51
|
Anstötz M, Lee SK, Maccaferri G. Expression of TRPV1 channels by Cajal-Retzius cells and layer-specific modulation of synaptic transmission by capsaicin in the mouse hippocampus. J Physiol 2018; 596:3739-3758. [PMID: 29806907 DOI: 10.1113/jp275685] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 05/16/2018] [Indexed: 12/27/2022] Open
Abstract
KEY POINTS By taking advantage of calcium imaging and electrophysiology, we provide direct pharmacological evidence for the functional expression of TRPV1 channels in hippocampal Cajal-Retzius cells. Application of the TRPV1 activator capsaicin powerfully enhances spontaneous synaptic transmission in the hippocampal layers that are innervated by the axons of Cajal-Retzius cells. Capsaicin-triggered calcium responses and membrane currents in Cajal-Retzius cells, as well as layer-specific modulation of spontaneous synaptic transmission, are absent when the drug is applied to slices prepared from TRPV1- /- animals. We discuss the implications of the functional expression of TRPV1 channels in Cajal-Retzius cells and of the observed TRPV1-dependent layer-specific modulation of synaptic transmission for physiological and pathological network processing. ABSTRACT The vanilloid receptor TRPV1 forms complex polymodal channels that are expressed by sensory neurons and play a critical role in nociception. Their distribution pattern and functions in cortical circuits are, however, much less understood. Although TRPV1 reporter mice have suggested that, in the hippocampus, TRPV1 is predominantly expressed by Cajal-Retzius cells (CRs), direct functional evidence is missing. As CRs powerfully excite GABAergic interneurons of the molecular layers, TRPV1 could play important roles in the regulation of layer-specific processing. Here, we have taken advantage of calcium imaging with the genetically encoded indicator GCaMP6s and patch-clamp techniques to study the responses of hippocampal CRs to the activation of TRPV1 by capsaicin, and have compared the effect of TRPV1 stimulation on synaptic transmission in layers innervated or non-innervated by CRs. Capsaicin induced both calcium responses and membrane currents in ∼50% of the cell tested. Neither increases of intracellular calcium nor whole-cell currents were observed in the presence of the TRPV1 antagonists capsazepine/Ruthenium Red or in slices prepared from TRPV1 knockout mice. We also report a powerful TRPV1-dependent enhancement of spontaneous synaptic transmission onto interneurons with dendritic trees confined to the layers innervated by CRs. In conclusion, our work establishes that functional TRPV1 is expressed by a significant fraction of CRs and we propose that TRPV1 activity may regulate layer-specific synaptic transmission in the hippocampus. Lastly, as CR density decreases during postnatal development, we also propose that functional TRPV1 receptors may be related to mechanisms involved in CR progressive reduction by calcium-dependent toxicity/apoptosis.
Collapse
Affiliation(s)
- Max Anstötz
- Department of Physiology, Northwestern University, Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL, 60611-3008, USA.,Institute for Neuroanatomy, University/University Hospital Hamburg, Martinistr. 52, 20246, Hamburg, Germany
| | - Sun Kyong Lee
- Department of Physiology, Northwestern University, Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL, 60611-3008, USA
| | - Gianmaria Maccaferri
- Department of Physiology, Northwestern University, Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL, 60611-3008, USA
| |
Collapse
|
52
|
Shiotani H, Miyata M, Itoh Y, Wang S, Kaito A, Mizoguchi A, Yamasaki M, Watanabe M, Mandai K, Mochizuki H, Takai Y. Localization of nectin-2α at the boundary between the adjacent somata of the clustered cholinergic neurons and its regulatory role in the subcellular localization of the voltage-gated A-type K+channel Kv4.2 in the medial habenula. J Comp Neurol 2018. [DOI: 10.1002/cne.24425] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Hajime Shiotani
- Division of Pathogenetic Signaling, Department of Biochemistry and Molecular Biology; Kobe University Graduate School of Medicine; Kobe Hyogo 650-0047 Japan
- Department of Neurology; Osaka University Graduate School of Medicine; Suita Osaka 565-0871 Japan
| | - Muneaki Miyata
- Division of Pathogenetic Signaling, Department of Biochemistry and Molecular Biology; Kobe University Graduate School of Medicine; Kobe Hyogo 650-0047 Japan
| | - Yu Itoh
- Division of Pathogenetic Signaling, Department of Biochemistry and Molecular Biology; Kobe University Graduate School of Medicine; Kobe Hyogo 650-0047 Japan
| | - Shujie Wang
- Department of Neural Regeneration and Cell Communication; Mie University Graduate School of Medicine; Tsu Mie 514-8507 Japan
| | - Aika Kaito
- Department of Neural Regeneration and Cell Communication; Mie University Graduate School of Medicine; Tsu Mie 514-8507 Japan
| | - Akira Mizoguchi
- Department of Neural Regeneration and Cell Communication; Mie University Graduate School of Medicine; Tsu Mie 514-8507 Japan
| | - Miwako Yamasaki
- Department of Anatomy, Faculty of Medicine; Hokkaido University; Sapporo Hokkaido 060-8638 Japan
| | - Masahiko Watanabe
- Department of Anatomy, Faculty of Medicine; Hokkaido University; Sapporo Hokkaido 060-8638 Japan
| | - Kenji Mandai
- Division of Pathogenetic Signaling, Department of Biochemistry and Molecular Biology; Kobe University Graduate School of Medicine; Kobe Hyogo 650-0047 Japan
| | - Hideki Mochizuki
- Department of Neurology; Osaka University Graduate School of Medicine; Suita Osaka 565-0871 Japan
| | - Yoshimi Takai
- Division of Pathogenetic Signaling, Department of Biochemistry and Molecular Biology; Kobe University Graduate School of Medicine; Kobe Hyogo 650-0047 Japan
| |
Collapse
|
53
|
Dewitz C, Pimpinella S, Hackel P, Akalin A, Jessell TM, Zampieri N. Nuclear Organization in the Spinal Cord Depends on Motor Neuron Lamination Orchestrated by Catenin and Afadin Function. Cell Rep 2018; 22:1681-1694. [DOI: 10.1016/j.celrep.2018.01.059] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Revised: 12/19/2017] [Accepted: 01/18/2018] [Indexed: 01/08/2023] Open
|
54
|
Abstract
The formation of the nervous system is a multistep process that yields a mature brain. Failure in any of the steps of this process may cause brain malfunction. In the early stages of embryonic development, neural progenitors quickly proliferate and then, at a specific moment, differentiate into neurons or glia. Once they become postmitotic neurons, they migrate to their final destinations and begin to extend their axons to connect with other neurons, sometimes located in quite distant regions, to establish different neural circuits. During the last decade, it has become evident that Zic genes, in addition to playing important roles in early development (e.g., gastrulation and neural tube closure), are involved in different processes of late brain development, such as neuronal migration, axon guidance, and refinement of axon terminals. ZIC proteins are therefore essential for the proper wiring and connectivity of the brain. In this chapter, we review our current knowledge of the role of Zic genes in the late stages of neural circuit formation.
Collapse
|
55
|
Adnani L, Han S, Li S, Mattar P, Schuurmans C. Mechanisms of Cortical Differentiation. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 336:223-320. [DOI: 10.1016/bs.ircmb.2017.07.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
56
|
Ruiz-Reig N, Studer M. Rostro-Caudal and Caudo-Rostral Migrations in the Telencephalon: Going Forward or Backward? Front Neurosci 2017; 11:692. [PMID: 29311773 PMCID: PMC5742585 DOI: 10.3389/fnins.2017.00692] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 11/23/2017] [Indexed: 11/13/2022] Open
Abstract
The generation and differentiation of an appropriate number of neurons, as well as its distribution in different parts of the brain, is crucial for the proper establishment, maintenance and plasticity of neural circuitries. Newborn neurons travel along the brain in a process known as neuronal migration, to finalize their correct position in the nervous system. Defects in neuronal migration produce abnormalities in the brain that can generate neurodevelopmental pathologies, such as autism, schizophrenia and intellectual disability. In this review, we present an overview of the developmental origin of the different telencephalic subdivisions and a description of migratory pathways taken by distinct neural populations traveling long distances before reaching their target position in the brain. In addition, we discuss some of the molecules implicated in the guidance of these migratory paths and transcription factors that contribute to the correct migration and integration of these neurons.
Collapse
|
57
|
The functions of Reelin in membrane trafficking and cytoskeletal dynamics: implications for neuronal migration, polarization and differentiation. Biochem J 2017; 474:3137-3165. [PMID: 28887403 DOI: 10.1042/bcj20160628] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 07/27/2017] [Accepted: 08/01/2017] [Indexed: 02/06/2023]
Abstract
Reelin is a large extracellular matrix protein with relevant roles in mammalian central nervous system including neurogenesis, neuronal polarization and migration during development; and synaptic plasticity with its implications in learning and memory, in the adult. Dysfunctions in reelin signaling are associated with brain lamination defects such as lissencephaly, but also with neuropsychiatric diseases like autism, schizophrenia and depression as well with neurodegeneration. Reelin signaling involves a core pathway that activates upon reelin binding to its receptors, particularly ApoER2 (apolipoprotein E receptor 2)/LRP8 (low-density lipoprotein receptor-related protein 8) and very low-density lipoprotein receptor, followed by Src/Fyn-mediated phosphorylation of the adaptor protein Dab1 (Disabled-1). Phosphorylated Dab1 (pDab1) is a hub in the signaling cascade, from which several other downstream pathways diverge reflecting the different roles of reelin. Many of these pathways affect the dynamics of the actin and microtubular cytoskeleton, as well as membrane trafficking through the regulation of the activity of small GTPases, including the Rho and Rap families and molecules involved in cell polarity. The complexity of reelin functions is reflected by the fact that, even now, the precise mode of action of this signaling cascade in vivo at the cellular and molecular levels remains unclear. This review addresses and discusses in detail the participation of reelin in the processes underlying neurogenesis, neuronal migration in the cerebral cortex and the hippocampus; and the polarization, differentiation and maturation processes that neurons experiment in order to be functional in the adult brain. In vivo and in vitro evidence is presented in order to facilitate a better understanding of this fascinating system.
