151
|
The Intricate Epigenetic and Transcriptional Alterations in Pediatric High-Grade Gliomas: Targeting the Crosstalk as the Oncogenic Achilles’ Heel. Biomedicines 2022; 10:biomedicines10061311. [PMID: 35740334 PMCID: PMC9219798 DOI: 10.3390/biomedicines10061311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 05/27/2022] [Accepted: 05/28/2022] [Indexed: 02/01/2023] Open
Abstract
Pediatric high-grade gliomas (pHGGs) are a deadly and heterogenous subgroup of gliomas for which the development of innovative treatments is urgent. Advances in high-throughput molecular techniques have shed light on key epigenetic components of these diseases, such as K27M and G34R/V mutations on histone 3. However, modification of DNA compaction is not sufficient by itself to drive those tumors. Here, we review molecular specificities of pHGGs subcategories in the context of epigenomic rewiring caused by H3 mutations and the subsequent oncogenic interplay with transcriptional signaling pathways co-opted from developmental programs that ultimately leads to gliomagenesis. Understanding how transcriptional and epigenetic alterations synergize in each cellular context in these tumors could allow the identification of new Achilles’ heels, thereby highlighting new levers to improve their therapeutic management.
Collapse
|
152
|
Brain Organization and Human Diseases. Cells 2022; 11:cells11101642. [PMID: 35626679 PMCID: PMC9139716 DOI: 10.3390/cells11101642] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 05/09/2022] [Accepted: 05/12/2022] [Indexed: 02/06/2023] Open
Abstract
The cortex is a highly organized structure that develops from the caudal regions of the segmented neural tube. Its spatial organization sets the stage for future functional arealization. Here, we suggest using a developmental perspective to describe and understand the etiology of common cortical malformations and their manifestation in the human brain.
Collapse
|
153
|
Alsagaby SA. Transcriptomics-Based Investigation of Molecular Mechanisms Underlying Apoptosis Induced by ZnO Nanoparticles in Human Diffuse Large B-Cell Lymphoma. Int J Nanomedicine 2022; 17:2261-2281. [PMID: 35611214 PMCID: PMC9124502 DOI: 10.2147/ijn.s355408] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 04/29/2022] [Indexed: 12/13/2022] Open
Abstract
Introduction Zinc oxide nanoparticles (ZnO NPs) show anti-cancer activity. Diffuse Large B-cell Lymphoma (DLBCL) is a type of B-cell malignancies with unsatisfying treatment outcomes. This study was set to assess the potential of ZnO NPs to selectively induce apoptosis in human DLBCL cells (OCI-LY3), and to describe possible molecular mechanisms of action. Methods The impact of ZnO NPs on DLBCL cells and normal peripheral blood mononuclear cells (PBMCs) was studied using cytotoxicity assay and flow-cytometry. Transcriptomics analysis was conducted to identify ZnO NPs-dependent changes in the transcriptomic profiles of DLBCL cells. Results ZnO NPs selectively induced apoptosis in DLBCL cells, and caused changes in their transcriptomes. Deferential gene expression (DGE) with fold change (FC) ≥3 and p ≤ 0.008 with corrected p ≤ 0.05 was identified for 528 genes; 125 genes were over-expressed and 403 genes were under-expressed in ZnO NPs-treated DLBCL cells. The over-expressed genes involved in biological processes and pathways like stress response to metal ion, cellular response to zinc ion, metallothioneins bind metals, oxidative stress, and negative regulation of growth. In contrast, the under-expressed genes were implicated in DNA packaging complex, signaling by NOTCH, negative regulation of gene expression by epigenetic, signaling by WNT, M phase of cell cycle, and telomere maintenance. Setting the FC to ≥1.5 with p ≤ 0.05 and corrected p ≤ 0.1 showed ZnO NPs to induce over-expression of anti-oxidant genes and under-expression of oncogenes; target B-cell receptor (BCR) signaling pathway and NF-κB pathway; and promote apoptosis by intrinsic and extrinsic pathways. Discussion Overall, ZnO NPs selectively induced apoptosis in DLBCL cells, and possible molecular mechanisms of action were described.
Collapse
Affiliation(s)
- Suliman A Alsagaby
- Department of Medical Laboratories Sciences, College of Applied Medical Sciences, Majmaah University, AL-Majmaah, 11932, Saudi Arabia
- Correspondence: Suliman A Alsagaby, Email
| |
Collapse
|
154
|
Yang Q, Hong Y, Zhao T, Song H, Ming GL. What Makes Organoids Good Models of Human Neurogenesis? Front Neurosci 2022; 16:872794. [PMID: 35495031 PMCID: PMC9048596 DOI: 10.3389/fnins.2022.872794] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 03/02/2022] [Indexed: 01/03/2023] Open
Abstract
Human neurogenesis occurs mainly in embryonic, fetal, and neonatal stages and generates tremendously diverse neural cell types that constitute the human nervous system. Studies on human neurogenesis have been limited due to a lack of access to human embryonic and fetal tissues. Brain organoids derived from human pluripotent stem cells not only recapitulate major developmental processes during neurogenesis, but also exhibit human-specific features, thus providing an unprecedented opportunity to study human neurodevelopment. First, three-dimensional brain organoids resemble early human neurogenesis with diverse stem cell pools, including the presence of primate-enriched outer radial glia cells. Second, brain organoids recapitulate human neurogenesis at the cellular level, generating diverse neuronal cell types and forming stratified cortical layers. Third, brain organoids also capture gliogenesis with the presence of human-specific astrocytes. Fourth, combined with genome-editing technologies, brain organoids are promising models for investigating functions of human-specific genes at different stages of human neurogenesis. Finally, human organoids derived from patient iPSCs can recapitulate specific disease phenotypes, providing unique models for studying developmental brain disorders of genetic and environmental causes, and for mechanistic studies and drug screening. The aim of this review is to illustrate why brain organoids are good models to study various steps of human neurogenesis, with a focus on corticogenesis. We also discuss limitations of current brain organoid models and future improvements.
Collapse
Affiliation(s)
- Qian Yang
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Yan Hong
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Ting Zhao
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Hongjun Song
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States,Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States,Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, United States,The Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Guo-li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States,Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States,Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, United States,Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States,*Correspondence: Guo-li Ming,
| |
Collapse
|
155
|
Mora-Bermúdez F, Huttner WB. What Are the Human-Specific Aspects of Neocortex Development? Front Neurosci 2022; 16:878950. [PMID: 35495057 PMCID: PMC9047014 DOI: 10.3389/fnins.2022.878950] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 03/18/2022] [Indexed: 11/13/2022] Open
Abstract
When considering what makes us human, the development of the neocortex, the seat of our higher cognitive abilities, is of central importance. Throughout this complex developmental process, neocortical stem and progenitor cells (NSPCs) exert a priming role in determining neocortical tissue fate, through a series of cellular and molecular events. In this Perspective article, we address five questions of relevance for potentially human-specific aspects of NSPCs, (i) Are there human-specific NSPC subtypes? (ii) What is the functional significance of the known temporal differences in NSPC dynamics between human and other great apes? (iii) Are there functional interactions between the human-specific genes preferentially expressed in NSPCs? (iv) Do humans amplify certain metabolic pathways for NSPC proliferation? and finally (v) Have differences evolved during human evolution, notably between modern humans and Neandertals, that affect the performance of key genes operating in NSPCs? We discuss potential implications inherent to these questions, and suggest experimental approaches on how to answer them, hoping to provide incentives to further understand key issues of human cortical development.
Collapse
Affiliation(s)
| | - Wieland B. Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| |
Collapse
|
156
|
Shinmyo Y, Hamabe-Horiike T, Saito K, Kawasaki H. Investigation of the Mechanisms Underlying the Development and Evolution of the Cerebral Cortex Using Gyrencephalic Ferrets. Front Cell Dev Biol 2022; 10:847159. [PMID: 35386196 PMCID: PMC8977464 DOI: 10.3389/fcell.2022.847159] [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: 01/01/2022] [Accepted: 02/23/2022] [Indexed: 11/13/2022] Open
Abstract
The mammalian cerebral cortex has changed significantly during evolution. As a result of the increase in the number of neurons and glial cells in the cerebral cortex, its size has markedly expanded. Moreover, folds, called gyri and sulci, appeared on its surface, and its neuronal circuits have become much more complicated. Although these changes during evolution are considered to have been crucial for the acquisition of higher brain functions, the mechanisms underlying the development and evolution of the cerebral cortex of mammals are still unclear. This is, at least partially, because it is difficult to investigate these mechanisms using mice only. Therefore, genetic manipulation techniques for the cerebral cortex of gyrencephalic carnivore ferrets were developed recently. Furthermore, gene knockout was achieved in the ferret cerebral cortex using the CRISPR/Cas9 system. These techniques enabled molecular investigations using the ferret cerebral cortex. In this review, we will summarize recent findings regarding the mechanisms underlying the development and evolution of the mammalian cerebral cortex, mainly focusing on research using ferrets.
Collapse
Affiliation(s)
- Yohei Shinmyo
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Toshihide Hamabe-Horiike
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Kengo Saito
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Hiroshi Kawasaki
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
157
|
Gershman A, Sauria MEG, Guitart X, Vollger MR, Hook PW, Hoyt SJ, Jain M, Shumate A, Razaghi R, Koren S, Altemose N, Caldas GV, Logsdon GA, Rhie A, Eichler EE, Schatz MC, O'Neill RJ, Phillippy AM, Miga KH, Timp W. Epigenetic patterns in a complete human genome. Science 2022; 376:eabj5089. [PMID: 35357915 PMCID: PMC9170183 DOI: 10.1126/science.abj5089] [Citation(s) in RCA: 166] [Impact Index Per Article: 55.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The completion of a telomere-to-telomere human reference genome, T2T-CHM13, has resolved complex regions of the genome, including repetitive and homologous regions. Here, we present a high-resolution epigenetic study of previously unresolved sequences, representing entire acrocentric chromosome short arms, gene family expansions, and a diverse collection of repeat classes. This resource precisely maps CpG methylation (32.28 million CpGs), DNA accessibility, and short-read datasets (166,058 previously unresolved chromatin immunoprecipitation sequencing peaks) to provide evidence of activity across previously unidentified or corrected genes and reveals clinically relevant paralog-specific regulation. Probing CpG methylation across human centromeres from six diverse individuals generated an estimate of variability in kinetochore localization. This analysis provides a framework with which to investigate the most elusive regions of the human genome, granting insights into epigenetic regulation.
Collapse
Affiliation(s)
- Ariel Gershman
- Department of Molecular Biology and Genetics, Johns Hopkins University, Baltimore, MD, USA
| | - Michael E G Sauria
- Department of Biology and Computer Science, Johns Hopkins University, Baltimore, MD, USA
| | - Xavi Guitart
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - Mitchell R Vollger
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - Paul W Hook
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Savannah J Hoyt
- Institute for Systems Genomics, University of Connecticut, Storrs, CT, USA
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT, USA
| | - Miten Jain
- UC Santa Cruz Genomics Institute, University of California Santa Cruz, Santa Cruz, CA, USA
| | - Alaina Shumate
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Roham Razaghi
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Sergey Koren
- Genome Informatics Section, Computational and Statistical Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Nicolas Altemose
- Department of Bioengineering, University of California Berkeley, Berkeley, CA, USA
| | - Gina V Caldas
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley CA, USA
| | - Glennis A Logsdon
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - Arang Rhie
- Genome Informatics Section, Computational and Statistical Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Evan E Eichler
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA
| | - Michael C Schatz
- Department of Biology and Computer Science, Johns Hopkins University, Baltimore, MD, USA
| | - Rachel J O'Neill
- Institute for Systems Genomics, University of Connecticut, Storrs, CT, USA
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT, USA
| | - Adam M Phillippy
- Genome Informatics Section, Computational and Statistical Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Karen H Miga
- UC Santa Cruz Genomics Institute, University of California Santa Cruz, Santa Cruz, CA, USA
| | - Winston Timp
- Department of Molecular Biology and Genetics, Johns Hopkins University, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
158
|
Vollger MR, Guitart X, Dishuck PC, Mercuri L, Harvey WT, Gershman A, Diekhans M, Sulovari A, Munson KM, Lewis AP, Hoekzema K, Porubsky D, Li R, Nurk S, Koren S, Miga KH, Phillippy AM, Timp W, Ventura M, Eichler EE. Segmental duplications and their variation in a complete human genome. Science 2022; 376:eabj6965. [PMID: 35357917 PMCID: PMC8979283 DOI: 10.1126/science.abj6965] [Citation(s) in RCA: 190] [Impact Index Per Article: 63.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Despite their importance in disease and evolution, highly identical segmental duplications (SDs) are among the last regions of the human reference genome (GRCh38) to be fully sequenced. Using a complete telomere-to-telomere human genome (T2T-CHM13), we present a comprehensive view of human SD organization. SDs account for nearly one-third of the additional sequence, increasing the genome-wide estimate from 5.4 to 7.0% [218 million base pairs (Mbp)]. An analysis of 268 human genomes shows that 91% of the previously unresolved T2T-CHM13 SD sequence (68.3 Mbp) better represents human copy number variation. Comparing long-read assemblies from human (n = 12) and nonhuman primate (n = 5) genomes, we systematically reconstruct the evolution and structural haplotype diversity of biomedically relevant and duplicated genes. This analysis reveals patterns of structural heterozygosity and evolutionary differences in SD organization between humans and other primates.
Collapse
Affiliation(s)
- Mitchell R Vollger
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - Xavi Guitart
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - Philip C Dishuck
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - Ludovica Mercuri
- Department of Biology, University of Bari, Aldo Moro, Bari 70125, Italy
| | - William T Harvey
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - Ariel Gershman
- Department of Molecular Biology and Genetics, Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Mark Diekhans
- UC Santa Cruz Genomics Institute, University of California Santa Cruz, Santa Cruz, CA, USA
| | - Arvis Sulovari
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - Katherine M Munson
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - Alexandra P Lewis
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - Kendra Hoekzema
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - David Porubsky
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - Ruiyang Li
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - Sergey Nurk
- Genome Informatics Section, Computational and Statistical Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sergey Koren
- Genome Informatics Section, Computational and Statistical Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Karen H Miga
- UC Santa Cruz Genomics Institute, University of California Santa Cruz, Santa Cruz, CA, USA
| | - Adam M Phillippy
- Genome Informatics Section, Computational and Statistical Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Winston Timp
- Department of Molecular Biology and Genetics, Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Mario Ventura
- Department of Biology, University of Bari, Aldo Moro, Bari 70125, Italy
| | - Evan E Eichler
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA
| |
Collapse
|
159
|
Nian FS, Hou PS. Evolving Roles of Notch Signaling in Cortical Development. Front Neurosci 2022; 16:844410. [PMID: 35422684 PMCID: PMC9001970 DOI: 10.3389/fnins.2022.844410] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 02/15/2022] [Indexed: 01/09/2023] Open
Abstract
Expansion of the neocortex is thought to pave the way toward acquisition of higher cognitive functions in mammals. The highly conserved Notch signaling pathway plays a crucial role in this process by regulating the size of the cortical progenitor pool, in part by controlling the balance between self-renewal and differentiation. In this review, we introduce the components of Notch signaling pathway as well as the different mode of molecular mechanisms, including trans- and cis-regulatory processes. We focused on the recent findings with regard to the expression pattern and levels in regulating neocortical formation in mammals and its interactions with other known signaling pathways, including Slit–Robo signaling and Shh signaling. Finally, we review the functions of Notch signaling pathway in different species as well as other developmental process, mainly somitogenesis, to discuss how modifications to the Notch signaling pathway can drive the evolution of the neocortex.
