Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 25
1.
Curr Opin Cell Biol ; 87: 102326, 2024 Apr.
Article En | MEDLINE | ID: mdl-38401181

Neurofilaments (NFs) are abundant cytoskeletal proteins that emerge as a critical hub for cell signalling within neurons. As we start to uncover essential roles of NFs in regulating microtubule and organelle dynamics, nerve conduction and neurotransmission, novel discoveries are expected to arise in genetics, with NFs identified as causal genes for various neurodegenerative diseases. This review will discuss how the latest advances in fundamental and translational research illuminate our understanding of NF biology, particularly their assembly, organisation, transport and degradation. We will emphasise the notion that filaments are not one entity and that future challenges will be to apprehend their diverse composition and structural heterogeneity and to scrutinize how this regulates signalling, sustains neuronal physiology and drives pathophysiology in disease.


Intermediate Filaments , Neurofilament Proteins , Intermediate Filaments/metabolism , Neurofilament Proteins/genetics , Neurofilament Proteins/metabolism , Cytoskeleton/metabolism , Neurons/metabolism , Microtubules/metabolism
2.
Cancer Res Commun ; 4(3): 706-722, 2024 03 08.
Article En | MEDLINE | ID: mdl-38421310

Gigaxonin is an E3 ubiquitin ligase that plays a role in cytoskeletal stability. Its role in cancer is not yet clearly understood. Our previous studies of head and neck cancer had identified gigaxonin interacting with p16 for NFκB ubiquitination. To explore its role in cancer cell growth suppression, we analyzed normal and tumor DNA from cervical and head and neck cancers. There was a higher frequency of exon 8 SNP (c.1293 C>T, rs2608555) in the tumor (46% vs. 25% normal, P = 0.011) pointing to a relationship to cancer. Comparison of primary tumor with recurrence and metastasis did not reveal a statistical significance. Two cervical cancer cell lines, ME180 and HT3 harboring exon 8 SNP and showing T allele expression correlated with higher gigaxonin expression, reduced in vitro cell growth and enhanced cisplatin sensitivity in comparison with C allele expressing cancer cell lines. Loss of gigaxonin expression in ME180 cells through CRISPR-Cas9 or siRNA led to aggressive cancer cell growth including increased migration and Matrigel invasion. The in vitro cell growth phenotypes were reversed with re-expression of gigaxonin. Suppression of cell growth correlated with reduced Snail and increased e-cadherin expression. Mouse tail vein injection studies showed increased lung metastasis of cells with low gigaxonin expression and reduced metastasis with reexpression of gigaxonin. We have found an association between C allele expression and RNA instability and absence of multimeric protein formation. From our results, we conclude that gigaxonin expression is associated with suppression of epithelial-mesenchymal transition through inhibition of Snail. SIGNIFICANCE: Our results suggest that GAN gene exon 8 SNP T allele expression correlates with higher gigaxonin expression and suppression of aggressive cancer cell growth. There is downregulation of Snail and upregulation of e-cadherin through NFκB ubiquitination. We hypothesize that exon 8 T allele and gigaxonin expression could serve as diagnostic markers of suppression of aggressive growth of head and neck cancer.


Head and Neck Neoplasms , Humans , Animals , Mice , Down-Regulation/genetics , Cell Line, Tumor , Head and Neck Neoplasms/drug therapy , Epithelial-Mesenchymal Transition/genetics , Cadherins/genetics
3.
Cells ; 13(1)2024 01 04.
Article En | MEDLINE | ID: mdl-38201307

Autophagy is a major degradative pathway that plays a key role in sustaining cell homeostasis, integrity, and physiological functions. Macroautophagy, which ensures the clearance of cytoplasmic components engulfed in a double-membrane autophagosome that fuses with lysosomes, is orchestrated by a complex cascade of events. Autophagy has a particularly strong impact on the nervous system, and mutations in core components cause numerous neurological diseases. We first review the regulation of autophagy, from autophagosome biogenesis to lysosomal degradation and associated neurodevelopmental/neurodegenerative disorders. We then describe how this process is specifically regulated in the axon and in the somatodendritic compartment and how it is altered in diseases. In particular, we present the neuronal specificities of autophagy, with the spatial control of autophagosome biogenesis, the close relationship of maturation with axonal transport, and the regulation by synaptic activity. Finally, we discuss the physiological functions of autophagy in the nervous system, during development and in adulthood.


