Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Cell ; 155(2): 333-44, 2013 Oct 10.
Article in English | MEDLINE | ID: mdl-24120134

ABSTRACT

Primary cilia are key sensory organelles that are thought to be disassembled prior to mitosis. Inheritance of the mother centriole, which nucleates the primary cilium, in relation to asymmetric daughter cell behavior has previously been studied. However, the fate of the ciliary membrane upon cell division is unknown. Here, we followed the ciliary membrane in dividing embryonic neocortical stem cells and cultured cells. Ciliary membrane attached to the mother centriole was endocytosed at mitosis onset, persisted through mitosis at one spindle pole, and was asymmetrically inherited by one daughter cell, which retained stem cell character. This daughter re-established a primary cilium harboring an activated signal transducer earlier than the noninheriting daughter. Centrosomal association of ciliary membrane in dividing neural stem cells decreased at late neurogenesis when these cells differentiate. Our data imply that centrosome-associated ciliary membrane acts as a determinant for the temporal-spatial control of ciliogenesis.


Subject(s)
Cell Division , Cilia/metabolism , Neural Stem Cells/cytology , ADP-Ribosylation Factors , Animals , Centrioles/metabolism , Centrosome/metabolism , HEK293 Cells , Humans , Mice , Mitosis , Neural Stem Cells/metabolism
2.
Nature ; 580(7802): 263-268, 2020 04.
Article in English | MEDLINE | ID: mdl-32269334

ABSTRACT

In cells, organs and whole organisms, nutrient sensing is key to maintaining homeostasis and adapting to a fluctuating environment1. In many animals, nutrient sensors are found within the enteroendocrine cells of the digestive system; however, less is known about nutrient sensing in their cellular siblings, the absorptive enterocytes1. Here we use a genetic screen in Drosophila melanogaster to identify Hodor, an ionotropic receptor in enterocytes that sustains larval development, particularly in nutrient-scarce conditions. Experiments in Xenopus oocytes and flies indicate that Hodor is a pH-sensitive, zinc-gated chloride channel that mediates a previously unrecognized dietary preference for zinc. Hodor controls systemic growth from a subset of enterocytes-interstitial cells-by promoting food intake and insulin/IGF signalling. Although Hodor sustains gut luminal acidity and restrains microbial loads, its effect on systemic growth results from the modulation of Tor signalling and lysosomal homeostasis within interstitial cells. Hodor-like genes are insect-specific, and may represent targets for the control of disease vectors. Indeed, CRISPR-Cas9 genome editing revealed that the single hodor orthologue in Anopheles gambiae is an essential gene. Our findings highlight the need to consider the instructive contributions of metals-and, more generally, micronutrients-to energy homeostasis.


Subject(s)
Chloride Channels/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Eating/physiology , Intestines/physiology , Zinc/metabolism , Animals , Drosophila melanogaster/genetics , Enterocytes/metabolism , Female , Food Preferences , Homeostasis , Insect Vectors , Insulin/metabolism , Ion Channel Gating , Larva/genetics , Larva/growth & development , Larva/metabolism , Lysosomes/metabolism , Male , Oocytes/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction , Xenopus
3.
EMBO J ; 40(19): e108041, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34431536

ABSTRACT

The role of WNT/ß-catenin signalling in mouse neocortex development remains ambiguous. Most studies demonstrate that WNT/ß-catenin regulates progenitor self-renewal but others suggest it can also promote differentiation. Here we explore the role of WNT/STOP signalling, which stabilizes proteins during G2/M by inhibiting glycogen synthase kinase (GSK3)-mediated protein degradation. We show that mice mutant for cyclin Y and cyclin Y-like 1 (Ccny/l1), key regulators of WNT/STOP signalling, display reduced neurogenesis in the developing neocortex. Specifically, basal progenitors, which exhibit delayed cell cycle progression, were drastically decreased. Ccny/l1-deficient apical progenitors show reduced asymmetric division due to an increase in apical-basal astral microtubules. We identify the neurogenic transcription factors Sox4 and Sox11 as direct GSK3 targets that are stabilized by WNT/STOP signalling in basal progenitors during mitosis and that promote neuron generation. Our work reveals that WNT/STOP signalling drives cortical neurogenesis and identifies mitosis as a critical phase for neural progenitor fate.