Collapse
Affiliation(s)
- Fang-Shin Nian
- Institute of Anatomy and Cell Biology, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Institute of Brain Science, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Pei-Shan Hou
- Institute of Anatomy and Cell Biology, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
- *Correspondence: Pei-Shan Hou,
| |
Collapse
|
160
|
Dyakonova VE. Origin and Evolution of the Nervous System: New Data from Comparative Whole Genome Studies of Multicellular Animals. Russ J Dev Biol 2022. [DOI: 10.1134/s1062360422010088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
161
|
Jaaro-Peled H, Landek-Salgado MA, Cascella NG, Nucifora FC, Coughlin JM, Nestadt G, Sedlak TW, Lavoie J, De Silva S, Lee S, Tajinda K, Hiyama H, Ishizuka K, Yang K, Sawa A. Sex-specific involvement of the Notch-JAG pathway in social recognition. Transl Psychiatry 2022; 12:99. [PMID: 35273151 PMCID: PMC8913639 DOI: 10.1038/s41398-022-01867-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 02/12/2022] [Accepted: 02/24/2022] [Indexed: 12/18/2022] Open
Abstract
Under the hypothesis that olfactory neural epithelium gene expression profiles may be useful to look for disease-relevant neuronal signatures, we examined microarray gene expression in olfactory neuronal cells and underscored Notch-JAG pathway molecules in association with schizophrenia (SZ). The microarray profiling study underscored JAG1 as the most promising candidate. Combined with further validation with real-time PCR, downregulation of NOTCH1 was statistically significant. Accordingly, we reverse-translated the significant finding from a surrogate tissue for neurons, and studied the behavioral profile of Notch1+/- mice. We found a specific impairment in social novelty recognition, whereas other behaviors, such as sociability, novel object recognition and olfaction of social odors, were normal. This social novelty recognition deficit was male-specific and was rescued by rapamycin treatment. Based on the results from the animal model, we next tested whether patients with psychosis might have male-specific alterations in social cognition in association with the expression of NOTCH1 or JAG1. In our first episode psychosis cohort, we observed a specific correlation between the expression of JAG1 and a face processing measure only in male patients. The expression of JAG1 was not correlated with any other cognitive and symptomatic scales in all subjects. Together, although we acknowledge the pioneering and exploratory nature, the present work that combines both human and animal studies in a reciprocal manner suggests a novel role for the Notch-JAG pathway in a behavioral dimension(s) related to social cognition in psychotic disorders in a male-specific manner.
Collapse
Affiliation(s)
- Hanna Jaaro-Peled
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Melissa A Landek-Salgado
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Nicola G Cascella
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Frederick C Nucifora
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Jennifer M Coughlin
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Gerald Nestadt
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Thomas W Sedlak
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Joelle Lavoie
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Sarah De Silva
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Somin Lee
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Katsunori Tajinda
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Hideki Hiyama
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Koko Ishizuka
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Kun Yang
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Akira Sawa
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
- Department of Mental Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, 21205, USA.
| |
Collapse
|
162
|
Zhu X, Guo Y, Chu C, Liu D, Duan K, Yin Y, Si C, Kang Y, Yao J, Du X, Li J, Zhao S, Ai Z, Zhu Q, Ji W, Niu Y, Li T. BRN2 as a key gene drives the early primate telencephalon development. SCIENCE ADVANCES 2022; 8:eabl7263. [PMID: 35245119 PMCID: PMC8896791 DOI: 10.1126/sciadv.abl7263] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 01/10/2022] [Indexed: 06/14/2023]
Abstract
Evolutionary mutations in primate-specific genes drove primate cortex expansion. However, whether conserved genes with previously unidentified functions also play a key role in primate brain expansion remains unknown. Here, we focus on BRN2 (POU3F2), a gene encoding a neural transcription factor commonly expressed in both primates and mice. Compared to the limited effects on mouse brain development, BRN2 biallelic knockout in cynomolgus monkeys (Macaca fascicularis) is lethal before midgestation. Histology analysis and single-cell transcriptome show that BRN2 deficiency decreases RGC expansion, induces precocious differentiation, and alters the trajectory of neurogenesis in the telencephalon. BRN2, serving as an upstream factor, controls specification and differentiation of ganglionic eminences. In addition, we identified the conserved function of BRN2 in cynomolgus monkeys to human RGCs. BRN2 may function by directly regulating SOX2 and STAT3 and maintaining HOPX. Our findings reveal a previously unknown mechanism that BRN2, a conserved gene, drives early primate telencephalon development by gaining novel mechanistic functions.
Collapse
Affiliation(s)
- Xiaoqing Zhu
- State Key Laboratory of Primate Biomedical Research and Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan 650500, China
| | - Yicheng Guo
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
| | - Chu Chu
- State Key Laboratory of Primate Biomedical Research and Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Dahai Liu
- Department of Basic Medicine and Biomedical Engineering, School of Stomatology and Medicine, Foshan University, Foshan, Guangdong 528000, China
| | - Kui Duan
- State Key Laboratory of Primate Biomedical Research and Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan 650500, China
| | - Yu Yin
- State Key Laboratory of Primate Biomedical Research and Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan 650500, China
| | - Chenyang Si
- State Key Laboratory of Primate Biomedical Research and Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan 650500, China
| | - Yu Kang
- State Key Laboratory of Primate Biomedical Research and Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan 650500, China
| | - Junjun Yao
- State Key Laboratory of Primate Biomedical Research and Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Xuewei Du
- State Key Laboratory of Primate Biomedical Research and Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Junliang Li
- State Key Laboratory of Primate Biomedical Research and Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Shumei Zhao
- State Key Laboratory of Primate Biomedical Research and Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Zongyong Ai
- State Key Laboratory of Primate Biomedical Research and Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Qingyuan Zhu
- State Key Laboratory of Primate Biomedical Research and Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Weizhi Ji
- State Key Laboratory of Primate Biomedical Research and Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan 650500, China
| | - Yuyu Niu
- State Key Laboratory of Primate Biomedical Research and Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan 650500, China
| | - Tianqing Li
- State Key Laboratory of Primate Biomedical Research and Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan 650500, China
| |
Collapse
|
163
|
Kang X, Zhang ZP, Song CG, Liu L, Zhao Y, Du JL, Lai YB, Cao XL, Ye WM, Zhang YF, Zheng MH, Zeng YH, Sun XL, Wu SX, Gao F. γ-secretase inhibitor disturbs the morphological development of differentiating neurons through affecting Notch/miR-342-5p. Neurosci Lett 2022; 778:136603. [DOI: 10.1016/j.neulet.2022.136603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 03/21/2022] [Accepted: 03/26/2022] [Indexed: 10/18/2022]
|
164
|
Boivin M, Charlet-Berguerand N. Trinucleotide CGG Repeat Diseases: An Expanding Field of Polyglycine Proteins? Front Genet 2022; 13:843014. [PMID: 35295941 PMCID: PMC8918734 DOI: 10.3389/fgene.2022.843014] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 01/31/2022] [Indexed: 12/30/2022] Open
Abstract
Microsatellites are repeated DNA sequences of 3–6 nucleotides highly variable in length and sequence and that have important roles in genomes regulation and evolution. However, expansion of a subset of these microsatellites over a threshold size is responsible of more than 50 human genetic diseases. Interestingly, some of these disorders are caused by expansions of similar sequences, sizes and localizations and present striking similarities in clinical manifestations and histopathological features, which suggest a common mechanism of disease. Notably, five identical CGG repeat expansions, but located in different genes, are the causes of fragile X-associated tremor/ataxia syndrome (FXTAS), neuronal intranuclear inclusion disease (NIID), oculopharyngodistal myopathy type 1 to 3 (OPDM1-3) and oculopharyngeal myopathy with leukoencephalopathy (OPML), which are neuromuscular and neurodegenerative syndromes with overlapping symptoms and similar histopathological features, notably the presence of characteristic eosinophilic ubiquitin-positive intranuclear inclusions. In this review we summarize recent finding in neuronal intranuclear inclusion disease and FXTAS, where the causing CGG expansions were found to be embedded within small upstream ORFs (uORFs), resulting in their translation into novel proteins containing a stretch of polyglycine (polyG). Importantly, expression of these polyG proteins is toxic in animal models and is sufficient to reproduce the formation of ubiquitin-positive intranuclear inclusions. These data suggest the existence of a novel class of human genetic pathology, the polyG diseases, and question whether a similar mechanism may exist in other diseases, notably in OPDM and OPML.
Collapse
|
165
|
Liu C, Pei M, Li Q, Zhang Y. Decellularized extracellular matrix mediates tissue construction and regeneration. Front Med 2022; 16:56-82. [PMID: 34962624 PMCID: PMC8976706 DOI: 10.1007/s11684-021-0900-3] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 09/23/2021] [Indexed: 02/05/2023]
Abstract
Contributing to organ formation and tissue regeneration, extracellular matrix (ECM) constituents provide tissue with three-dimensional (3D) structural integrity and cellular-function regulation. Containing the crucial traits of the cellular microenvironment, ECM substitutes mediate cell-matrix interactions to prompt stem-cell proliferation and differentiation for 3D organoid construction in vitro or tissue regeneration in vivo. However, these ECMs are often applied generically and have yet to be extensively developed for specific cell types in 3D cultures. Cultured cells also produce rich ECM, particularly stromal cells. Cellular ECM improves 3D culture development in vitro and tissue remodeling during wound healing after implantation into the host as well. Gaining better insight into ECM derived from either tissue or cells that regulate 3D tissue reconstruction or organ regeneration helps us to select, produce, and implant the most suitable ECM and thus promote 3D organoid culture and tissue remodeling for in vivo regeneration. Overall, the decellularization methodologies and tissue/cell-derived ECM as scaffolds or cellular-growth supplements used in cell propagation and differentiation for 3D tissue culture in vitro are discussed. Moreover, current preclinical applications by which ECM components modulate the wound-healing process are reviewed.
Collapse
Affiliation(s)
- Chuanqi Liu
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, 26506, USA
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Yuanyuan Zhang
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, 27109, USA.
| |
Collapse
|
166
|
Ossola C, Kalebic N. Roots of the Malformations of Cortical Development in the Cell Biology of Neural Progenitor Cells. Front Neurosci 2022; 15:817218. [PMID: 35069108 PMCID: PMC8766818 DOI: 10.3389/fnins.2021.817218] [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: 11/17/2021] [Accepted: 12/14/2021] [Indexed: 12/13/2022] Open
Abstract
The cerebral cortex is a structure that underlies various brain functions, including cognition and language. Mammalian cerebral cortex starts developing during the embryonic period with the neural progenitor cells generating neurons. Newborn neurons migrate along progenitors’ radial processes from the site of their origin in the germinal zones to the cortical plate, where they mature and integrate in the forming circuitry. Cell biological features of neural progenitors, such as the location and timing of their mitoses, together with their characteristic morphologies, can directly or indirectly regulate the abundance and the identity of their neuronal progeny. Alterations in the complex and delicate process of cerebral cortex development can lead to malformations of cortical development (MCDs). They include various structural abnormalities that affect the size, thickness and/or folding pattern of the developing cortex. Their clinical manifestations can entail a neurodevelopmental disorder, such as epilepsy, developmental delay, intellectual disability, or autism spectrum disorder. The recent advancements of molecular and neuroimaging techniques, along with the development of appropriate in vitro and in vivo model systems, have enabled the assessment of the genetic and environmental causes of MCDs. Here we broadly review the cell biological characteristics of neural progenitor cells and focus on those features whose perturbations have been linked to MCDs.
Collapse
|
167
|
Guo B, Zou M, Sakamoto T, Innan H. Functional Innovation through Gene Duplication Followed by Frameshift Mutation. Genes (Basel) 2022; 13:genes13020190. [PMID: 35205235 PMCID: PMC8872073 DOI: 10.3390/genes13020190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 01/14/2022] [Accepted: 01/18/2022] [Indexed: 11/16/2022] Open
Abstract
In his influential book “Evolution by Gene Duplication”, Ohno postulated that frameshift mutation could lead to a new function after duplication, but frameshift mutation is generally thought to be deleterious, and thus drew little attention in functional innovation in duplicate evolution. To this end, we here report an exhaustive survey of the genomes of human, mouse, zebrafish, and fruit fly. We identified 80 duplicate genes that involved frameshift mutations after duplication. The frameshift mutation preferentially located close to the C-terminus in most cases (55/88), which indicated that a frameshift mutation that changed the reading frame in a small part at the end of a duplicate may likely have contributed to adaptive evolution (e.g., human genes NOTCH2NL and ARHGAP11B) otherwise too deleterious to survive. A few cases (11/80) involved multiple frameshift mutations, exhibiting various patterns of modifications of the reading frame. Functionality of duplicate genes involving frameshift mutations was confirmed by sequence characteristics and expression profile, suggesting a potential role of frameshift mutation in creating functional novelty. We thus showed that genomes have non-negligible numbers of genes that have experienced frameshift mutations following gene duplication. Our results demonstrated the potential importance of frameshift mutations in molecular evolution, as Ohno verbally argued 50 years ago.
Collapse
Affiliation(s)
- Baocheng Guo
- Key Laboratory of Zoological Systematics and Evolution, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China;
- University of Chinese Academy of Sciences, Beijing 100049, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming 650223, China
- Correspondence: (B.G.); (H.I.)
| | - Ming Zou
- Key Laboratory of Zoological Systematics and Evolution, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China;
| | - Takahiro Sakamoto
- Department of Evolutionary Studies of Biosystems, Graduate University for Advanced Studies, Hayama 240-0193, Kanagawa, Japan;
| | - Hideki Innan
- Department of Evolutionary Studies of Biosystems, Graduate University for Advanced Studies, Hayama 240-0193, Kanagawa, Japan;
- Correspondence: (B.G.); (H.I.)
| |
Collapse
|
168
|
Chinnappa K, Cárdenas A, Prieto-Colomina A, Villalba A, Márquez-Galera Á, Soler R, Nomura Y, Llorens E, Tomasello U, López-Atalaya JP, Borrell V. Secondary loss of miR-3607 reduced cortical progenitor amplification during rodent evolution. SCIENCE ADVANCES 2022; 8:eabj4010. [PMID: 35020425 PMCID: PMC8754304 DOI: 10.1126/sciadv.abj4010] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
The evolutionary expansion and folding of the mammalian cerebral cortex resulted from amplification of progenitor cells during embryonic development. This process was reversed in the rodent lineage after splitting from primates, leading to smaller and smooth brains. Genetic mechanisms underlying this secondary loss in rodent evolution remain unknown. We show that microRNA miR-3607 is expressed embryonically in the large cortex of primates and ferret, distant from the primate-rodent lineage, but not in mouse. Experimental expression of miR-3607 in embryonic mouse cortex led to increased Wnt/β-catenin signaling, amplification of radial glia cells (RGCs), and expansion of the ventricular zone (VZ), via blocking the β-catenin inhibitor APC (adenomatous polyposis coli). Accordingly, loss of endogenous miR-3607 in ferret reduced RGC proliferation, while overexpression in human cerebral organoids promoted VZ expansion. Our results identify a gene selected for secondary loss during mammalian evolution to limit RGC amplification and, potentially, cortex size in rodents.
