
2
|
Dennis DJ, Wang BS, Karamboulas K, Kaplan DR, Miller FD. Single-cell approaches define two groups of mammalian oligodendrocyte precursor cells and their evolution over developmental time. Stem Cell Reports 2024; 19:654-672. [PMID: 38579710 PMCID: PMC11103788 DOI: 10.1016/j.stemcr.2024.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 03/04/2024] [Accepted: 03/05/2024] [Indexed: 04/07/2024] Open
Abstract
Here, we used single-cell RNA sequencing (scRNA-seq), single-cell ATAC sequencing (scATAC-seq), and single-cell spatial transcriptomics to characterize murine cortical OPCs throughout postnatal life. During development, we identified two groups of differentially localized PDGFRα+ OPCs that are transcriptionally and epigenetically distinct. One group (active, or actOPCs) is metabolically active and enriched in white matter. The second (homeostatic, or hOPCs) is less active, enriched in gray matter, and predicted to derive from actOPCs. In adulthood, these two groups are transcriptionally but not epigenetically distinct, and relative to developing OPCs are less active metabolically and have less open chromatin. When adult oligodendrogenesis is enhanced during experimentally induced remyelination, adult OPCs do not reacquire a developmental open chromatin state, and the oligodendrogenesis trajectory is distinct from that seen neonatally. These data suggest that there are two OPC groups subserving distinct postnatal functions and that neonatal and adult OPC-mediated oligodendrogenesis are fundamentally different.
Collapse
Affiliation(s)
- Daniel J Dennis
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Beatrix S Wang
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada; Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Konstantina Karamboulas
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - David R Kaplan
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Freda D Miller
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada.
| |
Collapse
|
3
|
Mukhtar T, Breda J, Adam MA, Boareto M, Grobecker P, Karimaddini Z, Grison A, Eschbach K, Chandrasekhar R, Vermeul S, Okoniewski M, Pachkov M, Harwell CC, Atanasoski S, Beisel C, Iber D, van Nimwegen E, Taylor V. Temporal and sequential transcriptional dynamics define lineage shifts in corticogenesis. EMBO J 2022; 41:e111132. [PMID: 36345783 PMCID: PMC9753470 DOI: 10.15252/embj.2022111132] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 09/09/2022] [Accepted: 09/26/2022] [Indexed: 11/11/2022] Open
Abstract
The cerebral cortex contains billions of neurons, and their disorganization or misspecification leads to neurodevelopmental disorders. Understanding how the plethora of projection neuron subtypes are generated by cortical neural stem cells (NSCs) is a major challenge. Here, we focused on elucidating the transcriptional landscape of murine embryonic NSCs, basal progenitors (BPs), and newborn neurons (NBNs) throughout cortical development. We uncover dynamic shifts in transcriptional space over time and heterogeneity within each progenitor population. We identified signature hallmarks of NSC, BP, and NBN clusters and predict active transcriptional nodes and networks that contribute to neural fate specification. We find that the expression of receptors, ligands, and downstream pathway components is highly dynamic over time and throughout the lineage implying differential responsiveness to signals. Thus, we provide an expansive compendium of gene expression during cortical development that will be an invaluable resource for studying neural developmental processes and neurodevelopmental disorders.
