Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Methods Mol Biol ; 2431: 325-350, 2022.
Article in English | MEDLINE | ID: mdl-35412285

ABSTRACT

Axonal transport is crucial for neuronal homeostasis, survival, and development. Indeed, axonal transport needs to be precisely regulated for developing axons to swiftly and accurately respond to their complex and evolving environment in space and time. A growing number of studies have started to unravel the diversity of regulatory and adaptor proteins required to orchestrate the axonal transport machinery. Despite some discrepancies between in vitro and in vivo axonal transport studies, most analyses aiming at deciphering these regulatory complexes, as well as their mode of action, were carried out in vitro in primary cultures of neurons, and mainly focused on their impact on axon specification and elongation, but rarely on axon navigation per se. Given the clear influence of the in vivo environment on axonal transport, including chemical and physical interactions with neighboring cells, it is essential to develop in vivo models to identify and characterize the molecular complexes involved in this key process. Here, we describe an experimental system to monitor axonal transport in vivo in developing axons of live zebrafish embryos with high spatial and temporal resolution. Due to its optical transparency and easy genetic manipulation, the zebrafish embryo is ideally suited to study such cellular dynamics at a single axon scale. Using this approach, we were able to unravel the key role of Fidgetin-like 1 in the regulation of bidirectional axonal transport required for motor axon targeting. Moreover, this protocol can be easily adapted to characterize a wide range of axonal transport regulators and components in physiological conditions and may additionally be used to screen new therapeutic compounds based on their ability to recue axonal transport defects in pathological conditions.


Subject(s)
Axonal Transport , Zebrafish , Animals , Axonal Transport/physiology , Axons/metabolism , Neurons/metabolism , Zebrafish/genetics , Zebrafish Proteins/metabolism
2.
J Cell Biol ; 218(10): 3290-3306, 2019 10 07.
Article in English | MEDLINE | ID: mdl-31541015

ABSTRACT

Neuronal connectivity relies on molecular motor-based axonal transport of diverse cargoes. Yet the precise players and regulatory mechanisms orchestrating such trafficking events remain largely unknown. We here report the ATPase Fignl1 as a novel regulator of bidirectional transport during axon navigation. Using a yeast two-hybrid screen and coimmunoprecipitation assays, we showed that Fignl1 binds the kinesin Kif1bß and the dynein/dynactin adaptor Bicaudal D-1 (Bicd1) in a molecular complex including the dynactin subunit dynactin 1. Fignl1 colocalized with Kif1bß and showed bidirectional mobility in zebrafish axons. Notably, Kif1bß and Fignl1 loss of function similarly altered zebrafish motor axon pathfinding and increased dynein-based transport velocity of Rab3 vesicles in these navigating axons, pinpointing Fignl1/Kif1bß as a dynein speed limiter complex. Accordingly, disrupting dynein/dynactin activity or Bicd1/Fignl1 interaction induced motor axon pathfinding defects characteristic of Fignl1 gain or loss of function, respectively. Finally, pharmacological inhibition of dynein activity partially rescued the axon pathfinding defects of Fignl1-depleted larvae. Together, our results identify Fignl1 as a key dynein regulator required for motor circuit wiring.


Subject(s)
ATPases Associated with Diverse Cellular Activities/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Axons/metabolism , Cytoskeletal Proteins/metabolism , Dyneins/metabolism , Kinesins/metabolism , Microtubule-Associated Proteins/metabolism , Nuclear Proteins/metabolism , Animals , Biological Transport , COS Cells , Cells, Cultured , Chlorocebus aethiops , Humans , Zebrafish
3.
Development ; 145(17)2018 09 12.
Article in English | MEDLINE | ID: mdl-30082270