Collapse
|
169
|
Neurodegenerative diseases associated with non-coding CGG tandem repeat expansions. Nat Rev Neurol 2022; 18:145-157. [PMID: 35022573 DOI: 10.1038/s41582-021-00612-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/15/2021] [Indexed: 02/07/2023]
Abstract
Non-coding CGG repeat expansions cause multiple neurodegenerative disorders, including fragile X-associated tremor/ataxia syndrome, neuronal intranuclear inclusion disease, oculopharyngeal myopathy with leukodystrophy, and oculopharyngodistal myopathy. The underlying genetic causes of several of these diseases have been identified only in the past 2-3 years. These expansion disorders have substantial overlapping clinical, neuroimaging and histopathological features. The shared features suggest common mechanisms that could have implications for the development of therapies for this group of diseases - similar therapeutic strategies or drugs may be effective for various neurodegenerative disorders induced by non-coding CGG expansions. In this Review, we provide an overview of clinical and pathological features of these CGG repeat expansion diseases and consider the likely pathological mechanisms, including RNA toxicity, CGG repeat-associated non-AUG-initiated translation, protein aggregation and mitochondrial impairment. We then discuss future research needed to improve the identification and diagnosis of CGG repeat expansion diseases, to improve modelling of these diseases and to understand their pathogenesis. We also consider possible therapeutic strategies. Finally, we propose that CGG repeat expansion diseases may represent manifestations of a single underlying neuromyodegenerative syndrome in which different organs are affected to different extents depending on the gene location of the repeat expansion.
Collapse
|
170
|
Schmidt ERE, Polleux F. Genetic Mechanisms Underlying the Evolution of Connectivity in the Human Cortex. Front Neural Circuits 2022; 15:787164. [PMID: 35069126 PMCID: PMC8777274 DOI: 10.3389/fncir.2021.787164] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/09/2021] [Indexed: 12/22/2022] Open
Abstract
One of the most salient features defining modern humans is our remarkable cognitive capacity, which is unrivaled by any other species. Although we still lack a complete understanding of how the human brain gives rise to these unique abilities, the past several decades have witnessed significant progress in uncovering some of the genetic, cellular, and molecular mechanisms shaping the development and function of the human brain. These features include an expansion of brain size and in particular cortical expansion, distinct physiological properties of human neurons, and modified synaptic development. Together they specify the human brain as a large primate brain with a unique underlying neuronal circuit architecture. Here, we review some of the known human-specific features of neuronal connectivity, and we outline how novel insights into the human genome led to the identification of human-specific genetic modifiers that played a role in the evolution of human brain development and function. Novel experimental paradigms are starting to provide a framework for understanding how the emergence of these human-specific genomic innovations shaped the structure and function of neuronal circuits in the human brain.
Collapse
Affiliation(s)
- Ewoud R. E. Schmidt
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States
- *Correspondence: Ewoud R. E. Schmidt
| | - Franck Polleux
- Department of Neuroscience, Columbia University, New York, NY, United States
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, United States
- Kavli Institute for Brain Science, Columbia University, New York, NY, United States
- Franck Polleux
| |
Collapse
|
171
|
Using induced pluripotent stem cells to investigate human neuronal phenotypes in 1q21.1 deletion and duplication syndrome. Mol Psychiatry 2022; 27:819-830. [PMID: 34112971 PMCID: PMC9054650 DOI: 10.1038/s41380-021-01182-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 05/17/2021] [Accepted: 05/27/2021] [Indexed: 01/08/2023]
Abstract
Copy Number Variation (CNV) at the 1q21.1 locus is associated with a range of neurodevelopmental and psychiatric disorders in humans, including abnormalities in head size and motor deficits. Yet, the functional consequences of these CNVs (both deletion and duplication) on neuronal development remain unknown. To determine the impact of CNV at the 1q21.1 locus on neuronal development, we generated induced pluripotent stem cells from individuals harbouring 1q21.1 deletion or duplication and differentiated them into functional cortical neurons. We show that neurons with 1q21.1 deletion or duplication display reciprocal phenotype with respect to proliferation, differentiation potential, neuronal maturation, synaptic density and functional activity. Deletion of the 1q21.1 locus was also associated with an increased expression of lower cortical layer markers. This difference was conserved in the mouse model of 1q21.1 deletion, which displayed altered corticogenesis. Importantly, we show that neurons with 1q21.1 deletion and duplication are associated with differential expression of calcium channels and demonstrate that physiological deficits in neurons with 1q21.1 deletion or duplication can be pharmacologically modulated by targeting Ca2+ channel activity. These findings provide biological insight into the neuropathological mechanism underlying 1q21.1 associated brain disorder and indicate a potential target for therapeutic interventions.
Collapse
|
172
|
Sønderby IE, Ching CRK, Thomopoulos SI, van der Meer D, Sun D, Villalon‐Reina JE, Agartz I, Amunts K, Arango C, Armstrong NJ, Ayesa‐Arriola R, Bakker G, Bassett AS, Boomsma DI, Bülow R, Butcher NJ, Calhoun VD, Caspers S, Chow EWC, Cichon S, Ciufolini S, Craig MC, Crespo‐Facorro B, Cunningham AC, Dale AM, Dazzan P, de Zubicaray GI, Djurovic S, Doherty JL, Donohoe G, Draganski B, Durdle CA, Ehrlich S, Emanuel BS, Espeseth T, Fisher SE, Ge T, Glahn DC, Grabe HJ, Gur RE, Gutman BA, Haavik J, Håberg AK, Hansen LA, Hashimoto R, Hibar DP, Holmes AJ, Hottenga J, Hulshoff Pol HE, Jalbrzikowski M, Knowles EEM, Kushan L, Linden DEJ, Liu J, Lundervold AJ, Martin‐Brevet S, Martínez K, Mather KA, Mathias SR, McDonald‐McGinn DM, McRae AF, Medland SE, Moberget T, Modenato C, Monereo Sánchez J, Moreau CA, Mühleisen TW, Paus T, Pausova Z, Prieto C, Ragothaman A, Reinbold CS, Reis Marques T, Repetto GM, Reymond A, Roalf DR, Rodriguez‐Herreros B, Rucker JJ, Sachdev PS, Schmitt JE, Schofield PR, Silva AI, Stefansson H, Stein DJ, Tamnes CK, Tordesillas‐Gutiérrez D, Ulfarsson MO, Vajdi A, van 't Ent D, van den Bree MBM, Vassos E, Vázquez‐Bourgon J, Vila‐Rodriguez F, Walters GB, Wen W, Westlye LT, Wittfeld K, Zackai EH, Stefánsson K, Jacquemont S, et alSønderby IE, Ching CRK, Thomopoulos SI, van der Meer D, Sun D, Villalon‐Reina JE, Agartz I, Amunts K, Arango C, Armstrong NJ, Ayesa‐Arriola R, Bakker G, Bassett AS, Boomsma DI, Bülow R, Butcher NJ, Calhoun VD, Caspers S, Chow EWC, Cichon S, Ciufolini S, Craig MC, Crespo‐Facorro B, Cunningham AC, Dale AM, Dazzan P, de Zubicaray GI, Djurovic S, Doherty JL, Donohoe G, Draganski B, Durdle CA, Ehrlich S, Emanuel BS, Espeseth T, Fisher SE, Ge T, Glahn DC, Grabe HJ, Gur RE, Gutman BA, Haavik J, Håberg AK, Hansen LA, Hashimoto R, Hibar DP, Holmes AJ, Hottenga J, Hulshoff Pol HE, Jalbrzikowski M, Knowles EEM, Kushan L, Linden DEJ, Liu J, Lundervold AJ, Martin‐Brevet S, Martínez K, Mather KA, Mathias SR, McDonald‐McGinn DM, McRae AF, Medland SE, Moberget T, Modenato C, Monereo Sánchez J, Moreau CA, Mühleisen TW, Paus T, Pausova Z, Prieto C, Ragothaman A, Reinbold CS, Reis Marques T, Repetto GM, Reymond A, Roalf DR, Rodriguez‐Herreros B, Rucker JJ, Sachdev PS, Schmitt JE, Schofield PR, Silva AI, Stefansson H, Stein DJ, Tamnes CK, Tordesillas‐Gutiérrez D, Ulfarsson MO, Vajdi A, van 't Ent D, van den Bree MBM, Vassos E, Vázquez‐Bourgon J, Vila‐Rodriguez F, Walters GB, Wen W, Westlye LT, Wittfeld K, Zackai EH, Stefánsson K, Jacquemont S, Thompson PM, Bearden CE, Andreassen OA. Effects of copy number variations on brain structure and risk for psychiatric illness: Large-scale studies from the ENIGMA working groups on CNVs. Hum Brain Mapp 2022; 43:300-328. [PMID: 33615640 PMCID: PMC8675420 DOI: 10.1002/hbm.25354] [Show More Authors] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 01/07/2021] [Accepted: 01/13/2021] [Indexed: 01/21/2023] Open
Abstract
The Enhancing NeuroImaging Genetics through Meta-Analysis copy number variant (ENIGMA-CNV) and 22q11.2 Deletion Syndrome Working Groups (22q-ENIGMA WGs) were created to gain insight into the involvement of genetic factors in human brain development and related cognitive, psychiatric and behavioral manifestations. To that end, the ENIGMA-CNV WG has collated CNV and magnetic resonance imaging (MRI) data from ~49,000 individuals across 38 global research sites, yielding one of the largest studies to date on the effects of CNVs on brain structures in the general population. The 22q-ENIGMA WG includes 12 international research centers that assessed over 533 individuals with a confirmed 22q11.2 deletion syndrome, 40 with 22q11.2 duplications, and 333 typically developing controls, creating the largest-ever 22q11.2 CNV neuroimaging data set. In this review, we outline the ENIGMA infrastructure and procedures for multi-site analysis of CNVs and MRI data. So far, ENIGMA has identified effects of the 22q11.2, 16p11.2 distal, 15q11.2, and 1q21.1 distal CNVs on subcortical and cortical brain structures. Each CNV is associated with differences in cognitive, neurodevelopmental and neuropsychiatric traits, with characteristic patterns of brain structural abnormalities. Evidence of gene-dosage effects on distinct brain regions also emerged, providing further insight into genotype-phenotype relationships. Taken together, these results offer a more comprehensive picture of molecular mechanisms involved in typical and atypical brain development. This "genotype-first" approach also contributes to our understanding of the etiopathogenesis of brain disorders. Finally, we outline future directions to better understand effects of CNVs on brain structure and behavior.
Collapse
Affiliation(s)
- Ida E. Sønderby
- Department of Medical GeneticsOslo University HospitalOsloNorway
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and AddictionOslo University Hospital and University of OsloOsloNorway
- KG Jebsen Centre for Neurodevelopmental DisordersUniversity of OsloOsloNorway
| | - Christopher R. K. Ching
- Imaging Genetics CenterMark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern CaliforniaMarina del ReyCaliforniaUSA
| | - Sophia I. Thomopoulos
- Imaging Genetics CenterMark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern CaliforniaMarina del ReyCaliforniaUSA
| | - Dennis van der Meer
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and AddictionOslo University Hospital and University of OsloOsloNorway
- School of Mental Health and Neuroscience, Faculty of Health, Medicine and Life SciencesMaastricht UniversityMaastrichtThe Netherlands
| | - Daqiang Sun
- Semel Institute for Neuroscience and Human Behavior, Departments of Psychiatry and Biobehavioral Sciences and PsychologyUniversity of California Los AngelesLos AngelesCaliforniaUSA
- Department of Mental HealthVeterans Affairs Greater Los Angeles Healthcare System, Los AngelesCaliforniaUSA
| | - Julio E. Villalon‐Reina
- Imaging Genetics CenterMark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern CaliforniaMarina del ReyCaliforniaUSA
| | - Ingrid Agartz
- NORMENT, Institute of Clinical PsychiatryUniversity of OsloOsloNorway
- Department of Psychiatric ResearchDiakonhjemmet HospitalOsloNorway
- Department of Clinical NeuroscienceKarolinska InstitutetStockholmSweden
| | - Katrin Amunts
- Institute of Neuroscience and Medicine (INM‐1)Research Centre JülichJülichGermany
- Cecile and Oskar Vogt Institute for Brain Research, Medical FacultyUniversity Hospital Düsseldorf, Heinrich‐Heine‐University DüsseldorfDüsseldorfGermany
| | - Celso Arango
- Department of Child and Adolescent PsychiatryInstitute of Psychiatry and Mental Health, Hospital General Universitario Gregorio Marañon, IsSGM, Universidad Complutense, School of MedicineMadridSpain
- Centro Investigación Biomédica en Red de Salud Mental (CIBERSAM)MadridSpain
| | | | - Rosa Ayesa‐Arriola
- Centro Investigación Biomédica en Red de Salud Mental (CIBERSAM)MadridSpain
- Department of PsychiatryMarqués de Valdecilla University Hospital, Valdecilla Biomedical Research Institute (IDIVAL)SantanderSpain
| | - Geor Bakker
- Department of Psychiatry and NeuropsychologyMaastricht UniversityMaastrichtThe Netherlands
- Department of Radiology and Nuclear MedicineVU University Medical CenterAmsterdamThe Netherlands
| | - Anne S. Bassett
- Clinical Genetics Research ProgramCentre for Addiction and Mental HealthTorontoOntarioCanada
- Dalglish Family 22q Clinic for Adults with 22q11.2 Deletion Syndrome, Toronto General HospitalUniversity Health NetworkTorontoOntarioCanada
- Department of PsychiatryUniversity of TorontoTorontoOntarioCanada
| | - Dorret I. Boomsma
- Department of Biological PsychologyVrije Universiteit AmsterdamAmsterdamThe Netherlands
- Amsterdam Public Health (APH) Research InstituteAmsterdam UMCAmsterdamThe Netherlands
| | - Robin Bülow
- Institute of Diagnostic Radiology and NeuroradiologyUniversity Medicine GreifswaldGreifswaldGermany
| | - Nancy J. Butcher
- Department of PsychiatryUniversity of TorontoTorontoOntarioCanada
- Child Health Evaluative SciencesThe Hospital for Sick Children Research InstituteTorontoOntarioCanada
| | - Vince D. Calhoun
- Tri‐institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS)Georgia State, Georgia Tech, EmoryAtlantaGeorgiaUSA
| | - Svenja Caspers
- Institute of Neuroscience and Medicine (INM‐1)Research Centre JülichJülichGermany
- Institute for Anatomy IMedical Faculty & University Hospital Düsseldorf, University of DüsseldorfDüsseldorfGermany
| | - Eva W. C. Chow
- Clinical Genetics Research ProgramCentre for Addiction and Mental HealthTorontoOntarioCanada
- Department of PsychiatryUniversity of TorontoTorontoOntarioCanada
| | - Sven Cichon
- Institute of Neuroscience and Medicine (INM‐1)Research Centre JülichJülichGermany
- Institute of Medical Genetics and PathologyUniversity Hospital BaselBaselSwitzerland
- Department of BiomedicineUniversity of BaselBaselSwitzerland
| | - Simone Ciufolini
- Department of Psychosis StudiesInstitute of Psychiatry, Psychology and Neuroscience, King's College LondonLondonUnited Kingdom
| | - Michael C. Craig
- Department of Forensic and Neurodevelopmental SciencesThe Sackler Institute for Translational Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's CollegeLondonUnited Kingdom
| | | | - Adam C. Cunningham
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical NeurosciencesCardiff UniversityCardiffUnited Kingdom
| | - Anders M. Dale
