151
|
Lesciotto KM, Richtsmeier JT. Craniofacial skeletal response to encephalization: How do we know what we think we know? AMERICAN JOURNAL OF PHYSICAL ANTHROPOLOGY 2019; 168 Suppl 67:27-46. [PMID: 30680710 PMCID: PMC6424107 DOI: 10.1002/ajpa.23766] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 11/16/2018] [Accepted: 11/21/2018] [Indexed: 12/17/2022]
Abstract
Dramatic changes in cranial capacity have characterized human evolution. Important evolutionary hypotheses, such as the spatial packing hypothesis, assert that increases in relative brain size (encephalization) have caused alterations to the modern human skull, resulting in a suite of traits unique among extant primates, including a domed cranial vault, highly flexed cranial base, and retracted facial skeleton. Most prior studies have used fossil or comparative primate data to establish correlations between brain size and cranial form, but the mechanistic basis for how changes in brain size impact the overall shape of the skull resulting in these cranial traits remains obscure and has only rarely been investigated critically. We argue that understanding how changes in human skull morphology could have resulted from increased encephalization requires the direct testing of hypotheses relating to interaction of embryonic development of the bones of the skull and the brain. Fossil and comparative primate data have thoroughly described the patterns of association between brain size and skull morphology. Here we suggest complementing such existing datasets with experiments focused on mechanisms responsible for producing the observed patterns to more thoroughly understand the role of encephalization in shaping the modern human skull.
Collapse
Affiliation(s)
- Kate M Lesciotto
- Department of Anthropology, Pennsylvania State University, University Park, Pennsylvania
| | - Joan T Richtsmeier
- Department of Anthropology, Pennsylvania State University, University Park, Pennsylvania
| |
Collapse
|
152
|
Kostović I, Sedmak G, Judaš M. Neural histology and neurogenesis of the human fetal and infant brain. Neuroimage 2018; 188:743-773. [PMID: 30594683 DOI: 10.1016/j.neuroimage.2018.12.043] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 12/18/2018] [Accepted: 12/20/2018] [Indexed: 01/11/2023] Open
Abstract
The human brain develops slowly and over a long period of time which lasts for almost three decades. This enables good spatio-temporal resolution of histogenetic and neurogenetic events as well as an appropriate and clinically relevant timing of these events. In order to successfully apply in vivo neuroimaging data, in analyzing both the normal brain development and the neurodevelopmental origin of major neurological and mental disorders, it is important to correlate these neuroimaging data with the existing data on morphogenetic, histogenetic and neurogenetic events. Furthermore, when performing such correlation, the genetic, genomic, and molecular biology data on phenotypic specification of developing brain regions, areas and neurons should also be included. In this review, we focus on early developmental periods (form 8 postconceptional weeks to the second postnatal year) and describe the microstructural organization and neural circuitry elements of the fetal and early postnatal human cerebrum.
Collapse
Affiliation(s)
- I Kostović
- University of Zagreb School of Medicine, Croatian Institute for Brain Research, Centre of Excellence for Basic, Clinical and Translational Neuroscience, Šalata 12, 10000, Zagreb, Croatia.
| | - G Sedmak
- University of Zagreb School of Medicine, Croatian Institute for Brain Research, Centre of Excellence for Basic, Clinical and Translational Neuroscience, Šalata 12, 10000, Zagreb, Croatia.
| | - M Judaš
- University of Zagreb School of Medicine, Croatian Institute for Brain Research, Centre of Excellence for Basic, Clinical and Translational Neuroscience, Šalata 12, 10000, Zagreb, Croatia.
| |
Collapse
|
153
|
Stephen J, Maddirevula S, Nampoothiri S, Burke JD, Herzog M, Shukla A, Steindl K, Eskin A, Patil SJ, Joset P, Lee H, Garrett LJ, Yokoyama T, Balanda N, Bodine SP, Tolman NJ, Zerfas PM, Zheng A, Ramantani G, Girisha KM, Rivas C, Suresh PV, Elkahloun A, Alsaif HS, Wakil SM, Mahmoud L, Ali R, Prochazkova M, Kulkarni AB, Ben-Omran T, Colak D, Morris HD, Rauch A, Martinez-Agosto JA, Nelson SF, Alkuraya FS, Gahl WA, Malicdan MCV, Malicdan MCV. Bi-allelic TMEM94 Truncating Variants Are Associated with Neurodevelopmental Delay, Congenital Heart Defects, and Distinct Facial Dysmorphism. Am J Hum Genet 2018; 103:948-967. [PMID: 30526868 DOI: 10.1016/j.ajhg.2018.11.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 11/02/2018] [Indexed: 02/06/2023] Open
Abstract
Neurodevelopmental disorders (NDD) are genetically and phenotypically heterogeneous conditions due to defects in genes involved in development and function of the nervous system. Individuals with NDD, in addition to their primary neurodevelopmental phenotype, may also have accompanying syndromic features that can be very helpful diagnostically especially those with recognizable facial appearance. In this study, we describe ten similarly affected individuals from six unrelated families of different ethnic origins having bi-allelic truncating variants in TMEM94, which encodes for an uncharacterized transmembrane nuclear protein that is highly conserved across mammals. The affected individuals manifested with global developmental delay/intellectual disability, and dysmorphic facial features including triangular face, deep set eyes, broad nasal root and tip and anteverted nostrils, thick arched eye brows, hypertrichosis, pointed chin, and hypertelorism. Birthweight in the upper normal range was observed in most, and all but one had congenital heart defects (CHD). Gene expression analysis in available cells from affected individuals showed reduced expression of TMEM94. Global transcriptome profiling using microarray and RNA sequencing revealed several dysregulated genes essential for cell growth, proliferation and survival that are predicted to have an impact on cardiotoxicity hematological system and neurodevelopment. Loss of Tmem94 in mouse model generated by CRISPR/Cas9 was embryonic lethal and led to craniofacial and cardiac abnormalities and abnormal neuronal migration pattern, suggesting that this gene is important in craniofacial, cardiovascular, and nervous system development. Our study suggests the genetic etiology of a recognizable dysmorphic syndrome with NDD and CHD and highlights the role of TMEM94 in early development.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - May Christine V Malicdan
- Section of Human Biochemical Genetics, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA; NIH Undiagnosed Diseases Program, NHGRI and the Common Fund, National Institutes of Health, Bethesda, MD 20892, USA; Office of the Clinical Director, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
154
|
Mancinelli S, Lodato S. Decoding neuronal diversity in the developing cerebral cortex: from single cells to functional networks. Curr Opin Neurobiol 2018; 53:146-155. [DOI: 10.1016/j.conb.2018.08.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 07/13/2018] [Accepted: 08/03/2018] [Indexed: 12/14/2022]
|
155
|
Transcriptional control of long-range cortical projections. Curr Opin Neurobiol 2018; 53:57-65. [DOI: 10.1016/j.conb.2018.05.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 05/23/2018] [Indexed: 12/11/2022]
|
156
|
Eid L, Raju PK, Rossignol E. PHACTRing in actin: actin deregulation in genetic epilepsies. Brain 2018; 141:3084-3088. [PMID: 30364981 DOI: 10.1093/brain/awy272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Lara Eid
- CHU Sainte-Justine Research Center, Montreal, Canada.,Department of Neurosciences, University of Montreal, Montreal, Canada
| | - Praveen K Raju
- CHU Sainte-Justine Research Center, Montreal, Canada.,Department of Neurosciences, University of Montreal, Montreal, Canada
| | - Elsa Rossignol
- CHU Sainte-Justine Research Center, Montreal, Canada.,Department of Neurosciences, University of Montreal, Montreal, Canada.,Department of Pediatrics, University of Montreal, Montreal, Canada
| |
Collapse
|
157
|
Zalucki O, Harris L, Harvey TJ, Harkins D, Widagdo J, Oishi S, Matuzelski E, Yong XLH, Schmidt H, Anggono V, Burne THJ, Gronostajski RM, Piper M. NFIX-Mediated Inhibition of Neuroblast Branching Regulates Migration Within the Adult Mouse Ventricular–Subventricular Zone. Cereb Cortex 2018; 29:3590-3604. [DOI: 10.1093/cercor/bhy233] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Revised: 08/26/2018] [Accepted: 08/29/2018] [Indexed: 12/13/2022] Open
Abstract
Abstract
Understanding the migration of newborn neurons within the brain presents a major challenge in contemporary biology. Neuronal migration is widespread within the developing brain but is also important within the adult brain. For instance, stem cells within the ventricular–subventricular zone (V-SVZ) and the subgranular zone of dentate gyrus of the adult rodent brain produce neuroblasts that migrate to the olfactory bulb and granule cell layer of the dentate gyrus, respectively, where they regulate key brain functions including innate olfactory responses, learning, and memory. Critically, our understanding of the factors mediating neuroblast migration remains limited. The transcription factor nuclear factor I X (NFIX) has previously been implicated in embryonic cortical development. Here, we employed conditional ablation of Nfix from the adult mouse brain and demonstrated that the removal of this gene from either neural stem and progenitor cells, or neuroblasts, within the V-SVZ culminated in neuroblast migration defects. Mechanistically, we identified aberrant neuroblast branching, due in part to increased expression of the guanylyl cyclase natriuretic peptide receptor 2 (Npr2), as a factor contributing to abnormal migration in Nfix-deficient adult mice. Collectively, these data provide new insights into how neuroblast migration is regulated at a transcriptional level within the adult brain.
Collapse
Affiliation(s)
- Oressia Zalucki
- The School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Lachlan Harris
- The School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Tracey J Harvey
- The School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Danyon Harkins
- The School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Jocelyn Widagdo
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
- Clem Jones Centre for Ageing Dementia Research, The University of Queensland, Brisbane, QLD, Australia
| | - Sabrina Oishi
- The School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Elise Matuzelski
- The School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Xuan Ling Hilary Yong
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
- Clem Jones Centre for Ageing Dementia Research, The University of Queensland, Brisbane, QLD, Australia
| | - Hannes Schmidt
- Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen, Germany
| | - Victor Anggono
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
- Clem Jones Centre for Ageing Dementia Research, The University of Queensland, Brisbane, QLD, Australia
| | - Thomas H J Burne
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
- Queensland Centre for Mental Health Research, The Park Centre for Mental Health, Wacol, QLD, Australia
| | - Richard M Gronostajski
- Department of Biochemistry, Program in Genetics, Genomics and Bioinformatics, Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Michael Piper
- The School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
158
|
Chang M, Kawai HD. A characterization of laminar architecture in mouse primary auditory cortex. Brain Struct Funct 2018; 223:4187-4209. [PMID: 30187193 DOI: 10.1007/s00429-018-1744-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 08/29/2018] [Indexed: 12/19/2022]
Abstract
Laminar architecture of primary auditory cortex (A1) has long been investigated by traditional histochemical techniques such as Nissl staining, retrograde and anterograde tracings. Uncertainty still remains, however, about laminar boundaries in mice. Here we investigated the cortical lamina structure by combining neuronal tracing and immunofluorochemistry for laminar specific markers. Most retrogradely labeled corticothalamic neurons expressed Forkhead box protein P2 (Foxp2) and distributed within the laminar band of Foxp2-expressing cells, identifying layer 6. Cut-like homeobox 1 (Cux1) expression in layer 2-4 neurons divided the upper layers into low expression layers 2/3 and high expression layers 3/4, which overlapped with the dense terminals of vesicular glutamate transporter 2 (vGluT2) and anterogradely labeled lemniscal thalamocortical axons. In layer 5, between Cux1-expressing layers 2-4 and Foxp2-defined layer 6, retrogradely labeled corticocollicular projection neurons mostly expressed COUP-TF interacting protein 2 (Ctip2). Ctip2-expressing neurons formed a laminar band in the middle of layer 5 distant from layer 6, creating a laminar gap between the two laminas. This gap contained a high population of commissural neurons projecting to contralateral A1 compared to other layers and received vGluT2-immunopositive, presumptive thalamocortical axon collateral inputs. Our study shows that layer 5 is much wider than layer 6, and layer 5 can be divided into at least three sublayers. The thalamorecipient layers 3/4 may be separated from layers 2/3 using Cux1 and can be also divided into layer 4 and layer 3 based on the neuronal soma size. These data provide a new insight for the laminar structure of mouse A1.