Collapse
|
58
|
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: 3.8] [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
|
59
|
Shah B, Püschel AW. Regulation of Rap GTPases in mammalian neurons. Biol Chem 2017; 397:1055-69. [PMID: 27186679 DOI: 10.1515/hsz-2016-0165] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 05/06/2016] [Indexed: 12/15/2022]
Abstract
Small GTPases are central regulators of many cellular processes. The highly conserved Rap GTPases perform essential functions in the mammalian nervous system during development and in mature neurons. During neocortical development, Rap1 is required to regulate cadherin- and integrin-mediated adhesion. In the adult nervous system Rap1 and Rap2 regulate the maturation and plasticity of dendritic spine and synapses. Although genetic studies have revealed important roles of Rap GTPases in neurons, their regulation by guanine nucleotide exchange factors (GEFs) that activate them and GTPase activating proteins (GAPs) that inactivate them by stimulating their intrinsic GTPase activity is just beginning to be explored in vivo. Here we review how GEFs and GAPs regulate Rap GTPases in the nervous system with a focus on their in vivo function.
Collapse
|
60
|
Hirota Y, Nakajima K. Control of Neuronal Migration and Aggregation by Reelin Signaling in the Developing Cerebral Cortex. Front Cell Dev Biol 2017; 5:40. [PMID: 28507985 PMCID: PMC5410752 DOI: 10.3389/fcell.2017.00040] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 03/31/2017] [Indexed: 11/13/2022] Open
Abstract
The mammalian cerebral neocortex has a well-organized laminar structure, achieved by the highly coordinated control of neuronal migration. During cortical development, excitatory neurons born near the lateral ventricle migrate radially to reach their final positions to form the cortical plate. During this process, dynamic changes are observed in the morphologies and migration modes, including multipolar migration, locomotion, and terminal translocation, of the newborn neurons. Disruption of these migration processes can result in neuronal disorders such as lissencephaly and periventricular heterotopia. The extracellular protein, Reelin, mainly secreted by the Cajal-Retzius neurons in the marginal zone during development, plays a crucial role in the neuronal migration and neocortical lamination. Reelin signaling, which exerts essential roles in the formation of the layered neocortex, is triggered by the binding of Reelin to its receptors, ApoER2 and VLDLR, followed by phosphorylation of the Dab1 adaptor protein. Accumulating evidence suggests that Reelin signaling controls multiple steps of neuronal migration, including the transition from multipolar to bipolar neurons, terminal translocation, and termination of migration beneath the marginal zone. In addition, it has been shown that ectopically expressed Reelin can cause neuronal aggregation via an N-cadherin-mediated manner. This review attempts to summarize our knowledge of the roles played by Reelin in neuronal migration and the underlying mechanisms.
Collapse
Affiliation(s)
- Yuki Hirota
- Department of Anatomy, Keio University School of MedicineTokyo, Japan
| | - Kazunori Nakajima
- Department of Anatomy, Keio University School of MedicineTokyo, Japan
| |
Collapse
|
61
|
Shiotani H, Maruo T, Sakakibara S, Miyata M, Mandai K, Mochizuki H, Takai Y. Aging-dependent expression of synapse-related proteins in the mouse brain. Genes Cells 2017; 22:472-484. [DOI: 10.1111/gtc.12489] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 03/08/2017] [Indexed: 01/13/2023]
Affiliation(s)
- Hajime Shiotani
- Division of Pathogenetic Signaling, Department of Biochemistry and Molecular Biology; Kobe University Graduate School of Medicine; Kobe 650-0047 Japan
- Department of Neurology; Osaka University Graduate School of Medicine; Suita 565-0871 Japan
| | - Tomohiko Maruo
- Division of Pathogenetic Signaling, Department of Biochemistry and Molecular Biology; Kobe University Graduate School of Medicine; Kobe 650-0047 Japan
| | - Shotaro Sakakibara
- Division of Pathogenetic Signaling, Department of Biochemistry and Molecular Biology; Kobe University Graduate School of Medicine; Kobe 650-0047 Japan
| | - Muneaki Miyata
- Division of Pathogenetic Signaling, Department of Biochemistry and Molecular Biology; Kobe University Graduate School of Medicine; Kobe 650-0047 Japan
| | - Kenji Mandai
- Division of Pathogenetic Signaling, Department of Biochemistry and Molecular Biology; Kobe University Graduate School of Medicine; Kobe 650-0047 Japan
| | - Hideki Mochizuki
- Department of Neurology; Osaka University Graduate School of Medicine; Suita 565-0871 Japan
| | - Yoshimi Takai
- Division of Pathogenetic Signaling, Department of Biochemistry and Molecular Biology; Kobe University Graduate School of Medicine; Kobe 650-0047 Japan
| |
Collapse
|
62
|
Optogenetic control of the Dab1 signaling pathway. Sci Rep 2017; 7:43760. [PMID: 28272509 PMCID: PMC5363252 DOI: 10.1038/srep43760] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 01/26/2017] [Indexed: 12/31/2022] Open
Abstract
The Reelin-Dab1 signaling pathway regulates development of the mammalian brain, including neuron migrations in various brain regions, as well as learning and memory in adults. Extracellular Reelin binds to cell surface receptors and activates phosphorylation of the intracellular Dab1 protein. Dab1 is required for most effects of Reelin, but Dab1-independent pathways may contribute. Here we developed a single-component, photoactivatable Dab1 (opto-Dab1) by using the blue light-sensitive dimerization/oligomerization property of A. thaliana Cryptochrome 2 (Cry2). Opto-Dab1 can activate downstream signals rapidly, locally, and reversibly upon blue light illumination. The high spatiotemporal resolution of the opto-Dab1 probe also allows us to control membrane protrusion, retraction and ruffling by local illumination in both COS7 cells and in primary neurons. This shows that Dab1 activation is sufficient to orient cell movement in the absence of other signals. Opto-Dab1 may be useful to study the biological functions of the Reelin-Dab1 signaling pathway both in vitro and in vivo.
Collapse
|
63
|
Miyata M, Maruo T, Kaito A, Wang S, Yamamoto H, Fujiwara T, Mizoguchi A, Mandai K, Takai Y. Roles of afadin in the formation of the cellular architecture of the mouse hippocampus and dentate gyrus. Mol Cell Neurosci 2017; 79:34-44. [DOI: 10.1016/j.mcn.2016.12.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 11/15/2016] [Accepted: 12/27/2016] [Indexed: 12/19/2022] Open
|
64
|
Reelin transiently promotes N-cadherin-dependent neuronal adhesion during mouse cortical development. Proc Natl Acad Sci U S A 2017; 114:2048-2053. [PMID: 28174271 DOI: 10.1073/pnas.1615215114] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Reelin is an essential glycoprotein for the establishment of the highly organized six-layered structure of neurons of the mammalian neocortex. Although the role of Reelin in the control of neuronal migration has been extensively studied at the molecular level, the mechanisms underlying Reelin-dependent neuronal layer organization are not yet fully understood. In this study, we directly showed that Reelin promotes adhesion among dissociated neocortical neurons in culture. The Reelin-mediated neuronal aggregation occurs in an N-cadherin-dependent manner, both in vivo and in vitro. Unexpectedly, however, in a rotation culture of dissociated neocortical cells that gradually reaggregated over time, we found that it was the neural progenitor cells [radial glial cells (RGCs)], rather than the neurons, that tended to form clusters in the presence of Reelin. Mathematical modeling suggested that this clustering of RGCs could be recapitulated if the Reelin-dependent promotion of neuronal adhesion were to occur only transiently. Thus, we directly measured the adhesive force between neurons and N-cadherin by atomic force microscopy, and found that Reelin indeed enhanced the adhesiveness of neurons to N-cadherin; this enhanced adhesiveness began to be observed at 30 min after Reelin stimulation, but declined by 3 h. These results suggest that Reelin transiently (and not persistently) promotes N-cadherin-mediated neuronal aggregation. When N-cadherin and stabilized β-catenin were overexpressed in the migrating neurons, the transfected neurons were abnormally distributed in the superficial region of the neocortex, suggesting that appropriate regulation of N-cadherin-mediated adhesion is important for correct positioning of the neurons during neocortical development.
Collapse
|
65
|
|
66
|
Ishii K, Kubo KI, Nakajima K. Reelin and Neuropsychiatric Disorders. Front Cell Neurosci 2016; 10:229. [PMID: 27803648 PMCID: PMC5067484 DOI: 10.3389/fncel.2016.00229] [Citation(s) in RCA: 138] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 09/22/2016] [Indexed: 12/22/2022] Open
Abstract
Proper neuronal migration and laminar formation during corticogenesis is essential for normal brain function. Disruption of these developmental processes is thought to be involved in the pathogenesis of some neuropsychiatric conditions. Especially, Reelin, a glycoprotein mainly secreted by the Cajal-Retzius cells and a subpopulation of GABAergic interneurons, has been shown to play a critical role, both during embryonic and postnatal periods. Indeed, animal studies have clearly revealed that Reelin is an essential molecule for proper migration of cortical neurons and finally regulates the cell positioning in the cortex during embryonic and early postnatal stages; by contrast, Reelin signaling is closely involved in synaptic function in adulthood. In humans, genetic studies have shown that the reelin gene (RELN) is associated with a number of psychiatric diseases, including Schizophrenia (SZ), bipolar disorder (BP) and autistic spectrum disorder. Indeed, Reln haploinsufficiency has been shown to cause cognitive impairment in rodents, suggesting the expression level of the Reelin protein is closely related to the higher brain functions. However, the molecular abnormalities in the Reelin pathway involved in the pathogenesis of psychiatric disorders are not yet fully understood. In this article, we review the current progress in the understanding of the Reelin functions that could be related to the pathogenesis of psychiatric disorders. Furthermore, we discuss the basis for selecting Reelin and molecules in its downstream signaling pathway as potential therapeutic targets for psychiatric illnesses.