Subject(s)
Mitosis , Neocortex/embryology , Neocortex/metabolism , Neurogenesis , Wnt Signaling Pathway , Amino Acid Sequence , Animals , Biomarkers , Cell Cycle , Cell Differentiation/genetics , Cyclins/genetics , Cyclins/metabolism , Embryo, Mammalian , Fluorescent Antibody Technique , Gene Expression , Glycogen Synthase Kinase 3/metabolism , Immunohistochemistry , Mice , Mice, Knockout , Mitosis/genetics , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis/genetics , Phosphorylation , SOXC Transcription Factors/genetics , SOXC Transcription Factors/metabolism
4.
Nature ; 567(7746): 113-117, 2019 03.
Article in English | MEDLINE | ID: mdl-30787442

ABSTRACT

The expansion of brain size is accompanied by a relative enlargement of the subventricular zone during development. Epithelial-like neural stem cells divide in the ventricular zone at the ventricles of the embryonic brain, self-renew and generate basal progenitors1 that delaminate and settle in the subventricular zone in enlarged brain regions2. The length of time that cells stay in the subventricular zone is essential for controlling further amplification and fate determination. Here we show that the interphase centrosome protein AKNA has a key role in this process. AKNA localizes at the subdistal appendages of the mother centriole in specific subtypes of neural stem cells, and in almost all basal progenitors. This protein is necessary and sufficient to organize centrosomal microtubules, and promote their nucleation and growth. These features of AKNA are important for mediating the delamination process in the formation of the subventricular zone. Moreover, AKNA regulates the exit from the subventricular zone, which reveals the pivotal role of centrosomal microtubule organization in enabling cells to both enter and remain in the subventricular zone. The epithelial-to-mesenchymal transition is also regulated by AKNA in other epithelial cells, demonstrating its general importance for the control of cell delamination.


Subject(s)
Centrosome/metabolism , DNA-Binding Proteins/metabolism , Lateral Ventricles/cytology , Lateral Ventricles/embryology , Microtubules/metabolism , Neurogenesis , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Animals , Cell Movement , Cells, Cultured , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition , Humans , Intercellular Junctions/metabolism , Interphase , Lateral Ventricles/anatomy & histology , Mammary Glands, Animal/cytology , Mice , Organ Size , Organoids/cytology
5.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Article in English | MEDLINE | ID: mdl-35042817

ABSTRACT

Biofilms are multicellular microbial communities that encase themselves in an extracellular matrix (ECM) of secreted biopolymers and attach to surfaces and interfaces. Bacterial biofilms are detrimental in hospital and industrial settings, but they can be beneficial, for example, in agricultural as well as in food technology contexts. An essential property of biofilms that grants them with increased survival relative to planktonic cells is phenotypic heterogeneity, the division of the biofilm population into functionally distinct subgroups of cells. Phenotypic heterogeneity in biofilms can be traced to the cellular level; however, the molecular structures and elemental distribution across whole biofilms, as well as possible linkages between them, remain unexplored. Mapping X-ray diffraction across intact biofilms in time and space, we revealed the dominant structural features in Bacillus subtilis biofilms, stemming from matrix components, spores, and water. By simultaneously following the X-ray fluorescence signal of biofilms and isolated matrix components, we discovered that the ECM preferentially binds calcium ions over other metal ions, specifically, zinc, manganese, and iron. These ions, remaining free to flow below macroscopic wrinkles that act as water channels, eventually accumulate and may possibly lead to sporulation. The possible link between ECM properties, regulation of metal ion distribution, and sporulation across whole, intact biofilms unravels the importance of molecular-level heterogeneity in shaping biofilm physiology and development.


