1
|
Goovaerts S, Hoskens H, Eller RJ, Herrick N, Musolf AM, Justice CM, Yuan M, Naqvi S, Lee MK, Vandermeulen D, Szabo-Rogers HL, Romitti PA, Boyadjiev SA, Marazita ML, Shaffer JR, Shriver MD, Wysocka J, Walsh S, Weinberg SM, Claes P. Joint multi-ancestry and admixed GWAS reveals the complex genetics behind human cranial vault shape. Nat Commun 2023; 14:7436. [PMID: 37973980 PMCID: PMC10654897 DOI: 10.1038/s41467-023-43237-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 11/01/2023] [Indexed: 11/19/2023] Open
Abstract
The cranial vault in humans is highly variable, clinically relevant, and heritable, yet its genetic architecture remains poorly understood. Here, we conduct a joint multi-ancestry and admixed multivariate genome-wide association study on 3D cranial vault shape extracted from magnetic resonance images of 6772 children from the ABCD study cohort yielding 30 genome-wide significant loci. Follow-up analyses indicate that these loci overlap with genomic risk loci for sagittal craniosynostosis, show elevated activity cranial neural crest cells, are enriched for processes related to skeletal development, and are shared with the face and brain. We present supporting evidence of regional localization for several of the identified genes based on expression patterns in the cranial vault bones of E15.5 mice. Overall, our study provides a comprehensive overview of the genetics underlying normal-range cranial vault shape and its relevance for understanding modern human craniofacial diversity and the etiology of congenital malformations.
Collapse
Affiliation(s)
- Seppe Goovaerts
- Department of Human Genetics, KU Leuven, Leuven, Belgium.
- Medical Imaging Research Center, University Hospitals Leuven, Leuven, Belgium.
| | - Hanne Hoskens
- Medical Imaging Research Center, University Hospitals Leuven, Leuven, Belgium
- Department of Electrical Engineering, ESAT/PSI, KU Leuven, Leuven, Belgium
| | - Ryan J Eller
- Department of Biology, Indiana University Indianapolis, Indianapolis, IN, USA
| | - Noah Herrick
- Department of Biology, Indiana University Indianapolis, Indianapolis, IN, USA
| | - Anthony M Musolf
- Statistical Genetics Section, Computational and Statistical Genomics Branch, NHGRI, NIH, MD, Baltimore, USA
| | - Cristina M Justice
- Genometrics Section, Computational and Statistical Genomics Branch, Division of Intramural Research, NHGRI, NIH, Baltimore, MD, USA
- Neurobehavioral Clinical Research Section, Social and Behavioral Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Meng Yuan
- Department of Human Genetics, KU Leuven, Leuven, Belgium
- Medical Imaging Research Center, University Hospitals Leuven, Leuven, Belgium
- Department of Electrical Engineering, ESAT/PSI, KU Leuven, Leuven, Belgium
| | - Sahin Naqvi
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
- Departments of Genetics and Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Myoung Keun Lee
- Department of Oral and Craniofacial Sciences, Center for Craniofacial and Dental Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Dirk Vandermeulen
- Medical Imaging Research Center, University Hospitals Leuven, Leuven, Belgium
- Department of Electrical Engineering, ESAT/PSI, KU Leuven, Leuven, Belgium
| | - Heather L Szabo-Rogers
- Department of Anatomy, Physiology and Pharmacology, University of Saskatchewan, Saskatchewan, Canada
| | - Paul A Romitti
- Department of Epidemiology, College of Public Health, The University of Iowa, Iowa City, IA, USA
| | - Simeon A Boyadjiev
- Department of Pediatrics, University of California Davis, Sacramento, CA, USA
| | - Mary L Marazita
- Department of Oral and Craniofacial Sciences, Center for Craniofacial and Dental Genetics, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - John R Shaffer
- Department of Oral and Craniofacial Sciences, Center for Craniofacial and Dental Genetics, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mark D Shriver
- Department of Anthropology, Pennsylvania State University, State College, PA, USA
| | - Joanna Wysocka
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Susan Walsh
- Department of Biology, Indiana University Indianapolis, Indianapolis, IN, USA
| | - Seth M Weinberg
- Department of Oral and Craniofacial Sciences, Center for Craniofacial and Dental Genetics, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Anthropology, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Peter Claes
- Department of Human Genetics, KU Leuven, Leuven, Belgium.
- Medical Imaging Research Center, University Hospitals Leuven, Leuven, Belgium.
- Department of Electrical Engineering, ESAT/PSI, KU Leuven, Leuven, Belgium.
- Murdoch Children's Research Institute, Melbourne, VIC, Australia.
| |
Collapse
|
2
|
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
|
3
|
Rudy MJ, Salois G, Cubello J, Newell R, Mayer-Proschel M. Gestational iron deficiency affects the ratio between interneuron subtypes in the postnatal cerebral cortex in mice. Development 2023; 150:dev201068. [PMID: 36805633 PMCID: PMC10110419 DOI: 10.1242/dev.201068] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 01/30/2023] [Indexed: 02/22/2023]
Abstract
Gestational iron deficiency (gID) is highly prevalent and associated with an increased risk of intellectual and developmental disabilities in affected individuals that are often defined by a disrupted balance of excitation and inhibition (E/I) in the brain. Using a nutritional mouse model of gID, we previously demonstrated a shift in the E/I balance towards increased inhibition in the brains of gID offspring that was refractory to postnatal iron supplementation. We thus tested whether gID affects embryonic progenitor cells that are fated towards inhibitory interneurons. We quantified relevant cell populations during embryonic inhibitory neuron specification and found an increase in the proliferation of Nkx2.1+ interneuron progenitors in the embryonic medial ganglionic eminence at E14 that was associated with increased Shh signaling in gID animals at E12. When we quantified the number of mature inhibitory interneurons that are known to originate from the MGE, we found a persistent disruption of differentiated interneuron subtypes in early adulthood. Our data identify a cellular target that links gID with a disruption of cortical interneurons which play a major role in the establishment of the E/I balance.
Collapse
Affiliation(s)
- Michael J. Rudy
- Department of Biomedical Genetics, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, USA
- Department of Neurology, University of Colorado Denver – Anschutz Medical Campus, 13001 East 17th Place, Aurora, CO 80045, USA
| | - Garrick Salois
- Department of Biomedical Genetics, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | - Janine Cubello
- Department of Biomedical Genetics, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | - Robert Newell
- Department of Biomedical Genetics, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | - Margot Mayer-Proschel
- Department of Biomedical Genetics, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, USA
| |
Collapse
|
4
|
Alcaide Martin A, Mayerl S. Local Thyroid Hormone Action in Brain Development. Int J Mol Sci 2023; 24:12352. [PMID: 37569727 PMCID: PMC10418487 DOI: 10.3390/ijms241512352] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 07/28/2023] [Accepted: 07/30/2023] [Indexed: 08/13/2023] Open
Abstract
Proper brain development essentially depends on the timed availability of sufficient amounts of thyroid hormone (TH). This, in turn, necessitates a tightly regulated expression of TH signaling components such as TH transporters, deiodinases, and TH receptors in a brain region- and cell-specific manner from early developmental stages onwards. Abnormal TH levels during critical stages, as well as mutations in TH signaling components that alter the global and/or local thyroidal state, result in detrimental consequences for brain development and neurological functions that involve alterations in central neurotransmitter systems. Thus, the question as to how TH signaling is implicated in the development and maturation of different neurotransmitter and neuromodulator systems has gained increasing attention. In this review, we first summarize the current knowledge on the regulation of TH signaling components during brain development. We then present recent advances in our understanding on how altered TH signaling compromises the development of cortical glutamatergic neurons, inhibitory GABAergic interneurons, cholinergic and dopaminergic neurons. Thereby, we highlight novel mechanistic insights and point out open questions in this evolving research field.
Collapse
Affiliation(s)
| | - Steffen Mayerl
- Department of Endocrinology Diabetes & Metabolism, University Hospital Essen, University of Duisburg-Essen, Hufelandstraße 55, 45147 Essen, Germany
| |
Collapse
|
5
|
Mann B, Crawford JC, Reddy K, Lott J, Youn YH, Gao G, Guy C, Chou CH, Darnell D, Trivedi S, Bomme P, Loughran AJ, Thomas PG, Han YG, Tuomanen EI. Bacterial TLR2/6 Ligands Block Ciliogenesis, Derepress Hedgehog Signaling, and Expand the Neocortex. mBio 2023; 14:e0051023. [PMID: 37052506 PMCID: PMC10294647 DOI: 10.1128/mbio.00510-23] [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: 02/27/2023] [Accepted: 03/21/2023] [Indexed: 04/14/2023] Open
Abstract
Microbial components have a range of direct effects on the fetal brain. However, little is known about the cellular targets and molecular mechanisms that mediate these effects. Neural progenitor cells (NPCs) control the size and architecture of the brain and understanding the mechanisms regulating NPCs is crucial to understanding brain developmental disorders. We identify ventricular radial glia (vRG), the primary NPC, as the target of bacterial cell wall (BCW) generated during the antibiotic treatment of maternal pneumonia. BCW enhanced proliferative potential of vRGs by shortening the cell cycle and increasing self-renewal. Expanded vRGs propagated to increase neuronal output in all cortical layers. Remarkably, Toll-like receptor 2 (TLR2), which recognizes BCW, localized at the base of primary cilia in vRGs and the BCW-TLR2 interaction suppressed ciliogenesis leading to derepression of Hedgehog (HH) signaling and expansion of vRGs. We also show that TLR6 is an essential partner of TLR2 in this process. Surprisingly, TLR6 alone was required to set the number of cortical neurons under healthy conditions. These findings suggest that an endogenous signal from TLRs suppresses cortical expansion during normal development of the neocortex and that BCW antagonizes that signal through the TLR2/cilia/HH signaling axis changing brain structure and function. IMPORTANCE Fetal brain development in early gestation can be impacted by transplacental infection, altered metabolites from the maternal microbiome, or maternal immune activation. It is less well understood how maternal microbial subcomponents that cross the placenta, such as bacterial cell wall (BCW), directly interact with fetal neural progenitors and neurons and affect development. This scenario plays out in the clinic when BCW debris released during antibiotic therapy of maternal infection traffics to the fetal brain. This study identifies the direct interaction of BCW with TLR2/6 present on the primary cilium, the signaling hub on fetal neural progenitor cells (NPCs). NPCs control the size and architecture of the brain and understanding the mechanisms regulating NPCs is crucial to understanding brain developmental disorders. Within a window of vulnerability before the appearance of fetal immune cells, the BCW-TLR2/6 interaction results in the inhibition of ciliogenesis, derepression of Sonic Hedgehog signaling, excess proliferation of neural progenitors, and abnormal cortical architecture. In the first example of TLR signaling linked to Sonic Hedgehog, BCW/TLR2/6 appears to act during fetal brain morphogenesis to play a role in setting the total cell number in the neocortex.
Collapse
Affiliation(s)
- Beth Mann
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Jeremy Chase Crawford
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Kavya Reddy
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Josi Lott
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Yong Ha Youn
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Geli Gao
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Cliff Guy
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Ching-Heng Chou
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Daniel Darnell
- Hartwell Center for Bioinformatics and Biotechnology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Sanchit Trivedi
- Hartwell Center for Bioinformatics and Biotechnology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Perrine Bomme
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Allister J. Loughran
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Paul G. Thomas
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Young-Goo Han
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Elaine I. Tuomanen
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
6
|
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
|
7
|
Hunt CPJ, Moriarty N, van Deursen CBJ, Gantner CW, Thompson LH, Parish CL. Understanding and modeling regional specification of the human ganglionic eminence. Stem Cell Reports 2023; 18:654-671. [PMID: 36801004 PMCID: PMC10031306 DOI: 10.1016/j.stemcr.2023.01.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 01/22/2023] [Accepted: 01/23/2023] [Indexed: 02/18/2023] Open
Abstract
Inhibitory neurons originating from the ventral forebrain are associated with several neurological conditions. Distinct ventral forebrain subpopulations are generated from topographically defined zones; lateral-, medial- and caudal ganglionic eminences (LGE, MGE and CGE), yet key specification factors often span across developing zones contributing to difficulty in defining unique LGE, MGE or CGE profiles. Here we use human pluripotent stem cell (hPSC) reporter lines (NKX2.1-GFP and MEIS2-mCherry) and manipulation of morphogen gradients to gain greater insight into regional specification of these distinct zones. We identified Sonic hedgehog (SHH)-WNT crosstalk in regulating LGE and MGE fate and uncovered a role for retinoic acid signaling in CGE development. Unraveling the influence of these signaling pathways permitted development of fully defined protocols that favored generation of the three GE domains. These findings provide insight into the context-dependent role of morphogens in human GE specification and are of value for in vitro disease modeling and advancement of new therapies.
