1
|
Iguchi H, Katsuzawa T, Saruta C, Sadakata T, Kobayashi S, Sato Y, Sato A, Sano Y, Maezawa S, Shinoda Y, Furuichi T. Calcium-dependent activator protein for secretion 2 is involved in dopamine release in mouse midbrain neurons. Front Mol Neurosci 2024; 17:1444629. [PMID: 39092202 PMCID: PMC11291307 DOI: 10.3389/fnmol.2024.1444629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 06/28/2024] [Indexed: 08/04/2024] Open
Abstract
The Ca2+-dependent activator protein for secretion (CAPS/CADPS) family protein facilitates catecholamine release through the dense-core vesicle exocytosis in model neuroendocrine cell lines. However, it remains unclear if it induces dopamine release in the central neurons. This study aimed to examine the expression and function of CADPS2, one of the two CADPS paralogs, in dopamine neurons of the mouse midbrain. This study shows that CADPS2 was expressed in tyrosine hydroxylase and the vesicular monoamine transporter 2 (VMAT2)-positive dopaminergic neurons of the midbrain samples and primary mesencephalic cell cultures. Subcellular fractions rich in dopamine were collected using immunoaffinity for CADPS2 from midbrain protein extracts. Cell imaging using fluorescent false neurotransmitter FFN511 as a substrate for VMAT2 showed decreased activity-dependent dopamine release in Cadps2-deficient cultures, compared to that in wild-type cultures. These results suggest that CADPS2 is involved in dopamine release from the central neurons, indicating its involvement in the central dopamine pathway.
Collapse
Affiliation(s)
- Hirotoshi Iguchi
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Noda, Japan
| | - Takumi Katsuzawa
- Department of Environmental Health, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Japan
| | - Chihiro Saruta
- Laboratory for Molecular Neurogenesis, RIKEN Brain Science Institute, Wako, Japan
| | - Tetsushi Sadakata
- Laboratory for Molecular Neurogenesis, RIKEN Brain Science Institute, Wako, Japan
- Education and Research Support Center, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Shota Kobayashi
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Noda, Japan
| | - Yumi Sato
- Laboratory for Molecular Neurogenesis, RIKEN Brain Science Institute, Wako, Japan
- Department of Basic Pathology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Akira Sato
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Noda, Japan
- Laboratory for Molecular Neurogenesis, RIKEN Brain Science Institute, Wako, Japan
| | - Yoshitake Sano
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Noda, Japan
| | - So Maezawa
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Noda, Japan
| | - Yo Shinoda
- Department of Environmental Health, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Japan
- Laboratory for Molecular Neurogenesis, RIKEN Brain Science Institute, Wako, Japan
| | - Teiichi Furuichi
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Noda, Japan
- Laboratory for Molecular Neurogenesis, RIKEN Brain Science Institute, Wako, Japan
- Laboratory for Molecular Mechanisms of Brain Development, Center for Brain Science, RIKEN, Wako, Japan
| |
Collapse
|
2
|
Guo Y, Xiang P, Sun X, Liu W, Zhou J, Yin B, Hou L, Qiang B, Li H, Shu P, Peng X. Docking protein 6 (DOK6) selectively docks the neurotrophic signaling transduction to restrain peripheral neuropathy. Signal Transduct Target Ther 2024; 9:32. [PMID: 38351062 PMCID: PMC10864363 DOI: 10.1038/s41392-024-01742-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 12/12/2023] [Accepted: 01/09/2024] [Indexed: 02/16/2024] Open
Abstract
The appropriate and specific response of nerve cells to various external cues is essential for the establishment and maintenance of neural circuits, and this process requires the proper recruitment of adaptor molecules to selectively activate downstream pathways. Here, we identified that DOK6, a member of the Dok (downstream of tyrosine kinases) family, is required for the maintenance of peripheral axons, and that loss of Dok6 can cause typical peripheral neuropathy symptoms in mice, manifested as impaired sensory, abnormal posture, paw deformities, blocked nerve conduction, and dysmyelination. Furthermore, Dok6 is highly expressed in peripheral neurons but not in Schwann cells, and genetic deletion of Dok6 in peripheral neurons led to typical peripheral myelin outfolding, axon destruction, and hindered retrograde axonal transport. Specifically, DOK6 acts as an adaptor protein for selectivity-mediated neurotrophic signal transduction and retrograde transport for TrkC and Ret but not for TrkA and TrkB. DOK6 interacts with certain proteins in the trafficking machinery and controls their phosphorylation, including MAP1B, Tau and Dynein for axonal transport, and specifically activates the downstream ERK1/2 kinase pathway to maintain axonal survival and homeostasis. This finding provides new clues to potential insights into the pathogenesis and treatment of hereditary peripheral neuropathies and other degenerative diseases.
Collapse
Affiliation(s)
- Yan Guo
- Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Pan Xiang
- Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Xiaojiao Sun
- Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Wei Liu
- Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Jiafeng Zhou
- Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Bin Yin
- Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
- State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing, 100005, China
| | - Lin Hou
- Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
- State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing, 100005, China
| | - Boqin Qiang
- Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
- State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing, 100005, China
| | - Huiliang Li
- Wolfson Institute for Biomedical Research, University College London, Gower Street, London, WC1E 6BT, UK
| | - Pengcheng Shu
- Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China.
- State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing, 100005, China.
- Chinese Institute for Brain Research, Beijing, 102206, China.
| | - Xiaozhong Peng
- Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China.
- State Key Laboratory of Respiratory Health and Multimorbidity, Beijing, 100005, China.
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
3
|
Reichlmeir M, Canet-Pons J, Koepf G, Nurieva W, Duecker RP, Doering C, Abell K, Key J, Stokes MP, Zielen S, Schubert R, Ivics Z, Auburger G. In Cerebellar Atrophy of 12-Month-Old ATM-Null Mice, Transcriptome Upregulations Concern Most Neurotransmission and Neuropeptide Pathways, While Downregulations Affect Prominently Itpr1, Usp2 and Non-Coding RNA. Cells 2023; 12:2399. [PMID: 37830614 PMCID: PMC10572167 DOI: 10.3390/cells12192399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 09/29/2023] [Accepted: 10/01/2023] [Indexed: 10/14/2023] Open
Abstract
The autosomal recessive disorder Ataxia-Telangiectasia is caused by a dysfunction of the stress response protein, ATM. In the nucleus of proliferating cells, ATM senses DNA double-strand breaks and coordinates their repair. This role explains T-cell dysfunction and tumour risk. However, it remains unclear whether this function is relevant for postmitotic neurons and underlies cerebellar atrophy, since ATM is cytoplasmic in postmitotic neurons. Here, we used ATM-null mice that survived early immune deficits via bone-marrow transplantation, and that reached initial neurodegeneration stages at 12 months of age. Global cerebellar transcriptomics demonstrated that ATM depletion triggered upregulations in most neurotransmission and neuropeptide systems. Downregulated transcripts were found for the ATM interactome component Usp2, many non-coding RNAs, ataxia genes Itpr1, Grid2, immediate early genes and immunity factors. Allelic splice changes affected prominently the neuropeptide machinery, e.g., Oprm1. Validation experiments with stressors were performed in human neuroblastoma cells, where ATM was localised only to cytoplasm, similar to the brain. Effect confirmation in SH-SY5Y cells occurred after ATM depletion and osmotic stress better than nutrient/oxidative stress, but not after ATM kinase inhibition or DNA stressor bleomycin. Overall, we provide pioneer observations from a faithful A-T mouse model, which suggest general changes in synaptic and dense-core vesicle stress adaptation.
Collapse
Affiliation(s)
- Marina Reichlmeir
- Goethe University Frankfurt, University Hospital, Clinic of Neurology, Exp. Neurology, Heinrich Hoffmann Str. 7, 60590 Frankfurt am Main, Germany; (M.R.); (J.C.-P.); (J.K.)
| | - Júlia Canet-Pons
- Goethe University Frankfurt, University Hospital, Clinic of Neurology, Exp. Neurology, Heinrich Hoffmann Str. 7, 60590 Frankfurt am Main, Germany; (M.R.); (J.C.-P.); (J.K.)
| | - Gabriele Koepf
- Goethe University Frankfurt, University Hospital, Clinic of Neurology, Exp. Neurology, Heinrich Hoffmann Str. 7, 60590 Frankfurt am Main, Germany; (M.R.); (J.C.-P.); (J.K.)
| | - Wasifa Nurieva
- Transposition and Genome Engineering, Research Centre of the Division of Hematology, Gene and Cell Therapy, Paul Ehrlich Institute, 63225 Langen, Germany; (W.N.); (Z.I.)
| | - Ruth Pia Duecker
- Division of Pediatrics, Pulmonology, Allergology, Infectious Diseases and Gastroenterology, Children’s Hospital, University Hospital, Goethe-University, 60590 Frankfurt am Main, Germany; (R.P.D.); (S.Z.); (R.S.)
| | - Claudia Doering
- Dr. Senckenberg Institute of Pathology, University Hospital Frankfurt, 60590 Frankfurt am Main, Germany;
| | - Kathryn Abell
- Cell Signaling Technology, Inc., Danvers, MA 01923, USA; (K.A.); (M.P.S.)
| | - Jana Key
- Goethe University Frankfurt, University Hospital, Clinic of Neurology, Exp. Neurology, Heinrich Hoffmann Str. 7, 60590 Frankfurt am Main, Germany; (M.R.); (J.C.-P.); (J.K.)
| | - Matthew P. Stokes
- Cell Signaling Technology, Inc., Danvers, MA 01923, USA; (K.A.); (M.P.S.)
| | - Stefan Zielen
- Division of Pediatrics, Pulmonology, Allergology, Infectious Diseases and Gastroenterology, Children’s Hospital, University Hospital, Goethe-University, 60590 Frankfurt am Main, Germany; (R.P.D.); (S.Z.); (R.S.)
- Respiratory Research Institute, Medaimun GmbH, 60596 Frankfurt am Main, Germany
| | - Ralf Schubert
- Division of Pediatrics, Pulmonology, Allergology, Infectious Diseases and Gastroenterology, Children’s Hospital, University Hospital, Goethe-University, 60590 Frankfurt am Main, Germany; (R.P.D.); (S.Z.); (R.S.)
| | - Zoltán Ivics
- Transposition and Genome Engineering, Research Centre of the Division of Hematology, Gene and Cell Therapy, Paul Ehrlich Institute, 63225 Langen, Germany; (W.N.); (Z.I.)
| | - Georg Auburger
- Goethe University Frankfurt, University Hospital, Clinic of Neurology, Exp. Neurology, Heinrich Hoffmann Str. 7, 60590 Frankfurt am Main, Germany; (M.R.); (J.C.-P.); (J.K.)
| |
Collapse
|
4
|
Hobson BD, Stanley AT, De Los Santos MB, Culbertson B, Mosharov EV, Sims PA, Sulzer D. Conserved and cell type-specific transcriptional responses to IFN-γ in the ventral midbrain. Brain Behav Immun 2023; 111:277-291. [PMID: 37100211 PMCID: PMC10460506 DOI: 10.1016/j.bbi.2023.04.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 03/28/2023] [Accepted: 04/23/2023] [Indexed: 04/28/2023] Open
Abstract
Dysregulated inflammation within the central nervous system (CNS) contributes to neuropathology in infectious, autoimmune, and neurodegenerative disease. With the exception of microglia, major histocompatibility complex (MHC) proteins are virtually undetectable in the mature, healthy central nervous system (CNS). Neurons have generally been considered incapable of antigen presentation, and although interferon gamma (IFN-γ) can elicit neuronal MHC class I (MHC-I) expression and antigen presentation in vitro, it has been unclear whether similar responses occur in vivo. Here we directly injected IFN-γ into the ventral midbrain of mature mice and analyzed gene expression profiles of specific CNS cell types. We found that IFN-γ upregulated MHC-I and associated mRNAs in ventral midbrain microglia, astrocytes, oligodendrocytes, and GABAergic, glutamatergic, and dopaminergic neurons. The core set of IFN-γ-induced genes and their response kinetics were similar in neurons and glia, but with a lower amplitude of expression in neurons. A diverse repertoire of genes was upregulated in glia, particularly microglia, which were the only cells to undergo cellular proliferation and express MHC classII (MHC-II) and associated genes. To determine if neurons respond directly via cell-autonomous IFN-γ receptor (IFNGR) signaling, we produced mutant mice with a deletion of the IFN-γ-binding domain of IFNGR1 in dopaminergic neurons, which resulted in a complete loss of dopaminergic neuronal responses to IFN-γ. Our results demonstrate that IFN-γ induces neuronal IFNGR signaling and upregulation of MHC-I and related genes in vivo, although the expression level is low compared to oligodendrocytes, astrocytes, and microglia.
Collapse
Affiliation(s)
- Benjamin D Hobson
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032, United States; Medical Scientist Training Program, Columbia University Irving Medical Center, New York, NY 10032, United States; Department of Psychiatry, Columbia University Irving Medical Center, New York, NY 10032, United States; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, United States
| | - Adrien T Stanley
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY 10032, United States; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, United States
| | - Mark B De Los Santos
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY 10032, United States; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, United States
| | - Bruce Culbertson
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032, United States; Medical Scientist Training Program, Columbia University Irving Medical Center, New York, NY 10032, United States
| | - Eugene V Mosharov
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY 10032, United States; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, United States
| | - Peter A Sims
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032, United States; Department of Biochemistry & Molecular Biophysics, Columbia University Irving Medical Center, New York, NY 10032, United States; Sulzberger Columbia Genome Center, Columbia University Irving Medical Center, New York, NY 10032, United States; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, United States.
| | - David Sulzer
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, United States; Department of Psychiatry, Columbia University Irving Medical Center, New York, NY 10032, United States; Department of Pharmacology, Columbia University Irving Medical Center, New York, NY 10032, United States; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, United States; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, United States.
| |
Collapse
|
5
|
Arévalo JC, Deogracias R. Mechanisms Controlling the Expression and Secretion of BDNF. Biomolecules 2023; 13:biom13050789. [PMID: 37238659 DOI: 10.3390/biom13050789] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/19/2023] [Accepted: 04/28/2023] [Indexed: 05/28/2023] Open
Abstract
Brain-derived nerve factor (BDNF), through TrkB receptor activation, is an important modulator for many different physiological and pathological functions in the nervous system. Among them, BDNF plays a crucial role in the development and correct maintenance of brain circuits and synaptic plasticity as well as in neurodegenerative diseases. The proper functioning of the central nervous system depends on the available BDNF concentrations, which are tightly regulated at transcriptional and translational levels but also by its regulated secretion. In this review we summarize the new advances regarding the molecular players involved in BDNF release. In addition, we will address how changes of their levels or function in these proteins have a great impact in those functions modulated by BDNF under physiological and pathological conditions.
