1
|
Jin L, Li Y, Luo S, Peng Q, Zhai QX, Zhai JX, Gao LD, Guo JJ, Song W, Yi YH, He N, Chen YJ. Reprint of: Recessive APC2 missense variants associated with epilepsies without neurodevelopmental disorders. Seizure 2024; 116:87-92. [PMID: 38523034 DOI: 10.1016/j.seizure.2024.03.006] [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: 05/22/2023] [Revised: 08/10/2023] [Accepted: 08/17/2023] [Indexed: 03/26/2024] Open
Abstract
OBJECTIVES The APC2 gene, encoding adenomatous polyposis coli protein-2, is involved in cytoskeletal regulation in neurons responding to endogenous extracellular signals and plays an important role in brain development. Previously, the APC2 variants have been reported to be associated with cortical dysplasia and intellectual disability. This study aims to explore the association between APC2 variants and epilepsy. METHODS Whole-exome sequencing (WES) was performed in cases (trios) with epilepsies of unknown causes. The damaging effects of variants were predicted by protein modeling and in silico tools. Previously reported APC2 variants were reviewed to analyze the genotype-phenotype correlations. RESULTS Four pairs of compound heterozygous missense variants were identified in four unrelated patients with epilepsy without brain malformation/intellectual disability. All variants presented no or low allele frequencies in the controls. The missense variants were predicted to be damaging by silico tools, and affect hydrogen bonding with surrounding amino acids or decreased protein stability. Patients with variants that resulted in significant changes in protein stability exhibited more severe and intractable epilepsy, whereas patients with variants that had minor effect on protein stability exhibited relatively mild phenotypes. The previously reported APC2 variants in patients with complex cortical dysplasia with other brain malformations-10 (CDCBM10; MIM: 618677) were all truncating variants; in contrast, the variants identified in epilepsy in this study were all missense variants, suggesting a potential genotype-phenotype correlation. SIGNIFICANCE This study suggests that APC2 is potentially associated with epilepsy without brain malformation/intellectual disability. The genotype-phenotype correlation helps to understand the underlying mechanisms of phenotypic heterogeneity.
Collapse
Affiliation(s)
- Liang Jin
- Department of Neurology, the Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Yun Li
- Department of Brain Function and Neuroelectrophysiology, the Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Sheng Luo
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Qian Peng
- Department of Pediatrics, Dongguan Maternal and Child Health Hospital, Southern Medical University Affiliated, Dongguang, China
| | - Qiong-Xiang Zhai
- Department of Pediatrics, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jin-Xia Zhai
- Department of Neurology, the Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Liang-Di Gao
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jia-Jun Guo
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Wang Song
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yong-Hong Yi
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Na He
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.
| | - Yong-Jun Chen
- Department of Neurology, the Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, China.
| |
Collapse
|
2
|
Santhanam A, Shihabeddin E, Wei H, Wu J, O'Brien J. Molecular basis of retinal remodeling in a zebrafish model of retinitis pigmentosa. Cell Mol Life Sci 2023; 80:362. [PMID: 37979052 PMCID: PMC10657301 DOI: 10.1007/s00018-023-05021-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 10/10/2023] [Accepted: 10/27/2023] [Indexed: 11/19/2023]
Abstract
A hallmark of inherited retinal degenerative diseases such as retinitis pigmentosa (RP) is progressive structural and functional remodeling of the remaining retinal cells as photoreceptors degenerate. Extensive remodeling of the retina stands as a barrier for the successful implementation of strategies to restore vision. To understand the molecular basis of remodeling, we performed analyses of single-cell transcriptome data from adult zebrafish retina of wild type AB strain (WT) and a P23H mutant rhodopsin transgenic model of RP with continuous degeneration and regeneration. Retinas from both female and male fish were pooled to generate each library, combining data from both sexes. We provide a benchmark atlas of retinal cell type transcriptomes in zebrafish and insight into how each retinal cell type is affected in the P23H model. Oxidative stress is found throughout the retina, with increases in reliance on oxidative metabolism and glycolysis in the affected rods as well as cones, bipolar cells, and retinal ganglion cells. There is also transcriptional evidence for widespread synaptic remodeling and enhancement of glutamatergic transmission in the inner retina. Notably, changes in circadian rhythm regulation are detected in cones, bipolar cells, and retinal pigmented epithelium. We also identify the transcriptomic signatures of retinal progenitor cells and newly formed rods essential for the regenerative process. This comprehensive transcriptomic analysis provides a molecular road map to understand how the retina remodels in the context of chronic retinal degeneration with ongoing regeneration.
Collapse
Affiliation(s)
- Abirami Santhanam
- Department of Ophthalmology & Visual Science, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
- University of Houston College of Optometry, Houston, TX, 77204, USA.
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, 77030, USA.
| | - Eyad Shihabeddin
- Department of Ophthalmology & Visual Science, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
- MD Anderson UT Health Graduate School of Biomedical Sciences, Houston, TX, 77030, USA
| | - Haichao Wei
- Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Jiaqian Wu
- MD Anderson UT Health Graduate School of Biomedical Sciences, Houston, TX, 77030, USA
- Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - John O'Brien
- Department of Ophthalmology & Visual Science, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
- University of Houston College of Optometry, Houston, TX, 77204, USA.
- MD Anderson UT Health Graduate School of Biomedical Sciences, Houston, TX, 77030, USA.
| |
Collapse
|
3
|
Jin L, Li Y, Luo S, Peng Q, Zhai QX, Zhai JX, Gao LD, Guo JJ, Song W, Yi YH, He N, Chen YJ. Recessive APC2 missense variants associated with epilepsies without neurodevelopmental disorders. Seizure 2023; 111:172-177. [PMID: 37657306 DOI: 10.1016/j.seizure.2023.08.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/10/2023] [Accepted: 08/17/2023] [Indexed: 09/03/2023] Open
Abstract
OBJECTIVES The APC2 gene, encoding adenomatous polyposis coli protein-2, is involved in cytoskeletal regulation in neurons responding to endogenous extracellular signals and plays an important role in brain development. Previously, the APC2 variants have been reported to be associated with cortical dysplasia and intellectual disability. This study aims to explore the association between APC2 variants and epilepsy. METHODS Whole-exome sequencing (WES) was performed in cases (trios) with epilepsies of unknown causes. The damaging effects of variants were predicted by protein modeling and in silico tools. Previously reported APC2 variants were reviewed to analyze the genotype-phenotype correlations. RESULTS Four pairs of compound heterozygous missense variants were identified in four unrelated patients with epilepsy without brain malformation/intellectual disability. All variants presented no or low allele frequencies in the controls. The missense variants were predicted to be damaging by silico tools, and affect hydrogen bonding with surrounding amino acids or decreased protein stability. Patients with variants that resulted in significant changes in protein stability exhibited more severe and intractable epilepsy, whereas patients with variants that had minor effect on protein stability exhibited relatively mild phenotypes. The previously reported APC2 variants in patients with complex cortical dysplasia with other brain malformations-10 (CDCBM10; MIM: 618677) were all truncating variants; in contrast, the variants identified in epilepsy in this study were all missense variants, suggesting a potential genotype-phenotype correlation. SIGNIFICANCE This study suggests that APC2 is potentially associated with epilepsy without brain malformation/intellectual disability. The genotype-phenotype correlation helps to understand the underlying mechanisms of phenotypic heterogeneity.
Collapse
Affiliation(s)
- Liang Jin
- Department of Neurology, the Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Yun Li
- Department of Brain Function and Neuroelectrophysiology, the Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Sheng Luo
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Qian Peng
- Department of Pediatrics, Dongguan Maternal and Child Health Hospital, Southern Medical University Affiliated, Dongguang, China
| | - Qiong-Xiang Zhai
- Department of Pediatrics, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jin-Xia Zhai
- Department of Neurology, the Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Liang-Di Gao
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jia-Jun Guo
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Wang Song
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yong-Hong Yi
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Na He
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.
| | - Yong-Jun Chen
- Department of Neurology, the Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, China.
| |
Collapse
|
4
|
Fries M, Brown TW, Jolicoeur C, Boulan B, Boudreau-Pinsonneault C, Javed A, Abram P, Cayouette M. Pou3f1 orchestrates a gene regulatory network controlling contralateral retinogeniculate projections. Cell Rep 2023; 42:112985. [PMID: 37590135 DOI: 10.1016/j.celrep.2023.112985] [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: 07/07/2022] [Revised: 05/26/2023] [Accepted: 08/01/2023] [Indexed: 08/19/2023] Open
Abstract
The balance of contralateral and ipsilateral retinogeniculate projections is critical for binocular vision, but the transcriptional programs regulating this process remain ill defined. Here we show that the Pou class homeobox protein POU3F1 is expressed in nascent mouse contralateral retinal ganglion cells (cRGCs) but not ipsilateral RGCs (iRGCs). Upon Pou3f1 inactivation, the proportion of cRGCs is reduced in favor of iRGCs, leading to abnormal projection ratios at the optic chiasm. Conversely, misexpression of Pou3f1 in progenitors increases the production of cRGCs. Using CUT&RUN and RNA sequencing in gain- and loss-of-function assays, we demonstrate that POU3F1 regulates expression of several key members of the cRGC gene regulatory network. Finally, we report that POU3F1 is sufficient to induce RGC-like cell production, even in late-stage retinal progenitors of Atoh7 knockout mice. This work uncovers POU3F1 as a regulator of the cRGC transcriptional program, opening possibilities for optic nerve regenerative therapies.