Collapse
Affiliation(s)
- Cameron P J Hunt
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3052, Australia.
| | - Niamh Moriarty
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Coen B J van Deursen
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Carlos W Gantner
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Lachlan H Thompson
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Clare L Parish
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3052, Australia.
| |
Collapse
|
8
|
Fontes-Dutra M, Righes Marafiga J, Santos-Terra J, Deckmann I, Brum Schwingel G, Rabelo B, Kazmierzak de Moraes R, Rockenbach M, Vendramin Pasquetti M, Gottfried C, Calcagnotto ME. GABAergic synaptic transmission and cortical oscillation patterns in the primary somatosensory area of a valproic acid rat model of autism spectrum disorder. Eur J Neurosci 2023; 57:527-546. [PMID: 36504470 DOI: 10.1111/ejn.15893] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 11/20/2022] [Accepted: 11/30/2022] [Indexed: 12/14/2022]
Abstract
Autism spectrum disorder (ASD) is characterized by impaired social communication and interaction associated with repetitive or stereotyped behaviour. Prenatal valproic acid (VPA) exposure in rodents is a commonly used model of ASD. Resveratrol (RSV) has been shown to prevent interneuronal and behavioural impairments in the VPA model. We investigated the effects of prenatal VPA exposure and RSV on the GABAergic synaptic transmission, brain oscillations and on the genic expression of interneuron-associated transcription factor LHX6 in the primary somatosensory area (PSSA). Prenatal VPA exposure decreased the sIPSC and mIPSC frequencies and the sIPSC decay kinetics onto layers 4/5 pyramidal cells of PSSA. About 40% of VPA animals exhibited absence-like spike-wave discharge (SWD) events associated with behaviour arrest and increased power spectrum density of delta, beta and gamma cortical oscillations. VPA animals had reduced LHX6 expression in PSSA, but VPA animals treated with RSV had no changes on synaptic inhibition or LHX6 expression in the PSSA. SWD events associated with behaviour arrest and the abnormal increment of cortical oscillations were also absent in VPA animals treated with RSV. These findings provide new venues to investigate the role of both RSV and VPA in the pathophysiology of ASD and highlight the VPA animal model as an interesting tool to investigate pathways related to the aetiology and possible future therapies to this neuropsychiatric disorder.
Collapse
Affiliation(s)
- Mellanie Fontes-Dutra
- Translational Research Group in Autism Spectrum Disorders (GETTEA), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Joseane Righes Marafiga
- Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Graduate Program in Biological Science: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Neurophysiology and Neurochemistry of Neuronal Excitability and Synaptic Plasticity Laboratory, Department of Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Júlio Santos-Terra
- Translational Research Group in Autism Spectrum Disorders (GETTEA), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Iohanna Deckmann
- Translational Research Group in Autism Spectrum Disorders (GETTEA), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Gustavo Brum Schwingel
- Translational Research Group in Autism Spectrum Disorders (GETTEA), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Bruna Rabelo
- Translational Research Group in Autism Spectrum Disorders (GETTEA), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Rafael Kazmierzak de Moraes
- Translational Research Group in Autism Spectrum Disorders (GETTEA), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Marília Rockenbach
- Translational Research Group in Autism Spectrum Disorders (GETTEA), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Mayara Vendramin Pasquetti
- Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Graduate Program in Biological Science: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Neurophysiology and Neurochemistry of Neuronal Excitability and Synaptic Plasticity Laboratory, Department of Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Carmem Gottfried
- Translational Research Group in Autism Spectrum Disorders (GETTEA), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Maria Elisa Calcagnotto
- Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Graduate Program in Biological Science: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Neurophysiology and Neurochemistry of Neuronal Excitability and Synaptic Plasticity Laboratory, Department of Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Graduate Program in Neuroscience, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
9
|
Mueller-Buehl C, Wegrzyn D, Bauch J, Faissner A. Regulation of the E/I-balance by the neural matrisome. Front Mol Neurosci 2023; 16:1102334. [PMID: 37143468 PMCID: PMC10151766 DOI: 10.3389/fnmol.2023.1102334] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 03/27/2023] [Indexed: 05/06/2023] Open
Abstract
In the mammalian cortex a proper excitatory/inhibitory (E/I) balance is fundamental for cognitive functions. Especially γ-aminobutyric acid (GABA)-releasing interneurons regulate the activity of excitatory projection neurons which form the second main class of neurons in the cortex. During development, the maturation of fast-spiking parvalbumin-expressing interneurons goes along with the formation of net-like structures covering their soma and proximal dendrites. These so-called perineuronal nets (PNNs) represent a specialized form of the extracellular matrix (ECM, also designated as matrisome) that stabilize structural synapses but prevent the formation of new connections. Consequently, PNNs are highly involved in the regulation of the synaptic balance. Previous studies revealed that the formation of perineuronal nets is accompanied by an establishment of mature neuronal circuits and by a closure of critical windows of synaptic plasticity. Furthermore, it has been shown that PNNs differentially impinge the integrity of excitatory and inhibitory synapses. In various neurological and neuropsychiatric disorders alterations of PNNs were described and aroused more attention in the last years. The following review gives an update about the role of PNNs for the maturation of parvalbumin-expressing interneurons and summarizes recent findings about the impact of PNNs in different neurological and neuropsychiatric disorders like schizophrenia or epilepsy. A targeted manipulation of PNNs might provide an interesting new possibility to indirectly modulate the synaptic balance and the E/I ratio in pathological conditions.
Collapse
|
10
|
Somaiya RD, Stebbins K, Gingrich EC, Xie H, Campbell JN, Garcia ADR, Fox MA. Sonic hedgehog-dependent recruitment of GABAergic interneurons into the developing visual thalamus. eLife 2022; 11:e79833. [PMID: 36342840 PMCID: PMC9640189 DOI: 10.7554/elife.79833] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 10/10/2022] [Indexed: 11/09/2022] Open
Abstract
Axons of retinal ganglion cells (RGCs) play critical roles in the development of inhibitory circuits in visual thalamus. We previously reported that RGC axons signal astrocytes to induce the expression of fibroblast growth factor 15 (FGF15), a motogen required for GABAergic interneuron migration into visual thalamus. However, how retinal axons induce thalamic astrocytes to generate Fgf15 and influence interneuron migration remains unknown. Here, we demonstrate that impairing RGC activity had little impact on interneuron recruitment into mouse visual thalamus. Instead, our data show that retinal-derived sonic hedgehog (SHH) is essential for interneuron recruitment. Specifically, we show that thalamus-projecting RGCs express SHH and thalamic astrocytes generate downstream components of SHH signaling. Deletion of RGC-derived SHH leads to a significant decrease in Fgf15 expression, as well as in the percentage of interneurons recruited into visual thalamus. Overall, our findings identify a morphogen-dependent neuron-astrocyte signaling mechanism essential for the migration of thalamic interneurons.
Collapse
Affiliation(s)
- Rachana Deven Somaiya
- Center for Neurobiology Research, Fralin Biomedical Research Institute at Virginia Tech CarilionRoanokeUnited States
- Graduate Program in Translational Biology, Medicine, and Health, Virginia TechBlacksburgUnited States
| | - Katelyn Stebbins
- Center for Neurobiology Research, Fralin Biomedical Research Institute at Virginia Tech CarilionRoanokeUnited States
- Graduate Program in Translational Biology, Medicine, and Health, Virginia TechBlacksburgUnited States
- Virginia Tech Carilion School of MedicineRoanokeUnited States
| | - Ellen C Gingrich
- Department of Biology, Drexel UniversityPhiladelphiaUnited States
- Department of Neurobiology and Anatomy, Drexel University College of MedicinePhiladelphiaUnited States
| | - Hehuang Xie
- Fralin Life Sciences Institute at Virginia TechBlacksburgUnited States
- School of Neuroscience, College of Science, Virginia TechBlacksburgUnited States
- Genetics, Bioinformatics and Computational Biology Program, Virginia TechBlacksburgUnited States
- Department of Biomedical Sciences and Pathobiology, Virginia–Maryland College of Veterinary MedicineBlacksburgUnited States
| | - John N Campbell
- Department of Biology, University of VirginiaCharlottesvilleUnited States
- Neuroscience Graduate Program, University of VirginiaCharlottesvilleUnited States
| | - A Denise R Garcia
- Department of Biology, Drexel UniversityPhiladelphiaUnited States
- Department of Neurobiology and Anatomy, Drexel University College of MedicinePhiladelphiaUnited States
| | - Michael A Fox
- Center for Neurobiology Research, Fralin Biomedical Research Institute at Virginia Tech CarilionRoanokeUnited States
- School of Neuroscience, College of Science, Virginia TechBlacksburgUnited States
- Department of Biological Sciences, College of Science, Virginia TechBlacksburgUnited States
- Department of Pediatrics, Virginia Tech Carilion School of MedicineRoanokeUnited States
| |
Collapse
|
11
|
Giffin-Rao Y, Sheng J, Strand B, Xu K, Huang L, Medo M, Risgaard KA, Dantinne S, Mohan S, Keshan A, Daley RA, Levesque B, Amundson L, Reese R, Sousa AMM, Tao Y, Wang D, Zhang SC, Bhattacharyya A. Altered patterning of trisomy 21 interneuron progenitors. Stem Cell Reports 2022; 17:1366-1379. [PMID: 35623352 PMCID: PMC9214050 DOI: 10.1016/j.stemcr.2022.05.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 05/01/2022] [Accepted: 05/02/2022] [Indexed: 11/17/2022] Open
Abstract
Individuals with Down syndrome (DS; Ts21), the most common genetic cause of intellectual disability, have smaller brains that reflect fewer neurons at pre- and post-natal stages, implicating impaired neurogenesis during development. Our stereological analysis of adult DS cortex indicates a reduction of calretinin-expressing interneurons. Using Ts21 human induced pluripotent stem cells (iPSCs) and isogenic controls, we find that Ts21 progenitors generate fewer COUP-TFII+ progenitors with reduced proliferation. Single-cell RNA sequencing of Ts21 progenitors confirms the altered specification of progenitor subpopulations and identifies reduced WNT signaling. Activation of WNT signaling partially restores the COUP-TFII+ progenitor population in Ts21, suggesting that altered WNT signaling contributes to the defective development of cortical interneurons in DS.
Collapse
Affiliation(s)
| | - Jie Sheng
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Bennett Strand
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Ke Xu
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Leslie Huang
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Margaret Medo
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | | | - Samuel Dantinne
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Sruti Mohan
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Aratrika Keshan
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Roger A Daley
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Bradley Levesque
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Lindsey Amundson
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Rebecca Reese
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - André M M Sousa
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Yunlong Tao
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Daifeng Wang
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Su-Chun Zhang
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neurology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Anita Bhattacharyya
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA.
| |
Collapse
|
12
|
Leung RF, George AM, Roussel EM, Faux MC, Wigle JT, Eisenstat DD. Genetic Regulation of Vertebrate Forebrain Development by Homeobox Genes. Front Neurosci 2022; 16:843794. [PMID: 35546872 PMCID: PMC9081933 DOI: 10.3389/fnins.2022.843794] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 03/14/2022] [Indexed: 01/19/2023] Open
Abstract
Forebrain development in vertebrates is regulated by transcription factors encoded by homeobox, bHLH and forkhead gene families throughout the progressive and overlapping stages of neural induction and patterning, regional specification and generation of neurons and glia from central nervous system (CNS) progenitor cells. Moreover, cell fate decisions, differentiation and migration of these committed CNS progenitors are controlled by the gene regulatory networks that are regulated by various homeodomain-containing transcription factors, including but not limited to those of the Pax (paired), Nkx, Otx (orthodenticle), Gsx/Gsh (genetic screened), and Dlx (distal-less) homeobox gene families. This comprehensive review outlines the integral role of key homeobox transcription factors and their target genes on forebrain development, focused primarily on the telencephalon. Furthermore, links of these transcription factors to human diseases, such as neurodevelopmental disorders and brain tumors are provided.
