1
|
Martin-Lopez E, Brennan B, Lefèvre M, Spence NJ, Han K, Greer CA. Laminar organization of the anterior olfactory nucleus-the interplay between neurogenesis timing and neuroblast migration. Front Neurosci 2025; 19:1546397. [PMID: 40370659 PMCID: PMC12075217 DOI: 10.3389/fnins.2025.1546397] [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: 12/16/2024] [Accepted: 03/31/2025] [Indexed: 05/16/2025] Open
Abstract
Introduction The anterior olfactory nucleus (AON) is a laminar structure embedded within the olfactory peduncle which serves as the conduit for connectivity between the olfactory bulb (OB) and the central processing centers of the brain. The largest portion of the AON is a ring of neurons and fibers that surround the core of the peduncle, the pars principalis (AONpP). The AONpP is further subdivided into an outer plexiform layer, or layer 1 (L1), that contains axons and dendrites, and an inner cell zone, or layer 2 (L2), formed by densely packed pyramidal cells. Relative to other regions of the olfactory system, the development of the AON remains poorly understood. Methods We performed injections of thymidine analogs in pregnant mice from E10 to E18 to determine the timeline of AON neurogenesis and used immunohistochemistry to study neuronal phenotypes both at adult and embryonic stages. To better understand migration and differentiation of the AON neurons, we labeled AON precursors using in utero electroporations with the piggyBac transposon into the rostral lateral ganglionic eminence, the embryonic source of AON neurons. Results Our analyses established that the earliest neurons targeted to the AON laminae arose at E10 with neurogenesis peaking at E13. In L1, we found a caudal-to-rostral neurogenic gradient not detected in L2. Quantification across the cardinal axes showed no gradients in L2 and a medial-to-lateral gradient for L1. Using immunohistochemistry, we found that AON neurons express the most common cortical markers Tbr1, Ctip2, NeuroD1, Sox5 and Cux1+2 at adult stages without laminar distinction. Tbr1 and NeuroD1 first appeared embryonically at E12, while Ctip2 and Sox5 were present at E13, following a dorsal-ventral pattern. Cux1+2 was not detected embryonically. Embryonically, our data on neuroblasts migration revealed that AON neuroblasts use a scaffold of radial glia to migrate to their final destinations in both L1 and L2 through a caudal-to-rostral migratory gradient. Conclusion For the first time, our data show a comprehensive timeline for the AON neurogenesis across the anatomical axes, and a detailed analysis on neuroblast migration in the mouse embryo. These data are crucial to understanding the embryonic formation and relationship of relay stations along the olfactory pathway.
Collapse
Affiliation(s)
- Eduardo Martin-Lopez
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT, United States
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, United States
| | - Bowen Brennan
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT, United States
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, United States
| | - Marion Lefèvre
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT, United States
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, United States
| | - Natalie J. Spence
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT, United States
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, United States
| | - Kimberly Han
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT, United States
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, United States
| | - Charles A. Greer
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT, United States
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, United States
- Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
2
|
Li W, Su D, Li X, Lu K, Huang Q, Zheng J, Luo X, Chen G, Fan X. Identification of the core regulatory program driving NEUROD1-induced neuronal reprogramming. Cell Rep 2025; 44:115523. [PMID: 40173039 DOI: 10.1016/j.celrep.2025.115523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 02/03/2025] [Accepted: 03/14/2025] [Indexed: 04/04/2025] Open
Abstract
NEUROD1 (ND1)-induced astrocyte-to-neuron (AtN) conversion shows promise for treating neurological disorders. To gain insight into the molecular mechanisms of neuronal reprogramming, we established an in vitro system using primary cortical astrocyte cultures from postnatal rats and employed single-cell and multiomics sequencing. Our findings indicate that the initial cultures primarily consisted of immature astrocytes (ImAs), with potentially a minor presence of radial glial cells. The ImAs initially went through an intermediate state, activating both astrocyte and neural progenitor genes. Subsequently, they mimic in vivo neurogenesis to acquire mature neuronal characteristics. We show that ND1 acted as a pioneer factor that reshapes the chromatin landscape of astrocytes to that of neurons. This restructuring promotes the expression of neurogenic genes via inducing H3K27ac modification. Through integrative analysis of various ND1-induced neuronal specification systems, we identified 25 ND1 targets, including Hes6, as key regulators. Thus, our work highlights the key role of ND1 and its downstream regulators in neuronal reprogramming.
Collapse
Affiliation(s)
- Wen Li
- Guangdong-HongKong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou 510632, China; Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong Key Laboratory of Non-Human Primate Research, GHM Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China
| | - Dan Su
- GMU-GIBH Joint School of Life Sciences, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou National Laboratory, Guangzhou Medical University, Guangzhou 510005, China; The Bioland Laboratory, Guangzhou 510700, China
| | - Xining Li
- GMU-GIBH Joint School of Life Sciences, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou National Laboratory, Guangzhou Medical University, Guangzhou 510005, China; The Bioland Laboratory, Guangzhou 510700, China
| | - Kang Lu
- Guangdong-HongKong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou 510632, China
| | - Qingpei Huang
- GMU-GIBH Joint School of Life Sciences, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou National Laboratory, Guangzhou Medical University, Guangzhou 510005, China; The Bioland Laboratory, Guangzhou 510700, China
| | - Jiajun Zheng
- Guangdong-HongKong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou 510632, China; Department of Anesthesiology, Guangzhou First People's Hospital, Guangzhou 510180, China
| | - Xiaopeng Luo
- Guangdong-HongKong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou 510632, China
| | - Gong Chen
- Guangdong-HongKong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou 510632, China; Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong Key Laboratory of Non-Human Primate Research, GHM Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China.
| | - Xiaoying Fan
- GMU-GIBH Joint School of Life Sciences, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou National Laboratory, Guangzhou Medical University, Guangzhou 510005, China; The Bioland Laboratory, Guangzhou 510700, China.
| |
Collapse
|
3
|
Wang T, Weng H, Li Y. Comparative study of the effects of prenatal sevoflurane exposure at different cortical stages on forebrain development and maturation in offspring. Front Neurosci 2025; 19:1556703. [PMID: 40248263 PMCID: PMC12003305 DOI: 10.3389/fnins.2025.1556703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 03/20/2025] [Indexed: 04/19/2025] Open
Abstract
Introduction Brain development involves several critical stages, such as proliferation, neuronal migration, axonal pathfinding, and connection formation. Sevoflurane, a γ-aminobutyric acid (GABA) receptor agonist, is widely used as an inhaled general anesthetic. However, its impact on brain development has raised increasing concerns, particularly regarding prenatal exposure. This study aims to investigate the effects of prenatal sevoflurane exposure (PSE) at different cortical stages, focusing on its impact on the migration of glutamatergic and GABAergic neurons and neuronal behavior in offspring. Methods PSE was administered at two critical prenatal stages: embryonic day (E) 12.5 and E18.5. Double in situ hybridization was used to identify the coexpression of GABA receptors in Pax6- and Mash1-positive cells in the forebrain. The radial migration of glutamatergic neurons and the tangential migration of GABAergic neurons were analyzed. Behavioral tests, including the open-field test, elevated plus-maze test, forced swim test, tail suspension test, sucrose preference test, and Morris water maze, were performed on offspring to assess anxiety-like behaviors, depression, and learning and memory impairments. Results PSE inhibits the radial migration of glutamatergic neurons and promotes the tangential migration of GABAergic neurons. Specifically, early exposure (E12.5) inhibited the expression of the Pax6-Tbr2-Tbr1 cascade and the radial migration of Tbr1 in the ventral prefrontal cortex (PFC), whereas late exposure (E18.5) inhibited this process on the dorsal side. In addition, offspring mice with PSE exhibited increased anxiety-like behaviors, rather than depression, as demonstrated by reduced time spent in the center of the open-field test and in the open arms of the elevated plus-maze test. No significant differences were observed in the forced swim test, tail suspension test, or sucrose preference test. Furthermore, learning and memory impairments were observed in the Morris water maze. Conclusion Our results indicate that PSE at E12.5 and E18.5 leads to abnormalities in the migration of glutamatergic and GABAergic neurons, affecting long-term anxiety-like behaviors and causing learning and memory impairments in offspring mice.
Collapse
Affiliation(s)
- Tianyuan Wang
- School of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia, China
- Department of Anesthesiology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Huandi Weng
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yalan Li
- Department of Anesthesiology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
4
|
Clarence T, Bendl J, Cao X, Wang X, Zheng S, Hoffman GE, Kozlenkov A, Hong A, Iskhakova M, Jaiswal MK, Murphy S, Yu A, Haroutunian V, Dracheva S, Akbarian S, Fullard JF, Yuan GC, Lee D, Roussos P. Multiomic single-cell profiling identifies critical regulators of postnatal brain. Nat Genet 2025; 57:591-603. [PMID: 39962241 DOI: 10.1038/s41588-025-02083-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 01/08/2025] [Indexed: 03/15/2025]
Abstract
Human brain development spans from embryogenesis to adulthood, with dynamic gene expression controlled by cell-type-specific cis-regulatory element activity and three-dimensional genome organization. To advance our understanding of postnatal brain development, we simultaneously profiled gene expression and chromatin accessibility in 101,924 single nuclei from four brain regions across ten donors, covering five key postnatal stages from infancy to late adulthood. Using this dataset and chromosome conformation capture data, we constructed enhancer-based gene regulatory networks to identify cell-type-specific regulators of brain development and interpret genome-wide association study loci for ten main brain disorders. Our analysis connected 2,318 cell-specific loci to 1,149 unique genes, representing 41% of loci linked to the investigated traits, and highlighted 55 genes influencing several disease phenotypes. Pseudotime analysis revealed distinct stages of postnatal oligodendrogenesis and their regulatory programs. These findings provide a comprehensive dataset of cell-type-specific gene regulation at critical timepoints in postnatal brain development.
Collapse
Affiliation(s)
- Tereza Clarence
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Jaroslav Bendl
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Xuan Cao
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Xinyi Wang
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shiwei Zheng
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gabriel E Hoffman
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexey Kozlenkov
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Aram Hong
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Marina Iskhakova
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Manoj K Jaiswal
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mental Illness Research Education and Clinical Center, James J. Peters VA Medical Center, Bronx, NY, USA
| | - Sarah Murphy
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexander Yu
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Vahram Haroutunian
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mental Illness Research Education and Clinical Center, James J. Peters VA Medical Center, Bronx, NY, USA
| | - Stella Dracheva
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mental Illness Research Education and Clinical Center, James J. Peters VA Medical Center, Bronx, NY, USA
| | - Schahram Akbarian
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John F Fullard
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Guo-Cheng Yuan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Donghoon Lee
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Panos Roussos
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Mental Illness Research Education and Clinical Center, James J. Peters VA Medical Center, Bronx, NY, USA.
- Center for Precision Medicine and Translational Therapeutics, James J. Peters VA Medical Center, Bronx, NY, USA.
| |
Collapse
|
5
|
Mseis-Jackson N, Jiang M, Sharma M, Ranchod A, Williams C, Chen X, Li H. Dynamic regulation of NeuroD1 expression level by a novel viral construct during astrocyte-to-neuron reprogramming. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.17.638625. [PMID: 40027739 PMCID: PMC11870611 DOI: 10.1101/2025.02.17.638625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Astrocyte-to-neuron reprogramming presents a viable approach for regenerative medicine. The reprogramming factor NeuroD1 has demonstrated capability of neuronal reprogramming with high efficiency both in culture and in the injured central nervous system. High level of NeuroD1 expression is required to break down the cellular identity barrier for a successful reprogramming, and yet persistence of this high level drives the reprogrammed neurons primarily to glutamatergic subtype. This is consistent with the critical role of NeuroD1 in determination of glutamatergic neuronal lineage during development. However, diversified neuronal subtypes are needed to establish appropriate neuronal connectivity in disease/injury conditions. We reason that continuously high level of NeuroD1 expression forces the reprogrammed neurons into glutamatergic subtype, and that reducing NeuroD1 level after reprogramming may allow generation of neurons with diversified subtypes. For this purpose, we engineered a novel viral expression vector by which NeuroD1 expression can be dynamically regulated during the reprogramming process. Specifically, the target site of a neuron-specific microRNA (miR-124) is incorporated in the expression system. Therefore, this novel construct would still achieve a high NeuroD1 expression level in astrocytes for reprogramming to occur and yet reduce its level in the reprogrammed neurons by suppression of endogenous miR-124. In this study, we demonstrated that this construct elicits a dynamic gene expression pattern with much reduced level of NeuroD1 at later stages of neuronal reprogramming. We also showed that this construct still retains relatively high reprogramming efficiency and can generate mature neurons with an enhanced GABAergic neuronal phenotype.
Collapse
|
6
|
Dokukin NV, Chudakova DA, Shkap MO, Kovalchuk AM, Kibirsky PD, Baklaushev VP. Direct Neural Reprogramming in situ: Existing Approaches and Their Optimization. BIOCHEMISTRY. BIOKHIMIIA 2025; 90:214-230. [PMID: 40254400 DOI: 10.1134/s000629792460426x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/28/2025] [Accepted: 01/31/2025] [Indexed: 04/22/2025]
Abstract
Direct in situ neuronal reprogramming (transdifferentiation) of glial cells (astrocytes and microglia) has attracted a significant interest as a potential approach for the treatment of a wide range of neurodegenerative diseases and damages of the central nervous system (CNS). The nervous system of higher mammals has a very limited capacity for repair. Disruption of CNS functioning due to traumatic injuries or neurodegenerative processes can significantly affect the quality of patients' life, lead to motor and cognitive impairments, and result in disability and, in some cases, death. Restoration of lost neurons in situ via direct reprogramming of glial cells without the intermediate stage of pluripotency seems to be the most attractive approach from the viewpoint of translational biomedicine. The ability of astroglia to actively proliferate in response to the damage of neural tissue supports the idea that these neuron-like cells, which are already present at the lesion site, are good candidates for transdifferentiation into neurons, considering that the possibility of direct neuronal reprogramming of astrocytes both in vitro and in vivo have demonstrated in many independent studies. Overexpression of proneuronal transcription factors, e.g., neurogenic differentiation factors 1-4 (NeuroD1-4), Neurogenin 2 (NeuroG2), Ascl1 (Achaete-Scute homolog 1), and Dlx2 (distal-less homeobox 2), including pioneer transcription factors that recognize target sequences in the compacted chromatin and activate transcription of silent genes, has already been proven as a potential therapeutic strategy. Other strategies, such as microRNA-mediated suppression of activity of PTB and REST transcription factors and application of small molecules or various biomaterials, are also utilized in neuronal reprogramming. However, the efficiency of direct in situ reprogramming is limited by a number of factors, including cell specificity of transgene delivery systems and promoters, brain regions in which transdifferentiation occurs, factors affecting cell metabolism, microenvironment, etc. Reprogramming in situ, which takes place in the presence of a large number of different cell types, requires monitoring and precise phenotypic characterization of subpopulations of cells undergoing transdifferentiation in order to confirm the reprogramming of the astroglia into neurons and subsequent integration of these neurons into the CNS. Here, we discussed the most efficient strategies of neuronal reprogramming and technologies used to visualize the transdifferentiation process, with special focus on the obstacles to efficient neuronal conversion, as well as approaches to overcome them.
