1
|
Tai NC, Shinmyo Y, Kawasaki H. Astrocyte diversity in the ferret cerebrum revealed with astrocyte-specific genetic manipulation. Glia 2024; 72:1862-1873. [PMID: 38884631 DOI: 10.1002/glia.24587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 06/08/2024] [Accepted: 06/12/2024] [Indexed: 06/18/2024]
Abstract
Astrocytes in the cerebrum play important roles such as the regulation of synaptic functions, homeostasis, water transport, and the blood-brain barrier. It has been proposed that astrocytes in the cerebrum acquired diversity and developed functionally during evolution. Here, we show that like human astrocytes, ferret astrocytes in the cerebrum exhibit various morphological subtypes which mice do not have. We found that layer 1 of the ferret cerebrum contained not only protoplasmic astrocytes but also pial interlaminar astrocytes and subpial interlaminar astrocytes. Morphologically polarized astrocytes, which have a long unbranched process, were found in layer 6. Like human white matter, ferret white matter exhibited four subtypes of astrocytes. Furthermore, our quantification showed that ferret astrocytes had a larger territory size and a longer radius length than mouse astrocytes. Thus, our results indicate that, similar to the human cerebrum, the ferret cerebrum has a well-developed diversity of astrocytes. Ferrets should be useful for investigating the molecular and cellular mechanisms leading to astrocyte diversity, the functions of each astrocyte subtype and the involvement of different astrocyte subtypes in various neurological diseases.
Collapse
Affiliation(s)
- Nguyen Chi Tai
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
- Sapiens Life Sciences, Evolution and Medicine Research Center, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Yohei Shinmyo
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
- Department of Neurophysiology, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Hiroshi Kawasaki
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
- Sapiens Life Sciences, Evolution and Medicine Research Center, Kanazawa University, Kanazawa, Ishikawa, Japan
| |
Collapse
|
2
|
Cossard A, Stam K, Smets A, Jossin Y. MKL/SRF and Bcl6 mutual transcriptional repression safeguards the fate and positioning of neocortical progenitor cells mediated by RhoA. SCIENCE ADVANCES 2023; 9:eadd0676. [PMID: 37967194 PMCID: PMC10651131 DOI: 10.1126/sciadv.add0676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 10/16/2023] [Indexed: 11/17/2023]
Abstract
During embryogenesis, multiple intricate and intertwined cellular signaling pathways coordinate cell behavior. Their slightest alterations can have dramatic consequences for the cells and the organs they form. The transcriptional repressor Bcl6 was recently found as important for brain development. However, its regulation and integration with other signals is unknown. Using in vivo functional approaches combined with molecular mechanistic analysis, we identified a reciprocal regulatory loop between B cell lymphoma 6 (Bcl6) and the RhoA-regulated transcriptional complex megakaryoblastic leukemia/serum response factor (MKL/SRF). We show that Bcl6 physically interacts with MKL/SRF, resulting in a down-regulation of the transcriptional activity of both Bcl6 and MKL/SRF. This molecular cross-talk is essential for the control of proliferation, neurogenesis, and spatial positioning of neural progenitors. Overall, our data highlight a regulatory mechanism that controls neuronal production and neocortical development and reveal an MKL/SRF and Bcl6 interaction that may have broader implications in other physiological functions and in diseases.
Collapse
Affiliation(s)
- Alexia Cossard
- Laboratory of Mammalian Development and Cell Biology, Institute of Neuroscience, Université Catholique de Louvain, Brussels 1200, Belgium
| | | | | | | |
Collapse
|
3
|
Jaylet T, Quintens R, Armant O, Audouze K. An integrative systems biology strategy to support the development of adverse outcome pathways (AOPs): a case study on radiation-induced microcephaly. Front Cell Dev Biol 2023; 11:1197204. [PMID: 37427375 PMCID: PMC10323360 DOI: 10.3389/fcell.2023.1197204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 06/15/2023] [Indexed: 07/11/2023] Open
Abstract
Adverse Outcome Pathways (AOPs) are useful tools for assessing the potential risks associated with exposure to various stressors, including chemicals and environmental contaminants. They provide a framework for understanding the causal relationships between different biological events that can lead to adverse outcomes (AO). However, developing an AOP is a challenging task, particularly in identifying the molecular initiating events (MIEs) and key events (KEs) that constitute it. Here, we propose a systems biology strategy that can assist in the development of AOPs by screening publicly available databases, literature with the text mining tool AOP-helpFinder, and pathway/network analyses. This approach is straightforward to use, requiring only the name of the stressor and adverse outcome to be studied. From this, it quickly identifies potential KEs and literature providing mechanistic information on the links between the KEs. The proposed approach was applied to the recently developed AOP 441 on radiation-induced microcephaly, resulting in the confirmation of the KEs that were already present and identification of new relevant KEs, thereby validating the strategy. In conclusion, our systems biology approach represents a valuable tool to simplify the development and enrichment of Adverse Outcome Pathways (AOPs), thus supporting alternative methods in toxicology.
Collapse
Affiliation(s)
| | - Roel Quintens
- Belgian Nuclear Research Centre, SCK CEN, Mol, Belgium
| | - Olivier Armant
- PSE-ENV/SRTE/LECO, Institut de Radioprotection et de Sûreté Nucléaire (IRSN), Saint-Paul-Lez-Durance, France
| | | |
Collapse
|
4
|
Aberrant Cortical Layer Development of Brain Organoids Derived from Noonan Syndrome-iPSCs. Int J Mol Sci 2022; 23:ijms232213861. [PMID: 36430334 PMCID: PMC9699065 DOI: 10.3390/ijms232213861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/01/2022] [Accepted: 11/09/2022] [Indexed: 11/12/2022] Open
Abstract
Noonan syndrome (NS) is a genetic disorder mainly caused by gain-of-function mutations in Src homology region 2-containing protein tyrosine phosphatase 2 (SHP2). Although diverse neurological manifestations are commonly diagnosed in NS patients, the mechanisms as to how SHP2 mutations induce the neurodevelopmental defects associated with NS remain elusive. Here, we report that cortical organoids (NS-COs) derived from NS-induced pluripotent stem cells (iPSCs) exhibit developmental abnormalities, especially in excitatory neurons (ENs). Although NS-COs develop normally in their appearance, single-cell transcriptomic analysis revealed an increase in the EN population and overexpression of cortical layer markers in NS-COs. Surprisingly, the EN subpopulation co-expressing the upper layer marker SATB2 and the deep layer maker CTIP2 was enriched in NS-COs during cortical development. In parallel with the developmental disruptions, NS-COs also exhibited reduced synaptic connectivity. Collectively, our findings suggest that perturbed cortical layer identity and impeded neuronal connectivity contribute to the neurological manifestations of NS.
Collapse
|
5
|
KIF5C deficiency causes abnormal cortical neuronal migration, dendritic branching, and spine morphology in mice. Pediatr Res 2022; 92:995-1002. [PMID: 34966180 DOI: 10.1038/s41390-021-01922-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 11/18/2021] [Accepted: 12/13/2021] [Indexed: 11/08/2022]
Abstract
BACKGROUND Malformation of cortical development (MCD) includes a variety of developmental disorders that are common causes of neurodevelopmental delay and epilepsy. Most recently, clinical studies found that patients carrying KIF5C mutations present early-onset MCD; however, the underlying mechanisms remain elusive. METHODS KIF5C expression level was examined in mouse primary cortical neurons and human ips-derived forebrain organoids. We studied the cortical neuronal migration, dendritic branching, and dendritic spine growth after knocking down the KIF5C gene by electroporation in vitro and in vivo. Then, we studied the transcriptome differences between the knockdown and control groups through RNA sequencing. RESULTS We observed high KIF5C expression in neurons during the early developmental stage in mice and the human brain. Kif5c deficiency results in disturbed cortical neuronal migration, dendritic, and spine growth. Finally, we found that Kif5c knockdown affected several genes associated with cortical neuronal development in vitro. CONCLUSIONS These results suggested a critical role for Kif5c in cortical development, providing insights into underlying pathogenic factors of kinesins in MCD. IMPACT KIF5C mutation-related MCD might be caused by abnormal early cortical neuronal development. Kif5c deficiency led to abnormal cortical neuronal dendritic and spine growth and neuronal migration. Our findings explain how Kif5c deficiency is involved in the aberrant development of cortical neurons and provide a new perspective for the pathology of MCD.
Collapse
|
6
|
Shafique S. Stem cell-based region-specific brain organoids: Novel models to understand neurodevelopmental defects. Birth Defects Res 2022; 114:1003-1013. [PMID: 35332709 DOI: 10.1002/bdr2.2004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 03/07/2022] [Accepted: 03/11/2022] [Indexed: 11/08/2022]
Abstract
The study of human brain development and neurodevelopmental defects has remained challenging so far due to unique, specific, and complex underlying processes. Recent advances in the technologies and protocols of in vitro human brain organoid development have led to immense possibilities of understanding these processes. Human brain organoids are stem-cell derived three-dimensional in vitro tissues that resemble the developing fetal brain. Major advances in stem cell techniques pioneering the development of in vitro human brain development include reprogramming human somatic cells into induced pluripotent cells (iPSCs) followed by the targeted differentiation of iPSCs into the cells of three embryonic germ cell layers. The neural progenitor cells produced by the directed differentiation of iPSCs undergo some level of self-organization to generate in vitro human brain like tissue. A three-dimensional differentiation approach applied to create region-specific brain organoids has successfully led to develop highly specialized cortical, forebrain, pallium, and subpallium in vitro human brain organoid models. These stem cell-based brain organoids are novel models to study human brain development, neurodevelopmental defects, chemical toxicity testing, and drug repurposing screening. This review focuses on the fundamentals of brain organoid development and applications. The novel applications of using cortical organoids in understanding the mechanisms of Zika virus-induced microcephaly, congenital microcephaly, and lissencephaly are also discussed.
Collapse
Affiliation(s)
- Sidra Shafique
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
7
|
Shinmyo Y, Saito K, Hamabe-Horiike T, Kameya N, Ando A, Kawasaki K, Duong TAD, Sakashita M, Roboon J, Hattori T, Kannon T, Hosomichi K, Slezak M, Holt MG, Tajima A, Hori O, Kawasaki H. Localized astrogenesis regulates gyrification of the cerebral cortex. SCIENCE ADVANCES 2022; 8:eabi5209. [PMID: 35275722 PMCID: PMC8916738 DOI: 10.1126/sciadv.abi5209] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 01/19/2022] [Indexed: 06/14/2023]
Abstract
The development and evolution of mammalian higher cognition are represented by gyrification of the laminar cerebral cortex and astrocyte development, but their mechanisms and interrelationships remain unknown. Here, we show that localized astrogenesis plays an important role in gyri formation in the gyrencephalic cerebral cortex. In functional genetic experiments, we show that reducing astrocyte number prevents gyri formation in the ferret cortex, while increasing astrocyte number in mice, which do not have cortical folds, can induce gyrus-like protrusions. Morphometric analyses demonstrate that the vertical expansion of deep pallial regions achieved by localized astrogenesis is crucial for gyri formation. Furthermore, our findings suggest that localized astrogenesis by a positive feedback loop of FGF signaling is an important mechanism underlying cortical folding in gyrencephalic mammalian brains. Our findings reveal both the cellular mechanisms and the mechanical principle of gyrification in the mammalian brain.
