1
|
Clément AE, Merdrignac C, Puiggros SR, Sévère D, Brionne A, Lafond T, Nguyen T, Montfort J, Guyomar C, Dauvé A, Herpin A, Jabaudon D, Colson V, Murat F, Bobe J. Parent-of-origin regulation by maternal auts2 shapes neurodevelopment and behavior in fish. Genome Biol 2025; 26:125. [PMID: 40346605 PMCID: PMC12063280 DOI: 10.1186/s13059-025-03600-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 04/29/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND Parental experience can influence progeny behavior through gamete-mediated non-genetic inheritance, that is, mechanisms that do not involve changes in inherited DNA sequence. However, underlying mechanisms remain poorly understood in vertebrates, especially for maternal effects. Here, we use the medaka, a model fish species, to investigate the role of auts2a, the ortholog of human AUTS2, a gene repressed in the fish oocyte following maternal stress and associated with neurodevelopmental disorders. RESULTS We show that auts2a expression in the oocyte influences long-term progeny behavior, including anxiety-like behavior and environment recognition capabilities. Using single-nuclei RNA-sequencing, we reveal that maternal auts2a influences gene expression in neural cell populations during neurodevelopment. We also show that maternal auts2a knock-out triggers differences in maternally inherited factors, including early embryonic transcriptional and post-transcriptional regulators. CONCLUSIONS Together, our results reveal the unsuspected role of an autism-related gene expressed in the mother's oocyte in shaping progeny neurodevelopment and behavior. Finally, we report that auts2a/AUTS2 is part of a group of evolutionarily conserved genes associated with human neurodevelopmental disorders and expressed in oocytes across species, from fish to mammals. These findings raise important questions about their potential role in the non-genetic regulation of progeny neurodevelopment and behavior in vertebrates.
Collapse
Affiliation(s)
| | | | - Sergi Roig Puiggros
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
| | - Dorine Sévère
- INRAE, LPGP UR1037, Fish Physiology and Genomics Institute, Rennes, France
| | - Aurélien Brionne
- INRAE, LPGP UR1037, Fish Physiology and Genomics Institute, Rennes, France
| | - Thomas Lafond
- INRAE, LPGP UR1037, Fish Physiology and Genomics Institute, Rennes, France
| | - Thaovi Nguyen
- INRAE, LPGP UR1037, Fish Physiology and Genomics Institute, Rennes, France
| | - Jérôme Montfort
- INRAE, LPGP UR1037, Fish Physiology and Genomics Institute, Rennes, France
| | - Cervin Guyomar
- Sigenae, GenPhySE, INRAE, ENVT, Université de Toulouse, Toulouse, Castanet Tolosan, France
| | - Alexandra Dauvé
- MGX-Montpellier GenomiX, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Amaury Herpin
- INRAE, LPGP UR1037, Fish Physiology and Genomics Institute, Rennes, France
| | - Denis Jabaudon
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
| | - Violaine Colson
- INRAE, LPGP UR1037, Fish Physiology and Genomics Institute, Rennes, France
| | - Florent Murat
- INRAE, LPGP UR1037, Fish Physiology and Genomics Institute, Rennes, France
| | - Julien Bobe
- INRAE, LPGP UR1037, Fish Physiology and Genomics Institute, Rennes, France.
| |
Collapse
|
2
|
Saloni, Sachan M, Rahul, Verma RS, Patel GK. SOXs: Master architects of development and versatile emulators of oncogenesis. Biochim Biophys Acta Rev Cancer 2025; 1880:189295. [PMID: 40058508 DOI: 10.1016/j.bbcan.2025.189295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 02/26/2025] [Accepted: 03/03/2025] [Indexed: 03/22/2025]
Abstract
Transcription factors regulate a variety of events and maintain cellular homeostasis. Several transcription factors involved in embryonic development, has been shown to be closely associated with carcinogenesis when deregulated. Sry-like high mobility group box (SOX) proteins are potential transcription factors which are evolutionarily conserved. They regulate downstream genes to determine cell fate, via various signaling pathways and cellular processes essential for tissue and organ development. Dysregulation of SOXs has been reported to promote or suppress tumorigenesis by modulating cellular reprogramming, growth, proliferation, angiogenesis, metastasis, apoptosis, immune modulation, lineage plasticity, maintenance of the stem cell pool, therapy resistance and cancer relapse. This review provides a crucial understanding of the molecular mechanism by which SOXs play multifaceted roles in embryonic development and carcinogenesis. It also highlights their potential in advancing therapeutic strategies aimed at targeting SOXs and their downstream effectors in various malignancies.
Collapse
Affiliation(s)
- Saloni
- Cancer and Stem Cell Laboratory, Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj 211004, India
| | - Manisha Sachan
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj 211004, India
| | - Rahul
- Department of Surgical Gastroenterology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow 226014, India
| | - Rama Shanker Verma
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj 211004, India.
| | - Girijesh Kumar Patel
- Cancer and Stem Cell Laboratory, Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj 211004, India.
| |
Collapse
|
3
|
Gillespie B, Dunn A, Sundram S, Hill RA. Investigating 7,8-Dihydroxyflavone to combat maternal immune activation effects on offspring gene expression and behaviour. Prog Neuropsychopharmacol Biol Psychiatry 2024; 134:111078. [PMID: 38950841 DOI: 10.1016/j.pnpbp.2024.111078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/03/2024]
Abstract
Infection during pregnancy is a substantial risk factor for the unborn child to develop autism or schizophrenia later in life, and is thought to be driven by maternal immune activation (MIA). MIA can be modelled by exposing pregnant mice to Polyinosinic: polycytidylic acid (Poly-I:C), a viral mimetic that induces an immune response and recapitulates in the offspring many neurochemical features of ASD and schizophrenia, including altered BDNF-TrkB signalling and disruptions to excitatory/inhibitory balance. Therefore, we hypothesised that a BDNF mimetic, 7,8-Dihydroxyflavone (7,8-DHF), administered prophylactically to the dam may prevent the neurobehavioural sequelae of disruptions induced by MIA. Dams were treated with 7,8-DHF in the drinking water (0.08 mg/ML) from gestational day (GD) 9-20 and were exposed to Poly-I:C at GD17 (20 mg/kg, i.p.). Foetal brains were collected 6 h post Poly-I:C exposure for RT-qPCR analysis of BDNF, cytokine, GABAergic and glutamatergic gene targets. A second adult cohort were tested in a battery of behavioural tests relevant to schizophrenia and the prefrontal cortex and ventral hippocampus dissected for RT-qPCR analysis. Foetal brains exposed to Poly-I:C showed increased IL-6, but reduced expression of Ntrk2 and multiple GABAergic and glutamatergic markers. Anxiety-like behaviour was observed in adult offspring prenatally exposed to poly-I:C, which was accompanied by altered expression of Gria2 in the prefrontal cortex and Gria4 in the ventral hippocampus. While 7-8 DHF normalised the expression of some glutamatergic (Grm5) and GABAergic (Gabra1) genes in Poly-I:C exposed offspring, it also led to substantial alterations in offspring not exposed to Poly-I:C. Furthermore, mice exposed to 7,8-DHF prenatally showed increased pre-pulse inhibition and reduced working memory in adulthood. These data advance understanding of how 7,8-DHF and MIA prenatal exposure impacts genes critical to excitatory/inhibitory pathways and related behaviour.
Collapse
Affiliation(s)
- Brendan Gillespie
- Department of Psychiatry, Monash University, Clayton, VIC 3168, Australia
| | - Ariel Dunn
- Department of Psychiatry, Monash University, Clayton, VIC 3168, Australia
| | - Suresh Sundram
- Department of Psychiatry, Monash University, Clayton, VIC 3168, Australia
| | - Rachel A Hill
- Department of Psychiatry, Monash University, Clayton, VIC 3168, Australia.
| |
Collapse
|
4
|
Bodea GO, Botto JM, Ferreiro ME, Sanchez-Luque FJ, de Los Rios Barreda J, Rasmussen J, Rahman MA, Fenlon LR, Jansz N, Gubert C, Gerdes P, Bodea LG, Ajjikuttira P, Da Costa Guevara DJ, Cumner L, Bell CC, Kozulin P, Billon V, Morell S, Kempen MJHC, Love CJ, Saha K, Palmer LM, Ewing AD, Jhaveri DJ, Richardson SR, Hannan AJ, Faulkner GJ. LINE-1 retrotransposons contribute to mouse PV interneuron development. Nat Neurosci 2024; 27:1274-1284. [PMID: 38773348 PMCID: PMC11239520 DOI: 10.1038/s41593-024-01650-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 04/14/2024] [Indexed: 05/23/2024]
Abstract
Retrotransposons are mobile DNA sequences duplicated via transcription and reverse transcription of an RNA intermediate. Cis-regulatory elements encoded by retrotransposons can also promote the transcription of adjacent genes. Somatic LINE-1 (L1) retrotransposon insertions have been detected in mammalian neurons. It is, however, unclear whether L1 sequences are mobile in only some neuronal lineages or therein promote neurodevelopmental gene expression. Here we report programmed L1 activation by SOX6, a transcription factor critical for parvalbumin (PV) interneuron development. Mouse PV interneurons permit L1 mobilization in vitro and in vivo, harbor unmethylated L1 promoters and express full-length L1 mRNAs and proteins. Using nanopore long-read sequencing, we identify unmethylated L1s proximal to PV interneuron genes, including a novel L1 promoter-driven Caps2 transcript isoform that enhances neuron morphological complexity in vitro. These data highlight the contribution made by L1 cis-regulatory elements to PV interneuron development and transcriptome diversity, uncovered due to L1 mobility in this milieu.
Collapse
Affiliation(s)
- Gabriela O Bodea
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia.
- Mater Research Institute - University of Queensland, TRI Building, Woolloongabba, Queensland, Australia.
| | - Juan M Botto
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Maria E Ferreiro
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Francisco J Sanchez-Luque
- Institute of Parasitology and Biomedicine 'López-Neyra', Spanish National Research Council, Granada, Spain
| | | | - Jay Rasmussen
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Muhammed A Rahman
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Laura R Fenlon
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Natasha Jansz
- Mater Research Institute - University of Queensland, TRI Building, Woolloongabba, Queensland, Australia
| | - Carolina Gubert
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Patricia Gerdes
- Mater Research Institute - University of Queensland, TRI Building, Woolloongabba, Queensland, Australia
| | - Liviu-Gabriel Bodea
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Prabha Ajjikuttira
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Darwin J Da Costa Guevara
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia
- Mater Research Institute - University of Queensland, TRI Building, Woolloongabba, Queensland, Australia
| | - Linda Cumner
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Charles C Bell
- Mater Research Institute - University of Queensland, TRI Building, Woolloongabba, Queensland, Australia
| | - Peter Kozulin
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Victor Billon
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia
- Biology Department, École Normale Supérieure Paris-Saclay, Gif-sur-Yvette, France
| | - Santiago Morell
- Mater Research Institute - University of Queensland, TRI Building, Woolloongabba, Queensland, Australia
- Department of Genetics, University of Cambridge, Cambridge, UK
| | - Marie-Jeanne H C Kempen
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - Chloe J Love
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Karabi Saha
- Department of Pharmaceutical Sciences, South Dakota State University, Brookings, SD, USA
| | - Lucy M Palmer
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Adam D Ewing
- Mater Research Institute - University of Queensland, TRI Building, Woolloongabba, Queensland, Australia
| | - Dhanisha J Jhaveri
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia
- Mater Research Institute - University of Queensland, TRI Building, Woolloongabba, Queensland, Australia
| | - Sandra R Richardson
- Mater Research Institute - University of Queensland, TRI Building, Woolloongabba, Queensland, Australia
| | - Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Geoffrey J Faulkner
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia.
- Mater Research Institute - University of Queensland, TRI Building, Woolloongabba, Queensland, Australia.
| |
Collapse
|
5
|
Chapman G, Determan J, Jetter H, Kaushik K, Prakasam R, Kroll KL. Defining cis-regulatory elements and transcription factors that control human cortical interneuron development. iScience 2024; 27:109967. [PMID: 38827400 PMCID: PMC11140214 DOI: 10.1016/j.isci.2024.109967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 03/08/2024] [Accepted: 05/09/2024] [Indexed: 06/04/2024] Open
Abstract
Although human cortical interneurons (cINs) are a minority population in the cerebral cortex, disruption of interneuron development is a frequent contributor to neurodevelopmental disorders. Here, we utilized a model for deriving cINs from human embryonic stem cells to profile chromatin state changes and generate an atlas of cis-regulatory elements (CREs) controlling human cIN development. We used these data to define candidate transcription factors (TFs) that may bind these CREs to regulate interneuron progenitor specification. Among these were RFX3 and RFX4, risk genes for autism spectrum disorder (ASD) with uncharacterized roles in human neuronal development. Using RFX3 and RFX4 knockdown models, we demonstrated new requirements for both genes in interneuron progenitor specification, with RFX3 deficiency causing precocious neuronal differentiation while RFX4 deficiency instead resulted in cessation of progenitor cell proliferation. Together, this work both defined central features of cis-regulatory control and identified new TF requirements for human interneuron development.
Collapse
Affiliation(s)
- Gareth Chapman
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Julianna Determan
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Haley Jetter
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Komal Kaushik
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ramachandran Prakasam
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Kristen L. Kroll
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
6
|
Topchiy I, Mohbat J, Folorunso OO, Wang ZZ, Lazcano-Etchebarne C, Engin E. GABA system as the cause and effect in early development. Neurosci Biobehav Rev 2024; 161:105651. [PMID: 38579901 PMCID: PMC11081854 DOI: 10.1016/j.neubiorev.2024.105651] [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: 01/03/2024] [Revised: 03/05/2024] [Accepted: 04/01/2024] [Indexed: 04/07/2024]
Abstract
GABA is the primary inhibitory neurotransmitter in the adult brain and through its actions on GABAARs, it protects against excitotoxicity and seizure activity, ensures temporal fidelity of neurotransmission, and regulates concerted rhythmic activity of neuronal populations. In the developing brain, the development of GABAergic neurons precedes that of glutamatergic neurons and the GABA system serves as a guide and framework for the development of other brain systems. Despite this early start, the maturation of the GABA system also continues well into the early postnatal period. In this review, we organize evidence around two scenarios based on the essential and protracted nature of GABA system development: 1) disruptions in the development of the GABA system can lead to large scale disruptions in other developmental processes (i.e., GABA as the cause), 2) protracted maturation of this system makes it vulnerable to the effects of developmental insults (i.e., GABA as the effect). While ample evidence supports the importance of GABA/GABAAR system in both scenarios, large gaps in existing knowledge prevent strong mechanistic conclusions.