Collapse
Affiliation(s)
- Nikita V Dokukin
- Federal Center for Brain and Neurotechnology, Federal Medical and Biological Agency of Russia, Moscow, 117513, Russia
| | - Daria A Chudakova
- Federal Center for Brain and Neurotechnology, Federal Medical and Biological Agency of Russia, Moscow, 117513, Russia
- National Medical Research Center of Children's Health, Ministry of Health of the Russian Federation, Moscow, 119991, Russia
| | - Matvey O Shkap
- Federal Center for Brain and Neurotechnology, Federal Medical and Biological Agency of Russia, Moscow, 117513, Russia
| | - Anna M Kovalchuk
- Federal Center for Brain and Neurotechnology, Federal Medical and Biological Agency of Russia, Moscow, 117513, Russia
| | - Pavel D Kibirsky
- Federal Center for Brain and Neurotechnology, Federal Medical and Biological Agency of Russia, Moscow, 117513, Russia
| | - Vladimir P Baklaushev
- Federal Center for Brain and Neurotechnology, Federal Medical and Biological Agency of Russia, Moscow, 117513, Russia.
- Federal Scientific and Clinical Center for Specialized Types of Medical Care and Medical Technologies, Federal Medical and Biological Agency of Russia, Moscow, 115682, Russia
- Research Institute of Pulmonology, Federal Medical and Biological Agency of Russia, Moscow, 115682, Russia
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
| |
Collapse
|
7
|
Shin D, Urbanek ME, Larson HH, Moussa AJ, Lee KY, Baker DL, Standen-Bloom E, Ramachandran S, Bogdanoff D, Cadwell CR, Nowakowski TJ. High-Complexity Barcoded Rabies Virus for Scalable Circuit Mapping Using Single-Cell and Single-Nucleus Sequencing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.01.616167. [PMID: 39713304 PMCID: PMC11661106 DOI: 10.1101/2024.10.01.616167] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Single cell genomics has revolutionized our understanding of neuronal cell types. However, scalable technologies for probing single-cell connectivity are lacking, and we are just beginning to understand how molecularly defined cell types are organized into functional circuits. Here, we describe a protocol to generate high-complexity barcoded rabies virus (RV) for scalable circuit mapping from tens of thousands of individual starter cells in parallel. In addition, we introduce a strategy for targeting RV-encoded barcode transcripts to the nucleus so that they can be read out using single-nucleus RNA sequencing (snRNA-seq). We apply this tool in organotypic slice cultures of the developing human cerebral cortex, which reveals the emergence of cell type-specific circuit motifs in midgestation. By leveraging the power and throughput of single cell genomics for mapping synaptic connectivity, we chart a path forward for scalable circuit mapping of molecularly-defined cell types in healthy and disease states.
Collapse
Affiliation(s)
- David Shin
- Biomedical Sciences Graduate Program, University of California, San Francisco, CA, USA
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - Madeleine E. Urbanek
- Biomedical Sciences Graduate Program, University of California, San Francisco, CA, USA
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - H. Hanh Larson
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - Anthony J. Moussa
- Medical Scientist Training Program, University of California, San Francisco, CA, USA
| | - Kevin Y. Lee
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - Donovan L. Baker
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - Elio Standen-Bloom
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - Sangeetha Ramachandran
- Biomedical Sciences Graduate Program, University of California, San Francisco, CA, USA
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - Derek Bogdanoff
- Tetrad Graduate Program, University of California, San Francisco, CA, USA
| | - Cathryn R. Cadwell
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
- Department of Pathology, University of California, San Francisco, CA, USA
- Weill Neurohub, University of California, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, CA, USA
| | - Tomasz J. Nowakowski
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
- Department of Anatomy, University of California, San Francisco, CA, USA
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA, USA
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, USA
| |
Collapse
|
8
|
Taufer NP, Santos-Souza C, Larentis LT, Santos CND, Creuzet SE, Garcez RC. Integrative analysis of molecular pathways and morphological anomalies associated with congenital Zika syndrome. J Neurol Sci 2024; 465:123190. [PMID: 39182423 DOI: 10.1016/j.jns.2024.123190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 08/09/2024] [Accepted: 08/18/2024] [Indexed: 08/27/2024]
Abstract
Congenital Zika syndrome (CZS) comprises a set of clinical manifestations that can be presented by neonates born to mothers infected by the Zika virus (ZIKV). CZS-associated phenotypes include neurological, skeletal, and systemic alterations and long-term developmental sequelae. One of the most frequently reported clinical conditions is microcephaly characterized by a reduction in head circumference and cognitive complications. Nevertheless, the associations among the diverse signaling pathways underlying CZS phenotypes remain to be elucidated. To shed light on CZS, we have extensively reviewed the morphological anomalies resulting from ZIKV infection, as well as genes and proteins of interest obtained from the published literature. With this list of genes or proteins, we performed computational analyses to explore the cellular processes, molecular mechanisms, and molecular pathways related to ZIKV infection. Therefore, in this review, we comprehensively describe the morphological abnormalities caused by congenital ZIKV infection and, through the analysis noted above, propose common molecular pathways altered by ZIKV that could explain both central nervous system and craniofacial skeletal alterations.
Collapse
Affiliation(s)
- Nathali Parise Taufer
- Graduate Program in Cell and Developmental Biology, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil; Laboratório de Células-Tronco e Regeneração Tecidual (LACERT), Department of Cell Biology, Embryology, and Genetics, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Camila Santos-Souza
- Graduate Program in Cell and Developmental Biology, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil; Laboratório de Células-Tronco e Regeneração Tecidual (LACERT), Department of Cell Biology, Embryology, and Genetics, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Lucas Trentin Larentis
- Graduate Program in Cell and Developmental Biology, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil; Laboratório de Células-Tronco e Regeneração Tecidual (LACERT), Department of Cell Biology, Embryology, and Genetics, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | | | - Sophie Emmanuelle Creuzet
- Institut des Neurosciences Paris-Saclay (NeuroPSI), Paris-Saclay University, Centre National de la Recherche Scientifique UMR 9197, Saclay, France.
| | - Ricardo Castilho Garcez
- Graduate Program in Cell and Developmental Biology, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil; Laboratório de Células-Tronco e Regeneração Tecidual (LACERT), Department of Cell Biology, Embryology, and Genetics, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil.
| |
Collapse
|
9
|
Liu MH, Xu YG, Bai XN, Lin JH, Xiang ZQ, Wang T, Xu L, Chen G. Efficient Dlx2-mediated astrocyte-to-neuron conversion and inhibition of neuroinflammation by NeuroD1. Dev Neurobiol 2024; 84:274-290. [PMID: 39034481 DOI: 10.1002/dneu.22951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 06/05/2024] [Accepted: 07/01/2024] [Indexed: 07/23/2024]
Abstract
In vivo astrocyte-to-neuron (AtN) conversion induced by overexpression of neural transcriptional factors has great potential for neural regeneration and repair. Here, we demonstrate that a single neural transcriptional factor, Dlx2, converts mouse striatal astrocytes into neurons in a dose-dependent manner. Lineage-tracing studies in Aldh1l1-CreERT2 mice confirm that Dlx2 can convert striatal astrocytes into DARPP32+ and Ctip2+ medium spiny neurons (MSNs). Time-course studies reveal a gradual conversion from astrocytes to neurons in 1 month, with a distinct intermediate state in between astrocytes and neurons. Interestingly, when Dlx2-infected astrocytes start to lose astrocytic markers, the other local astrocytes proliferate to maintain astrocytic levels in the converted areas. Unexpectedly, although Dlx2 efficiently reprograms astrocytes into neurons in the gray matter striatum, it also induces partial reprogramming of astrocytes in the white matter corpus callosum. Such partial reprogramming of white matter astrocytes is associated with neuroinflammation, which can be suppressed by the addition of NeuroD1. Our results highlight the importance of investigating AtN conversion in both the gray matter and white matter to thoroughly evaluate therapeutic potentials. This study also unveils the critical role of anti-inflammation by NeuroD1 during AtN conversion.
Collapse
Affiliation(s)
- Min-Hui Liu
- Guangdong-HongKong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
- VIB Center for Brain and Disease, KU Leuven, Herestraat 49, Leuven, Belgium
| | - Yu-Ge Xu
- Guangdong-HongKong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
| | - Xiao-Ni Bai
- Guangdong-HongKong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
| | - Jian-Hua Lin
- Guangdong-HongKong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
| | - Zong-Qin Xiang
- Guangdong-HongKong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
- Department of Neurosurgery, the First Affiliated Hospital, Jinan University, Guangzhou, Guangdong Province, China
| | - Tao Wang
- Guangdong-HongKong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
| | - Liang Xu
- Guangdong-HongKong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
| | - Gong Chen
- Guangdong-HongKong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
| |
Collapse
|
10
|
Shim HS, Iaconelli J, Shang X, Li J, Lan ZD, Jiang S, Nutsch K, Beyer BA, Lairson LL, Boutin AT, Bollong MJ, Schultz PG, DePinho RA. TERT activation targets DNA methylation and multiple aging hallmarks. Cell 2024; 187:4030-4042.e13. [PMID: 38908367 PMCID: PMC11552617 DOI: 10.1016/j.cell.2024.05.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 04/03/2024] [Accepted: 05/23/2024] [Indexed: 06/24/2024]
Abstract
Insufficient telomerase activity, stemming from low telomerase reverse transcriptase (TERT) gene transcription, contributes to telomere dysfunction and aging pathologies. Besides its traditional function in telomere synthesis, TERT acts as a transcriptional co-regulator of genes pivotal in aging and age-associated diseases. Here, we report the identification of a TERT activator compound (TAC) that upregulates TERT transcription via the MEK/ERK/AP-1 cascade. In primary human cells and naturally aged mice, TAC-induced elevation of TERT levels promotes telomere synthesis, blunts tissue aging hallmarks with reduced cellular senescence and inflammatory cytokines, and silences p16INK4a expression via upregulation of DNMT3B-mediated promoter hypermethylation. In the brain, TAC alleviates neuroinflammation, increases neurotrophic factors, stimulates adult neurogenesis, and preserves cognitive function without evident toxicity, including cancer risk. Together, these findings underscore TERT's critical role in aging processes and provide preclinical proof of concept for physiological TERT activation as a strategy to mitigate multiple aging hallmarks and associated pathologies.
Collapse
Affiliation(s)
- Hong Seok Shim
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jonathan Iaconelli
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Xiaoying Shang
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jiexi Li
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Zheng D Lan
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shan Jiang
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kayla Nutsch
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Brittney A Beyer
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Luke L Lairson
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Adam T Boutin
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Michael J Bollong
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Peter G Schultz
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Ronald A DePinho
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
11
|
Pavlinkova G, Smolik O. NEUROD1: transcriptional and epigenetic regulator of human and mouse neuronal and endocrine cell lineage programs. Front Cell Dev Biol 2024; 12:1435546. [PMID: 39105169 PMCID: PMC11298428 DOI: 10.3389/fcell.2024.1435546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 07/02/2024] [Indexed: 08/07/2024] Open
Abstract
Transcription factors belonging to the basic helix-loop-helix (bHLH) family are key regulators of cell fate specification and differentiation during development. Their dysregulation is implicated not only in developmental abnormalities but also in various adult diseases and cancers. Recently, the abilities of bHLH factors have been exploited in reprogramming strategies for cell replacement therapy. One such factor is NEUROD1, which has been associated with the reprogramming of the epigenetic landscape and potentially possessing pioneer factor abilities, initiating neuronal developmental programs, and enforcing pancreatic endocrine differentiation. The review aims to consolidate current knowledge on NEUROD1's multifaceted roles and mechanistic pathways in human and mouse cell differentiation and reprogramming, exploring NEUROD1 roles in guiding the development and reprogramming of neuroendocrine cell lineages. The review focuses on NEUROD1's molecular mechanisms, its interactions with other transcription factors, its role as a pioneer factor in chromatin remodeling, and its potential in cell reprogramming. We also show a differential potential of NEUROD1 in differentiation of neurons and pancreatic endocrine cells, highlighting its therapeutic potential and the necessity for further research to fully understand and utilize its capabilities.
Collapse
Affiliation(s)
- Gabriela Pavlinkova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology Czech Academy of Sciences, Vestec, Czechia
| | | |
Collapse
|
12
|
Mseis-Jackson N, Sharma M, Li H. Controlling the Expression Level of the Neuronal Reprogramming Factors for a Successful Reprogramming Outcome. Cells 2024; 13:1223. [PMID: 39056804 PMCID: PMC11274869 DOI: 10.3390/cells13141223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 07/28/2024] Open
Abstract
Neuronal reprogramming is a promising approach for making major advancement in regenerative medicine. Distinct from the approach of induced pluripotent stem cells, neuronal reprogramming converts non-neuronal cells to neurons without going through a primitive stem cell stage. In vivo neuronal reprogramming brings this approach to a higher level by changing the cell fate of glial cells to neurons in neural tissue through overexpressing reprogramming factors. Despite the ongoing debate over the validation and interpretation of newly generated neurons, in vivo neuronal reprogramming is still a feasible approach and has the potential to become clinical treatment with further optimization and refinement. Here, we discuss the major neuronal reprogramming factors (mostly pro-neurogenic transcription factors during development), especially the significance of their expression levels during neurogenesis and the reprogramming process focusing on NeuroD1. In the developing central nervous system, these pro-neurogenic transcription factors usually elicit distinct spatiotemporal expression patterns that are critical to their function in generating mature neurons. We argue that these dynamic expression patterns may be similarly needed in the process of reprogramming adult cells into neurons and further into mature neurons with subtype identities. We also summarize the existing approaches and propose new ones that control gene expression levels for a successful reprogramming outcome.
