1
|
Wojnacki J, Quassollo G, Bordenave MD, Unsain N, Martínez GF, Szalai AM, Pertz O, Gundersen GG, Bartolini F, Stefani FD, Cáceres A, Bisbal M. Dual spatio-temporal regulation of axon growth and microtubule dynamics by RhoA signaling pathways. J Cell Sci 2024; 137:jcs261970. [PMID: 38910449 DOI: 10.1242/jcs.261970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 06/17/2024] [Indexed: 06/25/2024] Open
Abstract
RhoA plays a crucial role in neuronal polarization, where its action restraining axon outgrowth has been thoroughly studied. We now report that RhoA has not only an inhibitory but also a stimulatory effect on axon development depending on when and where exerts its action and the downstream effectors involved. In cultured hippocampal neurons, FRET imaging revealed that RhoA activity selectively localized in growth cones of undifferentiated neurites, whereas in developing axons it displayed a biphasic pattern, being low in nascent axons and high in elongating ones. RhoA-Rho kinase (ROCK) signaling prevented axon initiation but had no effect on elongation, whereas formin inhibition reduced axon extension without significantly altering initial outgrowth. In addition, RhoA-mDia signaling promoted axon elongation by stimulating growth cone microtubule stability and assembly, as opposed to RhoA-ROCK signaling, which restrained growth cone microtubule assembly and protrusion.
Collapse
Affiliation(s)
- José Wojnacki
- Instituto de Investigación Médica Mercedes y Martín Ferreyra (INIMEC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Córdoba, Córdoba 5016, Argentina
| | - Gonzalo Quassollo
- Instituto de Investigación Médica Mercedes y Martín Ferreyra (INIMEC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Córdoba, Córdoba 5016, Argentina
| | - Martín D Bordenave
- Centro de Investigaciones en Bionanociencias (CIBION), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Godoy Cruz 2390, Ciudad Autónoma de Buenos Aires C1425FQD, Argentina
| | - Nicolás Unsain
- Instituto de Investigación Médica Mercedes y Martín Ferreyra (INIMEC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Córdoba, Córdoba 5016, Argentina
- Instituto Universitario Ciencias Biomédicas de Córdoba (IUCBC), Córdoba 5016, Argentina
| | - Gaby F Martínez
- Instituto de Investigación Médica Mercedes y Martín Ferreyra (INIMEC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Córdoba, Córdoba 5016, Argentina
| | - Alan M Szalai
- Centro de Investigaciones en Bionanociencias (CIBION), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Godoy Cruz 2390, Ciudad Autónoma de Buenos Aires C1425FQD, Argentina
| | - Olivier Pertz
- Institute of Cell Biology, University of Bern, Baltzerstrasse 4, Bern 3012, Switzerland
| | - Gregg G Gundersen
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Francesca Bartolini
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Fernando D Stefani
- Centro de Investigaciones en Bionanociencias (CIBION), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Godoy Cruz 2390, Ciudad Autónoma de Buenos Aires C1425FQD, Argentina
- Departamento de Física, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Güiraldes 2620, Ciudad Autónoma de Buenos Aires C1428EHA, Argentina
| | - Alfredo Cáceres
- Centro Investigación Medicina Traslacional Severo R Amuchástegui (CIMETSA), Instituto Universitario Ciencias Biomédicas Córdoba (IUCBC), Av. Naciones Unidas 440, Córdoba 5016, Argentina
| | - Mariano Bisbal
- Instituto de Investigación Médica Mercedes y Martín Ferreyra (INIMEC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Córdoba, Córdoba 5016, Argentina
- Instituto Universitario Ciencias Biomédicas de Córdoba (IUCBC), Córdoba 5016, Argentina
| |
Collapse
|
2
|
Goo BS, Mun DJ, Kim S, Nhung TTM, Lee SB, Woo Y, Kim SJ, Suh BK, Park SJ, Lee HE, Park K, Jang H, Rah JC, Yoon KJ, Baek ST, Park SY, Park SK. Schizophrenia-associated Mitotic Arrest Deficient-1 (MAD1) regulates the polarity of migrating neurons in the developing neocortex. Mol Psychiatry 2023; 28:856-870. [PMID: 36357673 PMCID: PMC9908555 DOI: 10.1038/s41380-022-01856-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/20/2022] [Accepted: 10/24/2022] [Indexed: 11/11/2022]
Abstract
Although large-scale genome-wide association studies (GWAS) have identified an association between MAD1L1 (Mitotic Arrest Deficient-1 Like 1) and the pathology of schizophrenia, the molecular mechanisms underlying this association remain unclear. In the present study, we aimed to address these mechanisms by examining the role of MAD1 (the gene product of MAD1L1) in key neurodevelopmental processes in mice and human organoids. Our findings indicated that MAD1 is highly expressed during active cortical development and that MAD1 deficiency leads to impairments in neuronal migration and neurite outgrowth. We also observed that MAD1 is localized to the Golgi apparatus and regulates vesicular trafficking from the Golgi apparatus to the plasma membrane, which is required for the growth and polarity of migrating neurons. In this process, MAD1 physically interacts and collaborates with the kinesin-like protein KIFC3 (kinesin family member C3) to regulate the morphology of the Golgi apparatus and neuronal polarity, thereby ensuring proper neuronal migration and differentiation. Consequently, our findings indicate that MAD1 is an essential regulator of neuronal development and that alterations in MAD1 may underlie schizophrenia pathobiology.
Collapse
Affiliation(s)
- Bon Seong Goo
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Dong Jin Mun
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Seunghyun Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Truong Thi My Nhung
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Su Been Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Youngsik Woo
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Soo Jeong Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Bo Kyoung Suh
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Sung Jin Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, 01655, USA
| | - Hee-Eun Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Kunyou Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Hyunsoo Jang
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Jong-Cheol Rah
- Korea Brain Research Institute, Daegu, 41062, Republic of Korea
| | - Ki-Jun Yoon
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Seung Tae Baek
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Seung-Yeol Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea.
| | - Sang Ki Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea.
| |
Collapse
|
3
|
Zhao X, Liang W, Wang W, Liu H, Zhang X, Liu C, Zhu C, Cui B, Tang Y, Liu S. Changes in and asymmetry of the proteome in the human fetal frontal lobe during early development. Commun Biol 2022; 5:1031. [PMID: 36175510 PMCID: PMC9522861 DOI: 10.1038/s42003-022-04003-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 09/15/2022] [Indexed: 12/05/2022] Open
Abstract
Inherent hemispheric asymmetry is important for cognition, language and other functions. Describing normal brain and asymmetry development during early development will improve our understanding of how different hemispheres prioritize specific functions, which is currently unknown. Here, we analysed developmental changes in and asymmetry of the proteome in the bilateral frontal lobes of three foetal specimens in the late first trimester of pregnancy. We found that during this period, the difference in expression between gestational weeks (GWs) increased, and the difference in asymmetric expression decreased. Changes in the patterns of protein expression differed in the bilateral frontal lobes. Our results show that brain asymmetry can be observed in early development. These findings can guide researchers in further investigations of the mechanisms of brain asymmetry. We propose that both sides of the brain should be analysed separately in future multiomics and human brain mapping studies.
Collapse
Affiliation(s)
- Xiaotian Zhao
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Provincial Key Laboratory of Mental Disorder, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, Shandong, China
| | - Wenjia Liang
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Provincial Key Laboratory of Mental Disorder, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, Shandong, China
| | - Wenjun Wang
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Provincial Key Laboratory of Mental Disorder, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, Shandong, China
| | - Hailan Liu
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Provincial Key Laboratory of Mental Disorder, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, Shandong, China
| | - Xiaolei Zhang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Chengxin Liu
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Provincial Key Laboratory of Mental Disorder, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, Shandong, China
| | - Caiting Zhu
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Provincial Key Laboratory of Mental Disorder, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, Shandong, China
| | - Baoxia Cui
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Yuchun Tang
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Provincial Key Laboratory of Mental Disorder, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.
- Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, Shandong, China.
| | - Shuwei Liu
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Provincial Key Laboratory of Mental Disorder, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.
- Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, Shandong, China.
| |
Collapse
|
4
|
Wilson C, Moyano AL, Cáceres A. Perspectives on Mechanisms Supporting Neuronal Polarity From Small Animals to Humans. Front Cell Dev Biol 2022; 10:878142. [PMID: 35517494 PMCID: PMC9062071 DOI: 10.3389/fcell.2022.878142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 04/04/2022] [Indexed: 11/23/2022] Open
Abstract
Axon-dendrite formation is a crucial milestone in the life history of neurons. During this process, historically referred as “the establishment of polarity,” newborn neurons undergo biochemical, morphological and functional transformations to generate the axonal and dendritic domains, which are the basis of neuronal wiring and connectivity. Since the implementation of primary cultures of rat hippocampal neurons by Gary Banker and Max Cowan in 1977, the community of neurobiologists has made significant achievements in decoding signals that trigger axo-dendritic specification. External and internal cues able to switch on/off signaling pathways controlling gene expression, protein stability, the assembly of the polarity complex (i.e., PAR3-PAR6-aPKC), cytoskeleton remodeling and vesicle trafficking contribute to shape the morphology of neurons. Currently, the culture of hippocampal neurons coexists with alternative model systems to study neuronal polarization in several species, from single-cell to whole-organisms. For instance, in vivo approaches using C. elegans and D. melanogaster, as well as in situ imaging in rodents, have refined our knowledge by incorporating new variables in the polarity equation, such as the influence of the tissue, glia-neuron interactions and three-dimensional development. Nowadays, we have the unique opportunity of studying neurons differentiated from human induced pluripotent stem cells (hiPSCs), and test hypotheses previously originated in small animals and propose new ones perhaps specific for humans. Thus, this article will attempt to review critical mechanisms controlling polarization compiled over decades, highlighting points to be considered in new experimental systems, such as hiPSC neurons and human brain organoids.