Collapse
Affiliation(s)
- Irina Topchiy
- Division of Basic Neuroscience, McLean Hospital, Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02215, USA
| | - Julie Mohbat
- Division of Basic Neuroscience, McLean Hospital, Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02215, USA; School of Life Sciences, Ecole Polytechnique Federale de Lausanne, Lausanne CH-1015, Switzerland
| | - Oluwarotimi O Folorunso
- Division of Basic Neuroscience, McLean Hospital, Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02215, USA
| | - Ziyi Zephyr Wang
- Division of Basic Neuroscience, McLean Hospital, Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02215, USA
| | | | - Elif Engin
- Division of Basic Neuroscience, McLean Hospital, Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
7
|
Troppoli TA, Yang C, Katsuki F, Uygun DS, Lin I, Aguilar DD, Spratt T, Basheer R, McNally JM, Savio Chan C, McKenna JT, Brown RE. Neuronal PAS domain 1 identifies a major subpopulation of wakefulness-promoting GABAergic neurons in the basal forebrain. Proc Natl Acad Sci U S A 2024; 121:e2321410121. [PMID: 38748575 PMCID: PMC11127008 DOI: 10.1073/pnas.2321410121] [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: 12/05/2023] [Accepted: 04/11/2024] [Indexed: 05/22/2024] Open
Abstract
Here, we describe a group of basal forebrain (BF) neurons expressing neuronal Per-Arnt-Sim (PAS) domain 1 (Npas1), a developmental transcription factor linked to neuropsychiatric disorders. Immunohistochemical staining in Npas1-cre-2A-TdTomato mice revealed BF Npas1+ neurons are distinct from well-studied parvalbumin or cholinergic neurons. Npas1 staining in GAD67-GFP knock-in mice confirmed that the vast majority of Npas1+ neurons are GABAergic, with minimal colocalization with glutamatergic neurons in vGlut1-cre-tdTomato or vGlut2-cre-tdTomato mice. The density of Npas1+ neurons was high, five to six times that of neighboring cholinergic, parvalbumin, or glutamatergic neurons. Anterograde tracing identified prominent projections of BF Npas1+ neurons to brain regions involved in sleep-wake control, motivated behaviors, and olfaction such as the lateral hypothalamus, lateral habenula, nucleus accumbens shell, ventral tegmental area, and olfactory bulb. Chemogenetic activation of BF Npas1+ neurons in the light period increased the amount of wakefulness and the latency to sleep for 2 to 3 h, due to an increase in long wake bouts and short NREM sleep bouts. NREM slow-wave and sigma power, as well as sleep spindle density, amplitude, and duration, were reduced, reminiscent of findings in several neuropsychiatric disorders. Together with previous findings implicating BF Npas1+ neurons in stress responsiveness, the anatomical projections of BF Npas1+ neurons and the effect of activating them suggest a possible role for BF Npas1+ neurons in motivationally driven wakefulness and stress-induced insomnia. Identification of this major subpopulation of BF GABAergic neurons will facilitate studies of their role in sleep disorders, dementia, and other neuropsychiatric conditions involving BF.
Collapse
Affiliation(s)
- Timothy A. Troppoli
- Department of Psychiatry, Veterans Affairs Boston Healthcare System, Boston, MA02132
- Department of Psychiatry, Harvard Medical School, Boston, MA02115
| | - Chun Yang
- Department of Psychiatry, Veterans Affairs Boston Healthcare System, Boston, MA02132
- Department of Psychiatry, Harvard Medical School, Boston, MA02115
- Boston Veterans Affairs (VA) Research Institute, Boston, MA02130
| | - Fumi Katsuki
- Department of Psychiatry, Veterans Affairs Boston Healthcare System, Boston, MA02132
- Department of Psychiatry, Harvard Medical School, Boston, MA02115
| | - David S. Uygun
- Department of Psychiatry, Veterans Affairs Boston Healthcare System, Boston, MA02132
- Department of Psychiatry, Harvard Medical School, Boston, MA02115
| | | | - David D. Aguilar
- Department of Psychiatry, Veterans Affairs Boston Healthcare System, Boston, MA02132
- Department of Psychiatry, Harvard Medical School, Boston, MA02115
| | - Tristan Spratt
- Department of Psychiatry, Veterans Affairs Boston Healthcare System, Boston, MA02132
- Department of Psychiatry, Harvard Medical School, Boston, MA02115
| | - Radhika Basheer
- Department of Psychiatry, Veterans Affairs Boston Healthcare System, Boston, MA02132
- Department of Psychiatry, Harvard Medical School, Boston, MA02115
- Boston Veterans Affairs (VA) Research Institute, Boston, MA02130
| | - James M. McNally
- Department of Psychiatry, Veterans Affairs Boston Healthcare System, Boston, MA02132
- Department of Psychiatry, Harvard Medical School, Boston, MA02115
- Boston Veterans Affairs (VA) Research Institute, Boston, MA02130
| | - C. Savio Chan
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL60611
| | - James T. McKenna
- Department of Psychiatry, Veterans Affairs Boston Healthcare System, Boston, MA02132
- Department of Psychiatry, Harvard Medical School, Boston, MA02115
- Boston Veterans Affairs (VA) Research Institute, Boston, MA02130
| | - Ritchie E. Brown
- Department of Psychiatry, Veterans Affairs Boston Healthcare System, Boston, MA02132
- Department of Psychiatry, Harvard Medical School, Boston, MA02115
- Boston Veterans Affairs (VA) Research Institute, Boston, MA02130
| |
Collapse
|
8
|
Ozkan A, Padmanabhan HK, Shipman SL, Azim E, Kumar P, Sadegh C, Basak AN, Macklis JD. Directed differentiation of functional corticospinal-like neurons from endogenous SOX6+/NG2+ cortical progenitors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.21.590488. [PMID: 38712174 PMCID: PMC11071355 DOI: 10.1101/2024.04.21.590488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Corticospinal neurons (CSN) centrally degenerate in amyotrophic lateral sclerosis (ALS), along with spinal motor neurons, and loss of voluntary motor function in spinal cord injury (SCI) results from damage to CSN axons. For functional regeneration of specifically affected neuronal circuitry in vivo , or for optimally informative disease modeling and/or therapeutic screening in vitro , it is important to reproduce the type or subtype of neurons involved. No such appropriate in vitro models exist with which to investigate CSN selective vulnerability and degeneration in ALS, or to investigate routes to regeneration of CSN circuitry for ALS or SCI, critically limiting the relevance of much research. Here, we identify that the HMG-domain transcription factor Sox6 is expressed by a subset of NG2+ endogenous cortical progenitors in postnatal and adult cortex, and that Sox6 suppresses a latent neurogenic program by repressing inappropriate proneural Neurog2 expression by progenitors. We FACS-purify these genetically accessible progenitors from postnatal mouse cortex and establish a pure culture system to investigate their potential for directed differentiation into CSN. We then employ a multi-component construct with complementary and differentiation-sharpening transcriptional controls (activating Neurog2, Fezf2 , while antagonizing Olig2 with VP16:Olig2 ). We generate corticospinal-like neurons from SOX6+/NG2+ cortical progenitors, and find that these neurons differentiate with remarkable fidelity compared with corticospinal neurons in vivo . They possess appropriate morphological, molecular, transcriptomic, and electrophysiological characteristics, without characteristics of the alternate intracortical or other neuronal subtypes. We identify that these critical specifics of differentiation are not reproduced by commonly employed Neurog2 -driven differentiation. Neurons induced by Neurog2 instead exhibit aberrant multi-axon morphology and express molecular hallmarks of alternate cortical projection subtypes, often in mixed form. Together, this developmentally-based directed differentiation from genetically accessible cortical progenitors sets a precedent and foundation for in vitro mechanistic and therapeutic disease modeling, and toward regenerative neuronal repopulation and circuit repair.
Collapse
|
9
|
Miyoshi G, Ueta Y, Yagasaki Y, Kishi Y, Fishell G, Machold RP, Miyata M. Developmental trajectories of GABAergic cortical interneurons are sequentially modulated by dynamic FoxG1 expression levels. Proc Natl Acad Sci U S A 2024; 121:e2317783121. [PMID: 38588430 PMCID: PMC11032493 DOI: 10.1073/pnas.2317783121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 03/04/2024] [Indexed: 04/10/2024] Open
Abstract
GABAergic inhibitory interneurons, originating from the embryonic ventral forebrain territories, traverse a convoluted migratory path to reach the neocortex. These interneuron precursors undergo sequential phases of tangential and radial migration before settling into specific laminae during differentiation. Here, we show that the developmental trajectory of FoxG1 expression is dynamically controlled in these interneuron precursors at critical junctures of migration. By utilizing mouse genetic strategies, we elucidate the pivotal role of precise changes in FoxG1 expression levels during interneuron specification and migration. Our findings underscore the gene dosage-dependent function of FoxG1, aligning with clinical observations of FOXG1 haploinsufficiency and duplication in syndromic forms of autism spectrum disorders. In conclusion, our results reveal the finely tuned developmental clock governing cortical interneuron development, driven by temporal dynamics and the dose-dependent actions of FoxG1.
Collapse
Affiliation(s)
- Goichi Miyoshi
- Department of Developmental Genetics and Behavioral Neuroscience, Gunma University Graduate School of Medicine, Maebashi city, Gunma371-8511, Japan
- Department of Neurophysiology, Tokyo Women’s Medical University, Shinjuku, Tokyo162-8666, Japan
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, New York, NY10016
| | - Yoshifumi Ueta
- Department of Neurophysiology, Tokyo Women’s Medical University, Shinjuku, Tokyo162-8666, Japan
| | - Yuki Yagasaki
- Department of Neurophysiology, Tokyo Women’s Medical University, Shinjuku, Tokyo162-8666, Japan
| | - Yusuke Kishi
- Laboratory of Molecular Neurobiology, Institute for Quantitative Biosciences, University of Tokyo, Bunkyo, Tokyo113-0032, Japan
- Laboratory of Molecular Biology, Graduate School of Pharmaceutical Sciences, University of Tokyo, Bunkyo, Tokyo113-0033, Japan
| | - Gord Fishell
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, New York, NY10016
- Department of Neurobiology, Blavatnik Institute, Harvard Medical School, Boston, MA02115
- Stanley Center at the Broad Institute, Cambridge, MA02142
| | - Robert P. Machold
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, New York, NY10016
| | - Mariko Miyata
- Department of Neurophysiology, Tokyo Women’s Medical University, Shinjuku, Tokyo162-8666, Japan
| |
Collapse
|
10
|
Liu Y, Yuan J, Dong Y, Jiang S, Zhang M, Zhao X. Interaction between Oligodendrocytes and Interneurons in Brain Development and Related Neuropsychiatric Disorders. Int J Mol Sci 2024; 25:3620. [PMID: 38612430 PMCID: PMC11011273 DOI: 10.3390/ijms25073620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 03/17/2024] [Accepted: 03/22/2024] [Indexed: 04/14/2024] Open
Abstract
A variety of neurological and psychiatric disorders have recently been shown to be highly associated with the abnormal development and function of oligodendrocytes (OLs) and interneurons. OLs are the myelin-forming cells in the central nervous system (CNS), while interneurons are important neural types gating the function of excitatory neurons. These two types of cells are of great significance for the establishment and function of neural circuits, and they share similar developmental origins and transcriptional architectures, and interact with each other in multiple ways during development. In this review, we compare the similarities and differences in these two cell types, providing an important reference and further revealing the pathogenesis of related brain disorders.
Collapse
Affiliation(s)
| | | | | | | | | | - Xianghui Zhao
- Department of Neuroscience, Air Force Medical University, Xi’an 710032, China
| |
Collapse
|
11
|
Puvogel S, Alsema A, North HF, Webster MJ, Weickert CS, Eggen BJL. Single-Nucleus RNA-Seq Characterizes the Cell Types Along the Neuronal Lineage in the Adult Human Subependymal Zone and Reveals Reduced Oligodendrocyte Progenitor Abundance with Age. eNeuro 2024; 11:ENEURO.0246-23.2024. [PMID: 38351133 PMCID: PMC10913050 DOI: 10.1523/eneuro.0246-23.2024] [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: 07/12/2023] [Revised: 01/15/2024] [Accepted: 01/23/2024] [Indexed: 03/06/2024] Open
Abstract
The subependymal zone (SEZ), also known as the subventricular zone (SVZ), constitutes a neurogenic niche that persists during postnatal life. In humans, the neurogenic potential of the SEZ declines after the first year of life. However, studies discovering markers of stem and progenitor cells highlight the neurogenic capacity of progenitors in the adult human SEZ, with increased neurogenic activity occurring under pathological conditions. In the present study, the complete cellular niche of the adult human SEZ was characterized by single-nucleus RNA sequencing, and compared between four youth (age 16-22) and four middle-aged adults (age 44-53). We identified 11 cellular clusters including clusters expressing marker genes for neural stem cells (NSCs), neuroblasts, immature neurons, and oligodendrocyte progenitor cells. The relative abundance of NSC and neuroblast clusters did not differ between the two age groups, indicating that the pool of SEZ NSCs does not decline in this age range. The relative abundance of oligodendrocyte progenitors and microglia decreased in middle-age, indicating that the cellular composition of human SEZ is remodeled between youth and adulthood. The expression of genes related to nervous system development was higher across different cell types, including NSCs, in youth as compared with middle-age. These transcriptional changes suggest ongoing central nervous system plasticity in the SEZ in youth, which declined in middle-age.
Collapse
Affiliation(s)
- Sofía Puvogel
- Section Molecular Neurobiology, Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, Groningen 9700 AD, The Netherlands
- Department of Human Genetics, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Nijmegen 6500 HB, The Netherlands
| | - Astrid Alsema
- Section Molecular Neurobiology, Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, Groningen 9700 AD, The Netherlands
| | - Hayley F North
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Sydney, New South Wales 2031, Australia
- School of Psychiatry, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Maree J Webster
- Laboratory of Brain Research, Stanley Medical Research Institute, Rockville 20850, Maryland
| | - Cynthia Shannon Weickert
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Sydney, New South Wales 2031, Australia
- School of Psychiatry, University of New South Wales, Sydney, New South Wales 2052, Australia
- Department of Neuroscience and Physiology, Upstate Medical University, Syracuse, New York 13201
| | - Bart J L Eggen
- Section Molecular Neurobiology, Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, Groningen 9700 AD, The Netherlands
| |
Collapse
|
12
|
Yakovchik AY, Tolynyova DV, Kashtanova DA, Sutulova ER, Ivanov MV, Mamchur AA, Erema VV, Matkava LR, Terekhov MV, Rumyantseva AM, Blinova OI, Akinshina AI, Mitrofanov SI, Yudin VS, Makarov VV, Keskinov AА, Kraevoy SA, Yudin SM. Genetics of psycho-emotional well-being: genome-wide association study and polygenic risk score analysis. Front Psychiatry 2024; 14:1188427. [PMID: 38328521 PMCID: PMC10847277 DOI: 10.3389/fpsyt.2023.1188427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 07/28/2023] [Indexed: 02/09/2024] Open
Abstract
Background Psycho-emotional well-being is essential for living a life of satisfaction and fulfillment. However, depression and anxiety have become the leading mental health issues worldwide, according to the World Health Organization. Both disorders have been linked to stress and other psychological factors. Their genetic basis remains understudied. Methods In 2020-2021, the psycho-emotional well-being of 30,063 Russians with no known psychiatric history was assessed using the Hospital Anxiety and Depression Scale (HADS) for general mental health and the HADS subscale A (anxiety) for anxiety. Following the original instructions, an anxiety score of ≥11 points was used as the anxiety threshold. A genome-wide association study was performed to find associations between anxiety and HADS/HADS-A scores using linear and logistic regressions based on HADS/HADS-A scores as binary and continuous variables, respectively. In addition, the links between anxiety, sociodemographic factors (such as age, sex, and employment), lifestyle (such as physical activity, sleep duration, and smoking), and markers of caffeine and alcohol metabolism were analyzed. To assess the risk of anxiety, polygenic risk score modeling was carried out using open-access software and principal component analysis (PCA) to simplify the calculations (ROC AUC = 89.4 ± 2.2% on the test set). Results There was a strong positive association between HADS/HADS-A scores and sociodemographic factors and lifestyle. New single-nucleotide polymorphisms (SNPs) with genome-wide significance were discovered, which had not been associated with anxiety or other stress-related conditions but were located in genes previously associated with bipolar disorder, schizophrenia, or emotional instability. The CACNA1C variant rs1205787230 was associated with clinical anxiety (a HADS-A score of ≥11 points). There was an association between anxiety levels (HADS-A scores) and genes involved in the activity of excitatory neurotransmitters: PTPRN2 (rs3857647), DLGAP4 (rs8114927), and STK24 (rs9517326). Conclusion Our results suggest that calcium channels and monoamine neurotransmitters, as well as SNPs in genes directly or indirectly affecting neurogenesis and synaptic functions, may be involved in the development of increased anxiety. The role of some non-genetic factors and the clinical significance of physiological markers such as lifestyle were also demonstrated.
