1
|
Percelay S, Lahogue C, Billard JM, Freret T, Boulouard M, Bouet V. The 3-hit animal models of schizophrenia: Improving strategy to decipher and treat the disease? Neurosci Biobehav Rev 2024; 157:105526. [PMID: 38176632 DOI: 10.1016/j.neubiorev.2023.105526] [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: 10/12/2023] [Revised: 12/08/2023] [Accepted: 12/23/2023] [Indexed: 01/06/2024]
Abstract
Schizophrenia is a complex disease related to combination and interactions between genetic and environmental factors, with an epigenetic influence. After the development of the first mono-factorial animal models of schizophrenia (1-hit), that reproduced patterns of either positive, negative and/or cognitive symptoms, more complex models combining two factors (2-hit) have been developed to better fit with the multifactorial etiology of the disease. In the two past decades, a new way to design animal models of schizophrenia have emerged by adding a third hit (3-hit). This review aims to discuss the relevance of the risk factors chosen for the tuning of the 3-hit animal models, as well as the validities measurements and their contribution to schizophrenia understanding. We intended to establish a comprehensive overview to help in the choice of factors for the design of multiple-hit animal models of schizophrenia.
Collapse
Affiliation(s)
- Solenn Percelay
- Normandie Univ, UNICAEN, INSERM, CYCERON, CHU Caen, COMETE UMR 1075, 14000 Caen, France
| | - Caroline Lahogue
- Normandie Univ, UNICAEN, INSERM, CYCERON, CHU Caen, COMETE UMR 1075, 14000 Caen, France.
| | - Jean-Marie Billard
- Normandie Univ, UNICAEN, INSERM, CYCERON, CHU Caen, COMETE UMR 1075, 14000 Caen, France
| | - Thomas Freret
- Normandie Univ, UNICAEN, INSERM, CYCERON, CHU Caen, COMETE UMR 1075, 14000 Caen, France
| | - Michel Boulouard
- Normandie Univ, UNICAEN, INSERM, CYCERON, CHU Caen, COMETE UMR 1075, 14000 Caen, France
| | - Valentine Bouet
- Normandie Univ, UNICAEN, INSERM, CYCERON, CHU Caen, COMETE UMR 1075, 14000 Caen, France.
| |
Collapse
|
2
|
Wales-McGrath B, Mercer H, Piontkivska H. Changes in ADAR RNA editing patterns in CMV and ZIKV congenital infections. BMC Genomics 2023; 24:685. [PMID: 37968596 PMCID: PMC10652522 DOI: 10.1186/s12864-023-09778-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 10/31/2023] [Indexed: 11/17/2023] Open
Abstract
BACKGROUND RNA editing is a process that increases transcriptome diversity, often through Adenosine Deaminases Acting on RNA (ADARs) that catalyze the deamination of adenosine to inosine. ADAR editing plays an important role in regulating brain function and immune activation, and is dynamically regulated during brain development. Additionally, the ADAR1 p150 isoform is induced by interferons in viral infection and plays a role in antiviral immune response. However, the question of how virus-induced ADAR expression affects host transcriptome editing remains largely unanswered. This question is particularly relevant in the context of congenital infections, given the dynamic regulation of ADAR editing during brain development, the importance of this editing for brain function, and subsequent neurological symptoms of such infections, including microcephaly, sensory issues, and other neurodevelopmental abnormalities. Here, we begin to address this question, examining ADAR expression in publicly available datasets of congenital infections of human cytomegalovirus (HCMV) microarray expression data, as well as mouse cytomegalovirus (MCMV) and mouse/ human induced pluripotent neuroprogenitor stem cell (hiNPC) Zika virus (ZIKV) RNA-seq data. RESULTS We found that in all three datasets, ADAR1 was overexpressed in infected samples compared to uninfected samples. In the RNA-seq datasets, editing rates were also analyzed. In all mouse infections cases, the number of editing sites was significantly increased in infected samples, albeit this was not the case for hiNPC ZIKV samples. Mouse ZIKV samples showed altered editing of well-established protein-recoding sites such as Gria3, Grik5, and Nova1, as well as editing sites that may impact miRNA binding. CONCLUSIONS Our findings provide evidence for changes in ADAR expression and subsequent dysregulation of ADAR editing of host transcriptomes in congenital infections. These changes in editing patterns of key neural genes have potential significance in the development of neurological symptoms, thus contributing to neurodevelopmental abnormalities. Further experiments should be performed to explore the full range of editing changes that occur in different congenital infections, and to confirm the specific functional consequences of these editing changes.
Collapse
Affiliation(s)
- Benjamin Wales-McGrath
- University of Pennsylvania, Perelman School of Medicine, Department of Genetics, Philadelphia, PA, USA
- Children's Hospital of Philadelphia, Division of Cancer Pathobiology, Philadelphia, PA, USA
| | - Heather Mercer
- Department of Biological and Environmental Sciences, University of Mount Union, Alliance, OH, USA
| | - Helen Piontkivska
- Department of Biological Sciences, Kent State University, Kent, OH, USA.
- School of Biomedical Sciences, Kent State University, Kent, OH, USA.
- Brain Health Research Institute, Kent State University, Kent, OH, USA.
- Healthy Communities Research Institute, Kent State University, Kent, OH, USA.
| |
Collapse
|
3
|
Lin Y, Roy K, Ioka S, Otani R, Amezawa M, Ishikawa Y, Cherasse Y, Kaushik MK, Klewe-Nebenius D, Zhou L, Yanagisawa M, Oishi Y, Saitoh T, Lazarus M. Positive allosteric adenosine A 2A receptor modulation suppresses insomnia associated with mania- and schizophrenia-like behaviors in mice. Front Pharmacol 2023; 14:1138666. [PMID: 37153764 PMCID: PMC10155833 DOI: 10.3389/fphar.2023.1138666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 04/10/2023] [Indexed: 05/10/2023] Open
Abstract
Background: Insomnia is associated with psychiatric illnesses such as bipolar disorder or schizophrenia. Treating insomnia improves psychotic symptoms severity, quality of life, and functional outcomes. Patients with psychiatric disorders are often dissatisfied with the available therapeutic options for their insomnia. In contrast, positive allosteric modulation of adenosine A2A receptors (A2ARs) leads to slow-wave sleep without cardiovascular side effects in contrast to A2AR agonists. Methods: We investigated the hypnotic effects of A2AR positive allosteric modulators (PAMs) in mice with mania-like behavior produced by ablating GABAergic neurons in the ventral medial midbrain/pons area and in a mouse model of schizophrenia by knocking out of microtubule-associated protein 6. We also compared the properties of sleep induced by A2AR PAMs in mice with mania-like behavior with those induced by DORA-22, a dual orexin receptor antagonist that improves sleep in pre-clinical models, and the benzodiazepine diazepam. Results: A2AR PAMs suppress insomnia associated with mania- or schizophrenia-like behaviors in mice. A2AR PAM-mediated suppression of insomnia in mice with mania-like behavior was similar to that mediated by DORA-22, and, unlike diazepam, did not result in abnormal sleep. Conclusion: A2AR allosteric modulation may represent a new therapeutic avenue for sleep disruption associated with bipolar disorder or psychosis.
Collapse
Affiliation(s)
- Yang Lin
- International Institute for Integrative Sleep Medicine (WPI-IIIS), Tsukuba, Ibaraki, Japan
| | - Koustav Roy
- International Institute for Integrative Sleep Medicine (WPI-IIIS), Tsukuba, Ibaraki, Japan
| | - Shuji Ioka
- International Institute for Integrative Sleep Medicine (WPI-IIIS), Tsukuba, Ibaraki, Japan
| | - Rintaro Otani
- International Institute for Integrative Sleep Medicine (WPI-IIIS), Tsukuba, Ibaraki, Japan
| | - Mao Amezawa
- International Institute for Integrative Sleep Medicine (WPI-IIIS), Tsukuba, Ibaraki, Japan
| | - Yukiko Ishikawa
- International Institute for Integrative Sleep Medicine (WPI-IIIS), Tsukuba, Ibaraki, Japan
| | - Yoan Cherasse
- International Institute for Integrative Sleep Medicine (WPI-IIIS), Tsukuba, Ibaraki, Japan
| | - Mahesh K. Kaushik
- International Institute for Integrative Sleep Medicine (WPI-IIIS), Tsukuba, Ibaraki, Japan
| | - Daniela Klewe-Nebenius
- International Institute for Integrative Sleep Medicine (WPI-IIIS), Tsukuba, Ibaraki, Japan
| | - Li Zhou
- International Institute for Integrative Sleep Medicine (WPI-IIIS), Tsukuba, Ibaraki, Japan
| | - Masashi Yanagisawa
- International Institute for Integrative Sleep Medicine (WPI-IIIS), Tsukuba, Ibaraki, Japan
| | - Yo Oishi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), Tsukuba, Ibaraki, Japan
- Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Tsuyoshi Saitoh
- International Institute for Integrative Sleep Medicine (WPI-IIIS), Tsukuba, Ibaraki, Japan
- Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- *Correspondence: Tsuyoshi Saitoh, ; Michael Lazarus,
| | - Michael Lazarus
- International Institute for Integrative Sleep Medicine (WPI-IIIS), Tsukuba, Ibaraki, Japan
- Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- *Correspondence: Tsuyoshi Saitoh, ; Michael Lazarus,
| |
Collapse
|
4
|
Song H, Chen C, Kelley B, Tomasevich A, Lee H, Dolle JP, Cheng J, Garcia B, Meaney DF, Smith DH. Traumatic brain injury recapitulates developmental changes of axons. Prog Neurobiol 2022; 217:102332. [PMID: 35870679 PMCID: PMC9454890 DOI: 10.1016/j.pneurobio.2022.102332] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 07/14/2022] [Accepted: 07/19/2022] [Indexed: 11/28/2022]
Abstract
During development, half of brain white matter axons are maintained for growth, while the remainder undergo developmental axon degeneration. After traumatic brain injury (TBI), injured axons also appear to follow pathways leading to either degeneration or repair. These observations raise the intriguing, but unexamined possibility that TBI recapitulates developmental axonal programs. Here, we examined axonal changes in the developing brain in young rats and after TBI in adult rat. Multiple shared changes in axonal microtubule (MT) through tubulin post-translational modifications and MT associated proteins (MAPs), tau and MAP6, were found in both development and TBI. Specifically, degenerating axons in both development and TBI underwent phosphorylation of tau and excessive tubulin tyrosination, suggesting MT instability and depolyermization. Conversely, nearby axons without degenerating morphologies, had increased MAP6 expression and maintenance of tubulin acetylation, suggesting enhanced MT stabilization, thereby supporting survival or repair. Quantitative proteomics revealed similar signaling pathways of axon degeneration and growth/repair, including protein clusters and networks. This comparison approach demonstrates how focused evaluation of developmental processes may provide insight into pathways initiated by TBI. In particular, the data suggest that TBI may reawaken dormant axonal programs that direct axons towards either degeneration or growth/repair, supporting further study in this area.
Collapse
Affiliation(s)
- Hailong Song
- Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Chen Chen
- Department of Computer Sciences, University of Missouri, Columbia, MO 65211, United States
| | - Brian Kelley
- Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Alexandra Tomasevich
- Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Hyoungjoo Lee
- Department of Biochemistry and Biophysics, Quantitative Proteomics Resource Core, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Jean-Pierre Dolle
- Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Jianlin Cheng
- Department of Computer Sciences, University of Missouri, Columbia, MO 65211, United States
| | - Benjamin Garcia
- Department of Biochemistry and Biophysics, Quantitative Proteomics Resource Core, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - David F Meaney
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Douglas H Smith
- Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania, Philadelphia, PA 19104, United States.
| |
Collapse
|
5
|
Lafanechère L. The microtubule cytoskeleton: An old validated target for novel therapeutic drugs. Front Pharmacol 2022; 13:969183. [PMID: 36188585 PMCID: PMC9521402 DOI: 10.3389/fphar.2022.969183] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 08/30/2022] [Indexed: 12/02/2022] Open
Abstract
Compounds targeting microtubules are widely used in cancer therapy with a proven efficacy. However, because they also target non-cancerous cells, their administration leads to numerous adverse effects. With the advancement of knowledge on the structure of tubulin, the regulation of microtubule dynamics and their deregulation in pathological processes, new therapeutic strategies are emerging, both for the treatment of cancer and for other diseases, such as neuronal or even heart diseases and parasite infections. In addition, a better understanding of the mechanism of action of well-known drugs such as colchicine or certain kinase inhibitors contributes to the development of these new therapeutic approaches. Nowadays, chemists and biologists are working jointly to select drugs which target the microtubule cytoskeleton and have improved properties. On the basis of a few examples this review attempts to depict the panorama of these recent advances.
