1
|
Shobudani M, Sakamaki Y, Karasawa A, Ojiro R, Zou X, Tang Q, Ozawa S, Jin M, Yoshida T, Shibutani M. Metabolic shift as a compensatory response to impaired hippocampal neurogenesis after developmental exposure to sodium fluoride in rats. Acta Histochem 2024; 126:152204. [PMID: 39413662 DOI: 10.1016/j.acthis.2024.152204] [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: 04/25/2024] [Revised: 09/30/2024] [Accepted: 09/30/2024] [Indexed: 10/18/2024]
Abstract
Fluoride affects neurodevelopment in children. In this study, we examined the effects of developmental exposure to sodium fluoride (NaF) on hippocampal neurogenesis in rats. Dams were given drinking water containing NaF at 0 (untreated controls), 30 or 100 ppm from gestational day 6 to day 21 post-delivery upon weaning, and offspring were reared until postnatal day (PND) 77. On PND 21, NaF at 100 ppm altered the numbers in subpopulations of granule cell lineages, including a decrease in type-3 neural progenitor cells (NPCs), as well as a compensatory increase in type-1 neural stem cells (NSCs) and type-2a NPCs. NaF exposure tended to increase GluR2+ mossy cells in the hilus of the dentate gyrus (DG) in a dose-dependent manner, suggesting that NaF exposure induces a compensatory neurogenic response. NaF also caused a dose-dependent increase in ARC+ granule cells, and it upregulated Ptgs2 in the DG at 100 ppm, suggesting that NaF exposure increases synaptic plasticity in granule cells. NaF at 100 ppm upregulated granule cell lineage marker genes (Nes, Eomes and Rbfox3) and an anti-apoptotic gene (Bcl2), suggesting ameliorating responses against the impaired neurogenesis during NaF exposure. Moreover, NaF at 100 ppm downregulated oxidative phosphorylation-related genes (Atp5f1b and Sdhd) and upregulated a glycolysis-related gene (Hk3), suggesting a metabolic shift in cells undergoing neurogenesis. By PND 77, the changes in granule cell lineages were no longer detected, and GABAergic interneuron marker genes (Calb2 and Reln) were upregulated, suggesting a persistent protective response in granule cell lineages. Together, these findings suggest that developmental NaF exposure causes transient disruption of hippocampal neurogenesis, which in turn induces a metabolic shift as a compensatory response.
Collapse
Affiliation(s)
- Momoka Shobudani
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan.
| | - Yuri Sakamaki
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan.
| | - Ayumi Karasawa
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan.
| | - Ryota Ojiro
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan; Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan.
| | - Xinyu Zou
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan; Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan.
| | - Qian Tang
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan; Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan.
| | - Shunsuke Ozawa
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan; Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan.
| | - Meilan Jin
- Laboratory of Veterinary Pathology, College of Veterinary Medicine, Southwest University, No. 2 Tiansheng Road, BeiBei District, Chongqing 400715, PR China.
| | - Toshinori Yoshida
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan; Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan.
| | - Makoto Shibutani
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan; Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan.
| |
Collapse
|
2
|
LaNoce E, Zhang DY, Garcia-Epelboim A, Su Y, Sun Y, Alepa G, Angelucci AR, Akay-Espinoza C, Jordan-Sciutto KL, Song H, Ming GL, Christian KM. Exposure to the antiretroviral drug dolutegravir impairs structure and neurogenesis in a forebrain organoid model of human embryonic cortical development. Front Mol Neurosci 2024; 17:1459877. [PMID: 39569018 PMCID: PMC11576471 DOI: 10.3389/fnmol.2024.1459877] [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: 07/05/2024] [Accepted: 09/13/2024] [Indexed: 11/22/2024] Open
Abstract
Introduction For many therapeutic drugs, including antiretroviral drugs used to treat people living with HIV-1 (PLWH), we have little data on the potential effects on the developing human brain due to limited access to tissue and historical constraints on the inclusion of pregnant populations in clinical trials. Human induced pluripotent stem cells (iPSCs) offer a new avenue to gain insight on how drugs may impact human cell types representative of the developing central nervous system. To prevent vertical transmission of HIV and promote the health of pregnant PLWH, antiretroviral therapy must be initiated and/or maintained throughout pregnancy. However, many antiretroviral drugs are approved for widespread use following clinical testing only in non-pregnant populations and there may be limited information on potential teratogenicity until pregnancy outcomes are evaluated. The integrase strand transfer inhibitor dolutegravir (DTG) is a frontline antiretroviral drug that is effective in viral suppression of HIV but was previously reported to be associated with a slight increase in the risk for neural tube defects in one study, although this has not been replicated in other cohorts. Methods To directly investigate the potential impact of DTG on human cortical neurogenesis, we measured the effects of daily drug exposure on the early stages of corticogenesis in a human iPSC-based forebrain organoid model. We quantified organoid size and structure and analyzed gene and protein expression to evaluate the impact of several doses of DTG on organoid development. Results We observed deficits in organoid structure and impaired neurogenesis in DTG-treated organoids compared to vehicle-treated control organoids after 20 or 40 days in culture. Our highest dose of DTG (10 μM) resulted in significantly smaller organoids with a reduced density of neural rosette structures compared to vehicle-treated controls. Mechanistically, RNA-sequencing and immunohistological analysis suggests dysregulated amino acid transport and activation of the integrated stress response in the DTG-treated organoids, and functionally, a small molecule integrated stress response inhibitor (ISRIB) could partially rescue increased expression of proteins related to cell cycle regulation. Discussion Together, these results illustrate the potential for human iPSC-based strategies to reveal biological processes during neurogenesis that may be affected by therapeutic drugs and provide complementary data in relevant human cell types to augment preclinical investigations of drug safety during pregnancy.
Collapse
Affiliation(s)
- Emma LaNoce
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Daniel Y Zhang
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Alan Garcia-Epelboim
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Yijing Su
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Oral Medicine, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Yusha Sun
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Neuroscience Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Giana Alepa
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Angelina R Angelucci
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Cagla Akay-Espinoza
- Department of Oral Medicine, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Kelly L Jordan-Sciutto
- Department of Oral Medicine, School of Dental 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 Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Institute for Regenerative 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
- The Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Guo-Li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Institute for Regenerative 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
| | - Kimberly M Christian
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
3
|
Souza LRQ, Pedrosa CGDS, Puig-Pijuan T, da Silva Dos Santos C, Vitória G, Delou JMA, Setti-Perdigão P, Higa LM, Tanuri A, Rehen SK, Guimarães MZP. Saxitoxin potentiates human neuronal cell death induced by Zika virus while sparing neural progenitors and astrocytes. Sci Rep 2024; 14:22809. [PMID: 39354036 PMCID: PMC11445263 DOI: 10.1038/s41598-024-73873-z] [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: 06/21/2024] [Accepted: 09/23/2024] [Indexed: 10/04/2024] Open
Abstract
The Zika virus (ZIKV) epidemic declared in Brazil between 2015 and 2016 was associated with an increased prevalence of severe congenital malformations, including microcephaly. The distribution of microcephaly cases was not uniform across the country, with a disproportionately higher incidence in the Northeast region (NE). Our previous work demonstrated that saxitoxin (STX), a toxin present in the drinking water reservoirs of the NE, exacerbated the damaging effects of ZIKV on the developing brain. We hypothesized that the impact of STX might vary among different neural cell types. While ZIKV infection caused severe damages on astrocytes and neural stem cells (NSCs), the addition of STX did not exacerbate these effects. We observed that neurons subjected to STX exposure were more prone to apoptosis and displayed higher ZIKV infection rate. These findings suggest that STX exacerbates the harmful effects of ZIKV on neurons, thereby providing a plausible explanation for the heightened severity of ZIKV-induced congenital malformations observed in Brazil's NE. This study highlights the importance of understanding the interactive effects of environmental toxins and infectious pathogens on neural development, with potential implications for public health policies.
Collapse
Affiliation(s)
- Leticia R Q Souza
- Instituto D'Or de Pesquisa e Ensino, Rua Diniz Cordeiro, 30, Rio de Janeiro, CEP 22281-100, Brazil
| | - Carolina G da S Pedrosa
- Instituto D'Or de Pesquisa e Ensino, Rua Diniz Cordeiro, 30, Rio de Janeiro, CEP 22281-100, Brazil
| | - Teresa Puig-Pijuan
- Instituto D'Or de Pesquisa e Ensino, Rua Diniz Cordeiro, 30, Rio de Janeiro, CEP 22281-100, Brazil
| | | | - Gabriela Vitória
- Instituto D'Or de Pesquisa e Ensino, Rua Diniz Cordeiro, 30, Rio de Janeiro, CEP 22281-100, Brazil
| | - João M A Delou
- Instituto D'Or de Pesquisa e Ensino, Rua Diniz Cordeiro, 30, Rio de Janeiro, CEP 22281-100, Brazil
| | - Pedro Setti-Perdigão
- Instituto D'Or de Pesquisa e Ensino, Rua Diniz Cordeiro, 30, Rio de Janeiro, CEP 22281-100, Brazil
| | - Luiza M Higa
- Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Amilcar Tanuri
- Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Stevens Kastrup Rehen
- Instituto D'Or de Pesquisa e Ensino, Rua Diniz Cordeiro, 30, Rio de Janeiro, CEP 22281-100, Brazil
- Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Marília Zaluar P Guimarães
- Instituto D'Or de Pesquisa e Ensino, Rua Diniz Cordeiro, 30, Rio de Janeiro, CEP 22281-100, Brazil.
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil.
| |
Collapse
|
4
|
Breton TS, Fike S, Francis M, Patnaude M, Murray CA, DiMaggio MA. Characterizing the SREB G protein-coupled receptor family in fish: Brain gene expression and genomic differences in upstream transcription factor binding sites. Comp Biochem Physiol A Mol Integr Physiol 2023; 285:111507. [PMID: 37611891 PMCID: PMC10529039 DOI: 10.1016/j.cbpa.2023.111507] [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: 06/07/2023] [Revised: 07/12/2023] [Accepted: 08/20/2023] [Indexed: 08/25/2023]
Abstract
The SREB (Super-conserved Receptors Expressed in Brain) family of orphan G protein-coupled receptors is highly conserved in vertebrates and consists of three members: SREB1 (orphan designation GPR27), SREB2 (GPR85), and SREB3 (GPR173). SREBs are associated with processes ranging from neuronal plasticity to reproductive control. Relatively little is known about similarities across the entire family, or how mammalian gene expression patterns compare to non-mammalian vertebrates. In fish, this system may be particularly complex, as some species have gained a fourth member (SREB3B) while others have lost genes. To better understand the system, the present study aimed to: 1) use qPCR to characterize sreb and related gene expression patterns in the brains of three fish species with different systems, and 2) identify possible differences in transcriptional regulation among the receptors, using upstream transcription factor binding sites across 70 ray-finned fish genomes. Overall, regional patterns of sreb expression were abundant in forebrain-related areas. However, some species-specific patterns were detected, such as abundant expression of receptors in zebrafish (Danio rerio) hypothalamic-containing sections, and divergence between sreb3a and sreb3b in pufferfish (Dichotomyctere nigroviridis). In addition, a gene possibly related to the system (dkk3a) was spatially correlated with the receptors in all three species. Genomic regions upstream of sreb2 and sreb3b, but largely not sreb1 or sreb3a, contained many highly conserved transcription factor binding sites. These results provide novel information about expression differences and transcriptional regulation across fish that may inform future research to better understand these receptors.