Collapse
Affiliation(s)
- Kazuhiro Ishii
- Department of Anatomy, Keio University School of Medicine Tokyo, Japan
| | - Ken-Ichiro Kubo
- Department of Anatomy, Keio University School of Medicine Tokyo, Japan
| | - Kazunori Nakajima
- Department of Anatomy, Keio University School of Medicine Tokyo, Japan
| |
Collapse
|
67
|
de Frutos C, Bouvier G, Arai Y, Thion M, Lokmane L, Keita M, Garcia-Dominguez M, Charnay P, Hirata T, Riethmacher D, Grove E, Tissir F, Casado M, Pierani A, Garel S. Reallocation of Olfactory Cajal-Retzius Cells Shapes Neocortex Architecture. Neuron 2016; 92:435-448. [DOI: 10.1016/j.neuron.2016.09.020] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 07/13/2016] [Accepted: 09/06/2016] [Indexed: 11/25/2022]
|
68
|
Chai X, Frotscher M. How does Reelin signaling regulate the neuronal cytoskeleton during migration? NEUROGENESIS 2016; 3:e1242455. [PMID: 28265585 DOI: 10.1080/23262133.2016.1242455] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 09/15/2016] [Accepted: 09/25/2016] [Indexed: 01/17/2023]
Abstract
Neuronal migration is an essential step in the formation of laminated brain structures. In the developing cerebral cortex, pyramidal neurons migrate toward the Reelin-containing marginal zone. Reelin is an extracellular matrix protein synthesized by Cajal-Retzius cells. In this review, we summarize our recent results and hypotheses on how Reelin might regulate neuronal migration by acting on the actin and microtubule cytoskeleton. By binding to ApoER2 receptors on the migrating neurons, Reelin induces stabilization of the leading processes extending toward the marginal zone, which involves Dab1 phosphorylation, adhesion molecule expression, cofilin phosphorylation and inhibition of tau phosphorylation. By binding to VLDLR and integrin receptors, Reelin interacts with Lis1 and induces nuclear translocation, accompanied by the ubiquitination of phosphorylated Dab1. Eventually Reelin induces clustering of its receptors resulting in the endocytosis of a Reelin/receptor complex (particularly VLDLR). The resulting decrease in Reelin contributes to neuronal arrest at the marginal zone.
Collapse
Affiliation(s)
- Xuejun Chai
- Institute for Structural Neurobiology, Center for Molecular Neurobiology Hamburg (ZMNH) , Hamburg, Germany
| | - Michael Frotscher
- Institute for Structural Neurobiology, Center for Molecular Neurobiology Hamburg (ZMNH) , Hamburg, Germany
| |
Collapse
|
69
|
ADP Ribosylation Factor 6 Regulates Neuronal Migration in the Developing Cerebral Cortex through FIP3/Arfophilin-1-dependent Endosomal Trafficking of N-cadherin. eNeuro 2016; 3:eN-NWR-0148-16. [PMID: 27622210 PMCID: PMC5002984 DOI: 10.1523/eneuro.0148-16.2016] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 07/29/2016] [Accepted: 08/02/2016] [Indexed: 12/15/2022] Open
Abstract
During neural development, endosomal trafficking controls cell shape and motility through the polarized transport of membrane proteins related to cell–cell and cell–extracellular matrix interactions. ADP ribosylation factor 6 (Arf6) is a critical small GTPase that regulates membrane trafficking between the plasma membrane and endosomes. We herein demonstrated that the knockdown of endogenous Arf6 in mouse cerebral cortices led to impaired neuronal migration in the intermediate zone and cytoplasmic retention of N-cadherin and syntaxin12 in migrating neurons. Rescue experiments with separation-of-function Arf6 mutants identified Rab11 family-interacting protein 3 (FIP3)/Arfophilin-1, a dual effector for Arf6 and Rab11, as a downstream effector of Arf6 in migrating neurons. The knockdown of FIP3 led to impaired neuronal migration in the intermediate zone and cytoplasmic retention of N-cadherin in migrating neurons, similar to that of Arf6, which could be rescued by the coexpression of wild-type FIP3 but not FIP3 mutants lacking the binding site for Arf6 or Rab11. These results suggest that Arf6 regulates cortical neuronal migration in the intermediate zone through the FIP3-dependent endosomal trafficking.
Collapse
|
70
|
Jiang X, Nardelli J. Cellular and molecular introduction to brain development. Neurobiol Dis 2016; 92:3-17. [PMID: 26184894 PMCID: PMC4720585 DOI: 10.1016/j.nbd.2015.07.007] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2015] [Revised: 07/07/2015] [Accepted: 07/09/2015] [Indexed: 12/13/2022] Open
Abstract
Advances in the study of brain development over the last decades, especially recent findings regarding the evolutionary expansion of the human neocortex, and large-scale analyses of the proteome/transcriptome in the human brain, have offered novel insights into the molecular mechanisms guiding neural maturation, and the pathophysiology of multiple forms of neurological disorders. As a preamble to reviews of this issue, we provide an overview of the cellular, molecular and genetic bases of brain development with an emphasis on the major mechanisms associated with landmarks of normal neural development in the embryonic stage and early postnatal life, including neural stem/progenitor cell proliferation, cortical neuronal migration, evolution and folding of the cerebral cortex, synaptogenesis and neural circuit development, gliogenesis and myelination. We will only briefly depict developmental disorders that result from perturbations of these cellular or molecular mechanisms, and the most common perinatal brain injuries that could disturb normal brain development.
Collapse
Affiliation(s)
- Xiangning Jiang
- Department of Pediatrics, University of California, San Francisco, CA 94158, USA
| | - Jeannette Nardelli
- Inserm, U1141, Paris 75019, France; Université Paris Diderot, Sorbonne Paris Cité, UMRS 1141, Paris 75019, France.
| |
Collapse
|
71
|
Bock HH, May P. Canonical and Non-canonical Reelin Signaling. Front Cell Neurosci 2016; 10:166. [PMID: 27445693 PMCID: PMC4928174 DOI: 10.3389/fncel.2016.00166] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 06/08/2016] [Indexed: 12/11/2022] Open
Abstract
Reelin is a large secreted glycoprotein that is essential for correct neuronal positioning during neurodevelopment and is important for synaptic plasticity in the mature brain. Moreover, Reelin is expressed in many extraneuronal tissues; yet the roles of peripheral Reelin are largely unknown. In the brain, many of Reelin's functions are mediated by a molecular signaling cascade that involves two lipoprotein receptors, apolipoprotein E receptor-2 (Apoer2) and very low density-lipoprotein receptor (Vldlr), the neuronal phosphoprotein Disabled-1 (Dab1), and members of the Src family of protein tyrosine kinases as crucial elements. This core signaling pathway in turn modulates the activity of adaptor proteins and downstream protein kinase cascades, many of which target the neuronal cytoskeleton. However, additional Reelin-binding receptors have been postulated or described, either as coreceptors that are essential for the activation of the "canonical" Reelin signaling cascade involving Apoer2/Vldlr and Dab1, or as receptors that activate alternative or additional signaling pathways. Here we will give an overview of canonical and alternative Reelin signaling pathways, molecular mechanisms involved, and their potential physiological roles in the context of different biological settings.
Collapse
Affiliation(s)
- Hans H Bock
- Clinic of Gastroenterology and Hepatology, Heinrich-Heine-University Düsseldorf Düsseldorf, Germany
| | - Petra May
- Clinic of Gastroenterology and Hepatology, Heinrich-Heine-University Düsseldorf Düsseldorf, Germany
| |
Collapse
|
72
|
Lee GH, D'Arcangelo G. New Insights into Reelin-Mediated Signaling Pathways. Front Cell Neurosci 2016; 10:122. [PMID: 27242434 PMCID: PMC4860420 DOI: 10.3389/fncel.2016.00122] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 04/27/2016] [Indexed: 11/21/2022] Open
Abstract
Reelin, a multifunctional extracellular protein that is important for mammalian brain development and function, is secreted by different cell types in the prenatal or postnatal brain. The spatiotemporal regulation of Reelin expression and distribution during development relates to its multifaceted function in the brain. Prenatally Reelin controls neuronal radial migration and proper positioning in cortical layers, whereas postnatally Reelin promotes neuronal maturation, synaptic formation and plasticity. The molecular mechanisms underlying the distinct biological functions of Reelin during and after brain development involve unique and overlapping signaling pathways that are activated following Reelin binding to its cell surface receptors. Distinct Reelin ligand isoforms, such as the full-length protein or fragments generated by proteolytic cleavage differentially affect the activity of downstream signaling pathways. In this review, we discuss recent advances in our understanding of the signaling transduction pathways activated by Reelin that regulate different aspects of brain development and function. A core signaling machinery, including ApoER2/VLDLR receptors, Src/Fyn kinases, and the adaptor protein Dab1, participates in all known aspects of Reelin biology. However, distinct downstream mechanisms, such as the Crk/Rap1 pathway and cell adhesion molecules, play crucial roles in the control of neuronal migration, whereas the PI3K/Akt/mTOR pathway appears to be more important for dendrite and spine development. Finally, the NMDA receptor (NMDAR) and an unidentified receptor contribute to the activation of the MEK/Erk1/2 pathway leading to the upregulation of genes involved in synaptic plasticity and learning. This knowledge may provide new insight into neurodevelopmental or neurodegenerative disorders that are associated with Reelin dysfunction.