Subject(s)
Bacillus subtilis/genetics , Bacillus subtilis/metabolism , Biofilms/growth & development , Amyloidogenic Proteins/metabolism , Bacterial Proteins/metabolism , Extracellular Matrix/physiology , Ions/metabolism , Scattering, Radiation , Scattering, Small Angle , X-Rays
6.
Cell Commun Signal ; 22(1): 57, 2024 01 19.
Article in English | MEDLINE | ID: mdl-38243233

ABSTRACT

BACKGROUND: The incidence of melanoma is increasing worldwide. Since metastatic melanoma is highly aggressive, it is important to decipher all the biological aspects of melanoma cells. In this context, we have previously shown that metastatic FEMX-I melanoma cells release small (< 150 nm) extracellular vesicles (EVs) known as exosomes and ectosomes containing the stem (and cancer stem) cell antigenic marker CD133. EVs play an important role in intercellular communication, which could have a micro-environmental impact on surrounding tissues. RESULTS: We report here a new type of large CD133+ EVs released by FEMX-I cells. Their sizes range from 2 to 6 µm and they contain lipid droplets and mitochondria. Real-time video microscopy revealed that these EVs originate from the lipid droplet-enriched cell extremities that did not completely retract during the cell division process. Once released, they can be taken up by other cells. Silencing CD133 significantly affected the cellular distribution of lipid droplets, with a re-localization around the nuclear compartment. As a result, the formation of large EVs containing lipid droplets was severely compromised. CONCLUSION: Given the biochemical effect of lipid droplets and mitochondria and/or their complexes on cell metabolism, the release and uptake of these new large CD133+ EVs from dividing aggressive melanoma cells can influence both donor and recipient cells, and therefore impact melanoma growth and dissemination.


Subject(s)
Extracellular Vesicles , Melanoma , Humans , Melanoma/pathology , Lipid Droplets/metabolism , Lipid Droplets/pathology , Extracellular Vesicles/metabolism , Cell Division , Mitochondria/metabolism
7.
Traffic ; 20(1): 39-60, 2019 01.
Article in English | MEDLINE | ID: mdl-30328220

ABSTRACT

Prominin-1 is a cell surface biomarker that allows the identification of stem and cancer stem cells from different organs. It is also expressed in several differentiated epithelial and non-epithelial cells. Irrespective of the cell type, prominin-1 is associated with plasma membrane protrusions. Here, we investigate its impact on the architecture of membrane protrusions using microvilli of Madin-Darby canine kidney cells as the main model. Our high-resolution analysis revealed that upon the overexpression of prominin-1 the number of microvilli and clusters of them increased. Microvilli with branched and/or knob-like morphologies were observed and stimulated by mutations in the ganglioside-binding site of prominin-1. The altered phenotypes were caused by the interaction of prominin-1 with phosphoinositide 3-kinase and Arp2/3 complex. Mutation of tyrosine 828 of prominin-1 impaired its phosphorylation and thereby inhibited the aforementioned interactions abolishing altered microvilli. This suggests that the interplay of prominin-1-ganglioside membrane complexes, phosphoinositide 3-kinase and cytoskeleton components regulates microvillar architecture. Lastly, the expression of prominin-1 and its mutants modified the structure of filopodia emerging from fibroblast-like cells and silencing human prominin-1 in primary hematopoietic stem cells resulted in the loss of uropod-associated microvilli. Altogether, these findings strengthen the role of prominin-1 as an organizer of cellular protrusions.