Collapse
Affiliation(s)
- Michel Fries
- Cellular Neurobiology Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, QC H2W 1R7, Canada; Molecular Biology Program, Université de Montréal, Montreal, QC H3C 3J7, Canada
| | - Thomas W Brown
- Cellular Neurobiology Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, QC H2W 1R7, Canada; Integrated Program in Neuroscience, McGill University, Montreal, QC H3A 1A1, Canada
| | - Christine Jolicoeur
- Cellular Neurobiology Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, QC H2W 1R7, Canada
| | - Benoit Boulan
- Cellular Neurobiology Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, QC H2W 1R7, Canada
| | - Camille Boudreau-Pinsonneault
- Cellular Neurobiology Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, QC H2W 1R7, Canada; Integrated Program in Neuroscience, McGill University, Montreal, QC H3A 1A1, Canada
| | - Awais Javed
- Cellular Neurobiology Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, QC H2W 1R7, Canada; Molecular Biology Program, Université de Montréal, Montreal, QC H3C 3J7, Canada
| | - Pénélope Abram
- Cellular Neurobiology Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, QC H2W 1R7, Canada
| | - Michel Cayouette
- Cellular Neurobiology Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, QC H2W 1R7, Canada; Molecular Biology Program, Université de Montréal, Montreal, QC H3C 3J7, Canada; Integrated Program in Neuroscience, McGill University, Montreal, QC H3A 1A1, Canada; Department of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada.
| |
Collapse
|
5
|
Atkins M, Nicol X, Fassier C. Microtubule remodelling as a driving force of axon guidance and pruning. Semin Cell Dev Biol 2023; 140:35-53. [PMID: 35710759 DOI: 10.1016/j.semcdb.2022.05.030] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/26/2022] [Accepted: 05/31/2022] [Indexed: 01/28/2023]
Abstract
The establishment of neuronal connectivity relies on the microtubule (MT) cytoskeleton, which provides mechanical support, roads for axonal transport and mediates signalling events. Fine-tuned spatiotemporal regulation of MT functions by tubulin post-translational modifications and MT-associated proteins is critical for the coarse wiring and subsequent refinement of neuronal connectivity. The defective regulation of these processes causes a wide range of neurodevelopmental disorders associated with connectivity defects. This review focuses on recent studies unravelling how MT composition, post-translational modifications and associated proteins influence MT functions in axon guidance and/or pruning to build functional neuronal circuits. We here summarise experimental evidence supporting the key role of this network as a driving force for growth cone steering and branch-specific axon elimination. We further provide a global overview of the MT-interactors that tune developing axon behaviours, with a special emphasis on their emerging versatility in the regulation of MT dynamics/structure. Recent studies establishing the key and highly selective role of the tubulin code in the regulation of MT functions in axon pathfinding are also reported. Finally, our review highlights the emerging molecular links between these MT regulation processes and guidance signals that wire the nervous system.
Collapse
Affiliation(s)
- Melody Atkins
- INSERM, UMR-S 1270, Institut du Fer à Moulin, Sorbonne Université, F-75005 Paris, France
| | - Xavier Nicol
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, F-75012 Paris, France
| | - Coralie Fassier
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, F-75012 Paris, France.
| |
Collapse
|
6
|
Ritchie FD, Lizarraga SB. The role of histone methyltransferases in neurocognitive disorders associated with brain size abnormalities. Front Neurosci 2023; 17:989109. [PMID: 36845425 PMCID: PMC9950662 DOI: 10.3389/fnins.2023.989109] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 01/17/2023] [Indexed: 02/12/2023] Open
Abstract
Brain size is controlled by several factors during neuronal development, including neural progenitor proliferation, neuronal arborization, gliogenesis, cell death, and synaptogenesis. Multiple neurodevelopmental disorders have co-morbid brain size abnormalities, such as microcephaly and macrocephaly. Mutations in histone methyltransferases that modify histone H3 on Lysine 36 and Lysine 4 (H3K36 and H3K4) have been identified in neurodevelopmental disorders involving both microcephaly and macrocephaly. H3K36 and H3K4 methylation are both associated with transcriptional activation and are proposed to sterically hinder the repressive activity of the Polycomb Repressor Complex 2 (PRC2). During neuronal development, tri-methylation of H3K27 (H3K27me3) by PRC2 leads to genome wide transcriptional repression of genes that regulate cell fate transitions and neuronal arborization. Here we provide a review of neurodevelopmental processes and disorders associated with H3K36 and H3K4 histone methyltransferases, with emphasis on processes that contribute to brain size abnormalities. Additionally, we discuss how the counteracting activities of H3K36 and H3K4 modifying enzymes vs. PRC2 could contribute to brain size abnormalities which is an underexplored mechanism in relation to brain size control.
Collapse
|
7
|
Fang X, Svitkina TM. Adenomatous polyposis coli (APC) in cell migration. Eur J Cell Biol 2022; 101:151228. [DOI: 10.1016/j.ejcb.2022.151228] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/15/2022] [Accepted: 04/20/2022] [Indexed: 12/22/2022] Open
|
8
|
Sánchez-Huertas C, Herrera E. With the Permission of Microtubules: An Updated Overview on Microtubule Function During Axon Pathfinding. Front Mol Neurosci 2021; 14:759404. [PMID: 34924953 PMCID: PMC8675249 DOI: 10.3389/fnmol.2021.759404] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 11/01/2021] [Indexed: 01/27/2023] Open
Abstract
During the establishment of neural circuitry axons often need to cover long distances to reach remote targets. The stereotyped navigation of these axons defines the connectivity between brain regions and cellular subtypes. This chemotrophic guidance process mostly relies on the spatio-temporal expression patterns of extracellular proteins and the selective expression of their receptors in projection neurons. Axon guidance is stimulated by guidance proteins and implemented by neuronal traction forces at the growth cones, which engage local cytoskeleton regulators and cell adhesion proteins. Different layers of guidance signaling regulation, such as the cleavage and processing of receptors, the expression of co-receptors and a wide variety of intracellular cascades downstream of receptors activation, have been progressively unveiled. Also, in the last decades, the regulation of microtubule (MT) assembly, stability and interactions with the submembranous actin network in the growth cone have emerged as crucial effector mechanisms in axon pathfinding. In this review, we will delve into the intracellular signaling cascades downstream of guidance receptors that converge on the MT cytoskeleton of the growing axon. In particular, we will focus on the microtubule-associated proteins (MAPs) network responsible of MT dynamics in the axon and growth cone. Complementarily, we will discuss new evidences that connect defects in MT scaffold proteins, MAPs or MT-based motors and axon misrouting during brain development.
Collapse
Affiliation(s)
- Carlos Sánchez-Huertas
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández (CSIC-UMH), Alicante, Spain
| | | |
Collapse
|
9
|
Unraveling Axon Guidance during Axotomy and Regeneration. Int J Mol Sci 2021; 22:ijms22158344. [PMID: 34361110 PMCID: PMC8347220 DOI: 10.3390/ijms22158344] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 02/06/2023] Open
Abstract
During neuronal development and regeneration axons extend a cytoskeletal-rich structure known as the growth cone, which detects and integrates signals to reach its final destination. The guidance cues “signals” bind their receptors, activating signaling cascades that result in the regulation of the growth cone cytoskeleton, defining growth cone advance, pausing, turning, or collapse. Even though much is known about guidance cues and their isolated mechanisms during nervous system development, there is still a gap in the understanding of the crosstalk between them, and about what happens after nervous system injuries. After neuronal injuries in mammals, only axons in the peripheral nervous system are able to regenerate, while the ones from the central nervous system fail to do so. Therefore, untangling the guidance cues mechanisms, as well as their behavior and characterization after axotomy and regeneration, are of special interest for understanding and treating neuronal injuries. In this review, we present findings on growth cone guidance and canonical guidance cues mechanisms, followed by a description and comparison of growth cone pathfinding mechanisms after axotomy, in regenerative and non-regenerative animal models.
Collapse
|
10
|
Chen J, Kholina E, Szyk A, Fedorov VA, Kovalenko I, Gudimchuk N, Roll-Mecak A. α-tubulin tail modifications regulate microtubule stability through selective effector recruitment, not changes in intrinsic polymer dynamics. Dev Cell 2021; 56:2016-2028.e4. [PMID: 34022132 PMCID: PMC8476856 DOI: 10.1016/j.devcel.2021.05.005] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/26/2021] [Accepted: 05/05/2021] [Indexed: 10/21/2022]
Abstract
Microtubules are non-covalent polymers of αβ-tubulin dimers. Posttranslational processing of the intrinsically disordered C-terminal α-tubulin tail produces detyrosinated and Δ2-tubulin. Although these are widely employed as proxies for stable cellular microtubules, their effect (and of the α-tail) on microtubule dynamics remains uncharacterized. Using recombinant, engineered human tubulins, we now find that neither detyrosinated nor Δ2-tubulin affect microtubule dynamics, while the α-tubulin tail is an inhibitor of microtubule growth. Consistent with the latter, molecular dynamics simulations show the α-tubulin tail transiently occluding the longitudinal microtubule polymerization interface. The marked differential in vivo stabilities of the modified microtubule subpopulations, therefore, must result exclusively from selective effector recruitment. We find that tyrosination quantitatively tunes CLIP-170 density at the growing plus end and that CLIP170 and EB1 synergize to selectively upregulate the dynamicity of tyrosinated microtubules. Modification-dependent recruitment of regulators thereby results in microtubule subpopulations with distinct dynamics, a tenet of the tubulin code hypothesis.