- Center for Multimodal Imaging and GeneticsUniversity of California San DiegoLa JollaCaliforniaUSA
- Department RadiologyUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Paola Dazzan
- Department of Psychological MedicineInstitute of Psychiatry, Psychology and Neuroscience, King's College LondonLondonUnited Kingdom
| | - Greig I. de Zubicaray
- Faculty of HealthQueensland University of Technology (QUT)BrisbaneQueenslandAustralia
| | - Srdjan Djurovic
- Department of Medical GeneticsOslo University HospitalOsloNorway
- NORMENT, Department of Clinical ScienceUniversity of BergenBergenNorway
| | - Joanne L. Doherty
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical NeurosciencesCardiff UniversityCardiffUnited Kingdom
- Cardiff University Brain Research Imaging Centre (CUBRIC)CardiffUnited Kingdom
| | - Gary Donohoe
- Center for Neuroimaging, Genetics and GenomicsSchool of Psychology, NUI GalwayGalwayIreland
| | - Bogdan Draganski
- LREN, Centre for Research in Neuroscience, Department of NeuroscienceUniversity Hospital Lausanne and University LausanneLausanneSwitzerland
- Neurology DepartmentMax‐Planck Institute for Human Brain and Cognitive SciencesLeipzigGermany
| | - Courtney A. Durdle
- MIND Institute and Department of Psychiatry and Behavioral SciencesUniversity of California DavisDavisCaliforniaUSA
| | - Stefan Ehrlich
- Division of Psychological and Social Medicine and Developmental NeurosciencesFaculty of Medicine, TU DresdenDresdenGermany
| | - Beverly S. Emanuel
- Department of PediatricsPerelman School of Medicine at the University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Thomas Espeseth
- Department of PsychologyUniversity of OsloOsloNorway
- Department of PsychologyBjørknes CollegeOsloNorway
| | - Simon E. Fisher
- Language and Genetics DepartmentMax Planck Institute for PsycholinguisticsNijmegenThe Netherlands
- Donders Institute for Brain, Cognition and BehaviourRadboud UniversityNijmegenThe Netherlands
| | - Tian Ge
- Psychiatric and Neurodevelopmental Genetics UnitCenter for Genomic Medicine, Massachusetts General HospitalBostonMassachusettsUSA
- Department of Psychiatry, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - David C. Glahn
- Tommy Fuss Center for Neuropsychiatric Disease ResearchBoston Children's HospitalBostonMassachusettsUSA
- Department of PsychiatryHarvard Medical SchoolBostonMassachusettsUSA
| | - Hans J. Grabe
- German Center for Neurodegenerative Diseases (DZNE)Site Rostock/GreifswaldGreifswaldGermany
- Department of Psychiatry and PsychotherapyUniversity Medicine GreifswaldGreifswaldGermany
| | - Raquel E. Gur
- Department of PsychiatryUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Youth Suicide Prevention, Intervention and Research CenterChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
| | - Boris A. Gutman
- Medical Imaging Research Center, Department of Biomedical EngineeringIllinois Institute of TechnologyChicagoIllinoisUSA
| | - Jan Haavik
- Department of BiomedicineUniversity of BergenBergenNorway
- Division of PsychiatryHaukeland University HospitalBergenNorway
| | - Asta K. Håberg
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health SciencesNorwegian University of Science and TechnologyTrondheimNorway
- Department of Radiology and Nuclear MedicineSt. Olavs HospitalTrondheimNorway
| | - Laura A. Hansen
- Department of Psychiatry and Biobehavioral SciencesUniversity of California Los AngelesLos AngelesCaliforniaUSA
| | - Ryota Hashimoto
- Department of Pathology of Mental DiseasesNational Institute of Mental Health, National Center of Neurology and PsychiatryTokyoJapan
- Department of PsychiatryOsaka University Graduate School of MedicineOsakaJapan
| | - Derrek P. Hibar
- Personalized Healthcare AnalyticsGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Avram J. Holmes
- Department of PsychologyYale UniversityNew HavenConnecticutUSA
- Department of PsychiatryYale UniversityNew HavenConnecticutUSA
| | - Jouke‐Jan Hottenga
- Department of Biological PsychologyVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Hilleke E. Hulshoff Pol
- Department of Psychiatry, UMC Utrecht Brain Center, University Medical Center UtrechtUtrecht UniversityUtrechtThe Netherlands
| | | | - Emma E. M. Knowles
- Department of Psychiatry, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
- Department of PsychiatryBoston Children's HospitalBostonMassachusettsUSA
| | - Leila Kushan
- Semel Institute for Neuroscience and Human BehaviorUniversity of California Los AngelesLos AngelesCaliforniaUSA
| | - David E. J. Linden
- School for Mental Health and NeuroscienceMaastricht UniversityMaastrichtThe Netherlands
- Neuroscience and Mental Health Research InstituteCardiff UniversityCardiffUnited Kingdom
| | - Jingyu Liu
- Tri‐institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS)Georgia State, Georgia Tech, EmoryAtlantaGeorgiaUSA
- Computer ScienceGeorgia State UniversityAtlantaGeorgiaUSA
| | - Astri J. Lundervold
- Department of Biological and Medical PsychologyUniversity of BergenBergenNorway
| | - Sandra Martin‐Brevet
- LREN, Centre for Research in Neuroscience, Department of NeuroscienceUniversity Hospital Lausanne and University LausanneLausanneSwitzerland
| | - Kenia Martínez
- Department of Child and Adolescent PsychiatryInstitute of Psychiatry and Mental Health, Hospital General Universitario Gregorio Marañon, IsSGM, Universidad Complutense, School of MedicineMadridSpain
- Centro Investigación Biomédica en Red de Salud Mental (CIBERSAM)MadridSpain
- Facultad de PsicologíaUniversidad Autónoma de MadridMadridSpain
| | - Karen A. Mather
- Centre for Healthy Brain Ageing (CHeBA), School of Psychiatry, Faculty of MedicineUniversity of New South WalesSydneyNew South WalesAustralia
- Neuroscience Research AustraliaSydneyNew South WalesAustralia
| | - Samuel R. Mathias
- Department of PsychiatryHarvard Medical SchoolBostonMassachusettsUSA
- Department of PsychiatryBoston Children's HospitalBostonMassachusettsUSA
| | - Donna M. McDonald‐McGinn
- Department of PediatricsPerelman School of Medicine at the University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Division of Human GeneticsChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
- Division of Human Genetics and 22q and You CenterChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
| | - Allan F. McRae
- Institute for Molecular BioscienceThe University of QueenslandBrisbaneQueenslandAustralia
| | - Sarah E. Medland
- Psychiatric GeneticsQIMR Berghofer Medical Research InstituteBrisbaneQueenslandAustralia
| | - Torgeir Moberget
- Department of Psychology, Faculty of Social SciencesUniversity of OsloOsloNorway
| | - Claudia Modenato
- LREN, Centre for Research in Neuroscience, Department of NeuroscienceUniversity Hospital Lausanne and University LausanneLausanneSwitzerland
- University of LausanneLausanneSwitzerland
| | - Jennifer Monereo Sánchez
- School for Mental Health and NeuroscienceMaastricht UniversityMaastrichtThe Netherlands
- Faculty of Health, Medicine and Life SciencesMaastricht UniversityMaastrichtThe Netherlands
- Department of Radiology and Nuclear MedicineMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Clara A. Moreau
- Sainte Justine Hospital Research CenterUniversity of Montreal, MontrealQCCanada
| | - Thomas W. Mühleisen
- Institute of Neuroscience and Medicine (INM‐1)Research Centre JülichJülichGermany
- Cecile and Oskar Vogt Institute for Brain Research, Medical FacultyUniversity Hospital Düsseldorf, Heinrich‐Heine‐University DüsseldorfDüsseldorfGermany
- Department of BiomedicineUniversity of BaselBaselSwitzerland
| | - Tomas Paus
- Bloorview Research InstituteHolland Bloorview Kids Rehabilitation HospitalTorontoOntarioCanada
- Departments of Psychology and PsychiatryUniversity of TorontoTorontoOntarioCanada
| | - Zdenka Pausova
- Translational Medicine, The Hospital for Sick ChildrenTorontoOntarioCanada
| | - Carlos Prieto
- Bioinformatics Service, NucleusUniversity of SalamancaSalamancaSpain
| | | | - Céline S. Reinbold
- Department of BiomedicineUniversity of BaselBaselSwitzerland
- Centre for Lifespan Changes in Brain and Cognition, Department of PsychologyUniversity of OsloOsloNorway
| | - Tiago Reis Marques
- Department of Psychosis StudiesInstitute of Psychiatry, Psychology and Neuroscience, King's College LondonLondonUnited Kingdom
- Psychiatric Imaging Group, MRC London Institute of Medical Sciences (LMS), Hammersmith HospitalImperial College LondonLondonUnited Kingdom
| | - Gabriela M. Repetto
- Center for Genetics and GenomicsFacultad de Medicina, Clinica Alemana Universidad del DesarrolloSantiagoChile
| | - Alexandre Reymond
- Center for Integrative GenomicsUniversity of LausanneLausanneSwitzerland
| | - David R. Roalf
- Department of PsychiatryUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | | | - James J. Rucker
- Department of Psychological MedicineInstitute of Psychiatry, Psychology and Neuroscience, King's College LondonLondonUnited Kingdom
| | - Perminder S. Sachdev
- Centre for Healthy Brain Ageing (CHeBA), School of Psychiatry, Faculty of MedicineUniversity of New South WalesSydneyNew South WalesAustralia
- Neuropsychiatric InstituteThe Prince of Wales HospitalSydneyNew South WalesAustralia
| | - James E. Schmitt
- Department of Radiology and PsychiatryUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Peter R. Schofield
- Neuroscience Research AustraliaSydneyNew South WalesAustralia
- School of Medical SciencesUNSW SydneySydneyNew South WalesAustralia
| | - Ana I. Silva
- Neuroscience and Mental Health Research InstituteCardiff UniversityCardiffUnited Kingdom
- School for Mental Health and Neuroscience, Department of Psychiatry and Neuropsychology, Faculty of Health, Medicine and Life SciencesMaastricht UniversityMaastrichtThe Netherlands
| | | | - Dan J. Stein
- SA MRC Unit on Risk & Resilience in Mental Disorders, Department of Psychiatry and Neuroscience InstituteUniversity of Cape TownCape TownSouth Africa
| | - Christian K. Tamnes
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and AddictionOslo University Hospital and University of OsloOsloNorway
- Department of Psychiatric ResearchDiakonhjemmet HospitalOsloNorway
- PROMENTA Research Center, Department of PsychologyUniversity of OsloOsloNorway
| | - Diana Tordesillas‐Gutiérrez
- Centro Investigación Biomédica en Red de Salud Mental (CIBERSAM)MadridSpain
- Neuroimaging Unit, Technological FacilitiesValdecilla Biomedical Research Institute (IDIVAL), SantanderSpain
| | - Magnus O. Ulfarsson
- Population Genomics, deCODE genetics/AmgenReykjavikIceland
- Faculty of Electrical and Computer EngineeringUniversity of Iceland, ReykjavikIceland
| | - Ariana Vajdi
- Semel Institute for Neuroscience and Human BehaviorUniversity of California Los AngelesLos AngelesCaliforniaUSA
| | - Dennis van 't Ent
- Department of Biological PsychologyVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Marianne B. M. van den Bree
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical NeurosciencesCardiff UniversityCardiffUnited Kingdom
| | - Evangelos Vassos
- Social, Genetic and Developmental Psychiatry CentreInstitute of Psychiatry, Psychology & Neuroscience, King's College LondonLondonUnited Kingdom
| | - Javier Vázquez‐Bourgon
- Centro Investigación Biomédica en Red de Salud Mental (CIBERSAM)MadridSpain
- Department of PsychiatryMarqués de Valdecilla University Hospital, Valdecilla Biomedical Research Institute (IDIVAL)SantanderSpain
- School of MedicineUniversity of CantabriaSantanderSpain
| | - Fidel Vila‐Rodriguez
- Department of PsychiatryThe University of British ColumbiaVancouverBritish ColumbiaCanada
| | - G. Bragi Walters
- Population Genomics, deCODE genetics/AmgenReykjavikIceland
- Faculty of MedicineUniversity of IcelandReykjavikIceland
| | - Wei Wen
- Centre for Healthy Brain Ageing (CHeBA), School of Psychiatry, Faculty of MedicineUniversity of New South WalesSydneyNew South WalesAustralia
| | - Lars T. Westlye
- KG Jebsen Centre for Neurodevelopmental DisordersUniversity of OsloOsloNorway
- Department of PsychologyUniversity of OsloOsloNorway
- NORMENT, Division of Mental Health and AddictionOslo University HospitalOsloNorway
| | - Katharina Wittfeld
- German Center for Neurodegenerative Diseases (DZNE)Site Rostock/GreifswaldGreifswaldGermany
- Department of Psychiatry and PsychotherapyUniversity Medicine GreifswaldGreifswaldGermany
| | - Elaine H. Zackai
- Department of PediatricsPerelman School of Medicine at the University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Division of Human GeneticsChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
| | - Kári Stefánsson
- Population Genomics, deCODE genetics/AmgenReykjavikIceland
- Faculty of MedicineUniversity of IcelandReykjavikIceland
| | - Sebastien Jacquemont
- Sainte Justine Hospital Research CenterUniversity of Montreal, MontrealQCCanada
- Department of PediatricsUniversity of Montreal, MontrealQCCanada
| | - Paul M. Thompson
- Imaging Genetics CenterMark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern CaliforniaMarina del ReyCaliforniaUSA
| | - Carrie E. Bearden
- Semel Institute for Neuroscience and Human Behavior, Departments of Psychiatry and Biobehavioral Sciences and PsychologyUniversity of California Los AngelesLos AngelesCaliforniaUSA
- Center for Neurobehavioral GeneticsUniversity of California Los AngelesLos AngelesCaliforniaUSA
| | - Ole A. Andreassen
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and AddictionOslo University Hospital and University of OsloOsloNorway
| |
Collapse
|
173
|
Salama SR. The Complexity of the Mammalian Transcriptome. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1363:11-22. [PMID: 35220563 DOI: 10.1007/978-3-030-92034-0_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Draft genome assemblies for multiple mammalian species combined with new technologies to map transcripts from diverse RNA samples to these genomes developed in the early 2000s revealed that the mammalian transcriptome was vastly larger and more complex than previously anticipated. Efforts to comprehensively catalog the identity and features of transcripts present in a variety of species, tissues and cell lines revealed that a large fraction of the mammalian genome is transcribed in at least some settings. A large number of these transcripts encode long non-coding RNAs (lncRNAs). Many lncRNAs overlap or are anti-sense to protein coding genes and others overlap small RNAs. However, a large number are independent of any previously known mRNA or small RNA. While the functions of a majority of these lncRNAs are unknown, many appear to play roles in gene regulation. Many lncRNAs have species-specific and cell type specific expression patterns and their evolutionary origins are varied. While technological challenges have hindered getting a full picture of the diversity and transcript structure of all of the transcripts arising from lncRNA loci, new technologies including single molecule nanopore sequencing and single cell RNA sequencing promise to generate a comprehensive picture of the mammalian transcriptome.