Collapse
|
6
|
Zhao M, Chang Q, Yang H, Wang M, Liu Y, Lv N, Lei Q, Wei H. Epothilone D modulates autism-like behaviors in the BTBR mouse model of autism spectrum disorder. Neuroscience 2022; 490:171-181. [DOI: 10.1016/j.neuroscience.2022.02.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 02/17/2022] [Accepted: 02/21/2022] [Indexed: 10/19/2022]
|
7
|
Maly IV, Morales MJ, Pletnikov MV. Astrocyte Bioenergetics and Major Psychiatric Disorders. ADVANCES IN NEUROBIOLOGY 2021; 26:173-227. [PMID: 34888836 DOI: 10.1007/978-3-030-77375-5_9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Ongoing research continues to add new elements to the emerging picture of involvement of astrocyte energy metabolism in the pathophysiology of major psychiatric disorders, including schizophrenia, mood disorders, and addictions. This review outlines what is known about the energy metabolism in astrocytes, the most numerous cell type in the brain, and summarizes the recent work on how specific perturbations of astrocyte bioenergetics may contribute to the neuropsychiatric conditions. The role of astrocyte energy metabolism in mental health and disease is reviewed on the organism, organ, and cell level. Data arising from genomic, metabolomic, in vitro, and neurobehavioral studies is critically analyzed to suggest future directions in research and possible metabolism-focused therapeutic interventions.
Collapse
Affiliation(s)
- Ivan V Maly
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY, USA
| | - Michael J Morales
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY, USA
| | - Mikhail V Pletnikov
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY, USA.
| |
Collapse
|
8
|
Percelay S, Freret T, Turnbull N, Bouet V, Boulouard M. Combination of MAP6 deficit, maternal separation and MK801 in female mice: A 3-hit animal model of neurodevelopmental disorder with cognitive deficits. Behav Brain Res 2021; 413:113473. [PMID: 34280461 DOI: 10.1016/j.bbr.2021.113473] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 07/08/2021] [Accepted: 07/14/2021] [Indexed: 11/26/2022]
Abstract
Schizophrenia is a major psychiatric disease still lacking efficient treatment, particularly for cognitive deficits. To go further in research of new treatments that would encompass all the symptoms associated with this pathology, preclinical animal models need to be improved. To date, the aetiology of schizophrenia is unknown, but there is increasing evidence to highlight its multifactorial nature. We built a new neurodevelopmental mouse model gathering a triple factor combination (3-M): a genetic factor (partial deletion of MAP6 gene), an early stress (maternal separation) and a late pharmacological factor (MK801 administration, 0.05 mg/kg, i.p., daily for 5 days). The effects of each factor and of their combination were investigated on several behaviours including cognitive functions. While each individual factor induced slight deficits in one or another behavioural test, 3-M conditioning induces a wider phenotype with hyperlocomotion and cognitive deficits (working memory and social recognition). This study confirms the hypothesis that genetic, environmental and pharmacological factors, even if not deleterious by themselves, could act synergistically to induce a deleterious behavioural phenotype. It moreover encourages the use of such combined models to improve translational research on neurodevelopmental disorders.
Collapse
Affiliation(s)
- Solenn Percelay
- Normandie Université, UNICAEN, INSERM, COMETE, CYCERON, CHU Caen, 14000, Caen, France.
| | - Thomas Freret
- Normandie Université, UNICAEN, INSERM, COMETE, CYCERON, CHU Caen, 14000, Caen, France
| | - Nicole Turnbull
- Normandie Université, UNICAEN, INSERM, COMETE, CYCERON, CHU Caen, 14000, Caen, France
| | - Valentine Bouet
- Normandie Université, UNICAEN, INSERM, COMETE, CYCERON, CHU Caen, 14000, Caen, France
| | - Michel Boulouard
- Normandie Université, UNICAEN, INSERM, COMETE, CYCERON, CHU Caen, 14000, Caen, France
| |
Collapse
|
9
|
Pérez-Martín E, Muñoz-Castañeda R, Moutin MJ, Ávila-Zarza CA, Muñoz-Castañeda JM, Del Pilar C, Alonso JR, Andrieux A, Díaz D, Weruaga E. Oleoylethanolamide Delays the Dysfunction and Death of Purkinje Cells and Ameliorates Behavioral Defects in a Mouse Model of Cerebellar Neurodegeneration. Neurotherapeutics 2021; 18:1748-1767. [PMID: 33829414 PMCID: PMC8609004 DOI: 10.1007/s13311-021-01044-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/16/2021] [Indexed: 02/04/2023] Open
Abstract
Oleoylethanolamide (OEA) is an endocannabinoid that has been proposed to prevent neuronal damage and neuroinflammation. In this study, we evaluated the effects of OEA on the disruption of both cerebellar structure and physiology and on the behavior of Purkinje cell degeneration (PCD) mutant mice. These mice exhibit cerebellar degeneration, displaying microtubule alterations that trigger the selective loss of Purkinje cells and consequent behavioral impairments. The effects of different doses (1, 5, and 10 mg/kg, i.p.) and administration schedules (chronic and acute) of OEA were assessed at the behavioral, histological, cellular, and molecular levels to determine the most effective OEA treatment regimen. Our in vivo results demonstrated that OEA treatment prior to the onset of the preneurodegenerative phase prevented morphological alterations in Purkinje neurons (the somata and dendritic arbors) and decreased Purkinje cell death. This effect followed an inverted U-shaped time-response curve, with acute administration on postnatal day 12 (10 mg/kg, i.p.) being the most effective treatment regimen tested. Indeed, PCD mice that received this specific OEA treatment regimen showed improvements in motor, cognitive and social functions, which were impaired in these mice. Moreover, these in vivo neuroprotective effects of OEA were mediated by the PPARα receptor, as pretreatment with the PPARα antagonist GW6471 (2.5 mg/kg, i.p.) abolished them. Finally, our in vitro results suggested that the molecular effect of OEA was related to microtubule stability and structure since OEA administration normalized some alterations in microtubule features in PCD-like cells. These findings provide strong evidence supporting the use of OEA as a pharmacological agent to limit severe cerebellar neurodegenerative processes.
Collapse
Affiliation(s)
- Ester Pérez-Martín
- Laboratory of Neuronal Plasticity and Neurorepair, Institute for Neurosciences of Castile and Leon (INCyL), University of Salamanca, 37007, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), 37007, Salamanca, Spain
| | - Rodrigo Muñoz-Castañeda
- Laboratory of Neuronal Plasticity and Neurorepair, Institute for Neurosciences of Castile and Leon (INCyL), University of Salamanca, 37007, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), 37007, Salamanca, Spain
| | - Marie-Jo Moutin
- GIN, Univ. Grenoble Alpes, CNRS, CEA, Grenoble Institute Neurosciences, Inserm, U121638000, Grenoble, France
| | - Carmelo A Ávila-Zarza
- Institute of Biomedical Research of Salamanca (IBSAL), 37007, Salamanca, Spain
- Department of Statistics, University of Salamanca, 37007, Salamanca, Spain
| | - José M Muñoz-Castañeda
- Department of Theoretical, Atomic and Optical Physics, University of Valladolid, 47071, Valladolid, Spain
| | - Carlos Del Pilar
- Laboratory of Neuronal Plasticity and Neurorepair, Institute for Neurosciences of Castile and Leon (INCyL), University of Salamanca, 37007, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), 37007, Salamanca, Spain
| | - José R Alonso
- Laboratory of Neuronal Plasticity and Neurorepair, Institute for Neurosciences of Castile and Leon (INCyL), University of Salamanca, 37007, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), 37007, Salamanca, Spain
- Universidad de Tarapacá, Arica, Chile
| | - Annie Andrieux
- GIN, Univ. Grenoble Alpes, CNRS, CEA, Grenoble Institute Neurosciences, Inserm, U121638000, Grenoble, France
| | - David Díaz
- Laboratory of Neuronal Plasticity and Neurorepair, Institute for Neurosciences of Castile and Leon (INCyL), University of Salamanca, 37007, Salamanca, Spain.
- Institute of Biomedical Research of Salamanca (IBSAL), 37007, Salamanca, Spain.
| | - Eduardo Weruaga
- Laboratory of Neuronal Plasticity and Neurorepair, Institute for Neurosciences of Castile and Leon (INCyL), University of Salamanca, 37007, Salamanca, Spain.
- Institute of Biomedical Research of Salamanca (IBSAL), 37007, Salamanca, Spain.
| |
Collapse
|
10
|
Cuveillier C, Boulan B, Ravanello C, Denarier E, Deloulme JC, Gory-Fauré S, Delphin C, Bosc C, Arnal I, Andrieux A. Beyond Neuronal Microtubule Stabilization: MAP6 and CRMPS, Two Converging Stories. Front Mol Neurosci 2021; 14:665693. [PMID: 34025352 PMCID: PMC8131560 DOI: 10.3389/fnmol.2021.665693] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/09/2021] [Indexed: 12/21/2022] Open
Abstract
The development and function of the central nervous system rely on the microtubule (MT) and actin cytoskeletons and their respective effectors. Although the structural role of the cytoskeleton has long been acknowledged in neuronal morphology and activity, it was recently recognized to play the role of a signaling platform. Following this recognition, research into Microtubule Associated Proteins (MAPs) diversified. Indeed, historically, structural MAPs—including MAP1B, MAP2, Tau, and MAP6 (also known as STOP);—were identified and described as MT-binding and -stabilizing proteins. Extensive data obtained over the last 20 years indicated that these structural MAPs could also contribute to a variety of other molecular roles. Among multi-role MAPs, MAP6 provides a striking example illustrating the diverse molecular and cellular properties of MAPs and showing how their functional versatility contributes to the central nervous system. In this review, in addition to MAP6’s effect on microtubules, we describe its impact on the actin cytoskeleton, on neuroreceptor homeostasis, and its involvement in signaling pathways governing neuron development and maturation. We also discuss its roles in synaptic plasticity, brain connectivity, and cognitive abilities, as well as the potential relationships between the integrated brain functions of MAP6 and its molecular activities. In parallel, the Collapsin Response Mediator Proteins (CRMPs) are presented as examples of how other proteins, not initially identified as MAPs, fall into the broader MAP family. These proteins bind MTs as well as exhibiting molecular and cellular properties very similar to MAP6. Finally, we briefly summarize the multiple similarities between other classical structural MAPs and MAP6 or CRMPs.In summary, this review revisits the molecular properties and the cellular and neuronal roles of the classical MAPs, broadening our definition of what constitutes a MAP.
Collapse
|
11
|
Parato J, Bartolini F. The microtubule cytoskeleton at the synapse. Neurosci Lett 2021; 753:135850. [PMID: 33775740 DOI: 10.1016/j.neulet.2021.135850] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 03/19/2021] [Accepted: 03/22/2021] [Indexed: 12/13/2022]
Abstract
In neurons, microtubules (MTs) provide routes for transport throughout the cell and structural support for dendrites and axons. Both stable and dynamic MTs are necessary for normal neuronal functions. Research in the last two decades has demonstrated that MTs play additional roles in synaptic structure and function in both pre- and postsynaptic elements. Here, we review current knowledge of the functions that MTs perform in excitatory and inhibitory synapses, as well as in the neuromuscular junction and other specialized synapses, and discuss the implications that this knowledge may have in neurological disease.
