1
|
Li X, Bao L, Wang X, Wu H, Chen T, Xie R, Sun H, Zhang D, Wang L, Chen L. TACC3 facilitates chondrocyte differentiation by attenuating abnormally activated FGFR3 signaling in achondroplasia - An in vitro study. Tissue Cell 2025; 96:102940. [PMID: 40373614 DOI: 10.1016/j.tice.2025.102940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 04/17/2025] [Accepted: 04/24/2025] [Indexed: 05/17/2025]
Abstract
BACKGROUND Achondroplasia is a common form of dwarfism. It is caused by mutations in the fibroblast growth factor receptor 3 (FGFR3), which inhibits chondrocyte proliferation and differentiation. AIM In this study, we intended to investigate the underlying mechanism of FGFR3 mutation-induced chondrocyte differentiation defection. METHOD Insulin-transferrin-selenium (ITS-G) stimulated ATDC5 cells was used as an in vitro model. Alcian Blue staining was performed to detect ATDC5 cell differentiation. RESULTS TACC3 expression was increased during ATDC5 cell differentiation. ITS-G induced ATDC5 cell differentiation was inhibited by the FGFR3 mutation, as evidenced by the decreased expression of ACAN and COL2A1. The downregulation of TACC3 expression induced by ITS-G stimulation was upregulated by FGFR3 overactivation. The TACC3 inhibitor (KHS101) promoted differentiation in FGFR3 mutant ATDC5 cells. The p38 signaling pathway has been implicated in FGFR3 mutation-induced chondrocyte differentiation defects. KHS101 promoted the expression of p38. KHS101-induced increase in ATDC5 cell differentiation was inhibited by the administration of a p38 inhibitor. These results suggest that TACC3 might play a role through the p38 signaling pathway in chondrocyte differentiation defects caused by FGFR3 mutations. CONCLUSION TACC3 might represent a novel target for achondroplasia.
Collapse
Affiliation(s)
- Xiang Li
- Department of Endocrinology, Genetics and Metabolism, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Long Bao
- Department of Neurosurgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Xiaoyan Wang
- Department of Endocrinology, Genetics and Metabolism, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Haiying Wu
- Department of Endocrinology, Genetics and Metabolism, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Ting Chen
- Department of Endocrinology, Genetics and Metabolism, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Rongrong Xie
- Department of Endocrinology, Genetics and Metabolism, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Hui Sun
- Department of Endocrinology, Genetics and Metabolism, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Dandan Zhang
- Department of Endocrinology, Genetics and Metabolism, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Lili Wang
- Department of Endocrinology, Genetics and Metabolism, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Linqi Chen
- Department of Endocrinology, Genetics and Metabolism, Children's Hospital of Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
2
|
Saade M, Martí E. Early spinal cord development: from neural tube formation to neurogenesis. Nat Rev Neurosci 2025; 26:195-213. [PMID: 39915695 DOI: 10.1038/s41583-025-00906-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2025] [Indexed: 03/26/2025]
Abstract
As one of the simplest and most evolutionarily conserved parts of the vertebrate nervous system, the spinal cord serves as a key model for understanding the principles of nervous system construction. During embryonic development, the spinal cord originates from a population of bipotent stem cells termed neuromesodermal progenitors, which are organized within a transient embryonic structure known as the neural tube. Neural tube morphogenesis differs along its anterior-to-posterior axis: most of the neural tube (including the regions that will develop into the brain and the anterior spinal cord) forms via the bending and dorsal fusion of the neural groove, but the establishment of the posterior region of the neural tube involves de novo formation of a lumen within a solid medullary cord. The early spinal cord primordium consists of highly polarized neural progenitor cells organized into a pseudostratified epithelium. Tight regulation of the cell division modes of these progenitors drives the embryonic growth of the neural tube and initiates primary neurogenesis. A rich history of observational and functional studies across various vertebrate models has advanced our understanding of the cellular events underlying spinal cord development, and these foundational studies are beginning to inform our knowledge of human spinal cord development.
Collapse
Affiliation(s)
- Murielle Saade
- Department of Cells and Tissues, Instituto de Biología Molecular de Barcelona CSIC, Barcelona, Spain.
| | - Elisa Martí
- Department of Cells and Tissues, Instituto de Biología Molecular de Barcelona CSIC, Barcelona, Spain.
| |
Collapse
|
3
|
Lin W, Liu Y, Liang CS, Yeh PK, Tsai CK, Hung KS, An YC, Yang FC. Syncope in Migraine: A Genome-Wide Association Study Revealing Distinct Genetic Susceptibility Variants Across Subtypes. J Clin Neurol 2024; 20:599-609. [PMID: 39505312 PMCID: PMC11543389 DOI: 10.3988/jcn.2024.0156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 07/13/2024] [Accepted: 09/04/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND AND PURPOSE Syncope is characterized by the temporary loss of consciousness and is commonly associated with migraine. However, the genetic factors that contribute to this association are not well understood. This study investigated the specific genetic loci that make patients with migraine more susceptible to syncope as well as the genetic factors contributing to syncope and migraine comorbidity in a Han Chinese population in Taiwan. METHODS A genome-wide association study was applied to 1,724 patients with migraine who visited a tertiary hospital in Taiwan. The patients were genotyped using the Affymetrix Axiom Genome-Wide TWB 2.0 array and categorized into the following subgroups based on migraine type: episodic migraine, chronic migraine, migraine with aura, and migraine without aura. Multivariate regression analyses were used to assess the relationships between specific single-nucleotide polymorphisms (SNPs) and the clinical characteristics in patients with syncope and migraine comorbidity. RESULTS In patients with migraine, SNPs were observed to be associated with syncope. In particular, the rs797384 SNP located in the intron region of LOC102724945 was associated with syncope in all patients with migraine. Additionally, four SNPs associated with syncope susceptibility were detected in the nonmigraine control group, and these SNPs differed from those in the migraine group, suggesting distinct underlying mechanisms. Furthermore, the rs797384 variant in the intron region of LOC102724945 was associated with the score on the Beck Depression Inventory. CONCLUSIONS The novel genetic loci identified in this study will improve our understanding of the genetic basis of syncope and migraine comorbidity.
Collapse
Affiliation(s)
- Wei Lin
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yi Liu
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chih-Sung Liang
- Department of Psychiatry, Beitou Branch, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Po-Kuan Yeh
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- Department of Psychiatry, Beitou Branch, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chia-Kuang Tsai
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Kuo-Sheng Hung
- Center for Precision Medicine and Genomics, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yu-Chin An
- Department of Emergency, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Fu-Chi Yang
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- Center for Precision Medicine and Genomics, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.
| |
Collapse
|
4
|
Deng X, Qiu Z, Chen X, Liu J, Wang X, Li J, Zhang J, Cui X, Fu Y, Jiang M. Exploring the potential mechanism of ginsenoside Rg1 to regulate ferroptosis in Alzheimer's disease based on network pharmacology. Eur J Pharmacol 2024; 979:176859. [PMID: 39067563 DOI: 10.1016/j.ejphar.2024.176859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 07/24/2024] [Accepted: 07/24/2024] [Indexed: 07/30/2024]
Abstract
OBJECTIVES To explore the pathogenesis of Alzheimer's disease (AD), the potential targets and signaling pathways of ginsenoside Rg1 against AD were investigated by network pharmacology. METHODS Ginsenoside Rg1 targets were identified through PubChem, PharmMapper, and Uniprot databases, while the GeneCards database was used to examine the respective targets of amyloid precursor protein (APP) and AD. Then, the common targets between ginsenoside Rg1 and APP were explored by the Venny tool, the interaction network diagram between the active components and the targets was built via Cytoscape software, as well as GO enrichment and KEGG pathway annotation analysis were performed. Furthermore, genes associated with ferroptosis were found by the GeneCards and FerrDb databases. Besides, the connection among ginsenoside Rg1, APP, ferroptosis, and AD was predicted and analyzed. Finally, the effects of ginsenosides Rg1 and liproxstain-1 on the proliferation and differentiation of APP/PS1 mice were evaluated by immunohistochemistry. RESULTS Ginsenoside Rg1, APP, ferroptosis, and AD had 12 hub genes. GO enrichment and KEGG pathway annotation analysis showed that EGFR, SRC, protein hydrolysis, protein phosphorylation, the Relaxin pathway, and the FoxO signaling pathway play an important role in the potential mechanism of ginsenoside Rg1's under regulation of ferroptosis anti-AD through the modulation of APP-related signaling pathways. The APP/PS1 mice experiment verified that ginsenosides Rg1 and liproxstain-1 can promote the proliferation and differentiation. CONCLUSION Ginsenoside Rg1, APP and ferroptosis may act on EGFR, SRC, the Relaxin and FoxO signaling pathways to regulate protein metabolism, protein phosphorylation and other pathways to improve AD symptoms.
Collapse
Affiliation(s)
- Xu Deng
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Zixiong Qiu
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Xiaoshuai Chen
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Jiangxiu Liu
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Xiaowei Wang
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Jie Li
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Jiankai Zhang
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Xiaojun Cui
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China.
| | - Yuan Fu
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China.
| | - Mei Jiang
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China.
| |
Collapse
|
5
|
Toudji I, Toumi A, Chamberland É, Rossignol E. Interneuron odyssey: molecular mechanisms of tangential migration. Front Neural Circuits 2023; 17:1256455. [PMID: 37779671 PMCID: PMC10538647 DOI: 10.3389/fncir.2023.1256455] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 08/21/2023] [Indexed: 10/03/2023] Open
Abstract
Cortical GABAergic interneurons are critical components of neural networks. They provide local and long-range inhibition and help coordinate network activities involved in various brain functions, including signal processing, learning, memory and adaptative responses. Disruption of cortical GABAergic interneuron migration thus induces profound deficits in neural network organization and function, and results in a variety of neurodevelopmental and neuropsychiatric disorders including epilepsy, intellectual disability, autism spectrum disorders and schizophrenia. It is thus of paramount importance to elucidate the specific mechanisms that govern the migration of interneurons to clarify some of the underlying disease mechanisms. GABAergic interneurons destined to populate the cortex arise from multipotent ventral progenitor cells located in the ganglionic eminences and pre-optic area. Post-mitotic interneurons exit their place of origin in the ventral forebrain and migrate dorsally using defined migratory streams to reach the cortical plate, which they enter through radial migration before dispersing to settle in their final laminar allocation. While migrating, cortical interneurons constantly change their morphology through the dynamic remodeling of actomyosin and microtubule cytoskeleton as they detect and integrate extracellular guidance cues generated by neuronal and non-neuronal sources distributed along their migratory routes. These processes ensure proper distribution of GABAergic interneurons across cortical areas and lamina, supporting the development of adequate network connectivity and brain function. This short review summarizes current knowledge on the cellular and molecular mechanisms controlling cortical GABAergic interneuron migration, with a focus on tangential migration, and addresses potential avenues for cell-based interneuron progenitor transplants in the treatment of neurodevelopmental disorders and epilepsy.
Collapse
Affiliation(s)
- Ikram Toudji
- Centre Hospitalier Universitaire (CHU) Sainte-Justine Research Center, Montréal, QC, Canada
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada
| | - Asmaa Toumi
- Centre Hospitalier Universitaire (CHU) Sainte-Justine Research Center, Montréal, QC, Canada
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, QC, Canada
| | - Émile Chamberland
- Centre Hospitalier Universitaire (CHU) Sainte-Justine Research Center, Montréal, QC, Canada
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada
| | - Elsa Rossignol
- Centre Hospitalier Universitaire (CHU) Sainte-Justine Research Center, Montréal, QC, Canada
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada
- Department of Pediatrics, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
6
|
Ghate PS, Vacharasin JM, Ward JA, Nowling D, Kay V, Cowen MH, Lawlor MK, McCord M, Xu H, Carmona E, Cheon SH, Chukwurah E, Walla M, Lizarraga SB. The Warburg micro syndrome protein RAB3GAP1 modulates neuronal morphogenesis and interacts with axon elongation end ER-Golgi trafficking factors. Neurobiol Dis 2023; 184:106215. [PMID: 37385458 DOI: 10.1016/j.nbd.2023.106215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 05/26/2023] [Accepted: 06/26/2023] [Indexed: 07/01/2023] Open
Abstract
RAB3GAP1 is GTPase activating protein localized to the ER and Golgi compartments. In humans, mutations in RAB3GAP1 are the most common cause of Warburg Micro syndrome, a neurodevelopmental disorder associated with intellectual disability, microcephaly, and agenesis of the corpus callosum. We found that downregulation of RAB3GAP1 leads to a reduction in neurite outgrowth and complexity in human stem cell derived neurons. To further define the cellular function of RAB3GAP1, we sought to identify novel interacting proteins. We used a combination of mass spectrometry, co-immunoprecipitation and colocalization analysis and identified two novel interactors of RAB3GAP1: the axon elongation factor Dedicator of cytokinesis 7 (DOCK7) and the TATA modulatory factor 1 (TMF1) a modulator of Endoplasmic Reticulum (ER) to Golgi trafficking. To define the relationship between RAB3GAP1 and its two novel interactors, we analyzed their localization to different subcellular compartments in neuronal and non-neuronal cells with loss of RAB3GAP1. We find that RAB3GAP1 is important for the sub-cellular localization of TMF1 and DOCK7 across different compartments of the Golgi and endoplasmic reticulum. In addition, we find that loss of function mutations in RAB3GAP1 lead to dysregulation of pathways that are activated in response to the cellular stress like ATF6, MAPK, and PI3-AKT signaling. In summary, our findings suggest a novel role for RAB3GAP1 in neurite outgrowth that could encompass the regulation of proteins that control axon elongation, ER-Golgi trafficking, as well as pathways implicated in response to cellular stress.
