Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
J Cell Sci ; 137(8)2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38525600

ABSTRACT

In neurons, the microtubule (MT) cytoskeleton forms the basis for long-distance protein transport from the cell body into and out of dendrites and axons. To maintain neuronal polarity, the axon initial segment (AIS) serves as a physical barrier, separating the axon from the somatodendritic compartment and acting as a filter for axonal cargo. Selective trafficking is further instructed by axonal enrichment of MT post-translational modifications, which affect MT dynamics and the activity of motor proteins. Here, we compared two knockout mouse lines lacking the respective enzymes for MT tyrosination and detyrosination, and found that both knockouts led to a shortening of the AIS. Neurons from both lines also showed an increased immobile fraction of endolysosomes present in the axon, whereas mobile organelles displayed shortened run distances in the retrograde direction. Overall, our results highlight the importance of maintaining the balance of tyrosinated and detyrosinated MTs for proper AIS length and axonal transport processes.


Subject(s)
Axonal Transport , Lysosomes , Mice, Knockout , Microtubules , Tyrosine , Animals , Microtubules/metabolism , Tyrosine/metabolism , Lysosomes/metabolism , Mice , Axons/metabolism , Endosomes/metabolism , Neurons/metabolism
2.
Semin Cell Dev Biol ; 137: 46-62, 2023 03 15.
Article in English | MEDLINE | ID: mdl-34924330

ABSTRACT

Among the variety of post-translational modifications to which microtubules are subjected, the detyrosination/re-tyrosination cycle is specific to tubulin. It is conserved by evolution and characterized by the enzymatic removal and re-addition of a gene-encoded tyrosine residue at the C-terminus of α-tubulin. Detyrosinated tubulin can be further converted to Δ2-tubulin by the removal of an additional C-terminal glutamate residue. Detyrosinated and Δ2-tubulin are carried by stable microtubules whereas tyrosinated microtubules are present on dynamic polymers. The cycle regulates trafficking of many cargo transporting molecular motors and is linked to the microtubule dynamics via regulation of microtubule interactions with specific cellular effectors such as kinesin-13. Here, we give an historical overview of the general features discovered for the cycle. We highlight the recent progress toward structure and functioning of the enzymes that keep the levels of tyrosinated and detyrosinated tubulin in cells, the long-known tubulin tyrosine ligase and the recently discovered vasohibin-SVBP complexes. We further describe how the cycle controls microtubule functions in healthy neurons and cardiomyocytes and how deregulations of the cycle are involved in dysfunctions of these highly differentiated cells, leading to neurodegeneration and heart failure in humans.


Subject(s)
Myocytes, Cardiac , Tubulin , Humans , Tubulin/metabolism , Myocytes, Cardiac/metabolism , Microtubules/metabolism , Neurons/metabolism , Tyrosine/chemistry , Tyrosine/metabolism , Protein Processing, Post-Translational , Carrier Proteins/metabolism
3.
Front Cell Dev Biol ; 10: 926914, 2022.
Article in English | MEDLINE | ID: mdl-36092705

ABSTRACT

Microtubules (MTs) support a variety of neuronal functions, such as maintenance of cell structure, transport, and synaptic plasticity. Neuronal MTs are highly heterogeneous due to several tubulin isotypes and the presence of multiple post-translational modifications, such as detyrosination and acetylation. The tubulin tyrosination/detyrosination cycle is a key player in the maintenance of MT dynamics, as tyrosinated tubulin is associated with more dynamic MTs, while detyrosinated tubulin is linked to longer lived, more stable MTs. Dysfunction of tubulin re-tyrosination was recently correlated to Alzheimer's disease progression. The implication of tubulin acetylation in Alzheimer's disease has, however, remained controversial. Here, we demonstrate that tubulin acetylation accumulates in post-mortem brain tissues from Alzheimer's disease patients and human neurons harboring the Alzheimer's familial APP-V717I mutation. We further show that tubulin re-tyrosination, which is defective in Alzheimer's disease, can control acetylated tubulin in primary neurons irrespective of the levels of the enzymes regulating tubulin acetylation, suggesting that reduced MT dynamics associated with impaired tubulin re-tyrosination might contribute to the accumulation of tubulin acetylation that we detected in Alzheimer's disease.