Collapse
Affiliation(s)
- Julie Parato
- Columbia University Medical Center, Department of Pathology & Cell Biology, 630 West 168(th)Street, P&S 15-421, NY, NY, 10032, United States; SUNY Empire State College, Department of Natural Sciences, 177 Livingston Street, Brooklyn, NY, 11201, United States
| | - Francesca Bartolini
- Columbia University Medical Center, Department of Pathology & Cell Biology, 630 West 168(th)Street, P&S 15-421, NY, NY, 10032, United States.
| |
Collapse
|
12
|
Gory-Fauré S, Powell R, Jonckheere J, Lanté F, Denarier E, Peris L, Nguyen CH, Buisson A, Lafanechère L, Andrieux A. Pyr1-Mediated Pharmacological Inhibition of LIM Kinase Restores Synaptic Plasticity and Normal Behavior in a Mouse Model of Schizophrenia. Front Pharmacol 2021; 12:627995. [PMID: 33790791 PMCID: PMC8006432 DOI: 10.3389/fphar.2021.627995] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 02/04/2021] [Indexed: 12/14/2022] Open
Abstract
The search for effective treatments for neuropsychiatric disorders is ongoing, with progress being made as brain structure and neuronal function become clearer. The central roles played by microtubules (MT) and actin in synaptic transmission and plasticity suggest that the cytoskeleton and its modulators could be relevant targets for the development of new molecules to treat psychiatric diseases. In this context, LIM Kinase - which regulates both the actin and MT cytoskeleton especially in dendritic spines, the post-synaptic compartment of the synapse - might be a good target. In this study, we analyzed the consequences of blocking LIMK1 pharmacologically using Pyr1. We investigated synaptic plasticity defects and behavioral disorders in MAP6 KO mice, an animal model useful for the study of psychiatric disorders, particularly schizophrenia. Our results show that Pyr1 can modulate MT dynamics in neurons. In MAP6 KO mice, chronic LIMK inhibition by long-term treatment with Pyr1 can restore normal dendritic spine density and also improves long-term potentiation, both of which are altered in these mice. Pyr1 treatment improved synaptic plasticity, and also reduced social withdrawal and depressive/anxiety-like behavior in MAP6 KO mice. Overall, the results of this study validate the hypothesis that modulation of LIMK activity could represent a new therapeutic strategy for neuropsychiatric diseases.
Collapse
Affiliation(s)
- Sylvie Gory-Fauré
- Department of Molecular and Cellular Neurosciences, Grenoble Institute Neuroscience, Inserm U1216, Grenoble, France.,Université Grenoble Alpes, Grenoble, France
| | - Rebecca Powell
- Department of Molecular and Cellular Neurosciences, Grenoble Institute Neuroscience, Inserm U1216, Grenoble, France.,Université Grenoble Alpes, Grenoble, France
| | - Julie Jonckheere
- Department of Molecular and Cellular Neurosciences, Grenoble Institute Neuroscience, Inserm U1216, Grenoble, France.,Université Grenoble Alpes, Grenoble, France
| | - Fabien Lanté
- Department of Molecular and Cellular Neurosciences, Grenoble Institute Neuroscience, Inserm U1216, Grenoble, France.,Université Grenoble Alpes, Grenoble, France
| | - Eric Denarier
- Department of Molecular and Cellular Neurosciences, Grenoble Institute Neuroscience, Inserm U1216, Grenoble, France.,Université Grenoble Alpes, Grenoble, France.,Health Department, Interdisciplinary Research Institute of Grenoble, CEA, Grenoble, France
| | - Leticia Peris
- Department of Molecular and Cellular Neurosciences, Grenoble Institute Neuroscience, Inserm U1216, Grenoble, France.,Université Grenoble Alpes, Grenoble, France
| | - Chi Hung Nguyen
- Chimie et Modélisation pour la Biologie du Cancer, Institut Curie, PSL Research University, CNRS UMR9187, Inserm U1196, Orsay, France
| | - Alain Buisson
- Department of Molecular and Cellular Neurosciences, Grenoble Institute Neuroscience, Inserm U1216, Grenoble, France.,Université Grenoble Alpes, Grenoble, France
| | - Laurence Lafanechère
- Université Grenoble Alpes, Grenoble, France.,Microenvironment, Cell Plasticity and Signaling Department, Institute for Advanced Biosciences, CNRS UMR5309, Inserm U1209, Grenoble, France
| | - Annie Andrieux
- Department of Molecular and Cellular Neurosciences, Grenoble Institute Neuroscience, Inserm U1216, Grenoble, France.,Université Grenoble Alpes, Grenoble, France.,Health Department, Interdisciplinary Research Institute of Grenoble, CEA, Grenoble, France
| |
Collapse
|
13
|
Percelay S, Billard JM, Freret T, Andrieux A, Boulouard M, Bouet V. Functional Dysregulations in CA1 Hippocampal Networks of a 3-Hit Mouse Model of Schizophrenia. Int J Mol Sci 2021; 22:2644. [PMID: 33807989 PMCID: PMC7961987 DOI: 10.3390/ijms22052644] [Citation(s) in RCA: 7] [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: 01/19/2021] [Revised: 03/01/2021] [Accepted: 03/02/2021] [Indexed: 12/12/2022] Open
Abstract
For a better translation from treatment designs of schizophrenia to clinical efficiency, there is a crucial need to refine preclinical animal models. In order to consider the multifactorial nature of the disorder, a new mouse model associating three factors (genetic susceptibility-partial deletion of the MAP6 gene, early-life stress-maternal separation, and pharmacological treatment-chronic Δ-9-tetrahydrocannabinol during adolescence) has recently been described. While this model depicts a schizophrenia-like phenotype, the neurobiological correlates remain unknown. Synaptic transmission and functional plasticity of the CA1 hippocampal region of male and female 3-hit mice were therefore investigated using electrophysiological recordings on the hippocampus slice. While basal excitatory transmission remained unaffected, NMDA receptor (NMDAr)-mediated long-term potentiation (LTP) triggered by theta-burst (TBS) but not by high-frequency (HFS) stimulation was impaired in 3-hit mice. Isolated NMDAr activation was not affected or even increased in female 3-hit mice, revealing a sexual dimorphism. Considering that the regulation of LTP is more prone to inhibitory tone if triggered by TBS than by HFS, the weaker potentiation in 3-hit mice suggests a deficiency of intrinsic GABA regulatory mechanisms. Indeed, NMDAr activation was increased by GABAA receptor blockade in wild-type but not in 3-hit mice. This electrophysiological study highlights dysregulations of functional properties and plasticity in hippocampal networks of 3-hit mice, one of the mechanisms suspected to contribute to the pathophysiology of schizophrenia. It also shows differences between males and females, supporting the sexual dimorphism observed in the disorder. Combined with the previously reported study, the present data reinforce the face validity of the 3-hit model that will help to consider new therapeutic strategies for psychosis.
Collapse
Affiliation(s)
- Solenn Percelay
- UNICAEN, INSERM, COMETE, CYCERON, CHU Caen, Normandie Université, 14000 Caen, France; (J.-M.B.); (T.F.); (M.B.); (V.B.)
| | - Jean-Marie Billard
- UNICAEN, INSERM, COMETE, CYCERON, CHU Caen, Normandie Université, 14000 Caen, France; (J.-M.B.); (T.F.); (M.B.); (V.B.)
| | - Thomas Freret
- UNICAEN, INSERM, COMETE, CYCERON, CHU Caen, Normandie Université, 14000 Caen, France; (J.-M.B.); (T.F.); (M.B.); (V.B.)
| | - Annie Andrieux
- Inserm U1216, CEA, Grenoble Institut Neurosciences, Université Grenoble Alpes, 38000 Grenoble, France;
| | - Michel Boulouard
- UNICAEN, INSERM, COMETE, CYCERON, CHU Caen, Normandie Université, 14000 Caen, France; (J.-M.B.); (T.F.); (M.B.); (V.B.)
| | - Valentine Bouet
- UNICAEN, INSERM, COMETE, CYCERON, CHU Caen, Normandie Université, 14000 Caen, France; (J.-M.B.); (T.F.); (M.B.); (V.B.)
| |
Collapse
|
14
|
Bouet V, Percelay S, Leroux E, Diarra B, Léger M, Delcroix N, Andrieux A, Dollfus S, Freret T, Boulouard M. A new 3-hit mouse model of schizophrenia built on genetic, early and late factors. Schizophr Res 2021; 228:519-528. [PMID: 33298334 DOI: 10.1016/j.schres.2020.11.043] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 11/12/2020] [Accepted: 11/23/2020] [Indexed: 02/06/2023]
Abstract
Whether the etiology of schizophrenia remains unknown, its multifactorial aspect is conversely now well admitted. However, most preclinical models of the disease still rely on a mono-factorial construction and do not allow discover unequivocal treatments, particularly for negative and cognitive symptoms. The main interaction factors that have been implicated in schizophrenia are a genetic predisposition and unfavorable environmental factors. Here we propose a new animal model combining a genetic predisposition (1st hit: partial deletion of MAP-6 (microtubule-associated protein)) with an early postnatal stress (2nd hit: 24 h maternal separation at post-natal day 9), and a late cannabinoid exposure during adolescence (3rd hit: tetrahydrocannabinol THC from post-natal day 32 to 52; 8 mg/kg/day). The 2-hit mice displayed spatial memory deficits, decreased cortical thickness and fractional anisotropy of callosal fibers. The 3-hit mice were more severely affected as attested by supplementary deficits such a decrease in spontaneous activity, sociability-related behavior, working memory performances, an increase in anxiety-like behavior, a decrease in hippocampus volume together with impaired integrity of corpus callosum fibers (less axons, less myelin). Taken together, these results show that the new 3-hit model displays several landmarks mimicking negative and cognitive symptoms of schizophrenia, conferring a high relevance for research of new treatments. Moreover, this 3-hit model possesses a strong construct validity, which fits with gene x environment interactions hypothesis of schizophrenia. The 2-hit model, which associates maternal separation with THC exposure in wild-type mice gives a less severe phenotype, and could be useful for research on other forms of psychiatric diseases.
Collapse
Affiliation(s)
- Valentine Bouet
- Normandie Université, UNICAEN, INSERM, COMETE, CYCERON, CHYU CAEN, 14000 Caen, France.
| | - Solenn Percelay
- Normandie Université, UNICAEN, INSERM, COMETE, CYCERON, CHYU CAEN, 14000 Caen, France
| | - Elise Leroux
- Normandie Université, UNICAEN, EA 7466 ISTS, GIP Cyceron, 14000 Caen, France
| | - Boubacar Diarra
- Normandie Université, UNICAEN, INSERM, COMETE, CYCERON, CHYU CAEN, 14000 Caen, France
| | - Marianne Léger
- Normandie Université, UNICAEN, INSERM, COMETE, CYCERON, CHYU CAEN, 14000 Caen, France
| | - Nicolas Delcroix
- CNRS, UMS 3408, GIP CYCERON, Bd Henri Becquerel, BP5229, 14074 Caen cedex, France
| | - Annie Andrieux
- Univ. Grenoble Alpes, Inserm U1216, CEA, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Sonia Dollfus
- Normandie Université, UNICAEN, EA 7466 ISTS, GIP Cyceron, 14000 Caen, France; CHU de Caen, Service de Psychiatrie Adulte, 14000 Caen, France
| | - Thomas Freret
- Normandie Université, UNICAEN, INSERM, COMETE, CYCERON, CHYU CAEN, 14000 Caen, France
| | - Michel Boulouard
- Normandie Université, UNICAEN, INSERM, COMETE, CYCERON, CHYU CAEN, 14000 Caen, France
| |
Collapse
|
15
|
Wang L, Yan M, Wong CKC, Ge R, Wu X, Sun F, Cheng CY. Microtubule-associated proteins (MAPs) in microtubule cytoskeletal dynamics and spermatogenesis. Histol Histopathol 2020; 36:249-265. [PMID: 33174615 DOI: 10.14670/hh-18-279] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The microtubule (MT) cytoskeleton in Sertoli cells, a crucial cellular structure in the seminiferous epithelium of adult mammalian testes that supports spermatogenesis, was studied morphologically decades ago. However, its biology, in particular the involving regulatory biomolecules and the underlying mechanism(s) in modulating MT dynamics, are only beginning to be revealed in recent years. This lack of studies in delineating the biology of MT cytoskeletal dynamics undermines other studies in the field, in particular the plausible therapeutic treatment and management of male infertility and fertility since studies have shown that the MT cytoskeleton is one of the prime targets of toxicants. Interestingly, much of the information regarding the function of actin-, MT- and intermediate filament-based cytoskeletons come from studies using toxicant models including some genetic models. During the past several years, there have been some advances in studying the biology of MT cytoskeleton in the testis, and many of these studies were based on the use of pharmaceutical/toxicant models. In this review, we summarize the results of these findings, illustrating the importance of toxicant/pharmaceutical models in unravelling the biology of MT dynamics, in particular the role of microtubule-associated proteins (MAPs), a family of regulatory proteins that modulate MT dynamics but also actin- and intermediate filament-based cytoskeletons. We also provide a timely hypothetical model which can serve as a guide to design functional experiments to study how the MT cytoskeleton is regulated during spermatogenesis through the use of toxicants and/or pharmaceutical agents.
