1
|
Paget-Blanc V, Pfeffer ME, Pronot M, Lapios P, Angelo MF, Walle R, Cordelières FP, Levet F, Claverol S, Lacomme S, Petrel M, Martin C, Pitard V, De Smedt Peyrusse V, Biederer T, Perrais D, Trifilieff P, Herzog E. A synaptomic analysis reveals dopamine hub synapses in the mouse striatum. Nat Commun 2022; 13:3102. [PMID: 35660742 PMCID: PMC9166739 DOI: 10.1038/s41467-022-30776-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 05/18/2022] [Indexed: 11/13/2022] Open
Abstract
Dopamine transmission is involved in reward processing and motor control, and its impairment plays a central role in numerous neurological disorders. Despite its strong pathophysiological relevance, the molecular and structural organization of the dopaminergic synapse remains to be established. Here, we used targeted labelling and fluorescence activated sorting to purify striatal dopaminergic synaptosomes. We provide the proteome of dopaminergic synapses with 57 proteins specifically enriched. Beyond canonical markers of dopamine neurotransmission such as dopamine biosynthetic enzymes and cognate receptors, we validated 6 proteins not previously described as enriched. Moreover, our data reveal the adhesion of dopaminergic synapses to glutamatergic, GABAergic or cholinergic synapses in structures we named “dopamine hub synapses”. At glutamatergic synapses, pre- and postsynaptic markers are significantly increased upon association with dopamine synapses. Dopamine hub synapses may thus support local dopaminergic signalling, complementing volume transmission thought to be the major mechanism by which monoamines modulate network activity. The neurotransmitter dopamine is an important regulator of brain function. Here the authors describe “dopamine hub synapses”, where dopamine transmission may act in synergy with other neurotransmitters.
Collapse
Affiliation(s)
- Vincent Paget-Blanc
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000, Bordeaux, France
| | - Marlene E Pfeffer
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000, Bordeaux, France
| | - Marie Pronot
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000, Bordeaux, France
| | - Paul Lapios
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000, Bordeaux, France
| | - Maria-Florencia Angelo
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000, Bordeaux, France
| | - Roman Walle
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France
| | - Fabrice P Cordelières
- Univ. Bordeaux, CNRS, INSERM, Bordeaux Imaging Center, BIC, UAR 3420, US 4, F-33000, Bordeaux, France
| | - Florian Levet
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000, Bordeaux, France.,Univ. Bordeaux, CNRS, INSERM, Bordeaux Imaging Center, BIC, UAR 3420, US 4, F-33000, Bordeaux, France
| | | | - Sabrina Lacomme
- Univ. Bordeaux, CNRS, INSERM, Bordeaux Imaging Center, BIC, UAR 3420, US 4, F-33000, Bordeaux, France
| | - Mélina Petrel
- Univ. Bordeaux, CNRS, INSERM, Bordeaux Imaging Center, BIC, UAR 3420, US 4, F-33000, Bordeaux, France
| | - Christelle Martin
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000, Bordeaux, France
| | - Vincent Pitard
- UB'FACSility CNRS UMS 3427, INSERM US 005, Univ. Bordeaux, F-33000, Bordeaux, France
| | | | - Thomas Biederer
- Department of Neurology, Yale School of Medicine, New Haven, CT, 06511, USA
| | - David Perrais
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000, Bordeaux, France
| | - Pierre Trifilieff
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France
| | - Etienne Herzog
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000, Bordeaux, France.
| |
Collapse
|
2
|
Mint3 is dispensable for pancreatic and kidney functions in mice. Biochem Biophys Rep 2020; 24:100872. [PMID: 33319072 PMCID: PMC7725678 DOI: 10.1016/j.bbrep.2020.100872] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 11/06/2020] [Accepted: 11/24/2020] [Indexed: 02/07/2023] Open
Abstract
Munc-18 interacting protein 3 (Mint3) is an activator of hypoxia-inducible factor-1 in cancer cells, macrophages, and cancer-associated fibroblasts under pathological conditions. However, exactly which cells highly express Mint3 in vivo and whether Mint3 depletion affects their physiological functions remain unclear. Here, we surveyed mouse tissues for specific expression of Mint3 by comparing Mint3 expression in wild-type and Mint3-knockout mice. Interestingly, immunohistochemical analyses revealed that Mint3 was highly expressed in islet cells of the pancreas, distal tubular epithelia of the kidney, choroid plexus ependymal cells of the cerebrum, medullary cells of the adrenal gland, and epithelial cells of the seminal gland. We also studied whether Mint3 depletion affects the physiological functions of the islets and kidneys. Mint3-knockout mice did not show any abnormalities in glucose-tolerance and urine-biochemical tests, indicating that Mint3 depletion was compensated for in these organs. Thus, loss of Mint3 might be compensated in the islets and kidneys under physiological conditions in mice. Specific expression of Mint3 in mouse tissues is surveyed. Mint3 is highly expressed in islet cells of the pancreas. Mint3 is highly expressed in distal tubular epithelia of the kidney. Mint3 KO mice do not show any abnormalities in glucose-tolerance tests. Mint3 KO mice do not show any abnormalities in urine-biochemical tests.
Collapse
|
3
|
Mossa A, Manzini MC. Molecular causes of sex-specific deficits in rodent models of neurodevelopmental disorders. J Neurosci Res 2019; 99:37-56. [PMID: 31872500 PMCID: PMC7754327 DOI: 10.1002/jnr.24577] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 11/02/2019] [Accepted: 12/09/2019] [Indexed: 12/12/2022]
Abstract
Neurodevelopmental disorders (NDDs) such as intellectual disability and autism spectrum disorder consistently show a male bias in prevalence, but it remains unclear why males and females are affected with different frequency. While many behavioral studies of transgenic NDD models have focused only on males, the requirement by the National Institutes of Health to consider sex as a biological variable has promoted the comparison of male and female performance in wild-type and mutant animals. Here, we review examples of rodent models of NDDs in which sex-specific deficits were identified in molecular, physiological, and/or behavioral responses, showing sex differences in susceptibility to disruption of genes mutated in NDDs. Haploinsufficiency in genes involved in mechanisms such as synaptic function (GABRB3 and NRXN1), chromatin remodeling (CHD8, EMHT1, and ADNP), and intracellular signaling (CC2D1A and ERK1) lead to more severe behavioral outcomes in males. However, in the absence of behavioral deficits, females can still present with cellular and electrophysiological changes that could be due to compensatory mechanisms or differential allocation of molecular and cellular functions in the two sexes. By contrasting these findings with mouse models where females are more severely affected (MTHFR and AMBRA1), we propose a framework to approach the study of sex-specific deficits possibly leading to sex bias in NDDs.
