Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 20
1.
Nat Cell Biol ; 2024 May 07.
Article En | MEDLINE | ID: mdl-38714853

During brain development, neural progenitors expand through symmetric divisions before giving rise to differentiating cell types via asymmetric divisions. Transition between those modes varies among individual neural stem cells, resulting in clones of different sizes. Imaging-based lineage tracing allows for lineage analysis at high cellular resolution but systematic approaches to analyse clonal behaviour of entire tissues are currently lacking. Here we implement whole-tissue lineage tracing by genomic DNA barcoding in 3D human cerebral organoids, to show that individual stem cell clones produce progeny on a vastly variable scale. By using stochastic modelling we find that variable lineage sizes arise because a subpopulation of lineages retains symmetrically dividing cells. We show that lineage sizes can adjust to tissue demands after growth perturbation via chemical ablation or genetic restriction of a subset of cells in chimeric organoids. Our data suggest that adaptive plasticity of stem cell populations ensures robustness of development in human brain organoids.

2.
Nat Cell Biol ; 2024 Mar 28.
Article En | MEDLINE | ID: mdl-38548890

The human neocortex has undergone strong evolutionary expansion, largely due to an increased progenitor population, the basal radial glial cells. These cells are responsible for the production of a diversity of cell types, but the successive cell fate decisions taken by individual progenitors remain unknown. Here we developed a semi-automated live/fixed correlative imaging method to map basal radial glial cell division modes in early fetal tissue and cerebral organoids. Through the live analysis of hundreds of dividing progenitors, we show that basal radial glial cells undergo abundant symmetric amplifying divisions, and frequent self-consuming direct neurogenic divisions, bypassing intermediate progenitors. These direct neurogenic divisions are more abundant in the upper part of the subventricular zone. We furthermore demonstrate asymmetric Notch activation in the self-renewing daughter cells, independently of basal fibre inheritance. Our results reveal a remarkable conservation of fate decisions in cerebral organoids, supporting their value as models of early human neurogenesis.

3.
Curr Opin Neurobiol ; 80: 102709, 2023 06.
Article En | MEDLINE | ID: mdl-37003105

A high number of genetic mutations associated with cortical malformations are found in genes coding for microtubule-related factors. This has stimulated research to understand how the various microtubule-based processes are regulated to build a functional cerebral cortex. Here, we focus our review on the radial glial progenitor cells, the stem cells of the developing neocortex, summarizing research mostly performed in rodents and humans. We highlight how the centrosomal and acentrosomal microtubule networks are organized during interphase to support polarized transport and proper attachment of the apical and basal processes. We describe the molecular mechanism for interkinetic nuclear migration (INM), a microtubule-dependent oscillation of the nucleus. Finally, we describe how the mitotic spindle is built to ensure proper chromosome segregation, with a strong focus on factors mutated in microcephaly.


Microtubules , Neocortex , Humans , Spindle Apparatus , Stem Cells , Cell Nucleus
5.
EMBO Rep ; 23(10): e54605, 2022 10 06.
Article En | MEDLINE | ID: mdl-35979738

Radial glial (RG) cells are the neural stem cells of the developing neocortex. Apical RG (aRG) cells can delaminate to generate basal RG (bRG) cells, a cell type associated with human brain expansion. Here, we report that aRG delamination is regulated by the post-Golgi secretory pathway. Using in situ subcellular live imaging, we show that post-Golgi transport of RAB6+ vesicles occurs toward the minus ends of microtubules and depends on dynein. We demonstrate that the apical determinant Crumbs3 (CRB3) is also transported by dynein. Double knockout of RAB6A/A' and RAB6B impairs apical localization of CRB3 and induces a retraction of aRG cell apical process, leading to delamination and ectopic division. These defects are phenocopied by knockout of the dynein activator LIS1. Overall, our results identify a RAB6-dynein-LIS1 complex for Golgi to apical surface transport in aRG cells, and highlights the role of this pathway in the maintenance of neuroepithelial integrity.


