Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Front Cell Dev Biol ; 11: 1136699, 2023.
Article in English | MEDLINE | ID: mdl-36875768

ABSTRACT

Microtubules are filamentous structures that play a critical role in a diverse array of cellular functions including, mitosis, nuclear translocation, trafficking of organelles and cell shape. They are composed of α/ß-tubulin heterodimers which are encoded by a large multigene family that has been implicated in an umbrella of disease states collectively known as the tubulinopathies. De novo mutations in different tubulin genes are known to cause lissencephaly, microcephaly, polymicrogyria, motor neuron disease, and female infertility. The diverse clinical features associated with these maladies have been attributed to the expression pattern of individual tubulin genes, as well as their distinct Functional repertoire. Recent studies, however, have highlighted the impact of tubulin mutations on microtubule-associated proteins (MAPs). MAPs can be classified according to their effect on microtubules and include polymer stabilizers (e.g., tau, MAP2, doublecortin), destabilizers (e.g., spastin, katanin), plus-end binding proteins (e.g., EB1-3, XMAP215, CLASPs) and motor proteins (e.g., dyneins, kinesins). In this review we analyse mutation-specific disease mechanisms that influence MAP binding and their phenotypic consequences, and discuss methods by which we can exploit genetic variation to identify novel MAPs.

2.
Sci Rep ; 13(1): 1215, 2023 01 21.
Article in English | MEDLINE | ID: mdl-36681692

ABSTRACT

The tubulinopathies are an umbrella of rare diseases that result from mutations in tubulin genes and are frequently characterised by severe brain malformations. The characteristics of a given disease reflect the expression pattern of the transcript, the function of a given tubulin gene, and the role microtubules play in a particular cell type. Mouse models have proved to be valuable tools that have provided insight into the molecular and cellular mechanisms that underlie the disease state. In this manuscript we compare two Tuba1a mouse models, both of which express wild-type TUBA1A protein but employ different codon usage. We show that modification of the Tuba1a mRNA sequence results in homozygous lethality and a severe neurodevelopmental phenotype. This is associated with a decrease in the number of post-mitotic neurons, PAX6 positive progenitors, and an increase in the number of apoptotic cells. We attribute this to a decrease in the stability of the modified Tuba1a transcript, and the absence of compensation by the other neurogenic tubulins. Our findings highlight the importance of maintaining the wild-type coding sequence when engineering mouse lines and the impact of synonymous genetic variation.


Subject(s)
Codon , Tubulin , Animals , Mice , Codon/genetics , Mutation , Phenotype , RNA, Messenger/genetics , Tubulin/genetics , Tubulin/metabolism
4.
J Exp Med ; 219(9)2022 09 05.
Article in English | MEDLINE | ID: mdl-35947077

ABSTRACT

The genetic causes of primary antibody deficiencies and autism spectrum disorder (ASD) are largely unknown. Here, we report a patient with hypogammaglobulinemia and ASD who carries biallelic mutations in the transcription factor PAX5. A patient-specific Pax5 mutant mouse revealed an early B cell developmental block and impaired immune responses as the cause of hypogammaglobulinemia. Pax5 mutant mice displayed behavioral deficits in all ASD domains. The patient and the mouse model showed aberrant cerebellar foliation and severely impaired sensorimotor learning. PAX5 deficiency also caused profound hypoplasia of the substantia nigra and ventral tegmental area due to loss of GABAergic neurons, thus affecting two midbrain hubs, controlling motor function and reward processing, respectively. Heterozygous Pax5 mutant mice exhibited similar anatomic and behavioral abnormalities. Lineage tracing identified Pax5 as a crucial regulator of cerebellar morphogenesis and midbrain GABAergic neurogenesis. These findings reveal new roles of Pax5 in brain development and unravel the underlying mechanism of a novel immunological and neurodevelopmental syndrome.


Subject(s)
Agammaglobulinemia , Autism Spectrum Disorder , Animals , Autism Spectrum Disorder/genetics , Heterozygote , Mice , Mutation/genetics , PAX5 Transcription Factor/genetics
5.
PLoS Genet ; 16(11): e1009104, 2020 11.
Article in English | MEDLINE | ID: mdl-33137126