Collapse
Affiliation(s)
- Adele Mossa
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - M Chiara Manzini
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA.,Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| |
Collapse
|
4
|
MITSUMOTO Y, SATO R, TAGAWA N, KATO I. Rubiscolin-6, a δ-Opioid Peptide from Spinach RuBisCO, Exerts Antidepressant-Like Effect in Restraint-Stressed Mice. J Nutr Sci Vitaminol (Tokyo) 2019; 65:202-204. [DOI: 10.3177/jnsv.65.202] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Yasuhide MITSUMOTO
- Laboratory of Alternative Medicine and Experimental Therapeutics, Department of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Hokuriku University
| | - Reina SATO
- Laboratory of Alternative Medicine and Experimental Therapeutics, Department of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Hokuriku University
| | - Noriko TAGAWA
- Department of Medical Biochemistry, Kobe Pharmaceutical University
| | - Ikuo KATO
- Department of Medical Biochemistry, Kobe Pharmaceutical University
| |
Collapse
|
5
|
Rezazadeh A, Uddin M, Snead OC, Lira V, Silberberg A, Weiss S, Donner EJ, Zak M, Bradbury L, Scherer SW, Fasano A, Andrade DM. STXBP1 encephalopathy is associated with awake bruxism. Epilepsy Behav 2019; 92:121-124. [PMID: 30654231 DOI: 10.1016/j.yebeh.2018.12.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 12/15/2018] [Accepted: 12/21/2018] [Indexed: 12/20/2022]
Abstract
Heterozygous mutations in syntaxin-binding protein 1 (STXBP1) gene are associated with early infantile epileptic encephalopathy 4 (EIEE4). This condition is characterized by epilepsy, developmental delay (DD), and various movement disorders. Herein, we will report 5 unrelated patients with different de novo mutations in STXBP1. In addition, we conducted an online survey through Facebook to identify the incidence of bruxism (BRX) in these patients. Four out of 5 patients (80%) presented with awake BRX (A-BRX). Bruxism was also reported in 81.4% (57/70) of the patients with STXBP1 encephalopathy through the online questionnaire. No consistent correlation was identified between the type of mutation and development of movement disorders or BRX. This is the first study to demonstrate A-BRX in patients with STXBP1 mutation. Given the role of STXBP1 in exocytosis of neurotransmitters and other manifestations of dopamine dysregulation in patients with STXBP1-EIEE4, we suggest that in patients with STXBP1 encephalopathy, A-BRX might be the result of the involvement of dopaminergic circuits.
Collapse
Affiliation(s)
- Arezoo Rezazadeh
- Krembil Neurosciences Epilepsy Genetics Program, Toronto Western Hospital, University of Toronto, Toronto, Ontario, Canada.
| | - Mohammed Uddin
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada.
| | - O Carter Snead
- Division of Neurology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada.
| | - Victor Lira
- Krembil Neurosciences Epilepsy Genetics Program, Toronto Western Hospital, University of Toronto, Toronto, Ontario, Canada
| | | | - Shelly Weiss
- Division of Neurology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada.
| | - Elizabeth J Donner
- Division of Neurology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada.
| | - Maria Zak
- Division of Neurology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada.
| | - Laura Bradbury
- Division of Neurology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada.
| | - Stephen W Scherer
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Molecular Genetics and McLaughlin Centre, University of Toronto, Toronto, Ontario, Canada.
| | - Alfonso Fasano
- Edmond J. Safra Program in Parkinson's Disease and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, Toronto, Ontario, Canada; Division of Neurology, Department of Medicine, Toronto Western Hospital, Krembil Neuroscience Centre, University of Toronto, Toronto, Ontario, Canada.
| | - Danielle M Andrade
- Krembil Neurosciences Epilepsy Genetics Program, Toronto Western Hospital, University of Toronto, Toronto, Ontario, Canada; Division of Neurology, Department of Medicine, Toronto Western Hospital, Krembil Neuroscience Centre, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
6
|
Motodate R, Saito H, Sobu Y, Hata S, Saito Y, Nakaya T, Suzuki T. X11 and X11-like proteins regulate the level of extrasynaptic glutamate receptors. J Neurochem 2018; 148:480-498. [PMID: 30411795 DOI: 10.1111/jnc.14623] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 10/30/2018] [Accepted: 11/01/2018] [Indexed: 12/11/2022]
Abstract
X11/Mint 1 and X11-like (X11L)/Mint 2 are neuronal adaptor protein to regulate trafficking and/or localization of various membrane proteins. By analyzing the localization of neuronal membrane proteins in X11-, X11L-, and X11/X11L doubly deficient mice with membrane fractionation procedures, we found that deficient of X11 and X11L decreased the level of glutamate receptors in non-PSD fraction. This finding suggests that X11 and X11L regulate the glutamate receptor micro-localization to the extrasynaptic region. In vitro coimmunoprecipitation studies of NMDA receptors lacking various cytoplasmic regions with X11 and X11L proteins harboring domain deletion suggest that extrasynaptic localization of NMDA receptor may be as a result of the multiple interactions of the receptor subunits with X11 and X11L regulated by protein phosphorylation, while that of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor subunits is not dependent on the binding with X11 and X11L proteins. Because the loss of X11 and X11L tends to impair the exocytosis, but not endocytosis, of glutamate receptors, NMDA receptors are likely to be supplied to the extrasynaptic plasma membrane with a way distinct from the mechanism regulating the localization of NMDA receptors into synaptic membrane region. Reduced localization of NMDA receptor into the extrasynaptic region increased slightly the phosphorylation level of cAMP responsible element binding protein in brain of X11/X11L doubly deficient mice compare to wild-type mice, suggesting a possible role of X11 and X11L in the regulation of signal transduction pathway through extrasynaptic glutamate receptors. OPEN SCIENCE BADGES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. The complete Open Science Disclosure form for this article can be found at the end of the article. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/.
Collapse
Affiliation(s)
- Rika Motodate
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita-ku, Sapporo, Japan
| | - Haruka Saito
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita-ku, Sapporo, Japan
| | - Yuriko Sobu
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita-ku, Sapporo, Japan
| | - Saori Hata
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita-ku, Sapporo, Japan
| | - Yuhki Saito
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita-ku, Sapporo, Japan
| | - Tadashi Nakaya
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita-ku, Sapporo, Japan
| | - Toshiharu Suzuki
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita-ku, Sapporo, Japan
| |
Collapse
|
7
|
Guénette S, Strecker P, Kins S. APP Protein Family Signaling at the Synapse: Insights from Intracellular APP-Binding Proteins. Front Mol Neurosci 2017; 10:87. [PMID: 28424586 PMCID: PMC5371672 DOI: 10.3389/fnmol.2017.00087] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 03/13/2017] [Indexed: 12/17/2022] Open
Abstract
Understanding the molecular mechanisms underlying amyloid precursor protein family (APP/APP-like proteins, APLP) function in the nervous system can be achieved by studying the APP/APLP interactome. In this review article, we focused on intracellular APP interacting proteins that bind the YENPTY internalization motif located in the last 15 amino acids of the C-terminal region. These proteins, which include X11/Munc-18-interacting proteins (Mints) and FE65/FE65Ls, represent APP cytosolic binding partners exhibiting different neuronal functions. A comparison of FE65 and APP family member mutant mice revealed a shared function for APP/FE65 protein family members in neurogenesis and neuronal positioning. Accumulating evidence also supports a role for membrane-associated APP/APLP proteins in synapse formation and function. Therefore, it is tempting to speculate that APP/APLP C-terminal interacting proteins transmit APP/APLP-dependent signals at the synapse. Herein, we compare our current knowledge of the synaptic phenotypes of APP/APLP mutant mice with those of mice lacking different APP/APLP interaction partners and discuss the possible downstream effects of APP-dependent FE65/FE65L or X11/Mint signaling on synaptic vesicle release, synaptic morphology and function. Given that the role of X11/Mint proteins at the synapse is well-established, we propose a model highlighting the role of FE65 protein family members for transduction of APP/APLP physiological function at the synapse.