Collapse
Affiliation(s)
- Gum Hwa Lee
- College of Pharmacy, Chosun University Gwangju, South Korea
| | - Gabriella D'Arcangelo
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey Piscataway, NJ, USA
| |
Collapse
|
73
|
Martinez-Garay I, Gil-Sanz C, Franco SJ, Espinosa A, Molnár Z, Mueller U. Cadherin 2/4 signaling via PTP1B and catenins is crucial for nucleokinesis during radial neuronal migration in the neocortex. Development 2016; 143:2121-34. [PMID: 27151949 PMCID: PMC4920171 DOI: 10.1242/dev.132456] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 04/26/2016] [Indexed: 11/20/2022]
Abstract
Cadherins are crucial for the radial migration of excitatory projection neurons into the developing neocortical wall. However, the specific cadherins and the signaling pathways that regulate radial migration are not well understood. Here, we show that cadherin 2 (CDH2) and CDH4 cooperate to regulate radial migration in mouse brain via the protein tyrosine phosphatase 1B (PTP1B) and α- and β-catenins. Surprisingly, perturbation of cadherin-mediated signaling does not affect the formation and extension of leading processes of migrating neocortical neurons. Instead, movement of the cell body and nucleus (nucleokinesis) is disrupted. This defect is partially rescued by overexpression of LIS1, a microtubule-associated protein that has previously been shown to regulate nucleokinesis. Taken together, our findings indicate that cadherin-mediated signaling to the cytoskeleton is crucial for nucleokinesis of neocortical projection neurons during their radial migration. Highlighted article: In radially migrating mouse cortical neurons, cadherin-mediated signaling to the cytoskeleton regulates the forward movement of the nucleus.
Collapse
Affiliation(s)
- Isabel Martinez-Garay
- Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Cristina Gil-Sanz
- Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Santos J Franco
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, USA Program of Pediatric Stem Cell Biology, Children's Hospital Colorado, Aurora, CO 80045, USA
| | - Ana Espinosa
- Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Zoltán Molnár
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK
| | - Ulrich Mueller
- Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| |
Collapse
|
74
|
Prox1 Regulates the Subtype-Specific Development of Caudal Ganglionic Eminence-Derived GABAergic Cortical Interneurons. J Neurosci 2016; 35:12869-89. [PMID: 26377473 DOI: 10.1523/jneurosci.1164-15.2015] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
UNLABELLED Neurogliaform (RELN+) and bipolar (VIP+) GABAergic interneurons of the mammalian cerebral cortex provide critical inhibition locally within the superficial layers. While these subtypes are known to originate from the embryonic caudal ganglionic eminence (CGE), the specific genetic programs that direct their positioning, maturation, and integration into the cortical network have not been elucidated. Here, we report that in mice expression of the transcription factor Prox1 is selectively maintained in postmitotic CGE-derived cortical interneuron precursors and that loss of Prox1 impairs the integration of these cells into superficial layers. Moreover, Prox1 differentially regulates the postnatal maturation of each specific subtype originating from the CGE (RELN, Calb2/VIP, and VIP). Interestingly, Prox1 promotes the maturation of CGE-derived interneuron subtypes through intrinsic differentiation programs that operate in tandem with extrinsically driven neuronal activity-dependent pathways. Thus Prox1 represents the first identified transcription factor specifically required for the embryonic and postnatal acquisition of CGE-derived cortical interneuron properties. SIGNIFICANCE STATEMENT Despite the recognition that 30% of GABAergic cortical interneurons originate from the caudal ganglionic eminence (CGE), to date, a specific transcriptional program that selectively regulates the development of these populations has not yet been identified. Moreover, while CGE-derived interneurons display unique patterns of tangential and radial migration and preferentially populate the superficial layers of the cortex, identification of a molecular program that controls these events is lacking.Here, we demonstrate that the homeodomain transcription factor Prox1 is expressed in postmitotic CGE-derived cortical interneuron precursors and is maintained into adulthood. We found that Prox1 function is differentially required during both embryonic and postnatal stages of development to direct the migration, differentiation, circuit integration, and maintenance programs within distinct subtypes of CGE-derived interneurons.
Collapse
|
75
|
Tang BL. Rab, Arf, and Arl-Regulated Membrane Traffic in Cortical Neuron Migration. J Cell Physiol 2015; 231:1417-23. [DOI: 10.1002/jcp.25261] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 11/18/2015] [Indexed: 12/13/2022]
Affiliation(s)
- Bor Luen Tang
- Department of Biochemistry; Yong Loo Lin School of Medicine; National University of Singapore; Singapore
- NUS Graduate School for Integrative Sciences and Engineering; National University of Singapore; Singapore
| |
Collapse
|
76
|
Ohtaka-Maruyama C, Okado H. Molecular Pathways Underlying Projection Neuron Production and Migration during Cerebral Cortical Development. Front Neurosci 2015; 9:447. [PMID: 26733777 PMCID: PMC4682034 DOI: 10.3389/fnins.2015.00447] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 11/09/2015] [Indexed: 12/25/2022] Open
Abstract
Glutamatergic neurons of the mammalian cerebral cortex originate from radial glia (RG) progenitors in the ventricular zone (VZ). During corticogenesis, neuroblasts migrate toward the pial surface using two different migration modes. One is multipolar (MP) migration with random directional movement, and the other is locomotion, which is a unidirectional movement guided by the RG fiber. After reaching their final destination, the neurons finalize their migration by terminal translocation, which is followed by maturation via dendrite extension to initiate synaptogenesis and thereby complete neural circuit formation. This switching of migration modes during cortical development is unique in mammals, which suggests that the RG-guided locomotion mode may contribute to the evolution of the mammalian neocortical 6-layer structure. Many factors have been reported to be involved in the regulation of this radial neuronal migration process. In general, the radial migration can be largely divided into four steps; (1) maintenance and departure from the VZ of neural progenitor cells, (2) MP migration and transition to bipolar cells, (3) RG-guided locomotion, and (4) terminal translocation and dendrite maturation. Among these, many different gene mutations or knockdown effects have resulted in failure of the MP to bipolar transition (step 2), suggesting that it is a critical step, particularly in radial migration. Moreover, this transition occurs at the subplate layer. In this review, we summarize recent advances in our understanding of the molecular mechanisms underlying each of these steps. Finally, we discuss the evolutionary aspects of neuronal migration in corticogenesis.
Collapse
Affiliation(s)
- Chiaki Ohtaka-Maruyama
- Neural Network Project, Department of Brain Development and Neural Regeneration, Tokyo Metropolitan Institute of Medical Science Tokyo, Japan
| | - Haruo Okado
- Neural Development Project, Department of Brain Development and Neural Regeneration, Tokyo Metropolitan Institute of Medical Science Tokyo, Japan
| |
Collapse
|
77
|
Barber M, Pierani A. Tangential migration of glutamatergic neurons and cortical patterning during development: Lessons from Cajal-Retzius cells. Dev Neurobiol 2015; 76:847-81. [PMID: 26581033 DOI: 10.1002/dneu.22363] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 11/12/2015] [Accepted: 11/13/2015] [Indexed: 12/14/2022]
Abstract
Tangential migration is a mode of cell movement, which in the developing cerebral cortex, is defined by displacement parallel to the ventricular surface and orthogonal to the radial glial fibers. This mode of long-range migration is a strategy by which distinct neuronal classes generated from spatially and molecularly distinct origins can integrate to form appropriate neural circuits within the cortical plate. While it was previously believed that only GABAergic cortical interneurons migrate tangentially from their origins in the subpallial ganglionic eminences to integrate in the cortical plate, it is now known that transient populations of glutamatergic neurons also adopt this mode of migration. These include Cajal-Retzius cells (CRs), subplate neurons (SPs), and cortical plate transient neurons (CPTs), which have crucial roles in orchestrating the radial and tangential development of the embryonic cerebral cortex in a noncell-autonomous manner. While CRs have been extensively studied, it is only in the last decade that the molecular mechanisms governing their tangential migration have begun to be elucidated. To date, the mechanisms of SPs and CPTs tangential migration remain unknown. We therefore review the known signaling pathways, which regulate parameters of CRs migration including their motility, contact-redistribution and adhesion to the pial surface, and discuss this in the context of how CR migration may regulate their signaling activity in a spatial and temporal manner. © 2015 Wiley Periodicals, Inc. Develop Neurobiol 76: 847-881, 2016.
Collapse
Affiliation(s)
- Melissa Barber
- Institut Jacques-Monod, CNRS, Université Paris Diderot, Sorbonne Cité, Paris, France.,Department of Cell and Developmental Biology, University College London, WC1E 6BT, United Kingdom
| | - Alessandra Pierani
- Institut Jacques-Monod, CNRS, Université Paris Diderot, Sorbonne Cité, Paris, France
| |
Collapse
|
78
|
Abstract
UNLABELLED Human mutations in ZIC2 have been identified in patients with holoprosencephaly and schizophrenia. Similarly, Zic2 mutant mice exhibit holoprosencephaly in homozygosis and behavioral and morphological schizophrenic phenotypes associated with forebrain defects in heterozygosis. Despite the devastating effects of mutations in Zic2, the cellular and molecular mechanisms that provoke Zic2-deficiency phenotypes are yet unclear. Here, we report a novel role for this transcription factor in the migration of three different types of forebrain neurons: the Cajal-Retzius cells that populate the surface of the telencephalic vesicles, an amygdaloid group of cells originated in the caudal pole of the telencephalic pallium, and a cell population that travels from the prethalamic neuroepithelium to the ventral lateral geniculate nucleus. Our results also suggest that the receptor EphB1, previously identified as a Zic2 target, may mediate, at least partially, Zic2-dependent migratory events. According to these results, we propose that deficiencies in cell motility and guidance contribute to most of the forebrain pathologies associated with Zic2 mutations. SIGNIFICANCE STATEMENT Although the phenotype of Zic2 mutant individuals was reported more than 10 years ago, until now, the main function of this transcription factor during early development has not been precisely defined. Here, we reveal a previously unknown role for Zic2 in the migration of forebrain neurons such as Cajal-Retzius cells, interneurons moving to the ventral lateral geniculate nucleus, and neocortical cells going to the amygdala. We believe that the role of this transcription factor in certain populations of migratory cells contributes to defects in cortical layering and hypocellularity in the ventral LGN and amygdala and will contribute to our understanding of the devastating phenotypes associated with Zic2 mutations in both humans and mice.