Collapse
Affiliation(s)
- Timothy S Breton
- Division of Natural Sciences, University of Maine at Farmington, Farmington, ME 04938, USA.
| | - Samantha Fike
- Division of Natural Sciences, University of Maine at Farmington, Farmington, ME 04938, USA
| | - Mullein Francis
- Division of Natural Sciences, University of Maine at Farmington, Farmington, ME 04938, USA
| | - Michael Patnaude
- Division of Natural Sciences, University of Maine at Farmington, Farmington, ME 04938, USA
| | - Casey A Murray
- Tropical Aquaculture Laboratory, Program in Fisheries and Aquatic Sciences, School of Forest, Fisheries, and Geomatics Sciences, Institute of Food and Agricultural Sciences, University of Florida, Ruskin, FL 33570, USA
| | - Matthew A DiMaggio
- Tropical Aquaculture Laboratory, Program in Fisheries and Aquatic Sciences, School of Forest, Fisheries, and Geomatics Sciences, Institute of Food and Agricultural Sciences, University of Florida, Ruskin, FL 33570, USA
| |
Collapse
|
5
|
Gil N, Perry RBT, Mukamel Z, Tuck A, Bühler M, Ulitsky I. Complex regulation of Eomes levels mediated through distinct functional features of the Meteor long non-coding RNA locus. Cell Rep 2023; 42:112569. [PMID: 37256750 PMCID: PMC10320833 DOI: 10.1016/j.celrep.2023.112569] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 03/07/2023] [Accepted: 05/12/2023] [Indexed: 06/02/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) are implicated in a plethora of cellular processes, but an in-depth understanding of their functional features or their mechanisms of action is currently lacking. Here we study Meteor, a lncRNA transcribed near the gene encoding EOMES, a pleiotropic transcription factor implicated in various processes throughout development and in adult tissues. Using a wide array of perturbation techniques, we show that transcription elongation through the Meteor locus is required for Eomes activation in mouse embryonic stem cells, with Meteor repression linked to a change in the subpopulation primed to differentiate to the mesoderm lineage. We further demonstrate that a distinct functional feature of the locus-namely, the underlying DNA element-is required for suppressing Eomes expression following neuronal differentiation. Our results demonstrate the complex regulation that can be conferred by a single locus and emphasize the importance of careful selection of perturbation techniques when studying lncRNA loci.
Collapse
Affiliation(s)
- Noa Gil
- Department of Immunology and Regenerative Biology and Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Rotem Ben-Tov Perry
- Department of Immunology and Regenerative Biology and Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Zohar Mukamel
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Alex Tuck
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland
| | - Marc Bühler
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland; University of Basel, Basel, Switzerland
| | - Igor Ulitsky
- Department of Immunology and Regenerative Biology and Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
6
|
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
|
7
|
Andrographolide inhibits murine embryonic neuronal development through PFKFB3-mediated glycolytic pathway. Eur J Pharmacol 2023; 940:175474. [PMID: 36549500 DOI: 10.1016/j.ejphar.2022.175474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 12/15/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022]
Abstract
Dysregulation of neuronal development may cause neurodevelopmental disorders. However, how to regulate embryonic neuronal development and whether this regulation can be medical interrupted are largely unknown. This study aimed to investigate whether and how andrographolide (ANP) regulates embryonic neuronal development. The pregnant mice at embryonic day 10.5 (E10.5) were administrated with ANP, and the embryonic brains were harvested at E17.5 or E18.5. Immunofluorescence (IF), Immunohistochemistry (IHC) performed to determine whether ANP is critical in regulating neuronal development. Real-time quantitative PCR, western blotting, cell counting kit-8 assay, Flow Cytometry assay, Boyden Chamber Migration assay carried out to evaluate whether ANP regulates neuronal proliferation and migration. Protein-protein interaction, CO-immunoprecipitation and IF staining carried out to evaluate whether ANP regulates the interaction between PFKFB3, NeuN and TBR1. Knockdown or overexpression of PFKFB3 by adenovirus infection were used to determine whether ANP inhibits neuronal development through PFKFB3 mediated glycolytic pathway. Our data indicated that ANP inhibited the maturation of embryonic neurons characterized by suppressing neuronal proliferation and migration. ANP regulated the interaction between PFKFB3, NeuN, and TBR1. Knockdown of PFKFB3 aggravated ANP mediated inhibition of neuronal proliferation and migration, while overexpression of PFKFB3 attenuated ANP mediated neuronal developmental suppression. In summary, ANP suppressed the expression of PFKFB3, and interrupted the interaction between TRB1 and NeuN, resulting in suppressing neuronal proliferation, migration and maturation and eventually inhibiting murine embryonic neuronal development.
Collapse
|
8
|
Protective Effects of Early Caffeine Administration in Hyperoxia-Induced Neurotoxicity in the Juvenile Rat. Antioxidants (Basel) 2023; 12:antiox12020295. [PMID: 36829854 PMCID: PMC9952771 DOI: 10.3390/antiox12020295] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [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
|
9
|
Powers RM, Hevner RF, Halpain S. The Neuron Navigators: Structure, function, and evolutionary history. Front Mol Neurosci 2023; 15:1099554. [PMID: 36710926 PMCID: PMC9877351 DOI: 10.3389/fnmol.2022.1099554] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 12/19/2022] [Indexed: 01/13/2023] Open
Abstract
Neuron navigators (Navigators) are cytoskeletal-associated proteins important for neuron migration, neurite growth, and axon guidance, but they also function more widely in other tissues. Recent studies have revealed novel cellular functions of Navigators such as macropinocytosis, and have implicated Navigators in human disorders of axon growth. Navigators are present in most or all bilaterian animals: vertebrates have three Navigators (NAV1-3), Drosophila has one (Sickie), and Caenorhabditis elegans has one (Unc-53). Structurally, Navigators have conserved N- and C-terminal regions each containing specific domains. The N-terminal region contains a calponin homology (CH) domain and one or more SxIP motifs, thought to interact with the actin cytoskeleton and mediate localization to microtubule plus-end binding proteins, respectively. The C-terminal region contains two coiled-coil domains, followed by a AAA+ family nucleoside triphosphatase domain of unknown activity. The Navigators appear to have evolved by fusion of N- and C-terminal region homologs present in simpler organisms. Overall, Navigators participate in the cytoskeletal response to extracellular cues via microtubules and actin filaments, in conjunction with membrane trafficking. We propose that uptake of fluid-phase cues and nutrients and/or downregulation of cell surface receptors could represent general mechanisms that explain Navigator functions. Future studies developing new models, such as conditional knockout mice or human cerebral organoids may reveal new insights into Navigator function. Importantly, further biochemical studies are needed to define the activities of the Navigator AAA+ domain, and to study potential interactions among different Navigators and their binding partners.
Collapse
Affiliation(s)
- Regina M. Powers
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA, United States,Sanford Consortium for Regenerative Medicine, La Jolla, CA, United States
| | - Robert F. Hevner
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, United States,Department of Pathology, UC San Diego School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Shelley Halpain
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA, United States,Sanford Consortium for Regenerative Medicine, La Jolla, CA, United States,*Correspondence: Shelley Halpain, ✉
| |
Collapse
|
10
|
Crespo I, Pignatelli J, Kinare V, Méndez-Gómez HR, Esgleas M, Román MJ, Canals JM, Tole S, Vicario C. Tbr1 Misexpression Alters Neuronal Development in the Cerebral Cortex. Mol Neurobiol 2022; 59:5750-5765. [PMID: 35781633 PMCID: PMC9395452 DOI: 10.1007/s12035-022-02936-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 06/10/2022] [Indexed: 11/26/2022]
Abstract
Changes in the transcription factor (TF) expression are critical for brain development, and they may also underlie neurodevelopmental disorders. Indeed, T-box brain1 (Tbr1) is a TF crucial for the formation of neocortical layer VI, and mutations and microdeletions in that gene are associated with malformations in the human cerebral cortex, alterations that accompany autism spectrum disorder (ASD). Interestingly, Tbr1 upregulation has also been related to the occurrence of ASD-like symptoms, although limited studies have addressed the effect of increased Tbr1 levels during neocortical development. Here, we analysed the impact of Tbr1 misexpression in mouse neural progenitor cells (NPCs) at embryonic day 14.5 (E14.5), when they mainly generate neuronal layers II-IV. By E18.5, cells accumulated in the intermediate zone and in the deep cortical layers, whereas they became less abundant in the upper cortical layers. In accordance with this, the proportion of Sox5+ cells in layers V-VI increased, while that of Cux1+ cells in layers II-IV decreased. On postnatal day 7, fewer defects in migration were evident, although a higher proportion of Sox5+ cells were seen in the upper and deep layers. The abnormal neuronal migration could be partially due to the altered multipolar-bipolar neuron morphologies induced by Tbr1 misexpression, which also reduced dendrite growth and branching, and disrupted the corpus callosum. Our results indicate that Tbr1 misexpression in cortical NPCs delays or disrupts neuronal migration, neuronal specification, dendrite development and the formation of the callosal tract. Hence, genetic changes that provoke ectopic Tbr1 upregulation during development could provoke cortical brain malformations.
Collapse
Affiliation(s)
- Inmaculada Crespo
- Instituto Cajal-Consejo Superior de Investigaciones Científicas (CSIC), Avenida Doctor Arce 37, 28002, Madrid, Spain
- CIBERNED-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- CES Cardenal Cisneros, Madrid, Spain
| | - Jaime Pignatelli
- Instituto Cajal-Consejo Superior de Investigaciones Científicas (CSIC), Avenida Doctor Arce 37, 28002, Madrid, Spain
- CIBERNED-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Veena Kinare
- Department of Life Sciences, Sophia College for Women, Mumbai, 400026, India
| | - Héctor R Méndez-Gómez
- Instituto Cajal-Consejo Superior de Investigaciones Científicas (CSIC), Avenida Doctor Arce 37, 28002, Madrid, Spain
- CIBERNED-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Miriam Esgleas
- CIBERNED-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Laboratory of Stem Cells and Regenerative Medicine, Department of Biomedical Sciences, Creatio, Production and Validation Center of Advanced Therapies, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- August Pi I Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - María José Román
- Instituto Cajal-Consejo Superior de Investigaciones Científicas (CSIC), Avenida Doctor Arce 37, 28002, Madrid, Spain
- CIBERNED-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Josep M Canals
- CIBERNED-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Laboratory of Stem Cells and Regenerative Medicine, Department of Biomedical Sciences, Creatio, Production and Validation Center of Advanced Therapies, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- August Pi I Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Shubha Tole
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, 400005, India
| | - Carlos Vicario
- Instituto Cajal-Consejo Superior de Investigaciones Científicas (CSIC), Avenida Doctor Arce 37, 28002, Madrid, Spain.
- CIBERNED-Instituto de Salud Carlos III (ISCIII), Madrid, Spain.
| |
Collapse
|
11
|
Vrba L, Futscher BW, Oshiro M, Watts GS, Menashi E, Hu C, Hammad H, Pennington DR, Golconda U, Gavini H, Roe DJ, Shroff RT, Nelson MA. Liquid biopsy, using a novel DNA methylation signature, distinguishes pancreatic adenocarcinoma from benign pancreatic disease. Clin Epigenetics 2022; 14:28. [PMID: 35193708 PMCID: PMC8864826 DOI: 10.1186/s13148-022-01246-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 02/09/2022] [Indexed: 12/17/2022] Open
Abstract
We tested the ability of a novel DNA methylation biomarker set to distinguish metastatic pancreatic cancer cases from benign pancreatic cyst patients and to monitor tumor dynamics using quantitative DNA methylation analysis of cell-free DNA (cfDNA) from blood samples. The biomarkers were able to distinguish malignant cases from benign disease with high sensitivity and specificity (AUC = 0.999). Furthermore, the biomarkers detected a consistent decline in tumor-derived cfDNA in samples from patients undergoing chemotherapy. The study indicates that our liquid biopsy assay could be useful for management of pancreatic cancer patients.