Collapse
Affiliation(s)
| | - Paul Strecker
- Department of Biology, Division of Human Biology, University of KaiserslauternKaiserslautern, Germany
| | - Stefan Kins
- Department of Biology, Division of Human Biology, University of KaiserslauternKaiserslautern, Germany
| |
Collapse
|
8
|
Motodate R, Saito Y, Hata S, Suzuki T. Expression and localization of X11 family proteins in neurons. Brain Res 2016; 1646:227-234. [DOI: 10.1016/j.brainres.2016.05.054] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 05/28/2016] [Accepted: 05/31/2016] [Indexed: 01/10/2023]
|
9
|
Laarakker MC, Reinders NR, Bruining H, Ophoff RA, Kas MJH. Sex-dependent novelty response in neurexin-1α mutant mice. PLoS One 2012; 7:e31503. [PMID: 22348092 PMCID: PMC3278455 DOI: 10.1371/journal.pone.0031503] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2011] [Accepted: 01/12/2012] [Indexed: 11/19/2022] Open
Abstract
Neurexin-1 alpha (NRXN1α) belongs to the family of cell adhesion molecules (CAMs), which are involved in the formation of neuronal networks and synapses. NRXN1α gene mutations have been identified in neuropsychiatric diseases including Schizophrenia (SCZ) and Autism Spectrum Disorder (ASD). In order to get a better understanding of the pleiotropic behavioral manifestations caused by NRXN1α gene mutations, we performed a behavioral study of Nrxn1α heterozygous knock-out (+/−) mice and observed increased responsiveness to novelty and accelerated habituation to novel environments compared to wild type (+/+) litter-mates. However, this effect was mainly observed in male mice, strongly suggesting that gender-specific mechanisms play an important role in Nrxn1α-induced phenotypes.
Collapse
Affiliation(s)
- Marijke C. Laarakker
- Department of Neurosciences & Pharmacology, Division of Neuroscience, Rudolf Magnus Institute, University Medical Center Utrecht, The Netherlands
| | - Niels R. Reinders
- Department of Neurosciences & Pharmacology, Division of Neuroscience, Rudolf Magnus Institute, University Medical Center Utrecht, The Netherlands
| | - Hilgo Bruining
- Department of Neurosciences & Pharmacology, Division of Neuroscience, Rudolf Magnus Institute, University Medical Center Utrecht, The Netherlands
- Department of Psychiatry, Division of Neuroscience, Rudolf Magnus Institute, University Medical Center Utrecht, The Netherlands
| | - Roel A. Ophoff
- Department of Psychiatry, Division of Neuroscience, Rudolf Magnus Institute, University Medical Center Utrecht, The Netherlands
- Center for Neurobehavioral Genetics, University of California Los Angeles, Los Angeles, California, United States of America
| | - Martien J. H. Kas
- Department of Neurosciences & Pharmacology, Division of Neuroscience, Rudolf Magnus Institute, University Medical Center Utrecht, The Netherlands
- * E-mail:
| |
Collapse
|
10
|
Impaired striatal dopamine output of homozygous Wfs1 mutant mice in response to [K+] challenge. J Physiol Biochem 2010; 67:53-60. [DOI: 10.1007/s13105-010-0048-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2010] [Accepted: 09/21/2010] [Indexed: 12/18/2022]
|
11
|
Mitchell JC, Perkinton MS, Yates DM, Lau KF, Rogelj B, Miller CC, McLoughlin DM. Expression of the neuronal adaptor protein X11alpha protects against memory dysfunction in a transgenic mouse model of Alzheimer's disease. J Alzheimers Dis 2010; 20:31-6. [PMID: 20378958 PMCID: PMC3023903 DOI: 10.3233/jad-2009-1341] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
X11alpha is a neuronal-specific adaptor protein that binds to the amyloid-beta protein precursor (AbetaPP). Overexpression of X11alpha reduces Abeta production but whether X11alpha also protects against Abeta-related memory dysfunction is not known. To test this possibility, we crossed X11alpha transgenic mice with AbetaPP-Tg2576 mice. AbetaPP-Tg2576 mice produce high levels of brain Abeta and develop age-related defects in memory function that correlate with increasing Abeta load. Overexpression of X11alpha alone had no detectable adverse effect upon behavior. However, X11alpha reduced brain Abeta levels and corrected spatial reference memory defects in aged X11alpha/AbetaPP double transgenics. Thus, X11alpha may be a therapeutic target for Alzheimer's disease.
Collapse
Affiliation(s)
- Jacqueline C. Mitchell
- MRC Centre for Neurodegeneration Research, King’s College London, Institute of Psychiatry, London, UK
| | - Michael S. Perkinton
- MRC Centre for Neurodegeneration Research, King’s College London, Institute of Psychiatry, London, UK
| | - Darran M. Yates
- MRC Centre for Neurodegeneration Research, King’s College London, Institute of Psychiatry, London, UK
| | - Kwok-Fai Lau
- Department of Biochemistry (Science), The Chinese University of Hong Kong, Shatin NT, Hong Kong SAR
| | - Boris Rogelj
- MRC Centre for Neurodegeneration Research, King’s College London, Institute of Psychiatry, London, UK
| | - Christopher C.J. Miller
- MRC Centre for Neurodegeneration Research, King’s College London, Institute of Psychiatry, London, UK
| | - Declan M. McLoughlin
- MRC Centre for Neurodegeneration Research, King’s College London, Institute of Psychiatry, London, UK
- Department of Psychiatry and Trinity College Institute of Neuroscience, Trinity College Dublin, St Patrick’s University Hospital, Dublin, Ireland
| |
Collapse
|
12
|
Mitchell JC, Ariff BB, Yates DM, Lau KF, Perkinton MS, Rogelj B, Stephenson JD, Miller CCJ, McLoughlin DM. X11beta rescues memory and long-term potentiation deficits in Alzheimer's disease APPswe Tg2576 mice. Hum Mol Genet 2009; 18:4492-500. [PMID: 19744962 DOI: 10.1093/hmg/ddp408] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Increased production and deposition of amyloid beta-protein (Abeta) are believed to be key pathogenic events in Alzheimer's disease. As such, routes for lowering cerebral Abeta levels represent potential therapeutic targets for Alzheimer's disease. X11beta is a neuronal adaptor protein that binds to the intracellular domain of the amyloid precursor protein (APP). Overexpression of X11beta inhibits Abeta production in a number of experimental systems. However, whether these changes to APP processing and Abeta production induced by X11beta overexpression also induce beneficial effects to memory and synaptic plasticity are not known. We report here that X11beta-mediated reduction in cerebral Abeta is associated with normalization of both cognition and in vivo long-term potentiation in aged APPswe Tg2576 transgenic mice that model the amyloid pathology of Alzheimer's disease. Overexpression of X11beta itself has no detectable adverse effects upon mouse behaviour. These findings support the notion that modulation of X11beta function represents a therapeutic target for Abeta-mediated neuronal dysfunction in Alzheimer's disease.