Collapse
Affiliation(s)
- Anna Yurievna Yakovchik
- Federal State Budgetary Institution Centre for Strategic Planning and Management of Biomedical Health Risks of the Federal Medical Biological Agency, Moscow, Russia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Pavon N, Diep K, Yang F, Sebastian R, Martinez-Martin B, Ranjan R, Sun Y, Pak C. Patterning ganglionic eminences in developing human brain organoids using a morphogen-gradient-inducing device. CELL REPORTS METHODS 2024; 4:100689. [PMID: 38228151 PMCID: PMC10831957 DOI: 10.1016/j.crmeth.2023.100689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 10/21/2023] [Accepted: 12/18/2023] [Indexed: 01/18/2024]
Abstract
In early neurodevelopment, the central nervous system is established through the coordination of various neural organizers directing tissue patterning and cell differentiation. Better recapitulation of morphogen gradient production and signaling will be crucial for establishing improved developmental models of the brain in vitro. Here, we developed a method by assembling polydimethylsiloxane devices capable of generating a sustained chemical gradient to produce patterned brain organoids, which we termed morphogen-gradient-induced brain organoids (MIBOs). At 3.5 weeks, MIBOs replicated dorsal-ventral patterning observed in the ganglionic eminences (GE). Analysis of mature MIBOs through single-cell RNA sequencing revealed distinct dorsal GE-derived CALB2+ interneurons, medium spiny neurons, and medial GE-derived cell types. Finally, we demonstrate long-term culturing capabilities with MIBOs maintaining stable neural activity in cultures grown up to 5.5 months. MIBOs demonstrate a versatile approach for generating spatially patterned brain organoids for embryonic development and disease modeling.
Collapse
Affiliation(s)
- Narciso Pavon
- Graduate Program in Neuroscience and Behavior, UMass Amherst, Amherst, MA 01003, USA; Department of Biochemistry and Molecular Biology, UMass Amherst, Amherst, MA 01003, USA
| | - Karmen Diep
- Department of Biochemistry and Molecular Biology, UMass Amherst, Amherst, MA 01003, USA
| | - Feiyu Yang
- Department of Mechanical and Industrial Engineering, UMass Amherst, Amherst, MA 01003, USA
| | - Rebecca Sebastian
- Graduate Program in Neuroscience and Behavior, UMass Amherst, Amherst, MA 01003, USA; Department of Biochemistry and Molecular Biology, UMass Amherst, Amherst, MA 01003, USA
| | - Beatriz Martinez-Martin
- Department of Biochemistry and Molecular Biology, UMass Amherst, Amherst, MA 01003, USA; Graduate Program in Molecular and Cellular Biology, UMass Amherst, Amherst, MA 01003, USA
| | - Ravi Ranjan
- Genomics Core, Institute of Applied Life Sciences, UMass Amherst, Amherst, MA 01003, USA
| | - Yubing Sun
- Department of Mechanical and Industrial Engineering, UMass Amherst, Amherst, MA 01003, USA.
| | - ChangHui Pak
- Department of Biochemistry and Molecular Biology, UMass Amherst, Amherst, MA 01003, USA.
| |
Collapse
|
14
|
Munguba H, Nikouei K, Hochgerner H, Oberst P, Kouznetsova A, Ryge J, Muñoz-Manchado AB, Close J, Batista-Brito R, Linnarsson S, Hjerling-Leffler J. Transcriptional maintenance of cortical somatostatin interneuron subtype identity during migration. Neuron 2023; 111:3590-3603.e5. [PMID: 37625400 DOI: 10.1016/j.neuron.2023.07.018] [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: 03/29/2019] [Revised: 06/08/2023] [Accepted: 07/27/2023] [Indexed: 08/27/2023]
Abstract
Although cardinal cortical interneuron identity is established upon cell-cycle exit, it remains unclear whether specific interneuron subtypes are pre-established, and if so, how their identity is maintained prior to circuit integration. We conditionally removed Sox6 (Sox6-cKO) in migrating somatostatin (Sst+) interneurons and assessed the effects on their mature identity. In adolescent mice, five of eight molecular Sst+ subtypes were nearly absent in the Sox6-cKO cortex without a reduction in cell number. Sox6-cKO cells displayed electrophysiological maturity and expressed genes enriched within the broad class of Sst+ interneurons. Furthermore, we could infer subtype identity prior to cortical integration (embryonic day 18.5), suggesting that the loss in subtype was due to disrupted subtype maintenance. Conversely, Sox6 removal at postnatal day 7 did not disrupt marker expression in the mature cortex. Therefore, Sox6 is necessary during migration for maintenance of Sst+ subtype identity, indicating that subtype maintenance requires active transcriptional programs.
Collapse
Affiliation(s)
- Hermany Munguba
- Laboratory of Molecular Neurobiology, Department Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Kasra Nikouei
- Laboratory of Molecular Neurobiology, Department Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Hannah Hochgerner
- Laboratory of Molecular Neurobiology, Department Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Polina Oberst
- Laboratory of Molecular Neurobiology, Department Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Alexandra Kouznetsova
- Laboratory of Molecular Neurobiology, Department Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Jesper Ryge
- Brain Mind Institute, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Ana Belén Muñoz-Manchado
- Laboratory of Molecular Neurobiology, Department Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden; Departamento de Anatomía Patológica, Biología Celular, Histología, Historia de la Ciencia, Medicina Legal y Forense y Toxicología, Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), Universidad de Cádiz, Cádiz, Spain
| | - Jennie Close
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Renata Batista-Brito
- Einstein College of Medicine, Dominick Purpura Department of Neuroscience, 1300 Morris Park Ave, The Bronx, NY 10461, USA; Einstein College of Medicine, Department of Psychiatry and Behavioral Sciences, 1300 Morris Park Ave, The Bronx, NY 10461, USA; Einstein College of Medicine, Department of Genetics, 1300 Morris Park Ave, The Bronx, NY 10461, USA
| | - Sten Linnarsson
- Laboratory of Molecular Neurobiology, Department Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Jens Hjerling-Leffler
- Laboratory of Molecular Neurobiology, Department Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
15
|
Troppoli TA, Yang C, Katsuki F, Uygun DS, Lin I, Aguilar D, Spratt T, Basheer R, McNally JM, Chan CS, McKenna JT, Brown RE. Neuronal PAS domain 1 identifies a major subpopulation of wakefulness-promoting GABAergic neurons in basal forebrain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.09.566065. [PMID: 37986953 PMCID: PMC10659409 DOI: 10.1101/2023.11.09.566065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Here we describe a novel group of basal forebrain (BF) neurons expressing neuronal PAS domain 1 (Npas1), a developmental transcription factor linked to neuropsychiatric disorders. Immunohistochemical staining in Npas1-cre-2A-TdTomato mice revealed BF Npas1 + neurons are distinct from well-studied parvalbumin or cholinergic neurons. Npas1 staining in GAD67-GFP knock-in mice confirmed that the vast majority of Npas1 + neurons are GABAergic, with minimal colocalization with glutamatergic neurons in vGlut1-cre-tdTomato or vGlut2-cre-tdTomato mice. The density of Npas1 + neurons was high, 5-6 times that of neighboring cholinergic, parvalbumin or glutamatergic neurons. Anterograde tracing identified prominent projections of BF Npas1 + neurons to brain regions involved in sleep-wake control, motivated behaviors and olfaction such as the lateral hypothalamus, lateral habenula, nucleus accumbens shell, ventral tegmental area and olfactory bulb. Chemogenetic activation of BF Npas1 + neurons in the light (inactive) period increased the amount of wakefulness and the latency to sleep for 2-3 hr, due to an increase in long wake bouts and short NREM sleep bouts. Non-REM slow-wave (0-1.5 Hz) and sigma (9-15 Hz) power, as well as sleep spindle density, amplitude and duration, were reduced, reminiscent of findings in several neuropsychiatric disorders. Together with previous findings implicating BF Npas1 + neurons in stress responsiveness, the anatomical projections of BF Npas1 + neurons and the effect of activating them suggest a possible role for BF Npas1 + neurons in motivationally-driven wakefulness and stress-induced insomnia. Identification of this major subpopulation of BF GABAergic neurons will facilitate studies of their role in sleep disorders, dementia and other neuropsychiatric conditions involving BF. SIGNIFICANCE STATEMENT We characterize a group of basal forebrain (BF) neurons in the mouse expressing neuronal PAS domain 1 (Npas1), a developmental transcription factor linked to neuropsychiatric disorders. BF Npas1 + neurons are a major subset of GABAergic neurons distinct and more numerous than cholinergic, parvalbumin or glutamate neurons. BF Npas1 + neurons target brain areas involved in arousal, motivation and olfaction. Activation of BF Npas1 + neurons in the light (inactive) period increased wakefulness and the latency to sleep due to increased long wake bouts. Non-REM sleep slow waves and spindles were reduced reminiscent of findings in several neuropsychiatric disorders. Identification of this major subpopulation of BF GABAergic wake-promoting neurons will allow studies of their role in insomnia, dementia and other conditions involving BF.
Collapse
|
16
|
Bechelli L, Tomasella E, Cardoso SL, Belmonte M, Gelman DM. Selective dopamine D2 receptor deletion from Nkx6.2 expressing cells causes impaired cognitive, motivation and anxiety phenotypes in mice. Sci Rep 2023; 13:19473. [PMID: 37945756 PMCID: PMC10636105 DOI: 10.1038/s41598-023-46954-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 11/07/2023] [Indexed: 11/12/2023] Open
Abstract
Abnormal dopamine neurotransmission is a common trait of some psychiatric diseases, like schizophrenia or bipolar disorder. Excessive dopaminergic tone in subcortical brain regions is associated with psychotic episodes, while reduced prefrontal dopaminergic activity is associated with impaired cognitive performance and reduced motivation, among other symptoms. Inhibitory interneurons expressing the calcium binding protein parvalbumin are particularly affected in both schizophrenia and bipolar disorder, as they set a fine-tuned physiological inhibitory/excitatory balance. Parvalbumin and somatostatin interneuron subtypes, are born from the medial ganglionic eminence and require the sequential expression of specific transcription factors for their specification, such as Nkx6.2. Here, we aimed at characterizing in detail interneuron subtypes derived from Nkx6.2 expressing progenitors by the generation of an Nkx6.2 Cre transgenic mouse line. We show that Nkx6.2 specifies over a third part of the total population of cortical somatostatin interneurons, preferentially at early developmental time points, whereas at late developmental stages, Nkx6.2 expressing progenitors shift to parvalbumin interneuron specification. Dopamine D2 receptor deletion from Nkx6.2 expressing progenitors causes abnormal phenotypes restricted to cognitive, motivation and anxiety domains. Our results show that Nkx6.2 have the potential to specify both somatostatin and parvalbumin interneurons in an opposite timed program and that DRD2 expression is required in Nkx6.2 expressing progenitors to avoid impaired phenotypes commonly associated to the pathophysiology of psychiatric diseases.
Collapse
Affiliation(s)
- Lucila Bechelli
- Instituto de Biología y Medicina Experimental (IBYME). Fundación IBYME., Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)., Vuelta de Obligado 2490, C1428ADN, Ciudad Autónoma de Buenos Aires, Argentina
| | - Eugenia Tomasella
- Instituto de Biología y Medicina Experimental (IBYME). Fundación IBYME., Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)., Vuelta de Obligado 2490, C1428ADN, Ciudad Autónoma de Buenos Aires, Argentina
| | - Sofia Lopez Cardoso
- Instituto de Biología y Medicina Experimental (IBYME). Fundación IBYME., Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)., Vuelta de Obligado 2490, C1428ADN, Ciudad Autónoma de Buenos Aires, Argentina
| | - Martina Belmonte
- Instituto de Biología y Medicina Experimental (IBYME). Fundación IBYME., Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)., Vuelta de Obligado 2490, C1428ADN, Ciudad Autónoma de Buenos Aires, Argentina
| | - Diego M Gelman
- Instituto de Biología y Medicina Experimental (IBYME). Fundación IBYME., Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)., Vuelta de Obligado 2490, C1428ADN, Ciudad Autónoma de Buenos Aires, Argentina.
- Universidad Argentina de la Empresa (UADE), Lima 757, C1073AAO, Ciudad Autónoma de Buenos Aires, Argentina.
| |
Collapse
|
17
|
Krokidis MG, Vrahatis AG, Lazaros K, Skolariki K, Exarchos TP, Vlamos P. Machine Learning Analysis of Alzheimer's Disease Single-Cell RNA-Sequencing Data across Cortex and Hippocampus Regions. Curr Issues Mol Biol 2023; 45:8652-8669. [PMID: 37998721 PMCID: PMC10670182 DOI: 10.3390/cimb45110544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 10/15/2023] [Accepted: 10/26/2023] [Indexed: 11/25/2023] Open
Abstract
Advancements in molecular biology have revolutionized our understanding of complex diseases, with Alzheimer's disease being a prime example. Single-cell sequencing, currently the most suitable technology, facilitates profoundly detailed disease analysis at the cellular level. Prior research has established that the pathology of Alzheimer's disease varies across different brain regions and cell types. In parallel, only machine learning has the capacity to address the myriad challenges presented by such studies, where the integration of large-scale data and numerous experiments is required to extract meaningful knowledge. Our methodology utilizes single-cell RNA sequencing data from healthy and Alzheimer's disease (AD) samples, focused on the cortex and hippocampus regions in mice. We designed three distinct case studies and implemented an ensemble feature selection approach through machine learning, also performing an analysis of distinct age-related datasets to unravel age-specific effects, showing differential gene expression patterns within each condition. Important evidence was reported, such as enrichment in central nervous system development and regulation of oligodendrocyte differentiation between the hippocampus and cortex of 6-month-old AD mice as well as regulation of epinephrine secretion and dendritic spine morphogenesis in 15-month-old AD mice. Our outcomes from all three of our case studies illustrate the capacity of machine learning strategies when applied to single-cell data, revealing critical insights into Alzheimer's disease.
Collapse
Affiliation(s)
- Marios G. Krokidis
- Bioinformatics and Human Electrophysiology Laboratory, Department of Informatics, Ionian University, 49100 Corfu, Greece; (A.G.V.); (K.L.); (K.S.); (T.P.E.); (P.V.)
| | | | | | | | | | | |
Collapse
|
18
|
Pai ELL, Stafford AM, Vogt D. Cellular signaling impacts upon GABAergic cortical interneuron development. Front Neurosci 2023; 17:1138653. [PMID: 36998738 PMCID: PMC10043199 DOI: 10.3389/fnins.2023.1138653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 02/28/2023] [Indexed: 03/18/2023] Open
Abstract
The development and maturation of cortical GABAergic interneurons has been extensively studied, with much focus on nuclear regulation via transcription factors. While these seminal events are critical for the establishment of interneuron developmental milestones, recent studies on cellular signaling cascades have begun to elucidate some potential contributions of cell signaling during development. Here, we review studies underlying three broad signaling families, mTOR, MAPK, and Wnt/beta-catenin in cortical interneuron development. Notably, each pathway harbors signaling factors that regulate a breadth of interneuron developmental milestones and properties. Together, these events may work in conjunction with transcriptional mechanisms and other events to direct the complex diversity that emerges during cortical interneuron development and maturation.