Collapse
|
79
|
Abstract
UNLABELLED The mechanisms controlling cortical dendrite initiation and targeting are poorly understood. Multiphoton imaging of developing mouse cortex reveals that apical dendrites emerge by direct transformation of the neuron's leading process during the terminal phase of neuronal migration. During this ∼110 min period, the dendritic arbor increases ∼2.5-fold in size and migration arrest occurs below the first stable branch point in the developing arbor. This dendritic outgrowth is triggered at the time of leading process contact with the marginal zone (MZ) and occurs primarily by neurite extension into the extracellular matrix of the MZ. In reeler cortices that lack the secreted glycoprotein Reelin, a subset of neurons completed migration but then retracted and reorganized their arbor in a tangential direction away from the MZ soon after migration arrest. For these reeler neurons, the tangential oriented primary neurites were longer lived than the radially oriented primary neurites, whereas the opposite was true of wild-type (WT) neurons. Application of Reelin protein to reeler cortices destabilized tangential neurites while stabilizing radial neurites and stimulating dendritic growth in the MZ. Therefore, Reelin functions as part of a polarity signaling system that links dendritogenesis in the MZ with cellular positioning and cortical lamination. SIGNIFICANCE STATEMENT Whether the apical dendrite emerges by transformation of the leading process of the migrating neuron or emerges de novo after migration is completed is unclear. Similarly, it is not clear whether the secreted glycoprotein Reelin controls migration and dendritic growth as related or separate processes. Here, multiphoton microscopy reveals the direct transformation of the leading process into the apical dendrite. This transformation is coupled to the successful completion of migration and neuronal soma arrest occurs below the first stable branch point of the nascent dendrite. Deficiency in Reelin causes the forming dendrite to avoid its normal target area and branch aberrantly, leading to improper cellular positioning. Therefore, this study links Reelin-dependent dendritogenesis with migration arrest and cortical lamination.
Collapse
|
80
|
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: 6.7] [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
|
81
|
Yamamoto H, Mandai K, Konno D, Maruo T, Matsuzaki F, Takai Y. Impairment of radial glial scaffold-dependent neuronal migration and formation of double cortex by genetic ablation of afadin. Brain Res 2015; 1620:139-52. [DOI: 10.1016/j.brainres.2015.05.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 05/04/2015] [Accepted: 05/09/2015] [Indexed: 12/28/2022]
|
82
|
Moon UY, Park JY, Park R, Cho JY, Hughes LJ, McKenna J, Goetzl L, Cho SH, Crino PB, Gambello MJ, Kim S. Impaired Reelin-Dab1 Signaling Contributes to Neuronal Migration Deficits of Tuberous Sclerosis Complex. Cell Rep 2015; 12:965-78. [PMID: 26235615 PMCID: PMC4536164 DOI: 10.1016/j.celrep.2015.07.013] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 06/01/2015] [Accepted: 07/07/2015] [Indexed: 01/06/2023] Open
Abstract
Tuberous sclerosis complex (TSC) is associated with neurodevelopmental abnormalities, including defects in neuronal migration. However, the alterations in cell signaling mechanisms critical for migration and final positioning of neurons in TSC remain unclear. Our detailed cellular analyses reveal that reduced Tsc2 in newborn neurons causes abnormalities in leading processes of migrating neurons, accompanied by significantly delayed migration. Importantly, we demonstrate that Reelin-Dab1 signaling is aberrantly regulated in TSC mouse models and in cortical tubers from TSC patients owing to enhanced expression of the E3 ubiquitin ligase Cul5, a known mediator of pDab1 ubiquitination. Likewise, mTORC1 activation by Rheb overexpression generates similar neuronal and Reelin-Dab1 signaling defects, and directly upregulates Cul5 expression. Inhibition of mTORC1 by rapamycin treatment or by reducing Cul5 largely restores normal leading processes and positioning of migrating neurons. Thus, disrupted Reelin-Dab1 signaling is critically involved in the neuronal migration defects of TSC.
Collapse
Affiliation(s)
- Uk Yeol Moon
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA; Department of Anatomy and Cell Biology, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Jun Young Park
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA; Department of Anatomy and Cell Biology, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Raehee Park
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA; Department of Anatomy and Cell Biology, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Jennifer Y Cho
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Lucinda J Hughes
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA; Graduate Program of Biomedical Sciences, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - James McKenna
- Department of Human Genetics, Emory University, School of Medicine, Atlanta, GA 30322, USA
| | - Laura Goetzl
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA; Department of Obstetrics Gynecology and Reproductive Sciences, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Seo-Hee Cho
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA; Department of Anatomy and Cell Biology, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Peter B Crino
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA; Department of Neurology, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Michael J Gambello
- Department of Human Genetics, Emory University, School of Medicine, Atlanta, GA 30322, USA
| | - Seonhee Kim
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA; Department of Anatomy and Cell Biology, Temple University School of Medicine, Philadelphia, PA 19140, USA.
| |
Collapse
|
83
|
Toma K, Hanashima C. Switching modes in corticogenesis: mechanisms of neuronal subtype transitions and integration in the cerebral cortex. Front Neurosci 2015; 9:274. [PMID: 26321900 PMCID: PMC4531338 DOI: 10.3389/fnins.2015.00274] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 07/21/2015] [Indexed: 12/16/2022] Open
Abstract
Information processing in the cerebral cortex requires the activation of diverse neurons across layers and columns, which are established through the coordinated production of distinct neuronal subtypes and their placement along the three-dimensional axis. Over recent years, our knowledge of the regulatory mechanisms of the specification and integration of neuronal subtypes in the cerebral cortex has progressed rapidly. In this review, we address how the unique cytoarchitecture of the neocortex is established from a limited number of progenitors featuring neuronal identity transitions during development. We further illuminate the molecular mechanisms of the subtype-specific integration of these neurons into the cerebral cortex along the radial and tangential axis, and we discuss these key features to exemplify how neocortical circuit formation accomplishes economical connectivity while maintaining plasticity and evolvability to adapt to environmental changes.
Collapse
Affiliation(s)
- Kenichi Toma
- Laboratory for Neocortical Development, RIKEN Center for Developmental Biology Kobe, Japan
| | - Carina Hanashima
- Laboratory for Neocortical Development, RIKEN Center for Developmental Biology Kobe, Japan ; Department of Biology, Graduate School of Science, Kobe University Kobe, Japan
| |
Collapse
|
84
|
Honda T, Nakajima K. Proper Level of Cytosolic Disabled-1, Which Is Regulated by Dual Nuclear Translocation Pathways, Is Important for Cortical Neuronal Migration. Cereb Cortex 2015. [PMID: 26209842 DOI: 10.1093/cercor/bhv162] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Disabled-1 (Dab1) is an essential intracellular protein in the Reelin pathway. It has a nuclear localization signal (NLS; hereafter referred to as "NLS1") and 2 nuclear export signals, and shuttles between the nucleus and the cytoplasm. In this study, we found that Dab1 has an additional unidentified NLS, and that the Dab1 NLS1 mutant could translocate to the nucleus in an unconventional ATP/temperature-dependent and cytoplasmic factor/RanGTP gradient-independent manner. Additional mutations in the NLS1 mutant revealed that K(67) and K(69) are important for the nuclear transport. Furthermore, an excess of the intracellular domain of the Reelin receptors inhibited the nuclear translocation of Dab1. An in utero electroporation study showed that a large amount of Dab1 in the cytoplasm in migrating neurons inhibited the migration, and that forced transport of Dab1 into the nucleus attenuated this inhibitory effect. In addition, rescue experiments using yotari, an autosomal recessive mutant of dab1, revealed that cells expressing Dab1 NLS1 mutant tend to distribute at more superficial positions than those expressing wild-type Dab1. Taken together, these findings suggest that Dab1 has at least 2 NLSs, and that the regulation of the subcellular localization of Dab1 is important for the proper migration of excitatory neurons.
Collapse
Affiliation(s)
- Takao Honda
- Department of Anatomy, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Kazunori Nakajima
- Department of Anatomy, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| |
Collapse
|
85
|
Dentate Gyrus Development Requires ERK Activity to Maintain Progenitor Population and MAPK Pathway Feedback Regulation. J Neurosci 2015; 35:6836-48. [PMID: 25926459 DOI: 10.1523/jneurosci.4196-14.2015] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The ERK/MAPK pathway is an important developmental signaling pathway. Mutations in upstream elements of this pathway result in neuro-cardio-facial cutaneous (NCFC) syndromes, which are typified by impaired neurocognitive abilities that are reliant upon hippocampal function. The role of ERK signaling during hippocampal development has not been examined and may provide critical insight into the cause of hippocampal dysfunction in NCFC syndromes. In this study, we have generated ERK1 and conditional ERK2 compound knock-out mice to determine the role of ERK signaling during development of the hippocampal dentate gyrus. We found that loss of both ERK1 and ERK2 resulted in 60% fewer granule cells and near complete absence of neural progenitor pools in the postnatal dentate gyrus. Loss of ERK1/2 impaired maintenance of neural progenitors as they migrate from the dentate ventricular zone to the dentate gyrus proper, resulting in premature depletion of neural progenitor cells beginning at E16.5, which prevented generation of granule cells later in development. Finally, loss of ERK2 alone does not impair development of the dentate gyrus as animals expressing only ERK1 developed a normal hippocampus. These findings establish that ERK signaling regulates maintenance of progenitor cells required for development of the dentate gyrus.
Collapse
|
86
|
Squarzoni P, Thion MS, Garel S. Neuronal and microglial regulators of cortical wiring: usual and novel guideposts. Front Neurosci 2015; 9:248. [PMID: 26236185 PMCID: PMC4505395 DOI: 10.3389/fnins.2015.00248] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 06/30/2015] [Indexed: 12/17/2022] Open
Abstract
Neocortex functioning relies on the formation of complex networks that begin to be assembled during embryogenesis by highly stereotyped processes of cell migration and axonal navigation. The guidance of cells and axons is driven by extracellular cues, released along by final targets or intermediate targets located along specific pathways. In particular, guidepost cells, originally described in the grasshopper, are considered discrete, specialized cell populations located at crucial decision points along axonal trajectories that regulate tract formation. These cells are usually early-born, transient and act at short-range or via cell-cell contact. The vast majority of guidepost cells initially identified were glial cells, which play a role in the formation of important axonal tracts in the forebrain, such as the corpus callosum, anterior, and post-optic commissures as well as optic chiasm. In the last decades, tangential migrating neurons have also been found to participate in the guidance of principal axonal tracts in the forebrain. This is the case for several examples such as guideposts for the lateral olfactory tract (LOT), corridor cells, which open an internal path for thalamo-cortical axons and Cajal-Retzius cells that have been involved in the formation of the entorhino-hippocampal connections. More recently, microglia, the resident macrophages of the brain, were specifically observed at the crossroads of important neuronal migratory routes and axonal tract pathways during forebrain development. We furthermore found that microglia participate to the shaping of prenatal forebrain circuits, thereby opening novel perspectives on forebrain development and wiring. Here we will review the last findings on already known guidepost cell populations and will discuss the role of microglia as a potentially new class of atypical guidepost cells.