Autophagy , Macroautophagy , Autophagosomes , Axonal Transport , Lysosomes
4.
EMBO Mol Med ; 15(7): e16267, 2023 07 10.
Article En | MEDLINE | ID: mdl-37144692

Giant axonal neuropathy (GAN) is a fatal neurodegenerative disorder for which there is currently no treatment. Affecting the nervous system, GAN starts in infancy with motor deficits that rapidly evolve toward total loss of ambulation. Using the gan zebrafish model that reproduces the loss of motility as seen in patients, we conducted the first pharmacological screening for the GAN pathology. Here, we established a multilevel pipeline to identify small molecules restoring both the physiological and the cellular deficits in GAN. We combined behavioral, in silico, and high-content imaging analyses to refine our Hits to five drugs restoring locomotion, axonal outgrowth, and stabilizing neuromuscular junctions in the gan zebrafish. The postsynaptic nature of the drug's cellular targets provides direct evidence for the pivotal role the neuromuscular junction holds in the restoration of motility. Our results identify the first drug candidates that can now be integrated in a repositioning approach to fasten therapy for the GAN disease. Moreover, we anticipate both our methodological development and the identified hits to be of benefit to other neuromuscular diseases.


Giant Axonal Neuropathy , Animals , Giant Axonal Neuropathy/diagnosis , Giant Axonal Neuropathy/pathology , Giant Axonal Neuropathy/therapy , Cytoskeletal Proteins , Zebrafish , Neuromuscular Junction
5.
Front Cell Dev Biol ; 11: 1275155, 2023.
Article En | MEDLINE | ID: mdl-38164457

Neurofilaments (NFs) are the most abundant component of mature neurons, that interconnect with actin and microtubules to form the cytoskeleton. Specifically expressed in the nervous system, NFs present the particularity within the Intermediate Filament family of being formed by four subunits, the neurofilament light (NF-L), medium (NF-M), heavy (NF-H) proteins and α-internexin or peripherin. Here, we review the current knowledge on NF proteins and neurofilaments, from their domain structures and their model of assembly to the dynamics of their transport and degradation along the axon. The formation of the filament and its behaviour are regulated by various determinants, including post-transcriptional (miRNA and RBP proteins) and post-translational (phosphorylation and ubiquitination) modifiers. Altogether, the complex set of modifications enable the neuron to establish a stable but elastic NF array constituting the structural scaffold of the axon, while permitting the local expression of NF proteins and providing the dynamics necessary to fulfil local demands and respond to stimuli and injury. Thus, in addition to their roles in mechano-resistance, radial axonal outgrowth and nerve conduction, NFs control microtubule dynamics, organelle distribution and neurotransmission at the synapse. We discuss how the studies of neurodegenerative diseases with NF aggregation shed light on the biology of NFs. In particular, the NEFL and NEFH genes are mutated in Charcot-Marie-Tooth (CMT) disease, the most common inherited neurological disorder of the peripheral nervous system. The clinical features of the CMT forms (axonal CMT2E, CMT2CC; demyelinating CMT1F; intermediate I-CMT) with symptoms affecting the central nervous system (CNS) will allow us to further investigate the physiological roles of NFs in the brain. Thus, NF-CMT mouse models exhibit various degrees of sensory-motor deficits associated with CNS symptoms. Cellular systems brought findings regarding the dominant effect of NF-L mutants on NF aggregation and transport, although these have been recently challenged. Neurofilament detection without NF-L in recessive CMT is puzzling, calling for a re-examination of the current model in which NF-L is indispensable for NF assembly. Overall, we discuss how the fundamental and translational fields are feeding each-other to increase but also challenge our knowledge of NF biology, and to develop therapeutic avenues for CMT and neurodegenerative diseases with NF aggregation.