Collapse
Affiliation(s)
- Juan Carlos Arévalo
- Department of Cell Biology and Pathology, Institute of Neurosciences of Castille and Leon (INCyL), University of Salamanca, 37007 Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Rubén Deogracias
- Department of Cell Biology and Pathology, Institute of Neurosciences of Castille and Leon (INCyL), University of Salamanca, 37007 Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| |
Collapse
|
6
|
Lorenzo-Betancor O, Galosi L, Bonfili L, Eleuteri AM, Cecarini V, Verin R, Dini F, Attili AR, Berardi S, Biagini L, Robino P, Stella MC, Yearout D, Dorschner MO, Tsuang DW, Rossi G, Zabetian CP. Homozygous CADPS2 Mutations Cause Neurodegenerative Disease with Lewy Body-like Pathology in Parrots. Mov Disord 2022; 37:2345-2354. [PMID: 36086934 PMCID: PMC9772200 DOI: 10.1002/mds.29211] [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: 05/08/2022] [Revised: 07/19/2022] [Accepted: 08/12/2022] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Several genetic models that recapitulate neurodegenerative features of Parkinson's disease (PD) exist, which have been largely based on genes discovered in monogenic PD families. However, spontaneous genetic mutations have not been linked to the pathological hallmarks of PD in non-human vertebrates. OBJECTIVE To describe the genetic and pathological findings of three Yellow-crowned parrot (Amazona ochrocepahala) siblings with a severe and rapidly progressive neurological phenotype. METHODS The phenotype of the three parrots included severe ataxia, rigidity, and tremor, while their parents were phenotypically normal. Tests to identify avian viral infections and brain imaging studies were all negative. Due to their severe impairment, they were all euthanized at age 3 months and their brains underwent neuropathological examination and proteasome activity assays. Whole genome sequencing (WGS) was performed on the three affected parrots and their parents. RESULTS The brains of affected parrots exhibited neuronal loss, spongiosis, and widespread Lewy body-like inclusions in many regions including the midbrain, basal ganglia, and neocortex. Proteasome activity was significantly reduced in these animals compared to a control (P < 0.05). WGS identified a single homozygous missense mutation (p.V559L) in a highly conserved amino acid within the pleckstrin homology (PH) domain of the calcium-dependent secretion activator 2 (CADPS2) gene. CONCLUSIONS Our data suggest that a homozygous mutation in the CADPS2 gene causes a severe neurodegenerative phenotype with Lewy body-like pathology in parrots. Although CADPS2 variants have not been reported to cause PD, further investigation of the gene might provide important insights into the pathophysiology of Lewy body disorders. © 2022 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society. This article has been contributed to by U.S. Government employees and their work is in the public domain in the USA.
Collapse
Affiliation(s)
- Oswaldo Lorenzo-Betancor
- Veterans Affairs Puget Sound Health Care System, Seattle,
Washington, USA,Department of Neurology, University of Washington School of
Medicine, Seattle, Washington, USA
| | - Livio Galosi
- School of Biosciences and Veterinary Medicine, University
of Camerino, Matelica, Italy
| | - Laura Bonfili
- School of Biosciences and Veterinary Medicine, University
of Camerino, Matelica, Italy
| | - Anna Maria Eleuteri
- School of Biosciences and Veterinary Medicine, University
of Camerino, Matelica, Italy
| | - Valentina Cecarini
- School of Biosciences and Veterinary Medicine, University
of Camerino, Matelica, Italy
| | - Ranieri Verin
- Department of Comparative Biomedicine and Food Science,
University of Padova “Agripolis”, Legnaro, Italy
| | - Fabrizio Dini
- School of Biosciences and Veterinary Medicine, University
of Camerino, Matelica, Italy
| | - Anna-Rita Attili
- School of Biosciences and Veterinary Medicine, University
of Camerino, Matelica, Italy
| | - Sara Berardi
- School of Biosciences and Veterinary Medicine, University
of Camerino, Matelica, Italy
| | - Lucia Biagini
- School of Biosciences and Veterinary Medicine, University
of Camerino, Matelica, Italy
| | - Patrizia Robino
- Department of Veterinary Sciences, University of Torino,
Torino, Italy
| | | | - Dora Yearout
- Veterans Affairs Puget Sound Health Care System, Seattle,
Washington, USA
| | - Michael O. Dorschner
- Department of Pathology, Center for Precision Diagnostics,
University of Washington, Seattle, Washington, USA
| | - Debby W. Tsuang
- Veterans Affairs Puget Sound Health Care System, Seattle,
Washington, USA,Department of Psychiatry, University of Washington School
of Medicine, Seattle, Washington, USA,Correspondence to: Dr. Cyrus P.
Zabetian, Veterans Affairs Puget Sound Health Care System, Seattle, Washington
98108, USA; ; Dr. Giacomo Rossi, School of
Biosciences and Veterinary Medicine, University of Camerino, Matelica, Italy;
; Dr. Debby W. Tsuang, Veterans
Affairs Puget Sound Health Care System, Seattle, Washington 98108, USA;
| | - Giacomo Rossi
- School of Biosciences and Veterinary Medicine, University
of Camerino, Matelica, Italy,Correspondence to: Dr. Cyrus P.
Zabetian, Veterans Affairs Puget Sound Health Care System, Seattle, Washington
98108, USA; ; Dr. Giacomo Rossi, School of
Biosciences and Veterinary Medicine, University of Camerino, Matelica, Italy;
; Dr. Debby W. Tsuang, Veterans
Affairs Puget Sound Health Care System, Seattle, Washington 98108, USA;
| | - Cyrus P. Zabetian
- Veterans Affairs Puget Sound Health Care System, Seattle,
Washington, USA,Department of Neurology, University of Washington School of
Medicine, Seattle, Washington, USA,Correspondence to: Dr. Cyrus P.
Zabetian, Veterans Affairs Puget Sound Health Care System, Seattle, Washington
98108, USA; ; Dr. Giacomo Rossi, School of
Biosciences and Veterinary Medicine, University of Camerino, Matelica, Italy;
; Dr. Debby W. Tsuang, Veterans
Affairs Puget Sound Health Care System, Seattle, Washington 98108, USA;
| |
Collapse
|
7
|
Duński E, Pękowska A. Keeping the balance: Trade-offs between human brain evolution, autism, and schizophrenia. Front Genet 2022; 13:1009390. [DOI: 10.3389/fgene.2022.1009390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 10/12/2022] [Indexed: 11/22/2022] Open
Abstract
The unique qualities of the human brain are a product of a complex evolutionary process. Evolution, famously described by François Jacob as a “tinkerer,” builds upon existing genetic elements by modifying and repurposing them for new functions. Genetic changes in DNA may lead to the emergence of new genes or cause altered gene expression patterns. Both gene and regulatory element mutations may lead to new functions. Yet, this process may lead to side-effects. An evolutionary trade-off occurs when an otherwise beneficial change, which is important for evolutionary success and is under strong positive selection, concurrently results in a detrimental change in another trait. Pleiotropy occurs when a gene affects multiple traits. Antagonistic pleiotropy is a phenomenon whereby a genetic variant leads to an increase in fitness at one life-stage or in a specific environment, but simultaneously decreases fitness in another respect. Therefore, it is conceivable that the molecular underpinnings of evolution of highly complex traits, including brain size or cognitive ability, under certain conditions could result in deleterious effects, which would increase the susceptibility to psychiatric or neurodevelopmental diseases. Here, we discuss possible trade-offs and antagonistic pleiotropies between evolutionary change in a gene sequence, dosage or activity and the susceptibility of individuals to autism spectrum disorders and schizophrenia. We present current knowledge about genes and alterations in gene regulatory landscapes, which have likely played a role in establishing human-specific traits and have been implicated in those diseases.
Collapse
|
8
|
Sato Y, Tsuyusaki M, Takahashi-Iwanaga H, Fujisawa R, Masamune A, Hamada S, Matsumoto R, Tanaka Y, Kakuta Y, Yamaguchi-Kabata Y, Furuse T, Wakana S, Shimura T, Kobayashi R, Shinoda Y, Goitsuka R, Maezawa S, Sadakata T, Sano Y, Furuichi T. Loss of CAPS2/Cadps2 leads to exocrine pancreatic cell injury and intracellular accumulation of secretory granules in mice. Front Mol Biosci 2022; 9:1040237. [DOI: 10.3389/fmolb.2022.1040237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 10/26/2022] [Indexed: 11/09/2022] Open
Abstract
The type 2 Ca2+-dependent activator protein for secretion (CAPS2/CADPS2) regulates dense-core vesicle trafficking and exocytosis and is involved in the regulated release of catecholamines, peptidergic hormones, and neuromodulators. CAPS2 is expressed in the pancreatic exocrine acinar cells that produce and secrete digestive enzymes. However, the functional role of CAPS2 in vesicular trafficking and/or exocytosis of non-regulatory proteins in the exocrine pancreas remains to be determined. Here, we analyzed the morpho-pathological indicators of the pancreatic exocrine pathway in Cadps2-deficient mouse models using histochemistry, biochemistry, and electron microscopy. We used whole exosome sequencing to identify CADPS2 variants in patients with chronic pancreatitis (CP). Caps2/Cadps2-knockout (KO) mice exhibited morphophysiological abnormalities in the exocrine pancreas, including excessive accumulation of secretory granules (zymogen granules) and their amylase content in the cytoplasm, deterioration of the fine intracellular membrane structures (disorganized rough endoplasmic reticulum, dilated Golgi cisternae, and the appearance of empty vesicles and autophagic-like vacuoles), as well as exocrine pancreatic cell injury, including acinar cell atrophy, increased fibrosis, and inflammatory cell infiltration. Pancreas-specific Cadps2 conditional KO mice exhibited pathological abnormalities in the exocrine pancreas similar to the global Cadps2 KO mice, indicating that these phenotypes were caused either directly or indirectly by CAPS2 deficiency in the pancreas. Furthermore, we identified a rare variant in the exon3 coding region of CADPS2 in a non-alcoholic patient with CP and showed that Cadps2-dex3 mice lacking CAPS2 exon3 exhibited symptoms similar to those exhibited by the Cadps2 KO and cKO mice. These results suggest that CAPS2 is critical for the proper functioning of the pancreatic exocrine pathway, and its deficiency is associated with a risk of pancreatic acinar cell pathology.
Collapse
|
9
|
Staudt A, Ratai O, Bouzouina A, Fecher-Trost C, Shaaban A, Bzeih H, Horn A, Shaib AH, Klose M, Flockerzi V, Lauterbach MA, Rettig J, Becherer U. Localization of the Priming Factors CAPS1 and CAPS2 in Mouse Sensory Neurons Is Determined by Their N-Termini. Front Mol Neurosci 2022; 15:674243. [PMID: 35493323 PMCID: PMC9049930 DOI: 10.3389/fnmol.2022.674243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 02/09/2022] [Indexed: 11/13/2022] Open
Abstract
Both paralogs of the calcium-dependent activator protein for secretion (CAPS) are required for exocytosis of synaptic vesicles (SVs) and large dense core vesicles (LDCVs). Despite approximately 80% sequence identity, CAPS1 and CAPS2 have distinct functions in promoting exocytosis of SVs and LDCVs in dorsal root ganglion (DRG) neurons. However, the molecular mechanisms underlying these differences remain enigmatic. In this study, we applied high- and super-resolution imaging techniques to systematically assess the subcellular localization of CAPS paralogs in DRG neurons deficient in both CAPS1 and CAPS2. CAPS1 was found to be more enriched at the synapses. Using – in-depth sequence analysis, we identified a unique CAPS1 N-terminal sequence, which we introduced into CAPS2. This CAPS1/2 chimera reproduced the pre-synaptic localization of CAPS1 and partially rescued synaptic transmission in neurons devoid of CAPS1 and CAPS2. Using immunoprecipitation combined with mass spectrometry, we identified CAPS1-specific interaction partners that could be responsible for its pre-synaptic enrichment. Taken together, these data suggest an important role of the CAPS1-N terminus in the localization of the protein at pre-synapses.
Collapse
Affiliation(s)
- Angelina Staudt
- Department of Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine, Saarland University, Homburg, Germany
| | - Olga Ratai
- Department of Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine, Saarland University, Homburg, Germany
| | - Aicha Bouzouina
- Department of Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine, Saarland University, Homburg, Germany
| | - Claudia Fecher-Trost
- Department of Experimental and Clinical Pharmacology and Toxicology, Preclinical Center for Molecular Signaling (PZMS), Saarland University, Homburg, Germany
| | - Ahmed Shaaban
- Department of Neuroscience, University of Copenhagen, København, Denmark
| | - Hawraa Bzeih
- Department of Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine, Saarland University, Homburg, Germany
| | - Alexander Horn
- Department of Organic Chemistry, Saarland University, Saarbrücken, Germany
| | - Ali H. Shaib
- Department of Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine, Saarland University, Homburg, Germany
- Institute for Neuro- and Sensory Physiology, University Medical Center Göttingen, Göttingen, Germany
| | - Margarete Klose
- Department of Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine, Saarland University, Homburg, Germany
| | - Veit Flockerzi
- Department of Experimental and Clinical Pharmacology and Toxicology, Preclinical Center for Molecular Signaling (PZMS), Saarland University, Homburg, Germany
| | - Marcel A. Lauterbach
- Department of Molecular Imaging, Center for Integrative Physiology and Molecular Medicine, Saarland University, Homburg, Germany
| | - Jens Rettig
- Department of Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine, Saarland University, Homburg, Germany
| | - Ute Becherer
- Department of Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine, Saarland University, Homburg, Germany
- *Correspondence: Ute Becherer,
| |
Collapse
|
10
|
Mapelli L, Soda T, D’Angelo E, Prestori F. The Cerebellar Involvement in Autism Spectrum Disorders: From the Social Brain to Mouse Models. Int J Mol Sci 2022; 23:ijms23073894. [PMID: 35409253 PMCID: PMC8998980 DOI: 10.3390/ijms23073894] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 02/04/2023] Open
Abstract
Autism spectrum disorders (ASD) are pervasive neurodevelopmental disorders that include a variety of forms and clinical phenotypes. This heterogeneity complicates the clinical and experimental approaches to ASD etiology and pathophysiology. To date, a unifying theory of these diseases is still missing. Nevertheless, the intense work of researchers and clinicians in the last decades has identified some ASD hallmarks and the primary brain areas involved. Not surprisingly, the areas that are part of the so-called “social brain”, and those strictly connected to them, were found to be crucial, such as the prefrontal cortex, amygdala, hippocampus, limbic system, and dopaminergic pathways. With the recent acknowledgment of the cerebellar contribution to cognitive functions and the social brain, its involvement in ASD has become unmistakable, though its extent is still to be elucidated. In most cases, significant advances were made possible by recent technological developments in structural/functional assessment of the human brain and by using mouse models of ASD. Mouse models are an invaluable tool to get insights into the molecular and cellular counterparts of the disease, acting on the specific genetic background generating ASD-like phenotype. Given the multifaceted nature of ASD and related studies, it is often difficult to navigate the literature and limit the huge content to specific questions. This review fulfills the need for an organized, clear, and state-of-the-art perspective on cerebellar involvement in ASD, from its connections to the social brain areas (which are the primary sites of ASD impairments) to the use of monogenic mouse models.
Collapse
Affiliation(s)
- Lisa Mapelli
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (T.S.); (E.D.)
- Correspondence: (L.M.); (F.P.)
| | - Teresa Soda
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (T.S.); (E.D.)
| | - Egidio D’Angelo
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (T.S.); (E.D.)
- Brain Connectivity Center, IRCCS Mondino Foundation, 27100 Pavia, Italy
| | - Francesca Prestori
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (T.S.); (E.D.)
- Correspondence: (L.M.); (F.P.)
| |
Collapse
|
11
|
Hobson BD, Kong L, Angelo MF, Lieberman OJ, Mosharov EV, Herzog E, Sulzer D, Sims PA. Subcellular and regional localization of mRNA translation in midbrain dopamine neurons. Cell Rep 2022; 38:110208. [PMID: 35021090 PMCID: PMC8844886 DOI: 10.1016/j.celrep.2021.110208] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 10/25/2021] [Accepted: 12/13/2021] [Indexed: 12/12/2022] Open
Abstract
Midbrain dopaminergic (mDA) neurons exhibit extensive dendritic and axonal arborizations, but local protein synthesis is not characterized in these neurons. Here, we investigate messenger RNA (mRNA) localization and translation in mDA neuronal axons and dendrites, both of which release dopamine (DA). Using highly sensitive ribosome-bound RNA sequencing and imaging approaches, we find no evidence for mRNA translation in mDA axons. In contrast, mDA neuronal dendrites in the substantia nigra pars reticulata (SNr) contain ribosomes and mRNAs encoding the major components of DA synthesis, release, and reuptake machinery. Surprisingly, we also observe dendritic localization of mRNAs encoding synaptic vesicle-related proteins, including those involved in exocytic fusion. Our results are consistent with a role for local translation in the regulation of DA release from dendrites, but not from axons. Our translatome data define a molecular signature of sparse mDA neurons in the SNr, including the enrichment of Atp2a3/SERCA3, an atypical ER calcium pump.
