1
|
Essandoh K, Teuber JP, Brody MJ. Regulation of cardiomyocyte intracellular trafficking and signal transduction by protein palmitoylation. Biochem Soc Trans 2024; 52:41-53. [PMID: 38385554 PMCID: PMC10903464 DOI: 10.1042/bst20221296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/08/2024] [Accepted: 02/12/2024] [Indexed: 02/23/2024]
Abstract
Despite the well-established functions of protein palmitoylation in fundamental cellular processes, the roles of this reversible post-translational lipid modification in cardiomyocyte biology remain poorly studied. Palmitoylation is catalyzed by a family of 23 zinc finger and Asp-His-His-Cys domain-containing S-acyltransferases (zDHHC enzymes) and removed by select thioesterases of the lysophospholipase and α/β-hydroxylase domain (ABHD)-containing families of serine hydrolases. Recently, studies utilizing genetic manipulation of zDHHC enzymes in cardiomyocytes have begun to unveil essential functions for these enzymes in regulating cardiac development, homeostasis, and pathogenesis. Palmitoylation co-ordinates cardiac electrophysiology through direct modulation of ion channels and transporters to impact their trafficking or gating properties as well as indirectly through modification of regulators of channels, transporters, and calcium handling machinery. Not surprisingly, palmitoylation has roles in orchestrating the intracellular trafficking of proteins in cardiomyocytes, but also dynamically fine-tunes cardiomyocyte exocytosis and natriuretic peptide secretion. Palmitoylation has emerged as a potent regulator of intracellular signaling in cardiomyocytes, with recent studies uncovering palmitoylation-dependent regulation of small GTPases through direct modification and sarcolemmal targeting of the small GTPases themselves or by modification of regulators of the GTPase cycle. In addition to dynamic control of G protein signaling, cytosolic DNA is sensed and transduced into an inflammatory transcriptional output through palmitoylation-dependent activation of the cGAS-STING pathway, which has been targeted pharmacologically in preclinical models of heart disease. Further research is needed to fully understand the complex regulatory mechanisms governed by protein palmitoylation in cardiomyocytes and potential emerging therapeutic targets.
Collapse
Affiliation(s)
- Kobina Essandoh
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, U.S.A
| | - James P. Teuber
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, U.S.A
| | - Matthew J. Brody
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, U.S.A
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, U.S.A
| |
Collapse
|
2
|
Akkuş N, Duman TA. First Clinical Report of Two RAB3GAP1 Pathogenic Variant in Warburg Micro Syndrome. J Pediatr Genet 2023; 12:193-198. [PMID: 37575647 PMCID: PMC10421685 DOI: 10.1055/s-0043-1768693] [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: 09/22/2021] [Accepted: 04/10/2023] [Indexed: 08/15/2023]
Abstract
Warburg micro (WARBM) syndrome is an autosomal recessive disease characterized by severe brain and eye abnormalities. Loss-of-function mutations in RAB18, RAB3GAP2, RAB3GAP1, or TBC1D20 can lead to this disease. Here, we present two unrelated WARBM syndrome patients who had an RAB3GAP1 c.559 C > T, (p.Arg187Ter) and c.520 C > T (p.Arg174Ter) homozygous state. Both patients had microcephaly, microphthalmia, microcornea, bilateral congenital cataracts, severe intellectual disability, and congenital hypotonia. Using the method of next-generation sequencing and sanger sequencing, we found two nonsense variations at the splice site in exon 7 of RAB3GAP1 in the WARBM syndrome patients. The mutations were predicted to cause the syndrome due to the early stop codon, and the patients had the WARBM1 syndrome. We present the first clinical report of two different unreported variants with RAB3GAP1 mutation in the literature.
Collapse
Affiliation(s)
- Nejmiye Akkuş
- Department of Medical Genetics, Faculty of Medicine, Tokat Gaziosmanpasa University, Tokat, Türkiye
| | - Tuğba Akın Duman
- Department of Medical Genetics, Haseki Training and Research Hospital, Ministry of Health, Istanbul, Türkiye
| |
Collapse
|
3
|
Ghate PS, Vacharasin JM, Ward JA, Nowling D, Kay V, Cowen MH, Lawlor MK, McCord M, Xu H, Carmona E, Cheon SH, Chukwurah E, Walla M, Lizarraga SB. The Warburg micro syndrome protein RAB3GAP1 modulates neuronal morphogenesis and interacts with axon elongation end ER-Golgi trafficking factors. Neurobiol Dis 2023; 184:106215. [PMID: 37385458 DOI: 10.1016/j.nbd.2023.106215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 05/26/2023] [Accepted: 06/26/2023] [Indexed: 07/01/2023] Open
Abstract
RAB3GAP1 is GTPase activating protein localized to the ER and Golgi compartments. In humans, mutations in RAB3GAP1 are the most common cause of Warburg Micro syndrome, a neurodevelopmental disorder associated with intellectual disability, microcephaly, and agenesis of the corpus callosum. We found that downregulation of RAB3GAP1 leads to a reduction in neurite outgrowth and complexity in human stem cell derived neurons. To further define the cellular function of RAB3GAP1, we sought to identify novel interacting proteins. We used a combination of mass spectrometry, co-immunoprecipitation and colocalization analysis and identified two novel interactors of RAB3GAP1: the axon elongation factor Dedicator of cytokinesis 7 (DOCK7) and the TATA modulatory factor 1 (TMF1) a modulator of Endoplasmic Reticulum (ER) to Golgi trafficking. To define the relationship between RAB3GAP1 and its two novel interactors, we analyzed their localization to different subcellular compartments in neuronal and non-neuronal cells with loss of RAB3GAP1. We find that RAB3GAP1 is important for the sub-cellular localization of TMF1 and DOCK7 across different compartments of the Golgi and endoplasmic reticulum. In addition, we find that loss of function mutations in RAB3GAP1 lead to dysregulation of pathways that are activated in response to the cellular stress like ATF6, MAPK, and PI3-AKT signaling. In summary, our findings suggest a novel role for RAB3GAP1 in neurite outgrowth that could encompass the regulation of proteins that control axon elongation, ER-Golgi trafficking, as well as pathways implicated in response to cellular stress.
Collapse
Affiliation(s)
- Pankaj S Ghate
- Department of Biology and Center for Childhood Neurotherapeutics, University of South Carolina, Columbia, SC, United States of America
| | - Janay M Vacharasin
- Department of Biology and Center for Childhood Neurotherapeutics, University of South Carolina, Columbia, SC, United States of America
| | - Joseph A Ward
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, United States of America; Center for Translational Neuroscience, Brown University, Providence, RI, United states of America
| | - Duncan Nowling
- Department of Biology and Center for Childhood Neurotherapeutics, University of South Carolina, Columbia, SC, United States of America
| | - Valerie Kay
- Department of Biology and Center for Childhood Neurotherapeutics, University of South Carolina, Columbia, SC, United States of America
| | - Mara H Cowen
- Department of Biology and Center for Childhood Neurotherapeutics, University of South Carolina, Columbia, SC, United States of America
| | - Mary-Kate Lawlor
- Department of Biology and Center for Childhood Neurotherapeutics, University of South Carolina, Columbia, SC, United States of America
| | - Mikayla McCord
- Department of Biology and Center for Childhood Neurotherapeutics, University of South Carolina, Columbia, SC, United States of America
| | - Hailey Xu
- Department of Biology and Center for Childhood Neurotherapeutics, University of South Carolina, Columbia, SC, United States of America
| | - Esteban Carmona
- Department of Biology and Center for Childhood Neurotherapeutics, University of South Carolina, Columbia, SC, United States of America
| | - Seon-Hye Cheon
- Department of Biology and Center for Childhood Neurotherapeutics, University of South Carolina, Columbia, SC, United States of America
| | - Evelyn Chukwurah
- Department of Biology and Center for Childhood Neurotherapeutics, University of South Carolina, Columbia, SC, United States of America
| | - Mike Walla
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, United States of America
| | - Sofia B Lizarraga
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, United States of America; Center for Translational Neuroscience, Brown University, Providence, RI, United states of America.
| |
Collapse
|
4
|
Sisley EK, Hale OJ, Hughes JW, Cooper HJ. Tissue Washing Improves Native Ambient Mass Spectrometry Detection of Membrane Proteins Directly from Tissue. J Am Chem Soc 2023; 145:15658-15662. [PMID: 37459360 PMCID: PMC10375469 DOI: 10.1021/jacs.3c03454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2023]
Abstract
Native ambient mass spectrometry enables the in situ analysis of proteins and their complexes directly from tissue, providing both structural and spatial information. Until recently, the approach was applied exclusively to the analysis of soluble proteins; however, there is a drive for new techniques that enable analysis of membrane proteins. Here we demonstrate native ambient mass spectrometry of membrane proteins, including β-barrel and α-helical (single and multipass) integral membrane proteins and membrane-associated proteins incorporating lipid anchors, by integration of a simple washing protocol to remove soluble proteins. Mass spectrometry imaging revealed that washing did not disrupt the spatial distributions of the membrane and membrane-associated proteins. Some delocalization of the remaining soluble proteins was observed.
Collapse
Affiliation(s)
- Emma K Sisley
- School of Biosciences, University of Birmingham, Birmingham B15 2TT, U.K
| | - Oliver J Hale
- School of Biosciences, University of Birmingham, Birmingham B15 2TT, U.K
| | - James W Hughes
- School of Biosciences, University of Birmingham, Birmingham B15 2TT, U.K
| | - Helen J Cooper
- School of Biosciences, University of Birmingham, Birmingham B15 2TT, U.K
| |
Collapse
|
5
|
Essandoh K, Subramani A, Ferro OA, Teuber JP, Koripella S, Brody MJ. zDHHC9 Regulates Cardiomyocyte Rab3a Activity and Atrial Natriuretic Peptide Secretion Through Palmitoylation of Rab3gap1. JACC Basic Transl Sci 2023; 8:518-542. [PMID: 37325411 PMCID: PMC10264568 DOI: 10.1016/j.jacbts.2022.11.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 11/09/2022] [Accepted: 11/15/2022] [Indexed: 02/25/2023]
Abstract
Production and release of natriuretic peptides by the stressed heart reduce cardiac workload by promoting vasodilation, natriuresis, and diuresis, which has been leveraged in the recent development of novel heart-failure pharmacotherapies, yet the mechanisms regulating cardiomyocyte exocytosis and natriuretic peptide release remain ill defined. We found that the Golgi S-acyltransferase zDHHC9 palmitoylates Rab3gap1 resulting in its spatial segregation from Rab3a, elevation of Rab3a-GTP levels, formation of Rab3a-positive peripheral vesicles, and impairment of exocytosis that limits atrial natriuretic peptide release. This novel pathway potentially can be exploited for targeting natriuretic peptide signaling in the treatment of heart failure.
Collapse
Affiliation(s)
- Kobina Essandoh
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Olivia A. Ferro
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan, USA
| | - James P. Teuber
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan, USA
| | - Sribharat Koripella
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan, USA
| | - Matthew J. Brody
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
6
|
Wu S, Fan J, Tang F, Chen L, Zhang X, Xiao D, Li X. The role of RIM in neurotransmitter release: promotion of synaptic vesicle docking, priming, and fusion. Front Neurosci 2023; 17:1123561. [PMID: 37179554 PMCID: PMC10169678 DOI: 10.3389/fnins.2023.1123561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 04/06/2023] [Indexed: 05/15/2023] Open
Abstract
There are many special sites at the end of a synapse called active zones (AZs). Synaptic vesicles (SVs) fuse with presynaptic membranes at these sites, and this fusion is an important step in neurotransmitter release. The cytomatrix in the active zone (CAZ) is made up of proteins such as the regulating synaptic membrane exocytosis protein (RIM), RIM-binding proteins (RIM-BPs), ELKS/CAST, Bassoon/Piccolo, Liprin-α, and Munc13-1. RIM is a scaffold protein that interacts with CAZ proteins and presynaptic functional components to affect the docking, priming, and fusion of SVs. RIM is believed to play an important role in regulating the release of neurotransmitters (NTs). In addition, abnormal expression of RIM has been detected in many diseases, such as retinal diseases, Asperger's syndrome (AS), and degenerative scoliosis. Therefore, we believe that studying the molecular structure of RIM and its role in neurotransmitter release will help to clarify the molecular mechanism of neurotransmitter release and identify targets for the diagnosis and treatment of the aforementioned diseases.
Collapse
Affiliation(s)
- Shanshan Wu
- Emergency Department, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Jiali Fan
- Emergency Department, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Fajuan Tang
- Emergency Department, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Lin Chen
- Emergency Department, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Xiaoyan Zhang
- Emergency Department, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Dongqiong Xiao
- Emergency Department, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Xihong Li
- Emergency Department, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| |
Collapse
|
7
|
Mora-Roldan GA, Galaviz-Hernandez C, Hiebert-Froese J, Hernandez A, Montes L, Duran-Pasten ML, Gazarian K, Zenteno JC. A new missense variant in RAB3GAP2 in a family with muscular dystrophy-short stature and defective autophagy: An expansion of the micro/Martsolf spectrum or a new phenotype? Am J Med Genet A 2022; 188:1972-1978. [PMID: 35274444 DOI: 10.1002/ajmg.a.62723] [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: 10/27/2021] [Revised: 01/27/2022] [Accepted: 02/26/2022] [Indexed: 11/06/2022]
Abstract
We describe a sibling pair of Mennonite origin born from consanguineous parentage with a likely new phenotype of limb-girdle muscular dystrophy, short stature, ptosis, and tracheomalacia. Exome sequencing in the affected subjects identified a novel homozygous RAB3GAP2 missense variant as the potential causal variant. As RAB3GAP2 has been recently shown to be involved in the autophagy process, we analyzed patient-derived fibroblasts by fluorescence microscopy and demonstrated defective autophagic flux under rapamycin and serum starvation conditions when compared with wild-type cells. The phenotype in the siblings described here is distinct from Martsolf and Warburg's micro syndromes, the currently known diseases arising from RAB3GAP2 pathogenic variants. Thus, this work describes a potentially novel recessive phenotype associated with a RAB3GAP2 defect and manifesting as a muscular dystrophy-short stature disorder with no ocular anomalies. Functional analyses indicated defective autophagy in patient-derived fibroblasts, supporting the involvement of RAB3GAP2 in the etiology of this disorder. Our results contribute to a better characterization of the Martsolf/micro spectrum phenotype.