6.
Front Mol Neurosci ; 15: 956582, 2022.
Article En | MEDLINE | ID: mdl-36204134

The zebrafish (Danio rerio) is a vertebrate species offering multitude of advantages for the study of conserved biological systems in human and has considerably enriched our knowledge in developmental biology and physiology. Being equally important in medical research, the zebrafish has become a critical tool in the fields of diagnosis, gene discovery, disease modeling, and pharmacology-based therapy. Studies on the zebrafish neuromuscular system allowed for deciphering key molecular pathways in this tissue, and established it as a model of choice to study numerous motor neurons, neuromuscular junctions, and muscle diseases. Starting with the similarities of the zebrafish neuromuscular system with the human system, we review disease models associated with the neuromuscular system to focus on current methodologies employed to study them and outline their caveats. In particular, we put in perspective the necessity to develop standardized and high-resolution methodologies that are necessary to deepen our understanding of not only fundamental signaling pathways in a healthy tissue but also the changes leading to disease phenotype outbreaks, and offer templates for high-content screening strategies. While the development of high-throughput methodologies is underway for motility assays, there is no automated approach to quantify the key molecular cues of the neuromuscular junction. Here, we provide a novel high-throughput imaging methodology in the zebrafish that is standardized, highly resolutive, quantitative, and fit for drug screening. By providing a proof of concept for its robustness in identifying novel molecular players and therapeutic drugs in giant axonal neuropathy (GAN) disease, we foresee that this new tool could be useful for both fundamental and biomedical research.

7.
Curr Opin Cell Biol ; 68: 181-191, 2021 02.
Article En | MEDLINE | ID: mdl-33454158

In the last two years, neurofilaments (NFs) have become one of the most blazing topics in clinical neuroscience. NFs are major cytoskeletal constituents of neurons, can be detected in body fluids, and have recently emerged as universal biomarkers of neuronal injury and neurological diseases. This review will examine the evolving landscape of NFs, from their specific cellular functions within neurons to their broad clinical value as biomarkers. Particular attention will be given to the dynamic nature of the NF network and its novel roles in microtubule regulation, neurotransmission, and nanomedicine. Building from the initial evidence of causative mutations in NF genes in Charcot-Marie-Tooth diseases, the latest advances at the frontiers of basic and clinical sciences have expanded the scope and relevance of NFs for human health remarkably and have poised to fuel innovation in cell biology and neuroscience.


Intermediate Filaments/metabolism , Neurons/metabolism , Animals , Biomarkers/analysis , Charcot-Marie-Tooth Disease/metabolism , Charcot-Marie-Tooth Disease/pathology , Cytoskeleton/chemistry , Cytoskeleton/metabolism , Humans , Intermediate Filaments/chemistry , Intermediate Filaments/genetics , Mutation , Neurons/chemistry , Neurons/pathology
8.
Front Physiol ; 11: 1022, 2020.
Article En | MEDLINE | ID: mdl-33192535

Ubiquitination is a dynamic post-translational modification that regulates the fate of proteins and therefore modulates a myriad of cellular functions. At the last step of this sophisticated enzymatic cascade, E3 ubiquitin ligases selectively direct ubiquitin attachment to specific substrates. Altogether, the ∼800 distinct E3 ligases, combined to the exquisite variety of ubiquitin chains and types that can be formed at multiple sites on thousands of different substrates confer to ubiquitination versatility and infinite possibilities to control biological functions. E3 ubiquitin ligases have been shown to regulate behaviors of proteins, from their activation, trafficking, subcellular distribution, interaction with other proteins, to their final degradation. Largely known for tagging proteins for their degradation by the proteasome, E3 ligases also direct ubiquitinated proteins and more largely cellular content (organelles, ribosomes, etc.) to destruction by autophagy. This multi-step machinery involves the creation of double membrane autophagosomes in which engulfed material is degraded after fusion with lysosomes. Cooperating in sustaining homeostasis, actors of ubiquitination, proteasome and autophagy pathways are impaired or mutated in wide range of human diseases. From initial discovery of pathogenic mutations in the E3 ligase encoding for E6-AP in Angelman syndrome and Parkin in juvenile forms of Parkinson disease, the number of E3 ligases identified as causal gene for neurological diseases has considerably increased within the last years. In this review, we provide an overview of these diseases, by classifying the E3 ubiquitin ligase types and categorizing the neurological signs. We focus on the Gigaxonin-E3 ligase, mutated in giant axonal neuropathy and present a comprehensive analysis of the spectrum of mutations and the recent biological models that permitted to uncover novel mechanisms of action. Then, we discuss the common functions shared by Gigaxonin and the other E3 ligases in cytoskeleton architecture, cell signaling and autophagy. In particular, we emphasize their pivotal roles in controlling multiple steps of the autophagy pathway. In light of the various targets and extending functions sustained by a single E3 ligase, we finally discuss the challenge in understanding the complex pathological cascade underlying disease and in designing therapeutic approaches that can apprehend this complexity.