Dyneins , rab GTP-Binding Proteins , Dyneins/genetics , Dyneins/metabolism , Golgi Apparatus/metabolism , Humans , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Neurons/metabolism , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism
6.
Cells ; 11(14)2022 07 07.
Article En | MEDLINE | ID: mdl-35883578

How the brain develops and achieves its final size is a fascinating issue that questions cortical evolution across species and man's place in the animal kingdom. Although animal models have so far been highly valuable in understanding the key steps of cortical development, many human specificities call for appropriate models. In particular, microcephaly, a neurodevelopmental disorder that is characterized by a smaller head circumference has been challenging to model in mice, which often do not fully recapitulate the human phenotype. The relatively recent development of brain organoid technology from induced pluripotent stem cells (iPSCs) now makes it possible to model human microcephaly, both due to genetic and environmental origins, and to generate developing cortical tissue from the patients themselves. These 3D tissues rely on iPSCs differentiation into cortical progenitors that self-organize into neuroepithelial rosettes mimicking the earliest stages of human neurogenesis in vitro. Over the last ten years, numerous protocols have been developed to control the identity of the induced brain areas, the reproducibility of the experiments and the longevity of the cultures, allowing analysis of the later stages. In this review, we describe the different approaches that instruct human iPSCs to form cortical organoids, summarize the different microcephalic conditions that have so far been modeled by organoids, and discuss the relevance of this model to decipher the cellular and molecular mechanisms of primary and secondary microcephalies.


Induced Pluripotent Stem Cells , Microcephaly , Animals , Humans , Mice , Neurogenesis , Organoids , Reproducibility of Results
7.
Nat Commun ; 13(1): 16, 2022 01 10.
Article En | MEDLINE | ID: mdl-35013230

Primary microcephaly and megalencephaly are severe brain malformations defined by reduced and increased brain size, respectively. Whether these two pathologies arise from related alterations at the molecular level is unclear. Microcephaly has been largely associated with centrosomal defects, leading to cell death. Here, we investigate the consequences of WDR81 loss of function, which causes severe microcephaly in patients. We show that WDR81 regulates endosomal trafficking of EGFR and that loss of function leads to reduced MAP kinase pathway activation. Mouse radial glial progenitor cells knocked-out for WDR81 exhibit reduced proliferation rate, subsequently leading to reduced brain size. These proliferation defects are rescued in vivo by expressing a megalencephaly-causing mutant form of Cyclin D2. Our results identify the endosomal machinery as an important regulator of proliferation rates and brain growth, demonstrating that microcephaly and megalencephaly can be caused by opposite effects on the proliferation rate of radial glial progenitors.


Cell Proliferation , Microcephaly , Nerve Tissue Proteins/metabolism , Neural Stem Cells/metabolism , Transport Vesicles , Animals , Brain/embryology , Brain/metabolism , Cells, Cultured , Endosomes/metabolism , Green Fluorescent Proteins/metabolism , Humans , MAP Kinase Signaling System , Megalencephaly/etiology , Megalencephaly/metabolism , Megalencephaly/pathology , Mice , Microcephaly/etiology , Microcephaly/metabolism , Microcephaly/pathology , Nervous System Malformations/etiology , Nervous System Malformations/metabolism , Nervous System Malformations/pathology , Neuroglia/metabolism , Protein Transport/physiology , Transport Vesicles/metabolism , Transport Vesicles/pathology
8.
Nat Commun ; 12(1): 6298, 2021 11 02.
Article En | MEDLINE | ID: mdl-34728600

Basal progenitors (BPs), including intermediate progenitors and basal radial glia, are generated from apical radial glia and are enriched in gyrencephalic species like humans, contributing to neuronal expansion. Shortly after generation, BPs delaminate towards the subventricular zone, where they further proliferate before differentiation. Gene expression alterations involved in BP delamination and function in humans are poorly understood. Here, we study the role of LGALS3BP, so far known as a cancer biomarker, which is a secreted protein enriched in human neural progenitors (NPCs). We show that individuals with LGALS3BP de novo variants exhibit altered local gyrification, sulcal depth, surface area and thickness in their cortex. Additionally, using cerebral organoids, human fetal tissues and mice, we show that LGALS3BP regulates the position of NPCs. Single-cell RNA-sequencing and proteomics reveal that LGALS3BP-mediated mechanisms involve the extracellular matrix in NPCs' anchoring and migration within the human brain. We propose that its temporal expression influences NPCs' delamination, corticogenesis and gyrification extrinsically.