Collapse
Affiliation(s)
- Ryan F. Leung
- Murdoch Children’s Research Institute, The Royal Children’s Hospital Melbourne, Parkville, VIC, Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
| | - Ankita M. George
- Murdoch Children’s Research Institute, The Royal Children’s Hospital Melbourne, Parkville, VIC, Australia
| | - Enola M. Roussel
- Murdoch Children’s Research Institute, The Royal Children’s Hospital Melbourne, Parkville, VIC, Australia
| | - Maree C. Faux
- Murdoch Children’s Research Institute, The Royal Children’s Hospital Melbourne, Parkville, VIC, Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - Jeffrey T. Wigle
- Department of Biochemistry and Medical Genetics, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada
| | - David D. Eisenstat
- Murdoch Children’s Research Institute, The Royal Children’s Hospital Melbourne, Parkville, VIC, Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
- Department of Medical Genetics, University of Alberta, Edmonton, AB, Canada
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
13
|
Korade Z, Heffer M, Mirnics K. Medication effects on developmental sterol biosynthesis. Mol Psychiatry 2022; 27:490-501. [PMID: 33820938 PMCID: PMC8490477 DOI: 10.1038/s41380-021-01074-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 03/01/2021] [Accepted: 03/19/2021] [Indexed: 02/01/2023]
Abstract
Cholesterol is essential for normal brain function and development. Genetic disruptions of sterol biosynthesis result in intellectual and developmental disabilities. Developing neurons synthesize their own cholesterol, and disruption of this process can occur by both genetic and chemical mechanisms. Many commonly prescribed medications interfere with sterol biosynthesis, including haloperidol, aripiprazole, cariprazine, fluoxetine, trazodone and amiodarone. When used during pregnancy, these compounds might have detrimental effects on the developing brain of the offspring. In particular, inhibition of dehydrocholesterol-reductase 7 (DHCR7), the last enzyme in the biosynthesis pathway, results in accumulation of the immediate cholesterol precursor, 7-dehydrocholesterol (7-DHC). 7-DHC is highly unstable, giving rise to toxic oxysterols; this is particularly pronounced in a mouse model when both the mother and the offspring carry the Dhcr7+/- genotype. Studies of human dermal fibroblasts from individuals who carry DCHR7+/- single allele mutations suggest that the same gene*medication interaction also occurs in humans. The public health relevance of these findings is high, as DHCR7-inhibitors can be considered teratogens, and are commonly used by pregnant women. In addition, sterol biosynthesis inhibiting medications should be used with caution in individuals with mutations in sterol biosynthesis genes. In an age of precision medicine, further research in this area could open opportunities to improve patient and fetal/infant safety by tailoring medication prescriptions according to patient genotype and life stage.
Collapse
Affiliation(s)
- Zeljka Korade
- Department of Pediatrics, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA, 68198.,Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA, 68198
| | - Marija Heffer
- J. J. Strossmayer University of Osijek, Faculty of Medicine Osijek, Department of Medical Biology and Genetics, Josipa Huttlera 4, 31000 Osijek, Croatia
| | - Károly Mirnics
- Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA. .,Munroe-Meyer Institute for Genetics and Rehabilitation, University of Nebraska Medical Center, Omaha, NE, 68105, USA.
| |
Collapse
|
14
|
Krajka V, Naujock M, Pauly MG, Stengel F, Meier B, Stanslowsky N, Klein C, Seibler P, Wegner F, Capetian P. Ventral Telencephalic Patterning Protocols for Induced Pluripotent Stem Cells. Front Cell Dev Biol 2021; 9:716249. [PMID: 34490265 PMCID: PMC8416478 DOI: 10.3389/fcell.2021.716249] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 07/30/2021] [Indexed: 11/25/2022] Open
Abstract
The differentiation of human induced pluripotent stem cells (hiPSCs) into specific cell types for disease modeling and restorative therapies is a key research agenda and offers the possibility to obtain patient-specific cells of interest for a wide range of diseases. Basal forebrain cholinergic neurons (BFCNs) play a particular role in the pathophysiology of Alzheimer’s dementia and isolated dystonias. In this work, various directed differentiation protocols based on monolayer neural induction were tested for their effectiveness in promoting a ventral telencephalic phenotype and generating BFCN. Ventralizing factors [i.e., purmorphamine and Sonic hedgehog (SHH)] were applied at different time points, time intervals, and concentrations. In addition, caudal identity was prevented by the use of a small molecule XAV-939 that inhibits the Wnt-pathway. After patterning, gene expression profiles were analyzed by quantitative PCR (qPCR). Rostro-ventral patterning is most effective when initiated simultaneously with neural induction. The most promising combination of patterning factors was 0.5 μM of purmorphamine and 1 μM of XAV-939, which induces the highest expression of transcription factors specific for the medial ganglionic eminence, the source of GABAergic inter- and cholinergic neurons in the telencephalon. Upon maturation of cells, the immune phenotype, as well as electrophysiological properties were investigated showing the presence of marker proteins specific for BFCN (choline acetyltransferase, ISL1, p75, and NKX2.1) and GABAergic neurons. Moreover, a considerable fraction of measured cells displayed mature electrophysiological properties. Synaptic boutons containing the vesicular acetylcholine transporter (VACHT) could be observed in the vicinity of the cells. This work will help to generate basal forebrain interneurons from hiPSCs, providing a promising platform for modeling neurological diseases, such as Alzheimer’s disease or Dystonia.
Collapse
Affiliation(s)
- Victor Krajka
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | | | - Martje G Pauly
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Felix Stengel
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Britta Meier
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | | | - Christine Klein
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Philip Seibler
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Florian Wegner
- Department of Neurology, Hannover Medical School, Hanover, Germany
| | - Philipp Capetian
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany.,Department of Neurology, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
15
|
Development, Diversity, and Death of MGE-Derived Cortical Interneurons. Int J Mol Sci 2021; 22:ijms22179297. [PMID: 34502208 PMCID: PMC8430628 DOI: 10.3390/ijms22179297] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/24/2021] [Accepted: 08/25/2021] [Indexed: 12/17/2022] Open
Abstract
In the mammalian brain, cortical interneurons (INs) are a highly diverse group of cells. A key neurophysiological question concerns how each class of INs contributes to cortical circuit function and whether specific roles can be attributed to a selective cell type. To address this question, researchers are integrating knowledge derived from transcriptomic, histological, electrophysiological, developmental, and functional experiments to extensively characterise the different classes of INs. Our hope is that such knowledge permits the selective targeting of cell types for therapeutic endeavours. This review will focus on two of the main types of INs, namely the parvalbumin (PV+) or somatostatin (SOM+)-containing cells, and summarise the research to date on these classes.
Collapse
|
16
|
Mayerl S, Chen J, Salveridou E, Boelen A, Darras VM, Heuer H. Thyroid Hormone Transporter Deficiency in Mice Impacts Multiple Stages of GABAergic Interneuron Development. Cereb Cortex 2021; 32:329-341. [PMID: 34339499 PMCID: PMC8754375 DOI: 10.1093/cercor/bhab211] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 05/11/2021] [Accepted: 06/01/2021] [Indexed: 11/14/2022] Open
Abstract
Cortical interneuron neurogenesis is strictly regulated and depends on the presence of thyroid hormone (TH). In particular, inhibitory interneurons expressing the calcium binding protein Parvalbumin are highly sensitive toward developmental hypothyroidism. Reduced numbers of Parvalbumin-positive interneurons are observed in mice due to the combined absence of the TH transporters Mct8 and Oatp1c1. To unravel if cortical Parvalbumin-positive interneurons depend on cell-autonomous action of Mct8/Oatp1c1, we compared Mct8/Oatp1c1 double knockout (dko) mice to conditional knockouts with abolished TH transporter expression in progenitors of Parvalbumin-positive interneurons. These conditional knockouts exhibited a transient delay in the appearance of Parvalbumin-positive interneurons in the early postnatal somatosensory cortex while cell numbers remained permanently reduced in Mct8/Oatp1c1 dko mice. Using fluorescence in situ hybridization on E12.5 embryonic brains, we detected reduced expression of sonic hedgehog signaling components in Mct8/Oatp1c1 dko embryos only. Moreover, we revealed spatially distinct expression patterns of both TH transporters at brain barriers at E12.5 by immunofluorescence. At later developmental stages, we uncovered a sequential expression of first Oatp1c1 in individual interneurons and then Mct8 in Parvalbumin-positive subtypes. Together, our results point to multiple cell-autonomous and noncell-autonomous mechanisms that depend on proper TH transport during cortical interneuron development.
Collapse
Affiliation(s)
- Steffen Mayerl
- Leibniz Institute on Aging/Fritz Lipmann Institute, 07745 Jena, Germany.,MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK.,Department of Endocrinology, Diabetes and Metabolism; University Duisburg-Essen, 45147 Essen, Germany
| | - Jiesi Chen
- Leibniz Institute on Aging/Fritz Lipmann Institute, 07745 Jena, Germany.,Leibniz Research Institute for Environmental Medicine, 40225 Düsseldorf, Germany
| | - Eva Salveridou
- Department of Endocrinology, Diabetes and Metabolism; University Duisburg-Essen, 45147 Essen, Germany.,Leibniz Research Institute for Environmental Medicine, 40225 Düsseldorf, Germany
| | - Anita Boelen
- Endocrinology Laboratory, Department of Clinical Chemistry, Amsterdam Gastroenterology & Metabolism, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
| | - Veerle M Darras
- Laboratory of Comparative Endocrinology, Animal Physiology and Neurobiology Section, Biology Department, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Heike Heuer
- Leibniz Institute on Aging/Fritz Lipmann Institute, 07745 Jena, Germany.,Department of Endocrinology, Diabetes and Metabolism; University Duisburg-Essen, 45147 Essen, Germany.,Leibniz Research Institute for Environmental Medicine, 40225 Düsseldorf, Germany
| |
Collapse
|
17
|
Martinez JL, Zammit MD, West NR, Christian BT, Bhattacharyya A. Basal Forebrain Cholinergic Neurons: Linking Down Syndrome and Alzheimer's Disease. Front Aging Neurosci 2021; 13:703876. [PMID: 34322015 PMCID: PMC8311593 DOI: 10.3389/fnagi.2021.703876] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/17/2021] [Indexed: 12/31/2022] Open
Abstract
Down syndrome (DS, trisomy 21) is characterized by intellectual impairment at birth and Alzheimer's disease (AD) pathology in middle age. As individuals with DS age, their cognitive functions decline as they develop AD pathology. The susceptibility to degeneration of a subset of neurons, known as basal forebrain cholinergic neurons (BFCNs), in DS and AD is a critical link between cognitive impairment and neurodegeneration in both disorders. BFCNs are the primary source of cholinergic innervation to the cerebral cortex and hippocampus, as well as the amygdala. They play a critical role in the processing of information related to cognitive function and are directly engaged in regulating circuits of attention and memory throughout the lifespan. Given the importance of BFCNs in attention and memory, it is not surprising that these neurons contribute to dysfunctional neuronal circuitry in DS and are vulnerable in adults with DS and AD, where their degeneration leads to memory loss and disturbance in language. BFCNs are thus a relevant cell target for therapeutics for both DS and AD but, despite some success, efforts in this area have waned. There are gaps in our knowledge of BFCN vulnerability that preclude our ability to effectively design interventions. Here, we review the role of BFCN function and degeneration in AD and DS and identify under-studied aspects of BFCN biology. The current gaps in BFCN relevant imaging studies, therapeutics, and human models limit our insight into the mechanistic vulnerability of BFCNs in individuals with DS and AD.
Collapse
Affiliation(s)
- Jose L. Martinez
- Cellular and Molecular Biology Graduate Program, University of Wisconsin, Madison, WI, United States
- Waisman Center, University of Wisconsin, Madison, WI, United States
| | - Matthew D. Zammit
- Waisman Center, University of Wisconsin, Madison, WI, United States
- Department of Medical Physics, School of Medicine and Public Health, University of Wisconsin, Madison, WI, United States
| | - Nicole R. West
- Cellular and Molecular Biology Graduate Program, University of Wisconsin, Madison, WI, United States
- Waisman Center, University of Wisconsin, Madison, WI, United States
| | - Bradley T. Christian
- Waisman Center, University of Wisconsin, Madison, WI, United States
- Department of Medical Physics, School of Medicine and Public Health, University of Wisconsin, Madison, WI, United States
- Department of Psychiatry, School of Medicine and Public Health, University of Wisconsin, Madison, WI, United States
| | - Anita Bhattacharyya
- Waisman Center, University of Wisconsin, Madison, WI, United States
- Department of Cellular and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, United States
| |
Collapse
|
18
|
Nasu M, Esumi S, Hatakeyama J, Tamamaki N, Shimamura K. Two-Phase Lineage Specification of Telencephalon Progenitors Generated From Mouse Embryonic Stem Cells. Front Cell Dev Biol 2021; 9:632381. [PMID: 33937233 PMCID: PMC8086603 DOI: 10.3389/fcell.2021.632381] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 03/09/2021] [Indexed: 12/16/2022] Open
Abstract
Proper brain development requires precisely controlled phases of stem cell proliferation, lineage specification, differentiation, and migration. Lineage specification depends partly on concentration gradients of chemical cues called morphogens. However, the rostral brain (telencephalon) expands prominently during embryonic development, dynamically altering local morphogen concentrations, and telencephalic subregional properties develop with a time lag. Here, we investigated how progenitor specification occurs under these spatiotemporally changing conditions using a three-dimensional in vitro differentiation model. We verified the critical contributions of three signaling factors for the lineage specification of subregional tissues in the telencephalon, ventralizing sonic hedgehog (Shh) and dorsalizing bone morphogenetic proteins (BMPs) and WNT proteins (WNTs). We observed that a short-lasting signal is sufficient to induce subregional progenitors and that the timing of signal exposure for efficient induction is specific to each lineage. Furthermore, early and late progenitors possess different Shh signal response capacities. This study reveals a novel developmental mechanism for telencephalon patterning that relies on the interplay of dose- and time-dependent signaling, including a time lag for specification and a temporal shift in cellular Shh sensitivity. This delayed fate choice through two-phase specification allows tissues with marked size expansion, such as the telencephalon, to compensate for the changing dynamics of morphogen signals.