4.
Brain ; 145(7): 2486-2506, 2022 07 29.
Article in English | MEDLINE | ID: mdl-35148384

ABSTRACT

Microtubules play fundamental roles in the maintenance of neuronal processes and in synaptic function and plasticity. While dynamic microtubules are mainly composed of tyrosinated tubulin, long-lived microtubules contain detyrosinated tubulin, suggesting that the tubulin tyrosination/detyrosination cycle is a key player in the maintenance of microtubule dynamics and neuronal homeostasis, conditions that go awry in neurodegenerative diseases. In the tyrosination/detyrosination cycle, the C-terminal tyrosine of α-tubulin is removed by tubulin carboxypeptidases and re-added by tubulin tyrosine ligase (TTL). Here we show that TTL heterozygous mice exhibit decreased tyrosinated microtubules, reduced dendritic spine density and both synaptic plasticity and memory deficits. We further report decreased TTL expression in sporadic and familial Alzheimer's disease, and reduced microtubule dynamics in human neurons harbouring the familial APP-V717I mutation. Finally, we show that synapses visited by dynamic microtubules are more resistant to oligomeric amyloid-ß peptide toxicity and that expression of TTL, by restoring microtubule entry into spines, suppresses the loss of synapses induced by amyloid-ß peptide. Together, our results demonstrate that a balanced tyrosination/detyrosination tubulin cycle is necessary for the maintenance of synaptic plasticity, is protective against amyloid-ß peptide-induced synaptic damage and that this balance is lost in Alzheimer's disease, providing evidence that defective tubulin retyrosination may contribute to circuit dysfunction during neurodegeneration in Alzheimer's disease.


Subject(s)
Alzheimer Disease , Tubulin , Alzheimer Disease/metabolism , Animals , Humans , Mice , Microtubules , Peptides/metabolism , Tubulin/metabolism , Tyrosine/metabolism
5.
Front Pharmacol ; 12: 627995, 2021.
Article in English | MEDLINE | ID: mdl-33790791

ABSTRACT

The search for effective treatments for neuropsychiatric disorders is ongoing, with progress being made as brain structure and neuronal function become clearer. The central roles played by microtubules (MT) and actin in synaptic transmission and plasticity suggest that the cytoskeleton and its modulators could be relevant targets for the development of new molecules to treat psychiatric diseases. In this context, LIM Kinase - which regulates both the actin and MT cytoskeleton especially in dendritic spines, the post-synaptic compartment of the synapse - might be a good target. In this study, we analyzed the consequences of blocking LIMK1 pharmacologically using Pyr1. We investigated synaptic plasticity defects and behavioral disorders in MAP6 KO mice, an animal model useful for the study of psychiatric disorders, particularly schizophrenia. Our results show that Pyr1 can modulate MT dynamics in neurons. In MAP6 KO mice, chronic LIMK inhibition by long-term treatment with Pyr1 can restore normal dendritic spine density and also improves long-term potentiation, both of which are altered in these mice. Pyr1 treatment improved synaptic plasticity, and also reduced social withdrawal and depressive/anxiety-like behavior in MAP6 KO mice. Overall, the results of this study validate the hypothesis that modulation of LIMK activity could represent a new therapeutic strategy for neuropsychiatric diseases.