Collapse
Affiliation(s)
- Emily Ling-Lin Pai
- Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| | - April M. Stafford
- Department of Pediatrics and Human Development, Michigan State University, Grand Rapids, MI, United States
| | - Daniel Vogt
- Department of Pediatrics and Human Development, Michigan State University, Grand Rapids, MI, United States
- Neuroscience Program, Michigan State University, East Lansing, MI, United States
- *Correspondence: Daniel Vogt,
| |
Collapse
|
19
|
Zeng H, Chen YX. MiR-19b-3p Inhibits Hypoxia-Ischemia Encephalopathy by Inhibiting SOX6 Expression via Activating Wnt/β-catenin Pathway. Neurochem Res 2023; 48:874-884. [PMID: 36369428 DOI: 10.1007/s11064-022-03812-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Hypoxic-ischemic encephalopathy (HIE) is a detrimental factor in infant death and chronic disease. The specific pathogenesis is not entirely clear. Therefore, exploring the pathogenesis of HIE is critical. The expression of miR-19b-3p and SOX6 in umbilical blood of HIE patients was detected by qRT-PCR assay. HT22 cells were triggered with oxygen-glucose deprivation/reoxygenation (OGD/R) to construct the HIE cell model. Cell Counting Kit-8 (CCK-8) assay was used to estimate viability. SOD and MDA levels were detected by enzyme linked immunosorbent assay. Flow cytometry was implemented to ascertain neurocyte apoptosis. Cellular β-catenin immunofluorescence staining was used to detect the expression and distribution of β-catenin protein. Wnt signaling pathway activation was detected by TOPFlash/FOPFlash luciferase reporter assay. The targeting correlation of SOX6 and miR-19b-3p was corroborated by dual-luciferase reporter gene assay and RNA pull-down assay. MiR-19b-3p expression was once down-regulated, whilst SOX6 expression was up-regulated in HIE patients. MiR-19b-3p overexpression promoted cell proliferation, repressed cell apoptosis, oxidative stress response, and Wnt/β-catenin pathway activation in OGD/R-triggered HT22 cells. MiR-19b-3p negatively regulated SOX6 expression. SOX6 knockdown improved OGD/R-triggered HT22 cells injury via Wnt/β-catenin pathway activation. MiR-19b-3p overexpression suppressed OGD/R-triggered HT22 cell injury via inhibiting SOX6 expression via activating Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Hao Zeng
- Department of Neurosurgery, Shenzhen Longhua District Central Hospital, Shenzhen, 518110, Guangdong, People's Republic of China
| | - Yu-Xia Chen
- Department of Neontal Development, Shenzhen Longhua District Central Hospital, No. 187, Guanlan Avenue, Longhua District, Shenzhen, 518110, Guangdong, People's Republic of China.
| |
Collapse
|
20
|
Tan Z, Li W, Cheng X, Zhu Q, Zhang X. Non-Coding RNAs in the Regulation of Hippocampal Neurogenesis and Potential Treatment Targets for Related Disorders. Biomolecules 2022; 13:biom13010018. [PMID: 36671403 PMCID: PMC9855933 DOI: 10.3390/biom13010018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/17/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Non-coding RNAs (ncRNAs), including miRNAs, lncRNAs, circRNAs, and piRNAs, do not encode proteins. Nonetheless, they have critical roles in a variety of cellular activities-such as development, neurogenesis, degeneration, and the response to injury to the nervous system-via protein translation, RNA splicing, gene activation, silencing, modifications, and editing; thus, they may serve as potential targets for disease treatment. The activity of adult neural stem cells (NSCs) in the subgranular zone of the hippocampal dentate gyrus critically influences hippocampal function, including learning, memory, and emotion. ncRNAs have been shown to be involved in the regulation of hippocampal neurogenesis, including proliferation, differentiation, and migration of NSCs and synapse formation. The interaction among ncRNAs is complex and diverse and has become a major topic within the life science. This review outlines advances in research on the roles of ncRNAs in modulating NSC bioactivity in the hippocampus and discusses their potential applications in the treatment of illnesses affecting the hippocampus.
Collapse
Affiliation(s)
- Zhengye Tan
- Department of Anatomy, Institute of Neurobiology, Medical School, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Wen Li
- Department of Anatomy, Institute of Neurobiology, Medical School, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Xiang Cheng
- Department of Anatomy, Institute of Neurobiology, Medical School, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Qing Zhu
- School of Pharmacy, Nantong University, Nantong 226001, China
- Key Laboratory of Inflammation and Molecular Drug Target of Jiangsu Province, Nantong 226001, China
| | - Xinhua Zhang
- Department of Anatomy, Institute of Neurobiology, Medical School, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
- Central Lab, Yancheng Third People’s Hospital, The Sixth Affiliated Hospital of Nantong University, Yancheng 224001, China
- Correspondence:
| |
Collapse
|
21
|
Algorithmic reconstruction of glioblastoma network complexity. iScience 2022; 25:104179. [PMID: 35479408 PMCID: PMC9036113 DOI: 10.1016/j.isci.2022.104179] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/16/2022] [Accepted: 03/24/2022] [Indexed: 12/16/2022] Open
Abstract
Glioblastoma is a complex disease that is difficult to treat. Network and data science offer alternative approaches to classical bioinformatics pipelines to study gene expression patterns from single-cell datasets, helping to distinguish genes associated with the control of differentiation and aggression. To identify the key molecular regulators of the networks driving glioblastoma/GSC and predict their cell fate dynamics, we applied a host of data theoretic techniques to gene expression patterns from pediatric and adult glioblastoma, and adult glioma-derived stem cells (GSCs). We identified eight transcription factors (OLIG1/2, TAZ, GATA2, FOXG1, SOX6, SATB2, and YY1) and four signaling genes (ATL3, MTSS1, EMP1, and TPT1) as coordinators of cell state transitions and, thus, clinically targetable putative factors differentiating pediatric and adult glioblastomas from adult GSCs. Our study provides strong evidence of complex systems approaches for inferring complex dynamics from reverse-engineering gene networks, bolstering the search for new clinically relevant targets in glioblastoma. Complex cell fate attractors capture glioblastoma differentiation dynamics Graph theoretic approaches decode master regulators of GBM glioblastoma cell fate decisions Network dynamics of pediatric glioblastoma resemble adult GSCs Transcriptional networks may help reprogram glioblastoma behavioral patterns
Collapse
|
22
|
Donega V, van der Geest AT, Sluijs JA, van Dijk RE, Wang CC, Basak O, Pasterkamp RJ, Hol EM. Single-cell profiling of human subventricular zone progenitors identifies SFRP1 as a target to re-activate progenitors. Nat Commun 2022; 13:1036. [PMID: 35210419 PMCID: PMC8873234 DOI: 10.1038/s41467-022-28626-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 01/28/2022] [Indexed: 12/13/2022] Open
Abstract
Following the decline of neurogenesis at birth, progenitors of the subventricular zone (SVZ) remain mostly in a quiescent state in the adult human brain. The mechanisms that regulate this quiescent state are still unclear. Here, we isolate CD271+ progenitors from the aged human SVZ for single-cell RNA sequencing analysis. Our transcriptome data reveal the identity of progenitors of the aged human SVZ as late oligodendrocyte progenitor cells. We identify the Wnt pathway antagonist SFRP1 as a possible signal that promotes quiescence of progenitors from the aged human SVZ. Administration of WAY-316606, a small molecule that inhibits SFRP1 function, stimulates activation of neural stem cells both in vitro and in vivo under homeostatic conditions. Our data unravel a possible mechanism through which progenitors of the adult human SVZ are maintained in a quiescent state and a potential target for stimulating progenitors to re-activate. The decline in neurogenesis following birth is accompanied with a quiescent state characteristic of neural progenitors of the adult brain. Here, the authors identify the Wnt pathway antagonist SFRP1 as a potential signal that promotes quiescence and show that its inhibition stimulates stem cell activation.
Collapse
Affiliation(s)
- Vanessa Donega
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands.
| | - Astrid T van der Geest
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Jacqueline A Sluijs
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Roland E van Dijk
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Chi Chiu Wang
- Department of Obstetrics and Gynecology, Li Ka Shing Institute of Health Sciences, School of Biomedical Sciences, and Chinese University of Hong Kong -Sichuan University Joint Laboratory in Reproductive Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong.,Institute of Biochemistry, Charite-University Medicine Berlin, Charitéplatz 1, Berlin, Germany
| | - Onur Basak
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Elly M Hol
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands.
| |
Collapse
|
23
|
Individual human cortical progenitors can produce excitatory and inhibitory neurons. Nature 2022; 601:397-403. [PMID: 34912114 PMCID: PMC8994470 DOI: 10.1038/s41586-021-04230-7] [Citation(s) in RCA: 104] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 11/10/2021] [Indexed: 01/19/2023]
Abstract
The cerebral cortex is a cellularly complex structure comprising a rich diversity of neuronal and glial cell types. Cortical neurons can be broadly categorized into two classes-excitatory neurons that use the neurotransmitter glutamate, and inhibitory interneurons that use γ-aminobutyric acid (GABA). Previous developmental studies in rodents have led to a prevailing model in which excitatory neurons are born from progenitors located in the cortex, whereas cortical interneurons are born from a separate population of progenitors located outside the developing cortex in the ganglionic eminences1-5. However, the developmental potential of human cortical progenitors has not been thoroughly explored. Here we show that, in addition to excitatory neurons and glia, human cortical progenitors are also capable of producing GABAergic neurons with the transcriptional characteristics and morphologies of cortical interneurons. By developing a cellular barcoding tool called 'single-cell-RNA-sequencing-compatible tracer for identifying clonal relationships' (STICR), we were able to carry out clonal lineage tracing of 1,912 primary human cortical progenitors from six specimens, and to capture both the transcriptional identities and the clonal relationships of their progeny. A subpopulation of cortically born GABAergic neurons was transcriptionally similar to cortical interneurons born from the caudal ganglionic eminence, and these cells were frequently related to excitatory neurons and glia. Our results show that individual human cortical progenitors can generate both excitatory neurons and cortical interneurons, providing a new framework for understanding the origins of neuronal diversity in the human cortex.
Collapse
|
24
|
Shi Y, Wang M, Mi D, Lu T, Wang B, Dong H, Zhong S, Chen Y, Sun L, Zhou X, Ma Q, Liu Z, Wang W, Zhang J, Wu Q, Marín O, Wang X. Mouse and human share conserved transcriptional programs for interneuron development. Science 2021; 374:eabj6641. [PMID: 34882453 DOI: 10.1126/science.abj6641] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Yingchao Shi
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Brain Science and Intelligence Technology (Shanghai), Institute of Biophysics, Chinese Academy of Sciences (CAS), BNU IDG/McGovern Institute for Brain Research, Beijing 100101, China
| | - Mengdi Wang
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Brain Science and Intelligence Technology (Shanghai), Institute of Biophysics, Chinese Academy of Sciences (CAS), BNU IDG/McGovern Institute for Brain Research, Beijing 100101, China.,College of Life Science, University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Da Mi
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK.,MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK.,Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Tian Lu
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Brain Science and Intelligence Technology (Shanghai), Institute of Biophysics, Chinese Academy of Sciences (CAS), BNU IDG/McGovern Institute for Brain Research, Beijing 100101, China.,College of Life Science, University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Bosong Wang
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China
| | - Hao Dong
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Brain Science and Intelligence Technology (Shanghai), Institute of Biophysics, Chinese Academy of Sciences (CAS), BNU IDG/McGovern Institute for Brain Research, Beijing 100101, China.,College of Life Science, University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Suijuan Zhong
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China.,Chinese Institute for Brain Research, Beijing 102206, China
| | - Youqiao Chen
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China
| | - Le Sun
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Xin Zhou
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Brain Science and Intelligence Technology (Shanghai), Institute of Biophysics, Chinese Academy of Sciences (CAS), BNU IDG/McGovern Institute for Brain Research, Beijing 100101, China
| | - Qiang Ma
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Brain Science and Intelligence Technology (Shanghai), Institute of Biophysics, Chinese Academy of Sciences (CAS), BNU IDG/McGovern Institute for Brain Research, Beijing 100101, China.,College of Life Science, University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Zeyuan Liu
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Brain Science and Intelligence Technology (Shanghai), Institute of Biophysics, Chinese Academy of Sciences (CAS), BNU IDG/McGovern Institute for Brain Research, Beijing 100101, China.,College of Life Science, University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Wei Wang
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Brain Science and Intelligence Technology (Shanghai), Institute of Biophysics, Chinese Academy of Sciences (CAS), BNU IDG/McGovern Institute for Brain Research, Beijing 100101, China.,College of Life Science, University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Junjing Zhang
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China
| | - Qian Wu
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China.,Chinese Institute for Brain Research, Beijing 102206, China
| | - Oscar Marín
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK.,MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| | - Xiaoqun Wang
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Brain Science and Intelligence Technology (Shanghai), Institute of Biophysics, Chinese Academy of Sciences (CAS), BNU IDG/McGovern Institute for Brain Research, Beijing 100101, China.,College of Life Science, University of the Chinese Academy of Sciences, Beijing 100049, China.,Chinese Institute for Brain Research, Beijing 102206, China.,Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China.,Guangdong Institute of Intelligence Science and Technology, Guangdong 519031, China
| |
Collapse
|
25
|
Lichtensteiger W, Bassetti-Gaille C, Rehrauer H, Georgijevic JK, Tresguerres JAF, Schlumpf M. Converging Effects of Three Different Endocrine Disrupters on Sox and Pou Gene Expression in Developing Rat Hippocampus: Possible Role of microRNA in Sex Differences. Front Genet 2021; 12:718796. [PMID: 34858468 PMCID: PMC8632217 DOI: 10.3389/fgene.2021.718796] [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] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 09/20/2021] [Indexed: 11/22/2022] Open
Abstract
Endocrine disrupting chemicals (EDCs) can impair hippocampus-dependent behaviors in rat offspring and in children. In search for key processes underlying this effect, we compared the transcriptomes of rat hippocampus on postnatal day 6 after gestational and lactational exposure to three different EDCs at doses known to impair development of learning and memory. Aroclor 1254, a commercial PCB mixture (5 mg/kg or 0.5 mg/kg), or bisphenol A (5 mg/kg or 0.5 mg/kg) were administered in chow, chlorpyrifos (3 mg/kg or 1 mg/kg) was injected subcutaneously. Male hippocampus exhibited a common effect of all three chemicals on genes involved in cell-autonomous processes, Sox6, Sox11, Pou2f2/Oct2, and Pou3f2/Brn2, all upregulated at the high dose. Additional genes of the Sox and Pou families were affected by only one or two of the chemicals. Real time RT PCR showed a comparable expression change for bisphenol A also at the lower dose. Female hippocampus exhibited much fewer genes with expression changes (almost none with false discovery rate <0.05), and none of the genes of the Sox and Pou families was affected. Since gene network analyses in male hippocampus suggested a link between Sox6 and miR-24, known to be repressed by activation of ER-alpha and to repress Sox6 in other tissues, this microRNA was measured. miR-24 was downregulated by all chemicals at the high dose in males. Values of Sox6 mRNA and miR-24 were inversely correlated in individual male hippocampus samples, supporting the hypothesis that the change in Sox6 expression resulted from an action of miR-24. In contrast, miR-24 levels remained unchanged in hippocampus of females. A sexually dimorphic response of miR-24 may thus be at the basis of the sex difference in Sox6 expression changes following exposure to the three chemicals. ER-alpha expression was also sex-dependent, but the expression changes did not parallel those of potential downstream genes such as Sox6. Sox6 is known to suppress differentiation of Parvalbumin (Pvalb)-expressing interneurons. Individual Sox6 levels (FPKM) were inversely correlated with levels of Pvalb, but not with markers of Sox6-independent interneuron subpopulations, Nos1 and 5HT3aR. Effects on interneuron development are further suggested, in males, by expression changes of Nrg1 and its receptor Erbb4, controlling interneuron migration. Our study disclosed new types of EDC-responsive morphogenetic genes, and illustrated the potential relevance of microRNAs in sexually dimorphic EDC actions.
Collapse
Affiliation(s)
- Walter Lichtensteiger
- GREEN Tox and Institute of Veterinary Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Catherine Bassetti-Gaille
- GREEN Tox and Institute of Veterinary Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Hubert Rehrauer
- Functional Genomics Center, Swiss Federal Institute of Technology and University of Zurich, Zurich, Switzerland
| | - Jelena Kühn Georgijevic
- Functional Genomics Center, Swiss Federal Institute of Technology and University of Zurich, Zurich, Switzerland
| | | | - Margret Schlumpf
- GREEN Tox and Institute of Veterinary Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
26
|
Cell Type-Specific Role of RNA Nuclease SMG6 in Neurogenesis. Cells 2021; 10:cells10123365. [PMID: 34943873 PMCID: PMC8699217 DOI: 10.3390/cells10123365] [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] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/22/2021] [Accepted: 11/26/2021] [Indexed: 12/11/2022] Open
Abstract
SMG6 is an endonuclease, which cleaves mRNAs during nonsense-mediated mRNA decay (NMD), thereby regulating gene expression and controling mRNA quality. SMG6 has been shown as a differentiation license factor of totipotent embryonic stem cells. To investigate whether it controls the differentiation of lineage-specific pluripotent progenitor cells, we inactivated Smg6 in murine embryonic neural stem cells. Nestin-Cre-mediated deletion of Smg6 in mouse neuroprogenitor cells (NPCs) caused perinatal lethality. Mutant mice brains showed normal structure at E14.5 but great reduction of the cortical NPCs and late-born cortical neurons during later stages of neurogenesis (i.e., E18.5). Smg6 inactivation led to dramatic cell death in ganglionic eminence (GE) and a reduction of interneurons at E14.5. Interestingly, neurosphere assays showed self-renewal defects specifically in interneuron progenitors but not in cortical NPCs. RT-qPCR analysis revealed that the interneuron differentiation regulators Dlx1 and Dlx2 were reduced after Smg6 deletion. Intriguingly, when Smg6 was deleted specifically in cortical and hippocampal progenitors, the mutant mice were viable and showed normal size and architecture of the cortex at E18.5. Thus, SMG6 regulates cell fate in a cell type-specific manner and is more important for neuroprogenitors originating from the GE than for progenitors from the cortex.