Collapse
Affiliation(s)
- Jacqueline C Mitchell
- MRC Centre for Neurodegeneration Research, King's College London, Institute of Psychiatry, London SE5 8AF, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Saluja I, Paulson H, Gupta A, Turner RS. X11alpha haploinsufficiency enhances Abeta amyloid deposition in Alzheimer's disease transgenic mice. Neurobiol Dis 2009; 36:162-8. [PMID: 19631749 DOI: 10.1016/j.nbd.2009.07.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2009] [Revised: 07/02/2009] [Accepted: 07/13/2009] [Indexed: 10/20/2022] Open
Abstract
The neuronal adaptor protein X11alpha/mint-1/APBA-1 binds to the cytoplasmic domain of the amyloid precursor protein (APP) to modulate its trafficking and metabolism. We investigated the consequences of reducing X11alpha in a mouse model of Alzheimer's disease (AD). We crossed hAPPswe/PS-1DeltaE9 transgenic (AD tg) mice with X11alpha heterozygous knockout mice in which X11alpha expression is reduced by approximately 50%. The APP C-terminal fragments C99 and C83, as well as soluble Abeta40 and Abeta42, were increased significantly in brain of X11alpha haploinsufficient mice. Abeta/amyloid plaque burden also increased significantly in the hippocampus and cortex of one year old AD tg/X11alpha (+/-) mice compared to AD tg mice. In contrast, the levels of sAPPalpha and sAPPbeta were not altered significantly in AD tg/X11alpha (+/-) mice. The increased neuropathological indices of AD in mice expressing reduced X11alpha suggest a normal suppressor role for X11alpha on CNS Abeta/amyloid deposition.
Collapse
|
14
|
Abstract
Understanding how emotion is generated, how conflicting emotions are regulated, and how emotional states relate to sophisticated behaviors is a crucial challenge in brain research. Model animals showing selective emotion-related phenotypes are highly useful for examining these issues. Here, we describe a novel mouse model that withdraws in approach-avoidance conflicts. X11-like (X11L)/Mint2 is a neuronal adapter protein with multiple protein-protein interaction domains that interacts with several proteins involved in modulating neuronal activity. X11L-knock-out (KO) mice were subordinate under competitive feeding conditions. X11L-KO mice lost significantly more weight than cohoused wild-type mice without signs of decreased motivation to eat or physical weakness. In a resident-intruder test, X11L-KO mice showed decreased intruder exploration behavior. Moreover, X11L-KO mice displayed decreased marble-burying, digging and burrowing behaviors, indicating aberrant ethological responses to attractive stimuli. In contrast, X11L-KO mice were indistinguishable from wild-type mice in the open field, elevated plus maze, and light/dark transition tests, which are often used to assess anxiety-like behavior. Neurochemical analysis revealed a monoamine imbalance in several forebrain regions. The defective ethological responses and social behaviors in X11L-KO mice were rescued by the expression of X11L under a Camk2a promoter using the Tet-OFF system during development. These findings suggest that X11L is involved in the development of neuronal circuits that contribute to conflict resolution.
Collapse
|
15
|
Lachman HM. Copy variations in schizophrenia and bipolar disorder. Cytogenet Genome Res 2009; 123:27-35. [PMID: 19287136 DOI: 10.1159/000184689] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/30/2008] [Indexed: 01/19/2023] Open
Abstract
The analysis of copy number variations (CNVs) is an emerging tool for identifying genetic factors underlying complex traits. In this chapter I will review studies that have been carried out showing that CNVs play a role in the development of two such complex traits; schizophrenia (SZ) and bipolar disorder (BD). There are two aspects to consider regarding the role of copy variations in these conditions. One is gene discovery in which DNA from patients is analyzed for the purpose of identifying rare, patient-specific CNVs that may be informative to a larger population of affected individuals. The model for this concept is based on the emergence of DISC1 as a SZ candidate gene, which was discovered in a single informative family with a rare chromosomal translocation. Another aspect revolves around the idea that polymorphic CNVs found in the general population, many of which appear to disrupt previously identified SZ and BD candidate genes, contribute to disease pathogenesis. Here, gene-disrupting CNVs are viewed in the same manner as functional SNPs and analyzed for involvement in disease susceptibility using genetic association. Although the analysis of CNVs in patients with psychiatric disorders is in its infancy, informative new findings have already been made, suggesting that this is a very promising line of research.
Collapse
Affiliation(s)
- H M Lachman
- Department of Psychiatry and Behavioral Sciences, Division of Basic Research Albert Einstein College of Medicine, Bronx, New York, USA.
| |
Collapse
|
16
|
Saito Y, Sano Y, Vassar R, Gandy S, Nakaya T, Yamamoto T, Suzuki T. X11 proteins regulate the translocation of amyloid beta-protein precursor (APP) into detergent-resistant membrane and suppress the amyloidogenic cleavage of APP by beta-site-cleaving enzyme in brain. J Biol Chem 2008; 283:35763-71. [PMID: 18845544 PMCID: PMC2602917 DOI: 10.1074/jbc.m801353200] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2008] [Revised: 09/22/2008] [Indexed: 11/06/2022] Open
Abstract
X11 and X11-like proteins (X11L) are neuronal adaptor proteins whose association to the cytoplasmic domain of amyloid beta-protein precursor (APP) suppresses the generation of amyloid beta-protein (Abeta) implicated in Alzheimer disease pathogenesis. The amyloidogenic, but not amyloidolytic, metabolism of APP was selectively increased in the brain of mutant mice lacking X11L (Sano, Y., Syuzo-Takabatake, A., Nakaya, T., Saito, Y., Tomita, S., Itohara, S., and Suzuki, T. (2006) J. Biol. Chem. 281, 37853-37860). To reveal the actual role of X11 proteins (X11s) in suppressing amyloidogenic cleavage of APP in vivo, we generated X11 and X11L double knock-out mice and analyzed the metabolism of APP. The mutant mice showed enhanced beta-site cleavage of APP along with increased accumulation of Abeta in brain and increased colocalization of APP with beta-site APP-cleaving enzyme (BACE). In the brains of mice deficient in both X11 and X11L, the apparent relative subcellular distributions of both mature APP and its beta-C-terminal fragment were shifted toward the detergent-resistant membrane (DRM) fraction, an organelle in which BACE is active and both X11s are not nearly found. These results indicate that X11s associate primarily with APP molecules that are outside of DRM, that the dissociation of APP-X11/X11L complexes leads to entry of APP into DRM, and that cleavage of uncomplexed APP by BACE within DRM is enhanced by X11s deficiency. Present results lead to an idea that the dysfunction of X11L in the interaction with APP may recruit more APP into DRM and increase the generation of Abeta even if BACE activity did not increase in brain.