Subject(s)
AC133 Antigen/metabolism , Microvilli/metabolism , AC133 Antigen/chemistry , AC133 Antigen/genetics , Animals , Binding Sites , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Dogs , Gangliosides/metabolism , Hematopoietic Stem Cells/metabolism , Humans , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred C57BL , Microvilli/ultrastructure , Mutation , Phosphatidylinositol 3-Kinases/metabolism , Protein Binding
8.
Biochem Soc Trans ; 49(5): 1997-2006, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34397081

ABSTRACT

The mammalian neocortex is the seat of higher cognitive functions, such as thinking and language in human. A hallmark of the neocortex are the cortical neurons, which are generated from divisions of neural progenitor cells (NPCs) during development, and which constitute a key feature of the well-organized layered structure of the neocortex. Proper formation of neocortex structure requires an orchestrated cellular behavior of different cortical NPCs during development, especially during the process of cortical neurogenesis. Here, we review the great diversity of NPCs and their contribution to the development of the neocortex. First, we review the categorization of NPCs into different classes and types based on their cell biological features, and discuss recent advances in characterizing marker expression and cell polarity features in the different types of NPCs. Second, we review the different modes of cell divisions that NPCs undergo and discuss the importance of the balance between proliferation and differentiation of NPCs in neocortical development. Third, we review the different proliferative capacities among different NPC types and among the same type of NPC in different mammalian species. Dissecting the differences between NPC types and differences among mammalian species is beneficial to further understand the development and the evolutionary expansion of the neocortex and may open up new therapeutic avenues for neurodevelopmental and psychiatric disorders.


Subject(s)
Neocortex/cytology , Neocortex/growth & development , Neural Stem Cells/cytology , Neurogenesis/physiology , Neurons/cytology , Animals , Biological Evolution , Cell Division/physiology , Cell Polarity/physiology , Humans , Neural Stem Cells/classification , Neural Stem Cells/metabolism , Neurons/metabolism , Signal Transduction/physiology
9.
EMBO J ; 35(9): 942-60, 2016 05 02.
Article in English | MEDLINE | ID: mdl-26933123

ABSTRACT

The generation of multiciliated cells (MCCs) is required for the proper function of many tissues, including the respiratory tract, brain, and germline. Defects in MCC development have been demonstrated to cause a subclass of mucociliary clearance disorders termed reduced generation of multiple motile cilia (RGMC). To date, only two genes, Multicilin (MCIDAS) and cyclin O (CCNO) have been identified in this disorder in humans. Here, we describe mice lacking GEMC1 (GMNC), a protein with a similar domain organization as Multicilin that has been implicated in DNA replication control. We have found that GEMC1-deficient mice are growth impaired, develop hydrocephaly with a high penetrance, and are infertile, due to defects in the formation of MCCs in the brain, respiratory tract, and germline. Our data demonstrate that GEMC1 is a critical regulator of MCC differentiation and a candidate gene for human RGMC or related disorders.


Subject(s)
Carrier Proteins/metabolism , Cell Differentiation , Cilia/genetics , Cilia/physiology , Growth Disorders/genetics , Growth Disorders/pathology , Animals , Carrier Proteins/genetics , Cell Cycle Proteins , Mice , Mice, Knockout
10.
Proc Natl Acad Sci U S A ; 112(51): 15672-7, 2015 Dec 22.
Article in English | MEDLINE | ID: mdl-26644564

ABSTRACT

Cerebral organoids-3D cultures of human cerebral tissue derived from pluripotent stem cells-have emerged as models of human cortical development. However, the extent to which in vitro organoid systems recapitulate neural progenitor cell proliferation and neuronal differentiation programs observed in vivo remains unclear. Here we use single-cell RNA sequencing (scRNA-seq) to dissect and compare cell composition and progenitor-to-neuron lineage relationships in human cerebral organoids and fetal neocortex. Covariation network analysis using the fetal neocortex data reveals known and previously unidentified interactions among genes central to neural progenitor proliferation and neuronal differentiation. In the organoid, we detect diverse progenitors and differentiated cell types of neuronal and mesenchymal lineages and identify cells that derived from regions resembling the fetal neocortex. We find that these organoid cortical cells use gene expression programs remarkably similar to those of the fetal tissue to organize into cerebral cortex-like regions. Our comparison of in vivo and in vitro cortical single-cell transcriptomes illuminates the genetic features underlying human cortical development that can be studied in organoid cultures.


