Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 23
1.
Proc Natl Acad Sci U S A ; 121(17): e2306382121, 2024 Apr 23.
Article En | MEDLINE | ID: mdl-38640347

Hippocampal somatostatin-expressing (Sst) GABAergic interneurons (INs) exhibit considerable anatomical and functional heterogeneity. Recent single-cell transcriptome analyses have provided a comprehensive Sst-IN subpopulations census, a plausible molecular ground truth of neuronal identity whose links to specific functionality remain incomplete. Here, we designed an approach to identify and access subpopulations of Sst-INs based on transcriptomic features. Four mouse models based on single or combinatorial Cre- and Flp- expression differentiated functionally distinct subpopulations of CA1 hippocampal Sst-INs that largely tiled the morpho-functional parameter space of the Sst-INs superfamily. Notably, the Sst;;Tac1 intersection revealed a population of bistratified INs that preferentially synapsed onto fast-spiking interneurons (FS-INs) and were sufficient to interrupt their firing. In contrast, the Ndnf;;Nkx2-1 intersection identified a population of oriens lacunosum-moleculare INs that predominantly targeted CA1 pyramidal neurons, avoiding FS-INs. Overall, our results provide a framework to translate neuronal transcriptomic identity into discrete functional subtypes that capture the diverse specializations of hippocampal Sst-INs.


Hippocampus , Interneurons , Mice , Animals , Interneurons/physiology , Hippocampus/metabolism , Neurons/metabolism , Pyramidal Cells/metabolism , Somatostatin/genetics , Somatostatin/metabolism
2.
Cell Rep ; 43(4): 113839, 2024 Apr 23.
Article En | MEDLINE | ID: mdl-38507409

Homeostatic regulation of synapses is vital for nervous system function and key to understanding a range of neurological conditions. Synaptic homeostasis is proposed to operate over hours to counteract the destabilizing influence of long-term potentiation (LTP) and long-term depression (LTD). The prevailing view holds that synaptic scaling is a slow first-order process that regulates postsynaptic glutamate receptors and fundamentally differs from LTP or LTD. Surprisingly, we find that the dynamics of scaling induced by neuronal inactivity are not exponential or monotonic, and the mechanism requires calcineurin and CaMKII, molecules dominant in LTD and LTP. Our quantitative model of these enzymes reconstructs the unexpected dynamics of homeostatic scaling and reveals how synapses can efficiently safeguard future capacity for synaptic plasticity. This mechanism of synaptic adaptation supports a broader set of homeostatic changes, including action potential autoregulation, and invites further inquiry into how such a mechanism varies in health and disease.


Calcineurin , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Homeostasis , Synapses , Animals , Synapses/metabolism , Synapses/physiology , Calcineurin/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Long-Term Potentiation/physiology , Neuronal Plasticity/physiology , Long-Term Synaptic Depression/physiology , Neurons/metabolism , Neurons/physiology , Mice
3.
bioRxiv ; 2023 Apr 27.
Article En | MEDLINE | ID: mdl-37162922

Hippocampal somatostatin-expressing (Sst) GABAergic interneurons (INs) exhibit considerable anatomical and functional heterogeneity. Recent single cell transcriptome analyses have provided a comprehensive Sst-IN subtype census, a plausible molecular ground truth of neuronal identity whose links to specific functionality remain incomplete. Here, we designed an approach to identify and access subpopulations of Sst-INs based on transcriptomic features. Four mouse models based on single or combinatorial Cre- and Flp- expression differentiated functionally distinct subpopulations of CA1 hippocampal Sst-INs that largely tiled the morpho-functional parameter space of the Sst-INs superfamily. Notably, the Sst;;Tac1 intersection revealed a population of bistratified INs that preferentially synapsed onto fast-spiking interneurons (FS-INs) and were both necessary and sufficient to interrupt their firing. In contrast, the Ndnf;;Nkx2-1 intersection identified a population of oriens lacunosum-moleculare (OLM) INs that predominantly targeted CA1 pyramidal neurons, avoiding FS-INs. Overall, our results provide a framework to translate neuronal transcriptomic identity into discrete functional subtypes that capture the diverse specializations of hippocampal Sst-INs.