Collapse
Affiliation(s)
- Sofie R Salama
- UC Santa Cruz Genomics Institute, Department of Biomolecular Engineering and Howard Hughes Medical Institute, University of California, Santa Cruz, Santa Cruz, CA, USA.
| |
Collapse
|
174
|
Wilder L, Semendeferi K. Infant Brain Development and Plasticity from an Evolutionary Perspective. EVOLUTIONARY PSYCHOLOGY 2022. [DOI: 10.1007/978-3-030-76000-7_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
175
|
Yanovsky-Dagan S, Frumkin A, Lupski JR, Harel T. CRISPR/Cas9-induced gene conversion between ATAD3 paralogs. HGG ADVANCES 2022; 3:100092. [PMID: 35199044 PMCID: PMC8844715 DOI: 10.1016/j.xhgg.2022.100092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 01/19/2022] [Indexed: 11/23/2022] Open
Abstract
Paralogs and pseudogenes are abundant within the human genome, and can mediate non-allelic homologous recombination (NAHR) or gene conversion events. The ATAD3 locus contains three paralogs situated in tandem, and is therefore prone to NAHR-mediated deletions and duplications associated with severe neurological phenotypes. To study this locus further, we aimed to generate biallelic loss-of-function variants in ATAD3A by CRISPR/Cas9 genome editing. Unexpectedly, two of the generated clones underwent gene conversion, as evidenced by replacement of the targeted sequence of ATAD3A by a donor sequence from its paralog ATAD3B. We highlight the complexity of CRISPR/Cas9 design, end-product formation, and recombination repair mechanisms for CRISPR/Cas9 delivery as a nucleic acid molecular therapy when targeting genes that have paralogs or pseudogenes, and advocate meticulous evaluation of resultant clones in model organisms. In addition, we suggest that endogenous gene conversion may be used to repair missense variants in genes with paralogs or pseudogenes.
Collapse
Affiliation(s)
| | - Ayala Frumkin
- Department of Genetics, Hadassah Medical Organization, Jerusalem, Israel
| | - James R. Lupski
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
- Texas Children's Hospital, Houston, TX, USA
| | - Tamar Harel
- Department of Genetics, Hadassah Medical Organization, Jerusalem, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- Corresponding author
| |
Collapse
|
176
|
Bhattacharya A, Choi WWY, Muffat J, Li Y. Modeling Developmental Brain Diseases Using Human Pluripotent Stem Cells-Derived Brain Organoids - Progress and Perspective. J Mol Biol 2021; 434:167386. [PMID: 34883115 DOI: 10.1016/j.jmb.2021.167386] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 11/26/2021] [Accepted: 11/30/2021] [Indexed: 02/07/2023]
Abstract
Developmental brain diseases encompass a group of conditions resulting from genetic or environmental perturbations during early development. Despite the increased research attention in recent years following recognition of the prevalence of these diseases, there is still a significant lack of knowledge of their etiology and treatment options. The genetic and clinical heterogeneity of these diseases, in addition to the limitations of experimental animal models, contribute to this difficulty. In this regard, the advent of brain organoid technology has provided a new means to study the cause and progression of developmental brain diseases in vitro. Derived from human pluripotent stem cells, brain organoids have been shown to recapitulate key developmental milestones of the early human brain. Combined with technological advancements in genome editing, tissue engineering, electrophysiology, and multi-omics analysis, brain organoids have expanded the frontiers of human neurobiology, providing valuable insight into the cellular and molecular mechanisms of normal and pathological brain development. This review will summarize the current progress of applying brain organoids to model human developmental brain diseases and discuss the challenges that need to be overcome to further advance their utility.
Collapse
Affiliation(s)
- Afrin Bhattacharya
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada; The University of Toronto, Department of Molecular Genetics, 1 King's College Circle, Toronto, ON M5S 1A8, Canada
| | - Wendy W Y Choi
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada; The University of Toronto, Department of Molecular Genetics, 1 King's College Circle, Toronto, ON M5S 1A8, Canada; Program in Genetics and Genome Biology, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada
| | - Julien Muffat
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada; The University of Toronto, Department of Molecular Genetics, 1 King's College Circle, Toronto, ON M5S 1A8, Canada; Program in Neurosciences and Mental Health, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada
| | - Yun Li
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada; The University of Toronto, Department of Molecular Genetics, 1 King's College Circle, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
177
|
Zhong S, Lian Y, Luo W, Luo R, Wu X, Ji J, Ji Y, Ding J, Wang X. Upstream open reading frame with NOTCH2NLC GGC expansion generates polyglycine aggregates and disrupts nucleocytoplasmic transport: implications for polyglycine diseases. Acta Neuropathol 2021; 142:1003-1023. [PMID: 34694469 DOI: 10.1007/s00401-021-02375-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 08/30/2021] [Accepted: 09/25/2021] [Indexed: 12/13/2022]
Abstract
Neuronal intranuclear inclusion disease (NIID) is neurodegenerative disease characterized by widespread inclusions. Despite the identification of GGC repeat expansion in 5'UTR of NOTCH2NLC gene in adult-onset NIIDs, its pathogenic mechanism remains unclear. Gain-of-function poly-amino-acid proteins generated by unconventional translation have been revealed in nucleotide repeat expansion disorders, inspiring us to explore the possibility of unconventional translation in NIID. Here we demonstrated that NOTCH2NLC 5'UTR triggers the translation of a polyglycine (polyG)-containing protein, N2NLCpolyG. N2NLCpolyG accumulates in p62-positive inclusions in cultured cells, mouse models, and NIID patient tissues with NOTCH2NLC GGC expansion. Translation of N2NLCpolyG is initiated by an upstream open reading frame (uORF) embedding the GGC repeats. N2NLCpolyG tends to aggregate with the increase of GGC repeat units, and displays phase separation properties. N2NLCpolyG aggregation impairs nuclear lamina and nucleocytoplasmic transport but does not necessarily cause acute death on neuronal cells. Our study suggests a similarity of pathogenic mechanisms between NIID and another GGC-repeat disease, fragile X-associated tremor ataxia syndrome. These findings expand our knowledge of protein gain-of-function in NIID, and further highlight evidence for a novel spectrum of diseases caused by aberrant polyG protein aggregation, namely the polyG diseases.
Collapse
Affiliation(s)
- Shaoping Zhong
- Department of Neurology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Yangye Lian
- Department of Neurology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Wenyi Luo
- Department of Neurology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Rongkui Luo
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaoling Wu
- Department of Neurology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Jun Ji
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuan Ji
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jing Ding
- Department of Neurology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.
- CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai, China.
| | - Xin Wang
- Department of Neurology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.
- Department of The State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China.
| |
Collapse
|
178
|
Xie S, Leung AWS, Zheng Z, Zhang D, Xiao C, Luo R, Luo M, Zhang S. Applications and potentials of nanopore sequencing in the (epi)genome and (epi)transcriptome era. Innovation (N Y) 2021; 2:100153. [PMID: 34901902 PMCID: PMC8640597 DOI: 10.1016/j.xinn.2021.100153] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 08/09/2021] [Indexed: 02/08/2023] Open
Abstract
The Human Genome Project opened an era of (epi)genomic research, and also provided a platform for the development of new sequencing technologies. During and after the project, several sequencing technologies continue to dominate nucleic acid sequencing markets. Currently, Illumina (short-read), PacBio (long-read), and Oxford Nanopore (long-read) are the most popular sequencing technologies. Unlike PacBio or the popular short-read sequencers before it, which, as examples of the second or so-called Next-Generation Sequencing platforms, need to synthesize when sequencing, nanopore technology directly sequences native DNA and RNA molecules. Nanopore sequencing, therefore, avoids converting mRNA into cDNA molecules, which not only allows for the sequencing of extremely long native DNA and full-length RNA molecules but also document modifications that have been made to those native DNA or RNA bases. In this review on direct DNA sequencing and direct RNA sequencing using Oxford Nanopore technology, we focus on their development and application achievements, discussing their challenges and future perspective. We also address the problems researchers may encounter applying these approaches in their research topics, and how to resolve them.
Collapse
Affiliation(s)
- Shangqian Xie
- Key Laboratory of Ministry of Education for Genetics and Germplasm Innovation of Tropical Special Trees and Ornamental Plants, College of Forestry, Hainan University, Haikou 570228, China
| | - Amy Wing-Sze Leung
- Department of Computer Science, The University of Hong Kong, Hong Kong 999077, China
| | - Zhenxian Zheng
- Department of Computer Science, The University of Hong Kong, Hong Kong 999077, China
| | - Dake Zhang
- Beijing Advanced Innovation Centre for Biomedical Engineering, Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Chuanle Xiao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Centre, Sun Yat-sen University, Guangzhou 510060, China
| | - Ruibang Luo
- Department of Computer Science, The University of Hong Kong, Hong Kong 999077, China
| | - Ming Luo
- Agriculture and Biotechnology Research Center, Guangdong Provincial Key Laboratory of Applied Botany, Center of Economic Botany, Core Botanical Gardens, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou 510650, China
| | - Shoudong Zhang
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong 999077, China
- Center for Soybean Research of the State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Shatin, Hong Kong 999077, China
| |
Collapse
|
179
|
Kelley KW, Pașca SP. Human brain organogenesis: Toward a cellular understanding of development and disease. Cell 2021; 185:42-61. [PMID: 34774127 DOI: 10.1016/j.cell.2021.10.003] [Citation(s) in RCA: 134] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/24/2021] [Accepted: 10/01/2021] [Indexed: 02/06/2023]
Abstract
The construction of the human nervous system is a distinctly complex although highly regulated process. Human tissue inaccessibility has impeded a molecular understanding of the developmental specializations from which our unique cognitive capacities arise. A confluence of recent technological advances in genomics and stem cell-based tissue modeling is laying the foundation for a new understanding of human neural development and dysfunction in neuropsychiatric disease. Here, we review recent progress on uncovering the cellular and molecular principles of human brain organogenesis in vivo as well as using organoids and assembloids in vitro to model features of human evolution and disease.
Collapse
Affiliation(s)
- Kevin W Kelley
- Department of Psychiatry and Behavioral Sciences, Stanford University, CA, USA; Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute, Stanford, CA, USA
| | - Sergiu P Pașca
- Department of Psychiatry and Behavioral Sciences, Stanford University, CA, USA; Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute, Stanford, CA, USA.
| |
Collapse
|
180
|
Voitiuk K, Geng J, Keefe MG, Parks DF, Sanso SE, Hawthorne N, Freeman DB, Currie R, Mostajo-Radji MA, Pollen AA, Nowakowski TJ, Salama SR, Teodorescu M, Haussler D. Light-weight electrophysiology hardware and software platform for cloud-based neural recording experiments. J Neural Eng 2021; 18:10.1088/1741-2552/ac310a. [PMID: 34666315 PMCID: PMC8667733 DOI: 10.1088/1741-2552/ac310a] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 10/19/2021] [Indexed: 11/12/2022]
Abstract
Objective.Neural activity represents a functional readout of neurons that is increasingly important to monitor in a wide range of experiments. Extracellular recordings have emerged as a powerful technique for measuring neural activity because these methods do not lead to the destruction or degradation of the cells being measured. Current approaches to electrophysiology have a low throughput of experiments due to manual supervision and expensive equipment. This bottleneck limits broader inferences that can be achieved with numerous long-term recorded samples.Approach.We developed Piphys, an inexpensive open source neurophysiological recording platform that consists of both hardware and software. It is easily accessed and controlled via a standard web interface through Internet of Things (IoT) protocols.Main results.We used a Raspberry Pi as the primary processing device along with an Intan bioamplifier. We designed a hardware expansion circuit board and software to enable voltage sampling and user interaction. This standalone system was validated with primary human neurons, showing reliability in collecting neural activity in near real-time.Significance.The hardware modules and cloud software allow for remote control of neural recording experiments as well as horizontal scalability, enabling long-term observations of development, organization, and neural activity at scale.
Collapse
Affiliation(s)
- Kateryna Voitiuk
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA 95060, United States of America
| | - Jinghui Geng
- Department of Electrical and Computer Engineering, University of California Santa Cruz, Santa Cruz, CA 95060, United States of America
| | - Matthew G Keefe
- Department of Anatomy, University of California San Francisco, San Francisco, CA 94143, United States of America
| | - David F Parks
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA 95060, United States of America
| | - Sebastian E Sanso
- UC Santa Cruz Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95060, United States of America
| | - Nico Hawthorne
- Department of Electrical and Computer Engineering, University of California Santa Cruz, Santa Cruz, CA 95060, United States of America
| | - Daniel B Freeman
- Universal Audio Inc., Scotts Valley, CA 95066, United States of America
| | - Rob Currie
- UC Santa Cruz Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95060, United States of America
| | - Mohammed A Mostajo-Radji
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA 94143, United States of America
- UC Santa Cruz Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95060, United States of America
- Department of Neurology, University of California San Francisco, San Francisco, CA 94143, United States of America
| | - Alex A Pollen
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA 94143, United States of America
- Department of Neurology, University of California San Francisco, San Francisco, CA 94143, United States of America
| | - Tomasz J Nowakowski
- Department of Anatomy, University of California San Francisco, San Francisco, CA 94143, United States of America
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA 94143, United States of America
| | - Sofie R Salama
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA 95060, United States of America
- Howard Hughes Medical Institute, University of California Santa Cruz, Santa Cruz, CA 95064, United States of America
- UC Santa Cruz Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95060, United States of America
| | - Mircea Teodorescu
- Department of Electrical and Computer Engineering, University of California Santa Cruz, Santa Cruz, CA 95060, United States of America
- UC Santa Cruz Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95060, United States of America
| | - David Haussler
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA 95060, United States of America
- Howard Hughes Medical Institute, University of California Santa Cruz, Santa Cruz, CA 95064, United States of America
- UC Santa Cruz Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95060, United States of America
| |
Collapse
|
181
|
Ma L, Du Y, Hui Y, Li N, Fan B, Zhang X, Li X, Hong W, Wu Z, Zhang S, Zhou S, Xu X, Zhou Z, Jiang C, Liu L, Zhang X. Developmental programming and lineage branching of early human telencephalon. EMBO J 2021; 40:e107277. [PMID: 34558085 DOI: 10.15252/embj.2020107277] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 08/21/2021] [Accepted: 08/24/2021] [Indexed: 01/02/2023] Open
Abstract
The dorsal and ventral human telencephalons contain different neuronal subtypes, including glutamatergic, GABAergic, and cholinergic neurons, and how these neurons are generated during early development is not well understood. Using scRNA-seq and stringent validations, we reveal here a developmental roadmap for human telencephalic neurons. Both dorsal and ventral telencephalic radial glial cells (RGs) differentiate into neurons via dividing intermediate progenitor cells (IPCs_div) and early postmitotic neuroblasts (eNBs). The transcription factor ASCL1 plays a key role in promoting fate transition from RGs to IPCs_div in both regions. RGs from the regionalized neuroectoderm show heterogeneity, with restricted glutamatergic, GABAergic, and cholinergic differentiation potencies. During neurogenesis, IPCs_div gradually exit the cell cycle and branch into sister eNBs to generate distinct neuronal subtypes. Our findings highlight a general RGs-IPCs_div-eNBs developmental scheme for human telencephalic progenitors and support that the major neuronal fates of human telencephalon are predetermined during dorsoventral regionalization with neuronal diversity being further shaped during neurogenesis and neural circuit integration.