Collapse
Affiliation(s)
- Lingling Wang
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.,The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, NY, USA.,Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Ming Yan
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Chris K C Wong
- Department of Biology, Croucher Institute for Environmental Sciences, Hong Kong Baptist University, Kowloon, Hong Kong, China
| | - Renshan Ge
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaolong Wu
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, Jiangsu, China
| | - Fei Sun
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, Jiangsu, China
| | - C Yan Cheng
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, NY, USA.,The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
16
|
Kim K, Gibboney S, Razy-Krajka F, Lowe EK, Wang W, Stolfi A. Regulation of Neurogenesis by FGF Signaling and Neurogenin in the Invertebrate Chordate Ciona. Front Cell Dev Biol 2020; 8:477. [PMID: 32656209 PMCID: PMC7324659 DOI: 10.3389/fcell.2020.00477] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 05/21/2020] [Indexed: 12/22/2022] Open
Abstract
Neurogenesis is a complex sequence of cellular processes and behaviors driven by the coordinated expression of conserved effectors. The bipolar tail neurons (BTNs) of Ciona develop according to a highly dynamic, yet highly stereotyped developmental program and thus could serve as an accessible model system for neurogenesis, including underlying cell behaviors like neuronal delamination, migration, and polarized axon outgrowth. Here we investigate both the upstream events that shape BTN neurogenesis through spatiotemporal regulation of the conserved proneural factor Neurog, spatiotemporal, and the gene expression profile of differentiating BTNs downstream of Neurog activity. We show that, although early FGF signaling is required for Neurog expression and BTN specification, Fgf8/17/18 is expressed in tail tip cells at later stages and suppresses sustained Neurog expression in the anterior BTN (aBTN) lineage, such that only one cell (the one furthest from the source of Fgf8/17/18) maintains Neurog expression and becomes a neuron. Curiously, Fgf8/17/18 might not affect neurogenesis of the posterior BTNs (pBTNs), which are in direct contact with the Fgf8/17/18-expressing cells. Finally, to profile gene expression associated with BTN neurogenesis we performed RNAseq of isolated BTN lineage cells in which BTN neurogenesis was enhanced or suppressed by perturbing Neurog function. This allowed us to identify several candidate genes that might play conserved roles in neurogenesis and neuronal migration in other animals, including mammals.
Collapse
Affiliation(s)
- Kwantae Kim
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, United States
| | - Susanne Gibboney
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, United States
| | - Florian Razy-Krajka
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, United States
| | - Elijah K. Lowe
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, United States
| | - Wei Wang
- Department of Biology, New York University, New York, NY, United States
| | - Alberto Stolfi
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, United States
| |
Collapse
|
17
|
Clark JA, Chuckowree JA, Dyer MS, Dickson TC, Blizzard CA. Epothilone D alters normal growth, viability and microtubule dependent intracellular functions of cortical neurons in vitro. Sci Rep 2020; 10:918. [PMID: 31969604 PMCID: PMC6976590 DOI: 10.1038/s41598-020-57718-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 12/18/2019] [Indexed: 01/09/2023] Open
Abstract
Brain penetrant microtubule stabilising agents (MSAs) are being increasingly validated as potential therapeutic strategies for neurodegenerative diseases and traumatic injuries of the nervous system. MSAs are historically used to treat malignancies to great effect. However, this treatment strategy can also cause adverse off-target impacts, such as the generation of debilitating neuropathy and axonal loss. Understanding of the effects that individual MSAs have on neurons of the central nervous system is still incomplete. Previous research has revealed that aberrant microtubule stabilisation can perturb many neuronal functions, such as neuronal polarity, neurite outgrowth, microtubule dependant transport and overall neuronal viability. In the current study, we evaluate the dose dependant impact of epothilone D, a brain penetrant MSA, on both immature and relatively mature mouse cortical neurons in vitro. We show that epothilone D reduces the viability, growth and complexity of immature cortical neurons in a dose dependant manner. Furthermore, in relatively mature cortical neurons, we demonstrate that while cellularly lethal doses of epothilone D cause cellular demise, low sub lethal doses can also affect mitochondrial transport over time. Our results reveal an underappreciated mitochondrial disruption over a wide range of epothilone D doses and reiterate the importance of understanding the dosage, timing and intended outcome of MSAs, with particular emphasis on brain penetrant MSAs being considered to target neurons in disease and trauma.
Collapse
Affiliation(s)
- J A Clark
- Menzies Institute for Medical Research, University of Tasmania 17 Liverpool Street Hobart, Tasmania, 7000, Australia
| | - J A Chuckowree
- Menzies Institute for Medical Research, University of Tasmania 17 Liverpool Street Hobart, Tasmania, 7000, Australia
| | - M S Dyer
- Menzies Institute for Medical Research, University of Tasmania 17 Liverpool Street Hobart, Tasmania, 7000, Australia
| | - T C Dickson
- Menzies Institute for Medical Research, University of Tasmania 17 Liverpool Street Hobart, Tasmania, 7000, Australia
| | - C A Blizzard
- Menzies Institute for Medical Research, University of Tasmania 17 Liverpool Street Hobart, Tasmania, 7000, Australia.
| |
Collapse
|
18
|
Cognitive impairment in a rat model of neuropathic pain: role of hippocampal microtubule stability. Pain 2019; 159:1518-1528. [PMID: 29613911 DOI: 10.1097/j.pain.0000000000001233] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Clinical evidence indicates that cognitive impairment is a common comorbid condition of chronic pain. However, the cellular basis for chronic pain-mediated cognitive impairment remains unclear. We report here that rats exhibited memory deficits after spared nerve injury (SNI). We found that levels of stable microtubule (MT) were increased in the hippocampus of the rats with memory deficits. This increase in stable MT is marked by α-tubulin hyperacetylation. Paclitaxel, a pharmacological MT stabilizer, increased the level of stable MT in the hippocampus and induced learning and memory deficits in normal rats. Furthermore, paclitaxel reduced long-term potentiation in hippocampal slices and increased stable MT (evidenced by α-tubulin hyperacetylation) levels in hippocampal neuronal cells. Intracerebroventricular infusion of nocodazole, an MT destabilizer, ameliorated memory deficits in rats with SNI-induced nociceptive behavior. Expression of HDAC6, an α-tubulin deacetylase, was reduced in the hippocampus in rats with cognitive impairment. These findings indicate that peripheral nerve injury (eg, SNI) affects the MT dynamic equilibrium, which is critical to neuronal structure and synaptic plasticity.
Collapse
|
19
|
Chang Q, Yang H, Wang M, Wei H, Hu F. Role of Microtubule-Associated Protein in Autism Spectrum Disorder. Neurosci Bull 2018; 34:1119-1126. [PMID: 29936584 PMCID: PMC6246838 DOI: 10.1007/s12264-018-0246-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 04/19/2018] [Indexed: 12/14/2022] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by deficits in social interaction and communication, along with repetitive and restrictive patterns of behaviors or interests. Normal brain development is crucial to behavior and cognition in adulthood. Abnormal brain development, such as synaptic and myelin dysfunction, is involved in the pathogenesis of ASD. Microtubules and microtubule-associated proteins (MAPs) are important in regulating the processes of brain development, including neuron production and synaptic formation, as well as myelination. Increasing evidence suggests that the level of MAPs are changed in autistic patients and mouse models of ASD. Here, we discuss the roles of MAPs.
Collapse
Affiliation(s)
- Qiaoqiao Chang
- Department of Neurology, Shanxi Provincial People's Hospital, Affiliate of Shanxi Medical University, Taiyuan, 030012, China
| | - Hua Yang
- Department of Neurology, Shanxi Provincial People's Hospital, Affiliate of Shanxi Medical University, Taiyuan, 030012, China
| | - Min Wang
- Department of Neurology, Shanxi Provincial People's Hospital, Affiliate of Shanxi Medical University, Taiyuan, 030012, China
| | - Hongen Wei
- Department of Neurology, Shanxi Provincial People's Hospital, Affiliate of Shanxi Medical University, Taiyuan, 030012, China.
| | - Fengyun Hu
- Department of Neurology, Shanxi Provincial People's Hospital, Affiliate of Shanxi Medical University, Taiyuan, 030012, China.