Collapse
Affiliation(s)
- Jiayi Chen
- Cell Biology and Biophysics Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20892, USA
| | - Ekaterina Kholina
- Department of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Agnieszka Szyk
- Cell Biology and Biophysics Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20892, USA
| | - Vladimir A Fedorov
- Department of Biology, Lomonosov Moscow State University, Moscow, Russia; Center for Theoretical Problems of Physicochemical Pharmacology, Russian Academy of Sciences, Moscow, Russia
| | - Ilya Kovalenko
- Department of Biology, Lomonosov Moscow State University, Moscow, Russia; Astrakhan State University, Astrakhan 414056, Russia; Sechenov University, Moscow 119991, Russia
| | - Nikita Gudimchuk
- Department of Physics, Lomonosov Moscow State University, Moscow, Russia; Center for Theoretical Problems of Physicochemical Pharmacology, Russian Academy of Sciences, Moscow, Russia; Dmitry Rogachev National Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia.
| | - Antonina Roll-Mecak
- Cell Biology and Biophysics Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20892, USA; Biochemistry and Biophysics Center, National Heart Lung and Blood Institute, Bethesda, MD 20892, USA.
| |
Collapse
|
11
|
Zou R, El Marroun H, Cecil C, Jaddoe VWV, Hillegers M, Tiemeier H, White T. Maternal folate levels during pregnancy and offspring brain development in late childhood. Clin Nutr 2020; 40:3391-3400. [PMID: 33279309 DOI: 10.1016/j.clnu.2020.11.025] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 11/13/2020] [Accepted: 11/15/2020] [Indexed: 01/13/2023]
Abstract
BACKGROUND Cumulative evidence shows that low maternal folate levels during pregnancy are associated with offspring neuropsychiatric disorders even in the absence of neural tube defects. However, the relationship between prenatal exposure to folate and brain development in late childhood has been rarely investigated. METHODS In 2095 children from a prospective population-based cohort in Rotterdam, the Netherlands, we examined the association of maternal folate levels during pregnancy with downstream brain development in offspring. Maternal folate concentrations were measured from venous blood in early gestation. Child structural neuroimaging data were measured at age 9-11 years. In addition, measures of child head circumference using fetal ultrasound in the third trimester and total brain volume using magnetic resonance imaging at age 6-8 years were used for analyses with repeated assessments of brain development. RESULTS Maternal folate deficiency (i.e., <7 nmol/L) during pregnancy was associated with smaller total brain volume (B = -18.7 cm3, 95% CI -37.2 to -0.2) and smaller cerebral white matter (B = -7.2 cm3, 95% CI -11.8 to -2.6) in children aged 9-11 years. No differences in cortical thickness or surface area were observed. Analysis of the repeated brain assessments showed that children exposed to deficient folate concentrations in utero had persistently smaller brains compared to controls from the third trimester to childhood (β = -0.4, 95% CI -0.6 to -0.1). CONCLUSIONS Low maternal folate levels during pregnancy are associated with altered offspring brain development in childhood, suggesting the importance of essential folate concentrations in early pregnancy.
Collapse
Affiliation(s)
- Runyu Zou
- Department of Child and Adolescent Psychiatry, Erasmus MC University Medical Center Rotterdam, Rotterdam, the Netherlands; The Generation R Study Group, Erasmus MC University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Hanan El Marroun
- Department of Child and Adolescent Psychiatry, Erasmus MC University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Pediatrics, Erasmus MC University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Psychology, Education and Child Studies, Erasmus School of Social and Behavioral Sciences, Erasmus University Rotterdam, Rotterdam, the Netherlands
| | - Charlotte Cecil
- Department of Child and Adolescent Psychiatry, Erasmus MC University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, the Netherlands; Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, 2333 ZC, Leiden, the Netherlands
| | - Vincent W V Jaddoe
- The Generation R Study Group, Erasmus MC University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Pediatrics, Erasmus MC University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Manon Hillegers
- Department of Child and Adolescent Psychiatry, Erasmus MC University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Henning Tiemeier
- Department of Child and Adolescent Psychiatry, Erasmus MC University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Social and Behavioral Sciences, T.H. Chan School of Public Health, Harvard University, Boston, MA, United States
| | - Tonya White
- Department of Child and Adolescent Psychiatry, Erasmus MC University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, the Netherlands.
| |
Collapse
|
12
|
Huybrechts Y, Mortier G, Boudin E, Van Hul W. WNT Signaling and Bone: Lessons From Skeletal Dysplasias and Disorders. Front Endocrinol (Lausanne) 2020; 11:165. [PMID: 32328030 PMCID: PMC7160326 DOI: 10.3389/fendo.2020.00165] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 03/09/2020] [Indexed: 12/15/2022] Open
Abstract
Skeletal dysplasias are a diverse group of heritable diseases affecting bone and cartilage growth. Throughout the years, the molecular defect underlying many of the diseases has been identified. These identifications led to novel insights in the mechanisms regulating bone and cartilage growth and homeostasis. One of the pathways that is clearly important during skeletal development and bone homeostasis is the Wingless and int-1 (WNT) signaling pathway. So far, three different WNT signaling pathways have been described, which are all activated by binding of the WNT ligands to the Frizzled (FZD) receptors. In this review, we discuss the skeletal disorders that are included in the latest nosology of skeletal disorders and that are caused by genetic defects involving the WNT signaling pathway. The number of skeletal disorders caused by defects in WNT signaling genes and the clinical phenotype associated with these disorders illustrate the importance of the WNT signaling pathway during skeletal development as well as later on in life to maintain bone mass. The knowledge gained through the identification of the genes underlying these monogenic conditions is used for the identification of novel therapeutic targets. For example, the genes underlying disorders with altered bone mass are all involved in the canonical WNT signaling pathway. Consequently, targeting this pathway is one of the major strategies to increase bone mass in patients with osteoporosis. In addition to increasing the insights in the pathways regulating skeletal development and bone homeostasis, knowledge of rare skeletal dysplasias can also be used to predict possible adverse effects of these novel drug targets. Therefore, this review gives an overview of the skeletal and extra-skeletal phenotype of the different skeletal disorders linked to the WNT signaling pathway.
Collapse
|
13
|
Fujikawa A, Noda Y, Yamamoto H, Tanga N, Sakaguchi G, Hattori S, Song WJ, Sora I, Nabeshima T, Katsuura G, Noda M. Mice deficient in protein tyrosine phosphatase receptor type Z (PTPRZ) show reduced responsivity to methamphetamine despite an enhanced response to novelty. PLoS One 2019; 14:e0221205. [PMID: 31430310 PMCID: PMC6701799 DOI: 10.1371/journal.pone.0221205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 08/02/2019] [Indexed: 12/05/2022] Open
Abstract
Methamphetamine (METH), a commonly abused drug, elevates extracellular dopamine (DA) levels by inducing DA efflux through the DA transporter (DAT). Emerging evidence in rodent models suggests that locomotor responses to a novel inescapable open field may predict behavioral responses to abused drugs; METH produces more potent stimulant effects in high responders to novelty than in low responders. We herein found that mice deficient in protein tyrosine phosphatase receptor type Z (Ptprz-KO) exhibited an enhanced behavioral response to novelty; however, METH-induced hyperlocomotion was significantly lower in Ptprz-KO than in wild-type mice when METH was administered at a non-toxic dose of 1 mg per kg body weight (bdw). Single-cell RT-PCR revealed that the majority of midbrain DA neurons expressed PTPRZ. No histological alterations were observed in the mesolimbic or nigrostriatal dopaminergic pathways in Ptprz-KO brains; however, a significant decrease was noted in brain DA turnover, suggesting functional alterations. In vivo microdialysis experiments revealed that METH-evoked DA release in the nucleus accumbens was significantly lower in Ptprz-KO mice than in wild-type mice. Consistent with this result, Ptprz-KO mice showed significantly fewer cell surface DAT as well as weaker DA uptake activity in striatal synaptosomes prepared 1 hr after the administration of METH than wild-type mice, while no significant differences were observed in the two groups treated with saline. These results indicate that the high response phenotype of Ptprz-KO mice to novelty may not be simply attributed to hyper-dopaminergic activity, and that deficits in PTPRZ reduce the effects of METH by reducing DAT activity.