Collapse
Affiliation(s)
- Yohei Shinmyo
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8640, Japan
| | - Kengo Saito
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8640, Japan
| | - Toshihide Hamabe-Horiike
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8640, Japan
| | - Narufumi Kameya
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8640, Japan
| | - Akitaka Ando
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8640, Japan
| | - Kanji Kawasaki
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8640, Japan
| | - Tung Anh Dinh Duong
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8640, Japan
| | - Masataka Sakashita
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8640, Japan
| | - Jureepon Roboon
- Department of Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8640, Japan
| | - Tsuyoshi Hattori
- Department of Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8640, Japan
| | - Takayuki Kannon
- Department of Bioinformatics and Genomics, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, Ishikawa 920-8640, Japan
| | - Kazuyoshi Hosomichi
- Department of Bioinformatics and Genomics, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, Ishikawa 920-8640, Japan
| | - Michal Slezak
- VIB Center for Brain and Disease Research, Herestraat 49, Leuven 3000, Belgium
- Łukasiewicz Research Network-PORT Polish Institute for Technology Development, 54-066 Wroclaw, Poland
| | - Matthew G. Holt
- VIB Center for Brain and Disease Research, Herestraat 49, Leuven 3000, Belgium
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal
| | - Atsushi Tajima
- Department of Bioinformatics and Genomics, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, Ishikawa 920-8640, Japan
| | - Osamu Hori
- Department of Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8640, Japan
| | - Hiroshi Kawasaki
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8640, Japan
| |
Collapse
|
8
|
Upadhyay J, Tiwari N, Ansari MN. Cerebral palsy: Aetiology, pathophysiology and therapeutic interventions. Clin Exp Pharmacol Physiol 2020; 47:1891-1901. [PMID: 32662125 DOI: 10.1111/1440-1681.13379] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 05/08/2020] [Accepted: 07/09/2020] [Indexed: 11/26/2022]
Abstract
Cerebral palsy (CP) is the most common non-progressive neurodevelopmental disorder in which the impairment of motor and posture functions occurs. This condition may be present in many different clinical spectra. Various aetiological and risk factors play a crucial role in the causation of CP. In various cases, the causes of CP may not be apparent. Interruption in the supply of oxygen to the fetus or brain asphyxia was considered to be the main causative factor explaining CP. Antenatal, perinatal, and postnatal factors could be involved in the origin of CP. Understanding its pathophysiology is also crucial for developing preventive and protective strategies. A major advancement in the brain stimulation techniques has emerged as a promising status in diagnostic and interventional approaches. This review provides a brief explanation about the various aetiological factors, pathophysiology, and recent therapeutic approaches in the treatment of cerebral palsy.
Collapse
Affiliation(s)
- Jyoti Upadhyay
- School of Health Sciences, University of Petroleum and Energy Studies, Dehradun, India
| | - Nidhi Tiwari
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Mohd Nazam Ansari
- Department of Pharmacology and Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| |
Collapse
|
9
|
Francis F, Cappello S. Neuronal migration and disorders - an update. Curr Opin Neurobiol 2020; 66:57-68. [PMID: 33096394 DOI: 10.1016/j.conb.2020.10.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/15/2020] [Accepted: 10/04/2020] [Indexed: 12/22/2022]
Abstract
This review highlights genes, proteins and subcellular mechanisms, recently shown to influence cortical neuronal migration. A current view on mechanisms which become disrupted in a diverse array of migration disorders is presented. The microtubule (MT) cytoskeleton is a major player in migrating neurons. Recently, variable impacts on MTs have been revealed in different cell compartments. Thus there are a multiplicity of effects involving centrosomal, microtubule-associated, as well as motor proteins. However, other causative factors also emerge, illuminating cortical neuronal migration research. These include disruptions of the actin cytoskeleton, the extracellular matrix, different adhesion molecules and signaling pathways, especially revealed in disorders such as periventricular heterotopia. These recent advances often involve the use of human in vitro models as well as model organisms. Focusing on cell-type specific knockouts and knockins, as well as generating omics and functional data, all seem critical for an integrated view on neuronal migration dysfunction.
Collapse
Affiliation(s)
- Fiona Francis
- INSERM U 1270, Paris, France; Sorbonne University, UMR-S 1270, F-75005 Paris, France; Institut du Fer à Moulin, Paris, France.
| | | |
Collapse
|
10
|
Ferent J, Zaidi D, Francis F. Extracellular Control of Radial Glia Proliferation and Scaffolding During Cortical Development and Pathology. Front Cell Dev Biol 2020; 8:578341. [PMID: 33178693 PMCID: PMC7596222 DOI: 10.3389/fcell.2020.578341] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 09/08/2020] [Indexed: 01/14/2023] Open
Abstract
During the development of the cortex, newly generated neurons migrate long-distances in the expanding tissue to reach their final positions. Pyramidal neurons are produced from dorsal progenitors, e.g., radial glia (RGs) in the ventricular zone, and then migrate along RG processes basally toward the cortex. These neurons are hence dependent upon RG extensions to support their migration from apical to basal regions. Several studies have investigated how intracellular determinants are required for RG polarity and subsequent formation and maintenance of their processes. Fewer studies have identified the influence of the extracellular environment on this architecture. This review will focus on extracellular factors which influence RG morphology and pyramidal neuronal migration during normal development and their perturbations in pathology. During cortical development, RGs are present in different strategic positions: apical RGs (aRGs) have their cell bodies located in the ventricular zone with an apical process contacting the ventricle, while they also have a basal process extending radially to reach the pial surface of the cortex. This particular conformation allows aRGs to be exposed to long range and short range signaling cues, whereas basal RGs (bRGs, also known as outer RGs, oRGs) have their cell bodies located throughout the cortical wall, limiting their access to ventricular factors. Long range signals impacting aRGs include secreted molecules present in the embryonic cerebrospinal fluid (e.g., Neuregulin, EGF, FGF, Wnt, BMP). Secreted molecules also contribute to the extracellular matrix (fibronectin, laminin, reelin). Classical short range factors include cell to cell signaling, adhesion molecules and mechano-transduction mechanisms (e.g., TAG1, Notch, cadherins, mechanical tension). Changes in one or several of these components influencing the RG extracellular environment can disrupt the development or maintenance of RG architecture on which neuronal migration relies, leading to a range of cortical malformations. First, we will detail the known long range signaling cues impacting RG. Then, we will review how short range cell contacts are also important to instruct the RG framework. Understanding how RG processes are structured by their environment to maintain and support radial migration is a critical part of the investigation of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Julien Ferent
- Inserm, U 1270, Paris, France.,Sorbonne University, UMR-S 1270, IFM, Paris, France.,Institut du Fer á Moulin, Paris, France
| | - Donia Zaidi
- Inserm, U 1270, Paris, France.,Sorbonne University, UMR-S 1270, IFM, Paris, France.,Institut du Fer á Moulin, Paris, France
| | - Fiona Francis
- Inserm, U 1270, Paris, France.,Sorbonne University, UMR-S 1270, IFM, Paris, France.,Institut du Fer á Moulin, Paris, France
| |
Collapse
|
11
|
Aleo S, Cinnante C, Avignone S, Prada E, Scuvera G, Ajmone PF, Selicorni A, Costantino MA, Triulzi F, Marchisio P, Gervasini C, Milani D. Olfactory Malformations in Mendelian Disorders of the Epigenetic Machinery. Front Cell Dev Biol 2020; 8:710. [PMID: 32850830 PMCID: PMC7417603 DOI: 10.3389/fcell.2020.00710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 07/13/2020] [Indexed: 11/13/2022] Open
Abstract
Usually overlooked by physicians, olfactory abnormalities are not uncommon. Olfactory malformations have recently been reported in an emerging group of genetic disorders called Mendelian Disorders of the Epigenetic Machinery (MDEM). This study aims to determine the prevalence of olfactory malformations in a heterogeneous group of subjects with MDEM. We reviewed the clinical data of 35 patients, 20 females and 15 males, with a mean age of 9.52 years (SD 4.99). All patients had a MDEM and an already available high-resolution brain MRI scan. Two experienced neuroradiologists reviewed the MR images, noting abnormalities and classifying olfactory malformations. Main findings included Corpus Callosum, Cerebellar vermis, and olfactory defects. The latter were found in 11/35 cases (31.4%), of which 7/11 had Rubinstein-Taybi syndrome (RSTS), 2/11 had CHARGE syndrome, 1/11 had Kleefstra syndrome (KLFS), and 1/11 had Weaver syndrome (WVS). The irregularities mainly concerned the olfactory bulbs and were bilateral in 9/11 patients. With over 30% of our sample having an olfactory malformation, this study reveals a possible new diagnostic marker for MDEM and links the epigenetic machinery to the development of the olfactory bulbs.
Collapse
Affiliation(s)
- Sebastiano Aleo
- Pediatric Highly Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Claudia Cinnante
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neuroradiology Unit, Università degli Studi di Milano, Milan, Italy
| | - Sabrina Avignone
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neuroradiology Unit, Università degli Studi di Milano, Milan, Italy
| | - Elisabetta Prada
- Pediatric Highly Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Giulietta Scuvera
- Pediatric Highly Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Paola Francesca Ajmone
- Child and Adolescent Neuropsychiatric Service (UONPIA), Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Maria Antonella Costantino
- Child and Adolescent Neuropsychiatric Service (UONPIA), Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Fabio Triulzi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neuroradiology Unit, Università degli Studi di Milano, Milan, Italy
| | - Paola Marchisio
- Pediatric Highly Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Cristina Gervasini
- Division of Medical Genetics, Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| | - Donatella Milani
- Pediatric Highly Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
12
|
ZEB1 Represses Neural Differentiation and Cooperates with CTBP2 to Dynamically Regulate Cell Migration during Neocortex Development. Cell Rep 2020; 27:2335-2353.e6. [PMID: 31116980 DOI: 10.1016/j.celrep.2019.04.081] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 02/28/2019] [Accepted: 04/16/2019] [Indexed: 01/08/2023] Open
Abstract
Zinc-finger E-box binding homeobox 1 (Zeb1) is a key regulator of epithelial-mesenchymal transition and cancer metastasis. Mutation of ZEB1 is associated with human diseases and defective brain development. Here we show that downregulation of Zeb1 expression in embryonic cortical neural progenitor cells (NPCs) is necessary for proper neuronal differentiation and migration. Overexpression of Zeb1 during neuronal differentiation, when its expression normally declines, blocks NPC lineage progression and disrupts multipolar-to-bipolar transition of differentiating neurons, leading to severe migration defects and subcortical heterotopia bands at postnatal stages. ZEB1 regulates a cohort of genes involved in cell differentiation and migration, including Neurod1 and Pard6b. The interaction between ZEB1 and CTBP2 in the embryonic cerebral cortex is required for ZEB1 to elicit its effect on the multipolar-to-bipolar transition, but not its suppression of Neurod1. These findings provide insights into understanding the complexity of transcriptional regulation during neuronal differentiation.
Collapse
|
13
|
Liu J, Liu S, Gao H, Han L, Chu X, Sheng Y, Shou W, Wang Y, Liu Y, Wan J, Yang L. Genome-wide studies reveal the essential and opposite roles of ARID1A in controlling human cardiogenesis and neurogenesis from pluripotent stem cells. Genome Biol 2020; 21:169. [PMID: 32646524 PMCID: PMC7350744 DOI: 10.1186/s13059-020-02082-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 06/22/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Early human heart and brain development simultaneously occur during embryogenesis. Notably, in human newborns, congenital heart defects strongly associate with neurodevelopmental abnormalities, suggesting a common gene or complex underlying both cardiogenesis and neurogenesis. However, due to lack of in vivo studies, the molecular mechanisms that govern both early human heart and brain development remain elusive. RESULTS Here, we report ARID1A, a DNA-binding subunit of the SWI/SNF epigenetic complex, controls both neurogenesis and cardiogenesis from human embryonic stem cells (hESCs) through distinct mechanisms. Knockout-of-ARID1A (ARID1A-/-) leads to spontaneous differentiation of neural cells together with globally enhanced expression of neurogenic genes in undifferentiated hESCs. Additionally, when compared with WT hESCs, cardiac differentiation from ARID1A -/- hESCs is prominently suppressed, whereas neural differentiation is significantly promoted. Whole genome-wide scRNA-seq, ATAC-seq, and ChIP-seq analyses reveal that ARID1A is required to open chromatin accessibility on promoters of essential cardiogenic genes, and temporally associated with key cardiogenic transcriptional factors T and MEF2C during early cardiac development. However, during early neural development, transcription of most essential neurogenic genes is dependent on ARID1A, which can interact with a known neural restrictive silencer factor REST/NRSF. CONCLUSIONS We uncover the opposite roles by ARID1A to govern both early cardiac and neural development from pluripotent stem cells. Global chromatin accessibility on cardiogenic genes is dependent on ARID1A, whereas transcriptional activity of neurogenic genes is under control by ARID1A, possibly through ARID1A-REST/NRSF interaction.