Collapse
Affiliation(s)
- Yuhki Saito
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita12-Nishi6, Sapporo 060-0812, Japan
| | | | | | | | | | | | | |
Collapse
|
17
|
A validated gas chromatographic-electron impact ionization mass spectrometric method for methamphetamine, methylenedioxymethamphetamine (MDMA), and metabolites in mouse plasma and brain. J Chromatogr B Analyt Technol Biomed Life Sci 2008; 876:266-76. [PMID: 19026602 DOI: 10.1016/j.jchromb.2008.11.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2008] [Revised: 10/27/2008] [Accepted: 11/02/2008] [Indexed: 11/23/2022]
Abstract
A method was developed and fully validated for simultaneous quantification of methamphetamine (MAMP), amphetamine, hydroxy-methamphetamine, methylenedioxymethamphetamine (MDMA, ecstasy), methylenedioxyamphetamine, 3-hydroxy-4-methoxy-methamphetamine, and 3-hydroxy-4-methoxy-amphetamine in 100 microL mouse plasma and 7.5mg brain. Solid phase extraction and gas chromatography-electron impact ionization mass spectrometry in selected-ion monitoring mode achieved plasma linear ranges of 10-20 to 20,000 ng/mL and 0.1-0.2 to 200 ng/mg in brain. Recoveries were greater than 91%, bias 92.3-110.4%, and imprecision less than 5.3% coefficient of variation. This method was used for measuring MAMP and MDMA and metabolites in plasma and brain during mouse neurotoxicity studies.
Collapse
|
18
|
The X11L/X11beta/MINT2 and X11L2/X11gamma/MINT3 scaffold proteins shuttle between the nucleus and cytoplasm. Exp Cell Res 2007; 314:1155-62. [PMID: 18201694 DOI: 10.1016/j.yexcr.2007.12.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2007] [Revised: 12/04/2007] [Accepted: 12/05/2007] [Indexed: 11/21/2022]
Abstract
The X11/MINT family proteins are adaptor scaffolding proteins involved in formation of multiprotein complexes, and trafficking and metabolism of membrane proteins such as the beta-amyloid precursor protein. We found that a significant portion of X11L and X11L2 are recovered in nuclear fraction of mouse brain homogenates. EGFP-X11s were not detected in the nucleus of N2a neuroblastoma cells; however, administration of leptomycin B (LMB) induced substantial nuclear accumulation of EGFP-X11L and EGFP-X11L2, while EGFP-X11 showed little accumulation. Fluorescence loss in photobleaching (FLIP) analysis indicated that EGFP-X11L2 and EGFP-X11L are shuttled between the cytoplasm and nucleus, the former more effectively than the latter. We identified a nuclear export signal (NES) in the N-terminus of X11L2, mutation of which induces nuclear accumulation of EGFP-X11L2 in the absence of LMB. X11L2 fused to the Gal4 DNA binding domain (DBD) showed transcriptional activity, suggesting that X11L2 could function as a transcriptional activator if tethered near a promoter. Interestingly, attenuation of the nucleo-cytoplasmic shuttling of GAL4-DBD-X11L2 by mutating the NES or attaching the SV40 nuclear localization signal significantly decreased the apparent transcriptional activity. Our observations suggest that X11L2 functions in the nucleus by a mechanism distinct from conventional transactivators.
Collapse
|
19
|
Phillips TJ, Kamens HM, Wheeler JM. Behavioral genetic contributions to the study of addiction-related amphetamine effects. Neurosci Biobehav Rev 2007; 32:707-59. [PMID: 18207241 PMCID: PMC2360482 DOI: 10.1016/j.neubiorev.2007.10.008] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2007] [Revised: 09/28/2007] [Accepted: 10/28/2007] [Indexed: 11/24/2022]
Abstract
Amphetamines, including methamphetamine, pose a significant cost to society due to significant numbers of amphetamine-abusing individuals who suffer major health-related consequences. In addition, methamphetamine use is associated with heightened rates of violent and property-related crimes. The current paper reviews the existing literature addressing genetic differences in mice that impact behavioral responses thought to be relevant to the abuse of amphetamine and amphetamine-like drugs. Summarized are studies that used inbred strains, selected lines, single-gene knockouts and transgenics, and quantitative trait locus (QTL) mapping populations. Acute sensitivity, neuroadaptive responses, rewarding and conditioned effects are among those reviewed. Some gene mapping work has been accomplished, and although no amphetamine-related complex trait genes have been definitively identified, translational work leading from results in the mouse to studies performed in humans is beginning to emerge. The majority of genetic investigations have utilized single-gene knockout mice and have concentrated on dopamine- and glutamate-related genes. Genes that code for cell support and signaling molecules are also well-represented. There is a large behavioral genetic literature on responsiveness to amphetamines, but a considerably smaller literature focused on genes that influence the development and acceleration of amphetamine use, withdrawal, relapse, and behavioral toxicity. Also missing are genetic investigations into the effects of amphetamines on social behaviors. This information might help to identify at-risk individuals and in the future to develop treatments that take advantage of individualized genetic information.