Collapse
Affiliation(s)
- Paola Squarzoni
- Centre National de la Recherche Scientifique UMR8197, Ecole Normale Supérieure, Institut de Biologie, Institut National de la Santé et de la Recherche Médicale U1024 Paris, France
| | - Morgane S Thion
- Centre National de la Recherche Scientifique UMR8197, Ecole Normale Supérieure, Institut de Biologie, Institut National de la Santé et de la Recherche Médicale U1024 Paris, France
| | - Sonia Garel
- Centre National de la Recherche Scientifique UMR8197, Ecole Normale Supérieure, Institut de Biologie, Institut National de la Santé et de la Recherche Médicale U1024 Paris, France
| |
Collapse
|
87
|
Nakagawa N, Yagi H, Kato K, Takematsu H, Oka S. Ectopic clustering of Cajal-Retzius and subplate cells is an initial pathological feature in Pomgnt2-knockout mice, a model of dystroglycanopathy. Sci Rep 2015; 5:11163. [PMID: 26060116 PMCID: PMC4461912 DOI: 10.1038/srep11163] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 05/18/2015] [Indexed: 11/09/2022] Open
Abstract
Aberrant glycosylation of dystroglycan causes congenital muscular dystrophies associated with cobblestone lissencephaly, classified as dystroglycanopathy. However, pathological features in the onset of brain malformations, including the precise timing and primary cause of the pial basement membrane disruption and abnormalities in the migration of pyramidal neurons, remain unexplored. Using the Pomgnt2-knockout (KO) mouse as a dystroglycanopathy model, we show that breaches of the pial basement membrane appeared at embryonic day 11.5, coinciding with the ectopic clustering of Cajal-Retzius cells and subplate neurons and prior to the migration onset of pyramidal neurons. Furthermore, in the Pomgnt2-KO cerebral cortex, preplate splitting failure likely occurred due to the aggregation of Cajal-Retzius and subplate cells, and migrating pyramidal neurons lost polarity and radial orientation. Our findings demonstrate the initial pathological events in dystroglycanopathy mice and contribute to our understanding of how dystroglycan dysfunction affects brain development and progresses to cobblestone lissencephaly.
Collapse
Affiliation(s)
- Naoki Nakagawa
- Department of Biological Chemistry, Human Health Sciences, Graduate School of Medicine, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Hirokazu Yagi
- Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya 467-8603, Japan
| | - Koichi Kato
- 1] Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya 467-8603, Japan [2] Okazaki Institute for Integrative Bioscience and Institute for Molecular Science, National Institutes of Natural Sciences, 5-1 Higashiyama Myodaiji, Okazaki 444-8787, Japan
| | - Hiromu Takematsu
- Department of Biological Chemistry, Human Health Sciences, Graduate School of Medicine, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Shogo Oka
- Department of Biological Chemistry, Human Health Sciences, Graduate School of Medicine, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| |
Collapse
|
88
|
Skorput AGJ, Yeh HH. Effects of ethanol exposure in utero on Cajal-Retzius cells in the developing cortex. Alcohol Clin Exp Res 2015; 39:853-62. [PMID: 25845402 DOI: 10.1111/acer.12696] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 01/31/2015] [Indexed: 11/28/2022]
Abstract
BACKGROUND Prenatal exposure to ethanol exerts teratogenic effects on the developing brain. Here, we tested the hypothesis that exposure to ethanol in utero alters the disposition of Cajal-Retzius cells that play a key role in orchestrating proliferation, migration, and laminar integration of cortical neurons in the embryonic cortex. METHODS Pregnant Ebf2-EGFP mice, harboring EGFP-fluorescent Cajal-Retzius cells, were subjected to a 2% w/w ethanol consumption regimen starting at neural tube closure and lasting throughout gestation. Genesis of Cajal-Retzius cells was assessed by means of 5-bromo-2-deoxyuridine (BrdU) immunofluorescence at embryonic day 12.5, their counts and distribution were determined between postnatal day (P)0 and P4, patch clamp electrophysiology was performed between P2 and P3 to analyze GABA-mediated synaptic activity, and open-field behavioral testing was conducted in P45-P50 adolescents. RESULTS In Ebf2-EGFP embryos exposed to ethanol in utero, we found increased BrdU labeling and expanded distribution of Cajal-Retzius cells in the cortical hem, pointing to increased genesis and proliferation. Postnatally, we found an increase in Cajal-Retzius cell number in cortical layer I. In addition, they displayed altered patterning of spontaneous GABA-mediated synaptic barrages and enhanced GABA-mediated synaptic activity, suggesting enhanced GABAergic tone. CONCLUSIONS These findings, together, underscore that Cajal-Retzius cells contribute to the ethanol-induced aberration of cortical development and abnormal GABAergic neurotransmission at the impactful time when intracortical circuits form.
Collapse
Affiliation(s)
- Alexander G J Skorput
- Department of Physiology and Neurobiology, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire
| | | |
Collapse
|
89
|
Li W, Tang QY, Jadhav AD, Narang A, Qian WX, Shi P, Pang SW. Large-scale topographical screen for investigation of physical neural-guidance cues. Sci Rep 2015; 5:8644. [PMID: 25728549 DOI: 10.1038/srep08644] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 01/29/2015] [Indexed: 12/23/2022] Open
Abstract
A combinatorial approach was used to present primary neurons with a large library of topographical features in the form of micropatterned substrate for high-throughput screening of physical neural-guidance cues that can effectively promote different aspects of neuronal development, including axon and dendritic outgrowth. Notably, the neuronal-guidance capability of specific features was automatically identified using a customized image processing software, thus significantly increasing the screening throughput with minimal subjective bias. Our results indicate that the anisotropic topographies promote axonal and in some cases dendritic extension relative to the isotropic topographies, while dendritic branching showed preference to plain substrates over the microscale features. The results from this work can be readily applied towards engineering novel biomaterials with precise surface topography that can serve as guidance conduits for neuro-regenerative applications. This novel topographical screening strategy combined with the automated processing capability can also be used for high-throughput screening of chemical or genetic regulatory factors in primary neurons.
Collapse
Affiliation(s)
- Wei Li
- 1] Department of Mechanical and Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR, China [2] Center for Biosystems, Neuroscience, and Nanotechnology, City University of Hong Kong, Kowloon, Hong Kong SAR, China
| | - Qing Yuan Tang
- 1] Department of Electronic Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR, China [2] Center for Biosystems, Neuroscience, and Nanotechnology, City University of Hong Kong, Kowloon, Hong Kong SAR, China
| | - Amol D Jadhav
- 1] Department of Mechanical and Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR, China [2] Center for Biosystems, Neuroscience, and Nanotechnology, City University of Hong Kong, Kowloon, Hong Kong SAR, China
| | - Ankit Narang
- 1] Department of Mechanical and Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR, China [2] Center for Biosystems, Neuroscience, and Nanotechnology, City University of Hong Kong, Kowloon, Hong Kong SAR, China
| | - Wei Xian Qian
- 1] Department of Electronic Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR, China [2] Center for Biosystems, Neuroscience, and Nanotechnology, City University of Hong Kong, Kowloon, Hong Kong SAR, China [3] School of Electronic and Optical Engineering, Nanjing University of Science and Technology, Nanjing, Jiangsu, China
| | - Peng Shi
- 1] Department of Mechanical and Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR, China [2] Center for Biosystems, Neuroscience, and Nanotechnology, City University of Hong Kong, Kowloon, Hong Kong SAR, China [3] Shenzhen Research Institute, City University of Hong Kong Shenzhen, Guangdong, China
| | - Stella W Pang
- 1] Department of Electronic Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR, China [2] Center for Biosystems, Neuroscience, and Nanotechnology, City University of Hong Kong, Kowloon, Hong Kong SAR, China
| |
Collapse
|
90
|
Marín-Padilla M. Human cerebral cortex Cajal-Retzius neuron: development, structure and function. A Golgi study. Front Neuroanat 2015; 9:21. [PMID: 25774125 PMCID: PMC4343007 DOI: 10.3389/fnana.2015.00021] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 02/10/2015] [Indexed: 11/27/2022] Open
Abstract
The development, morphology and possible functional activity of the Cajal-Retzius cell of the developing human cerebral cortex are explored herein. The C-RC, of extracortical origin, is the essential neuron of the neocortex first lamina. It receives inputs from afferent fibers that reach the first lamina early in development. Although the origin and function of these original afferent fibers remain unknown, their target is the first lamina sole neuron: the C-RC. This neuron orchestrates the arrival, size and stratification of all pyramidal neurons (of ependymal origin) of the neocortex gray matter. Its axonic terminals spread radially and horizontally throughout the entirety of the first lamina establishing contacts with the dendritic terminals of all gray matter pyramidal cells regardless of size, location and/or eventual functional roles. While the neuron axonic terminals spread radially and horizontally throughout the first lamina, the neuronal’ body undergoes progressive developmental dilution and locating any of them in the adult brain become quite difficult. The neuron bodies are probably retained in the older regions of the neocortex while their axonic collaterals will spread throughout its more recent ones and eventually will extend to great majority of the cortical surface. The neocortex first lamina evolution and composition and that of the C-RC are intertwined and mutually interdependent. It is not possible to understand the C-RC evolving morphology without understanding that of the first lamina. The first lamina composition and its structural and functional organizations obtained with different staining methods may be utterly different. These differences have added unnecessary confusion about its nature. The essential emptiness observed in hematoxylin and eosin preparations (most commonly used) contrast sharply with the concentration of dendrites (the cortex’ largest) obtained using special (MAP-2) stain for dendrites. Only Golgi preparations demonstrate the numerous dendritic and axonic terminals that compose the first lamina basic structure. High power microscopic views of Golgi preparations demonstrate the intimate anatomical and functional interrelationships among dendritic and axonic terminals as well as synaptic contacts between them. The C-RC’ essential morphology does not changes but it is progressively modified by the first lamina increase in thickness and in number of terminal dendrites and their subsequent maturation. This neuron variable morphologic appearance has been the source of controversy. Its morphology depends on the first lamina thickness that may be quite variable among different mammals. In rodents (most commonly used experimental mammal), the first lamina thickness, number and horizontal expansion of dendrites is but a fraction of those in humans. This differences are reflected in the C-RC’ morphology among mammals (including humans) and should not be thought as representing new types of neurons.
