1
|
Ma D, Gu C. Discovering functional interactions among schizophrenia-risk genes by combining behavioral genetics with cell biology. Neurosci Biobehav Rev 2024; 167:105897. [PMID: 39278606 DOI: 10.1016/j.neubiorev.2024.105897] [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: 06/15/2024] [Revised: 08/26/2024] [Accepted: 09/13/2024] [Indexed: 09/18/2024]
Abstract
Despite much progress in identifying risk genes for polygenic brain disorders, their core pathogenic mechanisms remain poorly understood. In particular, functions of many proteins encoded by schizophrenia risk genes appear diverse and unrelated, complicating the efforts to establish the causal relationship between genes and behavior. Using various mouse lines, recent studies indicate that alterations of parvalbumin-positive (PV+) GABAergic interneurons can lead to schizophrenia-like behavior. PV+ interneurons display fast spiking and contribute to excitation-inhibition balance and network oscillations via feedback and feedforward inhibition. Here, we first summarize different lines of genetically modified mice that display motor, cognitive, emotional, and social impairments used to model schizophrenia and related mental disorders. We highlight ten genes, encoding either a nuclear, cytosolic, or membrane protein. Next, we discuss their functional relationship in regulating fast spiking and other aspects of PV+ interneurons and in the context of other domains of schizophrenia. Future investigations combining behavioral genetics and cell biology should elucidate functional relationships among risk genes to identify the core pathogenic mechanisms underlying polygenic brain disorders.
Collapse
Affiliation(s)
- Di Ma
- Ohio State Biochemistry Graduate Program, The Ohio State University, Columbus, OH 43210, USA
| | - Chen Gu
- Ohio State Biochemistry Graduate Program, The Ohio State University, Columbus, OH 43210, USA; Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
2
|
Wang S, Xing X, Ma J, Zheng S, Song Q, Zhang P. Deacylases-structure, function, and relationship to diseases. FEBS Lett 2024; 598:959-977. [PMID: 38644468 DOI: 10.1002/1873-3468.14885] [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: 11/19/2023] [Revised: 01/28/2024] [Accepted: 03/20/2024] [Indexed: 04/23/2024]
Abstract
Reversible S-acylation plays a pivotal role in various biological processes, modulating protein functions such as subcellular localization, protein stability/activity, and protein-protein interactions. These modifications are mediated by acyltransferases and deacylases, among which the most abundant modification is S-palmitoylation. Growing evidence has shown that this rivalrous pair of modifications, occurring in a reversible cycle, is essential for various biological functions. Aberrations in this process have been associated with various diseases, including cancer, neurological disorders, and immune diseases. This underscores the importance of studying enzymes involved in acylation and deacylation to gain further insights into disease pathogenesis and provide novel strategies for disease treatment. In this Review, we summarize our current understanding of the structure and physiological function of deacylases, highlighting their pivotal roles in pathology. Our aim is to provide insights for further clinical applications.
Collapse
Affiliation(s)
- Shuxian Wang
- Cancer Center, Renmin Hospital of Wuhan University, China
| | - Xiaoke Xing
- Cancer Center, Renmin Hospital of Wuhan University, China
| | - Jialin Ma
- Cancer Center, Renmin Hospital of Wuhan University, China
| | - Sihao Zheng
- Cancer Center, Renmin Hospital of Wuhan University, China
| | - Qibin Song
- Cancer Center, Renmin Hospital of Wuhan University, China
| | - Pingfeng Zhang
- Cancer Center, Renmin Hospital of Wuhan University, China
| |
Collapse
|
3
|
Ma D, Sun C, Manne R, Guo T, Bosc C, Barry J, Magliery T, Andrieux A, Li H, Gu C. A cytoskeleton-membrane interaction conserved in fast-spiking neurons controls movement, emotion, and memory. Mol Psychiatry 2023; 28:3994-4010. [PMID: 37833406 PMCID: PMC10905646 DOI: 10.1038/s41380-023-02286-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 09/20/2023] [Accepted: 09/29/2023] [Indexed: 10/15/2023]
Abstract
The pathogenesis of schizophrenia is believed to involve combined dysfunctions of many proteins including microtubule-associated protein 6 (MAP6) and Kv3.1 voltage-gated K+ (Kv) channel, but their relationship and functions in behavioral regulation are often not known. Here we report that MAP6 stabilizes Kv3.1 channels in parvalbumin-positive (PV+ ) fast-spiking GABAergic interneurons, regulating behavior. MAP6-/- and Kv3.1-/- mice display similar hyperactivity and avoidance reduction. Their proteins colocalize in PV+ interneurons and MAP6 deletion markedly reduces Kv3.1 protein level. We further show that two microtubule-binding modules of MAP6 bind the Kv3.1 tetramerization domain with high affinity, maintaining the channel level in both neuronal soma and axons. MAP6 knockdown by AAV-shRNA in the amygdala or the hippocampus reduces avoidance or causes hyperactivity and recognition memory deficit, respectively, through elevating projection neuron activity. Finally, knocking down Kv3.1 or disrupting the MAP6-Kv3.1 binding in these brain regions causes avoidance reduction and hyperactivity, consistent with the effects of MAP6 knockdown. Thus, disrupting this conserved cytoskeleton-membrane interaction in fast-spiking neurons causes different degrees of functional vulnerability in various neural circuits.
Collapse
Affiliation(s)
- Di Ma
- Ohio State Biochemistry Program, The Ohio State University, Columbus, OH, USA
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH, 43210, USA
| | - Chao Sun
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH, 43210, USA
- MCDB graduate program, The Ohio State University, Columbus, OH, USA
| | - Rahul Manne
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH, 43210, USA
| | - Tianqi Guo
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, 43210, USA
| | - Christophe Bosc
- Univ. Grenoble Alpes, Inserm, U1216, CEA, Grenoble Institut Neurosciences, 38000, Grenoble, France
| | - Joshua Barry
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH, 43210, USA
- IDDRC, Jane and Terry Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Thomas Magliery
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, 43210, USA
| | - Annie Andrieux
- Univ. Grenoble Alpes, Inserm, U1216, CEA, Grenoble Institut Neurosciences, 38000, Grenoble, France
| | - Houzhi Li
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH, 43210, USA
| | - Chen Gu
- Ohio State Biochemistry Program, The Ohio State University, Columbus, OH, USA.
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH, 43210, USA.
- MCDB graduate program, The Ohio State University, Columbus, OH, USA.
| |
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: 4.5] [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
|
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: 5.0] [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
|
6
|
Protasi F, Pietrangelo L, Boncompagni S. Calcium entry units (CEUs): perspectives in skeletal muscle function and disease. J Muscle Res Cell Motil 2020; 42:233-249. [PMID: 32812118 PMCID: PMC8332569 DOI: 10.1007/s10974-020-09586-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 08/03/2020] [Indexed: 12/28/2022]
Abstract
In the last decades the term Store-operated Ca2+ entry (SOCE) has been used in the scientific literature to describe an ubiquitous cellular mechanism that allows recovery of calcium (Ca2+) from the extracellular space. SOCE is triggered by a reduction of Ca2+ content (i.e. depletion) in intracellular stores, i.e. endoplasmic or sarcoplasmic reticulum (ER and SR). In skeletal muscle the mechanism is primarily mediated by a physical interaction between stromal interaction molecule-1 (STIM1), a Ca2+ sensor located in the SR membrane, and ORAI1, a Ca2+-permeable channel of external membranes, located in transverse tubules (TTs), the invaginations of the plasma membrane (PM) deputed to propagation of action potentials. It is generally accepted that in skeletal muscle SOCE is important to limit muscle fatigue during repetitive stimulation. We recently discovered that exercise promotes the assembly of new intracellular junctions that contains colocalized STIM1 and ORAI1, and that the presence of these new junctions increases Ca2+ entry via ORAI1, while improving fatigue resistance during repetitive stimulation. Based on these findings we named these new junctions Ca2+ Entry Units (CEUs). CEUs are dynamic organelles that assemble during muscle activity and disassemble during recovery thanks to the plasticity of the SR (containing STIM1) and the elongation/retraction of TTs (bearing ORAI1). Interestingly, similar structures described as SR stacks were previously reported in different mouse models carrying mutations in proteins involved in Ca2+ handling (calsequestrin-null mice; triadin and junctin null mice, etc.) or associated to microtubules (MAP6 knockout mice). Mutations in Stim1 and Orai1 (and calsequestrin-1) genes have been associated to tubular aggregate myopathy (TAM), a muscular disease characterized by: (a) muscle pain, cramping, or weakness that begins in childhood and worsens over time, and (b) the presence of large accumulations of ordered SR tubes (tubular aggregates, TAs) that do not contain myofibrils, mitochondria, nor TTs. Interestingly, TAs are also present in fast twitch muscle fibers of ageing mice. Several important issues remain un-answered: (a) the molecular mechanisms and signals that trigger the remodeling of membranes and the functional activation of SOCE during exercise are unclear; and (b) how dysfunctional SOCE and/or mutations in Stim1, Orai1 and calsequestrin (Casq1) genes lead to the formation of tubular aggregates (TAs) in aging and disease deserve investigation.
Collapse
Affiliation(s)
- Feliciano Protasi
- CAST, Center for Advanced Studies and Technology, University G. d'Annunzio of Chieti-Pescara, 66100, Chieti, Italy.