ABSTRACT

Functional analyses of genes responsible for neurodegenerative disorders have unveiled crucial links between neurodegenerative processes and key developmental signalling pathways. Mutations in SPG4-encoding spastin cause hereditary spastic paraplegia (HSP). Spastin is involved in diverse cellular processes that couple microtubule severing to membrane remodelling. Two main spastin isoforms are synthesised from alternative translational start sites (M1 and M87). However, their specific roles in neuronal development and homeostasis remain largely unknown. To selectively unravel their neuronal function, we blocked spastin synthesis from each initiation codon during zebrafish development and performed rescue analyses. The knockdown of each isoform led to different motor neuron and locomotion defects, which were not rescued by the selective expression of the other isoform. Notably, both morphant neuronal phenotypes were observed in a CRISPR/Cas9 spastin mutant. We next showed that M1 spastin, together with HSP proteins atlastin 1 and NIPA1, drives motor axon targeting by repressing BMP signalling, whereas M87 spastin acts downstream of neuropilin 1 to control motor neuron migration. Our data therefore suggest that defective BMP and neuropilin 1 signalling may contribute to the motor phenotype in a vertebrate model of spastin depletion.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Motor Neurons/cytology , Neuropilin-1/metabolism , Spastin/genetics , Zebrafish Proteins/genetics , Zebrafish/embryology , Animals , Axons/metabolism , COS Cells , CRISPR-Cas Systems/genetics , Cell Line , Cell Movement/genetics , Chlorocebus aethiops , GTP-Binding Proteins/metabolism , Gene Knockout Techniques , Humans , Membrane Proteins/metabolism , Protein Isoforms/genetics , Spastic Paraplegia, Hereditary/genetics , Spastin/biosynthesis , Zebrafish Proteins/biosynthesis
4.
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
5.
J Neurosci ; 36(16): 4421-33, 2016 Apr 20.
Article in English | MEDLINE | ID: mdl-27098687

ABSTRACT

The axon initial segment (AIS) is required for generating action potentials and maintaining neuronal polarity. Significant progress has been made in deciphering the basic building blocks composing the AIS, but the underlying mechanisms required for AIS formation remains unclear. The scaffolding protein ankyrin-G is the master-organizer of the AIS. Microtubules and their interactors, particularly end-binding proteins (EBs), have emerged as potential key players in AIS formation. Here, we show that the longest isoform of ankyrin-G (480AnkG) selectively associates with EBs via its specific tail domain and that this interaction is crucial for AIS formation and neuronal polarity in cultured rodent hippocampal neurons. EBs are essential for 480AnkG localization and stabilization at the AIS, whereas 480AnkG is required for the specific accumulation of EBs in the proximal axon. Our findings thus provide a conceptual framework for understanding how the cooperative relationship between 480AnkG and EBs induces the assembly of microtubule-AIS structures in the proximal axon. SIGNIFICANCE STATEMENT: Neuronal polarity is crucial for the proper function of neurons. The assembly of the axon initial segment (AIS), which is the hallmark of early neuronal polarization, relies on the longest 480 kDa ankyrin-G isoform. The microtubule cytoskeleton and its interacting proteins were suggested to be early key players in the process of AIS formation. In this study, we show that the crosstalk between 480 kDa ankyrin-G and the microtubule plus-end tracking proteins, EBs, at the proximal axon is decisive for AIS assembly and neuronal polarity. Our work thus provides insight into the functional mechanisms used by 480 kDa ankyrin-G to drive the AIS formation and thereby to establish neuronal polarity.


Subject(s)
Ankyrins/metabolism , Axons/metabolism , Microtubule-Associated Proteins/metabolism , Animals , Axons/ultrastructure , COS Cells , Cell Polarity/physiology , Cells, Cultured , Chlorocebus aethiops , Female , Mice , Neurons/metabolism , Neurons/ultrastructure , Protein Binding/physiology , Rats , Rats, Sprague-Dawley
6.
J Cell Biol ; 202(3): 527-43, 2013 Aug 05.
Article in English | MEDLINE | ID: mdl-23897888