Collapse
Affiliation(s)
- Pankaj S Ghate
- Department of Biology and Center for Childhood Neurotherapeutics, University of South Carolina, Columbia, SC, United States of America
| | - Janay M Vacharasin
- Department of Biology and Center for Childhood Neurotherapeutics, University of South Carolina, Columbia, SC, United States of America
| | - Joseph A Ward
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, United States of America; Center for Translational Neuroscience, Brown University, Providence, RI, United states of America
| | - Duncan Nowling
- Department of Biology and Center for Childhood Neurotherapeutics, University of South Carolina, Columbia, SC, United States of America
| | - Valerie Kay
- Department of Biology and Center for Childhood Neurotherapeutics, University of South Carolina, Columbia, SC, United States of America
| | - Mara H Cowen
- Department of Biology and Center for Childhood Neurotherapeutics, University of South Carolina, Columbia, SC, United States of America
| | - Mary-Kate Lawlor
- Department of Biology and Center for Childhood Neurotherapeutics, University of South Carolina, Columbia, SC, United States of America
| | - Mikayla McCord
- Department of Biology and Center for Childhood Neurotherapeutics, University of South Carolina, Columbia, SC, United States of America
| | - Hailey Xu
- Department of Biology and Center for Childhood Neurotherapeutics, University of South Carolina, Columbia, SC, United States of America
| | - Esteban Carmona
- Department of Biology and Center for Childhood Neurotherapeutics, University of South Carolina, Columbia, SC, United States of America
| | - Seon-Hye Cheon
- Department of Biology and Center for Childhood Neurotherapeutics, University of South Carolina, Columbia, SC, United States of America
| | - Evelyn Chukwurah
- Department of Biology and Center for Childhood Neurotherapeutics, University of South Carolina, Columbia, SC, United States of America
| | - Mike Walla
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, United States of America
| | - Sofia B Lizarraga
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, United States of America; Center for Translational Neuroscience, Brown University, Providence, RI, United states of America.
| |
Collapse
|
7
|
Wang L, Li G, Zhou Z, Ge C, Chen Q, Liu Y, Zhang N, Zhang K, Niu M, Li W, Zhong X, Wu S, Zhang J, Liu Y. Chromatin-associated OGT promotes the malignant progression of hepatocellular carcinoma by activating ZNF263. Oncogene 2023:10.1038/s41388-023-02751-1. [PMID: 37353617 DOI: 10.1038/s41388-023-02751-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 06/01/2023] [Accepted: 06/12/2023] [Indexed: 06/25/2023]
Abstract
Reversible and dynamic O-GlcNAcylation regulates vast networks of highly coordinated cellular and nuclear processes. Although dysregulation of the sole enzyme O-GlcNAc transferase (OGT) was shown to be associated with the progression of hepatocellular carcinoma (HCC), the mechanisms by which OGT controls the cis-regulatory elements in the genome and performs transcriptional functions remain unclear. Here, we demonstrate that elevated OGT levels enhance HCC proliferation and metastasis, in vitro and in vivo, by orchestrating the transcription of numerous regulators of malignancy. Diverse transcriptional regulators are recruited by OGT in HCC cells undergoing malignant progression, which shapes genome-wide OGT chromatin cis-element occupation. Furthermore, an unrecognized cooperation between ZNF263 and OGT is crucial for activating downstream transcription in HCC cells. We reveal that O-GlcNAcylation of Ser662 is responsible for the chromatin association of ZNF263 at candidate gene promoters and the OGT-facilitated HCC malignant phenotypes. Our data establish the importance of aberrant OGT activity and ZNF263 O-GlcNAcylation in the malignant progression of HCC and support the investigation of OGT as a therapeutic target for HCC.
Collapse
Affiliation(s)
- Lingyan Wang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Guofang Li
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Ziyu Zhou
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Chang Ge
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Qiushi Chen
- Department of Chemistry, The University of Hong Kong, Hong Kong, China
- Laboratory for Synthetic Chemistry and Chemical Biology Limited, Hong Kong, China
| | - Yajie Liu
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Nana Zhang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Keren Zhang
- Department of Chemistry, College of Science, Southern University of Science and Technology, Shenzhen, China
| | - Mingshan Niu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Wenli Li
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Xiaomin Zhong
- Department of Oncology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Sijin Wu
- Shenzhen Jingtai Technology Co., Ltd. (XtalPi), Shenzhen, China.
| | - Jianing Zhang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China.
| | - Yubo Liu
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China.
| |
Collapse
|
8
|
Wimmer R, Baffet AD. The microtubule cytoskeleton of radial glial progenitor cells. Curr Opin Neurobiol 2023; 80:102709. [PMID: 37003105 DOI: 10.1016/j.conb.2023.102709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/14/2023] [Accepted: 02/23/2023] [Indexed: 04/01/2023]
Abstract
A high number of genetic mutations associated with cortical malformations are found in genes coding for microtubule-related factors. This has stimulated research to understand how the various microtubule-based processes are regulated to build a functional cerebral cortex. Here, we focus our review on the radial glial progenitor cells, the stem cells of the developing neocortex, summarizing research mostly performed in rodents and humans. We highlight how the centrosomal and acentrosomal microtubule networks are organized during interphase to support polarized transport and proper attachment of the apical and basal processes. We describe the molecular mechanism for interkinetic nuclear migration (INM), a microtubule-dependent oscillation of the nucleus. Finally, we describe how the mitotic spindle is built to ensure proper chromosome segregation, with a strong focus on factors mutated in microcephaly.
Collapse
Affiliation(s)
- Ryszard Wimmer
- Institut Curie, PSL Research University, CNRS UMR144, Paris, France. https://twitter.com/RyWim
| | - Alexandre D Baffet
- Institut Curie, PSL Research University, CNRS UMR144, Paris, France; Institut national de la santé et de la recherche médicale (INSERM), France.
| |
Collapse
|
9
|
Proteomic Analysis of Murine Bone Marrow Very Small Embryonic-like Stem Cells at Steady-State Conditions and after In Vivo Stimulation by Nicotinamide and Follicle-Stimulating Factor Reflects their Germ-Lineage Origin and Multi Germ Layer Differentiation Potential. Stem Cell Rev Rep 2023; 19:120-132. [PMID: 35986128 PMCID: PMC9823037 DOI: 10.1007/s12015-022-10445-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/09/2022] [Indexed: 01/29/2023]
Abstract
Very small embryonic-like stem cells (VSELs) are a dormant population of development early stem cells deposited in adult tissues that as demonstrated contribute to tissue/organ repair and regeneration. We postulated developmental relationship of these cells to migrating primordial germ cells (PGCs) and explained the quiescent state of these cells by the erasure of differently methylated regions (DMRs) at some of the paternally imprinted genes involved in embryogenesis. Recently, we reported that VSELs began to proliferate and expand in vivo in murine bone marrow (BM) after exposure to nicotinamide (NAM) and selected pituitary and gonadal sex hormones. In the current report, we performed proteomic analysis of VSELs purified from murine bone marrow (BM) after repeated injections of NAM + Follicle-Stimulating Hormone (FSH) that in our previous studies turned out to be an effective combination to expand these cells. By employing the Gene Ontology (GO) resources, we have performed a combination of standard GO annotations (GO-CAM) to produce a network between BM steady-state conditions VSELs (SSC-VSELS) and FSH + NAM expanded VSELs (FSH + NAM VSELs). We have identified several GO biological processes regulating development, organogenesis, gene expression, signal transduction, Wnt signaling, insulin signaling, cytoskeleton organization, cell adhesion, inhibiting apoptosis, responses to extra- and intracellular stimuli, protein transport and stabilization, protein phosphorylation and ubiquitination, DNA repair, immune response, and regulation of circadian rhythm. We report that VSELs express a unique panel of proteins that only partially overlapped with the proteome of BM - derived hematopoietic stem cells (HSCs) and hematopoietic mononuclear cells (MNCs) and respond to FSH + NAM stimulation by expressing proteins involved in the development of all three germ layers. Thus, our current data supports further germ-lineage origin and multi germ layer differentiation potential of these cells.
Collapse
|
10
|
Kivrak Pfiffner F, Koller S, Ménétrey A, Graf U, Bähr L, Maspoli A, Hackenberg A, Kottke R, Gerth-Kahlert C, Berger W. Homozygosity for a Novel DOCK7 Variant Due to Segmental Uniparental Isodisomy of Chromosome 1 Associated with Early Infantile Epileptic Encephalopathy (EIEE) and Cortical Visual Impairment. Int J Mol Sci 2022; 23:ijms23137382. [PMID: 35806387 PMCID: PMC9266905 DOI: 10.3390/ijms23137382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 06/19/2022] [Accepted: 06/29/2022] [Indexed: 11/16/2022] Open
Abstract
Early infantile epileptic encephalopathy (EIEE) is a severe neurologic and neurodevelopmental disease that manifests in the first year of life. It shows a high degree of genetic heterogeneity, but the genetic origin is only identified in half of the cases. We report the case of a female child initially diagnosed with Leber congenital amaurosis (LCA), an early-onset retinal dystrophy due to photoreceptor cell degeneration in the retina. The first examination at 9 months of age revealed no reaction to light or objects and showed wandering eye movements. Ophthalmological examination did not show any ocular abnormalities. The patient displayed mildly dysmorphic features and a global developmental delay. Brain MRI demonstrated pontine hypo-/dysplasia. The patient developed myoclonic epileptic seizures and epileptic spasms with focal and generalized epileptiform discharges on electroencephalogram (EEG) at the age of 16 months. Genetic screening for a potentially pathogenic DNA sequence variant by whole-exome sequencing (WES) revealed a novel, conserved, homozygous frameshift variant (c.5391delA, p.(Ala1798LeufsTer59)) in exon 42 of the DOCK7 gene (NM_001271999.1). Further analysis by SNP array (Karyomapping) showed loss of heterozygosity (LOH) in four segments of chromosome 1. WES data of the parents and the index patient (trio analysis) demonstrated that chromosome 1 was exclusively inherited from the mother. Four LOH segments of chromosome 1 alternately showed isodisomy (UPiD) and heterodisomy (UPhD). In WES data, the father was a noncarrier, and the mother was heterozygous for this DOCK7 variant. The DOCK7 gene is located in 1p31.3, a region situated in one of the four isodisomic segments of chromosome 1, explaining the homozygosity seen in the affected child. Finally, Sanger sequencing confirmed maternal UPiD for the DOCK7 variant. Homozygous or compound heterozygous pathogenic variants in the DOCK7 (dedicator of cytokinesis 7) gene are associated with autosomal recessive, early infantile epileptic encephalopathy 23 (EIEE23; OMIM #615,859), a rare and heterogeneous group of neurodevelopmental disorders diagnosed during early childhood. To our knowledge, this is the first report of segmental uniparental iso- and heterodisomy of chromosome 1, leading to homozygosity of the DOCK7 frameshift variant in the affected patient.
Collapse
Affiliation(s)
- Fatma Kivrak Pfiffner
- Institute of Medical Molecular Genetics, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland; (F.K.P.); (S.K.); (U.G.); (L.B.); (A.M.)
| | - Samuel Koller
- Institute of Medical Molecular Genetics, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland; (F.K.P.); (S.K.); (U.G.); (L.B.); (A.M.)
| | - Anika Ménétrey
- Department of Pediatric Neurology, University Children’s Hospital, University of Zurich, 8032 Zurich, Switzerland; (A.M.); (A.H.)
| | - Urs Graf
- Institute of Medical Molecular Genetics, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland; (F.K.P.); (S.K.); (U.G.); (L.B.); (A.M.)
| | - Luzy Bähr
- Institute of Medical Molecular Genetics, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland; (F.K.P.); (S.K.); (U.G.); (L.B.); (A.M.)
| | - Alessandro Maspoli
- Institute of Medical Molecular Genetics, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland; (F.K.P.); (S.K.); (U.G.); (L.B.); (A.M.)
| | - Annette Hackenberg
- Department of Pediatric Neurology, University Children’s Hospital, University of Zurich, 8032 Zurich, Switzerland; (A.M.); (A.H.)
| | - Raimund Kottke
- Department of Diagnostic Imaging, University Children’s Hospital, University of Zurich, 8032 Zurich, Switzerland;
| | | | - Wolfgang Berger
- Institute of Medical Molecular Genetics, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland; (F.K.P.); (S.K.); (U.G.); (L.B.); (A.M.)
- Neuroscience Center Zurich, University and ETH Zurich, 8057 Zurich, Switzerland
- Center for Integrative Human Physiology, University of Zurich, 8057 Zurich, Switzerland
- Correspondence:
| |
Collapse
|
11
|
Kapoor D, Anand A, Siddiqui S, Sharma S. A novel pathogenic variant in DOCK 7 gene in an infant with dysmorphism, epileptic encephalopathy and cortical blindness. Clin Dysmorphol 2022; 31:39-41. [PMID: 34561314 DOI: 10.1097/mcd.0000000000000393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Dipti Kapoor
- Department of Pediatrics, Lady Hardinge Medical College, New Delhi
| | - Aakanksha Anand
- Department of Pediatrics, Lady Hardinge Medical College, New Delhi
| | - Shahyan Siddiqui
- Department of Neuroimaging and Intervention Radiology, STAR Institute of Neurosciences, STAR Hospitals, Hyderabad
| | | |
Collapse
|
12
|
Bartlett T. Fusion of single-cell transcriptome and DNA-binding data, for genomic network inference in cortical development. BMC Bioinformatics 2021; 22:301. [PMID: 34088262 PMCID: PMC8176738 DOI: 10.1186/s12859-021-04201-9] [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: 11/05/2020] [Accepted: 05/12/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Network models are well-established as very useful computational-statistical tools in cell biology. However, a genomic network model based only on gene expression data can, by definition, only infer gene co-expression networks. Hence, in order to infer gene regulatory patterns, it is necessary to also include data related to binding of regulatory factors to DNA. RESULTS We propose a new dynamic genomic network model, for inferring patterns of genomic regulatory influence in dynamic processes such as development. Our model fuses experiment-specific gene expression data with publicly available DNA-binding data. The method we propose is computationally efficient, and can be applied to genome-wide data with tens of thousands of transcripts. Thus, our method is well suited for use as an exploratory tool for genome-wide data. We apply our method to data from human fetal cortical development, and our findings confirm genomic regulatory patterns which are recognised as being fundamental to neuronal development. CONCLUSIONS Our method provides a mathematical/computational toolbox which, when coupled with targeted experiments, will reveal and confirm important new functional genomic regulatory processes in mammalian development.