Collapse
Affiliation(s)
- Juli Liu
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, 1044 W Walnut Street, R4 272, Indianapolis, IN, 46202, USA
| | - Sheng Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Hongyu Gao
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Lei Han
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, 1044 W Walnut Street, R4 272, Indianapolis, IN, 46202, USA
| | - Xiaona Chu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Yi Sheng
- Department of Obstetrics, Gynecology & Reproductive Sciences, Magee-Women's Research Institute, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Weinian Shou
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, 1044 W Walnut Street, R4 272, Indianapolis, IN, 46202, USA
| | - Yue Wang
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Yunlong Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of BioHealth Informatics, Indiana University School of Informatics and Computing, Indiana University - Purdue University Indianapolis, Indianapolis, IN, 46202, USA
| | - Jun Wan
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
- Department of BioHealth Informatics, Indiana University School of Informatics and Computing, Indiana University - Purdue University Indianapolis, Indianapolis, IN, 46202, USA.
| | - Lei Yang
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, 1044 W Walnut Street, R4 272, Indianapolis, IN, 46202, USA.
| |
Collapse
|
14
|
Jossin Y. Molecular mechanisms of cell polarity in a range of model systems and in migrating neurons. Mol Cell Neurosci 2020; 106:103503. [PMID: 32485296 DOI: 10.1016/j.mcn.2020.103503] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 04/20/2020] [Accepted: 05/23/2020] [Indexed: 01/09/2023] Open
Abstract
Cell polarity is defined as the asymmetric distribution of cellular components along an axis. Most cells, from the simplest single-cell organisms to highly specialized mammalian cells, are polarized and use similar mechanisms to generate and maintain polarity. Cell polarity is important for cells to migrate, form tissues, and coordinate activities. During development of the mammalian cerebral cortex, cell polarity is essential for neurogenesis and for the migration of newborn but as-yet undifferentiated neurons. These oriented migrations include both the radial migration of excitatory projection neurons and the tangential migration of inhibitory interneurons. In this review, I will first describe the development of the cerebral cortex, as revealed at the cellular level. I will then define the core molecular mechanisms - the Par/Crb/Scrib polarity complexes, small GTPases, the actin and microtubule cytoskeletons, and phosphoinositides/PI3K signaling - that are required for asymmetric cell division, apico-basal and front-rear polarity in model systems, including C elegans zygote, Drosophila embryos and cultured mammalian cells. As I go through each core mechanism I will explain what is known about its importance in radial and tangential migration in the developing mammalian cerebral cortex.
Collapse
Affiliation(s)
- Yves Jossin
- Laboratory of Mammalian Development & Cell Biology, Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium.
| |
Collapse
|
15
|
Li S, Wang M, Zhou J. Brain Organoids: A Promising Living Biobank Resource for Neuroscience Research. Biopreserv Biobank 2020; 18:136-143. [PMID: 31977235 DOI: 10.1089/bio.2019.0111] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Biobanking plays an important role between clinical practice and translational research. In addition to the traditional biomolecular-based biobanks, there is a growing interest in establishing living biobanks, including organoid biobanks that can collect and store viable and functional tissues and proliferative cell types for long periods of time. An organoid is a three-dimensional cell complex derived by self-organization of small tissue blocks or stem cells, which can recapitulate the phenotypic and genetic characteristics of targeted human organs. Publications on brain organoids have increased recently, and several types of brain organoids have been reported to model normal and abnormal neural development, as well as different neurodegenerative diseases, neuropsychiatric disorders, and other neural conditions. Based on the current status of research, more exploration on brain organoids is needed, through technical advancements, to improve the reproducibility and scalability, as well as to decrease the diversity. Moreover, given their natural characteristics, more attention to ethical considerations is needed, considering the extent of maturation and complexity of brain organoids. Living biobanks that are engaged in collecting categories of brain organoids possessing different genetic backgrounds, and with spatial and temporal characteristics, will eventually contribute to the understanding of neural conditions and ultimately facilitate innovative treatment development.
Collapse
Affiliation(s)
- Shuang Li
- Department of Central Laboratory, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Min Wang
- Department of Central Laboratory, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China.,Department of Pediatric Hematology and Oncology, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Junmei Zhou
- Department of Central Laboratory, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
16
|
Groden M, Weigand M, Triesch J, Jedlicka P, Cuntz H. A Model of Brain Folding Based on Strong Local and Weak Long-Range Connectivity Requirements. Cereb Cortex 2019; 30:2434-2451. [DOI: 10.1093/cercor/bhz249] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 08/20/2019] [Accepted: 10/01/2019] [Indexed: 12/21/2022] Open
Abstract
Abstract
Throughout the animal kingdom, the structure of the central nervous system varies widely from distributed ganglia in worms to compact brains with varying degrees of folding in mammals. The differences in structure may indicate a fundamentally different circuit organization. However, the folded brain most likely is a direct result of mechanical forces when considering that a larger surface area of cortex packs into the restricted volume provided by the skull. Here, we introduce a computational model that instead of modeling mechanical forces relies on dimension reduction methods to place neurons according to specific connectivity requirements. For a simplified connectivity with strong local and weak long-range connections, our model predicts a transition from separate ganglia through smooth brain structures to heavily folded brains as the number of cortical columns increases. The model reproduces experimentally determined relationships between metrics of cortical folding and its pathological phenotypes in lissencephaly, polymicrogyria, microcephaly, autism, and schizophrenia. This suggests that mechanical forces that are known to lead to cortical folding may synergistically contribute to arrangements that reduce wiring. Our model provides a unified conceptual understanding of gyrification linking cellular connectivity and macroscopic structures in large-scale neural network models of the brain.
Collapse
Affiliation(s)
- Moritz Groden
- Ernst Strüngmann Institute (ESI) for Neuroscience in Cooperation with Max Planck Society, Frankfurt am Main D-60528, Germany
- Frankfurt Institute for Advanced Studies (FIAS), Frankfurt am Main D-60438, Germany
- ICAR3R—Interdisciplinary Centre for 3Rs in Animal Research, Justus Liebig University Giessen, Giessen D-35390, Germany
| | - Marvin Weigand
- Ernst Strüngmann Institute (ESI) for Neuroscience in Cooperation with Max Planck Society, Frankfurt am Main D-60528, Germany
- Frankfurt Institute for Advanced Studies (FIAS), Frankfurt am Main D-60438, Germany
- Faculty of Biological Sciences, Goethe University, Frankfurt am Main D-60438, Germany
| | - Jochen Triesch
- Frankfurt Institute for Advanced Studies (FIAS), Frankfurt am Main D-60438, Germany
- Faculty of Physics, Goethe University, Frankfurt am Main D-60438, Germany
- Faculty of Computer Science and Mathematics, Goethe University, Frankfurt am Main D-60438, Germany
| | - Peter Jedlicka
- Frankfurt Institute for Advanced Studies (FIAS), Frankfurt am Main D-60438, Germany
- ICAR3R—Interdisciplinary Centre for 3Rs in Animal Research, Justus Liebig University Giessen, Giessen D-35390, Germany
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe University, Frankfurt am Main D-60528, Germany
| | - Hermann Cuntz
- Ernst Strüngmann Institute (ESI) for Neuroscience in Cooperation with Max Planck Society, Frankfurt am Main D-60528, Germany
- Frankfurt Institute for Advanced Studies (FIAS), Frankfurt am Main D-60438, Germany
| |
Collapse
|
17
|
Loss of Tiparp Results in Aberrant Layering of the Cerebral Cortex. eNeuro 2019; 6:ENEURO.0239-19.2019. [PMID: 31704703 PMCID: PMC6883171 DOI: 10.1523/eneuro.0239-19.2019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 09/13/2019] [Accepted: 10/10/2019] [Indexed: 01/26/2023] Open
Abstract
2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-inducible poly-ADP-ribose polymerase (TIPARP) is an enzyme that adds a single ADP-ribose moiety to itself or other proteins. Tiparp is highly expressed in the brain; however, its function in this organ is unknown. Here, we used Tiparp–/– mice to determine Tiparp’s role in the development of the prefrontal cortex. 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-inducible poly-ADP-ribose polymerase (TIPARP) is an enzyme that adds a single ADP-ribose moiety to itself or other proteins. Tiparp is highly expressed in the brain; however, its function in this organ is unknown. Here, we used Tiparp–/– mice to determine Tiparp’s role in the development of the prefrontal cortex. Loss of Tiparp resulted in an aberrant organization of the mouse cortex, where the upper layers presented increased cell density in the knock-out mice compared with wild type. Tiparp loss predominantly affected the correct distribution and number of GABAergic neurons. Furthermore, neural progenitor cell proliferation was significantly reduced. Neural stem cells (NSCs) derived from Tiparp–/– mice showed a slower rate of migration. Cytoskeletal components, such as α-tubulin are key regulators of neuronal differentiation and cortical development. α-tubulin mono-ADP ribosylation (MAR) levels were reduced in Tiparp–/– cells, suggesting that Tiparp plays a role in the MAR of α-tubulin. Despite the mild phenotype presented by Tiparp–/– mice, our findings reveal an important function for Tiparp and MAR in the correct development of the cortex. Unravelling Tiparp’s role in the cortex, could pave the way to a better understanding of a wide spectrum of neurological diseases which are known to have increased expression of TIPARP.
Collapse
|
18
|
Kon E, Calvo-Jiménez E, Cossard A, Na Y, Cooper JA, Jossin Y. N-cadherin-regulated FGFR ubiquitination and degradation control mammalian neocortical projection neuron migration. eLife 2019; 8:47673. [PMID: 31577229 PMCID: PMC6786859 DOI: 10.7554/elife.47673] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Accepted: 10/01/2019] [Indexed: 12/18/2022] Open
Abstract
The functions of FGF receptors (FGFRs) in early development of the cerebral cortex are well established. Their functions in the migration of neocortical projection neurons, however, are unclear. We have found that FGFRs regulate multipolar neuron orientation and the morphological change into bipolar cells necessary to enter the cortical plate. Mechanistically, our results suggest that FGFRs are activated by N-Cadherin. N-Cadherin cell-autonomously binds FGFRs and inhibits FGFR K27- and K29-linked polyubiquitination and lysosomal degradation. Accordingly, FGFRs accumulate and stimulate prolonged Erk1/2 phosphorylation. Neurons inhibited for Erk1/2 are stalled in the multipolar zone. Moreover, Reelin, a secreted protein regulating neuronal positioning, prevents FGFR degradation through N-Cadherin, causing Erk1/2 phosphorylation. These findings reveal novel functions for FGFRs in cortical projection neuron migration, suggest a physiological role for FGFR and N-Cadherin interaction in vivo and identify Reelin as an extracellular upstream regulator and Erk1/2 as downstream effectors of FGFRs during neuron migration.
Collapse
Affiliation(s)
- Elif Kon
- Laboratory of Mammalian Development & Cell Biology, Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| | - Elisa Calvo-Jiménez
- Laboratory of Mammalian Development & Cell Biology, Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| | - Alexia Cossard
- Laboratory of Mammalian Development & Cell Biology, Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| | - Youn Na
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, United States
| | - Jonathan A Cooper
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, United States
| | - Yves Jossin
- Laboratory of Mammalian Development & Cell Biology, Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
19
|
Adams JW, Cugola FR, Muotri AR. Brain Organoids as Tools for Modeling Human Neurodevelopmental Disorders. Physiology (Bethesda) 2019; 34:365-375. [PMID: 31389776 PMCID: PMC6863377 DOI: 10.1152/physiol.00005.2019] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 04/12/2019] [Accepted: 04/12/2019] [Indexed: 12/15/2022] Open
Abstract
Brain organoids recapitulate in vitro the specific stages of in vivo human brain development, thus offering an innovative tool by which to model human neurodevelopmental disease. We review here how brain organoids have been used to study neurodevelopmental disease and consider their potential for both technological advancement and therapeutic development.