Collapse
|
5
|
BARS Influences Neuronal Development by Regulation of Post-Golgi Trafficking. Cells 2022; 11:cells11081320. [PMID: 35455998 PMCID: PMC9026897 DOI: 10.3390/cells11081320] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/28/2022] [Accepted: 04/08/2022] [Indexed: 02/01/2023] Open
Abstract
Neurons are highly polarized cells requiring precise regulation of trafficking and targeting of membrane proteins to generate and maintain different and specialized compartments, such as axons and dendrites. Disruption of the Golgi apparatus (GA) secretory pathway in developing neurons alters axon/dendritic formation. Therefore, detailed knowledge of the mechanisms underlying vesicles exiting from the GA is crucial for understanding neuronal polarity. In this study, we analyzed the role of Brefeldin A-Ribosylated Substrate (CtBP1-S/BARS), a member of the C-terminal-binding protein family, in the regulation of neuronal morphological polarization and the exit of membrane proteins from the Trans Golgi Network. Here, we show that BARS is expressed during neuronal development in vitro and that RNAi suppression of BARS inhibits axonal and dendritic elongation in hippocampal neuronal cultures as well as largely perturbed neuronal migration and multipolar-to-bipolar transition during cortical development in situ. In addition, using plasma membrane (PM) proteins fused to GFP and engineered with reversible aggregation domains, we observed that expression of fission dominant-negative BARS delays the exit of dendritic and axonal membrane protein-containing carriers from the GA. Taken together, these data provide the first set of evidence suggesting a role for BARS in neuronal development by regulating post-Golgi membrane trafficking.
Collapse
|
6
|
Lamb AK, Fernandez AN, Peersen OB, Di Pietro SM. The dynein light chain protein Tda2 functions as a dimerization engine to regulate actin capping protein during endocytosis. Mol Biol Cell 2021; 32:1459-1473. [PMID: 34081539 PMCID: PMC8351736 DOI: 10.1091/mbc.e21-01-0032] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Clathrin- and actin-mediated endocytosis is a fundamental process in eukaryotic cells. Previously, we discovered Tda2 as a new yeast dynein light chain (DLC) that works with Aim21 to regulate actin assembly during endocytosis. Here we show Tda2 functions as a dimerization engine bringing two Aim21 molecules together using a novel binding surface different than the canonical DLC ligand binding groove. Point mutations on either protein that diminish the Tda2-Aim21 interaction in vitro cause the same in vivo phenotype as TDA2 deletion showing reduced actin capping protein (CP) recruitment and increased filamentous actin at endocytic sites. Remarkably, chemically induced dimerization of Aim21 rescues the endocytic phenotype of TDA2 deletion. We also uncovered a CP interacting motif in Aim21, expanding its function to a fundamental cellular pathway and showing such motif exists outside mammalian cells. Furthermore, specific disruption of this motif causes the same deficit of actin CP recruitment and increased filamentous actin at endocytic sites as AIM21 deletion. Thus, the data indicate the Tda2-Aim21 complex functions in actin assembly primarily through CP regulation. Collectively, our results provide a mechanistic view of the Tda2-Aim21 complex and its function in actin network regulation at endocytic sites.
Collapse
Affiliation(s)
- Andrew K Lamb
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523-1870
| | - Andres N Fernandez
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523-1870
| | - Olve B Peersen
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523-1870
| | - Santiago M Di Pietro
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523-1870
| |
Collapse
|
7
|
Wilson C, Giono LE, Rozés-Salvador V, Fiszbein A, Kornblihtt AR, Cáceres A. The Histone Methyltransferase G9a Controls Axon Growth by Targeting the RhoA Signaling Pathway. Cell Rep 2021; 31:107639. [PMID: 32402271 DOI: 10.1016/j.celrep.2020.107639] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 03/18/2020] [Accepted: 04/21/2020] [Indexed: 12/19/2022] Open
Abstract
The generation of axonal and dendritic domains is critical for brain circuitry assembly and physiology. Negative players, such as the RhoA-Rho coiled-coil-associated protein kinase (ROCK) signaling pathway, restrain axon development and polarization. Surprisingly, the genetic control of neuronal polarity has remained largely unexplored. Here, we report that, in primary cultured neurons, expression of the histone methyltransferase G9a and nuclear translocation of its major splicing isoform (G9a/E10+) peak at the time of axon formation. RNAi suppression of G9a/E10+ or pharmacological blockade of G9a constrains neuronal migration, axon initiation, and the establishment of neuronal polarity in situ and in vitro. Inhibition of G9a function upregulates RhoA-ROCK activity by increasing the expression of Lfc, a guanine nucleotide exchange factor (GEF) for RhoA. Together, these results identify G9a as a player in neuronal polarization.
Collapse
Affiliation(s)
- Carlos Wilson
- Instituto de Investigación Médica Mercedes y Martín Ferreyra (INIMEC-CONICET-UNC) Friuli 2434, 5016 Córdoba, Argentina; Universidad Nacional de Córdoba (UNC), Av. Haya de la Torre s/n, 5000 Córdoba, Argentina; Centro de Investigación en Medicina Traslacional "Severo R Amuchástegui" (CIMETSA), Instituto Universitario Ciencias Biomédicas Córdoba (IUCBC), Av. Friuli 2786, 5016 Córdoba, Argentina
| | - Luciana E Giono
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE-UBA-CONICET) and Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, C1428EHA Buenos Aires, Argentina
| | - Victoria Rozés-Salvador
- Instituto de Investigación Médica Mercedes y Martín Ferreyra (INIMEC-CONICET-UNC) Friuli 2434, 5016 Córdoba, Argentina; Universidad Nacional de Córdoba (UNC), Av. Haya de la Torre s/n, 5000 Córdoba, Argentina
| | - Ana Fiszbein
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE-UBA-CONICET) and Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, C1428EHA Buenos Aires, Argentina
| | - Alberto R Kornblihtt
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE-UBA-CONICET) and Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, C1428EHA Buenos Aires, Argentina
| | - Alfredo Cáceres
- Instituto de Investigación Médica Mercedes y Martín Ferreyra (INIMEC-CONICET-UNC) Friuli 2434, 5016 Córdoba, Argentina; Universidad Nacional de Córdoba (UNC), Av. Haya de la Torre s/n, 5000 Córdoba, Argentina; Centro de Investigación en Medicina Traslacional "Severo R Amuchástegui" (CIMETSA), Instituto Universitario Ciencias Biomédicas Córdoba (IUCBC), Av. Friuli 2786, 5016 Córdoba, Argentina.
| |
Collapse
|
8
|
Wilson C, Cáceres A. New insights on epigenetic mechanisms supporting axonal development: histone marks and miRNAs. FEBS J 2020; 288:6353-6364. [PMID: 33332753 DOI: 10.1111/febs.15673] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/21/2020] [Accepted: 12/15/2020] [Indexed: 11/27/2022]
Abstract
Mechanisms supporting axon growth and the establishment of neuronal polarity have remained largely disconnected from their genetic and epigenetic fundamentals. Recently, post-transcriptional modifications of histones involved in chromatin folding and transcription, and microRNAs controlling translation have emerged as regulators of axonal specification, growth, and guidance. In this article, we review novel evidence supporting the concept that epigenetic mechanisms work at both transcriptional and post-transcriptional levels to shape axons. We also discuss the role of splicing on axonal growth, as one of the most (if not the most) powerful post-transcriptional mechanism to diversify genetic information. Overall, we think exploring the gap between epigenetics and axonal growth raises new questions and perspectives to the development of axons in physiological and pathological contexts.