Collapse
Affiliation(s)
- Minzi Chang
- Department of Bioinformatics, Graduate School of Engineering, Soka University, Hachioji, Tokyo, 192-8577, Japan
| | - Hideki Derek Kawai
- Department of Bioinformatics, Graduate School of Engineering, Soka University, Hachioji, Tokyo, 192-8577, Japan. .,Department of Science and Engineering for Sustainable Innovation, Faculty of Science and Engineering, Soka University, Hachioji, Tokyo, 192-8577, Japan.
| |
Collapse
|
159
|
Cheng X, Li K, Liu M, Xu M, Hu X, Yan R, Förster E, Zhao S. The effect of P85 on neuronal proliferation and differentiation during development of mouse cerebral cortex. Dev Biol 2018; 441:95-103. [DOI: 10.1016/j.ydbio.2018.06.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 05/23/2018] [Accepted: 06/23/2018] [Indexed: 12/28/2022]
|
160
|
Chang M, Suzuki N, Kawai HD. Laminar specific gene expression reveals differences in postnatal laminar maturation in mouse auditory, visual, and somatosensory cortex. J Comp Neurol 2018; 526:2257-2284. [DOI: 10.1002/cne.24481] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 05/03/2018] [Accepted: 05/21/2018] [Indexed: 01/01/2023]
Affiliation(s)
- Minzi Chang
- Department of Bioinformatics; Graduate School of Engineering; Hachioji Tokyo 192-8577 Japan
| | - Nobuko Suzuki
- Department of Bioinformatics; Graduate School of Engineering; Hachioji Tokyo 192-8577 Japan
| | - Hideki Derek Kawai
- Department of Bioinformatics; Graduate School of Engineering; Hachioji Tokyo 192-8577 Japan
- Department of Science and Engineering for Sustainable Innovation; Faculty of Science and Engineering; Hachioji Tokyo 192-8577 Japan
| |
Collapse
|
161
|
Zucco AJ, Pozzo VD, Afinogenova A, Hart RP, Devinsky O, D'Arcangelo G. Neural progenitors derived from Tuberous Sclerosis Complex patients exhibit attenuated PI3K/AKT signaling and delayed neuronal differentiation. Mol Cell Neurosci 2018; 92:149-163. [PMID: 30144504 DOI: 10.1016/j.mcn.2018.08.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 07/23/2018] [Accepted: 08/20/2018] [Indexed: 12/20/2022] Open
Abstract
Tuberous Sclerosis Complex (TSC) is a disease caused by autosomal dominant mutations in the TSC1 or TSC2 genes, and is characterized by tumor susceptibility, brain lesions, seizures and behavioral impairments. The TSC1 and TSC2 genes encode proteins forming a complex (TSC), which is a major regulator and suppressor of mammalian target of rapamycin complex 1 (mTORC1), a signaling complex that promotes cell growth and proliferation. TSC1/2 loss of heterozygosity (LOH) and the subsequent complete loss of TSC regulatory activity in null cells causes mTORC1 dysregulation and TSC-associated brain lesions or other tissue tumors. However, it is not clear whether TSC1/2 heterozygous brain cells are abnormal and contribute to TSC neuropathology. To investigate this issue, we generated induced pluripotent stem cells (iPSCs) from TSC patients and unaffected controls, and utilized these to obtain neural progenitor cells (NPCs) and differentiated neurons in vitro. These patient-derived TSC2 heterozygous NPCs were delayed in their ability to differentiate into neurons. Patient-derived progenitor cells also exhibited a modest activation of mTORC1 signaling downstream of TSC, and a marked attenuation of upstream PI3K/AKT signaling. We further show that pharmacologic PI3K or AKT inhibition, but not mTORC1 inhibition, causes a neuronal differentiation delay, mimicking the patient phenotype. Together these data suggest that heterozygous TSC2 mutations disrupt neuronal development, potentially contributing to the disease neuropathology, and that this defect may result from dysregulated PI3K/AKT signaling in neural progenitor cells.
Collapse
Affiliation(s)
- Avery J Zucco
- Graduate Program in Neuroscience, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, United States of America; Department of Cell Biology and Neuroscience, Rutgers, the State University of New Jersey, Piscataway, NJ, United States of America
| | - Valentina Dal Pozzo
- Graduate Program in Neuroscience, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, United States of America; Department of Cell Biology and Neuroscience, Rutgers, the State University of New Jersey, Piscataway, NJ, United States of America
| | - Alina Afinogenova
- Department of Cell Biology and Neuroscience, Rutgers, the State University of New Jersey, Piscataway, NJ, United States of America
| | - Ronald P Hart
- Department of Cell Biology and Neuroscience, Rutgers, the State University of New Jersey, Piscataway, NJ, United States of America; Human Genetics Institute of New Jersey, Piscataway, NJ, United States of America
| | - Orrin Devinsky
- NYU Comprehensive Epilepsy Center, NYU Langone School of Medicine, New York, NY, United States of America
| | - Gabriella D'Arcangelo
- Department of Cell Biology and Neuroscience, Rutgers, the State University of New Jersey, Piscataway, NJ, United States of America; Human Genetics Institute of New Jersey, Piscataway, NJ, United States of America.
| |
Collapse
|
162
|
Further studies about Coactosin-like protein-1 affecting the migration of mouse neocortical neurons. J Mol Histol 2018; 49:519-530. [PMID: 30128637 DOI: 10.1007/s10735-018-9790-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Accepted: 07/27/2018] [Indexed: 12/18/2022]
Abstract
During the development of mammalian cortex, late neurons generated by neuronal progenitors bypass earlier-born neurons and migrate to reach upper layers of cortical plate in an inner-to-outer fashion. Filamentous-actin (F-actin) can regulate neuronal migration, whereas Coactosin-like protein 1 (Cotl1) modulates F-actin. Lys 75 and Arg 73 of Cotl1 play an important role in binding F-actin; when they are mutated to Glu, Cotl1 cannot bind F-actin, called as a non-actin-binding mutant (ABM). The Lys 131 site of Cotl1, the 5-Lipoxygenase (5LO) binding site, is spatially close to Lys 75, leading to impact the binding of Cotl1 to F-actin. When Lys 131 is mutated to Ala (K131A), Cotl1 cannot bind to 5LO. We have demonstrated that overexpression of Cotl1 inhibited neuronal migration and increased the length of neuronal leading processes. To further explore cellular and molecular mechanisms of Cotl1's effect on neuronal migration, we constructed two mutant vectors-Cotl1-ABM and Cotl1-K131A and studied using in utero electroporation and primary neuronal culture technique. Results indicated that in the Cotl1-ABM group, the neuronal migration and length of the leading process both recovered as control neurons at the postnatal day 1 (P1), while in the Cotl1-K131A group, numerous neurons remained in deeper layers of cortical plate or intermediate zone. However, at P7, most Cotl1-K131A transfected neurons reached their destination. Moreover, we found that overexpression of Cotl1 inhibited the proliferation and mitotic activity of NPs. Therefore, These results demonstrated that Cotl1 played an important role in mouse neocortical development.
Collapse
|
163
|
Wu Y, Qi F, Song D, He Z, Zuo Z, Yang Y, Liu Q, Hu S, Wang X, Zheng X, Yang J, Yuan Q, Zou J, Guo K, Yao Z. Prenatal influenza vaccination rescues impairments of social behavior and lamination in a mouse model of autism. J Neuroinflammation 2018; 15:228. [PMID: 30103815 PMCID: PMC6090662 DOI: 10.1186/s12974-018-1252-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 07/11/2018] [Indexed: 12/14/2022] Open
Abstract
Background Prenatal infection is a substantial risk factor for neurodevelopmental disorders such as autism in offspring. We have previously reported that influenza vaccination (VAC) during early pregnancy contributes to neurogenesis and behavioral function in offspring. Results Here, we probe the efficacy of VAC pretreatment on autism-like behaviors in a lipopolysaccharide (LPS)-induced maternal immune activation (MIA) mouse model. We show that VAC improves abnormal fetal brain cytoarchitecture and lamination, an effect associated with promotion of intermediate progenitor cell differentiation in MIA fetal brain. These beneficial effects are sufficient to prevent social deficits in adult MIA offspring. Furthermore, whole-genome analysis suggests a strong interaction between Ikzf1 (IKAROS family zinc-finger 1) and neuronal differentiation. Intriguingly, VAC rescues excessive microglial Ikzf1 expression and attenuates microglial inflammatory responses in the MIA fetal brain. Conclusions Our study implies that a preprocessed influenza vaccination prevents maternal bacterial infection from causing neocortical lamination impairments and autism-related behaviors in offspring. Electronic supplementary material The online version of this article (10.1186/s12974-018-1252-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yingying Wu
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Fangfang Qi
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Dan Song
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Zitian He
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Zejie Zuo
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Yunjie Yang
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Qiongliang Liu
- Department of Thoracic Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Saisai Hu
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Xiao Wang
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Xiaona Zheng
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Junhua Yang
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Qunfang Yuan
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Juntao Zou
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Kaihua Guo
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Zhibin Yao
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou, 510080, China. .,Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou, 510080, China.
| |
Collapse
|
164
|
Vieira MS, Santos AK, Vasconcellos R, Goulart VAM, Parreira RC, Kihara AH, Ulrich H, Resende RR. Neural stem cell differentiation into mature neurons: Mechanisms of regulation and biotechnological applications. Biotechnol Adv 2018; 36:1946-1970. [PMID: 30077716 DOI: 10.1016/j.biotechadv.2018.08.002] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 07/31/2018] [Accepted: 08/01/2018] [Indexed: 02/07/2023]
Abstract
The abilities of stem cells to self-renew and form different mature cells expand the possibilities of applications in cell-based therapies such as tissue recomposition in regenerative medicine, drug screening, and treatment of neurodegenerative diseases. In addition to stem cells found in the embryo, various adult organs and tissues have niches of stem cells in an undifferentiated state. In the central nervous system of adult mammals, neurogenesis occurs in two regions: the subventricular zone and the dentate gyrus in the hippocampus. The generation of the different neural lines originates in adult neural stem cells that can self-renew or differentiate into astrocytes, oligodendrocytes, or neurons in response to specific stimuli. The regulation of the fate of neural stem cells is a finely controlled process relying on a complex regulatory network that extends from the epigenetic to the translational level and involves extracellular matrix components. Thus, a better understanding of the mechanisms underlying how the process of neurogenesis is induced, regulated, and maintained will provide elues for development of novel for strategies for neurodegenerative therapies. In this review, we focus on describing the mechanisms underlying the regulation of the neuronal differentiation process by transcription factors, microRNAs, and extracellular matrix components.
Collapse
Affiliation(s)
- Mariana S Vieira
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Instituto Nanocell, Divinopólis, MG, Brazil
| | - Anderson K Santos
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Rebecca Vasconcellos
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Instituto Nanocell, Divinopólis, MG, Brazil
| | - Vânia A M Goulart
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ricardo C Parreira
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Instituto Nanocell, Divinopólis, MG, Brazil
| | - Alexandre H Kihara
- Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
| | - Henning Ulrich
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, SP, Brazil.
| | - Rodrigo R Resende
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Instituto Nanocell, Divinopólis, MG, Brazil.
| |
Collapse
|
165
|
Ozair MZ, Kirst C, van den Berg BL, Ruzo A, Rito T, Brivanlou AH. hPSC Modeling Reveals that Fate Selection of Cortical Deep Projection Neurons Occurs in the Subplate. Cell Stem Cell 2018; 23:60-73.e6. [PMID: 29937203 DOI: 10.1016/j.stem.2018.05.024] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 03/13/2018] [Accepted: 05/23/2018] [Indexed: 01/29/2023]
Abstract
Cortical deep projection neurons (DPNs) are implicated in neurodevelopmental disorders. Although recent findings emphasize post-mitotic programs in projection neuron fate selection, the establishment of primate DPN identity during layer formation is not well understood. The subplate lies underneath the developing cortex and is a post-mitotic compartment that is transiently and disproportionately enlarged in primates in the second trimester. The evolutionary significance of subplate expansion, the molecular identity of its neurons, and its contribution to primate corticogenesis remain open questions. By modeling subplate formation with human pluripotent stem cells (hPSCs), we show that all classes of cortical DPNs can be specified from subplate neurons (SPNs). Post-mitotic WNT signaling regulates DPN class selection, and DPNs in the caudal fetal cortex appear to exclusively derive from SPNs. Our findings indicate that SPNs have evolved in primates as an important source of DPNs that contribute to cortical lamination prior to their known role in circuit formation.