Collapse
Affiliation(s)
- German A Mora-Roldan
- Research Unit, Genetics Department, Institute of Ophthalmology "Conde de Valenciana", Mexico City, Mexico.,Biomedical Research Institute, Department of Genomic Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| | | | | | - Arturo Hernandez
- Institute of Cellular Physiology, Department of Cognitive Neuroscience, National Autonomous University of Mexico, Mexico City, Mexico.,National Laboratory of Channelopathies, National Autonomous University of Mexico, Mexico City, Mexico
| | - Luis Montes
- Research Unit, Genetics Department, Institute of Ophthalmology "Conde de Valenciana", Mexico City, Mexico
| | - Maria L Duran-Pasten
- National Laboratory of Channelopathies, National Autonomous University of Mexico, Mexico City, Mexico
| | - Karlen Gazarian
- Biomedical Research Institute, Department of Genomic Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| | - Juan C Zenteno
- Research Unit, Genetics Department, Institute of Ophthalmology "Conde de Valenciana", Mexico City, Mexico.,Department of Biochemistry, Faculty of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| |
Collapse
|
8
|
Kerkeni N, Kharrat M, Maazoul F, Boudabous H, M’rad R, Trabelsi M. Novel RAB3GAP1 Mutation in the First Tunisian Family With Warburg Micro Syndrome. J Clin Neurol 2022; 18:214-222. [PMID: 35196747 PMCID: PMC8926778 DOI: 10.3988/jcn.2022.18.2.214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 08/24/2021] [Accepted: 08/24/2021] [Indexed: 11/17/2022] Open
Affiliation(s)
- Nesrine Kerkeni
- University of Tunis El Manar, Faculty of Medicine of Tunis, Laboratory of Human Genetics LR99ES10, Tunis, Tunisia
| | - Maher Kharrat
- University of Tunis El Manar, Faculty of Medicine of Tunis, Laboratory of Human Genetics LR99ES10, Tunis, Tunisia
| | - Faouzi Maazoul
- University of Tunis El Manar, Faculty of Medicine of Tunis, Laboratory of Human Genetics LR99ES10, Tunis, Tunisia
- Department of Congenital and Hereditary Diseases, Charles Nicolle Hospital, Tunis, Tunisia
| | - Hela Boudabous
- Department of Paediatrics, Rabta Hospital, Tunis, Tunisia
| | - Ridha M’rad
- University of Tunis El Manar, Faculty of Medicine of Tunis, Laboratory of Human Genetics LR99ES10, Tunis, Tunisia
- Department of Congenital and Hereditary Diseases, Charles Nicolle Hospital, Tunis, Tunisia
| | - Mediha Trabelsi
- University of Tunis El Manar, Faculty of Medicine of Tunis, Laboratory of Human Genetics LR99ES10, Tunis, Tunisia
- Department of Congenital and Hereditary Diseases, Charles Nicolle Hospital, Tunis, Tunisia
| |
Collapse
|
9
|
Global effects of RAB3GAP1 dysexpression on the proteome of mouse cortical neurons. Amino Acids 2021; 53:1339-1350. [PMID: 34363538 DOI: 10.1007/s00726-021-03058-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 07/30/2021] [Indexed: 12/15/2022]
Abstract
Mounting studies have demonstrated that RAB3GAP1 expression is modified in brain diseases with multiple neurobiological functions and processes and acts as a potentially significant target. However, the cellular and molecular events arising from RAB3GAP1 dysexpression are still incompletely understood. In this work, underexpression and overexpression of RAB3GAP1 were first induced into cultured mouse cortical neurons by transfection with lentivirus plasmids. Then we globally explored the effects of RAB3GAP1 dysexpression on the proteome of the neurons through the use of isobaric tag for relative and absolute quantitation (iTRAQ)-based quantitative proteomics with bioinformatics. A total of 364 proteins in the RAB3GAP1-underexpression group and 314 proteins in the RAB3GAP1-overexpression group were identified to be differentially expressed. Subsequent bioinformatics analysis indicated that the proteome functional expression profiles induced by RAB3GAP1 underexpression and overexpression were different, suggesting the potential differences in biological processes and cellular effects. Subsequent intergroup cross-comparison revealed some candidate target proteins regulated directly by RAB3GAP1. Further parallel reaction monitoring (PRM) analysis illustrated that Sub1, Ssrp1, and Top1 proteins might serve as new potentially important linkers in the RAB3GAP1-mediated autophagy pathway in the cortical neurons. Collectively, the current proteomics data furnished new valuable insights to better understand the regulatory molecular mechanism of neuronal RAB3GAP1.
Collapse
|
10
|
Zhang L, Qin Z, Sharmin F, Lin W, Ricke KM, Zasloff MA, Stewart AFR, Chen HH. Tyrosine phosphatase PTP1B impairs presynaptic NMDA receptor-mediated plasticity in a mouse model of Alzheimer's disease. Neurobiol Dis 2021; 156:105402. [PMID: 34044147 DOI: 10.1016/j.nbd.2021.105402] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/29/2021] [Accepted: 05/21/2021] [Indexed: 12/11/2022] Open
Abstract
Mutations in the beta-amyloid protein (APP) cause familial Alzheimer's disease. In hAPP-J20 mice expressing mutant APP, pharmacological inhibition or genetic ablation of the tyrosine phosphatase PTP1B prevents CA3 hippocampus neuron loss and cognitive decline. However, how targeting PTP1B affects the cellular mechanisms underlying these cognitive deficits remains unknown. Changes in synaptic strength at the hippocampus can affect information processing for learning and memory. While prior studies have focused on post-synaptic mechanisms to account for synaptic deficits in Alzheimer's disease models, presynaptic mechanisms may also be affected. Here, using whole cell patch-clamp recording, coefficient of variation (CV) analysis suggested a profound presynaptic deficit in long-term potentiation (LTP) of CA3:CA1 synapses in hAPP-J20 mice. While the membrane-impermeable ionotropic NMDA receptor (NMDAR) blocker norketamine in the post-synaptic recording electrode had no effect on LTP, additional bath application of the ionotropic NMDAR blockers MK801 could replicate the deficit in LTP in wild type mice. In contrast to LTP, the paired-pulse ratio and short-term facilitation (STF) were aberrantly increased in hAPP-J20 mice. These synaptic deficits in hAPP-J20 mice were associated with reduced phosphorylation of NMDAR GluN2B and the synaptic vesicle recycling protein NSF (N-ethylmaleimide sensitive factor). Phosphorylation of both proteins, together with synaptic plasticity and cognitive function, were restored by PTP1B ablation or inhibition by the PTP1B-selective inhibitor Trodusquemine. Taken together, our results indicate that PTP1B impairs presynaptic NMDAR-mediated synaptic plasticity required for spatial learning in a mouse model of Alzheimer's disease. Since Trodusquemine has undergone phase 1/2 clinical trials to treat obesity, it could be repurposed to treat Alzheimer's disease.
Collapse
Affiliation(s)
- Li Zhang
- Neuroscience, Ottawa Hospital Research Institute, Ottawa, ON K1H8M5, Canada; University of Ottawa Brain and Mind Institute, Ottawa, ON K1H8M5, Canada
| | - Zhaohong Qin
- Neuroscience, Ottawa Hospital Research Institute, Ottawa, ON K1H8M5, Canada; University of Ottawa Brain and Mind Institute, Ottawa, ON K1H8M5, Canada
| | - Fariba Sharmin
- Neuroscience, Ottawa Hospital Research Institute, Ottawa, ON K1H8M5, Canada; University of Ottawa Brain and Mind Institute, Ottawa, ON K1H8M5, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Wei Lin
- Neuroscience, Ottawa Hospital Research Institute, Ottawa, ON K1H8M5, Canada; University of Ottawa Brain and Mind Institute, Ottawa, ON K1H8M5, Canada
| | - Konrad M Ricke
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada; University of Ottawa Heart Institute, Ottawa, ON K1Y4W7, Canada
| | - Michael A Zasloff
- Georgetown University School of Medicine, MedStar Georgetown Transplant Institute, Washington, DC, 2007, USA
| | - Alexandre F R Stewart
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada; University of Ottawa Heart Institute, Ottawa, ON K1Y4W7, Canada.
| | - Hsiao-Huei Chen
- Neuroscience, Ottawa Hospital Research Institute, Ottawa, ON K1H8M5, Canada; University of Ottawa Brain and Mind Institute, Ottawa, ON K1H8M5, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; Department of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada.
| |
Collapse
|
11
|
Novel manifestations of Warburg micro syndrome type 1 caused by a new splicing variant of RAB3GAP1: a case report. BMC Neurol 2021; 21:180. [PMID: 33910511 PMCID: PMC8080372 DOI: 10.1186/s12883-021-02204-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 04/15/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The present study aimed to determine the underlying genetic factors causing the possible Warburg micro syndrome (WARBM) phenotype in two Iranian patients. CASE PRESENTATION A 5-year-old female and a 4.5-year-old male were referred due to microcephaly, global developmental delay, and dysmorphic features. After doing neuroimaging and clinical examinations, due to the heterogeneity of neurodevelopmental disorders, we subjected 7 family members to whole-exome sequencing. Three candidate variants were confirmed by Sanger sequencing and allele frequency of each variant was also determined in 300 healthy ethnically matched people using the tetra-primer amplification refractory mutation system-PCR and PCR-restriction fragment length polymorphism. To show the splicing effects, reverse transcription-PCR (RT-PCR) and RT-qPCR were performed, followed by Sanger sequencing. A novel homozygous variant-NM_012233.2: c.151-5 T > G; p.(Gly51IlefsTer15)-in the RAB3GAP1 gene was identified as the most likely disease-causing variant. RT-PCR/RT-qPCR showed that this variant can activate a cryptic site of splicing in intron 3, changing the splicing and gene expression processes. We also identified some novel manifestations in association with WARBM type 1 to touch upon abnormal philtrum, prominent antitragus, downturned corners of the mouth, malaligned teeth, scrotal hypoplasia, low anterior hairline, hypertrichosis of upper back, spastic diplegia to quadriplegia, and cerebral white matter signal changes. CONCLUSIONS Due to the common phenotypes between WARBMs and Martsolf syndrome (MIM: 212720), we suggest using the "RABopathies" term that can in turn cover a broad range of manifestations. This study can per se increase the genotype-phenotype spectrum of WARBM type 1.
Collapse
|
12
|
Environmental Enrichment Enhances Ca v 2.1 Channel-Mediated Presynaptic Plasticity in Hypoxic-Ischemic Encephalopathy. Int J Mol Sci 2021; 22:ijms22073414. [PMID: 33810296 PMCID: PMC8037860 DOI: 10.3390/ijms22073414] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 03/23/2021] [Indexed: 12/16/2022] Open
Abstract
Hypoxic–ischemic encephalopathy (HIE) is a devastating neonatal brain condition caused by lack of oxygen and limited blood flow. Environmental enrichment (EE) is a classic paradigm with a complex stimulation of physical, cognitive, and social components. EE can exert neuroplasticity and neuroprotective effects in immature brains. However, the exact mechanism of EE on the chronic condition of HIE remains unclear. HIE was induced by a permanent ligation of the right carotid artery, followed by an 8% O2 hypoxic condition for 1 h. At 6 weeks of age, HIE mice were randomly assigned to either standard cages or EE cages. In the behavioral assessments, EE mice showed significantly improved motor performances in rotarod tests, ladder walking tests, and hanging wire tests, compared with HIE control mice. EE mice also significantly enhanced cognitive performances in Y-maze tests. Particularly, EE mice showed a significant increase in Cav 2.1 (P/Q type) and presynaptic proteins by molecular assessments, and a significant increase of Cav 2.1 in histological assessments of the cerebral cortex and hippocampus. These results indicate that EE can upregulate the expression of the Cav 2.1 channel and presynaptic proteins related to the synaptic vesicle cycle and neurotransmitter release, which may be responsible for motor and cognitive improvements in HIE.
Collapse
|
13
|
Perić I, Costina V, Gass P, Findeisen P, Filipović D. Hippocampal synaptoproteomic changes of susceptibility and resilience of male rats to chronic social isolation. Brain Res Bull 2020; 166:128-141. [PMID: 33238171 DOI: 10.1016/j.brainresbull.2020.11.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 11/07/2020] [Accepted: 11/18/2020] [Indexed: 12/22/2022]
Abstract
The susceptibility of an individual to chronic social isolation (CSIS) stress may cause major depression (MD) whereby some individuals are resistant to the stress. Recent studies relate MD with altered expression of synaptic proteins in specific brain regions. To explore the neurobiological underpinnings and identify candidate biomarkers of susceptibility or resilience to CSIS, a comparative proteomic approach was used to map hippocampal synaptic protein alterations of rats exposed to 6 weeks of CSIS, an animal model of depression. This model generates two stress-response phenotypes: CSIS-sensitive (depressive-like behaviour) and CSIS-resilience assessed by means of sucrose preference and forced swim tests. Our aim was to characterize the synaptoproteome changes representative of potential long-term changes in protein expression underlying susceptibility or resilience to stress. Proteomic data showed increased expression of glycolytic enzymes, the energy-related mitochondrial proteins, actin cytoskeleton, signalling transduction and synaptic transmission proteins in CSIS-sensitive rats. Protein levels of glutamate-related enzymes such as glutamate dehydrogenase and glutamine synthetase were also increased. CSIS-resilient rats showed similar proteome changes, however with a weaker increase compared to CSIS-sensitive rats. The main difference was observed in the level of protein expression of vesicle-mediated transport proteins. Nonetheless, only few proteins were uniquely up-regulated in the CSIS-resilient rats, whereby Cytochrome b-c1 complex subunit 2, mitochondrial (Uqcrc2) and Voltage-dependent anion-selective channel protein 1 (Vdac1) were uniquely down-regulated. Identified altered activated pathways and potential protein biomarkers may help us better understand the molecular mechanisms underlying synaptic neurotransmission in MD or resilience, crucial for development of new therapeutics.