Collapse
|
91
|
Qiao S, Homayouni R. Dab2IP Regulates Neuronal Positioning, Rap1 Activity and Integrin Signaling in the Developing Cortex. Dev Neurosci 2015; 37:131-41. [PMID: 25721469 DOI: 10.1159/000369092] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 10/15/2014] [Indexed: 01/10/2023] Open
Abstract
Dab2IP (DOC-2/DAB2 interacting protein) is a GTPase-activating protein which is involved in various aspects of brain development in addition to its roles in tumor formation and apoptosis in other systems. In this study, we carefully examined the expression profile of Dab2IP and investigated its physiological role during brain development using a Dab2IP-knockdown (KD) mouse model created by retroviral insertion of a LacZ-encoding gene-trapping cassette. LacZ staining revealed that Dab2IP is expressed in the ventricular zone as well as the cortical plate and the intermediate zone. Immunohistochemical analysis showed that Dab2IP protein is localized in the leading process and proximal cytoplasmic regions of migrating neurons in the intermediate zone. Bromodeoxyuridine birth dating experiments in combination with immunohistochemical analysis using layer-specific markers showed that Dab2IP is important for proper positioning of a subset of layer II-IV neurons in the developing cortex. Notably, neuronal migration was not completely disrupted in the cerebral cortex of Dab2IP-KD mice and disruption of migration was not strictly layer specific. Previously, we found that Dab2IP regulates multipolar transition in cortical neurons. Others have shown that Rap1 regulates the transition from multipolar to bipolar morphology in migrating postmitotic neurons through N-cadherin signaling and somal translocation in the superficial layer of the cortical plate through integrin signaling. Therefore, we examined whether Rap1 and integrin signaling were affected in Dab2IP-KD brains. We found that Dab2IP-KD resulted in higher levels of activated Rap1 and integrin in the developing cortex. Taken together, our results suggest that Dab2IP plays an important role in the migration and positioning of a subpopulation of later-born (layers II-IV) neurons, likely through the regulation of Rap1 and integrin signaling.
Collapse
Affiliation(s)
- Shuhong Qiao
- Department of Biological Sciences, University of Memphis, Memphis Tenn., USA
| | | |
Collapse
|
92
|
Mandai K, Rikitake Y, Mori M, Takai Y. Nectins and nectin-like molecules in development and disease. Curr Top Dev Biol 2015; 112:197-231. [PMID: 25733141 DOI: 10.1016/bs.ctdb.2014.11.019] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Nectins and nectin-like molecules (Necls)/Cadms are Ca(2+)-independent immunoglobulin superfamily cell adhesion molecules, expressed in most cell types. Nectins mediate not only homotypic but also heterotypic cell-cell adhesion, in contrast to classic cadherins which participate only in homophilic adhesion. Nectins and Necls function in organogenesis of the eye, inner ear, tooth, and cerebral cortex and in a variety of developmental processes including spermatogenesis, axon guidance, synapse formation, and myelination. They are also involved in various diseases, such as viral infection, hereditary ectodermal dysplasia, Alzheimer's disease, autism spectrum disorder, and cancer. Thus, nectins and Necls are crucial for both physiology and pathology. This review summarizes recent advances in research on these cell adhesion molecules in development and pathogenesis.
Collapse
Affiliation(s)
- Kenji Mandai
- Division of Pathogenetic Signaling, Kobe University Graduate School of Medicine, Kobe, Japan; CREST, Japan Science and Technology Agency, Kobe, Japan
| | - Yoshiyuki Rikitake
- CREST, Japan Science and Technology Agency, Kobe, Japan; Division of Signal Transduction, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan; Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Masahiro Mori
- CREST, Japan Science and Technology Agency, Kobe, Japan; Division of Neurophysiology, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Japan; Faculty of Health Sciences, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Yoshimi Takai
- Division of Pathogenetic Signaling, Kobe University Graduate School of Medicine, Kobe, Japan; CREST, Japan Science and Technology Agency, Kobe, Japan.
| |
Collapse
|
93
|
Hirota Y, Kubo KI, Katayama KI, Honda T, Fujino T, Yamamoto TT, Nakajima K. Reelin receptors ApoER2 and VLDLR are expressed in distinct spatiotemporal patterns in developing mouse cerebral cortex. J Comp Neurol 2014; 523:463-78. [PMID: 25308109 DOI: 10.1002/cne.23691] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 10/07/2014] [Accepted: 10/07/2014] [Indexed: 01/31/2023]
Abstract
In mammalian developing brain, neuronal migration is regulated by a variety of signaling cascades, including Reelin signaling. Reelin is a glycoprotein that is mainly secreted by Cajal-Retzius neurons in the marginal zone, playing essential roles in the formation of the layered neocortex via its receptors, apolipoprotein E receptor 2 (ApoER2) and very low density lipoprotein receptor (VLDLR). However, the precise mechanisms by which Reelin signaling controls the neuronal migration process remain unclear. To gain insight into how Reelin signaling controls individual migrating neurons, we generated monoclonal antibodies against ApoER2 and VLDLR and examined the localization of Reelin receptors in the developing mouse cerebral cortex. Immunohistochemical analyses revealed that VLDLR is localized to the distal portion of leading processes in the marginal zone (MZ), whereas ApoER2 is mainly localized to neuronal processes and the cell membranes of multipolar cells in the multipolar cell accumulation zone (MAZ). These different expression patterns may contribute to the distinct actions of Reelin on migrating neurons during both the early and late migratory stages in the developing cerebral cortex.
Collapse
Affiliation(s)
- Yuki Hirota
- Department of Anatomy, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | | | | | | | | | | | | |
Collapse
|
94
|
Trivedi N, Ramahi JS, Karakaya M, Howell D, Kerekes RA, Solecki DJ. Leading-process actomyosin coordinates organelle positioning and adhesion receptor dynamics in radially migrating cerebellar granule neurons. Neural Dev 2014; 9:26. [PMID: 25467954 PMCID: PMC4289176 DOI: 10.1186/1749-8104-9-26] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 09/26/2014] [Indexed: 01/29/2023] Open
Abstract
Background During brain development, neurons migrate from germinal zones to their final positions to assemble neural circuits. A unique saltatory cadence involving cyclical organelle movement (e.g., centrosome motility) and leading-process actomyosin enrichment prior to nucleokinesis organizes neuronal migration. While functional evidence suggests that leading-process actomyosin is essential for centrosome motility, the role of the actin-enriched leading process in globally organizing organelle transport or traction forces remains unexplored. Results We show that myosin ii motors and F-actin dynamics are required for Golgi apparatus positioning before nucleokinesis in cerebellar granule neurons (CGNs) migrating along glial fibers. Moreover, we show that primary cilia are motile organelles, localized to the leading-process F-actin-rich domain and immobilized by pharmacological inhibition of myosin ii and F-actin dynamics. Finally, leading process adhesion dynamics are dependent on myosin ii and F-actin. Conclusions We propose that actomyosin coordinates the overall polarity of migrating CGNs by controlling asymmetric organelle positioning and cell-cell contacts as these cells move along their glial guides. Electronic supplementary material The online version of this article (doi:10.1186/1749-8104-9-26) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | | | - Ryan A Kerekes
- Department of Developmental Neurobiology, St, Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA.
| | | |
Collapse
|
95
|
Optogenetic activation of cajal-retzius cells reveals their glutamatergic output and a novel feedforward circuit in the developing mouse hippocampus. J Neurosci 2014; 34:13018-32. [PMID: 25253849 DOI: 10.1523/jneurosci.1407-14.2014] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Cajal-Retzius cells orchestrate the development of cortical circuits by secreting the glycoprotein reelin. However, their computational functions are still unknown. In fact, the nature of their postsynaptic targets, major neurotransmitter released, as well as the class of postsynaptic receptors activated by their firing remain unclear. Here, we have addressed these questions by activating Cajal-Retzius cells optogenetically in mouse hippocampal slices. Light delivered to stratum lacunosum-moleculare triggered EPSCs both on local interneurons and on pyramidal cells. Responses recorded under voltage-clamp conditions had identical short latencies and similar amplitudes, but were kinetically different (i.e., faster in interneurons vs pyramidal cells). In both cases, responses were blocked by TTX, indicating that they were generated by action potential-dependent release. Responses in interneurons were rescued by the addition of 4-AP to TTX, and decreased when presynaptic firing in Cajal-Retzius cells was reduced by the chemokine CXCL12, indicating the existence of a direct Cajal-Retzius cell-interneuron monosynaptic connection. Although the combined application of 4-AP and TTX did not rescue responses in pyramidal cells, neither were they affected by the GABAA receptor blocker gabazine, which would be expected if they were polysynaptic. Both connections showed physiological and pharmacological properties indicating the involvement of AMPA- and NMDA-type glutamate receptors. The connectivity from presynaptic Cajal-Retzius cells to interneurons was strong enough to generate long-latency feedforward GABAergic input onto pyramidal cells. We propose that this newly defined Cajal-Retzius cell-dependent microcircuit may regulate synaptic plasticity and dendritic development in stratum lacunosum-moleculare, thus impacting the integrative properties of the developing hippocampus.