Collapse
Affiliation(s)
- Makoto Nasu
- Department of Health Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Shigeyuki Esumi
- Department of Anatomy and Neurobiology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Jun Hatakeyama
- Department of Brain Morphogenesis, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Nobuaki Tamamaki
- Department of Morphological Neural Science, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kenji Shimamura
- Department of Brain Morphogenesis, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
19
|
Shqirat M, Kinoshita A, Kageyama R, Ohtsuka T. Sonic hedgehog expands neural stem cells in the neocortical region leading to an expanded and wrinkled neocortical surface. Genes Cells 2021; 26:399-410. [PMID: 33811429 DOI: 10.1111/gtc.12847] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 03/31/2021] [Indexed: 01/08/2023]
Abstract
An expanded and folded neocortex is characteristic of higher mammals, including humans and other primates. The neocortical surface area was dramatically enlarged during the course of mammalian brain evolution from lissencephalic to gyrencephalic mammals, and this bestowed higher cognitive functions especially to primates, including humans. In this study, we generated transgenic (Tg) mice in which the expression of Sonic hedgehog (Shh) could be controlled in neural stem cells (NSCs) and neural progenitors by using the Tet-on system. Shh overexpression during embryogenesis promoted the symmetric proliferative division of NSCs in the neocortical region, leading to the expansion of lateral ventricles and tangential extension of the ventricular zone. Moreover, Shh-overexpressing Tg mice showed dramatic expansion of the neocortical surface area and exhibited a wrinkled brain when overexpression was commenced at early stages of neural development. These results indicate that Shh is able to increase the neocortical NSCs and contribute to expansion of the neocortex.
Collapse
Affiliation(s)
- Mohammed Shqirat
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan.,Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akira Kinoshita
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan.,Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Ryoichiro Kageyama
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan.,Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Graduate School of Biostudies, Kyoto University, Kyoto, Japan.,Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Kyoto, Japan.,RIKEN Center for Brain Science, Wako, Japan
| | - Toshiyuki Ohtsuka
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan.,Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| |
Collapse
|
20
|
Sobrido-Cameán D, Yáñez-Guerra LA, Deber A, Freire-Delgado M, Cacheiro-Vázquez R, Rodicio MC, Tostivint H, Anadón R, Barreiro-Iglesias A. Differential expression of somatostatin genes in the central nervous system of the sea lamprey. Brain Struct Funct 2021; 226:1031-1052. [PMID: 33532926 DOI: 10.1007/s00429-021-02224-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 01/19/2021] [Indexed: 01/29/2023]
Abstract
The identification of three somatostatin (SST) genes (SSTa, SSTb, and SSTc) in lampreys (Tostivint et al. Gen Comp Endocrinol 237:89-97 https://doi.org/10.1016/j.ygcen.2016.08.006 , 2016) prompted us to study their expression in the brain and spinal cord of the sea lamprey by in situ hybridization. These three genes were only expressed in equivalent neuronal populations in the hypothalamus. In other regions, SST transcripts showed clear differential expression. In the telencephalon, SSTc-positive cells were observed in the medial pallium, ventral part of the lateral pallium, striatum, subhippocampal lobe, and preoptic region. In the diencephalon, SSTa-positive cells were observed in the thalamus and SSTc-positive cells in the prethalamus, posterior tubercle, pretectal area, and nucleus of the medial longitudinal fascicle. In the midbrain, SSTc-positive cells were observed in the torus semicircularis, lateral reticular area, and perioculomotor tegmentum. Different SSTa- and SSTc-positive populations were observed in the isthmus. SSTc neurons were also observed in the rostral octavolateralis area and caudal rhombencephalon. In the spinal cord, SSTa was expressed in cerebrospinal-fluid-contacting (CSF-c) neurons and SSTc in non-CSF-c interneurons. Comparison with previous immunohistochemical studies using anti-SST-14 antibodies strongly suggests that SST-14-like neurons correspond with the SSTa populations. Thus, the SSTc populations were not reported previously in immunohistochemical studies. Cluster-based analyses and alignments of mature peptides suggested that SSTa is an ortholog of SST1 and that SSTb is closely related to SST2 and SST6. These results provide important new insights into the evolution of the somatostatinergic system in vertebrates.
Collapse
Affiliation(s)
- D Sobrido-Cameán
- Department of Functional Biology, Faculty of Biology, CIBUS, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, A Coruña, Spain.,Department of Zoology, University of Cambridge, Cambridge, UK
| | | | - A Deber
- Department of Functional Biology, Faculty of Biology, CIBUS, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, A Coruña, Spain
| | - M Freire-Delgado
- Department of Functional Biology, Faculty of Biology, CIBUS, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, A Coruña, Spain
| | - R Cacheiro-Vázquez
- Department of Functional Biology, Faculty of Biology, CIBUS, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, A Coruña, Spain
| | - M C Rodicio
- Department of Functional Biology, Faculty of Biology, CIBUS, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, A Coruña, Spain
| | - H Tostivint
- Molecular Physiology and Adaptation, UMR7221, CNRS and Muséum National D'Histoire Naturelle, Paris, France
| | - R Anadón
- Department of Functional Biology, Faculty of Biology, CIBUS, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, A Coruña, Spain
| | - A Barreiro-Iglesias
- Department of Functional Biology, Faculty of Biology, CIBUS, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, A Coruña, Spain.
| |
Collapse
|
21
|
Rivera-González KS, Beames TG, Lipinski RJ. Examining the developmental toxicity of piperonyl butoxide as a Sonic hedgehog pathway inhibitor. CHEMOSPHERE 2021; 264:128414. [PMID: 33007564 PMCID: PMC9158378 DOI: 10.1016/j.chemosphere.2020.128414] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/17/2020] [Accepted: 09/18/2020] [Indexed: 05/05/2023]
Abstract
Piperonyl butoxide (PBO) is a semisynthetic chemical present in hundreds of pesticide formulations used in agricultural, commercial, and residential settings. PBO acts as a pesticide synergist by inhibiting insect cytochrome P450 enzymes and is often present at much higher concentrations than active insecticidal ingredients. PBO was recently discovered to also inhibit Sonic hedgehog (Shh) signaling, a key molecular pathway in embryonic development and in brain and face morphogenesis. Recent animal model studies have shown that in utero PBO exposure can cause overt craniofacial malformations or more subtle neurodevelopmental abnormalities. Related adverse developmental outcomes in humans are etiologically heterogeneous, and, while studies are limited, PBO exposure during pregnancy has been linked to neurodevelopmental deficits. Contextualized in PBO's newly recognized mechanism as a Shh signaling inhibitor, these findings support more rigorous examination of the developmental toxicity of PBO and its potential contribution to etiologically complex human birth defects. In this review, we highlight environmental sources of human PBO exposure and summarize existing animal studies examining the developmental impact of prenatal PBO exposure. Also presented are critical knowledge gaps in our understanding of PBO's pharmacokinetics and potential role in gene-environment and environment-environment interactions that should be addressed to better understand the human health impact of environmental PBO exposure.
Collapse
Affiliation(s)
- Kenneth S Rivera-González
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, 53706, USA; Molecular and Environmental Toxicology Graduate Training Program, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Tyler G Beames
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, 53706, USA; Molecular and Environmental Toxicology Graduate Training Program, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Robert J Lipinski
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, 53706, USA; Molecular and Environmental Toxicology Graduate Training Program, University of Wisconsin-Madison, Madison, WI, 53706, USA.
| |
Collapse
|
22
|
Turrero García M, Baizabal JM, Tran DN, Peixoto R, Wang W, Xie Y, Adam MA, English LA, Reid CM, Brito SI, Booker MA, Tolstorukov MY, Harwell CC. Transcriptional regulation of MGE progenitor proliferation by PRDM16 controls cortical GABAergic interneuron production. Development 2020; 147:dev187526. [PMID: 33060132 PMCID: PMC7687860 DOI: 10.1242/dev.187526] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 10/05/2020] [Indexed: 11/20/2022]
Abstract
The mammalian cortex is populated by neurons derived from neural progenitors located throughout the embryonic telencephalon. Excitatory neurons are derived from the dorsal telencephalon, whereas inhibitory interneurons are generated in its ventral portion. The transcriptional regulator PRDM16 is expressed by radial glia, neural progenitors present in both regions; however, its mechanisms of action are still not fully understood. It is unclear whether PRDM16 plays a similar role in neurogenesis in both dorsal and ventral progenitor lineages and, if so, whether it regulates common or unique networks of genes. Here, we show that Prdm16 expression in mouse medial ganglionic eminence (MGE) progenitors is required for maintaining their proliferative capacity and for the production of proper numbers of forebrain GABAergic interneurons. PRDM16 binds to cis-regulatory elements and represses the expression of region-specific neuronal differentiation genes, thereby controlling the timing of neuronal maturation. PRDM16 regulates convergent developmental gene expression programs in the cortex and MGE, which utilize both common and region-specific sets of genes to control the proliferative capacity of neural progenitors, ensuring the generation of correct numbers of cortical neurons.
Collapse
Affiliation(s)
| | | | - Diana N Tran
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Rui Peixoto
- Howard Hughes Medical Institute, Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Wengang Wang
- Howard Hughes Medical Institute, Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Yajun Xie
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Manal A Adam
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Lauren A English
- Summer Honors Undergraduate Research Program, Harvard Medical School, Boston, MA 02115, USA
| | - Christopher M Reid
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Salvador I Brito
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Matthew A Booker
- Department of Informatics and Analytics, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Michael Y Tolstorukov
- Department of Informatics and Analytics, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Corey C Harwell
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
23
|
Groves I, Placzek M, Fletcher AG. Of mitogens and morphogens: modelling Sonic Hedgehog mechanisms in vertebrate development. Philos Trans R Soc Lond B Biol Sci 2020; 375:20190660. [PMID: 32829689 PMCID: PMC7482217 DOI: 10.1098/rstb.2019.0660] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/10/2020] [Indexed: 12/22/2022] Open
Abstract
Sonic Hedgehog (Shh) Is a critical protein in vertebrate development, orchestrating patterning and growth in many developing systems. First described as a classic morphogen that patterns tissues through a spatial concentration gradient, subsequent studies have revealed a more complex mechanism, in which Shh can also regulate proliferation and differentiation. While the mechanism of action of Shh as a morphogen is well understood, it remains less clear how Shh might integrate patterning, proliferation and differentiation in a given tissue, to ultimately direct its morphogenesis. In tandem with experimental studies, mathematical modelling can help gain mechanistic insights into these processes and bridge the gap between Shh-regulated patterning and growth, by integrating these processes into a common theoretical framework. Here, we briefly review the roles of Shh in vertebrate development, focusing on its functions as a morphogen, mitogen and regulator of differentiation. We then discuss the contributions that modelling has made to our understanding of the action of Shh and highlight current challenges in using mathematical models in a quantitative and predictive way. This article is part of a discussion meeting issue 'Contemporary morphogenesis'.