Collapse
|
27
|
Allaway KC, Gabitto MI, Wapinski O, Saldi G, Wang CY, Bandler RC, Wu SJ, Bonneau R, Fishell G. Genetic and epigenetic coordination of cortical interneuron development. Nature 2021; 597:693-697. [PMID: 34552240 DOI: 10.1038/s41586-021-03933-1] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 08/18/2021] [Indexed: 11/09/2022]
Abstract
One of the hallmarks of the cerebral cortex is the extreme diversity of interneurons1-3. The two largest subtypes of cortical interneurons, parvalbumin- and somatostatin-positive cells, are morphologically and functionally distinct in adulthood but arise from common lineages within the medial ganglionic eminence4-11. This makes them an attractive model for studying the generation of cell diversity. Here we examine how developmental changes in transcription and chromatin structure enable these cells to acquire distinct identities in the mouse cortex. Generic interneuron features are first detected upon cell cycle exit through the opening of chromatin at distal elements. By constructing cell-type-specific gene regulatory networks, we observed that parvalbumin- and somatostatin-positive cells initiate distinct programs upon settling within the cortex. We used these networks to model the differential transcriptional requirement of a shared regulator, Mef2c, and confirmed the accuracy of our predictions through experimental loss-of-function experiments. We therefore reveal how a common molecular program diverges to enable these neuronal subtypes to acquire highly specialized properties by adulthood. Our methods provide a framework for examining the emergence of cellular diversity, as well as for quantifying and predicting the effect of candidate genes on cell-type-specific development.
Collapse
Affiliation(s)
- Kathryn C Allaway
- Neuroscience Institute, New York University, New York, NY, USA.,Department of Neurobiology, Harvard Medical School, Boston, MA, USA.,Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA, USA
| | - Mariano I Gabitto
- Center for Computational Biology, Flatiron Institute, Simons Foundation, New York, NY, USA
| | - Orly Wapinski
- Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA, USA
| | - Giuseppe Saldi
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA.,Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA, USA.,Department of Biology, New York University, New York, NY, USA
| | - Chen-Yu Wang
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA.,Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA, USA
| | - Rachel C Bandler
- Neuroscience Institute, New York University, New York, NY, USA.,Department of Neurobiology, Harvard Medical School, Boston, MA, USA.,Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA, USA
| | - Sherry Jingjing Wu
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA.,Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA, USA
| | - Richard Bonneau
- Center for Computational Biology, Flatiron Institute, Simons Foundation, New York, NY, USA. .,Department of Biology, New York University, New York, NY, USA. .,Center for Data Science, New York University, New York, NY, USA.
| | - Gord Fishell
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA. .,Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA, USA.
| |
Collapse
|
28
|
Yang J, Yang X, Tang K. Interneuron development and dysfunction. FEBS J 2021; 289:2318-2336. [PMID: 33844440 DOI: 10.1111/febs.15872] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 04/09/2021] [Indexed: 12/17/2022]
Abstract
Understanding excitation and inhibition balance in the brain begins with the tale of two basic types of neurons, glutamatergic projection neurons and GABAergic interneurons. The diversity of cortical interneurons is contributed by multiple origins in the ventral forebrain, various tangential migration routes, and complicated regulations of intrinsic factors, extrinsic signals, and activities. Abnormalities of interneuron development lead to dysfunction of interneurons and inhibitory circuits, which are highly associated with neurodevelopmental disorders including schizophrenia, autism spectrum disorders, and intellectual disability. In this review, we mainly discuss recent findings on the development of cortical interneuron and on neurodevelopmental disorders related to interneuron dysfunction.
Collapse
Affiliation(s)
- Jiaxin Yang
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, China
| | - Xiong Yang
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, China
| | - Ke Tang
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, China
| |
Collapse
|
29
|
Stevanovic M, Drakulic D, Lazic A, Ninkovic DS, Schwirtlich M, Mojsin M. SOX Transcription Factors as Important Regulators of Neuronal and Glial Differentiation During Nervous System Development and Adult Neurogenesis. Front Mol Neurosci 2021; 14:654031. [PMID: 33867936 PMCID: PMC8044450 DOI: 10.3389/fnmol.2021.654031] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/11/2021] [Indexed: 12/11/2022] Open
Abstract
The SOX proteins belong to the superfamily of transcription factors (TFs) that display properties of both classical TFs and architectural components of chromatin. Since the cloning of the Sox/SOX genes, remarkable progress has been made in illuminating their roles as key players in the regulation of multiple developmental and physiological processes. SOX TFs govern diverse cellular processes during development, such as maintaining the pluripotency of stem cells, cell proliferation, cell fate decisions/germ layer formation as well as terminal cell differentiation into tissues and organs. However, their roles are not limited to development since SOX proteins influence survival, regeneration, cell death and control homeostasis in adult tissues. This review summarized current knowledge of the roles of SOX proteins in control of central nervous system development. Some SOX TFs suspend neural progenitors in proliferative, stem-like state and prevent their differentiation. SOX proteins function as pioneer factors that occupy silenced target genes and keep them in a poised state for activation at subsequent stages of differentiation. At appropriate stage of development, SOX members that maintain stemness are down-regulated in cells that are competent to differentiate, while other SOX members take over their functions and govern the process of differentiation. Distinct SOX members determine down-stream processes of neuronal and glial differentiation. Thus, sequentially acting SOX TFs orchestrate neural lineage development defining neuronal and glial phenotypes. In line with their crucial roles in the nervous system development, deregulation of specific SOX proteins activities is associated with neurodevelopmental disorders (NDDs). The overview of the current knowledge about the link between SOX gene variants and NDDs is presented. We outline the roles of SOX TFs in adult neurogenesis and brain homeostasis and discuss whether impaired adult neurogenesis, detected in neurodegenerative diseases, could be associated with deregulation of SOX proteins activities. We present the current data regarding the interaction between SOX proteins and signaling pathways and microRNAs that play roles in nervous system development. Finally, future research directions that will improve the knowledge about distinct and various roles of SOX TFs in health and diseases are presented and discussed.
Collapse
Affiliation(s)
- Milena Stevanovic
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia.,Faculty of Biology, University of Belgrade, Belgrade, Serbia.,Serbian Academy of Sciences and Arts, Belgrade, Serbia
| | - Danijela Drakulic
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Andrijana Lazic
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Danijela Stanisavljevic Ninkovic
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Marija Schwirtlich
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Marija Mojsin
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
30
|
Adhya D, Swarup V, Nagy R, Dutan L, Shum C, Valencia-Alarcón EP, Jozwik KM, Mendez MA, Horder J, Loth E, Nowosiad P, Lee I, Skuse D, Flinter FA, Murphy D, McAlonan G, Geschwind DH, Price J, Carroll J, Srivastava DP, Baron-Cohen S. Atypical Neurogenesis in Induced Pluripotent Stem Cells From Autistic Individuals. Biol Psychiatry 2021; 89:486-496. [PMID: 32826066 PMCID: PMC7843956 DOI: 10.1016/j.biopsych.2020.06.014] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 05/12/2020] [Accepted: 06/06/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Autism is a heterogeneous collection of disorders with a complex molecular underpinning. Evidence from postmortem brain studies have indicated that early prenatal development may be altered in autism. Induced pluripotent stem cells (iPSCs) generated from individuals with autism with macrocephaly also indicate prenatal development as a critical period for this condition. But little is known about early altered cellular events during prenatal stages in autism. METHODS iPSCs were generated from 9 unrelated individuals with autism without macrocephaly and with heterogeneous genetic backgrounds, and 6 typically developing control individuals. iPSCs were differentiated toward either cortical or midbrain fates. Gene expression and high throughput cellular phenotyping was used to characterize iPSCs at different stages of differentiation. RESULTS A subset of autism-iPSC cortical neurons were RNA-sequenced to reveal autism-specific signatures similar to postmortem brain studies, indicating a potential common biological mechanism. Autism-iPSCs differentiated toward a cortical fate displayed impairments in the ability to self-form into neural rosettes. In addition, autism-iPSCs demonstrated significant differences in rate of cell type assignment of cortical precursors and dorsal and ventral forebrain precursors. These cellular phenotypes occurred in the absence of alterations in cell proliferation during cortical differentiation, differing from previous studies. Acquisition of cell fate during midbrain differentiation was not different between control- and autism-iPSCs. CONCLUSIONS Taken together, our data indicate that autism-iPSCs diverge from control-iPSCs at a cellular level during early stage of neurodevelopment. This suggests that unique developmental differences associated with autism may be established at early prenatal stages.
Collapse
Affiliation(s)
- Dwaipayan Adhya
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom; Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Vivek Swarup
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Roland Nagy
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Lucia Dutan
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Carole Shum
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Eva P Valencia-Alarcón
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | | | - Maria Andreina Mendez
- Department of Forensic and Neurodevelopmental Sciences, Sackler Institute for Translational Neurodevelopment, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Jamie Horder
- Department of Forensic and Neurodevelopmental Sciences, Sackler Institute for Translational Neurodevelopment, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Eva Loth
- Department of Forensic and Neurodevelopmental Sciences, Sackler Institute for Translational Neurodevelopment, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Paulina Nowosiad
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Irene Lee
- Behavioural and Brain Sciences Unit, Population Policy Practice Programme, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - David Skuse
- Behavioural and Brain Sciences Unit, Population Policy Practice Programme, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Frances A Flinter
- Department of Clinical Genetics, Guy's and St. Thomas' NHS Foundation Trust, London, United Kingdom
| | - Declan Murphy
- Department of Forensic and Neurodevelopmental Sciences, Sackler Institute for Translational Neurodevelopment, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Grainne McAlonan
- Department of Forensic and Neurodevelopmental Sciences, Sackler Institute for Translational Neurodevelopment, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Daniel H Geschwind
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, California; Department of Human Genetics, University of California, Los Angeles, Los Angeles, California
| | - Jack Price
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom; MRC Centre for Neurodevelopmental Disorders, King's College London, London, United Kingdom
| | - Jason Carroll
- Cancer Research UK Cambridge Institute, Cambridge, United Kingdom
| | - Deepak P Srivastava
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom; MRC Centre for Neurodevelopmental Disorders, King's College London, London, United Kingdom.
| | - Simon Baron-Cohen
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
31
|
Franchini LF. Genetic Mechanisms Underlying Cortical Evolution in Mammals. Front Cell Dev Biol 2021; 9:591017. [PMID: 33659245 PMCID: PMC7917222 DOI: 10.3389/fcell.2021.591017] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 01/08/2021] [Indexed: 12/13/2022] Open
Abstract
The remarkable sensory, motor, and cognitive abilities of mammals mainly depend on the neocortex. Thus, the emergence of the six-layered neocortex in reptilian ancestors of mammals constitutes a fundamental evolutionary landmark. The mammalian cortex is a columnar epithelium of densely packed cells organized in layers where neurons are generated mainly in the subventricular zone in successive waves throughout development. Newborn cells move away from their site of neurogenesis through radial or tangential migration to reach their specific destination closer to the pial surface of the same or different cortical area. Interestingly, the genetic programs underlying neocortical development diversified in different mammalian lineages. In this work, I will review several recent studies that characterized how distinct transcriptional programs relate to the development and functional organization of the neocortex across diverse mammalian lineages. In some primates such as the anthropoids, the neocortex became extremely large, especially in humans where it comprises around 80% of the brain. It has been hypothesized that the massive expansion of the cortical surface and elaboration of its connections in the human lineage, has enabled our unique cognitive capacities including abstract thinking, long-term planning, verbal language and elaborated tool making capabilities. I will also analyze the lineage-specific genetic changes that could have led to the modification of key neurodevelopmental events, including regulation of cell number, neuronal migration, and differentiation into specific phenotypes, in order to shed light on the evolutionary mechanisms underlying the diversity of mammalian brains including the human brain.
Collapse
Affiliation(s)
- Lucía Florencia Franchini
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
32
|
Poulin JF, Luppi MP, Hofer C, Caronia G, Hsu PK, Chan CS, Awatramani R. PRISM: A Progenitor-Restricted Intersectional Fate Mapping Approach Redefines Forebrain Lineages. Dev Cell 2021; 53:740-753.e3. [PMID: 32574593 DOI: 10.1016/j.devcel.2020.05.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 03/24/2020] [Accepted: 05/15/2020] [Indexed: 01/08/2023]
Abstract
Lineage tracing aims to identify the progeny of a defined population of dividing progenitor cells, a daunting task in the developing central nervous system where thousands of cell types are generated. In mice, lineage analysis has been accomplished using Cre recombinase to indelibly label a defined progenitor population and its progeny. However, the interpretation of historical recombination events is hampered by the fact that driver genes are often expressed in both progenitors and postmitotic cells. Genetically inducible approaches provide temporal specificity but are afflicted by mosaicism and toxicity. Here, we present PRISM, a progenitor-restricted intersectional fate mapping approach in which Flp recombinase expression is both dependent on Cre and restricted to neural progenitors, thus circumventing the aforementioned confounds. This tool can be used in conjunction with existing Cre lines making it broadly applicable. We applied PRISM to resolve two developmentally important, but contentious, lineages-Shh and Cux2.
Collapse
Affiliation(s)
- Jean-François Poulin
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University Montreal, Quebec H3A 0G4, Canada
| | - Milagros Pereira Luppi
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Caitlyn Hofer
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Giuliana Caronia
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Pei-Ken Hsu
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - C Savio Chan
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Rajeshwar Awatramani
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
33
|
Turrero García M, Stegmann SK, Lacey TE, Reid CM, Hrvatin S, Weinreb C, Adam MA, Nagy MA, Harwell CC. Transcriptional profiling of sequentially generated septal neuron fates. eLife 2021; 10:71545. [PMID: 34851821 PMCID: PMC8694698 DOI: 10.7554/elife.71545] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 11/22/2021] [Indexed: 01/11/2023] Open
Abstract
The septum is a ventral forebrain structure known to regulate innate behaviors. During embryonic development, septal neurons are produced in multiple proliferative areas from neural progenitors following transcriptional programs that are still largely unknown. Here, we use a combination of single-cell RNA sequencing, histology, and genetic models to address how septal neuron diversity is established during neurogenesis. We find that the transcriptional profiles of septal progenitors change along neurogenesis, coinciding with the generation of distinct neuron types. We characterize the septal eminence, an anatomically distinct and transient proliferative zone composed of progenitors with distinctive molecular profiles, proliferative capacity, and fate potential compared to the rostral septal progenitor zone. We show that Nkx2.1-expressing septal eminence progenitors give rise to neurons belonging to at least three morphological classes, born in temporal cohorts that are distributed across different septal nuclei in a sequential fountain-like pattern. Our study provides insight into the molecular programs that control the sequential production of different neuronal types in the septum, a structure with important roles in regulating mood and motivation.