Collapse
Affiliation(s)
- Ivana Perić
- Molecular Biology and Endocrinology MBE-090, "VINČA", Institute of Nuclear Sciences - National Institute of thе Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Victor Costina
- Institute for Clinical Chemistry, Medical Faculty Mannheim of the University of Heidelberg, University Hospital Mannheim, Mannheim, Germany
| | - Peter Gass
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Peter Findeisen
- Institute for Clinical Chemistry, Medical Faculty Mannheim of the University of Heidelberg, University Hospital Mannheim, Mannheim, Germany
| | - Dragana Filipović
- Molecular Biology and Endocrinology MBE-090, "VINČA", Institute of Nuclear Sciences - National Institute of thе Republic of Serbia, University of Belgrade, Belgrade, Serbia.
| |
Collapse
|
14
|
Öztürk Z, O’Kane CJ, Pérez-Moreno JJ. Axonal Endoplasmic Reticulum Dynamics and Its Roles in Neurodegeneration. Front Neurosci 2020; 14:48. [PMID: 32116502 PMCID: PMC7025499 DOI: 10.3389/fnins.2020.00048] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 01/13/2020] [Indexed: 12/13/2022] Open
Abstract
The physical continuity of axons over long cellular distances poses challenges for their maintenance. One organelle that faces this challenge is endoplasmic reticulum (ER); unlike other intracellular organelles, this forms a physically continuous network throughout the cell, with a single membrane and a single lumen. In axons, ER is mainly smooth, forming a tubular network with occasional sheets or cisternae and low amounts of rough ER. It has many potential roles: lipid biosynthesis, glucose homeostasis, a Ca2+ store, protein export, and contacting and regulating other organelles. This tubular network structure is determined by ER-shaping proteins, mutations in some of which are causative for neurodegenerative disorders such as hereditary spastic paraplegia (HSP). While axonal ER shares many features with the tubular ER network in other contexts, these features must be adapted to the long and narrow dimensions of axons. ER appears to be physically continuous throughout axons, over distances that are enormous on a subcellular scale. It is therefore a potential channel for long-distance or regional communication within neurons, independent of action potentials or physical transport of cargos, but involving its physiological roles such as Ca2+ or organelle homeostasis. Despite its apparent stability, axonal ER is highly dynamic, showing features like anterograde and retrograde transport, potentially reflecting continuous fusion and breakage of the network. Here we discuss the transport processes that must contribute to this dynamic behavior of ER. We also discuss the model that these processes underpin a homeostatic process that ensures both enough ER to maintain continuity of the network and repair breaks in it, but not too much ER that might disrupt local cellular physiology. Finally, we discuss how failure of ER organization in axons could lead to axon degenerative diseases, and how a requirement for ER continuity could make distal axons most susceptible to degeneration in conditions that disrupt ER continuity.
Collapse
Affiliation(s)
| | - Cahir J. O’Kane
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| | | |
Collapse
|
15
|
Wang Z, Zhao H, Yuan C, Zhao D, Sun Y, Wang X, Zhang H. The RBG-1-RBG-2 complex modulates autophagy activity by regulating lysosomal biogenesis and function in C. elegans. J Cell Sci 2019; 132:jcs.234195. [PMID: 31444285 DOI: 10.1242/jcs.234195] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 08/19/2019] [Indexed: 01/05/2023] Open
Abstract
Vici syndrome is a severe and progressive multisystem disease caused by mutations in the EPG5 gene. In patient tissues and animal models, loss of EPG5 function is associated with defective autophagy caused by accumulation of non-degradative autolysosomes, but very little is known about the mechanism underlying this cellular phenotype. Here, we demonstrate that loss of function of the RBG-1-RBG-2 complex ameliorates the autophagy defect in C. elegans epg-5 mutants. The suppression effect is independent of the complex's activity as a RAB-3 GAP and a RAB-18 GEF. Loss of rbg-1 activity promotes lysosomal biogenesis and function, and also suppresses the accumulation of non-functional autolysosomes in epg-5 mutants. The mobility of late endosome- and lysosome-associated RAB-7 is reduced in epg-5 mutants, and this defect is rescued by simultaneous loss of function of rbg-1 Expression of the GDP-bound form of RAB-7 also promotes lysosomal biogenesis and suppresses the autophagy defect in epg-5 mutants. Our study reveals that the RBG-1-RBG-2 complex acts by modulating the dynamics of membrane-associated RAB-7 to regulate lysosomal biogenesis, and provides insights into the pathogenesis of Vici syndrome.
Collapse
Affiliation(s)
- Zhaoyu Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Hongyu Zhao
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Chongzhen Yuan
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Dongfeng Zhao
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yanan Sun
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiaochen Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hong Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China .,College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
16
|
Wang Z, Hou L, Wang D. Effects of exercise-induced fatigue on the morphology of asymmetric synapse and synaptic protein levels in rat striatum. Neurochem Int 2019; 129:104476. [PMID: 31145967 DOI: 10.1016/j.neuint.2019.104476] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 04/20/2019] [Accepted: 05/26/2019] [Indexed: 12/12/2022]
Abstract
Corticostriatal synaptic plasticity is considered to be a cellular basis for somatic motor regulation and motor skill learning. Changes in synaptic transmission efficiency underlie functional plasticity, while structural plasticity involves changes in the ultrastructure of the synapse and the levels of synaptic proteins. Exercise-induced fatigue may impair corticostriatal synaptic plasticity, and this impairment may be an important mechanism for exercise-induced fatigue. However, prior research focused mainly on functional plasticity such that the structural plasticity was not well understood. Because corticostriatal synapses are typical asymmetric synapses, here we have used transmission electron microscopy to examine the changes of asymmetry synaptic ultrastructure in rat striatum before and after repetitive exercise-induced fatigue; we have also used western blotting to detect the levels of synaptic active region protein Munc 13, RIM1 and synaptic vesicle protein Rab3A and postsynaptic density PSD-95 protein in rat striatum before and after exercise-induced fatigue. The results showed that the ultrastructure of asymmetry corticostriatal synapses and synaptic protein levels in the striatum of rats were abnormally changed after repetitive exercise-induced fatigue. These abnormal changes in synaptic ultrastructure and related protein levels may be the structural basis for the corticostriatal plasticity impairment after exercise-induced fatigue.
Collapse
Affiliation(s)
- Zhifeng Wang
- Department of Physical Education, Xi'an Polytechnic University, Xi'an, Shanxi, 710048, China
| | - Lijuan Hou
- Physical Education and Sports College, Beijing Normal University, Beijing, 100875, China
| | - Dongmei Wang
- College of Sports Medicine and Rehabilitation, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, Shandong, 271000, China.
| |
Collapse
|
17
|
Nomiyama R, Emoto M, Fukuda N, Matsui K, Kondo M, Sakane A, Sasaki T, Tanizawa Y. Protein kinase C iota facilitates insulin-induced glucose transport by phosphorylation of soluble nSF attachment protein receptor regulator (SNARE) double C2 domain protein b. J Diabetes Investig 2019; 10:591-601. [PMID: 30369065 PMCID: PMC6497606 DOI: 10.1111/jdi.12965] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 09/25/2018] [Accepted: 10/11/2018] [Indexed: 12/24/2022] Open
Abstract
AIMS/INTRODUCTION Double C2 domain protein b (DOC2b), one of the synaptotagmins, has been shown to translocate to the plasma membrane, and to initiate membrane-fusion processes of vesicles containing glucose transporter 4 proteins on insulin stimulation. However, the mechanism by which DOC2b is regulated remains unclear. Herein, we identified the upstream regulatory factors of DOC2b in insulin signal transduction. We also examined the role of DOC2b on systemic homeostasis using DOC2b knockout (KO) mice. MATERIALS AND METHODS We first identified DOC2b binding proteins by immunoprecipitation and mutagenesis experiments. Then, DOC2b KO mice were generated by disrupting the first exon of the DOC2b gene. In addition to the histological examination, glucose metabolism was assessed by measuring parameters on glucose/insulin tolerance tests. Insulin-stimulated glucose uptake was also measured using isolated soleus muscle and epididymal adipose tissue. RESULTS We identified an isoform of atypical protein kinase C (protein kinase C iota) that can bind to DOC2b and phosphorylates one of the serine residues of DOC2b (S34). This phosphorylation is essential for DOC2b translocation. DOC2b KO mice showed insulin resistance and impaired oral glucose tolerance on insulin and glucose tolerance tests, respectively. Insulin-stimulated glucose uptake was impaired in isolated soleus muscle and epididymal adipose tissues from DOC2b KO mice. CONCLUSIONS We propose a novel insulin signaling mechanism by which protein kinase C iota phosphorylates DOC2b, leading to glucose transporter 4 vesicle translocation, fusion and facilitation of glucose uptake in response to insulin. The present results also showed DOC2b to play important roles in systemic glucose homeostasis.
Collapse
Affiliation(s)
- Ryuta Nomiyama
- Division of Endocrinology, Metabolism, Hematological Sciences and TherapeuticsYamaguchi University Graduate School of MedicineUbeJapan
| | - Masahiro Emoto
- Division of Endocrinology, Metabolism, Hematological Sciences and TherapeuticsYamaguchi University Graduate School of MedicineUbeJapan
- Emoto ClinicUbeJapan
| | - Naofumi Fukuda
- Division of Endocrinology, Metabolism, Hematological Sciences and TherapeuticsYamaguchi University Graduate School of MedicineUbeJapan
| | - Kumiko Matsui
- Division of Endocrinology, Metabolism, Hematological Sciences and TherapeuticsYamaguchi University Graduate School of MedicineUbeJapan
| | - Manabu Kondo
- Division of Endocrinology, Metabolism, Hematological Sciences and TherapeuticsYamaguchi University Graduate School of MedicineUbeJapan
| | - Ayuko Sakane
- Department of BiochemistryTokushima University Graduate School of Medical SciencesTokushimaJapan
| | - Takuya Sasaki
- Department of BiochemistryTokushima University Graduate School of Medical SciencesTokushimaJapan
| | - Yukio Tanizawa
- Division of Endocrinology, Metabolism, Hematological Sciences and TherapeuticsYamaguchi University Graduate School of MedicineUbeJapan
| |
Collapse
|
18
|
Koparir A, Karatas OF, Yilmaz SS, Suer I, Ozer B, Yuceturk B, Ozen M. Revealing the functions of novel mutations in RAB3GAP1
in Martsolf and Warburg micro syndromes. Am J Med Genet A 2019; 179:579-587. [DOI: 10.1002/ajmg.a.61065] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 01/17/2019] [Accepted: 01/17/2019] [Indexed: 12/30/2022]
Affiliation(s)
- Asuman Koparir
- Department of Internal Medicine, Division of Medical Genetics; Istanbul University; Istanbul Turkey
| | - Omer Faruk Karatas
- Molecular Biology and Genetics Department; Erzurum Technical University; Erzurum Turkey
| | - Seda Salman Yilmaz
- Department of Medical Genetics; Istanbul University, Cerrahpasa Medical School; Istanbul Turkey
| | - Ilknur Suer
- Department of Internal Medicine, Division of Medical Genetics; Istanbul University; Istanbul Turkey
| | - Bugra Ozer
- Advanced Genomics and Bioinformatics Research Center; The Scientific and Technological Research Council of Turkey (TUBITAK-BILGEM); Kocaeli Turkey
| | - Betul Yuceturk
- Advanced Genomics and Bioinformatics Research Center; The Scientific and Technological Research Council of Turkey (TUBITAK-BILGEM); Kocaeli Turkey
| | - Mustafa Ozen
- Department of Medical Genetics; Istanbul University, Cerrahpasa Medical School; Istanbul Turkey
- Department of Pathology and Immunology; Baylor College of Medicine; Houston Texas
| |
Collapse
|
19
|
Limanaqi F, Biagioni F, Gambardella S, Ryskalin L, Fornai F. Interdependency Between Autophagy and Synaptic Vesicle Trafficking: Implications for Dopamine Release. Front Mol Neurosci 2018; 11:299. [PMID: 30186112 PMCID: PMC6110820 DOI: 10.3389/fnmol.2018.00299] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 08/06/2018] [Indexed: 12/16/2022] Open
Abstract
Autophagy (ATG) and the Ubiquitin Proteasome (UP) are the main clearing systems of eukaryotic cells, in that being ultimately involved in degrading damaged and potentially harmful cytoplasmic substrates. Emerging evidence implicates that, in addition to their classic catalytic function in the cytosol, autophagy and the proteasome act as modulators of neurotransmission, inasmuch as they orchestrate degradation and turnover of synaptic vesicles (SVs) and related proteins. These findings are now defining a novel synaptic scenario, where clearing systems and secretory pathways may be considered as a single system, which senses alterations in quality and distribution (in time, amount and place) of both synaptic proteins and neurotransmitters. In line with this, in the present manuscript we focus on evidence showing that, a dysregulation of secretory and trafficking pathways is quite constant in the presence of an impairment of autophagy-lysosomal machinery, which eventually precipitates synaptic dysfunction. Such a dual effect appears not to be just incidental but it rather represents the natural evolution of archaic cell compartments. While discussing these issues, we pose a special emphasis on the role of autophagy upon dopamine (DA) neurotransmission, which is early affected in several neurological and psychiatric disorders. In detail, we discuss how autophagy is engaged not only in removing potentially dangerous proteins, which can interfere with the mechanisms of DA release, but also the fate of synaptic DA vesicles thus surveilling DA neurotransmission. These concepts contribute to shed light on early mechanisms underlying intersection of autophagy with DA-related synaptic disorders.