- DMSI, Department of Medicine and Aging Sciences, University G. d'Annunzio of Chieti-Pescara, 66100, Chieti, Italy.
| | - Laura Pietrangelo
- CAST, Center for Advanced Studies and Technology, University G. d'Annunzio of Chieti-Pescara, 66100, Chieti, Italy
- DMSI, Department of Medicine and Aging Sciences, University G. d'Annunzio of Chieti-Pescara, 66100, Chieti, Italy
| | - Simona Boncompagni
- CAST, Center for Advanced Studies and Technology, University G. d'Annunzio of Chieti-Pescara, 66100, Chieti, Italy
- DNICS, Department of Neuroscience, Imaging and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, 66100, Chieti, Italy
| |
Collapse
|
7
|
Ma L, Song J, Sun X, Ding W, Fan K, Qi M, Xu Y, Zhang W. Role of microtubule-associated protein 6 glycosylated with Gal-(β-1,3)-GalNAc in Parkinson's disease. Aging (Albany NY) 2020; 11:4597-4610. [PMID: 31289257 PMCID: PMC6660046 DOI: 10.18632/aging.102072] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 06/28/2019] [Indexed: 12/13/2022]
Abstract
Aberrant glycosylation of proteins has major implications for human diseases. To determine whether protein glycosylation contributes to the pathogenesis of Parkinson’s disease (PD), a mouse model of PD was established by injection of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Induction of PD-like features was verified by assessing motor impairment and confirming reductions in biological markers, including dopamine, 5-hydroxytryptamine and tyrosine hydroxylase, as well as the aggregation of α-synuclein. Altered glycosylation was detected using biotinylated agaracus bisporus lectin, which specifically binds exposed Gal-(β-1,3)-GalNAc linked to glycoproteins. Subsequent lectin affinity chromatography coupled with mass spectrometry revealed enhanced glycosylation of microtubule-associated protein 6 (MAP6) in PD mice as compared to healthy controls. In situ dual co-immunofluorescence analysis and immunoblotting confirmed that MAP6 is glycosylated with Gal-(β-1,3)-GalNAc oligosaccharides, which in turn alters the distribution and structure of MAP6 complexes within neurons. This is the first study to described MAP6 as a glycoprotein containing Gal-(β-1,3)-GalNAc oligosaccharides and to show that hyperglycosylation of MAP6 is strongly associated with the pathogenesis of PD. These findings provide potentially valuable information for developing new therapeutic targets for the treatment of PD as well as reliably prognostic biomarkers.
Collapse
Affiliation(s)
- Li Ma
- Department of Epidemiology, Dalian Medical University, Dalian 116044, China
| | - Jiaxin Song
- Department of Epidemiology, Dalian Medical University, Dalian 116044, China
| | - Xueying Sun
- Department of Epidemiology, Dalian Medical University, Dalian 116044, China
| | - Wenyong Ding
- Biochemistry and Molecular Biology Department of College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Kaiyang Fan
- Medical Administration Department, Affiliated Hospital of Jining Medical University, Jining 272000, China
| | - Minghua Qi
- Biochemistry and Molecular Biology Department of College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Yuefei Xu
- Biochemistry and Molecular Biology Department of College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Wenli Zhang
- Biochemistry and Molecular Biology Department of College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China
| |
Collapse
|
8
|
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: 2.5] [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
|
9
|
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: 2.8] [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
|
10
|
Hankey W, McIlhatton MA, Ebede K, Kennedy B, Hancioglu B, Zhang J, Brock GN, Huang K, Groden J. Mutational Mechanisms That Activate Wnt Signaling and Predict Outcomes in Colorectal Cancer Patients. Cancer Res 2017; 78:617-630. [PMID: 29212857 DOI: 10.1158/0008-5472.can-17-1357] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 09/14/2017] [Accepted: 11/28/2017] [Indexed: 11/16/2022]
Abstract
APC biallelic loss-of-function mutations are the most prevalent genetic changes in colorectal tumors, but it is unknown whether these mutations phenocopy gain-of-function mutations in the CTNNB1 gene encoding β-catenin that also activate canonical WNT signaling. Here we demonstrate that these two mutational mechanisms are not equivalent. Furthermore, we show how differences in gene expression produced by these different mechanisms can stratify outcomes in more advanced human colorectal cancers. Gene expression profiling in Apc-mutant and Ctnnb1-mutant mouse colon adenomas identified candidate genes for subsequent evaluation of human TCGA (The Cancer Genome Atlas) data for colorectal cancer outcomes. Transcriptional patterns exhibited evidence of activated canonical Wnt signaling in both types of adenomas, with Apc-mutant adenomas also exhibiting unique changes in pathways related to proliferation, cytoskeletal organization, and apoptosis. Apc-mutant adenomas were characterized by increased expression of the glial nexin Serpine2, the human ortholog, which was increased in advanced human colorectal tumors. Our results support the hypothesis that APC-mutant colorectal tumors are transcriptionally distinct from APC-wild-type colorectal tumors with canonical WNT signaling activated by other mechanisms, with possible implications for stratification and prognosis.Significance: These findings suggest that colon adenomas driven by APC mutations are distinct from those driven by WNT gain-of-function mutations, with implications for identifying at-risk patients with advanced disease based on gene expression patterns. Cancer Res; 78(3); 617-30. ©2017 AACR.
Collapse
Affiliation(s)
- William Hankey
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Michael A McIlhatton
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Kenechi Ebede
- Department of Anesthesiology, University of Florida, Gainesville, Florida
| | - Brian Kennedy
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Baris Hancioglu
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Jie Zhang
- Department of Medical and Molecular Genetics, Indiana University, School of Medicine, Indianapolis, Indiana
| | - Guy N Brock
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Kun Huang
- Department of Medical and Molecular Genetics, Indiana University, School of Medicine, Indianapolis, Indiana
| | - Joanna Groden
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, Ohio.
| |
Collapse
|
11
|
Establishing Neuronal Polarity with Environmental and Intrinsic Mechanisms. Neuron 2017; 96:638-650. [DOI: 10.1016/j.neuron.2017.10.021] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 10/14/2017] [Accepted: 10/15/2017] [Indexed: 12/31/2022]
|
12
|
Dynamic Palmitoylation Targets MAP6 to the Axon to Promote Microtubule Stabilization during Neuronal Polarization. Neuron 2017; 94:809-825.e7. [PMID: 28521134 DOI: 10.1016/j.neuron.2017.04.042] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 02/19/2017] [Accepted: 04/27/2017] [Indexed: 11/22/2022]
Abstract
Microtubule-associated proteins (MAPs) are main candidates to stabilize neuronal microtubules, playing an important role in establishing axon-dendrite polarity. However, how MAPs are selectively targeted to specific neuronal compartments remains poorly understood. Here, we show specific localization of microtubule-associated protein 6 (MAP6)/stable tubule-only polypeptide (STOP) throughout neuronal maturation and its role in axonal development. In unpolarized neurons, MAP6 is present at the Golgi complex and in secretory vesicles. As neurons mature, MAP6 is translocated to the proximal axon, where it binds and stabilizes microtubules. Further, we demonstrate that dynamic palmitoylation, mediated by the family of α/β Hydrolase domain-containing protein 17 (ABHD17A-C) depalmitoylating enzymes, controls shuttling of MAP6 between membranes and microtubules and is required for MAP6 retention in axons. We propose a model in which MAP6's palmitoylation mediates microtubule stabilization, allows efficient organelle trafficking, and controls axon maturation in vitro and in situ.
Collapse
|
13
|
Gu Y, Jukkola P, Wang Q, Esparza T, Zhao Y, Brody D, Gu C. Polarity of varicosity initiation in central neuron mechanosensation. J Cell Biol 2017; 216:2179-2199. [PMID: 28606925 PMCID: PMC5496611 DOI: 10.1083/jcb.201606065] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 01/17/2017] [Accepted: 04/20/2017] [Indexed: 12/22/2022] Open
Abstract
Little is known about mechanical regulation of morphological and functional polarity of central neurons. In this study, we report that mechanical stress specifically induces varicosities in the axons but not the dendrites of central neurons by activating TRPV4, a Ca2+/Na+-permeable mechanosensitive channel. This process is unexpectedly rapid and reversible, consistent with the formation of axonal varicosities in vivo induced by mechanical impact in a mouse model of mild traumatic brain injury. In contrast, prolonged stimulation of glutamate receptors induces varicosities in dendrites but not in axons. We further show that axonal varicosities are induced by persistent Ca2+ increase, disassembled microtubules (MTs), and subsequently reversible disruption of axonal transport, and are regulated by stable tubulin-only polypeptide, an MT-associated protein. Finally, axonal varicosity initiation can trigger action potentials to antidromically propagate to the soma in retrograde signaling. Therefore, our study demonstrates a new feature of neuronal polarity: axons and dendrites preferentially respond to physical and chemical stresses, respectively.
Collapse
Affiliation(s)
- Yuanzheng Gu
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH
| | - Peter Jukkola
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH
| | - Qian Wang
- Biomedical Engineering Department, The Ohio State University, Columbus, OH
| | - Thomas Esparza
- Department of Neurology, Washington University, St. Louis, MO
| | - Yi Zhao
- Biomedical Engineering Department, The Ohio State University, Columbus, OH
| | - David Brody
- Department of Neurology, Washington University, St. Louis, MO
| | - Chen Gu
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH
| |
Collapse
|
14
|
Marchisella F, Coffey ET, Hollos P. Microtubule and microtubule associated protein anomalies in psychiatric disease. Cytoskeleton (Hoboken) 2016; 73:596-611. [DOI: 10.1002/cm.21300] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 04/03/2016] [Accepted: 04/13/2016] [Indexed: 12/24/2022]
Affiliation(s)
- Francesca Marchisella
- Turku Centre for Biotechnology; Åbo Akademi University and University of Turku; Finland
| | - Eleanor T. Coffey
- Turku Centre for Biotechnology; Åbo Akademi University and University of Turku; Finland
| | - Patrik Hollos
- Turku Centre for Biotechnology; Åbo Akademi University and University of Turku; Finland
| |
Collapse
|
15
|
Penazzi L, Bakota L, Brandt R. Microtubule Dynamics in Neuronal Development, Plasticity, and Neurodegeneration. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2015; 321:89-169. [PMID: 26811287 DOI: 10.1016/bs.ircmb.2015.09.004] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Neurons are the basic information-processing units of the nervous system. In fulfilling their task, they establish a structural polarity with an axon that can be over a meter long and dendrites with a complex arbor, which can harbor ten-thousands of spines. Microtubules and their associated proteins play important roles during the development of neuronal morphology, the plasticity of neurons, and neurodegenerative processes. They are dynamic structures, which can quickly adapt to changes in the environment and establish a structural scaffold with high local variations in composition and stability. This review presents a comprehensive overview about the role of microtubules and their dynamic behavior during the formation and maturation of processes and spines in the healthy brain, during aging and under neurodegenerative conditions. The review ends with a discussion of microtubule-targeted therapies as a perspective for the supportive treatment of neurodegenerative disorders.