Collapse
Affiliation(s)
- Lin Ma
- Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China.,Key Laboratory of Neuroregeneration of Shanghai Universities, School of Medicine, Tongji University, Shanghai, China.,Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China.,Department of Pathology and Pathophysiology, School of Medicine, Tongji University, Shanghai, China
| | - Yanhua Du
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Hui
- Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China.,Key Laboratory of Neuroregeneration of Shanghai Universities, School of Medicine, Tongji University, Shanghai, China.,Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China
| | - Nan Li
- Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China.,Key Laboratory of Neuroregeneration of Shanghai Universities, School of Medicine, Tongji University, Shanghai, China.,Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China
| | - Beibei Fan
- Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China.,Key Laboratory of Neuroregeneration of Shanghai Universities, School of Medicine, Tongji University, Shanghai, China.,Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China
| | - Xiaojie Zhang
- Department of Obstetrics and Gynecology, Shanghai Baoshan Luodian Hospital, Shanghai, China
| | - Xiaocui Li
- Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wei Hong
- Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhiping Wu
- Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shuwei Zhang
- Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China.,Key Laboratory of Neuroregeneration of Shanghai Universities, School of Medicine, Tongji University, Shanghai, China.,Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China
| | - Shanshan Zhou
- Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China.,Key Laboratory of Neuroregeneration of Shanghai Universities, School of Medicine, Tongji University, Shanghai, China.,Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China
| | - Xiangjie Xu
- Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China.,Key Laboratory of Neuroregeneration of Shanghai Universities, School of Medicine, Tongji University, Shanghai, China.,Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China
| | - Zhongshu Zhou
- Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China.,Key Laboratory of Neuroregeneration of Shanghai Universities, School of Medicine, Tongji University, Shanghai, China.,Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China
| | - Cizhong Jiang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, Shanghai, China
| | - Ling Liu
- Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China.,Key Laboratory of Neuroregeneration of Shanghai Universities, School of Medicine, Tongji University, Shanghai, China.,Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China.,Department of Pathology and Pathophysiology, School of Medicine, Tongji University, Shanghai, China
| | - Xiaoqing Zhang
- Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China.,Key Laboratory of Neuroregeneration of Shanghai Universities, School of Medicine, Tongji University, Shanghai, China.,Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China.,Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, Shanghai, China.,Brain and Spinal Cord Innovative Research Center, School of Medicine, Tongji University, Shanghai, China.,Tsingtao Advanced Research Institute, Tongji University, Qingdao, China
| |
Collapse
|
182
|
Modenato C, Martin-Brevet S, Moreau CA, Rodriguez-Herreros B, Kumar K, Draganski B, Sønderby IE, Jacquemont S. Lessons Learned From Neuroimaging Studies of Copy Number Variants: A Systematic Review. Biol Psychiatry 2021; 90:596-610. [PMID: 34509290 DOI: 10.1016/j.biopsych.2021.05.028] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 05/28/2021] [Accepted: 05/30/2021] [Indexed: 01/06/2023]
Abstract
Pathogenic copy number variants (CNVs) and aneuploidies alter gene dosage and are associated with neurodevelopmental psychiatric disorders such as autism spectrum disorder and schizophrenia. Brain mechanisms mediating genetic risk for neurodevelopmental psychiatric disorders remain largely unknown, but there is a rapid increase in morphometry studies of CNVs using T1-weighted structural magnetic resonance imaging. Studies have been conducted one mutation at a time, leaving the field with a complex catalog of brain alterations linked to different genomic loci. Our aim was to provide a systematic review of neuroimaging phenotypes across CNVs associated with developmental psychiatric disorders including autism and schizophrenia. We included 76 structural magnetic resonance imaging studies on 20 CNVs at the 15q11.2, 22q11.2, 1q21.1 distal, 16p11.2 distal and proximal, 7q11.23, 15q11-q13, and 22q13.33 (SHANK3) genomic loci as well as aneuploidies of chromosomes X, Y, and 21. Moderate to large effect sizes on global and regional brain morphometry are observed across all genomic loci, which is in line with levels of symptom severity reported for these variants. This is in stark contrast with the much milder neuroimaging effects observed in idiopathic psychiatric disorders. Data also suggest that CNVs have independent effects on global versus regional measures as well as on cortical surface versus thickness. Findings highlight a broad diversity of regional morphometry patterns across genomic loci. This heterogeneity of brain patterns provides insight into the weak effects reported in magnetic resonance imaging studies of cognitive dimension and psychiatric conditions. Neuroimaging studies across many more variants will be required to understand links between gene function and brain morphometry.
Collapse
Affiliation(s)
- Claudia Modenato
- Laboratory for Research in Neuroimaging, Centre for Research in Neurosciences, Department of Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Sandra Martin-Brevet
- Laboratory for Research in Neuroimaging, Centre for Research in Neurosciences, Department of Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Clara A Moreau
- Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada; Human Genetics and Cognitive Functions, Centre National de la Recherche Scientifique UMR 3571, Department of Neuroscience, Université de Paris, Institut Pasteur, Paris, France
| | - Borja Rodriguez-Herreros
- Service des Troubles du Spectre de l'Autisme et Apparentés, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Kuldeep Kumar
- Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada; Department of Pediatrics, University of Montreal, Montreal, Quebec, Canada
| | - Bogdan Draganski
- Laboratory for Research in Neuroimaging, Centre for Research in Neurosciences, Department of Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland; Neurology Department, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Ida E Sønderby
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Norwegian Centre for Mental Disorders Research, Division of Mental Health and Addiction, Oslo University Hospital, University of Oslo, Oslo, Norway; KG Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway
| | - Sébastien Jacquemont
- Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada; Department of Pediatrics, University of Montreal, Montreal, Quebec, Canada.
| |
Collapse
|
183
|
Huang XR, Tang BS, Jin P, Guo JF. The Phenotypes and Mechanisms of NOTCH2NLC-Related GGC Repeat Expansion Disorders: a Comprehensive Review. Mol Neurobiol 2021; 59:523-534. [PMID: 34718964 DOI: 10.1007/s12035-021-02616-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 10/24/2021] [Indexed: 01/11/2023]
Abstract
The human-specific gene NOTCH2NLC is primarily expressed in radial glial cells and plays an important role in neuronal differentiation and cortical neurogenesis. Increasing studies were conducted to verify the relationship between NOTCH2NLC gene and many neurological diseases, such as neuronal intranuclear inclusion disease, essential tremor, multiple system atrophy, Parkinson's disease, Alzheimer's disease, and even oculopharyngodistal myopathy. Thus, we support the concept, NOTCH2NLC-related GGC repeat expansion disorders (NRED), to summarize all diseases with the GGC repeat expansion in the 5'UTR of NOTCH2NLC gene, regardless of their various clinical phenotypes. Here, we discuss the reported cases to analyze the clinical features of NOTCH2NLC-related GGC repeat expansion disorders, including dementia, parkinsonism, peripheral neuropathy and myopathy, leukoencephalopathy, and essential tremor. In addition, we outline radiological and pathological manifestations of NOTCH2NLC-related GGC repeat expansion disorders, and then present possible mechanisms, such as toxic polyG protein, toxic repeat RNA, the GGC repeat size, and the size and types of trinucleotide interruption. Therefore, this review provides a systematic description of NOTCH2NLC-related GGC repeat expansion disorders and emphasizes the significance for understanding this type of repeat expansion disease.
Collapse
Affiliation(s)
- Xiu-Rong Huang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Bei-Sha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Centre for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, Hunan, China
| | - Peng Jin
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Ji-Feng Guo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China. .,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China. .,Centre for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China. .,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, Hunan, China.
| |
Collapse
|
184
|
Fan Y, Xu Y, Shi C. NOTCH2NLC-related disorders: the widening spectrum and genotype-phenotype correlation. J Med Genet 2021; 59:1-9. [PMID: 34675123 DOI: 10.1136/jmedgenet-2021-107883] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 09/06/2021] [Indexed: 11/04/2022]
Abstract
GGC repeat expansion in the 5' untranslated region of NOTCH2NLC is the most common causative factor in neuronal intranuclear inclusion disease (NIID) in Asians. Such expanded GGC repeats have been identified in patients with leukoencephalopathy, essential tremor (ET), multiple system atrophy, Parkinson's disease (PD), amyotrophic lateral sclerosis and oculopharyngodistal myopathy (OPDM). Herein, we review the recently reported NOTCH2NLC-related disorders and potential disease-causing mechanisms. We found that visual abnormalities may be NOTCH2NLC-specific and should be investigated in other patients with NOTCH2NLC mutations. NOTCH2NLC GGC repeat expansion was rarely identified in patients of European ancestry, whereas the actual prevalence of the expansion in European patients may be potentially higher than reported, and the CGG repeats in LRP12/GIPC1 are suggested to be screened in European patients with NIID. The repeat size and interruptions in NOTCH2NLC GGC expansion confer pleiotropic effects on clinical phenotype, a pure and stable ET phenotype may be an early symptom of NIID, and GGC repeats in NOTCH2NLC possibly give rise to ET. An association may also exist between intermediate-length NOTCH2NLC GGC repeat expansion and patients affected by PD and ET. NOTCH2NLC-OPDM highly resembles NOTCH2NLC-NIID, the two disorders may be the variations of a single neurodegenerative disease, and there may be a disease-causing upper limit in size of GGC repeats in NOTCH2NLC, repeats over which may be non-pathogenic. The haploinsufficiency of NOTCH2NLC may not be primarily involved in NOTCH2NLC-related disorders and a toxic gain-of-function mechanism possibly drives the pathogenesis of neurodegeneration in patients with NOTCH2NLC-associated disorders.
Collapse
Affiliation(s)
- Yu Fan
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China.,Academy of Medical Sciences of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Yuming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China.,Henan Key Laboratory of Cerebrovascular Diseases, Zhengzhou University, Zhengzhou, Henan, People's Republic of China.,Institute of Neuroscience, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Changhe Shi
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China .,Henan Key Laboratory of Cerebrovascular Diseases, Zhengzhou University, Zhengzhou, Henan, People's Republic of China.,Institute of Neuroscience, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| |
Collapse
|
185
|
Wang Y, Zhao B, Choi J, Lee EA. Genomic approaches to trace the history of human brain evolution with an emerging opportunity for transposon profiling of ancient humans. Mob DNA 2021; 12:22. [PMID: 34663455 PMCID: PMC8525043 DOI: 10.1186/s13100-021-00250-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 09/27/2021] [Indexed: 12/17/2022] Open
Abstract
Transposable elements (TEs) significantly contribute to shaping the diversity of the human genome, and lines of evidence suggest TEs as one of driving forces of human brain evolution. Existing computational approaches, including cross-species comparative genomics and population genetic modeling, can be adapted for the study of the role of TEs in evolution. In particular, diverse ancient and archaic human genome sequences are increasingly available, allowing reconstruction of past human migration events and holding the promise of identifying and tracking TEs among other evolutionarily important genetic variants at an unprecedented spatiotemporal resolution. However, highly degraded short DNA templates and other unique challenges presented by ancient human DNA call for major changes in current experimental and computational procedures to enable the identification of evolutionarily important TEs. Ancient human genomes are valuable resources for investigating TEs in the evolutionary context, and efforts to explore ancient human genomes will potentially provide a novel perspective on the genetic mechanism of human brain evolution and inspire a variety of technological and methodological advances. In this review, we summarize computational and experimental approaches that can be adapted to identify and validate evolutionarily important TEs, especially for human brain evolution. We also highlight strategies that leverage ancient genomic data and discuss unique challenges in ancient transposon genomics.
Collapse
Affiliation(s)
- Yilan Wang
- Division of Genetics and Genomics, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
- The Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA, USA
| | - Boxun Zhao
- Division of Genetics and Genomics, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
- The Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA, USA
| | - Jaejoon Choi
- Division of Genetics and Genomics, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Eunjung Alice Lee
- Division of Genetics and Genomics, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA.
- The Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
186
|
Abstract
The human brain is characterized by the large size and intricate folding of its cerebral cortex, which are fundamental for our higher cognitive function and frequently altered in pathological dysfunction. Cortex folding is not unique to humans, nor even to primates, but is common across mammals. Cortical growth and folding are the result of complex developmental processes that involve neural stem and progenitor cells and their cellular lineages, the migration and differentiation of neurons, and the genetic programs that regulate and fine-tune these processes. All these factors combined generate mechanical stress and strain on the developing neural tissue, which ultimately drives orderly cortical deformation and folding. In this review we examine and summarize the current knowledge on the molecular, cellular, histogenic and mechanical mechanisms that are involved in and influence folding of the cerebral cortex, and how they emerged and changed during mammalian evolution. We discuss the main types of pathological malformations of human cortex folding, their specific developmental origin, and how investigating their genetic causes has illuminated our understanding of key events involved. We close our review by presenting the state-of-the-art animal and in vitro models of cortex folding that are currently used to study these devastating developmental brain disorders in children, and what are the main challenges that remain ahead of us to fully understand brain folding.
Collapse
Affiliation(s)
- Lucia Del Valle Anton
- Instituto de Neurociencias, Agencia Estatal Consejo Superior de Investigaciones Científicas, San Juan de Alicante, Alicante, Spain
| | - Victor Borrell
- Instituto de Neurociencias, Agencia Estatal Consejo Superior de Investigaciones Científicas, San Juan de Alicante, Alicante, Spain
| |
Collapse
|
187
|
Johansson PA, Brattås PL, Douse CH, Hsieh P, Adami A, Pontis J, Grassi D, Garza R, Sozzi E, Cataldo R, Jönsson ME, Atacho DAM, Pircs K, Eren F, Sharma Y, Johansson J, Fiorenzano A, Parmar M, Fex M, Trono D, Eichler EE, Jakobsson J. A cis-acting structural variation at the ZNF558 locus controls a gene regulatory network in human brain development. Cell Stem Cell 2021; 29:52-69.e8. [PMID: 34624206 DOI: 10.1016/j.stem.2021.09.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 07/20/2021] [Accepted: 09/13/2021] [Indexed: 12/20/2022]
Abstract
The human forebrain has expanded in size and complexity compared to chimpanzees despite limited changes in protein-coding genes, suggesting that gene expression regulation is an important driver of brain evolution. Here, we identify a KRAB-ZFP transcription factor, ZNF558, that is expressed in human but not chimpanzee forebrain neural progenitor cells. ZNF558 evolved as a suppressor of LINE-1 transposons but has been co-opted to regulate a single target, the mitophagy gene SPATA18. ZNF558 plays a role in mitochondrial homeostasis, and loss-of-function experiments in cerebral organoids suggests that ZNF558 influences developmental timing during early human brain development. Expression of ZNF558 is controlled by the size of a variable number tandem repeat that is longer in chimpanzees compared to humans, and variable in the human population. Thus, this work provides mechanistic insight into how a cis-acting structural variation establishes a regulatory network that affects human brain evolution.