Collapse
Affiliation(s)
- Akihiro Fujikawa
- Division of Molecular Neurobiology, National Institute for Basic Biology, Higashiyama, Myodaiji-cho, Okazaki, Japan
| | - Yukihiro Noda
- Division of Clinical Sciences and Neuropsychopharmacology, Faculty of Pharmacy, Meijo University, Shiogamaguchi, Tempaku-ku, Nagoya, Aichi, Japan
| | - Hideko Yamamoto
- Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Kamikitazawa, Setagaya-ku, Tokyo, Japan
| | - Naomi Tanga
- Division of Molecular Neurobiology, National Institute for Basic Biology, Higashiyama, Myodaiji-cho, Okazaki, Japan
- School of Life Sciences, The Graduate University for Advanced Studies (SOKENDAI), Higashiyama, Myodaiji-cho, Okazaki, Aichi, Japan
| | - Gaku Sakaguchi
- Biomarker R&D Dept., SHIONOGI & CO. LTD., Futaba-cho, Toyonaka, Osaka, Japan
| | - Satoko Hattori
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, Japan
| | - Wen-Jie Song
- Department of Sensory and Cognitive Physiology, Graduate School of Medical Sciences, Kumamoto University, Honjo, Chuo-ku, Kumamoto, Japan
| | - Ichiro Sora
- Department of Psychiatry Kobe University Graduate School of Medicine, Kusunoki-cho, Chuo-ku, Kobe, Hyogo, Japan
| | - Toshitaka Nabeshima
- Advanced Diagnostic System Research Laboratory, Fujita Health University Graduate School of Health Sciences, Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, Japan
| | - Goro Katsuura
- Department of Psychosomatic Internal Medicine, Kagoshima University Graduate School of Medical and Dental Sciences & University Hospital, Sakuragaoka, Kagoshima, Japan
| | - Masaharu Noda
- Division of Molecular Neurobiology, National Institute for Basic Biology, Higashiyama, Myodaiji-cho, Okazaki, Japan
- School of Life Sciences, The Graduate University for Advanced Studies (SOKENDAI), Higashiyama, Myodaiji-cho, Okazaki, Aichi, Japan
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa, Japan
- * E-mail:
| |
Collapse
|
14
|
Mohamed NE, Hay T, Reed KR, Smalley MJ, Clarke AR. APC2 is critical for ovarian WNT signalling control, fertility and tumour suppression. BMC Cancer 2019; 19:677. [PMID: 31291912 PMCID: PMC6617595 DOI: 10.1186/s12885-019-5867-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 06/24/2019] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Canonical WNT signalling plays a critical role in the regulation of ovarian development; mis-regulation of this key pathway in the adult ovary is associated with subfertility and tumourigenesis. The roles of Adenomatous polyposis coli 2 (APC2), a little-studied WNT signalling pathway regulator, in ovarian homeostasis, fertility and tumourigenesis have not previously been explored. Here, we demonstrate essential roles of APC2 in regulating ovarian WNT signalling and ovarian homeostasis. METHODS A detailed analysis of ovarian histology, gene expression, ovulation and hormone levels was carried out in 10 week old and in aged constitutive APC2-knockout (Apc2-/-) mice (mixed background). Statistical significance for qRT-PCR data was determined from 95% confidence intervals. Significance testing was performed using 2-tailed Student's t-test, when 2 experimental cohorts were compared. When more were compared, ANOVA test was used, followed by a post-hoc test (LSD or Games-Howell). P-values of < 0.05 were considered statistically significant. RESULTS APC2-deficiency resulted in activation of ovarian WNT signalling and sub-fertility driven by intra-ovarian defects. Follicular growth was perturbed, resulting in a reduced rate of ovulation and corpora lutea formation, which could not be rescued by administration of gonadotrophins. Defects in steroidogenesis and follicular vascularity contributed to the subfertility phenotype. Tumour incidence was assessed in aged APC2-deficient mice, which also carried a hypomorphic Apc allele. APC2-deficiency in these mice resulted in predisposition to granulosa cell tumour (GCT) formation, accompanied by acute tumour-associated WNT-signalling activation and a histologic pattern and molecular signature seen in human adult GCTs. CONCLUSIONS Our work adds APC2 to the growing list of WNT-signalling members that regulate ovarian homeostasis, fertility and suppress GCT formation. Importantly, given that the APC2-deficient mouse develops tumours that recapitulate the molecular signature and histological features of human adult GCTs, this mouse has excellent potential as a pre-clinical model to study ovarian subfertility and transitioning to GCT, tumour biology and for therapeutic testing.
Collapse
Affiliation(s)
- Noha-Ehssan Mohamed
- European Cancer Stem Cell Research Institute, Cardiff University School of Biosciences, Hadyn Ellis Building, Maindy, Road, Cardiff, CF24 4HQ UK
- Hormones Evaluation Department, National Organization for Drug Control and Research (NODCAR), Giza, Egypt
- Present address: CRUK Beatson Institute, Switchback road, Bearsden, Glasgow, G61 1BD UK
| | - Trevor Hay
- European Cancer Stem Cell Research Institute, Cardiff University School of Biosciences, Hadyn Ellis Building, Maindy, Road, Cardiff, CF24 4HQ UK
| | - Karen R. Reed
- European Cancer Stem Cell Research Institute, Cardiff University School of Biosciences, Hadyn Ellis Building, Maindy, Road, Cardiff, CF24 4HQ UK
| | - Matthew J. Smalley
- European Cancer Stem Cell Research Institute, Cardiff University School of Biosciences, Hadyn Ellis Building, Maindy, Road, Cardiff, CF24 4HQ UK
| | - Alan R. Clarke
- European Cancer Stem Cell Research Institute, Cardiff University School of Biosciences, Hadyn Ellis Building, Maindy, Road, Cardiff, CF24 4HQ UK
| |
Collapse
|
15
|
Wang Z, Xie J, Wu C, Xiao G. Correlation Between Smoking and Passive Smoking with Multiple Sclerosis and the Underlying Molecular Mechanisms. Med Sci Monit 2019; 25:893-902. [PMID: 30703074 PMCID: PMC6367889 DOI: 10.12659/msm.912863] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic immune-mediated disease of the spinal cord and brain. Many studies have shown that smoking and passive smoking are key environmental risk factors for MS. Here, we provide an overview of the human leukocyte antigen (HLA) gene studies on smoking and MS risk, and we discuss recent studies on between epigenetics and smoking-induced MS. In addition, in this review we also summarize current research advances in biological pathways and smoking-induced MS. This review provides an overview of studies on the association between smoking, passive smoking, and MS susceptibility, and the underlying molecular mechanism.
Collapse
Affiliation(s)
- Zhaowei Wang
- Department of Neurology, Shaoxing People's Hospital (Shaoxing Hospital of Zhejiang University), Shaoxing, Zhejiang, China (mainland)
| | - Jianpin Xie
- Department of Neurology, Shaoxing People's Hospital (Shaoxing Hospital of Zhejiang University), Shaoxing, Zhejiang, China (mainland)
| | - Chenglong Wu
- Department of Neurology, Shaoxing People's Hospital (Shaoxing Hospital of Zhejiang University), Shaoxing, Zhejiang, China (mainland)
| | - Guirong Xiao
- Department of Neurology, Shaoxing People's Hospital (Shaoxing Hospital of Zhejiang University), Shaoxing, Zhejiang, China (mainland)
| |
Collapse
|
16
|
Protein Tyrosine Phosphatase Receptor Type J (PTPRJ) Regulates Retinal Axonal Projections by Inhibiting Eph and Abl Kinases in Mice. J Neurosci 2018; 38:8345-8363. [PMID: 30082414 DOI: 10.1523/jneurosci.0128-18.2018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 07/05/2018] [Accepted: 07/30/2018] [Indexed: 12/28/2022] Open
Abstract
Eph receptors play pivotal roles in the axon guidance of retinal ganglion cells (RGCs) at the optic chiasm and the establishment of the topographic retinocollicular map. We previously demonstrated that protein tyrosine phosphatase receptor type O (PTPRO) is specifically involved in the control of retinotectal projections in chicks through the dephosphorylation of EphA and EphB receptors. We subsequently revealed that all the mouse R3 subfamily members (PTPRB, PTPRH, PTPRJ, and PTPRO) of the receptor protein tyrosine phosphatase (RPTP) family inhibited Eph receptors as their substrates in cultured mammalian cells. We herein investigated the functional roles of R3 RPTPs in the projection of mouse retinal axon of both sexes. Ptpro and Ptprj were expressed in mouse RGCs; however, Ptprj expression levels were markedly higher than those of Ptpro Consistent with their expression levels, Eph receptor activity was significantly enhanced in Ptprj-knock-out (Ptprj-KO) retinas. In Ptprj-KO and Ptprj/Ptpro-double-KO (DKO) mice, the number of retinal axons that projected ipsilaterally or to the contralateral eye was significantly increased. Furthermore, retinal axons in Ptprj-KO and DKO mice formed anteriorly shifted ectopic terminal zones in the superior colliculus (SC). We found that c-Abl (Abelson tyrosine kinase) was downstream of ephrin-Eph signaling for the repulsion of retinal axons at the optic chiasm and in the SC. c-Abl was identified as a novel substrate for PTPRJ and PTPRO, and the phosphorylation of c-Abl was upregulated in Ptprj-KO and DKO retinas. Thus, PTPRJ regulates retinocollicular projections in mice by controlling the activity of Eph and c-Abl kinases.SIGNIFICANCE STATEMENT Correct retinocollicular projection is a prerequisite for proper vision. Eph receptors have been implicated in retinal axon guidance at the optic chiasm and the establishment of the topographic retinocollicular map. We herein demonstrated that protein tyrosine phosphatase receptor type J (PTPRJ) regulated retinal axonal projections by controlling Eph activities. The retinas of Ptprj-knock-out (KO) and Ptpro/Ptprj double-KO mice exhibited significantly enhanced Eph activities over those in wild-type mice, and their axons showed defects in pathfinding at the chiasm and retinocollicular topographic map formation. We also revealed that c-Abl (Abelson tyrosine kinase) downstream of Eph receptors was regulated by PTPRJ. These results indicate that the regulation of the ephrin-Eph-c-Abl axis by PTPRJ plays pivotal roles in the proper central projection of retinal axons during development.