ABSTRACT

Mechanisms coordinating endosomal degradation and recycling are poorly understood, as are the cellular roles of microtubule (MT) severing. We show that cells lacking the MT-severing protein spastin had increased tubulation of and defective receptor sorting through endosomal tubular recycling compartments. Spastin required the ability to sever MTs and to interact with ESCRT-III (a complex controlling cargo degradation) proteins to regulate endosomal tubulation. Cells lacking IST1 (increased sodium tolerance 1), an endosomal sorting complex required for transport (ESCRT) component to which spastin binds, also had increased endosomal tubulation. Our results suggest that inclusion of IST1 into the ESCRT complex allows recruitment of spastin to promote fission of recycling tubules from the endosome. Thus, we reveal a novel cellular role for MT severing and identify a mechanism by which endosomal recycling can be coordinated with the degradative machinery. Spastin is mutated in the axonopathy hereditary spastic paraplegia. Zebrafish spinal motor axons depleted of spastin or IST1 also had abnormal endosomal tubulation, so we propose this phenotype is important for axonal degeneration.


Subject(s)
Adenosine Triphosphatases/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Endosomes/metabolism , Microtubules/metabolism , Oncogene Proteins/metabolism , Adenosine Triphosphatases/chemistry , Animals , Cells, Cultured , Endosomal Sorting Complexes Required for Transport/chemistry , HeLa Cells , Humans , Oncogene Proteins/chemistry , Spastin , Zebrafish
7.
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
8.
Nat Neurosci ; 13(11): 1380-7, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20935645

ABSTRACT

To better understand hereditary spastic paraplegia (HSP), we characterized the function of atlastin, a protein that is frequently involved in juvenile forms of HSP, by analyzing loss- and gain-of-function phenotypes in the developing zebrafish. We found that knockdown of the gene for atlastin (atl1) caused a severe decrease in larval mobility that was preceded by abnormal architecture of spinal motor axons and was associated with a substantial upregulation of the bone morphogenetic protein (BMP) signaling pathway. Overexpression analyses confirmed that atlastin inhibits BMP signaling. In primary cultures of zebrafish spinal neurons, Atlastin partially colocalized with type I BMP receptors in late endosomes distributed along neurites, which suggests that atlastin may regulate BMP receptor trafficking. Finally, genetic or pharmacological inhibition of BMP signaling was sufficient to rescue the loss of mobility and spinal motor axon defects of atl1 morphants, emphasizing the importance of fine-tuning the balance of BMP signaling for vertebrate motor axon architecture and stability.


Subject(s)
Axons/physiology , Bone Morphogenetic Proteins/metabolism , Cell Movement/physiology , Motor Neurons/cytology , Signal Transduction/physiology , Spinal Cord/cytology , Animals , Animals, Genetically Modified , Behavior, Animal , Bone Morphogenetic Proteins/genetics , Cells, Cultured , Embryo, Nonmammalian , Endosomes/metabolism , Gene Expression Regulation, Developmental/genetics , Glycoproteins/genetics , Green Fluorescent Proteins/genetics , Intercellular Signaling Peptides and Proteins/genetics , Larva , RNA, Messenger/physiology , Signal Transduction/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Tubulin/metabolism , Zebrafish , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
9.
Am J Med Genet B Neuropsychiatr Genet ; 144B(7): 854-61, 2007 Oct 05.
Article in English | MEDLINE | ID: mdl-17503452

ABSTRACT

Thirty-three different loci for hereditary spastic paraplegias (HSP) have been mapped, and 15 responsible genes have been identified. Autosomal recessive spastic paraplegias (ARHSPs) usually have clinically complex phenotypes but the SPG5, SPG24, and SPG28 loci are considered to be associated with pure forms of the disease. We performed a genome-wide scan in a large French family. Fine mapping of the refined SPG5 region on chromosome 8q12 was performed in another 17 ARHSP families with additional microsatellite markers. After exclusion of known ARHSP loci, the genome-wide screen provided evidence of linkage with a maximal multipoint lod score of 2.6 in the D8S1113-D8S1699 interval. This interval partially overlapped SPG5 and reduced it to a 5.9 megabase (Mb)-region between D8S1113 and D8S544. In a family of Algerian origin from a series of 17 other ARHSP kindreds, linkage to the SPG5 locus was supported by a multipoint lod score of 2.3. The direct sequencing of the coding exons of seven candidate genes did not detect mutations/polymorphisms in the index cases of both linked families. The phenotype of the two SPG5-linked families consisted of spastic paraparesis associated with deep sensory loss. In several patients with long disease durations, there were also mild cerebellar signs. The frequency of SPG5 was approximately 10% (2/18) in our series of ARHSP families with pure or complex forms. We have refined the SPG5 locus to a 3.8 cM interval and extended the phenotype of this form of ARHSP to include slight cerebellar signs.