9.
Brain ; 143(7): 1975-1998, 2020 07 01.
Article En | MEDLINE | ID: mdl-32408345

Interest in neurofilaments has risen sharply in recent years with recognition of their potential as biomarkers of brain injury or neurodegeneration in CSF and blood. This is in the context of a growing appreciation for the complexity of the neurobiology of neurofilaments, new recognition of specialized roles for neurofilaments in synapses and a developing understanding of mechanisms responsible for their turnover. Here we will review the neurobiology of neurofilament proteins, describing current understanding of their structure and function, including recently discovered evidence for their roles in synapses. We will explore emerging understanding of the mechanisms of neurofilament degradation and clearance and review new methods for future elucidation of the kinetics of their turnover in humans. Primary roles of neurofilaments in the pathogenesis of human diseases will be described. With this background, we then will review critically evidence supporting use of neurofilament concentration measures as biomarkers of neuronal injury or degeneration. Finally, we will reflect on major challenges for studies of the neurobiology of intermediate filaments with specific attention to identifying what needs to be learned for more precise use and confident interpretation of neurofilament measures as biomarkers of neurodegeneration.


Biomarkers , Intermediate Filaments , Nerve Degeneration , Synapses , Animals , Humans
10.
J Clin Invest ; 129(12): 5312-5326, 2019 12 02.
Article En | MEDLINE | ID: mdl-31503551

Growing evidence shows that alterations occurring at early developmental stages contribute to symptoms manifested in adulthood in the setting of neurodegenerative diseases. Here, we studied the molecular mechanisms causing giant axonal neuropathy (GAN), a severe neurodegenerative disease due to loss-of-function of the gigaxonin-E3 ligase. We showed that gigaxonin governs Sonic Hedgehog (Shh) induction, the developmental pathway patterning the dorso-ventral axis of the neural tube and muscles, by controlling the degradation of the Shh-bound Patched receptor. Similar to Shh inhibition, repression of gigaxonin in zebrafish impaired motor neuron specification and somitogenesis and abolished neuromuscular junction formation and locomotion. Shh signaling was impaired in gigaxonin-null zebrafish and was corrected by both pharmacological activation of the Shh pathway and human gigaxonin, pointing to an evolutionary-conserved mechanism regulating Shh signaling. Gigaxonin-dependent inhibition of Shh activation was also demonstrated in primary fibroblasts from patients with GAN and in a Shh activity reporter line depleted in gigaxonin. Our findings establish gigaxonin as a key E3 ligase that positively controls the initiation of Shh transduction, and reveal the causal role of Shh dysfunction in motor deficits, thus highlighting the developmental origin of GAN.


Cytoskeletal Proteins/genetics , Giant Axonal Neuropathy/etiology , Hedgehog Proteins/physiology , Mutation , Animals , Cytoskeletal Proteins/physiology , Hedgehog Proteins/antagonists & inhibitors , Humans , Mice , Motor Neurons/physiology , Muscles/innervation , NIH 3T3 Cells , Patched-1 Receptor/physiology , Signal Transduction , Somites/physiology , Zebrafish
11.
Autophagy ; 15(9): 1650-1652, 2019 09.
Article En | MEDLINE | ID: mdl-31179825

The sequential action of ATG proteins guarantees the formation of the autophagosome from the steps of the induction, nucleation, elongation and sealing of the phagophore membrane. Posttranslational modifications further add to the fine-tuning regulation of this highly ordered machinery and confer, in space and time, the dynamics necessary to respond to macroautophagy/autophagy activation, and to shut it down. Recently, we reported the discovery of GAN (gigaxonin), an E3 ubiquitin ligase adaptor as a key regulator of the elongation step of phagophore formation. GAN interacts, ubiquitinates and degrades ATG16L1, which forms a complex with the ATG12-ATG5 ubiquitin-like conjugation system, and specifies the site of lipidation of LC3 by this complex onto the nascent phagophore. Accordingly, depletion of GAN in primary neurons causes the accumulation of ATG16L1 and decreases the autophagy flux by impairing the net production of autophagosomes. Considering the pivotal role of ATG16L1 in autophagy, and the reversal of the deficits upon reintroduction of GAN, one can speculate that GAN constitutes a novel molecular switch to fine tune the autophagy machinery.