Collapse
Affiliation(s)
- Thomas Bartlett
- University College London, Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
13
|
Walgrave H, Balusu S, Snoeck S, Vanden Eynden E, Craessaerts K, Thrupp N, Wolfs L, Horré K, Fourne Y, Ronisz A, Silajdžić E, Penning A, Tosoni G, Callaerts-Vegh Z, D'Hooge R, Thal DR, Zetterberg H, Thuret S, Fiers M, Frigerio CS, De Strooper B, Salta E. Restoring miR-132 expression rescues adult hippocampal neurogenesis and memory deficits in Alzheimer's disease. Cell Stem Cell 2021; 28:1805-1821.e8. [PMID: 34033742 DOI: 10.1016/j.stem.2021.05.001] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/24/2021] [Accepted: 04/30/2021] [Indexed: 12/14/2022]
Abstract
Neural stem cells residing in the hippocampal neurogenic niche sustain lifelong neurogenesis in the adult brain. Adult hippocampal neurogenesis (AHN) is functionally linked to mnemonic and cognitive plasticity in humans and rodents. In Alzheimer's disease (AD), the process of generating new neurons at the hippocampal neurogenic niche is impeded, yet the mechanisms involved are unknown. Here we identify miR-132, one of the most consistently downregulated microRNAs in AD, as a potent regulator of AHN, exerting cell-autonomous proneurogenic effects in adult neural stem cells and their progeny. Using distinct AD mouse models, cultured human primary and established neural stem cells, and human patient material, we demonstrate that AHN is directly affected by AD pathology. miR-132 replacement in adult mouse AD hippocampus restores AHN and relevant memory deficits. Our findings corroborate the significance of AHN in mouse models of AD and reveal the possible therapeutic potential of targeting miR-132 in neurodegeneration.
Collapse
Affiliation(s)
- Hannah Walgrave
- VIB Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Sriram Balusu
- VIB Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Sarah Snoeck
- Laboratory of Neurogenesis and Neurodegeneration, Netherlands Institute for Neuroscience, 1105BA Amsterdam, the Netherlands
| | - Elke Vanden Eynden
- VIB Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Katleen Craessaerts
- VIB Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Nicky Thrupp
- VIB Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Leen Wolfs
- VIB Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Katrien Horré
- VIB Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Yannick Fourne
- VIB Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Alicja Ronisz
- KU Leuven, Leuven Brain Institute, 3000 Leuven, Belgium; Laboratory for Neuropathology, KU Leuven, and Department of Pathology, UZ Leuven, 3000 Leuven, Belgium
| | - Edina Silajdžić
- Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE5 9RX, UK
| | - Amber Penning
- Laboratory of Neurogenesis and Neurodegeneration, Netherlands Institute for Neuroscience, 1105BA Amsterdam, the Netherlands
| | - Giorgia Tosoni
- Laboratory of Neurogenesis and Neurodegeneration, Netherlands Institute for Neuroscience, 1105BA Amsterdam, the Netherlands
| | - Zsuzsanna Callaerts-Vegh
- KU Leuven, Leuven Brain Institute, 3000 Leuven, Belgium; Laboratory for Biological Psychology, KU Leuven, 3000 Leuven, Belgium
| | - Rudi D'Hooge
- KU Leuven, Leuven Brain Institute, 3000 Leuven, Belgium; Laboratory for Biological Psychology, KU Leuven, 3000 Leuven, Belgium
| | - Dietmar Rudolf Thal
- KU Leuven, Leuven Brain Institute, 3000 Leuven, Belgium; Laboratory for Neuropathology, KU Leuven, and Department of Pathology, UZ Leuven, 3000 Leuven, Belgium
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 431 80 Mölndal, Sweden; Department of Neurodegenerative Disease, UCL Institute of Neurology, London, WC1N 3BG, UK; Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, 431 80 Mölndal, Sweden; UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
| | - Sandrine Thuret
- Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE5 9RX, UK
| | - Mark Fiers
- VIB Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Leuven Brain Institute, 3000 Leuven, Belgium
| | | | - Bart De Strooper
- VIB Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Leuven Brain Institute, 3000 Leuven, Belgium; UK Dementia Research Institute at UCL, London, WC1E 6BT, UK.
| | - Evgenia Salta
- Laboratory of Neurogenesis and Neurodegeneration, Netherlands Institute for Neuroscience, 1105BA Amsterdam, the Netherlands.
| |
Collapse
|
14
|
Thompson AP, Bitsina C, Gray JL, von Delft F, Brennan PE. RHO to the DOCK for GDP disembarking: Structural insights into the DOCK GTPase nucleotide exchange factors. J Biol Chem 2021; 296:100521. [PMID: 33684443 PMCID: PMC8063744 DOI: 10.1016/j.jbc.2021.100521] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 03/04/2021] [Accepted: 03/04/2021] [Indexed: 01/16/2023] Open
Abstract
The human dedicator of cytokinesis (DOCK) family consists of 11 structurally conserved proteins that serve as atypical RHO guanine nucleotide exchange factors (RHO GEFs). These regulatory proteins act as mediators in numerous cellular cascades that promote cytoskeletal remodeling, playing roles in various crucial processes such as differentiation, migration, polarization, and axon growth in neurons. At the molecular level, DOCK DHR2 domains facilitate nucleotide dissociation from small GTPases, a process that is otherwise too slow for rapid spatiotemporal control of cellular signaling. Here, we provide an overview of the biological and structural characteristics for the various DOCK proteins and describe how they differ from other RHO GEFs and between DOCK subfamilies. The expression of the family varies depending on cell or tissue type, and they are consequently implicated in a broad range of disease phenotypes, particularly in the brain. A growing body of available structural information reveals the mechanism by which the catalytic DHR2 domain elicits nucleotide dissociation and also indicates strategies for the discovery and design of high-affinity small-molecule inhibitors. Such compounds could serve as chemical probes to interrogate the cellular function and provide starting points for drug discovery of this important class of enzymes.
Collapse
Affiliation(s)
- Andrew P Thompson
- Nuffield Department of Medicine, Alzheimer's Research UK Oxford Drug Discovery Institute, University of Oxford, Oxford, United Kingdom; Nuffield Department of Medicine, Target Discovery Institute, University of Oxford, Oxford, United Kingdom; Nuffield Department of Medicine, Centre for Medicines Discovery, University of Oxford, Oxford, United Kingdom
| | - Christina Bitsina
- Nuffield Department of Medicine, Alzheimer's Research UK Oxford Drug Discovery Institute, University of Oxford, Oxford, United Kingdom; Nuffield Department of Medicine, Target Discovery Institute, University of Oxford, Oxford, United Kingdom; Nuffield Department of Medicine, Centre for Medicines Discovery, University of Oxford, Oxford, United Kingdom
| | - Janine L Gray
- Nuffield Department of Medicine, Alzheimer's Research UK Oxford Drug Discovery Institute, University of Oxford, Oxford, United Kingdom; Nuffield Department of Medicine, Target Discovery Institute, University of Oxford, Oxford, United Kingdom; Nuffield Department of Medicine, Centre for Medicines Discovery, University of Oxford, Oxford, United Kingdom
| | - Frank von Delft
- Nuffield Department of Medicine, Alzheimer's Research UK Oxford Drug Discovery Institute, University of Oxford, Oxford, United Kingdom; Nuffield Department of Medicine, Centre for Medicines Discovery, University of Oxford, Oxford, United Kingdom; Diamond Light Source, Harwell Science and Innovation Campus, Didcot, United Kingdom; Department of Biochemistry, University of Johannesburg, Auckland Park, South Africa
| | - Paul E Brennan
- Nuffield Department of Medicine, Alzheimer's Research UK Oxford Drug Discovery Institute, University of Oxford, Oxford, United Kingdom; Nuffield Department of Medicine, Target Discovery Institute, University of Oxford, Oxford, United Kingdom; Nuffield Department of Medicine, Centre for Medicines Discovery, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
15
|
Haberlandt E, Valovka T, Janjic T, Müller T, Blatsios G, Karall D, Janecke AR. Characteristic facial features and cortical blindness distinguish the DOCK7-related epileptic encephalopathy. Mol Genet Genomic Med 2021; 9:e1607. [PMID: 33471954 PMCID: PMC8104163 DOI: 10.1002/mgg3.1607] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/26/2020] [Accepted: 01/04/2021] [Indexed: 12/24/2022] Open
Abstract
Background The epileptic encephalopathies display extensive locus and allelic heterogeneity. Biallelic truncating DOCK7 variants were recently reported in five children with early‐onset epilepsy, intellectual disability, and cortical blindness, indicating that DOCK7 deficiency causes a specific type of epileptic encephalopathy. Methods We identified 23‐ and 27‐year‐old siblings with the clinical pattern reported for DOCK7 deficiency, and conducted genome‐wide linkage analysis and WES. The consequences of a DOCK7 variant were analyzed on the transcript and protein level in patients’ fibroblasts. Results We identified a novel homozygous DOCK7 frameshift variant, an intragenic tandem duplication of 124‐kb, previously missed by CGH array, in adult patients. Patients display atrophy in the occipital lobe and pontine hypoplasia with marked pontobulbar sulcus, and focal atrophy of occasional cerebellar folia is a novel finding. Recognizable dysmorphic features include normo‐brachycephaly, narrow forehead, low anterior and posterior hairlines, prominent ears, full cheeks, and long eyelashes. Our patients function on the level of 4‐year‐old children, never showed signs of regression, and seizures are largely controlled with multi‐pharmacotherapy. Studies of patients’ fibroblasts showed nonsense‐mediated RNA decay and lack of DOCK7 protein. Conclusion DOCK7 deficiency causes a definable clinical entity, a recognizable type of epileptic encephalopathy.
Collapse
Affiliation(s)
- Edda Haberlandt
- Department of Pediatrics I, Medical University of Innsbruck, Innsbruck, Austria.,Krankenhaus der Stadt Dornbirn, Kinder- und Jugendheilkunde, Dornbirn, Austria
| | - Taras Valovka
- Department of Pediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - Tanja Janjic
- Department of Neuroradiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Thomas Müller
- Department of Pediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - Georgios Blatsios
- Department of Ophthalmology, Medical University of Innsbruck, Innsbruck, Austria
| | - Daniela Karall
- Department of Pediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - Andreas R Janecke
- Department of Pediatrics I, Medical University of Innsbruck, Innsbruck, Austria.,Division of Human Genetics, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
16
|
Wang J, Li T, Wang JL, Xu Z, Meng W, Wu QF. Talpid3-Mediated Centrosome Integrity Restrains Neural Progenitor Delamination to Sustain Neurogenesis by Stabilizing Adherens Junctions. Cell Rep 2020; 33:108495. [DOI: 10.1016/j.celrep.2020.108495] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 08/03/2020] [Accepted: 11/17/2020] [Indexed: 12/21/2022] Open
|
17
|
Emond MR, Biswas S, Morrow ML, Jontes JD. Proximity-dependent Proteomics Reveals Extensive Interactions of Protocadherin-19 with Regulators of Rho GTPases and the Microtubule Cytoskeleton. Neuroscience 2020; 452:26-36. [PMID: 33010346 DOI: 10.1016/j.neuroscience.2020.09.033] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 09/02/2020] [Accepted: 09/14/2020] [Indexed: 12/15/2022]
Abstract
Protocadherin-19 belongs to the cadherin family of cell surface receptors and has been shown to play essential roles in the development of the vertebrate nervous system. Mutations in human Protocadherin-19 (PCDH19) lead to PCDH19 Female-limited epilepsy (PCDH19 FLE) in humans, characterized by the early onset of epileptic seizures in children and a range of cognitive and behavioral problems in adults. Despite being considered the second most prevalent gene in epilepsy, very little is known about the intercellular pathways in which it participates. In order to characterize the protein complexes within which Pcdh19 functions, we generated Pcdh19-BioID fusion proteins and utilized proximity-dependent biotinylation to identify neighboring proteins. Proteomic identification and analysis revealed that the Pcdh19 interactome is enriched in proteins that regulate Rho family GTPases, microtubule binding proteins and proteins that regulate cell divisions. We cloned the centrosomal protein Nedd1 and the RacGEF Dock7 and verified their interactions with Pcdh19 in vitro. Our findings provide the first comprehensive insights into the interactome of Pcdh19, and provide a platform for future investigations into the cellular and molecular biology of this protein critical to the proper development of the nervous system.
Collapse
Affiliation(s)
- Michelle R Emond
- Department of Neuroscience, Ohio State University, United States
| | | | - Matthew L Morrow
- Department of Neuroscience, Ohio State University, United States
| | - James D Jontes
- Department of Neuroscience, Ohio State University, United States.
| |
Collapse
|
18
|
Chen Y, Chen Y, Yin W, Han H, Miller H, Li J, Herrada AA, Kubo M, Sui Z, Gong Q, Liu C. The regulation of DOCK family proteins on T and B cells. J Leukoc Biol 2020; 109:383-394. [PMID: 32542827 DOI: 10.1002/jlb.1mr0520-221rr] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 05/15/2020] [Accepted: 05/16/2020] [Indexed: 01/01/2023] Open
Abstract
The dedicator of cytokinesis (DOCK) family proteins consist of 11 members, each of which contains 2 domains, DOCK homology region (DHR)-1 and DHR-2, and as guanine nucleotide exchange factors, they mediate activation of small GTPases. Both DOCK2 and DOCK8 deficiencies in humans can cause severe combined immunodeficiency, but they have different characteristics. DOCK8 defect mainly causes high IgE, allergic disease, refractory skin virus infection, and increased incidence of malignant tumor, whereas DOCK2 defect mainly causes early-onset, invasive infection with less atopy and increased IgE. However, the underlying molecular mechanisms causing the disease remain unclear. This paper discusses the role of DOCK family proteins in regulating B and T cells, including development, survival, migration, activation, immune tolerance, and immune functions. Moreover, related signal pathways or molecule mechanisms are also described in this review. A greater understanding of DOCK family proteins and their regulation of lymphocyte functions may facilitate the development of new therapeutics for immunodeficient patients and improve their prognosis.