Collapse
Affiliation(s)
- Benjamin D Hobson
- Department of Systems Biology, Columbia University Irving Medical Center, New York 10032, NY, USA; Medical Scientist Training Program, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Psychiatry, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Linghao Kong
- Department of Systems Biology, Columbia University Irving Medical Center, New York 10032, NY, USA
| | - Maria Florencia Angelo
- Interdisciplinary Institute for Neuroscience, Université de Bordeaux, Bordeaux, France; Interdisciplinary Institute for Neuroscience, CNRS UMR 5297, Bordeaux, France
| | - Ori J Lieberman
- Medical Scientist Training Program, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Psychiatry, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Eugene V Mosharov
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY 10032, USA; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA
| | - Etienne Herzog
- Interdisciplinary Institute for Neuroscience, Université de Bordeaux, Bordeaux, France; Interdisciplinary Institute for Neuroscience, CNRS UMR 5297, Bordeaux, France.
| | - David Sulzer
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Psychiatry, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Pharmacology, Columbia University Irving Medical Center, New York, NY 10032, USA; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| | - Peter A Sims
- Department of Systems Biology, Columbia University Irving Medical Center, New York 10032, NY, USA; Department of Biochemistry & Molecular Biophysics, Columbia University Irving Medical Center, New York, NY 10032, USA; Sulzberger Columbia Genome Center, Columbia University Irving Medical Center, New York, NY 10032, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| |
Collapse
|
12
|
Pleiotropic effects of BDNF on the cerebellum and hippocampus: Implications for neurodevelopmental disorders. Neurobiol Dis 2022; 163:105606. [PMID: 34974125 DOI: 10.1016/j.nbd.2021.105606] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 12/22/2021] [Accepted: 12/29/2021] [Indexed: 12/17/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is one of the most studied neurotrophins in the mammalian brain, essential not only to the development of the central nervous system but also to synaptic plasticity. BDNF is present in various brain areas, but highest levels of expression are seen in the cerebellum and hippocampus. After birth, BDNF acts in the cerebellum as a mitogenic and chemotactic factor, stimulating the cerebellar granule cell precursors to proliferate, migrate and maturate, while in the hippocampus BDNF plays a fundamental role in synaptic transmission and plasticity, representing a key regulator for the long-term potentiation, learning and memory. Furthermore, the expression of BDNF is highly regulated and changes of its expression are associated with both physiological and pathological conditions. The purpose of this review is to provide an overview of the current state of knowledge on the BDNF biology and its neurotrophic role in the proper development and functioning of neurons and synapses in two important brain areas of postnatal neurogenesis, the cerebellum and hippocampus. Dysregulation of BDNF expression and signaling, resulting in alterations in neuronal maturation and plasticity in both systems, is a common hallmark of several neurodevelopmental diseases, such as autism spectrum disorder, suggesting that neuronal malfunction present in these disorders is the result of excessive or reduced of BDNF support. We believe that the more the relevance of the pathophysiological actions of BDNF, and its downstream signals, in early postnatal development will be highlighted, the more likely it is that new neuroprotective therapeutic strategies will be identified in the treatment of various neurodevelopmental disorders.
Collapse
|
13
|
Trinoskey-Rice GE, Woon EP, Pitts EG, Gourley SL. Cocaine Elevates Calcium-Dependent Activator Protein for Secretion 2 in the Mouse Orbitofrontal Cortex. Dev Neurosci 2021; 43:376-382. [PMID: 34695821 DOI: 10.1159/000519681] [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: 04/09/2021] [Accepted: 09/09/2021] [Indexed: 11/19/2022] Open
Abstract
Calcium-dependent activator protein for secretion 2 (CAPS2; also referred to as CADPS2) is a dense core vesicle-associated protein that promotes the activity-dependent release of neuropeptides including neurotrophins. Addictive drugs appear to prime neurotrophin release in multiple brain regions, but mechanistic factors are still being elucidated. Here, experimenters administered cocaine to adolescent mice at doses that potentiated later cocaine self-administration. Experimenter-administered cocaine elevated the CAPS2 protein content in the orbitofrontal cortex (OFC; but not striatum) multiple weeks after drug exposure. Meanwhile, proteins that are sensitive to brain-derived neurotrophic factor (BDNF) release and binding (phosphorylated protein kinase B and phosphoinositide 3-kinase, and GABAAα1 levels) did not differ between cocaine-exposed and naive mice in the OFC. This pattern is consistent with evidence that CAPS2 primes stimulated release of neurotrophins like BDNF, rather than basal levels. Thus, cocaine administered at behaviorally relevant doses elevates CAPS2 protein content in the OFC, and the effects are detected long after cocaine exposure.
Collapse
Affiliation(s)
- Gracy E Trinoskey-Rice
- Departments of Pediatrics and Psychiatry, Emory University School of Medicine, Atlanta, Georgia, USA.,Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Ellen P Woon
- Departments of Pediatrics and Psychiatry, Emory University School of Medicine, Atlanta, Georgia, USA.,Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.,Graduate Training Program in Neuroscience, Emory University, Atlanta, Georgia, USA
| | - Elizabeth G Pitts
- Departments of Pediatrics and Psychiatry, Emory University School of Medicine, Atlanta, Georgia, USA.,Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.,Graduate Training Program in Neuroscience, Emory University, Atlanta, Georgia, USA
| | - Shannon L Gourley
- Departments of Pediatrics and Psychiatry, Emory University School of Medicine, Atlanta, Georgia, USA.,Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.,Graduate Training Program in Neuroscience, Emory University, Atlanta, Georgia, USA.,Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| |
Collapse
|
14
|
Rodríguez-Fdez S, Lorenzo-Martín LF, Fabbiano S, Menacho-Márquez M, Sauzeau V, Dosil M, Bustelo XR. New Functions of Vav Family Proteins in Cardiovascular Biology, Skeletal Muscle, and the Nervous System. BIOLOGY 2021; 10:biology10090857. [PMID: 34571735 PMCID: PMC8472352 DOI: 10.3390/biology10090857] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 08/27/2021] [Accepted: 08/29/2021] [Indexed: 11/16/2022]
Abstract
Simple Summary In this review, we provide information on the role of Vav proteins, a group of signaling molecules that act as both Rho GTPase activators and adaptor molecules, in the cardiovascular system, skeletal muscle, and the nervous system. We also describe how these functions impact in other physiological and pathological processes such as sympathoregulation, blood pressure regulation, systemic metabolism, and metabolic syndrome. Abstract Vav proteins act as tyrosine phosphorylation-regulated guanosine nucleotide exchange factors for Rho GTPases and as molecular scaffolds. In mammals, this family of signaling proteins is composed of three members (Vav1, Vav2, Vav3) that work downstream of protein tyrosine kinases in a wide variety of cellular processes. Recent work with genetically modified mouse models has revealed that these proteins play key signaling roles in vascular smooth and skeletal muscle cells, specific neuronal subtypes, and glia cells. These functions, in turn, ensure the proper regulation of blood pressure levels, skeletal muscle mass, axonal wiring, and fiber myelination events as well as systemic metabolic balance. The study of these mice has also led to the discovery of new physiological interconnection among tissues that contribute to the ontogeny and progression of different pathologies such as, for example, hypertension, cardiovascular disease, and metabolic syndrome. Here, we provide an integrated view of all these new Vav family-dependent signaling and physiological functions.
Collapse
Affiliation(s)
- Sonia Rodríguez-Fdez
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain; (S.R.-F.); (L.F.L.-M.); (S.F.); (M.M.-M.); (V.S.); (M.D.)
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, 37007 Salamanca, Spain
| | - L. Francisco Lorenzo-Martín
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain; (S.R.-F.); (L.F.L.-M.); (S.F.); (M.M.-M.); (V.S.); (M.D.)
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, 37007 Salamanca, Spain
| | - Salvatore Fabbiano
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain; (S.R.-F.); (L.F.L.-M.); (S.F.); (M.M.-M.); (V.S.); (M.D.)
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain
| | - Mauricio Menacho-Márquez
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain; (S.R.-F.); (L.F.L.-M.); (S.F.); (M.M.-M.); (V.S.); (M.D.)
- Instituto de Inmunología Clínica y Experimental, CONICET, Rosario 3100, Argentina
| | - Vincent Sauzeau
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain; (S.R.-F.); (L.F.L.-M.); (S.F.); (M.M.-M.); (V.S.); (M.D.)
- Institut du Thorax, UMR1087 CNRS 6291, INSERM, Université de Nantes, 44096 Nantes, France
| | - Mercedes Dosil
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain; (S.R.-F.); (L.F.L.-M.); (S.F.); (M.M.-M.); (V.S.); (M.D.)
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, 37007 Salamanca, Spain
| | - Xosé R. Bustelo
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain; (S.R.-F.); (L.F.L.-M.); (S.F.); (M.M.-M.); (V.S.); (M.D.)
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, 37007 Salamanca, Spain
- Correspondence: ; Tel.: +34-663-194-634
| |
Collapse
|
15
|
CAPS1 is involved in hippocampal synaptic plasticity and hippocampus-associated learning. Sci Rep 2021; 11:8656. [PMID: 33883618 PMCID: PMC8060421 DOI: 10.1038/s41598-021-88009-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 04/07/2021] [Indexed: 12/14/2022] Open
Abstract
Calcium-dependent activator protein for secretion 1 (CAPS1) is a key molecule in vesicular exocytosis, probably in the priming step. However, CAPS1's role in synaptic plasticity and brain function is elusive. Herein, we showed that synaptic plasticity and learning behavior were impaired in forebrain and/or hippocampus-specific Caps1 conditional knockout (cKO) mice by means of molecular, physiological, and behavioral analyses. Neonatal Caps1 cKO mice showed a decrease in the number of docked vesicles in the hippocampal CA3 region, with no detectable changes in the distribution of other major exocytosis-related molecules. Additionally, long-term potentiation (LTP) was partially and severely impaired in the CA1 and CA3 regions, respectively. CA1 LTP was reinforced by repeated high-frequency stimuli, whereas CA3 LTP was completely abolished. Accordingly, hippocampus-associated learning was severely impaired in adeno-associated virus (AAV) infection-mediated postnatal Caps1 cKO mice. Collectively, our findings suggest that CAPS1 is a key protein involved in the cellular mechanisms underlying hippocampal synaptic release and plasticity, which is crucial for hippocampus-associated learning.
Collapse
|
16
|
CAPS2 Deficiency Impairs the Release of the Social Peptide Oxytocin, as Well as Oxytocin-Associated Social Behavior. J Neurosci 2021; 41:4524-4535. [PMID: 33846232 DOI: 10.1523/jneurosci.3240-20.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 03/04/2021] [Accepted: 04/02/2021] [Indexed: 11/21/2022] Open
Abstract
Ca2+-dependent activator protein for secretion 2 (CAPS2) regulates dense-core vesicle (DCV) exocytosis to facilitate peptidergic and catecholaminergic transmitter release. CAPS2 deficiency in mice has mild neuronal effects but markedly impairs social behavior. Rare de novo Caps2 alterations also occur in autism spectrum disorder, although whether CAPS2-mediated release influences social behavior remains unclear. Here, we demonstrate that CAPS2 is associated with DCV exocytosis-mediated release of the social interaction modulatory peptide oxytocin (OXT). CAPS2 is expressed in hypothalamic OXT neurons and localizes to OXT nerve projection and OXT release sites, such as the pituitary. Caps2 KO mice exhibited reduced plasma albeit increased hypothalamic and pituitary OXT levels, indicating insufficient release. OXT neuron-specific Caps2 conditional KO supported CAPS2 function in pituitary OXT release, also affording impaired social interaction and recognition behavior that could be ameliorated by exogenous OXT administered intranasally. Thus, CAPS2 appears critical for OXT release, thereby being associated with social behavior.SIGNIFICANCE STATEMENT The role of the neuropeptide oxytocin in enhancing social interaction and social bonding behavior has attracted considerable public and neuroscientific attention. A central issue in oxytocin biology concerns how oxytocin release is regulated. Our study provides an important insight into the understanding of oxytocin-dependent social behavior from the perspective of the CAPS2-regulated release mechanism.
Collapse
|
17
|
Anti-epileptogenic effect of NRP2945 in the pilocarpine model of temporal lobe epilepsy. Eur J Pharmacol 2021; 901:174068. [PMID: 33798600 DOI: 10.1016/j.ejphar.2021.174068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 03/19/2021] [Accepted: 03/25/2021] [Indexed: 11/21/2022]
Abstract
Innovative therapeutic strategies are highly needed to tackle the major medical needs of epilepsy, like prevention of epilepsy development in at-risk individuals, treatment of severe and drug-resistant forms, control of co-morbidities. The Neural Regeneration Peptide NRP2945 (a peptidomimetic analogue of the human CAPS-2 protein) has been recently found to exert many potentially anti-epileptic effects, for example increased neuronal survival and differentiation. In the present study, we tested the effects of NRP2945 on the development of epilepsy (epileptogenesis) and on chronic, spontaneous seizures, by using the pilocarpine model of temporal lobe epilepsy. We found that NRP2945 exerts a robust anti-epileptogenic effect, reducing the frequency of spontaneous seizures, exerting a significant neuroprotective effect and attenuating anxiety-like behaviors and cognitive impairment. These effects appear to depend on modulation of the epileptogenesis process and not on seizure suppression, because NRP2945 did not reduce frequency or duration of spontaneous seizures when administered to already epileptic animals. These findings may form the basis for a preventive therapy for individuals at-risk of developing epilepsy.
Collapse
|
18
|
Sugimoto H, Horii T, Hirota JN, Sano Y, Shinoda Y, Konno A, Hirai H, Ishizaki Y, Hirase H, Hatada I, Furuichi T, Sadakata T. The Ser19Stop single nucleotide polymorphism (SNP) of human PHYHIPL affects the cerebellum in mice. Mol Brain 2021; 14:52. [PMID: 33712038 PMCID: PMC7953787 DOI: 10.1186/s13041-021-00766-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 03/03/2021] [Indexed: 11/12/2022] Open
Abstract
The HapMap Project is a major international research effort to construct a resource to facilitate the discovery of relationships between human genetic variations and health and disease. The Ser19Stop single nucleotide polymorphism (SNP) of human phytanoyl-CoA hydroxylase-interacting protein-like (PHYHIPL) gene was detected in HapMap project and registered in the dbSNP. PHYHIPL gene expression is altered in global ischemia and glioblastoma multiforme. However, the function of PHYHIPL is unknown. We generated PHYHIPL Ser19Stop knock-in mice and found that PHYHIPL impacts the morphology of cerebellar Purkinje cells (PCs), the innervation of climbing fibers to PCs, the inhibitory inputs to PCs from molecular layer interneurons, and motor learning ability. Thus, the Ser19Stop SNP of the PHYHIPL gene may be associated with cerebellum-related diseases.