Collapse
|
17
|
Kahn OI, Schätzle P, van de Willige D, Tas RP, Lindhout FW, Portegies S, Kapitein LC, Hoogenraad CC. APC2 controls dendrite development by promoting microtubule dynamics. Nat Commun 2018; 9:2773. [PMID: 30018294 PMCID: PMC6050278 DOI: 10.1038/s41467-018-05124-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 06/12/2018] [Indexed: 12/12/2022] Open
Abstract
Mixed polarity microtubule organization is the signature characteristic of vertebrate dendrites. Oppositely oriented microtubules form the basis for selective cargo trafficking in neurons, however the mechanisms that establish and maintain this organization are unclear. Here, we show that APC2, the brain-specific homolog of tumor-suppressor protein adenomatous polyposis coli (APC), promotes dynamics of minus-end-out microtubules in dendrites. We found that APC2 localizes as distinct clusters along microtubule bundles in dendrites and that this localization is driven by LC8-binding and two separate microtubule-interacting domains. Depletion of APC2 reduces the plus end dynamics of minus-end-out oriented microtubules, increases microtubule sliding, and causes defects in dendritic morphology. We propose a model in which APC2 regulates dendrite development by promoting dynamics of minus-end-out microtubules.
Collapse
Affiliation(s)
- Olga I Kahn
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, 3584 CH, Utrecht, The Netherlands
| | - Philipp Schätzle
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, 3584 CH, Utrecht, The Netherlands
| | - Dieudonnée van de Willige
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, 3584 CH, Utrecht, The Netherlands
| | - Roderick P Tas
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, 3584 CH, Utrecht, The Netherlands
| | - Feline W Lindhout
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, 3584 CH, Utrecht, The Netherlands
| | - Sybren Portegies
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, 3584 CH, Utrecht, The Netherlands
| | - Lukas C Kapitein
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, 3584 CH, Utrecht, The Netherlands
| | - Casper C Hoogenraad
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, 3584 CH, Utrecht, The Netherlands.
| |
Collapse
|
18
|
Merkurjev D, Hong WT, Iida K, Oomoto I, Goldie BJ, Yamaguti H, Ohara T, Kawaguchi SY, Hirano T, Martin KC, Pellegrini M, Wang DO. Synaptic N6-methyladenosine (m6A) epitranscriptome reveals functional partitioning of localized transcripts. Nat Neurosci 2018; 21:1004-1014. [DOI: 10.1038/s41593-018-0173-6] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 05/14/2018] [Indexed: 01/21/2023]
|
19
|
Arbeille E, Bashaw GJ. Brain Tumor promotes axon growth across the midline through interactions with the microtubule stabilizing protein Apc2. PLoS Genet 2018; 14:e1007314. [PMID: 29617376 PMCID: PMC5902039 DOI: 10.1371/journal.pgen.1007314] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 04/16/2018] [Accepted: 03/19/2018] [Indexed: 11/20/2022] Open
Abstract
Commissural axons must cross the midline to establish reciprocal connections between the two sides of the body. This process is highly conserved between invertebrates and vertebrates and depends on guidance cues and their receptors to instruct axon trajectories. The DCC family receptor Frazzled (Fra) signals chemoattraction and promotes midline crossing in response to its ligand Netrin. However, in Netrin or fra mutants, the loss of crossing is incomplete, suggesting the existence of additional pathways. Here, we identify Brain Tumor (Brat), a tripartite motif protein, as a new regulator of midline crossing in the Drosophila CNS. Genetic analysis indicates that Brat acts independently of the Netrin/Fra pathway. In addition, we show that through its B-Box domains, Brat acts cell autonomously to regulate the expression and localization of Adenomatous polyposis coli-2 (Apc2), a key component of the Wnt canonical signaling pathway, to promote axon growth across the midline. Genetic evidence indicates that the role of Brat and Apc2 to promote axon growth across the midline is independent of Wnt and Beta-catenin-mediated transcriptional regulation. Instead, we propose that Brat promotes midline crossing through directing the localization or stability of Apc2 at the plus ends of microtubules in navigating commissural axons. These findings define a new mechanism in the coordination of axon growth and guidance at the midline. The establishment of neuronal connections that cross the midline of the animal is essential to generate neural circuits that coordinate the left and right sides of the body. Axons that cross the midline to form these connections are called commissural axons and the molecules and mechanisms that control midline axon crossing are remarkably conserved across animal evolution. In this study we have used a genetic screen in the fruit fly in an attempt to uncover additional players in this key developmental process, and have identified a novel role for the Brain Tumor (Brat) protein in promoting commissural axon growth across the midline. Unlike its previous described functions, in the context of midline axon guidance Brat cooperates with the microtubule stabilizing protein Apc2 to coordinate axon growth and guidance. Molecular and genetic analyses point to the conserved B box motifs of the Brat protein as key in promoting the association of Apc2 with the plus ends of microtubules. Brat is highly conserved and future studies will determine whether homologous genes play analogous roles in mammalian neural development.
Collapse
Affiliation(s)
- Elise Arbeille
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Greg J. Bashaw
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
- * E-mail:
| |
Collapse
|
20
|
DNA Methylation: a New Player in Multiple Sclerosis. Mol Neurobiol 2016; 54:4049-4059. [PMID: 27314687 DOI: 10.1007/s12035-016-9966-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 06/08/2016] [Indexed: 12/11/2022]
Abstract
Multiple sclerosis (MS) is a neurological and chronic inflammatory disease that is mediated by demyelination and axonal degeneration in the central nervous system (CNS). Studies have shown that immune system components such as CD4+, CD8+, CD44+ T cells, B lymphatic cells, and inflammatory cytokines play a critical role in inflammatory processes and myelin damage associated with MS. Nevertheless, the pathogenesis of MS remains poorly defined. DNA methylation, a significant epigenetic modification, is reported to be extensively involved in MS pathogenesis through the regulation of gene expression. This review focuses on DNA methylation involved in MS pathogenesis. Evidence showed the hypermethylation of human leukocyte antigen-DRB1 (HLA-DRB1) in CD4+ T cells, the genome-wide DNA methylation in CD8+ T cells, the hypermethylation of interleukin-4 (IL-4)/forkhead winged helix transcription factor 3 (Foxp3), and the demethylation of interferon-γ (IFN-γ)/IL-17a in CD44+ encephalitogenic T cells. Studies also showed the hypermethylation of SH2-containing protein tyrosine phosphatase-1 (SHP-1) in peripheral blood mononuclear cells (PBMCs) and methylated changes of genes regulating oligodendrocyte and neuronal function in normal-appearing white matter. Clarifying the mechanism of aberrant methylation on MS may explain part of the pathology and will lead to the development of a new therapeutic target for the treatment of MS in the future.
Collapse
|
21
|
Palumbo P, Palumbo O, Leone MP, Stallone R, Palladino T, Zelante L, Carella M. Clinical and molecular characterization of a de novo 19p13.3 microdeletion. Mol Cytogenet 2016; 9:40. [PMID: 27239227 PMCID: PMC4882821 DOI: 10.1186/s13039-016-0252-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 05/18/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Structural rearrangements of chromosome 19p13.3 are a rare condition, and their phenotypic consequences remain not well defined, because of the variability of clinical manifestations. Increasing knowledge of new 19p13.3 microdeletion is useful to clarify the phenotypic variability observed in some patients. In a small number of recent papers, patients with intellectual disabilities, multiple congenital anomalies and microdeletion of the chromosome band 19p13.3 have been described. However, little is known about genes responsible for clinical features in patients carriers of 19p13.3 microdeletion; thus, increasing number of reported cases will be helpful to investigate the contribution of candidate genes, providing bases for future investigations. CASE PRESENTATION Here, we report on a 10-years-old girl referred to our genetics clinic due to intellectual disability, attention deficit, behavioral and speech delay, hypotonia, facial dysmorphisms, eye anomalies and congenital malformations. Using an high resolution SNP array, we identified a de novo microdeletion of chromosome 19p13.3, resulting in the heterozygous loss of 27 RefSeq genes and a miRNA, partially overlapping with three others deletions already reported in literature, but extending downstream (centromeric) for additional 386 Kb. This chromosomal region includes 13 genes amongst of which we suggest for the first time the APC2, PLK5 and MBD3 genes as potential functional candidates for neurodevelopmental and behavioral phenotypes observed. CONCLUSIONS Here we describe a patient with a 19p13.3 microdeletion that spans to the downstream chromosomal region with respect to the overlapping deletions previously reported in several other cases. The neurobehavioral features observed in our case has extended the phenotypic spectrum associated with the 19p13.3 microdeletion. New candidate genes are proposed for the neurobehavioral phenotype observed in our case.