Collapse
Affiliation(s)
- Yuanyuan Chen
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Chen
- The Second Department of Pediatrics, Affiliated Hospital of Zunyi, Zunyi, Guizhou, China
| | - Wei Yin
- Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Han
- Department of Hematology of Liyuan Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Heather Miller
- The Laboratory of Intracellular Parasites, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Jianrong Li
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Andres A Herrada
- Lymphatic and Inflammation Research Laboratory, Facultad de Ciencias de la Salud, Instituto de Ciencias Biomedicas, Universidad Autonoma de Chile, Talca, Chile
| | - Masato Kubo
- Laboratory for Cytokine Regulation, Center for Integrative Medical Science (IMS), RIKEN Yokohama Institute, Yokohama, Kanagawa, Japan
| | - Zhiwei Sui
- Division of Medical and Biological Measurement, National Institute of Metrology, Beijing, China
| | - Quan Gong
- Department of immunology, School of Medicine, Yangtze University, Jingzhou, China.,Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, China
| | - Chaohong Liu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
19
|
Journiac N, Gilabert-Juan J, Cipriani S, Benit P, Liu X, Jacquier S, Faivre V, Delahaye-Duriez A, Csaba Z, Hourcade T, Melinte E, Lebon S, Violle-Poirsier C, Oury JF, Adle-Biassette H, Wang ZQ, Mani S, Rustin P, Gressens P, Nardelli J. Cell Metabolic Alterations due to Mcph1 Mutation in Microcephaly. Cell Rep 2020; 31:107506. [DOI: 10.1016/j.celrep.2020.03.070] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 12/21/2019] [Accepted: 03/21/2020] [Indexed: 12/13/2022] Open
|
20
|
Guimbal S, Morel A, Guérit D, Chardon M, Blangy A, Vives V. Dock5 is a new regulator of microtubule dynamic instability in osteoclasts. Biol Cell 2019; 111:271-283. [PMID: 31461543 DOI: 10.1111/boc.201900014] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 07/02/2019] [Accepted: 08/06/2019] [Indexed: 12/27/2022]
Abstract
BACKGROUND INFORMATION Osteoclast resorption is dependent on a podosome-rich structure called sealing zone. It tightly attaches the osteoclast to the bone creating a favourable acidic microenvironment for bone degradation. This adhesion structure needs to be stabilised by microtubules whose acetylation is maintained by down-regulation of deacetylase HDAC6 and/or of microtubule destabilising kinase GSK3β activities. We already established that Dock5 is a guanine nucleotide exchange factor for Rac1. As a consequence, Dock5 inhibition results in a decrease of the GTPase activity associated with impaired podosome assembly into sealing zones and resorbing activity in osteoclasts. More, administration of C21, a chemical compound that directly inhibits the exchange activity of Dock5, disrupts osteoclast podosome organisation and protects mice against bone degradation in models recapitulating major osteolytic diseases. RESULTS In this report, we show that Dock5 knockout osteoclasts also present a reduced acetylated tubulin level leading to a decreased length and duration of microtubule growth phases, whereas their growth speed remains unaffected. Dock5 does not act by direct interaction with the polymerised tubulin. Using specific Rac inhibitors, we showed that Dock5 regulates microtubule dynamic instability through Rac-dependent and -independent pathways. The latter involves GSK3β inhibitory serine 9 phosphorylation downstream of Akt activation but not HDAC6 activity. CONCLUSION We showed that Dock5 is a new regulator of microtubule dynamic instability in osteoclast. SIGNIFICANCE Dock5 dual role in the regulation of the actin cytoskeleton and microtubule, which both need to be intact for bone resorption, reinforces the fact that it is an interesting therapeutic target for osteolytic pathologies.
Collapse
Affiliation(s)
- Sarah Guimbal
- Centre de Recherche de Biologie Cellulaire (CRBM), CNRS UMR 5237, Montpellier, Cedex 5, 34293, France.,Montpellier University, Montpellier, Cedex 5, 34095, France
| | - Anne Morel
- Centre de Recherche de Biologie Cellulaire (CRBM), CNRS UMR 5237, Montpellier, Cedex 5, 34293, France.,Montpellier University, Montpellier, Cedex 5, 34095, France
| | - David Guérit
- Centre de Recherche de Biologie Cellulaire (CRBM), CNRS UMR 5237, Montpellier, Cedex 5, 34293, France.,Montpellier University, Montpellier, Cedex 5, 34095, France
| | - Manon Chardon
- Centre de Recherche de Biologie Cellulaire (CRBM), CNRS UMR 5237, Montpellier, Cedex 5, 34293, France.,Montpellier University, Montpellier, Cedex 5, 34095, France
| | - Anne Blangy
- Centre de Recherche de Biologie Cellulaire (CRBM), CNRS UMR 5237, Montpellier, Cedex 5, 34293, France.,Montpellier University, Montpellier, Cedex 5, 34095, France
| | - Virginie Vives
- Centre de Recherche de Biologie Cellulaire (CRBM), CNRS UMR 5237, Montpellier, Cedex 5, 34293, France.,Montpellier University, Montpellier, Cedex 5, 34095, France
| |
Collapse
|
21
|
Xu Z, Chen Y, Chen Y. Spatiotemporal Regulation of Rho GTPases in Neuronal Migration. Cells 2019; 8:cells8060568. [PMID: 31185627 PMCID: PMC6627650 DOI: 10.3390/cells8060568] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 06/01/2019] [Accepted: 06/04/2019] [Indexed: 12/17/2022] Open
Abstract
Neuronal migration is essential for the orchestration of brain development and involves several contiguous steps: interkinetic nuclear movement (INM), multipolar–bipolar transition, locomotion, and translocation. Growing evidence suggests that Rho GTPases, including RhoA, Rac, Cdc42, and the atypical Rnd members, play critical roles in neuronal migration by regulating both actin and microtubule cytoskeletal components. This review focuses on the spatiotemporal-specific regulation of Rho GTPases as well as their regulators and effectors in distinct steps during the neuronal migration process. Their roles in bridging extracellular signals and cytoskeletal dynamics to provide optimal structural support to the migrating neurons will also be discussed.
Collapse
Affiliation(s)
- Zhenyan Xu
- The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, Guangdong, China.
| | - Yuewen Chen
- The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, Guangdong, China.
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen 518057, Guangdong, China.
| | - Yu Chen
- The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, Guangdong, China.
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen 518057, Guangdong, China.
| |
Collapse
|
22
|
Elevated signature of a gene module coexpressed with CDC20 marks genomic instability in glioma. Proc Natl Acad Sci U S A 2019; 116:6975-6984. [PMID: 30877245 DOI: 10.1073/pnas.1814060116] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Genomic instability (GI) drives tumor heterogeneity and promotes tumor progression and therapy resistance. However, causative factors underlying GI and means for clinical detection of GI in glioma are inadequately identified. We describe here that elevated expression of a gene module coexpressed with CDC20 (CDC20-M), the activator of the anaphase-promoting complex in the cell cycle, marks GI in glioma. The CDC20-M, containing 139 members involved in cell proliferation, DNA damage response, and chromosome segregation, was found to be consistently coexpressed in glioma transcriptomes. The coexpression of these genes was conserved across multiple species and organ systems, particularly in human neural stem and progenitor cells. CDC20-M expression was not correlated with the morphological subtypes, nor with the recently defined molecular subtypes of glioma. CDC20-M signature was an independent and robust predictor for poorer prognosis in over 1,000 patients from four large databases. Elevated CDC20-M signature enabled the identification of individual glioma samples with severe chromosome instability and mutation burden and of primary glioma cell lines with extensive mitotic errors leading to chromosome mis-segregation. AURKA, a core member of CDC20-M, was amplified in one-third of CDC20-M-high gliomas with gene-dosage-dependent expression. MLN8237, a Food and Drug Administration-approved AURKA inhibitor, selectively killed temozolomide-resistant primary glioma cells in vitro and prolonged the survival of a patient-derived xenograft mouse model with a high-CDC20-M signature. Our findings suggest that application of the CDC20-M signature may permit more selective use of adjuvant therapies for glioma patients and that dysregulated CDC20-M members may provide a therapeutic vulnerability in glioma.
Collapse
|
23
|
Bai B, Guo YR, Zhang YH, Jin CC, Zhang JM, Chen H, Zhu BS. Novel DOCK7 mutations in a Chinese patient with early infantile epileptic encephalopathy 23. Chin Med J (Engl) 2019; 132:600-603. [PMID: 30807358 PMCID: PMC6415993 DOI: 10.1097/cm9.0000000000000100] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Affiliation(s)
- Bing Bai
- Department of Pediatrics, The First People's Hospital of Yunnan Province, Kunming, Yunnan 650032, China.,Medical Faculty, Kunming University of Science and Technology, Kunming, Yunnan 650032, China.,National Health Commission's Key Laboratory for Western Healthy Birth, Kunming, Yunnan 650032, China
| | - Yi-Ran Guo
- Center for Applied Genomics at the Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Yin-Hong Zhang
- Department of Pediatrics, The First People's Hospital of Yunnan Province, Kunming, Yunnan 650032, China.,Medical Faculty, Kunming University of Science and Technology, Kunming, Yunnan 650032, China.,National Health Commission's Key Laboratory for Western Healthy Birth, Kunming, Yunnan 650032, China
| | - Chan-Chan Jin
- Department of Pediatrics, The First People's Hospital of Yunnan Province, Kunming, Yunnan 650032, China.,Medical Faculty, Kunming University of Science and Technology, Kunming, Yunnan 650032, China.,National Health Commission's Key Laboratory for Western Healthy Birth, Kunming, Yunnan 650032, China
| | - Jin-Man Zhang
- Department of Pediatrics, The First People's Hospital of Yunnan Province, Kunming, Yunnan 650032, China.,Medical Faculty, Kunming University of Science and Technology, Kunming, Yunnan 650032, China.,National Health Commission's Key Laboratory for Western Healthy Birth, Kunming, Yunnan 650032, China
| | - Hong Chen
- Department of Pediatrics, The First People's Hospital of Yunnan Province, Kunming, Yunnan 650032, China.,Medical Faculty, Kunming University of Science and Technology, Kunming, Yunnan 650032, China.,National Health Commission's Key Laboratory for Western Healthy Birth, Kunming, Yunnan 650032, China
| | - Bao-Sheng Zhu
- Department of Pediatrics, The First People's Hospital of Yunnan Province, Kunming, Yunnan 650032, China.,Medical Faculty, Kunming University of Science and Technology, Kunming, Yunnan 650032, China.,National Health Commission's Key Laboratory for Western Healthy Birth, Kunming, Yunnan 650032, China
| |
Collapse
|
24
|
Tai Y, Gallo NB, Wang M, Yu JR, Van Aelst L. Axo-axonic Innervation of Neocortical Pyramidal Neurons by GABAergic Chandelier Cells Requires AnkyrinG-Associated L1CAM. Neuron 2019; 102:358-372.e9. [PMID: 30846310 DOI: 10.1016/j.neuron.2019.02.009] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 12/20/2018] [Accepted: 02/04/2019] [Indexed: 11/17/2022]
Abstract
Among the diverse interneuron subtypes in the neocortex, chandelier cells (ChCs) are the only population that selectively innervate pyramidal neurons (PyNs) at their axon initial segment (AIS), the site of action potential initiation, allowing them to exert powerful control over PyN output. Yet, mechanisms underlying their subcellular innervation of PyN AISs are unknown. To identify molecular determinants of ChC/PyN AIS innervation, we performed an in vivo RNAi screen of PyN-expressed axonal cell adhesion molecules (CAMs) and select Ephs/ephrins. Strikingly, we found the L1 family member L1CAM to be the only molecule required for ChC/PyN AIS innervation. Further, we show that L1CAM is required during both the establishment and maintenance of innervation, and that selective innervation of PyN AISs by ChCs requires AIS anchoring of L1CAM by the cytoskeletal ankyrin-G/βIV-spectrin complex. Thus, our findings identify PyN-expressed L1CAM as a critical CAM required for innervation of neocortical PyN AISs by ChCs. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Yilin Tai
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Nicholas B Gallo
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY 11794, USA
| | - Minghui Wang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Jia-Ray Yu
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Linda Van Aelst
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA.
| |
Collapse
|
25
|
Turkdogan D, Turkyilmaz A, Gormez Z, Sager G, Ekinci G. A novel truncating mutation of DOCK7 gene with an early-onset non-encephalopathic epilepsy. Seizure 2019; 66:12-14. [PMID: 30771731 DOI: 10.1016/j.seizure.2019.01.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 11/16/2018] [Accepted: 01/12/2019] [Indexed: 11/16/2022] Open
Affiliation(s)
- Dilsad Turkdogan
- Marmara University, Medical Faculty, Department of Pediatric Neurology, Pendik, Istanbul, Turkey.
| | - Ayberk Turkyilmaz
- Marmara University, Medical Faculty, Department of Medical Genetics, Pendik, Istanbul, Turkey
| | | | - Gunes Sager
- Marmara University, Medical Faculty, Department of Pediatric Neurology, Pendik, Istanbul, Turkey
| | - Gazanfer Ekinci
- Marmara University, Medical Faculty, Department of Neuroradiology, Pendik, Istanbul, Turkey
| |
Collapse
|
26
|
Tang LT, Diaz-Balzac CA, Rahman M, Ramirez-Suarez NJ, Salzberg Y, Lázaro-Peña MI, Bülow HE. TIAM-1/GEF can shape somatosensory dendrites independently of its GEF activity by regulating F-actin localization. eLife 2019; 8:38949. [PMID: 30694177 PMCID: PMC6370339 DOI: 10.7554/elife.38949] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 01/28/2019] [Indexed: 12/14/2022] Open
Abstract
Dendritic arbors are crucial for nervous system assembly, but the intracellular mechanisms that govern their assembly remain incompletely understood. Here, we show that the dendrites of PVD neurons in Caenorhabditis elegans are patterned by distinct pathways downstream of the DMA-1 leucine-rich transmembrane (LRR-TM) receptor. DMA-1/LRR-TM interacts through a PDZ ligand motif with the guanine nucleotide exchange factor TIAM-1/GEF in a complex with act-4/Actin to pattern higher order 4° dendrite branches by localizing F-actin to the distal ends of developing dendrites. Surprisingly, TIAM-1/GEF appears to function independently of Rac1 guanine nucleotide exchange factor activity. A partially redundant pathway, dependent on HPO-30/Claudin, regulates formation of 2° and 3° branches, possibly by regulating membrane localization and trafficking of DMA-1/LRR-TM. Collectively, our experiments suggest that HPO-30/Claudin localizes the DMA-1/LRR-TM receptor on PVD dendrites, which in turn can control dendrite patterning by directly modulating F-actin dynamics through TIAM-1/GEF.