Collapse
Affiliation(s)
- Natalie Mseis-Jackson
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA;
| | - Mehek Sharma
- Department of Biological Sciences, College of Science & Mathematics, Augusta University, Augusta, GA 30912, USA;
| | - Hedong Li
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA;
| |
Collapse
|
13
|
Lee M, Kim EJ, Yum MS. Early developmental changes in a rat model of malformations of cortical development: Abnormal neuronal migration and altered response to NMDA-induced excitotoxic injury. Exp Neurol 2024; 376:114759. [PMID: 38519010 DOI: 10.1016/j.expneurol.2024.114759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 02/28/2024] [Accepted: 03/18/2024] [Indexed: 03/24/2024]
Abstract
Malformations of cortical development (MCDs) are caused by abnormal neuronal migration processes during the fetal period and are a major cause of intractable epilepsy in infancy. However, the timing of hyperexcitability or epileptogenesis in MCDs remains unclear. To identify the early developmental changes in the brain of the MCD rat model, which exhibits increased seizure susceptibility during infancy (P12-15), we analyzed the pathological changes in the brains of MCD model rats during the neonatal period and tested NMDA-induced seizure susceptibility. Pregnant rats were injected with two doses of methylazoxymethanol acetate (MAM, 15 mg/kg, i.p.) to induce MCD, while controls were administered normal saline. The cortical development of the offspring was measured by performing magnetic resonance imaging (MRI) on postnatal days (P) 1, 5, and 8. At P8, some rats were sacrificed for immunofluorescence, Golgi staining, and Western analysis. In another set of rats, the number and latency to onset of spasms were monitored for 90 min after the NMDA (5 mg/kg i.p.) injection at P8. In MCD rats, in vivo MR imaging showed smaller brain volume and thinner cortex from day 1 after birth (p < 0.001). Golgi staining and immunofluorescence revealed abnormal neuronal migration, with a reduced number of neuronal cell populations and less dendritic arborization at P8. Furthermore, MCD rats exhibited a significant reduction in the expression of NMDA receptors and AMPAR4, along with an increase in AMPAR3 expression (p < 0.05). Although there was no difference in the latency to seizure onset between MCD rats and controls, the MCD rats survived significantly longer than the controls. These results provide insights into the early developmental changes in the cortex of a MCD rat model and suggest that delayed and abnormal neuronal development in the immature brain is associated with a blunted response to NMDA-induced excitotoxic injury. These developmental changes may be involved in the sudden onset of epilepsy in patients with MCD or prenatal brain injury.
Collapse
Affiliation(s)
- Minyoung Lee
- Department of Pediatrics, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea; Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Republic of Korea.
| | - Eun-Jin Kim
- Department of Pediatrics, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea; Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Republic of Korea
| | - Mi-Sun Yum
- Department of Pediatrics, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea; Department of Pediatrics, Asan Medical Center Children's Hospital, Seoul 05505, Republic of Korea.
| |
Collapse
|
14
|
Ojiro R, Ozawa S, Zou X, Tang Q, Woo GH, Shibutani M. Similar toxicity potential of glyphosate and glyphosate-based herbicide on cerebellar development after maternal exposure in rats. ENVIRONMENTAL TOXICOLOGY 2024; 39:3040-3054. [PMID: 38314887 DOI: 10.1002/tox.24163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/10/2023] [Accepted: 01/18/2024] [Indexed: 02/07/2024]
Abstract
Studies on the effects of glyphosate (GlyP) and glyphosate-based herbicides (GBHs) on cerebellar development are extremely limited. This study examined the effects of maternal exposure to GlyP and GBH on rat cerebellar development in male offspring. From day 6 of gestation until day 21 postpartum at weaning, dams were given GlyP at 1.5% or 3.0% in diet or GBH at 1.0% in drinking water (corresponding to 0.36% GlyP). At weaning, GBH exposure was linked to increased numbers of DCX+ migrating granule cells in the cortex and TUNEL+ apoptotic cells in the internal granular layer (IGL), suggesting the disappearance of mismigrated granule cells via apoptosis. GBH also upregulated Nr4a3 and downregulated Cdk5 in the cerebellar vermis, suggesting a causal relation with the impaired granule cell development at this time. GlyP (3.0%) tended to increase in the number of DCX+ migrating granule cells in the IGL and upregulated Nr4a3 at weaning. Both compounds also upregulated genes related to granule cell migration (Astn1, Astn2, Nfia, and/or Nfix) at weaning and in adulthood, which might be an ameliorative response to delayed granule cell migration. Moreover, GBH induced Purkinje cell misalignment at weaning, which could be the result of delayed granule cell migration. In adulthood, GBH was associated with upregulation of the reelin signaling-related genes Reln, Dab1, and Efnb1, suggesting a compensatory response to Purkinje cell misalignment. GlyP induced the same gene expression changes. These results suggest that GBH reversibly disrupts cerebellar development, primarily by targeting granule cell migration and differentiation, whereas GlyP exhibited similar toxic potential as GBH.
Collapse
Affiliation(s)
- Ryota Ojiro
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
- Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Shunsuke Ozawa
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
- Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Xinyu Zou
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
- Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Qian Tang
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
- Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Gye-Hyeong Woo
- Laboratory of Histopathology, Department of Clinical Laboratory Science, Semyung University, Jecheon-si, Chungbuk, Korea
| | - Makoto Shibutani
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
- Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
- Institute of Global Innovation Research, Tokyo University of Agriculture and Technology, Tokyo, Japan
| |
Collapse
|
15
|
Xing L, Gkini V, Nieminen AI, Zhou HC, Aquilino M, Naumann R, Reppe K, Tanaka K, Carmeliet P, Heikinheimo O, Pääbo S, Huttner WB, Namba T. Functional synergy of a human-specific and an ape-specific metabolic regulator in human neocortex development. Nat Commun 2024; 15:3468. [PMID: 38658571 PMCID: PMC11043075 DOI: 10.1038/s41467-024-47437-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 04/02/2024] [Indexed: 04/26/2024] Open
Abstract
Metabolism has recently emerged as a major target of genes implicated in the evolutionary expansion of human neocortex. One such gene is the human-specific gene ARHGAP11B. During human neocortex development, ARHGAP11B increases the abundance of basal radial glia, key progenitors for neocortex expansion, by stimulating glutaminolysis (glutamine-to-glutamate-to-alpha-ketoglutarate) in mitochondria. Here we show that the ape-specific protein GLUD2 (glutamate dehydrogenase 2), which also operates in mitochondria and converts glutamate-to-αKG, enhances ARHGAP11B's ability to increase basal radial glia abundance. ARHGAP11B + GLUD2 double-transgenic bRG show increased production of aspartate, a metabolite essential for cell proliferation, from glutamate via alpha-ketoglutarate and the TCA cycle. Hence, during human evolution, a human-specific gene exploited the existence of another gene that emerged during ape evolution, to increase, via concerted changes in metabolism, progenitor abundance and neocortex size.
Collapse
Affiliation(s)
- Lei Xing
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.
- Department of Biological Sciences, University of Manitoba, Winnipeg, MB, Canada.
| | - Vasiliki Gkini
- Neuroscience Center, HiLIFE - Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Anni I Nieminen
- FIMM Metabolomics Unit, Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Hui-Chao Zhou
- Center for Cancer Biology (CCB), VIB-KU Leuven, B-3000, Leuven, Belgium
| | - Matilde Aquilino
- Neuroscience Center, HiLIFE - Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Ronald Naumann
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Katrin Reppe
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Kohichi Tanaka
- Laboratory of Molecular Neuroscience, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, B-3000, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, B-3000, Leuven, Belgium
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Oskari Heikinheimo
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Svante Pääbo
- Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
- Human Evolutionary Genomics Unit, Okinawa Institute of Science and Technology, Okinawa, Onna-son, Japan
| | - Wieland B Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.
| | - Takashi Namba
- Neuroscience Center, HiLIFE - Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
16
|
Davila-Valencia I, Saad M, Olthoff G, Faulkner M, Charara M, Farnum A, Dysko RC, Zhang Z. Sex specific effects of buprenorphine on adult hippocampal neurogenesis and behavioral outcomes during the acute phase after pediatric traumatic brain injury in mice. Neuropharmacology 2024; 245:109829. [PMID: 38159797 DOI: 10.1016/j.neuropharm.2023.109829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 12/15/2023] [Accepted: 12/23/2023] [Indexed: 01/03/2024]
Abstract
Traumatic brain injury (TBI) in children often causes cognitive and mental dysfunctions, as well as acute and chronic pain. Adult hippocampal neurogenesis plays a key role in cognition, depression, and pain. Adult hippocampal neurogenesis can be modulated by genetic and environmental factors, such as TBI and opioids. Buprenorphine (BPN), a semisynthetic opioid, is commonly used for pain management in children, however, the effects of BPN on adult hippocampal neurogenesis after pediatric TBI are still unclear. This study investigated the sex-specific effects of BPN on adult hippocampal neurogenesis during acute phase after pediatric TBI. Male and female littermates were randomized on postnatal day 20-21(P20-21) into Sham, TBI+saline and TBI+BPN groups. BPN was administered intraperitoneally to the TBI+BPN mice at 30 min after injury, and then every 6-12 h (h) for 2 days (d). Bromodeoxyuridine (BrdU) was administered intraperitoneally to all groups at 2, 4, 6, and 8-h post-injury. All outcomes were evaluated at 3-d post-BrdU administration. We found that TBI induced significant cognitive impairment, depression, and reduced adult hippocampal neurogenesis in both male and female mice, with more prominent effects in females. BPN significantly improved adult hippocampal neurogenesis and depression in males, but not in females. We further demonstrated that differential expressions of opioid receptors, transcription factors and neuroinflammatory markers at the neurogenic niche might be responsible for the differential effects of BPN in males and females. In conclusion, this study elucidates the effects of BPN on adult hippocampal neurogenesis and behavioral outcomes at the acute phase after pediatric TBI.
Collapse
Affiliation(s)
- Ivan Davila-Valencia
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI, 48128, USA.
| | - Mark Saad
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI, 48128, USA.
| | - Grace Olthoff
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI, 48128, USA.
| | - Megan Faulkner
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI, 48128, USA.
| | - Maysoun Charara
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI, 48128, USA.
| | - Abigail Farnum
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI, 48128, USA.
| | - Robert C Dysko
- Unit for Laboratory Animal Medicine, University of Michigan-Ann Arbor, 2800 Plymouth Rd, Ann Arbor, MI, 48109, USA.
| | - Zhi Zhang
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI, 48128, USA.
| |
Collapse
|
17
|
Napoli AJ, Laderwager S, Zoodsma JD, Biju B, Mucollari O, Schubel SK, Aprea C, Sayed A, Morgan K, Napoli A, Flanagan S, Wollmuth LP, Sirotkin HI. Developmental loss of NMDA receptors results in supernumerary forebrain neurons through delayed maturation of transit-amplifying neuroblasts. Sci Rep 2024; 14:3395. [PMID: 38336823 PMCID: PMC10858180 DOI: 10.1038/s41598-024-53910-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 02/06/2024] [Indexed: 02/12/2024] Open
Abstract
Developmental neurogenesis is a tightly regulated spatiotemporal process with its dysregulation implicated in neurodevelopmental disorders. NMDA receptors are glutamate-gated ion channels that are widely expressed in the early nervous system, yet their contribution to neurogenesis is poorly understood. Notably, a variety of mutations in genes encoding NMDA receptor subunits are associated with neurodevelopmental disorders. To rigorously define the role of NMDA receptors in developmental neurogenesis, we used a mutant zebrafish line (grin1-/-) that lacks all NMDA receptors yet survives to 10 days post-fertilization, offering the opportunity to study post-embryonic neurodevelopment in the absence of NMDA receptors. Focusing on the forebrain, we find that these fish have a progressive supernumerary neuron phenotype confined to the telencephalon at the end of embryonic neurogenesis, but which extends to all forebrain regions during postembryonic neurogenesis. This enhanced neuron population does not arise directly from increased numbers or mitotic activity of radial glia cells, the principal neural stem cells. Rather, it stems from a lack of timely maturation of transit-amplifying neuroblasts into post-mitotic neurons, as indicated by a decrease in expression of the ontogenetically-expressed chloride transporter, KCC2. Pharmacological blockade with MK-801 recapitulates the grin1-/- supernumerary neuron phenotype, indicating a requirement for ionotropic signaling. Thus, NMDA receptors are required for suppression of indirect, transit amplifying cell-driven neurogenesis by promoting maturational termination of mitosis. Loss of suppression results in neuronal overpopulation that can fundamentally change brain circuitry and may be a key factor in pathogenesis of neurodevelopmental disorders caused by NMDA receptor dysfunction.
Collapse
Affiliation(s)
- Amalia J Napoli
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794-5230, USA
| | - Stephanie Laderwager
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794-5230, USA
- Graduate Program in Neuroscience, Stony Brook University, Stony Brook, NY, 11794-5230, USA
| | - Josiah D Zoodsma
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794-5230, USA
| | - Bismi Biju
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794-5230, USA
| | - Olgerta Mucollari
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794-5230, USA
| | - Sarah K Schubel
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794-5230, USA
| | - Christieann Aprea
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794-5230, USA
| | - Aaliya Sayed
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794-5230, USA
| | - Kiele Morgan
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794-5230, USA
| | - Annelysia Napoli
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794-5230, USA
| | - Stephanie Flanagan
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794-5230, USA
| | - Lonnie P Wollmuth
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794-5230, USA
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, 11794-5215, USA
- Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY, 11794-5230, USA
| | - Howard I Sirotkin
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794-5230, USA.
| |
Collapse
|
18
|
Sokpor G, Kerimoglu C, Ulmke PA, Pham L, Nguyen HD, Brand-Saberi B, Staiger JF, Fischer A, Nguyen HP, Tuoc T. H3 Acetylation-Induced Basal Progenitor Generation and Neocortex Expansion Depends on the Transcription Factor Pax6. BIOLOGY 2024; 13:68. [PMID: 38392287 PMCID: PMC10886678 DOI: 10.3390/biology13020068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 02/24/2024]
Abstract
Enrichment of basal progenitors (BPs) in the developing neocortex is a central driver of cortical enlargement. The transcription factor Pax6 is known as an essential regulator in generation of BPs. H3 lysine 9 acetylation (H3K9ac) has emerged as a crucial epigenetic mechanism that activates the gene expression program required for BP pool amplification. In this current work, we applied immunohistochemistry, RNA sequencing, chromatin immunoprecipitation and sequencing, and the yeast two-hybrid assay to reveal that the BP-genic effect of H3 acetylation is dependent on Pax6 functionality in the developing mouse cortex. In the presence of Pax6, increased H3 acetylation caused BP pool expansion, leading to enhanced neurogenesis, which evoked expansion and quasi-convolution of the mouse neocortex. Interestingly, H3 acetylation activation exacerbates the BP depletion and corticogenesis reduction effect of Pax6 ablation in cortex-specific Pax6 mutants. Furthermore, we found that H3K9 acetyltransferase KAT2A/GCN5 interacts with Pax6 and potentiates Pax6-dependent transcriptional activity. This explains a genome-wide lack of H3K9ac, especially in the promoter regions of BP-genic genes, in the Pax6 mutant cortex. Together, these findings reveal a mechanistic coupling of H3 acetylation and Pax6 in orchestrating BP production and cortical expansion through the promotion of a BP gene expression program during cortical development.