Collapse
Affiliation(s)
- Ian Groves
- School of Mathematics and Statistics, University of Sheffield, Hicks Building, Hounsfield Road, Sheffield S3 7RH, UK
- Department of Biomedical Science, University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, UK
- Bateson Centre, University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, UK
| | - Marysia Placzek
- Department of Biomedical Science, University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, UK
- Bateson Centre, University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, UK
| | - Alexander G. Fletcher
- School of Mathematics and Statistics, University of Sheffield, Hicks Building, Hounsfield Road, Sheffield S3 7RH, UK
- Bateson Centre, University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, UK
| |
Collapse
|
24
|
Fitzgerald M, Sotuyo N, Tischfield DJ, Anderson SA. Generation of cerebral cortical GABAergic interneurons from pluripotent stem cells. Stem Cells 2020; 38:1375-1386. [PMID: 32638460 DOI: 10.1002/stem.3252] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 05/18/2020] [Accepted: 06/11/2020] [Indexed: 11/11/2022]
Abstract
The cerebral cortex functions by the complex interactions of intrinsic and extrinsic neuronal activities, glial actions, and the effects of humoral factors. The intrinsic neuronal influences are mediated by two major subclasses: excitatory glutamatergic neurons that generally have axonal projections extending beyond the neuron's locality and inhibitory GABAergic neurons that generally project locally. These interneurons can be grouped based on morphological, neurochemical, electrophysiological, axonal targeting, and circuit influence characteristics. Cortical interneurons (CIns) can also be grouped based on their origins within the subcortical telencephalon. Interneuron subtypes, of which a dozen or more are thought to exist, are characterized by combinations of these subgrouping features. Due to their well-documented relevance to the causes of and treatments for neuropsychiatric disorders, and to their remarkable capacity to migrate extensively following transplantation, there has been tremendous interest in generating cortical GABAergic interneurons from human pluripotent stem cells. In this concise review, we discuss recent progress in understanding how interneuron subtypes are generated in vivo, and how that progress is being applied to the generation of rodent and human CIns in vitro. In addition, we will discuss approaches for the rigorous designation of interneuron subgroups or subtypes in transplantation studies, and challenges to this field, including the protracted maturation of human interneurons.
Collapse
Affiliation(s)
- Megan Fitzgerald
- The Children's Hospital of Philadelphia and the University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Nathaniel Sotuyo
- The Children's Hospital of Philadelphia and the University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - David J Tischfield
- The Children's Hospital of Philadelphia and the University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Stewart A Anderson
- The Children's Hospital of Philadelphia and the University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
25
|
Göngrich C, Krapacher FA, Munguba H, Fernández-Suárez D, Andersson A, Hjerling-Leffler J, Ibáñez CF. ALK4 coordinates extracellular and intrinsic signals to regulate development of cortical somatostatin interneurons. J Cell Biol 2020; 219:jcb.201905002. [PMID: 31676717 PMCID: PMC7039195 DOI: 10.1083/jcb.201905002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 09/03/2019] [Accepted: 10/21/2019] [Indexed: 02/07/2023] Open
Abstract
Göngrich et al. show that the activin receptor ALK4 is a key regulator of the specification of somatostatin interneurons. They find that intrinsic transcriptional programs interact with extracellular signals present in the environment of MGE cells to regulate cortical interneuron specification. Although the role of transcription factors in fate specification of cortical interneurons is well established, how these interact with extracellular signals to regulate interneuron development is poorly understood. Here we show that the activin receptor ALK4 is a key regulator of the specification of somatostatin interneurons. Mice lacking ALK4 in GABAergic neurons of the medial ganglionic eminence (MGE) showed marked deficits in distinct subpopulations of somatostatin interneurons from early postnatal stages of cortical development. Specific losses were observed among distinct subtypes of somatostatin+/Reelin+ double-positive cells, including Hpse+ layer IV cells targeting parvalbumin+ interneurons, leading to quantitative alterations in the inhibitory circuitry of this layer. Activin-mediated ALK4 signaling in MGE cells induced interaction of Smad2 with SATB1, a transcription factor critical for somatostatin interneuron development, and promoted SATB1 nuclear translocation and repositioning within the somatostatin gene promoter. These results indicate that intrinsic transcriptional programs interact with extracellular signals present in the environment of MGE cells to regulate cortical interneuron specification.
Collapse
Affiliation(s)
| | | | - Hermany Munguba
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | | | - Annika Andersson
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Jens Hjerling-Leffler
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Carlos F Ibáñez
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden.,Department of Physiology, National University of Singapore, Singapore.,Life Sciences Institute, National University of Singapore, Singapore.,Stellenbosch Institute for Advanced Study, Wallenberg Research Centre at Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
26
|
Jia S, Zhang L, Zhang K, Wang L, Khan A, Zhang J, Sun Y, Wang Y, Song M, Lyu Y, Li M, Lu X, Niu B, Liu Z, Xie J. Nkx2.1 downregulation is involved in brain abnormality induced by excess retinoic acid. Acta Biochim Biophys Sin (Shanghai) 2020; 52:683-690. [PMID: 32445470 DOI: 10.1093/abbs/gmaa037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 03/12/2020] [Accepted: 03/29/2020] [Indexed: 11/13/2022] Open
Abstract
Abnormal development of central nervous system (CNS) caused by neural tube defects is not only a major contributor in the prevalence of stillbirths and neonatal deaths but also causes lifelong physical disability in surviving infants. Due to insufficient known investigated causes, CNS developmental abnormality has brought sever burden on health around the world. From previous results of high throughput transcriptome sequencing, we selected transcription factor Nkx2.1 as a candidate to investigate its role on brain abnormalities induced by excessive retinoic acid. The result of in situ hybridization showed that Nkx2.1 was mainly expressed in mouse brain. After the Nkx2.1 gene was silenced, retarded proliferation and accelerated apoptosis were found in mouse Neuro-2a (N2a) cells. Furthermore, our results indicated that the main components of sonic hedgehog (Shh) signaling pathway were affected in Nkx2.1-silenced cells, implying that Nkx2.1 plays an important role in the development of mouse brain by regulating Shh signaling pathway.
Collapse
Affiliation(s)
- Sansan Jia
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Shanxi Medical University, Taiyuan 030001, China
- State Key Laboratory of Military Stomatology, Department of Oral & Maxillofacial Surgery, The Fourth Military Medical University, Xi’an 710032, China
| | - Li Zhang
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Shanxi Medical University, Taiyuan 030001, China
| | - Kaili Zhang
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Shanxi Medical University, Taiyuan 030001, China
| | - Lei Wang
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Shanxi Medical University, Taiyuan 030001, China
| | - Ajab Khan
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Shanxi Medical University, Taiyuan 030001, China
| | - Juan Zhang
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Shanxi Medical University, Taiyuan 030001, China
| | - Yuqing Sun
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Shanxi Medical University, Taiyuan 030001, China
| | - Yufei Wang
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Shanxi Medical University, Taiyuan 030001, China
| | - Meiyan Song
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Shanxi Medical University, Taiyuan 030001, China
| | - Yi Lyu
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Shanxi Medical University, Taiyuan 030001, China
| | - Meining Li
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Shanxi Medical University, Taiyuan 030001, China
| | - Xin Lu
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Shanxi Medical University, Taiyuan 030001, China
| | - Bo Niu
- Department of Biotechnology, Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing 100020, China
| | - Zhizhen Liu
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Shanxi Medical University, Taiyuan 030001, China
| | - Jun Xie
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Shanxi Medical University, Taiyuan 030001, China
| |
Collapse
|
27
|
Delgado RN, Mansky B, Ahanger SH, Lu C, Andersen RE, Dou Y, Alvarez-Buylla A, Lim DA. Maintenance of neural stem cell positional identity by mixed-lineage leukemia 1. Science 2020; 368:48-53. [PMID: 32241942 DOI: 10.1126/science.aba5960] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 03/09/2020] [Indexed: 12/22/2022]
Abstract
Neural stem cells (NSCs) in the developing and postnatal brain have distinct positional identities that dictate the types of neurons they generate. Although morphogens initially establish NSC positional identity in the neural tube, it is unclear how such regional differences are maintained as the forebrain grows much larger and more anatomically complex. We found that the maintenance of NSC positional identity in the murine brain requires a mixed-lineage leukemia 1 (Mll1)-dependent epigenetic memory system. After establishment by sonic hedgehog, ventral NSC identity became independent of this morphogen. Even transient MLL1 inhibition caused a durable loss of ventral identity, resulting in the generation of neurons with the characteristics of dorsal NSCs in vivo. Thus, spatial information provided by morphogens can be transitioned to epigenetic mechanisms that maintain regionally distinct developmental programs in the forebrain.
Collapse
Affiliation(s)
- Ryan N Delgado
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA.,Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA.,Medical Scientist Training Program, University of California, San Francisco, San Francisco, CA 94143, USA.,Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Benjamin Mansky
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA.,Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA.,Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Sajad Hamid Ahanger
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA.,Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA.,San Francisco Veterans Affairs Medical Center, San Francisco, CA 94121, USA
| | - Changqing Lu
- Department of Human Anatomy, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, Sichuan, P.R. China
| | - Rebecca E Andersen
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA.,Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA.,Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Yali Dou
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Arturo Alvarez-Buylla
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA.,Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Daniel A Lim
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA. .,Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA.,San Francisco Veterans Affairs Medical Center, San Francisco, CA 94121, USA
| |
Collapse
|
28
|
Everson JL, Sun MR, Fink DM, Heyne GW, Melberg CG, Nelson KF, Doroodchi P, Colopy LJ, Ulschmid CM, Martin AA, McLaughlin MT, Lipinski RJ. Developmental Toxicity Assessment of Piperonyl Butoxide Exposure Targeting Sonic Hedgehog Signaling and Forebrain and Face Morphogenesis in the Mouse: An in Vitro and in Vivo Study. ENVIRONMENTAL HEALTH PERSPECTIVES 2019; 127:107006. [PMID: 31642701 PMCID: PMC6867268 DOI: 10.1289/ehp5260] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
BACKGROUND Piperonyl butoxide (PBO) is a pesticide synergist used in residential, commercial, and agricultural settings. PBO was recently found to inhibit Sonic hedgehog (Shh) signaling, a key developmental regulatory pathway. Disruption of Shh signaling is linked to birth defects, including holoprosencephaly (HPE), a malformation of the forebrain and face thought to result from complex gene-environment interactions. OBJECTIVES The impact of PBO on Shh signaling in vitro and forebrain and face development in vivo was examined. METHODS The influence of PBO on Shh pathway transduction was assayed in mouse and human cell lines. To examine its teratogenic potential, a single dose of PBO (22-1,800mg/kg) was administered by oral gavage to C57BL/6J mice at gestational day 7.75, targeting the critical period for HPE. Gene-environment interactions were investigated using Shh+/- mice, which model human HPE-associated genetic mutations. RESULTS PBO attenuated Shh signaling in vitro through a mechanism similar to that of the known teratogen cyclopamine. In utero PBO exposure caused characteristic HPE facial dysmorphology including dose-dependent midface hypoplasia and hypotelorism, with a lowest observable effect level of 67mg/kg. Median forebrain deficiency characteristic of HPE was observed in severely affected animals, whereas all effective doses disrupted development of Shh-dependent transient forebrain structures that generate cortical interneurons. Normally silent heterozygous Shh null mutations exacerbated PBO teratogenicity at all doses tested, including 33mg/kg. DISCUSSION These findings demonstrate that prenatal PBO exposure can cause overt forebrain and face malformations or neurodevelopmental disruptions with subtle or no craniofacial dysmorphology in mice. By targeting Shh signaling as a sensitive mechanism of action and examining gene-environment interactions, this study defined a lowest observable effect level for PBO developmental toxicity in mice more than 30-fold lower than previously recognized. Human exposure to PBO and its potential contribution to etiologically complex birth defects should be rigorously examined. https://doi.org/10.1289/EHP5260.
Collapse
Affiliation(s)
- Joshua L. Everson
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Molecular and Environmental Toxicology Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Miranda R. Sun
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Dustin M. Fink
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Galen W. Heyne
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Cal G. Melberg
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Kia F. Nelson
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Padydeh Doroodchi
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Lydia J. Colopy
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Caden M. Ulschmid
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Alexander A. Martin
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Matthew T. McLaughlin
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Robert J. Lipinski
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Molecular and Environmental Toxicology Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
29
|
McKenzie MG, Cobbs LV, Dummer PD, Petros TJ, Halford MM, Stacker SA, Zou Y, Fishell GJ, Au E. Non-canonical Wnt Signaling through Ryk Regulates the Generation of Somatostatin- and Parvalbumin-Expressing Cortical Interneurons. Neuron 2019; 103:853-864.e4. [PMID: 31257105 DOI: 10.1016/j.neuron.2019.06.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 04/12/2019] [Accepted: 06/06/2019] [Indexed: 01/22/2023]
Abstract
GABAergic interneurons have many important functions in cortical circuitry, a reflection of their cell diversity. The developmental origins of this diversity are poorly understood. Here, we identify rostral-caudal regionality in Wnt exposure within the interneuron progenitor zone delineating the specification of the two main interneuron subclasses. Caudally situated medial ganglionic eminence (MGE) progenitors receive high levels of Wnt signaling and give rise to somatostatin (SST)-expressing cortical interneurons. By contrast, parvalbumin (PV)-expressing basket cells originate mostly from the rostral MGE, where Wnt signaling is attenuated. Interestingly, rather than canonical signaling through β-catenin, signaling via the non-canonical Wnt receptor Ryk regulates interneuron cell-fate specification in vivo and in vitro. Indeed, gain of function of Ryk intracellular domain signaling regulates SST and PV fate in a dose-dependent manner, suggesting that Ryk signaling acts in a graded fashion. These data reveal an important role for non-canonical Wnt-Ryk signaling in establishing the correct ratios of cortical interneuron subtypes.