Collapse
Affiliation(s)
- Hisako Sugimoto
- Education and Research Support Center, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Takuro Horii
- Laboratory of Genome Science, Biosignal Genome Resource Center, Institute for Molecular and Cellular Regulation, Gunma University, 3-39-15 Showa-machi, Maebashi, 371-8512, Japan
| | - Jun-Na Hirota
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Yoshitake Sano
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Yo Shinoda
- Department of Environmental Health, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, 192-0392, Japan
| | - Ayumu Konno
- Department of Neurophysiology and Neural Repair, Gunma University Graduate School of Medicine, Maebashi, Gunma, 371-8511, Japan
| | - Hirokazu Hirai
- Department of Neurophysiology and Neural Repair, Gunma University Graduate School of Medicine, Maebashi, Gunma, 371-8511, Japan
| | - Yasuki Ishizaki
- Department of Molecular and Cellular Neurobiology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Hajime Hirase
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen N, Denmark
| | - Izuho Hatada
- Laboratory of Genome Science, Biosignal Genome Resource Center, Institute for Molecular and Cellular Regulation, Gunma University, 3-39-15 Showa-machi, Maebashi, 371-8512, Japan
| | - Teiichi Furuichi
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Tetsushi Sadakata
- Education and Research Support Center, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan.
| |
Collapse
|
19
|
Yoo H, Yang SH, Kim JY, Yang E, Park HS, Lee SJ, Rhyu IJ, Turecki G, Lee HW, Kim H. Down-regulation of habenular calcium-dependent secretion activator 2 induces despair-like behavior. Sci Rep 2021; 11:3700. [PMID: 33580180 PMCID: PMC7881199 DOI: 10.1038/s41598-021-83310-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 01/29/2021] [Indexed: 01/07/2023] Open
Abstract
Calcium-dependent secretion activator 2 (CAPS2) regulates the trafficking and exocytosis of neuropeptide-containing dense-core vesicles (DCVs). CAPS2 is prominently expressed in the medial habenula (MHb), which is related to depressive behavior; however, how MHb neurons cause depressive symptoms and the role of CAPS2 remains unclear. We hypothesized that dysfunction of MHb CAPS neurons might cause defects in neuropeptide secretion and the activity of monoaminergic centers, resulting in depressive-like behaviors. In this study, we examined (1) CAPS2 expression in the habenula of depression animal models and major depressive disorder patients and (2) the effects of down-regulation of MHb CAPS2 on the animal behaviors, synaptic transmission in the interpeduncular nucleus (IPN), and neuronal activity of monoamine centers. Habenular CAPS2 expression was decreased in the rat chronic restraint stress model, mouse learned helplessness model, and showed tendency to decrease in depression patients who died by suicide. Knockdown of CAPS2 in the mouse habenula evoked despair-like behavior and a reduction of the release of DCVs in the IPN. Neuronal activity of IPN and monoaminergic centers was also reduced. These results implicate MHb CAPS2 as playing a pivotal role in depressive behavior through the regulation of neuropeptide secretion of the MHb-IPN pathway and the activity of monoaminergic centers.
Collapse
Affiliation(s)
- Hyeijung Yoo
- Department of Anatomy, College of Medicine, Korea University, Seoul, 02841, Korea
- Department of Biomedical Sciences, Brain Korea 21 FOUR, College of Medicine, Korea University, Seoul, 02841, Korea
| | - Soo Hyun Yang
- Department of Anatomy, College of Medicine, Korea University, Seoul, 02841, Korea
- Department of Biomedical Sciences, Brain Korea 21 FOUR, College of Medicine, Korea University, Seoul, 02841, Korea
| | - Jin Yong Kim
- Department of Anatomy, College of Medicine, Korea University, Seoul, 02841, Korea
| | - Esther Yang
- Department of Anatomy, College of Medicine, Korea University, Seoul, 02841, Korea
- Department of Biomedical Sciences, Brain Korea 21 FOUR, College of Medicine, Korea University, Seoul, 02841, Korea
| | - Hyung Sun Park
- Department of Anatomy, College of Medicine, Korea University, Seoul, 02841, Korea
| | - Se Jeong Lee
- Department of Anatomy, College of Medicine, Korea University, Seoul, 02841, Korea
| | - Im Joo Rhyu
- Department of Anatomy, College of Medicine, Korea University, Seoul, 02841, Korea
- Department of Biomedical Sciences, Brain Korea 21 FOUR, College of Medicine, Korea University, Seoul, 02841, Korea
| | - Gustavo Turecki
- Department of Psychiatry, McGill University, Douglas, Mental Health University Institute, Montreal, QC, H4H 1R3, Canada
| | - Hyun Woo Lee
- Department of Anatomy, College of Medicine, Korea University, Seoul, 02841, Korea.
- Department of Biomedical Sciences, Brain Korea 21 FOUR, College of Medicine, Korea University, Seoul, 02841, Korea.
| | - Hyun Kim
- Department of Anatomy, College of Medicine, Korea University, Seoul, 02841, Korea.
- Department of Biomedical Sciences, Brain Korea 21 FOUR, College of Medicine, Korea University, Seoul, 02841, Korea.
| |
Collapse
|
20
|
Huang WQ, Lin Q, Chen S, Sun L, Chen Q, Yi K, Li Z, Ma Q, Tzeng CM. Integrated analysis of microRNA and mRNA expression profiling identifies BAIAP3 as a novel target of dysregulated hsa-miR-1972 in age-related white matter lesions. Aging (Albany NY) 2021; 13:4674-4695. [PMID: 33561007 PMCID: PMC7906144 DOI: 10.18632/aging.202562] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 11/18/2020] [Indexed: 11/25/2022]
Abstract
White matter lesions known as leukoaraiosis (LA) are cerebral white matter hyperintensities observed in elderly individuals. Currently, no reliable molecular biomarkers are available for monitoring their progression over time. To identify biomarkers for the onset and progression of LA, we analyzed whole blood-based, microRNA expression profiles of leukoaraiosis, validated those exhibiting significant microRNA changes in clinical subjects by means of quantitative real-time polymerase chain reactions and determined the function of miRNA in cell lines by means of microRNA mimic transfection assays. A total of seven microRNAs were found to be significantly down-regulated in leukoaraiosis. Among the microRNAs, hsa-miR-1972 was downregulated during the early onset phase of leukoaraiosis, as confirmed in independent patients, and it was found to target leukoaraiosis-dependent BAIAP3, decreasing its expression in 293T cell lines. Functional enrichment analysis revealed that significantly dysregulated miRNAs-mRNAs changes associated with the onset of leukoaraiosis were involved in neurogenesis, neuronal development, and differentiation. Taken together, the study identified a set of candidate microRNA biomarkers that may usefully monitor the onset and progression of leukoaraiosis. Given the enrichment of leukoaraiosis-associated microRNAs and mRNAs in neuron part and membrane system, BAIAP3 could potentially represent a novel target of hsa-miR-1972 in leukoaraiosis through which microRNAs are involved in the pathogenesis of white matter lesions.
Collapse
Affiliation(s)
- Wen-Qing Huang
- Shanghai Institute of Precision Medicine (SHIPM), Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Translational Medicine Research Center (TMRC), School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Qing Lin
- Department of Neurology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China.,Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China.,School of Medicine, Xiamen University, Xiamen, Fujian, China.,The First Clinical College of Fujian Medical University, Fuzhou, Fujian, China
| | - Shuai Chen
- Department of Otolaryngology-Head and Neck Surgery, Xiamen Key Laboratory of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China.,Chen Zhi-nan Academician Workstation, Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, Shanxi, China
| | - Lixiang Sun
- Translational Medicine Research Center (TMRC), School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Qingjie Chen
- Department of Nuclear Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Kehui Yi
- Department of Neurology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China.,Department of Neurology, Zhongshan Xiamen Hospital, Fudan University, Xiamen, Fujian, China
| | - Zhi Li
- Translational Medicine Research Center (TMRC), School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Qilin Ma
- Department of Neurology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China.,Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China.,School of Medicine, Xiamen University, Xiamen, Fujian, China.,The First Clinical College of Fujian Medical University, Fuzhou, Fujian, China
| | - Chi-Meng Tzeng
- Translational Medicine Research Center (TMRC), School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China.,College of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, Jiangsu, China
| |
Collapse
|
21
|
Fujima S, Amemiya N, Arima T, Sano Y, Furuichi T. CAPS2 deficiency induces proopiomelanocortin accumulation in pituitary and affects food intake behavior in mice. Neurosci Lett 2020; 738:135335. [PMID: 32891671 DOI: 10.1016/j.neulet.2020.135335] [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: 03/16/2020] [Revised: 08/11/2020] [Accepted: 08/26/2020] [Indexed: 10/23/2022]
Abstract
Proopiomelanocortin (POMC) is a neuropeptide precursor produced in the anterior and intermediate pituitary lobes, the hypothalamic arcuate nucleus (ARC), and solitary tract nucleus. Alpha-melanocyte-stimulating hormone (α-MSH) is a cell type specific POMC derivative that is essential for regulating feeding, and energy homeostasis. However, the molecular mechanism underlying POMC/α-MSH secretion remains unclear. Ca2+-dependent activator protein for secretion 2 (CAPS2) is a regulatory protein involved in the exocytosis of dense-core vesicles containing neuropeptides. We previously reported CAPS2 localization in the intermediate pituitary lobe and reduced body weights in Caps2-knockout (Caps2-KO) mice, compared to control mice. Here, we aimed to investigate CAPS2 expression in POMC-expressing neurons and the effects of CAPS2 deficiency on the secretion of POMC-related peptides and feeding behavior phenotype. CAPS2 was localized in the POMC-expressing neurons of the intermediate pituitary lobe, hypothalamic ARC, and the paraventricular nucleus, which is innervated by hypothalamic neurons. POMC protein levels in the intermediate pituitary lobe of Caps2-KO mice were significantly higher than that in the control mice, suggesting a possible accumulation of POMC-derived peptides in the intermediate pituitary lobe of Caps2-KO mice. Moreover, administration of low-dose melanotan-2, an α-MSH receptor (MC4R) agonist, decreased food intake per body weight in Caps2-KO mice; no such effect was observed in the wildtype mice. Collectively, these results suggest that CAPS2 is involved in regulating the secretion of POMC-derived peptides, including α-MSH, is partially associated with feeding, and affects energy metabolism.
Collapse
Affiliation(s)
- Shuhei Fujima
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Noda, Chiba 278-8510, Japan
| | - Natsuki Amemiya
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Noda, Chiba 278-8510, Japan
| | - Tomoki Arima
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Noda, Chiba 278-8510, Japan
| | - Yoshitake Sano
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Noda, Chiba 278-8510, Japan
| | - Teiichi Furuichi
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Noda, Chiba 278-8510, Japan.
| |
Collapse
|
22
|
Journey of brain-derived neurotrophic factor: from intracellular trafficking to secretion. Cell Tissue Res 2020; 382:125-134. [PMID: 32897423 DOI: 10.1007/s00441-020-03274-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 08/07/2020] [Indexed: 12/14/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is known to control a wide variety of brain functions, ranging from memory formation to food intake. However, since the BDNF levels are extremely low in the nervous system, the dynamics in neurons from intracellular trafficking to secretion is absolutely complicated; the understanding is not fully promoted. We here review the findings of those critical mechanisms from intracellular trafficking to the secretion of BDNF. Furthermore, to solve this issue, technological advances for the detection, measurement, and imaging of this growth factor are essential. We believe that this review helps the study of these complex but critical mechanisms of BDNF.
Collapse
|
23
|
Refinement of Cerebellar Network Organization by Extracellular Signaling During Development. Neuroscience 2020; 462:44-55. [PMID: 32502568 DOI: 10.1016/j.neuroscience.2020.05.036] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 05/20/2020] [Accepted: 05/21/2020] [Indexed: 12/21/2022]
Abstract
The cerebellum forms regular neural network structures consisting of a few major types of neurons, such as Purkinje cells, granule cells, and molecular layer interneurons, and receives two major inputs from climbing fibers and mossy fibers. Its regular structures consist of three well-defined layers, with each type of neuron designated to a specific location and forming specific synaptic connections. During the first few weeks of postnatal development in rodents, the cerebellum goes through dynamic changes via proliferation, migration, differentiation, synaptogenesis, and maturation, to create such a network structure. The development of this organized network structure presumably relies on the communication between developing elements in the network, including not only individual neurons, but also their dendrites, axons, and synapses. Therefore, it is reasonable that extracellular signaling via synaptic transmission, secreted molecules, and cell adhesion molecules, plays important roles in cerebellar network development. Although it is not yet clear as to how overall cerebellar development is orchestrated, there is indeed accumulating lines of evidence that extracellular signaling acts toward the development of individual elements in the cerebellar networks. In this article, we introduce what we have learned from many studies regarding the extracellular signaling required for cerebellar network development, including our recent study suggesting the importance of unbiased synaptic inputs from parallel fibers.
Collapse
|
24
|
Grandi FC, De Tomasi L, Mustapha M. Single-Cell RNA Analysis of Type I Spiral Ganglion Neurons Reveals a Lmx1a Population in the Cochlea. Front Mol Neurosci 2020; 13:83. [PMID: 32523514 PMCID: PMC7261882 DOI: 10.3389/fnmol.2020.00083] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 04/24/2020] [Indexed: 12/13/2022] Open
Abstract
In the mature cochlea, each inner hair cell (IHC) is innervated by multiple spiral ganglion neurons of type I (SGNI). SGNIs are morphologically and electro-physiologically diverse. Also, they differ in their susceptibility to noise insult. However, the molecular underpinnings of their identity and physiological differences remain poorly understood. In this study, we developed a novel triple transgenic mouse, which enabled the isolation of pure populations of SGNIs and the analysis of a 96-gene panel via single-cell qPCR. We found three distinct populations of Type I SGNs, which were marked by their exclusive expression of Lmx1a, Slc4a4, or Mfap4/Fzd2, respectively, at postnatal days P3, P8, and P12. Our data suggest that afferent SGN subtypes are established genetically before the onset of hearing and that the expression of key physiological markers, such as ion channels, is heterogeneous and may be underlying the heterogeneous firing proprieties of SGNIs.
Collapse
Affiliation(s)
| | - Lara De Tomasi
- Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom
| | - Mirna Mustapha
- Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom.,Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
25
|
Xiao R, Zhong H, Li X, Ma Y, Zhang R, Wang L, Zang Z, Fan X. Abnormal Cerebellar Development Is Involved in Dystonia-Like Behaviors and Motor Dysfunction of Autistic BTBR Mice. Front Cell Dev Biol 2020; 8:231. [PMID: 32318573 PMCID: PMC7154340 DOI: 10.3389/fcell.2020.00231] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 03/18/2020] [Indexed: 12/12/2022] Open
Abstract
Motor control and learning impairments are common complications in individuals with autism spectrum disorder (ASD). Abnormal cerebellar development during critical phases may disrupt these motor functions and lead to autistic motor dysfunction. However, the underlying mechanisms behind these impairments are not clear. Here, we utilized BTBR T+ Itprtf/J (BTBR) mice, an animal model of autism, to investigate the involvement of abnormal cerebellar development in motor performance. We found BTBR mice exhibited severe dystonia-like behavior and motor coordination or motor learning impairments. The onset of these abnormal movements coincided with the increased proliferation of granule neurons and enhanced foliation, and Purkinje cells displayed morphological hypotrophy with increased dendritic spine formation but suppressed maturation. The migration of granule neurons seemed unaffected. Transcriptional analyses confirmed the differential expression of genes involved in abnormal neurogenesis and revealed TRPC as a critical regulator in proliferation and synaptic formation. Taken together, these findings indicate that abnormal cerebellar development is closely related to dystonia-like behavior and motor dysfunction of BTBR mice and that TRPC may be a novel risk gene for ASD that may participate in the pathological process of autistic movement disorders.