Collapse
Affiliation(s)
- Lukas Vrba
- The University of Arizona Cancer Center, Tucson, AZ, USA.,Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, USA
| | - Bernard W Futscher
- The University of Arizona Cancer Center, Tucson, AZ, USA.,Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, USA.,Precision Epigenomics Inc, Tucson, AZ, USA
| | - Marc Oshiro
- The University of Arizona Cancer Center, Tucson, AZ, USA
| | - George S Watts
- The University of Arizona Cancer Center, Tucson, AZ, USA.,Department of Medical Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | | | - Charles Hu
- Dignity Health Chandler Regional Medical Center, Chandler, AZ, USA
| | - Hytham Hammad
- The University of Arizona Cancer Center, Tucson, AZ, USA.,Division of Hematology/Oncology, Department of Medicine, University of Arizona Caner Center, Tucson, AZ, USA
| | - Daniel R Pennington
- The University of Arizona Cancer Center, Tucson, AZ, USA.,Division of Hematology/Oncology, Department of Medicine, University of Arizona Caner Center, Tucson, AZ, USA
| | | | - Hemanth Gavini
- Division of Hematology/Oncology, Department of Medicine, University of Arizona Caner Center, Tucson, AZ, USA
| | - Denise J Roe
- The University of Arizona Cancer Center, Tucson, AZ, USA
| | - Rachna T Shroff
- The University of Arizona Cancer Center, Tucson, AZ, USA.,Division of Hematology/Oncology, Department of Medicine, University of Arizona Caner Center, Tucson, AZ, USA
| | - Mark A Nelson
- The University of Arizona Cancer Center, Tucson, AZ, USA. .,Department of Pathology, College of Medicine, University of Arizona, Tucson, AZ, USA. .,Precision Epigenomics Inc, Tucson, AZ, USA.
| |
Collapse
|
12
|
Shin BY, Lee SH, Kim Y, An J, Park TY, Lee SK. Interactomic inhibition of Eomes in the nucleus alleviates EAE via blocking the conversion of Th17 cells into non-classic Th1 cells. Immunol Med 2022; 45:119-127. [PMID: 35130134 DOI: 10.1080/25785826.2022.2031812] [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] [Indexed: 10/19/2022] Open
Abstract
Th17 cells are implicated in the pathogenesis of several autoimmune diseases. During the inflammation, Th17 cells exposed to IL-12 can shift towards the Th1 phenotype. These shifted cells are defined as 'non-classic Th1 cells'. Th17-derived non-classic Th1 cells play a critical role in late-onset chronic inflammatory diseases and are more pathogenic than the unshifted Th17 cells. Eomes is a transcription factor highly expressed in non-classic Th1 cells. To study the functional role of Eomes without genetic alteration, novel recombinant protein, ntEomes-TMD, was generated by fusing TMD of Eomes and Hph-1-PTD that facilitate intracellular delivery of its cargo molecule. ntEomes-TMD was delivered into the nucleus of the cells without influencing the T cell activation and cytotoxicity. ntEomes-TMD specifically inhibited the Eomes- and ROR-γt-mediated transcription and suppressed the Th1 and Th17 differentiation. Interestingly, ntEomes-TMD blocked the generation of non-classic Th1 cells from Th17 cells, leading to the inhibition of IFN-γ and GM-CSF secretion. In EAE, ntEomes-TMD alleviated the symptoms of EAE, and the combination treatment using ntEomes-TMD and anti-IL-17 mAb together showed better therapeutic efficacy than anti-IL-17 mAb treatment. The results suggest that ntEomes-TMD can be a new therapeutic reagent for treating chronic inflammatory diseases associated with non-classic Th1 cells.
Collapse
Affiliation(s)
- Bo-Young Shin
- Department of Biotechnology, Yonsei University College of Life Science and Biotechnology, Seoul, Republic of Korea
| | - Su-Hyeon Lee
- Department of Biotechnology, Yonsei University College of Life Science and Biotechnology, Seoul, Republic of Korea
| | - Yuna Kim
- Department of Biotechnology, Yonsei University College of Life Science and Biotechnology, Seoul, Republic of Korea
| | - Jaekyeung An
- Department of Biotechnology, Yonsei University College of Life Science and Biotechnology, Seoul, Republic of Korea
| | - Tae-Yoon Park
- Molecular Neurobiology Laboratory, Department of Psychiatry and McLean Hospital, Harvard Medical School, Belmont, MA, USA
| | - Sang-Kyou Lee
- Department of Biotechnology, Yonsei University College of Life Science and Biotechnology, Seoul, Republic of Korea.,Good T cells, Inc, Seoul, Republic of Korea
| |
Collapse
|
13
|
Ito A, Imamura F. Expression of Maf family proteins in glutamatergic neurons of the mouse olfactory bulb. Dev Neurobiol 2021; 82:77-87. [PMID: 34679244 DOI: 10.1002/dneu.22859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 10/11/2021] [Accepted: 10/14/2021] [Indexed: 11/09/2022]
Abstract
The fate of neurons in the developing brain is largely determined by the combination of transcription factors they express. In particular, stem cells must follow different transcriptional cascades during differentiation in order to generate neurons with different neurotransmitter properties, such as glutamatergic and GABAergic neurons. In the mouse cerebral cortex, it has been shown that large Maf family proteins, MafA, MafB and c-Maf, regulate the development of specific types of GABAergic interneurons but are not expressed in glutamatergic neurons. In this study, we examined the expression of large Maf family proteins in the developing mouse olfactory bulb (OB) by immunohistochemistry and found that the cell populations expressing MafA and MafB are almost identical, and most of them express Tbr2. As Tbr2 is expressed in glutamatergic neurons in the OB, we further examined the expression of glutamatergic and GABAergic neuronal markers in MafA and MafB positive cells. The results showed that in the OB, MafA and MafB are expressed exclusively in glutamatergic neurons, but not in GABAergic neurons. We also found that few cells express c-Maf in the OB. These results indicate that, unlike the cerebral cortex, MafA and/or MafB may regulate the development of glutamatergic neurons in the developing OB. This study advances our knowledge about the development of glutamatergic neurons in the olfactory bulb, and also might suggest that mechanisms for the generation of projection neurons and interneurons differ between the cortex and the olfactory bulb, even though they both develop from the telencephalon.
Collapse
Affiliation(s)
- Ayako Ito
- Department of Pharmacology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Fumiaki Imamura
- Department of Pharmacology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| |
Collapse
|
14
|
Matho KS, Huilgol D, Galbavy W, He M, Kim G, An X, Lu J, Wu P, Di Bella DJ, Shetty AS, Palaniswamy R, Hatfield J, Raudales R, Narasimhan A, Gamache E, Levine JM, Tucciarone J, Szelenyi E, Harris JA, Mitra PP, Osten P, Arlotta P, Huang ZJ. Genetic dissection of the glutamatergic neuron system in cerebral cortex. Nature 2021; 598:182-187. [PMID: 34616069 PMCID: PMC8494647 DOI: 10.1038/s41586-021-03955-9] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 08/25/2021] [Indexed: 11/09/2022]
Abstract
Diverse types of glutamatergic pyramidal neurons mediate the myriad processing streams and output channels of the cerebral cortex1,2, yet all derive from neural progenitors of the embryonic dorsal telencephalon3,4. Here we establish genetic strategies and tools for dissecting and fate-mapping subpopulations of pyramidal neurons on the basis of their developmental and molecular programs. We leverage key transcription factors and effector genes to systematically target temporal patterning programs in progenitors and differentiation programs in postmitotic neurons. We generated over a dozen temporally inducible mouse Cre and Flp knock-in driver lines to enable the combinatorial targeting of major progenitor types and projection classes. Combinatorial strategies confer viral access to subsets of pyramidal neurons defined by developmental origin, marker expression, anatomical location and projection targets. These strategies establish an experimental framework for understanding the hierarchical organization and developmental trajectory of subpopulations of pyramidal neurons that assemble cortical processing networks and output channels.
Collapse
Affiliation(s)
- Katherine S Matho
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
| | - Dhananjay Huilgol
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
- Department of Neurobiology, Duke University Medical Center, Durham, NC, USA
| | - William Galbavy
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
- Program in Neuroscience, Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, USA
| | - Miao He
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Gukhan Kim
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
| | - Xu An
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
- Department of Neurobiology, Duke University Medical Center, Durham, NC, USA
| | - Jiangteng Lu
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
- Shanghai Jiaotong University Medical School, Shanghai, China
| | - Priscilla Wu
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
| | - Daniela J Di Bella
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Ashwin S Shetty
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | | | - Joshua Hatfield
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
- Department of Neurobiology, Duke University Medical Center, Durham, NC, USA
| | - Ricardo Raudales
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
- Program in Neuroscience, Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, USA
| | - Arun Narasimhan
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
| | - Eric Gamache
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
| | - Jesse M Levine
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
- Program in Neuroscience and Medical Scientist Training Program, Stony Brook University, New York, NY, USA
| | - Jason Tucciarone
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
- Program in Neuroscience and Medical Scientist Training Program, Stony Brook University, New York, NY, USA
- Department of Psychiatry, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Eric Szelenyi
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
| | - Julie A Harris
- Program in Neuroscience and Medical Scientist Training Program, Stony Brook University, New York, NY, USA
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Partha P Mitra
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
| | - Pavel Osten
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
| | - Paola Arlotta
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Z Josh Huang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA.
- Department of Neurobiology, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
15
|
Birkhoff JC, Brouwer RWW, Kolovos P, Korporaal AL, Bermejo-Santos A, Boltsis I, Nowosad K, van den Hout MCGN, Grosveld FG, van IJcken WFJ, Huylebroeck D, Conidi A. Targeted chromatin conformation analysis identifies novel distal neural enhancers of ZEB2 in pluripotent stem cell differentiation. Hum Mol Genet 2021; 29:2535-2550. [PMID: 32628253 PMCID: PMC7471508 DOI: 10.1093/hmg/ddaa141] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 06/29/2020] [Accepted: 06/30/2020] [Indexed: 12/25/2022] Open
Abstract
The transcription factor zinc finger E-box binding protein 2 (ZEB2) controls embryonic and adult cell fate decisions and cellular maturation in many stem/progenitor cell types. Defects in these processes in specific cell types underlie several aspects of Mowat–Wilson syndrome (MOWS), which is caused by ZEB2 haplo-insufficiency. Human ZEB2, like mouse Zeb2, is located on chromosome 2 downstream of a ±3.5 Mb-long gene-desert, lacking any protein-coding gene. Using temporal targeted chromatin capture (T2C), we show major chromatin structural changes based on mapping in-cis proximities between the ZEB2 promoter and this gene desert during neural differentiation of human-induced pluripotent stem cells, including at early neuroprogenitor cell (NPC)/rosette state, where ZEB2 mRNA levels increase significantly. Combining T2C with histone-3 acetylation mapping, we identified three novel candidate enhancers about 500 kb upstream of the ZEB2 transcription start site. Functional luciferase-based assays in heterologous cells and NPCs reveal co-operation between these three enhancers. This study is the first to document in-cis Regulatory Elements located in ZEB2’s gene desert. The results further show the usability of T2C for future studies of ZEB2 REs in differentiation and maturation of multiple cell types and the molecular characterization of newly identified MOWS patients that lack mutations in ZEB2 protein-coding exons.