Collapse
|
96
|
Cooper JA. Molecules and mechanisms that regulate multipolar migration in the intermediate zone. Front Cell Neurosci 2014; 8:386. [PMID: 25452716 PMCID: PMC4231986 DOI: 10.3389/fncel.2014.00386] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 10/29/2014] [Indexed: 01/05/2023] Open
Abstract
Most neurons migrate with an elongated, “bipolar” morphology, extending a long leading process that explores the environment. However, when immature projection neurons enter the intermediate zone (IZ) of the neocortex they become “multipolar”. Multipolar cells extend and retract cytoplasmic processes in different directions and move erratically—sideways, up and down. Multipolar cells extend axons while they are in the lower half of the IZ. Remarkably, the cells then resume radial migration: they reorient their centrosome and Golgi apparatus towards the pia, transform back to bipolar morphology, and commence locomotion along radial glia (RG) fibers. This reorientation implies the existence of directional signals in the IZ that are ignored during the multipolar stage but sensed after axonogenesis. In vivo genetic manipulation has implicated a variety of candidate directional signals, cell surface receptors, and signaling pathways, that may be involved in polarizing multipolar cells and stabilizing a pia-directed leading process for radial migration. Other signals are implicated in starting multipolar migration and triggering axon outgrowth. Here we review the molecules and mechanisms that regulate multipolar migration, and also discuss how multipolar migration affects the orderly arrangement of neurons in layers and columns in the developing neocortex.
Collapse
Affiliation(s)
- Jonathan A Cooper
- Fred Hutchinson Cancer Research Center, Division of Basic Sciences Seattle, Washington, USA
| |
Collapse
|
97
|
Kobayashi R, Kurita S, Miyata M, Maruo T, Mandai K, Rikitake Y, Takai Y. s-Afadin binds more preferentially to the cell adhesion molecules nectins than l-afadin. Genes Cells 2014; 19:853-63. [DOI: 10.1111/gtc.12185] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 08/25/2014] [Indexed: 12/30/2022]
Affiliation(s)
- Reiko Kobayashi
- Division of Pathogenetic Signaling; Department of Biochemistry and Molecular Biology; Kobe University Graduate School of Medicine; Kobe Hyogo 650-0047 Japan
- Division of Molecular and Cellular Biology; Department of Biochemistry and Molecular Biology; Kobe University Graduate School of Medicine; Kobe Hyogo 650-0017 Japan
| | - Souichi Kurita
- Division of Pathogenetic Signaling; Department of Biochemistry and Molecular Biology; Kobe University Graduate School of Medicine; Kobe Hyogo 650-0047 Japan
- Division of Molecular and Cellular Biology; Department of Biochemistry and Molecular Biology; Kobe University Graduate School of Medicine; Kobe Hyogo 650-0017 Japan
| | - Muneaki Miyata
- Division of Signal Transduction; Department of Biochemistry and Molecular Biology; Kobe University Graduate School of Medicine; Kobe Hyogo 650-0017 Japan
| | - Tomohiko Maruo
- Division of Pathogenetic Signaling; Department of Biochemistry and Molecular Biology; Kobe University Graduate School of Medicine; Kobe Hyogo 650-0047 Japan
- Division of Molecular and Cellular Biology; Department of Biochemistry and Molecular Biology; Kobe University Graduate School of Medicine; Kobe Hyogo 650-0017 Japan
| | - Kenji Mandai
- Division of Pathogenetic Signaling; Department of Biochemistry and Molecular Biology; Kobe University Graduate School of Medicine; Kobe Hyogo 650-0047 Japan
- Division of Molecular and Cellular Biology; Department of Biochemistry and Molecular Biology; Kobe University Graduate School of Medicine; Kobe Hyogo 650-0017 Japan
| | - Yoshiyuki Rikitake
- Division of Molecular and Cellular Biology; Department of Biochemistry and Molecular Biology; Kobe University Graduate School of Medicine; Kobe Hyogo 650-0017 Japan
- Division of Signal Transduction; Department of Biochemistry and Molecular Biology; Kobe University Graduate School of Medicine; Kobe Hyogo 650-0017 Japan
| | - Yoshimi Takai
- Division of Pathogenetic Signaling; Department of Biochemistry and Molecular Biology; Kobe University Graduate School of Medicine; Kobe Hyogo 650-0047 Japan
- Division of Molecular and Cellular Biology; Department of Biochemistry and Molecular Biology; Kobe University Graduate School of Medicine; Kobe Hyogo 650-0017 Japan
| |
Collapse
|
98
|
Ka M, Jung EM, Mueller U, Kim WY. MACF1 regulates the migration of pyramidal neurons via microtubule dynamics and GSK-3 signaling. Dev Biol 2014; 395:4-18. [PMID: 25224226 DOI: 10.1016/j.ydbio.2014.09.009] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 08/13/2014] [Accepted: 09/05/2014] [Indexed: 10/24/2022]
Abstract
Neuronal migration and subsequent differentiation play critical roles for establishing functional neural circuitry in the developing brain. However, the molecular mechanisms that regulate these processes are poorly understood. Here, we show that microtubule actin crosslinking factor 1 (MACF1) determines neuronal positioning by regulating microtubule dynamics and mediating GSK-3 signaling during brain development. First, using MACF1 floxed allele mice and in utero gene manipulation, we find that MACF1 deletion suppresses migration of cortical pyramidal neurons and results in aberrant neuronal positioning in the developing brain. The cell autonomous deficit in migration is associated with abnormal dynamics of leading processes and centrosomes. Furthermore, microtubule stability is severely damaged in neurons lacking MACF1, resulting in abnormal microtubule dynamics. Finally, MACF1 interacts with and mediates GSK-3 signaling in developing neurons. Our findings establish a cellular mechanism underlying neuronal migration and provide insights into the regulation of cytoskeleton dynamics in developing neurons.
Collapse
Affiliation(s)
- Minhan Ka
- Developmental Neuroscience, Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Eui-Man Jung
- Developmental Neuroscience, Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Ulrich Mueller
- Dorris Neuroscience Center and Department of Cell Biology, The Scripps Research Institute, La Jolla, CA 92037, United States
| | - Woo-Yang Kim
- Developmental Neuroscience, Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, NE 68198, United States.
| |
Collapse
|
99
|
Yu D, Fan W, Wu P, Deng J, Liu J, Niu Y, Li M, Deng J. Characterization of hippocampal Cajal-Retzius cells during development in a mouse model of Alzheimer's disease (Tg2576). Neural Regen Res 2014; 9:394-401. [PMID: 25206826 PMCID: PMC4146192 DOI: 10.4103/1673-5374.128243] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/22/2014] [Indexed: 12/05/2022] Open
Abstract
Cajal-Retzius cells are reelin-secreting neurons in the marginal zone of the neocortex and hippocampus. The aim of this study was to investigate Cajal-Retzius cells in Alzheimer's disease pathology. Results revealed that the number of Cajal-Retzius cells markedly reduced with age in both wild type and in mice over-expressing the Swedish double mutant form of amyloid precursor protein 695 (transgenic (Tg) 2576 mice). Numerous reelin-positive neurons were positive for activated caspase 3 in Tg2576 mice, suggesting that Cajal-Retzius neuronal loss occurred via apoptosis in this Alzheimer's disease model. Compared with wild type, the number of Cajal-Retzius cells was significantly lower in Tg2576 mice. Western blot analysis confirmed that reelin levels were markedly lower in Tg2576 mice than in wild-type mice. The decline in Cajal-Retzius cells in Tg2576 mice was found to occur concomitantly with the onset of Alzheimer's disease amyloid pathology and related behavioral deficits. Overall, these data indicated that Cajal-Retzius cell loss occurred with the onset and development of Alzheimer's disease.
Collapse
Affiliation(s)
- Dongming Yu
- Institute of Neurobiology, School of Life Science, Henan University, Kaifeng, Henan Province, China
| | - Wenjuan Fan
- Laboratory of Molecular Medicine, Luohe Medical College, Luohe, Henan Province, China
| | - Ping Wu
- Institute of Neurobiology, School of Life Science, Henan University, Kaifeng, Henan Province, China
| | - Jiexin Deng
- Institute of Neurobiology, School of Life Science, Henan University, Kaifeng, Henan Province, China
| | - Jing Liu
- Institute of Neurobiology, School of Life Science, Henan University, Kaifeng, Henan Province, China
| | - Yanli Niu
- Institute of Neurobiology, School of Life Science, Henan University, Kaifeng, Henan Province, China
| | - Mingshan Li
- Institute of Neurobiology, School of Life Science, Henan University, Kaifeng, Henan Province, China
| | - Jinbo Deng
- Institute of Neurobiology, School of Life Science, Henan University, Kaifeng, Henan Province, China
| |
Collapse
|
100
|
Cdk5-mediated phosphorylation of RapGEF2 controls neuronal migration in the developing cerebral cortex. Nat Commun 2014; 5:4826. [PMID: 25189171 PMCID: PMC4164783 DOI: 10.1038/ncomms5826] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 07/25/2014] [Indexed: 12/16/2022] Open
Abstract
During cerebral cortex development, pyramidal neurons migrate through the intermediate zone and integrate into the cortical plate. These neurons undergo the multipolar-bipolar transition to initiate radial migration. While perturbation of this polarity acquisition leads to cortical malformations, how this process is initiated and regulated is largely unknown. Here we report that the specific upregulation of the Rap1 guanine nucleotide exchange factor, RapGEF2, in migrating neurons corresponds to the timing of this polarity transition. In utero electroporation and live-imaging studies reveal that RapGEF2 acts on the multipolar-bipolar transition during neuronal migration via a Rap1/N-cadherin pathway. Importantly, activation of RapGEF2 is controlled via phosphorylation by a serine/threonine kinase Cdk5, whose activity is largely restricted to the radial migration zone. Thus, the specific expression and Cdk5-dependent phosphorylation of RapGEF2 during multipolar-bipolar transition within the intermediate zone are essential for proper neuronal migration and wiring of the cerebral cortex.
Collapse
|