Collapse
Affiliation(s)
- Rui Xiao
- Department of Military Cognitive Psychology, School of Psychology, Army Medical University, Chongqing, China
| | - Hongyu Zhong
- Department of Military Cognitive Psychology, School of Psychology, Army Medical University, Chongqing, China
| | - Xin Li
- Department of Military Cognitive Psychology, School of Psychology, Army Medical University, Chongqing, China
| | - Yuanyuan Ma
- Department of Military Cognitive Psychology, School of Psychology, Army Medical University, Chongqing, China.,Department of Basic Nursing, School of Nursing, Army Medical University, Chongqing, China
| | - Ruiyu Zhang
- Department of Military Cognitive Psychology, School of Psychology, Army Medical University, Chongqing, China
| | - Lian Wang
- Department of Military Cognitive Psychology, School of Psychology, Army Medical University, Chongqing, China
| | - Zhenle Zang
- Department of Military Cognitive Psychology, School of Psychology, Army Medical University, Chongqing, China
| | - Xiaotang Fan
- Department of Military Cognitive Psychology, School of Psychology, Army Medical University, Chongqing, China
| |
Collapse
|
26
|
Hong S, Lee SJ, Cho SR. Chromosomal Deletion in 7q31.2-31.32 Involving Ca2+-Dependent Activator Protein for Secretion Gene in a Patient with Cerebellar Ataxia: a Case Report. BRAIN & NEUROREHABILITATION 2019; 13:e9. [PMID: 36744273 PMCID: PMC9879524 DOI: 10.12786/bn.2020.13.e9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 11/12/2019] [Accepted: 11/20/2019] [Indexed: 11/08/2022] Open
Abstract
We present a 33-year-old male patient with cerebellar ataxia. He was first considered to have a psychiatric conversion disorder but finally found to have chromosomal deletion in 7q31.2-31.32 involving Ca2+-dependent activator protein for secretion (CADPS) gene. When a targeted gene sequencing using next-generation sequencing panel and chromosomal microarray analysis were performed, an 8.6 Mb deletion within chromosome 7q31.2-31.32 was discovered. Deletion of CADPS gene in the 7q31.2-31.32 was suggested as the causative factor of cerebellar ataxia. Functional levels evaluated by Berg balance scale and modified Barthel index were improved via comprehensive rehabilitation including balance training and a dopamine agonist medication. To the best of our knowledge, this is the first report of chromosomal deletion in 7q31.2-31.32 including CADPS gene detected in patients with cerebellar ataxia.
Collapse
Affiliation(s)
- Seungbeen Hong
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Su Ji Lee
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Sung-Rae Cho
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Korea
| |
Collapse
|
27
|
Pavone P, Corsello G, Marino SD, Ruggieri M, Falsaperla R. 7q31.32 partial duplication: First report of a child with dysmorphism, autistic spectrum disorder, moderate intellectual disability and, epilepsy. Literature review. Epilepsy Res 2019; 158:106223. [PMID: 31707317 DOI: 10.1016/j.eplepsyres.2019.106223] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 09/10/2019] [Accepted: 10/19/2019] [Indexed: 10/25/2022]
Abstract
INTRODUCTION Duplication of long arm of chromosome 7(q) is uncommon. It may occur as "pure", isolated anomaly or in association with other mutations involving the same or other chromosomes. "Pure" chromosome 7q duplication has recently been classified by segment involved: the interstitial, proximal, or distal segment of the arm. Attempts to correlate genotype with phenotype in each group has yielded questionable results even though intellective disability and minor dysmorphic features of variable types are typically seen. MATERIAL AND METHODS In a young boy showing minor facial dysmorphism, language delay, autistic spectrum disorder, epileptic seizures, behavioral disturbances and irritability an array-CGH analysis was carried out. RESULTS Array-CGH analysis found in the proband a de novo variant of partial duplication of 7q31.32 (122.254.792-122.376.908). DISCUSSION A very few cases of partial 7q duplication have been reported thus far mainly presenting with clinical signs of dysmorphic features, large head, developmental delay, epileptic seizures and skeletal anomalies. To our knowledge, this is the first report of a case of a de novo variant of 7q31.32 duplication, showing dysmorphic anomalies and neurologic impairment including ASD and seizures. In the 7q31.32 region is located the gene CADPS2, which has been associated to autistic spectrum disorder and other neurologic disorders. In the child, a genotype-phenotype correlation may be hypothesized. Further similar reports may be useful to confirm this observation.
Collapse
Affiliation(s)
- Piero Pavone
- Department of Pediatrics, University Hospital. "Vittorio Emanuele-Policlinico" Catania, Italy.
| | - Giovanni Corsello
- Department of Maternal and Child Health, University of Palermo, Italy
| | - Simona Domenica Marino
- Pediatrics and Pediatric Emergency Complex Unity, University-Hospital "Policlinico-Vittorio Emanuele", Catania, Italy
| | - Martino Ruggieri
- Unit of Rare Diseases of the Nervous System in childhood, Department of Clinical and Experimental Medicine, Section of Pediatrics and Child Neuropsychiatry, A.U.O. Vittorio Emanuele-Policlinico of Catania, Italy
| | - Raffaele Falsaperla
- Pediatrics and Pediatric Emergency Complex Unity, University-Hospital "Policlinico-Vittorio Emanuele", Catania, Italy
| |
Collapse
|
28
|
Alonso-Gonzalez A, Calaza M, Rodriguez-Fontenla C, Carracedo A. Gene-based analysis of ADHD using PASCAL: a biological insight into the novel associated genes. BMC Med Genomics 2019; 12:143. [PMID: 31651322 PMCID: PMC6813133 DOI: 10.1186/s12920-019-0593-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 09/24/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Attention-Deficit Hyperactivity Disorder (ADHD) is a complex neurodevelopmental disorder (NDD) which may significantly impact on the affected individual's life. ADHD is acknowledged to have a high heritability component (70-80%). Recently, a meta-analysis of GWAS (Genome Wide Association Studies) has demonstrated the association of several independent loci. Our main aim here, is to apply PASCAL (pathway scoring algorithm), a new gene-based analysis (GBA) method, to the summary statistics obtained in this meta-analysis. PASCAL will take into account the linkage disequilibrium (LD) across genomic regions in a different way than the most commonly employed GBA methods (MAGMA or VEGAS (Versatile Gene-based Association Study)). In addition to PASCAL analysis a gene network and an enrichment analysis for KEGG and GO terms were carried out. Moreover, GENE2FUNC tool was employed to create gene expression heatmaps and to carry out a (DEG) (Differentially Expressed Gene) analysis using GTEX v7 and BrainSpan data. RESULTS PASCAL results have revealed the association of new loci with ADHD and it has also highlighted other genes previously reported by MAGMA analysis. PASCAL was able to discover new associations at a gene level for ADHD: FEZF1 (p-value: 2.2 × 10- 7) and FEZF1-AS1 (p-value: 4.58 × 10- 7). In addition, PASCAL has been able to highlight association of other genes that share the same LD block with some previously reported ADHD susceptibility genes. Gene network analysis has revealed several interactors with the associated ADHD genes and different GO and KEGG terms have been associated. In addition, GENE2FUNC has demonstrated the existence of several up and down regulated expression clusters when the associated genes and their interactors were considered. CONCLUSIONS PASCAL has been revealed as an efficient tool to extract additional information from previous GWAS using their summary statistics. This study has identified novel ADHD associated genes that were not previously reported when other GBA methods were employed. Moreover, a biological insight into the biological function of the ADHD associated genes across brain regions and neurodevelopmental stages is provided.
Collapse
Affiliation(s)
- Aitana Alonso-Gonzalez
- Grupo de Medicina Xenómica, Fundación Instituto de Investigación Sanitaria de Santiago de Compostela (FIDIS), Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidad de Santiago de Compostela, Santiago de Compostela, Spain
| | - Manuel Calaza
- Grupo de Medicina Xenómica, Fundación Instituto de Investigación Sanitaria de Santiago de Compostela (FIDIS), Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidad de Santiago de Compostela, Santiago de Compostela, Spain
| | - Cristina Rodriguez-Fontenla
- Grupo de Medicina Genómica, CIBERER, CIMUS (Centre for Research in Molecular Medicine and Chronic Diseases), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Angel Carracedo
- Grupo de Medicina Xenómica, Fundación Instituto de Investigación Sanitaria de Santiago de Compostela (FIDIS), Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidad de Santiago de Compostela, Santiago de Compostela, Spain
- Grupo de Medicina Genómica, CIBERER, CIMUS (Centre for Research in Molecular Medicine and Chronic Diseases), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| |
Collapse
|
29
|
Castora FJ. Mitochondrial function and abnormalities implicated in the pathogenesis of ASD. Prog Neuropsychopharmacol Biol Psychiatry 2019; 92:83-108. [PMID: 30599156 DOI: 10.1016/j.pnpbp.2018.12.015] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 12/20/2018] [Accepted: 12/24/2018] [Indexed: 12/18/2022]
Abstract
Mitochondria are the powerhouse that generate over 90% of the ATP produced in cells. In addition to its role in energy production, the mitochondrion also plays a major role in carbohydrate, fatty acid, amino acid and nucleotide metabolism, programmed cell death (apoptosis), generation of and protection against reactive oxygen species (ROS), immune response, regulation of intracellular calcium ion levels and even maintenance of gut microbiota. With its essential role in bio-energetic as well as non-energetic biological processes, it is not surprising that proper cellular, tissue and organ function is dependent upon proper mitochondrial function. Accordingly, mitochondrial dysfunction has been shown to be directly linked to a variety of medical disorders, particularly neuromuscular disorders and increasing evidence has linked mitochondrial dysfunction to neurodegenerative and neurodevelopmental disorders such as Alzheimer's Disease (AD), Parkinson's Disease (PD), Rett Syndrome (RS) and Autism Spectrum Disorders (ASD). Over the last 40 years there has been a dramatic increase in the diagnosis of ASD and, more recently, an increasing body of evidence indicates that mitochondrial dysfunction plays an important role in ASD development. In this review, the latest evidence linking mitochondrial dysfunction and abnormalities in mitochondrial DNA (mtDNA) to the pathogenesis of autism will be presented. This review will also summarize the results of several recent `approaches used for improving mitochondrial function that may lead to new therapeutic approaches to managing and/or treating ASD.
Collapse
Affiliation(s)
- Frank J Castora
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA, USA; Department of Neurology, Eastern Virginia Medical School, Norfolk, VA, USA.
| |
Collapse
|
30
|
Crummy E, Mani M, Thellman JC, Martin TFJ. The priming factor CAPS1 regulates dense-core vesicle acidification by interacting with rabconnectin3β/WDR7 in neuroendocrine cells. J Biol Chem 2019; 294:9402-9415. [PMID: 31004036 PMCID: PMC6579465 DOI: 10.1074/jbc.ra119.007504] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 03/21/2019] [Indexed: 12/20/2022] Open
Abstract
Vacuolar-type H+-ATPases (V-ATPases) contribute to pH regulation and play key roles in secretory and endocytic pathways. Dense-core vesicles (DCVs) in neuroendocrine cells are maintained at an acidic pH, which is part of the electrochemical driving force for neurotransmitter loading and is required for hormonal propeptide processing. Genetic loss of CAPS1 (aka calcium-dependent activator protein for secretion, CADPS), a vesicle-bound priming factor required for DCV exocytosis, dissipates the pH gradient across DCV membranes and reduces neurotransmitter loading. However, the basis for CAPS1 binding to DCVs and for its regulation of vesicle pH has not been determined. Here, MS analysis of CAPS1 immunoprecipitates from brain membrane fractions revealed that CAPS1 associates with a rabconnectin3 (Rbcn3) complex comprising Dmx-like 2 (DMXL2) and WD repeat domain 7 (WDR7) proteins. Using immunofluorescence microscopy, we found that Rbcn3α/DMXL2 and Rbcn3β/WDR7 colocalize with CAPS1 on DCVs in human neuroendocrine (BON) cells. The shRNA-mediated knockdown of Rbcn3β/WDR7 redistributed CAPS1 from DCVs to the cytosol, indicating that Rbcn3β/WDR7 is essential for optimal DCV localization of CAPS1. Moreover, cell-free experiments revealed direct binding of CAPS1 to Rbcn3β/WDR7, and cell assays indicated that Rbcn3β/WDR7 recruits soluble CAPS1 to membranes. As anticipated by the reported association of Rbcn3 with V-ATPase, we found that knocking down CAPS1, Rbcn3α, or Rbcn3β in neuroendocrine cells impaired rates of DCV reacidification. These findings reveal a basis for CAPS1 binding to DCVs and for CAPS1 regulation of V-ATPase activity via Rbcn3β/WDR7 interactions.
Collapse
Affiliation(s)
- Ellen Crummy
- From the Department of Biochemistry, University of Wisconsin, Madison, Wisconsin 53706
| | - Muralidharan Mani
- From the Department of Biochemistry, University of Wisconsin, Madison, Wisconsin 53706
| | - John C Thellman
- From the Department of Biochemistry, University of Wisconsin, Madison, Wisconsin 53706
| | - Thomas F J Martin
- From the Department of Biochemistry, University of Wisconsin, Madison, Wisconsin 53706
| |
Collapse
|
31
|
Shinoda Y, Sadakata T, Yagishita K, Kinameri E, Katoh-Semba R, Sano Y, Furuichi T. Aspects of excitatory/inhibitory synapses in multiple brain regions are correlated with levels of brain-derived neurotrophic factor/neurotrophin-3. Biochem Biophys Res Commun 2018; 509:429-434. [PMID: 30594389 DOI: 10.1016/j.bbrc.2018.12.100] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 12/14/2018] [Indexed: 12/29/2022]
Abstract
Appropriate synapse formation during development is necessary for normal brain function, and synapse impairment is often associated with brain dysfunction. Brain-derived neurotrophic factor (BDNF) and neurotrophin-3 (NT-3) are key factors in regulating synaptic development. We previously reported that BDNF/NT-3 secretion was enhanced by calcium-dependent activator protein for secretion 2 (CADPS2). Although BDNF/NT-3 and CADPS2 are co-expressed in various brain regions, the effect of Cadps2-deficiency on brain region-specific BDNF/NT-3 levels and synaptic development remains elusive. Here, we show developmental changes of BDNF/NT-3 levels and we assess disruption of excitatory/inhibitory synapses in multiple brain regions (cerebellum, hypothalamus, striatum, hippocampus, parietal cortex and prefrontal cortex) of Cadps2 knockout (KO) mice compared with wild-type (WT) mice. Compared with WT, BDNF levels in KO mice were reduced in young/adult hippocampus, but increased in young hypothalamus, while NT-3 levels were reduced in adult cerebellum and young hippocampus, but increased in adult parietal cortex. Immunofluorescence of vGluT1, an excitatory synapse marker, and vGAT, an inhibitory synapse marker, in adult KO showed that vGluT1 was higher in the cerebellum and parietal cortex but lower in the hippocampus, whereas vGAT was lower in the hippocampus and parietal cortex compared with WT. Immunolabeling for both vGluT1 and vGAT was increased in the parietal cortex but vGAT was decreased in the cerebellum in adult KO compared with WT. These data suggest that CADPS2-mediated secretion of BDNF/NT-3 may be involved in development and maturation of synapses and in the balance between inhibitory and excitatory synapses.