Collapse
Affiliation(s)
| | - Sarah K Stegmann
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
| | - Tiara E Lacey
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States,Biological and Biomedical Sciences PhD program at Harvard UniversityCambridgeUnited States
| | - Christopher M Reid
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States,PhD Program in Neuroscience at Harvard UniversityCambridgeUnited States
| | - Sinisa Hrvatin
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
| | - Caleb Weinreb
- Department of Systems Biology, Harvard Medical SchoolBostonUnited States,PhD Program in Systems Biology at Harvard UniversityCambridgeUnited States
| | - Manal A Adam
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
| | - M Aurel Nagy
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States,PhD Program in Neuroscience at Harvard UniversityCambridgeUnited States
| | - Corey C Harwell
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
| |
Collapse
|
34
|
An evolutionarily acquired microRNA shapes development of mammalian cortical projections. Proc Natl Acad Sci U S A 2020; 117:29113-29122. [PMID: 33139574 PMCID: PMC7682328 DOI: 10.1073/pnas.2006700117] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The mammalian central nervous system contains unique projections from the cerebral cortex thought to underpin complex motor and cognitive skills, including the corticospinal tract and corpus callosum. The neurons giving rise to these projections—corticospinal and callosal projection neurons—develop from the same progenitors, but acquire strikingly different fates. The broad evolutionary conservation of known genes controlling cortical projection neuron fates raises the question of how the more narrowly conserved corticospinal and callosal projections evolved. We identify a microRNA cluster selectively expressed by corticospinal projection neurons and exclusive to placental mammals. One of these microRNAs promotes corticospinal fate via regulation of the callosal gene LMO4, suggesting a mechanism whereby microRNA regulation during development promotes evolution of neuronal diversity. The corticospinal tract is unique to mammals and the corpus callosum is unique to placental mammals (eutherians). The emergence of these structures is thought to underpin the evolutionary acquisition of complex motor and cognitive skills. Corticospinal motor neurons (CSMN) and callosal projection neurons (CPN) are the archetypal projection neurons of the corticospinal tract and corpus callosum, respectively. Although a number of conserved transcriptional regulators of CSMN and CPN development have been identified in vertebrates, none are unique to mammals and most are coexpressed across multiple projection neuron subtypes. Here, we discover 17 CSMN-enriched microRNAs (miRNAs), 15 of which map to a single genomic cluster that is exclusive to eutherians. One of these, miR-409-3p, promotes CSMN subtype identity in part via repression of LMO4, a key transcriptional regulator of CPN development. In vivo, miR-409-3p is sufficient to convert deep-layer CPN into CSMN. This is a demonstration of an evolutionarily acquired miRNA in eutherians that refines cortical projection neuron subtype development. Our findings implicate miRNAs in the eutherians’ increase in neuronal subtype and projection diversity, the anatomic underpinnings of their complex behavior.
Collapse
|
35
|
De León Reyes NS, Bragg-Gonzalo L, Nieto M. Development and plasticity of the corpus callosum. Development 2020; 147:147/18/dev189738. [PMID: 32988974 DOI: 10.1242/dev.189738] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The corpus callosum (CC) connects the cerebral hemispheres and is the major mammalian commissural tract. It facilitates bilateral sensory integration and higher cognitive functions, and is often affected in neurodevelopmental diseases. Here, we review the mechanisms that contribute to the development of CC circuits in animal models and humans. These species comparisons reveal several commonalities. First, there is an early period of massive axonal projection. Second, there is a postnatal temporal window, varying between species, in which early callosal projections are selectively refined. Third, sensory-derived activity influences axonal refinement. We also discuss how defects in CC formation can lead to mild or severe CC congenital malformations.
Collapse
Affiliation(s)
- Noelia S De León Reyes
- Department of Cellular and Molecular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, (CNB-CSIC) Campus de Cantoblanco, Darwin 3, 28049 Madrid, Spain
| | - Lorena Bragg-Gonzalo
- Department of Cellular and Molecular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, (CNB-CSIC) Campus de Cantoblanco, Darwin 3, 28049 Madrid, Spain
| | - Marta Nieto
- Department of Cellular and Molecular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, (CNB-CSIC) Campus de Cantoblanco, Darwin 3, 28049 Madrid, Spain
| |
Collapse
|
36
|
Noorani I, de la Rosa J, Choi YH, Strong A, Ponstingl H, Vijayabaskar MS, Lee J, Lee E, Richard-Londt A, Friedrich M, Furlanetto F, Fuente R, Banerjee R, Yang F, Law F, Watts C, Rad R, Vassiliou G, Kim JK, Santarius T, Brandner S, Bradley A. PiggyBac mutagenesis and exome sequencing identify genetic driver landscapes and potential therapeutic targets of EGFR-mutant gliomas. Genome Biol 2020; 21:181. [PMID: 32727536 PMCID: PMC7392733 DOI: 10.1186/s13059-020-02092-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 07/06/2020] [Indexed: 12/25/2022] Open
Abstract
Background Glioma is the most common intrinsic brain tumor and also occurs in the spinal cord. Activating EGFR mutations are common in IDH1 wild-type gliomas. However, the cooperative partners of EGFR driving gliomagenesis remain poorly understood. Results We explore EGFR-mutant glioma evolution in conditional mutant mice by whole-exome sequencing, transposon mutagenesis forward genetic screening, and transcriptomics. We show mutant EGFR is sufficient to initiate gliomagenesis in vivo, both in the brain and spinal cord. We identify significantly recurrent somatic alterations in these gliomas including mutant EGFR amplifications and Sub1, Trp53, and Tead2 loss-of-function mutations. Comprehensive functional characterization of 96 gliomas by genome-wide piggyBac insertional mutagenesis in vivo identifies 281 known and novel EGFR-cooperating driver genes, including Cdkn2a, Nf1, Spred1, and Nav3. Transcriptomics confirms transposon-mediated effects on expression of these genes. We validate the clinical relevance of new putative tumor suppressors by showing these are frequently altered in patients’ gliomas, with prognostic implications. We discover shared and distinct driver mutations in brain and spinal gliomas and confirm in vivo differential tumor suppressive effects of Pten between these tumors. Functional validation with CRISPR-Cas9-induced mutations in novel genes Tead2, Spred1, and Nav3 demonstrates heightened EGFRvIII-glioma cell proliferation. Chemogenomic analysis of mutated glioma genes reveals potential drug targets, with several investigational drugs showing efficacy in vitro. Conclusion Our work elucidates functional driver landscapes of EGFR-mutant gliomas, uncovering potential therapeutic strategies, and provides new tools for functional interrogation of gliomagenesis.
Collapse
Affiliation(s)
- Imran Noorani
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire, CB10 1SA, UK. .,Department of Neurosurgery, Addenbrookes Hospital, University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK.
| | - Jorge de la Rosa
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Yoon Ha Choi
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire, CB10 1SA, UK.,Department of New Biology, DGIST, 333, Techno Jungang Daero, Hyeonpung-Myeon, Dalseong-Gun, Daegu, 42988, South Korea
| | - Alexander Strong
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Hannes Ponstingl
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - M S Vijayabaskar
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Jusung Lee
- Department of New Biology, DGIST, 333, Techno Jungang Daero, Hyeonpung-Myeon, Dalseong-Gun, Daegu, 42988, South Korea
| | - Eunmin Lee
- Department of New Biology, DGIST, 333, Techno Jungang Daero, Hyeonpung-Myeon, Dalseong-Gun, Daegu, 42988, South Korea
| | - Angela Richard-Londt
- Division of Neuropathology and Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, Mailbox 126, London, WC1N 3BG, UK
| | - Mathias Friedrich
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire, CB10 1SA, UK.,Department of Internal Medicine II, Klinikum rechts der Isar, Technische Universität München, Ismaninger Strasse 22, 81675, Munich, Germany
| | - Federica Furlanetto
- Department of Internal Medicine II, Klinikum rechts der Isar, Technische Universität München, Ismaninger Strasse 22, 81675, Munich, Germany
| | - Rocio Fuente
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Ruby Banerjee
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Fengtang Yang
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Frances Law
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Colin Watts
- Department of Neurosurgery, Addenbrookes Hospital, University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK.,Birmingham Brain Cancer Program, Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Roland Rad
- Department of Internal Medicine II, Klinikum rechts der Isar, Technische Universität München, Ismaninger Strasse 22, 81675, Munich, Germany
| | - George Vassiliou
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Jong Kyoung Kim
- Department of New Biology, DGIST, 333, Techno Jungang Daero, Hyeonpung-Myeon, Dalseong-Gun, Daegu, 42988, South Korea
| | - Thomas Santarius
- Department of Neurosurgery, Addenbrookes Hospital, University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK
| | - Sebastian Brandner
- Division of Neuropathology and Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, Mailbox 126, London, WC1N 3BG, UK
| | - Allan Bradley
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire, CB10 1SA, UK.
| |
Collapse
|
37
|
Abstract
Cortical interneurons display striking differences in shape, physiology, and other attributes, challenging us to appropriately classify them. We previously suggested that interneuron types should be defined by their role in cortical processing. Here, we revisit the question of how to codify their diversity based upon their division of labor and function as controllers of cortical information flow. We suggest that developmental trajectories provide a guide for appreciating interneuron diversity and argue that subtype identity is generated using a configurational (rather than combinatorial) code of transcription factors that produce attractor states in the underlying gene regulatory network. We present our updated three-stage model for interneuron specification: an initial cardinal step, allocating interneurons into a few major classes, followed by definitive refinement, creating subclasses upon settling within the cortex, and lastly, state determination, reflecting the incorporation of interneurons into functional circuit ensembles. We close by discussing findings indicating that major interneuron classes are both evolutionarily ancient and conserved. We propose that the complexity of cortical circuits is generated by phylogenetically old interneuron types, complemented by an evolutionary increase in principal neuron diversity. This suggests that a natural neurobiological definition of interneuron types might be derived from a match between their developmental origin and computational function.
Collapse
Affiliation(s)
- Gord Fishell
- Department of Neurobiology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts 02115, USA;
- Stanley Center for Psychiatric Research, Broad Institute, Cambridge, Massachusetts 02142, USA
- Center for Genomics and Systems Biology, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Adam Kepecs
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
- Department of Neuroscience, Washington University in St. Louis, St. Louis, Missouri 63130, USA;
| |
Collapse
|
38
|
Bartholome O, de la Brassinne Bonardeaux O, Neirinckx V, Rogister B. A Composite Sketch of Fast-Spiking Parvalbumin-Positive Neurons. Cereb Cortex Commun 2020; 1:tgaa026. [PMID: 34296100 PMCID: PMC8153048 DOI: 10.1093/texcom/tgaa026] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/15/2020] [Accepted: 06/15/2020] [Indexed: 01/28/2023] Open
Abstract
Parvalbumin-positive neurons are inhibitory neurons that release GABA and are mostly represented by fast-spiking basket or chandelier cells. They constitute a minor neuronal population, yet their peculiar profiles allow them to react quickly to any event in the brain under normal or pathological conditions. In this review, we will summarize the current knowledge about the fundamentals of fast-spiking parvalbumin-positive neurons, focusing on their morphology and specific channel/protein content. Next, we will explore their development, maturation, and migration in the brain. Finally, we will unravel their potential contribution to the physiopathology of epilepsy.
Collapse
Affiliation(s)
| | | | | | - Bernard Rogister
- GIGA-Neurosciences, University of Liege, 4000 Liège, Belgium
- Neurology Department, CHU, Academic Hospital, University of Liege, 4000 Liège, Belgium
| |
Collapse
|
39
|
Tolchin D, Yeager JP, Prasad P, Dorrani N, Russi AS, Martinez-Agosto JA, Haseeb A, Angelozzi M, Santen GWE, Ruivenkamp C, Mercimek-Andrews S, Depienne C, Kuechler A, Mikat B, Ludecke HJ, Bilan F, Le Guyader G, Gilbert-Dussardier B, Keren B, Heide S, Haye D, Van Esch H, Keldermans L, Ortiz D, Lancaster E, Krantz ID, Krock BL, Pechter KB, Arkader A, Medne L, DeChene ET, Calpena E, Melistaccio G, Wilkie AOM, Suri M, Foulds N, Begtrup A, Henderson LB, Forster C, Reed P, McDonald MT, McConkie-Rosell A, Thevenon J, Le Tanno P, Coutton C, Tsai ACH, Stewart S, Maver A, Gorazd R, Pichon O, Nizon M, Cogné B, Isidor B, Martin-Coignard D, Stoeva R, Lefebvre V, Le Caignec C. De Novo SOX6 Variants Cause a Neurodevelopmental Syndrome Associated with ADHD, Craniosynostosis, and Osteochondromas. Am J Hum Genet 2020; 106:830-845. [PMID: 32442410 PMCID: PMC7273536 DOI: 10.1016/j.ajhg.2020.04.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 04/24/2020] [Indexed: 12/21/2022] Open
Abstract
SOX6 belongs to a family of 20 SRY-related HMG-box-containing (SOX) genes that encode transcription factors controlling cell fate and differentiation in many developmental and adult processes. For SOX6, these processes include, but are not limited to, neurogenesis and skeletogenesis. Variants in half of the SOX genes have been shown to cause severe developmental and adult syndromes, referred to as SOXopathies. We here provide evidence that SOX6 variants also cause a SOXopathy. Using clinical and genetic data, we identify 19 individuals harboring various types of SOX6 alterations and exhibiting developmental delay and/or intellectual disability; the individuals are from 17 unrelated families. Additional, inconstant features include attention-deficit/hyperactivity disorder (ADHD), autism, mild facial dysmorphism, craniosynostosis, and multiple osteochondromas. All variants are heterozygous. Fourteen are de novo, one is inherited from a mosaic father, and four offspring from two families have a paternally inherited variant. Intragenic microdeletions, balanced structural rearrangements, frameshifts, and nonsense variants are predicted to inactivate the SOX6 variant allele. Four missense variants occur in residues and protein regions highly conserved evolutionarily. These variants are not detected in the gnomAD control cohort, and the amino acid substitutions are predicted to be damaging. Two of these variants are located in the HMG domain and abolish SOX6 transcriptional activity in vitro. No clear genotype-phenotype correlations are found. Taken together, these findings concur that SOX6 haploinsufficiency leads to a neurodevelopmental SOXopathy that often includes ADHD and abnormal skeletal and other features.