Collapse
Affiliation(s)
- Leo Th Tang
- Department of Genetics, Albert Einstein College of Medicine, Bronx, United States
| | - Carlos A Diaz-Balzac
- Department of Genetics, Albert Einstein College of Medicine, Bronx, United States
| | - Maisha Rahman
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, United States
| | | | - Yehuda Salzberg
- Department of Genetics, Albert Einstein College of Medicine, Bronx, United States
| | - Maria I Lázaro-Peña
- Department of Genetics, Albert Einstein College of Medicine, Bronx, United States
| | - Hannes E Bülow
- Department of Genetics, Albert Einstein College of Medicine, Bronx, United States
| |
Collapse
|
27
|
Broix L, Asselin L, Silva CG, Ivanova EL, Tilly P, Gilet JG, Lebrun N, Jagline H, Muraca G, Saillour Y, Drouot N, Reilly ML, Francis F, Benmerah A, Bahi-Buisson N, Belvindrah R, Nguyen L, Godin JD, Chelly J, Hinckelmann MV. Ciliogenesis and cell cycle alterations contribute to KIF2A-related malformations of cortical development. Hum Mol Genet 2019; 27:224-238. [PMID: 29077851 DOI: 10.1093/hmg/ddx384] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 10/17/2017] [Indexed: 11/13/2022] Open
Abstract
Genetic findings reported by our group and others showed that de novo missense variants in the KIF2A gene underlie malformations of brain development called pachygyria and microcephaly. Though KIF2A is known as member of the Kinesin-13 family involved in the regulation of microtubule end dynamics through its ATP dependent MT-depolymerase activity, how KIF2A variants lead to brain malformations is still largely unknown. Using cellular and in utero electroporation approaches, we show here that KIF2A disease-causing variants disrupts projection neuron positioning and interneuron migration, as well as progenitors proliferation. Interestingly, further dissection of this latter process revealed that ciliogenesis regulation is also altered during progenitors cell cycle. Altogether, our data suggest that deregulation of the coupling between ciliogenesis and cell cycle might contribute to the pathogenesis of KIF2A-related brain malformations. They also raise the issue whether ciliogenesis defects are a hallmark of other brain malformations, such as those related to tubulins and MT-motor proteins variants.
Collapse
Affiliation(s)
- Loïc Broix
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch 67400, France.,CNRS U7104, Illkirch 67400, France.,INSERM U964, Illkirch 67400, France.,Université de Strasbourg, Illkirch 67400, France.,Institut Cochin, INSERM U1016, CNRS U8104, Paris Descartes University, Paris 75000, France
| | - Laure Asselin
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch 67400, France.,CNRS U7104, Illkirch 67400, France.,INSERM U964, Illkirch 67400, France.,Université de Strasbourg, Illkirch 67400, France
| | - Carla G Silva
- GIGA-Neurosciences, University of Liège, C.H.U. Sart Tilman, Liège 4000, Belgium
| | - Ekaterina L Ivanova
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch 67400, France.,CNRS U7104, Illkirch 67400, France.,INSERM U964, Illkirch 67400, France.,Université de Strasbourg, Illkirch 67400, France
| | - Peggy Tilly
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch 67400, France.,CNRS U7104, Illkirch 67400, France.,INSERM U964, Illkirch 67400, France.,Université de Strasbourg, Illkirch 67400, France
| | - Johan G Gilet
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch 67400, France.,CNRS U7104, Illkirch 67400, France.,INSERM U964, Illkirch 67400, France.,Université de Strasbourg, Illkirch 67400, France
| | - Nicolas Lebrun
- Institut Cochin, INSERM U1016, CNRS U8104, Paris Descartes University, Paris 75000, France
| | - Hélène Jagline
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch 67400, France.,CNRS U7104, Illkirch 67400, France.,INSERM U964, Illkirch 67400, France.,Université de Strasbourg, Illkirch 67400, France
| | - Giuseppe Muraca
- Institut Cochin, INSERM U1016, CNRS U8104, Paris Descartes University, Paris 75000, France
| | - Yoann Saillour
- Institut Cochin, INSERM U1016, CNRS U8104, Paris Descartes University, Paris 75000, France
| | - Nathalie Drouot
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch 67400, France.,CNRS U7104, Illkirch 67400, France.,INSERM U964, Illkirch 67400, France.,Université de Strasbourg, Illkirch 67400, France
| | - Madeline Louise Reilly
- Paris Diderot University, Paris 75013, France.,INSERM UMR 1163, Paris 75015, France.,Paris Descartes - Sorbonne Paris Cité University, Imagine Institute, Paris 75015, France
| | - Fiona Francis
- Inserm UMR-S 839, Paris 75005, France.,Sorbonne Université, Université Pierre et Marie Curie, Paris 75000, France.,Institut du Fer à Moulin, Paris 75000, France
| | - Alexandre Benmerah
- INSERM UMR 1163, Paris 75015, France.,Paris Descartes - Sorbonne Paris Cité University, Imagine Institute, Paris 75015, France
| | - Nadia Bahi-Buisson
- Paris Diderot University, Paris 75013, France.,INSERM UMR 1163, Paris 75015, France
| | - Richard Belvindrah
- Inserm UMR-S 839, Paris 75005, France.,Sorbonne Université, Université Pierre et Marie Curie, Paris 75000, France.,Institut du Fer à Moulin, Paris 75000, France
| | - Laurent Nguyen
- GIGA-Neurosciences, University of Liège, C.H.U. Sart Tilman, Liège 4000, Belgium
| | - Juliette D Godin
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch 67400, France.,CNRS U7104, Illkirch 67400, France.,INSERM U964, Illkirch 67400, France.,Université de Strasbourg, Illkirch 67400, France
| | - Jamel Chelly
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch 67400, France.,CNRS U7104, Illkirch 67400, France.,INSERM U964, Illkirch 67400, France.,Université de Strasbourg, Illkirch 67400, France.,Service de Diagnostic Génétique, Hôpital Civil de Strasbourg, Hôpitaux Universitaires de Strasbourg, Strasbourg 67000, France
| | - Maria-Victoria Hinckelmann
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch 67400, France.,CNRS U7104, Illkirch 67400, France.,INSERM U964, Illkirch 67400, France.,Université de Strasbourg, Illkirch 67400, France
| |
Collapse
|
28
|
Wang C, Ren H, Dong H, Liang M, Wu Q, Liao Y. 18q22.1-qter deletion and 4p16.3 microduplication in a boy with speech delay and mental retardation: case report and review of the literature. Mol Cytogenet 2018; 11:55. [PMID: 30377449 PMCID: PMC6194714 DOI: 10.1186/s13039-018-0404-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 10/01/2018] [Indexed: 11/13/2022] Open
Abstract
Background Deletions involving the long arm of chromosome 18 have been associated with a highly variable phenotypic spectrum that is related to the extent of the deleted region. Duplications in chromosomal region 4p16.3 have also been shown to cause 4p16.3 microduplication syndrome. Most reported patients of trisomy 4p16.3 have more duplications, including the Wolf-Hirschhorn critical region (WHSCR). Here, we present a patient with speech delay and mental retardation caused by a deletion of 18q (18q22.1-qter) and terminal microduplication of 4p (4p16.3-pter) distal to WHSCR. Case presentation The patient was a 23-month-old boy with moderate growth retardation, severe speech delay, mental retardation, and dysmorphic features. Single nucleotide polymorphism (SNP) array analysis confirmed an 11.2-Mb terminal deletion at 18q22.1 and revealed a 1.8-Mb terminal duplication of 4p16.3. Our patient showed clinical overlap with these two syndromes, although his overall features were milder than what had been previously described. Some dosage-sensitive genes on the 18q terminal deleted region and 4p16.3 duplicated region of the present case may have contributed to his phenotype. Conclusions This is the first report of a patient with combined terminal deletion of 18q22.1 and duplication of 4p16.3. In this report, we provide clinical and molecular evidence supporting that the microduplication in 4p16.3, distal to WHSCR, is pathogenic. The coexistence of two chromosome aberrations complicates the clinical picture and creates a chimeric phenotype. This report provides further information on the genotype-phenotype correlation of 18q terminal deletion and 4p microduplication.
Collapse
Affiliation(s)
- Chunjing Wang
- 1Department of Life Sciences, Bengbu Medical College, 2600 Donghai Avenue, Bengbu, Anhui 233030 People's Republic of China
| | - Huanhuan Ren
- 1Department of Life Sciences, Bengbu Medical College, 2600 Donghai Avenue, Bengbu, Anhui 233030 People's Republic of China
| | - Huaifu Dong
- 2Department of Paediatrics, The First Affiliated Hospital of Bengbu Medical College, 2600 Donghai Avenue, Bengbu, Anhui China
| | - Meng Liang
- 1Department of Life Sciences, Bengbu Medical College, 2600 Donghai Avenue, Bengbu, Anhui 233030 People's Republic of China
| | - Qi Wu
- 1Department of Life Sciences, Bengbu Medical College, 2600 Donghai Avenue, Bengbu, Anhui 233030 People's Republic of China
| | - Yaping Liao
- 1Department of Life Sciences, Bengbu Medical College, 2600 Donghai Avenue, Bengbu, Anhui 233030 People's Republic of China
| |
Collapse
|
29
|
Polarized Dock Activity Drives Shh-Mediated Axon Guidance. Dev Cell 2018; 46:410-425.e7. [PMID: 30078728 DOI: 10.1016/j.devcel.2018.07.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 05/18/2018] [Accepted: 07/06/2018] [Indexed: 11/23/2022]
Abstract
In the developing spinal cord, Sonic hedgehog (Shh) attracts commissural axons toward the floorplate. How Shh regulates the cytoskeletal remodeling that underlies growth cone turning is unknown. We found that Shh-mediated growth cone turning requires the activity of Docks, which are unconventional GEFs. Knockdown of Dock3 and 4, or their binding partner ELMO1 and 2, abolished commissural axon attraction by Shh in vitro. Dock3/4 and ELMO1/2 were also required for correct commissural axon guidance in vivo. Polarized Dock activity was sufficient to induce axon turning, indicating that Docks are instructive for axon guidance. Mechanistically, we show that Dock and ELMO interact with Boc, the Shh receptor, and that this interaction is reduced upon Shh stimulation. Furthermore, Shh stimulation translocates ELMO to the growth cone periphery and activates Rac1. This identifies Dock/ELMO as an effector complex of non-canonical Shh signaling and demonstrates the instructive role of GEFs in axon guidance.
Collapse
|
30
|
Le PT, Bishop KA, Maridas DE, Motyl KJ, Brooks DJ, Nagano K, Baron R, Bouxsein ML, Rosen CJ. Spontaneous mutation of Dock7 results in lower trabecular bone mass and impaired periosteal expansion in aged female Misty mice. Bone 2017; 105:103-114. [PMID: 28821457 PMCID: PMC5693233 DOI: 10.1016/j.bone.2017.08.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 07/08/2017] [Accepted: 08/14/2017] [Indexed: 01/17/2023]
Abstract
Misty mice (m/m) have a loss of function mutation in Dock7 gene, a guanine nucleotide exchange factor, resulting in low bone mineral density, uncoupled bone remodeling and reduced bone formation. Dock7 has been identified as a modulator of osteoblast number and in vitro osteogenic differentiation in calvarial osteoblast culture. In addition, m/m exhibit reduced preformed brown adipose tissue innervation and temperature as well as compensatory increase in beige adipocyte markers. While the low bone mineral density phenotype is in part due to higher sympathetic nervous system (SNS) drive in young mice, it is unclear what effect aging would have in mice homozygous for the mutation in the Dock7 gene. We hypothesized that age-related trabecular bone loss and periosteal envelope expansion would be altered in m/m. To test this hypothesis, we comprehensively characterized the skeletal phenotype of m/m at 16, 32, 52, and 78wks of age. When compared to age-matched wild-type control mice (+/+), m/m had lower areal bone mineral density (aBMD) and areal bone mineral content (aBMC). Similarly, both femoral and vertebral BV/TV, Tb.N, and Conn.D were decreased in m/m while there was also an increase in Tb.Sp. As low bone mineral density and decreased trabecular bone were already present at 16wks of age in m/m and persisted throughout life, changes in age-related trabecular bone loss were not observed highlighting the role of Dock7 in controlling trabecular bone acquisition or bone loss prior to 16wks of age. Cortical thickness was also lower in the m/m across all ages. Periosteal and endosteal circumferences were higher in m/m compared to +/+ at 16wks. However, endosteal and periosteal expansion were attenuated in m/m, resulting in m/m having lower periosteal and endosteal circumferences by 78wks of age compared to +/+, highlighting the critical role of Dock7 in appositional bone expansion. Histomorphometry revealed that osteoblasts were nearly undetectable in m/m and marrow adipocytes were elevated 3.5 fold over +/+ (p=0.014). Consistent with reduced bone formation, osteoblast gene expression of Alp, Col1a1, Runx-2, Sp7, and Bglap was significantly decreased in m/m whole bone. Furthermore, markers of osteoclasts were either unchanged or suppressed. Bone marrow stromal cell migration and motility were inhibited in culture and changes in senescence markers suggest that osteoblast function may also be inhibited with loss of Dock7 expression in m/m. Finally, increased Oil Red O staining in m/m ear mesenchymal stem cells during adipogenesis highlights a potential shift of cells from the osteogenic to adipogenic lineages. In summary, loss of Dock7 in the aging m/m resulted in an impairment of periosteal and endocortical envelope expansion, but did not alter age-related trabecular bone loss. These studies establish Dock7 as a critical regulator of both cortical and trabecular bone mass, and demonstrate for the first time a novel role of Dock7 in modulating compensatory changes in the periosteum with aging.