Collapse
Affiliation(s)
- Fiona Limanaqi
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | | | | | - Larisa Ryskalin
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Francesco Fornai
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy.,IRCCS Neuromed, Pozzilli, Italy
| |
Collapse
|
20
|
Genetics and mechanisms leading to human cortical malformations. Semin Cell Dev Biol 2018; 76:33-75. [DOI: 10.1016/j.semcdb.2017.09.031] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 09/21/2017] [Accepted: 09/21/2017] [Indexed: 02/06/2023]
|
21
|
Gallo LI, Dalghi MG, Clayton DR, Ruiz WG, Khandelwal P, Apodaca G. RAB27B requirement for stretch-induced exocytosis in bladder umbrella cells. Am J Physiol Cell Physiol 2017; 314:C349-C365. [PMID: 29167152 DOI: 10.1152/ajpcell.00218.2017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Umbrella cells, which must maintain a tight barrier, modulate their apical surface area during bladder filling by exocytosis of an abundant, subapical pool of discoidal- and/or fusiform-shaped vesicles (DFVs). Despite the importance of this trafficking event for bladder function, the pathways that promote DFV exocytosis remain to be identified. We previously showed that DFV exocytosis depends in part on a RAB11A-RAB8A-MYO5B network, but RAB27B is also reported to be associated with DFVs, and knockout mice lacking RAB27B have fewer DFVs. However, the RAB27B requirements for DFV exocytosis and the relationship between RAB27B and the other umbrella cell-expressed RABs remains unclear. Using a whole bladder preparation, we observed that filling-induced exocytosis of human growth hormone-loaded DFVs was significantly inhibited when RAB27B expression was downregulated using shRNA. RAB27A was also expressed in rat urothelium; however, RAB27A-specific shRNAs did not inhibit exocytosis, and the combination of RAB27A and RAB27B shRNAs did not significantly affect DFV exocytosis more than treatment with RAB27B shRNA alone. RAB27B and RAB11A showed a small degree of overlap when quantified using Squassh segmentation software, and expression of dominant-active or dominant-negative mutants of RAB11A or RAB8A, or expression of a RAB11A-specific shRNA, had no significant effect on the size, number, or intensity of RAB27B-positive DFVs. Likewise, treatment with RAB27B-specific shRNA had no effect on RAB11A-positive DFV parameters. We conclude that RAB27B, but not RAB27A, regulates DFV exocytosis in bladder umbrella cells in a manner that may be parallel to the previously described RAB11A-RAB8A-MYO5B pathway.
Collapse
Affiliation(s)
- Luciana I Gallo
- Department of Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Marianela G Dalghi
- Department of Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Dennis R Clayton
- Department of Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Wily G Ruiz
- Department of Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Puneet Khandelwal
- Department of Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Gerard Apodaca
- Department of Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania.,Department of Cell Biology, University of Pittsburgh , Pittsburgh, Pennsylvania
| |
Collapse
|
22
|
Abstract
Macroautophagy is a conserved degradative pathway in which a double-membrane compartment sequesters cytoplasmic cargo and delivers the contents to lysosomes for degradation. Efficient formation and maturation of autophagic vesicles, so-called phagophores that are precursors to autophagosomes, and their subsequent trafficking to lysosomes relies on the activity of small RAB GTPases, which are essential factors of cellular vesicle transport systems. The activity of RAB GTPases is coordinated by upstream factors, which include guanine nucleotide exchange factors (RAB GEFs) and RAB GTPase activating proteins (RAB GAPs). A role in macroautophagy regulation for different TRE2-BUB2-CDC16 (TBC) domain-containing RAB GAPs has been established. Recently, however, a positive modulation of macroautophagy has also been demonstrated for the TBC domain-free RAB3GAP1/2, adding to the family of RAB GAPs that coordinate macroautophagy and additional cellular trafficking pathways.
Collapse
Affiliation(s)
- Andreas Kern
- a Institute for Pathobiochemistry; University Medical Center of the Johannes Gutenberg University ; Mainz , Germany
| | - Ivan Dikic
- b Buchmann Institute for Molecular Life Sciences; Goethe University Frankfurt ; Frankfurt am Main , Germany
| | - Christian Behl
- a Institute for Pathobiochemistry; University Medical Center of the Johannes Gutenberg University ; Mainz , Germany
| |
Collapse
|
23
|
Fassio A, Fadda M, Benfenati F. Molecular Machines Determining the Fate of Endocytosed Synaptic Vesicles in Nerve Terminals. Front Synaptic Neurosci 2016; 8:10. [PMID: 27242505 PMCID: PMC4863888 DOI: 10.3389/fnsyn.2016.00010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 04/18/2016] [Indexed: 11/28/2022] Open
Abstract
The cycle of a synaptic vesicle (SV) within the nerve terminal is a step-by-step journey with the final goal of ensuring the proper synaptic strength under changing environmental conditions. The SV cycle is a precisely regulated membrane traffic event in cells and, because of this, a plethora of membrane-bound and cytosolic proteins are devoted to assist SVs in each step of the journey. The cycling fate of endocytosed SVs determines both the availability for subsequent rounds of release and the lifetime of SVs in the terminal and is therefore crucial for synaptic function and plasticity. Molecular players that determine the destiny of SVs in nerve terminals after a round of exo-endocytosis are largely unknown. Here we review the functional role in SV fate of phosphorylation/dephosphorylation of SV proteins and of small GTPases acting on membrane trafficking at the synapse, as they are emerging as key molecules in determining the recycling route of SVs within the nerve terminal. In particular, we focus on: (i) the cyclin-dependent kinase-5 (cdk5) and calcineurin (CN) control of the recycling pool of SVs; (ii) the role of small GTPases of the Rab and ADP-ribosylation factor (Arf) families in defining the route followed by SV in their nerve terminal cycle. These regulatory proteins together with their synaptic regulators and effectors, are molecular nanomachines mediating homeostatic responses in synaptic plasticity and potential targets of drugs modulating the efficiency of synaptic transmission.
Collapse
Affiliation(s)
- Anna Fassio
- Department of Experimental Medicine, University of GenoaGenoa, Italy; Center of Synaptic Neuroscience and Technology, Istituto Italiano di TecnologiaGenova, Italy
| | - Manuela Fadda
- Department of Experimental Medicine, University of Genoa Genoa, Italy
| | - Fabio Benfenati
- Department of Experimental Medicine, University of GenoaGenoa, Italy; Center of Synaptic Neuroscience and Technology, Istituto Italiano di TecnologiaGenova, Italy
| |
Collapse
|
24
|
Handley MT, Carpanini SM, Mali GR, Sidjanin DJ, Aligianis IA, Jackson IJ, FitzPatrick DR. Warburg Micro syndrome is caused by RAB18 deficiency or dysregulation. Open Biol 2016; 5:150047. [PMID: 26063829 PMCID: PMC4632505 DOI: 10.1098/rsob.150047] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
RAB18, RAB3GAP1, RAB3GAP2 and TBC1D20 are each mutated in Warburg Micro syndrome, a rare autosomal recessive multisystem disorder. RAB3GAP1 and RAB3GAP2 form a binary ‘RAB3GAP’ complex that functions as a guanine-nucleotide exchange factor (GEF) for RAB18, whereas TBC1D20 shows modest RAB18 GTPase-activating (GAP) activity in vitro. Here, we show that in the absence of functional RAB3GAP or TBC1D20, the level, localization and dynamics of cellular RAB18 is altered. In cell lines where TBC1D20 is absent from the endoplasmic reticulum (ER), RAB18 becomes more stably ER-associated and less cytosolic than in control cells. These data suggest that RAB18 is a physiological substrate of TBC1D20 and contribute to a model in which a Rab-GAP can be essential for the activity of a target Rab. Together with previous reports, this indicates that Warburg Micro syndrome can be caused directly by loss of RAB18, or indirectly through loss of RAB18 regulators RAB3GAP or TBC1D20.
Collapse
Affiliation(s)
- Mark T Handley
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Sarah M Carpanini
- Division of Neurobiology, The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK
| | - Girish R Mali
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Duska J Sidjanin
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Irene A Aligianis
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Ian J Jackson
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - David R FitzPatrick
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| |
Collapse
|
25
|
Novel RAB3GAP1 compound heterozygous mutations in Japanese siblings with Warburg Micro syndrome. Brain Dev 2016; 38:337-40. [PMID: 26421802 DOI: 10.1016/j.braindev.2015.09.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 09/11/2015] [Accepted: 09/12/2015] [Indexed: 11/22/2022]
Abstract
BACKGROUND Warburg Micro syndrome (WARBM) is a rare autosomal recessive disease characterized by postnatal growth retardation, microcephaly, severely delayed motor and intellectual development, microcornea, congenital cataracts, optic atrophy, and hypogonadism. While WARBM is a genetically heterogeneous condition, RAB3GAP1 mutations account for ∼40% of WARBM patients, and 69 different mutations of various types (nonsense, missense, frameshift, and splice site mutations) have been identified to date. PATIENTS Japanese siblings (a 7 years 3 months old male and a 2 years 1month old female) were found to have WARBM-compatible phenotypes. Direct sequencing of RAB3GAP1 revealed novel compound heterozygous mutations in the siblings: a paternally inherited missense mutation (c.560G>C; p.Arg187Pro) in exon 7 and a maternally derived nonsense mutation (c.1009C>T; p.Arg337Ter) in exon 12. CONCLUSION The siblings had WARBM caused by novel mutations in RAB3GAP1. Since molecular diagnosis permits adequate genetic counseling and appropriate management for predicted complications such as adequate sex steroid supplementation therapy for hypogonadism, in addition to standard supportive therapies for developmental delay and visual dysfunction, we recommend molecular studies for this rare condition.
Collapse
|
26
|
Spang N, Feldmann A, Huesmann H, Bekbulat F, Schmitt V, Hiebel C, Koziollek-Drechsler I, Clement AM, Moosmann B, Jung J, Behrends C, Dikic I, Kern A, Behl C. RAB3GAP1 and RAB3GAP2 modulate basal and rapamycin-induced autophagy. Autophagy 2015; 10:2297-309. [PMID: 25495476 PMCID: PMC4502700 DOI: 10.4161/15548627.2014.994359] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Macroautophagy is a degradative pathway that sequesters and transports cytosolic cargo in autophagosomes to lysosomes, and its deterioration affects intracellular proteostasis. Membrane dynamics accompanying autophagy are mostly elusive and depend on trafficking processes. RAB GTPase activating proteins (RABGAPs) are important factors for the coordination of cellular vesicle transport systems, and several TBC (TRE2-BUB2-CDC16) domain-containing RABGAPs are associated with autophagy. Employing C. elegans and human primary fibroblasts, we show that RAB3GAP1 and RAB3GAP2, which are components of the TBC domain-free RAB3GAP complex, influence protein aggregation and affect autophagy at basal and rapamycin-induced conditions. Correlating the activity of RAB3GAP1/2 with ATG3 and ATG16L1 and analyzing ATG5 punctate structures, we illustrate that the RAB3GAPs modulate autophagosomal biogenesis. Significant levels of RAB3GAP1/2 colocalize with members of the Atg8 family at lipid droplets, and their autophagy modulatory activity depends on the GTPase-activating activity of RAB3GAP1 but is independent of the RAB GTPase RAB3. Moreover, we analyzed RAB3GAP1/2 in relation to the previously reported suppressive autophagy modulators FEZ1 and FEZ2 and demonstrate that both reciprocally regulate autophagy. In conclusion, we identify RAB3GAP1/2 as novel conserved factors of the autophagy and proteostasis network.