Collapse
Affiliation(s)
- Jason W Adams
- Department of Pediatrics/Rady Children's Hospital San Diego, School of Medicine, University of California San Diego, San Diego, California
- Department of Cellular & Molecular Medicine, Stem Cell Program, Center for Academic Research and Training in Anthropogeny (CARTA), Kavli Institute for Brain and Mind, La Jolla, California
- Department of Neurosciences, School of Medicine, University of California San Diego, San Diego, California
| | - Fernanda R Cugola
- Department of Pediatrics/Rady Children's Hospital San Diego, School of Medicine, University of California San Diego, San Diego, California
- Department of Cellular & Molecular Medicine, Stem Cell Program, Center for Academic Research and Training in Anthropogeny (CARTA), Kavli Institute for Brain and Mind, La Jolla, California
| | - Alysson R Muotri
- Department of Pediatrics/Rady Children's Hospital San Diego, School of Medicine, University of California San Diego, San Diego, California
- Department of Cellular & Molecular Medicine, Stem Cell Program, Center for Academic Research and Training in Anthropogeny (CARTA), Kavli Institute for Brain and Mind, La Jolla, California
| |
Collapse
|
20
|
Insolia V, Priori EC, Gasperini C, Coppa F, Cocchia M, Iervasi E, Ferrari B, Besio R, Maruelli S, Bernocchi G, Forlino A, Bottone MG. Prolidase enzyme is required for extracellular matrix integrity and impacts on postnatal cerebellar cortex development. J Comp Neurol 2019; 528:61-80. [PMID: 31246278 DOI: 10.1002/cne.24735] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 06/11/2019] [Accepted: 06/20/2019] [Indexed: 12/12/2022]
Abstract
The extracellular matrix is essential for brain development, lamination, and synaptogenesis. In particular, the basement membrane below the pial meninx (pBM) is required for correct cortical development. The last step in the catabolism of the most abundant protein in pBM, collagen Type IV, requires prolidase, an exopeptidase cleaving the imidodipeptides containing pro or hyp at the C-terminal end. Mutations impairing prolidase activity lead in humans to the rare disease prolidase deficiency characterized by severe skin ulcers and mental impairment. Thus, the dark-like (dal) mouse, in which the prolidase is knocked-out, was used to investigate whether the deficiency of prolidase affects the neuronal maturation during development of a brain cortex area. Focusing on the cerebellar cortex, thinner collagen fibers and disorganized pBM were found. Aberrant cortical granule cell proliferation and migration occurred, associated to defects in brain lamination, and in particular in maturation of Purkinje neurons and formation of synaptic contacts. This study deeply elucidates a link between prolidase activity and neuronal maturation shedding new light on the molecular basis of functional aspects in the prolidase deficiency.
Collapse
Affiliation(s)
- Violetta Insolia
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Erica C Priori
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Caterina Gasperini
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Federica Coppa
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Marco Cocchia
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Erika Iervasi
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Beatrice Ferrari
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Roberta Besio
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Pavia, Italy
| | - Silvia Maruelli
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Pavia, Italy
| | | | - Antonella Forlino
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Pavia, Italy
| | - Maria G Bottone
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| |
Collapse
|
21
|
Minocha S, Herr W. Cortical and Commissural Defects Upon HCF-1 Loss in Nkx2.1-Derived Embryonic Neurons and Glia. Dev Neurobiol 2019; 79:578-595. [PMID: 31207118 PMCID: PMC6771735 DOI: 10.1002/dneu.22704] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 06/06/2019] [Accepted: 06/06/2019] [Indexed: 11/28/2022]
Abstract
Formation of the cerebral cortex and commissures involves a complex developmental process defined by multiple molecular mechanisms governing proliferation of neuronal and glial precursors, neuronal and glial migration, and patterning events. Failure in any of these processes can lead to malformations. Here, we study the role of HCF-1 in these processes. HCF-1 is a conserved metazoan transcriptional co-regulator long implicated in cell proliferation and more recently in human metabolic disorders and mental retardation. Loss of HCF-1 in a subset of ventral telencephalic Nkx2.1-positive progenitors leads to reduced numbers of GABAergic interneurons and glia, owing not to decreased proliferation but rather to increased apoptosis before cell migration. The loss of these cells leads to development of severe commissural and cortical defects in early postnatal mouse brains. These defects include mild and severe structural defects of the corpus callosum and anterior commissure, respectively, and increased folding of the cortex resembling polymicrogyria. Hence, in addition to its well-established role in cell proliferation, HCF-1 is important for organ development, here the brain.
Collapse
Affiliation(s)
- Shilpi Minocha
- Center for Integrative Genomics, GénopodeUniversity of LausanneLausanneCH‐1015Switzerland
| | - Winship Herr
- Center for Integrative Genomics, GénopodeUniversity of LausanneLausanneCH‐1015Switzerland
| |
Collapse
|
22
|
TUBG1 missense variants underlying cortical malformations disrupt neuronal locomotion and microtubule dynamics but not neurogenesis. Nat Commun 2019; 10:2129. [PMID: 31086189 PMCID: PMC6513894 DOI: 10.1038/s41467-019-10081-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 04/05/2019] [Indexed: 01/13/2023] Open
Abstract
De novo heterozygous missense variants in the γ-tubulin gene TUBG1 have been linked to human malformations of cortical development associated with intellectual disability and epilepsy. Here, we investigated through in-utero electroporation and in-vivo studies, how four of these variants affect cortical development. We show that TUBG1 mutants affect neuronal positioning, disrupting the locomotion of new-born neurons but without affecting progenitors’ proliferation. We further demonstrate that pathogenic TUBG1 variants are linked to reduced microtubule dynamics but without major structural nor functional centrosome defects in subject-derived fibroblasts. Additionally, we developed a knock-in Tubg1Y92C/+ mouse model and assessed consequences of the mutation. Although centrosomal positioning in bipolar neurons is correct, they fail to initiate locomotion. Furthermore, Tubg1Y92C/+ animals show neuroanatomical and behavioral defects and increased epileptic cortical activity. We show that Tubg1Y92C/+ mice partially mimic the human phenotype and therefore represent a relevant model for further investigations of the physiopathology of cortical malformations. New mutations and genes associated with malformations of cortical development keep being identified, yet there is little known about the underlying cellular mechanisms controlling these impairments. Here, authors generate and characterize a heterozygous TUBG1 knock-in mouse model bearing one of these known mutations and show that TUBG1 mutation leads to the miss-positioning of neurons in the cortical wall due to migration, because of defective microtubules dynamics, and not proliferation defects during corticogenesis.
Collapse
|
23
|
Takano T, Funahashi Y, Kaibuchi K. Neuronal Polarity: Positive and Negative Feedback Signals. Front Cell Dev Biol 2019; 7:69. [PMID: 31069225 PMCID: PMC6491837 DOI: 10.3389/fcell.2019.00069] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 04/09/2019] [Indexed: 12/21/2022] Open
Abstract
Establishment and maintenance of neuronal polarity are critical for neuronal development and function. One of the fundamental questions in neurodevelopment is how neurons generate only one axon and several dendrites from multiple minor neurites. Over the past few decades, molecular and cell biological approaches have unveiled a large number of signaling networks regulating neuronal polarity in cultured hippocampal neurons and the developing cortex. Emerging evidence reveals that positive and negative feedback signals play a crucial role in axon and dendrite specification. Positive feedback signals are continuously activated in one of minor neurites and result in axon specification and elongation, whereas negative feedback signals are propagated from a nascent axon terminal to all minor neurites and inhibit the formation of multiple axon, thereby leading to dendrite specification, and maintaining neuronal polarity. This current insight provides a holistic picture of the signaling mechanisms underlying neuronal polarization during neuronal development. Here, our review highlights recent advancements in this fascinating field, with a focus on the positive, and negative feedback signals as key regulatory mechanisms underlying neuronal polarization.
Collapse
Affiliation(s)
- Tetsuya Takano
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Department of Cell Biology, Duke University Medical School, Durham, NC, United States
| | - Yasuhiro Funahashi
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kozo Kaibuchi
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
24
|
Buchsbaum IY, Cappello S. Neuronal migration in the CNS during development and disease: insights from in vivo and in vitro models. Development 2019; 146:146/1/dev163766. [DOI: 10.1242/dev.163766] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
ABSTRACT
Neuronal migration is a fundamental process that governs embryonic brain development. As such, mutations that affect essential neuronal migration processes lead to severe brain malformations, which can cause complex and heterogeneous developmental and neuronal migration disorders. Our fragmented knowledge about the aetiology of these disorders raises numerous issues. However, many of these can now be addressed through studies of in vivo and in vitro models that attempt to recapitulate human-specific mechanisms of cortical development. In this Review, we discuss the advantages and limitations of these model systems and suggest that a complementary approach, using combinations of in vivo and in vitro models, will broaden our knowledge of the molecular and cellular mechanisms that underlie defective neuronal positioning in the human cerebral cortex.
Collapse
Affiliation(s)
- Isabel Yasmin Buchsbaum
- Developmental Neurobiology, Max Planck Institute of Psychiatry, 80804 Munich, Germany
- Graduate School of Systemic Neurosciences, Ludwig-Maximilians-University Munich, 82152 Planegg, Germany
| | - Silvia Cappello
- Developmental Neurobiology, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| |
Collapse
|
25
|
Marino BS, Tabbutt S, MacLaren G, Hazinski MF, Adatia I, Atkins DL, Checchia PA, DeCaen A, Fink EL, Hoffman GM, Jefferies JL, Kleinman M, Krawczeski CD, Licht DJ, Macrae D, Ravishankar C, Samson RA, Thiagarajan RR, Toms R, Tweddell J, Laussen PC. Cardiopulmonary Resuscitation in Infants and Children With Cardiac Disease: A Scientific Statement From the American Heart Association. Circulation 2018; 137:e691-e782. [PMID: 29685887 DOI: 10.1161/cir.0000000000000524] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Cardiac arrest occurs at a higher rate in children with heart disease than in healthy children. Pediatric basic life support and advanced life support guidelines focus on delivering high-quality resuscitation in children with normal hearts. The complexity and variability in pediatric heart disease pose unique challenges during resuscitation. A writing group appointed by the American Heart Association reviewed the literature addressing resuscitation in children with heart disease. MEDLINE and Google Scholar databases were searched from 1966 to 2015, cross-referencing pediatric heart disease with pertinent resuscitation search terms. The American College of Cardiology/American Heart Association classification of recommendations and levels of evidence for practice guidelines were used. The recommendations in this statement concur with the critical components of the 2015 American Heart Association pediatric basic life support and pediatric advanced life support guidelines and are meant to serve as a resuscitation supplement. This statement is meant for caregivers of children with heart disease in the prehospital and in-hospital settings. Understanding the anatomy and physiology of the high-risk pediatric cardiac population will promote early recognition and treatment of decompensation to prevent cardiac arrest, increase survival from cardiac arrest by providing high-quality resuscitations, and improve outcomes with postresuscitation care.
Collapse
|
26
|
Genetics and mechanisms leading to human cortical malformations. Semin Cell Dev Biol 2018; 76:33-75. [DOI: 10.1016/j.semcdb.2017.09.031] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 09/21/2017] [Accepted: 09/21/2017] [Indexed: 02/06/2023]
|
27
|
Iefremova V, Manikakis G, Krefft O, Jabali A, Weynans K, Wilkens R, Marsoner F, Brändl B, Müller FJ, Koch P, Ladewig J. An Organoid-Based Model of Cortical Development Identifies Non-Cell-Autonomous Defects in Wnt Signaling Contributing to Miller-Dieker Syndrome. Cell Rep 2017; 19:50-59. [PMID: 28380362 DOI: 10.1016/j.celrep.2017.03.047] [Citation(s) in RCA: 191] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 01/25/2017] [Accepted: 03/13/2017] [Indexed: 02/06/2023] Open
Abstract
Miller-Dieker syndrome (MDS) is caused by a heterozygous deletion of chromosome 17p13.3 involving the genes LIS1 and YWHAE (coding for 14.3.3ε) and leads to malformations during cortical development. Here, we used patient-specific forebrain-type organoids to investigate pathological changes associated with MDS. Patient-derived organoids are significantly reduced in size, a change accompanied by a switch from symmetric to asymmetric cell division of ventricular zone radial glia cells (vRGCs). Alterations in microtubule network organization in vRGCs and a disruption of cortical niche architecture, including altered expression of cell adhesion molecules, are also observed. These phenotypic changes lead to a non-cell-autonomous disturbance of the N-cadherin/β-catenin signaling axis. Reinstalling active β-catenin signaling rescues division modes and ameliorates growth defects. Our data define the role of LIS1 and 14.3.3ε in maintaining the cortical niche and highlight the utility of organoid-based systems for modeling complex cell-cell interactions in vitro.