Autophagosomes , Autophagy , Autophagy-Related Protein 12 , Autophagy-Related Proteins , Ubiquitin-Protein Ligases
12.
Nat Commun ; 10(1): 780, 2019 02 15.
Article En | MEDLINE | ID: mdl-30770803

Autophagy is an essential self-digestion machinery for cell survival and homoeostasis. Membrane elongation is fundamental, as it drives the formation of the double-membrane vesicles that engulf cytosolic material. LC3-lipidation, the signature of autophagosome formation, results from a complex ubiquitin-conjugating cascade orchestrated by the ATG16L1 protein, whose regulation is unknown. Here, we identify the Gigaxonin-E3 ligase as the first regulator of ATG16L1 turn-over and autophagosome production. Gigaxonin interacts with the WD40 domain of ATG16L1 to drive its ubiquitination and subsequent degradation. Gigaxonin depletion induces the formation of ATG16L1 aggregates and impairs LC3 lipidation, hence altering lysosomal fusion and degradation of the main autophagy receptor p62. Altogether, we demonstrate that the Gigaxonin-E3 ligase controls the production of autophagosomes by a reversible, ubiquitin-dependent process selective for ATG16L1. Our findings unveil the fundamental mechanisms of the control of autophagosome formation, and provide a molecular switch to fine-tune the activation of autophagy.


Autophagosomes/metabolism , Cytoskeletal Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Autophagy/physiology , Autophagy-Related Proteins/metabolism , COS Cells , Chlorocebus aethiops , HEK293 Cells , Humans
13.
Clin Exp Nephrol ; 22(6): 1251-1257, 2018 Dec.
Article En | MEDLINE | ID: mdl-29869755

BACKGROUND: Pseudohypoaldosteronism type II (PHAII) is a hereditary hypertensive disease caused by mutations in four genes: WNK1, WNK4, Kelch-like3 (KLHL3), and cullin3 (CUL3). Recently, it was revealed that CUL3-KLHL3 E3 ligase complex ubiquitinates WNK1 and WNK4, leading to their degradation, and that a common pathogenesis of PHAII is defective WNK degradation due to CUL3-KLHL3 E3 ligase complex impairment. PHAII-causing CUL3 mutations mediate exon9 skipping, producing a CUL3 protein with a 57-amino acid deletion (Δ403-459). However, the pathogenic effects of KLHL3, an adaptor protein that links WNKs with CUL3, in PHAII caused by CUL3 mutation remain unclear. METHODS: To clarify detailed pathophysiological mechanisms underlying PHAII caused by CUL3 mutation in vivo, we generated and analyzed knock-in mice carrying the same CUL3 exon9 deletion (CUL3WT/Δex9) as that reported in PHAII patients. RESULTS: CUL3WT/Δex9 mice exhibited a PHAII-like phenotype. Interestingly, we confirmed markedly decreased KLHL3 expression in CUL3WT/Δex9 mice by confirming the true KLHL3 band in vivo. However, the expression of other KLHL family proteins, such as KLHL2, was comparable between WT and mutant mice. CONCLUSION: KLHL3 expression was decreased in CUL3WT/Δex9 mice. However, expression levels of other KLHL family proteins were comparable between the wild-type and mutant mice. These findings indicate that the decreased abundance of KLHL3 is a specific phenomenon caused by mutant CUL3 (Δexon9). Our findings would improve our understanding of the pathogenesis of PHAII caused by CUL3 mutation in vivo.