Collapse
Key Words
- ATG, autophagy-related
- ATG16L1
- ATG3
- BSA, bovine serum albumin
- Bafi, bafilomycin A1
- C. elegans, Caenorhabditis elegans
- CALCOCO2, calcium binding and coiled-coil domain 2
- DAPI, 4’, 6-diamidino-2-phenylindole
- DMSO, dimethyl sulfoxide
- DPH, 1, 6-diphenyl-1, 3, 5-hexatriene
- FEZ, fasciculation and elongation protein zeta
- FEZ1
- FEZ2
- GABARAP, GABA(A) receptor-associated protein
- GEF, guanine nucleotide exchange factor
- GFP, green fluorescent protein
- MAP1LC3, microtubule-associated protein 1 light chain 3
- NBR1, neighbor of BRCA1 gene 1
- PBS, phosphate-buffered saline
- PE, phosphatidylethanolamine
- RAB3GAP1
- RAB3GAP2
- RABGAP, RAB GTPase activating protein
- SQSTM1, sequestosome 1
- TBC domain, TRE2-BUB2-CDC16 domain
- autophagy
- eV, empty vector
- lipid droplets
- proteostasis
- siRNA, small interfering RNA
Collapse
Affiliation(s)
- Natalie Spang
- a Institute for Pathobiochemistry ; University Medical Center of the Johannes Gutenberg University ; Mainz , Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Mhlanga-Mutangadura T, Johnson GS, Schnabel RD, Taylor JF, Johnson GC, Katz ML, Shelton GD, Lever TE, Giuliano E, Granger N, Shomper J, O'Brien DP. A mutation in the Warburg syndrome gene, RAB3GAP1, causes a similar syndrome with polyneuropathy and neuronal vacuolation in Black Russian Terrier dogs. Neurobiol Dis 2015; 86:75-85. [PMID: 26607784 DOI: 10.1016/j.nbd.2015.11.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2015] [Revised: 11/13/2015] [Accepted: 11/18/2015] [Indexed: 11/30/2022] Open
Abstract
An autosomal recessive disease of Black Russian Terriers was previously described as a juvenile-onset, laryngeal paralysis and polyneuropathy similar to Charcot Marie Tooth disease in humans. We found that in addition to an axonal neuropathy, affected dogs exhibit microphthalmia, cataracts, and miotic pupils. On histopathology, affected dogs exhibit a spongiform encephalopathy characterized by accumulations of abnormal, membrane-bound vacuoles of various sizes in neuronal cell bodies, axons and adrenal cells. DNA from an individual dog with this polyneuropathy with ocular abnormalities and neuronal vacuolation (POANV) was used to generate a whole genome sequence which contained a homozygous RAB3GAP1:c.743delC mutation that was absent from 73 control canine whole genome sequences. An additional 12 Black Russian Terriers with POANV were RAB3GAP1:c.743delC homozygotes. DNA samples from 249 Black Russian Terriers with no known signs of POANV were either heterozygotes or homozygous for the reference allele. Mutations in human RAB3GAP1 cause Warburg micro syndrome (WARBM), a severe developmental disorder characterized by abnormalities of the eye, genitals and nervous system including a predominantly axonal peripheral neuropathy. RAB3GAP1 encodes the catalytic subunit of a GTPase activator protein and guanine exchange factor for Rab3 and Rab18 respectively. Rab proteins are involved in membrane trafficking in the endoplasmic reticulum, axonal transport, autophagy and synaptic transmission. The neuronal vacuolation and membranous inclusions and vacuoles in axons seen in this canine disorder likely reflect alterations of these processes. Thus, this canine disease could serve as a model for WARBM and provide insight into its pathogenesis and treatment.
Collapse
Affiliation(s)
| | - Gary S Johnson
- Department of Veterinary Pathobiology, University of Missouri, Columbia, USA
| | - Robert D Schnabel
- Division of Animal Sciences, University of Missouri, Columbia, USA; Informatics Institute, University of Missouri, Columbia, USA
| | - Jeremy F Taylor
- Division of Animal Sciences, University of Missouri, Columbia, USA
| | - Gayle C Johnson
- Department of Veterinary Pathobiology, University of Missouri, Columbia, USA
| | - Martin L Katz
- Mason Eye Institute, University of Missouri, Columbia, USA
| | - G Diane Shelton
- Department of Pathology, University of California, San Diego, La Jolla, USA
| | - Teresa E Lever
- Department of Otolaryngology, University of Missouri, Columbia, USA
| | - Elizabeth Giuliano
- Department of Veterinary Medicine & Surgery, University of Missouri, Columbia, USA
| | - Nicolas Granger
- School of Veterinary Sciences, University of Bristol, Langford House, Langford, Somerset BS40 5DU, UK
| | - Jeremy Shomper
- Department of Veterinary Medicine & Surgery, University of Missouri, Columbia, USA
| | - Dennis P O'Brien
- Department of Veterinary Medicine & Surgery, University of Missouri, Columbia, USA.
| |
Collapse
|
28
|
A RAB3GAP1 SINE Insertion in Alaskan Huskies with Polyneuropathy, Ocular Abnormalities, and Neuronal Vacuolation (POANV) Resembling Human Warburg Micro Syndrome 1 (WARBM1). G3-GENES GENOMES GENETICS 2015; 6:255-62. [PMID: 26596647 PMCID: PMC4751546 DOI: 10.1534/g3.115.022707] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
We observed a hereditary phenotype in Alaskan Huskies that was characterized by polyneuropathy with ocular abnormalities and neuronal vacuolation (POANV). The affected dogs developed a progressive severe ataxia, which led to euthanasia between 8 and 16 months of age. The pedigrees were consistent with a monogenic autosomal recessive inheritance. We localized the causative genetic defect to a 4 Mb interval on chromosome 19 by a combined linkage and homozygosity mapping approach. Whole genome sequencing of one affected dog, an obligate carrier, and an unrelated control revealed a 218-bp SINE insertion into exon 7 of the RAB3GAP1 gene. The SINE insertion was perfectly associated with the disease phenotype in a cohort of 43 Alaskan Huskies, and it was absent from 541 control dogs of diverse other breeds. The SINE insertion induced aberrant splicing and led to a transcript with a greatly altered exon 7. RAB3GAP1 loss-of-function variants in humans cause Warburg Micro Syndrome 1 (WARBM1), which is characterized by additional developmental defects compared to canine POANV, whereas Rab3gap1-deficient mice have a much milder phenotype than either humans or dogs. Thus, the RAB3GAP1 mutant Alaskan Huskies provide an interesting intermediate phenotype that may help to better understand the function of RAB3GAP1 in development. Furthermore, the identification of the presumed causative genetic variant will enable genetic testing to avoid the nonintentional breeding of affected dogs.
Collapse
|
29
|
Zhou J, Liu Z, Yu J, Han X, Fan S, Shao W, Chen J, Qiao R, Xie P. Quantitative Proteomic Analysis Reveals Molecular Adaptations in the Hippocampal Synaptic Active Zone of Chronic Mild Stress-Unsusceptible Rats. Int J Neuropsychopharmacol 2015; 19:pyv100. [PMID: 26364272 PMCID: PMC4772275 DOI: 10.1093/ijnp/pyv100] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 08/31/2015] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND While stressful events are recognized as an important cause of major depressive disorder, some individuals exposed to life stressors maintain normal psychological functioning. The molecular mechanism(s) underlying this phenomenon remain unclear. Abnormal transmission and plasticity of hippocampal synapses have been implied to play a key role in the pathoetiology of major depressive disorder. METHODS A chronic mild stress protocol was applied to separate susceptible and unsusceptible rat subpopulations. Proteomic analysis using an isobaric tag for relative and absolute quantitation coupled with tandem mass spectrometry was performed to identify differential proteins in enriched hippocampal synaptic junction preparations. RESULTS A total of 4318 proteins were quantified, and 89 membrane proteins were present in differential amounts. Of these, SynaptomeDB identified 81 (91%) having a synapse-specific localization. The unbiased profiles identified several candidate proteins within the synaptic junction that may be associated with stress vulnerability or insusceptibility. Subsequent functional categorization revealed that protein systems particularly involved in membrane trafficking at the synaptic active zone exhibited a positive strain as potential molecular adaptations in the unsusceptible rats. Moreover, through STRING and immunoblotting analysis, membrane-associated GTP-bound Rab3a and Munc18-1 appear to coregulate syntaxin-1/SNAP25/VAMP2 assembly at the hippocampal presynaptic active zone of unsusceptible rats, facilitating SNARE-mediated membrane fusion and neurotransmitter release, and may be part of a stress-protection mechanism in actively maintaining an emotional homeostasis. CONCLUSIONS The present results support the concept that there is a range of potential protein adaptations in the hippocampal synaptic active zone of unsusceptible rats, revealing new investigative targets that may contribute to a better understanding of stress insusceptibility.
Collapse
Affiliation(s)
- Jian Zhou
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China (Drs Zhou, Liu, Yu, Han, Fan, Shao, Chen, Qiao, and Xie); Chongqing Key Laboratory of Neurobiology, Chongqing, China (Drs Zhou, Liu, Yu, Han, Fan, Shao, Chen, Qiao, and Xie); Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China (Drs Liu, Han, Fan, Shao, and Xie)
| | - Zhao Liu
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China (Drs Zhou, Liu, Yu, Han, Fan, Shao, Chen, Qiao, and Xie); Chongqing Key Laboratory of Neurobiology, Chongqing, China (Drs Zhou, Liu, Yu, Han, Fan, Shao, Chen, Qiao, and Xie); Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China (Drs Liu, Han, Fan, Shao, and Xie)
| | - Jia Yu
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China (Drs Zhou, Liu, Yu, Han, Fan, Shao, Chen, Qiao, and Xie); Chongqing Key Laboratory of Neurobiology, Chongqing, China (Drs Zhou, Liu, Yu, Han, Fan, Shao, Chen, Qiao, and Xie); Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China (Drs Liu, Han, Fan, Shao, and Xie)
| | - Xin Han
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China (Drs Zhou, Liu, Yu, Han, Fan, Shao, Chen, Qiao, and Xie); Chongqing Key Laboratory of Neurobiology, Chongqing, China (Drs Zhou, Liu, Yu, Han, Fan, Shao, Chen, Qiao, and Xie); Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China (Drs Liu, Han, Fan, Shao, and Xie)
| | - Songhua Fan
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China (Drs Zhou, Liu, Yu, Han, Fan, Shao, Chen, Qiao, and Xie); Chongqing Key Laboratory of Neurobiology, Chongqing, China (Drs Zhou, Liu, Yu, Han, Fan, Shao, Chen, Qiao, and Xie); Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China (Drs Liu, Han, Fan, Shao, and Xie)
| | - Weihua Shao
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China (Drs Zhou, Liu, Yu, Han, Fan, Shao, Chen, Qiao, and Xie); Chongqing Key Laboratory of Neurobiology, Chongqing, China (Drs Zhou, Liu, Yu, Han, Fan, Shao, Chen, Qiao, and Xie); Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China (Drs Liu, Han, Fan, Shao, and Xie)
| | - Jianjun Chen
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China (Drs Zhou, Liu, Yu, Han, Fan, Shao, Chen, Qiao, and Xie); Chongqing Key Laboratory of Neurobiology, Chongqing, China (Drs Zhou, Liu, Yu, Han, Fan, Shao, Chen, Qiao, and Xie); Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China (Drs Liu, Han, Fan, Shao, and Xie)
| | - Rui Qiao
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China (Drs Zhou, Liu, Yu, Han, Fan, Shao, Chen, Qiao, and Xie); Chongqing Key Laboratory of Neurobiology, Chongqing, China (Drs Zhou, Liu, Yu, Han, Fan, Shao, Chen, Qiao, and Xie); Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China (Drs Liu, Han, Fan, Shao, and Xie)
| | - Peng Xie
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China (Drs Zhou, Liu, Yu, Han, Fan, Shao, Chen, Qiao, and Xie); Chongqing Key Laboratory of Neurobiology, Chongqing, China (Drs Zhou, Liu, Yu, Han, Fan, Shao, Chen, Qiao, and Xie); Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China (Drs Liu, Han, Fan, Shao, and Xie).
| |
Collapse
|
30
|
Mutational Analysis of Rab3 Function for Controlling Active Zone Protein Composition at the Drosophila Neuromuscular Junction. PLoS One 2015; 10:e0136938. [PMID: 26317909 PMCID: PMC4552854 DOI: 10.1371/journal.pone.0136938] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 08/11/2015] [Indexed: 11/26/2022] Open
Abstract
At synapses, the release of neurotransmitter is regulated by molecular machinery that aggregates at specialized presynaptic release sites termed active zones. The complement of active zone proteins at each site is a determinant of release efficacy and can be remodeled to alter synapse function. The small GTPase Rab3 was previously identified as playing a novel role that controls the distribution of active zone proteins to individual release sites at the Drosophila neuromuscular junction. Rab3 has been extensively studied for its role in the synaptic vesicle cycle; however, the mechanism by which Rab3 controls active zone development remains unknown. To explore this mechanism, we conducted a mutational analysis to determine the molecular and structural requirements of Rab3 function at Drosophila synapses. We find that GTP-binding is required for Rab3 to traffick to synapses and distribute active zone components across release sites. Conversely, the hydrolytic activity of Rab3 is unnecessary for this function. Through a structure-function analysis we identify specific residues within the effector-binding switch regions that are required for Rab3 function and determine that membrane attachment is essential. Our findings suggest that Rab3 controls the distribution of active zone components via a vesicle docking mechanism that is consistent with standard Rab protein function.
Collapse
|
31
|
Namba T, Funahashi Y, Nakamuta S, Xu C, Takano T, Kaibuchi K. Extracellular and Intracellular Signaling for Neuronal Polarity. Physiol Rev 2015; 95:995-1024. [PMID: 26133936 DOI: 10.1152/physrev.00025.2014] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Neurons are one of the highly polarized cells in the body. One of the fundamental issues in neuroscience is how neurons establish their polarity; therefore, this issue fascinates many scientists. Cultured neurons are useful tools for analyzing the mechanisms of neuronal polarization, and indeed, most of the molecules important in their polarization were identified using culture systems. However, we now know that the process of neuronal polarization in vivo differs in some respects from that in cultured neurons. One of the major differences is their surrounding microenvironment; neurons in vivo can be influenced by extrinsic factors from the microenvironment. Therefore, a major question remains: How are neurons polarized in vivo? Here, we begin by reviewing the process of neuronal polarization in culture conditions and in vivo. We also survey the molecular mechanisms underlying neuronal polarization. Finally, we introduce the theoretical basis of neuronal polarization and the possible involvement of neuronal polarity in disease and traumatic brain injury.