Collapse
Affiliation(s)
- Dara Tolchin
- Department of Surgery, Division of Orthopaedic Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Jessica P Yeager
- Department of Surgery, Division of Orthopaedic Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Priya Prasad
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Naghmeh Dorrani
- Division of Medical Genetics, Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Alvaro Serrano Russi
- Division of Medical Genetics, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
| | - Julian A Martinez-Agosto
- Division of Medical Genetics, Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Abdul Haseeb
- Department of Surgery, Division of Orthopaedic Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Marco Angelozzi
- Department of Surgery, Division of Orthopaedic Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - G W E Santen
- Department of Clinical Genetics, Leiden University Medical Centre, 2300 LC Leiden, the Netherlands
| | - Claudia Ruivenkamp
- Department of Clinical Genetics, Leiden University Medical Centre, 2300 LC Leiden, the Netherlands
| | - Saadet Mercimek-Andrews
- Division of Clinical and Metabolic Genetics, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada; Institute of Medical Sciences, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Christel Depienne
- Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Alma Kuechler
- Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Barbara Mikat
- Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Hermann-Josef Ludecke
- Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; Institute für Humangenetik, Universitätsklinikum Düsseldorf, Heinrich-Heine-Universität, 40225 Düsseldorf, Germany
| | - Frederic Bilan
- Neurovascular Unit and Cognitive Disorders (EA-3808 NEUVACOD), Université de Poitiers, 86073 Poitiers, France; Service de Génétique Clinique, Centre Hospitalier Universitaire de Poitiers, 86021 Poitiers, France
| | - Gwenael Le Guyader
- Neurovascular Unit and Cognitive Disorders (EA-3808 NEUVACOD), Université de Poitiers, 86073 Poitiers, France; Service de Génétique Clinique, Centre Hospitalier Universitaire de Poitiers, 86021 Poitiers, France
| | - Brigitte Gilbert-Dussardier
- Neurovascular Unit and Cognitive Disorders (EA-3808 NEUVACOD), Université de Poitiers, 86073 Poitiers, France; Service de Génétique Clinique, Centre Hospitalier Universitaire de Poitiers, 86021 Poitiers, France
| | - Boris Keren
- Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Pitié-Salpêtrière, Département de Génétique, 75013 Paris, France
| | - Solveig Heide
- Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Pitié-Salpêtrière, Département de Génétique, 75013 Paris, France
| | - Damien Haye
- Service de Génétique, Centre Hospitalier Universitaire de Nice Hôpital de l'Archet 2,151 route Saint Antoine de la Ginestière, 062002 Nice, France
| | - Hilde Van Esch
- Center for Human Genetics, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Liesbeth Keldermans
- Laboratory for Molecular Diagnosis, Center for Human Genetics, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Damara Ortiz
- University of Pittsburgh Medical Center, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
| | - Emily Lancaster
- University of Pittsburgh Medical Center, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
| | - Ian D Krantz
- Roberts Individualized Medical Genetics Center, Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Bryan L Krock
- Division of Genomic Diagnostics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Kieran B Pechter
- Division of Genomic Diagnostics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Alexandre Arkader
- Department of Surgery, Division of Orthopaedic Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Livija Medne
- Roberts Individualized Medical Genetics Center, Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Elizabeth T DeChene
- Division of Genomic Diagnostics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Eduardo Calpena
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, UK
| | - Giada Melistaccio
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, UK
| | - Andrew O M Wilkie
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, UK; Clinical Genetics Service, Nottingham University Hospitals NHS Trust, City Hospital Campus, Nottingham NG5 1PB, UK
| | - Mohnish Suri
- Clinical Genetics Service, Nottingham University Hospitals NHS Trust, City Hospital Campus, Nottingham NG5 1PB, UK
| | - Nicola Foulds
- Wessex Clinical Genetics Services, University Hospital Southampton NHS Foundation Trust, Southampton SO16 5YA, UK
| | | | | | | | | | - Marie T McDonald
- Division of Medical Genetics, Department of Pediatrics, Duke University, Durham, NC 27707, USA
| | - Allyn McConkie-Rosell
- Division of Medical Genetics, Department of Pediatrics, Duke University, Durham, NC 27707, USA
| | - Julien Thevenon
- Service de Génétique, Génomique, et Procréation, Centre Hospitalier Universitaire Grenoble Alpes, 38700 La Tronche, France; INSERM 1209, CNRS UMR 5309, Institute for Advanced Biosciences, Université Grenoble Alpes, 38706 Grenoble, France
| | - Pauline Le Tanno
- Service de Génétique, Génomique, et Procréation, Centre Hospitalier Universitaire Grenoble Alpes, 38700 La Tronche, France; INSERM 1209, CNRS UMR 5309, Institute for Advanced Biosciences, Université Grenoble Alpes, 38706 Grenoble, France
| | - Charles Coutton
- Service de Génétique, Génomique, et Procréation, Centre Hospitalier Universitaire Grenoble Alpes, 38700 La Tronche, France; INSERM 1209, CNRS UMR 5309, Institute for Advanced Biosciences, Université Grenoble Alpes, 38706 Grenoble, France
| | - Anne C H Tsai
- Section of Genetics, Department of Pediatrics, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO 80045, USA
| | - Sarah Stewart
- Section of Genetics, Department of Pediatrics, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO 80045, USA
| | - Ales Maver
- Clinical Institute of Medical Genetics, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
| | - Rudolf Gorazd
- Clinical Institute of Medical Genetics, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
| | - Olivier Pichon
- Centre Hospitalier Universitaire Nantes, Service de Génétique Médicale, 44000 Nantes, France
| | - Mathilde Nizon
- Centre Hospitalier Universitaire Nantes, Service de Génétique Médicale, 44000 Nantes, France; Université de Nantes, CNRS, INSERM, L'Institut du Thorax, 44000 Nantes, France
| | - Benjamin Cogné
- Centre Hospitalier Universitaire Nantes, Service de Génétique Médicale, 44000 Nantes, France; Université de Nantes, CNRS, INSERM, L'Institut du Thorax, 44000 Nantes, France
| | - Bertrand Isidor
- Centre Hospitalier Universitaire Nantes, Service de Génétique Médicale, 44000 Nantes, France; Université de Nantes, CNRS, INSERM, L'Institut du Thorax, 44000 Nantes, France
| | | | - Radka Stoeva
- Service de Cytogénétique, Centre Hospitalier Universitaire de Le Mans, 72037 Le Mans, France
| | - Véronique Lefebvre
- Department of Surgery, Division of Orthopaedic Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA.
| | - Cédric Le Caignec
- Centre Hospitalier Universitaire Nantes, Service de Génétique Médicale, 44000 Nantes, France; Centre Hospitalier Universitaire Toulouse, Service de Génétique Médicale, 31000 Toulouse, France.
| |
Collapse
|
40
|
Deryckere A, Stappers E, Dries R, Peyre E, van den Berghe V, Conidi A, Zampeta FI, Francis A, Bresseleers M, Stryjewska A, Vanlaer R, Maas E, Smal IV, van IJcken WFJ, Grosveld FG, Nguyen L, Huylebroeck D, Seuntjens E. Multifaceted actions of Zeb2 in postnatal neurogenesis from the ventricular-subventricular zone to the olfactory bulb. Development 2020; 147:dev184861. [PMID: 32253238 DOI: 10.1242/dev.184861] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 03/23/2020] [Indexed: 03/01/2024]
Abstract
The transcription factor Zeb2 controls fate specification and subsequent differentiation and maturation of multiple cell types in various embryonic tissues. It binds many protein partners, including activated Smad proteins and the NuRD co-repressor complex. How Zeb2 subdomains support cell differentiation in various contexts has remained elusive. Here, we studied the role of Zeb2 and its domains in neurogenesis and neural differentiation in the young postnatal ventricular-subventricular zone (V-SVZ), in which neural stem cells generate olfactory bulb-destined interneurons. Conditional Zeb2 knockouts and separate acute loss- and gain-of-function approaches indicated that Zeb2 is essential for controlling apoptosis and neuronal differentiation of V-SVZ progenitors before and after birth, and we identified Sox6 as a potential downstream target gene of Zeb2. Zeb2 genetic inactivation impaired the differentiation potential of the V-SVZ niche in a cell-autonomous fashion. We also provide evidence that its normal function in the V-SVZ also involves non-autonomous mechanisms. Additionally, we demonstrate distinct roles for Zeb2 protein-binding domains, suggesting that Zeb2 partners co-determine neuronal output from the mouse V-SVZ in both quantitative and qualitative ways in early postnatal life.
Collapse
Affiliation(s)
- Astrid Deryckere
- Laboratory of Developmental Neurobiology, Department of Biology, KU Leuven, Leuven 3000, Belgium
| | - Elke Stappers
- Laboratory of Molecular Biology (Celgen), Department of Development and Regeneration, KU Leuven, Leuven 3000, Belgium
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam 3015 CN, The Netherlands
| | - Ruben Dries
- Laboratory of Molecular Biology (Celgen), Department of Development and Regeneration, KU Leuven, Leuven 3000, Belgium
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam 3015 CN, The Netherlands
| | - Elise Peyre
- GIGA-Stem Cells and GIGA-Neurosciences, Liège University, Liège 4000, Belgium
| | - Veronique van den Berghe
- Laboratory of Molecular Biology (Celgen), Department of Development and Regeneration, KU Leuven, Leuven 3000, Belgium
- Department of Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, and MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| | - Andrea Conidi
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam 3015 CN, The Netherlands
| | - F Isabella Zampeta
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam 3015 CN, The Netherlands
| | - Annick Francis
- Laboratory of Molecular Biology (Celgen), Department of Development and Regeneration, KU Leuven, Leuven 3000, Belgium
| | - Marjolein Bresseleers
- Laboratory of Molecular Biology (Celgen), Department of Development and Regeneration, KU Leuven, Leuven 3000, Belgium
| | - Agata Stryjewska
- Laboratory of Molecular Biology (Celgen), Department of Development and Regeneration, KU Leuven, Leuven 3000, Belgium
| | - Ria Vanlaer
- Laboratory of Developmental Neurobiology, Department of Biology, KU Leuven, Leuven 3000, Belgium
| | - Elke Maas
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KU Leuven, Leuven 3000, Belgium
| | - Ihor V Smal
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam 3015 CN, The Netherlands
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam 3015 CN, The Netherlands
| | - Wilfred F J van IJcken
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam 3015 CN, The Netherlands
- Center for Biomics-Genomics, Department of Cell Biology, Erasmus University Medical Center, Rotterdam 3015 CN, The Netherlands
| | - Frank G Grosveld
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam 3015 CN, The Netherlands
| | - Laurent Nguyen
- GIGA-Stem Cells and GIGA-Neurosciences, Liège University, Liège 4000, Belgium
| | - Danny Huylebroeck
- Laboratory of Molecular Biology (Celgen), Department of Development and Regeneration, KU Leuven, Leuven 3000, Belgium
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam 3015 CN, The Netherlands
| | - Eve Seuntjens
- Laboratory of Developmental Neurobiology, Department of Biology, KU Leuven, Leuven 3000, Belgium
- Laboratory of Molecular Biology (Celgen), Department of Development and Regeneration, KU Leuven, Leuven 3000, Belgium
| |
Collapse
|
41
|
Symmank J, Gölling V, Gerstmann K, Zimmer G. The Transcription Factor LHX1 Regulates the Survival and Directed Migration of POA-derived Cortical Interneurons. Cereb Cortex 2020; 29:1644-1658. [PMID: 29912395 DOI: 10.1093/cercor/bhy063] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Revised: 02/22/2018] [Accepted: 02/23/2018] [Indexed: 12/17/2022] Open
Abstract
The delicate balance of excitation and inhibition is crucial for proper function of the cerebral cortex, relying on the accurate number and subtype composition of inhibitory gamma-aminobutyric (GABA)-expressing interneurons. Various intrinsic and extrinsic factors precisely orchestrate their multifaceted development including the long-range migration from the basal telencephalon to cortical targets as well as interneuron survival throughout the developmental period. Particularly expressed guidance receptors were described to channel the migration of cortical interneurons deriving from the medial ganglionic eminence (MGE) and the preoptic area (POA) along distinct routes. Hence, unveiling the regulatory genetic networks controlling subtype-specific gene expression profiles is key to understand interneuron-specific developmental programs and to reveal causes for associated disorders. In contrast to MGE-derived interneurons, little is known about the transcriptional networks in interneurons born in the POA. Here, we provide first evidence for the LIM-homeobox transcription factor LHX1 as a crucial key player in the post-mitotic development of POA-derived cortical interneurons. By transcriptional regulation of related genes, LHX1 modulates their survival as well as the subtype-specific expression of guidance receptors of the Eph/ephrin family, thereby affecting directional migration and layer distribution in the adult cortex.
Collapse
Affiliation(s)
- Judit Symmank
- Institute of Human Genetics, University Hospital Jena, Jena, Germany
| | - Vanessa Gölling
- Institute of Human Genetics, University Hospital Jena, Jena, Germany
| | - Katrin Gerstmann
- Institute of Human Genetics, University Hospital Jena, Jena, Germany
| | - Geraldine Zimmer
- Institute of Human Genetics, University Hospital Jena, Jena, Germany
| |
Collapse
|
42
|
Shen W, Ba R, Su Y, Ni Y, Chen D, Xie W, Pleasure SJ, Zhao C. Foxg1 Regulates the Postnatal Development of Cortical Interneurons. Cereb Cortex 2020; 29:1547-1560. [PMID: 29912324 DOI: 10.1093/cercor/bhy051] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 01/23/2018] [Accepted: 02/15/2018] [Indexed: 12/12/2022] Open
Abstract
Abnormalities in cortical interneurons are closely associated with neurological diseases. Most patients with Foxg1 syndrome experience seizures, suggesting a possible role of Foxg1 in the cortical interneuron development. Here, by conditional deletion of Foxg1, which was achieved by crossing Foxg1fl/fl with the Gad2-CreER line, we found the postnatal distributions of somatostatin-, calretinin-, and neuropeptide Y-positive interneurons in the cortex were impaired. Further investigations revealed an enhanced dendritic complexity and decreased migration capacity of Foxg1-deficient interneurons, accompanied by remarkable downregulation of Dlx1 and CXCR4. Overexpression of Dlx1 or knock down its downstream Pak3 rescued the differentiation detects, demonstrated that Foxg1 functioned upstream of Dlx1-Pak3 signal pathway to regulate the postnatal development of cortical interneurons. Due to the imbalanced neural circuit, Foxg1 mutants showed increased seizure susceptibility. These findings will improve our understanding of the postnatal development of interneurons and help to elucidate the mechanisms underlying seizure in patients carrying Foxg1 mutations.
Collapse
Affiliation(s)
- Wei Shen
- Key Laboratory of Developmental Genes and Human Diseases, MOE, School of Medicine, Southeast University, Nanjing, P. R. China
| | - Ru Ba
- Key Laboratory of Developmental Genes and Human Diseases, MOE, School of Medicine, Southeast University, Nanjing, P. R. China
| | - Yan Su
- Key Laboratory of Developmental Genes and Human Diseases, MOE, School of Medicine, Southeast University, Nanjing, P. R. China
| | - Yang Ni
- Key Laboratory of Developmental Genes and Human Diseases, MOE, School of Medicine, Southeast University, Nanjing, P. R. China
| | - Dongsheng Chen
- Key Laboratory of Developmental Genes and Human Diseases, MOE, School of Medicine, Southeast University, Nanjing, P. R. China
| | - Wei Xie
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Institute of Life Science, Southeast University, Nanjing, P. R. China
| | - Samuel J Pleasure
- Department of Neurology, Weill Institute for Neuroscience, Programs in Neuroscience and Developmental Stem Cell Biology, UCSF, San Francisco, CA, USA
| | - Chunjie Zhao
- Key Laboratory of Developmental Genes and Human Diseases, MOE, School of Medicine, Southeast University, Nanjing, P. R. China.,Center of Depression, Beijing Institute for Brain Disorders, Beijing 100069, People's Republic of China
| |
Collapse
|
43
|
Abecassis ZA, Berceau BL, Win PH, García D, Xenias HS, Cui Q, Pamukcu A, Cherian S, Hernández VM, Chon U, Lim BK, Kim Y, Justice NJ, Awatramani R, Hooks BM, Gerfen CR, Boca SM, Chan CS. Npas1 +-Nkx2.1 + Neurons Are an Integral Part of the Cortico-pallido-cortical Loop. J Neurosci 2020; 40:743-768. [PMID: 31811030 PMCID: PMC6975296 DOI: 10.1523/jneurosci.1199-19.2019] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 11/21/2019] [Accepted: 11/26/2019] [Indexed: 11/21/2022] Open
Abstract
Within the basal ganglia circuit, the external globus pallidus (GPe) is critically involved in motor control. Aside from Foxp2+ neurons and ChAT+ neurons that have been established as unique neuron types, there is little consensus on the classification of GPe neurons. Properties of the remaining neuron types are poorly defined. In this study, we leverage new mouse lines, viral tools, and molecular markers to better define GPe neuron subtypes. We found that Sox6 represents a novel, defining marker for GPe neuron subtypes. Lhx6+ neurons that lack the expression of Sox6 were devoid of both parvalbumin and Npas1. This result confirms previous assertions of the existence of a unique Lhx6+ population. Neurons that arise from the Dbx1+ lineage were similarly abundant in the GPe and displayed a heterogeneous makeup. Importantly, tracing experiments revealed that Npas1+-Nkx2.1+ neurons represent the principal noncholinergic, cortically-projecting neurons. In other words, they form the pallido-cortical arm of the cortico-pallido-cortical loop. Our data further show that pyramidal-tract neurons in the cortex collateralized within the GPe, forming a closed-loop system between the two brain structures. Overall, our findings reconcile some of the discrepancies that arose from differences in techniques or the reliance on preexisting tools. Although spatial distribution and electrophysiological properties of GPe neurons reaffirm the diversification of GPe subtypes, statistical analyses strongly support the notion that these neuron subtypes can be categorized under the two principal neuron classes: PV+ neurons and Npas1+ neurons.SIGNIFICANCE STATEMENT The poor understanding of the neuronal composition in the external globus pallidus (GPe) undermines our ability to interrogate its precise behavioral and disease involvements. In this study, 12 different genetic crosses were used, hundreds of neurons were electrophysiologically characterized, and >100,000 neurons were histologically- and/or anatomically-profiled. Our current study further establishes the segregation of GPe neuron classes and illustrates the complexity of GPe neurons in adult mice. Our results support the idea that Npas1+-Nkx2.1+ neurons are a distinct GPe neuron subclass. By providing a detailed analysis of the organization of the cortico-pallidal-cortical projection, our findings establish the cellular and circuit substrates that can be important for motor function and dysfunction.