Subject(s)
Gene Expression Regulation, Developmental , Neocortex/embryology , Organoids/physiology , Cell Differentiation , Cell Lineage , Humans , Sequence Analysis, RNA , Single-Cell Analysis , Tissue Culture Techniques
11.
Development ; 141(4): 795-806, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24496617

ABSTRACT

Neocortex expansion during evolution is associated with the enlargement of the embryonic subventricular zone, which reflects an increased self-renewal and proliferation of basal progenitors. In contrast to human, the vast majority of mouse basal progenitors lack self-renewal capacity, possibly due to lack of a basal process contacting the basal lamina and downregulation of cell-autonomous production of extracellular matrix (ECM) constituents. Here we show that targeted activation of the ECM receptor integrin αvß3 on basal progenitors in embryonic mouse neocortex promotes their expansion. Specifically, integrin αvß3 activation causes an increased cell cycle re-entry of Pax6-negative, Tbr2-positive intermediate progenitors, rather than basal radial glia, and a decrease in the proportion of intermediate progenitors committed to neurogenic division. Interestingly, integrin αvß3 is the only known cell surface receptor for thyroid hormones. Remarkably, tetrac, a thyroid hormone analog that inhibits the binding of thyroid hormones to integrin αvß3, completely abolishes the intermediate progenitor expansion observed upon targeted integrin αvß3 activation, indicating that this expansion requires the binding of thyroid hormones to integrin αvß3. Convergence of ECM and thyroid hormones on integrin αvß3 thus appears to be crucial for cortical progenitor proliferation and self-renewal, and hence for normal brain development and the evolutionary expansion of the neocortex.


Subject(s)
Integrin alphaVbeta3/metabolism , Neocortex/embryology , Neural Stem Cells/physiology , Neurogenesis/physiology , Thyroid Hormones/metabolism , Animals , Extracellular Matrix/metabolism , Flow Cytometry , Fluorescence , Immunohistochemistry , Mice , Mice, Inbred C57BL , Neocortex/metabolism , Neural Stem Cells/metabolism , Phenylurea Compounds , Rats , Rats, Sprague-Dawley , T-Box Domain Proteins/metabolism , Thyroxine/analogs & derivatives
12.
Development ; 139(1): 95-105, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22096071

ABSTRACT

Delamination of neural progenitors from the apical adherens junction belt of the neuroepithelium is a hallmark of cerebral cortex development and evolution. Specific cell biological processes preceding this delamination are largely unknown. Here, we identify a novel, pre-delamination state of neuroepithelial cells in mouse embryonic neocortex. Specifically, in a subpopulation of neuroepithelial cells that, like all others, exhibit apical-basal polarity and apical adherens junctions, the re-establishing of the primary cilium after mitosis occurs at the basolateral rather than the apical plasma membrane. Neuroepithelial cells carrying basolateral primary cilia appear at the onset of cortical neurogenesis, increase in abundance with its progression, selectively express the basal (intermediate) progenitor marker Tbr2, and eventually delaminate from the apical adherens junction belt to become basal progenitors, translocating their nucleus from the ventricular to the subventricular zone. Overexpression of insulinoma-associated 1, a transcription factor known to promote the generation of basal progenitors, increases the proportion of basolateral cilia. Basolateral cilia in cells delaminating from the apical adherens junction belt are preferentially found near spot-like adherens junctions, suggesting that the latter provide positional cues to basolateral ciliogenesis. We conclude that re-establishing a basolateral primary cilium constitutes the first known cell biological feature preceding neural progenitor delamination.