Collapse
Affiliation(s)
- Tanzila Mukhtar
- Department of BiomedicineUniversity of BaselBaselSwitzerland
| | - Jeremie Breda
- BiozentrumUniversity of BaselBaselSwitzerland
- Swiss Institute of Bioinformatics (SIB)BaselSwitzerland
| | - Manal A Adam
- Eli and Edythe Broad Center of Regeneration Medicine and Stem cell ResearchUniversity of California, San FranciscoSan FranciscoCAUSA
- Weill Institute for NeuroscienceSan FranciscoCAUSA
- Department of NeurologyUniversity of California, San FranciscoSan FranciscoCAUSA
| | - Marcelo Boareto
- Swiss Institute of Bioinformatics (SIB)BaselSwitzerland
- Computational Biology Group, D‐BSSEETH ZürichBaselSwitzerland
| | - Pascal Grobecker
- BiozentrumUniversity of BaselBaselSwitzerland
- Swiss Institute of Bioinformatics (SIB)BaselSwitzerland
| | - Zahra Karimaddini
- Swiss Institute of Bioinformatics (SIB)BaselSwitzerland
- Computational Biology Group, D‐BSSEETH ZürichBaselSwitzerland
| | - Alice Grison
- Department of BiomedicineUniversity of BaselBaselSwitzerland
| | - Katja Eschbach
- Department of Biosystems Science and EngineeringETH ZürichBaselSwitzerland
| | | | - Swen Vermeul
- Scientific IT ServicesETH ZürichZürichSwitzerland
| | | | - Mikhail Pachkov
- BiozentrumUniversity of BaselBaselSwitzerland
- Swiss Institute of Bioinformatics (SIB)BaselSwitzerland
| | - Corey C Harwell
- Eli and Edythe Broad Center of Regeneration Medicine and Stem cell ResearchUniversity of California, San FranciscoSan FranciscoCAUSA
- Weill Institute for NeuroscienceSan FranciscoCAUSA
- Department of NeurologyUniversity of California, San FranciscoSan FranciscoCAUSA
| | - Suzana Atanasoski
- Department of BiomedicineUniversity of BaselBaselSwitzerland
- Faculty of MedicineUniversity of ZürichZürichSwitzerland
| | - Christian Beisel
- Department of Biosystems Science and EngineeringETH ZürichBaselSwitzerland
| | - Dagmar Iber
- Swiss Institute of Bioinformatics (SIB)BaselSwitzerland
- Weill Institute for NeuroscienceSan FranciscoCAUSA
| | - Erik van Nimwegen
- BiozentrumUniversity of BaselBaselSwitzerland
- Swiss Institute of Bioinformatics (SIB)BaselSwitzerland
| | - Verdon Taylor
- Department of BiomedicineUniversity of BaselBaselSwitzerland
| |
Collapse
|
4
|
Chowdhury R, Wang Y, Campbell M, Goderie SK, Doyle F, Tenenbaum SA, Kusek G, Kiehl TR, Ansari SA, Boles NC, Temple S. STAU2 binds a complex RNA cargo that changes temporally with production of diverse intermediate progenitor cells during mouse corticogenesis. Development 2021; 148:271165. [PMID: 34345913 DOI: 10.1242/dev.199376] [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: 12/20/2020] [Accepted: 07/05/2021] [Indexed: 12/18/2022]
Abstract
STAU2 is a double-stranded RNA-binding protein enriched in the nervous system. During asymmetric divisions in the developing mouse cortex, STAU2 preferentially distributes into the intermediate progenitor cell (IPC), delivering RNA molecules that can impact IPC behavior. Corticogenesis occurs on a precise time schedule, raising the hypothesis that the cargo STAU2 delivers into IPCs changes over time. To test this, we combine RNA-immunoprecipitation with sequencing (RIP-seq) over four stages of mouse cortical development, generating a comprehensive cargo profile for STAU2. A subset of the cargo was 'stable', present at all stages, and involved in chromosome organization, macromolecule localization, translation and DNA repair. Another subset was 'dynamic', changing with cortical stage, and involved in neurogenesis, cell projection organization, neurite outgrowth, and included cortical layer markers. Notably, the dynamic STAU2 cargo included determinants of IPC versus neuronal fates and genes contributing to abnormal corticogenesis. Knockdown of one STAU2 target, Taf13, previously linked to microcephaly and impaired myelination, reduced oligodendrogenesis in vitro. We conclude that STAU2 contributes to the timing of corticogenesis by binding and delivering complex and temporally regulated RNA cargo into IPCs.