Collapse
Affiliation(s)
- Pietro Palumbo
- Laboratorio di Genetica Medica, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo (FG), Italy
| | - Orazio Palumbo
- Laboratorio di Genetica Medica, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo (FG), Italy
| | - Maria Pia Leone
- Laboratorio di Genetica Medica, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo (FG), Italy ; Dipartimento di Scienze del suolo, della pianta e degli alimenti, Università degli Studi di Bari "Aldo Moro", Bari, Italy
| | - Raffaella Stallone
- Laboratorio di Genetica Medica, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo (FG), Italy
| | - Teresa Palladino
- Laboratorio di Genetica Medica, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo (FG), Italy
| | - Leopoldo Zelante
- Laboratorio di Genetica Medica, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo (FG), Italy
| | - Massimo Carella
- Laboratorio di Genetica Medica, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo (FG), Italy
| |
Collapse
|
22
|
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
|
23
|
Joubert BR, den Dekker HT, Felix JF, Bohlin J, Ligthart S, Beckett E, Tiemeier H, van Meurs JB, Uitterlinden AG, Hofman A, Håberg SE, Reese SE, Peters MJ, Andreassen BK, Steegers EAP, Nilsen RM, Vollset SE, Midttun Ø, Ueland PM, Franco OH, Dehghan A, de Jongste JC, Wu MC, Wang T, Peddada SD, Jaddoe VWV, Nystad W, Duijts L, London SJ. Maternal plasma folate impacts differential DNA methylation in an epigenome-wide meta-analysis of newborns. Nat Commun 2016; 7:10577. [PMID: 26861414 PMCID: PMC4749955 DOI: 10.1038/ncomms10577] [Citation(s) in RCA: 194] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2015] [Accepted: 12/31/2015] [Indexed: 12/13/2022] Open
Abstract
Folate is vital for fetal development. Periconceptional folic acid supplementation and food fortification are recommended to prevent neural tube defects. Mechanisms whereby periconceptional folate influences normal development and disease are poorly understood: epigenetics may be involved. We examine the association between maternal plasma folate during pregnancy and epigenome-wide DNA methylation using Illumina's HumanMethyl450 Beadchip in 1,988 newborns from two European cohorts. Here we report the combined covariate-adjusted results using meta-analysis and employ pathway and gene expression analyses. Four-hundred forty-three CpGs (320 genes) are significantly associated with maternal plasma folate levels during pregnancy (false discovery rate 5%); 48 are significant after Bonferroni correction. Most genes are not known for folate biology, including APC2, GRM8, SLC16A12, OPCML, PRPH, LHX1, KLK4 and PRSS21. Some relate to birth defects other than neural tube defects, neurological functions or varied aspects of embryonic development. These findings may inform how maternal folate impacts the developing epigenome and health outcomes in offspring.
Collapse
Affiliation(s)
- Bonnie R Joubert
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina 27709, USA
| | - Herman T den Dekker
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, Netherlands.,Department of Pediatrics, Division of Respiratory Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, Netherlands.,Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, Netherlands
| | - Janine F Felix
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, Netherlands.,Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, Netherlands.,Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, Netherlands
| | - Jon Bohlin
- Norwegian Institute of Public Health, Oslo 0403, Norway
| | - Symen Ligthart
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, Netherlands
| | - Emma Beckett
- Department of Applied Sciences, School of Environmental and Life Sciences, University of Newcastle, Ourimbah, New South Wales 2258, Australia.,Food and Nutrition Flagship, CSIRO, North Ryde, New South Wales 2113, Australia
| | - Henning Tiemeier
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, Netherlands.,Department of Psychiatry, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, Netherlands
| | - Joyce B van Meurs
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, Netherlands
| | - Andre G Uitterlinden
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, Netherlands.,Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, Netherlands
| | - Albert Hofman
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, Netherlands
| | - Siri E Håberg
- Norwegian Institute of Public Health, Oslo 0403, Norway
| | - Sarah E Reese
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina 27709, USA
| | - Marjolein J Peters
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, Netherlands
| | - Bettina Kulle Andreassen
- Department of Clinical Molecular Biology, Institute of Clinical Medicine, University of Oslo, Oslo 0316, Norway
| | - Eric A P Steegers
- Department of Obstetrics and Gynaecology, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, Netherlands
| | - Roy M Nilsen
- Department of Research and Development, Centre for Clinical Research, Haukeland University Hospital, Bergen 5021, Norway
| | - Stein E Vollset
- Norwegian Institute of Public Health, Oslo 0403, Norway.,Department of Global Public Health and Primary Care, University of Bergen, Bergen 5018, Norway
| | | | - Per M Ueland
- Department of Clinical Science, University of Bergen, Bergen 5018, Norway.,Laboratory of Clinical Biochemistry, Haukeland University Hospital, Bergen 5018, Norway
| | - Oscar H Franco
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, Netherlands
| | - Abbas Dehghan
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, Netherlands
| | - Johan C de Jongste
- Department of Pediatrics, Division of Respiratory Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, Netherlands
| | - Michael C Wu
- Public Health Sciences Program, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA
| | - Tianyuan Wang
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina 27709, USA
| | - Shyamal D Peddada
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina 27709, USA
| | - Vincent W V Jaddoe
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, Netherlands.,Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, Netherlands.,Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, Netherlands
| | - Wenche Nystad
- Norwegian Institute of Public Health, Oslo 0403, Norway
| | - Liesbeth Duijts
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, Netherlands.,Department of Pediatrics, Division of Respiratory Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, Netherlands.,Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, Netherlands
| | - Stephanie J London
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina 27709, USA
| |
Collapse
|
24
|
Yu H, Sun D, Wang N, Wang M, Lan Y, Fan W, Zhao Y, Guo W, Zhu X. Headless Myo10 is a regulator of microtubule stability during neuronal development. J Neurochem 2015; 135:261-73. [PMID: 26178610 DOI: 10.1111/jnc.13238] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Revised: 07/01/2015] [Accepted: 07/01/2015] [Indexed: 01/19/2023]
Abstract
Stabilized microtubules are required for neuronal morphogenesis and migration. However, the underlying mechanism is not fully understood. In this study, we demonstrate that myosin X (Myo10), which is composed of full-length myosin X (fMyo10) and headless myosin X (hMyo10), is important for axon development. fMyo10 is involved in axon elongation, whereas hMyo10 is critical for Tau-1 positive axon formation through stabilizing microtubules. Furthermore, in vivo studies reveal that hMyo10-mediated microtubule stability has a profound effect on both neuronal migration and dendritic arborization in the mammalian cerebral cortex. Taken together, our findings suggest that hMyo10 is involved in neuronal development both in vitro and in vivo by regulating microtubule stability.
Collapse
Affiliation(s)
- Huali Yu
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Cytology and Genetics, Northeast Normal University, Changchun, Jilin, China
| | - Dong Sun
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Cytology and Genetics, Northeast Normal University, Changchun, Jilin, China
| | - Nannan Wang
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Cytology and Genetics, Northeast Normal University, Changchun, Jilin, China
| | - Min Wang
- State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Yongsheng Lan
- School of Physical Education, Changchun Normal University, Changchun, Jilin, China
| | - Wenqiang Fan
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Cytology and Genetics, Northeast Normal University, Changchun, Jilin, China
| | - Yang Zhao
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Cytology and Genetics, Northeast Normal University, Changchun, Jilin, China
| | - Weixiang Guo
- State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Xiaojuan Zhu
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Cytology and Genetics, Northeast Normal University, Changchun, Jilin, China
| |
Collapse
|
25
|
Shintani T, Higashi S, Takeuchi Y, Gaudio E, Trapasso F, Fusco A, Noda M. The R3 receptor-like protein tyrosine phosphatase subfamily inhibits insulin signalling by dephosphorylating the insulin receptor at specific sites. J Biochem 2015; 158:235-43. [PMID: 26063811 DOI: 10.1093/jb/mvv045] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 03/19/2015] [Indexed: 12/28/2022] Open
Abstract
The autophosphorylation of specific tyrosine residues occurs in the cytoplasmic region of the insulin receptor (IR) upon insulin binding, and this in turn initiates signal transduction. The R3 subfamily (Ptprb, Ptprh, Ptprj and Ptpro) of receptor-like protein tyrosine phosphatases (RPTPs) is characterized by an extracellular region with 6-17 fibronectin type III-like repeats and a cytoplasmic region with a single phosphatase domain. We herein identified the IR as a substrate for R3 RPTPs by using the substrate-trapping mutants of R3 RPTPs. The co-expression of R3 RPTPs with the IR in HEK293T cells suppressed insulin-induced tyrosine phosphorylation of the IR. In vitro assays using synthetic phosphopeptides revealed that R3 RPTPs preferentially dephosphorylated a particular phosphorylation site of the IR: Y960 in the juxtamembrane region and Y1146 in the activation loop. Among four R3 members, only Ptprj was co-expressed with the IR in major insulin target tissues, such as the skeletal muscle, liver and adipose tissue. Importantly, the activation of IR and Akt by insulin was enhanced, and glucose and insulin tolerance was improved in Ptprj-deficient mice. These results demonstrated Ptprj as a physiological enzyme that attenuates insulin signalling in vivo, and indicate that an inhibitor of Ptprj may be an insulin-sensitizing agent.