| |
Collapse
|
20
|
Jonckheere J, Deloulme JC, Dall’Igna G, Chauliac N, Pelluet A, Nguon AS, Lentini C, Brocard J, Denarier E, Brugière S, Couté Y, Heinrich C, Porcher C, Holtzmann J, Andrieux A, Suaud-Chagny MF, Gory-Fauré S. Short- and long-term efficacy of electroconvulsive stimulation in animal models of depression: The essential role of neuronal survival. Brain Stimul 2018; 11:1336-1347. [DOI: 10.1016/j.brs.2018.08.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 07/12/2018] [Accepted: 08/03/2018] [Indexed: 12/28/2022] Open
|
21
|
Sébastien M, Giannesini B, Aubin P, Brocard J, Chivet M, Pietrangelo L, Boncompagni S, Bosc C, Brocard J, Rendu J, Gory-Fauré S, Andrieux A, Fourest-Lieuvin A, Fauré J, Marty I. Deletion of the microtubule-associated protein 6 (MAP6) results in skeletal muscle dysfunction. Skelet Muscle 2018; 8:30. [PMID: 30231928 PMCID: PMC6147105 DOI: 10.1186/s13395-018-0176-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 09/12/2018] [Indexed: 01/23/2023] Open
Abstract
Background The skeletal muscle fiber has a specific and precise intracellular organization which is at the basis of an efficient muscle contraction. Microtubules are long known to play a major role in the function and organization of many cells, but in skeletal muscle, the contribution of the microtubule cytoskeleton to the efficiency of contraction has only recently been studied. The microtubule network is dynamic and is regulated by many microtubule-associated proteins (MAPs). In the present study, the role of the MAP6 protein in skeletal muscle organization and function has been studied using the MAP6 knockout mouse line. Methods The presence of MAP6 transcripts and proteins was shown in mouse muscle homogenates and primary culture using RT-PCR and western blot. The in vivo evaluation of muscle force of MAP6 knockout (KO) mice was performed on anesthetized animals using electrostimulation coupled to mechanical measurement and multimodal magnetic resonance. The impact of MAP6 deletion on microtubule organization and intracellular structures was studied using immunofluorescent labeling and electron microscopy, and on calcium release for muscle contraction using Fluo-4 calcium imaging on cultured myotubes. Statistical analysis was performed using Student’s t test or the Mann-Whitney test. Results We demonstrate the presence of MAP6 transcripts and proteins in skeletal muscle. Deletion of MAP6 results in a large number of muscle modifications: muscle weakness associated with slight muscle atrophy, alterations of microtubule network and sarcoplasmic reticulum organization, and reduction in calcium release. Conclusion Altogether, our results demonstrate that MAP6 is involved in skeletal muscle function. Its deletion results in alterations in skeletal muscle contraction which contribute to the global deleterious phenotype of the MAP6 KO mice. As MAP6 KO mouse line is a model for schizophrenia, our work points to a possible muscle weakness associated to some forms of schizophrenia. Electronic supplementary material The online version of this article (10.1186/s13395-018-0176-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Muriel Sébastien
- INSERM 1216, Grenoble Institute of Neurosciences, F-38000, Grenoble, France.,University Grenoble Alpes, F-38000, Grenoble, France
| | | | - Perrine Aubin
- INSERM 1216, Grenoble Institute of Neurosciences, F-38000, Grenoble, France.,University Grenoble Alpes, F-38000, Grenoble, France
| | - Julie Brocard
- INSERM 1216, Grenoble Institute of Neurosciences, F-38000, Grenoble, France.,University Grenoble Alpes, F-38000, Grenoble, France
| | - Mathilde Chivet
- INSERM 1216, Grenoble Institute of Neurosciences, F-38000, Grenoble, France.,University Grenoble Alpes, F-38000, Grenoble, France
| | - Laura Pietrangelo
- CeSI-Met & DNICS, University G. d' Annunzio of Chieti, I-66100, Chieti, Italy
| | - Simona Boncompagni
- CeSI-Met & DNICS, University G. d' Annunzio of Chieti, I-66100, Chieti, Italy
| | - Christophe Bosc
- INSERM 1216, Grenoble Institute of Neurosciences, F-38000, Grenoble, France.,University Grenoble Alpes, F-38000, Grenoble, France
| | - Jacques Brocard
- INSERM 1216, Grenoble Institute of Neurosciences, F-38000, Grenoble, France.,University Grenoble Alpes, F-38000, Grenoble, France
| | - John Rendu
- INSERM 1216, Grenoble Institute of Neurosciences, F-38000, Grenoble, France.,University Grenoble Alpes, F-38000, Grenoble, France.,CHU Grenoble, Biochimie et Génétique Moléculaire, F-38000, Grenoble, France
| | - Sylvie Gory-Fauré
- INSERM 1216, Grenoble Institute of Neurosciences, F-38000, Grenoble, France.,University Grenoble Alpes, F-38000, Grenoble, France
| | - Annie Andrieux
- INSERM 1216, Grenoble Institute of Neurosciences, F-38000, Grenoble, France.,University Grenoble Alpes, F-38000, Grenoble, France.,CEA-Grenoble, BIG, F-38000, Grenoble, France
| | - Anne Fourest-Lieuvin
- INSERM 1216, Grenoble Institute of Neurosciences, F-38000, Grenoble, France.,University Grenoble Alpes, F-38000, Grenoble, France.,CEA-Grenoble, BIG, F-38000, Grenoble, France
| | - Julien Fauré
- INSERM 1216, Grenoble Institute of Neurosciences, F-38000, Grenoble, France.,University Grenoble Alpes, F-38000, Grenoble, France.,CHU Grenoble, Biochimie et Génétique Moléculaire, F-38000, Grenoble, France
| | - Isabelle Marty
- INSERM 1216, Grenoble Institute of Neurosciences, F-38000, Grenoble, France. .,University Grenoble Alpes, F-38000, Grenoble, France. .,GIN- Inserm U1216 - Bat EJ Safra, Chemin Fortuné Ferrini, 38700, La Tronche, France.
| |
Collapse
|
22
|
Dissociated features of social cognition altered in mouse models of schizophrenia: Focus on social dominance and acoustic communication. Neuropharmacology 2018; 159:107334. [PMID: 30236964 DOI: 10.1016/j.neuropharm.2018.09.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 09/05/2018] [Accepted: 09/08/2018] [Indexed: 02/07/2023]
Abstract
Social and communication impairments are common features of psychiatric disorders. Animal models of schizophrenia display various social deficits due to difference in tests, mouse strains and drugs. Moreover, communication deficits have not been studied. Our objectives were to assess and compare three major features of social cognition in different mouse models of schizophrenia: interest for a social stimulus, organization and acceptance of social contact, and acoustic communication to question whether mouse models for schizophrenia with social dysfunction also exhibit vocal communication defects. To achieve these aims we treated acutely C57BL/6J mice either with MK-801 or ketamine and tested WT and microtubule-associated protein 6 -MAP6- KO mice in two complementary social tasks: the 3-chamber test which measures social motivation and the social interaction task -SIT- which relies on prefrontal cortex activity and measures the ability to organize and respond to a real interaction, and which promotes ultrasonic vocalizations. Our results reveal that schizophrenia models have intact interest for a social stimulus in the 3-chamber test. However, thanks to principal component analyses of social interaction data, we demonstrate that social motivation and the ability to act socially rely on distinct mechanisms in revealing a decrease in dominance and communication in pharmacological schizophrenia models along with social withdraw, classically observed in schizophrenia, in MK-801 model. In this latter model, some social parameters can be significantly improved by aripiprazole, an atypical antipsychotic. Our social protocol, combined with fine-tuned analysis, is expected to provide an innovative framework for testing future treatments in preclinical models. This article is part of the Special Issue entitled 'The neuropharmacology of social behavior: from bench to bedside'.
Collapse
|
23
|
A key function for microtubule-associated-protein 6 in activity-dependent stabilisation of actin filaments in dendritic spines. Nat Commun 2018; 9:3775. [PMID: 30224655 PMCID: PMC6141585 DOI: 10.1038/s41467-018-05869-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 07/27/2018] [Indexed: 11/09/2022] Open
Abstract
Emerging evidence indicates that microtubule-associated proteins (MAPs) are implicated in synaptic function; in particular, mice deficient for MAP6 exhibit striking deficits in plasticity and cognition. How MAP6 connects to plasticity mechanisms is unclear. Here, we address the possible role of this protein in dendritic spines. We find that in MAP6-deficient cortical and hippocampal neurons, maintenance of mature spines is impaired, and can be restored by expressing a stretch of the MAP6 sequence called Mc modules. Mc modules directly bind actin filaments and mediate activity-dependent stabilisation of F-actin in dendritic spines, a key event of synaptic plasticity. In vitro, Mc modules enhance actin filament nucleation and promote the formation of stable, highly ordered filament bundles. Activity-induced phosphorylation of MAP6 likely controls its transfer to the spine cytoskeleton. These results provide a molecular explanation for the role of MAP6 in cognition, enlightening the connection between cytoskeletal dysfunction, synaptic impairment and neuropsychiatric illnesses. Microtubule-associated protein 6 (MAP6) is known to be important for synaptic plasticity and cognition, supposedly via interaction with microtubules. Here, the authors found that MAP6 is crucial for the stabilisation of enlarged synapses through its association with a different cytoskeletal element, actin.
Collapse
|
24
|
Varidaki A, Hong Y, Coffey ET. Repositioning Microtubule Stabilizing Drugs for Brain Disorders. Front Cell Neurosci 2018; 12:226. [PMID: 30135644 PMCID: PMC6092511 DOI: 10.3389/fncel.2018.00226] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 07/12/2018] [Indexed: 12/13/2022] Open
Abstract
Microtubule stabilizing agents are among the most clinically useful chemotherapeutic drugs. Mostly, they act to stabilize microtubules and inhibit cell division. While not without side effects, new generations of these compounds display improved pharmacokinetic properties and brain penetrance. Neurological disorders are intrinsically associated with microtubule defects, and efforts to reposition microtubule-targeting chemotherapeutic agents for treatment of neurodegenerative and psychiatric illnesses are underway. Here we catalog microtubule regulators that are associated with Alzheimer's and Parkinson's disease, amyotrophic lateral sclerosis, schizophrenia and mood disorders. We outline the classes of microtubule stabilizing agents used for cancer treatment, their brain penetrance properties and neuropathy side effects, and describe efforts to apply these agents for treatment of brain disorders. Finally, we summarize the current state of clinical trials for microtubule stabilizing agents under evaluation for central nervous system disorders.
Collapse
Affiliation(s)
- Artemis Varidaki
- Turku Centre for Biotechnology, Åbo Akademi University and University of Turku, Biocity, Tykistokatu, Turku, Finland
| | - Ye Hong
- Turku Centre for Biotechnology, Åbo Akademi University and University of Turku, Biocity, Tykistokatu, Turku, Finland
| | - Eleanor T Coffey
- Turku Centre for Biotechnology, Åbo Akademi University and University of Turku, Biocity, Tykistokatu, Turku, Finland
| |
Collapse
|
25
|
Corticosterone level and central dopaminergic activity involved in agile and exploratory behaviours in formosan wood mice (Apodemus semotus). J Comp Physiol A Neuroethol Sens Neural Behav Physiol 2018; 204:549-559. [PMID: 29589113 DOI: 10.1007/s00359-018-1259-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 03/21/2018] [Accepted: 03/22/2018] [Indexed: 01/19/2023]
Abstract
The native Formosan wood mouse (Apodemus semotus) is the dominant rodent in Taiwan. In their natural environment, Formosan wood mice exhibit high locomotor activity, including searching and exploratory behaviours, which is observed similarly in the laboratory environment. How the behavioural responses of Formosan wood mice exhibit in elevated plus maze and marble burying tests remains unclear. How corticosterone levels and central dopaminergic activities are related to the behaviours in these tests is also unclear. This study compared the behaviours of Formosan wood mice with that of C57BL/6J mice using the elevated plus maze and marble burying tests, and measured the corticosterone levels and central dopaminergic activities. Formosan wood mice showed greater locomotor and exploratory activity than the C57BL/6J mice. Similarly, the marble burying and rearing numbers were higher for Formosan wood mice. High locomotor and exploratory behaviours were strongly correlated with corticosterone levels after acute mild restraint stress in Formosan wood mice. The anxiolytic, diazepam, reduced the high exploratory activity, corticosterone levels and central dopaminergic activities. The high locomotor and exploratory behaviours of Formosan wood mice are related to the corticosterone levels and central dopaminergic activities. These data may explain Formosan wood mice dominance in the intermediate altitude of Taiwan.
Collapse
|
26
|
Pocivavsek A, Rowland LM. Basic Neuroscience Illuminates Causal Relationship Between Sleep and Memory: Translating to Schizophrenia. Schizophr Bull 2018; 44:7-14. [PMID: 29136236 PMCID: PMC5768044 DOI: 10.1093/schbul/sbx151] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Patients with schizophrenia are often plagued by sleep disturbances that can exacerbate the illness, including potentiating psychosis and cognitive impairments. Cognitive dysfunction is a core feature of schizophrenia with learning and memory being particularly impaired. Sleep disruptions often accompanying the illness and may be key mechanism that contribute to these core dysfunctions. In this special translational neuroscience feature, we highlight the role of sleep in mediating cognitive function, with a special focus on learning and memory. By defining dysfunctional sleep architecture and rhythms in schizophrenia, we focus on the disarray of mechanisms critical to learning and memory and postulate an association between sleep disturbances and cognitive impairments in the disorder. Lastly, we review preclinical models of schizophrenia and highlight exciting translational research that may lead to new therapeutic approaches to alleviating sleep disturbances and effectively improving cognitive function in schizophrenia.