Collapse
Affiliation(s)
- M Zeeshan Ozair
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Christoph Kirst
- Center for Studies in Physics and Biology and Kavli Neural Systems Institute, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Bastiaan L van den Berg
- Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Sciencepark 904, 1098XH Amsterdam, the Netherlands
| | - Albert Ruzo
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Tiago Rito
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Ali H Brivanlou
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
166
|
Nguyen LS, Fregeac J, Bole-Feysot C, Cagnard N, Iyer A, Anink J, Aronica E, Alibeu O, Nitschke P, Colleaux L. Role of miR-146a in neural stem cell differentiation and neural lineage determination: relevance for neurodevelopmental disorders. Mol Autism 2018; 9:38. [PMID: 29951184 PMCID: PMC6011198 DOI: 10.1186/s13229-018-0219-3] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 05/30/2018] [Indexed: 12/21/2022] Open
Abstract
Background MicroRNAs (miRNAs) are small, non-coding RNAs that regulate gene expression at the post-transcriptional level. miRNAs have emerged as important modulators of brain development and neuronal function and are implicated in several neurological diseases. Previous studies found miR-146a upregulation is the most common miRNA deregulation event in neurodevelopmental disorders such as autism spectrum disorder (ASD), epilepsy, and intellectual disability (ID). Yet, how miR-146a upregulation affects the developing fetal brain remains unclear. Methods We analyzed the expression of miR-146a in the temporal lobe of ASD children using Taqman assay. To assess the role of miR-146a in early brain development, we generated and characterized stably induced H9 human neural stem cell (H9 hNSC) overexpressing miR-146a using various cell and molecular biology techniques. Results We first showed that miR-146a upregulation occurs early during childhood in the ASD brain. In H9 hNSC, miR-146a overexpression enhances neurite outgrowth and branching and favors differentiation into neuronal like cells. Expression analyses revealed that 10% of the transcriptome was deregulated and organized into two modules critical for cell cycle control and neuronal differentiation. Twenty known or predicted targets of miR-146a were significantly deregulated in the modules, acting as potential drivers. The two modules also display distinct transcription profiles during human brain development, affecting regions relevant for ASD including the neocortex, amygdala, and hippocampus. Cell type analyses indicate markers for pyramidal, and interneurons are highly enriched in the deregulated gene list. Up to 40% of known markers of newly defined neuronal lineages were deregulated, suggesting that miR-146a could participate also in the acquisition of neuronal identities. Conclusion Our results demonstrate the dynamic roles of miR-146a in early neuronal development and provide new insight into the molecular events that link miR-146a overexpression to impaired neurodevelopment. This, in turn, may yield new therapeutic targets and strategies. Electronic supplementary material The online version of this article (10.1186/s13229-018-0219-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lam Son Nguyen
- 1INSERM UMR 1163, Laboratory of Molecular and pathophysiological bases of cognitive disorders, Imagine Institute, Necker-Enfants Malades Hospital, 24 Boulevard du Montparnasse, 75015 Paris, France.,Paris Descartes-Sorbonne Paris Cité University, 12 Rue de l'École de Médecine, 75006 Paris, France
| | - Julien Fregeac
- 1INSERM UMR 1163, Laboratory of Molecular and pathophysiological bases of cognitive disorders, Imagine Institute, Necker-Enfants Malades Hospital, 24 Boulevard du Montparnasse, 75015 Paris, France.,Paris Descartes-Sorbonne Paris Cité University, 12 Rue de l'École de Médecine, 75006 Paris, France
| | - Christine Bole-Feysot
- 1INSERM UMR 1163, Laboratory of Molecular and pathophysiological bases of cognitive disorders, Imagine Institute, Necker-Enfants Malades Hospital, 24 Boulevard du Montparnasse, 75015 Paris, France
| | - Nicolas Cagnard
- 1INSERM UMR 1163, Laboratory of Molecular and pathophysiological bases of cognitive disorders, Imagine Institute, Necker-Enfants Malades Hospital, 24 Boulevard du Montparnasse, 75015 Paris, France
| | - Anand Iyer
- 3Department of (Neuro) Pathology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Jasper Anink
- 3Department of (Neuro) Pathology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Eleonora Aronica
- 3Department of (Neuro) Pathology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Olivier Alibeu
- 1INSERM UMR 1163, Laboratory of Molecular and pathophysiological bases of cognitive disorders, Imagine Institute, Necker-Enfants Malades Hospital, 24 Boulevard du Montparnasse, 75015 Paris, France
| | - Patrick Nitschke
- 1INSERM UMR 1163, Laboratory of Molecular and pathophysiological bases of cognitive disorders, Imagine Institute, Necker-Enfants Malades Hospital, 24 Boulevard du Montparnasse, 75015 Paris, France
| | - Laurence Colleaux
- 1INSERM UMR 1163, Laboratory of Molecular and pathophysiological bases of cognitive disorders, Imagine Institute, Necker-Enfants Malades Hospital, 24 Boulevard du Montparnasse, 75015 Paris, France.,Paris Descartes-Sorbonne Paris Cité University, 12 Rue de l'École de Médecine, 75006 Paris, France
| |
Collapse
|
167
|
PARD3 dysfunction in conjunction with dynamic HIPPO signaling drives cortical enlargement with massive heterotopia. Genes Dev 2018; 32:763-780. [PMID: 29899142 PMCID: PMC6049519 DOI: 10.1101/gad.313171.118] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 05/07/2018] [Indexed: 12/22/2022]
Abstract
In this study, Liu et al. investigated the molecular mechanisms regulating the proper organization and orderly mitosis of radial glial progenitors (RGPs), which drive the formation of a laminated mammalian cortex in the correct size. They show that RGP behavior and cortical development are controlled by partitioning-defective 3 (PARD3) in concert with dynamic HIPPO and NOTCH signaling regulation, thus providing new insights into mammalian cortical development. Proper organization and orderly mitosis of radial glial progenitors (RGPs) drive the formation of a laminated mammalian cortex in the correct size. However, the molecular underpinnings of the intricate process remain largely unclear. Here we show that RGP behavior and cortical development are controlled by temporally distinct actions of partitioning-defective 3 (PARD3) in concert with dynamic HIPPO signaling. RGPs lacking PARD3 exhibit developmental stage-dependent abnormal switches in division mode, resulting in an initial overproduction of RGPs located largely outside the ventricular zone at the expense of deep-layer neurons. Ectopically localized RGPs subsequently undergo accelerated and excessive neurogenesis, leading to the formation of an enlarged cortex with massive heterotopia and increased seizure susceptibility. Simultaneous removal of HIPPO pathway effectors Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) suppresses cortical enlargement and heterotopia formation. These results define a dynamic regulatory program of mammalian cortical development and highlight a progenitor origin of megalencephaly with ribbon heterotopia and epilepsy.
Collapse
|
168
|
Abstract
This review by O'Brown et al. discusses the cellular nature of the blood–brain barrier (BBB) and the conservation and variation of BBB function across taxa. It compares the BBB across organisms in order to provide insight into the human BBB both under normal physiological conditions and in neurological diseases. The blood–brain barrier (BBB) restricts free access of molecules between the blood and the brain and is essential for regulating the neural microenvironment. Here, we describe how the BBB was initially characterized and how the current field evaluates barrier properties. We next detail the cellular nature of the BBB and discuss both the conservation and variation of BBB function across taxa. Finally, we examine our current understanding of mouse and zebrafish model systems, as we expect that comparison of the BBB across organisms will provide insight into the human BBB under normal physiological conditions and in neurological diseases.
Collapse
Affiliation(s)
- Natasha M O'Brown
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Sarah J Pfau
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Chenghua Gu
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
169
|
Baizabal JM, Mistry M, García MT, Gómez N, Olukoya O, Tran D, Johnson MB, Walsh CA, Harwell CC. The Epigenetic State of PRDM16-Regulated Enhancers in Radial Glia Controls Cortical Neuron Position. Neuron 2018; 98:945-962.e8. [PMID: 29779941 PMCID: PMC6667181 DOI: 10.1016/j.neuron.2018.04.033] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 03/14/2018] [Accepted: 04/24/2018] [Indexed: 01/09/2023]
Abstract
The epigenetic landscape is dynamically remodeled during neurogenesis. However, it is not understood how chromatin modifications in neural stem cells instruct the formation of complex structures in the brain. We report that the histone methyltransferase PRDM16 is required in radial glia to regulate lineage-autonomous and stage-specific gene expression programs that control number and position of upper layer cortical projection neurons. PRDM16 regulates the epigenetic state of transcriptional enhancers to activate genes involved in intermediate progenitor cell production and repress genes involved in cell migration. The histone methyltransferase domain of PRDM16 is necessary in radial glia to promote cortical neuron migration through transcriptional silencing. We show that repression of the gene encoding the E3 ubiquitin ligase PDZRN3 by PRDM16 determines the position of upper layer neurons. These findings provide insights into how epigenetic control of transcriptional enhancers in radial glial determines the organization of the mammalian cerebral cortex.
Collapse
Affiliation(s)
| | - Meeta Mistry
- Bioinformatics Core, Department of Biostatistics, Harvard T.H. Chan School of Public Health, Harvard Medical School, Boston, MA 02115, USA
| | | | - Nicolás Gómez
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Olubusola Olukoya
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Diana Tran
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Matthew B Johnson
- Division of Genetics and Genomics, Manton Center for Orphan Disease and Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA 02115, USA
| | - Christopher A Walsh
- Division of Genetics and Genomics, Manton Center for Orphan Disease and Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA 02115, USA
| | - Corey C Harwell
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
170
|
In utero electroporation-based translating ribosome affinity purification identifies age-dependent mRNA expression in cortical pyramidal neurons. Neurosci Res 2018; 143:44-52. [PMID: 29857015 DOI: 10.1016/j.neures.2018.05.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 05/10/2018] [Accepted: 05/28/2018] [Indexed: 02/02/2023]
Abstract
We combined translating ribosome affinity purification (TRAP) with in utero electroporation (IUE), called iTRAP to identify the molecular profile of specific neuronal populations during neonatal development without the need for viral approaches and FACS sorting. We electroporated a plasmid encoding EGFP-tagged ribosomal protein L10a at embryonic day (E) 14-15 to target layer 2-4 cortical neurons of the somatosensory cortex. At three postnatal (P) ages-P0, P7, and P14-when morphogenesis occurs and synapses are forming, TRAP and molecular profiling was performed from electroporated regions. We found that ribosome bound (Ribo)-mRNAs from ∼7300 genes were significantly altered over time and included classical neuronal genes known to decrease (e.g., Tbr1, Dcx) or increase (e.g., Eno2, Camk2a, Syn1) as neurons mature. This approach led to the identification of specific developmental patterns for Ribo-mRNAs not previously reported to be developmentally regulated in neurons, providing rationale for future examination of their role in selective biological processes. These include upregulation of Lynx1, Nrn1, Cntnap1 over time; downregulation of St8sia2 and Draxin; and bidirectional changes to Fkbp1b. iTRAP is a versatile approach that allows researchers to easily assess the molecular profile of specific neuronal populations in selective brain regions under various conditions, including overexpression and knockdown of target genes, and in disease settings.
Collapse
|
171
|
Bertacchi M, Parisot J, Studer M. The pleiotropic transcriptional regulator COUP-TFI plays multiple roles in neural development and disease. Brain Res 2018; 1705:75-94. [PMID: 29709504 DOI: 10.1016/j.brainres.2018.04.024] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 04/19/2018] [Accepted: 04/20/2018] [Indexed: 12/23/2022]
Abstract
Transcription factors are expressed in a dynamic fashion both in time and space during brain development, and exert their roles by activating a cascade of multiple target genes. This implies that understanding the precise function of a transcription factor becomes a challenging task. In this review, we will focus on COUP-TFI (or NR2F1), a nuclear receptor belonging to the superfamily of the steroid/thyroid hormone receptors, and considered to be one of the major transcriptional regulators orchestrating cortical arealization, cell-type specification and maturation. Recent data have unraveled the multi-faceted functions of COUP-TFI in the development of several mouse brain structures, including the neocortex, hippocampus and ganglionic eminences. Despite NR2F1 mutations and deletions in humans have been linked to a complex neurodevelopmental disease mainly associated to optic atrophy and intellectual disability, its role during the formation of the retina and optic nerve remains unclear. In light of its major influence in cortical development, we predict that its haploinsufficiency might be the cause of other cognitive diseases, not identified so far. Mouse models offer a unique opportunity of dissecting COUP-TFI function in different regions during brain assembly; hence, the importance of comparing and discussing common points linking mouse models to human patients' symptoms.