Collapse
|
20
|
Ho A, Morishita W, Atasoy D, Liu X, Tabuchi K, Hammer RE, Malenka RC, Südhof TC. Genetic analysis of Mint/X11 proteins: essential presynaptic functions of a neuronal adaptor protein family. J Neurosci 2007; 26:13089-101. [PMID: 17167098 PMCID: PMC6674967 DOI: 10.1523/jneurosci.2855-06.2006] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Mints/X11s are adaptor proteins composed of three isoforms: neuron-specific Mints 1 and 2, and the ubiquitously expressed Mint 3. We have now analyzed constitutive and conditional knock-out mice for all three Mints/X11s. We found that approximately 80% of mice lacking both neuron-specific Mint isoforms (Mints 1 and 2) die at birth, whereas mice lacking any other combination of Mint isoforms survive normally. The approximately 20% surviving Mint 1/2 double knock-out mice exhibit a decrease in weight and deficits in motor behaviors. Hippocampal slice electrophysiology uncovered a decline in spontaneous neurotransmitter release, lowered synaptic strength, and enhanced paired-pulse facilitation in Mint-deficient mice, suggesting a decreased presynaptic release probability. Acute ablation of Mint expression in cultured neurons from conditional Mint 1/2/3 triple knock-in mice also revealed a decline in spontaneous release, confirming that deletion of Mints impair presynaptic function. Quantitation of synaptic proteins showed that acute deletion of Mints caused a selective increase in Munc18-1 and Fe65 proteins, and overexpression of Munc18-1 in wild-type neurons also produced a decrease in spontaneous release, suggesting that the interaction of Mints with Munc18-1 may contribute to the presynaptic phenotype observed in Mint-deficient mice. Our studies thus indicate that Mints are important regulators of presynaptic neurotransmitter release that are essential for mouse survival.
Collapse
Affiliation(s)
| | - Wade Morishita
- Department of Psychiatry and Behavioral Sciences, Nancy Friend Pritzker Laboratory, Stanford University School of Medicine, Stanford, California 94304
| | | | | | | | - Robert E. Hammer
- Biochemistry, and
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9111, and
| | - Robert C. Malenka
- Department of Psychiatry and Behavioral Sciences, Nancy Friend Pritzker Laboratory, Stanford University School of Medicine, Stanford, California 94304
| | - Thomas C. Südhof
- Center for Basic Neuroscience
- Departments of Molecular Genetics and
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9111, and
| |
Collapse
|
21
|
Sano Y, Syuzo-Takabatake A, Nakaya T, Saito Y, Tomita S, Itohara S, Suzuki T. Enhanced Amyloidogenic Metabolism of the Amyloid β-Protein Precursor in the X11L-deficient Mouse Brain. J Biol Chem 2006; 281:37853-60. [PMID: 17032642 DOI: 10.1074/jbc.m609312200] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
X11L, a neuronal adaptor protein, associates with the cytoplasmic domain of APP and suppresses APP cellular metabolism. APP is the precursor of Abeta, whose metabolism is strongly implicated in Alzheimer disease pathogenesis. To examine the roles of X11L function in APP metabolism, including the generation of Abeta in the brain, we produced X11L-deficient mutant mice on the C57BL/6 background. The mutant mice did not exhibit histopathological alterations or compensatory changes in the expression of other X11 family proteins, X11 and X11L2. The expression level and distribution of APP in the brain of mutant mice were also identical to those in wild-type mice. However, in the hippocampus, where substantial levels of X11L and APP are expressed, the mutant mice exhibited a significant increase in the level of the C-terminal fragments of APP produced by cleavage with beta-secretase but not alpha-secretase. The levels of Abeta were increased in the hippocampus of aged mutant mice as compared with age-matched controls. These observations clearly indicate that X11L suppresses the amyloidogenic but not amyloidolytic processing of APP in regions of the brain such as the hippocampus, which express significant levels of X11L.
Collapse
Affiliation(s)
- Yoshitake Sano
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita-12 Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | | | | | | | | | | | | |
Collapse
|
22
|
Rogelj B, Mitchell JC, Miller CCJ, McLoughlin DM. The X11/Mint family of adaptor proteins. ACTA ACUST UNITED AC 2006; 52:305-15. [PMID: 16764936 DOI: 10.1016/j.brainresrev.2006.04.005] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2006] [Revised: 03/27/2006] [Accepted: 04/16/2006] [Indexed: 12/25/2022]
Abstract
The X11 protein family are multidomain proteins composed of a conserved PTB domain and two C-terminal PDZ domains. They are involved in formation of multiprotein complexes and two of the family members, X11alpha and X11beta, are expressed primarily in neurones. Not much is known about the principal function of X11s, but through interactions with other neuronal proteins, they are believed to be involved in regulating neuronal signaling, trafficking and plasticity. Furthermore, they have been shown to modulate processing of APP and accumulation of Abeta, making them potential therapeutic targets for Alzheimer's disease. This article reviews the known interactions of the different X11s and their involvement in Alzheimer's disease.
Collapse
Affiliation(s)
- Boris Rogelj
- King's College London, MRC Centre for Neurodegeneration Research, Department of Neuroscience, Institute of Psychiatry, De Crespigny Park, London SE5 8AF, UK
| | | | | | | |
Collapse
|
23
|
Vishnu S, Hertenstein A, Betschinger J, Knoblich JA, Gert de Couet H, Fischbach KF. The adaptor protein X11Lα/Dmint1 interacts with the PDZ-binding domain of the cell recognition protein Rst in Drosophila. Dev Biol 2006; 289:296-307. [PMID: 16380111 DOI: 10.1016/j.ydbio.2005.09.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2004] [Revised: 07/04/2005] [Accepted: 09/09/2005] [Indexed: 12/22/2022]
Abstract
The Drosophila cell adhesion molecule Rst plays key roles during the development of the embryonic musculature, spacing of ommatidia in the compound eye and of sensory organs on the antenna, as well as in the neuronal wiring of the optic lobe. In rst(CT) mutants lacking the cytoplasmic domain of the Rst protein, cell sorting and apoptosis in the eye are affected, suggesting a requirement of this domain for Rst function. To identify potential interacting proteins, yeast two-hybrid screens were performed using the cytoplasmic domains of Rst and its paralogue Kirre as baits. Among several putative interactors, two paralogous Drosophila PDZ motif proteins related to X11/Mint were identified. X11/Mint family members in C. elegans (LIN-10) and vertebrates are believed to function as adaptor proteins and to regulate the assembly of multi-subunit complexes at the synapse, thereby linking the vesicle cycle to cell adhesion. Using genetic, cell biological, and biochemical approaches, we show that the interaction of Rst with X11Lalpha is of biological significance. The proteins interact, for example, in the context of cell sorting in the pupal retina.