Collapse
Affiliation(s)
- Melissa G McKenzie
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA; NYU Neuroscience Institute, New York University Langone Medical Center, New York, NY 10016, USA
| | - Lucy V Cobbs
- NYU Neuroscience Institute, New York University Langone Medical Center, New York, NY 10016, USA
| | - Patrick D Dummer
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA
| | - Timothy J Petros
- NYU Neuroscience Institute, New York University Langone Medical Center, New York, NY 10016, USA
| | - Michael M Halford
- Tumour Angiogenesis and Microenvironment Program, Department of Oncology, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, Victoria 3000, Australia
| | - Steven A Stacker
- Tumour Angiogenesis and Microenvironment Program, Department of Oncology, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, Victoria 3000, Australia
| | - Yimin Zou
- Neurobiology Section, Biological Sciences Division, University of California, San Diego, CA 92093, USA
| | - Gord J Fishell
- NYU Neuroscience Institute, New York University Langone Medical Center, New York, NY 10016, USA; Department of Neurobiology, Harvard Medical School, Boston, MA 04115, USA; The Stanley Center at the Broad, Cambridge, MA 02142, USA
| | - Edmund Au
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA; NYU Neuroscience Institute, New York University Langone Medical Center, New York, NY 10016, USA; Department of Rehabilitation and Regenerative Medicine, Columbia University Medical Center, New York, NY 10032, USA; Columbia Translational Neuroscience Initiative Scholar, Columbia University Medical Center, New York, NY 10032, USA.
| |
Collapse
|
30
|
Pluripotent stem cell-derived interneuron progenitors mature and restore memory deficits but do not suppress seizures in the epileptic mouse brain. Stem Cell Res 2018; 33:83-94. [DOI: 10.1016/j.scr.2018.10.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Revised: 08/27/2018] [Accepted: 10/03/2018] [Indexed: 01/11/2023] Open
|
31
|
Chen CY, Anderson NC, Becker S, Schicht M, Stoddard C, Bräuer L, Paulsen F, Grabel L. Examining the role of the surfactant family member SFTA3 in interneuron specification. PLoS One 2018; 13:e0198703. [PMID: 30408033 PMCID: PMC6224035 DOI: 10.1371/journal.pone.0198703] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 10/23/2018] [Indexed: 01/31/2023] Open
Abstract
The transcription factor NKX2.1, expressed at high levels in the medial ganglionic eminence (MGE), is a master regulator of cortical interneuron progenitor development. To identify gene candidates with expression profiles similar to NKX2.1, previous transcriptome analysis of human embryonic stem cell (hESC)-derived MGE-like progenitors revealed SFTA3 as the strongest candidate. Quantitative real-time PCR analysis of hESC-derived NKX2.1-positive progenitors and transcriptome data available from the Allen Institute for Brain Science revealed comparable expression patterns for NKX2.1 and SFTA3 during interneuron differentiation in vitro and demonstrated high SFTA3 expression in the human MGE. Although SFTA3 has been well studied in the lung, the possible role of this surfactant protein in the MGE during embryonic development remains unexamined. To determine if SFTA3 plays a role in MGE specification, SFTA3-/- and NKX2.1 -/- hESC lines were generated using custom designed CRISPRs. We show that NKX2.1 KOs have a significantly diminished capacity to differentiate into MGE interneuron subtypes. SFTA3 KOs also demonstrated a somewhat reduced ability to differentiate down the MGE-like lineage, although not as severe relative to NKX2.1 deficiency. These results suggest NKX2.1 and SFTA3 are co-regulated genes, and that deletion of SFTA3 does not lead to a major change in the specification of MGE derivatives.
Collapse
Affiliation(s)
- Christopher Y. Chen
- Department of Biology, Wesleyan University, Middletown, Connecticut, United States of America
| | - Nickesha C. Anderson
- Department of Biology, Wesleyan University, Middletown, Connecticut, United States of America
| | - Sandy Becker
- Department of Biology, Wesleyan University, Middletown, Connecticut, United States of America
| | - Martin Schicht
- Institute of Functional and Clinical Anatomy, Friedrich Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Christopher Stoddard
- Genome Sciences, University of Connecticut Health, Farmington, Connecticut, United States of America
| | - Lars Bräuer
- Institute of Functional and Clinical Anatomy, Friedrich Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Friedrich Paulsen
- Institute of Functional and Clinical Anatomy, Friedrich Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Laura Grabel
- Department of Biology, Wesleyan University, Middletown, Connecticut, United States of America
| |
Collapse
|
32
|
Yuan F, Chen X, Fang KH, Wang Y, Lin M, Xu SB, Huo HQ, Xu M, Ma L, Chen Y, He S, Liu Y. Induction of human somatostatin and parvalbumin neurons by expressing a single transcription factor LIM homeobox 6. eLife 2018; 7:37382. [PMID: 30251953 PMCID: PMC6181563 DOI: 10.7554/elife.37382] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 09/24/2018] [Indexed: 01/22/2023] Open
Abstract
Human GABAergic interneurons (GIN) are implicated in normal brain function and in numerous mental disorders. However, the generation of functional human GIN subtypes from human pluripotent stem cells (hPSCs) has not been established. By expressing LHX6, a transcriptional factor that is critical for GIN development, we induced hPSCs to form GINs, including somatostatin (SST, 29%) and parvalbumin (PV, 21%) neurons. Our RNAseq results also confirmed the alteration of GIN identity with the overexpression of LHX6. Five months after transplantation into the mouse brain, the human GABA precursors generated increased population of SST and PV neurons by overexpressing LHX6. Importantly, the grafted human GINs exhibited functional electrophysiological properties and even fast-spiking-like action potentials. Thus, expression of the single transcription factor LHX6 under our GIN differentiation condition is sufficient to robustly induce human PV and SST subtypes.
Collapse
Affiliation(s)
- Fang Yuan
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing, China.,State Key Laboratory of Reproductive Medicine, Nanjing Medical Unveristy, Nanjing, China
| | - Xin Chen
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Kai-Heng Fang
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Yuanyuan Wang
- Department of Neuroscience, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Mingyan Lin
- Department of Neuroscience, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Shi-Bo Xu
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Hai-Qin Huo
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Min Xu
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Lixiang Ma
- Department of Human Anatomy and Histology, Institute of Stem Cells and Regenerative Medicine, Fudan University Shanghai Medical School, Shanghai, China
| | - Yuejun Chen
- Institute of Neuroscience, Chinese Academy of Sciences, Beijing, China
| | - Shuijin He
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yan Liu
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing, China.,State Key Laboratory of Reproductive Medicine, Nanjing Medical Unveristy, Nanjing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
33
|
Yabut OR, Pleasure SJ. Sonic Hedgehog Signaling Rises to the Surface: Emerging Roles in Neocortical Development. Brain Plast 2018; 3:119-128. [PMID: 30151337 PMCID: PMC6091060 DOI: 10.3233/bpl-180064] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The mammalian neocortex is composed of a diverse population of neuronal and glial cells that are crucial for cognition and consciousness. Orchestration of molecular events that lead to the production of distinct cell lineages is now a major research focus. Recent studies in mammalian animal models reveal that Sonic Hedgehog (Shh) signaling plays crucial roles in this process. In this review, we will evaluate these studies and provide insights on how Shh signaling specifically influence cortical development, beyond its established roles in telencephalic patterning, by specifically focusing on its impact on cells derived from the cortical radial glial (RG) cells. We will also assess how these findings further advance our knowledge of neurological diseases and discuss potential roles of targeting Shh signaling in therapies.
Collapse
Affiliation(s)
- Odessa R Yabut
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA.,Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Samuel J Pleasure
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA.,Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA.,Programs in Neuroscience and Developmental Biology, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Kavli Institute for Fundamental Neuroscience, University of California San Francisco, CA, USA
| |
Collapse
|
34
|
Adams KV, Morshead CM. Neural stem cell heterogeneity in the mammalian forebrain. Prog Neurobiol 2018; 170:2-36. [PMID: 29902499 DOI: 10.1016/j.pneurobio.2018.06.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 05/23/2018] [Accepted: 06/07/2018] [Indexed: 12/21/2022]
Abstract
The brain was long considered an organ that underwent very little change after development. It is now well established that the mammalian central nervous system contains neural stem cells that generate progeny that are capable of making new neurons, astrocytes, and oligodendrocytes throughout life. The field has advanced rapidly as it strives to understand the basic biology of these precursor cells, and explore their potential to promote brain repair. The purpose of this review is to present current knowledge about the diversity of neural stem cells in vitro and in vivo, and highlight distinctions between neural stem cell populations, throughout development, and within the niche. A comprehensive understanding of neural stem cell heterogeneity will provide insights into the cellular and molecular regulation of neural development and lifelong neurogenesis, and will guide the development of novel strategies to promote regeneration and neural repair.
Collapse
Affiliation(s)
- Kelsey V Adams
- Institute of Medical Science, Terrence Donnelly Centre, University of Toronto, Toronto ON, M5S 3E2, Canada.
| | - Cindi M Morshead
- Institute of Medical Science, Terrence Donnelly Centre, University of Toronto, Toronto ON, M5S 3E2, Canada; Department of Surgery, Division of Anatomy, Canada; Institute of Biomaterials and Biomedical Engineering, Canada; Rehabilitation Science Institute, University of Toronto, Canada.
| |
Collapse
|
35
|
Memi F, Zecevic N, Radonjić N. Multiple roles of Sonic Hedgehog in the developing human cortex are suggested by its widespread distribution. Brain Struct Funct 2018; 223:2361-2375. [PMID: 29492654 PMCID: PMC5968052 DOI: 10.1007/s00429-018-1621-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 01/25/2018] [Indexed: 01/01/2023]
Abstract
Sonic Hedgehog (Shh) plays an instrumental role in brain development, fine-tuning processes such as cell proliferation, patterning, and fate specification. Although, mutations in the SHH pathway in humans are associated with various neurodevelopmental disorders, ranging from holoprosencephaly to schizophrenia, its expression pattern in the developing human brain is not well established. We now determined the previously not reported wide expression of SHH in the human fetal cerebral cortex during most of the gestation period (10–40 gestational weeks). This spatiotemporal distribution puts Shh in a position to influence the fundamental processes involved in corticogenesis. SHH expression increased during development, shifting from progenitor cells in the proliferative zones to neurons, both glutamatergic and GABAergic, and astrocytes in upper cortical compartments. Importantly, the expression of its downstream effectors and complementary receptors revealed evolutionary differences in SHH-pathway gene expression between humans and rodents.
Collapse
Affiliation(s)
- Fani Memi
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, 06030, USA. .,Department of Cell and Developmental Biology, University College London, 21 University Street, London, WC1E 6DE, UK.
| | - Nada Zecevic
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Nevena Radonjić
- Department of Psychiatry, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT, 06030, USA.
| |
Collapse
|
36
|
Yang Y, Shen W, Ni Y, Su Y, Yang Z, Zhao C. Impaired Interneuron Development after Foxg1 Disruption. Cereb Cortex 2018; 27:793-808. [PMID: 26620267 DOI: 10.1093/cercor/bhv297] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Interneurons play pivotal roles in the modulation of cortical function; however, the mechanisms that control interneuron development remain unclear. This study aimed to explore a new role for Foxg1 in interneuron development. By crossing Foxg1fl/fl mice with a Dlx5/6-Cre line, we determined that conditional disruption of Foxg1 in the subpallium results in defects in interneuron development. In developing interneurons, the expression levels of several receptors, including roundabout-1, Eph receptor A4, and C-X-C motif receptor 4/7, were strongly downregulated, which led to migration defects after Foxg1 ablation. The transcription factors Dlx1/2 and Mash1, which have been reported to be involved in interneuron development, were significantly upregulated at the mRNA levels. Foxg1 mutant cells developed shorter neurites and fewer branches and displayed severe migration defects in vitro. Notably, Prox1, which is a transcription factor that functions as a key regulator in the development of excitatory neurons, was also dramatically upregulated at both the mRNA and protein levels, suggesting that Prox1 is also important for interneuron development. Our work demonstrates that Foxg1 may act as a critical upstream regulator of Dlx1/2, Mash1, and Prox1 to control interneuron development. These findings will further our understanding of the molecular mechanisms of interneuron development.