Antigens, Neoplasm/metabolism , Biomarkers, Tumor/metabolism , Cerebral Cortex/cytology , Extracellular Vesicles/metabolism , Induced Pluripotent Stem Cells/cytology , Neocortex/cytology , Neural Stem Cells/cytology , Neuroglia/metabolism , Animals , Cell Differentiation , Cerebral Cortex/metabolism , Female , Humans , Induced Pluripotent Stem Cells/metabolism , Lateral Ventricles/cytology , Lateral Ventricles/metabolism , Mice , Mice, Inbred C57BL , Models, Animal , Neocortex/metabolism , Neural Stem Cells/metabolism
9.
J Cell Biol ; 220(8)2021 08 02.
Article En | MEDLINE | ID: mdl-34019079

Neurons of the neocortex are generated by stem cells called radial glial cells. These polarized cells extend a short apical process toward the ventricular surface and a long basal fiber that acts as a scaffold for neuronal migration. How the microtubule cytoskeleton is organized in these cells to support long-range transport is unknown. Using subcellular live imaging within brain tissue, we show that microtubules in the apical process uniformly emanate for the pericentrosomal region, while microtubules in the basal fiber display a mixed polarity, reminiscent of the mammalian dendrite. We identify acentrosomal microtubule organizing centers localized in varicosities of the basal fiber. CAMSAP family members accumulate in these varicosities, where they control microtubule growth. Double knockdown of CAMSAP1 and 2 leads to a destabilization of the entire basal process. Finally, using live imaging of human fetal cortex, we reveal that this organization is conserved in basal radial glial cells, a related progenitor cell population associated with human brain size expansion.


Ependymoglial Cells/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Neocortex/metabolism , Neural Stem Cells/metabolism , Neurogenesis , Animals , Cell Lineage , Cell Movement , Gestational Age , Humans , Mice , Microscopy, Fluorescence , Microtubule-Associated Proteins/genetics , Microtubules/genetics , Neocortex/embryology , Signal Transduction , Time Factors , Time-Lapse Imaging
10.
Curr Biol ; 29(18): 2993-3005.e9, 2019 09 23.
Article En | MEDLINE | ID: mdl-31495584

A functional bipolar spindle is essential to segregate chromosomes correctly during mitosis. Across organisms and cell types, spindle architecture should be optimized to promote error-free divisions. However, it remains to be investigated whether mitotic spindle morphology adapts to changes in tissue properties, typical of embryonic development, in order to ensure different tasks, such as spindle positioning and chromosome segregation. We have characterized mitotic spindles in neural stem cells (NSCs) of the embryonic developing mouse neocortex. Surprisingly, we found a switch in spindle morphology from early to late neurogenic stages, which relies on an increase in inner spindle microtubule density and stability. Mechanistically, we identified the microtubule-associated protein TPX2 as one determinant of spindle shape, contributing not only to its robustness but also to correct chromosome segregation upon mitotic challenge. Our findings highlight a possible causal relationship between spindle architecture and mitotic accuracy with likely implications in brain size regulation.


Chromosome Segregation/physiology , Microtubules/metabolism , Spindle Apparatus/metabolism , Animals , Brain/metabolism , Cell Cycle Proteins/metabolism , Chromosome Segregation/genetics , Female , Kinetochores/metabolism , Male , Mammals/genetics , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Microtubules/physiology , Mitosis/physiology , Neural Stem Cells/metabolism , Neurogenesis/physiology , Pregnancy , Spindle Apparatus/physiology
11.
Nat Commun ; 9(1): 2443, 2018 06 22.
Article En | MEDLINE | ID: mdl-29934494

Cancer cells' ability to migrate through constricting pores in the tissue matrix is limited by nuclear stiffness. MT1-MMP contributes to metastasis by widening matrix pores, facilitating confined migration. Here, we show that modulation of matrix pore size or of lamin A expression known to modulate nuclear stiffness directly impinges on levels of MT1-MMP-mediated pericellular collagenolysis by cancer cells. A component of this adaptive response is the centrosome-centered distribution of MT1-MMP intracellular storage compartments ahead of the nucleus. We further show that this response, including invadopodia formation in association with confining matrix fibrils, requires an intact connection between the nucleus and the centrosome via the linker of nucleoskeleton and cytoskeleton (LINC) complex protein nesprin-2 and dynein adaptor Lis1. Our results uncover a digest-on-demand strategy for nuclear translocation through constricted spaces whereby confined migration triggers polarization of MT1-MMP storage compartments and matrix proteolysis in front of the nucleus depending on nucleus-microtubule linkage.