Collapse
Affiliation(s)
- Rebecca Chowdhury
- Neural Stem Cell Institute, Regenerative Research Foundation, Rensselaer, NY 12144, USA
| | - Yue Wang
- Neural Stem Cell Institute, Regenerative Research Foundation, Rensselaer, NY 12144, USA
| | - Melissa Campbell
- Neural Stem Cell Institute, Regenerative Research Foundation, Rensselaer, NY 12144, USA
| | - Susan K Goderie
- Neural Stem Cell Institute, Regenerative Research Foundation, Rensselaer, NY 12144, USA
| | - Francis Doyle
- Nanobioscience Constellation, College of Nanoscale Science and Engineering, SUNY Polytechnic Institute, Albany, NY 12203, USA
| | - Scott A Tenenbaum
- Nanobioscience Constellation, College of Nanoscale Science and Engineering, SUNY Polytechnic Institute, Albany, NY 12203, USA
| | - Gretchen Kusek
- Neural Stem Cell Institute, Regenerative Research Foundation, Rensselaer, NY 12144, USA
| | - Thomas R Kiehl
- Neural Stem Cell Institute, Regenerative Research Foundation, Rensselaer, NY 12144, USA
| | - Suraiya A Ansari
- Department of Biochemistry and Molecular Biology, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, United Arab Emirates
| | - Nathan C Boles
- Neural Stem Cell Institute, Regenerative Research Foundation, Rensselaer, NY 12144, USA
| | - Sally Temple
- Neural Stem Cell Institute, Regenerative Research Foundation, Rensselaer, NY 12144, USA
| |
Collapse
|
5
|
Jin T, Rehani P, Ying M, Huang J, Liu S, Roussos P, Wang D. scGRNom: a computational pipeline of integrative multi-omics analyses for predicting cell-type disease genes and regulatory networks. Genome Med 2021; 13:95. [PMID: 34044854 PMCID: PMC8161957 DOI: 10.1186/s13073-021-00908-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Accepted: 05/13/2021] [Indexed: 02/06/2023] Open
Abstract
Understanding cell-type-specific gene regulatory mechanisms from genetic variants to diseases remains challenging. To address this, we developed a computational pipeline, scGRNom (single-cell Gene Regulatory Network prediction from multi-omics), to predict cell-type disease genes and regulatory networks including transcription factors and regulatory elements. With applications to schizophrenia and Alzheimer's disease, we predicted disease genes and regulatory networks for excitatory and inhibitory neurons, microglia, and oligodendrocytes. Further enrichment analyses revealed cross-disease and disease-specific functions and pathways at the cell-type level. Our machine learning analysis also found that cell-type disease genes improved clinical phenotype predictions. scGRNom is a general-purpose tool available at https://github.com/daifengwanglab/scGRNom .
Collapse
Affiliation(s)
- Ting Jin
- Department of Biostatistics and Medical Informatics, University of Wisconsin - Madison, Madison, WI, 53706, USA
- Waisman Center, University of Wisconsin - Madison, Madison, WI, 53705, USA
| | - Peter Rehani
- Waisman Center, University of Wisconsin - Madison, Madison, WI, 53705, USA
- Department of Integrative Biology, University of Wisconsin - Madison, Madison, WI, 53706, USA
- Present address: Morgridge Institute for Research, Madison, WI, 53715, USA
| | - Mufang Ying
- Department of Statistics, University of Wisconsin - Madison, Madison, WI, 53706, USA
- Present address: Department of Statistics, Rutgers University, Piscataway, NJ, 08854, USA
| | - Jiawei Huang
- Department of Statistics, University of Wisconsin - Madison, Madison, WI, 53706, USA
| | - Shuang Liu
- Waisman Center, University of Wisconsin - Madison, Madison, WI, 53705, USA
| | - Panagiotis Roussos
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Daifeng Wang
- Department of Biostatistics and Medical Informatics, University of Wisconsin - Madison, Madison, WI, 53706, USA.
- Waisman Center, University of Wisconsin - Madison, Madison, WI, 53705, USA.
- Department of Computer Sciences, University of Wisconsin - Madison, Madison, WI, 53706, USA.
| |
Collapse
|
6
|
Gruchot J, Weyers V, Göttle P, Förster M, Hartung HP, Küry P, Kremer D. The Molecular Basis for Remyelination Failure in Multiple Sclerosis. Cells 2019; 8:cells8080825. [PMID: 31382620 PMCID: PMC6721708 DOI: 10.3390/cells8080825] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 07/31/2019] [Accepted: 08/01/2019] [Indexed: 12/13/2022] Open
Abstract
Myelin sheaths in the central nervous system (CNS) insulate axons and thereby allow saltatory nerve conduction, which is a prerequisite for complex brain function. Multiple sclerosis (MS), the most common inflammatory autoimmune disease of the CNS, leads to the destruction of myelin sheaths and the myelin-producing oligodendrocytes, thus leaving behind demyelinated axons prone to injury and degeneration. Clinically, this process manifests itself in significant neurological symptoms and disability. Resident oligodendroglial precursor cells (OPCs) and neural stem cells (NSCs) are present in the adult brain, and can differentiate into mature oligodendrocytes which then remyelinate the demyelinated axons. However, for multiple reasons, in MS the regenerative capacity of these cell populations diminishes significantly over time, ultimately leading to neurodegeneration, which currently remains untreatable. In addition, microglial cells, the resident innate immune cells of the CNS, can contribute further to inflammatory and degenerative axonal damage. Here, we review the molecular factors contributing to remyelination failure in MS by inhibiting OPC and NSC differentiation or modulating microglial behavior.