Collapse
Affiliation(s)
- Ana Pocivavsek
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD
| | - Laura M Rowland
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD
| |
Collapse
|
27
|
Ramkumar A, Jong BY, Ori-McKenney KM. ReMAPping the microtubule landscape: How phosphorylation dictates the activities of microtubule-associated proteins. Dev Dyn 2017; 247:138-155. [PMID: 28980356 DOI: 10.1002/dvdy.24599] [Citation(s) in RCA: 136] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 09/11/2017] [Accepted: 09/19/2017] [Indexed: 12/12/2022] Open
Abstract
Classical microtubule-associated proteins (MAPs) were originally identified based on their co-purification with microtubules assembled from mammalian brain lysate. They have since been found to perform a range of functions involved in regulating the dynamics of the microtubule cytoskeleton. Most of these MAPs play integral roles in microtubule organization during neuronal development, microtubule remodeling during neuronal activity, and microtubule stabilization during neuronal maintenance. As a result, mutations in MAPs contribute to neurodevelopmental disorders, psychiatric conditions, and neurodegenerative diseases. MAPs are post-translationally regulated by phosphorylation depending on developmental time point and cellular context. Phosphorylation can affect the microtubule affinity, cellular localization, or overall function of a particular MAP and can thus have profound implications for neuronal health. Here we review MAP1, MAP2, MAP4, MAP6, MAP7, MAP9, tau, and DCX, and how each is regulated by phosphorylation in neuronal physiology and disease. Developmental Dynamics 247:138-155, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Amrita Ramkumar
- Department of Molecular and Cellular Biology, University of California, Davis, CA
| | - Brigette Y Jong
- Department of Molecular and Cellular Biology, University of California, Davis, CA
| | | |
Collapse
|
28
|
Gimenez U, Boulan B, Mauconduit F, Taurel F, Leclercq M, Denarier E, Brocard J, Gory-Fauré S, Andrieux A, Lahrech H, Deloulme JC. 3D imaging of the brain morphology and connectivity defects in a model of psychiatric disorders: MAP6-KO mice. Sci Rep 2017; 7:10308. [PMID: 28871106 PMCID: PMC5583184 DOI: 10.1038/s41598-017-10544-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 08/10/2017] [Indexed: 11/03/2022] Open
Abstract
In the central nervous system, microtubule-associated protein 6 (MAP6) is expressed at high levels and is crucial for cognitive abilities. The large spectrum of social and cognitive impairments observed in MAP6-KO mice are reminiscent of the symptoms observed in psychiatric diseases, such as schizophrenia, and respond positively to long-term treatment with antipsychotics. MAP6-KO mice have therefore been proposed to be a useful animal model for these diseases. Here, we explored the brain anatomy in MAP6-KO mice using high spatial resolution 3D MRI, including a volumetric T1w method to image brain structures, and Diffusion Tensor Imaging (DTI) for white matter fiber tractography. 3D DTI imaging of neuronal tracts was validated by comparing results to optical images of cleared brains. Changes to brain architecture included reduced volume of the cerebellum and the thalamus and altered size, integrity and spatial orientation of some neuronal tracks such as the anterior commissure, the mammillary tract, the corpus callosum, the corticospinal tract, the fasciculus retroflexus and the fornix. Our results provide information on the neuroanatomical defects behind the neurological phenotype displayed in the MAP6-KO mice model and especially highlight a severe damage of the corticospinal tract with defasciculation at the location of the pontine nuclei.
Collapse
Affiliation(s)
- Ulysse Gimenez
- INSERM, U1205, BrainTech Lab, F-38000, Grenoble, France.,Univ. Grenoble Alpes, F-38000, Grenoble, France
| | - Benoit Boulan
- Univ. Grenoble Alpes, F-38000, Grenoble, France.,INSERM, U1216, Grenoble Institut des Neurosciences, F-38000, Grenoble, France
| | - Franck Mauconduit
- INSERM, U1205, BrainTech Lab, F-38000, Grenoble, France.,Univ. Grenoble Alpes, F-38000, Grenoble, France
| | - Fanny Taurel
- INSERM, U1205, BrainTech Lab, F-38000, Grenoble, France.,Univ. Grenoble Alpes, F-38000, Grenoble, France
| | - Maxime Leclercq
- INSERM, U1205, BrainTech Lab, F-38000, Grenoble, France.,Univ. Grenoble Alpes, F-38000, Grenoble, France
| | - Eric Denarier
- Univ. Grenoble Alpes, F-38000, Grenoble, France.,INSERM, U1216, Grenoble Institut des Neurosciences, F-38000, Grenoble, France.,Commissariat à l'Energie Atomique, BIG-GPC, F-38000, Grenoble, France
| | - Jacques Brocard
- Univ. Grenoble Alpes, F-38000, Grenoble, France.,INSERM, U1216, Grenoble Institut des Neurosciences, F-38000, Grenoble, France
| | - Sylvie Gory-Fauré
- Univ. Grenoble Alpes, F-38000, Grenoble, France.,INSERM, U1216, Grenoble Institut des Neurosciences, F-38000, Grenoble, France
| | - Annie Andrieux
- Univ. Grenoble Alpes, F-38000, Grenoble, France.,INSERM, U1216, Grenoble Institut des Neurosciences, F-38000, Grenoble, France.,Commissariat à l'Energie Atomique, BIG-GPC, F-38000, Grenoble, France
| | - Hana Lahrech
- INSERM, U1205, BrainTech Lab, F-38000, Grenoble, France. .,Univ. Grenoble Alpes, F-38000, Grenoble, France.
| | - Jean Christophe Deloulme
- Univ. Grenoble Alpes, F-38000, Grenoble, France. .,INSERM, U1216, Grenoble Institut des Neurosciences, F-38000, Grenoble, France.
| |
Collapse
|
29
|
Omics analysis of mouse brain models of human diseases. Gene 2017; 600:90-100. [DOI: 10.1016/j.gene.2016.11.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 11/04/2016] [Accepted: 11/10/2016] [Indexed: 01/24/2023]
|
30
|
Voelzmann A, Hahn I, Pearce SP, Sánchez-Soriano N, Prokop A. A conceptual view at microtubule plus end dynamics in neuronal axons. Brain Res Bull 2016; 126:226-237. [PMID: 27530065 PMCID: PMC5090033 DOI: 10.1016/j.brainresbull.2016.08.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 08/08/2016] [Accepted: 08/11/2016] [Indexed: 12/02/2022]
Abstract
Axons are the cable-like protrusions of neurons which wire up the nervous system. Polar bundles of microtubules (MTs) constitute their structural backbones and are highways for life-sustaining transport between proximal cell bodies and distal synapses. Any morphogenetic changes of axons during development, plastic rearrangement, regeneration or degeneration depend on dynamic changes of these MT bundles. A key mechanism for implementing such changes is the coordinated polymerisation and depolymerisation at the plus ends of MTs within these bundles. To gain an understanding of how such regulation can be achieved at the cellular level, we provide here an integrated overview of the extensive knowledge we have about the molecular mechanisms regulating MT de/polymerisation. We first summarise insights gained from work in vitro, then describe the machinery which supplies the essential tubulin building blocks, the protein complexes associating with MT plus ends, and MT shaft-based mechanisms that influence plus end dynamics. We briefly summarise the contribution of MT plus end dynamics to important cellular functions in axons, and conclude by discussing the challenges and potential strategies of integrating the existing molecular knowledge into conceptual understanding at the level of axons.
Collapse
Affiliation(s)
- André Voelzmann
- The University of Manchester, Faculty of Biology, Medicine and Health, Michael Smith Building, Oxford Road, Manchester M13 9PT, UK
| | - Ines Hahn
- The University of Manchester, Faculty of Biology, Medicine and Health, Michael Smith Building, Oxford Road, Manchester M13 9PT, UK
| | - Simon P Pearce
- The University of Manchester, Faculty of Biology, Medicine and Health, Michael Smith Building, Oxford Road, Manchester M13 9PT, UK; The University of Manchester, School of Mathematics, Alan Turing Building, Oxford Road, Manchester M13 9PL, UK
| | - Natalia Sánchez-Soriano
- University of Liverpool, Institute of Translational Medicine, Department of Cellular and Molecular Physiology, Crown Street, Liverpool, L69 3BX, UK
| | - Andreas Prokop
- The University of Manchester, Faculty of Biology, Medicine and Health, Michael Smith Building, Oxford Road, Manchester M13 9PT, UK.
| |
Collapse
|
31
|
Baas PW, Rao AN, Matamoros AJ, Leo L. Stability properties of neuronal microtubules. Cytoskeleton (Hoboken) 2016; 73:442-60. [PMID: 26887570 PMCID: PMC5541393 DOI: 10.1002/cm.21286] [Citation(s) in RCA: 217] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 02/02/2016] [Accepted: 02/12/2016] [Indexed: 01/12/2023]
Abstract
Neurons are terminally differentiated cells that use their microtubule arrays not for cell division but rather as architectural elements required for the elaboration of elongated axons and dendrites. In addition to acting as compression-bearing struts that provide for the shape of the neuron, microtubules also act as directional railways for organelle transport. The stability properties of neuronal microtubules are commonly discussed in the biomedical literature as crucial to the development and maintenance of the nervous system, and have recently gained attention as central to the etiology of neurodegenerative diseases. Drugs that affect microtubule stability are currently under investigation as potential therapies for disease and injury of the nervous system. There is often a lack of consistency, however, in how the issue of microtubule stability is discussed in the literature, and this can affect the design and interpretation of experiments as well as potential therapeutic regimens. Neuronal microtubules are considered to be more stable than microtubules in dividing cells. On average, this is true, but in addition to an abundant stable microtubule fraction in neurons, there is also an abundant labile microtubule fraction. Both are functionally important. Individual microtubules consist of domains that differ in their stability properties, and these domains can also differ markedly in their composition as well as how they interact with various microtubule-related proteins in the neuron. Myriad proteins and pathways have been discussed as potential contributors to microtubule stability in neurons. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Peter W Baas
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA.
| | - Anand N Rao
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Andrew J Matamoros
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Lanfranco Leo
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
32
|
Profitt MF, Deurveilher S, Robertson GS, Rusak B, Semba K. Disruptions of Sleep/Wake Patterns in the Stable Tubule Only Polypeptide (STOP) Null Mouse Model of Schizophrenia. Schizophr Bull 2016; 42:1207-15. [PMID: 26940700 PMCID: PMC4988734 DOI: 10.1093/schbul/sbw017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Disruption of sleep/wake cycles is common in patients with schizophrenia and correlates with cognitive and affective abnormalities. Mice deficient in stable tubule only polypeptide (STOP) show cognitive, behavioral, and neurobiological deficits that resemble those seen in patients with schizophrenia, but little is known about their sleep phenotype. We characterized baseline sleep/wake patterns and recovery sleep following sleep deprivation in STOP null mice. Polysomnography was conducted in adult male STOP null and wild-type (WT) mice under a 12:12 hours light:dark cycle before, during, and after 6 hours of sleep deprivation during the light phase. At baseline, STOP null mice spent more time awake and less time in non-rapid eye movement sleep (NREMS) over a 24-hour period, with more frequent transitions between wake and NREMS, compared to WT mice, especially during the dark phase. The distributions of wake, NREMS and REMS across the light and the dark phases differed by genotype, and so did features of the electroencephalogram (EEG). Following sleep deprivation, both genotypes showed homeostatic increases in sleep duration, with no significant genotype differences in the initial compensatory increase in sleep intensity (EEG delta power). These results indicate that STOP null mice sleep less overall, and their sleep and wake periods are more fragmented than those of WT mice. These features in STOP null mice are consistent with the sleep patterns observed in patients with schizophrenia.