Collapse
Affiliation(s)
- Lorène Penazzi
- Department of Neurobiology, University of Osnabrück, Osnabrück, Germany
| | - Lidia Bakota
- Department of Neurobiology, University of Osnabrück, Osnabrück, Germany
| | - Roland Brandt
- Department of Neurobiology, University of Osnabrück, Osnabrück, Germany
| |
Collapse
|
16
|
Dubey J, Ratnakaran N, Koushika SP. Neurodegeneration and microtubule dynamics: death by a thousand cuts. Front Cell Neurosci 2015; 9:343. [PMID: 26441521 PMCID: PMC4563776 DOI: 10.3389/fncel.2015.00343] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2015] [Accepted: 08/18/2015] [Indexed: 12/11/2022] Open
Abstract
Microtubules form important cytoskeletal structures that play a role in establishing and maintaining neuronal polarity, regulating neuronal morphology, transporting cargo, and scaffolding signaling molecules to form signaling hubs. Within a neuronal cell, microtubules are found to have variable lengths and can be both stable and dynamic. Microtubule associated proteins, post-translational modifications of tubulin subunits, microtubule severing enzymes, and signaling molecules are all known to influence both stable and dynamic pools of microtubules. Microtubule dynamics, the process of interconversion between stable and dynamic pools, and the proportions of these two pools have the potential to influence a wide variety of cellular processes. Reduced microtubule stability has been observed in several neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), Amyotrophic Lateral Sclerosis (ALS), and tauopathies like Progressive Supranuclear Palsy. Hyperstable microtubules, as seen in Hereditary Spastic Paraplegia (HSP), also lead to neurodegeneration. Therefore, the ratio of stable and dynamic microtubules is likely to be important for neuronal function and perturbation in microtubule dynamics might contribute to disease progression.
Collapse
Affiliation(s)
- Jyoti Dubey
- Department of Biological Sciences, Tata Institute of Fundamental Research Mumbai, India ; InStem Bangalore, India
| | - Neena Ratnakaran
- Department of Biological Sciences, Tata Institute of Fundamental Research Mumbai, India
| | - Sandhya P Koushika
- Department of Biological Sciences, Tata Institute of Fundamental Research Mumbai, India
| |
Collapse
|
17
|
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: 5.6] [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
|
18
|
Gory-Fauré S, Windscheid V, Brocard J, Montessuit S, Tsutsumi R, Denarier E, Fukata Y, Bosc C, Delaroche J, Collomb N, Fukata M, Martinou JC, Pernet-Gallay K, Andrieux A. Non-microtubular localizations of microtubule-associated protein 6 (MAP6). PLoS One 2014; 9:e114905. [PMID: 25526643 PMCID: PMC4272302 DOI: 10.1371/journal.pone.0114905] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 11/11/2014] [Indexed: 01/29/2023] Open
Abstract
MAP6 proteins (MAP6s), which include MAP6-N (also called Stable Tubule Only Polypeptide, or STOP) and MAP6d1 (MAP6 domain-containing protein 1, also called STOP-Like protein 21 kD, or SL21), bind to and stabilize microtubules. MAP6 deletion in mice severely alters integrated brain functions and is associated with synaptic defects, suggesting that MAP6s may also have alternative cellular roles. MAP6s reportedly associate with the Golgi apparatus through palmitoylation of their N-terminal domain, and specific isoforms have been shown to bind actin. Here, we use heterologous systems to investigate several biochemical properties of MAP6 proteins. We demonstrate that the three N-terminal cysteines of MAP6d1 are palmitoylated by a subset of DHHC-type palmitoylating enzymes. Analysis of the subcellular localization of palmitoylated MAP6d1, including electron microscopic analysis, reveals possible localization to the Golgi and the plasma membrane but no association with the endoplasmic reticulum. Moreover, we observed localization of MAP6d1 to mitochondria, which requires the N-terminus of the protein but does not require palmitoylation. We show that endogenous MAP6d1 localized at mitochondria in mature mice neurons as well as at the outer membrane and in the intermembrane space of purified mouse mitochondria. Last, we found that MAP6d1 can multimerize via a microtubule-binding module. Interestingly, most of these properties of MAP6d1 are shared by MAP6-N. Together, these results describe several properties of MAP6 proteins, including their intercellular localization and multimerization activity, which may be relevant to neuronal differentiation and synaptic functions.
Collapse
Affiliation(s)
- Sylvie Gory-Fauré
- Inserm, U836, Physiopathologie du cytosquelette, BP170, Grenoble, France
- University Grenoble Alpes, Grenoble Institut des Neurosciences, BP170, Grenoble, France
- * E-mail: (SGF); (AA)
| | - Vanessa Windscheid
- Inserm, U836, Physiopathologie du cytosquelette, BP170, Grenoble, France
- University Grenoble Alpes, Grenoble Institut des Neurosciences, BP170, Grenoble, France
| | - Jacques Brocard
- Inserm, U836, Physiopathologie du cytosquelette, BP170, Grenoble, France
- University Grenoble Alpes, Grenoble Institut des Neurosciences, BP170, Grenoble, France
| | - Sylvie Montessuit
- Department of Cell Biology, University of Geneva, Sciences III, Geneva, Switzerland
| | - Ryouhei Tsutsumi
- Division of Membrane Physiology, Department of Cell Physiology, National Institute for Physiological Sciences, Aichi, Japan
| | - Eric Denarier
- Inserm, U836, Physiopathologie du cytosquelette, BP170, Grenoble, France
- University Grenoble Alpes, Grenoble Institut des Neurosciences, BP170, Grenoble, France
- Commissariat à l'énergie atomique, Institut de Recherches en Technologies et Sciences pour le Vivant, Groupe Physiopathologie du Cytosquelette, Grenoble, France
| | - Yuko Fukata
- Division of Membrane Physiology, Department of Cell Physiology, National Institute for Physiological Sciences, Aichi, Japan
| | - Christophe Bosc
- Inserm, U836, Physiopathologie du cytosquelette, BP170, Grenoble, France
- University Grenoble Alpes, Grenoble Institut des Neurosciences, BP170, Grenoble, France
| | - Julie Delaroche
- Inserm, U836, Physiopathologie du cytosquelette, BP170, Grenoble, France
- University Grenoble Alpes, Grenoble Institut des Neurosciences, BP170, Grenoble, France
| | - Nora Collomb
- Inserm, U836, Physiopathologie du cytosquelette, BP170, Grenoble, France
- University Grenoble Alpes, Grenoble Institut des Neurosciences, BP170, Grenoble, France
| | - Masaki Fukata
- Division of Membrane Physiology, Department of Cell Physiology, National Institute for Physiological Sciences, Aichi, Japan
| | - Jean-Claude Martinou
- Department of Cell Biology, University of Geneva, Sciences III, Geneva, Switzerland
| | - Karin Pernet-Gallay
- Inserm, U836, Physiopathologie du cytosquelette, BP170, Grenoble, France
- University Grenoble Alpes, Grenoble Institut des Neurosciences, BP170, Grenoble, France
| | - Annie Andrieux
- Inserm, U836, Physiopathologie du cytosquelette, BP170, Grenoble, France
- University Grenoble Alpes, Grenoble Institut des Neurosciences, BP170, Grenoble, France
- Commissariat à l'énergie atomique, Institut de Recherches en Technologies et Sciences pour le Vivant, Groupe Physiopathologie du Cytosquelette, Grenoble, France
- * E-mail: (SGF); (AA)
| |
Collapse
|
19
|
Schwenk BM, Lang CM, Hogl S, Tahirovic S, Orozco D, Rentzsch K, Lichtenthaler SF, Hoogenraad CC, Capell A, Haass C, Edbauer D. The FTLD risk factor TMEM106B and MAP6 control dendritic trafficking of lysosomes. EMBO J 2013; 33:450-67. [PMID: 24357581 DOI: 10.1002/embj.201385857] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
TMEM106B is a major risk factor for frontotemporal lobar degeneration with TDP-43 pathology. TMEM106B localizes to lysosomes, but its function remains unclear. We show that TMEM106B knockdown in primary neurons affects lysosomal trafficking and blunts dendritic arborization. We identify microtubule-associated protein 6 (MAP6) as novel interacting protein for TMEM106B. MAP6 over-expression inhibits dendritic branching similar to TMEM106B knockdown. MAP6 knockdown fully rescues the dendritic phenotype of TMEM106B knockdown, supporting a functional interaction between TMEM106B and MAP6. Live imaging reveals that TMEM106B knockdown and MAP6 overexpression strongly increase retrograde transport of lysosomes in dendrites. Downregulation of MAP6 in TMEM106B knockdown neurons restores the balance of anterograde and retrograde lysosomal transport and thereby prevents loss of dendrites. To strengthen the link, we enhanced anterograde lysosomal transport by expressing dominant-negative Rab7-interacting lysosomal protein (RILP), which also rescues the dendrite loss in TMEM106B knockdown neurons. Thus, TMEM106B/MAP6 interaction is crucial for controlling dendritic trafficking of lysosomes, presumably by acting as a molecular brake for retrograde transport. Lysosomal misrouting may promote neurodegeneration in patients with TMEM106B risk variants.