Collapse
Affiliation(s)
- Pia A Johansson
- Laboratory of Molecular Neurogenetics, Department of Experimental Medical Science, Wallenberg Neuroscience Center and Lund Stem Cell Center, BMC A11, Lund University, 221 84 Lund, Sweden
| | - Per Ludvik Brattås
- Laboratory of Molecular Neurogenetics, Department of Experimental Medical Science, Wallenberg Neuroscience Center and Lund Stem Cell Center, BMC A11, Lund University, 221 84 Lund, Sweden
| | - Christopher H Douse
- Laboratory of Molecular Neurogenetics, Department of Experimental Medical Science, Wallenberg Neuroscience Center and Lund Stem Cell Center, BMC A11, Lund University, 221 84 Lund, Sweden
| | - PingHsun Hsieh
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Anita Adami
- Laboratory of Molecular Neurogenetics, Department of Experimental Medical Science, Wallenberg Neuroscience Center and Lund Stem Cell Center, BMC A11, Lund University, 221 84 Lund, Sweden
| | - Julien Pontis
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Daniela Grassi
- Laboratory of Molecular Neurogenetics, Department of Experimental Medical Science, Wallenberg Neuroscience Center and Lund Stem Cell Center, BMC A11, Lund University, 221 84 Lund, Sweden
| | - Raquel Garza
- Laboratory of Molecular Neurogenetics, Department of Experimental Medical Science, Wallenberg Neuroscience Center and Lund Stem Cell Center, BMC A11, Lund University, 221 84 Lund, Sweden
| | - Edoardo Sozzi
- Developmental and Regenerative Neurobiology, Department of Experimental Medical Science, Wallenberg Neuroscience Center and Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Rodrigo Cataldo
- Lund University Diabetes Centre, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Marie E Jönsson
- Laboratory of Molecular Neurogenetics, Department of Experimental Medical Science, Wallenberg Neuroscience Center and Lund Stem Cell Center, BMC A11, Lund University, 221 84 Lund, Sweden
| | - Diahann A M Atacho
- Laboratory of Molecular Neurogenetics, Department of Experimental Medical Science, Wallenberg Neuroscience Center and Lund Stem Cell Center, BMC A11, Lund University, 221 84 Lund, Sweden
| | - Karolina Pircs
- Laboratory of Molecular Neurogenetics, Department of Experimental Medical Science, Wallenberg Neuroscience Center and Lund Stem Cell Center, BMC A11, Lund University, 221 84 Lund, Sweden
| | - Feride Eren
- Laboratory of Molecular Neurogenetics, Department of Experimental Medical Science, Wallenberg Neuroscience Center and Lund Stem Cell Center, BMC A11, Lund University, 221 84 Lund, Sweden
| | - Yogita Sharma
- Laboratory of Molecular Neurogenetics, Department of Experimental Medical Science, Wallenberg Neuroscience Center and Lund Stem Cell Center, BMC A11, Lund University, 221 84 Lund, Sweden
| | - Jenny Johansson
- Laboratory of Molecular Neurogenetics, Department of Experimental Medical Science, Wallenberg Neuroscience Center and Lund Stem Cell Center, BMC A11, Lund University, 221 84 Lund, Sweden
| | - Alessandro Fiorenzano
- Developmental and Regenerative Neurobiology, Department of Experimental Medical Science, Wallenberg Neuroscience Center and Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Malin Parmar
- Developmental and Regenerative Neurobiology, Department of Experimental Medical Science, Wallenberg Neuroscience Center and Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Malin Fex
- Lund University Diabetes Centre, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Didier Trono
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Evan E Eichler
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA; Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA
| | - Johan Jakobsson
- Laboratory of Molecular Neurogenetics, Department of Experimental Medical Science, Wallenberg Neuroscience Center and Lund Stem Cell Center, BMC A11, Lund University, 221 84 Lund, Sweden.
| |
Collapse
|
188
|
Troskie RL, Faulkner GJ, Cheetham SW. Processed pseudogenes: A substrate for evolutionary innovation: Retrotransposition contributes to genome evolution by propagating pseudogene sequences with rich regulatory potential throughout the genome. Bioessays 2021; 43:e2100186. [PMID: 34569081 DOI: 10.1002/bies.202100186] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/09/2021] [Accepted: 09/13/2021] [Indexed: 11/08/2022]
Abstract
Processed pseudogenes may serve as a genetic reservoir for evolutionary innovation. Here, we argue that through the activity of long interspersed element-1 retrotransposons, processed pseudogenes disperse coding and noncoding sequences rich with regulatory potential throughout the human genome. While these sequences may appear to be non-functional, a lack of contemporary function does not prohibit future development of biological activity. Here, we discuss the dynamic evolution of certain processed pseudogenes into coding and noncoding genes and regulatory elements, and their implication in wide-ranging biological and pathological processes. Also see the video abstract here: https://youtu.be/iUY_mteVoPI.
Collapse
Affiliation(s)
- Robin-Lee Troskie
- Mater Research Institute, University of Queensland, Woolloongabba, Australia
| | - Geoffrey J Faulkner
- Mater Research Institute, University of Queensland, Woolloongabba, Australia.,Queensland Brain Institute, University of Queensland, Brisbane, Australia
| | - Seth W Cheetham
- Mater Research Institute, University of Queensland, Woolloongabba, Australia
| |
Collapse
|
189
|
Akbaba TH, Bekircan-Kurt CE, Balci-Peynircioglu B, Balci-Hayta B. Biologia Futura: the importance of 3D organoids-a new approach for research on neurological and rare diseases. Biol Futur 2021; 72:281-290. [PMID: 34554549 DOI: 10.1007/s42977-021-00070-8] [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] [Received: 09/17/2020] [Accepted: 01/12/2021] [Indexed: 12/14/2022]
Abstract
3D cell cultures and organoid approach are increasingly being used for basic research and drug discovery of several diseases. Recent advances in these technologies, enabling research on tissue-like structures created in vitro is very important for the value of the data produced. Application of 3D cultures will not only contribute to advancing basic research, but also help to reduce animal usage in biomedical science. The 3D organoid approach is important for research on diseases where patient tissue is difficult to obtain. Therefore, this review aims to show recent advances in the 3D organoid technology in disease modeling and potential usage in translational and personalized medicine of diseases with limited patient material such as neurological diseases and rare diseases.
Collapse
Affiliation(s)
- Tayfun Hilmi Akbaba
- Department of Medical Biology, Faculty of Medicine, Hacettepe University, Ankara, 06100, Turkey
| | - Can Ebru Bekircan-Kurt
- Department of Medical Biology, Faculty of Medicine, Hacettepe University, Ankara, 06100, Turkey
- Department of Neurology, Faculty of Medicine, Hacettepe University, Ankara, 06100, Turkey
| | - Banu Balci-Peynircioglu
- Department of Medical Biology, Faculty of Medicine, Hacettepe University, Ankara, 06100, Turkey
| | - Burcu Balci-Hayta
- Department of Medical Biology, Faculty of Medicine, Hacettepe University, Ankara, 06100, Turkey.
| |
Collapse
|
190
|
Libé-Philippot B, Vanderhaeghen P. Cellular and Molecular Mechanisms Linking Human Cortical Development and Evolution. Annu Rev Genet 2021; 55:555-581. [PMID: 34535062 DOI: 10.1146/annurev-genet-071719-020705] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The cerebral cortex is at the core of brain functions that are thought to be particularly developed in the human species. Human cortex specificities stem from divergent features of corticogenesis, leading to increased cortical size and complexity. Underlying cellular mechanisms include prolonged patterns of neuronal generation and maturation, as well as the amplification of specific types of stem/progenitor cells. While the gene regulatory networks of corticogenesis appear to be largely conserved among all mammals including humans, they have evolved in primates, particularly in the human species, through the emergence of rapidly divergent transcriptional regulatory elements, as well as recently duplicated novel genes. These human-specific molecular features together control key cellular milestones of human corticogenesis and are often affected in neurodevelopmental disorders, thus linking human neural development, evolution, and diseases. Expected final online publication date for the Annual Review of Genetics, Volume 55 is November 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Baptiste Libé-Philippot
- VIB-KU Leuven Center for Brain & Disease Research, KU Leuven Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium; .,Institut de Recherches Interdisciplinaires en Biologie Humaine et Moléculaire (IRIBHM) and ULB Neuroscience Institute (UNI), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Pierre Vanderhaeghen
- VIB-KU Leuven Center for Brain & Disease Research, KU Leuven Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium; .,Institut de Recherches Interdisciplinaires en Biologie Humaine et Moléculaire (IRIBHM) and ULB Neuroscience Institute (UNI), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| |
Collapse
|
191
|
Kerimoglu C, Pham L, Tonchev AB, Sakib MS, Xie Y, Sokpor G, Ulmke PA, Kaurani L, Abbas E, Nguyen H, Rosenbusch J, Michurina A, Capece V, Angelova M, Maricic N, Brand-Saberi B, Esgleas M, Albert M, Minkov R, Kovachev E, Teichmann U, Seong RH, Huttner WB, Nguyen HP, Stoykova A, Staiger JF, Fischer A, Tuoc T. H3 acetylation selectively promotes basal progenitor proliferation and neocortex expansion. SCIENCE ADVANCES 2021; 7:eabc6792. [PMID: 34524839 PMCID: PMC8443185 DOI: 10.1126/sciadv.abc6792] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 07/23/2021] [Indexed: 06/13/2023]
Abstract
Increase in the size of human neocortex―acquired in evolution―accounts for the unique cognitive capacity of humans. This expansion reflects the evolutionarily enhanced proliferative ability of basal progenitors (BPs), including the basal radial glia and basal intermediate progenitors (bIPs) in mammalian cortex, which may have been acquired through epigenetic alterations in BPs. However, how the epigenome in BPs differs across species is not known. Here, we report that histone H3 acetylation is a key epigenetic regulation in bIP amplification and cortical expansion. Through epigenetic profiling of sorted bIPs, we show that histone H3 lysine 9 acetylation (H3K9ac) is low in murine bIPs and high in human bIPs. Elevated H3K9ac preferentially increases bIP proliferation, increasing the size and folding of the normally smooth mouse neocortex. H3K9ac drives bIP amplification by increasing expression of the evolutionarily regulated gene, Trnp1, in developing cortex. Our findings demonstrate a previously unknown mechanism that controls cortical architecture.
Collapse
Affiliation(s)
- Cemil Kerimoglu
- German Center for Neurodegenerative Diseases, 37077 Goettingen, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center, Georg-August-University Goettingen, 37075 Goettingen, Germany
| | - Linh Pham
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, 37075 Goettingen, Germany
- Department of Human Genetics, Ruhr University of Bochum, 44791 Bochum, Germany
| | - Anton B. Tonchev
- Max Planck Institute for Biophysical Chemistry, 37077 Goettingen, Germany
- Departments of Anatomy and Cell Biology and Stem Cell Biology, Research Institute, Medical University of Varna, Varna 9002, Bulgaria
| | - M. Sadman Sakib
- German Center for Neurodegenerative Diseases, 37077 Goettingen, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center, Georg-August-University Goettingen, 37075 Goettingen, Germany
| | - Yuanbin Xie
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, 37075 Goettingen, Germany
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Gannan Medical University, Ganzhou 341000, The People’s Republic of China
| | - Godwin Sokpor
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, 37075 Goettingen, Germany
- Department of Human Genetics, Ruhr University of Bochum, 44791 Bochum, Germany
| | - Pauline Antonie Ulmke
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, 37075 Goettingen, Germany
| | - Lalit Kaurani
- German Center for Neurodegenerative Diseases, 37077 Goettingen, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center, Georg-August-University Goettingen, 37075 Goettingen, Germany
| | - Eman Abbas
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, 37075 Goettingen, Germany
- Zoology Department, Faculty of Science, Alexandria University, Alexandria, Egypt
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, 01307 Dresden, Germany
| | - Huong Nguyen
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, 37075 Goettingen, Germany
- Faculty of Biotechnology, Thai Nguyen University of Sciences, Thai Nguyen, Vietnam
| | - Joachim Rosenbusch
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, 37075 Goettingen, Germany
| | | | - Vincenzo Capece
- German Center for Neurodegenerative Diseases, 37077 Goettingen, Germany
| | - Meglena Angelova
- Departments of Anatomy and Cell Biology and Stem Cell Biology, Research Institute, Medical University of Varna, Varna 9002, Bulgaria
| | - Nenad Maricic
- Institute of Anatomy and Molecular Embryology, Ruhr University of Bochum, 44791 Bochum, Germany
| | - Beate Brand-Saberi
- Institute of Anatomy and Molecular Embryology, Ruhr University of Bochum, 44791 Bochum, Germany
| | - Miriam Esgleas
- Institute of Stem Cell Research, Helmholtz Center Munich, German Research Center for Environmental Health, Munich, Germany
| | - Mareike Albert
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, 01307 Dresden, Germany
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Radoslav Minkov
- Specialized Hospital for Obstetrics and Gynecology “Prof. Dimitar Stamatov” –Varna, Medical University of Varna, Varna 9002, Bulgaria
| | - Emil Kovachev
- Specialized Hospital for Obstetrics and Gynecology “Prof. Dimitar Stamatov” –Varna, Medical University of Varna, Varna 9002, Bulgaria
| | - Ulrike Teichmann
- Max Planck Institute for Biophysical Chemistry, 37077 Goettingen, Germany
| | - Rho H. Seong
- School of Biological Sciences and Institute for Molecular Biology and Genetics, Seoul National University, Seoul, South Korea
| | - Wieland B. Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Huu Phuc Nguyen
- Department of Human Genetics, Ruhr University of Bochum, 44791 Bochum, Germany
| | - Anastassia Stoykova
- Max Planck Institute for Biophysical Chemistry, 37077 Goettingen, Germany
- DFG Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), 37075 Goettingen, Germany
| | - Jochen F. Staiger
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, 37075 Goettingen, Germany
- DFG Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), 37075 Goettingen, Germany
| | - Andre Fischer
- German Center for Neurodegenerative Diseases, 37077 Goettingen, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center, Georg-August-University Goettingen, 37075 Goettingen, Germany
- DFG Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), 37075 Goettingen, Germany
- Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells” (MBExC), Georg-August-University Goettingen, 37075 Goettingen, Germany
| | - Tran Tuoc
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, 37075 Goettingen, Germany
- Department of Human Genetics, Ruhr University of Bochum, 44791 Bochum, Germany
- DFG Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), 37075 Goettingen, Germany
| |
Collapse
|
192
|
Han S, Okawa S, Wilkinson GA, Ghazale H, Adnani L, Dixit R, Tavares L, Faisal I, Brooks MJ, Cortay V, Zinyk D, Sivitilli A, Li S, Malik F, Ilnytskyy Y, Angarica VE, Gao J, Chinchalongporn V, Oproescu AM, Vasan L, Touahri Y, David LA, Raharjo E, Kim JW, Wu W, Rahmani W, Chan JAW, Kovalchuk I, Attisano L, Kurrasch D, Dehay C, Swaroop A, Castro DS, Biernaskie J, Del Sol A, Schuurmans C. Proneural genes define ground-state rules to regulate neurogenic patterning and cortical folding. Neuron 2021; 109:2847-2863.e11. [PMID: 34407390 PMCID: PMC12080610 DOI: 10.1016/j.neuron.2021.07.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 05/19/2021] [Accepted: 07/08/2021] [Indexed: 02/06/2023]
Abstract
Asymmetric neuronal expansion is thought to drive evolutionary transitions between lissencephalic and gyrencephalic cerebral cortices. We report that Neurog2 and Ascl1 proneural genes together sustain neurogenic continuity and lissencephaly in rodent cortices. Using transgenic reporter mice and human cerebral organoids, we found that Neurog2 and Ascl1 expression defines a continuum of four lineage-biased neural progenitor cell (NPC) pools. Double+ NPCs, at the hierarchical apex, are least lineage restricted due to Neurog2-Ascl1 cross-repression and display unique features of multipotency (more open chromatin, complex gene regulatory network, G2 pausing). Strikingly, selectively eliminating double+ NPCs by crossing Neurog2-Ascl1 split-Cre mice with diphtheria toxin-dependent "deleter" strains locally disrupts Notch signaling, perturbs neurogenic symmetry, and triggers cortical folding. In support of our discovery that double+ NPCs are Notch-ligand-expressing "niche" cells that control neurogenic periodicity and cortical folding, NEUROG2, ASCL1, and HES1 transcript distribution is modular (adjacent high/low zones) in gyrencephalic macaque cortices, prefiguring future folds.