Subject(s)
Cilia/physiology , Neocortex/embryology , Neuroepithelial Cells/physiology , Stem Cells/physiology , Adherens Junctions , Animals , Cell Polarity/physiology , DNA-Binding Proteins/metabolism , Mice , Microscopy, Electron, Transmission , Microscopy, Immunoelectron , Neocortex/ultrastructure , Repressor Proteins , Stem Cells/cytology , T-Box Domain Proteins/metabolism , Transcription Factors/metabolism
13.
Nature ; 457(7227): 322-6, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-18971929

ABSTRACT

The nephron is the basic structural and functional unit of the vertebrate kidney. It is composed of a glomerulus, the site of ultrafiltration, and a renal tubule, along which the filtrate is modified. Although widely regarded as a vertebrate adaptation, 'nephron-like' features can be found in the excretory systems of many invertebrates, raising the possibility that components of the vertebrate excretory system were inherited from their invertebrate ancestors. Here we show that the insect nephrocyte has remarkable anatomical, molecular and functional similarity to the glomerular podocyte, a cell in the vertebrate kidney that forms the main size-selective barrier as blood is ultrafiltered to make urine. In particular, both cell types possess a specialized filtration diaphragm, known as the slit diaphragm in podocytes or the nephrocyte diaphragm in nephrocytes. We find that fly (Drosophila melanogaster) orthologues of the major constituents of the slit diaphragm, including nephrin, NEPH1 (also known as KIRREL), CD2AP, ZO-1 (TJP1) and podocin, are expressed in the nephrocyte and form a complex of interacting proteins that closely mirrors the vertebrate slit diaphragm complex. Furthermore, we find that the nephrocyte diaphragm is completely lost in flies lacking the orthologues of nephrin or NEPH1-a phenotype resembling loss of the slit diaphragm in the absence of either nephrin (as in human congenital nephrotic syndrome of the Finnish type, NPHS1) or NEPH1. These changes markedly impair filtration function in the nephrocyte. The similarities we describe between invertebrate nephrocytes and vertebrate podocytes provide evidence suggesting that the two cell types are evolutionarily related, and establish the nephrocyte as a simple model in which to study podocyte biology and podocyte-associated diseases.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Podocytes/cytology , Podocytes/physiology , Animals , Cell Line , Drosophila Proteins/genetics , Drosophila melanogaster/anatomy & histology , Drosophila melanogaster/physiology , Immunoglobulins/genetics , Immunoglobulins/metabolism , Membrane Proteins/deficiency , Membrane Proteins/genetics , Membrane Proteins/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , Podocytes/metabolism
14.
Exp Cell Res ; 319(6): 810-9, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23318676

ABSTRACT

Prominin-1 (CD133) is the first identified gene of a novel class of pentaspan membrane glycoproteins. It is expressed by various epithelial and non-epithelial cells, and notably by stem and cancer stem cells. In non-cancerous cells such as neuro-epithelial and hematopoietic stem cells, prominin-1 is selectively concentrated in plasma membrane protrusions, and released into the extracellular milieu in association with small vesicles. Previously, we demonstrated that prominin-1 contributes to melanoma cells pro-metastatic properties and suggested that it may constitute a molecular target to prevent prominin-1-expressing melanomas from colonizing and growing in lymph nodes and distant organs. Here, we report that three distinct pools of prominin-1 co-exist in cultures of human FEMX-I metastatic melanoma. Morphologically, in addition to the plasma membrane localization, prominin-1 is found within the intracellular compartments, (e.g., Golgi apparatus) and in association with extracellular membrane vesicles. The latter prominin-1-positive structures appeared in three sizes (small, ≤40 nm; intermediates ~40-80 nm, and large, >80 nm). Functionally, the down-regulation of prominin-1 in FEMX-I cells resulted in a significant reduction of number of lipid droplets as observed by coherent anti-Stokes Raman scattering image analysis and Oil red O staining, and surprisingly in a decrease in the nuclear localization of beta-catenin, a surrogate marker of Wnt activation. Moreover, the T-cell factor/lymphoid enhancer factor (TCF/LEF) promoter activity was 2 to 4 times higher in parental than in prominin-1-knockdown cells. Collectively, our results point to Wnt signaling and/or release of prominin-1-containing membrane vesicles as mediators of the pro-metastatic activity of prominin-1 in FEMX-I melanoma.