4.
Cell ; 181(7): 1547-1565.e15, 2020 06 25.
Article En | MEDLINE | ID: mdl-32492405

Homeostasis of neural firing properties is important in stabilizing neuronal circuitry, but how such plasticity might depend on alternative splicing is not known. Here we report that chronic inactivity homeostatically increases action potential duration by changing alternative splicing of BK channels; this requires nuclear export of the splicing factor Nova-2. Inactivity and Nova-2 relocation were connected by a novel synapto-nuclear signaling pathway that surprisingly invoked mechanisms akin to Hebbian plasticity: Ca2+-permeable AMPA receptor upregulation, L-type Ca2+ channel activation, enhanced spine Ca2+ transients, nuclear translocation of a CaM shuttle, and nuclear CaMKIV activation. These findings not only uncover commonalities between homeostatic and Hebbian plasticity but also connect homeostatic regulation of synaptic transmission and neuronal excitability. The signaling cascade provides a full-loop mechanism for a classic autoregulatory feedback loop proposed ∼25 years ago. Each element of the loop has been implicated previously in neuropsychiatric disease.


Large-Conductance Calcium-Activated Potassium Channels/metabolism , Long-Term Potentiation/physiology , Nerve Tissue Proteins/metabolism , RNA-Binding Proteins/metabolism , Action Potentials/physiology , Alternative Splicing/genetics , Alternative Splicing/physiology , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 1/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Female , HEK293 Cells , Homeostasis/physiology , Humans , Large-Conductance Calcium-Activated Potassium Channels/genetics , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/physiology , Neuro-Oncological Ventral Antigen , Neuronal Plasticity/physiology , Neurons/metabolism , RNA-Binding Proteins/physiology , Rats , Rats, Sprague-Dawley , Signal Transduction , Synapses/metabolism , Synaptic Transmission/physiology
5.
Neuron ; 100(6): 1354-1368.e5, 2018 12 19.
Article En | MEDLINE | ID: mdl-30449657

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.


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
6.
Am J Hum Genet ; 101(6): 1006-1012, 2017 Dec 07.
Article En | MEDLINE | ID: mdl-29198720

Leber congenital amaurosis (LCA) is a neurodegenerative disease of photoreceptor cells that causes blindness within the first year of life. It occasionally occurs in syndromic metabolic diseases and plurisystemic ciliopathies. Using exome sequencing in a multiplex family and three simplex case subjects with an atypical association of LCA with early-onset hearing loss, we identified two heterozygous mutations affecting Arg391 in ß-tubulin 4B isotype-encoding (TUBB4B). Inspection of the atomic structure of the microtubule (MT) protofilament reveals that the ß-tubulin Arg391 residue contributes to a binding pocket that interacts with α-tubulin contained in the longitudinally adjacent αß-heterodimer, consistent with a role in maintaining MT stability. Functional analysis in cultured cells overexpressing FLAG-tagged wild-type or mutant TUBB4B as well as in primary skin-derived fibroblasts showed that the mutant TUBB4B is able to fold, form αß-heterodimers, and co-assemble into the endogenous MT lattice. However, the dynamics of growing MTs were consistently altered, showing that the mutations have a significant dampening impact on normal MT growth. Our findings provide a link between sensorineural disease and anomalies in MT behavior and describe a syndromic LCA unrelated to ciliary dysfunction.


Leber Congenital Amaurosis/genetics , Microtubules/genetics , Tubulin/genetics , Adult , Binding Sites/genetics , Cells, Cultured , Child , DNA Mutational Analysis , Female , Humans , Male , Microtubules/metabolism , Middle Aged , Mutation, Missense/genetics , Photoreceptor Cells/metabolism , Tubulin/metabolism , Exome Sequencing
7.
Hum Mol Genet ; 26(2): 258-269, 2017 01 15.
Article En | MEDLINE | ID: mdl-28013290

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.


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
8.
Mol Genet Genomic Med ; 4(6): 599-603, 2016 Nov.
Article En | MEDLINE | ID: mdl-27896282

BACKGROUND: Microtubules are dynamic polymers of α/ß tubulin heterodimers that play a critical role in cerebral cortical development, by regulating neuronal migration, differentiation, and morphogenesis. Mutations in genes that encode either α- or ß-tubulin or a spectrum of proteins involved in the regulation of microtubule dynamics lead to clinically devastating malformations of cortical development, including lissencephaly. METHODS: This is a single case report or a patient with lissencephaly, developmental delay, nystagmus, persistent hyperplastic primary vitreous, and infantile spasms, and undertook a neurogenetic workup. We include studies of mutant function in Escherichia coli and HeLa cells. RESULTS: The patient was found to have a novel de novo mutation in kinesin family member 2A (KIF2A). This mutation results in a substitution of isoleucine at a highly conserved threonine residue within the ATP-binding domain. The KIF2A p.Thr320Ile mutant protein exhibited abnormal solubility, and KIF2A p.Thr320Ile overexpression in cultured cells led to the formation of aberrant microtubule networks. CONCLUSION: Findings support the pathogenic link between KIF2A mutation and lissencephaly, and expand the range of presentation to include infantile spasms and congenital anomalies.