Collapse
Affiliation(s)
- Smitha Vishnu
- Institut für Biologie III, Albert-Ludwigs Universität, D-79104 Freiburg im Breisgau, Germany
| | | | | | | | | | | |
Collapse
|
24
|
Lockyer S, Okuyama K, Begum S, Le S, Sun B, Watanabe T, Matsumoto Y, Yoshitake M, Kambayashi J, Tandon NN. GPVI-deficient mice lack collagen responses and are protected against experimentally induced pulmonary thromboembolism. Thromb Res 2006; 118:371-80. [PMID: 16139873 DOI: 10.1016/j.thromres.2005.08.001] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2005] [Revised: 07/14/2005] [Accepted: 08/02/2005] [Indexed: 11/24/2022]
Abstract
Platelet glycoprotein VI (GPVI) is now considered to be a major player in platelet-collagen adhesive interactions leading to thrombus formation. GPVI blockade, or its depletion, has been shown in mice to result in complete protection against arterial thrombosis, without significant prolongation of bleeding time. GPVI may therefore represent a useful antithrombotic target. In order to reaffirm the role of GPVI in platelet-collagen interactions, we developed GPVI(null) mice by targeted disruption methodology. GPVI(null) mice platelets failed to respond to a high dose of fibrillar collagen, or convulxin, a GPVI agonist, but showed a normal response to other agonists such as ADP, PMA and arachidonic acid. We report, for the first time, that a proportion of GPVI(null) mice is protected against lethal thromboembolism, induced by the infusion of a mixture of collagen and epinephrine. Greater than 55% of GPVI(null) mice survived the challenge, whereas the maximal survival from the other genotypes was 17% (n=18 per genotype). Washed platelets obtained from GPVI(null) mice showed >90% reduction in adhesion to fibrillar collagen under static conditions. Platelet adhesion to collagen under dynamic conditions using a high shear rate (2600 s(-1)) was dramatically reduced using blood from GPVI(null) mice, while platelets from wild-type and heterozygous animals showed a similar amount of adhesion. Animals from each genotype had essentially similar tail bleeding time, suggesting that a complete deficiency of GPVI, at least in mice, does not result in an enhanced bleeding tendency. These observations clearly establish that blockade of GPVI may attenuate platelet-collagen interactions without adversely affecting the bleeding time.
Collapse
Affiliation(s)
- Simon Lockyer
- Otsuka Maryland Medicinal Laboratories, 9900 Medical Center Drive, Rockville, MD 20850, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Jacobs EH, Williams RJ, Francis PT. Cyclin-dependent kinase 5, Munc18a and Munc18-interacting protein 1/X11α protein up-regulation in Alzheimer’s disease. Neuroscience 2006; 138:511-22. [PMID: 16413130 DOI: 10.1016/j.neuroscience.2005.11.017] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2005] [Revised: 11/11/2005] [Accepted: 11/15/2005] [Indexed: 12/01/2022]
Abstract
Besides formation of neurofibrillary tangles and neuron loss, the Alzheimer's disease brain is characterized by neuritic plaques consisting of beta-amyloid peptide deposits and impaired neurotransmission. The proteins Munc18a, Munc18-interacting protein 1 and Munc18-interacting protein 2 mediate exocytosis and decrease beta-amyloid peptide formation. Cyclin-dependent kinase 5 and its activator p35 disrupt Munc18a-syntaxin 1 binding, thereby promoting synaptic vesicle fusion during exocytosis. We investigated protein levels of the signaling pathway: p35, cyclin-dependent kinase 5, Munc18a, syntaxin 1A and 1B, Munc18-interacting protein 1 and Munc18-interacting protein 2 in Alzheimer's disease cortex and found that this pathway was up-regulated in the Alzheimer's disease parietal and occipital cortex. In the cortex of transgenic Tg2576 mice over-expressing human beta-amyloid precursor protein with the Swedish mutation known to lead to familial Alzheimer's disease, which have substantial levels of beta-amyloid peptide but lack neurofibrillary tangles and neuron loss, no alterations of protein levels were detected. These data suggest that the pathway is enhanced in dying or surviving neurons and might serve a protective role by compensating for decreased neurotransmission and decreasing beta-amyloid peptide levels early during the progression of Alzheimer's disease.
Collapse
Affiliation(s)
- E H Jacobs
- Wolfson Centre for Age-Related Diseases, Guy's, King's and St Thomas' Schools of Biomedical Sciences, King's College London, St. Thomas Street, London SE1 1UL, UK.
| | | | | |
Collapse
|
26
|
Kimura K, Kitano J, Nakajima Y, Nakanishi S. Hyperpolarization-activated, cyclic nucleotide-gated HCN2 cation channel forms a protein assembly with multiple neuronal scaffold proteins in distinct modes of protein-protein interaction. Genes Cells 2005; 9:631-40. [PMID: 15265006 DOI: 10.1111/j.1356-9597.2004.00752.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Hyperpolarization-activated cation currents, termed Ih, are non-uniformly distributed along dendritic arbors with current density increasing with increasing distance from the soma. The non-uniform distribution of Ih currents contributes to normalization of location-dependent variability in temporal integration of synaptic input, but the molecular basis for the graded HCN distribution remains to be investigated. The hyperpolarization-activated, cyclic nucleotide-gated cation channels (HCNs) underlie Ih currents and consist of four members (HCN1-HCN4) of the gene family in mammals. In this investigation, we report that HCN2 forms a protein assembly with tamalin, S-SCAM and Mint2 scaffold proteins, using several different approaches including immunoprecipitation of rat brain and heterologously expressing cell extracts and glutathione S-transferase pull-down assays. The PDZ domain of tamalin interacts with HCN2 at both the PDZ-binding motif and the internal carboxy-terminal tail of HCN2, whereas binding of the PDZ domain of S-SCAM occurs at the cyclic nucleotide-binding domain (CNBD) and the CNBD-downstream sequence of the carboxy-terminal tail of HCN2. A protein assembly between HCN2 and Mint2 is formed by the interaction of the munc18-interacting domain of Mint2 with the CNBD-downstream sequence of HCN2. The results demonstrate that HCN2 forms a protein complex with multiple neuronal scaffold proteins in distinct modes of protein-protein interaction.
Collapse
Affiliation(s)
- Kouji Kimura
- Department of Biological Sciences, Faculty of Medicine, and Department of Molecular and System Biology, Graduate School of Biostudies, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | | | | | | |
Collapse
|
27
|
Mori A, Ohashi S, Nakai M, Moriizumi T, Mitsumoto Y. Neural mechanisms underlying motor dysfunction as detected by the tail suspension test in MPTP-treated C57BL/6 mice. Neurosci Res 2005; 51:265-74. [PMID: 15710490 DOI: 10.1016/j.neures.2004.11.008] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2004] [Revised: 11/24/2004] [Accepted: 11/29/2004] [Indexed: 10/25/2022]
Abstract
Contradictory data on behavioral changes in MPTP-treated C57BL/6 mice have been reported, even though the toxin-treated mice have been widely used for non-clinical studies as an in vivo model of Parkinson's disease (PD). We found that the duration of immobility in the tail suspension test (TST) was significantly increased in MPTP-treated C57BL/6 mice as compared with control mice without a significant change in the locomotor activity (LA). Dopamine (DA) contents and protein levels of tyrosine hydroxylase and dopamine transporter in the striatum were profoundly decreased in the toxin-treated mice. These behavioral and neurobiochemical changes were almost completely inhibited by a pretreatment with deprenyl, a monoamine oxidase-B inhibitor. The stimulation of dopaminergic neurotransmission induced by L-dopa or a dopamine D2 receptor agonist ameliorated the increase in immobility time. Threshold level of striatal DA that produced the increase in immobility time in MPTP-treated mice was estimated to be between 11 and 27% of control level. We concluded that the increase in immobility time in the TST was induced by the nigrostriatal dopaminergic degeneration and was thought to be a consequence of motor dysfunction in this mouse model of PD.