6.
Dev Neurobiol ; 81(3): 253-272, 2021 04.
Article in English | MEDLINE | ID: mdl-33325152

ABSTRACT

Microtubules (MTs) are an essential component of the neuronal cytoskeleton; they are involved in various aspects of neuron development, maintenance, and functions including polarization, synaptic plasticity, and transport. Neuronal MTs are highly heterogeneous due to the presence of multiple tubulin isotypes and extensive post-translational modifications (PTMs). These PTMs-most notably detyrosination, acetylation, and polyglutamylation-have emerged as important regulators of the neuronal microtubule cytoskeleton. With this review, we summarize what is currently known about the impact of tubulin PTMs on microtubule dynamics, neuronal differentiation, plasticity, and transport as well as on brain function in normal and pathological conditions, in particular during neuro-degeneration. The main therapeutic approaches to neuro-diseases based on the modulation of tubulin PTMs are also summarized. Overall, the review indicates how tubulin PTMs can generate a large number of functionally specialized microtubule sub-networks, each of which is crucial to specific neuronal features.


Subject(s)
Microtubules , Tubulin , Acetylation , Cytoskeleton/metabolism , Microtubules/metabolism , Protein Processing, Post-Translational , Tubulin/metabolism
7.
PLoS Biol ; 18(12): e3000621, 2020 12.
Article in English | MEDLINE | ID: mdl-33351792

ABSTRACT

Neurons extend long axons that require maintenance and are susceptible to degeneration. Long-term integrity of axons depends on intrinsic mechanisms including axonal transport and extrinsic support from adjacent glial cells. The mechanisms of support provided by myelinating oligodendrocytes to underlying axons are only partly understood. Oligodendrocytes release extracellular vesicles (EVs) with properties of exosomes, which upon delivery to neurons improve neuronal viability in vitro. Here, we show that oligodendroglial exosome secretion is impaired in 2 mouse mutants exhibiting secondary axonal degeneration due to oligodendrocyte-specific gene defects. Wild-type oligodendroglial exosomes support neurons by improving the metabolic state and promoting axonal transport in nutrient-deprived neurons. Mutant oligodendrocytes release fewer exosomes, which share a common signature of underrepresented proteins. Notably, mutant exosomes lack the ability to support nutrient-deprived neurons and to promote axonal transport. Together, these findings indicate that glia-to-neuron exosome transfer promotes neuronal long-term maintenance by facilitating axonal transport, providing a novel mechanistic link between myelin diseases and secondary loss of axonal integrity.


Subject(s)
Axonal Transport/physiology , Neurons/metabolism , Oligodendroglia/metabolism , Animals , Axonal Transport/genetics , Axons/physiology , Exosomes/metabolism , Exosomes/physiology , Extracellular Vesicles/metabolism , Extracellular Vesicles/physiology , Female , HEK293 Cells , Humans , Maintenance , Male , Mice , Mice, Inbred C57BL , Myelin Sheath/metabolism , Neuroglia , Neurons/physiology , Oligodendroglia/physiology , Signal Transduction/physiology
8.
Front Pharmacol ; 11: 543, 2020.
Article in English | MEDLINE | ID: mdl-32425788

ABSTRACT

Agents able to modify microtubule dynamics are important anticancer drugs. The absence of microtubules resulting from drug-induced depolymerization is easy to detect. However the detection of a stabilized microtubule network needs specific assays since there is not a significant visual difference between normal and stabilized microtubule networks. Here, we describe a quantitative cell-based assay, suitable for automation, which allows the detection of stabilized microtubules without the need of microscopic examination. The rationale of this assay is based on the drug-induced resistance of the microtubule network to the depolymerizing agent combretastatin A4 and the subsequent detection of the residual microtubules by immunoluminescence. Using this assay to screen a kinase inhibitor library allowed the selection of seven known kinase inhibitors: selonsertib, masatinib, intedanib, PF0477736, SNS-314 mesylate, MPI0479605, and ponatinib. The yet undescribed ability of these inhibitors to stabilize cellular microtubules was confirmed using additional markers of stable microtubules and time-lapse video-microscopy to track individual microtubules in living cells. None of the compounds interacted, however, directly with tubulin. By employing other inhibitors of the same kinases, which have structurally unrelated scaffolds, we determined if the microtubule stabilizing effect was due to the inhibition of the targeted kinase, or to an off-target effect. Many of these inhibitors are clinically approved or currently assayed in phase 2 or phase 3 clinical trials. Their microtubule-stabilizing effect may account for their therapeutic effect as well as for some of their adverse side effects. These results indicate also a possible repurposing of some of these drugs.