Collapse
Affiliation(s)
- Zachary A Abecassis
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Brianna L Berceau
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Phyo H Win
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Daniela García
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Harry S Xenias
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Qiaoling Cui
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Arin Pamukcu
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Suraj Cherian
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Vivian M Hernández
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Uree Chon
- Department of Neural and Behavioral Sciences, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania
| | - Byung Kook Lim
- Neurobiology Section, Biological Sciences Division, University of California San Diego, La Jolla, California
| | - Yongsoo Kim
- Department of Neural and Behavioral Sciences, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania
| | - Nicholas J Justice
- Center for Metabolic and degenerative disease, Institute of Molecular Medicine, University of Texas, Houston, Texas
- Department of Integrative Pharmacology, University of Texas, Houston, Texas
| | - Raj Awatramani
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Bryan M Hooks
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Charles R Gerfen
- Laboratory of Systems Neuroscience, National Institute of Mental Health, Bethesda, Maryland, and
| | - Simina M Boca
- Innovation Center for Biomedical Informatics, Georgetown University Medical Center, Washington, District of Columbia
| | - C Savio Chan
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois,
| |
Collapse
|
44
|
Saleem M, Hodgkinson CP, Xiao L, Gimenez-Bastida JA, Rasmussen ML, Foss J, Payne AJ, Mirotsou M, Gama V, Dzau VJ, Gomez JA. Sox6 as a new modulator of renin expression in the kidney. Am J Physiol Renal Physiol 2019; 318:F285-F297. [PMID: 31760770 DOI: 10.1152/ajprenal.00095.2019] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Juxtaglomerular (JG) cells, major sources of renin, differentiate from metanephric mesenchymal cells that give rise to JG cells or a subset of smooth muscle cells of the renal afferent arteriole. During periods of dehydration and salt deprivation, renal mesenchymal stromal cells (MSCs) differentiate from JG cells. JG cells undergo expansion and smooth muscle cells redifferentiate to express renin along the afferent arteriole. Gene expression profiling comparing resident renal MSCs with JG cells indicates that the transcription factor Sox6 is highly expressed in JG cells in the adult kidney. In vitro, loss of Sox6 expression reduces differentiation of renal MSCs to renin-producing cells. In vivo, Sox6 expression is upregulated after a low-Na+ diet and furosemide. Importantly, knockout of Sox6 in Ren1d+ cells halts the increase in renin-expressing cells normally seen during a low-Na+ diet and furosemide as well as the typical increase in renin. Furthermore, Sox6 ablation in renin-expressing cells halts the recruitment of smooth muscle cells along the afferent arteriole, which normally express renin under these conditions. These results support a previously undefined role for Sox6 in renin expression.
Collapse
Affiliation(s)
- Mohammad Saleem
- Department of Medicine/Clinical Pharmacology Division, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Conrad P Hodgkinson
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Liang Xiao
- Department of Medicine/Clinical Pharmacology Division, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Juan A Gimenez-Bastida
- Department of Medicine/Clinical Pharmacology Division, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Megan L Rasmussen
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee
| | - Jason Foss
- Department of Medicine/Clinical Pharmacology Division, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Alan J Payne
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Maria Mirotsou
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Vivian Gama
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee
| | - Victor J Dzau
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Jose A Gomez
- Department of Medicine/Clinical Pharmacology Division, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
45
|
Raina A, Mahajani S, Bähr M, Kügler S. Neuronal Trans-differentiation by Transcription Factors Ascl1 and Nurr1: Induction of a Dopaminergic Neurotransmitter Phenotype in Cortical GABAergic Neurons. Mol Neurobiol 2019; 57:249-260. [PMID: 31317490 DOI: 10.1007/s12035-019-01701-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 07/09/2019] [Indexed: 12/17/2022]
Abstract
Neurons with a desired neurotransmitter phenotype can be differentiated from induced pluripotent stem cells or from somatic cells only through tedious protocols with relatively low yield. Readily available cortical neurons isolated from embryonic rat brain, which have already undergone a complete neuronal differentiation process, might serve as alternative template source. These cultures consist of 85% glutamatergic and 15% GABAergic neurons, and we attempted to trans-differentiate them into dopaminergic neurons. Transcription factors Nurr1, Lmx1A and Pitx3, essential determinants of a dopaminergic cell fate during CNS development, were not sufficient to induce tyrosine hydroxylase expression in a significant number of cells. Combining Nurr1 with the generic neuronal differentiator and re-programming factor Ascl1, however, resulted in generation of neurons which express dopaminergic markers TH, AADC, VMAT2 and DAT. Only neurons of GABAergic phenotype could be trans-differentiated towards a dopaminergic neurotransmitter phenotype, while for glutamatergic neurons, this process proved to be neurotoxic. Intriguingly, GABAergic neurons isolated from embryonal midbrain could not be trans-differentiated into dopaminergic neurons by Ascl1 and Nurr1. Thus, in principle, post-mitotic embryonal neurons can serve as templates for neurons with a desired neurotransmitter phenotype. However, neurotransmitter phenotype plasticity critically depends on the differentiation history of the template neurons, which can result in relatively low yields of dopaminergic neurons.
Collapse
Affiliation(s)
- Anupam Raina
- Department of Neurology, University Medicine Göttingen, Waldweg 33, 37073, Göttingen, Germany.,Center Nanoscale Microscopy and Physiology of the Brain (CNMPB), Göttingen, Germany
| | - Sameehan Mahajani
- Department of Neurology, University Medicine Göttingen, Waldweg 33, 37073, Göttingen, Germany.,Center Nanoscale Microscopy and Physiology of the Brain (CNMPB), Göttingen, Germany
| | - Mathias Bähr
- Department of Neurology, University Medicine Göttingen, Waldweg 33, 37073, Göttingen, Germany.,Center Nanoscale Microscopy and Physiology of the Brain (CNMPB), Göttingen, Germany
| | - Sebastian Kügler
- Department of Neurology, University Medicine Göttingen, Waldweg 33, 37073, Göttingen, Germany. .,Center Nanoscale Microscopy and Physiology of the Brain (CNMPB), Göttingen, Germany.
| |
Collapse
|
46
|
Angelozzi M, Lefebvre V. SOXopathies: Growing Family of Developmental Disorders Due to SOX Mutations. Trends Genet 2019; 35:658-671. [PMID: 31288943 DOI: 10.1016/j.tig.2019.06.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 06/12/2019] [Accepted: 06/17/2019] [Indexed: 12/15/2022]
Abstract
The SRY-related (SOX) transcription factor family pivotally contributes to determining cell fate and identity in many lineages. Since the original discovery that SRY deletions cause sex reversal, mutations in half of the 20 human SOX genes have been associated with rare congenital disorders, henceforward called SOXopathies. Mutations are generally de novo, heterozygous, and inactivating, revealing gene haploinsufficiency, but other types, including duplications, have been reported too. Missense variants primarily target the HMG domain, the SOX hallmark that mediates DNA binding and bending, nuclear trafficking, and protein-protein interactions. We here review key clinical and molecular features of SOXopathies and discuss the prospect that the disease family likely involves more SOX genes and larger clinical and genetic spectrums than currently appreciated.
Collapse
Affiliation(s)
- Marco Angelozzi
- Department of Surgery/Division of Orthopaedic Surgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Véronique Lefebvre
- Department of Surgery/Division of Orthopaedic Surgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA.
| |
Collapse
|
47
|
Chen D, Wang C, Li M, She X, Yuan Y, Chen H, Zhang W, Zhao C. Loss of Foxg1 Impairs the Development of Cortical SST-Interneurons Leading to Abnormal Emotional and Social Behaviors. Cereb Cortex 2019; 29:3666-3682. [DOI: 10.1093/cercor/bhz114] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 04/10/2019] [Accepted: 05/05/2019] [Indexed: 12/19/2022] Open
Abstract
Abstract
FOXG1 syndrome is a severe encephalopathy that exhibit intellectual disability, emotional disorder, and limited social communication. To elucidate the contribution of somatostatin-expressing interneurons (SST-INs) to the cellular basis underlying FOXG1 syndrome, here, by crossing SST-cre with a Foxg1fl/fl line, we selectively ablated Foxg1. Loss of Foxg1 resulted in an obvious reduction in the number of SST-INs, accompanied by an altered ratio of subtypes. Foxg1-deficient SST-INs exhibited decreased membrane excitability and a changed ratio of electrophysiological firing patterns, which subsequently led to an excitatory/inhibitory imbalance. Moreover, cognitive defects, limited social interactions, and depression-like behaviors were detected in Foxg1 cKO mice. Treatment with low-dose of clonazepam effectively alleviated the defects. These results identify a link of SST-IN development to the aberrant emotion, cognition, and social capacities in patients. Our findings identify a novel role of Foxg1 in SST-IN development and put new insights into the cellular basis of FOXG1 syndrome.
Collapse
Affiliation(s)
- Dongsheng Chen
- Key Laboratory of Developmental Genes and Human Diseases, MOE, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
| | - Chunlian Wang
- Key Lab of Cognition and Personality, MOE, School of Psychology, Southwest University, Chongqing, China
| | - Meiyi Li
- Key Lab of Cognition and Personality, MOE, School of Psychology, Southwest University, Chongqing, China
| | - Xinyu She
- Key Laboratory of Developmental Genes and Human Diseases, MOE, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
| | - Yonggui Yuan
- Department of Psychosomatics and Psychiatry, ZhongDa Hospital, Medical School of Southeast University, Nanjing, Jiangsu Province, China
| | - Huanxin Chen
- Key Lab of Cognition and Personality, MOE, School of Psychology, Southwest University, Chongqing, China
| | - Weining Zhang
- School of Medicine, Jiangsu University, ZhenJiang, Jiangsu Province, China
| | - Chunjie Zhao
- Key Laboratory of Developmental Genes and Human Diseases, MOE, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
| |
Collapse
|
48
|
Denaxa M, Neves G, Rabinowitz A, Kemlo S, Liodis P, Burrone J, Pachnis V. Modulation of Apoptosis Controls Inhibitory Interneuron Number in the Cortex. Cell Rep 2019; 22:1710-1721. [PMID: 29444425 PMCID: PMC6230259 DOI: 10.1016/j.celrep.2018.01.064] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 12/18/2017] [Accepted: 01/22/2018] [Indexed: 12/20/2022] Open
Abstract
Cortical networks are composed of excitatory projection neurons and inhibitory interneurons. Finding the right balance between the two is important for controlling overall cortical excitation and network dynamics. However, it is unclear how the correct number of cortical interneurons (CIs) is established in the mammalian forebrain. CIs are generated in excess from basal forebrain progenitors, and their final numbers are adjusted via an intrinsically determined program of apoptosis that takes place during an early postnatal window. Here, we provide evidence that the extent of CI apoptosis during this critical period is plastic and cell-type specific and can be reduced in a cell-autonomous manner by acute increases in neuronal activity. We propose that the physiological state of the emerging neural network controls the activity levels of local CIs to modulate their numbers in a homeostatic manner. Lhx6 is required for survival of CIs generated in the MGE MGE-derived CI loss is compensated for by a decrease in CGE-derived interneuron apoptosis Increases in cortical network activity are correlated with improved CI survival Transient, cell-autonomous depolarization improves the survival of grafted CIs
Collapse
Affiliation(s)
- Myrto Denaxa
- Nervous System Development and Homeostasis Laboratory, Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK.
| | - Guilherme Neves
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK.
| | - Adam Rabinowitz
- Bioinformatics and Biostatistics Laboratory, Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Sarah Kemlo
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| | - Petros Liodis
- Molecular Neurobiology, National Institute for Medical Research, the Ridgeway, Mill Hill, London NW7 1AA, UK
| | - Juan Burrone
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK.
| | - Vassilis Pachnis
- Nervous System Development and Homeostasis Laboratory, Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK.
| |
Collapse
|
49
|
Mukamel EA, Ngai J. Perspectives on defining cell types in the brain. Curr Opin Neurobiol 2019; 56:61-68. [PMID: 30530112 PMCID: PMC6551297 DOI: 10.1016/j.conb.2018.11.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 11/10/2018] [Accepted: 11/21/2018] [Indexed: 12/11/2022]
Abstract
The diversity of brain cell types was one of the earliest observations in modern neuroscience and continues to be one of the central concerns of current neuroscience research. Despite impressive recent progress, including single cell transcriptome and epigenome profiling as well as anatomical methods, we still lack a complete census or taxonomy of brain cell types. We argue this is due partly to the conceptual difficulty in defining a cell type. By considering the biological drivers of cell identity, such as networks of genes and gene regulatory elements, we propose a definition of cell type that emphasizes self-stabilizing regulation. We explore the predictions and hypotheses that arise from this definition. Integration of data from multiple modalities, including molecular profiling of genes and gene products, epigenetic landscape, cellular morphology, connectivity, and physiology, will be essential for a meaningful and broadly useful definition of brain cell types.
Collapse
Affiliation(s)
- Eran A Mukamel
- Department of Cognitive Science, University of California, San Diego, CA 92037, United States.
| | - John Ngai
- Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, QB3 Functional Genomics Laboratory, University of California, Berkeley, CA 94720, United States.
| |
Collapse
|
50
|
Xu R, Brawner AT, Li S, Liu JJ, Kim H, Xue H, Pang ZP, Kim WY, Hart RP, Liu Y, Jiang P. OLIG2 Drives Abnormal Neurodevelopmental Phenotypes in Human iPSC-Based Organoid and Chimeric Mouse Models of Down Syndrome. Cell Stem Cell 2019; 24:908-926.e8. [PMID: 31130512 DOI: 10.1016/j.stem.2019.04.014] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 10/05/2018] [Accepted: 04/17/2019] [Indexed: 02/06/2023]
Abstract
Down syndrome (DS) is a common neurodevelopmental disorder, and cognitive defects in DS patients may arise from imbalances in excitatory and inhibitory neurotransmission. Understanding the mechanisms underlying such imbalances may provide opportunities for therapeutic intervention. Here, we show that human induced pluripotent stem cells (hiPSCs) derived from DS patients overproduce OLIG2+ ventral forebrain neural progenitors. As a result, DS hiPSC-derived cerebral organoids excessively produce specific subclasses of GABAergic interneurons and cause impaired recognition memory in neuronal chimeric mice. Increased OLIG2 expression in DS cells directly upregulates interneuron lineage-determining transcription factors. shRNA-mediated knockdown of OLIG2 largely reverses abnormal gene expression in early-stage DS neural progenitors, reduces interneuron production in DS organoids and chimeric mouse brains, and improves behavioral deficits in DS chimeric mice. Thus, altered OLIG2 expression may underlie neurodevelopmental abnormalities and cognitive defects in DS patients.
Collapse
Affiliation(s)
- Ranjie Xu
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA; Department of Developmental Neuroscience, Munroe-Meyer Institute and Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Andrew T Brawner
- Department of Developmental Neuroscience, Munroe-Meyer Institute and Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Shenglan Li
- Department of Neurosurgery and Center for Stem Cell and Regenerative Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Jing-Jing Liu
- Department of Neuroscience and Cell Biology and Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Hyosung Kim
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Haipeng Xue
- Department of Neurosurgery and Center for Stem Cell and Regenerative Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Zhiping P Pang
- Department of Neuroscience and Cell Biology and Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Woo-Yang Kim
- Department of Biological Sciences, Kent State University, Kent, OH 44242, USA
| | - Ronald P Hart
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Ying Liu
- Department of Neurosurgery and Center for Stem Cell and Regenerative Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Peng Jiang
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA; Department of Developmental Neuroscience, Munroe-Meyer Institute and Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|