ABSTRACT

Microtubules play a critical role in multiple aspects of neurodevelopment, including the generation, migration and differentiation of neurons. A recurrent mutation (R402H) in the α-tubulin gene TUBA1A is known to cause lissencephaly with cerebellar and striatal phenotypes. Previous work has shown that this mutation does not perturb the chaperone-mediated folding of tubulin heterodimers, which are able to assemble and incorporate into the microtubule lattice. To explore the molecular mechanisms that cause the disease state we generated a new conditional mouse line that recapitulates the R402H variant. We show that heterozygous mutants present with laminar phenotypes in the cortex and hippocampus, as well as a reduction in striatal size and cerebellar abnormalities. We demonstrate that homozygous expression of the R402H allele causes neuronal death and exacerbates a cell intrinsic defect in cortical neuronal migration. Microtubule sedimentation assays coupled with quantitative mass spectrometry demonstrated that the binding and/or levels of multiple microtubule associated proteins (MAPs) are perturbed by the R402H mutation including VAPB, REEP1, EZRIN, PRNP and DYNC1l1/2. Consistent with these data we show that the R402H mutation impairs dynein-mediated transport which is associated with a decoupling of the nucleus to the microtubule organising center. Our data support a model whereby the R402H variant is able to fold and incorporate into microtubules, but acts as a gain of function by perturbing the binding of MAPs.


Subject(s)
Brain/pathology , Lissencephaly/pathology , Microtubule-Associated Proteins/metabolism , Tubulin/genetics , Animals , Brain/cytology , Brain/embryology , Cell Movement , Cytoplasmic Dyneins/metabolism , Disease Models, Animal , Embryo, Mammalian , Female , Heterozygote , Humans , Lissencephaly/genetics , Mice , Mice, Transgenic , Microtubules/metabolism , Mutation, Missense , Neurons/metabolism , Neurons/pathology , Protein Binding/genetics , Proteomics , Tubulin/metabolism
6.
Neuron ; 100(6): 1354-1368.e5, 2018 12 19.
Article in English | MEDLINE | ID: mdl-30449657

ABSTRACT

Corpus callosum malformations are associated with a broad range of neurodevelopmental diseases. We report that de novo mutations in MAST1 cause mega-corpus-callosum syndrome with cerebellar hypoplasia and cortical malformations (MCC-CH-CM) in the absence of megalencephaly. We show that MAST1 is a microtubule-associated protein that is predominantly expressed in post-mitotic neurons and is present in both dendritic and axonal compartments. We further show that Mast1 null animals are phenotypically normal, whereas the deletion of a single amino acid (L278del) recapitulates the distinct neurological phenotype observed in patients. In animals harboring Mast1 microdeletions, we find that the PI3K/AKT3/mTOR pathway is unperturbed, whereas Mast2 and Mast3 levels are diminished, indicative of a dominant-negative mode of action. Finally, we report that de novo MAST1 substitutions are present in patients with autism and microcephaly, raising the prospect that mutations in this gene give rise to a spectrum of neurodevelopmental diseases.


Subject(s)
Agenesis of Corpus Callosum/genetics , Cerebellum/abnormalities , Gene Expression Regulation, Developmental/genetics , Malformations of Cortical Development/genetics , Microtubule-Associated Proteins/genetics , Mutation/genetics , Nervous System Malformations/genetics , Agenesis of Corpus Callosum/complications , Agenesis of Corpus Callosum/diagnostic imaging , Agenesis of Corpus Callosum/pathology , Animals , Animals, Newborn , Apoptosis/genetics , Brain/metabolism , Brain/pathology , Cells, Cultured , Cerebellum/diagnostic imaging , Child , Developmental Disabilities/complications , Developmental Disabilities/diagnostic imaging , Developmental Disabilities/genetics , Disease Models, Animal , Embryo, Mammalian , Female , Humans , Male , Malformations of Cortical Development/complications , Malformations of Cortical Development/diagnostic imaging , Mice , Mice, Inbred C57BL , Mice, Knockout , Microtubule-Associated Proteins/deficiency , Nerve Tissue Proteins/metabolism , Nervous System Malformations/complications , Nervous System Malformations/diagnostic imaging , PAX6 Transcription Factor/metabolism
7.
Nat Neurosci ; 21(8): 1139, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29875394

ABSTRACT

In the supplementary information PDF originally posted, there were discrepancies from the integrated supplementary information that appeared in the HTML; the former has been corrected as follows. In the legend to Supplementary Fig. 2c, "major organs of the mouse" has been changed to "major organs of the adult mouse." In the legend to Supplementary Fig. 6d,h, "At E14.5 Mbe/Mbe mutants have a smaller percentage of Brdu positive cells in bin 3" has been changed to "At E14.5 Mbe/Mbe mutants have a higher percentage of Brdu positive cells in bin 3."

8.
Nat Neurosci ; 21(2): 207-217, 2018 02.
Article in English | MEDLINE | ID: mdl-29311744

ABSTRACT

The formation of the vertebrate brain requires the generation, migration, differentiation and survival of neurons. Genetic mutations that perturb these critical cellular events can result in malformations of the telencephalon, providing a molecular window into brain development. Here we report the identification of an N-ethyl-N-nitrosourea-induced mouse mutant characterized by a fractured hippocampal pyramidal cell layer, attributable to defects in neuronal migration. We show that this is caused by a hypomorphic mutation in Vps15 that perturbs endosomal-lysosomal trafficking and autophagy, resulting in an upregulation of Nischarin, which inhibits Pak1 signaling. The complete ablation of Vps15 results in the accumulation of autophagic substrates, the induction of apoptosis and severe cortical atrophy. Finally, we report that mutations in VPS15 are associated with cortical atrophy and epilepsy in humans. These data highlight the importance of the Vps15-Vps34 complex and the Nischarin-Pak1 signaling hub in the development of the telencephalon.