Collapse
Affiliation(s)
- Carlos Wilson
- Centro de Investigación en Medicina Traslacional "Severo R Amuchástegui" (CIMETSA), Instituto Universitario Ciencias Biomédicas Córdoba (IUCBC), Córdoba, Argentina.,Instituto de Investigación Médica Mercedes y Martín Ferreyra (INIMEC-CONICET-UNC), Córdoba, Argentina.,Universidad Nacional de Córdoba (UNC), Argentina
| | - Alfredo Cáceres
- Centro de Investigación en Medicina Traslacional "Severo R Amuchástegui" (CIMETSA), Instituto Universitario Ciencias Biomédicas Córdoba (IUCBC), Córdoba, Argentina
| |
Collapse
|
9
|
Sekiguchi M, Sobue A, Kushima I, Wang C, Arioka Y, Kato H, Kodama A, Kubo H, Ito N, Sawahata M, Hada K, Ikeda R, Shinno M, Mizukoshi C, Tsujimura K, Yoshimi A, Ishizuka K, Takasaki Y, Kimura H, Xing J, Yu Y, Yamamoto M, Okada T, Shishido E, Inada T, Nakatochi M, Takano T, Kuroda K, Amano M, Aleksic B, Yamomoto T, Sakuma T, Aida T, Tanaka K, Hashimoto R, Arai M, Ikeda M, Iwata N, Shimamura T, Nagai T, Nabeshima T, Kaibuchi K, Yamada K, Mori D, Ozaki N. ARHGAP10, which encodes Rho GTPase-activating protein 10, is a novel gene for schizophrenia risk. Transl Psychiatry 2020; 10:247. [PMID: 32699248 PMCID: PMC7376022 DOI: 10.1038/s41398-020-00917-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 06/12/2020] [Accepted: 07/03/2020] [Indexed: 02/07/2023] Open
Abstract
Schizophrenia (SCZ) is known to be a heritable disorder; however, its multifactorial nature has significantly hampered attempts to establish its pathogenesis. Therefore, in this study, we performed genome-wide copy-number variation (CNV) analysis of 2940 patients with SCZ and 2402 control subjects and identified a statistically significant association between SCZ and exonic CNVs in the ARHGAP10 gene. ARHGAP10 encodes a member of the RhoGAP superfamily of proteins that is involved in small GTPase signaling. This signaling pathway is one of the SCZ-associated pathways and may contribute to neural development and function. However, the ARHGAP10 gene is often confused with ARHGAP21, thus, the significance of ARHGAP10 in the molecular pathology of SCZ, including the expression profile of the ARHGAP10 protein, remains poorly understood. To address this issue, we focused on one patient identified to have both an exonic deletion and a missense variant (p.S490P) in ARHGAP10. The missense variant was found to be located in the RhoGAP domain and was determined to be relevant to the association between ARHGAP10 and the active form of RhoA. We evaluated ARHGAP10 protein expression in the brains of reporter mice and generated a mouse model to mimic the patient case. The model exhibited abnormal emotional behaviors, along with reduced spine density in the medial prefrontal cortex (mPFC). In addition, primary cultured neurons prepared from the mouse model brain exhibited immature neurites in vitro. Furthermore, we established induced pluripotent stem cells (iPSCs) from this patient, and differentiated them into tyrosine hydroxylase (TH)-positive neurons in order to analyze their morphological phenotypes. TH-positive neurons differentiated from the patient-derived iPSCs exhibited severe defects in both neurite length and branch number; these defects were restored by the addition of the Rho-kinase inhibitor, Y-27632. Collectively, our findings suggest that rare ARHGAP10 variants may be genetically and biologically associated with SCZ and indicate that Rho signaling represents a promising drug discovery target for SCZ treatment.
Collapse
Affiliation(s)
- Mariko Sekiguchi
- grid.27476.300000 0001 0943 978XDepartment of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan ,grid.27476.300000 0001 0943 978XDepartment of Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Akira Sobue
- grid.27476.300000 0001 0943 978XDepartment of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University, Graduate School of Medicine, Nagoya, Aichi Japan
| | - Itaru Kushima
- grid.27476.300000 0001 0943 978XDepartment of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan ,grid.437848.40000 0004 0569 8970Medical Genomics Center, Nagoya University Hospital, Nagoya, Aichi Japan
| | - Chenyao Wang
- grid.27476.300000 0001 0943 978XDepartment of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Yuko Arioka
- grid.27476.300000 0001 0943 978XDepartment of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan ,grid.437848.40000 0004 0569 8970Center for Advanced Medicine and Clinical Research, Nagoya University Hospital, Nagoya, Aichi Japan
| | - Hidekazu Kato
- grid.27476.300000 0001 0943 978XDepartment of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Akiko Kodama
- grid.27476.300000 0001 0943 978XDepartment of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan ,grid.27476.300000 0001 0943 978XDepartment of Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Hisako Kubo
- grid.27476.300000 0001 0943 978XDepartment of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Norimichi Ito
- grid.27476.300000 0001 0943 978XDepartment of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University, Graduate School of Medicine, Nagoya, Aichi Japan
| | - Masahito Sawahata
- grid.27476.300000 0001 0943 978XDepartment of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University, Graduate School of Medicine, Nagoya, Aichi Japan
| | - Kazuhiro Hada
- grid.27476.300000 0001 0943 978XDepartment of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University, Graduate School of Medicine, Nagoya, Aichi Japan
| | - Ryosuke Ikeda
- grid.27476.300000 0001 0943 978XDepartment of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan ,grid.27476.300000 0001 0943 978XDepartment of Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan ,grid.27476.300000 0001 0943 978XDepartment of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University, Graduate School of Medicine, Nagoya, Aichi Japan
| | - Mio Shinno
- grid.27476.300000 0001 0943 978XDepartment of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan ,grid.27476.300000 0001 0943 978XDepartment of Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan ,grid.27476.300000 0001 0943 978XDepartment of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University, Graduate School of Medicine, Nagoya, Aichi Japan
| | - Chikara Mizukoshi
- grid.27476.300000 0001 0943 978XDepartment of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Keita Tsujimura
- grid.27476.300000 0001 0943 978XDepartment of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Akira Yoshimi
- grid.27476.300000 0001 0943 978XDepartment of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Kanako Ishizuka
- grid.27476.300000 0001 0943 978XDepartment of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Yuto Takasaki
- grid.27476.300000 0001 0943 978XDepartment of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Hiroki Kimura
- grid.27476.300000 0001 0943 978XDepartment of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Jingrui Xing
- grid.27476.300000 0001 0943 978XDepartment of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Yanjie Yu
- grid.27476.300000 0001 0943 978XDepartment of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Maeri Yamamoto
- grid.27476.300000 0001 0943 978XDepartment of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Takashi Okada
- grid.27476.300000 0001 0943 978XDepartment of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Emiko Shishido
- grid.27476.300000 0001 0943 978XDepartment of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Toshiya Inada
- grid.27476.300000 0001 0943 978XDepartment of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Masahiro Nakatochi
- grid.27476.300000 0001 0943 978XDivision of Data Science, Department of Nursing, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Tetsuya Takano
- grid.27476.300000 0001 0943 978XDepartment of Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Keisuke Kuroda
- grid.27476.300000 0001 0943 978XDepartment of Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Mutsuki Amano
- grid.27476.300000 0001 0943 978XDepartment of Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Branko Aleksic
- grid.27476.300000 0001 0943 978XDepartment of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Takashi Yamomoto
- grid.257022.00000 0000 8711 3200Division of Integrated Sciences for Life, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Tetsushi Sakuma
- grid.257022.00000 0000 8711 3200Division of Integrated Sciences for Life, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Tomomi Aida
- grid.265073.50000 0001 1014 9130Laboratory of Molecular Neuroscience, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kohichi Tanaka
- grid.265073.50000 0001 1014 9130Laboratory of Molecular Neuroscience, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Ryota Hashimoto
- grid.419280.60000 0004 1763 8916Department of Pathology of Mental Diseases, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan ,grid.136593.b0000 0004 0373 3971Molecular Research Center for Children’s Mental Development, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan ,grid.136593.b0000 0004 0373 3971Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Makoto Arai
- grid.272456.0Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Masashi Ikeda
- grid.256115.40000 0004 1761 798XDepartment of Psychiatry, Fujita Health University School of Medicine, Toyoake, Japan
| | - Nakao Iwata
- grid.256115.40000 0004 1761 798XDepartment of Psychiatry, Fujita Health University School of Medicine, Toyoake, Japan
| | - Teppei Shimamura
- grid.27476.300000 0001 0943 978XDivision of Systems Biology, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Taku Nagai
- grid.27476.300000 0001 0943 978XDepartment of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University, Graduate School of Medicine, Nagoya, Aichi Japan
| | - Toshitaka Nabeshima
- Advanced Diagnostic System Research Laboratory Fujita Health University, Graduate School of Health Sciences & Aino University, Toyoake, Aichi Japan
| | - Kozo Kaibuchi
- grid.27476.300000 0001 0943 978XDepartment of Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Kiyofumi Yamada
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University, Graduate School of Medicine, Nagoya, Aichi, Japan.
| | - Daisuke Mori
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan. .,Department of Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan. .,Brain and Mind Research Center, Nagoya University, Nagoya, Aichi, Japan.
| | - Norio Ozaki
- grid.27476.300000 0001 0943 978XDepartment of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| |
Collapse
|
10
|
Siri SO, Rozés-Salvador V, de la Villarmois EA, Ghersi MS, Quassollo G, Pérez MF, Conde C. Decrease of Rab11 prevents the correct dendritic arborization, synaptic plasticity and spatial memory formation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118735. [PMID: 32389643 DOI: 10.1016/j.bbamcr.2020.118735] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 04/27/2020] [Accepted: 05/02/2020] [Indexed: 12/30/2022]
Abstract
Emerging evidence shows that Rab11 recycling endosomes (REs Rab11) are essential for several neuronal processes, including the proper functioning of growth cones, synapse architecture regulation and neuronal migration. However, several aspects of REs Rab11 remain unclear, such as its sub-cellular distribution across neuronal development, contribution to dendritic tree organization and its consequences in memory formation. In this work we show a spatio-temporal correlation between the endogenous localization of REs Rab11 and developmental stage of neurons. Furthermore, Rab11-suppressed neurons showed an increase on dendritic branching (without altering total dendritic length) and misdistribution of dendritic proteins in cultured neurons. In addition, suppression of Rab11 in adult rat brains in vivo (by expressing shRab11 through lentiviral infection), showed a decrease on both the sensitivity to induce long-term potentiation and hippocampal-dependent memory acquisition. Taken together, our results suggest that REs Rab11 expression is required for a proper dendritic architecture and branching, controlling key aspects of synaptic plasticity and spatial memory formation.