Collapse
Affiliation(s)
- Michele Bertacchi
- Université Côte d'Azur, CNRS, Inserm, iBV - Institut de Biologie Valrose, 06108 Nice, France.
| | - Josephine Parisot
- Université Côte d'Azur, CNRS, Inserm, iBV - Institut de Biologie Valrose, 06108 Nice, France
| | - Michèle Studer
- Université Côte d'Azur, CNRS, Inserm, iBV - Institut de Biologie Valrose, 06108 Nice, France.
| |
Collapse
|
172
|
Ouyang M, Dubois J, Yu Q, Mukherjee P, Huang H. Delineation of early brain development from fetuses to infants with diffusion MRI and beyond. Neuroimage 2018; 185:836-850. [PMID: 29655938 DOI: 10.1016/j.neuroimage.2018.04.017] [Citation(s) in RCA: 165] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 04/01/2018] [Accepted: 04/08/2018] [Indexed: 02/08/2023] Open
Abstract
Dynamic macrostructural and microstructural changes take place from the mid-fetal stage to 2 years after birth. Delineating structural changes of the brain during early development provides new insights into the complicated processes of both typical development and the pathological mechanisms underlying various psychiatric and neurological disorders including autism, attention deficit hyperactivity disorder and schizophrenia. Decades of histological studies have identified strong spatial and functional maturation gradients in human brain gray and white matter. The recent improvements in magnetic resonance imaging (MRI) techniques, especially diffusion MRI (dMRI), relaxometry imaging, and magnetization transfer imaging (MTI) have provided unprecedented opportunities to non-invasively quantify and map the early developmental changes at whole brain and regional levels. Here, we review the recent advances in understanding early brain structural development during the second half of gestation and the first two postnatal years using modern MR techniques. Specifically, we review studies that delineate the emergence and microstructural maturation of white matter tracts, as well as dynamic mapping of inhomogeneous cortical microstructural organization unique to fetuses and infants. These imaging studies converge into maturational curves of MRI measurements that are distinctive across different white matter tracts and cortical regions. Furthermore, contemporary models offering biophysical interpretations of the dMRI-derived measurements are illustrated to infer the underlying microstructural changes. Collectively, this review summarizes findings that contribute to charting spatiotemporally heterogeneous gray and white matter structural development, offering MRI-based biomarkers of typical brain development and setting the stage for understanding aberrant brain development in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Minhui Ouyang
- Radiology Research, Children's Hospital of Philadelphia, PA, United States
| | - Jessica Dubois
- INSERM, UMR992, CEA, NeuroSpin Center, University Paris Saclay, Gif-sur-Yvette, France
| | - Qinlin Yu
- Radiology Research, Children's Hospital of Philadelphia, PA, United States
| | - Pratik Mukherjee
- Department of Radiology & Biomedical Imaging, University of California, San Francisco, CA, United States
| | - Hao Huang
- Radiology Research, Children's Hospital of Philadelphia, PA, United States; Department of Radiology, Perelman School of Medicine, University of Pennsylvania, PA, United States.
| |
Collapse
|
173
|
Mukhtar T, Taylor V. Untangling Cortical Complexity During Development. J Exp Neurosci 2018; 12:1179069518759332. [PMID: 29551911 PMCID: PMC5846925 DOI: 10.1177/1179069518759332] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 01/23/2018] [Indexed: 12/23/2022] Open
Abstract
The cerebral cortex is composed of billions of morphologically and functionally distinct neurons. These neurons are produced and organized in a regimental fashion during development. The ability of neurons to encode and elicit complex cognitive and motor functions depends on their precise molecular processes, identity, and connectivity established during development. Elucidating the cellular and molecular mechanisms that regulate development of the neocortex has been a challenge for many years. The cerebral cortical neuronal subtypes are classified based on morphology, function, intrinsic synaptic properties, location, connectivity, and marker gene expression. Development of the neocortex requires an orchestration of a series of processes including the appropriate determination, migration and positioning of the neurons, acquisition of layer-specific transcriptional hallmarks, and formation of precise axonal projections and networks. Historically, fate mapping, genome-wide analysis, and transcriptome profiling have provided many opportunities for the characterization of neuronal subtypes. During the course of this review, we will address the regimental organization of the cerebral cortex, dissect the cellular subtypes that contribute to cortical complexity, and outline their molecular hallmarks to understand cellular diversity in the cerebral cortex with a focus on the excitatory neurons.
Collapse
Affiliation(s)
- Tanzila Mukhtar
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Verdon Taylor
- Department of Biomedicine, University of Basel, Basel, Switzerland
| |
Collapse
|
174
|
Mehta S, Lo Cascio C. Developmentally regulated signaling pathways in glioma invasion. Cell Mol Life Sci 2018; 75:385-402. [PMID: 28821904 PMCID: PMC5765207 DOI: 10.1007/s00018-017-2608-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 07/18/2017] [Accepted: 08/03/2017] [Indexed: 01/06/2023]
Abstract
Malignant gliomas are the most common, infiltrative, and lethal primary brain tumors affecting the adult population. The grim prognosis for this disease is due to a combination of the presence of highly invasive tumor cells that escape surgical resection and the presence of a population of therapy-resistant cancer stem cells found within these tumors. Several studies suggest that glioma cells have cleverly hijacked the normal developmental program of neural progenitor cells, including their transcriptional programs, to enhance gliomagenesis. In this review, we summarize the role of developmentally regulated signaling pathways that have been found to facilitate glioma growth and invasion. Furthermore, we discuss how the microenvironment and treatment-induced perturbations of these highly interconnected signaling networks can trigger a shift in cellular phenotype and tumor subtype.
Collapse
Affiliation(s)
- Shwetal Mehta
- Division of Neurobiology, Barrow Brain Tumor Research Center, Barrow Neurological Institute, Phoenix, AZ, 85013, USA.
| | - Costanza Lo Cascio
- Division of Neurobiology, Barrow Brain Tumor Research Center, Barrow Neurological Institute, Phoenix, AZ, 85013, USA
| |
Collapse
|
175
|
Zahr SK, Yang G, Kazan H, Borrett MJ, Yuzwa SA, Voronova A, Kaplan DR, Miller FD. A Translational Repression Complex in Developing Mammalian Neural Stem Cells that Regulates Neuronal Specification. Neuron 2018; 97:520-537.e6. [PMID: 29395907 DOI: 10.1016/j.neuron.2017.12.045] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 11/22/2017] [Accepted: 12/28/2017] [Indexed: 01/28/2023]
Abstract
The mechanisms instructing genesis of neuronal subtypes from mammalian neural precursors are not well understood. To address this issue, we have characterized the transcriptional landscape of radial glial precursors (RPs) in the embryonic murine cortex. We show that individual RPs express mRNA, but not protein, for transcriptional specifiers of both deep and superficial layer cortical neurons. Some of these mRNAs, including the superficial versus deep layer neuron transcriptional regulators Brn1 and Tle4, are translationally repressed by their association with the RNA-binding protein Pumilio2 (Pum2) and the 4E-T protein. Disruption of these repressive complexes in RPs mid-neurogenesis by knocking down 4E-T or Pum2 causes aberrant co-expression of deep layer neuron specification proteins in newborn superficial layer neurons. Thus, cortical RPs are transcriptionally primed to generate diverse types of neurons, and a Pum2/4E-T complex represses translation of some of these neuronal identity mRNAs to ensure appropriate temporal specification of daughter neurons.
Collapse
Affiliation(s)
- Siraj K Zahr
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON M5G 1L7, Canada; Institute of Medical Science, University of Toronto, Toronto, ON M5G 1A8, Canada
| | - Guang Yang
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON M5G 1L7, Canada
| | - Hilal Kazan
- Department of Computer Engineering, Antalya Bilim University, Antalya, Turkey
| | - Michael J Borrett
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON M5G 1L7, Canada; Institute of Medical Science, University of Toronto, Toronto, ON M5G 1A8, Canada
| | - Scott A Yuzwa
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON M5G 1L7, Canada
| | - Anastassia Voronova
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON M5G 1L7, Canada
| | - David R Kaplan
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON M5G 1L7, Canada; Institute of Medical Science, University of Toronto, Toronto, ON M5G 1A8, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1A8, Canada
| | - Freda D Miller
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON M5G 1L7, Canada; Institute of Medical Science, University of Toronto, Toronto, ON M5G 1A8, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1A8, Canada; Department of Physiology, University of Toronto, Toronto, ON M5G 1A8, Canada.
| |
Collapse
|
176
|
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]
|
177
|
Huang B, Li X, Tu X, Zhao W, Zhu D, Feng Y, Si X, Chen JG. OTX1 regulates cell cycle progression of neural progenitors in the developing cerebral cortex. J Biol Chem 2017; 293:2137-2148. [PMID: 29273633 DOI: 10.1074/jbc.ra117.001249] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Indexed: 01/01/2023] Open
Abstract
The progenitor cells in the cerebral cortex coordinate proliferation and mitotic exit to generate the correct number of neurons and glial cells during development. However, mechanisms for regulating the mitotic cycle of cortical progenitors are not fully understood. Otx1 is one of the homeobox-containing transcription factors frequently implicated in the development of the central nervous system. Mice bearing a targeted deletion of Otx1 exhibit brain hypoplasia and a decrease in the number of cortical neurons. We hypothesized that Otx1 might be crucial to the proliferation and differentiation of cortical progenitors. Otx1 knockdown by in utero electroporation in the mouse brain reduced the proportion of the G1 phase while increasing the S and M phases of progenitor cells. The knockdown diminished Tbr1+ neurons but increased GFAP+ astrocytes in the early postnatal cortex as revealed by lineage tracing study. Tbr2+ basal progenitors lacking Otx1 were held at the transit-amplifying stage. In contrast, overexpression of wildtype Otx1 but not an astrocytoma-related mutant Y320C inhibited proliferation of the progenitor cells in embryonic cortex. This study demonstrates that Otx1 is one of the key elements regulating cortical neurogenesis, and a loss-of-function in Otx1 may contribute to the overproduction of astrocytes in vivo.
Collapse
Affiliation(s)
- Baoshan Huang
- From the School of Ophthalmology and Optometry and Eye Hospital.,the State Key Laboratory of Optometry, Ophthalmology and Vision Science, and.,the Zhejiang Provincial Key Laboratory of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Xue Li
- From the School of Ophthalmology and Optometry and Eye Hospital.,the State Key Laboratory of Optometry, Ophthalmology and Vision Science, and.,the Zhejiang Provincial Key Laboratory of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Xiaomeng Tu
- From the School of Ophthalmology and Optometry and Eye Hospital.,the State Key Laboratory of Optometry, Ophthalmology and Vision Science, and.,the Zhejiang Provincial Key Laboratory of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Wei Zhao
- From the School of Ophthalmology and Optometry and Eye Hospital.,the State Key Laboratory of Optometry, Ophthalmology and Vision Science, and.,the Zhejiang Provincial Key Laboratory of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Dan Zhu
- From the School of Ophthalmology and Optometry and Eye Hospital.,the State Key Laboratory of Optometry, Ophthalmology and Vision Science, and.,the Zhejiang Provincial Key Laboratory of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Yue Feng
- From the School of Ophthalmology and Optometry and Eye Hospital.,the State Key Laboratory of Optometry, Ophthalmology and Vision Science, and.,the Zhejiang Provincial Key Laboratory of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Xiang Si
- From the School of Ophthalmology and Optometry and Eye Hospital.,the State Key Laboratory of Optometry, Ophthalmology and Vision Science, and.,the Zhejiang Provincial Key Laboratory of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Jie-Guang Chen
- From the School of Ophthalmology and Optometry and Eye Hospital, .,the State Key Laboratory of Optometry, Ophthalmology and Vision Science, and.,the Zhejiang Provincial Key Laboratory of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| |
Collapse
|
178
|
Oishi K, Nakajima K. Subtype Specification of Cerebral Cortical Neurons in Their Immature Stages. Neurochem Res 2017; 43:238-244. [PMID: 29185180 DOI: 10.1007/s11064-017-2441-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 11/21/2017] [Accepted: 11/23/2017] [Indexed: 10/18/2022]
Abstract
The diversification of neuronal subtypes during corticogenesis is fundamental to the establishment of the complex cortical structure. Although subtype specification has been assumed to occur in neural progenitor cells, increasing evidence has begun to reveal the plasticity of subtype determination in immature neurons. Here, we summarize recent findings regarding the regulation of subtype specification during later periods of neuronal differentiation, such as the post-mitotic and post-migratory stages. We also discuss thalamocortical axons as an extra-cortical cue that provides information on the subtype determination of immature cortical neurons.