9.
J Med Genet ; 53(10): 662-71, 2016 10.
Article En | MEDLINE | ID: mdl-27273344

BACKGROUND: TUBB8 is a primate-specific ß-tubulin isotype whose expression is confined to oocytes and the early embryo. We previously found that mutations in TUBB8 caused oocyte maturation arrest. The objective was to describe newly discovered mutations in TUBB8 and to characterise the accompanying spectrum of phenotypes and modes of inheritance. METHODS AND RESULTS: Patients with oocyte maturation arrest were sequenced with respect to TUBB8. We investigated the effects of identified mutations in vitro, in cultured cells and in mouse oocytes. Seven heterozygous missense and two homozygous mutations were identified. These mutations cause a range of folding defects in vitro, different degrees of microtubule disruption upon expression in cultured cells and interfere to varying extents in the proper assembly of the meiotic spindle in mouse oocytes. Several of the newly discovered TUBB8 mutations result in phenotypic variability. For example, oocytes harbouring any of three missense mutations (I210V, T238M and N348S) could extrude the first polar body. Moreover, they could be fertilised, although the ensuing embryos became developmentally arrested. Surprisingly, oocytes from patients harbouring homozygous TUBB8 mutations that in either case preclude the expression of a functional TUBB8 polypeptide nonetheless contained identifiable spindles. CONCLUSIONS: Our data substantially expand the range of dysfunctional oocyte phenotypes incurred by mutation in TUBB8, underscore the independent nature of human oocyte meiosis and differentiation, extend the class of genetic diseases known as the tubulinopathies and provide new criteria for the qualitative evaluation of meiosis II (MII) oocytes for in vitro fertilization (IVF).


Infertility, Female/metabolism , Mutation , Oocytes/metabolism , Phenotype , Tubulin/genetics , Animals , Embryo, Mammalian/metabolism , Female , Humans , Infertility, Female/genetics , Mice , Spindle Apparatus
10.
N Engl J Med ; 374(3): 223-32, 2016 Jan 21.
Article En | MEDLINE | ID: mdl-26789871

Background Human reproduction depends on the fusion of a mature oocyte with a sperm cell to form a fertilized egg. The genetic events that lead to the arrest of human oocyte maturation are unknown. Methods We sequenced the exomes of five members of a four-generation family, three of whom had infertility due to oocyte meiosis I arrest. We performed Sanger sequencing of a candidate gene, TUBB8, in DNA samples from these members, additional family members, and members of 23 other affected families. The expression of TUBB8 and all other ß-tubulin isotypes was assessed in human oocytes, early embryos, sperm cells, and several somatic tissues by means of a quantitative reverse-transcriptase-polymerase-chain-reaction assay. We evaluated the effect of the TUBB8 mutations on the assembly of the heterodimer consisting of one α-tubulin polypeptide and one ß-tubulin polypeptide (α/ß-tubulin heterodimer) in vitro, on microtubule architecture in HeLa cells, on microtubule dynamics in yeast cells, and on spindle assembly in mouse and human oocytes. Results We identified seven mutations in the primate-specific gene TUBB8 that were responsible for oocyte meiosis I arrest in 7 of the 24 families. TUBB8 expression is unique to oocytes and the early embryo, in which this gene accounts for almost all the expressed ß-tubulin. The mutations affect chaperone-dependent folding and assembly of the α/ß-tubulin heterodimer, disrupt microtubule behavior on expression in cultured cells, alter microtubule dynamics in vivo, and cause catastrophic spindle-assembly defects and maturation arrest on expression in mouse and human oocytes. Conclusions TUBB8 mutations have dominant-negative effects that disrupt microtubule behavior and oocyte meiotic spindle assembly and maturation, causing female infertility. (Funded by the National Basic Research Program of China and others.).