Collapse
Affiliation(s)
- Joel Gruchot
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, 40225 Düsseldorf, Germany
| | - Vivien Weyers
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, 40225 Düsseldorf, Germany
| | - Peter Göttle
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, 40225 Düsseldorf, Germany
| | - Moritz Förster
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, 40225 Düsseldorf, Germany
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, 40225 Düsseldorf, Germany
| | - Patrick Küry
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, 40225 Düsseldorf, Germany
| | - David Kremer
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, 40225 Düsseldorf, Germany.
| |
Collapse
|
7
|
Ge J, Dong H, Yang Y, Liu B, Zheng M, Cheng Q, Peng L, Li J. NFIX downregulation independently predicts poor prognosis in lung adenocarcinoma, but not in squamous cell carcinoma. Future Oncol 2018; 14:3135-3144. [PMID: 30418046 DOI: 10.2217/fon-2018-0164] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
AIM To study the expression profile of NFIX, its prognostic value and the mechanism of its dysregulation in lung adenocarcinoma (LUAD). Patients & materials: A retrospective study was performed by using data from the Cancer Genome Atlas and the Human Protein Atlas. RESULTS High NFIX RNA expression was an independent prognostic factor of favorable overall survival (HR: 0.687, 95% CI: 0.496-0.951; p = 0.024) and recurrence-free survival (HR: 0.700, 95% CI: 0.493-0.994, p = 0.046) in LUAD, but not in lung squamous cell carcinoma. NFIX DNA hypermethylation was associated with significantly decreased NFIX expression and shorter overall survival and recurrence-free survival in LUAD. CONCLUSION NFIX downregulation might independently predict poor prognosis in LUAD. DNA hypermethylation might be an important cause of the downregulation.
Collapse
Affiliation(s)
- Jun Ge
- Department of Medical Oncology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science & Technology of China, Chengdu, 610041, Sichuan, PR China
| | - Hang Dong
- Department of Cancer Emergency, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science & Technology of China, Chengdu, 610041, Sichuan, PR China
| | - Ye Yang
- Department of Medical Oncology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science & Technology of China, Chengdu, 610041, Sichuan, PR China
| | - Bin Liu
- Department of Medical Oncology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science & Technology of China, Chengdu, 610041, Sichuan, PR China
| | - Min Zheng
- Department of Medical Oncology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science & Technology of China, Chengdu, 610041, Sichuan, PR China
| | - Qing Cheng
- Department of Medical Oncology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science & Technology of China, Chengdu, 610041, Sichuan, PR China
| | - Li Peng
- Department of Medical Oncology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science & Technology of China, Chengdu, 610041, Sichuan, PR China
| | - Juan Li
- Department of Medical Oncology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science & Technology of China, Chengdu, 610041, Sichuan, PR China
| |
Collapse
|
8
|
Piper M, Gronostajski R, Messina G. Nuclear Factor One X in Development and Disease. Trends Cell Biol 2018; 29:20-30. [PMID: 30287093 DOI: 10.1016/j.tcb.2018.09.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 09/03/2018] [Accepted: 09/06/2018] [Indexed: 02/06/2023]
Abstract
The past decade has seen incredible advances in the field of stem cell biology that have greatly improved our understanding of development and provided important insights into pathological processes. Transcription factors (TFs) play a central role in mediating stem cell proliferation, quiescence, and differentiation. One TF that contributes to these processes is Nuclear Factor One X (NFIX). Recently, NFIX activity has been shown to be essential in multiple organ systems and to have important translational impacts for human health. Here, we describe recent studies showing the contribution of NFIX to muscle development and muscular dystrophies, hematopoiesis, cancer, and neural stem cell biology, highlighting the importance of this knowledge in the development of therapeutic targets.
Collapse
Affiliation(s)
- Michael Piper
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia.
| | - Richard Gronostajski
- Department of Biochemistry, Genetics, Genomics & Bioinformatics Graduate Program, New York State Center of Excellence in Bioinformatics and Life Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Graziella Messina
- Department of Biosciences, University of Milan, via Celoria 26, 20133, Milan, Italy.