Collapse
Affiliation(s)
- Sebastian O Siri
- Universidad Nacional de Córdoba (UNC), Av. Haya de la Torre s/n Ciudad Universitaria, 5000 Córdoba, Argentina; Instituto de Investigación Médica Mercedes y Martıín Ferreyra (INIMEC-CONICET-UNC), Av. Friuli 2434, 5016 Córdoba, Argentina
| | - Victoria Rozés-Salvador
- Universidad Nacional de Córdoba (UNC), Av. Haya de la Torre s/n Ciudad Universitaria, 5000 Córdoba, Argentina; Instituto A.P. de Ciencias Básicas y Aplicadas, Universidad Nacional de Villa María (UNVM), Arturo Jauretche 1555, Ciudad Universitaria, Villa María, Argentina
| | - Emilce Artur de la Villarmois
- IFEC, CONICET, Departamento de Farmacologia, Facultad de Ciencias Químicas, UNC, Haya de la Torre Y Medina Allende, 5000 Córdoba, Argentina
| | - Marisa S Ghersi
- IFEC, CONICET, Departamento de Farmacologia, Facultad de Ciencias Químicas, UNC, Haya de la Torre Y Medina Allende, 5000 Córdoba, Argentina
| | - Gonzalo Quassollo
- Universidad Nacional de Córdoba (UNC), Av. Haya de la Torre s/n Ciudad Universitaria, 5000 Córdoba, Argentina; Instituto de Investigación Médica Mercedes y Martıín Ferreyra (INIMEC-CONICET-UNC), Av. Friuli 2434, 5016 Córdoba, Argentina
| | - Mariela F Pérez
- IFEC, CONICET, Departamento de Farmacologia, Facultad de Ciencias Químicas, UNC, Haya de la Torre Y Medina Allende, 5000 Córdoba, Argentina
| | - Cecilia Conde
- Universidad Nacional de Córdoba (UNC), Av. Haya de la Torre s/n Ciudad Universitaria, 5000 Córdoba, Argentina; Instituto de Investigación Médica Mercedes y Martıín Ferreyra (INIMEC-CONICET-UNC), Av. Friuli 2434, 5016 Córdoba, Argentina.
| |
Collapse
|
11
|
Dupraz S, Hilton BJ, Husch A, Santos TE, Coles CH, Stern S, Brakebusch C, Bradke F. RhoA Controls Axon Extension Independent of Specification in the Developing Brain. Curr Biol 2019; 29:3874-3886.e9. [PMID: 31679934 DOI: 10.1016/j.cub.2019.09.040] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 08/22/2019] [Accepted: 09/16/2019] [Indexed: 12/22/2022]
Abstract
The specification of an axon and its subsequent outgrowth are key steps during neuronal polarization, a prerequisite to wire the brain. The Rho-guanosine triphosphatase (GTPase) RhoA is believed to be a central player in these processes. However, its physiological role has remained undefined. Here, genetic loss- and gain-of-function experiments combined with time-lapse microscopy, cell culture, and in vivo analysis show that RhoA is not involved in axon specification but confines the initiation of neuronal polarization and axon outgrowth during development. Biochemical analysis and super-resolution microscopy together with molecular and pharmacological manipulations reveal that RhoA restrains axon growth by activating myosin-II-mediated actin arc formation in the growth cone to prevent microtubules from protruding toward the leading edge. Through this mechanism, RhoA regulates the duration of axon growth and pause phases, thus controlling the tightly timed extension of developing axons. Thereby, this work unravels physiologically relevant players coordinating actin-microtubule interactions during axon growth.
Collapse
Affiliation(s)
- Sebastian Dupraz
- Axonal Growth and Regeneration Group, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Building 99, 53127 Bonn, Germany
| | - Brett J Hilton
- Axonal Growth and Regeneration Group, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Building 99, 53127 Bonn, Germany
| | - Andreas Husch
- Axonal Growth and Regeneration Group, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Building 99, 53127 Bonn, Germany
| | - Telma E Santos
- Axonal Growth and Regeneration Group, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Building 99, 53127 Bonn, Germany
| | - Charlotte H Coles
- Axonal Growth and Regeneration Group, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Building 99, 53127 Bonn, Germany
| | - Sina Stern
- Axonal Growth and Regeneration Group, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Building 99, 53127 Bonn, Germany
| | - Cord Brakebusch
- Biotech Research & Innovation Centre, University of Copenhagen, Ole Maaløes Vej 5, 2200 Copenhagen, Denmark
| | - Frank Bradke
- Axonal Growth and Regeneration Group, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Building 99, 53127 Bonn, Germany.
| |
Collapse
|
12
|
Tedeschi A, Dupraz S, Curcio M, Laskowski CJ, Schaffran B, Flynn KC, Santos TE, Stern S, Hilton BJ, Larson MJE, Gurniak CB, Witke W, Bradke F. ADF/Cofilin-Mediated Actin Turnover Promotes Axon Regeneration in the Adult CNS. Neuron 2019; 103:1073-1085.e6. [PMID: 31400829 DOI: 10.1016/j.neuron.2019.07.007] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 06/06/2019] [Accepted: 07/08/2019] [Indexed: 11/27/2022]
Abstract
Injured axons fail to regenerate in the adult CNS, which contrasts with their vigorous growth during embryonic development. We explored the potential of re-initiating axon extension after injury by reactivating the molecular mechanisms that drive morphogenetic transformation of neurons during development. Genetic loss- and gain-of-function experiments followed by time-lapse microscopy, in vivo imaging, and whole-mount analysis show that axon regeneration is fueled by elevated actin turnover. Actin depolymerizing factor (ADF)/cofilin controls actin turnover to sustain axon regeneration after spinal cord injury through its actin-severing activity. This pinpoints ADF/cofilin as a key regulator of axon growth competence, irrespective of developmental stage. These findings reveal the central role of actin dynamics regulation in this process and elucidate a core mechanism underlying axon growth after CNS trauma. Thereby, neurons maintain the capacity to stimulate developmental programs during adult life, expanding their potential for plasticity. Thus, actin turnover is a key process for future regenerative interventions.
Collapse
Affiliation(s)
- Andrea Tedeschi
- Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Str. 27, 53127 Bonn, Germany
| | - Sebastian Dupraz
- Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Str. 27, 53127 Bonn, Germany
| | - Michele Curcio
- Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Str. 27, 53127 Bonn, Germany
| | - Claudia J Laskowski
- Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Str. 27, 53127 Bonn, Germany
| | - Barbara Schaffran
- Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Str. 27, 53127 Bonn, Germany
| | - Kevin C Flynn
- Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Str. 27, 53127 Bonn, Germany
| | - Telma E Santos
- Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Str. 27, 53127 Bonn, Germany
| | - Sina Stern
- Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Str. 27, 53127 Bonn, Germany
| | - Brett J Hilton
- Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Str. 27, 53127 Bonn, Germany
| | - Molly J E Larson
- Center for Brain and Spinal Cord Repair, Department of Neuroscience, Wexner Medical Center, The Ohio State University, 460 W. 12th Ave., Columbus, OH 43210, USA
| | - Christine B Gurniak
- Institute of Genetics, University of Bonn, Karlrobert-Kreiten-Str. 13, 53115 Bonn, Germany
| | - Walter Witke
- Institute of Genetics, University of Bonn, Karlrobert-Kreiten-Str. 13, 53115 Bonn, Germany
| | - Frank Bradke
- Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Str. 27, 53127 Bonn, Germany.
| |
Collapse
|
13
|
Takano T, Funahashi Y, Kaibuchi K. Neuronal Polarity: Positive and Negative Feedback Signals. Front Cell Dev Biol 2019; 7:69. [PMID: 31069225 PMCID: PMC6491837 DOI: 10.3389/fcell.2019.00069] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 04/09/2019] [Indexed: 12/21/2022] Open
Abstract
Establishment and maintenance of neuronal polarity are critical for neuronal development and function. One of the fundamental questions in neurodevelopment is how neurons generate only one axon and several dendrites from multiple minor neurites. Over the past few decades, molecular and cell biological approaches have unveiled a large number of signaling networks regulating neuronal polarity in cultured hippocampal neurons and the developing cortex. Emerging evidence reveals that positive and negative feedback signals play a crucial role in axon and dendrite specification. Positive feedback signals are continuously activated in one of minor neurites and result in axon specification and elongation, whereas negative feedback signals are propagated from a nascent axon terminal to all minor neurites and inhibit the formation of multiple axon, thereby leading to dendrite specification, and maintaining neuronal polarity. This current insight provides a holistic picture of the signaling mechanisms underlying neuronal polarization during neuronal development. Here, our review highlights recent advancements in this fascinating field, with a focus on the positive, and negative feedback signals as key regulatory mechanisms underlying neuronal polarization.