Infertility, Female/genetics , Meiosis/genetics , Microtubules/pathology , Mutation , Oocytes/physiology , Spindle Apparatus/physiology , Tubulin/genetics , Adult , Animals , Female , Humans , Meiosis/physiology , Mice , Microtubules/physiology , RNA
11.
Hum Mol Genet ; 25(21): 4635-4648, 2016 11 01.
Article En | MEDLINE | ID: mdl-28158450

Mutation in a growing spectrum of genes is known to either cause or contribute to primary or secondary microcephaly. In primary microcephaly the genetic determinants frequently involve mutations that contribute to or modulate the microtubule cytoskeleton by causing perturbations of neuronal proliferation and migration. Here we describe four patients from two unrelated families each with an infantile neurodegenerative disorder characterized by loss of developmental milestones at 9­24 months of age followed by seizures, dystonia and acquired microcephaly. The patients harboured homozygous missense mutations (A475T and A586V) in TBCD, a gene encoding one of five tubulin-specific chaperones (termed TBCA-E) that function in concert as a nanomachine required for the de novo assembly of the α/ß tubulin heterodimer. The latter is the subunit from which microtubule polymers are assembled. We found a reduced intracellular abundance of TBCD in patient fibroblasts to about 10% (in the case of A475T) or 40% (in the case of A586V) compared to age-matched wild type controls. Functional analyses of the mutant proteins revealed a partially compromised ability to participate in the heterodimer assembly pathway. We show via in utero shRNA-mediated suppression that a balanced supply of tbcd is critical for cortical cell proliferation and radial migration in the developing mouse brain. We conclude that TBCD is a novel functional contributor to the mammalian cerebral cortex development, and that the pathological mechanism resulting from the mutations we describe is likely to involve compromised interactions with one or more TBCD-interacting effectors that influence the dynamics and behaviour of the neuronal cytoskeleton.


Heredodegenerative Disorders, Nervous System/genetics , Microcephaly/genetics , Microtubule-Associated Proteins/genetics , Animals , Brain/metabolism , Cytoskeleton/metabolism , Fibroblasts/metabolism , Heredodegenerative Disorders, Nervous System/metabolism , Humans , Infant , Infant, Newborn , Mice , Mice, Inbred C57BL/embryology , Microtubule-Associated Proteins/metabolism , Microtubules/genetics , Microtubules/physiology , Tubulin/genetics , Tubulin/metabolism , Exome Sequencing/methods
12.
Am J Hum Genet ; 97(6): 790-800, 2015 Dec 03.
Article En | MEDLINE | ID: mdl-26637975

Circumferential skin creases Kunze type (CSC-KT) is a specific congenital entity with an unknown genetic cause. The disease phenotype comprises characteristic circumferential skin creases accompanied by intellectual disability, a cleft palate, short stature, and dysmorphic features. Here, we report that mutations in either MAPRE2 or TUBB underlie the genetic origin of this syndrome. MAPRE2 encodes a member of the microtubule end-binding family of proteins that bind to the guanosine triphosphate cap at growing microtubule plus ends, and TUBB encodes a ß-tubulin isotype that is expressed abundantly in the developing brain. Functional analyses of the TUBB mutants show multiple defects in the chaperone-dependent tubulin heterodimer folding and assembly pathway that leads to a compromised yield of native heterodimers. The TUBB mutations also have an impact on microtubule dynamics. For MAPRE2, we show that the mutations result in enhanced MAPRE2 binding to microtubules, implying an increased dwell time at microtubule plus ends. Further, in vivo analysis of MAPRE2 mutations in a zebrafish model of craniofacial development shows that the variants most likely perturb the patterning of branchial arches, either through excessive activity (under a recessive paradigm) or through haploinsufficiency (dominant de novo paradigm). Taken together, our data add CSC-KT to the growing list of tubulinopathies and highlight how multiple inheritance paradigms can affect dosage-sensitive biological systems so as to result in the same clinical defect.