Collapse
Affiliation(s)
- Phuong T Le
- Maine Medical Center Research Institute, Maine Medical Center, Scarborough, ME 04074, United States
| | - Kathleen A Bishop
- Maine Medical Center Research Institute, Maine Medical Center, Scarborough, ME 04074, United States.
| | - David E Maridas
- Maine Medical Center Research Institute, Maine Medical Center, Scarborough, ME 04074, United States; University of Maine Graduate School of Biomedical Science and Engineering, Orono, ME 04469, United States
| | - Katherine J Motyl
- Maine Medical Center Research Institute, Maine Medical Center, Scarborough, ME 04074, United States
| | - Daniel J Brooks
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States
| | - Kenichi Nagano
- Harvard School of Dental Medicine, Boston, MA 02215, United States
| | - Roland Baron
- Harvard School of Dental Medicine, Boston, MA 02215, United States; Harvard School of Medicine, Boston, MA 02215, United States
| | - Mary L Bouxsein
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States
| | - Clifford J Rosen
- Maine Medical Center Research Institute, Maine Medical Center, Scarborough, ME 04074, United States; University of Maine Graduate School of Biomedical Science and Engineering, Orono, ME 04469, United States
| |
Collapse
|
31
|
Nakamuta S, Yang YT, Wang CL, Gallo NB, Yu JR, Tai Y, Van Aelst L. Dual role for DOCK7 in tangential migration of interneuron precursors in the postnatal forebrain. J Cell Biol 2017; 216:4313-4330. [PMID: 29089377 PMCID: PMC5716287 DOI: 10.1083/jcb.201704157] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 09/01/2017] [Accepted: 09/15/2017] [Indexed: 12/14/2022] Open
Abstract
Throughout life, stem cells in the ventricular-subventricular zone generate neuroblasts that migrate via the rostral migratory stream (RMS) to the olfactory bulb, where they differentiate into local interneurons. Although progress has been made toward identifying extracellular factors that guide the migration of these cells, little is known about the intracellular mechanisms that govern the dynamic reshaping of the neuroblasts' morphology required for their migration along the RMS. In this study, we identify DOCK7, a member of the DOCK180-family, as a molecule essential for tangential neuroblast migration in the postnatal mouse forebrain. DOCK7 regulates the migration of these cells by controlling both leading process (LP) extension and somal translocation via distinct pathways. It controls LP stability/growth via a Rac-dependent pathway, likely by modulating microtubule networks while also regulating F-actin remodeling at the cell rear to promote somal translocation via a previously unrecognized myosin phosphatase-RhoA-interacting protein-dependent pathway. The coordinated action of both pathways is required to ensure efficient neuroblast migration along the RMS.
Collapse
Affiliation(s)
| | - Yu-Ting Yang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY
| | - Chia-Lin Wang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY
| | - Nicholas B Gallo
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY.,Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY
| | - Jia-Ray Yu
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY
| | - Yilin Tai
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY
| | | |
Collapse
|
32
|
Merfeld E, Ben‐Avi L, Kennon M, Cerveny KL. Potential mechanisms of Zika-linked microcephaly. WILEY INTERDISCIPLINARY REVIEWS. DEVELOPMENTAL BIOLOGY 2017; 6:e273. [PMID: 28383800 PMCID: PMC5516183 DOI: 10.1002/wdev.273] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 02/20/2017] [Accepted: 02/23/2017] [Indexed: 01/01/2023]
Abstract
A recent outbreak of Zika virus (ZIKV) in Brazil is associated with microcephaly in infants born of infected mothers. As this pandemic spreads, rapid scientific investigation is shedding new light on how prenatal infection with ZIKV causes microcephaly. In this analysis we provide an overview of both microcephaly and ZIKV, explore the connection between prenatal ZIKV infection and microcephaly, and highlight recent insights into how prenatal ZIKV infection depletes the pool of neural progenitors in the developing brain. WIREs Dev Biol 2017, 6:e273. doi: 10.1002/wdev.273 For further resources related to this article, please visit the WIREs website.
Collapse
|
33
|
Sun D, Zhou X, Yu HL, He XX, Guo WX, Xiong WC, Zhu XJ. Regulation of neural stem cell proliferation and differentiation by Kinesin family member 2a. PLoS One 2017; 12:e0179047. [PMID: 28591194 PMCID: PMC5462413 DOI: 10.1371/journal.pone.0179047] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Accepted: 05/23/2017] [Indexed: 12/03/2022] Open
Abstract
In the developing neocortex, cells in the ventricular/subventricular zone are largely multipotent neural stem cells and neural progenitor cells. These cells undergo self-renewal at the early stage of embryonic development to amplify the progenitor pool and subsequently differentiate into neurons. It is thus of considerable interest to investigate mechanisms controlling the switch from neural stem cells or neural progenitor cells to neurons. Here, we present evidence that Kif2a, a member of the Kinesin-13 family, plays a role in regulating the proliferation and differentiation of neural stem cells or neural progenitor cells at embryonic day 13.5. Silencing Kif2a by use of in utero electroporation of Kif2a shRNA reduced neural stem cells proliferation or self-renewal but increased neuronal differentiation. We further found that knockdown of Kif2a decreased the protein level of β-catenin, which is a critical molecule for neocortical neurogenesis. Together, these results reveal an important function of Kif2a in embryonic neocortical neurogenesis.
Collapse
Affiliation(s)
- Dong Sun
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Cytology and Genetics, Northeast Normal University, Changchun, Jilin, China
- Department of Neuroscience & Regenerative Medicine and Department of Neurology, Augusta University, Augusta, Georgia, United States of America
| | - Xue Zhou
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Cytology and Genetics, Northeast Normal University, Changchun, Jilin, China
| | - Hua-Li Yu
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Cytology and Genetics, Northeast Normal University, Changchun, Jilin, China
- Department of Neuroscience & Regenerative Medicine and Department of Neurology, Augusta University, Augusta, Georgia, United States of America
| | - Xiao-Xiao He
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Cytology and Genetics, Northeast Normal University, Changchun, Jilin, China
| | - Wei-Xiang Guo
- State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Wen-Cheng Xiong
- Department of Neuroscience & Regenerative Medicine and Department of Neurology, Augusta University, Augusta, Georgia, United States of America
- * E-mail: (X-JZ); (W-CX)
| | - Xiao-Juan Zhu
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Cytology and Genetics, Northeast Normal University, Changchun, Jilin, China
- * E-mail: (X-JZ); (W-CX)
| |
Collapse
|
34
|
Genetic and Molecular Approaches to Study Neuronal Migration in the Developing Cerebral Cortex. Brain Sci 2017; 7:brainsci7050053. [PMID: 28475113 PMCID: PMC5447935 DOI: 10.3390/brainsci7050053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 04/21/2017] [Accepted: 05/02/2017] [Indexed: 11/17/2022] Open
Abstract
The migration of neuronal cells in the developing cerebral cortex is essential for proper development of the brain and brain networks. Disturbances in this process, due to genetic abnormalities or exogenous factors, leads to aberrant brain formation, brain network formation, and brain function. In the last decade, there has been extensive research in the field of neuronal migration. In this review, we describe different methods and approaches to assess and study neuronal migration in the developing cerebral cortex. First, we discuss several genetic methods, techniques and genetic models that have been used to study neuronal migration in the developing cortex. Second, we describe several molecular approaches to study aberrant neuronal migration in the cortex which can be used to elucidate the underlying mechanisms of neuronal migration. Finally, we describe model systems to investigate and assess the potential toxicity effect of prenatal exposure to environmental chemicals on proper brain formation and neuronal migration.
Collapse
|
35
|
van de Willige D, Hoogenraad CC, Akhmanova A. Microtubule plus-end tracking proteins in neuronal development. Cell Mol Life Sci 2016; 73:2053-77. [PMID: 26969328 PMCID: PMC4834103 DOI: 10.1007/s00018-016-2168-3] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Revised: 02/04/2016] [Accepted: 02/22/2016] [Indexed: 11/28/2022]
Abstract
Regulation of the microtubule cytoskeleton is of pivotal importance for neuronal development and function. One such regulatory mechanism centers on microtubule plus-end tracking proteins (+TIPs): structurally and functionally diverse regulatory factors, which can form complex macromolecular assemblies at the growing microtubule plus-ends. +TIPs modulate important properties of microtubules including their dynamics and their ability to control cell polarity, membrane transport and signaling. Several neurodevelopmental and neurodegenerative diseases are associated with mutations in +TIPs or with misregulation of these proteins. In this review, we focus on the role and regulation of +TIPs in neuronal development and associated disorders.
Collapse
Affiliation(s)
- Dieudonnée van de Willige
- Cell Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| | - Casper C Hoogenraad
- Cell Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands.
| | - Anna Akhmanova
- Cell Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands.
| |
Collapse
|
36
|
Strzyz PJ, Matejcic M, Norden C. Heterogeneity, Cell Biology and Tissue Mechanics of Pseudostratified Epithelia: Coordination of Cell Divisions and Growth in Tightly Packed Tissues. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 325:89-118. [PMID: 27241219 DOI: 10.1016/bs.ircmb.2016.02.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Pseudostratified epithelia (PSE) are tightly packed proliferative tissues that are important precursors of the development of diverse organs in a plethora of species, invertebrate and vertebrate. PSE consist of elongated epithelial cells that are attached to the apical and basal side of the tissue. The nuclei of these cells undergo interkinetic nuclear migration (IKNM) which leads to all mitotic events taking place at the apical surface of the epithelium. In this review, we discuss the intricacies of proliferation in PSE, considering cell biological, as well as the physical aspects. First, we summarize the principles governing the invariability of apical nuclear migration and apical cell division as well as the importance of apical mitoses for tissue proliferation. Then, we focus on the mechanical and structural features of these tissues. Here, we discuss how the overall architecture of pseudostratified tissues changes with increased cell packing. Lastly, we consider possible mechanical cues resulting from these changes and their potential influence on cell proliferation.
Collapse
Affiliation(s)
- P J Strzyz
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.
| | - M Matejcic
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - C Norden
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.
| |
Collapse
|
37
|
Dantas TJ, Carabalona A, Hu DJK, Vallee RB. Emerging roles for motor proteins in progenitor cell behavior and neuronal migration during brain development. Cytoskeleton (Hoboken) 2016; 73:566-576. [PMID: 26994401 DOI: 10.1002/cm.21293] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 03/11/2016] [Accepted: 03/13/2016] [Indexed: 12/21/2022]
Abstract
Over the past two decades, substantial progress has been made in visualizing and understanding neuronal cell migration and morphogenesis during brain development. Distinct mechanisms have evolved to support migration of the various cell types that compose the developing neocortex. A specific subset of molecular motors, so far consisting of cytoplasmic dynein 1, Kif1a and myosin II, are responsible for cytoskeletal and nuclear transport in these cells. This review focuses on the emerging roles for each of these motor proteins in the migratory mechanisms of neocortical cell types. We discuss how migration can be cell cycle regulated and how coordination of motor activity is required to ensure migratory direction. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Tiago J Dantas
- Department of Pathology and Cell Biology, Columbia University, New York, NY.
| | - Aurelie Carabalona
- Department of Pathology and Cell Biology, Columbia University, New York, NY
| | - Daniel Jun Kit Hu
- Department of Pathology and Cell Biology, Columbia University, New York, NY
| | - Richard B Vallee
- Department of Pathology and Cell Biology, Columbia University, New York, NY.
| |
Collapse
|
38
|
Sobczak M, Chumak V, Pomorski P, Wojtera E, Majewski Ł, Nowak J, Yamauchi J, Rędowicz MJ. Interaction of myosin VI and its binding partner DOCK7 plays an important role in NGF-stimulated protrusion formation in PC12 cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:1589-600. [PMID: 27018747 DOI: 10.1016/j.bbamcr.2016.03.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 03/05/2016] [Accepted: 03/22/2016] [Indexed: 10/22/2022]
Abstract
DOCK7 (dedicator of cytokinesis 7) is a guanidine nucleotide exchange factor (GEF) for Rac1 GTPase that is involved in neuronal polarity and axon generation as well in Schwann cell differentiation and myelination. Recently, we identified DOCK7 as the binding partner of unconventional myosin VI (MVI) in neuronal-lineage PC12 cells and postulated that this interaction could be important in vivo [Majewski et al. (2012) Biochem Cell Biol., 90:565-574]. Herein, we found that MVI-DOCK7 interaction takes also place in other cell lines and demonstrated that MVI cargo domain via its RRL motif binds to DOCK7 C-terminal M2 and DHR2 domains. In MVI knockdown cells, lower Rac1 activity and a decrease of DOCK7 phosphorylation on Tyr1118 were observed, indicating that MVI could contribute to DOCK7 activity. MVI and DOCK7 co-localization was maintained during NGF-stimulated PC12 cell differentiation and observed also in the outgrowths. Also, during differentiation an increase in phosphorylation of DOCK7 as well as of its downstream effector JNK kinase was detected. Interestingly, overexpression of GFP-tagged MVI cargo domain (GFP-GT) impaired protrusion formation indicating that full length protein is important for this process. Moreover, a transient increase in Rac activity observed at 5min of NGF-stimulated differentiation of PC12 cells (overexpressing either GFP or GFP-MVI) was not detected in cells overexpressing the cargo domain. These data indicate that MVI-DOCK7 interaction could have functional implications in the protrusion outgrowth, and full length MVI seems to be important for delivery and maintenance of DOCK7 along the protrusions, and exerting its GEF activity.