Carrier Proteins/physiology , Cullin Proteins/genetics , Mutation , Pseudohypoaldosteronism/etiology , Adaptor Proteins, Signal Transducing , Animals , Carrier Proteins/analysis , Humans , Mice , Microfilament Proteins , Pseudohypoaldosteronism/genetics
14.
Am J Hum Genet ; 99(3): 695-703, 2016 09 01.
Article En | MEDLINE | ID: mdl-27545681

Via whole-exome sequencing, we identified rare autosomal-recessive variants in UBA5 in five children from four unrelated families affected with a similar pattern of severe intellectual deficiency, microcephaly, movement disorders, and/or early-onset intractable epilepsy. UBA5 encodes the E1-activating enzyme of ubiquitin-fold modifier 1 (UFM1), a recently identified ubiquitin-like protein. Biochemical studies of mutant UBA5 proteins and studies in fibroblasts from affected individuals revealed that UBA5 mutations impair the process of ufmylation, resulting in an abnormal endoplasmic reticulum structure. In Caenorhabditis elegans, knockout of uba-5 and of human orthologous genes in the UFM1 cascade alter cholinergic, but not glutamatergic, neurotransmission. In addition, uba5 silencing in zebrafish decreased motility while inducing abnormal movements suggestive of seizures. These clinical, biochemical, and experimental findings support our finding of UBA5 mutations as a pathophysiological cause for early-onset encephalopathies due to abnormal protein ufmylation.


Alleles , Brain Diseases/genetics , Mutation/genetics , Proteins/metabolism , Ubiquitin-Activating Enzymes/genetics , Age of Onset , Animals , Brain Mapping , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Child , Child, Preschool , Cholinergic Neurons/metabolism , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/pathology , Epilepsy/genetics , Exome/genetics , Female , Fibroblasts , Genes, Recessive/genetics , Humans , Intellectual Disability/genetics , Magnetic Resonance Imaging , Male , Microcephaly/genetics , Movement Disorders , Proteins/genetics , Synaptic Transmission/genetics , Ubiquitin/genetics , Ubiquitin/metabolism , Ubiquitin-Activating Enzymes/deficiency , Ubiquitin-Activating Enzymes/metabolism , Ubiquitins/genetics , Ubiquitins/metabolism , Zebrafish/genetics , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
15.
Methods Enzymol ; 569: 215-31, 2016.
Article En | MEDLINE | ID: mdl-26778561

Intermediate filament turnover is a highly dynamic process required to maintain tissue integrity and is implicated in degenerative and regenerative processes. Despite these essential roles, little is known about the mechanisms that cause the degradation of intermediate filaments. Nevertheless, the last decade has seen the emergence of the ubiquitin proteasome system, in particular E3 ubiquitin ligases, as important regulators. Here, we will focus on the first identified factor controlling the degradation of the entire intermediate filament family, the gigaxonin-E3 ligase. We will present the scientific achievements and the methodologies to study gigaxonin and its crucial role in intermediate filament turnover.


Cytoskeletal Proteins/physiology , Intermediate Filaments/metabolism , Animals , Humans , Proteolysis
16.
Am J Hum Genet ; 97(5): 754-60, 2015 Nov 05.
Article En | MEDLINE | ID: mdl-26593267

Autosomal-recessive optic neuropathies are rare blinding conditions related to retinal ganglion cell (RGC) and optic-nerve degeneration, for which only mutations in TMEM126A and ACO2 are known. In four families with early-onset recessive optic neuropathy, we identified mutations in RTN4IP1, which encodes a mitochondrial ubiquinol oxydo-reductase. RTN4IP1 is a partner of RTN4 (also known as NOGO), and its ortholog Rad8 in C. elegans is involved in UV light response. Analysis of fibroblasts from affected individuals with a RTN4IP1 mutation showed loss of the altered protein, a deficit of mitochondrial respiratory complex I and IV activities, and increased susceptibility to UV light. Silencing of RTN4IP1 altered the number and morphogenesis of mouse RGC dendrites in vitro and the eye size, neuro-retinal development, and swimming behavior in zebrafish in vivo. Altogether, these data point to a pathophysiological mechanism responsible for RGC early degeneration and optic neuropathy and linking RTN4IP1 functions to mitochondrial physiology, response to UV light, and dendrite growth during eye maturation.