Collapse
Affiliation(s)
- Tetsuya Takano
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Department of Cell Biology, Duke University Medical School, Durham, NC, United States
| | - Yasuhiro Funahashi
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kozo Kaibuchi
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
14
|
Bisbal M, Sanchez M. Neurotoxicity of the pesticide rotenone on neuronal polarization: a mechanistic approach. Neural Regen Res 2019; 14:762-766. [PMID: 30688258 PMCID: PMC6375050 DOI: 10.4103/1673-5374.249847] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Neurons are the most extensive and polarized cells that display a unique single long axon and multiple dendrites, which are compartments exhibiting structural and functional differences. Polarity occurs early in neuronal development and it is maintained by complex subcellular mechanisms throughout cell life. A well-defined and controlled spatio-temporal program of cellular and molecular events strictly regulates the formation of the axon and dendrites from a non-polarized cell. This event is critical for an adequate neuronal wiring and therefore for the normal functioning of the nervous system. Neuronal polarity is very sensitive to the harmful effects of different factors present in the environment. In this regard, rotenone is a crystalline, colorless and odorless isoflavone used as insecticide, piscicide and broad spectrum pesticide commonly used earlier in agriculture. In the present review we will summarize the toxicity mechanism caused by this pesticide in different neuronal cell types, focusing on a particular biological mechanism whereby rotenone could impair neuronal polarization in cultured hippocampal neurons. Recent advances suggest that the inhibition of axonogenesis produced by rotenone could be related with its effect on microtubule dynamics, the actin cytoskeleton and their regulatory pathways, particularly affecting the small RhoGTPase RhoA. Unveiling the mechanism by which rotenone produces neurotoxicity will be instrumental to understand the cellular mechanisms involved in neurodegenerative diseases influenced by this environmental pollutant, which may lead to research focused on the design of new therapeutic strategies.
Collapse
Affiliation(s)
- Mariano Bisbal
- Laboratory of Neurobiology, Instituto Mercedes y Martín Ferreyra (INIMEC-CONICET); Universidad Nacional de Córdoba; Instituto Universitario Ciencias Biomédicas Córdoba, Córdoba, Argentina
| | - Mónica Sanchez
- Laboratory of Neurobiology, Instituto Mercedes y Martín Ferreyra (INIMEC-CONICET); Universidad Nacional de Córdoba; Instituto Universitario Ciencias Biomédicas Córdoba, Córdoba, Argentina
| |
Collapse
|
15
|
Bisbal M, Remedi M, Quassollo G, Cáceres A, Sanchez M. Rotenone inhibits axonogenesis via an Lfc/RhoA/
ROCK
pathway in cultured hippocampal neurons. J Neurochem 2018; 146:570-584. [DOI: 10.1111/jnc.14547] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 05/24/2018] [Accepted: 06/25/2018] [Indexed: 12/24/2022]
Affiliation(s)
- Mariano Bisbal
- Laboratory of Neurobiology INIMEC‐CONICET Córdoba Argentina
- Universidad Nacional de Córdoba Córdoba Argentina
- Instituto Universitario Ciencias Biomédicas Córdoba Córdoba Argentina
| | - Mónica Remedi
- Laboratory of Neurobiology INIMEC‐CONICET Córdoba Argentina
- Universidad Nacional de Córdoba Córdoba Argentina
- Instituto Universitario Ciencias Biomédicas Córdoba Córdoba Argentina
| | - Gonzalo Quassollo
- Laboratory of Neurobiology INIMEC‐CONICET Córdoba Argentina
- Universidad Nacional de Córdoba Córdoba Argentina
- Instituto Universitario Ciencias Biomédicas Córdoba Córdoba Argentina
| | - Alfredo Cáceres
- Laboratory of Neurobiology INIMEC‐CONICET Córdoba Argentina
- Universidad Nacional de Córdoba Córdoba Argentina
- Instituto Universitario Ciencias Biomédicas Córdoba Córdoba Argentina
| | - Mónica Sanchez
- Laboratory of Neurobiology INIMEC‐CONICET Córdoba Argentina
- Universidad Nacional de Córdoba Córdoba Argentina
- Instituto Universitario Ciencias Biomédicas Córdoba Córdoba Argentina
| |
Collapse
|
16
|
Rho GTPases in Intellectual Disability: From Genetics to Therapeutic Opportunities. Int J Mol Sci 2018; 19:ijms19061821. [PMID: 29925821 PMCID: PMC6032284 DOI: 10.3390/ijms19061821] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 06/14/2018] [Accepted: 06/16/2018] [Indexed: 12/22/2022] Open
Abstract
Rho-class small GTPases are implicated in basic cellular processes at nearly all brain developmental steps, from neurogenesis and migration to axon guidance and synaptic plasticity. GTPases are key signal transducing enzymes that link extracellular cues to the neuronal responses required for the construction of neuronal networks, as well as for synaptic function and plasticity. Rho GTPases are highly regulated by a complex set of activating (GEFs) and inactivating (GAPs) partners, via protein:protein interactions (PPI). Misregulated RhoA, Rac1/Rac3 and cdc42 activity has been linked with intellectual disability (ID) and other neurodevelopmental conditions that comprise ID. All genetic evidences indicate that in these disorders the RhoA pathway is hyperactive while the Rac1 and cdc42 pathways are consistently hypoactive. Adopting cultured neurons for in vitro testing and specific animal models of ID for in vivo examination, the endophenotypes associated with these conditions are emerging and include altered neuronal networking, unbalanced excitation/inhibition and altered synaptic activity and plasticity. As we approach a clearer definition of these phenotype(s) and the role of hyper- and hypo-active GTPases in the construction of neuronal networks, there is an increasing possibility that selective inhibitors and activators might be designed via PPI, or identified by screening, that counteract the misregulation of small GTPases and result in alleviation of the cognitive condition. Here we review all knowledge in support of this possibility.
Collapse
|
17
|
Ismail OZ, Sriranganathan S, Zhang X, Bonventre JV, Zervos AS, Gunaratnam L. Tctex-1, a novel interaction partner of Kidney Injury Molecule-1, is required for efferocytosis. J Cell Physiol 2018; 233:6877-6895. [PMID: 29693725 DOI: 10.1002/jcp.26578] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 03/01/2018] [Indexed: 02/04/2023]
Abstract
Kidney injury molecule-1 (KIM-1) is a phosphatidylserine receptor that is specifically upregulated on proximal tubular epithelial cells (PTECs) during acute kidney injury and mitigates tissue damage by mediating efferocytosis (the phagocytic clearance of apoptotic cells). The signaling molecules that regulate efferocytosis in TECs are not well understood. Using a yeast two-hybrid screen, we identified the dynein light chain protein, Tctex-1, as a novel KIM-1-interacting protein. Immunoprecipitation and confocal imaging studies suggested that Tctex-1 associates with KIM-1 in cells at baseline, but, dissociates from KIM-1 within 90 min of initiation of efferocytosis. Interfering with actin or microtubule polymerization interestingly prevented the dissociation of KIM-1 from Tctex-1. Moreover, the subcellular localization of Tctex-1 changed from being microtubule-associated to mainly cytosolic upon expression of KIM-1. Short hairpin RNA-mediated silencing of endogenous Tctex-1 in cells significantly inhibited efferocytosis to levels comparable to that of knock down of KIM-1 in the same cells. Importantly, Tctex-1 was not involved in the delivery of KIM-1 to the cell-surface. On the other hand, KIM-1 expression significantly inhibited the phosphorylation of Tctex-1 at threonine 94 (T94), a post-translational modification which is known to disrupt the binding of Tctex-1 to dynein on microtubules. In keeping with this, we found that KIM-1 bound less efficiently to the phosphomimic (T94E) mutant of Tctex-1 compared to wild type Tctex-1. Surprisingly, expression of Tctex-1 T94E did not influence KIM-1-mediated efferocytosis. Our studies uncover a previously unknown role for Tctex-1 in KIM-1-dependent efferocytosis in epithelial cells.
Collapse
Affiliation(s)
- Ola Z Ismail
- Matthew Mailing Center for Translational Transplant Studies, London Health Sciences Center, Lawson Health Research Institute, London, Ontario, Canada
| | - Saranga Sriranganathan
- Matthew Mailing Center for Translational Transplant Studies, London Health Sciences Center, Lawson Health Research Institute, London, Ontario, Canada
| | - Xizhong Zhang
- Matthew Mailing Center for Translational Transplant Studies, London Health Sciences Center, Lawson Health Research Institute, London, Ontario, Canada
| | - Joseph V Bonventre
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Antonis S Zervos
- Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, Orlando, Florida
| | - Lakshman Gunaratnam
- Matthew Mailing Center for Translational Transplant Studies, London Health Sciences Center, Lawson Health Research Institute, London, Ontario, Canada.,Division of Nephrology, Department of Medicine, Schulich School of Medicine and Dentistry, London, Western University, Ontario, Canada
| |
Collapse
|
18
|
Farrell KB, McDonald S, Lamb AK, Worcester C, Peersen OB, Di Pietro SM. Novel function of a dynein light chain in actin assembly during clathrin-mediated endocytosis. J Cell Biol 2017; 216:2565-2580. [PMID: 28706108 PMCID: PMC5551697 DOI: 10.1083/jcb.201604123] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 01/04/2017] [Accepted: 05/12/2017] [Indexed: 11/22/2022] Open
Abstract
Actin-capping protein is a key component of the actin cytoskeleton at sites of clathrin-mediated endocytosis. Farrell et al. show that a newly discovered component of the endocytic machinery belongs to the dynein light chain family and regulates the recruitment of actin-capping protein in a dynein motor–independent manner. Clathrin- and actin-mediated endocytosis is essential in eukaryotic cells. In this study, we demonstrate that Tda2 is a novel protein of the endocytic machinery necessary for normal internalization of native cargo in yeast. Tda2 has not been classified in any protein family. Unexpectedly, solving the crystal structure of Tda2 revealed it belongs to the dynein light chain family. However, Tda2 works independently of the dynein motor complex and microtubules. Tda2 forms a tight complex with the polyproline motif–rich protein Aim21, which interacts physically with the SH3 domain of the Arp2/3 complex regulator Bbc1. The Tda2–Aim21 complex localizes to endocytic sites in a Bbc1- and filamentous actin–dependent manner. Importantly, the Tda2–Aim21 complex interacts directly with and facilitates the recruitment of actin-capping protein, revealing barbed-end filament capping at endocytic sites to be a regulated event. Thus, we have uncovered a new layer of regulation of the actin cytoskeleton by a member of a conserved protein family that has not been previously associated with a function in endocytosis.