Collapse
Affiliation(s)
- Koji Oishi
- Department of Anatomy, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| | - Kazunori Nakajima
- Department of Anatomy, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| |
Collapse
|
179
|
Ma YX, Wang XL, Chen JQ, Li B, Hur EM, Saijilafu. Differential Roles of Glycogen Synthase Kinase 3 Subtypes Alpha and Beta in Cortical Development. Front Mol Neurosci 2017; 10:391. [PMID: 29234272 PMCID: PMC5712306 DOI: 10.3389/fnmol.2017.00391] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 11/09/2017] [Indexed: 11/13/2022] Open
Abstract
Glycogen synthase kinases 3 (GSK3) α and β are expressed in the nervous system, and disruption of GSK3 signaling has been implicated in a wide range of neurodevelopmental and psychiatric disorders. Although several studies have established a role of GSK3 signaling in the nervous system, much less is known about isoform-specific functions. Here, we have examined the role of GSK3α and GSK3β in the developing neocortex by performing in utero electroporation with specific small interfering RNAs targeting each isoform. We found that depletion of either GSK3α or GSK3β commonly promoted the proliferation of neural progenitor cells in the ventricular zone, but at later stages, knocking down of each isoform resulted in distinct outcomes. In particular, the transformation of radial progenitors to intermediate progenitor cells was promoted in GSK3α-depleted cells, but markedly prevented in GSK3β-depleted cells. Moreover, knocking down of GSK3β but not GSK3α prevented the generation of upper-layer Cux1+ neurons. Consistent with the distinct outcomes, protein levels of c-Myc and β-catenin, well-known substrates of GSK3, were differentially affected by depletion of GSK3α and GSK3β. Together, these results suggest that GSK3α and GSK3β might play distinct roles in the genesis and differentiation of neuronal lineage cells during neocortex development by differential regulation of downstream signaling pathways.
Collapse
Affiliation(s)
- Yan-Xia Ma
- Department of Orthopaedics, The First Affiliated Hospital, Orthopaedic Institute, Soochow University, Suzhou, China
| | - Xiu-Li Wang
- Department of Orthopaedics, The First Affiliated Hospital, Orthopaedic Institute, Soochow University, Suzhou, China
| | - Jian-Quan Chen
- Department of Orthopaedics, The First Affiliated Hospital, Orthopaedic Institute, Soochow University, Suzhou, China
| | - Bin Li
- Department of Orthopaedics, The First Affiliated Hospital, Orthopaedic Institute, Soochow University, Suzhou, China
| | - Eun-Mi Hur
- Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology, Seoul, South Korea.,Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology, Seoul, South Korea.,Department of Neuroscience, Korea University of Science and Technology, Daejeon, South Korea
| | - Saijilafu
- Department of Orthopaedics, The First Affiliated Hospital, Orthopaedic Institute, Soochow University, Suzhou, China
| |
Collapse
|
180
|
Ledonne F, Orduz D, Mercier J, Vigier L, Grove EA, Tissir F, Angulo MC, Pierani A, Coppola E. Targeted Inactivation of Bax Reveals a Subtype-Specific Mechanism of Cajal-Retzius Neuron Death in the Postnatal Cerebral Cortex. Cell Rep 2017; 17:3133-3141. [PMID: 28009284 DOI: 10.1016/j.celrep.2016.11.074] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 09/26/2016] [Accepted: 11/23/2016] [Indexed: 12/31/2022] Open
Abstract
Cajal-Retzius cells (CRs), the first-born neurons in the developing cerebral cortex, coordinate crucial steps in the construction of functional circuits. CRs are thought to be transient, as they disappear during early postnatal life in both mice and humans, where their abnormal persistence is associated with pathological conditions. Embryonic CRs comprise at least three molecularly and functionally distinct subtypes: septum, ventral pallium/pallial-subpallial boundary (PSB), and hem. However, whether subtype-specific features exist postnatally and through which mechanisms they disappear remain unknown. We report that CR subtypes display unique distributions and dynamics of death in the postnatal mouse cortex. Surprisingly, although all CR subtypes undergo cell death, septum, but not hem, CRs die in a Bax-dependent manner. Bax-inactivated rescued septum-CRs maintain immature electrophysiological properties. These results underlie the existence of an exquisitely refined control of developmental cell death and provide a model to test the effect of maintaining immature circuits in the adult neocortex.
Collapse
Affiliation(s)
- Fanny Ledonne
- Institut Jacques Monod, CNRS UMR 7592, Université Paris Diderot, Sorbonne Paris Cité, 75205 Paris Cedex 13, France
| | - David Orduz
- INSERM U1128, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, 75006 Paris, France
| | - Judith Mercier
- Institut Jacques Monod, CNRS UMR 7592, Université Paris Diderot, Sorbonne Paris Cité, 75205 Paris Cedex 13, France
| | - Lisa Vigier
- Institut Jacques Monod, CNRS UMR 7592, Université Paris Diderot, Sorbonne Paris Cité, 75205 Paris Cedex 13, France
| | | | - Fadel Tissir
- Université Catholique de Louvain, Institute of Neuroscience, 1200 Brussels, Belgium
| | - Maria Cecilia Angulo
- INSERM U1128, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, 75006 Paris, France
| | - Alessandra Pierani
- Institut Jacques Monod, CNRS UMR 7592, Université Paris Diderot, Sorbonne Paris Cité, 75205 Paris Cedex 13, France.
| | - Eva Coppola
- Institut Jacques Monod, CNRS UMR 7592, Université Paris Diderot, Sorbonne Paris Cité, 75205 Paris Cedex 13, France.
| |
Collapse
|
181
|
Nitarska J, Smith JG, Sherlock WT, Hillege MMG, Nott A, Barshop WD, Vashisht AA, Wohlschlegel JA, Mitter R, Riccio A. A Functional Switch of NuRD Chromatin Remodeling Complex Subunits Regulates Mouse Cortical Development. Cell Rep 2017; 17:1683-1698. [PMID: 27806305 PMCID: PMC5149529 DOI: 10.1016/j.celrep.2016.10.022] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 08/03/2016] [Accepted: 10/07/2016] [Indexed: 12/31/2022] Open
Abstract
Histone modifications and chromatin remodeling represent universal mechanisms by which cells adapt their transcriptional response to rapidly changing environmental conditions. Extensive chromatin remodeling takes place during neuronal development, allowing the transition of pluripotent cells into differentiated neurons. Here, we report that the NuRD complex, which couples ATP-dependent chromatin remodeling with histone deacetylase activity, regulates mouse brain development. Subunit exchange of CHDs, the core ATPase subunits of the NuRD complex, is required for distinct aspects of cortical development. Whereas CHD4 promotes the early proliferation of progenitors, CHD5 facilitates neuronal migration and CHD3 ensures proper layer specification. Inhibition of each CHD leads to defects of neuronal differentiation and migration, which cannot be rescued by expressing heterologous CHDs. Finally, we demonstrate that NuRD complexes containing specific CHDs are recruited to regulatory elements and modulate the expression of genes essential for brain development. The ATPases CHD3, CHD4, and CHD5 are mutually exclusive subunits of the NuRD complex CHD3, CHD4, and CHD5 regulate distinct and non-redundant aspects of cortical development Loss of each CHD leads to specific defects of neuronal proliferation and migration CHD3, CHD4, and CHD5 regulate distinct set of genes essential for brain development
Collapse
Affiliation(s)
- Justyna Nitarska
- MRC Laboratory for Molecular and Cell Biology, University College London, London WC1E 6BT, UK
| | - Jacob G Smith
- MRC Laboratory for Molecular and Cell Biology, University College London, London WC1E 6BT, UK
| | - William T Sherlock
- MRC Laboratory for Molecular and Cell Biology, University College London, London WC1E 6BT, UK
| | - Michele M G Hillege
- MRC Laboratory for Molecular and Cell Biology, University College London, London WC1E 6BT, UK
| | - Alexi Nott
- MRC Laboratory for Molecular and Cell Biology, University College London, London WC1E 6BT, UK
| | - William D Barshop
- Department of Biological Chemistry, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1737 USA
| | - Ajay A Vashisht
- Department of Biological Chemistry, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1737 USA
| | - James A Wohlschlegel
- Department of Biological Chemistry, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1737 USA
| | - Richard Mitter
- Lincoln's Inn Fields Laboratory, The Francis Crick Institute, London WC2A 3LY, UK
| | - Antonella Riccio
- MRC Laboratory for Molecular and Cell Biology, University College London, London WC1E 6BT, UK.
| |
Collapse
|
182
|
Cheng X, Li K, Liu M, Hu X, Xu M, Yan R, Zhao S. P85 regulates neuronal migration through affecting neuronal morphology during mouse corticogenesis. Cell Tissue Res 2017; 372:23-31. [PMID: 29130119 DOI: 10.1007/s00441-017-2707-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Accepted: 10/04/2017] [Indexed: 11/25/2022]
Abstract
In mammalian developing embryonic cortex, projection neurons migrate from the ventricular zone to the cortical plate, guided by radial glial cells with a transformation between bipolar and multipolar morphology. Previous studies have demonstrated that the PI3K-Akt-mTOR signal plays a critical role in brain development. However, the function of P85 in cortical development is still unclear. In the present study, we found that overexpression of P85 impaired cortical neuronal migration. Using in utero electroporation, we revealed that the length of the leading process in P85 overexpressed neurons became shorter than that in the control group but with more branches. Using markers for new-born neurons, we further found that overexpression of P85 did not affect the ultimate fate of these cortical neurons. These findings indicated that the P85 subunit plays an essential role in neuronal migration and neuronal morphology during mouse corticogenesis.
Collapse
Affiliation(s)
- Xinran Cheng
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, People's Republic of China
| | - Kaikai Li
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, People's Republic of China
| | - MengMeng Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, People's Republic of China
| | - Xinde Hu
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, People's Republic of China
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Mingrui Xu
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100, Shaanxi, People's Republic of China
| | - Runchuan Yan
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, People's Republic of China
| | - Shanting Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, People's Republic of China.
| |
Collapse
|
183
|
Popovitchenko T, Rasin MR. Transcriptional and Post-Transcriptional Mechanisms of the Development of Neocortical Lamination. Front Neuroanat 2017; 11:102. [PMID: 29170632 PMCID: PMC5684109 DOI: 10.3389/fnana.2017.00102] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 10/25/2017] [Indexed: 12/31/2022] Open
Abstract
The neocortex is a laminated brain structure that is the seat of higher cognitive capacity and responses, long-term memory, sensory and emotional functions, and voluntary motor behavior. Proper lamination requires that progenitor cells give rise to a neuron, that the immature neuron can migrate away from its mother cell and past other cells, and finally that the immature neuron can take its place and adopt a mature identity characterized by connectivity and gene expression; thus lamination proceeds through three steps: genesis, migration, and maturation. Each neocortical layer contains pyramidal neurons that share specific morphological and molecular characteristics that stem from their prenatal birth date. Transcription factors are dynamic proteins because of the cohort of downstream factors that they regulate. RNA-binding proteins are no less dynamic, and play important roles in every step of mRNA processing. Indeed, recent screens have uncovered post-transcriptional mechanisms as being integral regulatory mechanisms to neocortical development. Here, we summarize major aspects of neocortical laminar development, emphasizing transcriptional and post-transcriptional mechanisms, with the aim of spurring increased understanding and study of its intricacies.