| |
Collapse
|
9
|
Patel R, Muir M, Cvetkovic C, Krencik R. Concepts toward directing human astroplasticity to promote neuroregeneration. Dev Dyn 2018; 248:21-33. [DOI: 10.1002/dvdy.24655] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 06/19/2018] [Accepted: 06/19/2018] [Indexed: 12/20/2022] Open
Affiliation(s)
| | | | - Caroline Cvetkovic
- Center for Neuroregeneration, Department of Neurosurgery; Houston Methodist Research Institute; Houston Texas
| | - Robert Krencik
- Center for Neuroregeneration, Department of Neurosurgery; Houston Methodist Research Institute; Houston Texas
| |
Collapse
|
10
|
Stem Cells as Potential Targets of Polyphenols in Multiple Sclerosis and Alzheimer's Disease. BIOMED RESEARCH INTERNATIONAL 2018; 2018:1483791. [PMID: 30112360 PMCID: PMC6077677 DOI: 10.1155/2018/1483791] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 06/19/2018] [Indexed: 12/16/2022]
Abstract
Alzheimer's disease (AD) and multiple sclerosis are major neurodegenerative diseases, which are characterized by the accumulation of abnormal pathogenic proteins due to oxidative stress, mitochondrial dysfunction, impaired autophagy, and pathogens, leading to neurodegeneration and behavioral deficits. Herein, we reviewed the utility of plant polyphenols in regulating proliferation and differentiation of stem cells for inducing brain self-repair in AD and multiple sclerosis. Firstly, we discussed the genetic, physiological, and environmental factors involved in the pathophysiology of both the disorders. Next, we reviewed various stem cell therapies available and how they have proved useful in animal models of AD and multiple sclerosis. Lastly, we discussed how polyphenols utilize the potential of stem cells, either complementing their therapeutic effects or stimulating endogenous and exogenous neurogenesis, against these diseases. We suggest that polyphenols could be a potential candidate for stem cell therapy against neurodegenerative disorders.
Collapse
|
11
|
Harris L, Zalucki O, Clément O, Fraser J, Matuzelski E, Oishi S, Harvey TJ, Burne THJ, Heng JIT, Gronostajski RM, Piper M. Neurogenic differentiation by hippocampal neural stem and progenitor cells is biased by NFIX expression. Development 2018; 145:145/3/dev155689. [DOI: 10.1242/dev.155689] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Accepted: 12/22/2017] [Indexed: 12/29/2022]
Abstract
ABSTRACT
Our understanding of the transcriptional programme underpinning adult hippocampal neurogenesis is incomplete. In mice, under basal conditions, adult hippocampal neural stem cells (AH-NSCs) generate neurons and astrocytes, but not oligodendrocytes. The factors limiting oligodendrocyte production, however, remain unclear. Here, we reveal that the transcription factor NFIX plays a key role in this process. NFIX is expressed by AH-NSCs, and its expression is sharply upregulated in adult hippocampal neuroblasts. Conditional ablation of Nfix from AH-NSCs, coupled with lineage tracing, transcriptomic sequencing and behavioural studies collectively reveal that NFIX is cell-autonomously required for neuroblast maturation and survival. Moreover, a small number of AH-NSCs also develop into oligodendrocytes following Nfix deletion. Remarkably, when Nfix is deleted specifically from intermediate progenitor cells and neuroblasts using a Dcx-creERT2 driver, these cells also display elevated signatures of oligodendrocyte gene expression. Together, these results demonstrate the central role played by NFIX in neuroblasts within the adult hippocampal stem cell neurogenic niche in promoting the maturation and survival of these cells, while concomitantly repressing oligodendrocyte gene expression signatures.
Collapse
Affiliation(s)
- Lachlan Harris
- The School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia 4072
| | - Oressia Zalucki
- The School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia 4072
| | - Olivier Clément
- Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia 6102
| | - James Fraser
- The School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia 4072
| | - Elise Matuzelski
- The School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia 4072
| | - Sabrina Oishi
- The School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia 4072
| | - Tracey J. Harvey
- The School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia 4072
| | - Thomas H. J. Burne
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia 4072
- Queensland Centre for Mental Health Research, The Park Centre for Mental Health, Wacol, Queensland, Australia 4076
| | - Julian Ik-Tsen Heng
- Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia 6102
| | - Richard M. Gronostajski
- Department of Biochemistry, Program in Genetics, Genomics and Bioinformatics, Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Michael Piper
- The School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia 4072
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia 4072
| |
Collapse
|
12
|
19p13 microduplications encompassing NFIX are responsible for intellectual disability, short stature and small head circumference. Eur J Hum Genet 2017; 26:85-93. [PMID: 29184170 DOI: 10.1038/s41431-017-0037-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Revised: 10/11/2017] [Accepted: 10/17/2017] [Indexed: 01/17/2023] Open
Abstract
Syndromes caused by copy number variations are described as reciprocal when they result from deletions or duplications of the same chromosomal region. When comparing the phenotypes of these syndromes, various clinical features could be described as reversed, probably due to the opposite effect of these imbalances on the expression of genes located at this locus. The NFIX gene codes for a transcription factor implicated in neurogenesis and chondrocyte differentiation. Microdeletions and loss of function variants of NFIX are responsible for Sotos syndrome-2 (also described as Malan syndrome), a syndromic form of intellectual disability associated with overgrowth and macrocephaly. Here, we report a cohort of nine patients harboring microduplications encompassing NFIX. These patients exhibit variable intellectual disability, short stature and small head circumference, which can be described as a reversed Sotos syndrome-2 phenotype. Strikingly, such a reversed phenotype has already been described in patients harboring microduplications encompassing NSD1, the gene whose deletions and loss-of-function variants are responsible for classical Sotos syndrome. Even though the type/contre-type concept has been criticized, this model seems to give a plausible explanation for the pathogenicity of 19p13 microduplications, and the common phenotype observed in our cohort.