Collapse
Affiliation(s)
- Ying Yang
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Department of Anatomy and Neuroscience, School of Medicine, Southeast University, Nanjing 210009, China
| | - Wei Shen
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Department of Anatomy and Neuroscience, School of Medicine, Southeast University, Nanjing 210009, China
| | - Yang Ni
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Department of Anatomy and Neuroscience, School of Medicine, Southeast University, Nanjing 210009, China
| | - Yan Su
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Department of Anatomy and Neuroscience, School of Medicine, Southeast University, Nanjing 210009, China
| | - Zhengang Yang
- Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Chunjie Zhao
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Department of Anatomy and Neuroscience, School of Medicine, Southeast University, Nanjing 210009, China.,Center of Depression, Beijing Institute for Brain Disorders, Beijing 100069, China
| |
Collapse
|
37
|
Adnani L, Han S, Li S, Mattar P, Schuurmans C. Mechanisms of Cortical Differentiation. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 336:223-320. [DOI: 10.1016/bs.ircmb.2017.07.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
38
|
Tischfield DJ, Anderson SA. Differentiation of Mouse Embryonic Stem Cells into Cortical Interneuron Precursors. J Vis Exp 2017. [PMID: 29286389 DOI: 10.3791/56358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
GABAergic cortical interneurons are a heterogeneous population of cells that play critical roles in regulating the output of excitatory pyramidal neurons as well as synchronizing the outputs of pyramidal neuron ensembles. Deficits in interneuron function have been implicated in a variety of neuropsychiatric disorders, including schizophrenia, autism, and epilepsy. The derivation of cortical interneurons from embryonic stem cells not only allows for the study of their development and function, but provides insight into the molecular mechanisms underlying the pathogenesis of cortical interneuron-related disorders. Interneurons also have the remarkable capacity to survive, migrate, and integrate into host cortical circuitry post-transplantation, making them ideal candidates for use in cell-based therapies. Here, we present a scalable, highly efficient, modified embryoid body-to-monolayer method for the derivation of Nkx2.1-expressing interneuron progenitors and their progeny from mouse embryonic stem cells (mESCs). Using a Nkx2.1::mCherry:Lhx6::GFP dual reporter mESC line, Nkx2.1 progenitors or their Lhx6-expressing post-mitotic progeny can be isolated via fluorescence-activated cell sorting (FACS) and subsequently used in a number of downstream applications. We also provide methods to enrich for parvalbumin (PV) or somatostatin (SST) interneuron subgroups, which may be helpful for studying aspects of fate determination or for use in therapeutic applications that would benefit from interneuron subgroup-enriched transplantations.
Collapse
Affiliation(s)
- David J Tischfield
- Neuroscience Graduate Group, Perelman School of Medicine, University of Pennsylvania; Department of Psychiatry, Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine
| | - Stewart A Anderson
- Neuroscience Graduate Group, Perelman School of Medicine, University of Pennsylvania;
| |
Collapse
|
39
|
Delgado RN, Lim DA. Maintenance of Positional Identity of Neural Progenitors in the Embryonic and Postnatal Telencephalon. Front Mol Neurosci 2017; 10:373. [PMID: 29180952 PMCID: PMC5693875 DOI: 10.3389/fnmol.2017.00373] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 10/26/2017] [Indexed: 12/27/2022] Open
Abstract
Throughout embryonic development and into postnatal life, regionally distinct populations of neural progenitor cells (NPCs) collectively generate the many different types of neurons that underlie the complex structure and function of the adult mammalian brain. At very early stages of telencephalic development, NPCs become organized into regional domains that each produce different subsets of neurons. This positional identity of NPCs relates to the regional expression of specific, fate-determining homeodomain transcription factors. As development progresses, the brain undergoes vast changes in both size and shape, yet important aspects of NPC positional identity persist even into the postnatal brain. How can NPC positional identity, which is established so early in brain development, endure the many dynamic, large-scale and complex changes that occur over a relatively long period of time? In this Perspective article, we review data and concepts derived from studies in Drosophila regarding the function of homeobox (Hox) genes, Polycomb group (PcG) and trithorax group (trxG) chromatin regulators. We then discuss how this knowledge may contribute to our understanding of the maintenance of positional identity of NPCs in the mammalian telencephalon. Similar to the axial body plan of Drosophila larvae, there is a segmental nature to NPC positional identity, with loss of specific homeodomain transcription factors causing homeotic-like shifts in brain development. Finally, we speculate about the role of mammalian PcG and trxG factors in the long-term maintenance of NPC positional identity and certain neurodevelopmental disorders.
Collapse
Affiliation(s)
- Ryan N Delgado
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA,, United States.,Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA,, United States.,Biomedical Sciences Program, University of California, San Francisco, San Francisco, CA,, United States.,Medical Scientist Training Program, University of California, San Francisco, San Francisco, CA,, United States
| | - Daniel A Lim
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA,, United States.,Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA,, United States.,San Francisco Veterans Affairs Medical Center, San Francisco, CA,, United States
| |
Collapse
|
40
|
Chen CY, Plocik A, Anderson NC, Moakley D, Boyi T, Dundes C, Lassiter C, Graveley BR, Grabel L. Transcriptome and in Vitro Differentiation Profile of Human Embryonic Stem Cell Derived NKX2.1-Positive Neural Progenitors. Stem Cell Rev Rep 2017; 12:744-756. [PMID: 27539622 DOI: 10.1007/s12015-016-9676-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The generation of inhibitory interneuron progenitors from human embryonic stem cells (ESCs) is of great interest due to their potential use in transplantation therapies designed to treat central nervous system disorders. The medial ganglionic eminence (MGE) is a transient embryonic structure in the ventral telencephalon that is a major source of cortical GABAergic inhibitory interneuron progenitors. These progenitors migrate tangentially to sites in the cortex and differentiate into a variety of interneuron subtypes, forming local synaptic connections with excitatory projection neurons to modulate activity of the cortical circuitry. The homeobox domain-containing transcription factor NKX2.1 is highly expressed in the MGE and pre-optic area of the ventral subpallium and is essential for specifying cortical interneuron fate. Using a combination of growth factor agonists and antagonists to specify ventral telencephalic fates, we previously optimized a protocol for the efficient generation of NKX2.1-positive MGE-like neural progenitors from human ESCs. To establish their identity, we now characterize the transcriptome of these MGE-like neural progenitors using RNA sequencing and demonstrate the capacity of these cells to differentiate into inhibitory interneurons in vitro using a neuron-astrocyte co-culture system. These data provide information on the potential origin of interneurons in the human brain.
Collapse
Affiliation(s)
- Christopher Y Chen
- Department of Biology, Wesleyan University, 52 Lawn Avenue, Middletown, CT, 06459, USA.
| | - Alex Plocik
- Department of Genetics and Genome Sciences, Institute for Systems Genomics, 400 Farmington Avenue, UCONN Health, Farmington, CT, 06030, USA
| | - Nickesha C Anderson
- Department of Biology, Wesleyan University, 52 Lawn Avenue, Middletown, CT, 06459, USA
| | - Daniel Moakley
- Department of Biology, Wesleyan University, 52 Lawn Avenue, Middletown, CT, 06459, USA
| | - Trinithas Boyi
- Department of Biology, Wesleyan University, 52 Lawn Avenue, Middletown, CT, 06459, USA
| | - Carolyn Dundes
- Department of Biology, Wesleyan University, 52 Lawn Avenue, Middletown, CT, 06459, USA
| | - Chelsea Lassiter
- Department of Biology, Wesleyan University, 52 Lawn Avenue, Middletown, CT, 06459, USA
| | - Brenton R Graveley
- Department of Genetics and Genome Sciences, Institute for Systems Genomics, 400 Farmington Avenue, UCONN Health, Farmington, CT, 06030, USA
| | - Laura Grabel
- Department of Biology, Wesleyan University, 52 Lawn Avenue, Middletown, CT, 06459, USA
| |
Collapse
|
41
|
Floruta CM, Du R, Kang H, Stein JL, Weick JP. Default Patterning Produces Pan-cortical Glutamatergic and CGE/LGE-like GABAergic Neurons from Human Pluripotent Stem Cells. Stem Cell Reports 2017; 9:1463-1476. [PMID: 29107596 PMCID: PMC5831028 DOI: 10.1016/j.stemcr.2017.09.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 09/25/2017] [Accepted: 09/26/2017] [Indexed: 10/25/2022] Open
Abstract
Default differentiation of human pluripotent stem cells has been promoted as a model of cortical development. In this study, a developmental transcriptome analysis of default-differentiated hPSNs revealed a gene expression program resembling in vivo CGE/LGE subpallial domains and GABAergic signaling. A combination of bioinformatic, functional, and immunocytochemical analysis further revealed that hPSNs consist of both cortical glutamatergic and CGE-like GABAergic neurons. This study provides a comprehensive characterization of the heterogeneous group of neurons produced by default differentiation and insight into future directed differentiation strategies.
Collapse
Affiliation(s)
- Crina M. Floruta
- Department of Neurosciences, University of New Mexico-Health Science Center, Albuquerque, NM 87131, USA
| | - Ruofei Du
- UNM Comprehensive Cancer Center, University of New Mexico-Health Science Center, Albuquerque, NM 87131, USA
| | - Huining Kang
- UNM Comprehensive Cancer Center, University of New Mexico-Health Science Center, Albuquerque, NM 87131, USA,Department of Internal Medicine, University of New Mexico-Health Science Center, Albuquerque, NM 87131, USA
| | - Jason L. Stein
- Department of Genetics, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA,UNC Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Jason P. Weick
- Department of Neurosciences, University of New Mexico-Health Science Center, Albuquerque, NM 87131, USA,Corresponding author
| |
Collapse
|
42
|
Donegan JJ, Tyson JA, Branch SY, Beckstead MJ, Anderson SA, Lodge DJ. Stem cell-derived interneuron transplants as a treatment for schizophrenia: preclinical validation in a rodent model. Mol Psychiatry 2017; 22:1492-1501. [PMID: 27480492 PMCID: PMC5290293 DOI: 10.1038/mp.2016.121] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 06/17/2016] [Accepted: 06/21/2016] [Indexed: 01/28/2023]
Abstract
An increasing literature suggests that schizophrenia is associated with a reduction in hippocampal interneuron function. Thus, we posit that stem cell-derived interneuron transplants may be an effective therapeutic strategy to reduce hippocampal hyperactivity and attenuate behavioral deficits in schizophrenia. Here we used a dual-reporter embryonic stem cell line to generate enriched populations of parvalbumin (PV)- or somatostatin (SST)-positive interneurons, which were transplanted into the ventral hippocampus of the methylazoxymethanol rodent model of schizophrenia. These interneuron transplants integrate within the existing circuitry, reduce hippocampal hyperactivity and normalize aberrant dopamine neuron activity. Further, interneuron transplants alleviate behaviors that model negative and cognitive symptoms, including deficits in social interaction and cognitive inflexibility. Interestingly, PV- and SST-enriched transplants produced differential effects on behavior, with PV-enriched populations effectively normalizing all the behaviors examined. These data suggest that the stem cell-derived interneuron transplants may represent a novel therapeutic strategy for schizophrenia.
Collapse
Affiliation(s)
- J J Donegan
- Department of Pharmacology and Center for Biomedical Neuroscience, University of Texas Health Science Center, San Antonio, TX, USA
| | - J A Tyson
- Department of Psychiatry, Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - S Y Branch
- Department of Physiology and Center for Biomedical Neuroscience, University of Texas Health Science Center, San Antonio, TX, USA
| | - M J Beckstead
- Department of Physiology and Center for Biomedical Neuroscience, University of Texas Health Science Center, San Antonio, TX, USA
| | - S A Anderson
- Department of Psychiatry, Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - D J Lodge
- Department of Pharmacology and Center for Biomedical Neuroscience, University of Texas Health Science Center, San Antonio, TX, USA
| |
Collapse
|
43
|
Pelkey KA, Chittajallu R, Craig MT, Tricoire L, Wester JC, McBain CJ. Hippocampal GABAergic Inhibitory Interneurons. Physiol Rev 2017; 97:1619-1747. [PMID: 28954853 PMCID: PMC6151493 DOI: 10.1152/physrev.00007.2017] [Citation(s) in RCA: 569] [Impact Index Per Article: 71.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 05/16/2017] [Accepted: 05/26/2017] [Indexed: 12/11/2022] Open
Abstract
In the hippocampus GABAergic local circuit inhibitory interneurons represent only ~10-15% of the total neuronal population; however, their remarkable anatomical and physiological diversity allows them to regulate virtually all aspects of cellular and circuit function. Here we provide an overview of the current state of the field of interneuron research, focusing largely on the hippocampus. We discuss recent advances related to the various cell types, including their development and maturation, expression of subtype-specific voltage- and ligand-gated channels, and their roles in network oscillations. We also discuss recent technological advances and approaches that have permitted high-resolution, subtype-specific examination of their roles in numerous neural circuit disorders and the emerging therapeutic strategies to ameliorate such pathophysiological conditions. The ultimate goal of this review is not only to provide a touchstone for the current state of the field, but to help pave the way for future research by highlighting where gaps in our knowledge exist and how a complete appreciation of their roles will aid in future therapeutic strategies.