Collapse
Affiliation(s)
- Maxine F. Profitt
- Department of Medical Neuroscience, Dalhousie University, Halifax, NS, Canada
| | - Samuel Deurveilher
- Department of Medical Neuroscience, Dalhousie University, Halifax, NS, Canada
| | - George S. Robertson
- Department of Psychiatry, Dalhousie University, Halifax, NS, Canada;,Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
| | - Benjamin Rusak
- Department of Psychiatry, Dalhousie University, Halifax, NS, Canada;,Department of Pharmacology, Dalhousie University, Halifax, NS, Canada;,Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS, Canada
| | - Kazue Semba
- Department of Medical Neuroscience, Dalhousie University, Halifax, NS, Canada; Department of Psychiatry, Dalhousie University, Halifax, NS, Canada; Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
33
|
Benítez-Burraco A, Uriagereka J. The Immune Syntax Revisited: Opening New Windows on Language Evolution. Front Mol Neurosci 2016; 8:84. [PMID: 26793054 PMCID: PMC4707268 DOI: 10.3389/fnmol.2015.00084] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 12/14/2015] [Indexed: 01/29/2023] Open
Abstract
Recent research has added new dimensions to our understanding of classical evolution, according to which evolutionary novelties result from gene mutations inherited from parents to offspring. Language is surely one such novelty. Together with specific changes in our genome and epigenome, we suggest that two other (related) mechanisms may have contributed to the brain rewiring underlying human cognitive evolution and, specifically, the changes in brain connectivity that prompted the emergence of our species-specific linguistic abilities: the horizontal transfer of genetic material by viral and non-viral vectors and the brain/immune system crosstalk (more generally, the dialogue between the microbiota, the immune system, and the brain).
Collapse
Affiliation(s)
| | - Juan Uriagereka
- Department of Linguistics, University of Maryland College Park, MD, USA
| |
Collapse
|
34
|
Merenlender-Wagner A, Shemer Z, Touloumi O, Lagoudaki R, Giladi E, Andrieux A, Grigoriadis NC, Gozes I. New horizons in schizophrenia treatment: autophagy protection is coupled with behavioral improvements in a mouse model of schizophrenia. Autophagy 2015; 10:2324-32. [PMID: 25484074 DOI: 10.4161/15548627.2014.984274] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Autophagy plays a key role in the pathophysiology of schizophrenia as manifested by a 40% decrease in BECN1/Beclin 1 mRNA in postmortem hippocampal tissues relative to controls. This decrease was coupled with the deregulation of the essential ADNP (activity-dependent neuroprotector homeobox), a binding partner of MAP1LC3B/LC3B (microtubule-associated protein 1 light chain 3 β) another major constituent of autophagy. The drug candidate NAP (davunetide), a peptide fragment from ADNP, enhanced the ADNP-LC3B interaction. Parallel genetic studies have linked allelic variation in the gene encoding MAP6/STOP (microtubule-associated protein 6) to schizophrenia, along with altered MAP6/STOP protein expression in the schizophrenic brain and schizophrenic-like behaviors in Map6-deficient mice. In this study, for the first time, we reveal significant decreases in hippocampal Becn1 mRNA and reversal by NAP but not by the antipsychotic clozapine (CLZ) in Map6-deficient (Map6(+/-)) mice. Normalization of Becn1 expression by NAP was coupled with behavioral protection against hyperlocomotion and cognitive deficits measured in the object recognition test. CLZ reduced hyperlocomotion below control levels and did not significantly affect object recognition. The combination of CLZ and NAP resulted in normalized outcome behaviors. Phase II clinical studies have shown NAP-dependent augmentation of functional activities of daily living coupled with brain protection. The current studies provide a new mechanistic pathway and a novel avenue for drug development.
Collapse
Key Words
- ADNP, activity-dependent neuroprotector homeobox (human)
- Adnp, activity-dependent neuroprotective protein (mouse)
- Adnp2 (mouse), ADNP2 (human), ADNP homeobox 2
- Becn1 (mouse), BECN1 (human), Beclin 1, autophagy-related
- CLZ, clozapine
- HUGO gene nomenclature committee database)
- Hprt/Hprt1, hypoxanthine phosphoribosyl transferase
- MGI database)
- Map1lc3b (mouse), MAP1LC3B (human), microtubule-associated protein 1 light chain 3 β
- Map6 (mouse), MAP6 (human), microtubule-associated protein 6
- NAP (davunetide); object recognition
- activity-dependent neuroprotective protein (ADNP
- activity-dependent neuroprotector homeobox (ADNP
- hyperactivity; immunohistochemistry
- microtubule-associated protein 6 (MAP6)/stable tubule only polypeptide (STOP) deficiency
- real-time PCR
Collapse
Affiliation(s)
- Avia Merenlender-Wagner
- a The Adams Super Center for Brain Studies; The Lily and Avraham Gildor Chair for the Investigation of Growth Factors; The Elton Laboratory for Neuroendocrinology; Department of Human Molecular Genetics and Biochemistry; Sagol School of Neuroscience; Sackler Faculty of Medicine ; Tel Aviv University ; Tel Aviv ; Israel
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Deloulme JC, Gory-Fauré S, Mauconduit F, Chauvet S, Jonckheere J, Boulan B, Mire E, Xue J, Jany M, Maucler C, Deparis AA, Montigon O, Daoust A, Barbier EL, Bosc C, Deglon N, Brocard J, Denarier E, Le Brun I, Pernet-Gallay K, Vilgrain I, Robinson PJ, Lahrech H, Mann F, Andrieux A. Microtubule-associated protein 6 mediates neuronal connectivity through Semaphorin 3E-dependent signalling for axonal growth. Nat Commun 2015; 6:7246. [PMID: 26037503 PMCID: PMC4468860 DOI: 10.1038/ncomms8246] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 04/22/2015] [Indexed: 01/07/2023] Open
Abstract
Structural microtubule associated proteins (MAPs) stabilize microtubules, a property that was thought to be essential for development, maintenance and function of neuronal circuits. However, deletion of the structural MAPs in mice does not lead to major neurodevelopment defects. Here we demonstrate a role for MAP6 in brain wiring that is independent of microtubule binding. We find that MAP6 deletion disrupts brain connectivity and is associated with a lack of post-commissural fornix fibres. MAP6 contributes to fornix development by regulating axonal elongation induced by Semaphorin 3E. We show that MAP6 acts downstream of receptor activation through a mechanism that requires a proline-rich domain distinct from its microtubule-stabilizing domains. We also show that MAP6 directly binds to SH3 domain proteins known to be involved in neurite extension and semaphorin function. We conclude that MAP6 is critical to interface guidance molecules with intracellular signalling effectors during the development of cerebral axon tracts. Loss of the structural microtubule-associated protein 6 (MAP6) leads to neuronal differentiation defects that are independent of MAP6's microtubule-binding properties. Here the authors establish a functional link between MAP6 and Semaphorin 3E signalling for proper formation of the fornix of the brain.
Collapse
Affiliation(s)
- Jean-Christophe Deloulme
- 1] INSERM, U836, F-38000 Grenoble, France [2] Univ. Grenoble Alpes, Grenoble Institut Neurosciences, F-38000 Grenoble, France
| | - Sylvie Gory-Fauré
- 1] INSERM, U836, F-38000 Grenoble, France [2] Univ. Grenoble Alpes, Grenoble Institut Neurosciences, F-38000 Grenoble, France
| | - Franck Mauconduit
- 1] INSERM, U836, F-38000 Grenoble, France [2] Univ. Grenoble Alpes, Grenoble Institut Neurosciences, F-38000 Grenoble, France
| | - Sophie Chauvet
- Aix-Marseille Université, CNRS, IBDM UMR 7288, 13288 Marseille, France
| | - Julie Jonckheere
- 1] INSERM, U836, F-38000 Grenoble, France [2] Univ. Grenoble Alpes, Grenoble Institut Neurosciences, F-38000 Grenoble, France
| | - Benoit Boulan
- 1] INSERM, U836, F-38000 Grenoble, France [2] Univ. Grenoble Alpes, Grenoble Institut Neurosciences, F-38000 Grenoble, France
| | - Erik Mire
- Aix-Marseille Université, CNRS, IBDM UMR 7288, 13288 Marseille, France
| | - Jing Xue
- Cell Signalling Unit, Children's Medical Research Institute, University of Sydney, Wentworthville, New South Wales 2145, Australia
| | - Marion Jany
- 1] INSERM, U836, F-38000 Grenoble, France [2] Univ. Grenoble Alpes, Grenoble Institut Neurosciences, F-38000 Grenoble, France
| | - Caroline Maucler
- 1] INSERM, U836, F-38000 Grenoble, France [2] Univ. Grenoble Alpes, Grenoble Institut Neurosciences, F-38000 Grenoble, France
| | - Agathe A Deparis
- 1] INSERM, U836, F-38000 Grenoble, France [2] Univ. Grenoble Alpes, Grenoble Institut Neurosciences, F-38000 Grenoble, France
| | - Olivier Montigon
- 1] INSERM, U836, F-38000 Grenoble, France [2] Univ. Grenoble Alpes, Grenoble Institut Neurosciences, F-38000 Grenoble, France [3] Centre Hospitalier Universitaire de Grenoble, IRMaGe, 38043 Grenoble, France [4] CNRS, UMS 3552, 38042 Grenoble, France
| | - Alexia Daoust
- 1] INSERM, U836, F-38000 Grenoble, France [2] Univ. Grenoble Alpes, Grenoble Institut Neurosciences, F-38000 Grenoble, France
| | - Emmanuel L Barbier
- 1] INSERM, U836, F-38000 Grenoble, France [2] Univ. Grenoble Alpes, Grenoble Institut Neurosciences, F-38000 Grenoble, France
| | - Christophe Bosc
- 1] INSERM, U836, F-38000 Grenoble, France [2] Univ. Grenoble Alpes, Grenoble Institut Neurosciences, F-38000 Grenoble, France
| | - Nicole Deglon
- 1] Lausanne University Hospital (CHUV), Department of Clinical Neurosciences (DNC), Laboratory of Cellular and Molecular Neurotherapies (LCMN), 1011 Lausanne, Switzerland [2] Lausanne University Hospital (CHUV), Neuroscience Research Center (CRN), 1011 Lausanne, Switzerland
| | - Jacques Brocard
- 1] INSERM, U836, F-38000 Grenoble, France [2] Univ. Grenoble Alpes, Grenoble Institut Neurosciences, F-38000 Grenoble, France
| | - Eric Denarier
- 1] INSERM, U836, F-38000 Grenoble, France [2] Univ. Grenoble Alpes, Grenoble Institut Neurosciences, F-38000 Grenoble, France [3] CEA, iRTSV, F-38000 Grenoble, France
| | - Isabelle Le Brun
- 1] Univ. Grenoble Alpes, Grenoble Institut Neurosciences, F-38000 Grenoble, France [2] INSERM, U1036, 38054 Grenoble, France [3] CEA, iRTSV, F-38000 Grenoble, France
| | - Karin Pernet-Gallay
- 1] INSERM, U836, F-38000 Grenoble, France [2] Univ. Grenoble Alpes, Grenoble Institut Neurosciences, F-38000 Grenoble, France
| | - Isabelle Vilgrain
- 1] Univ. Grenoble Alpes, Grenoble Institut Neurosciences, F-38000 Grenoble, France [2] INSERM, U1036, 38054 Grenoble, France [3] INSERM, U1036, 38054 Grenoble, France
| | - Phillip J Robinson
- Cell Signalling Unit, Children's Medical Research Institute, University of Sydney, Wentworthville, New South Wales 2145, Australia
| | - Hana Lahrech
- 1] INSERM, U836, F-38000 Grenoble, France [2] Univ. Grenoble Alpes, Grenoble Institut Neurosciences, F-38000 Grenoble, France [3] CEA, LETI, CLINATEC, MINATEC Campus, F-38054 Grenoble, France
| | - Fanny Mann
- Aix-Marseille Université, CNRS, IBDM UMR 7288, 13288 Marseille, France
| | - Annie Andrieux
- 1] INSERM, U836, F-38000 Grenoble, France [2] Univ. Grenoble Alpes, Grenoble Institut Neurosciences, F-38000 Grenoble, France [3] CEA, iRTSV, F-38000 Grenoble, France
| |
Collapse
|
36
|
Venkatasubramanian G. Understanding schizophrenia as a disorder of consciousness: biological correlates and translational implications from quantum theory perspectives. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE : THE OFFICIAL SCIENTIFIC JOURNAL OF THE KOREAN COLLEGE OF NEUROPSYCHOPHARMACOLOGY 2015; 13:36-47. [PMID: 25912536 PMCID: PMC4423156 DOI: 10.9758/cpn.2015.13.1.36] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Revised: 12/07/2014] [Accepted: 12/08/2014] [Indexed: 11/18/2022]
Abstract
From neurophenomenological perspectives, schizophrenia has been conceptualized as "a disorder with heterogeneous manifestations that can be integrally understood to involve fundamental perturbations in consciousness". While these theoretical constructs based on consciousness facilitate understanding the 'gestalt' of schizophrenia, systematic research to unravel translational implications of these models is warranted. To address this, one needs to begin with exploration of plausible biological underpinnings of "perturbed consciousness" in schizophrenia. In this context, an attractive proposition to understand the biology of consciousness is "the orchestrated object reduction (Orch-OR) theory" which invokes quantum processes in the microtubules of neurons. The Orch-OR model is particularly important for understanding schizophrenia especially due to the shared 'scaffold' of microtubules. The initial sections of this review focus on the compelling evidence to support the view that "schizophrenia is a disorder of consciousness" through critical summary of the studies that have demonstrated self-abnormalities, aberrant time perception as well as dysfunctional intentional binding in this disorder. Subsequently, these findings are linked with 'Orch-OR theory' through the research evidence for aberrant neural oscillations as well as microtubule abnormalities observed in schizophrenia. Further sections emphasize the applicability and translational implications of Orch-OR theory in the context of schizophrenia and elucidate the relevance of quantum biology to understand the origins of this puzzling disorder as "fundamental disturbances in consciousness".