Collapse
Affiliation(s)
- Vira Iefremova
- Institute of Reconstructive Neurobiology, University of Bonn, Bonn 53127, Germany
| | - George Manikakis
- Institute of Reconstructive Neurobiology, University of Bonn, Bonn 53127, Germany
| | - Olivia Krefft
- Institute of Reconstructive Neurobiology, University of Bonn, Bonn 53127, Germany
| | - Ammar Jabali
- Institute of Reconstructive Neurobiology, University of Bonn, Bonn 53127, Germany
| | - Kevin Weynans
- Institute of Reconstructive Neurobiology, University of Bonn, Bonn 53127, Germany
| | - Ruven Wilkens
- Institute of Reconstructive Neurobiology, University of Bonn, Bonn 53127, Germany
| | - Fabio Marsoner
- Institute of Reconstructive Neurobiology, University of Bonn, Bonn 53127, Germany
| | - Björn Brändl
- Department of Psychiatry and Psychotherapy, University Hospital Schleswig Holstein, Kiel 24105, Germany
| | - Franz-Josef Müller
- Department of Psychiatry and Psychotherapy, University Hospital Schleswig Holstein, Kiel 24105, Germany
| | - Philipp Koch
- Institute of Reconstructive Neurobiology, University of Bonn, Bonn 53127, Germany.
| | - Julia Ladewig
- Institute of Reconstructive Neurobiology, University of Bonn, Bonn 53127, Germany.
| |
Collapse
|
28
|
Abstract
A 1-year-old neutered male Pekingese was presented for evaluation and further treatment
of cluster seizures. The dog had behavioral abnormalities, and a prosencephalic lesion was
suspected following neurological examination. The dog showed signs of learning difficulty.
Magnetic resonance imaging of the brain revealed a remarkably smooth cerebral cortex with
a reduced number of gyri, as well as a cystic lesion associated with the quadrigeminal
cistern. A diagnosis of lissencephaly, concurrent with a quadrigeminal cisternal cyst, was
made. High-dose and multiple anticonvulsants were necessary to control the seizures. This
is the first report of lissencephaly in a Pekingese.
Collapse
Affiliation(s)
- Genya Shimbo
- Veterinary Medical Center, Obihiro University of Agriculture and Veterinary Medicine, Inada, Obihiro, Hokkaido 080-8555, Japan
| | - Michihito Tagawa
- Veterinary Medical Center, Obihiro University of Agriculture and Veterinary Medicine, Inada, Obihiro, Hokkaido 080-8555, Japan
| | - Eiji Oohashi
- Akashiya Animal Clinic, Satsunai Sakura-machi, Makubetsu-cho, Nakagawa-gun, Hokkaido 089-0535, Japan
| | - Masashi Yanagawa
- Department of Applied Veterinary Medicine, Obihiro University of Agriculture and Veterinary Medicine, Inada, Obihiro, Hokkaido 080-8555, Japan
| | - Kazuro Miyahara
- Veterinary Medical Center, Obihiro University of Agriculture and Veterinary Medicine, Inada, Obihiro, Hokkaido 080-8555, Japan
| |
Collapse
|
29
|
Shinmyo Y, Terashita Y, Dinh Duong TA, Horiike T, Kawasumi M, Hosomichi K, Tajima A, Kawasaki H. Folding of the Cerebral Cortex Requires Cdk5 in Upper-Layer Neurons in Gyrencephalic Mammals. Cell Rep 2017; 20:2131-2143. [DOI: 10.1016/j.celrep.2017.08.024] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Revised: 06/08/2017] [Accepted: 08/03/2017] [Indexed: 02/06/2023] Open
|
30
|
Kon E, Cossard A, Jossin Y. Neuronal Polarity in the Embryonic Mammalian Cerebral Cortex. Front Cell Neurosci 2017; 11:163. [PMID: 28670267 PMCID: PMC5472699 DOI: 10.3389/fncel.2017.00163] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 05/26/2017] [Indexed: 11/13/2022] Open
Abstract
The cerebral cortex is composed of billions of neurons that can grossly be subdivided into two broad classes: inhibitory GABAergic interneurons and excitatory glutamatergic neurons. The majority of cortical neurons in mammals are the excitatory type and they are the main focus of this review article. Like many of the cells in multicellular organisms, fully differentiated neurons are both morphologically and functionally polarized. However, they go through several changes in polarity before reaching this final mature differentiated state. Neurons are derived from polarized neuronal progenitor/stem cells and their commitment to neuronal fate is decided by cellular and molecular asymmetry during their last division in the neurogenic zone. They migrate from their birthplace using so-called multipolar migration, during which they switch direction of movement several times, and repolarize for bipolar migration when the axon is specified. Therefore, neurons have to break their previous symmetry, change their morphology and adequately respond to polarizing signals during migration in order to reach the correct position in the cortex and start making connections. Finally, the dendritic tree is elaborated and the axon/dendrite morphological polarity is set. Here we will describe the function, establishment and maintenance of polarity during the different developmental steps starting from neural stem cell (NSC) division, neuronal migration and axon specification at embryonic developmental stages.
Collapse
Affiliation(s)
- Elif Kon
- Mammalian Development and Cell Biology Unit, Institute of Neuroscience, Université catholique de LouvainBrussels, Belgium
| | - Alexia Cossard
- Mammalian Development and Cell Biology Unit, Institute of Neuroscience, Université catholique de LouvainBrussels, Belgium
| | - Yves Jossin
- Mammalian Development and Cell Biology Unit, Institute of Neuroscience, Université catholique de LouvainBrussels, Belgium
| |
Collapse
|
31
|
Jossin Y, Lee M, Klezovitch O, Kon E, Cossard A, Lien WH, Fernandez TE, Cooper JA, Vasioukhin V. Llgl1 Connects Cell Polarity with Cell-Cell Adhesion in Embryonic Neural Stem Cells. Dev Cell 2017; 41:481-495.e5. [PMID: 28552558 DOI: 10.1016/j.devcel.2017.05.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 04/11/2017] [Accepted: 05/01/2017] [Indexed: 10/19/2022]
Abstract
Malformations of the cerebral cortex (MCCs) are devastating developmental disorders. We report here that mice with embryonic neural stem-cell-specific deletion of Llgl1 (Nestin-Cre/Llgl1fl/fl), a mammalian ortholog of the Drosophila cell polarity gene lgl, exhibit MCCs resembling severe periventricular heterotopia (PH). Immunohistochemical analyses and live cortical imaging of PH formation revealed that disruption of apical junctional complexes (AJCs) was responsible for PH in Nestin-Cre/Llgl1fl/fl brains. While it is well known that cell polarity proteins govern the formation of AJCs, the exact mechanisms remain unclear. We show that LLGL1 directly binds to and promotes internalization of N-cadherin, and N-cadherin/LLGL1 interaction is inhibited by atypical protein kinase C-mediated phosphorylation of LLGL1, restricting the accumulation of AJCs to the basolateral-apical boundary. Disruption of the N-cadherin-LLGL1 interaction during cortical development in vivo is sufficient for PH. These findings reveal a mechanism responsible for the physical and functional connection between cell polarity and cell-cell adhesion machineries in mammalian cells.
Collapse
Affiliation(s)
- Yves Jossin
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Mammalian Development & Cell Biology Unit, Institute of Neuroscience, Université Catholique de Louvain, 1200 Brussels, Belgium.
| | - Minhui Lee
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
| | - Olga Klezovitch
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Elif Kon
- Mammalian Development & Cell Biology Unit, Institute of Neuroscience, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Alexia Cossard
- Mammalian Development & Cell Biology Unit, Institute of Neuroscience, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Wen-Hui Lien
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
| | - Tania E Fernandez
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Jonathan A Cooper
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
| | - Valera Vasioukhin
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA; Department of Pathology, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
32
|
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: 2.0] [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
|
33
|
Zollo M, Ahmed M, Ferrucci V, Salpietro V, Asadzadeh F, Carotenuto M, Maroofian R, Al-Amri A, Singh R, Scognamiglio I, Mojarrad M, Musella L, Duilio A, Di Somma A, Karaca E, Rajab A, Al-Khayat A, Mohan Mohapatra T, Eslahi A, Ashrafzadeh F, Rawlins LE, Prasad R, Gupta R, Kumari P, Srivastava M, Cozzolino F, Kumar Rai S, Monti M, Harlalka GV, Simpson MA, Rich P, Al-Salmi F, Patton MA, Chioza BA, Efthymiou S, Granata F, Di Rosa G, Wiethoff S, Borgione E, Scuderi C, Mankad K, Hanna MG, Pucci P, Houlden H, Lupski JR, Crosby AH, Baple EL. PRUNE is crucial for normal brain development and mutated in microcephaly with neurodevelopmental impairment. Brain 2017; 140:940-952. [PMID: 28334956 PMCID: PMC5382943 DOI: 10.1093/brain/awx014] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 12/13/2016] [Indexed: 12/22/2022] Open
Abstract
PRUNE is a member of the DHH (Asp-His-His) phosphoesterase protein superfamily of molecules important for cell motility, and implicated in cancer progression. Here we investigated multiple families from Oman, India, Iran and Italy with individuals affected by a new autosomal recessive neurodevelopmental and degenerative disorder in which the cardinal features include primary microcephaly and profound global developmental delay. Our genetic studies identified biallelic mutations of PRUNE1 as responsible. Our functional assays of disease-associated variant alleles revealed impaired microtubule polymerization, as well as cell migration and proliferation properties, of mutant PRUNE. Additionally, our studies also highlight a potential new role for PRUNE during microtubule polymerization, which is essential for the cytoskeletal rearrangements that occur during cellular division and proliferation. Together these studies define PRUNE as a molecule fundamental for normal human cortical development and define cellular and clinical consequences associated with PRUNE mutation.