Subject(s)
Cell Movement/genetics , Gene Expression Regulation, Developmental/drug effects , Mutation/drug effects , Neurodevelopmental Disorders , Neurons/pathology , Vacuolar Proton-Translocating ATPases/genetics , Alkylating Agents/toxicity , Animals , Animals, Newborn , Atrophy/chemically induced , Atrophy/genetics , Atrophy/pathology , Autophagy/drug effects , Autophagy/genetics , Brain/drug effects , Brain/pathology , Cell Movement/drug effects , Disease Models, Animal , Embryo, Mammalian , Ethylnitrosourea/toxicity , Female , Gene Expression Regulation, Developmental/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurodevelopmental Disorders/chemically induced , Neurodevelopmental Disorders/diagnostic imaging , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/pathology , Neurons/drug effects , Neurons/ultrastructure , Signal Transduction/drug effects , Signal Transduction/genetics , Vacuolar Proton-Translocating ATPases/drug effects
9.
Mol Cell Neurosci ; 84: 58-67, 2017 10.
Article in English | MEDLINE | ID: mdl-28347630

ABSTRACT

The development of the vertebrate central nervous system is reliant on a complex cascade of biological processes that include mitotic division, relocation of migrating neurons, and the extension of dendritic and axonal processes. Each of these cellular events requires the diverse functional repertoire of the microtubule cytoskeleton for the generation of forces, assembly of macromolecular complexes and transport of molecules and organelles. The tubulins are a multi-gene family that encode for the constituents of microtubules, and have been implicated in a spectrum of neurological disorders. Evidence is building that different tubulins tune the functional properties of the microtubule cytoskeleton dependent on the cell type, developmental profile and subcellular localisation. Here we review of the origins of the functional specification of the tubulin gene family in the developing brain at a transcriptional, translational, and post-transcriptional level. We remind the reader that tubulins are not just loading controls for your average Western blot.


Subject(s)
Brain/growth & development , Cytoskeleton/metabolism , Microtubules/metabolism , Tubulin/metabolism , Animals , Humans , Protein Processing, Post-Translational/physiology , Proteomics
10.
Hum Mol Genet ; 26(2): 258-269, 2017 01 15.
Article in English | MEDLINE | ID: mdl-28013290

ABSTRACT

The integrity and dynamic properties of the microtubule cytoskeleton are indispensable for the development of the mammalian brain. Consequently, mutations in the genes that encode the structural component (the α/ß-tubulin heterodimer) can give rise to severe, sporadic neurodevelopmental disorders. These are commonly referred to as the tubulinopathies. Here we report the addition of recessive quadrupedalism, also known as Uner Tan syndrome (UTS), to the growing list of diseases caused by tubulin variants. Analysis of a consanguineous UTS family identified a biallelic TUBB2B mutation, resulting in a p.R390Q amino acid substitution. In addition to the identifying quadrupedal locomotion, all three patients showed severe cerebellar hypoplasia. None, however, displayed the basal ganglia malformations typically associated with TUBB2B mutations. Functional analysis of the R390Q substitution revealed that it did not affect the ability of ß-tubulin to fold or become assembled into the α/ß-heterodimer, nor did it influence the incorporation of mutant-containing heterodimers into microtubule polymers. The 390Q mutation in S. cerevisiae TUB2 did not affect growth under basal conditions, but did result in increased sensitivity to microtubule-depolymerizing drugs, indicative of a mild impact of this mutation on microtubule function. The TUBB2B mutation described here represents an unusual recessive mode of inheritance for missense-mediated tubulinopathies and reinforces the sensitivity of the developing cerebellum to microtubule defects.


Subject(s)
Cerebellum/abnormalities , Malformations of Cortical Development/genetics , Microtubules/genetics , Nervous System Malformations/genetics , Tubulin/genetics , Adult , Amino Acid Substitution/genetics , Basal Ganglia/pathology , Brain/growth & development , Brain/pathology , Cerebellum/physiopathology , Developmental Disabilities/genetics , Developmental Disabilities/physiopathology , Female , Homozygote , Humans , Male , Malformations of Cortical Development/physiopathology , Microtubules/pathology , Mutation , Nervous System Malformations/physiopathology , Phenotype , Saccharomyces cerevisiae/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...