Collapse
Affiliation(s)
- Ganesan Venkatasubramanian
- The Schizophrenia Clinic, Department of Psychiatry and Translational Psychiatry Laboratory, Neurobiology Research Centre, National Institute of Mental Health and Neurosciences, Bangalore,
India
| |
Collapse
|
37
|
Dacheux D, Roger B, Bosc C, Landrein N, Roche E, Chansel L, Trian T, Andrieux A, Papaxanthos-Roche A, Marthan R, Robinson DR, Bonhivers M. Human FAM154A (SAXO1) is a microtubule-stabilizing protein specific to cilia and related structures. J Cell Sci 2015; 128:1294-307. [PMID: 25673876 DOI: 10.1242/jcs.155143] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Cilia and flagella are microtubule-based organelles present at the surface of most cells, ranging from protozoa to vertebrates, in which these structures are implicated in processes from morphogenesis to cell motility. In vertebrate neurons, microtubule-associated MAP6 proteins stabilize cold-resistant microtubules through their Mn and Mc modules, and play a role in synaptic plasticity. Although centrioles, cilia and flagella have cold-stable microtubules, MAP6 proteins have not been identified in these organelles, suggesting that additional proteins support this role in these structures. Here, we characterize human FAM154A (hereafter referred to as hSAXO1) as the first human member of a widely conserved family of MAP6-related proteins specific to centrioles and cilium microtubules. Our data demonstrate that hSAXO1 binds specifically to centriole and cilium microtubules. We identify, in vivo and in vitro, hSAXO1 Mn modules as responsible for microtubule binding and stabilization as well as being necessary for ciliary localization. Finally, overexpression and knockdown studies show that hSAXO1 modulates axoneme length. Taken together, our findings suggest a fine regulation of hSAXO1 localization and important roles in cilium biogenesis and function.
Collapse
Affiliation(s)
- Denis Dacheux
- University Bordeaux, Microbiologie Fondamentale et Pathogénicité, UMR 5234, F-33000 Bordeaux, France CNRS, Microbiologie Fondamentale et Pathogénicité, UMR 5234, F-33000 Bordeaux, France Institut Polytechnique de Bordeaux, Microbiologie Fondamentale et Pathogénicité, UMR 5234, F-33000 Bordeaux, France
| | - Benoit Roger
- University Bordeaux, Microbiologie Fondamentale et Pathogénicité, UMR 5234, F-33000 Bordeaux, France CNRS, Microbiologie Fondamentale et Pathogénicité, UMR 5234, F-33000 Bordeaux, France
| | - Christophe Bosc
- INSERM, Centre de Recherche U836, F-38000, Grenoble, France University Grenoble Alpes, Grenoble Institut des Neurosciences, F-38000, Grenoble, France
| | - Nicolas Landrein
- University Bordeaux, Microbiologie Fondamentale et Pathogénicité, UMR 5234, F-33000 Bordeaux, France CNRS, Microbiologie Fondamentale et Pathogénicité, UMR 5234, F-33000 Bordeaux, France
| | - Emmanuel Roche
- University Bordeaux, Microbiologie Fondamentale et Pathogénicité, UMR 5234, F-33000 Bordeaux, France CNRS, Microbiologie Fondamentale et Pathogénicité, UMR 5234, F-33000 Bordeaux, France
| | - Lucie Chansel
- CHU de Bordeaux, Centre Aliénor d'Aquitaine, Laboratoire de Biologie de la Reproduction, F-33000 Bordeaux, France
| | - Thomas Trian
- University Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, F-33000 Bordeaux, France INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, F-33000 Bordeaux, France
| | - Annie Andrieux
- INSERM, Centre de Recherche U836, F-38000, Grenoble, France University Grenoble Alpes, Grenoble Institut des Neurosciences, F-38000, Grenoble, France CEA, Institut de Recherches en Technologies et Sciences pour le Vivant, GPC, F-38000 Grenoble, France
| | - Aline Papaxanthos-Roche
- CHU de Bordeaux, Centre Aliénor d'Aquitaine, Laboratoire de Biologie de la Reproduction, F-33000 Bordeaux, France
| | - Roger Marthan
- University Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, F-33000 Bordeaux, France INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, F-33000 Bordeaux, France
| | - Derrick R Robinson
- University Bordeaux, Microbiologie Fondamentale et Pathogénicité, UMR 5234, F-33000 Bordeaux, France CNRS, Microbiologie Fondamentale et Pathogénicité, UMR 5234, F-33000 Bordeaux, France
| | - Mélanie Bonhivers
- University Bordeaux, Microbiologie Fondamentale et Pathogénicité, UMR 5234, F-33000 Bordeaux, France CNRS, Microbiologie Fondamentale et Pathogénicité, UMR 5234, F-33000 Bordeaux, France
| |
Collapse
|
38
|
Daoust A, Bohic S, Saoudi Y, Debacker C, Gory-Fauré S, Andrieux A, Barbier EL, Deloulme JC. Neuronal transport defects of the MAP6 KO mouse - a model of schizophrenia - and alleviation by Epothilone D treatment, as observed using MEMRI. Neuroimage 2014; 96:133-42. [PMID: 24704457 DOI: 10.1016/j.neuroimage.2014.03.071] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Revised: 03/18/2014] [Accepted: 03/25/2014] [Indexed: 11/28/2022] Open
Abstract
The MAP6 (microtubule-associated protein 6) KO mouse is a microtubule-deficient model of schizophrenia that exhibits severe behavioral disorders that are associated with synaptic plasticity anomalies. These defects are alleviated not only by neuroleptics, which are the gold standard molecules for the treatment of schizophrenia, but also by Epothilone D (Epo D), which is a microtubule-stabilizing molecule. To compare the neuronal transport between MAP6 KO and wild-type mice and to measure the effect of Epo D treatment on neuronal transport in KO mice, MnCl2 was injected in the primary somatosensory cortex. Then, using manganese-enhanced magnetic resonance imaging (MEMRI), we followed the propagation of Mn(2+) through axonal tracts and brain regions that are connected to the somatosensory cortex. In MAP6 KO mice, the measure of the MRI relative signal intensity over 24h revealed that the Mn(2+) transport rate was affected with a stronger effect on long-range and polysynaptic connections than in short-range and monosynaptic tracts. The chronic treatment of MAP6 KO mice with Epo D strongly increased Mn(2+) propagation within both mono- and polysynaptic connections. Our results clearly indicate an in vivo deficit in neuronal Mn(2+) transport in KO MAP6 mice, which might be due to both axonal transport defects and synaptic transmission impairments. Epo D treatment alleviated the axonal transport defects, and this improvement most likely contributes to the positive effect of Epo D on behavioral defects in KO MAP6 mice.
Collapse
Affiliation(s)
- Alexia Daoust
- Inserm U836, Equipe NeuroImagerie Fonctionnelle et Perfusion Cérébrale, BP170, Grenoble 38042, France; Université Joseph Fourier, Grenoble Institut des Neurosciences, Grenoble, France
| | - Sylvain Bohic
- Inserm U836, Equipe NeuroImagerie Fonctionnelle et Perfusion Cérébrale, BP170, Grenoble 38042, France; Université Joseph Fourier, Grenoble Institut des Neurosciences, Grenoble, France; European Synchrotron Radiation Facility (ESRF), Grenoble, France
| | - Yasmina Saoudi
- Université Joseph Fourier, Grenoble Institut des Neurosciences, Grenoble, France; Inserm U836, Equipe Physiopathologie du Cytosquelette, Grenoble, France; Commissariat à l'Energie Atomique et aux Energies Alternatives, iRTSV-GPC, Grenoble, France
| | - Clément Debacker
- Inserm U836, Equipe NeuroImagerie Fonctionnelle et Perfusion Cérébrale, BP170, Grenoble 38042, France; Université Joseph Fourier, Grenoble Institut des Neurosciences, Grenoble, France; Bruker Biospin MRI, Ettlingen, Germany
| | - Sylvie Gory-Fauré
- Université Joseph Fourier, Grenoble Institut des Neurosciences, Grenoble, France; Inserm U836, Equipe Physiopathologie du Cytosquelette, Grenoble, France; Commissariat à l'Energie Atomique et aux Energies Alternatives, iRTSV-GPC, Grenoble, France
| | - Annie Andrieux
- Université Joseph Fourier, Grenoble Institut des Neurosciences, Grenoble, France; Inserm U836, Equipe Physiopathologie du Cytosquelette, Grenoble, France; Commissariat à l'Energie Atomique et aux Energies Alternatives, iRTSV-GPC, Grenoble, France
| | - Emmanuel Luc Barbier
- Inserm U836, Equipe NeuroImagerie Fonctionnelle et Perfusion Cérébrale, BP170, Grenoble 38042, France; Université Joseph Fourier, Grenoble Institut des Neurosciences, Grenoble, France.
| | - Jean-Christophe Deloulme
- Université Joseph Fourier, Grenoble Institut des Neurosciences, Grenoble, France; Inserm U836, Equipe Physiopathologie du Cytosquelette, Grenoble, France; Commissariat à l'Energie Atomique et aux Energies Alternatives, iRTSV-GPC, Grenoble, France.
| |
Collapse
|
39
|
Hong I, Kang T, Yun KN, Yoo Y, Park S, Kim J, An B, Song S, Lee S, Kim J, Song B, Kwon KH, Kim JY, Park YM, Choi S. Quantitative proteomics of auditory fear conditioning. Biochem Biophys Res Commun 2013; 434:87-94. [PMID: 23542466 DOI: 10.1016/j.bbrc.2013.03.060] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Accepted: 03/20/2013] [Indexed: 10/27/2022]
Abstract
Auditory fear conditioning is a well-characterized rodent learning model where a neutral auditory cue is paired with an aversive outcome to induce associative fear memory. The storage of long-term auditory fear memory requires long-term potentiation (LTP) in the lateral amygdala and de novo protein synthesis. Although many studies focused on individual proteins have shown their contribution to LTP and fear conditioning, non-biased genome-wide studies have only recently been possible with microarrays, which nevertheless fall short of measuring changes at the level of proteins. Here we employed quantitative proteomics to examine the expression of hundreds of proteins in the lateral amygdala in response to auditory fear conditioning. We found that various proteins previously implicated in LTP, learning and axon/dendrite growth were regulated by fear conditioning. A substantial number of proteins that were regulated by fear conditioning have not yet been studied specifically in learning or synaptic plasticity.
Collapse
Affiliation(s)
- Ingie Hong
- School of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 151-742, South Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|