Subject(s)
Genes, Recessive , Genetic Linkage , Spastic Paraplegia, Hereditary/genetics , Chromosome Mapping , Chromosomes, Human, Pair 8 , Family Health , Genome, Human , Humans , Microsatellite Repeats , Pedigree , Phenotype , Sequence Analysis, DNA , Spastic Paraplegia, Hereditary/diagnosis
10.
Gene Expr Patterns ; 7(3): 289-96, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17045851

ABSTRACT

The drg11 gene is a member of the vertebrate aristaless-related gene family and encodes a paired homeodomain transcription factor. Its expression is largely restricted to PNS neurons subserving somatosensory functions and their CNS targets in rodents. The phenotype of drg11 null mice suggests that it is crucial for the proper development in the embryo of nociceptive circuits. To allow functional studies in the zebrafish, a simple vertebrate model organism, we have cloned the homologous gene and studied its expression throughout embryonic development. drg11 transcripts are first detected at neurula stage in the developing trigeminal ganglion, where it persists throughout development. This is followed by transient expression in spinal cord mechanosensory Rohon-Beard neurons shortly before axogenesis. Expression is later evident in neuronal populations of the dorsal spinal cord and in the dorsal root ganglia. In the developing brain, drg11 expression is mainly restricted to sensory neuron populations of the midbrain and hindbrain, in cranial sensory ganglia and in the habenula. Unlike rodents, however, trochlear motor neurons transiently express drg11. Our results suggest that drg11 expression in the developing zebrafish is, in common with its mammalian homologous gene, predominantly localised to neurons in sensory processing areas of the embryonic nervous system and is both spatially and temporally regulated.


Subject(s)
Gene Expression Regulation, Developmental , Neurons, Afferent/metabolism , Transcription Factors/genetics , Zebrafish Proteins/genetics , Zebrafish/embryology , Amino Acid Sequence , Animals , Central Nervous System , Cloning, Molecular , Embryo, Nonmammalian/metabolism , Embryonic Development , Ganglia, Sensory/embryology , Ganglia, Sensory/metabolism , Mesencephalon/embryology , Mesencephalon/metabolism , Molecular Sequence Data , Organ Specificity , Prosencephalon/embryology , Prosencephalon/metabolism , Rhombencephalon/embryology , Rhombencephalon/metabolism , Sequence Alignment , Spinal Cord/embryology , Spinal Cord/metabolism , Transcription Factors/metabolism , Zebrafish/genetics , Zebrafish Proteins/metabolism
11.
Dev Biol ; 277(2): 417-24, 2005 Jan 15.
Article in English | MEDLINE | ID: mdl-15617684

ABSTRACT

Opitz G/BBB syndrome (OS) is a congenital defect characterized by hypertelorism and hypospadias, but additional midline malformations are also common in OS patients. X-linked OS is caused by mutations in the ubiquitin ligase MID1. In chick, MID1 is involved in left-right determination: a mutually repressive relationship between Shh and cMid1 in Hensen's node plays a key role in establishing the avian left-right axis. We have utilized our existing knowledge of the molecular basis of avian L/R determination to investigate the possible existence of functional redundancy between MID1 and its close homologue MID2. The expression of cMid2 overlaps with that of cMid1 in the node, and we demonstrate that MID2 can both mimic MID1 function as a right side determinant and rescue the laterality defects caused by knocking down endogenous MID proteins in the node. Our results show that MID2 is able to compensate for an absence in MID1 during chick left-right determination and may explain why OS patients do not suffer laterality defects despite the association between midline and L/R development. The demonstration of functional redundancy between MID1 and MID2 in the node provides supports for the hypothesis that partial functional redundancy between MID proteins in other developing structures contributes to the wide variability of OS phenotype.