Collapse
Affiliation(s)
- Yo Shinoda
- Department of Environmental Health, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, 192-0392, Japan; Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Noda, Chiba, 278-8510, Japan; Laboratory for Molecular Neurogenesis, RIKEN Brain Science Institute, Wako, Saitama, 351-0198, Japan.
| | - Tetsushi Sadakata
- Laboratory for Molecular Neurogenesis, RIKEN Brain Science Institute, Wako, Saitama, 351-0198, Japan; Education and Research Support Center, Gunma University Graduate School of Medicine, Maebashi, Gunma, 371-8511, Japan
| | - Kaori Yagishita
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Noda, Chiba, 278-8510, Japan
| | - Emi Kinameri
- Laboratory for Molecular Neurogenesis, RIKEN Brain Science Institute, Wako, Saitama, 351-0198, Japan
| | - Ritsuko Katoh-Semba
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Noda, Chiba, 278-8510, Japan; Laboratory for Molecular Neurogenesis, RIKEN Brain Science Institute, Wako, Saitama, 351-0198, Japan
| | - Yoshitake Sano
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Noda, Chiba, 278-8510, Japan
| | - Teiichi Furuichi
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Noda, Chiba, 278-8510, Japan; Laboratory for Molecular Neurogenesis, RIKEN Brain Science Institute, Wako, Saitama, 351-0198, Japan.
| |
Collapse
|
32
|
Hökfelt T, Barde S, Xu ZQD, Kuteeva E, Rüegg J, Le Maitre E, Risling M, Kehr J, Ihnatko R, Theodorsson E, Palkovits M, Deakin W, Bagdy G, Juhasz G, Prud’homme HJ, Mechawar N, Diaz-Heijtz R, Ögren SO. Neuropeptide and Small Transmitter Coexistence: Fundamental Studies and Relevance to Mental Illness. Front Neural Circuits 2018; 12:106. [PMID: 30627087 PMCID: PMC6309708 DOI: 10.3389/fncir.2018.00106] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 11/05/2018] [Indexed: 12/31/2022] Open
Abstract
Neuropeptides are auxiliary messenger molecules that always co-exist in nerve cells with one or more small molecule (classic) neurotransmitters. Neuropeptides act both as transmitters and trophic factors, and play a role particularly when the nervous system is challenged, as by injury, pain or stress. Here neuropeptides and coexistence in mammals are reviewed, but with special focus on the 29/30 amino acid galanin and its three receptors GalR1, -R2 and -R3. In particular, galanin's role as a co-transmitter in both rodent and human noradrenergic locus coeruleus (LC) neurons is addressed. Extensive experimental animal data strongly suggest a role for the galanin system in depression-like behavior. The translational potential of these results was tested by studying the galanin system in postmortem human brains, first in normal brains, and then in a comparison of five regions of brains obtained from depressed people who committed suicide, and from matched controls. The distribution of galanin and the four galanin system transcripts in the normal human brain was determined, and selective and parallel changes in levels of transcripts and DNA methylation for galanin and its three receptors were assessed in depressed patients who committed suicide: upregulation of transcripts, e.g., for galanin and GalR3 in LC, paralleled by a decrease in DNA methylation, suggesting involvement of epigenetic mechanisms. It is hypothesized that, when exposed to severe stress, the noradrenergic LC neurons fire in bursts and release galanin from their soma/dendrites. Galanin then acts on somato-dendritic, inhibitory galanin autoreceptors, opening potassium channels and inhibiting firing. The purpose of these autoreceptors is to act as a 'brake' to prevent overexcitation, a brake that is also part of resilience to stress that protects against depression. Depression then arises when the inhibition is too strong and long lasting - a maladaption, allostatic load, leading to depletion of NA levels in the forebrain. It is suggested that disinhibition by a galanin antagonist may have antidepressant activity by restoring forebrain NA levels. A role of galanin in depression is also supported by a recent candidate gene study, showing that variants in genes for galanin and its three receptors confer increased risk of depression and anxiety in people who experienced childhood adversity or recent negative life events. In summary, galanin, a neuropeptide coexisting in LC neurons, may participate in the mechanism underlying resilience against a serious and common disorder, MDD. Existing and further results may lead to an increased understanding of how this illness develops, which in turn could provide a basis for its treatment.
Collapse
Affiliation(s)
- Tomas Hökfelt
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Swapnali Barde
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Zhi-Qing David Xu
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neurobiology, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Laboratory of Brain Disorders (Ministry of Science and Technology), Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Eugenia Kuteeva
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Joelle Rüegg
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- The Center for Molecular Medicine, Stockholm, Sweden
- Swedish Toxicology Sciences Research Center, Swetox, Södertälje, Sweden
| | - Erwan Le Maitre
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Mårten Risling
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Jan Kehr
- Pronexus Analytical AB, Solna, Sweden
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Robert Ihnatko
- Department of Clinical Chemistry, Linköping University, Linköping, Sweden
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Elvar Theodorsson
- Department of Clinical Chemistry, Linköping University, Linköping, Sweden
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Miklos Palkovits
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - William Deakin
- Neuroscience and Psychiatry Unit, University of Manchester, Manchester, United Kingdom
| | - Gyorgy Bagdy
- Department of Pharmacodynamics, Semmelweis University, Budapest, Hungary
- MTA-SE Neuropsychopharmacology and Neurochemistry Research Group, Hungarian Academy of Sciences, Semmelweis University, Budapest, Hungary
- NAP 2-SE New Antidepressant Target Research Group, Hungarian Brain Research Program, Semmelweis University, Budapest, Hungary
| | - Gabriella Juhasz
- Neuroscience and Psychiatry Unit, University of Manchester, Manchester, United Kingdom
- Department of Pharmacodynamics, Semmelweis University, Budapest, Hungary
- SE-NAP2 Genetic Brain Imaging Migraine Research Group, Hungarian Brain Research Program, Semmelweis University, Budapest, Hungary
| | | | - Naguib Mechawar
- Douglas Hospital Research Centre, Verdun, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | | | - Sven Ove Ögren
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
33
|
Shaib AH, Staudt A, Harb A, Klose M, Shaaban A, Schirra C, Mohrmann R, Rettig J, Becherer U. Paralogs of the Calcium-Dependent Activator Protein for Secretion Differentially Regulate Synaptic Transmission and Peptide Secretion in Sensory Neurons. Front Cell Neurosci 2018; 12:304. [PMID: 30254567 PMCID: PMC6141663 DOI: 10.3389/fncel.2018.00304] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 08/21/2018] [Indexed: 12/29/2022] Open
Abstract
The two paralogs of the calcium-dependent activator protein for secretion (CAPS) are priming factors for synaptic vesicles (SVs) and neuropeptide containing large dense-core vesicles (LDCVs). Yet, it is unclear whether CAPS1 and CAPS2 regulate exocytosis of these two vesicle types differentially in dorsal root ganglion (DRG) neurons, wherein synaptic transmission and neuropeptide release are of equal importance. These sensory neurons transfer information from the periphery to the spinal cord (SC), releasing glutamate as the primary neurotransmitter, with co-transmission via neuropeptides in a subset of so called peptidergic neurons. Neuropeptides are key components of the information-processing machinery of pain perception and neuropathic pain generation. Here, we compared the ability of CAPS1 and CAPS2 to support priming of both vesicle types in single and double knock-out mouse (DRG) neurons using a variety of high-resolution live cell imaging methods. While CAPS1 was localized to synapses of all DRG neurons and promoted synaptic transmission, CAPS2 was found exclusively in peptidergic neurons and mediated LDCV exocytosis. Intriguingly, ectopic expression of CAPS2 empowered non-peptidergic neurons to drive LDCV fusion, thereby identifying CAPS2 as an essential molecular determinant for peptidergic signaling. Our results reveal that these distinct functions of both CAPS paralogs are based on their differential subcellular localization in DRG neurons. Our data suggest a major role for CAPS2 in neuropathic pain via control of neuropeptide release.
Collapse
Affiliation(s)
- Ali H. Shaib
- Department of Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, Homburg, Germany
| | - Angelina Staudt
- Department of Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, Homburg, Germany
| | - Ali Harb
- Department of Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, Homburg, Germany
- ZHMB Junior Group, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, Homburg, Germany
| | - Margarete Klose
- Department of Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, Homburg, Germany
| | - Ahmed Shaaban
- ZHMB Junior Group, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, Homburg, Germany
| | - Claudia Schirra
- Department of Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, Homburg, Germany
| | - Ralf Mohrmann
- ZHMB Junior Group, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, Homburg, Germany
| | - Jens Rettig
- Department of Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, Homburg, Germany
| | - Ute Becherer
- Department of Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, Homburg, Germany
| |
Collapse
|
34
|
Regulation of BDNF Release by ARMS/Kidins220 through Modulation of Synaptotagmin-IV Levels. J Neurosci 2018; 38:5415-5428. [PMID: 29769266 DOI: 10.1523/jneurosci.1653-17.2018] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 04/07/2018] [Accepted: 05/03/2018] [Indexed: 11/21/2022] Open
Abstract
BDNF is a growth factor with important roles in the nervous system in both physiological and pathological conditions, but the mechanisms controlling its secretion are not completely understood. Here, we show that ARMS/Kidins220 negatively regulates BDNF secretion in neurons from the CNS and PNS. Downregulation of the ARMS/Kidins220 protein in the adult mouse brain increases regulated BDNF secretion, leading to its accumulation in the striatum. Interestingly, two mouse models of Huntington's disease (HD) showed increased levels of ARMS/Kidins220 in the hippocampus and regulated BDNF secretion deficits. Importantly, reduction of ARMS/Kidins220 in hippocampal slices from HD mice reversed the impaired regulated BDNF release. Moreover, there are increased levels of ARMS/Kidins220 in the hippocampus and PFC of patients with HD. ARMS/Kidins220 regulates Synaptotagmin-IV levels, which has been previously observed to modulate BDNF secretion. These data indicate that ARMS/Kidins220 controls the regulated secretion of BDNF and might play a crucial role in the pathogenesis of HD.SIGNIFICANCE STATEMENT BDNF is an important growth factor that plays a fundamental role in the correct functioning of the CNS. The secretion of BDNF must be properly controlled to exert its functions, but the proteins regulating its release are not completely known. Using neuronal cultures and a new conditional mouse to modulate ARMS/Kidins220 protein, we report that ARMS/Kidins220 negatively regulates BDNF secretion. Moreover, ARMS/Kidins220 is overexpressed in two mouse models of Huntington's disease (HD), causing an impaired regulation of BDNF secretion. Furthermore, ARMS/Kidins220 levels are increased in brain samples from HD patients. Future studies should address whether ARMS/Kidins220 has any function on the pathophysiology of HD.
Collapse
|
35
|
Shinoda Y, Sadakata T, Akagi T, Sakamaki Y, Hashikawa T, Sano Y, Furuichi T. Calcium-dependent activator protein for secretion 2 (CADPS2) deficiency causes abnormal synapse development in hippocampal mossy fiber terminals. Neurosci Lett 2018; 677:65-71. [PMID: 29689341 DOI: 10.1016/j.neulet.2018.04.036] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 04/06/2018] [Accepted: 04/18/2018] [Indexed: 12/27/2022]
Abstract
Hippocampal mossy fibers (MFs) project from dentate gyrus granule cells onto the CA2-CA3 region. MF-mediated synaptic transmission plays an important role in hippocampal learning and memory. However, the molecular mechanisms underlying MF synaptic development and subsequent functional organization are not fully understood. We previously reported that calcium-dependent activator protein for secretion 2 (CADPS2, also known as CAPS2) regulates the secretion of dense-core vesicles (DCVs). Because CADPS2 is strongly expressed in MF terminals, we hypothesized that CADPS2 regulates the development and functional organization of MF synapses by controlling the secretion of DCVs and their contents. To test this, we compared the synaptic microstructures of hippocampal MF terminals in Cadps2 knockout (KO) mice and wild-type (WT) mice by electron microscopy (EM). On postnatal day 15 (P15), KO mice exhibited morphological abnormalities in MF boutons, including smaller bouton size, a larger number of DCVs and a smaller number of post-synaptic densities (PSDs), compared with WT mice. In adults (P56), MF boutons were larger in KO mice. Synaptic vesicles (SVs) were increased but with a lower density compared with the WT. Furthermore, the number of SVs was decreased near the active zone. Moreover, MF-innervated CA3 postsynapses in KO mice displayed aberrant structures at the postsynaptic density (PSD), with an increased number of PSDs (likely because of a larger number of perforated PSDs), compared with WT mice. Taken together, our findings suggest that CADPS2 plays a critical role in MF synaptic development and functional organization.
Collapse
Affiliation(s)
- Yo Shinoda
- Department of Environmental Health, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan; Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Noda, Chiba 278-8510, Japan; Laboratory for Molecular Neurogenesis, RIKEN Brain Science Institute, Wako, Saitama 351-0198, Japan.
| | - Tetsushi Sadakata
- Laboratory for Molecular Neurogenesis, RIKEN Brain Science Institute, Wako, Saitama 351-0198, Japan; Education and Research Support Center, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Takumi Akagi
- Research Resource Center, RIKEN Brain Science Institute, Wako, Saitama 351-0106, Japan; Department of Physiology, Nippon Medical School, Bunkyo-ku, Tokyo 113-8602, Japan
| | - Yuriko Sakamaki
- Research Resource Center, RIKEN Brain Science Institute, Wako, Saitama 351-0106, Japan; Research Core, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Tsutomu Hashikawa
- Research Resource Center, RIKEN Brain Science Institute, Wako, Saitama 351-0106, Japan; Laboratory for Molecular Mechanisms of Thalamus Development, RIKEN Brain Science Institute, Wako, Saitama 351-0198, Japan
| | - Yoshitake Sano
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Noda, Chiba 278-8510, Japan
| | - Teiichi Furuichi
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Noda, Chiba 278-8510, Japan; Laboratory for Molecular Neurogenesis, RIKEN Brain Science Institute, Wako, Saitama 351-0198, Japan.
| |
Collapse
|
36
|
Hou H, Uusküla-Reimand L, Makarem M, Corre C, Saleh S, Metcalf A, Goldenberg A, Palmert MR, Wilson MD. Gene expression profiling of puberty-associated genes reveals abundant tissue and sex-specific changes across postnatal development. Hum Mol Genet 2018; 26:3585-3599. [PMID: 28911201 DOI: 10.1093/hmg/ddx246] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 06/20/2017] [Indexed: 12/12/2022] Open
Abstract
The timing of human puberty is highly variable, sexually dimorphic, and associated with adverse health outcomes. Over 20 genes carrying rare mutations have been identified in known pubertal disorders, many of which encode critical components of the hypothalamic-pituitary-gonadal (HPG) axis. Recent genome-wide association studies (GWAS) have identified more than 100 candidate genes at loci associated with age at menarche or voice breaking in males. We know little about the spatial, temporal or postnatal expression patterns of the majority of these puberty-associated genes. Using a high-throughput and sensitive microfluidic quantitative PCR strategy, we profiled the gene expression patterns of the mouse orthologs of 178 puberty-associated genes in male and female mouse HPG axis tissues, the pineal gland, and the liver at five postnatal ages spanning the pubertal transition. The most dynamic gene expression changes were observed prior to puberty in all tissues. We detected known and novel tissue-enhanced gene expression patterns, with the hypothalamus expressing the largest number of the puberty-associated genes. Notably, over 40 puberty-associated genes in the pituitary gland showed sex-biased gene expression, most of which occurred peri-puberty. These sex-biased genes included the orthologs of candidate genes at GWAS loci that show sex-discordant effects on pubertal timing. Our findings provide new insight into the expression of puberty-associated genes and support the possibility that the pituitary plays a role in determining sex differences in the timing of puberty.