Collapse
Affiliation(s)
- Judith C Birkhoff
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam, CN 3015, The Netherlands
| | - Rutger W W Brouwer
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam, CN 3015, The Netherlands.,Center for Biomics, Erasmus University Medical Center, Rotterdam, CN 3015, The Netherlands
| | - Petros Kolovos
- Department of Molecular Biology and Genetics, Democritus University of Thrace, Alexandroupolis 68100, Greece
| | - Anne L Korporaal
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam, CN 3015, The Netherlands
| | - Ana Bermejo-Santos
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam, CN 3015, The Netherlands
| | - Ilias Boltsis
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam, CN 3015, The Netherlands
| | - Karol Nowosad
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam, CN 3015, The Netherlands.,Department of Biochemistry and Molecular Biology, Medical University of Lublin, Lublin 20-093, Poland
| | - Mirjam C G N van den Hout
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam, CN 3015, The Netherlands.,Center for Biomics, Erasmus University Medical Center, Rotterdam, CN 3015, The Netherlands
| | - Frank G Grosveld
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam, CN 3015, The Netherlands
| | - Wilfred F J van IJcken
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam, CN 3015, The Netherlands.,Center for Biomics, Erasmus University Medical Center, Rotterdam, CN 3015, The Netherlands
| | - Danny Huylebroeck
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam, CN 3015, The Netherlands.,Department of Development and Regeneration, KU Leuven, Leuven B-3000, Belgium
| | - Andrea Conidi
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam, CN 3015, The Netherlands
| |
Collapse
|
16
|
Wang H, Zhou C, Hou M, Huang H, Sun Y. Neurogenesis Potential Evaluation and Transcriptome Analysis of Fetal Hypothalamic Neural Stem/Progenitor Cells With Prenatal High Estradiol Exposure. Front Genet 2021; 12:677935. [PMID: 34239542 PMCID: PMC8258253 DOI: 10.3389/fgene.2021.677935] [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: 03/08/2021] [Accepted: 05/10/2021] [Indexed: 11/13/2022] Open
Abstract
High maternal estradiol is reported to induce metabolic disorders by modulating hypothalamic gene expression in offspring. Since neurogenesis plays a crucial role during hypothalamus development, we explored whether prenatal high estradiol exposure (HE) affects proliferation and differentiation of fetal hypothalamic neural stem/progenitor cells (NSC/NPCs) in mice and performed RNA sequencing to identify the critical genes involved. NSC/NPCs in HE mice presented attenuated cell proliferation but increased neuronal differentiation in vitro compared with control (NC) cells. Gene set enrichment analysis of mRNA profiles indicated that genes downregulated in HE NSC/NPCs were enriched in neurogenesis-related Gene Ontology (GO) terms, while genes upregulated in HE NSC/NPCs were enriched in response to estradiol. Protein-protein interaction analysis of genes with core enrichment in GO terms of neurogenesis and response to estradiol identified 10 Hub mRNAs, among which three were potentially correlated with six differentially expressed (DE) lncRNAs based on lncRNA profiling and co-expression analysis. These findings offer important insights into developmental modifications in hypothalamic NSC/NPCs and may provide new clues for further investigation on maternal environment programmed neural development disorders.
Collapse
Affiliation(s)
- Huihui Wang
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China.,Animal Laboratory, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chengliang Zhou
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Min Hou
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Hefeng Huang
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China.,Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Yun Sun
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China.,Animal Laboratory, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
17
|
Herrero-Navarro Á, Puche-Aroca L, Moreno-Juan V, Sempere-Ferràndez A, Espinosa A, Susín R, Torres-Masjoan L, Leyva-Díaz E, Karow M, Figueres-Oñate M, López-Mascaraque L, López-Atalaya JP, Berninger B, López-Bendito G. Astrocytes and neurons share region-specific transcriptional signatures that confer regional identity to neuronal reprogramming. SCIENCE ADVANCES 2021; 7:7/15/eabe8978. [PMID: 33827819 PMCID: PMC8026135 DOI: 10.1126/sciadv.abe8978] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 02/18/2021] [Indexed: 05/15/2023]
Abstract
Neural cell diversity is essential to endow distinct brain regions with specific functions. During development, progenitors within these regions are characterized by specific gene expression programs, contributing to the generation of diversity in postmitotic neurons and astrocytes. While the region-specific molecular diversity of neurons and astrocytes is increasingly understood, whether these cells share region-specific programs remains unknown. Here, we show that in the neocortex and thalamus, neurons and astrocytes express shared region-specific transcriptional and epigenetic signatures. These signatures not only distinguish cells across these two brain regions but are also detected across substructures within regions, such as distinct thalamic nuclei, where clonal analysis reveals the existence of common nucleus-specific progenitors for neurons and astrocytes. Consistent with their shared molecular signature, regional specificity is maintained following astrocyte-to-neuron reprogramming. A detailed understanding of these regional-specific signatures may thus inform strategies for future cell-based brain repair.
Collapse
Affiliation(s)
- Álvaro Herrero-Navarro
- Instituto de Neurociencias, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), Sant Joan d'Alacant, Spain
| | - Lorenzo Puche-Aroca
- Instituto de Neurociencias, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), Sant Joan d'Alacant, Spain
| | - Verónica Moreno-Juan
- Instituto de Neurociencias, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), Sant Joan d'Alacant, Spain
| | - Alejandro Sempere-Ferràndez
- Instituto de Neurociencias, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), Sant Joan d'Alacant, Spain
| | - Ana Espinosa
- Instituto de Neurociencias, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), Sant Joan d'Alacant, Spain
| | - Rafael Susín
- Instituto de Neurociencias, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), Sant Joan d'Alacant, Spain
| | - Laia Torres-Masjoan
- Institute of Psychiatry, Psychology, and Neuroscience, Centre for Developmental Neurobiology, and MRC Centre for Neurodevelopmental Disorders, King's College London, SE1 1UL London, UK
| | - Eduardo Leyva-Díaz
- Instituto de Neurociencias, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), Sant Joan d'Alacant, Spain
| | - Marisa Karow
- Institute of Biochemistry, Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany
- Physiological Genomics, Biomedical Center, Ludwig Maximilians University Munich, 82152 Planegg/Munich, Germany
| | | | | | - José P López-Atalaya
- Instituto de Neurociencias, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), Sant Joan d'Alacant, Spain
| | - Benedikt Berninger
- Institute of Psychiatry, Psychology, and Neuroscience, Centre for Developmental Neurobiology, and MRC Centre for Neurodevelopmental Disorders, King's College London, SE1 1UL London, UK
- Institute of Physiological Chemistry, University Medical Center Johannes Gutenberg University Mainz, 55128 Mainz, Germany
| | - Guillermina López-Bendito
- Instituto de Neurociencias, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), Sant Joan d'Alacant, Spain.
| |
Collapse
|
18
|
Oldham Green N, Maniam J, Riese J, Morris MJ, Voineagu I. Transcriptomic signature of early life stress in male rat prefrontal cortex. Neurobiol Stress 2021; 14:100316. [PMID: 33796639 PMCID: PMC7995657 DOI: 10.1016/j.ynstr.2021.100316] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 01/02/2023] Open
Abstract
Early life stress (ELS) is associated with adverse mental health outcomes including anxiety, depression and addiction-like behaviours. While ELS is known to affect the developing brain, leading to increased stress responsiveness and increased glucocorticoid levels, the molecular mechanisms underlying the detrimental effects of ELS remain incompletely characterised. Rodent models have been instrumental in beginning to uncover the molecular and cellular underpinnings of ELS. Limited nesting (LN), an ELS behavioural paradigm with significant improvements over maternal separation, mimics human maternal neglect. We have previously shown that LN leads to an increase in one of the behavioural measures of anxiety like-behaviours in rats (percent of entries in the EPM open arm). Here we assessed gene expression changes induced by ELS in rat prefrontal cortex by RNA-sequencing. We show that LN leads primarily to transcriptional repression and identify a molecular signature of LN in rat PFC that is observed across ELS protocols and replicable across rodent species (mouse and rat).
Collapse
Affiliation(s)
- Nicole Oldham Green
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Jayanthi Maniam
- School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Jessica Riese
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Margaret J Morris
- School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Irina Voineagu
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
19
|
Abstract
Mitochondria are signaling hubs responsible for the generation of energy through oxidative phosphorylation, the production of key metabolites that serve the bioenergetic and biosynthetic needs of the cell, calcium (Ca2+) buffering and the initiation/execution of apoptosis. The ability of mitochondria to coordinate this myriad of functions is achieved through the exquisite regulation of fundamental dynamic properties, including remodeling of the mitochondrial network via fission and fusion, motility and mitophagy. In this Review, we summarize the current understanding of the mechanisms by which these dynamic properties of the mitochondria support mitochondrial function, review their impact on human cortical development and highlight areas in need of further research.
Collapse
Affiliation(s)
- Tierney Baum
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Vivian Gama
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
- Vanderbilt Center for Stem Cell Biology, Vanderbilt University, Nashville, TN 37232, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, USA
- Vanderbilt Ingram Cancer Center, Vanderbilt University, Nashville, TN 37232, USA
| |
Collapse
|
20
|
McDonough A, Elsen GE, Daza RM, Bachleda AR, Pizzo D, DelleTorri OM, Hevner RF. Unipolar (Dendritic) Brush Cells Are Morphologically Complex and Require Tbr2 for Differentiation and Migration. Front Neurosci 2021; 14:598548. [PMID: 33488348 PMCID: PMC7820753 DOI: 10.3389/fnins.2020.598548] [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: 08/25/2020] [Accepted: 12/04/2020] [Indexed: 01/21/2023] Open
Abstract
Previous studies demonstrated specific expression of transcription factor Tbr2 in unipolar brush cells (UBCs) of the cerebellum during development and adulthood. To further study UBCs and the role of Tbr2 in their development we examined UBC morphology in transgenic mouse lines (reporter and lineage tracer) and also examined the effects of Tbr2 deficiency in Tbr2 (MGI: Eomes) conditional knock-out (cKO) mice. In Tbr2 reporter and lineage tracer cerebellum, UBCs exhibited more complex morphologies than previously reported including multiple dendrites, bifurcating dendrites, and up to four dendritic brushes. We propose that “dendritic brush cells” (DBCs) may be a more apt nomenclature. In Tbr2 cKO cerebellum, mature UBCs were completely absent. Migration of UBC precursors from rhombic lip to cerebellar cortex and other nuclei was impaired in Tbr2 cKO mice. Our results indicate that UBC migration and differentiation are sensitive to Tbr2 deficiency. To investigate whether UBCs develop similarly in humans as in rodents, we studied Tbr2 expression in mid-gestational human cerebellum. Remarkably, Tbr2+ UBC precursors migrate along the same pathways in humans as in rodent cerebellum and disperse to create the same “fountain-like” appearance characteristic of UBCs exiting the rhombic lip.
Collapse
Affiliation(s)
- Ashley McDonough
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States
| | - Gina E Elsen
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States
| | - Ray M Daza
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States.,Department of Pathology, University of California, San Diego, CA, United States
| | - Amelia R Bachleda
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States
| | - Donald Pizzo
- Department of Pathology, University of California, San Diego, CA, United States
| | - Olivia M DelleTorri
- California Institute for Regenerative Medicine, California State University San Marcos, San Marcos, CA, United States
| | - Robert F Hevner
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States.,Department of Pathology, University of California, San Diego, CA, United States.,Department of Neurological Surgery, University of Washington, Seattle, WA, United States
| |
Collapse
|
21
|
Cole TB, Chang YC, Dao K, Daza R, Hevner R, Costa LG. Developmental exposure to diesel exhaust upregulates transcription factor expression, decreases hippocampal neurogenesis, and alters cortical lamina organization: relevance to neurodevelopmental disorders. J Neurodev Disord 2020; 12:41. [PMID: 33327933 PMCID: PMC7745370 DOI: 10.1186/s11689-020-09340-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 11/13/2020] [Indexed: 12/20/2022] Open
Abstract
Background Exposure to traffic-related air pollution (TRAP) during development and/or in adulthood has been associated in many human studies with both neurodevelopmental and neurodegenerative diseases, such as autism spectrum disorder (ASD) and Alzheimer’s disease (AD) or Parkinson’s disease (PD). Methods In the present study, C57BL/6 J mice were exposed to environmentally relevant levels (250+/−50 μg/m3) of diesel exhaust (DE) or filtered air (FA) during development (E0 to PND21). The expression of several transcription factors relevant for CNS development was assessed on PND3. To address possible mechanistic underpinnings of previously observed behavioral effects of DE exposure, adult neurogenesis in the hippocampus and laminar organization of neurons in the somatosensory cortex were analyzed on PND60. Results were analyzed separately for male and female mice. Results Developmental DE exposure caused a male-specific upregulation of Pax6, Tbr1, Tbr2, Sp1, and Creb1 on PND3. In contrast, in both males and females, Tbr2+ intermediate progenitor cells in the PND60 hippocampal dentate gyrus were decreased, as an indication of reduced adult neurogenesis. In the somatosensory region of the cerebral cortex, laminar distribution of Trb1, calbindin, and parvalbumin (but not of Ctip2 or Cux1) was altered by developmental DE exposure. Conclusions These results provide additional evidence to previous findings indicating the ability of developmental DE exposure to cause biochemical/molecular and behavioral alterations that may be involved in neurodevelopmental disorders such as ASD.