Collapse
|
20
|
Volle J, Brocard J, Saoud M, Gory-Faure S, Brunelin J, Andrieux A, Suaud-Chagny MF. Reduced expression of STOP/MAP6 in mice leads to cognitive deficits. Schizophr Bull 2013; 39:969-78. [PMID: 23002183 PMCID: PMC3756782 DOI: 10.1093/schbul/sbs113] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND STOP/MAP6 null (KO) mice recapitulate behavioral abnormalities related to positive and negative symptoms and cognitive deficits of schizophrenia. Here, we investigated whether decreased expression of STOP/MAP6 proteins in heterozygous mice (only one allele expressed) would result in abnormal behavior related to those displayed by STOP null mice. METHODS Using a comprehensive test battery, we investigated the behavioral phenotype of STOP heterozygous (Het) mice compared with STOP KO and wild type (WT) mice on animals raised either in standard conditions (controls) or submitted to maternal deprivation. RESULTS Control Het mice displayed prominent deficits in social interaction and learning, resembling KO mice. In contrast, they exhibited short-lasting locomotor hyperreactivity to acute mild stress and no impaired locomotor response to amphetamine, much like WT mice. Additionally, perinatal stress deteriorated Het mouse phenotype by exacerbating alterations related to positive symptoms such as their locomotor reactivity to acute mild stress and psychostimulant challenge. CONCLUSION Results show that the dosage of susceptibility genes modulates their putative phenotypic contribution and that STOP expression has a high penetrance on cognitive abilities. Hence, STOP Het mice might be useful to investigate cognitive defects related to those observed in mental diseases and ultimately might be a valuable experimental model to evaluate preventive treatments.
Collapse
Affiliation(s)
- Julien Volle
- Université de Lyon, Lyon, F-69003, France; Université Lyon 1, Lyon, EA 4615
| | - Jacques Brocard
- Institut National de la Santé et de la Recherche Médicale Unité 836, Institut des Neurosciences de Grenoble, Université Joseph Fourier, 38042 Grenoble Cedex 9, France;,Groupe Physiopathologie du Cytosquelette, Institut de Recherches en Technologies et Sciences pour le Vivant Direction des Sciences du Vivant, Commissariat à l’Énergie Atomique, 38054 Grenoble Cedex 9, France
| | - Mohamed Saoud
- Université de Lyon, Lyon, F-69003, France; Université Lyon 1, Lyon, EA 4615;,Centre Hospitalier le Vinatier, F-69677 Bron Cedex, France
| | - Sylvie Gory-Faure
- Institut National de la Santé et de la Recherche Médicale Unité 836, Institut des Neurosciences de Grenoble, Université Joseph Fourier, 38042 Grenoble Cedex 9, France;,Groupe Physiopathologie du Cytosquelette, Institut de Recherches en Technologies et Sciences pour le Vivant Direction des Sciences du Vivant, Commissariat à l’Énergie Atomique, 38054 Grenoble Cedex 9, France
| | - Jérôme Brunelin
- Université de Lyon, Lyon, F-69003, France; Université Lyon 1, Lyon, EA 4615;,Centre Hospitalier le Vinatier, F-69677 Bron Cedex, France
| | - Annie Andrieux
- Institut National de la Santé et de la Recherche Médicale Unité 836, Institut des Neurosciences de Grenoble, Université Joseph Fourier, 38042 Grenoble Cedex 9, France;,Groupe Physiopathologie du Cytosquelette, Institut de Recherches en Technologies et Sciences pour le Vivant Direction des Sciences du Vivant, Commissariat à l’Énergie Atomique, 38054 Grenoble Cedex 9, France
| | - Marie-Françoise Suaud-Chagny
- Université de Lyon, Lyon, F-69003, France; Université Lyon 1, Lyon, EA 4615;,Centre Hospitalier le Vinatier, F-69677 Bron Cedex, France;,To whom correspondence should be addressed; EA 4615, Pôle Est - Pr d’Amato, CH le vinatier, 95 bd Pinel, 69677 Bron cedex, France; tel: +33 4 37 91 55 65, fax: +33 4 37 91 55 49, e-mail:
| |
Collapse
|
21
|
Lefèvre J, Savarin P, Gans P, Hamon L, Clément MJ, David MO, Bosc C, Andrieux A, Curmi PA. Structural basis for the association of MAP6 protein with microtubules and its regulation by calmodulin. J Biol Chem 2013; 288:24910-22. [PMID: 23831686 DOI: 10.1074/jbc.m113.457267] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Microtubules are highly dynamic αβ-tubulin polymers. In vitro and in living cells, microtubules are most often cold- and nocodazole-sensitive. When present, the MAP6/STOP family of proteins protects microtubules from cold- and nocodazole-induced depolymerization but the molecular and structure determinants by which these proteins stabilize microtubules remain under debate. We show here that a short protein fragment from MAP6-N, which encompasses its Mn1 and Mn2 modules (MAP6(90-177)), recapitulates the function of the full-length MAP6-N protein toward microtubules, i.e. its ability to stabilize microtubules in vitro and in cultured cells in ice-cold conditions or in the presence of nocodazole. We further show for the first time, using biochemical assays and NMR spectroscopy, that these effects result from the binding of MAP6(90-177) to microtubules with a 1:1 MAP6(90-177):tubulin heterodimer stoichiometry. NMR data demonstrate that the binding of MAP6(90-177) to microtubules involve its two Mn modules but that a single one is also able to interact with microtubules in a closely similar manner. This suggests that the Mn modules represent each a full microtubule binding domain and that MAP6 proteins may stabilize microtubules by bridging tubulin heterodimers from adjacent protofilaments or within a protofilament. Finally, we demonstrate that Ca(2+)-calmodulin competes with microtubules for MAP6(90-177) binding and that the binding mode of MAP6(90-177) to microtubules and Ca(2+)-calmodulin involves a common stretch of amino acid residues on the MAP6(90-177) side. This result accounts for the regulation of microtubule stability in cold condition by Ca(2+)-calmodulin.
Collapse
Affiliation(s)
- Julien Lefèvre
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR829, Laboratoire Structure-Activité des Biomolécules Normales et Pathologiques, Université Evry-Val d'Essonne, Evry 91025, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
|
23
|
Morderer D, Nikolaienko O, Skrypkina I, Cherkas V, Tsyba L, Belan P, Rynditch A. Endocytic adaptor protein intersectin 1 forms a complex with microtubule stabilizer STOP in neurons. Gene 2012; 505:360-4. [DOI: 10.1016/j.gene.2012.06.061] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Accepted: 06/18/2012] [Indexed: 12/17/2022]
|
24
|
Kulkarni VA, Firestein BL. The dendritic tree and brain disorders. Mol Cell Neurosci 2012; 50:10-20. [DOI: 10.1016/j.mcn.2012.03.005] [Citation(s) in RCA: 242] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Accepted: 03/09/2012] [Indexed: 01/21/2023] Open
|
25
|
Arama J, Boulay AC, Bosc C, Delphin C, Loew D, Rostaing P, Amigou E, Ezan P, Wingertsmann L, Guillaud L, Andrieux A, Giaume C, Cohen-Salmon M. Bmcc1s, a novel brain-isoform of Bmcc1, affects cell morphology by regulating MAP6/STOP functions. PLoS One 2012; 7:e35488. [PMID: 22523599 PMCID: PMC3327665 DOI: 10.1371/journal.pone.0035488] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Accepted: 03/16/2012] [Indexed: 12/21/2022] Open
Abstract
The BCH (BNIP2 and Cdc42GAP Homology) domain-containing protein Bmcc1/Prune2 is highly enriched in the brain and is involved in the regulation of cytoskeleton dynamics and cell survival. However, the molecular mechanisms accounting for these functions are poorly defined. Here, we have identified Bmcc1s, a novel isoform of Bmcc1 predominantly expressed in the mouse brain. In primary cultures of astrocytes and neurons, Bmcc1s localized on intermediate filaments and microtubules and interacted directly with MAP6/STOP, a microtubule-binding protein responsible for microtubule cold stability. Bmcc1s overexpression inhibited MAP6-induced microtubule cold stability by displacing MAP6 away from microtubules. It also resulted in the formation of membrane protrusions for which MAP6 was a necessary cofactor of Bmcc1s. This study identifies Bmcc1s as a new MAP6 interacting protein able to modulate MAP6-induced microtubule cold stability. Moreover, it illustrates a novel mechanism by which Bmcc1 regulates cell morphology.