Collapse
Affiliation(s)
- Tatiana Popovitchenko
- Neuroscience and Cell Biology, Robert Wood Johnson Medical School, New Brunswick, NJ, United States
| | - Mladen-Roko Rasin
- Neuroscience and Cell Biology, Robert Wood Johnson Medical School, New Brunswick, NJ, United States
| |
Collapse
|
184
|
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
|
185
|
Pfurr S, Chu YH, Bohrer C, Greulich F, Beattie R, Mammadzada K, Hils M, Arnold SJ, Taylor V, Schachtrup K, Uhlenhaut NH, Schachtrup C. The E2A splice variant E47 regulates the differentiation of projection neurons via p57(KIP2) during cortical development. Development 2017; 144:3917-3931. [PMID: 28939666 DOI: 10.1242/dev.145698] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 09/11/2017] [Indexed: 02/01/2023]
Abstract
During corticogenesis, distinct classes of neurons are born from progenitor cells located in the ventricular and subventricular zones, from where they migrate towards the pial surface to assemble into highly organized layer-specific circuits. However, the precise and coordinated transcriptional network activity defining neuronal identity is still not understood. Here, we show that genetic depletion of the basic helix-loop-helix (bHLH) transcription factor E2A splice variant E47 increased the number of Tbr1-positive deep layer and Satb2-positive upper layer neurons at E14.5, while depletion of the alternatively spliced E12 variant did not affect layer-specific neurogenesis. While ChIP-Seq identified a big overlap for E12- and E47-specific binding sites in embryonic NSCs, including sites at the cyclin-dependent kinase inhibitor (CDKI) Cdkn1c gene locus, RNA-Seq revealed a unique transcriptional regulation by each splice variant. E47 activated the expression of the CDKI Cdkn1c through binding to a distal enhancer. Finally, overexpression of E47 in embryonic NSCs in vitro impaired neurite outgrowth, and overexpression of E47 in vivo by in utero electroporation disturbed proper layer-specific neurogenesis and upregulated p57(KIP2) expression. Overall, this study identifies E2A target genes in embryonic NSCs and demonstrates that E47 regulates neuronal differentiation via p57(KIP2).
Collapse
Affiliation(s)
- Sabrina Pfurr
- Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg 79104, Germany.,Faculty of Biology, University of Freiburg, Freiburg 79104, Germany
| | - Yu-Hsuan Chu
- Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg 79104, Germany.,Faculty of Biology, University of Freiburg, Freiburg 79104, Germany
| | - Christian Bohrer
- Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg 79104, Germany.,Faculty of Biology, University of Freiburg, Freiburg 79104, Germany
| | - Franziska Greulich
- Helmholtz Diabetes Center (HDC) and German Center for Diabetes Research (DZD), Helmholtz Zentrum München, Neuherberg 85764, Germany
| | - Robert Beattie
- Department of Biomedicine, Embryology and Stem Cell Biology, University of Basel, Basel 4058, Switzerland
| | - Könül Mammadzada
- Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg 79104, Germany.,Faculty of Biology, University of Freiburg, Freiburg 79104, Germany
| | - Miriam Hils
- Faculty of Biology, University of Freiburg, Freiburg 79104, Germany.,Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg 79106, Germany
| | - Sebastian J Arnold
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg 79104, Germany.,BIOSS Centre of Biological Signalling Studies, Albert-Ludwigs-University, Freiburg 79104, Germany
| | - Verdon Taylor
- Department of Biomedicine, Embryology and Stem Cell Biology, University of Basel, Basel 4058, Switzerland
| | - Kristina Schachtrup
- Faculty of Biology, University of Freiburg, Freiburg 79104, Germany.,Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg 79106, Germany
| | - N Henriette Uhlenhaut
- Helmholtz Diabetes Center (HDC) and German Center for Diabetes Research (DZD), Helmholtz Zentrum München, Neuherberg 85764, Germany
| | - Christian Schachtrup
- Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg 79104, Germany
| |
Collapse
|
186
|
Clare AJ, Wicky HE, Empson RM, Hughes SM. RNA-Sequencing Analysis Reveals a Regulatory Role for Transcription Factor Fezf2 in the Mature Motor Cortex. Front Mol Neurosci 2017; 10:283. [PMID: 28936162 PMCID: PMC5594072 DOI: 10.3389/fnmol.2017.00283] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 08/22/2017] [Indexed: 12/14/2022] Open
Abstract
Forebrain embryonic zinc finger (Fezf2) encodes a transcription factor essential for the specification of layer 5 projection neurons (PNs) in the developing cerebral cortex. As with many developmental transcription factors, Fezf2 continues to be expressed into adulthood, suggesting it remains crucial to the maintenance of neuronal phenotypes. Despite the continued expression, a function has yet to be explored for Fezf2 in the PNs of the developed cortex. Here, we investigated the role of Fezf2 in mature neurons, using lentiviral-mediated delivery of a shRNA to conditionally knockdown the expression of Fezf2 in the mouse primary motor cortex (M1). RNA-sequencing analysis of Fezf2-reduced M1 revealed significant changes to the transcriptome, identifying a regulatory role for Fezf2 in the mature M1. Kyoto Encyclopedia Genes and Genomes (KEGG) pathway analyses of Fezf2-regulated genes indicated a role in neuronal signaling and plasticity, with significant enrichment of neuroactive ligand-receptor interaction, cell adhesion molecules and calcium signaling pathways. Gene Ontology analysis supported a functional role for Fezf2-regulated genes in neuronal transmission and additionally indicated an importance in the regulation of behavior. Using the mammalian phenotype ontology database, we identified a significant overrepresentation of Fezf2-regulated genes associated with specific behavior phenotypes, including associative learning, social interaction, locomotor activation and hyperactivity. These roles were distinct from that of Fezf2-regulated genes identified in development, indicating a dynamic transition in Fezf2 function. Together our findings demonstrate a regulatory role for Fezf2 in the mature brain, with Fezf2-regulated genes having functional roles in sustaining normal neuronal and behavioral phenotypes. These results support the hypothesis that developmental transcription factors are important for maintaining neuron transcriptomes and that disruption of their expression could contribute to the progression of disease phenotypes.
Collapse
Affiliation(s)
- Alison J Clare
- Department of Biochemistry, School of Biomedical Sciences, University of OtagoDunedin, New Zealand.,Brain Health Research Centre, University of OtagoDunedin, New Zealand.,Genetics Otago, University of OtagoDunedin, New Zealand
| | - Hollie E Wicky
- Department of Biochemistry, School of Biomedical Sciences, University of OtagoDunedin, New Zealand.,Brain Health Research Centre, University of OtagoDunedin, New Zealand.,Genetics Otago, University of OtagoDunedin, New Zealand
| | - Ruth M Empson
- Brain Health Research Centre, University of OtagoDunedin, New Zealand.,Department of Physiology, School of Biomedical Sciences, University of OtagoDunedin, New Zealand
| | - Stephanie M Hughes
- Department of Biochemistry, School of Biomedical Sciences, University of OtagoDunedin, New Zealand.,Brain Health Research Centre, University of OtagoDunedin, New Zealand.,Genetics Otago, University of OtagoDunedin, New Zealand
| |
Collapse
|
187
|
Johnson CA, Wright CE, Ghashghaei HT. Regulation of cytokinesis during corticogenesis: focus on the midbody. FEBS Lett 2017; 591:4009-4026. [PMID: 28493553 DOI: 10.1002/1873-3468.12676] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 04/23/2017] [Accepted: 05/07/2017] [Indexed: 12/21/2022]
Abstract
Development of the cerebral cortices depends on tight regulation of cell divisions. In this system, stem and progenitor cells undergo symmetric and asymmetric divisions to ultimately produce neurons that establish the layers of the cortex. Cell division culminates with the formation of the midbody, a transient organelle that establishes the site of abscission between nascent daughter cells. During cytokinetic abscission, the final stage of cell division, one daughter cell will inherit the midbody remnant, which can then maintain or expel the remnant, but mechanisms and circumstances influencing this decision are unclear. This review describes the midbody and its constituent proteins, as well as the known consequences of their manipulation during cortical development. The potential functional relevance of midbody mechanisms is discussed.
Collapse
Affiliation(s)
- Caroline A Johnson
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA.,Comparative Biomedical Sciences Graduate Program, Neurosciences Concentration Area, North Carolina State University, Raleigh, NC, USA
| | - Catherine E Wright
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA
| | - H Troy Ghashghaei
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA.,Comparative Biomedical Sciences Graduate Program, Neurosciences Concentration Area, North Carolina State University, Raleigh, NC, USA.,Program in Genetics, North Carolina State University, Raleigh, NC, USA.,Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC, USA
| |
Collapse
|
188
|
Kimura E, Kubo KI, Endo T, Ling W, Nakajima K, Kakeyama M, Tohyama C. Impaired dendritic growth and positioning of cortical pyramidal neurons by activation of aryl hydrocarbon receptor signaling in the developing mouse. PLoS One 2017; 12:e0183497. [PMID: 28820910 PMCID: PMC5562321 DOI: 10.1371/journal.pone.0183497] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 08/05/2017] [Indexed: 11/24/2022] Open
Abstract
The basic helix-loop-helix (bHLH) transcription factors exert multiple functions in mammalian cerebral cortex development. The aryl hydrocarbon receptor (AhR), a member of the bHLH-Per-Arnt-Sim subfamily, is a ligand-activated transcription factor reported to regulate nervous system development in both invertebrates and vertebrates, but the functions that AhR signaling pathway may have for mammalian cerebral cortex development remains elusive. Although the endogenous ligand involved in brain developmental process has not been identified, the environmental pollutant dioxin potently binds AhR and induces abnormalities in higher brain function of laboratory animals. Thus, we studied how activation of AhR signaling influences cortical development in mice. To this end, we produced mice expressing either constitutively active-AhR (CA-AhR), which has the capacity for ligand-independent activation of downstream genes, or AhR, which requires its ligands for activation. In brief, CA-AhR-expressing plasmid and AhR-expressing plasmid were each transfected into neural stems cells in the developing cerebrum by in utero electroporation on embryonic day 14.5. On postnatal day 14, mice transfected in utero with CA-AhR, but not those transfected with AhR, exhibited drastically reduced dendritic arborization of layer II/III pyramidal neurons and impaired neuronal positioning in the developing somatosensory cortex. The effects of CA-AhR were observed for dendrite development but not for the commissural fiber projection, suggesting a preferential influence on dendrites. The present results indicate that over-activation of AhR perturbs neuronal migration and morphological development in mammalian cortex, supporting previous observations of impaired dendritic structure, cortical dysgenesis, and behavioral abnormalities following perinatal dioxin exposure.
Collapse
Affiliation(s)
- Eiki Kimura
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Center for Health and Environmental Risk Research, National Institute for Environmental Studies, Tsukuba, Japan
- Research Fellow of Japan Society for the Promotion of Science, Tokyo, Japan
| | - Ken-ichiro Kubo
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| | - Toshihiro Endo
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Wenting Ling
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kazunori Nakajima
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| | - Masaki Kakeyama
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Laboratory for Systems Neuroscience and Preventive Medicine, Faculty of Human Sciences, Waseda University, Tokorozawa, Japan
| | - Chiharu Tohyama
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
- * E-mail:
| |
Collapse
|
189
|
Wiegreffe C, Feldmann S, Gaessler S, Britsch S. Time-lapse Confocal Imaging of Migrating Neurons in Organotypic Slice Culture of Embryonic Mouse Brain Using In Utero Electroporation. J Vis Exp 2017. [PMID: 28784978 DOI: 10.3791/55886] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
In utero electroporation is a rapid and powerful approach to study the process of radial migration in the cerebral cortex of developing mouse embryos. It has helped to describe the different steps of radial migration and characterize the molecular mechanisms controlling this process. To directly and dynamically analyze migrating neurons they have to be traced over time. This protocol describes a workflow that combines in utero electroporation with organotypic slice culture and time-lapse confocal imaging, which allows for a direct examination and dynamic analysis of radially migrating cortical neurons. Furthermore, detailed characterization of migrating neurons, such as migration speed, speed profiles, as well as radial orientation changes, is possible. The method can easily be adapted to perform functional analyses of genes of interest in radially migrating cortical neurons by loss and gain of function as well as rescue experiments. Time-lapse imaging of migrating neurons is a state-of-the-art technique that once established is a potent tool to study the development of the cerebral cortex in mouse models of neuronal migration disorders.
Collapse
Affiliation(s)
| | | | | | - Stefan Britsch
- Institute of Molecular and Cellular Anatomy, Ulm University
| |
Collapse
|
190
|
Azzarelli R, Oleari R, Lettieri A, Andre' V, Cariboni A. In Vitro, Ex Vivo and In Vivo Techniques to Study Neuronal Migration in the Developing Cerebral Cortex. Brain Sci 2017; 7:brainsci7050048. [PMID: 28448448 PMCID: PMC5447930 DOI: 10.3390/brainsci7050048] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 04/21/2017] [Accepted: 04/24/2017] [Indexed: 11/16/2022] Open
Abstract
Neuronal migration is a fundamental biological process that underlies proper brain development and neuronal circuit formation. In the developing cerebral cortex, distinct neuronal populations, producing excitatory, inhibitory and modulatory neurotransmitters, are generated in different germinative areas and migrate along various routes to reach their final positions within the cortex. Different technical approaches and experimental models have been adopted to study the mechanisms regulating neuronal migration in the cortex. In this review, we will discuss the most common in vitro, ex vivo and in vivo techniques to visualize and study cortical neuronal migration.