Collapse
Affiliation(s)
- Toby B Cole
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA. .,Center on Human Development and Disability, University of Washington, Seattle, WA, USA.
| | - Yu-Chi Chang
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA.,Gradient Corporation, Seattle, WA, USA
| | - Khoi Dao
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA
| | - Ray Daza
- Department of Pathology, University of California at San Diego, San Diego, CA, USA.,Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Robert Hevner
- Department of Pathology, University of California at San Diego, San Diego, CA, USA.,Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA.,Department of Neurological Surgery, University of Washington, Seattle, WA, USA
| | - Lucio G Costa
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA.,Department of Medicine and Surgery, University of Parma, Parma, Italy
| |
Collapse
|
22
|
Bueichekú E, Aznárez-Sanado M, Diez I, d'Oleire Uquillas F, Ortiz-Terán L, Qureshi AY, Suñol M, Basaia S, Ortiz-Terán E, Pastor MA, Sepulcre J. Central neurogenetic signatures of the visuomotor integration system. Proc Natl Acad Sci U S A 2020; 117:6836-6843. [PMID: 32144139 PMCID: PMC7104395 DOI: 10.1073/pnas.1912429117] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Visuomotor impairments characterize numerous neurological disorders and neurogenetic syndromes, such as autism spectrum disorder (ASD) and Dravet, Fragile X, Prader-Willi, Turner, and Williams syndromes. Despite recent advances in systems neuroscience, the biological basis underlying visuomotor functional impairments associated with these clinical conditions is poorly understood. In this study, we used neuroimaging connectomic approaches to map the visuomotor integration (VMI) system in the human brain and investigated the topology approximation of the VMI network to the Allen Human Brain Atlas, a whole-brain transcriptome-wide atlas of cortical genetic expression. We found the genetic expression of four genes-TBR1, SCN1A, MAGEL2, and CACNB4-to be prominently associated with visuomotor integrators in the human cortex. TBR1 gene transcripts, an ASD gene whose expression is related to neural development of the cortex and the hippocampus, showed a central spatial allocation within the VMI system. Our findings delineate gene expression traits underlying the VMI system in the human cortex, where specific genes, such as TBR1, are likely to play a central role in its neuronal organization, as well as on specific phenotypes of neurogenetic syndromes.
Collapse
Affiliation(s)
- Elisenda Bueichekú
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115
- Department of Basic Psychology, Clinical Psychology and Psychobiology, Jaume I University, 12071 Castelló de la Plana, Spain
| | - Maite Aznárez-Sanado
- Neuroimaging Laboratory, School of Medicine, University of Navarra, 31008 Pamplona, Spain
- School of Education and Psychology, University of Navarra, 31008 Pamplona, Spain
| | - Ibai Diez
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115
- Neurotechnology Laboratory, Health Department, Tecnalia, E-48160 Derio, Spain
| | - Federico d'Oleire Uquillas
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, 08540
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129
| | - Laura Ortiz-Terán
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115
- Department of Radiology, Brigham and Women's Hospital, Boston MA 02115
| | - Abid Y Qureshi
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129
- Department of Neurology, University of Kansas Medical Center, Kansas City, MO 66160
| | - Maria Suñol
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115
- Psychiatry Department, Bellvitge University Hospital, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), 08907 L'Hospitalet de Llobregat, Barcelona, Spain
- Center for Biomedical Research in Mental Health Network, Carlos III Health Institute, 08907 L'Hospitalet de Llobregat, Barcelona, Spain
- Department of Clinical Sciences, School of Medicine, University of Barcelona, 08036 Barcelona, Spain
| | - Silvia Basaia
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115
- Neuroimaging Research Unit, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Elena Ortiz-Terán
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115
| | - Maria A Pastor
- Neuroimaging Laboratory, School of Medicine, University of Navarra, 31008 Pamplona, Spain
| | - Jorge Sepulcre
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115;
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129
| |
Collapse
|
23
|
Nasirova N, Quina LA, Agosto-Marlin IM, Ramirez JM, Lambe EK, Turner EE. Dual recombinase fate mapping reveals a transient cholinergic phenotype in multiple populations of developing glutamatergic neurons. J Comp Neurol 2020; 528:283-307. [PMID: 31396962 PMCID: PMC6889053 DOI: 10.1002/cne.24753] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 07/16/2019] [Accepted: 08/02/2019] [Indexed: 01/13/2023]
Abstract
Cholinergic transmission shapes the maturation of glutamatergic circuits, yet the developmental sources of acetylcholine have not been systematically explored. Here, we have used Cre-recombinase-mediated genetic labeling to identify and map both mature and developing CNS neurons that express choline acetyltransferase (ChAT). Correction of a significant problem with a widely used ChatCre transgenic line ensures that this map does not contain expression artifacts. ChatCre marks all known cholinergic systems in the adult brain, but also identifies several brain areas not usually regarded as cholinergic, including specific thalamic and hypothalamic neurons, the subiculum, the lateral parabrachial nucleus, the cuneate/gracilis nuclei, and the pontocerebellar system. This ChatCre fate map suggests transient developmental expression of a cholinergic phenotype in areas important for cognition, motor control, and respiration. We therefore examined expression of ChAT and the vesicular acetylcholine transporter in the embryonic and early postnatal brain to determine the developmental timing of this transient cholinergic phenotype, and found that it mirrored the establishment of relevant glutamatergic projection pathways. We then used an intersectional genetic strategy combining ChatCre with Vglut2Flp to show that these neurons adopt a glutamatergic fate in the adult brain. The transient cholinergic phenotype of these glutamatergic neurons suggests a homosynaptic source of acetylcholine for the maturation of developing glutamatergic synapses. These findings thus define critical windows during which specific glutamatergic circuits may be vulnerable to disruption by nicotine in utero, and suggest new mechanisms for pediatric disorders associated with maternal smoking, such as sudden infant death syndrome.
Collapse
Affiliation(s)
- Nailyam Nasirova
- Center for Integrative Brain Research, Seattle Children’s Research Institute
| | - Lely A. Quina
- Center for Integrative Brain Research, Seattle Children’s Research Institute
| | | | - Jan-Marino Ramirez
- Center for Integrative Brain Research, Seattle Children’s Research Institute
| | - Evelyn K. Lambe
- Departments of Physiology, Obstetrics and Gynecology, and Psychiatry, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Eric E. Turner
- Center for Integrative Brain Research, Seattle Children’s Research Institute
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle WA, 98101
| |
Collapse
|
24
|
Lee YY, Choi HJ, Lee SY, Park SY, Kang MJ, Han J, Han JS. Bcl-2 Overexpression Induces Neurite Outgrowth via the Bmp4/Tbx3/NeuroD1 Cascade in H19-7 Cells. Cell Mol Neurobiol 2020; 40:153-166. [PMID: 31493044 DOI: 10.1007/s10571-019-00732-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 08/29/2019] [Indexed: 12/25/2022]
Abstract
Bcl-2 is overexpressed in the nervous system during neural development and plays an important role in modulating cell survival. In addition to its anti-apoptotic function, it has been suggested previously that Bcl-2 might act as a mediator of neuronal differentiation. However, the mechanism by which Bcl-2 might influence neurogenesis is not sufficiently understood. In this study, we aimed to determine the non-apoptotic functions of Bcl-2 during neuronal differentiation. First, we used microarrays to analyze the whole-genome expression patterns of rat neural stem cells overexpressing Bcl-2 and found that Bcl-2 overexpression induced the expression of various neurogenic genes. Moreover, Bcl-2 overexpression increased the neurite length as well as expression of Bmp4, Tbx3, and proneural basic helix-loop-helix genes, such as NeuroD1, NeuroD2, and Mash1, in H19-7 rat hippocampal precursor cells. To determine the hierarchy of these molecules, we selectively depleted Bmp4, Tbx3, and NeuroD1 in Bcl-2-overexpressing cells. Bmp4 depletion suppressed the upregulation of Tbx3 and NeuroD1 as well as neurite outgrowth, which was induced by Bcl-2 overexpression. Although Tbx3 knockdown repressed Bcl-2-mediated neurite elaboration and downregulated NeuroD1 expression, it did not affect Bcl-2-induced Bmp4 expression. While the depletion of NeuroD1 had no effect on the expression of Bcl-2, Bmp4, or Tbx3, Bcl-2-mediated neurite outgrowth was suppressed. Taken together, these results demonstrate that Bcl-2 regulates neurite outgrowth through the Bmp4/Tbx3/NeuroD1 cascade in H19-7 cells, indicating that Bcl-2 may have a direct role in neuronal development in addition to its well-known anti-apoptotic function in response to environmental insults.
Collapse
Affiliation(s)
- Yun Young Lee
- Department of Biomedical Sciences, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, 04763, Republic of Korea
- Biomedical Research Institute and Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul, 04763, Republic of Korea
| | - Hye-Jin Choi
- Biomedical Research Institute and Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul, 04763, Republic of Korea
| | - So Young Lee
- Department of Biomedical Sciences, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, 04763, Republic of Korea
| | - Shin-Young Park
- Biomedical Research Institute and Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul, 04763, Republic of Korea
| | - Min-Jeong Kang
- Department of Biomedical Sciences, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, 04763, Republic of Korea
| | - Jinil Han
- Department of Biomedical Sciences, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, 04763, Republic of Korea
- Gencurix, Inc, Hanwha Bizmetro 1, Guro 3-dong, Guro-gu, Seoul, 08394, Republic of Korea
| | - Joong-Soo Han
- Department of Biomedical Sciences, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, 04763, Republic of Korea.
- Biomedical Research Institute and Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul, 04763, Republic of Korea.
| |
Collapse
|
25
|
Molnár Z, Clowry GJ, Šestan N, Alzu'bi A, Bakken T, Hevner RF, Hüppi PS, Kostović I, Rakic P, Anton ES, Edwards D, Garcez P, Hoerder‐Suabedissen A, Kriegstein A. New insights into the development of the human cerebral cortex. J Anat 2019; 235:432-451. [PMID: 31373394 PMCID: PMC6704245 DOI: 10.1111/joa.13055] [Citation(s) in RCA: 206] [Impact Index Per Article: 41.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/11/2019] [Indexed: 12/12/2022] Open
Abstract
The cerebral cortex constitutes more than half the volume of the human brain and is presumed to be responsible for the neuronal computations underlying complex phenomena, such as perception, thought, language, attention, episodic memory and voluntary movement. Rodent models are extremely valuable for the investigation of brain development, but cannot provide insight into aspects that are unique or highly derived in humans. Many human psychiatric and neurological conditions have developmental origins but cannot be studied adequately in animal models. The human cerebral cortex has some unique genetic, molecular, cellular and anatomical features, which need to be further explored. The Anatomical Society devoted its summer meeting to the topic of Human Brain Development in June 2018 to tackle these important issues. The meeting was organized by Gavin Clowry (Newcastle University) and Zoltán Molnár (University of Oxford), and held at St John's College, Oxford. The participants provided a broad overview of the structure of the human brain in the context of scaling relationships across the brains of mammals, conserved principles and recent changes in the human lineage. Speakers considered how neuronal progenitors diversified in human to generate an increasing variety of cortical neurons. The formation of the earliest cortical circuits of the earliest generated neurons in the subplate was discussed together with their involvement in neurodevelopmental pathologies. Gene expression networks and susceptibility genes associated to neurodevelopmental diseases were discussed and compared with the networks that can be identified in organoids developed from induced pluripotent stem cells that recapitulate some aspects of in vivo development. New views were discussed on the specification of glutamatergic pyramidal and γ-aminobutyric acid (GABA)ergic interneurons. With the advancement of various in vivo imaging methods, the histopathological observations can be now linked to in vivo normal conditions and to various diseases. Our review gives a general evaluation of the exciting new developments in these areas. The human cortex has a much enlarged association cortex with greater interconnectivity of cortical areas with each other and with an expanded thalamus. The human cortex has relative enlargement of the upper layers, enhanced diversity and function of inhibitory interneurons and a highly expanded transient subplate layer during development. Here we highlight recent studies that address how these differences emerge during development focusing on diverse facets of our evolution.