Carrier Proteins/genetics , Fibroblasts/pathology , Mitochondria/pathology , Mitochondrial Proteins/genetics , Mutation/genetics , Optic Nerve Diseases/genetics , Optic Nerve Diseases/pathology , Retinal Ganglion Cells/pathology , Amino Acid Sequence , Animals , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/metabolism , Case-Control Studies , Cells, Cultured , Electron Transport Complex I , Female , Fibroblasts/metabolism , Follow-Up Studies , Genes, Recessive , Humans , Male , Mice , Mitochondria/genetics , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/metabolism , Molecular Sequence Data , Nerve Degeneration , Pedigree , Prognosis , Retinal Ganglion Cells/metabolism , Sequence Homology, Amino Acid , Zebrafish/genetics , Zebrafish/growth & development , Zebrafish/metabolism
17.
Acta Neuropathol Commun ; 2: 47, 2014 Apr 24.
Article En | MEDLINE | ID: mdl-24758703

BACKGROUND: The BTB-KELCH protein Gigaxonin plays key roles in sustaining neuron survival and cytoskeleton architecture. Indeed, recessive mutations in the Gigaxonin-encoding gene cause Giant Axonal Neuropathy (GAN), a severe neurodegenerative disorder characterized by a wide disorganization of the Intermediate Filament network. Growing evidences suggest that GAN is a continuum with the peripheral neuropathy Charcot-Marie-Tooth diseases type 2 (CMT2). Sharing similar sensory-motor alterations and aggregation of Neurofilaments, few reports have revealed that GAN and some CMT2 forms can be misdiagnosed on clinical and histopathological examination. The goal of this study is to propose a new differential diagnostic test for GAN/CMT2. Moreover, we aim at identifying the mechanisms causing the loss-of-function of Gigaxonin, which has been proposed to bind CUL3 and substrates as part of an E3 ligase complex. RESULTS: We establish that determining Gigaxonin level constitutes a very valuable diagnostic test in discriminating new GAN cases from clinically related inherited neuropathies. Indeed, in a set of seven new families presenting a neuropathy resembling GAN/CMT2, only five exhibiting a reduced Gigaxonin abundance have been subsequently genetically linked to GAN. Generating the homology modeling of Gigaxonin, we suggest that disease mutations would lead to a range of defects in Gigaxonin stability, impairing its homodimerization, BTB or KELCH domain folding, or CUL3 and substrate binding. We further demonstrate that regardless of the mutations or the severity of the disease, Gigaxonin abundance is severely reduced in all GAN patients due to both mRNA and protein instability mechanisms. CONCLUSIONS: In this study, we developed a new penetrant and specific test to diagnose GAN among a set of individuals exhibiting CMT2 of unknown etiology to suggest that the prevalence of GAN is probably under-evaluated among peripheral neuropathies. We propose to use this new test in concert with the clinical examination and prior to the systematic screening of GAN mutations that has shown strong limitations for large deletions. Combining the generation of the structural modeling of Gigaxonin to an analysis of Gigaxonin transcripts and proteins in patients, we provide the first evidences of the instability of this E3 ligase adaptor in disease.


Cytoskeletal Proteins/metabolism , Giant Axonal Neuropathy/genetics , Giant Axonal Neuropathy/metabolism , Mutation/genetics , Adult , Animals , COS Cells , Child , Chlorocebus aethiops , DNA Mutational Analysis , Datasets as Topic , Female , Gene Expression Regulation/genetics , Giant Axonal Neuropathy/pathology , Humans , Male , Models, Molecular , Phenotype , Transfection , Young Adult
18.
J Clin Invest ; 123(5): 1964-75, 2013 May.
Article En | MEDLINE | ID: mdl-23585478

Giant axonal neuropathy (GAN) is an early-onset neurological disorder caused by mutations in the GAN gene (encoding for gigaxonin), which is predicted to be an E3 ligase adaptor. In GAN, aggregates of intermediate filaments (IFs) represent the main pathological feature detected in neurons and other cell types, including patients' dermal fibroblasts. The molecular mechanism by which these mutations cause IFs to aggregate is unknown. Using fibroblasts from patients and normal individuals, as well as Gan-/- mice, we demonstrated that gigaxonin was responsible for the degradation of vimentin IFs. Gigaxonin was similarly involved in the degradation of peripherin and neurofilament IF proteins in neurons. Furthermore, proteasome inhibition by MG-132 reversed the clearance of IF proteins in cells overexpressing gigaxonin, demonstrating the involvement of the proteasomal degradation pathway. Together, these findings identify gigaxonin as a major factor in the degradation of cytoskeletal IFs and provide an explanation for IF aggregate accumulation, the subcellular hallmark of this devastating human disease.