Collapse
Affiliation(s)
- Jessica Arama
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale U1050, Paris, France
- University Pierre et Marie Curie, ED, N°158, Paris, France
- MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, Paris, France
| | - Anne-Cécile Boulay
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale U1050, Paris, France
- University Pierre et Marie Curie, ED, N°158, Paris, France
- MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, Paris, France
| | - Christophe Bosc
- Equipe Physiopathologie du Cytosquelette, Institut National de la Santé et de la Recherche Médicale U836, Institut des Neurosciences, Université Joseph Fourier, Faculté de Médecine, Domaine de la Merci, La Tronche, France
| | - Christian Delphin
- Equipe Physiopathologie du Cytosquelette, Institut National de la Santé et de la Recherche Médicale U836, Institut des Neurosciences, Université Joseph Fourier, Faculté de Médecine, Domaine de la Merci, La Tronche, France
| | - Damarys Loew
- Institut Curie, Laboratory of Proteomic Mass Spectrometry, Paris, France
| | - Philippe Rostaing
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Institut National de la Santé et de la Recherche Médicale U1024, Paris, France
| | - Edwige Amigou
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale U1050, Paris, France
- University Pierre et Marie Curie, ED, N°158, Paris, France
- MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, Paris, France
| | - Pascal Ezan
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale U1050, Paris, France
- University Pierre et Marie Curie, ED, N°158, Paris, France
- MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, Paris, France
| | - Laure Wingertsmann
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Institut National de la Santé et de la Recherche Médicale U1024, Paris, France
| | - Laurent Guillaud
- Cell and Molecular Synaptic Function Unit, Okinawa Institute of Science and Technology, Okinawa, Japan
| | - Annie Andrieux
- Equipe Physiopathologie du Cytosquelette, Institut National de la Santé et de la Recherche Médicale U836, Institut des Neurosciences, Université Joseph Fourier, Faculté de Médecine, Domaine de la Merci, La Tronche, France
| | - Christian Giaume
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale U1050, Paris, France
- University Pierre et Marie Curie, ED, N°158, Paris, France
- MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, Paris, France
| | - Martine Cohen-Salmon
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale U1050, Paris, France
- University Pierre et Marie Curie, ED, N°158, Paris, France
- MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, Paris, France
- * E-mail:
| |
Collapse
|
26
|
Lin PC, Chan PM, Hall C, Manser E. Collapsin response mediator proteins (CRMPs) are a new class of microtubule-associated protein (MAP) that selectively interacts with assembled microtubules via a taxol-sensitive binding interaction. J Biol Chem 2011; 286:41466-41478. [PMID: 21953449 DOI: 10.1074/jbc.m111.283580] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Collapsin response mediator proteins are ubiquitously expressed from multiple genes (CRMPs 1-5) and play important roles in dividing cells and during semaphorin 3A (Sema3A) signaling. Nonetheless, their mode of action remains opaque. Here we carried out in vivo and in vitro assays that demonstrate that CRMPs are a new class of microtubule-associated protein (MAP). In experiments with CRMP1 or CRMP2 and their derivatives, only the C-terminal region (residues 490-572) mediated microtubule binding. The in vivo microtubule association of CRMPs was abolished by taxol or epothilone B, which is highly unusual. CRMP2-depleted cells exhibited destabilized anaphase astral microtubules and altered spindle position. In a cell-based assay, all CRMPs stabilized interphase microtubules against nocodazole-mediated depolymerization, with CRMP1 being the most potent. Remarkably, a 82-residue C-terminal region of CRMP1 or CRMP2, unrelated to other microtubule binding motifs, is sufficient to stabilize microtubules. In cells, we demonstrate that glycogen synthase kinase-3β (GSK3β) inhibition potentiates this activity. Thus, CRMPs are a new class of MAP that binds through a unique motif, but in common with others such as Tau, is antagonized by GSK3β. This regulation is consistent with such kinases being critical for the Sema3A (collapsin) pathway. These findings have implications for cancer and neurodegeneration.
Collapse
Affiliation(s)
- Pao-Chun Lin
- Institute of Medical Biology, 8A Biomedical Grove, Singapore 138648
| | - Perry M Chan
- Small G-protein Signaling and Kinases (sGSK-NRP) Group, Neuroscience Research Partnership, 61 Biopolis Drive, Singapore 138673
| | - Christine Hall
- Institute of Neurology, University College London, 1 Wakefield Street, London WC1N 1PJ, United Kingdom
| | - Ed Manser
- Small G-protein Signaling and Kinases (sGSK-NRP) Group, Neuroscience Research Partnership, 61 Biopolis Drive, Singapore 138673; Institute of Medical Biology, 8A Biomedical Grove, Singapore 138648.
| |
Collapse
|
27
|
Cytoskeletal remodeling by C3G to induce neurite-like extensions and inhibit motility in highly invasive breast carcinoma cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2011; 1813:456-65. [DOI: 10.1016/j.bbamcr.2011.01.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2010] [Revised: 12/29/2010] [Accepted: 01/03/2011] [Indexed: 11/24/2022]
|
28
|
Fournet V, Jany M, Fabre V, Chali F, Orsal D, Schweitzer A, Andrieux A, Messanvi F, Giros B, Hamon M, Lanfumey L, Deloulme JC, Martres MP. The deletion of the microtubule-associated STOP protein affects the serotonergic mouse brain network. J Neurochem 2010; 115:1579-94. [PMID: 20969568 DOI: 10.1111/j.1471-4159.2010.07064.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The deletion of microtubule-associated protein stable tubule only polypeptide (STOP) leads to neuroanatomical, biochemical and severe behavioral alterations in mice, partly alleviated by antipsychotics. Therefore, STOP knockout (KO) mice have been proposed as a model of some schizophrenia-like symptoms. Preliminary data showed decreased brain serotonin (5-HT) tissue levels in STOP KO mice. As literature data demonstrate various interactions between microtubule-associated proteins and 5-HT, we characterized some features of the serotonergic neurotransmission in STOP KO mice. In the brainstem, mutant mice displayed higher tissue 5-HT levels and in vivo synthesis rate, together with marked increases in 5-HT transporter densities and 5-HT1A autoreceptor levels and electrophysiological sensitivity, without modification of the serotonergic soma number. Conversely, in projection areas, STOP KO mice exhibited lower 5-HT levels and in vivo synthesis rate, associated with severe decreases in 5-HT transporter densities, possibly related to reduced serotonergic terminals. Mutant mice also displayed a deficit of adult hippocampal neurogenesis, probably related to both STOP deletion and 5-HT depletion. Finally, STOP KO mice exhibited a reduced anxiety- and, probably, an increased helpness-status, that could be because of the strong imbalance of the serotonin neurotransmission between somas and terminals. Altogether, these data suggested that STOP deletion elicited peculiar 5-HT disconnectivity.
Collapse
Affiliation(s)
- Vincent Fournet
- INSERM UMRS 952, CNRS UMR 7224, Université Pierre et Marie Curie, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Vikhreva PN, Korobko EV, Korobko IV. Identification of novel proteins, possible interaction partners for guanine nucleotide exchange factor varp. DOKL BIOCHEM BIOPHYS 2009; 429:323-5. [DOI: 10.1134/s160767290906009x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
30
|
Bisig CG, Chesta ME, Zampar GG, Purro SA, Santander VS, Arce CA. Lack of stabilized microtubules as a result of the absence of major maps in CAD cells does not preclude neurite formation. FEBS J 2009; 276:7110-23. [DOI: 10.1111/j.1742-4658.2009.07422.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
31
|
The microtubule network and neuronal morphogenesis: Dynamic and coordinated orchestration through multiple players. Mol Cell Neurosci 2009; 43:15-32. [PMID: 19660553 DOI: 10.1016/j.mcn.2009.07.012] [Citation(s) in RCA: 116] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2009] [Accepted: 07/27/2009] [Indexed: 11/24/2022] Open
Abstract
Nervous system function and plasticity rely on the complex architecture of neuronal networks elaborated during development, when neurons acquire their specific and complex shape. During neuronal morphogenesis, the formation and outgrowth of functionally and structurally distinct axons and dendrites require a coordinated and dynamic reorganization of the microtubule cytoskeleton involving numerous regulators. While most of these factors act directly on microtubules to stabilize them or promote their assembly, depolymerization or fragmentation, others are now emerging as essential regulators of neuronal differentiation by controlling tubulin availability and modulating microtubule dynamics. In this review, we recapitulate how the microtubule network is actively regulated during the successive phases of neuronal morphogenesis, and what are the specific roles of the various microtubule-regulating proteins in that process. We then describe the specific signaling pathways and inter-regulations that coordinate the different activities of these proteins to sustain neuronal development in response to environmental cues.
Collapse
|
32
|
Ajuba: a new microtubule-associated protein that interacts with BUBR1 and Aurora B at kinetochores in metaphase. Biol Cell 2009; 101:221-35. [PMID: 18710370 DOI: 10.1042/bc20080060] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND INFORMATION The role of the LIM-domain-containing protein Ajuba was initially described in cell adhesion and migration processes and recently in mitosis as an activator of the Aurora A kinase. RESULTS In the present study, we show that Ajuba localizes to centrosomes and kinetochores during mitosis. This localization is microtubule-dependent and Ajuba binds microtubules in vitro. A microtubule regrowth assay showed that Ajuba follows nascent microtubules from centrosomes to kinetochores. Owing to its contribution to mitotic commitment and its microtubule-dependent localization, Ajuba could also play a role during the metaphase-anaphase transition. We show that Ajuba interacts with Aurora B and BUBR1 [BUB (budding uninhibited by benomyl)-related 1], two major components of the mitotic checkpoint. Inhibition of BUBR1 by siRNA (small interfering RNA) disrupts chromosome alignment at the metaphase plate and modifies Ajuba localization due to premature mitotic exit. CONCLUSIONS Ajuba is a microtubule-associated protein that collaborates with Aurora B and BUBR1 at the metaphase-anaphase transition and this may be important to ensure proper chromosome segregation.