Collapse
Affiliation(s)
- Takafumi Shintani
- Division of Molecular Neurobiology, Department of Neurobiology, National Institute for Basic Biology, Okazaki 444-8787, Japan; Department of Basic Biology, School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), Okazaki 444-8787, Japan
| | - Satoru Higashi
- Division of Molecular Neurobiology, Department of Neurobiology, National Institute for Basic Biology, Okazaki 444-8787, Japan; Department of Basic Biology, School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), Okazaki 444-8787, Japan
| | - Yasushi Takeuchi
- Division of Molecular Neurobiology, Department of Neurobiology, National Institute for Basic Biology, Okazaki 444-8787, Japan
| | - Eugenio Gaudio
- Dipartimento di Medicina Sperimentale e Clinica, University Magna Græcia, Campus "S. Venuta", Catanzaro 88100, Italy; and
| | - Francesco Trapasso
- Dipartimento di Medicina Sperimentale e Clinica, University Magna Græcia, Campus "S. Venuta", Catanzaro 88100, Italy; and
| | - Alfredo Fusco
- Istituto di Endocrinologia ed Oncologia Sperimentale del CNR c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Scuola di Medicina e Chirurgia di Napoli, Università degli Studi di Napoli Federico II, Napoli 80138, Italy
| | - Masaharu Noda
- Division of Molecular Neurobiology, Department of Neurobiology, National Institute for Basic Biology, Okazaki 444-8787, Japan; Department of Basic Biology, School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), Okazaki 444-8787, Japan;
| |
Collapse
|
26
|
Zheng X, Demirci FY, Barmada MM, Richardson GA, Lopez OL, Sweet RA, Kamboh MI, Feingold E. Genome-wide copy-number variation study of psychosis in Alzheimer's disease. Transl Psychiatry 2015; 5:e574. [PMID: 26035058 PMCID: PMC4490277 DOI: 10.1038/tp.2015.64] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 01/23/2015] [Accepted: 02/08/2015] [Indexed: 01/17/2023] Open
Abstract
About 40-60% of patients with late-onset Alzheimer's disease (AD) develop psychosis, which represents a distinct phenotype of more severe cognitive and functional deficits. The estimated heritability of AD+P is ~61%, which makes it a good target for genetic mapping. We performed a genome-wide copy-number variation (CNV) study on 496 AD cases with psychosis (AD+P), 639 AD subjects with intermediate psychosis (AD intermediate P) and 156 AD subjects without psychosis (AD-P) who were recruited at the University of Pittsburgh Alzheimer's Disease Research Center using over 1 million single-nucleotide polymorphisms (SNPs) and CNV markers. CNV load analysis found no significant difference in total and average CNV length and CNV number in the AD+P or AD intermediate P groups compared with the AD-P group. Our analysis revealed a marginally significant lower number of duplication events in AD+P cases compared with AD-P controls (P=0.059) using multivariable regression model. The most interesting finding was the presence of a genome-wide significant duplication in the APC2 gene on chromosome 19, which was protective against developing AD+P (odds ratio=0.42; P=7.2E-10). We also observed suggestive associations of duplications with AD+P in the SET (P=1.95E-06), JAG2 (P=5.01E-07) and ZFPM1 (P=2.13E-07) genes and marginal association of a deletion in CNTLN (P=8.87E-04). We have identified potential novel loci for psychosis in Alzheimer's disease that warrant follow-up in large-scale independent studies.
Collapse
Affiliation(s)
- X Zheng
- Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA,Department of Pediatrics, School of Medicine, University of North Carolina, Chapel Hill, NC, USA,Department of Pediatrics, School of Medicine, University of North Carolina, Chapel Hill, NC 27514, USA. E-mail:
| | - F Y Demirci
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - M M Barmada
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - G A Richardson
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA,Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - O L Lopez
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA,VISN 4 Mental Illness Research, Education and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh, PA, USA
| | - R A Sweet
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA,Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA,VISN 4 Mental Illness Research, Education and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh, PA, USA
| | - M I Kamboh
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA,Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA,Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - E Feingold
- Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA,Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
27
|
Almuriekhi M, Shintani T, Fahiminiya S, Fujikawa A, Kuboyama K, Takeuchi Y, Nawaz Z, Nadaf J, Kamel H, Kitam AK, Samiha Z, Mahmoud L, Ben-Omran T, Majewski J, Noda M. Loss-of-Function Mutation in APC2 Causes Sotos Syndrome Features. Cell Rep 2015; 10:1585-1598. [PMID: 25753423 DOI: 10.1016/j.celrep.2015.02.011] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 01/14/2015] [Accepted: 01/31/2015] [Indexed: 01/24/2023] Open
Abstract
Sotos syndrome, characterized by intellectual disability and characteristic facial features, is caused by haploinsufficiency in the NSD1 gene. We conducted an etiological study on two siblings with Sotos features without mutations in NSD1 and detected a homozygous frameshift mutation in the APC2 gene by whole-exome sequencing, which resulted in the loss of function of cytoskeletal regulation in neurons. Apc2-deficient (Apc2-/-) mice exhibited impaired learning and memory abilities along with an abnormal head shape. Endogenous Apc2 expression was downregulated by the knockdown of Nsd1, indicating that APC2 is a downstream effector of NSD1 in neurons. Nsd1 knockdown in embryonic mouse brains impaired the migration and laminar positioning of cortical neurons, as observed in Apc2-/- mice, and this defect was rescued by the forced expression of Apc2. Thus, APC2 is a crucial target of NSD1, which provides an explanation for the intellectual disability associated with Sotos syndrome.
Collapse
Affiliation(s)
- Mariam Almuriekhi
- Section of Clinical and Metabolic Genetics, Department of Pediatrics, Hamad Medical Corporation, P.O. Box 3050, Doha, Qatar
| | - Takafumi Shintani
- Division of Molecular Neurobiology, National Institute for Basic Biology, Okazaki 444-8787, Japan; School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), Okazaki 444-8787, Japan
| | - Somayyeh Fahiminiya
- Department of Human Genetics, Faculty of Medicine, McGill University, Montreal, QC H3A 1B1, Canada; McGill University and Génome Québec Innovation Centre, Montreal, QC H3A 0G1, Canada
| | - Akihiro Fujikawa
- Division of Molecular Neurobiology, National Institute for Basic Biology, Okazaki 444-8787, Japan
| | - Kazuya Kuboyama
- Division of Molecular Neurobiology, National Institute for Basic Biology, Okazaki 444-8787, Japan
| | - Yasushi Takeuchi
- Division of Molecular Neurobiology, National Institute for Basic Biology, Okazaki 444-8787, Japan
| | - Zafar Nawaz
- Cytogenetic and Molecular Cytogenetic Laboratory, Department of Laboratory Medicine and Pathology, Hamad Medical Corporation, P.O. Box 3050, Doha, Qatar
| | - Javad Nadaf
- Department of Human Genetics, Faculty of Medicine, McGill University, Montreal, QC H3A 1B1, Canada; McGill University and Génome Québec Innovation Centre, Montreal, QC H3A 0G1, Canada
| | - Hussein Kamel
- Department of Radiology, Hamad Medical Corporation, P.O. Box 3050, Doha, Qatar
| | - Abu Khadija Kitam
- Cytogenetic and Molecular Cytogenetic Laboratory, Department of Laboratory Medicine and Pathology, Hamad Medical Corporation, P.O. Box 3050, Doha, Qatar
| | - Zaineddin Samiha
- Cytogenetic and Molecular Cytogenetic Laboratory, Department of Laboratory Medicine and Pathology, Hamad Medical Corporation, P.O. Box 3050, Doha, Qatar
| | - Laila Mahmoud
- Section of Clinical and Metabolic Genetics, Department of Pediatrics, Hamad Medical Corporation, P.O. Box 3050, Doha, Qatar
| | - Tawfeg Ben-Omran
- Section of Clinical and Metabolic Genetics, Department of Pediatrics, Hamad Medical Corporation, P.O. Box 3050, Doha, Qatar
| | - Jacek Majewski
- Department of Human Genetics, Faculty of Medicine, McGill University, Montreal, QC H3A 1B1, Canada; McGill University and Génome Québec Innovation Centre, Montreal, QC H3A 0G1, Canada
| | - Masaharu Noda
- Division of Molecular Neurobiology, National Institute for Basic Biology, Okazaki 444-8787, Japan; School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), Okazaki 444-8787, Japan.
| |
Collapse
|
28
|
Beta M, Chitipothu S, Khetan V, Biswas J, Krishnakumar S. Hypermethylation of adenomatosis polyposis coli-2 and its tumor suppressor role in retinoblastoma. Curr Eye Res 2014; 40:719-28. [PMID: 25207834 DOI: 10.3109/02713683.2014.954673] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
PURPOSE Retinoblastoma (RB) is a progressive eye cancer of infancy and childhood. Hypermethylation, epigenetic silencing of genes is one of the key events in tumorigenesis. The purpose of this study is to investigate hypermethylation of adenomatosis polyposis coli homologue, APC-2 and possible interaction of APC-2 with Wnt signaling β-catenin protein in Retinoblastoma. METHODOLOGY Primary RB tumor samples and cell line were used for the study. DNA isolation, bisulfite conversion, methylation specific PCR and DNA sequencing analysis of PCR products were performed to identify CpG islands and methylation in primary RB tumor samples (n = 30). Chemical demethylation and retrieval of APC-2 expression was studied using 5-Azacytidine (5'-AZC). Flow cytometry, immunofluorescence, western blot were performed for APC-2 expression analysis in demethylated Y79 cells. Co-localization study was conducted to understand the interaction between APC-2 and β-catenin. RESULTS APC-2 gene was methylated and down regulated in primary RB tumors. We observed that 70% of RB tumors (21/30) showed positivity with APC-2 methylation. The RB Y79 cells after treatment with demethylating agent 5'-AZC retrieved APC-2 expression, which was confirmed by immunofluorescence and Western blot. Flow cytometry showed APC-2 expression of 29.22% in 5'-AZC treated cells. Co-localization study showed interaction of APC-2 and RB upregulated β-catenin in Y79 cells. CONCLUSION We report that APC-2 gene is hypermethylated in both RB tumor samples and Y79 cells. Reduced APC-2 lead to increased Wnt signaling pathway protein, β-catenin suggesting tumor suppressive role of APC-2 gene.