Brain/metabolism , Cutis Laxa/congenital , Hamartoma/genetics , Microtubule-Associated Proteins/genetics , Microtubules/genetics , Mutation , Skin Abnormalities/genetics , Skin/metabolism , Tubulin/genetics , Adolescent , Animals , Brain/growth & development , Brain/pathology , Child , Cutis Laxa/genetics , Cutis Laxa/metabolism , Cutis Laxa/pathology , Female , Gene Dosage , Gene Expression Regulation, Developmental , Genes, Recessive , Hamartoma/metabolism , Hamartoma/pathology , Haploinsufficiency , Humans , Infant , Inheritance Patterns , Male , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Microtubules/pathology , Protein Folding , Protein Multimerization , Skin/growth & development , Skin/pathology , Skin Abnormalities/metabolism , Skin Abnormalities/pathology , Tubulin/metabolism , Young Adult , Zebrafish
13.
Methods Cell Biol ; 115: 155-71, 2013.
Article En | MEDLINE | ID: mdl-23973072

The tubulin heterodimer consists of one α- and one ß-tubulin polypeptide. Neither protein can partition to the native state or assemble into polymerization competent heterodimers without the concerted action of a series of chaperone proteins including five tubulin-specific chaperones (TBCs) termed TBCA-TBCE. TBCA and TBCB bind to and stabilize newly synthesized quasi-native ß- and α-tubulin polypeptides, respectively, following their generation via multiple rounds of ATP-dependent interaction with the cytosolic chaperonin. There is free exchange of ß-tubulin between TBCA and TBCD, and of α-tubulin between TBCB and TBCE, resulting in the formation of TBCD/ß and TBCE/α, respectively. The latter two complexes interact, forming a supercomplex (TBCE/α/TBCD/ß). Discharge of the native α/ß heterodimer occurs via interaction of the supercomplex with TBCC, which results in the triggering of TBC-bound ß-tubulin (E-site) GTP hydrolysis. This reaction acts as a switch for disassembly of the supercomplex and the release of E-site GDP-bound heterodimer, which becomes polymerization competent following spontaneous exchange with GTP. The tubulin-specific chaperones thus function together as a tubulin assembly machine, marrying the α- and ß-tubulin subunits into a tightly associated heterodimer. The existence of this evolutionarily conserved pathway explains why it has never proved possible to isolate α- or ß-tubulin as stable independent entities in the absence of their cognate partners, and implies that each exists and is maintained in the heterodimer in a nonminimal energy state. Here, we describe methods for the purification of recombinant TBCs as biologically active proteins following their expression in a variety of host/vector systems.


Molecular Chaperones/analysis , Tubulin/biosynthesis , Tubulin/metabolism , Cell Line, Tumor , Chromatography/methods , HeLa Cells , Humans , Microtubule-Associated Proteins/metabolism , Molecular Chaperones/metabolism , Protein Multimerization , Sf9 Cells/metabolism
14.
Nat Genet ; 45(6): 639-47, 2013 Jun.
Article En | MEDLINE | ID: mdl-23603762

The genetic causes of malformations of cortical development (MCD) remain largely unknown. Here we report the discovery of multiple pathogenic missense mutations in TUBG1, DYNC1H1 and KIF2A, as well as a single germline mosaic mutation in KIF5C, in subjects with MCD. We found a frequent recurrence of mutations in DYNC1H1, implying that this gene is a major locus for unexplained MCD. We further show that the mutations in KIF5C, KIF2A and DYNC1H1 affect ATP hydrolysis, productive protein folding and microtubule binding, respectively. In addition, we show that suppression of mouse Tubg1 expression in vivo interferes with proper neuronal migration, whereas expression of altered γ-tubulin proteins in Saccharomyces cerevisiae disrupts normal microtubule behavior. Our data reinforce the importance of centrosomal and microtubule-related proteins in cortical development and strongly suggest that microtubule-dependent mitotic and postmitotic processes are major contributors to the pathogenesis of MCD.


Cytoplasmic Dyneins/genetics , Kinesins/genetics , Microcephaly/genetics , Mutation, Missense , Tubulin/genetics , Animals , COS Cells , Cell Movement , Chlorocebus aethiops , Exome , Genetic Association Studies , Germ-Line Mutation , Humans , Lissencephaly/genetics , Lissencephaly/pathology , Magnetic Resonance Imaging , Malformations of Cortical Development/genetics , Malformations of Cortical Development/pathology , Mice , Microcephaly/pathology , Models, Molecular , Neuroimaging , Pedigree , Sequence Analysis, DNA
15.
Cell Rep ; 2(6): 1554-62, 2012 Dec 27.
Article En | MEDLINE | ID: mdl-23246003