Collapse
Affiliation(s)
- Takashi Namba
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yasuhiro Funahashi
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shinichi Nakamuta
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Chundi Xu
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tetsuya Takano
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kozo Kaibuchi
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
32
|
Cheng CY, Wu JC, Tsai JW, Nian FS, Wu PC, Kao LS, Fann MJ, Tsai SJ, Liou YJ, Tai CY, Hong CJ. ENU mutagenesis identifies mice modeling Warburg Micro Syndrome with sensory axon degeneration caused by a deletion in Rab18. Exp Neurol 2015; 267:143-51. [PMID: 25779931 DOI: 10.1016/j.expneurol.2015.03.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Accepted: 03/05/2015] [Indexed: 11/18/2022]
Abstract
Mutations in the gene of RAB18, a member of Ras superfamily of small G-proteins, cause Warburg Micro Syndrome (WARBM) which is characterized by defective neurodevelopmental and ophthalmological phenotypes. Despite loss of Rab18 had been reported to induce disruption of the endoplasmic reticulum structure and neuronal cytoskeleton organization, parts of the pathogenic mechanism caused by RAB18 mutation remain unclear. From the N-ethyl-N-nitrosourea (ENU)-induced mutagenesis library, we identified a mouse line whose Rab18 was knocked out. This Rab18(-/-) mouse exhibited stomping gait, smaller testis and eyes, mimicking several features of WARBM. Rab18(-/-) mice were obviously less sensitive to pain and touch than WT mice. Histological examinations on Rab18(-/-) mice revealed progressive axonal degeneration in the optic nerves, dorsal column of the spinal cord and sensory roots of the spinal nerves while the motor roots were spared. All the behavioral and pathological changes that resulted from abnormalities in the sensory axons were prevented by introducing an extra copy of Rab18 transgene in Rab18(-/-) mice. Our results reveal that sensory axonal degeneration is the primary cause of stomping gait and progressive weakness of the hind limbs in Rab18(-/-) mice, and optic nerve degeneration should be the major pathology of progressive optic atrophy in children with WARBM. Our results indicate that the sensory nervous system is more vulnerable to Rab18 deficiency and WARBM is not only a neurodevelopmental but also neurodegenerative disease.
Collapse
Affiliation(s)
- Chih-Ya Cheng
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Jaw-Ching Wu
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan; Institute of Clinical Medicine and Cancer Research Center, National Yang-Ming University, Taipei, Taiwan; Department of Medical Research and Education, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Jin-Wu Tsai
- Institute of Brain Science, National Yang-Ming University, Taipei, Taiwan; Brain Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Fang-Shin Nian
- Institute of Brain Science, National Yang-Ming University, Taipei, Taiwan
| | - Pei-Chun Wu
- Brain Research Center, National Yang-Ming University, Taipei, Taiwan; Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Lung-Sen Kao
- Brain Research Center, National Yang-Ming University, Taipei, Taiwan; Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Ming-Ji Fann
- Brain Research Center, National Yang-Ming University, Taipei, Taiwan; Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Shih-Jen Tsai
- Division of Psychiatry, School of Medicine, National Yang-Ming University, Taipei, Taiwan; Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ying-Jay Liou
- Division of Psychiatry, School of Medicine, National Yang-Ming University, Taipei, Taiwan; Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chin-Yin Tai
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei, Taiwan
| | - Chen-Jee Hong
- Institute of Brain Science, National Yang-Ming University, Taipei, Taiwan; Division of Psychiatry, School of Medicine, National Yang-Ming University, Taipei, Taiwan; Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan.
| |
Collapse
|
33
|
Park AK, Liegel RP, Ronchetti A, Ebert AD, Geurts A, Sidjanin DJ. Targeted disruption of Tbc1d20 with zinc-finger nucleases causes cataracts and testicular abnormalities in mice. BMC Genet 2014; 15:135. [PMID: 25476608 PMCID: PMC4266191 DOI: 10.1186/s12863-014-0135-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 11/24/2014] [Indexed: 12/27/2022] Open
Abstract
Background Loss-of-function mutations in TBC1D20 cause Warburg Micro syndrome 4 (WARBM4), which is an autosomal recessive syndromic disorder characterized by eye, brain, and genital abnormalities. Blind sterile (bs) mice carry a Tbc1d20-null mutation and exhibit cataracts and testicular phenotypes similar to those observed in WARBM4 patients. In addition to TBC1D20, mutations in RAB3GAP1, RAB3GAP2 and RAB18 cause WARBM1-3 respectively. However, regardless of which gene harbors the causative mutation, all individuals affected with WARBM exhibit indistinguishable clinical presentations. In contrast, bs, Rab3gap1-/-, and Rab18-/- mice exhibit distinct phenotypes; this phenotypic variability of WARBM mice was previously attributed to potential compensatory mechanisms. Rab3gap1-/- and Rab18-/- mice were genetically engineered using standard approaches, whereas the Tbc1d20 mutation in the bs mice arose spontaneously. There is the possibility that another unidentified mutation within the bs linkage disequilibrium may be contributing to the bs phenotypes and thus contributing to the phenotypic variability in WARBM mice. The goal of this study was to establish the phenotypic consequences in mice caused by the disruption of the Tbc1d20 gene. Results The zinc finger nuclease (ZFN) mediated genomic editing generated a Tbc1d20 c.[418_426del] deletion encoding a putative TBC1D20-ZFN protein with an in-frame p.[H140_Y143del] deletion within the highly conserved TBC domain. The evaluation of Tbc1d20ZFN/ZFN eyes identified severe cataracts and thickened pupillary sphincter muscle. Tbc1d20ZFN/ZFN males are infertile and the analysis of the seminiferous tubules identified disrupted acrosomal development. The compound heterozygote Tbc1d20ZFN/bs mice, generated from an allelic bs/+ X Tbc1d20ZFN/+ cross, exhibited cataracts and aberrant acrosomal development indicating a failure to complement. Conclusions Our findings show that the disruption of Tbc1d20 in mice results in cataracts and aberrant acrosomal formation, thus establishing bs and Tbc1d20ZFN/ZFN as allelic variants. Although the WARBM molecular disease etiology remains unclear, both the bs and Tbc1d20ZFN/ZFN mice are excellent model organisms for future studies to establish TBC1D20-mediated molecular and cellular functions.
Collapse
Affiliation(s)
- Anna Kyunglim Park
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank, Milwaukee, WI, 53226, USA.
| | - Ryan P Liegel
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank, Milwaukee, WI, 53226, USA.
| | - Adam Ronchetti
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank, Milwaukee, WI, 53226, USA.
| | - Allison D Ebert
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank, Milwaukee, WI, 53226, USA.
| | - Aron Geurts
- Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank, Milwaukee, WI, 53226, USA. .,Human and Molecular Genetics Center, Medical College of Wisconsin, 8701 Watertown Plank, Milwaukee, WI, 53226, USA.
| | - Duska J Sidjanin
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank, Milwaukee, WI, 53226, USA. .,Human and Molecular Genetics Center, Medical College of Wisconsin, 8701 Watertown Plank, Milwaukee, WI, 53226, USA.
| |
Collapse
|
34
|
Tata B, Huijbregts L, Jacquier S, Csaba Z, Genin E, Meyer V, Leka S, Dupont J, Charles P, Chevenne D, Carel JC, Léger J, de Roux N. Haploinsufficiency of Dmxl2, encoding a synaptic protein, causes infertility associated with a loss of GnRH neurons in mouse. PLoS Biol 2014; 12:e1001952. [PMID: 25248098 PMCID: PMC4172557 DOI: 10.1371/journal.pbio.1001952] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 08/12/2014] [Indexed: 12/20/2022] Open
Abstract
Characterization of the genetic defects causing gonadotropic deficiency has made a major contribution to elucidation of the fundamental role of Kisspeptins and Neurokinin B in puberty onset and reproduction. The absence of puberty may also reveal neurodevelopmental disorders caused by molecular defects in various cellular pathways. Investigations of these neurodevelopmental disorders may provide information about the neuronal processes controlling puberty onset and reproductive capacity. We describe here a new syndrome observed in three brothers, which involves gonadotropic axis deficiency, central hypothyroidism, peripheral demyelinating sensorimotor polyneuropathy, mental retardation, and profound hypoglycemia, progressing to nonautoimmune insulin-dependent diabetes mellitus. High-throughput sequencing revealed a homozygous in-frame deletion of 15 nucleotides in DMXL2 in all three affected patients. This homozygous deletion was associated with lower DMXL2 mRNA levels in the blood lymphocytes of the patients. DMXL2 encodes the synaptic protein rabconnectin-3α, which has been identified as a putative scaffold protein for Rab3-GAP and Rab3-GEP, two regulators of the GTPase Rab3a. We found that rabconnectin-3α was expressed in exocytosis vesicles in gonadotropin-releasing hormone (GnRH) axonal extremities in the median eminence of the hypothalamus. It was also specifically expressed in cells expressing luteinizing hormone (LH) and follicle-stimulating hormone (FSH) within the pituitary. The conditional heterozygous deletion of Dmxl2 from mouse neurons delayed puberty and resulted in very low fertility. This reproductive phenotype was associated with a lower number of GnRH neurons in the hypothalamus of adult mice. Finally, Dmxl2 knockdown in an insulin-secreting cell line showed that rabconnectin-3α controlled the constitutive and glucose-induced secretion of insulin. In conclusion, this study shows that low levels of DMXL2 expression cause a complex neurological phenotype, with abnormal glucose metabolism and gonadotropic axis deficiency due to a loss of GnRH neurons. Our findings identify rabconectin-3α as a key controller of neuronal and endocrine homeostatic processes.
Collapse
Affiliation(s)
- Brooke Tata
- Inserm, U1141, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Hôpital Robert Debré, Paris, France
| | - Lukas Huijbregts
- Inserm, U1141, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Hôpital Robert Debré, Paris, France
| | | | | | | | | | | | - Joelle Dupont
- INRA, UMR85 Physiologie de la Reproduction et des Comportements, Nouzilly, France
| | - Perrine Charles
- Genetics Department and Inserm US975, Université Pierre et Marie Curie, Hôpital la Pitié-Salpêtrière, Paris, France
| | - Didier Chevenne
- AP-HP, Laboratoire de Biochimie, Hôpital Robert Debré, Paris, France
| | - Jean-Claude Carel
- Inserm, U1141, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Hôpital Robert Debré, Paris, France
- AP-HP, Service d'Endocrinologie Diabétologie Pédiatrique et Centre de Référence des Maladies Endocriniennes Rares de la Croissance, Hôpital Robert Debré, Paris, France
| | - Juliane Léger
- Inserm, U1141, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Hôpital Robert Debré, Paris, France
- AP-HP, Service d'Endocrinologie Diabétologie Pédiatrique et Centre de Référence des Maladies Endocriniennes Rares de la Croissance, Hôpital Robert Debré, Paris, France
| | - Nicolas de Roux
- Inserm, U1141, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Hôpital Robert Debré, Paris, France
- AP-HP, Laboratoire de Biochimie, Hôpital Robert Debré, Paris, France
- * E-mail:
| |
Collapse
|
35
|
Cazares VA, Subramani A, Saldate JJ, Hoerauf W, Stuenkel EL. Distinct actions of Rab3 and Rab27 GTPases on late stages of exocytosis of insulin. Traffic 2014; 15:997-1015. [PMID: 24909540 DOI: 10.1111/tra.12182] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 06/04/2014] [Accepted: 06/04/2014] [Indexed: 12/16/2022]
Abstract
Rab GTPases associated with insulin-containing secretory granules (SGs) are key in targeting, docking and assembly of molecular complexes governing pancreatic β-cell exocytosis. Four Rab3 isoforms along with Rab27A are associated with insulin granules, yet elucidation of the distinct roles of these Rab families on exocytosis remains unclear. To define specific actions of these Rab families we employ Rab3GAP and/or EPI64A GTPase-activating protein overexpression in β-cells from wild-type or Ashen mice to selectively transit the entire Rab3 family or Rab27A to a GDP-bound state. Ashen mice carry a spontaneous mutation that eliminates Rab27A expression. Using membrane capacitance measurements we find that GTP/GDP nucleotide cycling of Rab27A is essential for generation of the functionally defined immediately releasable pool (IRP) and central to regulating the size of the readily releasable pool (RRP). By comparison, nucleotide cycling of Rab3 GTPases, but not of Rab27A, is essential for a kinetically rapid filling of the RRP with SGs. Aside from these distinct functions, Rab3 and Rab27A GTPases demonstrate considerable functional overlap in building the readily releasable granule pool. Hence, while Rab3 and Rab27A cooperate to generate release-ready SGs in β-cells, they also direct unique kinetic and functional properties of the exocytotic pathway.
Collapse
Affiliation(s)
- Victor A Cazares
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA; Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, 48109, USA
| | | | | | | | | |
Collapse
|
36
|
Gerondopoulos A, Bastos RN, Yoshimura SI, Anderson R, Carpanini S, Aligianis I, Handley MT, Barr FA. Rab18 and a Rab18 GEF complex are required for normal ER structure. ACTA ACUST UNITED AC 2014; 205:707-20. [PMID: 24891604 PMCID: PMC4050724 DOI: 10.1083/jcb.201403026] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The ancestral Rab GTPase Rab18 and both subunits of the Rab3GAP complex are mutated in the human neurological and developmental disorder Warburg Micro syndrome. Here, we demonstrate that the Rab3GAP complex is a specific Rab18 guanine nucleotide exchange factor (GEF). The Rab3GAP complex localizes to the endoplasmic reticulum (ER) and is necessary for ER targeting of Rab18. It is also sufficient to promote membrane recruitment of Rab18. Disease-associated point mutations of conserved residues in either the Rab3GAP1 (T18P and E24V) or Rab3GAP2 (R426C) subunits result in loss of the Rab18 GEF and membrane-targeting activities. Supporting the view that Rab18 activity is important for ER structure, in the absence of either Rab3GAP subunit or Rab18 function, ER tubular networks marked by reticulon 4 were disrupted, and ER sheets defined by CLIMP-63 spread out into the cell periphery. Micro syndrome is therefore a disease characterized by direct loss of Rab18 function or loss of Rab18 activation at the ER by its GEF Rab3GAP.