Collapse
Affiliation(s)
- Madhu Beta
- L & T Ocular Pathology Department, Vision Research Foundation , Chennai, Tamil Nadu , India
| | | | | | | | | |
Collapse
|
29
|
Igarashi M. Proteomic identification of the molecular basis of mammalian CNS growth cones. Neurosci Res 2014; 88:1-15. [PMID: 25066522 DOI: 10.1016/j.neures.2014.07.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 06/13/2014] [Accepted: 07/02/2014] [Indexed: 11/28/2022]
Abstract
The growth cone, which is a unique structure with high motility that forms at the tips of extending axons and dendrites, is crucial to neuronal network formation. Axonal growth of the mammalian CNS is most likely achieved by the complicated coordination of cytoskeletal rearrangement and vesicular trafficking via many proteins. Before recent advances, no methods to identify numerous proteins existed; however, proteomics revolutionarily resolved such problems. In this review, I summarize the profiles of the mammalian growth cone proteins revealed by proteomics as the molecular basis of the growth cone functions, with molecular mapping. These results should be used as a basis for understanding the mechanisms of the complex mammalian CNS developmental process.
Collapse
Affiliation(s)
- Michihiro Igarashi
- Department of Neurochemistry and Molecular Cell Biology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan; Trans-disciplinary Program, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan.
| |
Collapse
|
30
|
Liu G, Dwyer T. Microtubule dynamics in axon guidance. Neurosci Bull 2014; 30:569-83. [PMID: 24968808 DOI: 10.1007/s12264-014-1444-6] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 04/10/2014] [Indexed: 12/18/2022] Open
Abstract
Precise modulation of the cytoskeleton is involved in a variety of cellular processes including cell division, migration, polarity, and adhesion. In developing post-mitotic neurons, extracellular guidance cues not only trigger signaling cascades that act at a distance to indirectly regulate microtubule distribution, and assembly and disassembly in the growth cone, but also directly modulate microtubule stability and dynamics through coupling of guidance receptors with microtubules to control growth-cone turning. Microtubule-associated proteins including classical microtubule-associated proteins and microtubule plus-end tracking proteins are required for modulating microtubule dynamics to influence growth-cone steering. Multiple key signaling components, such as calcium, small GTPases, glycogen synthase kinase-3β, and c-Jun N-terminal kinase, link upstream signal cascades to microtubule stability and dynamics in the growth cone to control axon outgrowth and projection. Understanding the functions and regulation of microtubule dynamics in the growth cone provides new insights into the molecular mechanisms of axon guidance.
Collapse
Affiliation(s)
- Guofa Liu
- Department of Biological Sciences, University of Toledo, Toledo, OH, 43606, USA,
| | | |
Collapse
|
31
|
Abstract
The expression of the gut tumor suppressor gene adenomatous polyposis coli (Apc) and its role in the oligodendroglial lineage are poorly understood. We found that immunoreactive APC is transiently induced in the oligodendroglial lineage during both normal myelination and remyelination following toxin-induced, genetic, or autoimmune demyelination murine models. Using the Cre/loxP system to conditionally ablate APC from the oligodendroglial lineage, we determined that APC enhances proliferation of oligodendroglial progenitor cells (OPCs) and is essential for oligodendrocyte differentiation in a cell-autonomous manner. Biallelic Apc disruption caused translocation of β-catenin into the nucleus and upregulated β-catenin-mediated Wnt signaling in early postnatal but not adult oligodendroglial lineage cells. The results of conditional ablation of Apc or Ctnnb1 (the gene encoding β-catenin) and of simultaneous conditional ablation of Apc and Ctnnb1 revealed that β-catenin is dispensable for postnatal oligodendroglial differentiation, that Apc one-allele deficiency is not sufficient to dysregulate β-catenin-mediated Wnt signaling in oligodendroglial lineage cells, and that APC regulates oligodendrocyte differentiation through β-catenin-independent, as well as β-catenin-dependent, mechanisms. Gene ontology analysis of microarray data suggested that the β-catenin-independent mechanism involves APC regulation of the cytoskeleton, a result compatible with established APC functions in neural precursors and with our observation that Apc-deleted OPCs develop fewer, shorter processes in vivo. Together, our data support the hypothesis that APC regulates oligodendrocyte differentiation through both β-catenin-dependent and additional β-catenin-independent mechanisms.
Collapse
|
32
|
Nishimura Y, Applegate K, Davidson MW, Danuser G, Waterman CM. Automated screening of microtubule growth dynamics identifies MARK2 as a regulator of leading edge microtubules downstream of Rac1 in migrating cells. PLoS One 2012; 7:e41413. [PMID: 22848487 PMCID: PMC3404095 DOI: 10.1371/journal.pone.0041413] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Accepted: 06/21/2012] [Indexed: 12/21/2022] Open
Abstract
Polarized microtubule (MT) growth in the leading edge is critical to directed cell migration, and is mediated by Rac1 GTPase. To find downstream targets of Rac1 that affect MT assembly dynamics, we performed an RNAi screen of 23 MT binding and regulatory factors and identified RNAi treatments that suppressed changes in MT dynamics induced by constitutively activated Rac1. By analyzing fluorescent EB3 dynamics with automated tracking, we found that RNAi treatments targeting p150glued, APC2, spastin, EB1, Op18, or MARK2 blocked Rac1-mediated MT growth in lamellipodia. MARK2 was the only protein whose RNAi targeting additionally suppressed Rac1 effects on MT orientation in lamellipodia, and thus became the focus of further study. We show that GFP-MARK2 rescued effects of MARK2 depletion on MT growth lifetime and orientation, and GFP-MARK2 localized in lamellipodia in a Rac1-activity-dependent manner. In a wound-edge motility assay, MARK2-depleted cells failed to polarize their centrosomes or exhibit oriented MT growth in the leading edge, and displayed defects in directional cell migration. Thus, automated image analysis of MT assembly dynamics identified MARK2 as a target regulated downstream of Rac1 that promotes oriented MT growth in the leading edge to mediate directed cell migration.
Collapse
Affiliation(s)
- Yukako Nishimura
- Cell Biology and Physiology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Kathryn Applegate
- Department of Cell Biology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Michael W. Davidson
- National High Magnetic Field Laboratory, Florida State University, Tallahassee Florida, United States of America
| | - Gaudenz Danuser
- Department of Cell Biology, The Scripps Research Institute, La Jolla, California, United States of America
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Clare M. Waterman
- Cell Biology and Physiology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
33
|
Abstract
Adenomatous polyposis coli 2 (APC2) is a family member of APC and mainly expressed in the nervous system. We previously reported that APC2 plays a critical role in axonal projection through the regulation of microtubule stability. Here, we show that a lack of Apc2 induces severe laminary defects in some regions of the mouse brain, including the cerebral cortex and cerebellum. In vivo BrdU labeling and immunohistochemical analyses with specific markers revealed that the laminary abnormalities are a result of dysregulated neuronal migration by a cell-autonomous mechanism. Using total internal reflection fluorescent microscopy, we found that APC2 is distributed along actin fibers as well as microtubules. Cerebellar granule cells in dissociated cultures and in vivo showed that BDNF-stimulated directional migration is impaired in Apc2-deficient neurons. We revealed that this impairment stems from the dysregulations of Rho family GTPase activation and TrkB localization, which disrupts the formation of BDNF-stimulated F-actin at the leading edge. Thus, APC2 is an essential mediator of the cytoskeletal regulation at leading edges in response to extracellular signals.
Collapse
|
34
|
Dent EW, Gupton SL, Gertler FB. The growth cone cytoskeleton in axon outgrowth and guidance. Cold Spring Harb Perspect Biol 2011; 3:cshperspect.a001800. [PMID: 21106647 DOI: 10.1101/cshperspect.a001800] [Citation(s) in RCA: 421] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Axon outgrowth and guidance to the proper target requires the coordination of filamentous (F)-actin and microtubules (MTs), the dynamic cytoskeletal polymers that promote shape change and locomotion. Over the past two decades, our knowledge of the many guidance cues, receptors, and downstream signaling cascades involved in neuronal outgrowth and guidance has increased dramatically. Less is known, however, about how those cascades of information converge and direct appropriate remodeling and interaction of cytoskeletal polymers, the ultimate effectors of movement and guidance. During development, much of the communication that occurs between environmental guidance cues and the cytoskeleton takes place at the growing tip of the axon, the neuronal growth cone. Several articles on this topic focus on the "input" to the growth cone, the myriad of receptor types, and their corresponding cognate ligands. Others investigate the signaling cascades initiated by receptors and propagated by second messenger pathways (i.e., kinases, phosphatases, GTPases). Ultimately, this plethora of information converges on proteins that associate directly with the actin and microtubule cytoskeletons. The role of these cytoskeletal-associated proteins, as well as the cytoskeleton itself in axon outgrowth and guidance, is the subject of this article.
Collapse
Affiliation(s)
- Erik W Dent
- Department of Anatomy, University of Wisconsin-Madison, 53706, USA
| | | | | |
Collapse
|
35
|
Gouveia SM, Akhmanova A. Cell and Molecular Biology of Microtubule Plus End Tracking Proteins. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2010; 285:1-74. [DOI: 10.1016/b978-0-12-381047-2.00001-3] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|