Collapse
Affiliation(s)
- Kristen B Farrell
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO
| | - Seth McDonald
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO
| | - Andrew K Lamb
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO
| | - Colette Worcester
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO
| | - Olve B Peersen
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO
| | - Santiago M Di Pietro
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO
| |
Collapse
|
19
|
Discovery of long-range inhibitory signaling to ensure single axon formation. Nat Commun 2017; 8:33. [PMID: 28652571 PMCID: PMC5484694 DOI: 10.1038/s41467-017-00044-2] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 04/27/2017] [Indexed: 01/12/2023] Open
Abstract
A long-standing question in neurodevelopment is how neurons develop a single axon and multiple dendrites from common immature neurites. Long-range inhibitory signaling from the growing axon is hypothesized to prevent outgrowth of other immature neurites and to differentiate them into dendrites, but the existence and nature of this inhibitory signaling remains unknown. Here, we demonstrate that axonal growth triggered by neurotrophin-3 remotely inhibits neurite outgrowth through long-range Ca2+ waves, which are delivered from the growing axon to the cell body. These Ca2+ waves increase RhoA activity in the cell body through calcium/calmodulin-dependent protein kinase I. Optogenetic control of Rho-kinase combined with computational modeling reveals that active Rho-kinase diffuses to growing other immature neurites and inhibits their outgrowth. Mechanistically, calmodulin-dependent protein kinase I phosphorylates a RhoA-specific GEF, GEF-H1, whose phosphorylation enhances its GEF activity. Thus, our results reveal that long-range inhibitory signaling mediated by Ca2+ wave is responsible for neuronal polarization. Emerging evidence suggests that gut microbiota influences immune function in the brain and may play a role in neurological diseases. Here, the authors offer in vivo evidence from a Drosophila model that supports a role for gut microbiota in modulating the progression of Alzheimer’s disease.
Collapse
|
20
|
Rozés Salvador V, Heredia F, Berardo A, Palandri A, Wojnacki J, Vivinetto AL, Sheikh KA, Caceres A, Lopez PHH. Anti-glycan antibodies halt axon regeneration in a model of Guillain Barrè Syndrome axonal neuropathy by inducing microtubule disorganization via RhoA-ROCK-dependent inactivation of CRMP-2. Exp Neurol 2016; 278:42-53. [PMID: 26804001 DOI: 10.1016/j.expneurol.2016.01.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 01/19/2016] [Indexed: 10/22/2022]
Abstract
Several reports have linked the presence of high titers of anti-Gg Abs with delayed recovery/poor prognosis in GBS. In most cases, failure to recover is associated with halted/deficient axon regeneration. Previous work identified that monoclonal and patient-derived anti-Gg Abs can act as inhibitory factors in an animal model of axon regeneration. Further studies using primary dorsal root ganglion neuron (DRGn) cultures demonstrated that anti-Gg Abs can inhibit neurite outgrowth by targeting gangliosides via activation of the small GTPase RhoA and its associated kinase (ROCK), a signaling pathway common to other established inhibitors of axon regeneration. We aimed to study the molecular basis of the inhibitory effect of anti-Gg abs on neurite outgrowth by dissecting the molecular dynamics of growth cones (GC) cytoskeleton in relation to the spatial-temporal analysis of RhoA activity. We now report that axon growth inhibition in DRGn induced by a well characterized mAb targeting gangliosides GD1a/GT1b involves: i) an early RhoA/ROCK-independent collapse of lamellipodia; ii) a RhoA/ROCK-dependent shrinking of filopodia; and iii) alteration of GC microtubule organization/and presumably dynamics via RhoA/ROCK-dependent phosphorylation of CRMP-2 at threonine 555. Our results also show that mAb 1B7 inhibits peripheral axon regeneration in an animal model via phosphorylation/inactivation of CRMP-2 at threonine 555. Overall, our data may help to explain the molecular mechanisms underlying impaired nerve repair in GBS. Future work should define RhoA-independent pathway/s and effectors regulating actin cytoskeleton, thus providing an opportunity for the design of a successful therapy to guarantee an efficient target reinnervation.
Collapse
Affiliation(s)
- Victoria Rozés Salvador
- Laboratory of Neurobiology, Instituto de Investigación Médica Mercedes y Martin Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Argentina
| | - Florencia Heredia
- Laboratory of Neurobiology, Instituto de Investigación Médica Mercedes y Martin Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Argentina
| | - Andrés Berardo
- Laboratory of Neurobiology, Instituto de Investigación Médica Mercedes y Martin Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Argentina
| | - Anabela Palandri
- Laboratory of Neurobiology, Instituto de Investigación Médica Mercedes y Martin Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Argentina
| | - Jose Wojnacki
- Laboratory of Neurobiology, Instituto de Investigación Médica Mercedes y Martin Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Argentina
| | - Ana L Vivinetto
- Laboratory of Neurobiology, Instituto de Investigación Médica Mercedes y Martin Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Argentina
| | - Kazim A Sheikh
- Department of Neurology, University of Texas Medical School at Houston, Houston, USA
| | - Alfredo Caceres
- Laboratory of Neurobiology, Instituto de Investigación Médica Mercedes y Martin Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Argentina
| | - Pablo H H Lopez
- Laboratory of Neurobiology, Instituto de Investigación Médica Mercedes y Martin Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Argentina; Facultad de Psicología, Universidad Nacional de Córdoba, Argentina.
| |
Collapse
|
21
|
Menon S, Gupton SL. Building Blocks of Functioning Brain: Cytoskeletal Dynamics in Neuronal Development. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 322:183-245. [PMID: 26940519 PMCID: PMC4809367 DOI: 10.1016/bs.ircmb.2015.10.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neural connectivity requires proper polarization of neurons, guidance to appropriate target locations, and establishment of synaptic connections. From when neurons are born to when they finally reach their synaptic partners, neurons undergo constant rearrangment of the cytoskeleton to achieve appropriate shape and polarity. Of particular importance to neuronal guidance to target locations is the growth cone at the tip of the axon. Growth-cone steering is also dictated by the underlying cytoskeleton. All these changes require spatiotemporal control of the cytoskeletal machinery. This review summarizes the proteins that are involved in modulating the actin and microtubule cytoskeleton during the various stages of neuronal development.
Collapse
Affiliation(s)
- Shalini Menon
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, United States of America
| | - Stephanie L Gupton
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, United States of America; Neuroscience Center and Curriculum in Neurobiology, University of North Carolina, Chapel Hill, NC, United States of America; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, United States of America.
| |
Collapse
|
22
|
Abstract
Neurons are highly polarized cells with structurally and functionally distinct processes called axons and dendrites. This polarization underlies the directional flow of information in the central nervous system, so the establishment and maintenance of neuronal polarization is crucial for correct development and function. Great progress in our understanding of how neurons establish their polarity has been made through the use of cultured hippocampal neurons, while recent technological advances have enabled in vivo analysis of axon specification and elongation. This short review and accompanying poster highlight recent advances in this fascinating field, with an emphasis on the signaling mechanisms underlying axon and dendrite specification in vitro and in vivo.
Collapse
Affiliation(s)
- Tetsuya Takano
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Chundi Xu
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Yasuhiro Funahashi
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Takashi Namba
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Kozo Kaibuchi
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| |
Collapse
|
23
|
Copf T. Importance of gene dosage in controlling dendritic arbor formation during development. Eur J Neurosci 2015; 42:2234-49. [PMID: 26108333 DOI: 10.1111/ejn.13002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Revised: 06/05/2015] [Accepted: 06/18/2015] [Indexed: 12/11/2022]
Abstract
Proper dendrite morphology is crucial for normal nervous system functioning. While a number of genes have been implicated in dendrite morphogenesis in both invertebrates and mammals, it remains unclear how developing dendrites respond to changes in gene dosage and what type of patterns their responses may follow. To understand this, I review here evidence from the recent literature, focusing on the genetic studies performed in the Drosophila larval dendritic arborization class IV neuron, an excellent cell type to understand dendrite morphogenesis. I summarize how class IV arbors change morphology in response to developmental fluctuations in the expression levels of 47 genes, studied by means of genetic manipulations such as loss-of-function and gain-of-function, and for which sufficient information is available. I find that arbors can respond to changing gene dosage in several distinct ways, each characterized by a singular dose-response curve. Interestingly, in 72% of cases arbors are sensitive, and thus adjust their morphology, in response to both decreases and increases in the expression of a given gene, indicating that dendrite morphogenesis is a process particularly sensitive to gene dosage. By summarizing the parallels between Drosophila and mammals, I show that many Drosophila dendrite morphogenesis genes have orthologs in mammals, and that some of these are associated with mammalian dendrite outgrowth and human neurodevelopmental disorders. One notable disease-related molecule is kinase Dyrk1A, thought to be a causative factor in Down syndrome. Both increases and decreases in Dyrk1A gene dosage lead to impaired dendrite morphogenesis, which may contribute to Down syndrome pathoetiology.