Collapse
Affiliation(s)
| | | | - Shin-Ichiro Yoshimura
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Rachel Anderson
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, England, UK
| | - Sarah Carpanini
- Division of Neurobiology, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh EH25 9RG, Scotland, UK
| | - Irene Aligianis
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, Scotland, UK
| | - Mark T Handley
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, Scotland, UK
| | - Francis A Barr
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, England, UK
| |
Collapse
|
37
|
Carpanini SM, McKie L, Thomson D, Wright AK, Gordon SL, Roche SL, Handley MT, Morrison H, Brownstein D, Wishart TM, Cousin MA, Gillingwater TH, Aligianis IA, Jackson IJ. A novel mouse model of Warburg Micro syndrome reveals roles for RAB18 in eye development and organisation of the neuronal cytoskeleton. Dis Model Mech 2014; 7:711-22. [PMID: 24764192 PMCID: PMC4036478 DOI: 10.1242/dmm.015222] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Mutations in RAB18 have been shown to cause the heterogeneous autosomal recessive disorder Warburg Micro syndrome (WARBM). Individuals with WARBM present with a range of clinical symptoms, including ocular and neurological abnormalities. However, the underlying cellular and molecular pathogenesis of the disorder remains unclear, largely owing to the lack of any robust animal models that phenocopy both the ocular and neurological features of the disease. We report here the generation and characterisation of a novel Rab18-mutant mouse model of WARBM. Rab18-mutant mice are viable and fertile. They present with congenital nuclear cataracts and atonic pupils, recapitulating the characteristic ocular features that are associated with WARBM. Additionally, Rab18-mutant cells exhibit an increase in lipid droplet size following treatment with oleic acid. Lipid droplet abnormalities are a characteristic feature of cells taken from WARBM individuals, as well as cells taken from individuals with other neurodegenerative conditions. Neurological dysfunction is also apparent in Rab18-mutant mice, including progressive weakness of the hind limbs. We show that the neurological defects are, most likely, not caused by gross perturbations in synaptic vesicle recycling in the central or peripheral nervous system. Rather, loss of Rab18 is associated with widespread disruption of the neuronal cytoskeleton, including abnormal accumulations of neurofilament and microtubule proteins in synaptic terminals, and gross disorganisation of the cytoskeleton in peripheral nerves. Global proteomic profiling of peripheral nerves in Rab18-mutant mice reveals significant alterations in several core molecular pathways that regulate cytoskeletal dynamics in neurons. The apparent similarities between the WARBM phenotype and the phenotype that we describe here indicate that the Rab18-mutant mouse provides an important platform for investigation of the disease pathogenesis and therapeutic interventions.
Collapse
Affiliation(s)
- Sarah M. Carpanini
- MRC Human Genetics Unit, Institute for Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK.,The Roslin Institute, University of Edinburgh, Edinburgh EH25 9RG, UK
| | - Lisa McKie
- MRC Human Genetics Unit, Institute for Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Derek Thomson
- Euan MacDonald Centre for Motor Neurone Disease Research and Centre for Integrative Physiology, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Ann K. Wright
- Euan MacDonald Centre for Motor Neurone Disease Research and Centre for Integrative Physiology, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Sarah L. Gordon
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Sarah L. Roche
- Euan MacDonald Centre for Motor Neurone Disease Research and Centre for Integrative Physiology, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Mark T. Handley
- MRC Human Genetics Unit, Institute for Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Harris Morrison
- MRC Human Genetics Unit, Institute for Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - David Brownstein
- Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Thomas M. Wishart
- The Roslin Institute, University of Edinburgh, Edinburgh EH25 9RG, UK.,Euan MacDonald Centre for Motor Neurone Disease Research and Centre for Integrative Physiology, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Michael A. Cousin
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Thomas H. Gillingwater
- Euan MacDonald Centre for Motor Neurone Disease Research and Centre for Integrative Physiology, University of Edinburgh, Edinburgh EH8 9XD, UK.,Authors for correspondence (; )
| | - Irene A. Aligianis
- MRC Human Genetics Unit, Institute for Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Ian J. Jackson
- MRC Human Genetics Unit, Institute for Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK.,The Roslin Institute, University of Edinburgh, Edinburgh EH25 9RG, UK.,Authors for correspondence (; )
| |
Collapse
|
38
|
Stutterd CA, Leventer RJ. Polymicrogyria: a common and heterogeneous malformation of cortical development. AMERICAN JOURNAL OF MEDICAL GENETICS PART C-SEMINARS IN MEDICAL GENETICS 2014; 166C:227-39. [PMID: 24888723 DOI: 10.1002/ajmg.c.31399] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Polymicrogyria (PMG) is one of the most common malformations of cortical development. It is characterized by overfolding of the cerebral cortex and abnormal cortical layering. It is a highly heterogeneous malformation with variable clinical and imaging features, pathological findings, and etiologies. It may occur as an isolated cortical malformation, or in association with other malformations within the brain or body as part of a multiple congenital anomaly syndrome. Polymicrogyria shows variable topographic patterns with the bilateral perisylvian pattern being most common. Schizencephaly is a subtype of PMG in which the overfolded cortex lines full-thickness clefts connecting the subarachnoid space with the cerebral ventricles. Both genetic and non-genetic causes of PMG have been identified. Non-genetic causes include congenital cytomegalovirus infection and in utero ischemia. Genetic causes include metabolic conditions such as peroxisomal disorders and the 22q11.2 and 1p36 continguous gene deletion syndromes. Mutations in over 30 genes have been found in association with PMG, especially mutations in the tubulin family of genes. Mutations in the (PI3K)-AKT pathway have been found in association PMG and megalencephaly. Despite recent genetic advances, the mechanisms by which polymicrogyric cortex forms and causes of the majority of cases remain unknown, making diagnostic and prenatal testing and genetic counseling challenging. This review summarizes the clinical, imaging, pathologic, and etiologic features of PMG, highlighting recent genetic advances.
Collapse
|
39
|
Liegel R, Handley M, Ronchetti A, Brown S, Langemeyer L, Linford A, Chang B, Morris-Rosendahl D, Carpanini S, Posmyk R, Harthill V, Sheridan E, Abdel-Salam G, Terhal P, Faravelli F, Accorsi P, Giordano L, Pinelli L, Hartmann B, Ebert A, Barr F, Aligianis I, Sidjanin D. Loss-of-function mutations in TBC1D20 cause cataracts and male infertility in blind sterile mice and Warburg micro syndrome in humans. Am J Hum Genet 2013; 93:1001-14. [PMID: 24239381 DOI: 10.1016/j.ajhg.2013.10.011] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Revised: 09/20/2013] [Accepted: 10/11/2013] [Indexed: 12/22/2022] Open
Abstract
blind sterile (bs) is a spontaneous autosomal-recessive mouse mutation discovered more than 30 years ago. Phenotypically, bs mice exhibit nuclear cataracts and male infertility; genetic analyses assigned the bs locus to mouse chromosome 2. In this study, we first positionally cloned the bs locus and identified a putative causative mutation in the Tbc1d20 gene. Functional analysis established the mouse TBC1D20 protein as a GTPase-activating protein (GAP) for RAB1 and RAB2, and bs as a TBC1D20 loss-of-function mutation. Evaluation of bs mouse embryonic fibroblasts (mEFs) identified enlarged Golgi morphology and aberrant lipid droplet (LD) formation. Based on the function of TBC1D20 as a RABGAP and the bs cataract and testicular phenotypes, we hypothesized that mutations in TBC1D20 may contribute to Warburg micro syndrome (WARBM); WARBM constitutes a spectrum of disorders characterized by eye, brain, and endocrine abnormalities caused by mutations in RAB3GAP1, RAB3GAP2, and RAB18. Sequence analysis of a cohort of 77 families affected by WARBM identified five distinct TBC1D20 loss-of-function mutations, thereby establishing these mutations as causative of WARBM. Evaluation of human fibroblasts deficient in TBC1D20 function identified aberrant LDs similar to those identified in the bs mEFs. Additionally, our results show that human fibroblasts deficient in RAB18 and RAB3GAP1 function also exhibit aberrant LD formation. These findings collectively indicate that a defect in LD formation/metabolism may be a common cellular abnormality associated with WARBM, although it remains unclear whether abnormalities in LD metabolism are contributing to WARBM disease pathology.
Collapse
|
40
|
Giorgini F, Steinert JR. Rab11 as a modulator of synaptic transmission. Commun Integr Biol 2013; 6:e26807. [PMID: 24563714 PMCID: PMC3922788 DOI: 10.4161/cib.26807] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Revised: 10/12/2013] [Indexed: 01/07/2023] Open
Abstract
Many neurodegenerative disorders are characterized by synaptic dysfunction preceding general neuronal loss and subsequent cognitive or behavioral anomalies. Much recent research has been aimed at understanding the early underlying processes leading to dysfunction at the synapse, as this knowledge would likely inform interventions that could potentially slow progression and delay onset of disease. We have recently reported that synaptic dysfunction in a Drosophila melanogaster model of Huntington's disease (HD) can be prevented by enhanced neuronal expression of Rab11, a Rab family GTPase involved in endosomal recycling, which complements studies that have found disrupted Rab11 activity in several models of this disorder. Indeed, inhibition of Rab11 function in fibroblasts of HD patients has been observed to perturb vesicle formation from recycling endosomes. Therefore, our study investigated a potential role of Rab11 in synaptic dysfunction prior to the onset of HD symptoms, with the aim of finding a possible early intervention to disease progression. We found that Rab11 ameliorates synaptic dysfunction due to expression of mutant huntingtin-the causative protein in HD-by normalizing synaptic vesicle size, which consequently ameliorates locomotor deficits in Drosophila larvae. Here we further consider these results and the implications this work has on potential therapeutic intervention in HD and other neurodegenerative disorders.
Collapse
|
41
|
Handley MT, Morris-Rosendahl DJ, Brown S, Macdonald F, Hardy C, Bem D, Carpanini SM, Borck G, Martorell L, Izzi C, Faravelli F, Accorsi P, Pinelli L, Basel-Vanagaite L, Peretz G, Abdel-Salam GMH, Zaki MS, Jansen A, Mowat D, Glass I, Stewart H, Mancini G, Lederer D, Roscioli T, Giuliano F, Plomp AS, Rolfs A, Graham JM, Seemanova E, Poo P, García-Cazorla A, Edery P, Jackson IJ, Maher ER, Aligianis IA. Mutation spectrum in RAB3GAP1, RAB3GAP2, and RAB18 and genotype-phenotype correlations in warburg micro syndrome and Martsolf syndrome. Hum Mutat 2013; 34:686-96. [PMID: 23420520 DOI: 10.1002/humu.22296] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Accepted: 02/07/2013] [Indexed: 01/10/2023]
Abstract
Warburg Micro syndrome and Martsolf syndrome (MS) are heterogeneous autosomal-recessive developmental disorders characterized by brain, eye, and endocrine abnormalities. Causative biallelic germline mutations have been identified in RAB3GAP1, RAB3GAP2, or RAB18, each of which encode proteins involved in membrane trafficking. This report provides an up to date overview of all known disease variants identified in 29 previously published families and 52 new families. One-hundred and forty-four Micro and nine Martsolf families were investigated, identifying mutations in RAB3GAP1 in 41% of cases, mutations in RAB3GAP2 in 7% of cases, and mutations in RAB18 in 5% of cases. These are listed in Leiden Open source Variation Databases, which was created by us for all three genes. Genotype-phenotype correlations for these genes have now established that the clinical phenotypes in Micro syndrome and MS represent a phenotypic continuum related to the nature and severity of the mutations present in the disease genes, with more deleterious mutations causing Micro syndrome and milder mutations causing MS. RAB18 has not yet been linked to the RAB3 pathways, but mutations in all three genes cause an indistinguishable phenotype, making it likely that there is some overlap. There is considerable genetic heterogeneity for these disorders and further gene identification will help delineate these pathways.
Collapse
Affiliation(s)
- Mark T Handley
- MRC Human Genetics Unit, Medical Research Council and Institute of Genetics and Molecular Medicine, University of Edinburgh, Scotland, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Frank CA. Homeostatic plasticity at the Drosophila neuromuscular junction. Neuropharmacology 2013; 78:63-74. [PMID: 23806804 DOI: 10.1016/j.neuropharm.2013.06.015] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Revised: 05/31/2013] [Accepted: 06/11/2013] [Indexed: 02/07/2023]
Abstract
In biology, homeostasis refers to how cells maintain appropriate levels of activity. This concept underlies a balancing act in the nervous system. Synapses require flexibility (i.e. plasticity) to adjust to environmental challenges. Yet there must also exist regulatory mechanisms that constrain activity within appropriate physiological ranges. An abundance of evidence suggests that homeostatic regulation is critical in this regard. In recent years, important progress has been made toward identifying molecules and signaling processes required for homeostatic forms of neuroplasticity. The Drosophila melanogaster third instar larval neuromuscular junction (NMJ) has been an important experimental system in this effort. Drosophila neuroscientists combine genetics, pharmacology, electrophysiology, imaging, and a variety of molecular techniques to understand how homeostatic signaling mechanisms take shape at the synapse. At the NMJ, homeostatic signaling mechanisms couple retrograde (muscle-to-nerve) signaling with changes in presynaptic calcium influx, changes in the dynamics of the readily releasable vesicle pool, and ultimately, changes in presynaptic neurotransmitter release. Roles in these processes have been demonstrated for several molecules and signaling systems discussed here. This review focuses primarily on electrophysiological studies or data. In particular, attention is devoted to understanding what happens when NMJ function is challenged (usually through glutamate receptor inhibition) and the resulting homeostatic responses. A significant area of study not covered in this review, for the sake of simplicity, is the homeostatic control of synapse growth, which naturally, could also impinge upon synapse function in myriad ways. This article is part of the Special Issue entitled 'Homeostatic Synaptic Plasticity'.