Collapse
Affiliation(s)
- Godwin Sokpor
- Department of Human Genetics, Ruhr University of Bochum, 44791 Bochum, Germany
- Lincoln Medical School, University of Lincoln, Lincoln LN6 7TS, UK
| | - Cemil Kerimoglu
- German Center for Neurodegenerative Diseases, 37077 Goettingen, Germany
| | | | - Linh Pham
- Department of Human Genetics, Ruhr University of Bochum, 44791 Bochum, Germany
| | - Hoang Duy Nguyen
- Department of Human Genetics, Ruhr University of Bochum, 44791 Bochum, Germany
| | - Beate Brand-Saberi
- Department of Anatomy and Molecular Embryology, Institute of Anatomy, Medical Faculty, Ruhr University Bochum, 44801 Bochum, Germany
| | - Jochen F Staiger
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, 37075 Goettingen, Germany
| | - Andre Fischer
- German Center for Neurodegenerative Diseases, 37077 Goettingen, Germany
| | - Huu Phuc Nguyen
- Department of Human Genetics, Ruhr University of Bochum, 44791 Bochum, Germany
| | - Tran Tuoc
- Department of Human Genetics, Ruhr University of Bochum, 44791 Bochum, Germany
| |
Collapse
|
19
|
Péron S, Miyakoshi LM, Brill MS, Manzano-Franco D, Serrano-López J, Fan W, Marichal N, Ghanem A, Conzelmann KK, Karow M, Ortega F, Gascón S, Berninger B. Programming of neural progenitors of the adult subependymal zone towards a glutamatergic neuron lineage by neurogenin 2. Stem Cell Reports 2023; 18:2418-2433. [PMID: 37995703 PMCID: PMC10724369 DOI: 10.1016/j.stemcr.2023.10.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 10/26/2023] [Accepted: 10/27/2023] [Indexed: 11/25/2023] Open
Abstract
Although adult subependymal zone (SEZ) neural stem cells mostly generate GABAergic interneurons, a small progenitor population expresses the proneural gene Neurog2 and produces glutamatergic neurons. Here, we determined whether Neurog2 could respecify SEZ neural stem cells and their progeny toward a glutamatergic fate. Retrovirus-mediated expression of Neurog2 induced the glutamatergic lineage markers TBR2 and TBR1 in cultured SEZ progenitors, which differentiated into functional glutamatergic neurons. Likewise, Neurog2-transduced SEZ progenitors acquired glutamatergic neuron hallmarks in vivo. Intriguingly, they failed to migrate toward the olfactory bulb and instead differentiated within the SEZ or the adjacent striatum, where they received connections from local neurons, as indicated by rabies virus-mediated monosynaptic tracing. In contrast, lentivirus-mediated expression of Neurog2 failed to reprogram early SEZ neurons, which maintained GABAergic identity and migrated to the olfactory bulb. Our data show that NEUROG2 can program SEZ progenitors toward a glutamatergic identity but fails to reprogram their neuronal progeny.
Collapse
Affiliation(s)
- Sophie Péron
- Research Group "Adult Neurogenesis and Cellular Reprogramming", Institute of Physiological Chemistry, University Medical Center Johannes Gutenberg University, Mainz, Germany; Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Leo M Miyakoshi
- Physiological Genomics, Institute of Physiology, Ludwig-Maximilians University Munich, Munich, Germany
| | - Monika S Brill
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany; Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Diana Manzano-Franco
- Department of Molecular, Cellular and Developmental Neurobiology, Cajal Institute - CSIC, Madrid, Spain
| | - Julia Serrano-López
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary, Universidad Complutense de Madrid (UCM), Madrid, Spain; Instituto Universitario de Investigación en Neuroquímica (IUIN), Madrid, Spain; Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain
| | - Wenqiang Fan
- Research Group "Adult Neurogenesis and Cellular Reprogramming", Institute of Physiological Chemistry, University Medical Center Johannes Gutenberg University, Mainz, Germany
| | - Nicolás Marichal
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Alexander Ghanem
- Max von Pettenkofer Institute and Gene Center, Ludwig Maximilians-University Munich, Munich, Germany
| | - Karl-Klaus Conzelmann
- Max von Pettenkofer Institute and Gene Center, Ludwig Maximilians-University Munich, Munich, Germany
| | - Marisa Karow
- Institute of Biochemistry, Friedrich-Alexander Universität Nürnberg-Erlangen, Erlangen, Germany
| | - Felipe Ortega
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary, Universidad Complutense de Madrid (UCM), Madrid, Spain; Instituto Universitario de Investigación en Neuroquímica (IUIN), Madrid, Spain; Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain
| | - Sergio Gascón
- Department of Molecular, Cellular and Developmental Neurobiology, Cajal Institute - CSIC, Madrid, Spain.
| | - Benedikt Berninger
- Research Group "Adult Neurogenesis and Cellular Reprogramming", Institute of Physiological Chemistry, University Medical Center Johannes Gutenberg University, Mainz, Germany; Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; Focus Program Translational Neurosciences, Johannes Gutenberg University, Mainz, Germany.
| |
Collapse
|
20
|
Lagunas T, Plassmeyer SP, Fischer AD, Friedman RZ, Rieger MA, Selmanovic D, Sarafinovska S, Sol YK, Kasper MJ, Fass SB, Aguilar Lucero AF, An JY, Sanders SJ, Cohen BA, Dougherty JD. A Cre-dependent massively parallel reporter assay allows for cell-type specific assessment of the functional effects of non-coding elements in vivo. Commun Biol 2023; 6:1151. [PMID: 37953348 PMCID: PMC10641075 DOI: 10.1038/s42003-023-05483-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 10/18/2023] [Indexed: 11/14/2023] Open
Abstract
The function of regulatory elements is highly dependent on the cellular context, and thus for understanding the function of elements associated with psychiatric diseases these would ideally be studied in neurons in a living brain. Massively Parallel Reporter Assays (MPRAs) are molecular genetic tools that enable functional screening of hundreds of predefined sequences in a single experiment. These assays have not yet been adapted to query specific cell types in vivo in a complex tissue like the mouse brain. Here, using a test-case 3'UTR MPRA library with genomic elements containing variants from autism patients, we developed a method to achieve reproducible measurements of element effects in vivo in a cell type-specific manner, using excitatory cortical neurons and striatal medium spiny neurons as test cases. This targeted technique should enable robust, functional annotation of genetic elements in the cellular contexts most relevant to psychiatric disease.
Collapse
Affiliation(s)
- Tomas Lagunas
- Department of Genetics, Washington University School of Medicine, 660 S. Euclid Ave, Saint Louis, MO, 63108, USA
- Department of Psychiatry, Washington University School of Medicine., 660 S. Euclid Ave, Saint Louis, MO, 63108, USA
- Division of Biology and Biomedical Sciences, Washington University School of Medicine, 660 S. Euclid Ave, Saint Louis, MO, 63108, USA
| | - Stephen P Plassmeyer
- Department of Genetics, Washington University School of Medicine, 660 S. Euclid Ave, Saint Louis, MO, 63108, USA
- Department of Psychiatry, Washington University School of Medicine., 660 S. Euclid Ave, Saint Louis, MO, 63108, USA
| | - Anthony D Fischer
- Department of Genetics, Washington University School of Medicine, 660 S. Euclid Ave, Saint Louis, MO, 63108, USA
- Department of Psychiatry, Washington University School of Medicine., 660 S. Euclid Ave, Saint Louis, MO, 63108, USA
| | - Ryan Z Friedman
- Department of Genetics, Washington University School of Medicine, 660 S. Euclid Ave, Saint Louis, MO, 63108, USA
- Division of Biology and Biomedical Sciences, Washington University School of Medicine, 660 S. Euclid Ave, Saint Louis, MO, 63108, USA
| | - Michael A Rieger
- Department of Genetics, Washington University School of Medicine, 660 S. Euclid Ave, Saint Louis, MO, 63108, USA
- Department of Psychiatry, Washington University School of Medicine., 660 S. Euclid Ave, Saint Louis, MO, 63108, USA
- Division of Biology and Biomedical Sciences, Washington University School of Medicine, 660 S. Euclid Ave, Saint Louis, MO, 63108, USA
| | - Din Selmanovic
- Department of Genetics, Washington University School of Medicine, 660 S. Euclid Ave, Saint Louis, MO, 63108, USA
- Department of Psychiatry, Washington University School of Medicine., 660 S. Euclid Ave, Saint Louis, MO, 63108, USA
- Division of Biology and Biomedical Sciences, Washington University School of Medicine, 660 S. Euclid Ave, Saint Louis, MO, 63108, USA
| | - Simona Sarafinovska
- Department of Genetics, Washington University School of Medicine, 660 S. Euclid Ave, Saint Louis, MO, 63108, USA
- Department of Psychiatry, Washington University School of Medicine., 660 S. Euclid Ave, Saint Louis, MO, 63108, USA
| | - Yvette K Sol
- Department of Genetics, Washington University School of Medicine, 660 S. Euclid Ave, Saint Louis, MO, 63108, USA
- Department of Psychiatry, Washington University School of Medicine., 660 S. Euclid Ave, Saint Louis, MO, 63108, USA
| | - Michael J Kasper
- Department of Genetics, Washington University School of Medicine, 660 S. Euclid Ave, Saint Louis, MO, 63108, USA
- Department of Psychiatry, Washington University School of Medicine., 660 S. Euclid Ave, Saint Louis, MO, 63108, USA
| | - Stuart B Fass
- Department of Genetics, Washington University School of Medicine, 660 S. Euclid Ave, Saint Louis, MO, 63108, USA
- Department of Psychiatry, Washington University School of Medicine., 660 S. Euclid Ave, Saint Louis, MO, 63108, USA
| | - Alessandra F Aguilar Lucero
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neuroscience, University of California San Francisco, San Francisco, CA, 94518, USA
| | - Joon-Yong An
- Department of Integrated Biomedical and Life Science, Korea University, Seoul, 02841, Republic of Korea
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul, 02841, Republic of Korea
| | - Stephan J Sanders
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neuroscience, University of California San Francisco, San Francisco, CA, 94518, USA
| | - Barak A Cohen
- Department of Genetics, Washington University School of Medicine, 660 S. Euclid Ave, Saint Louis, MO, 63108, USA
| | - Joseph D Dougherty
- Department of Genetics, Washington University School of Medicine, 660 S. Euclid Ave, Saint Louis, MO, 63108, USA.
- Department of Psychiatry, Washington University School of Medicine., 660 S. Euclid Ave, Saint Louis, MO, 63108, USA.
| |
Collapse
|
21
|
Puelles L, Stühmer T, Rubenstein JLR, Diaz C. Critical test of the assumption that the hypothalamic entopeduncular nucleus of rodents is homologous with the primate internal pallidum. J Comp Neurol 2023; 531:1715-1750. [PMID: 37695031 PMCID: PMC11418882 DOI: 10.1002/cne.25536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/26/2023] [Accepted: 08/03/2023] [Indexed: 09/12/2023]
Abstract
The globus pallidus (GP) of primates is divided conventionally into distinct internal and external parts. The literature repeats since 1930 the opinion that the homolog of the primate internal pallidum in rodents is the hypothalamic entopeduncular nucleus (embedded within fiber tracts of the cerebral peduncle). To test this idea, we explored its historic fundaments, checked the development and genoarchitecture of mouse entopeduncular and pallidal neurons, and examined relevant comparative connectivity data. We found that the extratelencephalic mouse entopeduncular structure consists of four different components arrayed along a dorsoventral sequence in the alar hypothalamus. The ventral entopeduncular nucleus (EPV), with GABAergic neurons expressing Dlx5&6 and Nkx2-1, lies within the hypothalamic peduncular subparaventricular area. Three other formations-the dorsal entopeduncular nucleus (EPD), the prereticular entopeduncular nucleus (EPPRt ), and the preeminential entopeduncular nucleus (EPPEm )-lie within the overlying paraventricular area, under the subpallium. EPD contains glutamatergic neurons expressing Tbr1, Otp, and Pax6. The EPPRt has GABAergic cells expressing Isl1 and Meis2, whereas the EPPEm population expresses Foxg1 and may be glutamatergic. Genoarchitectonic observations on relevant areas of the mouse pallidal/diagonal subpallium suggest that the GP of rodents is constituted as in primates by two adjacent but molecularly and hodologically differentiable telencephalic portions (both expressing Foxg1). These and other reported data oppose the notion that the rodent extratelencephalic entopeduncular nucleus is homologous to the primate internal pallidum. We suggest instead that all mammals, including rodents, have dual subpallial GP components, whereas primates probably also have a comparable set of hypothalamic entopeduncular nuclei. Remarkably, there is close similarity in some gene expression properties of the telencephalic internal GP and the hypothalamic EPV. This apparently underlies their notable functional analogy, sharing GABAergic neurons and thalamopetal connectivity.
Collapse
Affiliation(s)
- Luis Puelles
- Department of Human Anatomy and Psychobiology and IMIB-Arrixaca Institute, University of Murcia, El Palmar (Murcia), 30120, Spain
| | - Thorsten Stühmer
- Nina Ireland Laboratory of Developmental Neurobiology, Department of Psychiatry, UCSF Medical School, San Francisco, California
| | - John L. R. Rubenstein
- Nina Ireland Laboratory of Developmental Neurobiology, Department of Psychiatry, UCSF Medical School, San Francisco, California
| | - Carmen Diaz
- School of Medicine and Institute for Research in Neurological Disabilities, University of Castilla-La Mancha, Albacete, 02006, Spain
| |
Collapse
|
22
|
Jun S, Kim M, Park H, Hwang E, Yamamoto Y, Tanaka-Yamamoto K. Organization of Purkinje cell development by neuronal MEGF11 in cerebellar granule cells. Cell Rep 2023; 42:113137. [PMID: 37708022 DOI: 10.1016/j.celrep.2023.113137] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 07/24/2023] [Accepted: 08/31/2023] [Indexed: 09/16/2023] Open
Abstract
As cerebellar granule cells (GCs) coordinate the formation of regular cerebellar networks during postnatal development, molecules in GCs are expected to be involved. Here, we test the effects of the knockdown (KD) of multiple epidermal growth factor-like domains protein 11 (MEGF11), which is a homolog of proteins mediating astrocytic phagocytosis but is substantially increased at the later developmental stages of GCs on cerebellar development. MEGF11-KD in GCs of developing mice results in abnormal cerebellar structures, including extensively ectopic Purkinje cell (PC) somas, and in impaired motor functions. MEGF11-KD also causes abnormally asynchronous synaptic release from GC axons, parallel fibers, before the appearance of abnormal cerebellar structures. Interestingly, blockade of this abnormal synaptic release restores most of the cerebellar structures. Thus, apart from phagocytic functions of its related homologs in astrocytes, MEGF11 in GCs promotes proper PC development and cerebellar network formation by regulating immature synaptic transmission.