Collapse
Affiliation(s)
- Massimo Zollo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche DMMBM, Università di Napoli Federico II, Via Sergio Pansini 5, Naples, 80131, Italy.,CEINGE Biotecnologie Avanzate, Via Gaetano Salvatore 486, Naples, Italy.,European School of Molecular Medicine, SEMM, University of Milan, Italy
| | - Mustafa Ahmed
- Medical Research (Level 4), RILD Wellcome Wolfson Centre, University of Exeter Medical School, Royal Devon & Exeter NHS Foundation Trust, Barrack Road, Exeter, EX2 5DW, UK
| | - Veronica Ferrucci
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche DMMBM, Università di Napoli Federico II, Via Sergio Pansini 5, Naples, 80131, Italy.,CEINGE Biotecnologie Avanzate, Via Gaetano Salvatore 486, Naples, Italy.,European School of Molecular Medicine, SEMM, University of Milan, Italy
| | - Vincenzo Salpietro
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - Fatemeh Asadzadeh
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche DMMBM, Università di Napoli Federico II, Via Sergio Pansini 5, Naples, 80131, Italy.,CEINGE Biotecnologie Avanzate, Via Gaetano Salvatore 486, Naples, Italy
| | - Marianeve Carotenuto
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche DMMBM, Università di Napoli Federico II, Via Sergio Pansini 5, Naples, 80131, Italy.,CEINGE Biotecnologie Avanzate, Via Gaetano Salvatore 486, Naples, Italy
| | - Reza Maroofian
- Medical Research (Level 4), RILD Wellcome Wolfson Centre, University of Exeter Medical School, Royal Devon & Exeter NHS Foundation Trust, Barrack Road, Exeter, EX2 5DW, UK
| | - Ahmed Al-Amri
- Section of Ophthalmology and Neuroscience, Leeds Institute of Biomedical and Clinical Sciences, University of Leeds, UK.,National Genetic Centre, Directorate General of Royal Hospital, Ministry of Health, Muscat, Sultanate of Oman
| | - Royana Singh
- Molecular Genetics, Department of Anatomy, Institute of Medical Sciences, Banaras Hindu University, Varanasi -221005, UP, India
| | - Iolanda Scognamiglio
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche DMMBM, Università di Napoli Federico II, Via Sergio Pansini 5, Naples, 80131, Italy.,CEINGE Biotecnologie Avanzate, Via Gaetano Salvatore 486, Naples, Italy
| | - Majid Mojarrad
- Department of Medical Genetics, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Medical Genetics Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Luca Musella
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche DMMBM, Università di Napoli Federico II, Via Sergio Pansini 5, Naples, 80131, Italy.,CEINGE Biotecnologie Avanzate, Via Gaetano Salvatore 486, Naples, Italy
| | - Angela Duilio
- Dipartimento di Scienze Chimiche, Università Federico II, Naples, Italy
| | - Angela Di Somma
- Dipartimento di Scienze Chimiche, Università Federico II, Naples, Italy
| | - Ender Karaca
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Anna Rajab
- National Genetic Centre, Directorate General of Royal Hospital, Ministry of Health, Muscat, Sultanate of Oman
| | - Aisha Al-Khayat
- Department of Biology, Sultan Qaboos University, PO Box 36, Post code 123, Sultanate of Oman
| | - Tribhuvan Mohan Mohapatra
- Molecular Genetics, Department of Anatomy, Institute of Medical Sciences, Banaras Hindu University, Varanasi -221005, UP, India
| | - Atieh Eslahi
- Department of Medical Genetics, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farah Ashrafzadeh
- Department of Medical Genetics, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pediatric Neurology, Ghaem Medical Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Zip Code- 9919991766, Iran
| | - Lettie E Rawlins
- Medical Research (Level 4), RILD Wellcome Wolfson Centre, University of Exeter Medical School, Royal Devon & Exeter NHS Foundation Trust, Barrack Road, Exeter, EX2 5DW, UK
| | - Rajniti Prasad
- Department of Pediatrics, Institute of Medical Sciences, Banaras Hindu University, Varanasi -221005, UP, India
| | - Rashmi Gupta
- Molecular Genetics, Department of Anatomy, Institute of Medical Sciences, Banaras Hindu University, Varanasi -221005, UP, India
| | - Preeti Kumari
- Molecular Genetics, Department of Anatomy, Institute of Medical Sciences, Banaras Hindu University, Varanasi -221005, UP, India
| | - Mona Srivastava
- Molecular Genetics, Department of Anatomy, Institute of Medical Sciences, Banaras Hindu University, Varanasi -221005, UP, India.,Department of Psychiatry, Institute of Medical Sciences, Banaras Hindu University, Varanasi -221005, UP, India
| | - Flora Cozzolino
- CEINGE Biotecnologie Avanzate, Via Gaetano Salvatore 486, Naples, Italy
| | - Sunil Kumar Rai
- Molecular Genetics, Department of Anatomy, Institute of Medical Sciences, Banaras Hindu University, Varanasi -221005, UP, India
| | - Maria Monti
- CEINGE Biotecnologie Avanzate, Via Gaetano Salvatore 486, Naples, Italy.,Dipartimento di Scienze Chimiche, Università Federico II, Naples, Italy
| | - Gaurav V Harlalka
- Medical Research (Level 4), RILD Wellcome Wolfson Centre, University of Exeter Medical School, Royal Devon & Exeter NHS Foundation Trust, Barrack Road, Exeter, EX2 5DW, UK
| | - Michael A Simpson
- Department of Medical and Molecular Genetics, Division of Genetics and Molecular Medicine, King's College London, London, UK
| | - Philip Rich
- Department of Neuroradiology, St. George's Hospital, London, UK
| | - Fatema Al-Salmi
- Department of Biology, Sultan Qaboos University, PO Box 36, Post code 123, Sultanate of Oman
| | - Michael A Patton
- Medical Research (Level 4), RILD Wellcome Wolfson Centre, University of Exeter Medical School, Royal Devon & Exeter NHS Foundation Trust, Barrack Road, Exeter, EX2 5DW, UK.,Department of Biology, Sultan Qaboos University, PO Box 36, Post code 123, Sultanate of Oman.,Genetics Research Centre, St. George's, University of London, London, SW17 0RE, UK
| | - Barry A Chioza
- Medical Research (Level 4), RILD Wellcome Wolfson Centre, University of Exeter Medical School, Royal Devon & Exeter NHS Foundation Trust, Barrack Road, Exeter, EX2 5DW, UK
| | - Stephanie Efthymiou
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - Francesca Granata
- Unit of Neuroradiology, Department of Biomedical Science and Morphological and Functional Images, University of Messina, Messina, Italy
| | - Gabriella Di Rosa
- Unit of Child Neurology and Psychiatry, Department of Human Pathology of the Adult and Developmental Age, University of Messina, Messina, Italy
| | - Sarah Wiethoff
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - Eugenia Borgione
- Unit of Neuromuscular disorders, IRCCS Oasi Maria SS Troina, Enna, Italy
| | - Carmela Scuderi
- Unit of Neuromuscular disorders, IRCCS Oasi Maria SS Troina, Enna, Italy
| | - Kshitij Mankad
- Department of Neuroradiology, Great Ormond Street Hospital for Children, London WC1N 3JH, UK
| | - Michael G Hanna
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK.,MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology, London WC1N 3BG, UK
| | - Piero Pucci
- CEINGE Biotecnologie Avanzate, Via Gaetano Salvatore 486, Naples, Italy.,Dipartimento di Scienze Chimiche, Università Federico II, Naples, Italy
| | - Henry Houlden
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - James R Lupski
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA.,Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA.,Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA.,Texas Children's Hospital, Houston, TX 77030, USA
| | - Andrew H Crosby
- Medical Research (Level 4), RILD Wellcome Wolfson Centre, University of Exeter Medical School, Royal Devon & Exeter NHS Foundation Trust, Barrack Road, Exeter, EX2 5DW, UK
| | - Emma L Baple
- Medical Research (Level 4), RILD Wellcome Wolfson Centre, University of Exeter Medical School, Royal Devon & Exeter NHS Foundation Trust, Barrack Road, Exeter, EX2 5DW, UK
| |
Collapse
|
34
|
Chen Y, Xu Y, Li G, Li N, Yu T, Yao RE, Wang X, Shen Y, Wang J. Exome Sequencing Identifies De Novo DYNC1H1 Mutations Associated With Distal Spinal Muscular Atrophy and Malformations of Cortical Development. J Child Neurol 2017; 32:379-386. [PMID: 28193117 DOI: 10.1177/0883073816683083] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Exome sequencing has become a formidable tool for identifying potential de novo variants in causative genes of human diseases, such as neurodegenerative disorders. This article describes a 16-month-old girl with spinal muscular atrophy with lower extremity predominance and a 13-month-old girl with malformations of cortical development. Exome sequencing identified a novel de novo heterozygous missense mutation c.3395G>A (p.Gly1132Glu) and a previously reported de novo heterozygous missense mutation c.10151G>A (p.Arg3384Gln) in the DYNC1H1 gene. Bioinformatics predictions for c.3395G>A and c.10151G>A indicated pathogenicity of the mutations. DYNC1H1 is a pivotal component of cytoplasmic dynein complex, which is a microtubule-related motor involved in retrograde transport. Previous studies indicated that mutant dynein showed decreased run-length of the motor proteins and diminished retrograde transport, which were clearly associated with neuronal death and neurologic diseases. The present findings expand the mutational spectrum of the DYNC1H1 gene, reemphasizing the significance of the DYNC1H1 protein in the functioning of neurons.
Collapse
Affiliation(s)
- Yulin Chen
- 1 Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, PR China
| | - Yufei Xu
- 1 Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, PR China
| | - Guoqiang Li
- 1 Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, PR China
| | - Niu Li
- 1 Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, PR China
| | - Tingting Yu
- 1 Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, PR China
| | - Ru-En Yao
- 1 Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, PR China
| | - Xiumin Wang
- 1 Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, PR China
| | - Yiping Shen
- 1 Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, PR China
- 2 Department of Laboratory Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Jian Wang
- 1 Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, PR China
| |
Collapse
|
35
|
Mutations in the HECT domain of NEDD4L lead to AKT-mTOR pathway deregulation and cause periventricular nodular heterotopia. Nat Genet 2016; 48:1349-1358. [PMID: 27694961 PMCID: PMC5086093 DOI: 10.1038/ng.3676] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 08/24/2016] [Indexed: 12/16/2022]
Abstract
Neurodevelopmental disorders with periventricular nodular heterotopia (PNH) are etiologically heterogeneous, and their genetic causes remain in many cases unknown. Here we show that missense mutations in NEDD4L mapping to the HECT domain of the encoded E3 ubiquitin ligase lead to PNH associated with toe syndactyly, cleft palate and neurodevelopmental delay. Cellular and expression data showed sensitivity of PNH-associated mutants to proteasome degradation. Moreover, an in utero electroporation approach showed that PNH-related mutants and excess wild-type NEDD4L affect neurogenesis, neuronal positioning and terminal translocation. Further investigations, including rapamycin-based experiments, found differential deregulation of pathways involved. Excess wild-type NEDD4L leads to disruption of Dab1 and mTORC1 pathways, while PNH-related mutations are associated with deregulation of mTORC1 and AKT activities. Altogether, these data provide insights into the critical role of NEDD4L in the regulation of mTOR pathways and their contributions in cortical development.
Collapse
|
36
|
Clark JA, Yeaman EJ, Blizzard CA, Chuckowree JA, Dickson TC. A Case for Microtubule Vulnerability in Amyotrophic Lateral Sclerosis: Altered Dynamics During Disease. Front Cell Neurosci 2016; 10:204. [PMID: 27679561 PMCID: PMC5020100 DOI: 10.3389/fncel.2016.00204] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 08/15/2016] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is an aggressive multifactorial disease converging on a common pathology: the degeneration of motor neurons (MNs), their axons and neuromuscular synapses. This vulnerability and dysfunction of MNs highlights the dependency of these large cells on their intracellular machinery. Neuronal microtubules (MTs) are intracellular structures that facilitate a myriad of vital neuronal functions, including activity dependent axonal transport. In ALS, it is becoming increasingly apparent that MTs are likely to be a critical component of this disease. Not only are disruptions in this intracellular machinery present in the vast majority of seemingly sporadic cases, recent research has revealed that mutation to a microtubule protein, the tubulin isoform TUBA4A, is sufficient to cause a familial, albeit rare, form of disease. In both sporadic and familial disease, studies have provided evidence that microtubule mediated deficits in axonal transport are the tipping point for MN survivability. Axonal transport deficits would lead to abnormal mitochondrial recycling, decreased vesicle and mRNA transport and limited signaling of key survival factors from the neurons peripheral synapses, causing the characteristic peripheral "die back". This disruption to microtubule dependant transport in ALS has been shown to result from alterations in the phenomenon of microtubule dynamic instability: the rapid growth and shrinkage of microtubule polymers. This is accomplished primarily due to aberrant alterations to microtubule associated proteins (MAPs) that regulate microtubule stability. Indeed, the current literature would argue that microtubule stability, particularly alterations in their dynamics, may be the initial driving force behind many familial and sporadic insults in ALS. Pharmacological stabilization of the microtubule network offers an attractive therapeutic strategy in ALS; indeed it has shown promise in many neurological disorders, ALS included. However, the pathophysiological involvement of MTs and their functions is still poorly understood in ALS. Future investigations will hopefully uncover further therapeutic targets that may aid in combating this awful disease.