Collapse
|
13
|
Transcriptional regulation of Nfix by NFIB drives astrocytic maturation within the developing spinal cord. Dev Biol 2017; 432:286-297. [PMID: 29106906 DOI: 10.1016/j.ydbio.2017.10.019] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 10/23/2017] [Accepted: 10/23/2017] [Indexed: 01/15/2023]
Abstract
During mouse spinal cord development, ventricular zone progenitor cells transition from producing neurons to producing glia at approximately embryonic day 11.5, a process known as the gliogenic switch. The transcription factors Nuclear Factor I (NFI) A and B initiate this developmental transition, but the contribution of a third NFI member, NFIX, remains unknown. Here, we reveal that ventricular zone progenitor cells within the spinal cord express NFIX after the onset of NFIA and NFIB expression, and after the gliogenic switch has occurred. Mice lacking NFIX exhibit normal neurogenesis within the spinal cord, and, while early astrocytic differentiation proceeds normally, aspects of terminal astrocytic differentiation are impaired. Finally, we report that, in the absence of Nfia or Nfib, there is a marked reduction in the spinal cord expression of NFIX, and that NFIB can transcriptionally activate Nfix expression in vitro. These data demonstrate that NFIX is part of the downstream transcriptional program through which NFIA and NFIB coordinate gliogenesis within the spinal cord. This hierarchical organisation of NFI protein expression and function during spinal cord gliogenesis reveals a previously unrecognised auto-regulatory mechanism within this gene family.
Collapse
|
14
|
Abstract
Tooth is made of an enamel-covered crown and a cementum-covered root. Studies on crown dentin formation have been a major focus in tooth development for several decades. Interestingly, the population prevalence for genetic short root anomaly (SRA) with no apparent defects in crown is close to 1.3%. Furthermore, people with SRA itself are predisposed to root resorption during orthodontic treatment. The discovery of the unique role of Nfic (nuclear factor I C; a transcriptional factor) in controlling root but not crown dentin formation points to a new concept: tooth crown and root have different control mechanisms. Further genetic mechanism studies have identified more key molecules (including Osterix, β-catenin, and sonic hedgehog) that play a critical role in root formation. Extensive studies have also revealed the critical role of Hertwig's epithelial root sheath in tooth root formation. In addition, Wnt10a has recently been found to be linked to multirooted tooth furcation formation. These exciting findings not only fill the critical gaps in our understanding about tooth root formation but will aid future research regarding the identifying factors controlling tooth root size and the generation of a whole "bio-tooth" for therapeutic purposes. This review starts with human SRA and mainly focuses on recent progress on the roles of NFIC-dependent and NFIC-independent signaling pathways in tooth root formation. Finally, this review includes a list of the various Cre transgenic mouse lines used to achieve tooth root formation-related gene deletion or overexpression, as well as strengths and limitations of each line.