Collapse
Affiliation(s)
- Sisu Han
- Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Satoshi Okawa
- Computational Biology Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 4362 Esch-sur-Alzette, Luxembourg; Integrated BioBank of Luxembourg, 3555, 3531 Dudelange, Luxembourg
| | - Grey Atteridge Wilkinson
- Department of Biochemistry and Molecular Biology, ACHRI, HBI, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Hussein Ghazale
- Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Lata Adnani
- Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada; Department of Biochemistry and Molecular Biology, ACHRI, HBI, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Rajiv Dixit
- Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Ligia Tavares
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - Imrul Faisal
- Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Matthew J Brooks
- Neurobiology-Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD 20892-1204, USA
| | - Veronique Cortay
- Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, 69500 Bron, France
| | - Dawn Zinyk
- Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada
| | - Adam Sivitilli
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada; Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Saiqun Li
- Department of Biochemistry and Molecular Biology, ACHRI, HBI, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Faizan Malik
- Department of Medical Genetics, ACHRI, HBI, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Yaroslav Ilnytskyy
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| | - Vladimir Espinosa Angarica
- Computational Biology Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 4362 Esch-sur-Alzette, Luxembourg
| | - Jinghua Gao
- Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Vorapin Chinchalongporn
- Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Ana-Maria Oproescu
- Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Lakshmy Vasan
- Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Yacine Touahri
- Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Luke Ajay David
- Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Eko Raharjo
- Department of Comparative Biology and Experimental Medicine, HBI, ACHRI, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Jung-Woong Kim
- Neurobiology-Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD 20892-1204, USA
| | - Wei Wu
- Department of Pathology and Laboratory Medicine, Charbonneau Cancer Institute, HBI, University of Calgary, Calgary, AB T2N 4Z6, Canada
| | - Waleed Rahmani
- Department of Comparative Biology and Experimental Medicine, HBI, ACHRI, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Jennifer Ai-Wen Chan
- Department of Pathology and Laboratory Medicine, Charbonneau Cancer Institute, HBI, University of Calgary, Calgary, AB T2N 4Z6, Canada
| | - Igor Kovalchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| | - Liliana Attisano
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada; Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Deborah Kurrasch
- Department of Medical Genetics, ACHRI, HBI, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Colette Dehay
- Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, 69500 Bron, France
| | - Anand Swaroop
- Neurobiology-Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD 20892-1204, USA
| | - Diogo S Castro
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - Jeff Biernaskie
- Department of Comparative Biology and Experimental Medicine, HBI, ACHRI, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Antonio Del Sol
- Computational Biology Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 4362 Esch-sur-Alzette, Luxembourg; CIC bioGUNE, Bizkaia Technology Park, 48160 Derio, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao 48013, Spain
| | - Carol Schuurmans
- Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Biochemistry and Molecular Biology, ACHRI, HBI, University of Calgary, Calgary, AB T2N 4N1, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
193
|
Pseudogenes and their potential functions in hematopoiesis. Exp Hematol 2021; 103:24-29. [PMID: 34517065 DOI: 10.1016/j.exphem.2021.09.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 09/01/2021] [Accepted: 09/05/2021] [Indexed: 11/24/2022]
Abstract
Pseudogenes are DNA regions comprising defective copies of functional genes, the majority of which were generated by RNA- or DNA-level duplications. They exist across almost all forms of life and account for about one-quarter of the annotated genes in the human genome. Although these have been considered nonfunctional for decades, a growing number of pseudogenes have been found to be transcribed and to play crucial regulatory roles. Accumulating evidence indicates that they regulate gene expression through molecular interactions at the protein, RNA, and DNA levels. However, pseudogenes are often excluded in multiple genomewide analyses and functional screening, and their biological activities remain to be systematically disclosed. Here, we summarize the features of and progress of research on pseudogenes, in addition to discussing what is unknown about these genetic elements. Our previous findings, together with evidence of their poor conservation, prompted us to propose that pseudogenes may contribute to primate- or human-specific regulation, especially in hematopoiesis.
Collapse
|
194
|
Berto S, Liu Y, Konopka G. Genomics at cellular resolution: insights into cognitive disorders and their evolution. Hum Mol Genet 2021; 29:R1-R9. [PMID: 32566943 DOI: 10.1093/hmg/ddaa117] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 06/05/2020] [Accepted: 06/11/2020] [Indexed: 02/06/2023] Open
Abstract
High-throughput genomic sequencing approaches have held the promise of understanding and ultimately leading to treatments for cognitive disorders such as autism spectrum disorders, schizophrenia and Alzheimer's disease. Although significant progress has been made into identifying genetic variants associated with these diseases, these studies have also uncovered that these disorders are mostly genetically complex and thus challenging to model in non-human systems. Improvements in such models might benefit from understanding the evolution of the human genome and how such modifications have affected brain development and function. The intersection of genome-wide variant information with cell-type-specific expression and epigenetic information will further assist in resolving the contribution of particular cell types in evolution or disease. For example, the role of non-neuronal cells in brain evolution and cognitive disorders has gone mostly underappreciated until the recent availability of single-cell transcriptomic approaches. In this review, we discuss recent studies that carry out cell-type-specific assessments of gene expression in brain tissue across primates and between healthy and disease populations. The emerging results from these studies are beginning to elucidate how specific cell types in the evolved human brain are contributing to cognitive disorders.
Collapse
|
195
|
Gill PS, Clothier JL, Veerapandiyan A, Dweep H, Porter-Gill PA, Schaefer GB. Molecular Dysregulation in Autism Spectrum Disorder. J Pers Med 2021; 11:848. [PMID: 34575625 PMCID: PMC8466026 DOI: 10.3390/jpm11090848] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/21/2021] [Accepted: 08/26/2021] [Indexed: 12/14/2022] Open
Abstract
Autism Spectrum Disorder (ASD) comprises a heterogeneous group of neurodevelopmental disorders with a strong heritable genetic component. At present, ASD is diagnosed solely by behavioral criteria. Advances in genomic analysis have contributed to numerous candidate genes for the risk of ASD, where rare mutations and s common variants contribute to its susceptibility. Moreover, studies show rare de novo variants, copy number variation and single nucleotide polymorphisms (SNPs) also impact neurodevelopment signaling. Exploration of rare and common variants involved in common dysregulated pathways can provide new diagnostic and therapeutic strategies for ASD. Contributions of current innovative molecular strategies to understand etiology of ASD will be explored which are focused on whole exome sequencing (WES), whole genome sequencing (WGS), microRNA, long non-coding RNAs and CRISPR/Cas9 models. Some promising areas of pharmacogenomic and endophenotype directed therapies as novel personalized treatment and prevention will be discussed.
Collapse
Affiliation(s)
- Pritmohinder S. Gill
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA;
- Arkansas Children’s Research Institute, 13 Children’s Way, Little Rock, AR 72202, USA;
| | - Jeffery L. Clothier
- Psychiatric Research Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Aravindhan Veerapandiyan
- Pediatric Neurology, Arkansas Children’s Hospital, 1 Children’s Way, Little Rock, AR 72202, USA;
| | - Harsh Dweep
- The Wistar Institute, 3601 Spruce St., Philadelphia, PA 19104, USA;
| | | | - G. Bradley Schaefer
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA;
- Genetics and Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA
- Arkansas Children’s Hospital NW, Springdale, AR 72762, USA
| |
Collapse
|
196
|
Anand H, Ende V, Singh G, Qureshi I, Duong TQ, Mehler MF. Nervous System-Systemic Crosstalk in SARS-CoV-2/COVID-19: A Unique Dyshomeostasis Syndrome. Front Neurosci 2021; 15:727060. [PMID: 34512253 PMCID: PMC8430330 DOI: 10.3389/fnins.2021.727060] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 07/30/2021] [Indexed: 01/05/2023] Open
Abstract
SARS-CoV-2 infection is associated with a spectrum of acute neurological syndromes. A subset of these syndromes promotes higher in-hospital mortality than is predicted by traditional parameters defining critical care illness. This suggests that deregulation of components of the central and peripheral nervous systems compromises the interplay with systemic cellular, tissue and organ interfaces to mediate numerous atypical manifestations of COVID-19 through impairments in organismal homeostasis. This unique dyshomeostasis syndrome involves components of the ACE-2/1 lifecycles, renin-angiotensin system regulatory axes, integrated nervous system functional interactions and brain regions differentially sculpted by accelerated evolutionary processes and more primordial homeostatic functions. These biological contingencies suggest a mechanistic blueprint to define long-term neurological sequelae and systemic manifestations such as premature aging phenotypes, including organ fibrosis, tissue degeneration and cancer. Therapeutic initiatives must therefore encompass innovative combinatorial agents, including repurposing FDA-approved drugs targeting components of the autonomic nervous system and recently identified products of SARS-CoV-2-host interactions.
Collapse
Affiliation(s)
- Harnadar Anand
- The Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Victoria Ende
- Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, United States
| | - Gurinder Singh
- Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, United States
| | - Irfan Qureshi
- The Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, United States
- Biohaven Pharmaceuticals, New Haven, CT, United States
| | - Tim Q. Duong
- Department of Radiology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, United States
- Department of Physiology and Biophysics, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Mark F. Mehler
- The Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, United States
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, United States
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY, United States
- Institute for Brain Disorders and Neural Regeneration, Albert Einstein College of Medicine, Bronx, NY, United States
- Rose F. Kennedy Center for Intellectual and Developmental Disabilities, Albert Einstein College of Medicine, Bronx, NY, United States
- Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY, United States
- Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, United States
- Center for Epigenomics, Albert Einstein College of Medicine, Bronx, NY, United States
| |
Collapse
|
197
|
Evidence for opposing selective forces operating on human-specific duplicated TCAF genes in Neanderthals and humans. Nat Commun 2021; 12:5118. [PMID: 34433829 PMCID: PMC8387397 DOI: 10.1038/s41467-021-25435-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 08/04/2021] [Indexed: 11/30/2022] Open
Abstract
TRP channel-associated factor 1/2 (TCAF1/TCAF2) proteins antagonistically regulate the cold-sensor protein TRPM8 in multiple human tissues. Understanding their significance has been complicated given the locus spans a gap-ridden region with complex segmental duplications in GRCh38. Using long-read sequencing, we sequence-resolve the locus, annotate full-length TCAF models in primate genomes, and show substantial human-specific TCAF copy number variation. We identify two human super haplogroups, H4 and H5, and establish that TCAF duplications originated ~1.7 million years ago but diversified only in Homo sapiens by recurrent structural mutations. Conversely, in all archaic-hominin samples the fixation for a specific H4 haplotype without duplication is likely due to positive selection. Here, our results of TCAF copy number expansion, selection signals in hominins, and differential TCAF2 expression between haplogroups and high TCAF2 and TRPM8 expression in liver and prostate in modern-day humans imply TCAF diversification among hominins potentially in response to cold or dietary adaptations. Duplications of gene segments can allow novel physiological adaptations to evolve. A detailed analysis of the TCAF gene family in primates and archaic humans suggest rapid duplication and diversification in this gene family is associated with cold or dietary adaptations.
Collapse
|
198
|
Abstract
Although rodent research provides important insights into neural correlates of human psychology, new cortical areas, connections, and cognitive abilities emerged during primate evolution, including human evolution. Comparison of human brains with those of nonhuman primates reveals two aspects of human brain evolution particularly relevant to emotional disorders: expansion of homotypical association areas and expansion of the hippocampus. Two uniquely human cognitive capacities link these phylogenetic developments with emotion: a subjective sense of participating in and reexperiencing remembered events and a limitless capacity to imagine details of future events. These abilities provided evolving humans with selective advantages, but they also created proclivities for emotional problems. The first capacity evokes the "reliving" of past events in the "here-and-now," accompanied by emotional responses that occurred during memory encoding. It contributes to risk for stress-related syndromes, such as posttraumatic stress disorder. The second capacity, an ability to imagine future events without temporal limitations, facilitates flexible, goal-related behavior by drawing on and creating a uniquely rich array of mental representations. It promotes goal achievement and reduces errors, but the mental construction of future events also contributes to developmental aspects of anxiety and mood disorders. With maturation of homotypical association areas, the concrete concerns of childhood expand to encompass the abstract apprehensions of adolescence and adulthood. These cognitive capacities and their dysfunction are amenable to a research agenda that melds experimental therapeutic interventions, cognitive neuropsychology, and developmental psychology in both humans and nonhuman primates.
Collapse
Affiliation(s)
- Daniel S. Pine
- Section on Development and Affective Neuroscience, Emotion and Development Branch, National Institute of Mental Health, Bethesda, MD 20892
| | - Steven P. Wise
- Olschefskie Institute for the Neurobiology of Knowledge, Bethesda, MD 20814
| | - Elisabeth A. Murray
- Section on the Neurobiology of Learning and Memory, Laboratory of Neuropsychology, National Institute of Mental Health, Bethesda, MD 20892
| |
Collapse
|
199
|
Rayon T, Maizels RJ, Barrington C, Briscoe J. Single-cell transcriptome profiling of the human developing spinal cord reveals a conserved genetic programme with human-specific features. Development 2021; 148:dev199711. [PMID: 34351410 PMCID: PMC8353162 DOI: 10.1242/dev.199711] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 07/05/2021] [Indexed: 12/11/2022]
Abstract
The spinal cord receives input from peripheral sensory neurons and controls motor output by regulating muscle innervating motor neurons. These functions are carried out by neural circuits comprising molecularly distinct neuronal subtypes generated in a characteristic spatiotemporal arrangement from progenitors in the embryonic neural tube. To gain insight into the diversity and complexity of cells in the developing human neural tube, we used single-cell mRNA sequencing to profile cervical and thoracic regions in four human embryos of Carnegie stages (CS) CS12, CS14, CS17 and CS19 from gestational weeks 4-7. Analysis of progenitor and neuronal populations from the neural tube and dorsal root ganglia identified dozens of distinct cell types and facilitated the reconstruction of the differentiation pathways of specific neuronal subtypes. Comparison with mouse revealed overall similarity of mammalian neural tube development while highlighting some human-specific features. These data provide a catalogue of gene expression and cell type identity in the human neural tube that will support future studies of sensory and motor control systems. The data can be explored at https://shiny.crick.ac.uk/scviewer/neuraltube/.
Collapse
|
200
|
Cao L, Yan Y, Zhao G. NOTCH2NLC-related repeat expansion disorders: an expanding group of neurodegenerative disorders. Neurol Sci 2021; 42:4055-4062. [PMID: 34333668 DOI: 10.1007/s10072-021-05498-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 07/18/2021] [Indexed: 12/22/2022]
Abstract
The NOTCH2NLC gene 5' untranslated region (UTR) GGC repeat expansion mutations were identified as a genetic contributor of neuronal intranuclear inclusion disease (NIID) in 2019. Since then, the number of reported cases with NOTCH2NLC GGC repeat expansion in Asian and European populations has increased rapidly, indicating that the expanded mutation not only leads to the onset or progression of the NIID, but also may play an important role in multiple progressive neurological disorders, including Parkinson's disease, essential tremor, multiple system atrophy, Alzheimer's disease, frontotemporal dementia, amyotrophic lateral sclerosis, leukoencephalopathy, and oculopharyngodistal myopathy type 3. Nevertheless, the underlying pathogenic mechanism of the NOTCH2NLC 5' UTR region GGC repeat expansion in these disorders remains largely unknown. This review aims to present recent breakthroughs on this mutation and improve our knowledge of a newly defined spectrum of disease: NOTCH2NLC-related repeat expansion disorder.
Collapse
Affiliation(s)
- Lanxiao Cao
- Department of Neurology, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, N1 Shangcheng Avenue, Yiwu, 322000, Zhejiang Province, China
| | - Yaping Yan
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, No.88 Jiefang Road, Hangzhou, 310009, Zhejiang Province, China.
| | - Guohua Zhao
- Department of Neurology, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, N1 Shangcheng Avenue, Yiwu, 322000, Zhejiang Province, China.
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, No.88 Jiefang Road, Hangzhou, 310009, Zhejiang Province, China.
| |
Collapse
|