Collapse
Affiliation(s)
- Soyoung Jun
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology (UST), Seoul 02792, Republic of Korea
| | - Muwoong Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology (UST), Seoul 02792, Republic of Korea
| | - Heeyoun Park
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Eunmi Hwang
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology (UST), Seoul 02792, Republic of Korea
| | - Yukio Yamamoto
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea.
| | - Keiko Tanaka-Yamamoto
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology (UST), Seoul 02792, Republic of Korea.
| |
Collapse
|
23
|
Napoli AJ, Laderwager S, Zoodsma JD, Biju B, Mucollari O, Schubel SK, Aprea C, Sayed A, Morgan K, Napoli A, Flanagan S, Wollmuth LP, Sirotkin HI. Loss of NMDA receptor function during development results in decreased KCC2 expression and increased neurons in the zebrafish forebrain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.25.554812. [PMID: 37786708 PMCID: PMC10541604 DOI: 10.1101/2023.08.25.554812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2023]
Abstract
Developmental neurogenesis is a tightly regulated spatiotemporal process with its dysregulation implicated in neurodevelopmental disorders. NMDA receptors are glutamate-gated ion channels that are widely expressed in the early nervous system, yet their contribution to neurogenesis is poorly understood. Notably, a variety of mutations in genes encoding NMDA receptor subunits are associated with neurodevelopmental disorders. To rigorously define the role of NMDA receptors in developmental neurogenesis, we used a mutant zebrafish line ( grin1 -/- ) that lacks all NMDA receptors yet survives to 10 days post-fertilization, offering the opportunity to study post-embryonic neurodevelopment in the absence of NMDA receptors. Focusing on the forebrain, we find that these fish have a progressive supernumerary neuron phenotype confined to the telencephalon at the end of embryonic neurogenesis, but which extends to all forebrain regions during postembryonic neurogenesis. This enhanced neuron population does not arise directly from increased numbers or mitotic activity of radial glia cells, the principal neural stem cells. Rather, it stems from a lack of timely maturation of transit-amplifying neuroblasts into post-mitotic neurons, as indicated by a decrease in expression of the ontogenetically-expressed chloride transporter, KCC2. Pharmacological blockade with MK-801 recapitulates the grin1 -/- supernumerary neuron phenotype, indicating a requirement for ionotropic signaling. Thus, NMDA receptors are required for suppression of indirect, transit amplifying cell-driven neurogenesis by promoting maturational termination of mitosis. Loss of suppression results in neuronal overpopulation that can fundamentally change brain circuitry and may be a key factor in pathogenesis of neurodevelopmental disorders caused by NMDA receptor dysfunction.
Collapse
|
24
|
Yang C, Shitamukai A, Yang S, Kawaguchi A. Advanced Techniques Using In Vivo Electroporation to Study the Molecular Mechanisms of Cerebral Development Disorders. Int J Mol Sci 2023; 24:14128. [PMID: 37762431 PMCID: PMC10531473 DOI: 10.3390/ijms241814128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/12/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
The mammalian cerebral cortex undergoes a strictly regulated developmental process. Detailed in situ visualizations, imaging of these dynamic processes, and in vivo functional gene studies significantly enhance our understanding of brain development and related disorders. This review introduces basic techniques and recent advancements in in vivo electroporation for investigating the molecular mechanisms underlying cerebral diseases. In utero electroporation (IUE) is extensively used to visualize and modify these processes, including the forced expression of pathological mutants in human diseases; thus, this method can be used to establish animal disease models. The advent of advanced techniques, such as genome editing, including de novo knockout, knock-in, epigenetic editing, and spatiotemporal gene regulation, has further expanded our list of investigative tools. These tools include the iON expression switch for the precise control of timing and copy numbers of exogenous genes and TEMPO for investigating the temporal effects of genes. We also introduce the iGONAD method, an improved genome editing via oviductal nucleic acid delivery approach, as a novel genome-editing technique that has accelerated brain development exploration. These advanced in vivo electroporation methods are expected to provide valuable insights into pathological conditions associated with human brain disorders.
Collapse
Affiliation(s)
- Chen Yang
- Human Anatomy and Histology and Embryology, School of Basic Medicine, Harbin Medical University, Harbin 150081, China
- Department of Human Morphology, Okayama University Graduate School of Medicine, Density and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Atsunori Shitamukai
- Department of Human Morphology, Okayama University Graduate School of Medicine, Density and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Shucai Yang
- Human Anatomy and Histology and Embryology, School of Basic Medicine, Harbin Medical University, Harbin 150081, China
| | - Ayano Kawaguchi
- Department of Human Morphology, Okayama University Graduate School of Medicine, Density and Pharmaceutical Sciences, Okayama 700-8558, Japan
| |
Collapse
|
25
|
Bohuslavova R, Fabriciova V, Smolik O, Lebrón-Mora L, Abaffy P, Benesova S, Zucha D, Valihrach L, Berkova Z, Saudek F, Pavlinkova G. NEUROD1 reinforces endocrine cell fate acquisition in pancreatic development. Nat Commun 2023; 14:5554. [PMID: 37689751 PMCID: PMC10492842 DOI: 10.1038/s41467-023-41306-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 08/30/2023] [Indexed: 09/11/2023] Open
Abstract
NEUROD1 is a transcription factor that helps maintain a mature phenotype of pancreatic β cells. Disruption of Neurod1 during pancreatic development causes severe neonatal diabetes; however, the exact role of NEUROD1 in the differentiation programs of endocrine cells is unknown. Here, we report a crucial role of the NEUROD1 regulatory network in endocrine lineage commitment and differentiation. Mechanistically, transcriptome and chromatin landscape analyses demonstrate that Neurod1 inactivation triggers a downregulation of endocrine differentiation transcription factors and upregulation of non-endocrine genes within the Neurod1-deficient endocrine cell population, disturbing endocrine identity acquisition. Neurod1 deficiency altered the H3K27me3 histone modification pattern in promoter regions of differentially expressed genes, which resulted in gene regulatory network changes in the differentiation pathway of endocrine cells, compromising endocrine cell potential, differentiation, and functional properties.
Collapse
Affiliation(s)
- Romana Bohuslavova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology CAS, 25250, Vestec, Czechia
| | - Valeria Fabriciova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology CAS, 25250, Vestec, Czechia
| | - Ondrej Smolik
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology CAS, 25250, Vestec, Czechia
| | - Laura Lebrón-Mora
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology CAS, 25250, Vestec, Czechia
| | - Pavel Abaffy
- Laboratory of Gene Expression, Institute of Biotechnology CAS, 25250, Vestec, Czechia
| | - Sarka Benesova
- Laboratory of Gene Expression, Institute of Biotechnology CAS, 25250, Vestec, Czechia
| | - Daniel Zucha
- Laboratory of Gene Expression, Institute of Biotechnology CAS, 25250, Vestec, Czechia
| | - Lukas Valihrach
- Laboratory of Gene Expression, Institute of Biotechnology CAS, 25250, Vestec, Czechia
| | - Zuzana Berkova
- Diabetes Centre, Experimental Medicine Centre, Institute for Clinical and Experimental Medicine, 14021, Prague, Czechia
| | - Frantisek Saudek
- Diabetes Centre, Experimental Medicine Centre, Institute for Clinical and Experimental Medicine, 14021, Prague, Czechia
| | - Gabriela Pavlinkova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology CAS, 25250, Vestec, Czechia.
| |
Collapse
|
26
|
Kim M, Jun S, Park H, Tanaka-Yamamoto K, Yamamoto Y. Regulation of cerebellar network development by granule cells and their molecules. Front Mol Neurosci 2023; 16:1236015. [PMID: 37520428 PMCID: PMC10375027 DOI: 10.3389/fnmol.2023.1236015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 06/27/2023] [Indexed: 08/01/2023] Open
Abstract
The well-organized cerebellar structures and neuronal networks are likely crucial for their functions in motor coordination, motor learning, cognition, and emotion. Such cerebellar structures and neuronal networks are formed during developmental periods through orchestrated mechanisms, which include not only cell-autonomous programs but also interactions between the same or different types of neurons. Cerebellar granule cells (GCs) are the most numerous neurons in the brain and are generated through intensive cell division of GC precursors (GCPs) during postnatal developmental periods. While GCs go through their own developmental processes of proliferation, differentiation, migration, and maturation, they also play a crucial role in cerebellar development. One of the best-characterized contributions is the enlargement and foliation of the cerebellum through massive proliferation of GCPs. In addition to this contribution, studies have shown that immature GCs and GCPs regulate multiple factors in the developing cerebellum, such as the development of other types of cerebellar neurons or the establishment of afferent innervations. These studies have often found impairments of cerebellar development in animals lacking expression of certain molecules in GCs, suggesting that the regulations are mediated by molecules that are secreted from or present in GCs. Given the growing recognition of GCs as regulators of cerebellar development, this review will summarize our current understanding of cerebellar development regulated by GCs and molecules in GCs, based on accumulated studies and recent findings, and will discuss their potential further contributions.
Collapse
Affiliation(s)
- Muwoong Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul, Republic of Korea
| | - Soyoung Jun
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul, Republic of Korea
| | - Heeyoun Park
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Keiko Tanaka-Yamamoto
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul, Republic of Korea
| | - Yukio Yamamoto
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| |
Collapse
|
27
|
Hong H, Yoon SB, Park JE, Lee JI, Kim HY, Nam HJ, Cho H. MeCP2 dysfunction prevents proper BMP signaling and neural progenitor expansion in brain organoid. Ann Clin Transl Neurol 2023. [PMID: 37302988 DOI: 10.1002/acn3.51799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 04/26/2023] [Accepted: 05/10/2023] [Indexed: 06/13/2023] Open
Abstract
OBJECTIVES Sporadic mutations in MeCP2 are a hallmark of Rett syndrome (RTT). Many RTT brain organoid models have exhibited pathogenic phenotypes such as decreased spine density and small size of soma with altered electrophysiological signals. However, previous models are mainly focused on the phenotypes observed in the late phase and rarely provide clues for the defect of neural progenitors which generate different types of neurons and glial cells. METHODS We newly established the RTT brain organoid model derived from MeCP2-truncated iPS cells which were genetically engineered by CRISPR/Cas9 technology. By immunofluorescence imaging, we studied the development of NPC pool and its fate specification into glutamatergic neurons or astrocytes in RTT organoids. By total RNA sequencing, we investigated which signaling pathways were altered during the early brain development in RTT organoids. RESULTS Dysfunction of MeCP2 caused the defect of neural rosette formation in the early phase of cortical development. In total transcriptome analysis, BMP pathway-related genes are highly associated with MeCP2 depletion. Moreover, levels of pSMAD1/5 and BMP target genes are excessively increased, and treatment of BMP inhibitors partially rescues the cell cycle progression of neural progenitors. Subsequently, MeCP2 dysfunction reduced the glutamatergic neurogenesis and induced overproduction of astrocytes. Nevertheless, early inhibition of BMP pathway rescued VGLUT1 expression and suppressed astrocyte maturation. INTERPRETATION Our results demonstrate that MeCP2 is required for the expansion of neural progenitor cells by modulating BMP pathway at early stages of development, and this influence persists during neurogenesis and gliogenesis at later stages of brain organoid development.
Collapse
Affiliation(s)
- Hyowon Hong
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong-gu, Daejeon, Republic of Korea
| | - Sae-Bom Yoon
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong-gu, Daejeon, Republic of Korea
| | - Jung Eun Park
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong-gu, Daejeon, Republic of Korea
| | - Jung In Lee
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong-gu, Daejeon, Republic of Korea
| | - Hyun Young Kim
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong-gu, Daejeon, Republic of Korea
| | - Hye Jin Nam
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong-gu, Daejeon, Republic of Korea
| | - Heeyeong Cho
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong-gu, Daejeon, Republic of Korea
| |
Collapse
|
28
|
McHale-Matthews AC, DeCampo DM, Love T, Cameron JL, Fudge JL. Immature neurons in the primate amygdala: Changes with early development and disrupted early environment. Dev Cogn Neurosci 2023; 61:101248. [PMID: 37120994 PMCID: PMC10173404 DOI: 10.1016/j.dcn.2023.101248] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 03/11/2023] [Accepted: 04/21/2023] [Indexed: 05/02/2023] Open
Abstract
In human and nonhuman primates, the amygdala paralaminar nucleus (PL) contains immature neurons. To explore the PL's potential for cellular growth during development, we compared PL neurons in (1) infant and adolescent macaques (control, maternally-reared), and in (2) infant macaques that experienced separation from their mother in the first month of life compared to control maternally-reared infants. In maternally-reared animals, the adolescent PL had fewer immature neurons, more mature neurons, and larger immature soma volumes compared to infant PL. There were also fewer total neurons (immature plus mature) in adolescent versus infant PL, suggesting that some neurons move out of the PL by adolescence. Maternal separation did not change mean immature or mature neuron counts in infant PL. However, across all infant animals, immature neuron soma volume was strongly correlated with mature neuron counts. TBR1 mRNA, a transcript required for glutamatergic neuron maturation, is significantly reduced in the maternally-separated infant PL (DeCampo et al., 2017), and was also positively correlated with mature neuron counts in infant PL. We conclude that immature neurons gradually mature by adolescence, and that the stress of maternal separation may shift this trajectory, as revealed by correlations between TBR1 mRNA and mature neuron numbers across animals.