Collapse
Affiliation(s)
- Roberta Azzarelli
- Department of Oncology, University of Cambridge, Hutchison-MRC Research Centre, Hills Road, Cambridge CB2 0XZ, UK.
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK.
- Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge CB3 0HE, UK.
| | - Roberto Oleari
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti, 9, Milan 20133, Italy.
| | - Antonella Lettieri
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti, 9, Milan 20133, Italy.
| | - Valentina Andre'
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti, 9, Milan 20133, Italy.
| | - Anna Cariboni
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti, 9, Milan 20133, Italy.
- Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK.
| |
Collapse
|
191
|
Yang Z, Li PF, Chen RC, Wang J, Wang S, Shen Y, Wu X, Fang B, Cheng X, Xiong ZQ. ADAM10-Initiated Release of Notch Intracellular Domain Regulates Microtubule Stability and Radial Migration of Cortical Neurons. Cereb Cortex 2017; 27:919-932. [PMID: 28158408 PMCID: PMC6093323 DOI: 10.1093/cercor/bhx006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2016] [Indexed: 11/24/2022] Open
Abstract
Proper neuronal migration is orchestrated by combined membrane signal paradigms, whereas the role and mechanism of regulated intramembrane proteolysis (RIP) remain to be illustrated. We show here that the disintegrin and metalloprotease-domain containing protein 10 (ADAM10) regulates cortical neurons migration by initiating the RIP of Notch. We found that Notch intracellular domain (NICD) significantly rescued the migration defect of ADAM10-deficient neurons. Moreover, ADAM10 deficiency led to reduced neuronal motility and disrupted microtubule (MT) structure, which were associated with downregulated expression of acetylated tubulin and MT-associated proteins. Specifically, the NICD/RBPJ complex bound directly to the promoter, and regulated the neuronal expression level of doublecortin (DCX), a modulator of the MT cytoskeleton. Functionally, DCX overexpression largely restored neuron motility and reversed migration defect caused by ADAM10 knockout. Taken together, these findings demonstrate the direct requirement of ADAM10 in cortical radial migration and reveal the underlying mechanism by linking ADAM10-initiated RIP of Notch to the regulation of MT cytoskeleton through transcriptional control of Dcx expression.
Collapse
Affiliation(s)
- Zhi Yang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China.,Shanghai Key Laboratory of Stomatology, Department of Oral and Cranio-Maxillofacial Science, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200011, China
| | - Peng-Fei Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ren-Chao Chen
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jie Wang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shaoran Wang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ya Shen
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiaohui Wu
- Institute of Developmental Biology and Molecular Medicine, School of Life Science, Fudan University, Shanghai 200433, China
| | - Bing Fang
- Shanghai Key Laboratory of Stomatology, Department of Oral and Cranio-Maxillofacial Science, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200011, China
| | - Xuewen Cheng
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhi-Qi Xiong
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
192
|
Teoh JJ, Iwano T, Kunii M, Atik N, Avriyanti E, Yoshimura SI, Moriwaki K, Harada A. BIG1 is required for the survival of deep layer neurons, neuronal polarity, and the formation of axonal tracts between the thalamus and neocortex in developing brain. PLoS One 2017; 12:e0175888. [PMID: 28414797 PMCID: PMC5393877 DOI: 10.1371/journal.pone.0175888] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 04/02/2017] [Indexed: 12/17/2022] Open
Abstract
BIG1, an activator protein of the small GTPase, Arf, and encoded by the Arfgef1 gene, is one of candidate genes for epileptic encephalopathy. To know the involvement of BIG1 in epileptic encephalopathy, we analyzed BIG1-deficient mice and found that BIG1 regulates neurite outgrowth and brain development in vitro and in vivo. The loss of BIG1 decreased the size of the neocortex and hippocampus. In BIG1-deficient mice, the neuronal progenitor cells (NPCs) and the interneurons were unaffected. However, Tbr1+ and Ctip2+ deep layer (DL) neurons showed spatial-temporal dependent apoptosis. This apoptosis gradually progressed from the piriform cortex (PIR), peaked in the neocortex, and then progressed into the hippocampus from embryonic day 13.5 (E13.5) to E17.5. The upper layer (UL) and DL order in the neocortex was maintained in BIG1-deficient mice, but the excitatory neurons tended to accumulate before their destination layers. Further pulse-chase migration assay showed that the migration defect was non-cell autonomous and secondary to the progression of apoptosis into the BIG1-deficient neocortex after E15.5. In BIG1-deficient mice, we observed an ectopic projection of corticothalamic axons from the primary somatosensory cortex (S1) into the dorsal lateral geniculate nucleus (dLGN). The thalamocortical axons were unable to cross the diencephalon-telencephalon boundary (DTB). In vitro, BIG1-deficient neurons showed a delay in neuronal polarization. BIG1-deficient neurons were also hypersensitive to low dose glutamate (5 μM), and died via apoptosis. This study showed the role of BIG1 in the survival of DL neurons in developing embryonic brain and in the generation of neuronal polarity.
Collapse
Affiliation(s)
- Jia-Jie Teoh
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Tomohiko Iwano
- Department of Anatomy and Cell Biology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Masataka Kunii
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Nur Atik
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Department of Anatomy and Cell Biology, Faculty of Medicine, Padjadjaran University, Bandung, Indonesia
| | - Erda Avriyanti
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Department of Dermatology and Venereology, Faculty of Medicine, Padjadjaran University, Bandung, Indonesia
| | - Shin-ichiro Yoshimura
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Kenta Moriwaki
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Akihiro Harada
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
193
|
Ascl1 promotes tangential migration and confines migratory routes by induction of Ephb2 in the telencephalon. Sci Rep 2017; 7:42895. [PMID: 28276447 PMCID: PMC5343589 DOI: 10.1038/srep42895] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 01/18/2017] [Indexed: 01/13/2023] Open
Abstract
During development, cortical interneurons generated from the ventral telencephalon migrate tangentially into the dorsal telencephalon. Although Achaete-scute family bHLH transcription factor 1 (Ascl1) plays important roles in the developing telencephalon, whether Ascl1 regulates tangential migration remains unclear. Here, we found that Ascl1 promoted tangential migration along the ventricular zone/subventricular zone (VZ/SVZ) and intermediate zone (IZ) of the dorsal telencephalon. Distal-less homeobox 2 (Dlx2) acted downstream of Ascl1 in promoting tangential migration along the VZ/SVZ but not IZ. We further identified Eph receptor B2 (Ephb2) as a direct target of Ascl1. Knockdown of EphB2 disrupted the separation of the VZ/SVZ and IZ migratory routes. Ephrin-A5, a ligand of EphB2, was sufficient to repel both Ascl1-expressing cells in vitro and tangentially migrating cortical interneurons in vivo. Together, our results demonstrate that Ascl1 induces expression of Dlx2 and Ephb2 to maintain distinct tangential migratory routes in the dorsal telencephalon.
Collapse
|
194
|
NEUROD2 Regulates Stim1 Expression and Store-Operated Calcium Entry in Cortical Neurons. eNeuro 2017; 4:eN-NWR-0255-16. [PMID: 28303257 PMCID: PMC5343279 DOI: 10.1523/eneuro.0255-16.2017] [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: 08/28/2016] [Revised: 01/18/2017] [Accepted: 01/25/2017] [Indexed: 12/26/2022] Open
Abstract
Calcium signaling controls many key processes in neurons, including gene expression, axon guidance, and synaptic plasticity. In contrast to calcium influx through voltage- or neurotransmitter-gated channels, regulatory pathways that control store-operated calcium entry (SOCE) in neurons are poorly understood. Here, we report a transcriptional control of Stim1 (stromal interaction molecule 1) gene, which is a major sensor of endoplasmic reticulum (ER) calcium levels and a regulator of SOCE. By using a genome-wide chromatin immunoprecipitation and sequencing approach in mice, we find that NEUROD2, a neurogenic transcription factor, binds to an intronic element within the Stim1 gene. We show that NEUROD2 limits Stim1 expression in cortical neurons and consequently fine-tunes the SOCE response upon depletion of ER calcium. Our findings reveal a novel mechanism that regulates neuronal calcium homeostasis during cortical development.
Collapse
|
195
|
Neonatal CX26 removal impairs neocortical development and leads to elevated anxiety. Proc Natl Acad Sci U S A 2017; 114:3228-3233. [PMID: 28265099 DOI: 10.1073/pnas.1613237114] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Electrical coupling between excitatory neurons in the neocortex is developmentally regulated. It is initially prominent but eliminated at later developmental stages when chemical synapses emerge. However, it remains largely unclear whether early electrical coupling networks broadly contribute to neocortical circuit formation and animal behavior. Here, we report that neonatal electrical coupling between neocortical excitatory neurons is critical for proper neuronal development, synapse formation, and animal behavior. Conditional deletion of Connexin 26 (CX26) in the superficial layer excitatory neurons of the mouse neocortex around birth significantly reduces spontaneous firing activity and the frequency and size of spontaneous network oscillations at postnatal day 5-6. Moreover, CX26-conditional knockout (CX26-cKO) neurons tend to have simpler dendritic trees and lower spine density compared with wild-type neurons. Importantly, early, but not late, postnatal deletion of CX26, decreases the frequency of miniature excitatory postsynaptic currents (mEPSCs) in both young and adult mice, whereas miniature inhibitory postsynaptic currents (mIPSCs) were unaffected. Furthermore, CX26-cKO mice exhibit increased anxiety-related behavior. These results suggest that electrical coupling between excitatory neurons at early postnatal stages is a critical step for neocortical development and function.
Collapse
|
196
|
Ma H, Yu H, Li T, Zhao Y, Hou M, Chen Z, Wang Y, Sun T. JIP3 regulates neuronal radial migration by mediating TrkB axonal anterograde transport in the developing cerebral cortex. Biochem Biophys Res Commun 2017; 485:790-795. [PMID: 28259553 DOI: 10.1016/j.bbrc.2017.02.132] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Accepted: 02/27/2017] [Indexed: 11/27/2022]
Abstract
Radial migration is essential for the precise lamination and the coordinated function of the cerebral cortex. However, the molecular mechanisms for neuronal radial migration are not clear. Here, we report that c-Jun NH2-terminal kinase (JNK)-interacting protein-3 (JIP3) is highly expressed in the brain of embryonic mice and essential for radial migration. Knocking down JIP3 by in utero electroporation specifically perturbs the radial migration of cortical neurons but has no effect on neurogenesis and neuronal differentiation. Furthermore, we illustrate that JIP3 knockdown delays but does not block the migration of cortical neurons by investigating the distribution of neurons with JIP3 knocked down in the embryo and postnatal mouse. Finally, we find that JIP3 regulates cortical neuronal migration by mediating TrkB axonal anterograde transport during brain development. These findings deepen our understanding of the regulation of neuronal development by JIP3 and provide us a novel view on the regulating mechanisms of neuronal radial migration.