Collapse
Affiliation(s)
- Jayden A Clark
- Menzies Institute for Medical Research, University of Tasmania Hobart, TAS, Australia
| | - Elise J Yeaman
- Menzies Institute for Medical Research, University of Tasmania Hobart, TAS, Australia
| | - Catherine A Blizzard
- Menzies Institute for Medical Research, University of Tasmania Hobart, TAS, Australia
| | - Jyoti A Chuckowree
- Menzies Institute for Medical Research, University of Tasmania Hobart, TAS, Australia
| | - Tracey C Dickson
- Menzies Institute for Medical Research, University of Tasmania Hobart, TAS, Australia
| |
Collapse
|
37
|
Abstract
The most striking structure in the nervous system is the complex yet stereotyped morphology of the neuronal dendritic tree. Dendritic morphologies and the connections they make govern information flow and integration in the brain. The fundamental mechanisms that regulate dendritic outgrowth and branching are subjects of extensive study. In this review, we summarize recent advances in the molecular and cellular mechanisms for routing dendrites in layers and columns, prevalent organizational structures in the brain. We highlight how dendritic patterning influences the formation of synaptic circuits.
Collapse
Affiliation(s)
- Jiangnan Luo
- a Section on Neuronal Connectivity, Cell Biology and Neurobiology Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development , Bethesda , MD , USA
| | - Philip G McQueen
- b Mathematical and Statistical Computing Laboratory, Office of Intramural Research, Center for Information Technology , National Institutes of Health , Bethesda , MD , USA
| | - Bo Shi
- a Section on Neuronal Connectivity, Cell Biology and Neurobiology Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development , Bethesda , MD , USA ;,c Biological Sciences Graduate Program, College of Computer, Mathematical, and Natural Sciences , University of Maryland , College Park , MD , USA
| | - Chi-Hon Lee
- a Section on Neuronal Connectivity, Cell Biology and Neurobiology Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development , Bethesda , MD , USA
| | - Chun-Yuan Ting
- a Section on Neuronal Connectivity, Cell Biology and Neurobiology Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development , Bethesda , MD , USA
| |
Collapse
|
38
|
Tetramethylpyrazine Promotes Migration of Neural Precursor Cells via Activating the Phosphatidylinositol 3-Kinase Pathway. Mol Neurobiol 2015; 53:6526-6539. [DOI: 10.1007/s12035-015-9551-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 11/18/2015] [Indexed: 12/31/2022]
|
39
|
Wagener RJ, Witte M, Guy J, Mingo-Moreno N, Kügler S, Staiger JF. Thalamocortical Connections Drive Intracortical Activation of Functional Columns in the Mislaminated Reeler Somatosensory Cortex. Cereb Cortex 2015; 26:820-37. [PMID: 26564256 PMCID: PMC4712806 DOI: 10.1093/cercor/bhv257] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Neuronal wiring is key to proper neural information processing. Tactile information from the rodent's whiskers reaches the cortex via distinct anatomical pathways. The lemniscal pathway relays whisking and touch information from the ventral posteromedial thalamic nucleus to layer IV of the primary somatosensory “barrel” cortex. The disorganized neocortex of the reeler mouse is a model system that should severely compromise the ingrowth of thalamocortical axons (TCAs) into the cortex. Moreover, it could disrupt intracortical wiring. We found that neuronal intermingling within the reeler barrel cortex substantially exceeded previous descriptions, leading to the loss of layers. However, viral tracing revealed that TCAs still specifically targeted transgenically labeled spiny layer IV neurons. Slice electrophysiology and optogenetics proved that these connections represent functional synapses. In addition, we assessed intracortical activation via immediate-early-gene expression resulting from a behavioral exploration task. The cellular composition of activated neuronal ensembles suggests extensive similarities in intracolumnar information processing in the wild-type and reeler brains. We conclude that extensive ectopic positioning of neuronal partners can be compensated for by cell-autonomous mechanisms that allow for the establishment of proper connectivity. Thus, genetic neuronal fate seems to be of greater importance for correct cortical wiring than radial neuronal position.
Collapse
Affiliation(s)
- Robin J Wagener
- Institute for Neuroanatomy, Universitätsmedizin Göttingen, Georg-August-Universität Göttingen, 37075 Göttingen, Germany
| | - Mirko Witte
- Institute for Neuroanatomy, Universitätsmedizin Göttingen, Georg-August-Universität Göttingen, 37075 Göttingen, Germany
| | - Julien Guy
- Institute for Neuroanatomy, Universitätsmedizin Göttingen, Georg-August-Universität Göttingen, 37075 Göttingen, Germany
| | - Nieves Mingo-Moreno
- Institute for Neuroanatomy, Universitätsmedizin Göttingen, Georg-August-Universität Göttingen, 37075 Göttingen, Germany Center Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
| | - Sebastian Kügler
- Center Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany Department of Neurology, Universitätsmedizin Göttingen, Georg-August-Universität Göttingen, 37075 Göttingen, Germany
| | - Jochen F Staiger
- Institute for Neuroanatomy, Universitätsmedizin Göttingen, Georg-August-Universität Göttingen, 37075 Göttingen, Germany Center Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
| |
Collapse
|
40
|
Pathophysiological analyses of cortical malformation using gyrencephalic mammals. Sci Rep 2015; 5:15370. [PMID: 26482531 PMCID: PMC4613358 DOI: 10.1038/srep15370] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 09/23/2015] [Indexed: 12/12/2022] Open
Abstract
One of the most prominent features of the cerebral cortex of higher mammals is the presence of gyri. Because malformations of the cortical gyri are associated with severe disability in brain function, the mechanisms underlying malformations of the cortical gyri have been of great interest. Combining gyrencephalic carnivore ferrets and genetic manipulations using in utero electroporation, here we successfully recapitulated the cortical phenotypes of thanatophoric dysplasia (TD) by expressing fibroblast growth factor 8 in the ferret cerebral cortex. Strikingly, in contrast to TD mice, our TD ferret model showed not only megalencephaly but also polymicrogyria. We further uncovered that outer radial glial cells (oRGs) and intermediate progenitor cells (IPs) were markedly increased. Because it has been proposed that increased oRGs and/or IPs resulted in the appearance of cortical gyri during evolution, it seemed possible that increased oRGs and IPs underlie the pathogenesis of polymicrogyria. Our findings should help shed light on the molecular mechanisms underlying the formation and malformation of cortical gyri in higher mammals.
Collapse
|
41
|
Kawauchi T. Cellullar insights into cerebral cortical development: focusing on the locomotion mode of neuronal migration. Front Cell Neurosci 2015; 9:394. [PMID: 26500496 PMCID: PMC4595654 DOI: 10.3389/fncel.2015.00394] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 09/22/2015] [Indexed: 02/01/2023] Open
Abstract
The mammalian brain consists of numerous compartments that are closely connected with each other via neural networks, comprising the basis of higher order brain functions. The highly specialized structure originates from simple pseudostratified neuroepithelium-derived neural progenitors located near the ventricle. A long journey by neurons from the ventricular side is essential for the formation of a sophisticated brain structure, including a mammalian-specific six-layered cerebral cortex. Neuronal migration consists of several contiguous steps, but the locomotion mode comprises a large part of the migration. The locomoting neurons exhibit unique features; a radial glial fiber-dependent migration requiring the endocytic recycling of N-cadherin and a neuron-specific migration mode with dilation/swelling formation that requires the actin and microtubule organization possibly regulated by cyclin-dependent kinase 5 (Cdk5), Dcx, p27(kip1), Rac1, and POSH. Here I will introduce the roles of various cellular events, such as cytoskeletal organization, cell adhesion, and membrane trafficking, in the regulation of the neuronal migration, with particular focus on the locomotion mode.
Collapse
Affiliation(s)
- Takeshi Kawauchi
- Department of Physiology, Keio University School of Medicine Tokyo, Japan ; Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science and Technology Agency Saitama, Japan ; Laboratory of Molecular Life Science, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation Kobe, Japan
| |
Collapse
|
42
|
CHCHD2 is down-regulated in neuronal cells differentiated from iPS cells derived from patients with lissencephaly. Genomics 2015; 106:196-203. [DOI: 10.1016/j.ygeno.2015.07.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 06/17/2015] [Accepted: 07/01/2015] [Indexed: 12/20/2022]
|
43
|
Stouffer MA, Golden JA, Francis F. Neuronal migration disorders: Focus on the cytoskeleton and epilepsy. Neurobiol Dis 2015; 92:18-45. [PMID: 26299390 DOI: 10.1016/j.nbd.2015.08.003] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 08/05/2015] [Accepted: 08/12/2015] [Indexed: 01/28/2023] Open
Abstract
A wide spectrum of focal, regional, or diffuse structural brain abnormalities, collectively known as malformations of cortical development (MCDs), frequently manifest with intellectual disability (ID), epilepsy, and/or autistic spectrum disorder (ASD). As the acronym suggests, MCDs are perturbations of the normal architecture of the cerebral cortex and hippocampus. The pathogenesis of these disorders remains incompletely understood; however, one area that has provided important insights has been the study of neuronal migration. The amalgamation of human genetics and experimental studies in animal models has led to the recognition that common genetic causes of neurodevelopmental disorders, including many severe epilepsy syndromes, are due to mutations in genes regulating the migration of newly born post-mitotic neurons. Neuronal migration genes often, though not exclusively, code for proteins involved in the function of the cytoskeleton. Other cellular processes, such as cell division and axon/dendrite formation, which similarly depend on cytoskeletal functions, may also be affected. We focus here on how the susceptibility of the highly organized neocortex and hippocampus may be due to their laminar organization, which involves the tight regulation, both temporally and spatially, of gene expression, specialized progenitor cells, the migration of neurons over large distances and a birthdate-specific layering of neurons. Perturbations in neuronal migration result in abnormal lamination, neuronal differentiation defects, abnormal cellular morphology and circuit formation. Ultimately this results in disorganized excitatory and inhibitory activity leading to the symptoms observed in individuals with these disorders.
Collapse
Affiliation(s)
- Melissa A Stouffer
- INSERM UMRS 839, Paris, France; Sorbonne Universités, Université Pierre et Marie Curie, Paris, France; Institut du Fer à Moulin, Paris, France
| | - Jeffrey A Golden
- Department of Pathology, Brigham & Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Fiona Francis
- INSERM UMRS 839, Paris, France; Sorbonne Universités, Université Pierre et Marie Curie, Paris, France; Institut du Fer à Moulin, Paris, France.
| |
Collapse
|
44
|
Akhtar AA, Breunig JJ. Lost highway(s): barriers to postnatal cortical neurogenesis and implications for brain repair. Front Cell Neurosci 2015; 9:216. [PMID: 26136658 PMCID: PMC4468390 DOI: 10.3389/fncel.2015.00216] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 05/21/2015] [Indexed: 11/13/2022] Open
Abstract
The genesis of the cerebral cortex is a highly complex and tightly-orchestrated process of cell division, migration, maturation, and integration. Developmental missteps often have catastrophic consequences on cortical function. Further, the cerebral cortex, in which neurogenesis takes place almost exclusively prenatally, has a very poor capacity for replacement of neurons lost to injury or disease. A multitude of factors underlie this deficit, including the depletion of radial glia, the gliogenic switch which mitigates continued neurogenesis, diminished neuronal migratory streams, and inflammatory processes associated with disease. Despite this, there are glimmers of hope that new approaches may allow for more significant cortical repair. Herein, we review corticogenesis from the context of regeneration and detail the strategies to promote neurogenesis, including interneuron transplants and glial reprogramming. Such strategies circumvent the "lost highways" which are critical for cortical development but are absent in the adult. These new approaches may provide for the possibility of meaningful clinical regeneration of elements of cortical circuitry lost to trauma and disease.