Collapse
Affiliation(s)
- C Andrew Frank
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA.
| |
Collapse
|
43
|
Youssef G, Gerner L, Naeem AS, Ralph O, Ono M, O'Neill CA, O'Shaughnessy RFL. Rab3Gap1 mediates exocytosis of Claudin-1 and tight junction formation during epidermal barrier acquisition. Dev Biol 2013; 380:274-85. [PMID: 23685254 PMCID: PMC3995087 DOI: 10.1016/j.ydbio.2013.04.034] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Revised: 04/26/2013] [Accepted: 04/30/2013] [Indexed: 12/01/2022]
Abstract
Epidermal barrier acquisition during late murine gestation is accompanied by an increase in Akt kinase activity and cJun dephosphorlyation. The latter is directed by the Ppp2r2a regulatory subunit of the Pp2a phosphatase. This was accompanied by a change of Claudin-1 localisation to the cell surface and interaction between Occludin and Claudin-1 which are thought to be required for tight junction formation. The aim of this study was to determine the nature of the barrier defect caused by the loss of AKT/Ppp2r2a function. There was a paracellular barrier defect in rat epidermal keratinocytes expressing a Ppp2r2a siRNA. In Ppp2r2a knockdown cells, Claudin-1 was located to the cytoplasm and its expression was increased. Inhibiting cJun phosphorylation restored barrier function and plasma membrane localisation of Claudin-1. Expression of the Rab3 GTPase activating protein, Rab3Gap1, was restored in Ppp2r2a siRNA cells when cJun phosphorylation was inhibited. During normal mouse epidermal development, Claudin-1 plasma membrane localisation and Rab3Gap1 cell surface expression were co-incident with Akt activation in mouse epidermis, strongly suggesting a role of Rab3Gap1 in epidermal barrier acquisition. Supporting this hypothesis, siRNA knockdown of Rab3Gap1 prevented plasma membrane Claudin-1 expression and the formation of a barrier competent epithelium. Replacing Rab3Gap1 in Ppp2r2a knockdown cells was sufficient to rescue Claudin-1 transport to the cell surface. Therefore these data suggest Rab3Gap1 mediated exocytosis of Claudin-1 is an important component of epidermal barrier acquisition during epidermal development. Barrier acquisition correlates with Ppp2r2a and cell surface Claudin-1 expression. Ppp2r2a knockdown results in a paracellular barrier defect. Ppp2r2a knockdown prevents cell-surface claudin-1 expression in a c-Jun dependent fashion. Barrier rescue by inhibition of c-Jun phosphorylation involves exocytosis and Rab3Gap1. Rab3Gap1 is induced during barrier acquisition and is necessary for cell surface claudin-1.
Collapse
Affiliation(s)
- G Youssef
- Livingstone Skin Research Centre for Children, UCL Institute of Child Health, London WC1N 1EH, UK
| | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
Micro syndrome (OMIM 60018) and Martsolf syndrome (OMIM 21270) are related rare autosomal recessive disorders characterized by ocular and neurological abnormalities and hypothalamic hypogonadism. Micro syndrome has been associated with causative mutations in three disease genes: RAB3GAP1, RAB3GAP2 and RAB18. Martsolf syndrome has been associated with a mutation in RAB3GAP2. The present review summarizes the current literature on these genes and the proteins they encode.
Collapse
|
45
|
Seixas E, Barros M, Seabra MC, Barral DC. Rab and Arf proteins in genetic diseases. Traffic 2013; 14:871-85. [PMID: 23565987 DOI: 10.1111/tra.12072] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2012] [Revised: 04/03/2013] [Accepted: 04/08/2013] [Indexed: 01/29/2023]
Abstract
Rab and ADP-ribosylation factor (Arf) family proteins are master regulators of membrane trafficking and are involved in all steps of vesicular transport. These families of small guanine-nucleotide-binding (G) proteins are well suited to regulate membrane trafficking processes since their nucleotide state determines their conformation and the capacity to bind to a multitude of effectors, which mediate their functions. In recent years, several inherited diseases have been associated with mutations in genes encoding proteins belonging to these two families or in proteins that regulate their GTP-binding cycle. The genetic diseases that are caused by defects in Rabs, Arfs or their regulatory proteins are heterogeneous and display diverse symptoms. However, these diseases mainly affect two types of subcellular compartments, namely lysosome-related organelles and cilia. Also, several of these diseases affect the nervous system. Thus, the study of these diseases represents an opportunity to understand their etiology and the molecular mechanisms involved, as well as to develop novel therapeutic strategies.
Collapse
Affiliation(s)
- Elsa Seixas
- CEDOC, Faculdade de Ciências Médicas, FCM, Universidade Nova de Lisboa, 1169-056, Lisboa, Portugal
| | | | | | | |
Collapse
|
46
|
Marrocco J, Mairesse J, Ngomba RT, Silletti V, Van Camp G, Bouwalerh H, Summa M, Pittaluga A, Nicoletti F, Maccari S, Morley-Fletcher S. Anxiety-like behavior of prenatally stressed rats is associated with a selective reduction of glutamate release in the ventral hippocampus. J Neurosci 2012; 32:17143-54. [PMID: 23197707 PMCID: PMC6621858 DOI: 10.1523/jneurosci.1040-12.2012] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Revised: 09/26/2012] [Accepted: 10/01/2012] [Indexed: 02/05/2023] Open
Abstract
Abnormalities of synaptic transmission and plasticity in the hippocampus represent an integral part of the altered programming triggered by early life stress. Prenatally restraint stressed (PRS) rats develop long-lasting biochemical and behavioral changes, which are the expression of an anxious/depressive-like phenotype. We report here that PRS rats showed a selective impairment of depolarization- or kainate-stimulated glutamate and [(3)H]d-aspartate release in the ventral hippocampus, a region encoding memories related to stress and emotions. GABA release was unaffected in PRS rats. As a consequence of reduced glutamate release, PRS rats were also highly resistant to kainate-induced seizures. Abnormalities of glutamate release were associated with large reductions in the levels of synaptic vesicle-related proteins, such as VAMP (synaptobrevin), syntaxin-1, synaptophysin, synapsin Ia/b and IIa, munc-18, and Rab3A in the ventral hippocampus of PRS rats. Anxiety-like behavior in male PRS (and control) rats was inversely related to the extent of depolarization-evoked glutamate release in the ventral hippocampus. A causal relationship between anxiety-like behavior and reduction in glutamate release was demonstrated using a mixture of the mGlu2/3 receptor antagonist, LY341495, and the GABA(B) receptor antagonist, CGP52432, which was shown to amplify depolarization-evoked [(3)H]d-aspartate release in the ventral hippocampus. Bilateral microinfusion of CGP52432 plus LY341495 in the ventral hippocampus abolished anxiety-like behavior in PRS rats. These findings indicate that an impairment of glutamate release in the ventral hippocampus is a key component of the neuroplastic program induced by PRS, and that strategies aimed at enhancing glutamate release in the ventral hippocampus correct the "anxious phenotype" caused by early life stress.
Collapse
Affiliation(s)
- Jordan Marrocco
- Neural Plasticity Team-UMR CNRS/USTL 8576 Structural and Functional Glycobiology Unit, North University of Lille, 59655 Villeneuve d'Ascq France
| | - Jérôme Mairesse
- Neural Plasticity Team-UMR CNRS/USTL 8576 Structural and Functional Glycobiology Unit, North University of Lille, 59655 Villeneuve d'Ascq France
| | | | - Viviana Silletti
- Neural Plasticity Team-UMR CNRS/USTL 8576 Structural and Functional Glycobiology Unit, North University of Lille, 59655 Villeneuve d'Ascq France
| | - Gilles Van Camp
- Neural Plasticity Team-UMR CNRS/USTL 8576 Structural and Functional Glycobiology Unit, North University of Lille, 59655 Villeneuve d'Ascq France
| | - Hammou Bouwalerh
- Neural Plasticity Team-UMR CNRS/USTL 8576 Structural and Functional Glycobiology Unit, North University of Lille, 59655 Villeneuve d'Ascq France
| | - Maria Summa
- Department of Experimental Medicine, University of Genoa, 16126 Genoa, Italy, and
| | - Anna Pittaluga
- Department of Experimental Medicine, University of Genoa, 16126 Genoa, Italy, and
| | - Ferdinando Nicoletti
- IRCCS Neuromed, 86077 Pozzilli, Italy
- Department of Pharmacology, Sapienza University of Rome, 00185 Rome, Italy
| | - Stefania Maccari
- Neural Plasticity Team-UMR CNRS/USTL 8576 Structural and Functional Glycobiology Unit, North University of Lille, 59655 Villeneuve d'Ascq France
| | - Sara Morley-Fletcher
- Neural Plasticity Team-UMR CNRS/USTL 8576 Structural and Functional Glycobiology Unit, North University of Lille, 59655 Villeneuve d'Ascq France
| |
Collapse
|
47
|
Attention-deficit hyperactivity disorder and binge eating disorder in a patient with 2q21.1-q22.2 deletion. Psychiatr Genet 2012; 22:202-5. [PMID: 22517382 DOI: 10.1097/ypg.0b013e3283539604] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
We report the case of a young male with attention-deficit hyperactivity disorder, oppositional defiant disorder, eating problems and overweight, and mild mental retardation. Karyotype analysis detected an apparently balanced translocation: t(1;2)(p34.1;q21.1) de novo. Array comparative genomic hybridization analysis defined a de-novo cryptic deletion of 2q21.1-q22.2 bands. The deletion, here first associated with this complex phenotype, encompasses several genes with a putative role in different domains of behavioral control and neurocognitive functions; their deregulated expression may influence metabolic pathways and the role of dopamine in reward, explaining the complex psychiatric phenotype and the pharmacotherapy response described in our patient.
Collapse
|
48
|
Hernan AE, Holmes GL, Isaev D, Scott RC, Isaeva E. Altered short-term plasticity in the prefrontal cortex after early life seizures. Neurobiol Dis 2012; 50:120-6. [PMID: 23064435 DOI: 10.1016/j.nbd.2012.10.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Revised: 08/30/2012] [Accepted: 10/03/2012] [Indexed: 01/31/2023] Open
Abstract
Seizures during development are a relatively common occurrence and are often associated with poor cognitive outcomes. Recent studies show that early life seizures alter the function of various brain structures and have long-term consequences on seizure susceptibility and behavioral regulation. While many neocortical functions could be disrupted by epileptic seizures, we have concentrated on studying the prefrontal cortex (PFC) as disturbance of PFC functions is involved in numerous co-morbid disorders associated with epilepsy. In the present work we report an alteration of short-term plasticity in the PFC in rats that have experienced early life seizures. The most robust alteration occurs in the layer II/III to layer V network of neurons. However short-term plasticity of layer V to layer V network was also affected, indicating that the PFC function is broadly influenced by early life seizures. These data strongly suggest that repetitive seizures early in development cause substantial alteration in PFC function, which may be an important component underlying cognitive deficits in individuals with a history of seizures during development.
Collapse
Affiliation(s)
- A E Hernan
- Department of Neurology, Neuroscience Center at Dartmouth, Geisel School of Medicine at Dartmouth, Lebanon, NH 03766, USA.
| | | | | | | | | |
Collapse
|
49
|
Kimura N, Okabayashi S, Ono F. Dynein dysfunction disrupts intracellular vesicle trafficking bidirectionally and perturbs synaptic vesicle docking via endocytic disturbances a potential mechanism underlying age-dependent impairment of cognitive function. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 180:550-61. [PMID: 22182700 DOI: 10.1016/j.ajpath.2011.10.037] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Revised: 10/18/2011] [Accepted: 10/25/2011] [Indexed: 01/09/2023]
Abstract
Although genetic studies have demonstrated that β-amyloid protein (Aβ) plays a pivotal role in Alzheimer's disease (AD) pathogenesis, how aging contributes to AD onset remains unclear. Moreover, growing evidence suggests that Aβ-independent mechanisms, such as altered intracellular signaling cascades and impaired neurotransmitter release, also are likely involved in this process. Cytoplasmic dynein, a microtubule-based motor protein, mediates minus end-directed vesicle transport via interactions with dynactin, another microtubule-associated protein. We previously showed that normal aging attenuates the interaction between dynein-dynactin complexes in monkey brain and that dynein dysfunction reproduces age-dependent endocytic disturbances, resulting in intracellular Aβ accumulation. In this study, we report that dynein dysfunction disrupts not only retrograde transport of neurotrophic receptors but also anterograde transport of synaptic vesicles, which occurs concomitantly with an increase in Rab3 GTPase levels. Additionally, synaptic vesicle docking was perturbed via enhanced endocytosis. Dynein dysfunction also induced neuritic swelling, which is accompanied by a significant accumulation of neurofilaments. Moreover, we also confirmed that the dynein dysfunction-related disturbances are associated with aging in monkey brains and that age-dependent endocytic disturbances precede Aβ abnormality. These findings suggest that dynein dysfunction can alter neuronal activity via endocytic disturbances and may underlie age-dependent impairment of cognitive function. Moreover, in the presence of other risk factors, such as intracellular Aβ accumulation, dynein dysfunction may contribute to the development of AD.
Collapse
Affiliation(s)
- Nobuyuki Kimura
- Laboratory of Disease Control, Tsukuba Primate Research Center, National Institute of Biomedical Innovation, Ibaraki, Japan.
| | | | | |
Collapse
|
50
|
Esposito G, Ana Clara F, Verstreken P. Synaptic vesicle trafficking and Parkinson's disease. Dev Neurobiol 2011; 72:134-44. [DOI: 10.1002/dneu.20916] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|