Subject(s)
Abnormalities, Multiple/genetics , Body Patterning/genetics , Deglutition Disorders/genetics , Gene Expression Regulation, Developmental , Hypospadias/genetics , Microtubule Proteins/genetics , Microtubule-Associated Proteins/genetics , Nuclear Proteins/genetics , Transcription Factors/genetics , Animals , Chick Embryo , Cloning, Molecular , Disease Models, Animal , Electroporation , Functional Laterality/genetics , Genetic Diseases, X-Linked/genetics , In Situ Hybridization , Male , Microtubule Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Mutation/genetics , Nuclear Proteins/metabolism , Organizers, Embryonic/metabolism , Syndrome , Transcription Factors/metabolism , Ubiquitin-Protein Ligases
12.
Neurogenetics ; 5(4): 239-43, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15517445

ABSTRACT

Hereditary spastic paraplegia (HSP) is a group of neurodegenerative disorders mainly characterized by progressive spasticity of the lower limbs. The major features of HSP are a marked phenotypic variability both among and within families and an extended genetic heterogeneity. More than 20 HSP loci and 10 spastic paraplegia genes (SPG) have been identified to date, including the genes responsible for the two most frequent forms of autosomal dominant spastic paraplegia (AD-HSP), encoding spastin (SPG4) and atlastin (SPG3A), respectively. To date, only eight mutations have been described in the atlastin gene, which was reported to account for about 10% of all AD-HSP families. We investigated 15 German and French AD-HSP families, including the 3 large pedigrees that allowed the mapping and subsequent refinement of the SPG3A locus. Three novel mutations were found in exons 4, 9, and 12 of the atlastin gene and the common R239C mutation located in exon 7 was confirmed in a 7th family of European origin. Overall, the comparison of the clinical data for all SPG3A-HSP families reported to date failed to reveal any genotype/phenotype correlation as demonstrated for other forms of AD-HSP. However, it confirmed the early onset of this form of HSP, which was observed in almost all affected individuals with a mutation in the atlastin gene.


Subject(s)
GTP Phosphohydrolases/genetics , Mutation, Missense , Spastic Paraplegia, Hereditary/genetics , Adolescent , Adult , Age of Onset , Amino Acid Sequence , Child , Exons , Family Health , Female , France , GTP-Binding Proteins , Genes, Dominant , Germany , Humans , Male , Membrane Proteins , Molecular Sequence Data
13.
Hum Mol Genet ; 12(1): 71-8, 2003 Jan 01.
Article in English | MEDLINE | ID: mdl-12490534

ABSTRACT

Mutations of spastin are responsible for the most common autosomal dominant form of hereditary spastic paraplegia (AD-HSP), a disease characterized by axonal degeneration of corticospinal tracts and posterior columns. Generation of polyclonal antibodies specific to spastin has revealed two isoforms of 75 and 80 kDa in both human and mouse tissues with a tissue-specific variability of the isoform ratio. Spastin is an abundant protein in neural tissues and immunolabeling experiments have shown that spastin is expressed in neurons but not in glial cells. These data indicate that axonal degeneration linked to spastin mutations is caused by a primary defect of neurons. Protein and transcript analyses of patients carrying either nonsense or frameshift spastin mutations revealed neither truncated protein nor mutated transcripts, providing evidence that these mutations are responsible for a loss of spastin function. Identifying agents able to induce the expression of the non-mutated spastin allele should represent an attractive therapeutic strategy in this disease.


Subject(s)
Adenosine Triphosphatases/metabolism , Calcium-Binding Proteins/physiology , Cell Nucleus/metabolism , Mutation/genetics , Spastic Paraplegia, Hereditary/genetics , Spastic Paraplegia, Hereditary/pathology , Adenosine Triphosphatases/genetics , Alleles , Animals , Cell Line , Gene Expression , HeLa Cells , Humans , Spastin
SELECTION OF CITATIONS
SEARCH DETAIL
...