9.
Hum Mol Genet ; 28(20): 3391-3405, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31363758

ABSTRACT

Reversible detyrosination of tubulin, the building block of microtubules, is crucial for neuronal physiology. Enzymes responsible for detyrosination were recently identified as complexes of vasohibins (VASHs) one or two with small VASH-binding protein (SVBP). Here we report three consanguineous families, each containing multiple individuals with biallelic inactivation of SVBP caused by truncating variants (p.Q28* and p.K13Nfs*18). Affected individuals show brain abnormalities with microcephaly, intellectual disability and delayed gross motor and speech development. Immunoblot testing in cells with pathogenic SVBP variants demonstrated that the encoded proteins were unstable and non-functional, resulting in a complete loss of VASH detyrosination activity. Svbp knockout mice exhibit drastic accumulation of tyrosinated tubulin and a reduction of detyrosinated tubulin in brain tissue. Similar alterations in tubulin tyrosination levels were observed in cultured neurons and associated with defects in axonal differentiation and architecture. Morphological analysis of the Svbp knockout mouse brains by anatomical magnetic resonance imaging showed a broad impact of SVBP loss, with a 7% brain volume decrease, numerous structural defects and a 30% reduction of some white matter tracts. Svbp knockout mice display behavioural defects, including mild hyperactivity, lower anxiety and impaired social behaviour. They do not, however, show prominent memory defects. Thus, SVBP-deficient mice recapitulate several features observed in human patients. Altogether, our data demonstrate that deleterious variants in SVBP cause this neurodevelopmental pathology, by leading to a major change in brain tubulin tyrosination and alteration of microtubule dynamics and neuron physiology.


Subject(s)
Brain/abnormalities , Brain/metabolism , Cell Cycle Proteins/metabolism , Neurons/metabolism , Tubulin/metabolism , Animals , Carrier Proteins/metabolism , Cognitive Dysfunction/genetics , Cognitive Dysfunction/metabolism , Female , Humans , Immunoblotting , Magnetic Resonance Imaging , Mice , Microcephaly/genetics , Microcephaly/metabolism , Tyrosine/metabolism
10.
Nat Struct Mol Biol ; 26(7): 571-582, 2019 07.
Article in English | MEDLINE | ID: mdl-31235911

ABSTRACT

Vasohibins are tubulin tyrosine carboxypeptidases that are important in neuron physiology. We examined the crystal structures of human vasohibin 1 and 2 in complex with small vasohibin-binding protein (SVBP) in the absence and presence of different inhibitors and a C-terminal α-tubulin peptide. In combination with functional data, we propose that SVBP acts as an activator of vasohibins. An extended groove and a distinctive surface residue patch of vasohibins define the specific determinants for recognizing and cleaving the C-terminal tyrosine of α-tubulin and for binding microtubules, respectively. The vasohibin-SVBP interaction and the ability of the enzyme complex to associate with microtubules regulate axon specification of neurons. Our results define the structural basis of tubulin detyrosination by vasohibins and show the relevance of this process for neuronal development. Our findings offer a unique platform for developing drugs against human conditions with abnormal tubulin tyrosination levels, such as cancer, heart defects and possibly brain disorders.