Collapse
Affiliation(s)
- Huayun Hou
- Program in Genetics and Genome Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Liis Uusküla-Reimand
- Program in Genetics and Genome Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada.,Department of Gene Technology, Tallinn University of Technology, 12616 Tallinn, Estonia
| | - Maisam Makarem
- Program in Genetics and Genome Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Christina Corre
- Program in Genetics and Genome Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Shems Saleh
- Program in Genetics and Genome Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada.,Department of Computer Science, University of Toronto, Toronto, ON M5S 2E5, Canada
| | - Ariane Metcalf
- Program in Genetics and Genome Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Anna Goldenberg
- Program in Genetics and Genome Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada.,Department of Computer Science, University of Toronto, Toronto, ON M5S 2E5, Canada
| | - Mark R Palmert
- Program in Genetics and Genome Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada.,Division of Endocrinology, Hospital for Sick Children, Toronto, ON M5G 1X8, Canada.,Departments of Paediatrics and Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Michael D Wilson
- Program in Genetics and Genome Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
37
|
Sharma K, Singh J, Frost EE, Pillai PP. MeCP2 in central nervous system glial cells: current updates. Acta Neurobiol Exp (Wars) 2018. [DOI: 10.21307/ane-2018-007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
38
|
Deletion Involving the 7q31-32 Band at the CADPS2 Gene Locus in a Patient with Autism Spectrum Disorder and Recurrent Psychotic Syndrome Triggered by Stress. Case Rep Psychiatry 2017; 2017:4254152. [PMID: 29201482 PMCID: PMC5671701 DOI: 10.1155/2017/4254152] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 09/13/2017] [Indexed: 11/17/2022] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder marked by impairments in social functioning, language, communication, and behavior. Recent genome-wide association studies show some microdeletions on the 7q31-32 region, including the CADPS2 locus in autistic patients. This paper reports the case of a patient with ASD and recurrent psychotic syndrome, in which a deletion on the 7q31-32 band at the CADPS2 gene locus was evidenced, as well as a brief review of the literature on the CADPS2 gene and its association with ASD.
Collapse
|
39
|
CAPS2 deficiency affects environmental enrichment-induced adult neurogenesis and differentiation/survival of newborn neurons in the hippocampal dentate gyrus. Neurosci Lett 2017; 661:121-125. [PMID: 28963059 DOI: 10.1016/j.neulet.2017.09.047] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 09/05/2017] [Accepted: 09/25/2017] [Indexed: 11/23/2022]
Abstract
Hippocampal adult neurogenesis is observed in the subgranular zone of the dentate gyrus (DG), and is associated with hippocampal memory formation and several psychiatric disorders including autism spectrum disorder (ASD). Calcium-dependent activator protein for secretion 2 (CAPS2) is a candidate gene related to ASD, and is highly expressed in the hippocampal DG region, with Caps2 knockout (KO) mice exhibiting ASD-like behavior. Accordingly, CAPS2 is potentially associated with hippocampal adult neurogenesis, the relationship between CAPS2 and adult neurogenesis has not yet been investigated. Here, we determined whether deficit of the Caps2 gene affects hippocampal adult neurogenesis and maturation of newborn neurons. To induce adult neurogenesis, we used the environmental enrichment (EE) condition. Both wild-type (WT) and Caps2 KO mice were housed in control or EE conditions for 3 or 14days. Hippocampal levels of brain-derived neurotrophic factor (BDNF) can be used as a physiological EE conditioned marker, and were increased at 14days in the EE condition in both WT and KO mice. Newborn cells during control and EE conditions were labeled by BrdU, and the labeled cells co-immunostained with the immature and mature neuron markers, calretinin (CR) and NeuN. The ratio of CR/BrdU and NeuN/BrdU double positive cells to all of BrdU positive cells were significantly increased in WT mice housed in the EE condition for 14days compared with the control condition. Whereas KO mice in the EE condition showed no significant increase of newborn neurons. These findings suggest that CAPS2 deficiency strongly impairs hippocampal adult neurogenesis and maturation of newborn neurons.
Collapse
|
40
|
van Keimpema L, Kooistra R, Toonen RF, Verhage M. CAPS-1 requires its C2, PH, MHD1 and DCV domains for dense core vesicle exocytosis in mammalian CNS neurons. Sci Rep 2017; 7:10817. [PMID: 28883501 PMCID: PMC5589909 DOI: 10.1038/s41598-017-10936-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 08/16/2017] [Indexed: 01/11/2023] Open
Abstract
CAPS (calcium-dependent activator protein for secretion) are multi-domain proteins involved in regulated exocytosis of synaptic vesicles (SVs) and dense core vesicles (DCVs). Here, we assessed the contribution of different CAPS-1 domains to its subcellular localization and DCV exocytosis by expressing CAPS-1 mutations in four functional domains in CAPS-1/-2 null mutant (CAPS DKO) mouse hippocampal neurons, which are severely impaired in DCV exocytosis. CAPS DKO neurons showed normal development and no defects in DCV biogenesis and their subcellular distribution. Truncation of the CAPS-1 C-terminus (CAPS Δ654-1355) impaired CAPS-1 synaptic enrichment. Mutations in the C2 (K428E or G476E) or pleckstrin homology (PH; R558D/K560E/K561E) domain did not. However, all mutants rescued DCV exocytosis in CAPS DKO neurons to only 20% of wild type CAPS-1 exocytosis capacity. To assess the relative importance of CAPS for both secretory pathways, we compared effect sizes of CAPS-1/-2 deficiency on SV and DCV exocytosis. Using the same (intense) stimulation, DCV exocytosis was impaired relatively strong (96% inhibition) compared to SV exocytosis (39%). Together, these data show that the CAPS-1 C-terminus regulates synaptic enrichment of CAPS-1. All CAPS-1 functional domains are required, and the C2 and PH domain together are not sufficient, for DCV exocytosis in mammalian CNS neurons.
Collapse
Affiliation(s)
- Linda van Keimpema
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, VU University, 1081 HV, Amsterdam, The Netherlands
- Sylics (Synaptologics BV), PO box 71033, 1008 BA, Amsterdam, The Netherlands
| | - Robbelien Kooistra
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, VU University, 1081 HV, Amsterdam, The Netherlands
| | - Ruud F Toonen
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, VU University, 1081 HV, Amsterdam, The Netherlands.
| | - Matthijs Verhage
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, VU University, 1081 HV, Amsterdam, The Netherlands.
| |
Collapse
|
41
|
Xu B, Kumazawa A, Kobayashi S, Hisanaga SI, Inoue T, Ohshima T. Cdk5 activity is required for Purkinje cell dendritic growth in cell-autonomous and non-cell-autonomous manners. Dev Neurobiol 2017; 77:1175-1187. [PMID: 28589675 DOI: 10.1002/dneu.22507] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 03/27/2017] [Accepted: 06/02/2017] [Indexed: 12/25/2022]
Abstract
Cyclin-dependent kinase 5 (Cdk5) is recognized as a unique member among other Cdks due to its versatile roles in many biochemical processes in the nervous system. The proper development of neuronal dendrites is required for the formation of complex neural networks providing the physiological basis of various neuronal functions. We previously reported that sparse dendrites were observed on cultured Cdk5-null Purkinje cells and Purkinje cells in Wnt1cre -mediated Cdk5 conditional knockout (KO) mice. In the present study, we generated L7cre -mediated p35; p39 double KO (L7cre -p35f/f ; p39-/- ) mice whose Cdk5 activity was eliminated specifically in Purkinje cells of the developing cerebellum. Consequently, these mice exhibited defective Purkinje cell migration, motor coordination deficiency and a Purkinje dendritic abnormality similar to what we have observed before, suggesting that dendritic growth of Purkinje cells was cell-autonomous in vivo. We found that mixed and overlay cultures of WT cerebellar cells rescued the dendritic deficits in Cdk5-null Purkinje cells, however, indicating that Purkinje cell dendritic development was also supported by non-cell-autonomous factors. We then again rescued these abnormalities in vitro by applying exogenous brain-derived neurotrophic factor (BDNF). Based on the results from culture experiments, we attempted to rescue the developmental defects of Purkinje cells in L7cre -p35f/f ; p39-/- mice by using a TrkB agonist. We observed partial rescue of morphological defects of dendritic structures of Purkinje cells. These results suggest that Cdk5 activity is required for Purkinje cell dendritic growth in cell-autonomous and non-cell-autonomous manners. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 77: 1175-1187, 2017.
Collapse
Affiliation(s)
- Bozong Xu
- Department of Life Science and Medical Bioscience, Laboratory for Molecular Brain Science, Waseda University, Tokyo, 162-8480, Japan
| | - Ayumi Kumazawa
- Department of Life Science and Medical Bioscience, Laboratory for Molecular Brain Science, Waseda University, Tokyo, 162-8480, Japan.,Department of Biological Science, Tokyo Metropolitan University, Hachioji, Tokyo, 192-0397, Japan
| | - Shunsuke Kobayashi
- Department of Life Science and Medical Bioscience, Laboratory for Molecular Brain Science, Waseda University, Tokyo, 162-8480, Japan
| | - Shin-Ichi Hisanaga
- Department of Biological Science, Tokyo Metropolitan University, Hachioji, Tokyo, 192-0397, Japan
| | - Takafumi Inoue
- Department of Life Science and Medical Bioscience, Laboratory for Neurophysiology, Waseda University, Tokyo, 162-8480, Japan
| | - Toshio Ohshima
- Department of Life Science and Medical Bioscience, Laboratory for Molecular Brain Science, Waseda University, Tokyo, 162-8480, Japan
| |
Collapse
|
42
|
Dezsi G, Sieg F, Thomas M, O’Brien TJ, van der Hart M, Jones NC. Disease-Modifying Effects of Neural Regeneration Peptide 2945 in the GAERS Model of Absence Epilepsy. Neurochem Res 2017; 42:2055-2064. [DOI: 10.1007/s11064-017-2305-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 05/11/2017] [Accepted: 05/12/2017] [Indexed: 01/23/2023]
|
43
|
Analysis of gene expression in Ca2+-dependent activator protein for secretion 2 (Cadps2) knockout cerebellum using GeneChip and KEGG pathways. Neurosci Lett 2017; 639:88-93. [DOI: 10.1016/j.neulet.2016.12.068] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 12/09/2016] [Accepted: 12/28/2016] [Indexed: 11/20/2022]
|
44
|
Shinoda Y, Ishii C, Fukazawa Y, Sadakata T, Ishii Y, Sano Y, Iwasato T, Itohara S, Furuichi T. CAPS1 stabilizes the state of readily releasable synaptic vesicles to fusion competence at CA3-CA1 synapses in adult hippocampus. Sci Rep 2016; 6:31540. [PMID: 27545744 PMCID: PMC4992871 DOI: 10.1038/srep31540] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 07/21/2016] [Indexed: 01/06/2023] Open
Abstract
Calcium-dependent activator protein for secretion 1 (CAPS1) regulates exocytosis of dense-core vesicles in neuroendocrine cells and of synaptic vesicles in neurons. However, the synaptic function of CAPS1 in the mature brain is unclear because Caps1 knockout (KO) results in neonatal death. Here, using forebrain-specific Caps1 conditional KO (cKO) mice, we demonstrate, for the first time, a critical role of CAPS1 in adult synapses. The amplitude of synaptic transmission at CA3–CA1 synapses was strongly reduced, and paired-pulse facilitation was significantly increased, in acute hippocampal slices from cKO mice compared with control mice, suggesting a perturbation in presynaptic function. Morphological analysis revealed an accumulation of synaptic vesicles in the presynapse without any overall morphological change. Interestingly, however, the percentage of docked vesicles was markedly decreased in the Caps1 cKO. Taken together, our findings suggest that CAPS1 stabilizes the state of readily releasable synaptic vesicles, thereby enhancing neurotransmitter release at hippocampal synapses.
Collapse
Affiliation(s)
- Yo Shinoda
- Department of Applied Biological Science, Tokyo University of Science, Noda, Chiba 278-8510, Japan.,School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Chiaki Ishii
- Department of Applied Biological Science, Tokyo University of Science, Noda, Chiba 278-8510, Japan
| | - Yugo Fukazawa
- Department of Brain Structure and Function, Faculty of Medical Sciences, University of Fukui, Yoshida-gun, Fukui 910-1193, Japan
| | - Tetsushi Sadakata
- Advanced Scientific Research Leaders Development Unit, Gunma University, Maebashi, Gunma 371-8511, Japan
| | - Yuki Ishii
- Department of Applied Biological Science, Tokyo University of Science, Noda, Chiba 278-8510, Japan
| | - Yoshitake Sano
- Department of Applied Biological Science, Tokyo University of Science, Noda, Chiba 278-8510, Japan
| | - Takuji Iwasato
- Division of Neurogenetics, National Institute of Genetics, Mishima, Shizuoka 411-8540, Japan.,Department of Genetics, SOKENDAI, Mishima, Shizuoka 411-8540, Japan
| | - Shigeyoshi Itohara
- Laboratory for Behavioral Genetics, RIKEN Brain Science Institute, Wako, Saitama 351-0198, Japan
| | - Teiichi Furuichi
- Department of Applied Biological Science, Tokyo University of Science, Noda, Chiba 278-8510, Japan
| |
Collapse
|
45
|
Interaction of Ca(2+)-dependent activator protein for secretion 1 (CAPS1) with septin family proteins in mouse brain. Neurosci Lett 2016; 617:232-5. [PMID: 26917099 DOI: 10.1016/j.neulet.2016.02.035] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 02/18/2016] [Accepted: 02/19/2016] [Indexed: 01/22/2023]
Abstract
The Ca(2+)-dependent activator protein for secretion 1 (CAPS1) protein plays a regulatory role in the dense-core vesicle exocytosis pathway. To clarify the functions of this protein in the brain, we searched for novel interaction partners of CAPS1 by mass spectrometry. We identified a specific interaction of CAPS1 with septin family proteins. We also demonstrated that the C-terminal region of the CAPS1 protein binds to part of the deduced GTP-binding domain of septin proteins. It is possible that a tertiary complex of septin, CAPS, and syntaxin contributes to dense-core vesicle trafficking and exocytosis in neurons.
Collapse
|
46
|
Delépine C, Nectoux J, Letourneur F, Baud V, Chelly J, Billuart P, Bienvenu T. Astrocyte Transcriptome from the Mecp2(308)-Truncated Mouse Model of Rett Syndrome. Neuromolecular Med 2015. [PMID: 26208914 DOI: 10.1007/s12017-015-8363-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Mutations in the gene encoding the transcriptional modulator methyl-CpG binding protein 2 (MeCP2) are responsible for the neurodevelopmental disorder Rett syndrome which is one of the most frequent sources of intellectual disability in women. Recent studies showed that loss of Mecp2 in astrocytes contributes to Rett-like symptoms and restoration of Mecp2 can rescue some of these defects. The goal of this work is to compare gene expression profiles of wild-type and mutant astrocytes from Mecp2(308/y) mice (B6.129S-MeCP2<tm1Heto>/J) by using Affymetrix mouse 2.0 microarrays. Results were confirmed by quantitative real-time RT-PCR and by Western blot analysis. Gene set enrichment analysis utilizing Ingenuity Pathways was employed to identify pathways disrupted by Mecp2 deficiency. A total of 2152 genes were statistically differentially expressed between wild-type and mutated samples, including 1784 coding transcripts. However, only 257 showed fold changes >1.2. We confirmed our data by replicative studies in independent primary cultures of cortical astrocytes from Mecp2-deficient mice. Interestingly, two genes known to encode secreted proteins, chromogranin B and lipocalin-2, showed significant dysregulation. These proteins secreted from Mecp2-deficient glia may exert negative non-cell autonomous effects on neuronal properties, including dendritic morphology. Moreover, transcriptional profiling revealed altered Nr2f2 expression which may explain down- and upregulation of several target genes in astrocytes such as Ccl2, Lcn2 and Chgb. Unraveling Nr2f2 involvement in Mecp2-deficient astrocytes could pave the way for a better understanding of Rett syndrome pathophysiology and offers new therapeutic perspectives.