Collapse
Affiliation(s)
- Tijana Copf
- Institute of Molecular Biology and Biotechnology, Nikolaou Plastira 100, PO Box 1385, Heraklion, GR-70013, Crete, Greece
| |
Collapse
|
24
|
Namba T, Funahashi Y, Nakamuta S, Xu C, Takano T, Kaibuchi K. Extracellular and Intracellular Signaling for Neuronal Polarity. Physiol Rev 2015; 95:995-1024. [PMID: 26133936 DOI: 10.1152/physrev.00025.2014] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Neurons are one of the highly polarized cells in the body. One of the fundamental issues in neuroscience is how neurons establish their polarity; therefore, this issue fascinates many scientists. Cultured neurons are useful tools for analyzing the mechanisms of neuronal polarization, and indeed, most of the molecules important in their polarization were identified using culture systems. However, we now know that the process of neuronal polarization in vivo differs in some respects from that in cultured neurons. One of the major differences is their surrounding microenvironment; neurons in vivo can be influenced by extrinsic factors from the microenvironment. Therefore, a major question remains: How are neurons polarized in vivo? Here, we begin by reviewing the process of neuronal polarization in culture conditions and in vivo. We also survey the molecular mechanisms underlying neuronal polarization. Finally, we introduce the theoretical basis of neuronal polarization and the possible involvement of neuronal polarity in disease and traumatic brain injury.
Collapse
Affiliation(s)
- Takashi Namba
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yasuhiro Funahashi
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shinichi Nakamuta
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Chundi Xu
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tetsuya Takano
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kozo Kaibuchi
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
25
|
Luo T, Roman P, Liu C, Sun X, Park Y, Hu B. Upregulation of the GEF-H1 pathway after transient cerebral ischemia. Exp Neurol 2014; 263:306-13. [PMID: 25447939 DOI: 10.1016/j.expneurol.2014.10.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 10/10/2014] [Accepted: 10/22/2014] [Indexed: 01/10/2023]
Abstract
The microtubule-dependent GEF-H1 pathway controls synaptic re-networking and overall gene expression via regulating cytoskeleton dynamics. Understanding this pathway after ischemia is essential to developing new therapies for neuronal function recovery. However, how the GEF-H1 pathway is regulated following transient cerebral ischemia remains unknown. This study employed a rat model of transient forebrain ischemia to investigate alterations of the GEF-H1 pathway using Western blotting, confocal and electron microscopy, dephosphorylation analysis, and pull-down assay. The GEF-H1 activity was significantly upregulated by: (i) dephosphorylation and (ii) translocation to synaptic membrane and nuclear structures during the early phase of reperfusion. GEF-H1 protein was then downregulated in the brain regions where neurons were destined to undergo delayed neuronal death, but markedly upregulated in neurons that were resistant to the same episode of cerebral ischemia. Consistently, GTP-RhoA, a GEF-H1 substrate, was significantly upregulated after brain ischemia. Electron microscopy further showed that neuronal microtubules were persistently depolymerized in the brain region where GEF-H1 protein was downregulated after brain ischemia. The results demonstrate that the GEF-H1 activity is significantly upregulated in both vulnerable and resistant brain regions in the early phase of reperfusion. However, GEF-H1 protein is downregulated in the vulnerable neurons but upregulated in the ischemic resistant neurons during the recovery phase after ischemia. The initial upregulation of GEF-H1 activity may contribute to excitotoxicity, whereas the late upregulation of GEF-H1 protein may promote neuroplasticity after brain ischemia.
Collapse
Affiliation(s)
- Tianfei Luo
- Neurochemistry Laboratory of Brain Injury, Shock Trauma & Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD 21201, United States; Department of Neurology, The First Teaching Hospital, Jilin University, Changchun, China
| | - Philip Roman
- Neurochemistry Laboratory of Brain Injury, Shock Trauma & Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD 21201, United States
| | - Chunli Liu
- Neurochemistry Laboratory of Brain Injury, Shock Trauma & Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD 21201, United States
| | - Xin Sun
- Neurochemistry Laboratory of Brain Injury, Shock Trauma & Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD 21201, United States; Department of Neurology, The First Teaching Hospital, Jilin University, Changchun, China
| | - Yujung Park
- Neurochemistry Laboratory of Brain Injury, Shock Trauma & Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD 21201, United States
| | - Bingren Hu
- Neurochemistry Laboratory of Brain Injury, Shock Trauma & Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD 21201, United States.
| |
Collapse
|
26
|
Wojnacki J, Quassollo G, Marzolo MP, Cáceres A. Rho GTPases at the crossroad of signaling networks in mammals: impact of Rho-GTPases on microtubule organization and dynamics. Small GTPases 2014; 5:e28430. [PMID: 24691223 DOI: 10.4161/sgtp.28430] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Microtubule (MT) organization and dynamics downstream of external cues is crucial for maintaining cellular architecture and the generation of cell asymmetries. In interphase cells RhoA, Rac, and Cdc42, conspicuous members of the family of small Rho GTPases, have major roles in modulating MT stability, and hence polarized cell behaviors. However, MTs are not mere targets of Rho GTPases, but also serve as signaling platforms coupling MT dynamics to Rho GTPase activation in a variety of cellular conditions. In this article, we review some of the key studies describing the reciprocal relationship between small Rho-GTPases and MTs during migration and polarization.
Collapse
Affiliation(s)
- José Wojnacki
- Laboratory of Neurobiology; Instituto Mercedes y Martin Ferreyra (INIMEC) CONICET; Córdoba, Argentina
| | - Gonzalo Quassollo
- Laboratory of Neurobiology; Instituto Mercedes y Martin Ferreyra (INIMEC) CONICET; Córdoba, Argentina
| | - María-Paz Marzolo
- Laboratorio de Tráfico Intracelular y Señalización; Departamento de Biología Celular y Molecular; Facultad de Ciencias Biológicas; Pontificia Universidad Católica de Chile; Santiago, Chile
| | - Alfredo Cáceres
- Laboratory of Neurobiology; Instituto Mercedes y Martin Ferreyra (INIMEC) CONICET; Córdoba, Argentina; Universidad Nacional Córdoba (UNC); Córdoba, Argentina; Instituto Universitario Ciencias Biomédicas Córdoba (IUCBC); Córdoba-Argentina
| |
Collapse
|
27
|
Slater PG, Ramirez VT, Gonzalez-Billault C, Varela-Nallar L, Inestrosa NC. Frizzled-5 receptor is involved in neuronal polarity and morphogenesis of hippocampal neurons. PLoS One 2013; 8:e78892. [PMID: 24205342 PMCID: PMC3800132 DOI: 10.1371/journal.pone.0078892] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Accepted: 09/17/2013] [Indexed: 01/09/2023] Open
Abstract
The Wnt signaling pathway plays important roles during different stages of neuronal development, including neuronal polarization and dendritic and axonal outgrowth. However, little is known about the identity of the Frizzled receptors mediating these processes. In the present study, we investigated the role of Frizzled-5 (Fzd5) on neuronal development in cultured Sprague-Dawley rat hippocampal neurons. We found that Fzd5 is expressed early in cultured neurons on actin-rich structures localized at minor neurites and axonal growth cones. At 4 DIV, Fzd5 polarizes towards the axon, where its expression is detected mainly at the peripheral zone of axonal growth cones, with no obvious staining at dendrites; suggesting a role of Fzd5 in neuronal polarization. Overexpression of Fzd5 during the acquisition of neuronal polarity induces mislocalization of the receptor and a loss of polarized axonal markers. Fzd5 knock-down leads to loss of axonal proteins, suggesting an impaired neuronal polarity. In contrast, overexpression of Fzd5 in neurons that are already polarized did not alter polarity, but decreased the total length of axons and increased total dendrite length and arborization. Fzd5 activated JNK in HEK293 cells and the effects triggered by Fzd5 overexpression in neurons were partially prevented by inhibition of JNK, suggesting that a non-canonical Wnt signaling mechanism might be involved. Our results suggest that, Fzd5 has a role in the establishment of neuronal polarity, and in the morphogenesis of neuronal processes, in part through the activation of the non-canonical Wnt mechanism involving JNK.
Collapse
Affiliation(s)
- Paula G. Slater
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, P. Universidad Católica de Chile, Santiago, Chile
| | - Valerie T. Ramirez
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, P. Universidad Católica de Chile, Santiago, Chile
| | | | - Lorena Varela-Nallar
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, P. Universidad Católica de Chile, Santiago, Chile
| | - Nibaldo C. Inestrosa
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, P. Universidad Católica de Chile, Santiago, Chile
- * . E-mail:
| |
Collapse
|
28
|
Sabirzhanova I, Liu C, Zhao J, Bramlett H, Dietrich WD, Hu B. Changes in the GEF-H1 pathways after traumatic brain injury. J Neurotrauma 2013; 30:1449-56. [PMID: 23611588 DOI: 10.1089/neu.2012.2673] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Brains undergo significant remodeling after traumatic brain injury (TBI). The Rho guanine triphosphate (GTP)ase pathways control brain remodeling during development and under pathological conditions. How the Rho GTPase pathways are regulated in the brain after TBI remains largely unknown, however. This study used the rat fluid percussion injury model to investigate changes in the Rho GTPase pathways after TBI. The results showed that TBI leads to activation and translocation of RhoA and Rac1 proteins from cytosolic fraction to the membrane fraction after injury. Consistently, the Rho guanine nucleotide exchange factors GEF-H1 and Cool-2/αPix are significantly activated by dephosphorylation and accumulation in the cytosolic fractions during the post-TBI phase. Because the Rho GTPase pathways are key regulators of brain remodeling, these results depict regulatory mechanisms of the Rho GTPase pathways after TBI, and pave the way for the study of therapeutic agents targeting the Rho GTPase pathways for functional recovery after TBI.