Collapse
Affiliation(s)
- Aslam Abbasi Akhtar
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center Los Angeles, CA, USA ; Department of Biomedical Sciences, Cedars-Sinai Medical Center Los Angeles, CA, USA
| | - Joshua J Breunig
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center Los Angeles, CA, USA ; Department of Biomedical Sciences, Cedars-Sinai Medical Center Los Angeles, CA, USA ; Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center Los Angeles, CA, USA
| |
Collapse
|
45
|
El Fatimy R, Miozzo F, Le Mouël A, Abane R, Schwendimann L, Sabéran-Djoneidi D, de Thonel A, Massaoudi I, Paslaru L, Hashimoto-Torii K, Christians E, Rakic P, Gressens P, Mezger V. Heat shock factor 2 is a stress-responsive mediator of neuronal migration defects in models of fetal alcohol syndrome. EMBO Mol Med 2015; 6:1043-61. [PMID: 25027850 PMCID: PMC4154132 DOI: 10.15252/emmm.201303311] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Fetal alcohol spectrum disorder (FASD) is a frequent cause of mental retardation. However, the molecular mechanisms underlying brain development defects induced by maternal alcohol consumption during pregnancy are unclear. We used normal and Hsf2-deficient mice and cell systems to uncover a pivotal role for heat shock factor 2 (HSF2) in radial neuronal migration defects in the cortex, a hallmark of fetal alcohol exposure. Upon fetal alcohol exposure, HSF2 is essential for the triggering of HSF1 activation, which is accompanied by distinctive post-translational modifications, and HSF2 steers the formation of atypical alcohol-specific HSF1-HSF2 heterocomplexes. This perturbs the in vivo binding of HSF2 to heat shock elements (HSEs) in genes that control neuronal migration in normal conditions, such as p35 or the MAPs (microtubule-associated proteins, such as Dclk1 and Dcx), and alters their expression. In the absence of HSF2, migration defects as well as alterations in gene expression are reduced. Thus, HSF2, as a sensor for alcohol stress in the fetal brain, acts as a mediator of the neuronal migration defects associated with FASD.
Collapse
Affiliation(s)
- Rachid El Fatimy
- CNRS UMR7216 Épigénétique et Destin Cellulaire, Paris Cedex 13, France Univ Paris Diderot Sorbonne Paris Cité, Paris Cedex 13, France ED 387 iViv UPMC Univ Paris 06, Paris, France Univ Paris Diderot, Paris Cedex 13, France
| | - Federico Miozzo
- CNRS UMR7216 Épigénétique et Destin Cellulaire, Paris Cedex 13, France Univ Paris Diderot Sorbonne Paris Cité, Paris Cedex 13, France ED 387 iViv UPMC Univ Paris 06, Paris, France Univ Paris Diderot, Paris Cedex 13, France
| | - Anne Le Mouël
- CNRS UMR7216 Épigénétique et Destin Cellulaire, Paris Cedex 13, France Univ Paris Diderot Sorbonne Paris Cité, Paris Cedex 13, France
| | - Ryma Abane
- CNRS UMR7216 Épigénétique et Destin Cellulaire, Paris Cedex 13, France Univ Paris Diderot Sorbonne Paris Cité, Paris Cedex 13, France ED 387 iViv UPMC Univ Paris 06, Paris, France Univ Paris Diderot, Paris Cedex 13, France
| | - Leslie Schwendimann
- INSERM U1141, Hôpital Robert Debré, Paris, France Faculté de Médecine Denis Diderot, Univ Paris Diderot Sorbonne Paris Cité, Paris, France
| | - Délara Sabéran-Djoneidi
- CNRS UMR7216 Épigénétique et Destin Cellulaire, Paris Cedex 13, France Univ Paris Diderot Sorbonne Paris Cité, Paris Cedex 13, France
| | - Aurélie de Thonel
- INSERM UMR 866, Dijon, France Faculty of Medicine and Pharmacy, Univ Burgundy, Dijon, France
| | - Illiasse Massaoudi
- CNRS UMR7216 Épigénétique et Destin Cellulaire, Paris Cedex 13, France Univ Paris Diderot Sorbonne Paris Cité, Paris Cedex 13, France
| | - Liliana Paslaru
- Carol Davila University of Medicine and Pharmacy Fundeni Hospital, Bucharest, Romania
| | - Kazue Hashimoto-Torii
- Department of Neurobiology and Kavli Institute for Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| | - Elisabeth Christians
- Laboratoire de Biologie du Développement de Villefranche-sur-mer, Observatoire Océanologique, CNRS, Villefranche-sur-mer, France Sorbonne Universités UPMC Univ Paris 06, Villefranche-sur-mer, France
| | - Pasko Rakic
- Department of Neurobiology and Kavli Institute for Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| | - Pierre Gressens
- INSERM U1141, Hôpital Robert Debré, Paris, France Faculté de Médecine Denis Diderot, Univ Paris Diderot Sorbonne Paris Cité, Paris, France
| | - Valérie Mezger
- CNRS UMR7216 Épigénétique et Destin Cellulaire, Paris Cedex 13, France Univ Paris Diderot Sorbonne Paris Cité, Paris Cedex 13, France
| |
Collapse
|
46
|
Involvement of GSK3 in the formation of the leading process and migration of neurons from the embryonic rat medial ganglionic eminence in vitro. Neuroreport 2015; 26:179-85. [PMID: 25646586 DOI: 10.1097/wnr.0000000000000333] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Migrating neurons have leading processes that direct cell movement in response to guidance cues. We investigated the involvement of glycogen synthase kinase 3 (GSK3) in the formation of leading processes and migration of neurons in vitro. We used embryonic rat medial ganglionic eminence (MGE) neurons, which are precursors of inhibitory neurons that migrate into the cerebral cortex. When MGE neurons were placed on an astrocyte layer, they migrated freely with the highest speed among neurons from other parts of the embryonic forebrain. When they were cultured alone, they showed bipolar morphology and extended leading processes within 20 h. Their leading processes had large growth cones, but did not elongate during 3 days in culture, indicating that leading processes are distinct from short axons. Next, we examined the effect of GSK3 inhibitors on leading processes and the migratory behavior of MGE neurons. MGE neurons treated with GSK3 inhibitors showed multipolar morphology and altered process shapes. Moreover, migration of MGE neurons on the astrocyte layer was significantly decreased in the presence of GSK3 inhibitors. These data suggest that GSK3 is involved in the formation of leading processes and in the migration of MGE neurons.
Collapse
|
47
|
Bizzotto S, Francis F. Morphological and functional aspects of progenitors perturbed in cortical malformations. Front Cell Neurosci 2015; 9:30. [PMID: 25729350 PMCID: PMC4325918 DOI: 10.3389/fncel.2015.00030] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Accepted: 01/18/2015] [Indexed: 11/13/2022] Open
Abstract
In this review, we discuss molecular and cellular mechanisms important for the function of neuronal progenitors during development, revealed by their perturbation in different cortical malformations. We focus on a class of neuronal progenitors, radial glial cells (RGCs), which are renowned for their unique morphological and behavioral characteristics, constituting a key element during the development of the mammalian cerebral cortex. We describe how the particular morphology of these cells is related to their roles in the orchestration of cortical development and their influence on other progenitor types and post-mitotic neurons. Important for disease mechanisms, we overview what is currently known about RGC cellular components, cytoskeletal mechanisms, signaling pathways and cell cycle characteristics, focusing on how defects lead to abnormal development and cortical malformation phenotypes. The multiple recent entry points from human genetics and animal models are contributing to our understanding of this important cell type. Combining data from phenotypes in the mouse reveals molecules which potentially act in common pathways. Going beyond this, we discuss future directions that may provide new data in this expanding area.
Collapse
Affiliation(s)
- Sara Bizzotto
- INSERM UMRS 839 Paris, France ; Sorbonne Universités, Université Pierre et Marie Curie Paris, France ; Institut du Fer à Moulin Paris, France
| | - Fiona Francis
- INSERM UMRS 839 Paris, France ; Sorbonne Universités, Université Pierre et Marie Curie Paris, France ; Institut du Fer à Moulin Paris, France
| |
Collapse
|
48
|
Kielar M, Phan Dinh Tuy F, Bizzotto S, Belvindrah R, Croquelois A, Francis F. [Mutations in Eml1/EML1 lead to ectopic progenitors and neuronal heterotopia in mouse and human]. Med Sci (Paris) 2014; 30:1087-90. [PMID: 25537038 DOI: 10.1051/medsci/20143012011] [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)
- Michel Kielar
- Department of clinical neuroscience, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Suisse - Department of fundamental neuroscience, University of Lausanne, 1005 Lausanne, Suisse
| | - Françoise Phan Dinh Tuy
- Inserm UMR-S 839, 75005 Paris, France - Sorbonne Universités, Université Pierre et Marie Curie, 75005 Paris, France - Institut du Fer à Moulin, 17, rue du Fer à Moulin 75005 Paris, France
| | - Sara Bizzotto
- Inserm UMR-S 839, 75005 Paris, France - Sorbonne Universités, Université Pierre et Marie Curie, 75005 Paris, France - Institut du Fer à Moulin, 17, rue du Fer à Moulin 75005 Paris, France
| | - Richard Belvindrah
- Inserm UMR-S 839, 75005 Paris, France - Sorbonne Universités, Université Pierre et Marie Curie, 75005 Paris, France - Institut du Fer à Moulin, 17, rue du Fer à Moulin 75005 Paris, France
| | - Alexandre Croquelois
- Department of clinical neuroscience, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Suisse - Department of fundamental neuroscience, University of Lausanne, 1005 Lausanne, Suisse
| | - Fiona Francis
- Inserm UMR-S 839, 75005 Paris, France - Sorbonne Universités, Université Pierre et Marie Curie, 75005 Paris, France - Institut du Fer à Moulin, 17, rue du Fer à Moulin 75005 Paris, France
| |
Collapse
|
49
|
Fallet-Bianco C, Laquerrière A, Poirier K, Razavi F, Guimiot F, Dias P, Loeuillet L, Lascelles K, Beldjord C, Carion N, Toussaint A, Revencu N, Addor MC, Lhermitte B, Gonzales M, Martinovich J, Bessieres B, Marcy-Bonnière M, Jossic F, Marcorelles P, Loget P, Chelly J, Bahi-Buisson N. Mutations in tubulin genes are frequent causes of various foetal malformations of cortical development including microlissencephaly. Acta Neuropathol Commun 2014; 2:69. [PMID: 25059107 PMCID: PMC4222268 DOI: 10.1186/2051-5960-2-69] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 06/04/2014] [Indexed: 01/18/2023] Open
Abstract
Complex cortical malformations associated with mutations in tubulin genes are commonly referred to as “Tubulinopathies”. To further characterize the mutation frequency and phenotypes associated with tubulin mutations, we studied a cohort of 60 foetal cases. Twenty-six tubulin mutations were identified, of which TUBA1A mutations were the most prevalent (19 cases), followed by TUBB2B (6 cases) and TUBB3 (one case). Three subtypes clearly emerged. The most frequent (n = 13) was microlissencephaly with corpus callosum agenesis, severely hypoplastic brainstem and cerebellum. The cortical plate was either absent (6/13), with a 2–3 layered pattern (5/13) or less frequently thickened (2/13), often associated with neuroglial overmigration (4/13). All cases had voluminous germinal zones and ganglionic eminences. The second subtype was lissencephaly (n = 7), either classical (4/7) or associated with cerebellar hypoplasia (3/7) with corpus callosum agenesis (6/7). All foetuses with lissencephaly and cerebellar hypoplasia carried distinct TUBA1A mutations, while those with classical lissencephaly harbored recurrent mutations in TUBA1A (3 cases) or TUBB2B (1 case). The third group was polymicrogyria-like cortical dysplasia (n = 6), consisting of asymmetric multifocal or generalized polymicrogyria with inconstant corpus callosum agenesis (4/6) and hypoplastic brainstem and cerebellum (3/6). Polymicrogyria was either unlayered or 4-layered with neuronal heterotopias (5/6) and occasional focal neuroglial overmigration (2/6). Three had TUBA1A mutations and 3 TUBB2B mutations. Foetal TUBA1A tubulinopathies most often consist in microlissencephaly or classical lissencephaly with corpus callosum agenesis, but polymicrogyria may also occur. Conversely, TUBB2B mutations are responsible for either polymicrogyria (4/6) or microlissencephaly (2/6).
Collapse
|
50
|
|