Collapse
Affiliation(s)
- Zoltán Molnár
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
| | - Gavin J. Clowry
- Institute of NeuroscienceNewcastle UniversityNewcastle upon TyneUK
| | - Nenad Šestan
- Department of Neuroscience, Yale University School of MedicineNew HavenCTUSA
| | - Ayman Alzu'bi
- Department of Basic Medical SciencesFaculty of MedicineYarmouk UniversityIrbidJordan
| | | | | | - Petra S. Hüppi
- Dept. de l'enfant et de l'adolescentHôpitaux Universitaires de GenèveGenèveSwitzerland
| | - Ivica Kostović
- Croatian Institute for Brain ResearchSchool of MedicineUniversity of ZagrebZagrebCroatia
| | - Pasko Rakic
- Department of Neuroscience, Yale University School of MedicineNew HavenCTUSA
| | - E. S. Anton
- UNC Neuroscience CenterDepartment of Cell and Molecular PhysiologyThe University of North Carolina School of MedicineChapel HillNCUSA
| | - David Edwards
- Centre for the Developing BrainBiomedical Engineering and Imaging Sciences,King's College LondonLondonUK
| | - Patricia Garcez
- Federal University of Rio de Janeiro, UFRJInstitute of Biomedical SciencesRio de JaneiroBrazil
| | | | - Arnold Kriegstein
- Department of NeurologyUniversity of California, San Francisco (UCSF)San FranciscoCAUSA
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell ResearchUCSFSan FranciscoCAUSA
| |
Collapse
|
26
|
Mouse induced pluripotent stem cells-derived Alzheimer's disease cerebral organoid culture and neural differentiation disorders. Neurosci Lett 2019; 711:134433. [PMID: 31421155 DOI: 10.1016/j.neulet.2019.134433] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 08/11/2019] [Accepted: 08/13/2019] [Indexed: 11/20/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease, characterized by cognitive impairment. However, the pathogenesis of AD are very complicated, and the theories of Aβ and neurofibrillary tangles cannot explain all pathological alterations and clinical symptoms. Here, we used three-dimensional (3D) neural organoids culture derived from mouse induced pluripotent stem cells (iPSCs) to investigate the pathological mechanisms of AD. In this study, AD cerebral organoids were generated by overexpressing familial AD mutations (APP and PS1 genes) in mouse induced pluripotent stem cells, so that the early pathogenesis of AD could be investigated well with protein and cellular phenotype analyses. The results showed that AD cerebral organoids appeared some AD pathological alterations, and high levels of Aβ and p-Tau were induced as well. Furthermore, the number of GFAP-positive astrocytes and glutamatergic excitatory neurons increased significantly, but the number of GABAergic interneurons decreased. In conclusion, we suggest that cerebral organoids are a suitable AD model for scientific study, and that will provide us a novel insight into the understanding of the pathogenesis of AD.
Collapse
|
27
|
Docampo-Seara A, Lanoizelet M, Lagadec R, Mazan S, Candal E, Rodríguez MA. Mitral cell development in the olfactory bulb of sharks: evidences of a conserved pattern of glutamatergic neurogenesis. Brain Struct Funct 2019; 224:2325-2341. [PMID: 31203451 PMCID: PMC6698271 DOI: 10.1007/s00429-019-01906-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 06/07/2019] [Indexed: 12/18/2022]
Abstract
In mammals, the development of the olfactory bulb (OB) relies in part on the expression of transcription factors involved in the specifications/differentiation of glutamatergic cells. In a previous study from our group, a high molecular similarity was reported between mammals and cartilaginous fishes regarding the neurogenic mechanisms underlying the development of glutamatergic cells in the telencephalon. However, information about the transcriptional program operating in the development of the glutamatergic system (mainly represented by mitral cells) in the OB is lacking in the catshark Scyliorhinus canicula, a cartilaginous fish. Using immunohistochemistry and in situ hybridization techniques, we have found that, previously to the appearance of the olfactory primordium (OP), proliferating cells expressing Pax6 with molecular hallmarks of progenitor radial glia were located in the ventrolateral pallial ventricular zone. Later in development, when the OP is recognizable, a stream of Pax6-positive cells were observed between the ventricular zone and the OP, where transcription factors involved in mitral cell development in mammals (ScTbr2, ScNeuroD, Tbr1) are expressed. Later in development, these transcription factors became expressed in a layered-like structure where ScVglut1, a marker of mitral cells, is also present. Our data suggest that the transcriptional program related with the specification/differentiation of glutamatergic cells in the telencephalon has been conserved throughout the evolution of vertebrates. These results, in combination with previous studies concerning GABAergic neurogenesis in sharks, have evidenced that the OB of mammals and sharks shares similarities in the timing and molecular programs of development.
Collapse
Affiliation(s)
- A Docampo-Seara
- Departamento de Bioloxía Funcional, Centro de Investigación en Bioloxía (CIBUS), Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - M Lanoizelet
- CNRS, Sorbonne Universités, UPMC Univ Paris 06, UMR7232, Observatoire Océanologique, Banyuls sur Mer, France
| | - R Lagadec
- CNRS, Sorbonne Universités, UPMC Univ Paris 06, UMR7232, Observatoire Océanologique, Banyuls sur Mer, France
| | - S Mazan
- CNRS, Sorbonne Universités, UPMC Univ Paris 06, UMR7232, Observatoire Océanologique, Banyuls sur Mer, France
| | - E Candal
- Departamento de Bioloxía Funcional, Centro de Investigación en Bioloxía (CIBUS), Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - M A Rodríguez
- Departamento de Bioloxía Funcional, Centro de Investigación en Bioloxía (CIBUS), Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain.
| |
Collapse
|
28
|
Chang YC, Daza R, Hevner R, Costa LG, Cole TB. Prenatal and early life diesel exhaust exposure disrupts cortical lamina organization: Evidence for a reelin-related pathogenic pathway induced by interleukin-6. Brain Behav Immun 2019; 78:105-115. [PMID: 30668980 PMCID: PMC6557404 DOI: 10.1016/j.bbi.2019.01.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 01/10/2019] [Accepted: 01/16/2019] [Indexed: 12/20/2022] Open
Abstract
Several epidemiological studies have shown associations between developmental exposure to traffic-related air pollution and increased risk for autism spectrum disorders (ASD), a spectrum of neurodevelopmental disorders with increasing prevalence rate in the United States. Though animal studies have provided support for these associations, little is known regarding possible underlying mechanisms. In a previous study we found that exposure of C57BL/6J mice of both sexes to environmentally relevant levels (250-300 µg/m3) of diesel exhaust (DE) from embryonic day 0 to postnatal day 21 (E0 to PND21) caused significant changes in all three characteristic behavioral domains of ASD in the offspring. In the present study we investigated a potential mechanistic pathway that may be of relevance for ASD-like changes associated with developmental DE exposure. Using the same DE exposure protocol (250-300 µg/m3 DE from E0 to PND21) several molecular markers were examined in the brains of male and female mice at PND3, 21, and 60. Exposure to DE as above increased levels of interleukin-6 (IL-6) in placenta and in neonatal brain. The JAK2/STAT3 pathway, a target for IL-6, was activated by STAT3 phosphorylation, and the expression of DNA methyltransferase 1 (DNMT1), a STAT3 target gene, was increased in DE-exposed neonatal brain. DNMT1 has been reported to down-regulate expression of reelin (RELN), an extracellular matrix glycoprotein important in regulating the processes of neuronal migration. RELN is considered an important modulator for ASD, since there are several polymorphisms in this gene linked to the disease, and since lower levels of RELN have been reported in brains of ASD patients. We observed decreased RELN expression in brains of the DE-exposed mice at PND3. Since disorganized patches in the prefrontal cortex have been reported in ASD patients and disrupted cortical organization has been found in RELN-deficient mice, we also assessed cortical organization, by labeling cells expressing the lamina-specific-markers RELN and calretinin. In DE-exposed mice we found increased cell density in deeper cortex (lamina layers VI-IV) for cells expressing either RELN or calretinin. These findings demonstrate that developmental DE exposure is associated with subtle disorganization of the cerebral cortex at PND60, and suggest a pathway involving IL-6, STAT3, and DNMT1 leading to downregulation of RELN expression that could be contributing to this long-lasting disruption in cortical laminar organization.
Collapse
Affiliation(s)
- Yu-Chi Chang
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA.
| | - Ray Daza
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA.
| | - Robert Hevner
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA; Department of Neurological Surgery, University of Washington, Seattle, WA, USA.
| | - Lucio G. Costa
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA,Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Toby B. Cole
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA,Center on Human Development and Disability, University of Washington, Seattle, WA, USA,Corresponding author at: Department of Environmental and Occupational Health Sciences, University of Washington, Box 357234, 1959 NE Pacific St., Seattle, WA, USA. (Y.-C. Chang), , (R. Daza), , (R. Hevner), (L.G. Costa), (T.B. Cole)
| |
Collapse
|
29
|
Kiyama T, Long Y, Chen CK, Whitaker CM, Shay A, Wu H, Badea TC, Mohsenin A, Parker-Thornburg J, Klein WH, Mills SL, Massey SC, Mao CA. Essential Roles of Tbr1 in the Formation and Maintenance of the Orientation-Selective J-RGCs and a Group of OFF-Sustained RGCs in Mouse. Cell Rep 2019; 27:900-915.e5. [PMID: 30995485 PMCID: PMC6542366 DOI: 10.1016/j.celrep.2019.03.077] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 02/10/2019] [Accepted: 03/20/2019] [Indexed: 01/27/2023] Open
Abstract
In the mouse retina, more than 30 retinal ganglion cell (RGC) subtypes have been classified based on a combined metric of morphological and functional characteristics. RGCs arise from a common pool of retinal progenitor cells during embryonic stages and differentiate into mature subtypes in adult retinas. However, the cellular and molecular mechanisms controlling formation and maturation of such remarkable cellular diversity remain unknown. Here, we demonstrate that T-box transcription factor T-brain 1 (Tbr1) is expressed in two groups of morphologically and functionally distinct RGCs: the orientation-selective J-RGCs and a group of OFF-sustained RGCs with symmetrical dendritic arbors. When Tbr1 is genetically ablated during retinal development, these two RGC groups cannot develop. Ectopically expressing Tbr1 in M4 ipRGCs during development alters dendritic branching and density but not the inner plexiform layer stratification level. Our data indicate that Tbr1 plays critical roles in regulating the formation and dendritic morphogenesis of specific RGC types.
Collapse
Affiliation(s)
- Takae Kiyama
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA
| | - Ye Long
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA
| | - Ching-Kang Chen
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Christopher M Whitaker
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA
| | - Allison Shay
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hongyu Wu
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA
| | - Tudor C Badea
- National Eye Institute, NIH, Bethesda, MD 20892, USA
| | - Amir Mohsenin
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA; Robert Cizik Eye Clinic, Houston, TX 77030, USA
| | - Jan Parker-Thornburg
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - William H Klein
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Stephen L Mills
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA
| | - Stephen C Massey
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA
| | - Chai-An Mao
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA.
| |
Collapse
|
30
|
Adamo AM, Liu X, Mathieu P, Nuttall JR, Supasai S, Oteiza PI. Early Developmental Marginal Zinc Deficiency Affects Neurogenesis Decreasing Neuronal Number and Altering Neuronal Specification in the Adult Rat Brain. Front Cell Neurosci 2019; 13:62. [PMID: 30890920 PMCID: PMC6414196 DOI: 10.3389/fncel.2019.00062] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 02/08/2019] [Indexed: 12/03/2022] Open
Abstract
During pregnancy, a decreased availability of zinc to the fetus can disrupt the development of the central nervous system leading to defects ranging from severe malformations to subtle neurological and cognitive effects. We previously found that marginal zinc deficiency down-regulates the extracellular signal-regulated kinase 1/2 (ERK1/2) signaling pathway and affects neural progenitor cell (NPC) proliferation. This study investigated if marginal zinc deficiency during gestation in rats could disrupt fetal neurogenesis and affect the number and specification of neurons in the adult offspring brain cortex. Rats were fed a marginal zinc deficient or adequate diet throughout gestation and until postnatal day (P) 2, and subsequently the zinc adequate diet until P56. Neurogenesis was evaluated in the offspring at embryonic day (E)14, E19, P2, and P56 measuring parameters of NPC proliferation and differentiation by Western blot and/or immunofluorescence. At E14 and E19, major signals (i.e., ERK1/2, Sox2, and Pax6) that stimulate NPC proliferation and self-renewal were markedly downregulated in the marginal zinc deficient fetal brain. These alterations were associated to a lower number of Ki67 positive cells in the ventricular (VZs) and subventricular zones (SVZs). Following the progression of NPCs into intermediate progenitor cells (IPCs) and into neurons, Pax6, Tbr2 and Tbr1 were affected in the corresponding areas of the brain at E19 and P2. The above signaling alterations led to a lower density of neurons and a selective decrease of glutamatergic neurons in the young adult brain cortex exposed to maternal marginal zinc deficiency from E14 to P2. Current results supports the concept that marginal zinc deficiency during fetal development can disrupt neurogenesis and alter cortical structure potentially leading to irreversible neurobehavioral impairments later in life.