Collapse
Affiliation(s)
- J Wang
- 1 Biomedical Sciences, Texas A&M College of Dentistry, Dallas, TX, USA
- 2 State Key Laboratory of Oral Diseases, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - J Q Feng
- 1 Biomedical Sciences, Texas A&M College of Dentistry, Dallas, TX, USA
| |
Collapse
|
15
|
Chen KS, Harris L, Lim JWC, Harvey TJ, Piper M, Gronostajski RM, Richards LJ, Bunt J. Differential neuronal and glial expression of nuclear factor I proteins in the cerebral cortex of adult mice. J Comp Neurol 2017; 525:2465-2483. [PMID: 28295292 DOI: 10.1002/cne.24206] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 02/23/2017] [Accepted: 03/02/2017] [Indexed: 12/31/2022]
Abstract
The nuclear factor I (NFI) family of transcription factors plays an important role in the development of the cerebral cortex in humans and mice. Disruption of nuclear factor IA (NFIA), nuclear factor IB (NFIB), or nuclear factor IX (NFIX) results in abnormal development of the corpus callosum, lateral ventricles, and hippocampus. However, the expression or function of these genes has not been examined in detail in the adult brain, and the cell type-specific expression of NFIA, NFIB, and NFIX is currently unknown. Here, we demonstrate that the expression of each NFI protein shows a distinct laminar pattern in the adult mouse neocortex and that their cell type-specific expression differs depending on the family member. NFIA expression was more frequently observed in astrocytes and oligodendroglia, whereas NFIB expression was predominantly localized to astrocytes and neurons. NFIX expression was most commonly observed in neurons. The NFI proteins were equally distributed within microglia, and the ependymal cells lining the ventricles of the brain expressed all three proteins. In the hippocampus, the NFI proteins were expressed during all stages of neural stem cell differentiation in the dentate gyrus, with higher expression intensity in neuroblast cells as compared to quiescent stem cells and mature granule neurons. These findings suggest that the NFI proteins may play distinct roles in cell lineage specification or maintenance, and establish the basis for further investigation of their function in the adult brain and their emerging role in disease.
Collapse
Affiliation(s)
- Kok-Siong Chen
- The Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Lachlan Harris
- The School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Jonathan W C Lim
- The Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Tracey J Harvey
- The School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Michael Piper
- The Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia.,The School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Richard M Gronostajski
- Department of Biochemistry, Program in Genetics, Genomics and Bioinformatics, Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, New York
| | - Linda J Richards
- The Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia.,The School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Jens Bunt
- The Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
16
|
Cell-type-specific expression of NFIX in the developing and adult cerebellum. Brain Struct Funct 2016; 222:2251-2270. [PMID: 27878595 DOI: 10.1007/s00429-016-1340-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 11/16/2016] [Indexed: 12/13/2022]
Abstract
Transcription factors from the nuclear factor one (NFI) family have been shown to play a central role in regulating neural progenitor cell differentiation within the embryonic and post-natal brain. NFIA and NFIB, for instance, promote the differentiation and functional maturation of granule neurons within the cerebellum. Mice lacking Nfix exhibit delays in the development of neuronal and glial lineages within the cerebellum, but the cell-type-specific expression of this transcription factor remains undefined. Here, we examined the expression of NFIX, together with various cell-type-specific markers, within the developing and adult cerebellum using both chromogenic immunohistochemistry and co-immunofluorescence labelling and confocal microscopy. In embryos, NFIX was expressed by progenitor cells within the rhombic lip and ventricular zone. After birth, progenitor cells within the external granule layer, as well as migrating and mature granule neurons, expressed NFIX. Within the adult cerebellum, NFIX displayed a broad expression profile, and was evident within granule cells, Bergmann glia, and interneurons, but not within Purkinje neurons. Furthermore, transcriptomic profiling of cerebellar granule neuron progenitor cells showed that multiple splice variants of Nfix are expressed within this germinal zone of the post-natal brain. Collectively, these data suggest that NFIX plays a role in regulating progenitor cell biology within the embryonic and post-natal cerebellum, as well as an ongoing role within multiple neuronal and glial populations within the adult cerebellum.
Collapse
|
17
|
Akkermann R, Jadasz JJ, Azim K, Küry P. Taking Advantage of Nature's Gift: Can Endogenous Neural Stem Cells Improve Myelin Regeneration? Int J Mol Sci 2016; 17:ijms17111895. [PMID: 27854261 PMCID: PMC5133894 DOI: 10.3390/ijms17111895] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 10/28/2016] [Accepted: 11/09/2016] [Indexed: 01/18/2023] Open
Abstract
Irreversible functional deficits in multiple sclerosis (MS) are directly correlated to axonal damage and loss. Neurodegeneration results from immune-mediated destruction of myelin sheaths and subsequent axonal demyelination. Importantly, oligodendrocytes, the myelinating glial cells of the central nervous system, can be replaced to some extent to generate new myelin sheaths. This endogenous regeneration capacity has so far mainly been attributed to the activation and recruitment of resident oligodendroglial precursor cells. As this self-repair process is limited and increasingly fails while MS progresses, much interest has evolved regarding the development of remyelination-promoting strategies and the presence of alternative cell types, which can also contribute to the restoration of myelin sheaths. The adult brain comprises at least two neurogenic niches harboring life-long adult neural stem cells (NSCs). An increasing number of investigations are beginning to shed light on these cells under pathological conditions and revealed a significant potential of NSCs to contribute to myelin repair activities. In this review, these emerging investigations are discussed with respect to the importance of stimulating endogenous repair mechanisms from germinal sources. Moreover, we present key findings of NSC-derived oligodendroglial progeny, including a comprehensive overview of factors and mechanisms involved in this process.