Collapse
|
33
|
Richard M, Sacquet J, Jany M, Schweitzer A, Jourdan F, Andrieux A, Pellier-Monnin V. STOP proteins contribute to the maturation of the olfactory system. Mol Cell Neurosci 2009; 41:120-34. [PMID: 19236915 DOI: 10.1016/j.mcn.2009.02.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2008] [Revised: 01/13/2009] [Accepted: 02/06/2009] [Indexed: 01/09/2023] Open
Abstract
Regulation of microtubule dynamics is crucial for axon growth and guidance as well as for the establishment of synaptic connections. STOPs (Stable Tubule Only Polypeptides) are microtubule-associated proteins that regulate microtubule stabilization but are also able to interact with actin or Golgi membranes. Here, we have investigated the involvement of STOPs during the development of the olfactory system. We first describe the spatio-temporal expression patterns of N- and E-STOP, the two neuronal-specific isoforms of STOP. E- and N-STOP are expressed in the axonal compartment of olfactory sensory neurons, but are differentially regulated during development. Interestingly, each neuronal isoform displays a specific gradient distribution within the olfactory nerve layer. Then, we have examined the development of the olfactory system in the absence of STOPs. Olfactory axons display a normal outgrowth and targeting in STOP-null mice, but maturation of the synapses in the glomerular neuropil is altered.
Collapse
Affiliation(s)
- Marion Richard
- Laboratoire Neurosciences Sensorielles, Comportement, Cognition, CNRS-UMR 5020, Université de Lyon, Lyon 1, F-69366, France.
| | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
Axons are occupied by dense arrays of cytoskeletal elements called microtubules, which are critical for generating and maintaining the architecture of the axon, and for acting as railways for the transport of organelles in both directions within the axon. Microtubules are organized and regulated by molecules that affect their assembly and disassembly, their stabilization, their association with other cytoskeletal elements, and their alignment and bundling with one another. Recent studies have accentuated the role of molecular motor proteins and microtubule-severing proteins in the establishment and maintenance of the axonal microtubule array. The growing body of knowledge on the proteins and mechanisms that regulate axonal microtubules has fostered a better understanding of how many debilitating diseases cause axons to degenerate. The purpose of this chapter is to provide an update on current knowledge of axonal microtubules and the proteins that regulate them, and to reflect on cutting-edge findings linking these proteins and mechanisms to diseases that afflict the human population.
Collapse
|
35
|
The stop null mice model for schizophrenia displays [corrected] cognitive and social deficits partly alleviated by neuroleptics. Neuroscience 2008; 157:29-39. [PMID: 18804150 DOI: 10.1016/j.neuroscience.2008.07.080] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2008] [Revised: 07/31/2008] [Accepted: 07/31/2008] [Indexed: 11/20/2022]
Abstract
Recently evidence has accumulated that schizophrenia can arise from primary synaptic defects involving structural proteins particularly, microtubule associated proteins. Previous experiments have demonstrated that a STOP (stable tubule only peptide) gene deletion in mice leads to a phenotype mimicking some aspects of positive symptoms classically observed in schizophrenic patients. In the current study, we determined if STOP null mice demonstrate behavioral abnormalities related to the social and cognitive impairments of schizophrenia. Compared with wild-type mice, STOP null mice exhibited deficits in the non-aggressive component of social recognition, short term working memory and social and spatial learning. As described in humans, learning deficits in STOP null mice were poorly sensitive to long term treatment with typical neuroleptics. Since social and cognitive dysfunction have consistently been considered as central features of schizophrenia, we propose that STOP null mice may provide a useful model to understand the neurobiological correlates of social and cognitive defects in schizophrenia and to develop treatments that better target these symptoms.
Collapse
|
36
|
Perrot R, Berges R, Bocquet A, Eyer J. Review of the Multiple Aspects of Neurofilament Functions, and their Possible Contribution to Neurodegeneration. Mol Neurobiol 2008; 38:27-65. [DOI: 10.1007/s12035-008-8033-0] [Citation(s) in RCA: 205] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2008] [Accepted: 06/14/2008] [Indexed: 10/21/2022]
|
37
|
Hanaya R, Koning E, Ferrandon A, Schweitzer A, Andrieux A, Nehlig A. Deletion of the STOP gene, a microtubule stabilizing factor, leads only to discrete cerebral metabolic changes in mice. J Neurosci Res 2008; 86:813-20. [PMID: 17969102 DOI: 10.1002/jnr.21550] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
In mice, deletion of the STOP protein leads to subtle anatomic changes and induces depleted synaptic vesicle pools, impaired synaptic plasticity, hyperdopaminergy, and major behavioral disorders alleviated by neuroleptics, hence leading to a schizophrenic-like phenotype. In this study, we applied the quantitative autoradiographic [(14)C]2-deoxyglucose technique to study to what extent the basal rate of cerebral glucose utilization in STOP-knockout (STOP-KO) mice occurs in regions where metabolic changes have been reported in schizophrenic patients. Studies were performed on wild-type, heterozygous, and homozygous STOP-KO mice (7-8 per group). Mice were implanted with femoral artery and vein catheters, and cerebral glucose utilization was quantified over 45 min. Compared with that in wild-type mice, glucose utilization in STOP-KO mice was significantly increased in the olfactory cortex, ventromedial and anterolateral hypothalamus, ventral tegmental area, and substantia nigra pars compacta. Nonsignificant increases, ranging between 9% and 19%, were recorded in the whole auditory system, CA1 pyramidal cell layer, and dorsal raphe. Glucose utilization was also significantly increased in heterozygous mice compared with that in wild-type mice in olfactory cortex. These data might reflect hyperdopaminergic activity, olfactory deficits, and sleep disturbances in STOP-KO mice that have also been reported in schizophrenic patients.
Collapse
|
38
|
Bouvrais-Veret C, Weiss S, Hanoun N, Andrieux A, Schweitzer A, Job D, Hamon M, Giros B, Martres MP. Microtubule-associated STOP protein deletion triggers restricted changes in dopaminergic neurotransmission. J Neurochem 2008; 104:745-56. [PMID: 18199119 DOI: 10.1111/j.1471-4159.2007.05025.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The microtubule-associated stable tubule only polypeptide (STOP) protein plays a key-role in neuron architecture and synaptic plasticity. Recent studies suggest that schizophrenia is associated with alterations in the synaptic connectivity. Mice invalidated for the STOP gene display phenotype reminiscent of some schizophrenic-like symptoms, such as behavioral disturbances, dopamine (DA) hyper-reactivity, and possible hypoglutamatergia, partly improved by antipsychotic treatment. In the present work, we examined potential alterations in some DAergic key proteins and behaviors in STOP knockout mice. Whereas the densities of the DA transporter, the vesicular monoamine transporter and the D(1) receptor were not modified, the densities of the D(2) and D(3) receptors were decreased in some DAergic regions in mutant versus wild-type mice. Endogenous DA levels were selectively decreased in DAergic terminals areas, although the in vivo DA synthesis was diminished both in cell bodies and terminal areas. The DA uptake was decreased in accumbic synaptosomes, but not significantly altered in striatal synaptosomes. Finally, STOP knockout mice were hypersensitive to acute and subchronic locomotor effects of cocaine, although the drug equally inhibited DA uptake in mutant and wild-type mice. Altogether, these data showed that deletion of the ubiquitous STOP protein elicited restricted alterations in DAergic neurotransmission, preferentially in the meso-limbic pathway.
Collapse
Affiliation(s)
- Caroline Bouvrais-Veret
- Inserm, U513, Créteil, France, and Univ Paris 12, Faculté de Médecine Henri Mondor, Créteil, France
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Bégou M, Brun P, Bertrand JB, Job D, Schweitzer A, D'Amato T, Saoud M, Andrieux A, Suaud-Chagny MF. Post-pubertal emergence of alterations in locomotor activity in stop null mice. Synapse 2007; 61:689-97. [PMID: 17559095 DOI: 10.1002/syn.20409] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Overt schizophrenia is preceded by a prodromal phase during which juvenile patients display attenuated schizophrenia-related symptoms. Here, we have looked for evidence of a prodromal phase in juvenile STOP null mice, which, during adulthood, imitate features of schizophrenia. We have principally examined locomotor activity, which is abnormal in adult STOP null mice, and its apparent relationship with perturbed glutamatergic and dopaminergic transmission. When compared to corresponding wild-type mice, juvenile STOP null mice did not exhibit the basal hyperlocomotion or locomotor hypersensitivity to mild stress observed in adult mice. Juvenile STOP null mice also lacked disturbed locomotor sensitivity to MK-801, which was evident in adult mice. In contrast, juvenile STOP null mice exhibited a similar hypersensitivity to amphetamine as that found in adult mice. Thus, STOP null mice exhibited both a progression of locomotor activity defects over time and subtle alterations in the prepubertal period. We suggest that the pattern of locomotor disturbances observed in this study is related to altered dopaminergic reactivity in juvenile mice without major disturbance in glutamatergic transmission, whereas both neurotransmitter systems are impaired in adult mice.
Collapse
Affiliation(s)
- Mélina Bégou
- Institut Fédératif des Neurosciences de Lyon, Bron, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Bouvrais-Veret C, Weiss S, Andrieux A, Schweitzer A, McIntosh JM, Job D, Giros B, Martres MP. Sustained increase of alpha7 nicotinic receptors and choline-induced improvement of learning deficit in STOP knock-out mice. Neuropharmacology 2007; 52:1691-700. [PMID: 17512560 DOI: 10.1016/j.neuropharm.2007.03.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2006] [Revised: 03/16/2007] [Accepted: 03/30/2007] [Indexed: 11/16/2022]
Abstract
Mice deficient in the microtubule stabilizing protein STOP (stable tubule only polypeptide) show synaptic plasticity anomalies in hippocampus, dopamine hyper-reactivity in the limbic system and severe behavioral deficits. Some of these disturbances are alleviated by long-term antipsychotic treatment. Therefore, this mouse line represents a pertinent model for some aspects of schizophrenia symptomatology. Numerous data support dysfunction of nicotinic neurotransmission in schizophrenia and epidemiological studies show increased tobacco use in schizophrenic patients, in whom nicotine has been reported to improve cognitive deficits and impairment in sensory gating. In this study, we examined potential alterations in cholinergic (ACh) and nicotinic components and functions in STOP mutant mice. STOP KO mice displayed no variation of the density of ACh esterase and beta2* nicotinic receptors (nAChRs), large reductions in the density of vesicular ACh transporter and alpha6* nAChRs and marked increases in the density of alpha7 nAChRs, in some brain areas. STOP KO mice were hypersensitive to the stimulating locomotor effect of nicotine and, interestingly, their impaired performance in learning the cued version of the water maze were improved by administration of the preferential alpha7 nAChR agonist choline. Altogether, our data show that the deletion of the ubiquitous STOP protein elicited restricted alterations in ACh components. They also suggest that nicotinic neurotransmission can be deficient in STOP KO mice and that mutant mice can represent a meaningful model to study some nicotinic dysfunctions and therapeutic treatments.