1-Alkyl-2-acetylglycerophosphocholine Esterase/metabolism , Cell Movement , Matrix Metalloproteinase 14/metabolism , Microfilament Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Neoplasms/pathology , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Centrosome/metabolism , Humans , Lamin Type A/metabolism , Neoplasm Invasiveness/pathology , Podosomes/metabolism , Proteolysis
12.
Brain ; 140(10): 2597-2609, 2017 Oct 01.
Article En | MEDLINE | ID: mdl-28969387

Microlissencephaly is a rare brain malformation characterized by congenital microcephaly and lissencephaly. Microlissencephaly is suspected to result from abnormalities in the proliferation or survival of neural progenitors. Despite the recent identification of six genes involved in microlissencephaly, the pathophysiological basis of this condition remains poorly understood. We performed trio-based whole exome sequencing in seven subjects from five non-consanguineous families who presented with either microcephaly or microlissencephaly. This led to the identification of compound heterozygous mutations in WDR81, a gene previously associated with cerebellar ataxia, intellectual disability and quadrupedal locomotion. Patient phenotypes ranged from severe microcephaly with extremely reduced gyration with pontocerebellar hypoplasia to moderate microcephaly with cerebellar atrophy. In patient fibroblast cells, WDR81 mutations were associated with increased mitotic index and delayed prometaphase/metaphase transition. Similarly, in vivo, we showed that knockdown of the WDR81 orthologue in Drosophila led to increased mitotic index of neural stem cells with delayed mitotic progression. In summary, we highlight the broad phenotypic spectrum of WDR81-related brain malformations, which include microcephaly with moderate to extremely reduced gyration and cerebellar anomalies. Our results suggest that WDR81 might have a role in mitosis that is conserved between Drosophila and humans.


Fibroblasts/cytology , Microcephaly/genetics , Microcephaly/pathology , Mitosis/genetics , Mutation/genetics , Nerve Tissue Proteins/genetics , Neural Stem Cells/cytology , Animals , Animals, Genetically Modified , Brain/diagnostic imaging , Brain/pathology , Cells, Cultured , Child, Preschool , Drosophila , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Female , Fibroblasts/pathology , Gene Expression Regulation/genetics , Humans , Ki-67 Antigen/metabolism , Male , Microcephaly/diagnostic imaging , Neural Stem Cells/pathology , RNA Interference/physiology , Young Adult
13.
EBioMedicine ; 10: 71-6, 2016 Aug.
Article En | MEDLINE | ID: mdl-27453325

The recent Zika outbreak in South America and French Polynesia was associated with an epidemic of microcephaly, a disease characterized by a reduced size of the cerebral cortex. Other members of the Flavivirus genus, including West Nile virus (WNV), can cause encephalitis but were not demonstrated to cause microcephaly. It remains unclear whether Zika virus (ZIKV) and other flaviviruses may infect different cell populations in the developing neocortex and lead to distinct developmental defects. Here, we describe an assay to infect mouse E15 embryonic brain slices with ZIKV, WNV and dengue virus serotype 4 (DENV-4). We show that this tissue is able to support viral replication of ZIKV and WNV, but not DENV-4. Cell fate analysis reveals a remarkable tropism of ZIKV infection for neural stem cells. Closely related WNV displays a very different tropism of infection, with a bias towards neurons. We further show that ZIKV infection, but not WNV infection, impairs cell cycle progression of neural stem cells. Both viruses inhibited apoptosis at early stages of infection. This work establishes a powerful comparative approach to identify ZIKV-specific alterations in the developing neocortex and reveals specific preferential infection of neural stem cells by ZIKV.