Subject(s)
Angiogenic Proteins/metabolism , Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Tubulin/metabolism , Angiogenic Proteins/chemistry , Animals , Carrier Proteins/chemistry , Cell Cycle Proteins/chemistry , Cells, Cultured , Crystallography, X-Ray , HEK293 Cells , Humans , Mice , Models, Molecular , Protein Conformation , Protein Interaction Maps , Tubulin/chemistry
11.
Nat Commun ; 9(1): 3775, 2018 09 17.
Article in English | MEDLINE | ID: mdl-30224655

ABSTRACT

Emerging evidence indicates that microtubule-associated proteins (MAPs) are implicated in synaptic function; in particular, mice deficient for MAP6 exhibit striking deficits in plasticity and cognition. How MAP6 connects to plasticity mechanisms is unclear. Here, we address the possible role of this protein in dendritic spines. We find that in MAP6-deficient cortical and hippocampal neurons, maintenance of mature spines is impaired, and can be restored by expressing a stretch of the MAP6 sequence called Mc modules. Mc modules directly bind actin filaments and mediate activity-dependent stabilisation of F-actin in dendritic spines, a key event of synaptic plasticity. In vitro, Mc modules enhance actin filament nucleation and promote the formation of stable, highly ordered filament bundles. Activity-induced phosphorylation of MAP6 likely controls its transfer to the spine cytoskeleton. These results provide a molecular explanation for the role of MAP6 in cognition, enlightening the connection between cytoskeletal dysfunction, synaptic impairment and neuropsychiatric illnesses.


Subject(s)
Actin Cytoskeleton/metabolism , Dendrites/metabolism , Microtubule-Associated Proteins/metabolism , Neurons/cytology , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cells, Cultured , Fluorescence Resonance Energy Transfer , Hippocampus/cytology , Humans , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microtubule-Associated Proteins/genetics , Microtubules/metabolism , Neurons/metabolism , Phosphorylation , Photobleaching
12.
J Cell Biol ; 217(5): 1719-1738, 2018 05 07.
Article in English | MEDLINE | ID: mdl-29535193

ABSTRACT

During neural circuit assembly, extrinsic signals are integrated into changes in growth cone (GC) cytoskeleton underlying axon guidance decisions. Microtubules (MTs) were shown to play an instructive role in GC steering. However, the numerous actors required for MT remodeling during axon navigation and their precise mode of action are far from being deciphered. Using loss- and gain-of-function analyses during zebrafish development, we identify in this study the meiotic clade adenosine triphosphatase Fidgetin-like 1 (Fignl1) as a key GC-enriched MT-interacting protein in motor circuit wiring and larval locomotion. We show that Fignl1 controls GC morphology and behavior at intermediate targets by regulating MT plus end dynamics and growth directionality. We further reveal that alternative translation of Fignl1 transcript is a sophisticated mechanism modulating MT dynamics: a full-length isoform regulates MT plus end-tracking protein binding at plus ends, whereas shorter isoforms promote their depolymerization beneath the cell cortex. Our study thus pinpoints Fignl1 as a multifaceted key player in MT remodeling underlying motor circuit connectivity.


Subject(s)
Adenosine Triphosphatases/metabolism , Axon Guidance , Axons/metabolism , Microtubules/metabolism , Nuclear Proteins/metabolism , Adenosine Triphosphatases/chemistry , Animals , Cytoskeleton/metabolism , Gene Knockdown Techniques , Growth Cones/metabolism , Humans , Larva/metabolism , Locomotion , Microtubule-Associated Proteins/metabolism , Motor Neurons/metabolism , Nuclear Proteins/chemistry , Polymerization , Protein Isoforms/metabolism , Spinal Cord/metabolism
13.
Sci Rep ; 8(1): 3072, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29449678