Collapse
Affiliation(s)
- Ana M Adamo
- Department of Biological Chemistry and IQUIFIB (UBA-CONICET), Facultad de Farmacia y Bioquimica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Xiuzhen Liu
- Department of Nutrition and Department of Environmental Toxicology, University of California, Davis, Davis, CA, United States
| | - Patricia Mathieu
- Department of Biological Chemistry and IQUIFIB (UBA-CONICET), Facultad de Farmacia y Bioquimica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Johnathan R Nuttall
- Department of Nutrition and Department of Environmental Toxicology, University of California, Davis, Davis, CA, United States
| | - Suangsuda Supasai
- Department of Nutrition and Department of Environmental Toxicology, University of California, Davis, Davis, CA, United States.,Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Patricia I Oteiza
- Department of Nutrition and Department of Environmental Toxicology, University of California, Davis, Davis, CA, United States
| |
Collapse
|
31
|
Huang TN, Yen TL, Qiu LR, Chuang HC, Lerch JP, Hsueh YP. Haploinsufficiency of autism causative gene Tbr1 impairs olfactory discrimination and neuronal activation of the olfactory system in mice. Mol Autism 2019; 10:5. [PMID: 30792833 PMCID: PMC6371489 DOI: 10.1186/s13229-019-0257-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 01/25/2019] [Indexed: 12/21/2022] Open
Abstract
Background Autism spectrum disorders (ASD) exhibit two clusters of core symptoms, i.e., social and communication impairment, and repetitive behaviors and sensory abnormalities. Our previous study demonstrated that TBR1, a causative gene of ASD, controls axonal projection and neuronal activation of amygdala and regulates social interaction and vocal communication in a mouse model. Behavioral defects caused by Tbr1 haploinsufficiency can be ameliorated by increasing neural activity via D-cycloserine treatment, an N-methyl-D-aspartate receptor (NMDAR) coagonist. In this report, we investigate the role of TBR1 in regulating olfaction and test whether D-cycloserine can also improve olfactory defects in Tbr1 mutant mice. Methods We used Tbr1+/− mice as a model to investigate the function of TBR1 in olfactory sensation and discrimination of non-social odors. We employed a behavioral assay to characterize the olfactory defects of Tbr1+/− mice. Magnetic resonance imaging (MRI) and histological analysis were applied to characterize anatomical features. Immunostaining was performed to further analyze differences in expression of TBR1 subfamily members (namely TBR1, TBR2, and TBX21), interneuron populations, and dendritic abnormalities in olfactory bulbs. Finally, C-FOS staining was used to monitor neuronal activation of the olfactory system upon odor stimulation. Results Tbr1+/− mice exhibited smaller olfactory bulbs and anterior commissures, reduced interneuron populations, and an abnormal dendritic morphology of mitral cells in the olfactory bulbs. Tbr1 haploinsufficiency specifically impaired olfactory discrimination but not olfactory sensation. Neuronal activation upon odorant stimulation was reduced in the glomerular layer of Tbr1+/− olfactory bulbs. Furthermore, although the sizes of piriform and perirhinal cortices were not affected by Tbr1 deficiency, neuronal activation was reduced in these two cortical regions in response to odorant stimulation. These results suggest an impairment of neuronal activation in olfactory bulbs and defective connectivity from olfactory bulbs to the upper olfactory system in Tbr1+/− mice. Systemic administration of D-cycloserine, an NMDAR co-agonist, ameliorated olfactory discrimination in Tbr1+/− mice, suggesting that increased neuronal activity has a beneficial effect on Tbr1 deficiency. Conclusions Tbr1 regulates neural circuits and activity in the olfactory system to control olfaction. Tbr1+/− mice can serve as a suitable model for revealing how an autism causative gene controls neuronal circuits, neural activity, and autism-related behaviors. Electronic supplementary material The online version of this article (10.1186/s13229-019-0257-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tzyy-Nan Huang
- 1Institute of Molecular Biology, Academia Sinica, 128, Academia Rd., Sec. 2, Taipei, 11529 Taiwan
| | - Tzu-Li Yen
- 1Institute of Molecular Biology, Academia Sinica, 128, Academia Rd., Sec. 2, Taipei, 11529 Taiwan
| | - Lily R Qiu
- 2Mouse Imaging Centre, Hospital for Sick Children, Toronto, Canada
| | - Hsiu-Chun Chuang
- 1Institute of Molecular Biology, Academia Sinica, 128, Academia Rd., Sec. 2, Taipei, 11529 Taiwan.,4Present address: Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720 USA
| | - Jason P Lerch
- 2Mouse Imaging Centre, Hospital for Sick Children, Toronto, Canada.,3Department of Medical Biophysics, The University of Toronto, Toronto, Canada
| | - Yi-Ping Hsueh
- 1Institute of Molecular Biology, Academia Sinica, 128, Academia Rd., Sec. 2, Taipei, 11529 Taiwan
| |
Collapse
|
32
|
Elsen GE, Bedogni F, Hodge RD, Bammler TK, MacDonald JW, Lindtner S, Rubenstein JLR, Hevner RF. The Epigenetic Factor Landscape of Developing Neocortex Is Regulated by Transcription Factors Pax6→ Tbr2→ Tbr1. Front Neurosci 2018; 12:571. [PMID: 30186101 PMCID: PMC6113890 DOI: 10.3389/fnins.2018.00571] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 07/30/2018] [Indexed: 12/12/2022] Open
Abstract
Epigenetic factors (EFs) regulate multiple aspects of cerebral cortex development, including proliferation, differentiation, laminar fate, and regional identity. The same neurodevelopmental processes are also regulated by transcription factors (TFs), notably the Pax6→ Tbr2→ Tbr1 cascade expressed sequentially in radial glial progenitors (RGPs), intermediate progenitors, and postmitotic projection neurons, respectively. Here, we studied the EF landscape and its regulation in embryonic mouse neocortex. Microarray and in situ hybridization assays revealed that many EF genes are expressed in specific cortical cell types, such as intermediate progenitors, or in rostrocaudal gradients. Furthermore, many EF genes are directly bound and transcriptionally regulated by Pax6, Tbr2, or Tbr1, as determined by chromatin immunoprecipitation-sequencing and gene expression analysis of TF mutant cortices. Our analysis demonstrated that Pax6, Tbr2, and Tbr1 form a direct feedforward genetic cascade, with direct feedback repression. Results also revealed that each TF regulates multiple EF genes that control DNA methylation, histone marks, chromatin remodeling, and non-coding RNA. For example, Tbr1 activates Rybp and Auts2 to promote the formation of non-canonical Polycomb repressive complex 1 (PRC1). Also, Pax6, Tbr2, and Tbr1 collectively drive massive changes in the subunit isoform composition of BAF chromatin remodeling complexes during differentiation: for example, a novel switch from Bcl7c (Baf40c) to Bcl7a (Baf40a), the latter directly activated by Tbr2. Of 11 subunits predominantly in neuronal BAF, 7 were transcriptionally activated by Pax6, Tbr2, or Tbr1. Using EFs, Pax6→ Tbr2→ Tbr1 effect persistent changes of gene expression in cell lineages, to propagate features such as regional and laminar identity from progenitors to neurons.
Collapse
Affiliation(s)
- Gina E. Elsen
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States
| | - Francesco Bedogni
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States
| | - Rebecca D. Hodge
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States
| | - Theo K. Bammler
- Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA, United States
| | - James W. MacDonald
- Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA, United States
| | - Susan Lindtner
- Nina Ireland Laboratory of Developmental Neurobiology, University of California, San Francisco, San Francisco, CA, United States
- Department of Psychiatry, University of California, San Francisco, San Francisco, CA, United States
| | - John L. R. Rubenstein
- Nina Ireland Laboratory of Developmental Neurobiology, University of California, San Francisco, San Francisco, CA, United States
- Department of Psychiatry, University of California, San Francisco, San Francisco, CA, United States
| | - Robert F. Hevner
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States
- Department of Neurological Surgery, School of Medicine, University of Washington, Seattle, WA, United States
| |
Collapse
|
33
|
Bagnoli E, FitzGerald U. Mitral cells and the glucagon-like peptide 1 receptor: The sweet smell of success? Eur J Neurosci 2018; 49:422-439. [PMID: 30120857 DOI: 10.1111/ejn.14115] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 07/19/2018] [Accepted: 08/03/2018] [Indexed: 12/11/2022]
Abstract
The olfactory bulb (OB) is often affected at very early stages of neurodegenerative disorders, in the so-called "prodromal" phase. In Parkinson's disease (PD), olfactory disturbances appear years before motor symptoms arise. Additionally, pathological alpha-synuclein aggregates are found in olfactory regions before spreading to other areas of the brain. Being positioned at the frontier between the brain and a potentially hostile environment, could explain the particular vulnerability of the OB. Mitral cells (MCs), the principal projecting neurons of the olfactory system, are involved in the pathogenesis and in the prion-like progression of PD. They are affected by Lewy pathology and are thought to contribute to the axonal transport of misfolded alpha-synuclein to other regions of the brain. Here, we first describe the main markers reported to distinguish MCs from other olfactory neurons. We focus on the glucagon-like peptide 1 receptor (GLP-1R), a membrane protein specifically expressed in MCs. After summarizing OB pathology, we explore the idea of targeting specifically MCs with GLP-1 or its analogues. Exenatide has shown great promise as a neuroprotective and neurorestorative agent and has been used in a clinical trial for clinical PD. Since GLP-1R activation has the ability to mitigate many facets of prodromal PD pathology, we postulate that once a robust biomarker is in place that is capable of identifying individuals in the prodromal phase of PD, homing in on GLP-1R could assist in deferring, or eradicating to a significant degree, the clinical manifestation of this debilitating human disorder.
Collapse
Affiliation(s)
- Enrico Bagnoli
- CÚRAM, Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland.,Galway Neuroscience Centre, School of Natural Sciences, National University of Ireland Galway, Galway, Ireland
| | - Una FitzGerald
- CÚRAM, Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland.,Galway Neuroscience Centre, School of Natural Sciences, National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
34
|
Apitz H, Salecker I. Spatio-temporal relays control layer identity of direction-selective neuron subtypes in Drosophila. Nat Commun 2018; 9:2295. [PMID: 29895891 PMCID: PMC5997761 DOI: 10.1038/s41467-018-04592-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 05/04/2018] [Indexed: 11/09/2022] Open
Abstract
Visual motion detection in sighted animals is essential to guide behavioral actions ensuring their survival. In Drosophila, motion direction is first detected by T4/T5 neurons. Their axons innervate one of the four lobula plate layers. How T4/T5 neurons with layer-specific representation of motion-direction preferences are specified during development is unknown. We show that diffusible Wingless (Wg) between adjacent neuroepithelia induces its own expression to form secondary signaling centers. These activate Decapentaplegic (Dpp) signaling in adjacent lateral tertiary neuroepithelial domains dedicated to producing layer 3/4-specific T4/T5 neurons. T4/T5 neurons derived from the core domain devoid of Dpp signaling adopt the default layer 1/2 fate. Dpp signaling induces the expression of the T-box transcription factor Optomotor-blind (Omb), serving as a relay to postmitotic neurons. Omb-mediated repression of Dachshund transforms layer 1/2- into layer 3/4-specific neurons. Hence, spatio-temporal relay mechanisms, bridging the distances between neuroepithelial domains and their postmitotic progeny, implement T4/T5 neuron-subtype identity.