Subject(s)
Antigens, CD/metabolism , Glycoproteins/metabolism , Melanoma/metabolism , Peptides/metabolism , Protein Interaction Mapping , Wnt Signaling Pathway , beta Catenin/metabolism , AC133 Antigen , Antigens, CD/genetics , Azo Compounds/metabolism , Biomarkers, Tumor/metabolism , Cell Adhesion , Cell Line, Tumor , Cell Membrane/metabolism , Cell Membrane/pathology , Cell Movement , Cell Nucleus/genetics , Cell Nucleus/metabolism , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Glycoproteins/genetics , Golgi Apparatus/metabolism , Humans , Immunohistochemistry , Lipids/analysis , Melanoma/pathology , Neoplasm Invasiveness/pathology , Peptides/genetics , Promoter Regions, Genetic , Spectrum Analysis, Raman , TCF Transcription Factors/genetics , TCF Transcription Factors/metabolism , Transcription, Genetic , Transfection , beta Catenin/genetics
15.
Front Cell Dev Biol ; 12: 1344734, 2024.
Article in English | MEDLINE | ID: mdl-38500687

ABSTRACT

The development of the neocortex involves an interplay between neural cells and the vasculature. However, little is known about this interplay at the ultrastructural level. To gain a 3D insight into the ultrastructure of the developing neocortex, we have analyzed the embryonic mouse neocortex by serial block-face scanning electron microscopy (SBF-SEM). In this study, we report a first set of findings that focus on the interaction of blood vessels, notably endothelial tip cells (ETCs), and the neural cells in this tissue. A key observation was that the processes of ETCs, located either in the ventricular zone (VZ) or subventricular zone (SVZ)/intermediate zone (IZ), can enter, traverse the cytoplasm, and even exit via deep plasma membrane invaginations of the host cells, including apical progenitors (APs), basal progenitors (BPs), and newborn neurons. More than half of the ETC processes were found to enter the neural cells. Striking examples of this ETC process "invasion" were (i) protrusions of apical progenitors or newborn basal progenitors into the ventricular lumen that contained an ETC process inside and (ii) ETC process-containing protrusions of neurons that penetrated other neurons. Our observations reveal a - so far unknown - complexity of the ETC-neural cell interaction.

16.
Macromol Biosci ; 24(3): e2300464, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37925629

ABSTRACT

Towards the goal of building synthetic cells from the bottom-up, the establishment of micrometer-sized compartments that contain and support cell free transcription and translation that couple cellular structure to function is of critical importance. Proteinosomes, formed from crosslinked cationized protein-polymer conjugates offer a promising solution to membrane-bound compartmentalization with an open, semi-permeable membrane. Critically, to date, there has been no demonstration of cell free transcription and translation within water-in-water proteinosomes. Herein, a novel approach to generate proteinosomes that can support cell free transcription and translation is presented. This approach generates proteinosomes directly from native protein-polymer (BSA-PNIPAAm) conjugates. These native proteinosomes offer an excellent alternative as a synthetic cell chassis to other membrane bound compartments. Significantly, the native proteinosomes are stable under high salt conditions that enables the ability to support cell free transcription and translation and offer enhanced protein expression compared to proteinosomes prepared from traditional methodologies. Furthermore, the integration of native proteinosomes into higher order synthetic cellular architectures with membrane free compartments such as liposomes is demonstrated. The integration of bioinspired architectural elements with the central dogma is an essential building block for realizing minimal synthetic cells and is key for exploiting artificial cells in real-world applications.