Collapse
Affiliation(s)
- Magdalena Sobczak
- Laboratory of Molecular Basis of Cell Motility, Department of Biochemistry, Nencki Institute of Experimental Biology, 3 Pasteur St., 02-093 Warsaw, Poland
| | - Vira Chumak
- Laboratory of Molecular Basis of Cell Motility, Department of Biochemistry, Nencki Institute of Experimental Biology, 3 Pasteur St., 02-093 Warsaw, Poland; Laboratory of Regulation of Cell Proliferation and Apoptosis, Institute of Cell Biology, National Academy of Sciences of Ukraine, 14-16 Drahomanov St., 79005 Lviv, Ukraine
| | - Paweł Pomorski
- Laboratory of Molecular Basis of Cell Motility, Department of Biochemistry, Nencki Institute of Experimental Biology, 3 Pasteur St., 02-093 Warsaw, Poland
| | - Emilia Wojtera
- Laboratory of Molecular Basis of Cell Motility, Department of Biochemistry, Nencki Institute of Experimental Biology, 3 Pasteur St., 02-093 Warsaw, Poland
| | - Łukasz Majewski
- Laboratory of Molecular Basis of Cell Motility, Department of Biochemistry, Nencki Institute of Experimental Biology, 3 Pasteur St., 02-093 Warsaw, Poland
| | - Jolanta Nowak
- Laboratory of Molecular Basis of Cell Motility, Department of Biochemistry, Nencki Institute of Experimental Biology, 3 Pasteur St., 02-093 Warsaw, Poland
| | - Junji Yamauchi
- Department of Pharmacology, National Research Institute for Child Health and Development, Setagaya, Tokyo, Japan; Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo, Tokyo, Japan
| | - Maria Jolanta Rędowicz
- Laboratory of Molecular Basis of Cell Motility, Department of Biochemistry, Nencki Institute of Experimental Biology, 3 Pasteur St., 02-093 Warsaw, Poland.
| |
Collapse
|
39
|
Zhang W, Kim PJ, Chen Z, Lokman H, Qiu L, Zhang K, Rozen SG, Tan EK, Je HS, Zeng L. MiRNA-128 regulates the proliferation and neurogenesis of neural precursors by targeting PCM1 in the developing cortex. eLife 2016; 5. [PMID: 26883496 PMCID: PMC4769165 DOI: 10.7554/elife.11324] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 01/22/2016] [Indexed: 12/18/2022] Open
Abstract
During the development, tight regulation of the expansion of neural progenitor cells (NPCs) and their differentiation into neurons is crucial for normal cortical formation and function. In this study, we demonstrate that microRNA (miR)-128 regulates the proliferation and differentiation of NPCs by repressing pericentriolar material 1 (PCM1). Specifically, overexpression of miR-128 reduced NPC proliferation but promoted NPC differentiation into neurons both in vivo and in vitro. In contrast, the reduction of endogenous miR-128 elicited the opposite effects. Overexpression of miR-128 suppressed the translation of PCM1, and knockdown of endogenous PCM1 phenocopied the observed effects of miR-128 overexpression. Furthermore, concomitant overexpression of PCM1 and miR-128 in NPCs rescued the phenotype associated with miR-128 overexpression, enhancing neurogenesis but inhibiting proliferation, both in vitro and in utero. Taken together, these results demonstrate a novel mechanism by which miR-128 regulates the proliferation and differentiation of NPCs in the developing neocortex. DOI:http://dx.doi.org/10.7554/eLife.11324.001 The neurons that transmit information around the brain develop from cells called neural progenitor cells. These cells can either divide to form more progenitor cells or to become specific types of neurons. If these carefully regulated processes go wrong – for example, if progenitors fail to stop dividing in order to mature – a range of neurodevelopmental conditions may develop, including autism spectrum disorders. Small RNA molecules called microRNAs control gene activity and protein formation by targeting certain other RNA molecules for destruction. One such microRNA, called miR-128, helps newly formed neurons to move to the correct region of the cortex – the outer layer of the brain, which is essential for many cognitive processes including thought and language. However, it was not clear whether miR-128 plays any other roles in the development of neurons. Zhang, Kim et al. have now analysed the role of miR-128 in the developing cortex of mice. The findings suggest that miR-128 prevents cortical neural progenitor cells from dividing and supports their development into more specialized cells. Causing miR-128 to be over-produced in the progenitor cells caused the cells to divide less often and encouraged them to mature into neurons. Conversely, removing miR-128 from the progenitor cells caused them to divide more and resulted in fewer neurons forming. Further investigation revealed that miR-128 works by causing less of a protein called PCM1 to be produced. Without this protein, cells cannot divide properly. Future studies could now investigate in more detail how miR-128 and PCM1 affect how the neurons in the cortex develop and work. DOI:http://dx.doi.org/10.7554/eLife.11324.002
Collapse
Affiliation(s)
- Wei Zhang
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore, Singapore
| | - Paul Jong Kim
- Molecular Neurophysiology Laboratory, Signature Program in Neuroscience and Behavioral Disorders, Duke-NUS Graduate Medical School, Singapore, Singapore
| | - Zhongcan Chen
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore, Singapore
| | - Hidayat Lokman
- Molecular Neurophysiology Laboratory, Signature Program in Neuroscience and Behavioral Disorders, Duke-NUS Graduate Medical School, Singapore, Singapore
| | - Lifeng Qiu
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore, Singapore
| | - Ke Zhang
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore, Singapore
| | - Steven George Rozen
- Center for Computational Biology, Duke-NUS Graduate Medical School, Singapore, Singapore
| | - Eng King Tan
- Department of Neurology, National Neuroscience Institute, Singapore, Singapore.,Neuroscience and Behavioral Disorders program, Duke-NUS Graduate Medical School, Singapore, Singapore
| | - Hyunsoo Shawn Je
- Molecular Neurophysiology Laboratory, Signature Program in Neuroscience and Behavioral Disorders, Duke-NUS Graduate Medical School, Singapore, Singapore.,Neuroscience and Behavioral Disorders program, Duke-NUS Graduate Medical School, Singapore, Singapore.,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Li Zeng
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore, Singapore.,Neuroscience and Behavioral Disorders program, Duke-NUS Graduate Medical School, Singapore, Singapore
| |
Collapse
|
40
|
Guo T, Yin RX, Lin WX, Wang W, Huang F, Pan SL. Association of the variants and haplotypes in the DOCK7, PCSK9 and GALNT2 genes and the risk of hyperlipidaemia. J Cell Mol Med 2016; 20:243-65. [PMID: 26493351 PMCID: PMC4727555 DOI: 10.1111/jcmm.12713] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Accepted: 09/07/2015] [Indexed: 01/09/2023] Open
Abstract
Little is known about the association between the single nucleotide polymorphisms (SNPs) and haplotypes of the dedicator of cytokinesis 7 (DOCK7), pro-protein convertase subtilisin/kexin type 9 (PCSK9) and polypeptide N-acetylgalactosaminyltransferase 2 (GALNT2) and serum lipid traits in the Chinese populations. This study was to determine the association between nine SNPs in the three genes and their haplotypes and hypercholesterolaemia (HCH)/hypertriglyceridaemia (HTG), and to identify the possible gene-gene interactions among these SNPs. Genotyping was performed in 733 HCH and 540 HTG participants. The haplotype of C-C-G-C-T-G-C-C-G [in the order of DOCK7 rs1168013 (G>C), rs10889332 (C>T); PCSK9 rs615563 (G>A), rs7552841 (C>T), rs11206517 (T>G); and GALNT2 rs1997947 (G>A), rs2760537 (C>T), rs4846913 (C>A) and rs11122316 (G>A) SNPs] was associated with increased risk of HCH and HTG. The haplotypes of C-C-G-C-T-G-C-C-A and G-C-G-T-T-G-T-C-G were associated with a reduced risk of HCH and HTG. The haplotypes of G-C-G-C-T-G-C-C-A and G-C-G-C-T-G-T-C-G were associated with increased risk of HCH. The haplotypes of C-T-G-C-T-G-C-C-G, G-C-A-C-T-G-C-C-G and G-C-G-C-T-G-C-C-A were associated with an increased risk of HTG. The haplotypes of G-C-G-C-T-G-T-C-A and G-C-G-T-T-G-T-C-G were associated with a reduced risk of HTG. In addition, possible inter-locus interactions among the DOCK7, PCSK9 and GALNT2 SNPs were also noted. However, further functional studies of these genes are still required to clarify which SNPs are functional and how these genes actually affect the serum lipid levels.
Collapse
Affiliation(s)
- Tao Guo
- Department of Cardiology, Institute of Cardiovascular Diseases, The First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China
| | - Rui-Xing Yin
- Department of Cardiology, Institute of Cardiovascular Diseases, The First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China
| | - Wei-Xiong Lin
- Department of Molecular Genetics, Medical Scientific Research Center, Guangxi Medical University, Nanning, Guangxi, China
| | - Wei Wang
- Department of Cardiology, Institute of Cardiovascular Diseases, The First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China
| | - Feng Huang
- Department of Cardiology, Institute of Cardiovascular Diseases, The First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China
| | - Shang-Ling Pan
- Department of Pathophysiology, School of Premedical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
41
|
Drago A, Crisafulli C, Sidoti A, Calabrò M, Serretti A. The microtubule-associated molecular pathways may be genetically disrupted in patients with Bipolar Disorder. Insights from the molecular cascades. J Affect Disord 2016; 190:429-438. [PMID: 26551401 DOI: 10.1016/j.jad.2015.10.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 09/24/2015] [Accepted: 10/10/2015] [Indexed: 01/15/2023]
Abstract
Bipolar Disorder is a severe disease characterized by pathological mood swings from major depressive episodes to manic ones and vice versa. The biological underpinnings of Bipolar Disorder have yet to be defined. As a consequence, pharmacological treatments are suboptimal. In the present paper we test the hypothesis that the molecular pathways involved with the direct targets of lithium, hold significantly more genetic variations associated with BD. A molecular pathway approach finds its rationale in the polygenic nature of the disease. The pathways were tested in a sample of ∼ 7,000 patients and controls. Data are available from the public NIMH database. The definition of the pathways was conducted according to the National Cancer Institute (http://pid.nci.nih.gov/). As a result, 3 out of the 18 tested pathways related to lithium action resisted the permutation analysis and were found to be associated with BD. These pathways were related to Reelin, Integrins and Aurora. A pool of genes selected from the ones linked with the above pathways was further investigated in order to identify the fine molecular mechanics shared by our significant pathways and also their link with lithium mechanism of action. The data obtained point out to a possible involvement of microtubule-related mechanics.
Collapse
Affiliation(s)
- Antonio Drago
- Department of Biomedical and Neuromotor Sciences - DIBINEM - University of Bologna, Bologna, Italy
| | - Concetta Crisafulli
- Department of Biomedical Science and Morphological and Functional Images, University of Messina, Via Consolare Valeria, 98125 Messina, Italy.
| | - Antonina Sidoti
- Department of Biomedical Science and Morphological and Functional Images, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | - Marco Calabrò
- Department of Biomedical Science and Morphological and Functional Images, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | - Alessandro Serretti
- Department of Biomedical and Neuromotor Sciences - DIBINEM - University of Bologna, Bologna, Italy
| |
Collapse
|
42
|
Guo T, Yin RX, Huang F, Yao LM, Lin WX, Pan SL. Association between the DOCK7, PCSK9 and GALNT2 Gene Polymorphisms and Serum Lipid levels. Sci Rep 2016; 6:19079. [PMID: 26744084 PMCID: PMC4705473 DOI: 10.1038/srep19079] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 12/04/2015] [Indexed: 01/02/2023] Open
Abstract
This study was to determine the association between several single nucleotide polymorphisms (SNPs) in the dedicator of cytokinesis 7 (DOCK7), proprotein convertase subtilisin/kexin type 9 (PCSK9) and polypeptide N-acetylgalactosaminyltransferase 2 (GALNT2) and serum lipid levels. Genotyping of 9 SNPs was performed in 881 Jing subjects and 988 Han participants. Allele and genotype frequencies of the detected SNPs were different between the two populations. Several SNPs were associated with triglyceride (TG, rs10889332, rs615563, rs7552841, rs1997947, rs2760537, rs4846913 and rs11122316), high-density lipoprotein (HDL) cholesterol (rs1997947), low-density lipoprotein (LDL) cholesterol (rs1168013 and rs7552841), apolipoprotein (Apo) A1 (rs1997947), ApoB (rs10889332 and rs7552841), and ApoA1/ApoB ratio (rs7552841) in Jing minority; and with TG (rs10889332, rs615563, rs7552841, rs11206517, rs1997947, rs4846913 and rs11122316), HDL cholesterol (rs11206517 and rs4846913), LDL cholesterol (rs1168013), ApoA1 (rs11206517 and rs4846913), ApoB (rs7552841), and ApoA1/ApoB ratio (rs4846913) in Han nationality. Strong linkage disequilibria were noted among the SNPs. The commonest haplotype was G-C-G-C-T-G-C-C-G (>10%). The frequencies of C-C-G-C-T-G-T-C-G, G-C-A-C-T-G-C-C-G, G-C-G-C-T-A-C-C-A, G-C-G-C-T-G-C-C-A, G-C-G-C-T-G-T-C-A haplotypes were different between the two populations. Haplotypes could explain much more serum lipid variation than any single SNP alone especially for TG. Differences in lipid profiles between the two populations might partially attribute to these SNPs and their haplotypes.