Collapse
Affiliation(s)
- Inna Sabirzhanova
- Neurochemistry Laboratory of Brain Injury, Department of Anesthesiology, and Shock Trauma and Anesthesiology Research Center, University of Maryland School of Medicine , Baltimore, MD 21201, USA
| | | | | | | | | | | |
Collapse
|
29
|
Jausoro I, Mestres I, Quassollo G, Masseroni L, Heredia F, Caceres A. Regulation of spine density and morphology by IQGAP1 protein domains. PLoS One 2013; 8:e56574. [PMID: 23441206 PMCID: PMC3575492 DOI: 10.1371/journal.pone.0056574] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Accepted: 01/15/2013] [Indexed: 11/25/2022] Open
Abstract
IQGAP1 is a scaffolding protein that regulates spine number. We now show a differential role for IQGAP1 domains in spine morphogenesis, in which a region of the N-terminus that promotes Arp2/3-mediated actin polymerization and branching stimulates spine head formation while a region that binds to Cdc42 and Rac is required for stalk extension. Conversely, IQGAP1 rescues spine deficiency induced by expression of dominant negative Cdc42 by stimulating formation of stubby spines. Together, our observations place IQGAP1 as a crucial regulator of spine number and shape acting through the N-Wasp Arp2/3 complex, as well as upstream and downstream of Cdc42.
Collapse
Affiliation(s)
- Ignacio Jausoro
- Laboratory of Neurobiology, Instituto Mercedes y Martín Ferreyra, INIMEC-CONICET, Córdoba, Argentina
- Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Ivan Mestres
- Laboratory of Neurobiology, Instituto Mercedes y Martín Ferreyra, INIMEC-CONICET, Córdoba, Argentina
- Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Gonzalo Quassollo
- Laboratory of Neurobiology, Instituto Mercedes y Martín Ferreyra, INIMEC-CONICET, Córdoba, Argentina
- Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Lujan Masseroni
- Laboratory of Neurobiology, Instituto Mercedes y Martín Ferreyra, INIMEC-CONICET, Córdoba, Argentina
- Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Florencia Heredia
- Laboratory of Neurobiology, Instituto Mercedes y Martín Ferreyra, INIMEC-CONICET, Córdoba, Argentina
- Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Alfredo Caceres
- Laboratory of Neurobiology, Instituto Mercedes y Martín Ferreyra, INIMEC-CONICET, Córdoba, Argentina
- Universidad Nacional de Córdoba, Córdoba, Argentina
- * E-mail:
| |
Collapse
|
30
|
Neuronal polarity: demarcation, growth and commitment. Curr Opin Cell Biol 2012; 24:547-53. [PMID: 22726583 DOI: 10.1016/j.ceb.2012.05.011] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Accepted: 05/23/2012] [Indexed: 02/03/2023]
Abstract
In a biological sense, polarity refers to the extremity of the main axis of an organelle, cell, or organism. In neurons, morphological polarity begins with the appearance of the first neurite from the cell body. In multipolar neurons, a second phase of polarization occurs when a single neurite initiates a phase of rapid growth to become the neuron's axon, while the others later differentiate as dendrites. Finally, during a third phase, axons and dendrites develop an elaborate architecture, acquiring special morphological and molecular features that commit them to their final identities. Mechanistically, each phase must be preceded by spatial restriction of growth activity. We will review recent work on the mechanisms underlying the polarized growth of neurons.
Collapse
|
31
|
Gonzalez-Billault C, Muñoz-Llancao P, Henriquez DR, Wojnacki J, Conde C, Caceres A. The role of small GTPases in neuronal morphogenesis and polarity. Cytoskeleton (Hoboken) 2012; 69:464-85. [PMID: 22605667 DOI: 10.1002/cm.21034] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2011] [Revised: 04/12/2012] [Accepted: 04/16/2012] [Indexed: 12/21/2022]
Abstract
The highly dynamic remodeling and cross talk of the microtubule and actin cytoskeleton support neuronal morphogenesis. Small RhoGTPases family members have emerged as crucial regulators of cytoskeletal dynamics. In this review we will comprehensively analyze findings that support the participation of RhoA, Rac, Cdc42, and TC10 in different neuronal morphogenetic events ranging from migration to synaptic plasticity. We will specifically address the contribution of these GTPases to support neuronal polarity and axonal elongation.
Collapse
Affiliation(s)
- Christian Gonzalez-Billault
- Faculty of Sciences, Laboratory of Cell and Neuronal Dynamics, Department of Biology and Institute for Cell Dynamics and Biotechnology, Universidad de Chile, Santiago, Chile.
| | | | | | | | | | | |
Collapse
|
32
|
Meiri D, Marshall CB, Greeve MA, Kim B, Balan M, Suarez F, Bakal C, Wu C, Larose J, Fine N, Ikura M, Rottapel R. Mechanistic insight into the microtubule and actin cytoskeleton coupling through dynein-dependent RhoGEF inhibition. Mol Cell 2012; 45:642-55. [PMID: 22405273 DOI: 10.1016/j.molcel.2012.01.027] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2011] [Revised: 09/01/2011] [Accepted: 01/20/2012] [Indexed: 11/16/2022]
Abstract
Actin-based stress fiber formation is coupled to microtubule depolymerization through the local activation of RhoA. While the RhoGEF Lfc has been implicated in this cytoskeleton coupling process, it has remained elusive how Lfc is recruited to microtubules and how microtubule recruitment moderates Lfc activity. Here, we demonstrate that the dynein light chain protein Tctex-1 is required for localization of Lfc to microtubules. Lfc residues 139-161 interact with Tctex-1 at a site distinct from the cleft that binds dynein intermediate chain. An NMR-based GEF assay revealed that interaction with Tctex-1 represses Lfc nucleotide exchange activity in an indirect manner that requires both polymerized microtubules and phosphorylation of S885 by PKA. We show that inhibition of Lfc by Tctex-1 is dynein dependent. These studies demonstrate a pivotal role of Tctex-1 as a negative regulator of actin filament organization through its control of Lfc in the crosstalk between microtubule and actin cytoskeletons.
Collapse
Affiliation(s)
- David Meiri
- Ontario Cancer Institute and the Campbell Family Cancer Research Institute, 101 College Street, Room 8-703 Toronto Medical Discovery Tower, University of Toronto, Toronto, ON M5G 1L7, Canada
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Duguay D, Bélanger-Nelson E, Mongrain V, Beben A, Khatchadourian A, Cermakian N. Dynein light chain Tctex-type 1 modulates orexin signaling through its interaction with orexin 1 receptor. PLoS One 2011; 6:e26430. [PMID: 22028875 PMCID: PMC3197643 DOI: 10.1371/journal.pone.0026430] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Accepted: 09/27/2011] [Indexed: 11/19/2022] Open
Abstract
Orexins (OX-A, OX-B) are neuropeptides involved in the regulation of the sleep-wake cycle, feeding and reward, via activation of orexin receptors 1 and 2 (OX1R, OX2R). The loss of orexin peptides or functional OX2R has been shown to cause the sleep disorder, narcolepsy. Since the regulation of orexin receptors remains largely undefined, we searched for novel protein partners of the intracellular tail of orexin receptors. Using a yeast two-hybrid screening strategy in combination with co-immunoprecipitation experiments, we found interactions between OX1R and the dynein light chains Tctex-type 1 and 3 (Dynlt1, Dynlt3). These interactions were mapped to the C-terminal region of the dynein light chains and to specific residues within the last 10 amino acids of OX1R. Hence, we hypothesized that dynein light chains could regulate orexin signaling. In HEK293 cells expressing OX1R, stimulation with OX-A produced a less sustained extracellular signal-regulated kinases 1/2 (ERK1/2) activation when Dynlt1 was co-expressed, while it was prolonged under reduced Dynlt1 expression. The amount of OX1R located at the plasma membrane as well as the kinetics and extent of OX-A-induced internalization of OX1R (disappearance from membrane) were not altered by Dynlt1. However, Dynlt1 reduced the localization of OX1R in early endosomes following initial internalization. Taken together, these data suggest that Dynlt1 modulates orexin signaling by regulating OX1R, namely its intracellular localization following ligand-induced internalization.
Collapse
Affiliation(s)
- David Duguay
- Laboratory of Molecular Chronobiology, Douglas Mental Health University Institute, Montréal, Quebec, Canada
- Department of Psychiatry, McGill University, Montréal, Quebec, Canada
| | - Erika Bélanger-Nelson
- Laboratory of Molecular Chronobiology, Douglas Mental Health University Institute, Montréal, Quebec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, Quebec, Canada
| | - Valérie Mongrain
- Laboratory of Molecular Chronobiology, Douglas Mental Health University Institute, Montréal, Quebec, Canada
- Department of Psychiatry, McGill University, Montréal, Quebec, Canada
| | - Anna Beben
- Laboratory of Molecular Chronobiology, Douglas Mental Health University Institute, Montréal, Quebec, Canada
| | - Armen Khatchadourian
- Laboratory of Molecular Chronobiology, Douglas Mental Health University Institute, Montréal, Quebec, Canada
| | - Nicolas Cermakian
- Laboratory of Molecular Chronobiology, Douglas Mental Health University Institute, Montréal, Quebec, Canada
- Department of Psychiatry, McGill University, Montréal, Quebec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, Quebec, Canada
| |
Collapse
|