Collapse
Affiliation(s)
- Alexandra C McHale-Matthews
- University of Rochester, School of Medicine and Dentistry, Department of Neuroscience, Rochester, NY 14642, USA
| | | | - Tanzy Love
- University of Rochester, School of Medicine and Dentistry, Department of Biostatistics, Rochester, NY 14642, USA
| | - Judy L Cameron
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| | - Julie L Fudge
- University of Rochester, School of Medicine and Dentistry, Department of Neuroscience, Rochester, NY 14642, USA; University of Rochester, School of Medicine and Dentistry, Department of Psychiatry, Rochester, NY 14642, USA
| |
Collapse
|
29
|
McHale-Matthews AC, DeCampo DM, Love T, Cameron JL, Fudge JL. Immature neurons in the primate amygdala: changes with early development and disrupted early environment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.10.528076. [PMID: 36798176 PMCID: PMC9934690 DOI: 10.1101/2023.02.10.528076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/14/2023]
Abstract
In human and nonhuman primates, the amygdala paralaminar nucleus (PL) contains immature neurons. To explore the PL’s potential for cellular growth during development, we compared PL cells in 1) infant and adolescent macaques (control, maternally-reared), and in 2) infant macaques that experienced separation from their mother in the first month of life. In maternally-reared animals, the adolescent PL had fewer immature neurons, more mature neurons, and larger immature soma volumes compared to infant PL. There were also fewer total neurons (immature plus mature) in adolescent versus infant PL, suggesting that some neurons move out of the PL by adolescence. Maternal separation did not change mean immature or mature neuron counts in infant PL. However, across all infant animals, immature neuron soma volume was strongly correlated with mature neuron counts. tbr-1 mRNA, a transcript required for glutamatergic neuron maturation, is significantly reduced in the maternally-separated infant PL (DeCampo et al, 2017), and was also positively correlated with mature neuron counts in infant PL. We conclude that immature neurons gradually mature by adolescence, and that the stress of maternal separation may shift this trajectory, as revealed by correlations between tbr1mRNA and mature neuron numbers across animals.
Collapse
Affiliation(s)
| | | | - Tanzy Love
- University of Rochester, School of Medicine and Dentistry, Department of Biostatistics, Rochester, NY 14642
| | - Judy L Cameron
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213
| | - Julie L Fudge
- University of Rochester, School of Medicine and Dentistry Department of Neuroscience Rochester, NY 14642
- University of Rochester, School of Medicine and Dentistry, Department of Psychiatry Rochester, NY 14642
| |
Collapse
|
30
|
Heise J, Schmitz T, Bührer C, Endesfelder S. Protective Effects of Early Caffeine Administration in Hyperoxia-Induced Neurotoxicity in the Juvenile Rat. Antioxidants (Basel) 2023; 12:295. [PMID: 36829854 PMCID: PMC9952771 DOI: 10.3390/antiox12020295] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/12/2023] [Accepted: 01/24/2023] [Indexed: 01/31/2023] Open
Abstract
High-risk preterm infants are affected by a higher incidence of cognitive developmental deficits due to the unavoidable risk factor of oxygen toxicity. Caffeine is known to have a protective effect in preventing bronchopulmonary dysplasia associated with improved neurologic outcomes, although very early initiation of therapy is controversial. In this study, we used newborn rats in an oxygen injury model to test the hypothesis that near-birth caffeine administration modulates neuronal maturation and differentiation in the hippocampus of the developing brain. For this purpose, newborn Wistar rats were exposed to 21% or 80% oxygen on the day of birth for 3 or 5 days and treated with vehicle or caffeine (10 mg/kg/48 h). Postnatal exposure to 80% oxygen resulted in a drastic reduction of associated neuronal mediators for radial glia, mitotic/postmitotic neurons, and impaired cell-cycle regulation, predominantly persistent even after recovery to room air until postnatal day 15. Systemic caffeine administration significantly counteracted the effects of oxygen insult on neuronal maturation in the hippocampus. Interestingly, under normoxia, caffeine inhibited the transcription of neuronal mediators of maturing and mature neurons. The early administration of caffeine modulated hyperoxia-induced decreased neurogenesis in the hippocampus and showed neuroprotective properties in the neonatal rat oxygen toxicity model.
Collapse
Affiliation(s)
| | | | | | - Stefanie Endesfelder
- Department of Neonatology, Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany
| |
Collapse
|
31
|
Abd-Nikfarjam B, Dolati-Somarin A, Baradaran Rahimi V, Askari VR. Cannabinoids in neuroinflammatory disorders: Focusing on multiple sclerosis, Parkinsons, and Alzheimers diseases. Biofactors 2023. [PMID: 36637897 DOI: 10.1002/biof.1936] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 12/29/2022] [Indexed: 01/14/2023]
Abstract
The medicinal properties of cannabis and cannabinoid-derivative are entirely investigated and known. In addition, the identification of psychotropic plant cannabinoids has led to more studies regarding the cannabinoid system and its therapeutic features in the treatment and management of clinical symptoms of neuroinflammatory disorders, such as multiple sclerosis (MS), Parkinsons disease (PD), and Alzheimers disease (AD). In fact, cannabinoid agonists are able to control and regulate inflammatory responses. In contrast to the cannabinoid receptor type 1 (CB1) and its unwanted adverse effects, the cannabinoid receptor type 2 (CB2) and its ligands hold promise for new and effective therapeutic approaches. So far, some successes have been achieved in this field. This review will discuss an outline of the endocannabinoid system's involvement in neuroinflammatory disorders. Moreover, the pharmacological efficacy of different natural and synthetic preparations of phytocannabinoids acting on cannabinoid receptors, particularly in MS, PD, and AD, will be updated. Also, the reasons for targeting CB2 for neurodegeneration will be explained.
Collapse
Affiliation(s)
- Bahareh Abd-Nikfarjam
- Department of Immunology, School of Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | | | - Vafa Baradaran Rahimi
- Department of Cardiovascular Diseases, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vahid Reza Askari
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
32
|
Liu X, Adamo AM, Oteiza PI. Di-2-ethylhexyl phthalate affects zinc metabolism and neurogenesis in the developing rat brain. Arch Biochem Biophys 2022; 727:109351. [PMID: 35841924 DOI: 10.1016/j.abb.2022.109351] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/07/2022] [Accepted: 07/08/2022] [Indexed: 12/28/2022]
Abstract
We previously observed that developmental marginal zinc deficiency affects neurogenesis. Maternal phthalate exposure could disrupt fetal zinc homeostasis by triggering an acute phase response, causing maternal liver zinc retention that limits zinc availability to the fetus. Thus, we currently investigated whether exposure to di-2-ethylhexyl phthalate (DEHP) during gestation in rats alters fetal brain neurogenesis by impairing zinc homeostasis. Dams consumed an adequate (25 μg zinc/g diet) (C) or a marginal zinc deficient (MZD) (10 μg zinc/g diet) diet, without or with DEHP (300 mg/kg BW) (C + DEHP, MZD + DEHP) from embryonic day (E) 0 to E19. To evaluate neurogenesis we measured parameters of neural progenitor cells (NPC) proliferation and differentiation. Maternal exposure to DEHP and/or zinc deficiency lowered fetal brain cortical tissue (CT) zinc concentrations. Transcription factors involved in NPC proliferation (PAX6, SOX2, EMX1), differentiation (TBR2, TBR1) and mature neurons (NeuN) were lower in MZD, MZD + DEHP and C + DEHP than in C E19 brain CT, being the lowest in the MZD + DEHP group. VGLUT1 levels, a marker of glutamatergic neurons, showed a similar pattern. Levels of a marker of GABAergic neurons, GAD65, did not vary among groups. Phosphorylated ERK1/2 levels were reduced by both MZD and DEHP, and particularly in the MZD + DEHP group. MEHP-treated human neuroblastoma IMR-32 cells and E19 brains from DEHP-treated dams showed that the zinc-regulated phosphatase PP2A can be in part responsible for DEHP-mediated ERK1/2 downregulation and impaired neurogenesis. Overall, gestational exposure to DEHP caused secondary zinc deficiency and impaired neurogenesis. These harmful effects could have long-term consequences on the adult offspring brain structure and function.
Collapse
Affiliation(s)
- Xiuzhen Liu
- Department of Nutrition, University of California, Davis, Davis, CA, USA; Department of Environmental Toxicology, University of California, Davis, Davis, CA, USA
| | - Ana M Adamo
- Departamento de Química Biológica and IQUIFIB (UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Patricia I Oteiza
- Department of Nutrition, University of California, Davis, Davis, CA, USA; Department of Environmental Toxicology, University of California, Davis, Davis, CA, USA.
| |
Collapse
|
33
|
Kelenis DP, Rodarte KE, Kollipara RK, Pozo K, Choudhuri SP, Spainhower KB, Wait SJ, Stastny V, Oliver TG, Johnson JE. Inhibition of Karyopherin β1-Mediated Nuclear Import Disrupts Oncogenic Lineage-Defining Transcription Factor Activity in Small Cell Lung Cancer. Cancer Res 2022; 82:3058-3073. [PMID: 35748745 PMCID: PMC9444950 DOI: 10.1158/0008-5472.can-21-3713] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 04/29/2022] [Accepted: 06/15/2022] [Indexed: 11/16/2022]
Abstract
Genomic studies support the classification of small cell lung cancer (SCLC) into subtypes based on the expression of lineage-defining transcription factors ASCL1 and NEUROD1, which together are expressed in ∼86% of SCLC. ASCL1 and NEUROD1 activate SCLC oncogene expression, drive distinct transcriptional programs, and maintain the in vitro growth and oncogenic properties of ASCL1 or NEUROD1-expressing SCLC. ASCL1 is also required for tumor formation in SCLC mouse models. A strategy to inhibit the activity of these oncogenic drivers may therefore provide both a targeted therapy for the predominant SCLC subtypes and a tool to investigate the underlying lineage plasticity of established SCLC tumors. However, there are no known agents that inhibit ASCL1 or NEUROD1 function. In this study, we identify a novel strategy to pharmacologically target ASCL1 and NEUROD1 activity in SCLC by exploiting the nuclear localization required for the function of these transcription factors. Karyopherin β1 (KPNB1) was identified as a nuclear import receptor for both ASCL1 and NEUROD1 in SCLC, and inhibition of KPNB1 led to impaired ASCL1 and NEUROD1 nuclear accumulation and transcriptional activity. Pharmacologic targeting of KPNB1 preferentially disrupted the growth of ASCL1+ and NEUROD1+ SCLC cells in vitro and suppressed ASCL1+ tumor growth in vivo, an effect mediated by a combination of impaired ASCL1 downstream target expression, cell-cycle activity, and proteostasis. These findings broaden the support for targeting nuclear transport as an anticancer therapeutic strategy and have implications for targeting lineage-transcription factors in tumors beyond SCLC. SIGNIFICANCE The identification of KPNB1 as a nuclear import receptor for lineage-defining transcription factors in SCLC reveals a viable therapeutic strategy for cancer treatment.
Collapse
Affiliation(s)
- Demetra P. Kelenis
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Kathia E. Rodarte
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Rahul K. Kollipara
- McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Karine Pozo
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA,Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | | | - Kyle B. Spainhower
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Sarah J. Wait
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Victor Stastny
- Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Trudy G. Oliver
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Jane E. Johnson
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
34
|
Hussan MT, Sakai A, Matsui H. Glutamatergic pathways in the brains of turtles: A comparative perspective among reptiles, birds, and mammals. Front Neuroanat 2022; 16:937504. [PMID: 36059432 PMCID: PMC9428285 DOI: 10.3389/fnana.2022.937504] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 07/22/2022] [Indexed: 11/13/2022] Open
Abstract
Glutamate acts as the main excitatory neurotransmitter in the brain and plays a vital role in physiological and pathological neuronal functions. In mammals, glutamate can cause detrimental excitotoxic effects under anoxic conditions. In contrast, Trachemys scripta, a freshwater turtle, is one of the most anoxia-tolerant animals, being able to survive up to months without oxygen. Therefore, turtles have been investigated to assess the molecular mechanisms of neuroprotective strategies used by them in anoxic conditions, such as maintaining low levels of glutamate, increasing adenosine and GABA, upregulating heat shock proteins, and downregulating KATP channels. These mechanisms of anoxia tolerance of the turtle brain may be applied to finding therapeutics for human glutamatergic neurological disorders such as brain injury or cerebral stroke due to ischemia. Despite the importance of glutamate as a neurotransmitter and of the turtle as an ideal research model, the glutamatergic circuits in the turtle brain remain less described whereas they have been well studied in mammalian and avian brains. In reptiles, particularly in the turtle brain, glutamatergic neurons have been identified by examining the expression of vesicular glutamate transporters (VGLUTs). In certain areas of the brain, some ionotropic glutamate receptors (GluRs) have been immunohistochemically studied, implying that there are glutamatergic target areas. Based on the expression patterns of these glutamate-related molecules and fiber connection data of the turtle brain that is available in the literature, many candidate glutamatergic circuits could be clarified, such as the olfactory circuit, hippocampal–septal pathway, corticostriatal pathway, visual pathway, auditory pathway, and granule cell–Purkinje cell pathway. This review summarizes the probable glutamatergic pathways and the distribution of glutamatergic neurons in the pallium of the turtle brain and compares them with those of avian and mammalian brains. The integrated knowledge of glutamatergic pathways serves as the fundamental basis for further functional studies in the turtle brain, which would provide insights on physiological and pathological mechanisms of glutamate regulation as well as neural circuits in different species.
Collapse
Affiliation(s)
- Mohammad Tufazzal Hussan
- Department of Neuroscience of Disease, Brain Research Institute, Niigata University, Niigata, Japan
- Department of Anatomy and Histology, Patuakhali Science and Technology University, Barishal, Bangladesh
- *Correspondence: Mohammad Tufazzal Hussan,
| | - Akiko Sakai
- Department of Neuroscience of Disease, Brain Research Institute, Niigata University, Niigata, Japan
| | - Hideaki Matsui
- Department of Neuroscience of Disease, Brain Research Institute, Niigata University, Niigata, Japan
- Hideaki Matsui,
| |
Collapse
|
35
|
Benedetti V, Banfi F, Zaghi M, Moll-Diaz R, Massimino L, Argelich L, Bellini E, Bido S, Muggeo S, Ordazzo G, Mastrototaro G, Moneta M, Sessa A, Broccoli V. A SOX2-engineered epigenetic silencer factor represses the glioblastoma genetic program and restrains tumor development. SCIENCE ADVANCES 2022; 8:eabn3986. [PMID: 35921410 PMCID: PMC9348799 DOI: 10.1126/sciadv.abn3986] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 06/14/2022] [Indexed: 06/15/2023]
Abstract
Current therapies remain unsatisfactory in preventing the recurrence of glioblastoma multiforme (GBM), which leads to poor patient survival. By rational engineering of the transcription factor SOX2, a key promoter of GBM malignancy, together with the Kruppel-associated box and DNA methyltransferase3A/L catalytic domains, we generated a synthetic repressor named SOX2 epigenetic silencer (SES), which induces the transcriptional silencing of its original targets. By doing so, SES kills both glioma cell lines and patient-derived cancer stem cells in vitro and in vivo. SES expression, through local viral delivery in mouse xenografts, induces strong regression of human tumors and survival rescue. Conversely, SES is not harmful to neurons and glia, also thanks to a minimal promoter that restricts its expression in mitotically active cells, rarely present in the brain parenchyma. Collectively, SES produces a significant silencing of a large fraction of the SOX2 transcriptional network, achieving high levels of efficacy in repressing aggressive brain tumors.