Collapse
|
41
|
Rodriguez de la Vega M, Sevilla RG, Hermoso A, Lorenzo J, Tanco S, Diez A, Fricker LD, Bautista JM, Avilés FX. Nna1-like proteins are active metallocarboxypeptidases of a new and diverse M14 subfamily. FASEB J 2007; 21:851-65. [PMID: 17244817 DOI: 10.1096/fj.06-7330com] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Nna1 has some sequence similarity to metallocarboxypeptidases, but the biochemical characterization of Nna1 has not previously been reported. In this work we performed a detailed genomic scan and found >100 Nna1 homologues in bacteria, Protista, and Animalia, including several paralogs in most eukaryotic species. Phylogenetic analysis of the Nna1-like sequences demonstrates a major divergence between Nna1-like peptidases and the previously known metallocarboxypeptidases subfamilies: M14A, M14B, and M14C. Conformational modeling of representative Nna1-like proteins from a variety of species indicates an unusually open active site, a property that might facilitate its action on a wide variety of peptide and protein substrates. To test this, we expressed a recombinant form of one of the Nna1-like peptidases from Caenorhabditis elegans and demonstrated that this protein is a fully functional metallocarboxypeptidase that cleaves a range of C-terminal amino acids from synthetic peptides. The enzymatic activity is activated by ATP/ADP and salt-inactivated, and is preferentially inhibited by Z-Glu-Tyr dipeptide, which is without precedent in metallocarboxypeptidases and resembles tubulin carboxypeptidase functioning; this hypothesis is strongly reinforced by the results depicted in Kalinina et al. published as accompanying paper in this journal. Our findings demonstrate that the M14 family of metallocarboxypeptidases is more complex and diverse than expected, and that Nna1-like peptidases are functional variants of such enzymes, representing a novel subfamily (we propose the name M14D) that contributes substantially to such diversity.
Collapse
Affiliation(s)
- Monica Rodriguez de la Vega
- Institut de Biotecnologia i de Biomedicina, Universitat Autonoma de Barcelona, 08193 Bellaterra (Barcelona), Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Andrieux A, Salin P, Schweitzer A, Bégou M, Pachoud B, Brun P, Gory-Fauré S, Kujala P, Suaud-Chagny MF, Höfle G, Job D. Microtubule stabilizer ameliorates synaptic function and behavior in a mouse model for schizophrenia. Biol Psychiatry 2006; 60:1224-30. [PMID: 16806091 DOI: 10.1016/j.biopsych.2006.03.048] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2005] [Revised: 03/13/2006] [Accepted: 03/14/2006] [Indexed: 10/24/2022]
Abstract
BACKGROUND Recent data suggest that cytoskeletal defects may play a role in schizophrenia. We previously imitated features of schizophrenia in an animal model by disrupting gene coding for a microtubule-associated protein called STOP. STOP-null mice display synaptic defects in glutamatergic neurons, hyper-dopaminergy, and severe behavioral disorders. Synaptic and behavioral deficits are amended by neuroleptic treatment in STOP-null mice, providing an attractive model to test new antipsychotic agents. We examined the effects of a taxol-related microtubule stabilizer, epothilone D. METHODS Mice were treated either with vehicle alone or with epothilone D. Treatment effects on synaptic function were assessed using electron-microscopy quantification of synaptic vesicle pools and electrophysiology in the CA1 region of the hippocampus. Dopamine transmission was investigated using electrochemical assays. Behavior was principally assessed using tests of maternal skills. RESULTS In STOP-null mice, treatment with epothilone D increased synaptic vesicle pools, ameliorated both short- and long-term forms of synaptic plasticity in glutamatergic neurons, and had a dramatic beneficial effect on mouse behavior. CONCLUSIONS A microtubule stabilizer can have a beneficial effect on synaptic function and behavior, suggesting new possibilities for treatment of schizophrenia.
Collapse
Affiliation(s)
- Annie Andrieux
- Laboratoire du Cytosquelette, INSERM U366 CEA, Département Réponse et Dynamique Cellulaire, Grenoble, France.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Gory-Fauré S, Windscheid V, Bosc C, Peris L, Proietto D, Franck R, Denarier E, Job D, Andrieux A. STOP-like Protein 21 Is a Novel Member of the STOP Family, Revealing a Golgi Localization of STOP Proteins. J Biol Chem 2006; 281:28387-96. [PMID: 16837464 DOI: 10.1074/jbc.m603380200] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Neuronal microtubules are stabilized by two calmodulin-regulated microtubule-associated proteins, E-STOP and N-STOP, which when suppressed in mice induce severe synaptic and behavioral deficits. Here we show that mature neurons also contain a 21-kDa STOP-like protein, SL21, which shares calmodulin-binding and microtubule-stabilizing homology domains with STOP proteins. Accordingly, in different biochemical or cellular assays, SL21 has calmodulin binding and microtubule stabilizing activity. However, in cultured hippocampal neurons, SL21 antibodies principally stain the somatic Golgi and punctate Golgi material in neurites. In cycling cells, transfected SL21 decorates microtubules when expressed at high levels but is otherwise principally visible at the Golgi. The Golgi targeting of SL21 depends on the presence of cysteine residues located within the SL21 N-terminal domain, suggesting that Golgi targeting may require SL21 palmitoylation. Accordingly we find that SL21 is palmitoylated in vivo. N-STOP and E-STOP, which contain the Golgi targeting sequences present in SL21, also display distinct Golgi staining when expressed at low level in cycling cells. Thus neuronal proteins of the STOP family have the capacity to associate with Golgi material, which could be important for STOP synaptic functions.
Collapse
Affiliation(s)
- Sylvie Gory-Fauré
- INSERM U366, Département Recherche et Dynamique Cellulaire/Cytosquelette, Commissariat à l'Energie Atomique Grenoble, 17 rue des Martyrs, 38054 Grenoble, cedex 9, France
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Galiano MR, Andrieux A, Deloulme JC, Bosc C, Schweitzer A, Job D, Hallak ME. Myelin basic protein functions as a microtubule stabilizing protein in differentiated oligodendrocytes. J Neurosci Res 2006; 84:534-41. [PMID: 16773649 DOI: 10.1002/jnr.20960] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Myelin basic protein (MBP) is an oligodendrocyte-specific protein essential for oligodendrocyte morphogenesis at late stages of cell differentiation. There is evidence that the morphogenetic function of MBP is mediated by MBP interaction with the cytoskeleton. Thus, an MBP/cytoplasmic microtubule association has been reported, and MBP has Ca(2+)/calmodulin-regulated microtubule cold-stabilizing activity in vitro. However, the unambiguous demonstration of a microtubule-stabilizing activity for MBP in cells has been difficult because oligodendrocytes contain variants of STOP (stable tubule only polypeptide) proteins, which are responsible for microtubule cold stability in different cell types. Herein, we have used genetic mouse models and RNA interference to assay independently the microtubule cold-stabilizing activities of MBP and of STOP in developing oligodendrocytes. In wild-type oligodendrocytes, microtubules were cold stable throughout maturation, which is consistent with the presence of STOP proteins from early stages of differentiation. In contrast, in oligodendrocytes from STOP-deficient mice, microtubules were cold labile in the absence of MBP expression or when MBP expression was restricted to the cell body and became stable in fully differentiated oligodendrocytes, where MBP is expressed in cell extensions. The suppression of MBP by RNA interference in STOP-deficient oligodendrocytes suppressed microtubule cold stability. Additionally, STOP suppression in oligodendrocytes derived from shiverer mice that lack MBP led to the complete suppression of microtubule cold stability at all stages of cell differentiation. These results demonstrate that both STOP and MBP function as microtubule-stabilizing proteins in differentiating oligodendrocytes and could be important for the morphogenetic function of MBP.