Cytoskeletal Proteins/genetics , Giant Axonal Neuropathy/pathology , Intermediate Filament Proteins/metabolism , Mutation , Animals , Cytoskeletal Proteins/metabolism , Cytoskeleton/metabolism , Enzyme-Linked Immunosorbent Assay , Fibroblasts/cytology , Fibroblasts/metabolism , HEK293 Cells , Humans , Leupeptins/pharmacology , Mice , Mice, Transgenic , Microscopy, Fluorescence , NIH 3T3 Cells , Neurons/metabolism , Proteasome Inhibitors/pharmacology , RNA, Messenger/metabolism , Sequence Analysis, DNA , Ubiquitin-Protein Ligases/metabolism , Vimentin/metabolism
19.
Mol Neurodegener ; 6: 25, 2011 Apr 12.
Article En | MEDLINE | ID: mdl-21486449

BACKGROUND: Giant Axonal Neuropathy (GAN) is a fatal neurodegenerative disorder with early onset characterized by a severe deterioration of the peripheral and central nervous system, involving both the motor and the sensory tracts and leading to ataxia, speech defect and intellectual disabilities. The broad deterioration of the nervous system is accompanied by a generalized disorganization of the intermediate filaments, including neurofilaments in neurons, but the implication of this defect in disease onset or progression remains unknown. The identification of gigaxonin, the substrate adaptor of an E3 ubiquitin ligase, as the defective protein in GAN allows us to now investigate the crucial role of the gigaxonin-E3 ligase in sustaining neuronal and intermediate filament integrity. To study the mechanisms controlled by gigaxonin in these processes and to provide a relevant model to test the therapeutic approaches under development for GAN, we generated a Gigaxonin-null mouse by gene targeting. RESULTS: We investigated for the first time in Gigaxonin-null mice the deterioration of the motor and sensory functions over time as well as the spatial disorganization of neurofilaments. We showed that gigaxonin depletion in mice induces mild but persistent motor deficits starting at 60 weeks of age in the 129/SvJ-genetic background, while sensory deficits were demonstrated in C57BL/6 animals. In our hands, another gigaxonin-null mouse did not display the early and severe motor deficits reported previously. No apparent neurodegeneration was observed in our knock-out mice, but dysregulation of neurofilaments in proximal and distal axons was massive. Indeed, neurofilaments were not only more abundant but they also showed the abnormal increase in diameter and misorientation that are characteristics of the human pathology. CONCLUSIONS: Together, our results show that gigaxonin depletion in mice induces mild motor and sensory deficits but recapitulates the severe neurofilament dysregulation seen in patients. Our model will allow investigation of the role of the gigaxonin-E3 ligase in organizing neurofilaments and may prove useful in understanding the pathological processes engaged in other neurodegenerative disorders characterized by accumulation of neurofilaments and dysfunction of the Ubiquitin Proteasome System, such as Amyotrophic Lateral Sclerosis, Huntington's, Alzheimer's and Parkinson's diseases.

20.
Nat Genet ; 41(6): 746-52, 2009 Jun.
Article En | MEDLINE | ID: mdl-19465910

Polymicrogyria is a relatively common but poorly understood defect of cortical development characterized by numerous small gyri and a thick disorganized cortical plate lacking normal lamination. Here we report de novo mutations in a beta-tubulin gene, TUBB2B, in four individuals and a 27-gestational-week fetus with bilateral asymmetrical polymicrogyria. Neuropathological examination of the fetus revealed an absence of cortical lamination associated with the presence of ectopic neuronal cells in the white matter and in the leptomeningeal spaces due to breaches in the pial basement membrane. In utero RNAi-based inactivation demonstrates that TUBB2B is required for neuronal migration. We also show that two disease-associated mutations lead to impaired formation of tubulin heterodimers. These observations, together with previous data, show that disruption of microtubule-based processes underlies a large spectrum of neuronal migration disorders that includes not only lissencephaly and pachygyria, but also polymicrogyria malformations.


Cerebral Cortex/abnormalities , Malformations of Cortical Development/genetics , Mutation , Tubulin/genetics , Adolescent , Adult , Amino Acid Substitution , Cerebral Cortex/embryology , Cerebral Cortex/pathology , Child, Preschool , Female , Fetal Diseases/genetics , Genetic Variation , Humans , Lissencephaly/genetics , Malformations of Cortical Development/pathology , Pia Mater/abnormalities , Pia Mater/embryology , Pia Mater/pathology , Pregnancy
...