Collapse
Affiliation(s)
- Huixian Ma
- Department of Neurobiology, Shandong Provincial Key Laboratory of Mental Disorders, School of Medicine and the Collaborative Innovation Center for Brain Science, Shandong University, No.44 Wenhua Xi Road, Jinan, Shandong 250012, PR China
| | - Hui Yu
- Department of Neurobiology, Shandong Provincial Key Laboratory of Mental Disorders, School of Medicine and the Collaborative Innovation Center for Brain Science, Shandong University, No.44 Wenhua Xi Road, Jinan, Shandong 250012, PR China
| | - Ting Li
- Department of Neurobiology, Shandong Provincial Key Laboratory of Mental Disorders, School of Medicine and the Collaborative Innovation Center for Brain Science, Shandong University, No.44 Wenhua Xi Road, Jinan, Shandong 250012, PR China
| | - Yan Zhao
- Shandong Provincial Key Laboratory of Immunohematology, Qilu Hospital, Shandong University, Jinan, Shandong 250012, PR China
| | - Ming Hou
- Shandong Provincial Key Laboratory of Immunohematology, Qilu Hospital, Shandong University, Jinan, Shandong 250012, PR China
| | - Zheyu Chen
- Department of Neurobiology, Shandong Provincial Key Laboratory of Mental Disorders, School of Medicine and the Collaborative Innovation Center for Brain Science, Shandong University, No.44 Wenhua Xi Road, Jinan, Shandong 250012, PR China
| | - Yue Wang
- Department of Neurobiology, Shandong Provincial Key Laboratory of Mental Disorders, School of Medicine and the Collaborative Innovation Center for Brain Science, Shandong University, No.44 Wenhua Xi Road, Jinan, Shandong 250012, PR China.
| | - Tao Sun
- Department of Neurobiology, Shandong Provincial Key Laboratory of Mental Disorders, School of Medicine and the Collaborative Innovation Center for Brain Science, Shandong University, No.44 Wenhua Xi Road, Jinan, Shandong 250012, PR China; Shandong Provincial Key Laboratory of Immunohematology, Qilu Hospital, Shandong University, Jinan, Shandong 250012, PR China.
| |
Collapse
|
197
|
Bagshaw ATM, Horwood LJ, Fergusson DM, Gemmell NJ, Kennedy MA. Microsatellite polymorphisms associated with human behavioural and psychological phenotypes including a gene-environment interaction. BMC MEDICAL GENETICS 2017; 18:12. [PMID: 28158988 PMCID: PMC5291968 DOI: 10.1186/s12881-017-0374-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 01/25/2017] [Indexed: 02/05/2023]
Abstract
Background The genetic and environmental influences on human personality and behaviour are a complex matter of ongoing debate. Accumulating evidence indicates that short tandem repeats (STRs) in regulatory regions are good candidates to explain heritability not accessed by genome-wide association studies. Methods We tested for associations between the genotypes of four selected repeats and 18 traits relating to personality, behaviour, cognitive ability and mental health in a well-studied longitudinal birth cohort (n = 458-589) using one way analysis of variance. The repeats were a highly conserved poly-AC microsatellite in the upstream promoter region of the T-box brain 1 (TBR1) gene and three previously studied STRs in the activating enhancer-binding protein 2-beta (AP2-β) and androgen receptor (AR) genes. Where significance was found we used multiple regression to assess the influence of confounding factors. Results Carriers of the shorter, most common, allele of the AR gene’s GGN microsatellite polymorphism had fewer anxiety-related symptoms, which was consistent with previous studies, but in our study this was not significant following Bonferroni correction. No associations with two repeats in the AP2-β gene withstood this correction. A novel finding was that carriers of the minor allele of the TBR1 AC microsatellite were at higher risk of conduct problems in childhood at age 7-9 (p = 0.0007, which did pass Bonferroni correction). Including maternal smoking during pregnancy (MSDP) in models controlling for potentially confounding influences showed that an interaction between TBR1 genotype and MSDP was a significant predictor of conduct problems in childhood and adolescence (p < 0.001), and of self-reported criminal behaviour up to age 25 years (p ≤ 0.02). This interaction remained significant after controlling for possible confounders including maternal age at birth, socio-economic status and education, and offspring birth weight. Conclusions The potential functional importance of the TBR1 gene’s promoter microsatellite deserves further investigation. Our results suggest that it participates in a gene-environment interaction with MDSP and antisocial behaviour. However, previous evidence that mothers who smoke during pregnancy carry genes for antisocial behaviour suggests that epistasis may influence the interaction. Electronic supplementary material The online version of this article (doi:10.1186/s12881-017-0374-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Andrew T M Bagshaw
- Department of Pathology, University of Otago, Christchurch, PO Box 4345, Christchurch, New Zealand.
| | - L John Horwood
- Department of Psychological Medicine, University of Otago, Christchurch, New Zealand
| | - David M Fergusson
- Department of Psychological Medicine, University of Otago, Christchurch, New Zealand
| | - Neil J Gemmell
- Department of Anatomy, University of Otago, Dunedin, New Zealand.,Gravida - National Centre for Growth and Development, University of Otago, Dunedin, New Zealand
| | - Martin A Kennedy
- Department of Pathology, University of Otago, Christchurch, PO Box 4345, Christchurch, New Zealand
| |
Collapse
|
198
|
Caubit X, Gubellini P, Andrieux J, Roubertoux PL, Metwaly M, Jacq B, Fatmi A, Had-Aissouni L, Kwan KY, Salin P, Carlier M, Liedén A, Rudd E, Shinawi M, Vincent-Delorme C, Cuisset JM, Lemaitre MP, Abderrehamane F, Duban B, Lemaitre JF, Woolf AS, Bockenhauer D, Severac D, Dubois E, Zhu Y, Sestan N, Garratt AN, Lydia Kerkerian-Le G, Fasano L. TSHZ3 deletion causes an autism syndrome and defects in cortical projection neurons. Nat Genet 2016; 48:1359-1369. [PMID: 27668656 PMCID: PMC5083212 DOI: 10.1038/ng.3681] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 08/29/2016] [Indexed: 12/12/2022]
Abstract
TSHZ3, which encodes a zinc-finger transcription factor, was recently positioned as a hub gene in a module of the genes with the highest expression in the developing human neocortex, but its functions remained unknown. Here we identify TSHZ3 as the critical region for a syndrome associated with heterozygous deletions at 19q12-q13.11, which includes autism spectrum disorder (ASD). In Tshz3-null mice, differentially expressed genes include layer-specific markers of cerebral cortical projection neurons (CPNs), and the human orthologs of these genes are strongly associated with ASD. Furthermore, mice heterozygous for Tshz3 show functional changes at synapses established by CPNs and exhibit core ASD-like behavioral abnormalities. These findings highlight essential roles for Tshz3 in CPN development and function, whose alterations can account for ASD in the newly defined TSHZ3 deletion syndrome.
Collapse
Affiliation(s)
| | | | - Joris Andrieux
- Institut de génétique médicale, Hôpital Jeanne de Flandre, CHRU Lille, France
| | | | | | - Bernard Jacq
- Aix Marseille Univ, CNRS, IBDM, Marseille, France
| | - Ahmed Fatmi
- Aix Marseille Univ, CNRS, IBDM, Marseille, France
| | | | - Kenneth Y Kwan
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
- Molecular & Behavioral Neuroscience Institute (MBNI), Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Pascal Salin
- Aix Marseille Univ, CNRS, IBDM, Marseille, France
| | | | - Agne Liedén
- Karolinska University Hospital Solna, Clinical Genetics Unit, Stockholm, Sweden
| | - Eva Rudd
- Karolinska University Hospital Solna, Clinical Genetics Unit, Stockholm, Sweden
| | - Marwan Shinawi
- Department of Pediatrics, Division of Genetics and Genomic Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | | | | | | | | | - Bénédicte Duban
- Centre de cytogénétique, Hôpital Saint Vincent de Paul, GHICL, UCL, Lille, France
| | | | - Adrian S Woolf
- Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, Manchester Academic Health Science Centre and the Royal Manchester Children's and St Mary's Hospitals, Manchester, UK
| | | | - Dany Severac
- MGX-Montpellier GenomiX, c/o Institut de Génomique Fonctionnelle, Montpellier, France
| | - Emeric Dubois
- MGX-Montpellier GenomiX, c/o Institut de Génomique Fonctionnelle, Montpellier, France
| | - Ying Zhu
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Nenad Sestan
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Alistair N Garratt
- Institute of Cell Biology and Neurobiology, Center for Anatomy, Charité University Hospital Berlin, Berlin, Germany
| | | | | |
Collapse
|
199
|
Yuryev M, Ferreira MPA, Balasubramanian V, Correia AMR, Mäkilä EM, Jokinen V, Andriichuk L, Kemell M, Salonen JJ, Hirvonen JT, Santos HA, Rivera C. Active diffusion of nanoparticles of maternal origin within the embryonic brain. Nanomedicine (Lond) 2016; 11:2471-81. [DOI: 10.2217/nnm-2016-0207] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Aim: To investigate porous silicon (PSi) nanoparticles (NPs) behavior in the embryonic brain. Materials & methods: Fluorescently labeled PSi NPs were injected into the embryonic brains intraventricularly and to the mother intravenously (iv.). Brain histology from different time points up to 3 days was analyzed and live brains imaged with two-photon microscopy. Results: PSi NPs were able to penetrate 80% of the embryonic cortical depth. Particle motility was confirmed in real-time in vivo. PSi NPs were able to penetrate the embryonic cortex after either iv. maternal or intraventricular injection. No developmental of macromorphological changes or increased cell apoptosis was observed. Conclusion: PSi NPs penetrate deep in the brain tissues of embryos after intraventricular injection and after iv. injection to the mother.
Collapse
Affiliation(s)
- Mikhail Yuryev
- Neuroscience Center, University of Helsinki, FI-00014 Helsinki, Finland
| | - Mónica PA Ferreira
- Division of Pharmaceutical Chemistry & Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Vimalkumar Balasubramanian
- Division of Pharmaceutical Chemistry & Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Alexandra MR Correia
- Division of Pharmaceutical Chemistry & Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Ermei M Mäkilä
- Laboratory of Industrial Physics, Department of Physics & Astronomy, University of Turku, FI-20014 Turku, Finland
| | - Ville Jokinen
- Aalto University, School of Chemical Technology, FI-02150 Espoo, Finland
| | - Liliia Andriichuk
- Neuroscience Center, University of Helsinki, FI-00014 Helsinki, Finland
| | - Marianna Kemell
- Department of Chemistry, University of Helsinki, PO Box 55, FI-00014 Helsinki, Finland
| | - Jarno J Salonen
- Laboratory of Industrial Physics, Department of Physics & Astronomy, University of Turku, FI-20014 Turku, Finland
| | - Jouni T Hirvonen
- Division of Pharmaceutical Chemistry & Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Hélder A Santos
- Division of Pharmaceutical Chemistry & Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Claudio Rivera
- Neuroscience Center, University of Helsinki, FI-00014 Helsinki, Finland
- Institut de Neurobiologie de la Méditerranée, Institut National de la Santé et de la Recherche Médicale Unité 901, 13009 Marseille, France Aix-Marseille Université, Unité Mixte de Recherche 901, 13273 Marseille, France
| |
Collapse
|
200
|
Chao MW, Chen CP, Yang YH, Chuang YC, Chu TY, Tseng CY. N-acetylcysteine attenuates lipopolysaccharide-induced impairment in lamination of Ctip2-and Tbr1- expressing cortical neurons in the developing rat fetal brain. Sci Rep 2016; 6:32373. [PMID: 27577752 PMCID: PMC5006028 DOI: 10.1038/srep32373] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 08/03/2016] [Indexed: 02/02/2023] Open
Abstract
Oxidative stress and inflammatory insults are the major instigating events of bacterial intrauterine infection that lead to fetal brain injury. The purpose of this study is to investigate the remedial effects of N-acetyl-cysteine (NAC) for inflammation-caused deficits in brain development. We found that lipopolysaccharide (LPS) induced reactive oxygen species (ROS) production by RAW264.7 cells. Macrophage-conditioned medium caused noticeable cortical cell damage, specifically in cortical neurons. LPS at 25 μg/kg caused more than 75% fetal loss in rats. An increase in fetal cortical thickness was noted in the LPS-treated group. In the enlarged fetal cortex, laminar positioning of the early born cortical cells expressing Tbr1 and Ctip2 was disrupted, with a scattered distribution. The effect was similar, but minor, in later born Satb2-expressing cortical cells. NAC protected against LPS-induced neuron toxicity in vitro and counteracted pregnancy loss and alterations in thickness and lamination of the neocortex in vivo. Fetal loss and abnormal fetal brain development were due to LPS-induced ROS production. NAC is an effective protective agent against LPS-induced damage. This finding highlights the key therapeutic impact of NAC in LPS-caused abnormal neuronal laminar distribution during brain development.
Collapse
Affiliation(s)
- Ming-Wei Chao
- Department of Bioscience Technology, Chung Yuan Christian University, Zhongli district, Taoyuan City, Taiwan
| | - Chie-Pein Chen
- Division of High Risk Pregnancy, Mackay Memorial Hospital, Taipei, Taiwan
| | - Yu-Hsiu Yang
- Department of Biomedical Engineering, Chung Yuan Christian University, Zhongli district, Taoyuan City, Taiwan
| | - Yu-Chen Chuang
- Department of Biomedical Engineering, Chung Yuan Christian University, Zhongli district, Taoyuan City, Taiwan
| | - Tzu-Yun Chu
- Division of High Risk Pregnancy, Mackay Memorial Hospital, Taipei, Taiwan
| | - Chia-Yi Tseng
- Department of Biomedical Engineering, Chung Yuan Christian University, Zhongli district, Taoyuan City, Taiwan
- International Master Program of Biomedical Material and Technology, Chung Yuan Christian University, Zhongli district, Taoyuan City, Taiwan
- Center for Nano-Technology, Chung Yuan Christian University, Zhongli district, Taoyuan City, Taiwan
| |
Collapse
|