Collapse
Affiliation(s)
- Atsushi Mori
- Research Unit for Neurological Diseases, Second Institute of New Drug Discovery, Otsuka Pharmaceutical Co., Ltd., Tokushima-city, Tokushima 771-0192, Japan
| | | | | | | | | |
Collapse
|
28
|
Zhang W, Lilja L, Bark C, Berggren PO, Meister B. Mint1, a Munc-18-interacting protein, is expressed in insulin-secreting beta-cells. Biochem Biophys Res Commun 2004; 320:717-21. [PMID: 15240107 DOI: 10.1016/j.bbrc.2004.05.208] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2004] [Indexed: 11/17/2022]
Abstract
Munc-18-interacting (Mint) proteins are adaptors involved in regulation of synaptic vesicle exocytosis. We have investigated expression and cellular localization of Mint1 in pancreatic islets with special reference to insulin-secreting beta-cells. Western blotting showed that Mint1 was expressed in hamster (HIT-T15) and rat (RINm5F) beta-cell lines. Mint1 immunoreactivity was preferentially localized to the periphery of individual islet cells. RT-PCR analysis revealed that apart from Mint1, RINm5F cells and rat islets also transcribed the mRNAs for Mint2 and Mint3. Expression of Mint proteins in pancreatic beta-cells suggests a functional role for these proteins in insulin granule exocytosis.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Neuroscience, Karolinska Institutet, SE-171 77, Stockholm, Sweden
| | | | | | | | | |
Collapse
|
29
|
King GD, Scott Turner R. Adaptor protein interactions: modulators of amyloid precursor protein metabolism and Alzheimer's disease risk? Exp Neurol 2004; 185:208-19. [PMID: 14736502 DOI: 10.1016/j.expneurol.2003.10.011] [Citation(s) in RCA: 124] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The cytoplasmic C-terminus of APP plays critical roles in its cellular trafficking and delivery to proteases. Adaptor proteins with phosphotyrosine-binding (PTB) domains, including those in the X11, Fe65, and c-Jun N-terminal kinase (JNK)-interacting protein (JIP) families, bind specifically to the absolutely conserved -YENPTY- motif in the APP C-terminus to regulate its trafficking and processing. Compounds that modulate APP-adaptor protein interactions may inhibit Abeta generation by specifically targeting the substrate (APP) instead of the enzyme (beta- or gamma-secretase). Genetic polymorphisms in (or near) adaptor proteins may influence risk of sporadic AD by interacting with APP in vivo to modulate its trafficking and processing to Abeta.
Collapse
Affiliation(s)
- Gwendalyn D King
- Neuroscience Program, University of Michigan, Ann Arbor, MI 48105, USA
| | | |
Collapse
|
30
|
Scorza CA, Garrido YDC, Arida RM, Amado D, Cavalheiro EA, Naffah-Mazzacoratti MDG. Levels of the synaptic protein X11 alpha/mint1 are increased in hippocampus of rats with epilepsy. Epilepsy Res 2003; 57:49-57. [PMID: 14706732 DOI: 10.1016/j.eplepsyres.2003.10.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
X11 alpha or Mint1 is a protein containing an N-terminal sequence, which binds to Munc-18 protein, a middle phosphotyrosine-binding domain (PTB) and two C-terminal PDZ (Post-synaptic density/Discs large/Zone Occludens-1) domains. The PDZ domains, which mediate protein-protein interactions have been shown to be involved in the organization of synaptic signaling pathways. Mint1 plays an important role in vesicle synaptic transport toward the active zone at the pre-synaptic site, and also participates in the transport of NR2B subunit of the NMDA receptor, to the post-synaptic site. To investigate the participation and distribution of this protein in the hippocampal subfield of rats submitted to the pilocarpine model of epilepsy, Mint1 was analyzed using Western blotting and immunohistochemistry. Animals of 5 h of status epilepticus showed decreased levels of this protein in the hippocampus when compared to the control animals. In contrast, animals from seizure-free period (silent group) and during spontaneous seizures phase (chronic group) showed increased Mint1 immunostaining in all hippocampal subfields, mainly in the dentate gyrus, when compared to the control group. The blotting confirmed the results obtained by immunohistochemistry. The present work suggests that Mint1 may be related to hippocampal plasticity during epileptogenesis in the pilocarpine model of temporal lobe epilepsy.
Collapse
Affiliation(s)
- Carla Alessandra Scorza
- Laboratório de Neurociência, Disciplina de Neurologia Experimental, Universidade Federal de São Paulo, Rua Botucatu 862, Ed Leal Prado CEP-04023-900, São Paulo, Brazil
| | | | | | | | | | | |
Collapse
|
31
|
Ho A, Morishita W, Hammer RE, Malenka RC, Sudhof TC. A role for Mints in transmitter release: Mint 1 knockout mice exhibit impaired GABAergic synaptic transmission. Proc Natl Acad Sci U S A 2003; 100:1409-14. [PMID: 12547917 PMCID: PMC298786 DOI: 10.1073/pnas.252774899] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Mints (also called X11-like proteins) are adaptor proteins composed of divergent N-terminal sequences that bind to synaptic proteins such as CASK (Mint 1 only) and Munc18-1 (Mints 1 and 2) and conserved C-terminal PTB- and PDZ-domains that bind to widely distributed proteins such as APP, presenilins, and Ca(2+) channels (all Mints). We find that Mints 1 and 2 are similarly expressed in most neurons except for inhibitory interneurons that contain selectively high levels of Mint 1. Using knockout mice, we show that deletion of Mint 1 does not impair survival or alter the overall brain architecture, arguing against an essential developmental function of the Mint 1-CASK complex. In electrophysiological recordings in the hippocampus, we observed no changes in short- or long-term synaptic plasticity in excitatory synapses from Mint 1-deficient mice and detected no alterations in the ratio of alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) to N-methyl-d-aspartate (NMDA) receptor-mediated synaptic currents. Thus the Mint 1-CASK complex is not required for AMPA- and NMDA-receptor functions or for synaptic plasticity in excitatory synapses. In inhibitory synapses, however, we uncovered an approximately 3-fold increase in presynaptic paired-pulse depression, suggesting that deletion of Mint 1 impairs the regulation of gamma-aminobutyric acid release. Our data indicate that Mints 1 and 2 perform redundant synaptic functions that become apparent in Mint 1-deficient mice in inhibitory interneurons because these neurons selectively express higher levels of Mint 1 than Mint 2.
Collapse
Affiliation(s)
- Angela Ho
- Center for Basic Neuroscience, Department of Molecular Genetics, and Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA
| | | | | | | | | |
Collapse
|