Collapse
Affiliation(s)
- Tao Guo
- Department of Cardiology, Institute of Cardiovascular Diseases, the First Affiliated Hospital, Nanning 530021, Guangxi, China
| | - Rui-Xing Yin
- Department of Cardiology, Institute of Cardiovascular Diseases, the First Affiliated Hospital, Nanning 530021, Guangxi, China
| | - Feng Huang
- Department of Cardiology, Institute of Cardiovascular Diseases, the First Affiliated Hospital, Nanning 530021, Guangxi, China
| | - Li-Mei Yao
- Department of Cardiology, Institute of Cardiovascular Diseases, the First Affiliated Hospital, Nanning 530021, Guangxi, China
| | - Wei-Xiong Lin
- Department of Molecular Genetics, Medical Scientific Research Center, Nanning 530021, Guangxi, China
| | - Shang-Ling Pan
- Department of Pathophysiology, School of Premedical Sciences, Guangxi Medical University, Nanning 530021, Guangxi, China
| |
Collapse
|
43
|
Ohtaka-Maruyama C, Okado H. Molecular Pathways Underlying Projection Neuron Production and Migration during Cerebral Cortical Development. Front Neurosci 2015; 9:447. [PMID: 26733777 PMCID: PMC4682034 DOI: 10.3389/fnins.2015.00447] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 11/09/2015] [Indexed: 12/25/2022] Open
Abstract
Glutamatergic neurons of the mammalian cerebral cortex originate from radial glia (RG) progenitors in the ventricular zone (VZ). During corticogenesis, neuroblasts migrate toward the pial surface using two different migration modes. One is multipolar (MP) migration with random directional movement, and the other is locomotion, which is a unidirectional movement guided by the RG fiber. After reaching their final destination, the neurons finalize their migration by terminal translocation, which is followed by maturation via dendrite extension to initiate synaptogenesis and thereby complete neural circuit formation. This switching of migration modes during cortical development is unique in mammals, which suggests that the RG-guided locomotion mode may contribute to the evolution of the mammalian neocortical 6-layer structure. Many factors have been reported to be involved in the regulation of this radial neuronal migration process. In general, the radial migration can be largely divided into four steps; (1) maintenance and departure from the VZ of neural progenitor cells, (2) MP migration and transition to bipolar cells, (3) RG-guided locomotion, and (4) terminal translocation and dendrite maturation. Among these, many different gene mutations or knockdown effects have resulted in failure of the MP to bipolar transition (step 2), suggesting that it is a critical step, particularly in radial migration. Moreover, this transition occurs at the subplate layer. In this review, we summarize recent advances in our understanding of the molecular mechanisms underlying each of these steps. Finally, we discuss the evolutionary aspects of neuronal migration in corticogenesis.
Collapse
Affiliation(s)
- Chiaki Ohtaka-Maruyama
- Neural Network Project, Department of Brain Development and Neural Regeneration, Tokyo Metropolitan Institute of Medical Science Tokyo, Japan
| | - Haruo Okado
- Neural Development Project, Department of Brain Development and Neural Regeneration, Tokyo Metropolitan Institute of Medical Science Tokyo, Japan
| |
Collapse
|
44
|
Nulty J, Alsaffar M, Barry D. Radial glial cells organize the central nervous system via microtubule dependant processes. Brain Res 2015; 1625:171-9. [DOI: 10.1016/j.brainres.2015.08.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 08/21/2015] [Accepted: 08/22/2015] [Indexed: 11/16/2022]
|
45
|
Razafsky D, Hodzic D. Nuclear envelope: positioning nuclei and organizing synapses. Curr Opin Cell Biol 2015; 34:84-93. [PMID: 26079712 DOI: 10.1016/j.ceb.2015.06.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 05/28/2015] [Accepted: 06/01/2015] [Indexed: 10/23/2022]
Abstract
The nuclear envelope plays an essential role in nuclear positioning within cells and tissues. This review highlights advances in understanding the mechanisms of nuclear positioning during skeletal muscle and central nervous system development. New findings, particularly about A-type lamins and Nesprin1, may link nuclear envelope integrity to synaptic integrity. Thus synaptic defects, rather than nuclear mispositioning, may underlie human pathologies associated with mutations of nuclear envelope proteins.
Collapse
Affiliation(s)
- David Razafsky
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, 660 S. Euclid Avenue, St Louis, MO 63110, USA
| | - Didier Hodzic
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, 660 S. Euclid Avenue, St Louis, MO 63110, USA.
| |
Collapse
|
46
|
Guedj F, Pennings JLA, Ferres MA, Graham LC, Wick HC, Miczek KA, Slonim DK, Bianchi DW. The fetal brain transcriptome and neonatal behavioral phenotype in the Ts1Cje mouse model of Down syndrome. Am J Med Genet A 2015; 167A:1993-2008. [PMID: 25975229 DOI: 10.1002/ajmg.a.37156] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 04/27/2015] [Indexed: 11/07/2022]
Abstract
Human fetuses with Down syndrome demonstrate abnormal brain growth and reduced neurogenesis. Despite the prenatal onset of the phenotype, most therapeutic trials have been conducted in adults. Here, we present evidence for fetal brain molecular and neonatal behavioral alterations in the Ts1Cje mouse model of Down syndrome. Embryonic day 15.5 brain hemisphere RNA from Ts1Cje embryos (n = 5) and wild type littermates (n = 5) was processed and hybridized to mouse gene 1.0 ST arrays. Bioinformatic analyses were implemented to identify differential gene and pathway regulation during Ts1Cje fetal brain development. In separate experiments, the Fox scale, ultrasonic vocalization and homing tests were used to investigate behavioral deficits in Ts1Cje pups (n = 29) versus WT littermates (n = 64) at postnatal days 3-21. Ts1Cje fetal brains displayed more differentially regulated genes (n = 71) than adult (n = 31) when compared to their age-matched euploid brains. Ts1Cje embryonic brains showed up-regulation of cell cycle markers and down-regulation of the solute-carrier amino acid transporters. Several cellular processes were dysregulated at both stages, including apoptosis, inflammation, Jak/Stat signaling, G-protein signaling, and oxidoreductase activity. In addition, early behavioral deficits in surface righting, cliff aversion, negative geotaxis, forelimb grasp, ultrasonic vocalization, and the homing tests were observed. The Ts1Cje mouse model exhibits abnormal gene expression during fetal brain development, and significant neonatal behavioral deficits in the pre-weaning period. In combination with human studies, this suggests that the Down syndrome phenotype manifests prenatally and provides a rationale for prenatal therapy to improve perinatal brain development and postnatal neurocognition.
Collapse
Affiliation(s)
- Faycal Guedj
- Mother Infant Research Institute, Tufts Medical Center and the Floating Hospital for Children, Boston, Massachusetts
| | - Jeroen L A Pennings
- Center for Health Protection (GZB), National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Millie A Ferres
- Mother Infant Research Institute, Tufts Medical Center and the Floating Hospital for Children, Boston, Massachusetts
| | - Leah C Graham
- Mother Infant Research Institute, Tufts Medical Center and the Floating Hospital for Children, Boston, Massachusetts
| | - Heather C Wick
- Department of Computer Science, Tufts University, Medford, Massachusetts
| | - Klaus A Miczek
- Department of Psychology, Tufts University, Medford, Massachusetts
| | - Donna K Slonim
- Department of Computer Science, Tufts University, Medford, Massachusetts
| | - Diana W Bianchi
- Mother Infant Research Institute, Tufts Medical Center and the Floating Hospital for Children, Boston, Massachusetts
| |
Collapse
|
47
|
Protein expression profiles characterize distinct features of mouse cerebral cortices at different developmental stages. PLoS One 2015; 10:e0125608. [PMID: 25915664 PMCID: PMC4411115 DOI: 10.1371/journal.pone.0125608] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 03/25/2015] [Indexed: 01/18/2023] Open
Abstract
The proper development of the mammalian cerebral cortex requires precise protein synthesis and accurate regulation of protein expression levels. To reveal signatures of protein expression in developing mouse cortices, we here generate proteomic profiles of cortices at embryonic and postnatal stages using tandem mass spectrometry (MS/MS). We found that protein expression profiles are mostly consistent with biological features of the developing cortex. Gene Ontology (GO) and KEGG pathway analyses demonstrate conserved molecules that maintain cortical development such as proteins involved in metabolism. GO and KEGG pathway analyses further identify differentially expressed proteins that function at specific stages, for example proteins regulating the cell cycle in the embryonic cortex, and proteins controlling axon guidance in the postnatal cortex, suggesting that distinct protein expression profiles determine biological events in the developing cortex. Furthermore, the STRING network analysis has revealed that many proteins control a single biological event, such as the cell cycle regulation, through cohesive interactions, indicating a complex network regulation in the cortex. Our study has identified protein networks that control the cortical development and has provided a protein reference for further investigation of protein interactions in the cortex.
Collapse
|
48
|
Regulating Rac in the nervous system: molecular function and disease implication of Rac GEFs and GAPs. BIOMED RESEARCH INTERNATIONAL 2015; 2015:632450. [PMID: 25879033 PMCID: PMC4388020 DOI: 10.1155/2015/632450] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 03/06/2015] [Indexed: 12/11/2022]
Abstract
Rho family GTPases, including RhoA, Rac1, and Cdc42 as the most studied members, are master regulators of actin cytoskeletal organization. Rho GTPases control various aspects of the nervous system and are associated with a number of neuropsychiatric and neurodegenerative diseases. The activity of Rho GTPases is controlled by two families of regulators, guanine nucleotide exchange factors (GEFs) as the activators and GTPase-activating proteins (GAPs) as the inhibitors. Through coordinated regulation by GEFs and GAPs, Rho GTPases act as converging signaling molecules that convey different upstream signals in the nervous system. So far, more than 70 members of either GEFs or GAPs of Rho GTPases have been identified in mammals, but only a small subset of them have well-known functions. Thus, characterization of important GEFs and GAPs in the nervous system is crucial for the understanding of spatiotemporal dynamics of Rho GTPase activity in different neuronal functions. In this review, we summarize the current understanding of GEFs and GAPs for Rac1, with emphasis on the molecular function and disease implication of these regulators in the nervous system.
Collapse
|
49
|
Palumbo O, Palumbo P, Ferri E, Riviello FN, Cloroformio L, Carella M, Di Giacomo MC. Report of a patient and further clinical and molecular characterization of interstitial 4p16.3 microduplication. Mol Cytogenet 2015; 8:15. [PMID: 25774220 PMCID: PMC4359789 DOI: 10.1186/s13039-015-0119-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 02/16/2015] [Indexed: 11/15/2022] Open
Abstract
Background Pure interstitial duplications of chromosome band 4p16.3 represent an infrequent chromosomal finding with, to the best of our knowledge, only two patients to date reported. Case presentation We report on a 13-year-old boy showing a set of dysmorphic facial features, attention deficit hyperactivity disorders, learning difficulties, speech and cognitive delays, overgrowth and musculoskeletal anomalies in whom an interstitial duplication of about 400 kb in 4p16.3 was detected by SNP-array analysis. The duplication includes the complete coding sequence of FAM53A, SLBP, TMEM129 and TACC3 genes and the first exon of the FGFR3 gene. Phenotypic comparison with previously described patients harboring a microduplication of similar size and position contributes to better define the clinical correlation of 4p16.3 microduplications, suggesting the existence of a novel distinct and phenotypically recognizable syndrome. In addition, being the duplication identified in our case the smallest so far reported, it allowed us to refine the smallest region of overlap among patients to 222 kb, enabling a more accurate genotype-phenotype correlation for 4p16.3 microduplications. Conclusions Our case report provide clinical and molecular evidences supporting the existence of a novel 4p16.3 microduplication syndrome. The genes FAM53A, TACC3 and FGFR3 seems to play a key role in the etiology of the clinical phenotype. Interestingly, our patient is the oldest described so far and for this reason useful to delineate the long-term prognosis of these patients.
Collapse
Affiliation(s)
- Orazio Palumbo
- Laboratorio di Genetica Medica, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, FG Italy
| | - Pietro Palumbo
- Laboratorio di Genetica Medica, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, FG Italy
| | - Emanuela Ferri
- U.O.C Anatomia Patologica, AOR Ospedale "San Carlo", Potenza, Italy
| | | | - Lea Cloroformio
- U.O.C Anatomia Patologica, AOR Ospedale "San Carlo", Potenza, Italy
| | - Massimo Carella
- Laboratorio di Genetica Medica, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, FG Italy
| | | |
Collapse
|
50
|
Bizzotto S, Francis F. Morphological and functional aspects of progenitors perturbed in cortical malformations. Front Cell Neurosci 2015; 9:30. [PMID: 25729350 PMCID: PMC4325918 DOI: 10.3389/fncel.2015.00030] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Accepted: 01/18/2015] [Indexed: 11/13/2022] Open
Abstract
In this review, we discuss molecular and cellular mechanisms important for the function of neuronal progenitors during development, revealed by their perturbation in different cortical malformations. We focus on a class of neuronal progenitors, radial glial cells (RGCs), which are renowned for their unique morphological and behavioral characteristics, constituting a key element during the development of the mammalian cerebral cortex. We describe how the particular morphology of these cells is related to their roles in the orchestration of cortical development and their influence on other progenitor types and post-mitotic neurons. Important for disease mechanisms, we overview what is currently known about RGC cellular components, cytoskeletal mechanisms, signaling pathways and cell cycle characteristics, focusing on how defects lead to abnormal development and cortical malformation phenotypes. The multiple recent entry points from human genetics and animal models are contributing to our understanding of this important cell type. Combining data from phenotypes in the mouse reveals molecules which potentially act in common pathways. Going beyond this, we discuss future directions that may provide new data in this expanding area.
Collapse
Affiliation(s)
- Sara Bizzotto
- INSERM UMRS 839 Paris, France ; Sorbonne Universités, Université Pierre et Marie Curie Paris, France ; Institut du Fer à Moulin Paris, France
| | - Fiona Francis
- INSERM UMRS 839 Paris, France ; Sorbonne Universités, Université Pierre et Marie Curie Paris, France ; Institut du Fer à Moulin Paris, France
| |
Collapse
|