Collapse
Affiliation(s)
- M R Galiano
- CIQUIBIC-Dpto. Química Biológica, Facultad Ciencias Químicas, Haya de la Torre S/N, Córdoba, Argentina
| | | | | | | | | | | | | |
Collapse
|
45
|
Baratier J, Peris L, Brocard J, Gory-Fauré S, Dufour F, Bosc C, Fourest-Lieuvin A, Blanchoin L, Salin P, Job D, Andrieux A. Phosphorylation of Microtubule-associated Protein STOP by Calmodulin Kinase II. J Biol Chem 2006; 281:19561-9. [PMID: 16651267 DOI: 10.1074/jbc.m509602200] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
STOP proteins are microtubule-associated, calmodulin-regulated proteins responsible for the high degree of stabilization displayed by neuronal microtubules. STOP suppression in mice induces synaptic defects affecting both short and long term synaptic plasticity in hippocampal neurons. Interestingly, STOP has been identified as a component of synaptic structures in neurons, despite the absence of microtubules in nerve terminals, indicating the existence of mechanisms able to induce a translocation of STOP from microtubules to synaptic compartments. Here we have tested STOP phosphorylation as a candidate mechanism for STOP relocalization. We show that, both in vitro and in vivo, STOP is phosphorylated by the multifunctional enzyme calcium/calmodulin-dependent protein kinase II (CaMKII), which is a key enzyme for synaptic plasticity. This phosphorylation occurs on at least two independent sites. Phosphorylated forms of STOP do not bind microtubules in vitro and do not co-localize with microtubules in cultured differentiating neurons. Instead, phosphorylated STOP co-localizes with actin assemblies along neurites or at branching points. Correlatively, we find that STOP binds to actin in vitro. Finally, in differentiated neurons, phosphorylated STOP co-localizes with clusters of synaptic proteins, whereas unphosphorylated STOP does not. Thus, STOP phosphorylation by CaMKII may promote STOP translocation from microtubules to synaptic compartments where it may interact with actin, which could be important for STOP function in synaptic plasticity.
Collapse
Affiliation(s)
- Julie Baratier
- Laboratoire du Cytosquelette, INSERM U366, DRDC/CS, CEA-Grenoble, 17 Rue des Martyrs, 38054 Grenoble cedex 9, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Vassal E, Barette C, Fonrose X, Dupont R, Sans-Soleilhac E, Lafanechère L. Miniaturization and validation of a sensitive multiparametric cell-based assay for the concomitant detection of microtubule-destabilizing and microtubule-stabilizing agents. ACTA ACUST UNITED AC 2006; 11:377-89. [PMID: 16751334 DOI: 10.1177/1087057106286210] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The authors describe a cell-based assay for anti-microtubule compounds suitable for automation. This assay allows the identification, in a single screening campaign, of both microtubule-destabilizing and microtubule-stabilizing agents. Its rationale is based on the substrate properties of the tubulin-modifying enzymes involved in the tubulin tyrosination cycle. This cycle involves the removal of the C-terminal tyrosine of the tubulin alpha-subunit by an ill-defined tubulin carboxypeptidase and its readdition by tubulin tyrosine ligase. Because of the substrate properties of these enzymes, dynamic microtubules, sensitive to depolymerizing drugs, are composed of tyrosinated tubulin, whereas non-dynamic, stabilized microtubules are composed of detyrosinated tubulin. Thus depolymerization or stabilization of the microtubule network can easily be detected with double-immunofluorescence staining using antibodies specific to tyrosinated and detyrosinated tubulin. The authors have scaled this assay to the 96-well plate format and adapted its process for an automated handling, including a readout using a microplate reader. They describe the different steps of this adaptation. This assay was validated using known compounds. This new cell-based assay represents an alternative to both global cytotoxicity assays and in vitro tubulin assembly assays commonly used for the detection of microtubule poisons.
Collapse
Affiliation(s)
- Emilie Vassal
- Université Joseph Fourier, INSERM U366-CS/DRDC/CEA Grenoble, France
| | | | | | | | | | | |
Collapse
|
47
|
Cottam DM, Tucker JB, Rogers-Bald MM, Mackie JB, Macintyre J, Scarborough JA, Ohkura H, Milner MJ. Non-centrosomal microtubule-organising centres in cold-treated cultured Drosophila cells. ACTA ACUST UNITED AC 2006; 63:88-100. [PMID: 16385467 DOI: 10.1002/cm.20103] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
In this paper we describe a new type of non-centrosomal microtubule-organising centre (MTOC), which is induced by cold treatment of certain cultured Drosophila cells and allows rapid reassembly of microtubule (MT) arrays. Prolonged cooling of two types of cultured Drosophila cells, muscle cells in primary culture and a wing imaginal disc cell line Cl.8+ results in disassembly of MT arrays and induces the formation of clusters of short MTs that have not been described before. Upon rewarming, the clusters are lost and the MT array is re-established within 1 h. In Cl.8+ cells, gamma-tubulin-containing centrosomes are detected, both in cell extensions and in the expected juxtanuclear position, and gamma-tubulin co-localises with the cold-induced MT clusters. The MT plus-end-binding protein, Drosophila EB1, decorates growing tips of MTs extending from clusters. We conclude that the cold-induced MT clusters represent acentrosomal MTOCs, allowing rapid reassembly of MT arrays following exposure to cold.
Collapse
Affiliation(s)
- Deborah M Cottam
- School of Biology, Bute Medical Building, University of St. Andrews, St. Andrews, Fife KY16 9TS, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Dougherty GW, Adler HJ, Rzadzinska A, Gimona M, Tomita Y, Lattig MC, Merritt RC, Kachar B. CLAMP, a novel microtubule-associated protein with EB-type calponin homology. CELL MOTILITY AND THE CYTOSKELETON 2005; 62:141-56. [PMID: 16206169 DOI: 10.1002/cm.20093] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Microtubules (MTs) are polymers of alpha and beta tubulin dimers that mediate many cellular functions, including the establishment and maintenance of cell shape. The dynamic properties of MTs may be influenced by tubulin isotype, posttranslational modifications of tubulin, and interaction with microtubule-associated proteins (MAPs). End-binding (EB) family proteins affect MT dynamics by stabilizing MTs, and are the only MAPs reported that bind MTs via a calponin-homology (CH) domain (J Biol Chem 278 (2003) 49721-49731; J Cell Biol 149 (2000) 761-766). Here, we describe a novel 27 kDa protein identified from an inner ear organ of Corti library. Structural homology modeling demonstrates a CH domain in this protein similar to EB proteins. Northern and Western blottings confirmed expression of this gene in other tissues, including brain, lung, and testis. In the organ of Corti, this protein localized throughout distinctively large and well-ordered MT bundles that support the elongated body of mechanically stiff pillar cells of the auditory sensory epithelium. When ectopically expressed in Cos-7 cells, this protein localized along cytoplasmic MTs, promoted MT bundling, and efficiently stabilized MTs against depolymerization in response to high concentration of nocodazole and cold temperature. We propose that this protein, designated CLAMP, is a novel MAP and represents a new member of the CH domain protein family.
Collapse
Affiliation(s)
- Gerard W Dougherty
- Section on Structural Cell Biology, NIDCD, NIH, Bethesda, Maryland 20892-8027, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Brun P, Bégou M, Andrieux A, Mouly-Badina L, Clerget M, Schweitzer A, Scarna H, Renaud B, Job D, Suaud-Chagny MF. Dopaminergic transmission in STOP null mice. J Neurochem 2005; 94:63-73. [PMID: 15953350 DOI: 10.1111/j.1471-4159.2005.03166.x] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Neuroleptics are thought to exert their anti-psychotic effects by counteracting a hyper-dopaminergic transmission. Here, we have examined the dopaminergic status of STOP (stable tubule only polypeptide) null mice, which lack a microtubule-stabilizing protein and which display neuroleptic-sensitive behavioural disorders. Dopamine transmission was investigated using both behavioural analysis and measurements of dopamine efflux in different conditions. Compared to wild-type mice in basal conditions or following mild stress, STOP null mice showed a hyper-locomotor activity, which was erased by neuroleptic treatment, and an increased locomotor reactivity to amphetamine. Such a behavioural profile is indicative of an increased dopaminergic transmission. In STOP null mice, the basal dopamine concentrations, measured by quantitative microdialysis, were normal in both the nucleus accumbens and the striatum. When measured by electrochemical techniques, the dopamine efflux evoked by electrical stimulations mimicking physiological stimuli was dramatically increased in the nucleus accumbens of STOP null mice, apparently due to an increased dopamine release, whereas dopaminergic uptake and auto-inhibition mechanisms were normal. In contrast, dopamine effluxes were slightly diminished in the striatum. Together with previous results, the present study indicates the association in STOP null mice of hippocampal hypo-glutamatergy and of limbic hyper-dopaminergy. Such neurotransmission defects are thought to be central to mental diseases such as schizophrenia.
Collapse
Affiliation(s)
- Philippe Brun
- Institut Fédératif des Neurosciences de Lyon; UCBL, Faculté de Pharmacie, Laboratoire de Neuropharmacologie et Neurochimie, Lyon, France.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Rong R, Jin W, Zhang J, Sheikh MS, Huang Y. Tumor suppressor RASSF1A is a microtubule-binding protein that stabilizes microtubules and induces G2/M arrest. Oncogene 2004; 23:8216-30. [PMID: 15378022 DOI: 10.1038/sj.onc.1207901] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
RASSF1A is a putative tumor suppressor gene that is inactivated in a variety of human tumors. Expression of exogenous RASSF1A has been shown to inhibit tumor growth in vitro and in animals. However, the molecular mechanisms by which RASSF1A mediates its tumor suppressive effects remain to be elucidated. Here, we report that RASSF1A is a microtubule-binding protein that interacts with and stabilizes microtubules. We have identified the RASSF1A region harboring a basic domain that appears to mediate the interactions between RASSF1A and microtubules. The basic domain-containing RASSF1C isoform also interacts with and stabilizes microtubules. We further show that in addition to G1 arrest, RASSF1A promotes growth arrest in the G2/M phase of the cell cycle and endogenous RASSF1A also interacts with microtubules. Based on our results, we propose that RASSF1A may mediate its tumor suppressive effects by inducing growth arrest in the G1 and G2/M phases. Together, these results provide important new insights into the molecular mechanisms by which this novel tumor suppressor mediates its biological effects.
Collapse
Affiliation(s)
- Rong Rong
- Department of Pharmacology, State University of New York, Upstate Medical University, 750 E Adams Street Syracuse, NY 13210, USA
| | | | | | | | | |
Collapse
|