Collapse
Affiliation(s)
- Kenneth A Pelkey
- Porter Neuroscience Center, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratories, University of Exeter, Exeter, United Kingdom; and Sorbonne Universités, UPMC University of Paris, INSERM, CNRS, Neurosciences Paris Seine-Institut de Biologie Paris Seine, Paris, France
| | - Ramesh Chittajallu
- Porter Neuroscience Center, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratories, University of Exeter, Exeter, United Kingdom; and Sorbonne Universités, UPMC University of Paris, INSERM, CNRS, Neurosciences Paris Seine-Institut de Biologie Paris Seine, Paris, France
| | - Michael T Craig
- Porter Neuroscience Center, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratories, University of Exeter, Exeter, United Kingdom; and Sorbonne Universités, UPMC University of Paris, INSERM, CNRS, Neurosciences Paris Seine-Institut de Biologie Paris Seine, Paris, France
| | - Ludovic Tricoire
- Porter Neuroscience Center, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratories, University of Exeter, Exeter, United Kingdom; and Sorbonne Universités, UPMC University of Paris, INSERM, CNRS, Neurosciences Paris Seine-Institut de Biologie Paris Seine, Paris, France
| | - Jason C Wester
- Porter Neuroscience Center, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratories, University of Exeter, Exeter, United Kingdom; and Sorbonne Universités, UPMC University of Paris, INSERM, CNRS, Neurosciences Paris Seine-Institut de Biologie Paris Seine, Paris, France
| | - Chris J McBain
- Porter Neuroscience Center, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratories, University of Exeter, Exeter, United Kingdom; and Sorbonne Universités, UPMC University of Paris, INSERM, CNRS, Neurosciences Paris Seine-Institut de Biologie Paris Seine, Paris, France
| |
Collapse
|
44
|
Suzuki T, Sato M. Inter-progenitor pool wiring: An evolutionarily conserved strategy that expands neural circuit diversity. Dev Biol 2017; 431:101-110. [PMID: 28958816 DOI: 10.1016/j.ydbio.2017.09.029] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 09/15/2017] [Accepted: 09/23/2017] [Indexed: 11/28/2022]
Abstract
Diversification of neuronal types is key to establishing functional variations in neural circuits. The first critical step to generate neuronal diversity is to organize the compartmental domains of developing brains into spatially distinct neural progenitor pools. Neural progenitors in each pool then generate a unique set of diverse neurons through specific spatiotemporal specification processes. In this review article, we focus on an additional mechanism, 'inter-progenitor pool wiring', that further expands the diversity of neural circuits. After diverse types of neurons are generated in one progenitor pool, a fraction of these neurons start migrating toward a remote brain region containing neurons that originate from another progenitor pool. Finally, neurons of different origins are intermingled and eventually form complex but precise neural circuits. The developing cerebral cortex of mammalian brains is one of the best examples of inter-progenitor pool wiring. However, Drosophila visual system development has revealed similar mechanisms in invertebrate brains, suggesting that inter-progenitor pool wiring is an evolutionarily conserved strategy that expands neural circuit diversity. Here, we will discuss how inter-progenitor pool wiring is accomplished in mammalian and fly brain systems.
Collapse
Affiliation(s)
- Takumi Suzuki
- Lab of Developmental Neurobiology, Graduate School of Medical Sciences, Mathematical Neuroscience Unit, Institute for Frontier Science Initiative, Kanazawa University, 13-1, Takaramachi, Kanazawa, Ishikawa 920-8640, Japan
| | - Makoto Sato
- Lab of Developmental Neurobiology, Graduate School of Medical Sciences, Mathematical Neuroscience Unit, Institute for Frontier Science Initiative, Kanazawa University, 13-1, Takaramachi, Kanazawa, Ishikawa 920-8640, Japan.
| |
Collapse
|
45
|
Guo J, Otis JM, Higginbotham H, Monckton C, Cheng J, Asokan A, Mykytyn K, Caspary T, Stuber GD, Anton ES. Primary Cilia Signaling Shapes the Development of Interneuronal Connectivity. Dev Cell 2017; 42:286-300.e4. [PMID: 28787594 DOI: 10.1016/j.devcel.2017.07.010] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 05/18/2017] [Accepted: 07/12/2017] [Indexed: 01/06/2023]
Abstract
Appropriate growth and synaptic integration of GABAergic inhibitory interneurons are essential for functional neural circuits in the brain. Here, we demonstrate that disruption of primary cilia function following the selective loss of ciliary GTPase Arl13b in interneurons impairs interneuronal morphology and synaptic connectivity, leading to altered excitatory/inhibitory activity balance. The altered morphology and connectivity of cilia mutant interneurons and the functional deficits are rescued by either chemogenetic activation of ciliary G-protein-coupled receptor (GPCR) signaling or the selective induction of Sstr3, a ciliary GPCR, in Arl13b-deficient cilia. Our results thus define a specific requirement for primary cilia-mediated GPCR signaling in interneuronal connectivity and inhibitory circuit formation.
Collapse
Affiliation(s)
- Jiami Guo
- UNC Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA; Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - James M Otis
- UNC Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA; Department of Psychiatry, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Holden Higginbotham
- UNC Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA; Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Chase Monckton
- UNC Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - JrGang Cheng
- UNC Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Aravind Asokan
- Department of Genetics and Gene Therapy Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Kirk Mykytyn
- Department of Biological Chemistry and Pharmacology, Neuroscience Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Tamara Caspary
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Garret D Stuber
- UNC Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA; Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA; Department of Psychiatry, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - E S Anton
- UNC Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA; Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA.
| |
Collapse
|
46
|
Turrero García M, Harwell CC. Radial glia in the ventral telencephalon. FEBS Lett 2017; 591:3942-3959. [PMID: 28862741 DOI: 10.1002/1873-3468.12829] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 08/23/2017] [Accepted: 08/24/2017] [Indexed: 12/31/2022]
Abstract
The ventral telencephalon is the developmental origin of the basal ganglia and the source of neuronal and glial cells that integrate into developing circuits in other areas of the brain. Radial glia in the embryonic subpallium give rise to an enormous diversity of mature cell types, either directly or through other transit-amplifying progenitors. Here, we review current knowledge about these subpallial neural stem cells and their progeny, focusing on the period of neurogenesis. We describe their cell biological features and the extrinsic and intrinsic molecular codes that guide their fate specification in defined temporal and spatial sequences. We also discuss the role of clonal lineage in the organization and specification of mature neurons.
Collapse
Affiliation(s)
| | - Corey C Harwell
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
47
|
Laclef C, Métin C. Conserved rules in embryonic development of cortical interneurons. Semin Cell Dev Biol 2017; 76:86-100. [PMID: 28918121 DOI: 10.1016/j.semcdb.2017.09.017] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 09/08/2017] [Accepted: 09/11/2017] [Indexed: 12/24/2022]
Abstract
This review will focus on early aspects of cortical interneurons (cIN) development from specification to migration and final positioning in the human cerebral cortex. These mechanisms have been largely studied in the mouse model, which provides unique possibilities of genetic analysis, essential to dissect the molecular and cellular events involved in cortical development. An important goal here is to discuss the conservation and the potential divergence of these mechanisms, with a particular interest for the situation in the human embryo. We will thus cover recent works, but also revisit older studies in the light of recent data to better understand the developmental mechanisms underlying cIN differentiation in human. Because cIN are implicated in severe developmental disorders, understanding the molecular and cellular mechanisms controlling their differentiation might clarify some causes and potential therapeutic approaches to these important clinical conditions.
Collapse
Affiliation(s)
- Christine Laclef
- INSERM, UMR-S839, Paris, France; Sorbonne Universités, UPMC University Paris 6, UMR-S839, Paris, France; Institut du Fer à Moulin, Paris, France
| | - Christine Métin
- INSERM, UMR-S839, Paris, France; Sorbonne Universités, UPMC University Paris 6, UMR-S839, Paris, France; Institut du Fer à Moulin, Paris, France.
| |
Collapse
|
48
|
Tischfield DJ, Kim J, Anderson SA. Atypical PKC and Notch Inhibition Differentially Modulate Cortical Interneuron Subclass Fate from Embryonic Stem Cells. Stem Cell Reports 2017; 8:1135-1143. [PMID: 28416285 PMCID: PMC5829278 DOI: 10.1016/j.stemcr.2017.03.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 03/12/2017] [Accepted: 03/13/2017] [Indexed: 11/21/2022] Open
Abstract
Recent studies indicate that the location of neurogenesis within the medial ganglionic eminence (MGE) critically influences the fate determination of cortical interneuron subgroups, with parvalbumin (Pv) interneurons originating from subventricular zone divisions and somatostatin (Sst) interneurons primarily arising from apical divisions. The aPKC-CBP and Notch signaling pathways regulate the transition from apical to basal progenitor and their differentiation into post-mitotic neurons. We find that aPKC inhibition enhances intermediate neurogenesis from stem cell-derived MGE progenitors, resulting in a markedly increased ratio of Pv- to Sst-expressing interneurons. Conversely, inhibition of Notch signaling enriches for Sst subtypes at the expense of Pv fates. These findings confirm that the mode of neurogenesis influences the fate of MGE-derived interneurons and provide a means of further enrichment for the generation of specific interneuron subgroups from pluripotent stem cells.
Collapse
Affiliation(s)
- David J Tischfield
- Neuroscience Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6085, USA; Department of Psychiatry, Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine ARC 517, Philadelphia, PA 19104-5127, USA
| | - Junho Kim
- Department of Psychiatry, Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine ARC 517, Philadelphia, PA 19104-5127, USA
| | - Stewart A Anderson
- Neuroscience Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6085, USA; Department of Psychiatry, Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine ARC 517, Philadelphia, PA 19104-5127, USA.
| |
Collapse
|
49
|
Abstract
The proper construction of neural circuits requires the generation of diverse cell types, their distribution to defined regions, and their specific and appropriate wiring. A major objective in neurobiology has been to understand the molecular determinants that link neural birth to terminal specification and functional connectivity, a task that is especially daunting in the case of cortical interneurons. Considerable evidence supports the idea that an interplay of intrinsic and environmental signalling is crucial to the sequential steps of interneuron specification, including migration, selection of a settling position, morphogenesis and synaptogenesis. However, when and how these influences merge to support the appropriate terminal differentiation of different classes of interneurons remains uncertain. In this Review, we discuss a wealth of recent findings that have advanced our understanding of the developmental mechanisms that contribute to the diversification of interneurons and suggest areas of particular promise for further investigation.
Collapse
|
50
|
Donegan JJ, Lodge DJ. Cell-based therapies for the treatment of schizophrenia. Brain Res 2017; 1655:262-269. [PMID: 27544423 PMCID: PMC5474910 DOI: 10.1016/j.brainres.2016.08.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 08/04/2016] [Accepted: 08/08/2016] [Indexed: 02/08/2023]
Abstract
Schizophrenia is a devastating psychiatric disorder characterized by positive, negative and cognitive symptoms. While aberrant dopamine system function is typically associated with the positive symptoms of the disease, it is thought that this is secondary to pathology in afferent regions. Indeed, schizophrenia patients show dysregulated activity in the hippocampus and prefrontal cortex, two regions known to regulate dopamine neuron activity. These deficits in hippocampal and prefrontal cortical function are thought to result, in part, from reductions in inhibitory interneuron function in these brain regions. Therefore, it has been hypothesized that restoring interneuron function in the hippocampus and/or prefrontal cortex may be an effective treatment strategy for schizophrenia. In this article, we will discuss the evidence for interneuron pathology in schizophrenia and review recent advances in our understanding of interneuron development. Finally, we will explore how these advances have allowed us to test the therapeutic value of interneuron transplants in multiple preclinical models of schizophrenia. This article is part of a Special Issue entitled SI:StemsCellsinPsychiatry.
Collapse
Affiliation(s)
- Jennifer J Donegan
- Department of Pharmacology and Center for Biomedical Neuroscience, University of Texas Health Science Center, San Antonio, TX 78229, USA.
| | - Daniel J Lodge
- Department of Pharmacology and Center for Biomedical Neuroscience, University of Texas Health Science Center, San Antonio, TX 78229, USA
| |
Collapse
|