ABSTRACT

The cerebellum plays a key role in motor tasks, but its involvement in cognition is still being considered. Although there is an association of different psychiatric and cognitive disorders with cerebellar impairments, the lack of time-course studies has hindered the understanding of the involvement of cerebellum in cognitive and non-motor functions. Such association was here studied using the Purkinje Cell Degeneration mutant mouse, a model of selective and progressive cerebellar degeneration that lacks the cytosolic carboxypeptidase 1 (CCP1). The effects of the absence of this enzyme on the cerebellum of mutant mice were analyzed both in vitro and in vivo. These analyses were carried out longitudinally (throughout both the pre-neurodegenerative and neurodegenerative stages) and different motor and non-motor tests were performed. We demonstrate that the lack of CCP1 affects microtubule dynamics and flexibility, defects that contribute to the morphological alterations of the Purkinje cells (PCs), and to progressive cerebellar breakdown. Moreover, this degeneration led not only to motor defects but also to gradual cognitive impairments, directly related to the progression of cellular damage. Our findings confirm the cerebellar implication in non-motor tasks, where the formation of the healthy, typical PCs structure is necessary for normal cognitive and affective behavior.


Subject(s)
GTP-Binding Proteins/physiology , Microtubules/physiology , Purkinje Cells/metabolism , Serine-Type D-Ala-D-Ala Carboxypeptidase/physiology , Animals , Cerebellum/metabolism , Cerebellum/physiology , Cognition/physiology , Cognition Disorders/metabolism , Cytoskeleton/metabolism , Cytoskeleton/physiology , Female , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Longitudinal Studies , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microtubules/metabolism , Motor Disorders/genetics , Purkinje Cells/physiology , Serine-Type D-Ala-D-Ala Carboxypeptidase/genetics , Serine-Type D-Ala-D-Ala Carboxypeptidase/metabolism
15.
Eur J Neurosci ; 46(11): 2754-2767, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29094416

ABSTRACT

MAP6 proteins were first described as microtubule-stabilizing agents, whose properties were thought to be essential for neuronal development and maintenance of complex neuronal networks. However, deletion of all MAP6 isoforms in MAP6 KO mice does not lead to dramatic morphological aberrations of the brain but rather to alterations in multiple neurotransmissions and severe behavioural impairments. A search for protein partners of MAP6 proteins identified Tctex1 - a dynein light chain with multiple non-microtubule-related functions. The involvement of Tctex1 in calcium signalling led to investigate it in MAP6 KO neurons. In this study, we show that functional Cav 2.2/N-type calcium channels are deficient in MAP6 KO neurons, due to improper location. We also show that MAP6 proteins interact directly with both Tctex1 and the C-terminus of Cav 2.2/N-type calcium channels. A balance of these two interactions seems to be crucial for MAP6 to modulate calcium signalling in neurons.


Subject(s)
Calcium Channels, N-Type/metabolism , Calcium Signaling/physiology , Dyneins/metabolism , Microtubule-Associated Proteins/metabolism , Neurons/metabolism , Animals , Binding Sites , Cells, Cultured , Female , Hippocampus/cytology , Mice , Mice, Knockout , Microtubule-Associated Proteins/genetics , Protein Binding
16.
Science ; 358(6369): 1448-1453, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29146868

ABSTRACT

Reversible detyrosination of α-tubulin is crucial to microtubule dynamics and functions, and defects have been implicated in cancer, brain disorganization, and cardiomyopathies. The identity of the tubulin tyrosine carboxypeptidase (TCP) responsible for detyrosination has remained unclear. We used chemical proteomics with a potent irreversible inhibitor to show that the major brain TCP is a complex of vasohibin-1 (VASH1) with the small vasohibin binding protein (SVBP). VASH1 and its homolog VASH2, when complexed with SVBP, exhibited robust and specific Tyr/Phe carboxypeptidase activity on microtubules. Knockdown of vasohibins or SVBP and/or inhibitor addition in cultured neurons reduced detyrosinated α-tubulin levels and caused severe differentiation defects. Furthermore, knockdown of vasohibins disrupted neuronal migration in developing mouse neocortex. Thus, vasohibin/SVBP complexes represent long-sought TCP enzymes.