Collapse
Affiliation(s)
- Rainer Akkermann
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, 40225 Düsseldorf, Germany.
| | - Janusz Joachim Jadasz
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, 40225 Düsseldorf, Germany.
| | - Kasum Azim
- Focus Translational Neuroscience, Institute of Physiological Chemistry, University of Mainz, 55122 Mainz, Germany.
| | - Patrick Küry
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, 40225 Düsseldorf, Germany.
| |
Collapse
|
18
|
Human cerebral organoids recapitulate gene expression programs of fetal neocortex development. Proc Natl Acad Sci U S A 2015; 112:15672-7. [PMID: 26644564 DOI: 10.1073/pnas.1520760112] [Citation(s) in RCA: 749] [Impact Index Per Article: 74.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Cerebral organoids-3D cultures of human cerebral tissue derived from pluripotent stem cells-have emerged as models of human cortical development. However, the extent to which in vitro organoid systems recapitulate neural progenitor cell proliferation and neuronal differentiation programs observed in vivo remains unclear. Here we use single-cell RNA sequencing (scRNA-seq) to dissect and compare cell composition and progenitor-to-neuron lineage relationships in human cerebral organoids and fetal neocortex. Covariation network analysis using the fetal neocortex data reveals known and previously unidentified interactions among genes central to neural progenitor proliferation and neuronal differentiation. In the organoid, we detect diverse progenitors and differentiated cell types of neuronal and mesenchymal lineages and identify cells that derived from regions resembling the fetal neocortex. We find that these organoid cortical cells use gene expression programs remarkably similar to those of the fetal tissue to organize into cerebral cortex-like regions. Our comparison of in vivo and in vitro cortical single-cell transcriptomes illuminates the genetic features underlying human cortical development that can be studied in organoid cultures.
Collapse
|
19
|
Aghajanirefah A, Nguyen LN, Ohadi M. BEND3 is involved in the human-specific repression of calreticulin: Implication for the evolution of higher brain functions in human. Gene 2015; 576:577-80. [PMID: 26481236 DOI: 10.1016/j.gene.2015.10.040] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 10/01/2015] [Accepted: 10/15/2015] [Indexed: 01/27/2023]
Abstract
Recent emerging evidence indicates that changes in gene expression levels are linked to human evolution. We have previously reported a human-specific nucleotide in the promoter sequence of the calreticulin (CALR) gene at position -220C, which is the site of action of valproic acid. Reversion of this nucleotide to the ancestral A-allele has been detected in patients with degrees of deficit in higher brain cognitive functions. This mutation has since been reported in the 1000 genomes database at an approximate frequency of <0.0004 in humans (rs138452745). In the study reported here, we present update on the status of rs138452745 across evolution, based on the Ensembl and NCBI databases. The DNA pulldown assay was also used to identify the proteins binding to the C- and A-alleles, using two cell lines, SK-N-BE and HeLa. Consistent with our previous findings, the C-allele is human-specific, and the A-allele is the rule across all other species (N=38). This nucleotide resides in a block of 12-nucleotides that is strictly conserved across evolution. The DNA pulldown experiments revealed that in both SK-N-BE and HeLa cells, the transcription repressor BEN domain containing 3 (BEND3) binds to the human-specific C-allele, whereas the nuclear factor I (NFI) family members, NF1A, B, C, and X, specifically bind to the ancestral A-allele. This binding pattern is consistent with a previously reported decreased promoter activity of the C-allele vs. the A-allele. We propose that there is a link between binding of BEND3 to the CALR rs138452745 C-allele and removal of NFI binding site from this nucleotide, and the evolution of human-specific higher brain functions. To our knowledge, CALR rs138452745 is the first instance of enormous nucleotide conservation across evolution, except in the human species.
Collapse
Affiliation(s)
- A Aghajanirefah
- Department of Molecular Biology, Faculty of Science and Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen, Nijmegen, The Netherlands
| | - L N Nguyen
- Department of Molecular Biology, Faculty of Science and Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen, Nijmegen, The Netherlands
| | - M Ohadi
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran.
| |
Collapse
|