The formation of the mammalian cortex requires the generation, migration, and differentiation of neurons. The vital role that the microtubule cytoskeleton plays in these cellular processes is reflected by the discovery that mutations in various tubulin isotypes cause different neurodevelopmental diseases, including lissencephaly (TUBA1A), polymicrogyria (TUBA1A, TUBB2B, TUBB3), and an ocular motility disorder (TUBB3). Here, we show that Tubb5 is expressed in neurogenic progenitors in the mouse and that its depletion in vivo perturbs the cell cycle of progenitors and alters the position of migrating neurons. We report the occurrence of three microcephalic patients with structural brain abnormalities harboring de novo mutations in TUBB5 (M299V, V353I, and E401K). These mutant proteins, which affect the chaperone-dependent assembly of tubulin heterodimers in different ways, disrupt neurogenic division and/or migration in vivo. Our results provide insight into the functional repertoire of the tubulin gene family, specifically implicating TUBB5 in embryonic neurogenesis and microcephaly.


Brain/abnormalities , Brain/metabolism , Microcephaly/metabolism , Mutation, Missense , Neural Stem Cells/metabolism , Tubulin/metabolism , Amino Acid Substitution , Animals , Brain/embryology , Brain/pathology , Female , Humans , Male , Mice , Mice, Mutant Strains , Microcephaly/embryology , Microcephaly/genetics , Microcephaly/pathology , Neural Stem Cells/pathology , Neurogenesis/genetics , Tubulin/genetics
16.
Cytoskeleton (Hoboken) ; 67(11): 706-14, 2010 Nov.
Article En | MEDLINE | ID: mdl-20740604

Assembly of the α/ß tubulin heterodimer requires the participation of a series of chaperone proteins (TBCA-E) that function downstream of the cytosolic chaperonin (CCT) as a heterodimer assembly machine. TBCD and TBCE are also capable of acting in a reverse reaction in which they disrupt native heterodimers. Homologs of TBCA-E exist in all eukaryotes, and the amino acid sequences of α- and ß-tubulin isotypes are rigidly conserved among vertebrates. However, the efficiency with which TBCD effects tubulin disruption in vivo depends on its origin: bovine (but not human) TBCD efficiently destroys tubulin and microtubules upon overexpression in cultured cells. Here we show that recombinant bovine TBCD is produced in HeLa cells as a stoichiometric cocomplex with ß-tubulin, consistent with its behavior in vitro and in vivo. In contrast, expression of human TBCD using the same host/vector system results in the generation of TBCD that is not complexed with ß-tubulin. We show that recombinant human TBCD functions indistinguishably from its nonrecombinant bovine counterpart in in vitro CCT-driven folding reactions, in tubulin disruption reactions, and in tubulin GTPase activating protein assays in which TBCD and TBCC stimulate GTP hydrolysis by ß-tubulin at a heterodimer concentration far below that required for polymerization into microtubules. We conclude that bovine and human TBCD have functionally identical roles in de novo tubulin heterodimer assembly, and show that the inability of human TBCD to disrupt microtubule integrity upon overexpression in vivo can be overcome by siRNA-mediated suppression of expression of the TBCD regulator Arl2 (ADP ribosylation factor-like protein).


GTP-Binding Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Tubulin/metabolism , Animals , Cattle , GTP-Binding Proteins/genetics , HeLa Cells , Humans , Microtubule-Associated Proteins/genetics , Molecular Chaperones/metabolism , Protein Conformation , Protein Folding , Protein Isoforms/chemistry , Protein Isoforms/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Tubulin/chemistry
17.
Hum Mol Genet ; 19(18): 3599-613, 2010 Sep 15.
Article En | MEDLINE | ID: mdl-20603323

Malformations of cortical development are characteristic of a plethora of diseases that includes polymicrogyria, periventricular and subcortical heterotopia and lissencephaly. Mutations in TUBA1A and TUBB2B, each a member of the multigene families that encode alpha- and beta-tubulins, have recently been implicated in these diseases. Here we examine the defects that result from nine disease-causing mutations (I188L, I238V, P263T, L286F, V303G, L397P, R402C, 402H, S419L) in TUBA1A. We show that the expression of all the mutant proteins in vitro results in the generation of tubulin heterodimers in varying yield and that these can co-polymerize with microtubules in vitro. We identify several kinds of defects that result from these mutations. Among these are various defects in the chaperone-dependent pathway leading to de novo tubulin heterodimer formation. These include a defective interaction with the chaperone prefoldin, a reduced efficiency in the generation of productive folding intermediates as a result of inefficient interaction with the cytosolic chaperonin, CCT, and, in several cases, a failure to stably interact with TBCB, one of five tubulin-specific chaperones that act downstream of CCT in the tubulin heterodimer assembly pathway. Other defects include structural instability in vitro, diminished stability in vivo, a compromised ability to co-assemble with microtubules in vivo and a suppression of microtubule growth rate in the neurites (but not the soma) of cultured neurons. Our data are consistent with the notion that some mutations in TUBA1A result in tubulin deficit, whereas others reflect compromised interactions with one or more MAPs that are essential to proper neuronal migration.