Collapse
Affiliation(s)
- Chloé Delépine
- Inserm, U1016, Faculté de Médecine, Laboratoire de Génétique et de Physiopathologie des Maladies Mentales, Institut Cochin, 24 Rue du Faubourg Saint Jacques, 75014, Paris, France.,Cnrs, UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Juliette Nectoux
- Inserm, U1016, Faculté de Médecine, Laboratoire de Génétique et de Physiopathologie des Maladies Mentales, Institut Cochin, 24 Rue du Faubourg Saint Jacques, 75014, Paris, France.,Cnrs, UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Groupe Universitaire Paris Centre, Site Cochin, Laboratoire de Biochimie et Génétique Moléculaire, Assistance Publique - Hôpitaux de Paris, 27 rue du Faubourg Saint Jacques, 75014, Paris, France
| | - Franck Letourneur
- Inserm, U1016, Faculté de Médecine, Laboratoire de Génétique et de Physiopathologie des Maladies Mentales, Institut Cochin, 24 Rue du Faubourg Saint Jacques, 75014, Paris, France.,Cnrs, UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Véronique Baud
- Inserm, U1016, Faculté de Médecine, Laboratoire de Génétique et de Physiopathologie des Maladies Mentales, Institut Cochin, 24 Rue du Faubourg Saint Jacques, 75014, Paris, France.,Cnrs, UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Jamel Chelly
- Inserm, U1016, Faculté de Médecine, Laboratoire de Génétique et de Physiopathologie des Maladies Mentales, Institut Cochin, 24 Rue du Faubourg Saint Jacques, 75014, Paris, France.,Cnrs, UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Pierre Billuart
- Inserm, U1016, Faculté de Médecine, Laboratoire de Génétique et de Physiopathologie des Maladies Mentales, Institut Cochin, 24 Rue du Faubourg Saint Jacques, 75014, Paris, France.,Cnrs, UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Thierry Bienvenu
- Inserm, U1016, Faculté de Médecine, Laboratoire de Génétique et de Physiopathologie des Maladies Mentales, Institut Cochin, 24 Rue du Faubourg Saint Jacques, 75014, Paris, France. .,Cnrs, UMR8104, Paris, France. .,Université Paris Descartes, Sorbonne Paris Cité, Paris, France. .,Groupe Universitaire Paris Centre, Site Cochin, Laboratoire de Biochimie et Génétique Moléculaire, Assistance Publique - Hôpitaux de Paris, 27 rue du Faubourg Saint Jacques, 75014, Paris, France.
| |
Collapse
|
47
|
Lack of stress responses to long-term effects of corticosterone in Caps2 knockout mice. Sci Rep 2015; 5:8932. [PMID: 25754523 PMCID: PMC4354153 DOI: 10.1038/srep08932] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 02/11/2015] [Indexed: 11/08/2022] Open
Abstract
Chronic stress is associated with anxiety and depressive disorders, and can cause weight gain. Ca(2+)-dependent activator protein for secretion 2 (CAPS2) is involved in insulin release. Caps2 knockout (KO) mice exhibit decreased body weight, reduced glucose-induced insulin release, and abnormal psychiatric behaviors. We chronically administered the stress hormone corticosterone (CORT), which induces anxiety/depressive-like behavior and normally increases plasma insulin levels, via the drinking water for 10 weeks, and we examined the stress response in KO mice. Chronic CORT exposure inhibited stress-induced serum CORT elevation in wild-type (WT) mice, but not in KO mice. Poor weight gain in CORT-treated animals was observed until week 6 in WT mice, but persisted for the entire duration of the experiment in KO mice, although there is no difference in drug*genotype interaction. Among KO mice, food consumption was unchanged, while water consumption was higher, over the duration of the experiment in CORT-treated animals, compared with untreated animals. Moreover, serum insulin and leptin levels were increased in CORT-treated WT mice, but not in KO mice. Lastly, both WT and KO mice displayed anxiety/depressive-like behavior after CORT administration. These results suggest that Caps2 KO mice have altered endocrine responses to CORT administration, while maintaining CORT-induced anxiety/depressive-like behavior.
Collapse
|
48
|
Farina M, van de Bospoort R, He E, Persoon CM, van Weering JRT, Broeke JH, Verhage M, Toonen RF. CAPS-1 promotes fusion competence of stationary dense-core vesicles in presynaptic terminals of mammalian neurons. eLife 2015; 4. [PMID: 25719439 PMCID: PMC4341531 DOI: 10.7554/elife.05438] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 02/09/2015] [Indexed: 01/03/2023] Open
Abstract
Neuropeptides released from dense-core vesicles (DCVs) modulate neuronal activity, but the molecules driving DCV secretion in mammalian neurons are largely unknown. We studied the role of calcium-activator protein for secretion (CAPS) proteins in neuronal DCV secretion at single vesicle resolution. Endogenous CAPS-1 co-localized with synaptic markers but was not enriched at every synapse. Deletion of CAPS-1 and CAPS-2 did not affect DCV biogenesis, loading, transport or docking, but DCV secretion was reduced by 70% in CAPS-1/CAPS-2 double null mutant (DKO) neurons and remaining fusion events required prolonged stimulation. CAPS deletion specifically reduced secretion of stationary DCVs. CAPS-1-EYFP expression in DKO neurons restored DCV secretion, but CAPS-1-EYFP and DCVs rarely traveled together. Synaptic localization of CAPS-1-EYFP in DKO neurons was calcium dependent and DCV fusion probability correlated with synaptic CAPS-1-EYFP expression. These data indicate that CAPS-1 promotes fusion competence of immobile (tethered) DCVs in presynaptic terminals and that CAPS-1 localization to DCVs is probably not essential for this role. DOI:http://dx.doi.org/10.7554/eLife.05438.001 Our ability to think and act is due to the remarkable capacity of the brain to process complex information. This involves nerve cells (or neurons) communicating with each other in a rapid and precise manner by releasing synaptic vesicles containing neurotransmitters across the gaps—called synapses—between neurons. In addition to this fast neurotransmitter signalling, neurons can transmit signals by releasing chemical signals called neuropeptides. Neuropeptides are major regulators of human brain function, including mood, anxiety, and social interactions. Neuropeptides and other neuromodulators such as serotonin and dopamine are normally packaged into bubble-like compartments called dense-core vesicles. Compared to synaptic vesicles we know much less about how dense-core vesicles are trafficked and released. Dense-core vesicles are generally mobile and move around the inside of cells to release neuropeptides where and when they are needed. However, some vesicles are stationary and may even be loosely tethered to the cell membrane. Most of the sites where dense-core vesicles can fuse with the cell membrane are at synapses. Previous work has suggested that the protein CAPS-1 is important for moving dense-core vesicles to the correct sites on the cell membrane, and for releasing neuropeptides across the synapses of worms and flies. However, detailed insights into this process in mammalian neurons are lacking. By examining neurons from both normal mice and mice lacking the CAPS-1 protein, Farina et al. have now analyzed the role CAPS-1 plays in releasing neuropeptides. In cells lacking CAPS-1 fewer dense-core vesicles merged with the cell membrane than in cells containing the protein. However, a new technique that tracks the movement of individual vesicles revealed that only stationary dense-core vesicles had difficulties fusing; mobile vesicles continued to fuse with the cell membrane in the normal manner. Introducing CAPS-1 into cells lacking this protein corrected the fusion defect experienced by the stationary vesicles. Farina et al. also showed that CAPS-1 was present at most—but not all—synapses, and synapses that had more CAPS-1 released more neuropeptides. This work shows that CAPS proteins strongly influence the probability of dense-core vesicle release and that neurons can tune this probability at individual synapses by controlling the expression of CAPS. Future work will be aimed at understanding how neurons can achieve this and which protein domains in CAPS are required. DOI:http://dx.doi.org/10.7554/eLife.05438.002
Collapse
Affiliation(s)
- Margherita Farina
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, VU University Amsterdam, Amsterdam, Netherlands
| | - Rhea van de Bospoort
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, VU University Amsterdam, Amsterdam, Netherlands
| | - Enqi He
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, VU University Amsterdam, Amsterdam, Netherlands
| | - Claudia M Persoon
- Department of Clinical Genetics, VU University Medical Center, Amsterdam, Netherlands
| | - Jan R T van Weering
- Department of Clinical Genetics, VU University Medical Center, Amsterdam, Netherlands
| | - Jurjen H Broeke
- Department of Clinical Genetics, VU University Medical Center, Amsterdam, Netherlands
| | - Matthijs Verhage
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, VU University Amsterdam, Amsterdam, Netherlands
| | - Ruud F Toonen
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, VU University Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
49
|
Sajadian A, Esteghamat S, Karimzadeh F, Eshaghabadi A, Sieg F, Speckmann EJ, Meuth S, Seidenbecher T, Budde T, Gorji A. Anticonvulsant effect of neural regeneration peptide 2945 on pentylenetetrazol-induced seizures in rats. Neuropeptides 2015; 49:15-23. [PMID: 25481799 DOI: 10.1016/j.npep.2014.11.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 11/05/2014] [Accepted: 11/19/2014] [Indexed: 11/30/2022]
Abstract
Neuron regeneration peptides (NRPs) are small synthetic peptides that stimulate neural proliferation, migration, and differentiation with no apparent toxicity and high target specificity in CNS. The aim of this study was to investigate the effect of NRP2945 on seizure activity induced by pentylenetetrazol (PTZ) in rats. Using behavioural assessment and electrocorticographical recordings, the effects of different doses of NRP2945 (5-20 µg/kg) were tested on seizure attacks induced by PTZ injection. In addition, the effect of NRP2945 was evaluated on the production of dark neurons and expression of GABAA receptor α and β subunits and GAD-65 in the hippocampus and somatosensory cortex of the rat brain. Intraperitoneal injection of NRP2945 at 20 µg/kg prevented seizure attacks after PTZ injection. NRP2945 at doses of 5 and 10 µg/kg significantly decreased the total duration of seizure attacks and reduced the amplitude, duration and latency of epileptiform burst discharges induced by PTZ. In addition, the peptide significantly inhibited the production of dark neurons in the hippocampus and somatosensory cortex of epileptic rats. NRP2945 also significantly increased the expression of GABAA receptor α and β subunits and GAD-65 in the hippocampus and somatosensory cortex compared with PTZ treated rats. This study indicates that NRP2945 is able to prevent the seizure attacks and neuronal injuries induced by PTZ, likely by stimulating GABAA and GAD-65 protein expression and/or protecting these components of GABAergic signalling from PTZ-induced alteration. Further studies are needed to elucidate the potential role of NRP2945 as an antiepileptic drug.
Collapse
Affiliation(s)
| | | | - Fariba Karimzadeh
- Departments of Neuroscience, School of Advanced Technology of Medical, Tehran Medical University, Tehran, Iran
| | | | - Frank Sieg
- CuroNZ Ltd, 29 Nugent Street, Grafton, Auckland, New Zealand
| | - Erwin-Josef Speckmann
- Institute of Physiology I, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - Sven Meuth
- Department of Neurology, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - Thomas Seidenbecher
- Institute of Physiology I, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - Thomas Budde
- Institute of Physiology I, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - Ali Gorji
- Shefa Neuroscience Research Center, Tehran, Iran; Department of Neurology, Westfälische Wilhelms-Universität Münster, Münster, Germany; Institute of Physiology I, Westfälische Wilhelms-Universität Münster, Münster, Germany; Epilepsy Research Center, Westfälische Wilhelms-Universität Münster, Münster, Germany; Klinik und Poliklinik für Neurochirurgie, Westfälische Wilhelms-Universität Münster, Münster, Germany.
| |
Collapse
|
50
|
Dittrich F, Ramenda C, Grillitsch D, Frankl-Vilches C, Ko MC, Hertel M, Goymann W, ter Maat A, Gahr M. Regulatory mechanisms of testosterone-stimulated song in the sensorimotor nucleus HVC of female songbirds. BMC Neurosci 2014; 15:128. [PMID: 25442096 PMCID: PMC4261767 DOI: 10.1186/s12868-014-0128-0] [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: 07/15/2014] [Accepted: 11/13/2014] [Indexed: 01/04/2023] Open
Abstract
Background In male birds, influence of the sex steroid hormone testosterone and its estrogenic metabolites on seasonal song behavior has been demonstrated for many species. In contrast, female song was only recently recognized to be widespread among songbird species, and to date, sex hormone effects on singing and brain regions controlling song development and production (song control nuclei) have been studied in females almost exclusively using domesticated canaries (Serinus canaria). However, domesticated female canaries hardly sing at all in normal circumstances and exhibit only very weak, if any, song seasonally under the natural photoperiod. By contrast, adult female European robins (Erithacus rubecula) routinely sing during the winter season, a time when they defend feeding territories and show elevated circulating testosterone levels. We therefore used wild female European robins captured in the fall to examine the effects of testosterone administration on song as well as on the anatomy and the transcriptome of the song control nucleus HVC (sic). The results obtained from female robins were compared to outcomes of a similar experiment done in female domesticated canaries. Results Testosterone treatment induced abundant song in female robins. Examination of HVC transcriptomes and histological analyses of song control nuclei showed testosterone-induced differentiation processes related to neuron growth and spacing, angiogenesis and neuron projection morphogenesis. Similar effects were found in female canaries treated with testosterone. In contrast, the expression of genes related to synaptic transmission was not enhanced in the HVC of testosterone treated female robins but was strongly up-regulated in female canaries. A comparison of the testosterone-stimulated transcriptomes indicated that brain-derived neurotrophic factor (BDNF) likely functions as a common mediator of the testosterone effects in HVC. Conclusions Testosterone-induced singing of female robins correlated with cellular differentiation processes in the HVC that were partially similar to those seen in the HVC of testosterone-treated female canaries. Other modes of testosterone action, notably related to synaptic transmission, appeared to be regulated in a more species-specific manner in the female HVC. Divergent effects of testosterone on the HVC of different species might be related to differences between species in regulatory mechanisms of the singing behavior. Electronic supplementary material The online version of this article (doi:10.1186/s12868-014-0128-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Falk Dittrich
- Max Planck Institute for Ornithology, Department of Behavioural Neurobiology, Eberhard-Gwinner Strasse, Haus 6a, Seewiesen, 82319, Germany.
| | - Claudia Ramenda
- Max Planck Institute for Ornithology, Department of Behavioural Neurobiology, Eberhard-Gwinner Strasse, Haus 6a, Seewiesen, 82319, Germany.
| | - Doris Grillitsch
- Max Planck Institute for Ornithology, Department of Behavioural Neurobiology, Eberhard-Gwinner Strasse, Haus 6a, Seewiesen, 82319, Germany.
| | - Carolina Frankl-Vilches
- Max Planck Institute for Ornithology, Department of Behavioural Neurobiology, Eberhard-Gwinner Strasse, Haus 6a, Seewiesen, 82319, Germany.
| | - Meng-Ching Ko
- Max Planck Institute for Ornithology, Department of Behavioural Neurobiology, Eberhard-Gwinner Strasse, Haus 6a, Seewiesen, 82319, Germany.
| | - Moritz Hertel
- Max Planck Institute for Ornithology, Department of Behavioural Neurobiology, Eberhard-Gwinner Strasse, Haus 6a, Seewiesen, 82319, Germany.
| | - Wolfgang Goymann
- Max Planck Institute for Ornithology, Department of Behavioural Neurobiology, Eberhard-Gwinner Strasse, Haus 6a, Seewiesen, 82319, Germany.
| | - Andries ter Maat
- Max Planck Institute for Ornithology, Department of Behavioural Neurobiology, Eberhard-Gwinner Strasse, Haus 6a, Seewiesen, 82319, Germany.
| | - Manfred Gahr
- Max Planck Institute for Ornithology, Department of Behavioural Neurobiology, Eberhard-Gwinner Strasse, Haus 6a, Seewiesen, 82319, Germany.
| |
Collapse
|