Collapse
Affiliation(s)
- Valerio Benedetti
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Federica Banfi
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- CNR Institute of Neuroscience, 20129 Milan, Italy
| | - Mattia Zaghi
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Raquel Moll-Diaz
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Luca Massimino
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Laura Argelich
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Edoardo Bellini
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Simone Bido
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Sharon Muggeo
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Gabriele Ordazzo
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Giuseppina Mastrototaro
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Matteo Moneta
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Alessandro Sessa
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Vania Broccoli
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- CNR Institute of Neuroscience, 20129 Milan, Italy
| |
Collapse
|
36
|
Brancaccio P, Anzilotti S, Cuomo O, Vinciguerra A, Campanile M, Herchuelz A, Amoroso S, Annunziato L, Pignataro G. Preconditioning in hypoxic-ischemic neonate mice triggers Na +-Ca 2+ exchanger-dependent neurogenesis. Cell Death Dis 2022; 8:318. [PMID: 35831286 PMCID: PMC9279453 DOI: 10.1038/s41420-022-01089-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 06/08/2022] [Accepted: 06/13/2022] [Indexed: 11/09/2022]
Abstract
To identify alternative interventions in neonatal hypoxic-ischemic encephalopathy, researchers’ attention has been focused to the study of endogenous neuroprotective strategies. Based on the preconditioning concept that a subthreshold insult may protect from a subsequent harmful event, we aimed at identifying a new preconditioning protocol able to enhance Ca2+-dependent neurogenesis in a mouse model of neonatal hypoxia ischemia (HI). To this purpose, we also investigated the role of the preconditioning-linked protein controlling ionic homeostasis, Na+/Ca2+ exchanger (NCX). Hypoxic Preconditioning (HPC) was reproduced by exposing P7 mice to 20’ hypoxia. HI was induced by isolating and cutting the right common carotid artery. A significant reduction in ischemic damage was observed in mice subjected to 20’ hypoxia followed,3 days later, by 60’ HI, thus suggesting that 20’ hypoxia functions as preconditioning stimulus. HPC promoted neuroblasts proliferation in the dentate gyrus mirrored by an increase of NCX1 and NCX3-positive cells and an improvement of behavioral motor performances in HI mice. An attenuation of HPC neuroprotection as well as a reduction in the expression of neurogenesis markers, including p57 and NeuroD1, was observed in preconditioned mice lacking NCX1 or NCX3. In summary, PC in neonatal mice triggers a neurogenic process linked to ionic homeostasis maintenance, regulated by NCX1 and NCX3.
Collapse
Affiliation(s)
- P Brancaccio
- Division of Pharmacology, Department of Neuroscience, School of Medicine, University of Naples "Federico II", 80131, Naples, Italy
| | - S Anzilotti
- Department of Science and Technology, University of Sannio, 82100, Benevento, Italy
| | - O Cuomo
- Division of Pharmacology, Department of Neuroscience, School of Medicine, University of Naples "Federico II", 80131, Naples, Italy
| | - A Vinciguerra
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", 60126, Ancona, Italy
| | - M Campanile
- Division of Pharmacology, Department of Neuroscience, School of Medicine, University of Naples "Federico II", 80131, Naples, Italy
| | - A Herchuelz
- Laboratoire de Pharmacodynamie et de Therapeutique-Faculté de Médecine Université Libre de Bruxelles, Bruxelles, Belgium
| | - S Amoroso
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", 60126, Ancona, Italy
| | - L Annunziato
- IRCCS Synlab SDN S.p.A, via Gianturco 113, 80143, Naples, Italy
| | - G Pignataro
- Division of Pharmacology, Department of Neuroscience, School of Medicine, University of Naples "Federico II", 80131, Naples, Italy.
| |
Collapse
|
37
|
Fernández Acosta FJ, Luque-Molina I, Vecino R, Díaz-Guerra E, Defterali Ç, Pignatelli J, Vicario C. Morphological Diversity of Calretinin Interneurons Generated From Adult Mouse Olfactory Bulb Core Neural Stem Cells. Front Cell Dev Biol 2022; 10:932297. [PMID: 35846352 PMCID: PMC9277347 DOI: 10.3389/fcell.2022.932297] [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: 04/29/2022] [Accepted: 06/08/2022] [Indexed: 11/19/2022] Open
Abstract
Neural stem cells (NSCs) in the olfactory bulb (OB) core can generate mature interneurons in the adult mice brain. The vast majority of these adult generated cells express the calcium-binding protein Calretinin (CalR), and they migrate towards different OB layers. However, these cells have yet to be fully characterized and hence, to achieve this we injected retroviral particles expressing GFP into the OB core of adult animals and found that the CalR+ neurons generated from NSCs mainly migrate to the granule cell layer (GCL) and glomerular layer (GL) in similar proportions. In addition, since morphology and function are closely related, we used three-dimensional imaging techniques to analyze the morphology of these adult born cells, describing new subtypes of CalR+ interneurons based on their dendritic arborizations and projections, as well as their localization in the GCL or GL. We also show that the migration and morphology of these newly generated neurons can be altered by misexpressing the transcription factor Tbr1 in the OB core. Therefore, the morphology acquired by neurons located in a specific OB layer is the result of a combination of both extrinsic (e.g., layer allocation) and intrinsic mechanisms (e.g., transcription factors). Defining the cellular processes and molecular mechanisms that govern adult neurogenesis might help better understand brain circuit formation and plasticity, as well as eventually opening the way to develop strategies for brain repair.
Collapse
Affiliation(s)
| | - Inma Luque-Molina
- Instituto Cajal (IC), CSIC, Madrid, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Rebeca Vecino
- Instituto Cajal (IC), CSIC, Madrid, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Eva Díaz-Guerra
- Instituto Cajal (IC), CSIC, Madrid, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Çagla Defterali
- Instituto Cajal (IC), CSIC, Madrid, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Jaime Pignatelli
- Instituto Cajal (IC), CSIC, Madrid, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Carlos Vicario
- Instituto Cajal (IC), CSIC, Madrid, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- *Correspondence: Carlos Vicario,
| |
Collapse
|
38
|
Early Signs of Molecular Defects in iPSC-Derived Neural Stems Cells from Patients with Familial Parkinson’s Disease. Biomolecules 2022; 12:biom12070876. [PMID: 35883433 PMCID: PMC9313424 DOI: 10.3390/biom12070876] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/14/2022] [Accepted: 06/18/2022] [Indexed: 11/17/2022] Open
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disorder, classically associated with extensive loss of dopaminergic neurons of the substantia nigra pars compacta. The hallmark of the disease is the accumulation of pathogenic conformations of the presynaptic protein, α-synuclein (αSyn), and the formation of intraneuronal protein aggregate inclusions. Neurodegeneration of dopamine neurons leads to a prominent dopaminergic deficiency in the basal ganglia, responsible for motor disturbances. However, it is now recognized that the disease involves more widespread neuronal dysfunction, leading to early and late non-motor symptoms. The development of in vitro systems based on the differentiation of human-induced pluripotent stem cells provides us the unique opportunity to monitor alterations at the cellular and molecular level throughout the differentiation procedure and identify perturbations that occur early, even at the neuronal precursor stage. Here we aim to identify whether p.A53T-αSyn induced disturbances at the molecular level are already present in neural precursors. Towards this, we present data from transcriptomics analysis of control and p.A53T-αSyn NPCs showing altered expression in transcripts involved in axon guidance, adhesion, synaptogenesis, ion transport, and metabolism. The comparative analysis with the transcriptomics profile of p.A53T-αSyn neurons shows both distinct and overlapping pathways leading to neurodegeneration while meta-analysis with transcriptomics data from both neurodegenerative and neurodevelopmental disorders reveals that p.A53T-pathology has a significant overlap with the latter category. This is the first study showing that molecular dysregulation initiates early at the p.A53T-αSyn NPC level, suggesting that synucleinopathies may have a neurodevelopmental component.
Collapse
|
39
|
Ochi S, Manabe S, Kikkawa T, Osumi N. Thirty Years' History since the Discovery of Pax6: From Central Nervous System Development to Neurodevelopmental Disorders. Int J Mol Sci 2022; 23:6115. [PMID: 35682795 PMCID: PMC9181425 DOI: 10.3390/ijms23116115] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 05/19/2022] [Accepted: 05/27/2022] [Indexed: 12/23/2022] Open
Abstract
Pax6 is a sequence-specific DNA binding transcription factor that positively and negatively regulates transcription and is expressed in multiple cell types in the developing and adult central nervous system (CNS). As indicated by the morphological and functional abnormalities in spontaneous Pax6 mutant rodents, Pax6 plays pivotal roles in various biological processes in the CNS. At the initial stage of CNS development, Pax6 is responsible for brain patterning along the anteroposterior and dorsoventral axes of the telencephalon. Regarding the anteroposterior axis, Pax6 is expressed inversely to Emx2 and Coup-TF1, and Pax6 mutant mice exhibit a rostral shift, resulting in an alteration of the size of certain cortical areas. Pax6 and its downstream genes play important roles in balancing the proliferation and differentiation of neural stem cells. The Pax6 gene was originally identified in mice and humans 30 years ago via genetic analyses of the eye phenotypes. The human PAX6 gene was discovered in patients who suffer from WAGR syndrome (i.e., Wilms tumor, aniridia, genital ridge defects, mental retardation). Mutations of the human PAX6 gene have also been reported to be associated with autism spectrum disorder (ASD) and intellectual disability. Rodents that lack the Pax6 gene exhibit diverse neural phenotypes, which might lead to a better understanding of human pathology and neurodevelopmental disorders. This review describes the expression and function of Pax6 during brain development, and their implications for neuropathology.
Collapse
Affiliation(s)
| | | | | | - Noriko Osumi
- Department of Developmental Neuroscience, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan; (S.O.); (S.M.); (T.K.)
| |
Collapse
|
40
|
Bond AM, Ming GL, Song H. What Is the Relationship Between Hippocampal Neurogenesis Across Different Stages of the Lifespan? Front Neurosci 2022; 16:891713. [PMID: 35685774 PMCID: PMC9173723 DOI: 10.3389/fnins.2022.891713] [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: 03/08/2022] [Accepted: 04/25/2022] [Indexed: 12/02/2022] Open
Abstract
Hippocampal neurogenesis has typically been studied during embryonic development or in adulthood, promoting the perception of two distinct phenomena. We propose a perspective that hippocampal neurogenesis in the mammalian brain is one continuous, lifelong developmental process. We summarize the common features of hippocampal neurogenesis that are maintained across the lifespan, as well as dynamic age-dependent properties. We highlight that while the progression of hippocampal neurogenesis across the lifespan is conserved between mammalian species, the timing of this progression is species-dependent. Finally, we discuss some current challenges in the hippocampus neurogenesis field, and future research directions to address them, such as time course analysis across the lifespan, mechanisms regulating neurogenesis progression, and interspecies comparisons. We hope that this new perspective of hippocampal neurogenesis will prompt fresh insight into previous research and inspire new directions to advance the field to identify biologically significant ways to harness the endogenous capacity for neurogenesis in the hippocampus.
Collapse
Affiliation(s)
- Allison M. Bond
- Department of Neuroscience and Mahoney Institute for Neurosciences, 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,Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States,Institute for Regenerative Medicine, 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, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States,The Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States,*Correspondence: Hongjun Song,
| |
Collapse
|
41
|
Early Life Events and Maturation of the Dentate Gyrus: Implications for Neurons and Glial Cells. Int J Mol Sci 2022; 23:ijms23084261. [PMID: 35457079 PMCID: PMC9031216 DOI: 10.3390/ijms23084261] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 04/08/2022] [Accepted: 04/11/2022] [Indexed: 12/15/2022] Open
Abstract
The dentate gyrus (DG), an important part of the hippocampus, plays a significant role in learning, memory, and emotional behavior. Factors potentially influencing normal development of neurons and glial cells in the DG during its maturation can exert long-lasting effects on brain functions. Early life stress may modify maturation of the DG and induce lifelong alterations in its structure and functioning, underlying brain pathologies in adults. In this paper, maturation of neurons and glial cells (microglia and astrocytes) and the effects of early life events on maturation processes in the DG have been comprehensively reviewed. Early postnatal interventions affecting the DG eventually result in an altered number of granule neurons in the DG, ectopic location of neurons and changes in adult neurogenesis. Adverse events in early life provoke proinflammatory changes in hippocampal glia at cellular and molecular levels immediately after stress exposure. Later, the cellular changes may disappear, though alterations in gene expression pattern persist. Additional stressful events later in life contribute to manifestation of glial changes and behavioral deficits. Alterations in the maturation of neuronal and glial cells induced by early life stress are interdependent and influence the development of neural nets, thus predisposing the brain to the development of cognitive and psychiatric disorders.
Collapse
|
42
|
Enzymatic Degradation of Cortical Perineuronal Nets Reverses GABAergic Interneuron Maturation. Mol Neurobiol 2022; 59:2874-2893. [PMID: 35233718 PMCID: PMC9016038 DOI: 10.1007/s12035-022-02772-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 02/16/2022] [Indexed: 12/03/2022]
Abstract
Perineuronal nets (PNNs) are specialised extracellular matrix structures which preferentially enwrap fast-spiking (FS) parvalbumin interneurons and have diverse roles in the cortex. PNN maturation coincides with closure of the critical period of cortical plasticity. We have previously demonstrated that BDNF accelerates interneuron development in a c-Jun-NH2-terminal kinase (JNK)–dependent manner, which may involve upstream thousand-and-one amino acid kinase 2 (TAOK2). Chondroitinase-ABC (ChABC) enzymatic digestion of PNNs reportedly reactivates ‘juvenile-like’ plasticity in the adult CNS. However, the mechanisms involved are unclear. We show that ChABC produces an immature molecular phenotype in cultured cortical neurons, corresponding to the phenotype prior to critical period closure. ChABC produced different patterns of PNN-related, GABAergic and immediate early (IE) gene expression than well-characterised modulators of mature plasticity and network activity (GABAA-R antagonist, bicuculline, and sodium-channel blocker, tetrodotoxin (TTX)). ChABC downregulated JNK activity, while this was upregulated by bicuculline. Bicuculline, but not ChABC, upregulated Bdnf expression and ERK activity. Furthermore, we found that BDNF upregulation of semaphorin-3A and IE genes was TAOK mediated. Our data suggest that ChABC heightens structural flexibility and network disinhibition, potentially contributing to ‘juvenile-like’ plasticity. The molecular phenotype appears to be distinct from heightened mature synaptic plasticity and could relate to JNK signalling. Finally, we highlight that BDNF regulation of plasticity and PNNs involves TAOK signalling.