Flavivirus/physiology , Neocortex/cytology , Neocortex/virology , Neural Stem Cells/virology , Viral Tropism , Zika Virus Infection/virology , Zika Virus/physiology , Animals , Apoptosis , Cell Cycle , Disease Models, Animal , Flavivirus/classification , Mice , Phylogeny , Vero Cells
14.
Methods Cell Biol ; 131: 349-63, 2016.
Article En | MEDLINE | ID: mdl-26794523

Development of the cerebral cortex is a very dynamic process, involving a series of complex morphogenetic events. Following division of progenitor cells in the ventricular zone, neurons undergo a series of morphological changes and migrate outward toward the cortical plate, where they differentiate and integrate into functional circuits. Errors at several of stages during neurogenesis and migration cause a variety of severe cortical malformations. A number of disease genes encode factors associated with the cytoskeleton, which plays a crucial role throughout cortical development. Methods for regulating gene expression coupled with imaging of subcellular structures have provided important insight into the mechanisms governing normal and abnormal brain development. We describe here a series of protocols for imaging motor protein-dependent processes in real time in the developing rat brain.


Cerebral Cortex/metabolism , Molecular Motor Proteins/genetics , Neural Stem Cells/metabolism , Animals , Cell Movement/physiology , Cerebral Cortex/cytology , Electroporation/methods , Embryo, Mammalian/innervation , Ependymoglial Cells/cytology , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Luminescent Proteins/genetics , Microtubules/metabolism , Neural Stem Cells/cytology , Protein Transport/physiology , RNA Interference , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley , Red Fluorescent Protein
15.
Dev Cell ; 33(6): 703-16, 2015 Jun 22.
Article En | MEDLINE | ID: mdl-26051540

Dynein recruitment to the nuclear envelope is required for pre-mitotic nucleus-centrosome interactions in nonneuronal cells and for apical nuclear migration in neural stem cells. In each case, dynein is recruited to the nuclear envelope (NE) specifically during G2 via two nuclear pore-mediated mechanisms involving RanBP2-BicD2 and Nup133-CENP-F. The mechanisms responsible for cell-cycle control of this behavior are unknown. We now find that Cdk1 serves as a direct master controller for NE dynein recruitment in neural stem cells and HeLa cells. Cdk1 phosphorylates conserved sites within RanBP2 and activates BicD2 binding and early dynein recruitment. Late recruitment is triggered by a Cdk1-induced export of CENP-F from the nucleus. Forced NE targeting of BicD2 overrides Cdk1 inhibition, fully rescuing dynein recruitment and nuclear migration in neural stem cells. These results reveal how NE dynein recruitment is cell-cycle regulated and identify the trigger mechanism for apical nuclear migration in the brain.


Cyclin-Dependent Kinases/metabolism , Dyneins/metabolism , Neural Stem Cells/metabolism , Active Transport, Cell Nucleus , Amino Acid Sequence , Animals , Brain/cytology , Brain/embryology , Brain/metabolism , CDC2 Protein Kinase , Cyclin-Dependent Kinases/antagonists & inhibitors , HeLa Cells , Humans , Microtubule-Associated Proteins/metabolism , Mitosis , Models, Neurological , Molecular Chaperones/chemistry , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Molecular Sequence Data , Neural Stem Cells/cytology , Nuclear Envelope/metabolism , Nuclear Pore Complex Proteins/chemistry , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/metabolism , Phosphorylation , RNA, Small Interfering/genetics , Rats , Rats, Transgenic
17.
Cell ; 154(6): 1300-13, 2013 Sep 12.
Article En | MEDLINE | ID: mdl-24034252

Radial glial progenitors (RGPs) are elongated epithelial cells that give rise to neurons, glia, and adult stem cells during brain development. RGP nuclei migrate basally during G1, apically using cytoplasmic dynein during G2, and undergo mitosis at the ventricular surface. By live imaging of in utero electroporated rat brain, we find that two distinct G2-specific mechanisms for dynein nuclear pore recruitment are essential for apical nuclear migration. The "RanBP2-BicD2" and "Nup133-CENP-F" pathways act sequentially, with Nup133 or CENP-F RNAi arresting nuclei close to the ventricular surface in a premitotic state. Forced targeting of dynein to the nuclear envelope rescues nuclear migration and cell-cycle progression, demonstrating that apical nuclear migration is not simply correlated with cell-cycle progression from G2 to mitosis, but rather, is a required event. These results reveal that cell-cycle control of apical nuclear migration occurs by motor protein recruitment and identify a role for nucleus- and centrosome-associated forces in mitotic entry. PAPERCLIP:


Brain/embryology , Cell Nucleus/metabolism , Dyneins/metabolism , Mitosis , Neural Stem Cells/cytology , Nuclear Pore/metabolism , Animals , Brain/cytology , Carrier Proteins/metabolism , Centrosome/metabolism , Embryo, Mammalian/metabolism , Membrane Proteins/metabolism , Microtubule-Associated Proteins , Neural Stem Cells/metabolism , Neurogenesis , Rats
18.
J Cell Sci ; 126(Pt 15): 3259-62, 2013 Aug 01.
Article En | MEDLINE | ID: mdl-23908378

Located in the 16th century Wiston House in West Sussex, UK, the 'Building a Centrosome' Workshop was organised by The Company of Biologists and chaired by Fanni Gergely and David Glover (University of Cambridge). Held in March 2013, the Workshop gathered together many of the leaders in the field of centrosome biology, as well as postdocs and students who were given the opportunity to meet and interact with many of the scientists who inspired their early careers. The diverse range of speakers provided a multi-disciplinary forum for the exchange of ideas, and gave fresh impetus to tackling outstanding questions related to centrosome biology. Here, we provide an overview of the meeting and highlight the main themes that were discussed.


Centrosome/physiology , Animals , Humans
19.
Mol Biol Cell ; 23(18): 3591-601, 2012 Sep.
Article En | MEDLINE | ID: mdl-22855530

Microtubules (MTs) are essential for cell division, shape, intracellular transport, and polarity. MT stability is regulated by many factors, including MT-associated proteins and proteins controlling the amount of free tubulin heterodimers available for polymerization. Tubulin-binding cofactors are potential key regulators of free tubulin concentration, since they are required for α-ß-tubulin dimerization in vitro. In this paper, we show that mutation of the Drosophila tubulin-binding cofactor B (dTBCB) affects the levels of both α- and ß-tubulins and dramatically destabilizes the MT network in different fly tissues. However, we find that dTBCB is dispensable for the early MT-dependent steps of oogenesis, including cell division, and that dTBCB is not required for mitosis in several tissues. In striking contrast, the absence of dTBCB during later stages of oogenesis causes major defects in cell polarity. We show that dTBCB is required for the polarized localization of the axis-determining mRNAs within the oocyte and for the apico-basal polarity of the surrounding follicle cells. These results establish a developmental function for the dTBCB gene that is essential for viability and MT-dependent cell polarity, but not cell division.


Cell Polarity/physiology , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Animals , Animals, Genetically Modified , Cell Cycle/genetics , Cell Cycle/physiology , Cell Line , Cell Polarity/genetics , Cell Proliferation , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Female , Gene Expression Regulation, Developmental , Immunohistochemistry , Larva/genetics , Larva/growth & development , Larva/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Microscopy, Confocal , Microtubule-Associated Proteins/genetics , Mutation , Oocytes/cytology , Oocytes/growth & development , Oocytes/metabolism , Oogenesis/genetics , Oogenesis/physiology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Signal Transduction/physiology , Tubulin/genetics , Tubulin/metabolism
20.
Dev Cell ; 18(5): 790-801, 2010 May 18.
Article En | MEDLINE | ID: mdl-20493812

Microtubules (MTs) are essential for many cell features, such as polarity, motility, shape, and vesicle trafficking. Therefore, in a multicellular organism, their organization differs between cell types and during development; however, the control of this process remains elusive. Here, we show that during Drosophila tracheal morphogenesis, MT reorganization is coupled to relocalization of the microtubule organizing centers (MTOC) components from the centrosome to the apical cell domain from where MTs then grow. We reveal that this process is controlled by the trachealess patterning gene in a two-step mechanism. MTOC components are first released from the centrosome by the activity of the MT-severing protein Spastin, and then anchored apically through the transmembrane protein Piopio. We further show that these changes are essential for tracheal development, thus stressing the functional relevance of MT reorganization for morphogenesis.


Drosophila/growth & development , Microtubules/physiology , Trachea/growth & development , Adenosine Triphosphatases/physiology , Animals , Carrier Proteins/physiology , Cell Differentiation , Cell Nucleus/physiology , Cell Nucleus/ultrastructure , Centrioles/physiology , Centrioles/ultrastructure , Centrosome/physiology , Drosophila/embryology , Drosophila Proteins/physiology , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/physiology , Microtubule-Associated Proteins/physiology , Morphogenesis/physiology , Trachea/cytology
...