Collapse
Affiliation(s)
- Holger Apitz
- The Francis Crick Institute, Visual Circuit Assembly Laboratory, 1 Midland Road, London, NW1 1AT, UK
| | - Iris Salecker
- The Francis Crick Institute, Visual Circuit Assembly Laboratory, 1 Midland Road, London, NW1 1AT, UK.
| |
Collapse
|
35
|
Liu J, Reggiani JDS, Laboulaye MA, Pandey S, Chen B, Rubenstein JLR, Krishnaswamy A, Sanes JR. Tbr1 instructs laminar patterning of retinal ganglion cell dendrites. Nat Neurosci 2018; 21:659-670. [PMID: 29632360 PMCID: PMC5920715 DOI: 10.1038/s41593-018-0127-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 02/20/2018] [Indexed: 12/20/2022]
Abstract
Visual information is delivered to the brain by >40 types of retinal ganglion cells (RGCs). Diversity in this representation arises within the inner plexiform layer (IPL), where dendrites of each RGC type are restricted to specific sublaminae, limiting the interneuronal types that can innervate them. How such dendritic restriction arises is unclear. We show that the transcription factor Tbr1 is expressed by four mouse RGC types with dendrites in the outer IPL and is required for their laminar specification. Loss of Tbr1 results in elaboration of dendrites within the inner IPL, while misexpression in other cells retargets their neurites to the outer IPL. Two transmembrane molecules, Sorcs3 and Cdh8, act as effectors of the Tbr1-controlled lamination program. However, they are expressed in just one Tbr1+ RGC type, supporting a model in which a single transcription factor implements similar laminar choices in distinct cell types by recruiting partially non-overlapping effectors.
Collapse
Affiliation(s)
- Jinyue Liu
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA.,Center for Brain Science, Harvard University, Cambridge, MA, USA.,Program in Neuroscience, Harvard Medical School, Boston, MA, USA
| | - Jasmine D S Reggiani
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA.,Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Mallory A Laboulaye
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA.,Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Shristi Pandey
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA.,Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Bin Chen
- Department of Molecular, Cell and Developmental Biology, University of California at Santa Cruz, Santa Cruz, CA, USA
| | - John L R Rubenstein
- Department of Psychiatry, University of California at San Francisco, San Francisco, CA, USA
| | - Arjun Krishnaswamy
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA.,Center for Brain Science, Harvard University, Cambridge, MA, USA.,Department of Physiology, McGill University, Montreal, QC, Canada
| | - Joshua R Sanes
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA. .,Center for Brain Science, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
36
|
Lo FS, Erzurumlu RS. Insulin receptor sensitization restores neocortical excitation/inhibition balance in a mouse model of autism. Mol Autism 2018; 9:13. [PMID: 29484150 PMCID: PMC5824550 DOI: 10.1186/s13229-018-0196-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 01/19/2018] [Indexed: 01/01/2023] Open
Abstract
Background Met receptor tyrosine kinase regulates neurogenesis, differentiation, migration, connectivity, and synaptic plasticity. The human Met gene has been identified as a prominent risk factor for autism spectrum disorder (ASD). Met gene-altered mice serve as useful models for mechanistic studies of ASD. Inactivation of Met in excitatory cortical neurons in mice (Emx1cre/Metflox mice) yields a phenotype in which significantly decreased GABAA receptor-mediated inhibition shifts the excitation/inhibition (E/I) balance toward excitation in the somatosensory cortex. Further, unlike that seen in wild-type mice, insulin does not increase inhibition in the mutant cortex, suggesting that one of the consequences of kinase inactive Met gene could be desensitization of insulin receptors. To test this hypothesis, we investigated the effects of insulin receptor sensitizer, pioglitazone, on inhibition in the somatosensory thalamocortical circuitry. Methods We used whole-cell patch clamp electrophysiology and analyzed excitatory and inhibitory responses of cortical layer IV excitatory cells following stimulation of their thalamic input in thalamocortical pathway intact brain slices. We applied insulin alone and insulin + a thiazolidinedione, pioglitazone (PIO), to test the effects of sensitizing insulin receptors on inhibitory responses mediated by GABAA receptors in the somatosensory cortex of Emx1cre/Metflox mice. Results In WT brain slices, application of insulin together with PIO did not enhance the effect of insulin alone. In contrast, PIO application induced a much larger inhibition than that of insulin alone in Met-defective cortex. Thus, insulin resistance of GABAA receptor-mediated response in Met mutant mice may result from desensitized insulin receptors. Conclusions Sporadic clinical studies reported improved behavioral symptoms in children with autism following PIO treatment. We show that PIO can aid in normalization of the E/I balance in the primary somatosensory cortex, a potential physiological mechanism underlying the positive effects of PIO treatment.
Collapse
Affiliation(s)
- Fu-Sun Lo
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, 22 Penn Street HSFII-S259, Baltimore, MD 21201 USA
| | - Reha S Erzurumlu
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, 22 Penn Street HSFII-S259, Baltimore, MD 21201 USA
| |
Collapse
|
37
|
Kaplan ES, Ramos-Laguna KA, Mihalas AB, Daza RAM, Hevner RF. Neocortical Sox9+ radial glia generate glutamatergic neurons for all layers, but lack discernible evidence of early laminar fate restriction. Neural Dev 2017; 12:14. [PMID: 28814327 DOI: 10.1186/s13064-017-0091-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 08/07/2017] [Indexed: 11/10/2022] Open
Abstract
Glutamatergic neurons in the cerebral cortex are derived from embryonic neural stem cells known as radial glial progenitors (RGPs). Early RGPs, present at the onset of cortical neurogenesis, are classically thought to produce columnar clones of glutamatergic neurons spanning the cortical layers. Recently, however, it has been reported that a subset of early RGPs may undergo early commitment to upper layer neuron fates, thus bypassing genesis of deep layer neurons. However, the latter mode of early RGP differentiation was not confirmed in some other studies, and remains controversial. To further investigate the clonal output from early RGPs, we employed genetic lineage tracing driven by Sox9, a transcription factor gene that is expressed in all early RGPs. We found that early RGPs produced columnar clones spanning all cortical layers, with no evidence of significant laminar fate restriction. These data support the classic progressive restriction model of cortical neurogenesis, and suggest that early RGPs do not undergo early commitment to only upper or lower layer fates.
Collapse
Affiliation(s)
- E S Kaplan
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, 98101, USA
| | - K A Ramos-Laguna
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, 98101, USA
| | - A B Mihalas
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, 98101, USA
| | - R A M Daza
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, 98101, USA
| | - R F Hevner
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, 98101, USA. .,Department of Neurological Surgery, University of Washington School of Medicine, Seattle, WA, 98104, USA.
| |
Collapse
|
38
|
Probst S, Daza RA, Bader N, Hummel JF, Weiß M, Tanriver Y, Hevner RF, Arnold SJ. A dual-fluorescence reporter in the Eomes locus for live imaging and medium-term lineage tracing. Genesis 2017. [PMID: 28646547 DOI: 10.1002/dvg.23043] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The T-box transcription factor Eomes (also known as Tbr2) shows short-lived expression in various localized domains of the embryo, including epiblast cells during gastrulation and intermediate progenitor cells in the cerebral cortex. In these tissues Eomes fulfills crucial roles for lineage specification of progenitors. To directly observe Eomes-dependent cell lineages in the living embryo, we generated a novel dual-fluorescence reporter allele that expresses a membrane-bound tdTomato protein for investigation of cell morphology and a nuclear GFP for cell tracing. This allele recapitulates endogenous EOMES protein expression and is suitable for live imaging. We found that the allele can also be used as a short-to-medium-term lineage tracer, as GFP persists in cells longer than EOMES protein and marks Eomes-dependent lineages with a timeframe of days to weeks depending on the proliferation rate. In summary, we present a novel genetic tool for investigation of Eomes-dependent cell types by live imaging and lineage tracing.
Collapse
Affiliation(s)
- Simone Probst
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ray A Daza
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington, 98101
| | - Natalie Bader
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Jonas F Hummel
- Institute of Medical Microbiology and Hygiene, Faculty of Medicine, University Medical Center, Freiburg, Germany
| | - Matthias Weiß
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Yakup Tanriver
- Institute of Medical Microbiology and Hygiene, Faculty of Medicine, University Medical Center, Freiburg, Germany.,Department of Internal Medicine IV, Faculty of Medicine, University Medical Center, Freiburg, Germany
| | - Robert F Hevner
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington, 98101
| | - Sebastian J Arnold
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,BIOSS Centre of Biological Signalling Studies, Albert-Ludwigs-University, Freiburg, Germany
| |
Collapse
|
39
|
Higgins GA, Georgoff P, Nikolian V, Allyn-Feuer A, Pauls B, Higgins R, Athey BD, Alam HE. Network Reconstruction Reveals that Valproic Acid Activates Neurogenic Transcriptional Programs in Adult Brain Following Traumatic Injury. Pharm Res 2017; 34:1658-1672. [PMID: 28271248 PMCID: PMC5498621 DOI: 10.1007/s11095-017-2130-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 02/17/2017] [Indexed: 12/22/2022]
Abstract
Objectives To determine the mechanism of action of valproic acid (VPA) in the adult central nervous system (CNS) following traumatic brain injury (TBI) and hemorrhagic shock (HS). Methods Data were analyzed from different sources, including experiments in a porcine model, data from postmortem human brain, published studies, public and commercial databases. Results The transcriptional program in the CNS following TBI, HS, and VPA treatment includes activation of regulatory pathways that enhance neurogenesis and suppress gliogenesis. Genes which encode the transcription factors (TFs) that specify neuronal cell fate, including MEF2D, MYT1L, NEUROD1, PAX6 and TBR1, and their target genes, are induced by VPA. VPA represses genes responsible for oligodendrogenesis, maintenance of white matter, T-cell activation, angiogenesis, and endothelial cell proliferation, adhesion and chemotaxis. NEUROD1 has regulatory interactions with 38% of the genes regulated by VPA in a swine model of TBI and HS in adult brain. Hi-C spatial mapping of a VPA pharmacogenomic SNP in the GRIN2B gene shows it is part of a transcriptional hub that contacts 12 genes that mediate chromatin-mediated neurogenesis and neuroplasticity. Conclusions Following TBI and HS, this study shows that VPA administration acts in the adult brain through differential activation of TFs responsible for neurogenesis, genes responsible for neuroplasticity, and repression of TFs that specify oligodendrocyte cell fate, endothelial cell chemotaxis and angiogenesis. Short title: Mechanism of action of valproic acid in traumatic brain injury Electronic supplementary material The online version of this article (doi:10.1007/s11095-017-2130-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Gerald A. Higgins
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan USA
| | - Patrick Georgoff
- Department of Surgery, University of Michigan Medical School, Ann Arbor, Michigan USA
| | - Vahagn Nikolian
- Department of Surgery, University of Michigan Medical School, Ann Arbor, Michigan USA
| | - Ari Allyn-Feuer
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan USA
| | - Brian Pauls
- College of Pharmacy, University of Michigan, Ann Arbor, Michigan USA
| | - Richard Higgins
- Department of Computer Science, University of Maryland, College Park, Maryland USA
| | - Brian D. Athey
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan USA
- Michigan Institute for Data Science (MIDAS), Ann Arbor, Michigan USA
| | - Hasan E. Alam
- Department of Surgery, University of Michigan Medical School, Ann Arbor, Michigan USA
| |
Collapse
|