Collapse
|
43
|
Transit Amplifying Progenitors in the Cerebellum: Similarities to and Differences from Transit Amplifying Cells in Other Brain Regions and between Species. Cells 2022; 11:cells11040726. [PMID: 35203375 PMCID: PMC8870322 DOI: 10.3390/cells11040726] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 02/07/2022] [Accepted: 02/15/2022] [Indexed: 12/15/2022] Open
Abstract
Transit amplification of neural progenitors/precursors is widely used in the development of the central nervous system and for tissue homeostasis. In most cases, stem cells, which are relatively less proliferative, first differentiate into transit amplifying cells, which are more proliferative, losing their stemness. Subsequently, transit amplifying cells undergo a limited number of mitoses and differentiation to expand the progeny of differentiated cells. This step-by-step proliferation is considered an efficient system for increasing the number of differentiated cells while maintaining the stem cells. Recently, we reported that cerebellar granule cell progenitors also undergo transit amplification in mice. In this review, we summarize our and others’ recent findings and the prospective contribution of transit amplification to neural development and evolution, as well as the molecular mechanisms regulating transit amplification.
Collapse
|
44
|
Filova I, Bohuslavova R, Tavakoli M, Yamoah EN, Fritzsch B, Pavlinkova G. Early Deletion of Neurod1 Alters Neuronal Lineage Potential and Diminishes Neurogenesis in the Inner Ear. Front Cell Dev Biol 2022; 10:845461. [PMID: 35252209 PMCID: PMC8894106 DOI: 10.3389/fcell.2022.845461] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 01/25/2022] [Indexed: 11/13/2022] Open
Abstract
Neuronal development in the inner ear is initiated by expression of the proneural basic Helix-Loop-Helix (bHLH) transcription factor Neurogenin1 that specifies neuronal precursors in the otocyst. The initial specification of the neuroblasts within the otic epithelium is followed by the expression of an additional bHLH factor, Neurod1. Although NEUROD1 is essential for inner ear neuronal development, the different aspects of the temporal and spatial requirements of NEUROD1 for the inner ear and, mainly, for auditory neuron development are not fully understood. In this study, using Foxg1Cre for the early elimination of Neurod1 in the mouse otocyst, we showed that Neurod1 deletion results in a massive reduction of differentiating neurons in the otic ganglion at E10.5, and in the diminished vestibular and rudimental spiral ganglia at E13.5. Attenuated neuronal development was associated with reduced and disorganized sensory epithelia, formation of ectopic hair cells, and the shortened cochlea in the inner ear. Central projections of inner ear neurons with conditional Neurod1 deletion are reduced, unsegregated, disorganized, and interconnecting the vestibular and auditory systems. In line with decreased afferent input from auditory neurons, the volume of cochlear nuclei was reduced by 60% in Neurod1 mutant mice. Finally, our data demonstrate that early elimination of Neurod1 affects the neuronal lineage potential and alters the generation of inner ear neurons and cochlear afferents with a profound effect on the first auditory nuclei, the cochlear nuclei.
Collapse
Affiliation(s)
- Iva Filova
- Laboratory of Molecular Pathogenesis, Institute of Biotechnology CAS, Vestec, Czechia
| | - Romana Bohuslavova
- Laboratory of Molecular Pathogenesis, Institute of Biotechnology CAS, Vestec, Czechia
| | - Mitra Tavakoli
- Laboratory of Molecular Pathogenesis, Institute of Biotechnology CAS, Vestec, Czechia
| | - Ebenezer N. Yamoah
- Department of Physiology and Cell Biology, Institute for Neuroscience, University of Nevada, Reno, NV, United States
| | - Bernd Fritzsch
- Department of Biology, University of Iowa, Iowa City, IA, United States
| | - Gabriela Pavlinkova
- Laboratory of Molecular Pathogenesis, Institute of Biotechnology CAS, Vestec, Czechia
| |
Collapse
|
45
|
Magaletta ME, Lobo M, Kernfeld EM, Aliee H, Huey JD, Parsons TJ, Theis FJ, Maehr R. Integration of single-cell transcriptomes and chromatin landscapes reveals regulatory programs driving pharyngeal organ development. Nat Commun 2022; 13:457. [PMID: 35075189 PMCID: PMC8786836 DOI: 10.1038/s41467-022-28067-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 01/07/2022] [Indexed: 12/13/2022] Open
Abstract
Maldevelopment of the pharyngeal endoderm, an embryonic tissue critical for patterning of the pharyngeal region and ensuing organogenesis, ultimately contributes to several classes of human developmental syndromes and disorders. Such syndromes are characterized by a spectrum of phenotypes that currently cannot be fully explained by known mutations or genetic variants due to gaps in characterization of critical drivers of normal and dysfunctional development. Despite the disease-relevance of pharyngeal endoderm, we still lack a comprehensive and integrative view of the molecular basis and gene regulatory networks driving pharyngeal endoderm development. To close this gap, we apply transcriptomic and chromatin accessibility single-cell sequencing technologies to generate a multi-omic developmental resource spanning pharyngeal endoderm patterning to the emergence of organ-specific epithelia in the developing mouse embryo. We identify cell-type specific gene regulation, distill GRN models that define developing organ domains, and characterize the role of an immunodeficiency-associated forkhead box transcription factor.
Collapse
Affiliation(s)
- Margaret E Magaletta
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
- Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, MA, USA
| | - Macrina Lobo
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
- Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, MA, USA
| | - Eric M Kernfeld
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
- Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, MA, USA
| | - Hananeh Aliee
- Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany
| | - Jack D Huey
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
- Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, MA, USA
| | - Teagan J Parsons
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
- Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, MA, USA
| | - Fabian J Theis
- Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany
- Department of Mathematics, Technische Universität München, Munich, Germany
- School of Life Sciences Weihenstephan, Technische Universität München, Freising, Germany
| | - René Maehr
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA.
- Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
46
|
Bokulić E, Medenica T, Knezović V, Štajduhar A, Almahariq F, Baković M, Judaš M, Sedmak G. The Stereological Analysis and Spatial Distribution of Neurons in the Human Subthalamic Nucleus. Front Neuroanat 2022; 15:749390. [PMID: 34970124 PMCID: PMC8712451 DOI: 10.3389/fnana.2021.749390] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 11/17/2021] [Indexed: 11/13/2022] Open
Abstract
The subthalamic nucleus (STN) is a small, ovoid structure, and an important site of deep brain stimulation (DBS) for the treatment of Parkinson’s disease. Although the STN is a clinically important structure, there are many unresolved issues with regard to it. These issues are especially related to the anatomical subdivision, neuronal phenotype, neuronal composition, and spatial distribution. In this study, we have examined the expression pattern of 8 neuronal markers [nNOS, NeuN, parvalbumin (PV), calbindin (CB), calretinin (CR), FOXP2, NKX2.1, and PAX6] in the adult human STN. All of the examined markers, except CB, were present in the STN. To determine the neuronal density, we have performed stereological analysis on Nissl-stained and immunohistochemical slides of positive markers. The stereology data were also used to develop a three-dimensional map of the spatial distribution of neurons within the STN. The nNOS population exhibited the largest neuronal density. The estimated total number of nNOS STN neurons is 281,308 ± 38,967 (± 13.85%). The STN neuronal subpopulations can be divided into two groups: one with a neuronal density of approximately 3,300 neurons/mm3 and the other with a neuronal density of approximately 2,200 neurons/mm3. The largest density of STN neurons was observed along the ventromedial border of the STN and the density gradually decreased toward the dorsolateral border. In this study, we have demonstrated the presence of 7 neuronal markers in the STN, three of which were not previously described in the human STN. The human STN is a collection of diverse, intermixed neuronal subpopulations, and our data, as far as the cytoarchitectonics is concerned, did not support the tripartite STN subdivision.
Collapse
Affiliation(s)
- Ema Bokulić
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia.,Centre of Excellence for Basic, Clinical and Translational Neuroscience, Zagreb, Croatia
| | - Tila Medenica
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia.,Centre of Excellence for Basic, Clinical and Translational Neuroscience, Zagreb, Croatia
| | - Vinka Knezović
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia.,Centre of Excellence for Basic, Clinical and Translational Neuroscience, Zagreb, Croatia
| | - Andrija Štajduhar
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia.,Centre of Excellence for Basic, Clinical and Translational Neuroscience, Zagreb, Croatia.,School of Public Health "Andrija Štampar," University of Zagreb School of Medicine, Zagreb, Croatia
| | - Fadi Almahariq
- Centre of Excellence for Basic, Clinical and Translational Neuroscience, Zagreb, Croatia.,Department of Neurosurgery, Clinical Hospital "Dubrava," Zagreb, Croatia
| | - Marija Baković
- Department of Forensic Medicine, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Miloš Judaš
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia.,Centre of Excellence for Basic, Clinical and Translational Neuroscience, Zagreb, Croatia
| | - Goran Sedmak
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia.,Centre of Excellence for Basic, Clinical and Translational Neuroscience, Zagreb, Croatia
| |
Collapse
|
47
|
Aghanoori MR, Burns KM, Subha M, Williams L, Hua M, Nobakht F, Krawec T, Yang G. Immunohistochemical analysis of the developing mouse cortex. Methods Cell Biol 2022; 170:31-46. [DOI: 10.1016/bs.mcb.2022.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
48
|
Wang T, Liao JC, Wang X, Wang QS, Wan KY, Yang YY, He Q, Zhang JX, Chen G, Li W. Unexpected BrdU inhibition on astrocyte-to-neuron conversion. Neural Regen Res 2021; 17:1526-1534. [PMID: 34916438 PMCID: PMC8771121 DOI: 10.4103/1673-5374.325747] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
5-Bromo-2′-deoxyuridine (BrdU) is a halogenated pyrimidine that can be incorporated into newly synthesized DNA during the S phase of the cell cycle. BrdU is widely used in fate-mapping studies of embryonic and adult neurogenesis to identify newborn neurons, however side effects on neural stem cells and their progeny have been reported. In vivo astrocyte-to-neuron (AtN) conversion is a new approach for generating newborn neurons by directly converting endogenous astrocytes into neurons. The BrdU-labeling strategy has been used to trace astrocyte-converted neurons, but whether BrdU has any effect on the AtN conversion is unknown. Here, while conducting a NeuroD1-mediated AtN conversion study using BrdU to label dividing reactive astrocytes following ischemic injury, we accidentally discovered that BrdU inhibited AtN conversion. We initially found a gradual reduction in BrdU-labeled astrocytes during NeuroD1-mediated AtN conversion in the mouse cortex. Although most NeuroD1-infected astrocytes were converted into neurons, the number of BrdU-labeled neurons was surprisingly low. To exclude the possibility that this BrdU inhibition was caused by the ischemic injury, we conducted an in vitro AtN conversion study by overexpressing NeuroD1 in cultured cortical astrocytes in the presence or absence of BrdU. Surprisingly, we also found a significantly lower conversion rate and a smaller number of converted neurons in the BrdU-treated group compared with the untreated group. These results revealed an unexpected inhibitory effect of BrdU on AtN conversion, suggesting more caution is needed when using BrdU in AtN conversion studies and in data interpretation.
Collapse
Affiliation(s)
- Tao Wang
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong Province, China
| | - Jian-Cheng Liao
- Department of Neurosurgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China
| | - Xu Wang
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong Province, China
| | - Qing-Song Wang
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong Province, China
| | - Kai-Ying Wan
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong Province, China
| | - Yi-Yi Yang
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong Province, China
| | - Qing He
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong Province, China
| | - Jia-Xuan Zhang
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong Province, China
| | - Gong Chen
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong Province, China
| | - Wen Li
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong Province, China
| |
Collapse
|
49
|
Abstract
Spinal cord injury represents a devastating central nervous system injury that could impair the mobility and sensory function of afflicted patients. The hallmarks of spinal cord injury include neuroinflammation, axonal degeneration, neuronal loss, and reactive gliosis. Furthermore, the formation of a glial scar at the injury site elicits an inhibitory environment for potential neuroregeneration. Besides axonal regeneration, a significant challenge in treating spinal cord injury is to replenish the neurons lost during the pathological process. However, despite decades of research efforts, current strategies including stem cell transplantation have not resulted in a successful clinical therapy. Furthermore, stem cell transplantation faces serious hurdles such as immunorejection of the transplanted cells and ethical issues. In vivo neuronal reprogramming is a recently developed technology and leading a major breakthrough in regenerative medicine. This innovative technology converts endogenous glial cells into functional neurons for injury repair in the central nervous system. The feasibility of in vivo neuronal reprogramming has been demonstrated successfully in models of different neurological disorders including spinal cord injury by numerous laboratories. Several reprogramming factors, mainly the pro-neural transcription factors, have been utilized to reprogram endogenous glial cells into functional neurons with distinct phenotypes. So far, the literature on in vivo neuronal reprogramming in the model of spinal cord injury is still small. In this review, we summarize a limited number of such reports and discuss several questions that we think are important for applying in vivo neuronal reprogramming in the research field of spinal cord injury as well as other central nervous system disorders.
Collapse
Affiliation(s)
- Xuanyu Chen
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Hedong Li
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| |
Collapse
|
50
|
Direct neuronal reprogramming: Fast forward from new concepts toward therapeutic approaches. Neuron 2021; 110:366-393. [PMID: 34921778 DOI: 10.1016/j.neuron.2021.11.023] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/25/2021] [Accepted: 11/19/2021] [Indexed: 12/21/2022]
Abstract
Differentiated cells have long been considered fixed in their identity. However, about 20 years ago, the first direct conversion of glial cells into neurons in vitro opened the field of "direct neuronal reprogramming." Since then, neuronal reprogramming has achieved the generation of fully functional, mature neurons with remarkable efficiency, even in diseased brain environments. Beyond their clinical implications, these discoveries provided basic insights into crucial mechanisms underlying conversion of specific cell types into neurons and maintenance of neuronal identity. Here we discuss such principles, including the importance of the starter cell for shaping the outcome of neuronal reprogramming. We further highlight technical concerns for in vivo reprogramming and propose a code of conduct to avoid artifacts and pitfalls. We end by pointing out next challenges for development of less invasive cell replacement therapies for humans.
Collapse
|