Malformations of Cortical Development/genetics , Mutation, Missense , Tubulin/chemistry , Tubulin/genetics , Animals , COS Cells , Cell Line , Chlorocebus aethiops , Dimerization , Humans , Malformations of Cortical Development/metabolism , Mice , Molecular Conformation , Mutation , Protein Folding , Protein Stability , Tubulin/metabolism
18.
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
19.
Mol Biol Cell ; 19(3): 1152-61, 2008 Mar.
Article En | MEDLINE | ID: mdl-18199681

The agyria (lissencephaly)/pachygyria phenotypes are catastrophic developmental diseases characterized by abnormal folds on the surface of the brain and disorganized cortical layering. In addition to mutations in at least four genes--LIS1, DCX, ARX and RELN--mutations in a human alpha-tubulin gene, TUBA1A, have recently been identified that cause these diseases. Here, we show that one such mutation, R264C, leads to a diminished capacity of de novo tubulin heterodimer formation. We identify the mechanisms that contribute to this defect. First, there is a reduced efficiency whereby quasinative alpha-tubulin folding intermediates are generated via ATP-dependent interaction with the cytosolic chaperonin CCT. Second, there is a failure of CCT-generated folding intermediates to stably interact with TBCB, one of the five tubulin chaperones (TBCA-E) that participate in the pathway leading to the de novo assembly of the tubulin heterodimer. We describe the behavior of the R264C mutation in terms of its effect on the structural integrity of alpha-tubulin and its interaction with TBCB. In spite of its compromised folding efficiency, R264C molecules that do productively assemble into heterodimers are capable of copolymerizing into dynamic microtubules in vivo. The diminished production of TUBA1A tubulin in R264C individuals is consistent with haploinsufficiency as a cause of the disease phenotype.


Chaperonins/metabolism , Lissencephaly/genetics , Microtubule-Associated Proteins/metabolism , Mutation/genetics , Tubulin/genetics , Animals , Cattle , Chaperonin Containing TCP-1 , Dimerization , Guanosine Triphosphate/metabolism , HeLa Cells , Humans , Microtubules/metabolism , Mutant Proteins/metabolism , Protein Binding , Protein Processing, Post-Translational , Protein Structure, Tertiary , Reelin Protein , Transcription, Genetic , Tubulin/chemistry
20.
Cell ; 128(1): 45-57, 2007 Jan 12.
Article En | MEDLINE | ID: mdl-17218254

The development of the mammalian brain is dependent on extensive neuronal migration. Mutations in mice and humans that affect neuronal migration result in abnormal lamination of brain structures with associated behavioral deficits. Here, we report the identification of a hyperactive N-ethyl-N-nitrosourea (ENU)-induced mouse mutant with abnormalities in the laminar architecture of the hippocampus and cortex, accompanied by impaired neuronal migration. We show that the causative mutation lies in the guanosine triphosphate (GTP) binding pocket of alpha-1 tubulin (Tuba1) and affects tubulin heterodimer formation. Phenotypic similarity with existing mouse models of lissencephaly led us to screen a cohort of patients with developmental brain anomalies. We identified two patients with de novo mutations in TUBA3, the human homolog of Tuba1. This study demonstrates the utility of ENU mutagenesis in the mouse as a means to discover the basis of human neurodevelopmental disorders.


Cell Movement , Cerebral Cortex/abnormalities , Hippocampus/abnormalities , Mutation/genetics , Neurons/pathology , Tubulin/genetics , Tubulin/metabolism , Amino Acid Sequence , Animals , Anxiety/genetics , Anxiety/pathology , Behavior, Animal , Cerebral Cortex/pathology , Chromosome Mapping , DNA Mutational Analysis , Dimerization , Female , Glutamic Acid/genetics , Guanosine Triphosphate/metabolism , Hippocampus/pathology , Humans , Male , Memory Disorders/genetics , Memory Disorders/pathology , Mice , Mice, Mutant Strains , Molecular Sequence Data , Phenotype , Serine/genetics , Tubulin/chemistry
...