Subject(s)
Angiogenic Proteins/metabolism , Carboxypeptidases/metabolism , Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Neurogenesis , Neurons/cytology , Tyrosine/metabolism , Angiogenic Proteins/genetics , Animals , Carboxypeptidases/genetics , Carrier Proteins/genetics , Cell Cycle Proteins/genetics , Cell Movement , Female , Gene Knockdown Techniques , HEK293 Cells , Humans , Male , Mice , Neocortex/cytology , Neocortex/embryology , Neurons/enzymology , Proteomics , Tubulin/metabolism
17.
Mol Biol Cell ; 27(19): 2924-34, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27466319

ABSTRACT

Proper regulation of microtubule dynamics is essential for cell functions and involves various microtubule-associated proteins (MAPs). Among them, end-binding proteins (EBs) accumulate at microtubule plus ends, whereas structural MAPs bind along the microtubule lattice. Recent data indicate that the structural MAP tau modulates EB subcellular localization in neurons. However, the molecular determinants of EB/tau interaction remain unknown, as is the effect of this interplay on microtubule dynamics. Here we investigate the mechanisms governing EB/tau interaction in cell-free systems and cellular models. We find that tau inhibits EB tracking at microtubule ends. Tau and EBs form a complex via the C-terminal region of EBs and the microtubule-binding sites of tau. These two domains are required for the inhibitory activity of tau on EB localization to microtubule ends. Moreover, the phosphomimetic mutation S262E within tau microtubule-binding sites impairs EB/tau interaction and prevents the inhibitory effect of tau on EB comets. We further show that microtubule dynamic parameters vary, depending on the combined activities of EBs and tau proteins. Overall our results demonstrate that tau directly antagonizes EB function through a phosphorylation-dependent mechanism. This study highlights a novel role for tau in EB regulation, which might be impaired in neurodegenerative disorders.


Subject(s)
Microtubule-Associated Proteins/metabolism , tau Proteins/genetics , tau Proteins/metabolism , Cell-Free System/metabolism , Humans , Microtubule-Associated Proteins/antagonists & inhibitors , Microtubules/metabolism , Neurons/metabolism , Phosphorylation , Protein Binding , Protein Domains , Protein Transport
18.
Mol Biol Cell ; 27(4): 640-53, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26739754

ABSTRACT

Cellular α-tubulin can bear various carboxy-terminal sequences: full-length tubulin arising from gene neosynthesis is tyrosinated, and two truncated variants, corresponding to detyrosinated and Δ2 α­tubulin, result from the sequential cleavage of one or two C-terminal residues, respectively. Here, by using a novel antibody named 3EG that is highly specific to the -EEEG C-terminal sequence, we demonstrate the occurrence in neuronal tissues of a new αΔ3­tubulin variant corresponding to α1A/B­tubulin deleted of its last three residues (EEY). αΔ3­tubulin has a specific distribution pattern: its quantity in the brain is similar to that of αΔ2-tubulin around birth but is much lower in adult tissue. This truncated α1A/B-tubulin variant can be generated from αΔ2-tubulin by the deglutamylases CCP1, CCP4, CCP5, and CCP6 but not by CCP2 and CCP3. Moreover, using 3EG antibody, we identify a C­terminally truncated ß-tubulin form with the same -EEEG C-terminal sequence. Using mass spectrometry, we demonstrate that ß2A/B-tubulin is modified by truncation of the four C-terminal residues (EDEA). We show that this newly identified ßΔ4-tubulin is ubiquitously present in cells and tissues and that its level is constant throughout the cell cycle. These new C-terminally truncated α- and ß-tubulin variants, both ending with -EEEG sequence, are expected to regulate microtubule physiology. Of interest, the αΔ3-tubulin seems to be related to dynamic microtubules, resembling tyrosinated-tubulin rather than the other truncated variants, and may have critical function(s) in neuronal development.


Subject(s)
Brain/metabolism , Carboxypeptidases/metabolism , Neurons/metabolism , Protein Processing, Post-Translational , Tubulin/metabolism , Amino Acid Sequence , Animals , Brain/cytology , Cell Cycle , Gene Knockdown Techniques , HEK293 Cells , HeLa Cells , Humans , Mass Spectrometry , Mice , Microtubules/metabolism , Molecular Sequence Data , Neurogenesis , Neurons/physiology , Peptide Synthases/genetics , Peptide Synthases/metabolism , Tyrosine/metabolism
19.
Dis Model Mech ; 6(1): 72-83, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22773755

ABSTRACT

Mutations in SPG4, encoding the microtubule-severing protein spastin, are responsible for the most frequent form of hereditary spastic paraplegia (HSP), a heterogeneous group of genetic diseases characterized by degeneration of the corticospinal tracts. We previously reported that mice harboring a deletion in Spg4, generating a premature stop codon, develop progressive axonal degeneration characterized by focal axonal swellings associated with impaired axonal transport. To further characterize the molecular and cellular mechanisms underlying this mutant phenotype, we have assessed microtubule dynamics and axonal transport in primary cultures of cortical neurons from spastin-mutant mice. We show an early and marked impairment of microtubule dynamics all along the axons of spastin-deficient cortical neurons, which is likely to be responsible for the occurrence of axonal swellings and cargo stalling. Our analysis also reveals that a modulation of microtubule dynamics by microtubule-targeting drugs rescues the mutant phenotype of cortical neurons. Together, these results contribute to a better understanding of the pathogenesis of SPG4-linked HSP and ascertain the influence of microtubule-targeted drugs on the early axonal phenotype in a mouse model of the disease.


Subject(s)
Adenosine Triphosphatases/deficiency , Adenosine Triphosphatases/genetics , Animals , Axonal Transport , Axons/drug effects , Axons/pathology , Cells, Cultured , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Disease Models, Animal , Hippocampus/metabolism , Hippocampus/pathology , Humans , Mice , Mice, Knockout , Microtubules/drug effects , Microtubules/metabolism , Models, Neurological , Mutation , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Nocodazole/pharmacology , Paclitaxel/pharmacology , Spastic Paraplegia, Hereditary/drug therapy , Spastic Paraplegia, Hereditary/genetics , Spastic Paraplegia, Hereditary/metabolism , Spastic Paraplegia, Hereditary/pathology , Spastin , Vinblastine/pharmacology
20.
Cell ; 143(4): 564-78, 2010 Nov 12.
Article in English | MEDLINE | ID: mdl-21074048

ABSTRACT

Polyglutamylation is a posttranslational modification that generates glutamate side chains on tubulins and other proteins. Although this modification has been shown to be reversible, little is known about the enzymes catalyzing deglutamylation. Here we describe the enzymatic mechanism of protein deglutamylation by members of the cytosolic carboxypeptidase (CCP) family. Three enzymes (CCP1, CCP4, and CCP6) catalyze the shortening of polyglutamate chains and a fourth (CCP5) specifically removes the branching point glutamates. In addition, CCP1, CCP4, and CCP6 also remove gene-encoded glutamates from the carboxyl termini of proteins. Accordingly, we show that these enzymes convert detyrosinated tubulin into Δ2-tubulin and also modify other substrates, including myosin light chain kinase 1. We further analyze Purkinje cell degeneration (pcd) mice that lack functional CCP1 and show that microtubule hyperglutamylation is directly linked to neurodegeneration. Taken together, our results reveal that controlling the length of the polyglutamate side chains on tubulin is critical for neuronal survival.


Subject(s)
Carboxypeptidases/metabolism , GTP-Binding Proteins/metabolism , Nerve Degeneration/metabolism , Polyglutamic Acid/metabolism , Serine-Type D-Ala-D-Ala Carboxypeptidase/metabolism , Amino Acid Sequence , Animals , Cell Line , Cell Survival , Cerebellum/pathology , Humans , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Olfactory Bulb/pathology , Sequence Alignment , Tubulin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...