Subject(s)
Artificial Cells , Proteins , Acrylic Resins/chemistry , Artificial Cells/chemistry , Artificial Cells/metabolism , Water
17.
Proc Natl Acad Sci U S A ; 107(38): 16595-600, 2010 Sep 21.
Article in English | MEDLINE | ID: mdl-20823249

ABSTRACT

Mutations in ASPM (abnormal spindle-like microcephaly associated) cause primary microcephaly in humans, a disorder characterized by a major reduction in brain size in the apparent absence of nonneurological anomalies. The function of the Aspm protein in neural progenitor cell expansion, as well as its localization to the mitotic spindle and midbody, suggest that it regulates brain development by a cell division-related mechanism. Furthermore, evidence that positive selection affected ASPM during primate evolution has led to suggestions that such a function changed during primate evolution. Here, we report that in Aspm mutant mice, truncated Aspm proteins similar to those causing microcephaly in humans fail to localize to the midbody during M-phase and cause mild microcephaly. A human ASPM transgene rescues this phenotype but, interestingly, does not cause a gain of function. Strikingly, truncated Aspm proteins also cause a massive loss of germ cells, resulting in a severe reduction in testis and ovary size accompanied by reduced fertility. These germline effects, too, are fully rescued by the human ASPM transgene, indicating that ASPM is functionally similar in mice and humans. Our findings broaden the spectrum of phenotypic effects of ASPM mutations and raise the possibility that positive selection of ASPM during primate evolution reflects its function in the germline.


Subject(s)
Microcephaly/genetics , Mutation , Nerve Tissue Proteins/genetics , Animals , Animals, Newborn , Base Sequence , Brain/abnormalities , Calmodulin-Binding Proteins , DNA Primers/genetics , Disease Models, Animal , Embryonic Stem Cells/pathology , Female , Germ-Line Mutation , Humans , Infertility/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Transgenic , Microcephaly/pathology , Nerve Tissue Proteins/physiology , Neurons/pathology , Oligospermia/genetics , Ovary/abnormalities , Peptide Fragments/genetics , Peptide Fragments/physiology , Phenotype , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Deletion , Sperm Motility/genetics , Testis/abnormalities
18.
Dev Cell ; 58(2): 139-154.e8, 2023 01 23.
Article in English | MEDLINE | ID: mdl-36693320

ABSTRACT

WNT signaling is important in development, stem cell maintenance, and disease. WNT ligands typically signal via receptor activation across the plasma membrane to induce ß-catenin-dependent gene activation. Here, we show that in mammalian primary cilia, WNT receptors relay a WNT/GSK3 signal that ß-catenin-independently promotes ciliogenesis. Characterization of a LRP6 ciliary targeting sequence and monitoring of acute WNT co-receptor activation (phospho-LRP6) support this conclusion. Ciliary WNT signaling inhibits protein phosphatase 1 (PP1) activity, a negative regulator of ciliogenesis, by preventing GSK3-mediated phosphorylation of the PP1 regulatory inhibitor subunit PPP1R2. Concordantly, deficiency of WNT/GSK3 signaling by depletion of cyclin Y and cyclin-Y-like protein 1 induces primary cilia defects in mouse embryonic neuronal precursors, kidney proximal tubules, and adult mice preadipocytes.


Subject(s)
Wnt Proteins , beta Catenin , Animals , Mice , beta Catenin/metabolism , Wnt Proteins/metabolism , Cilia/metabolism , Glycogen Synthase Kinase 3/metabolism , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Wnt Signaling Pathway , Phosphorylation , Cyclins/metabolism , Mammals/metabolism
19.
J Cell Biol ; 176(4): 483-95, 2007 Feb 12.
Article in English | MEDLINE | ID: mdl-17283184

ABSTRACT

Expansion of the neocortex requires symmetric divisions of neuroepithelial cells, the primary progenitor cells of the developing mammalian central nervous system. Symmetrically dividing neuroepithelial cells are known to form a midbody at their apical (rather than lateral) surface. We show that apical midbodies of neuroepithelial cells concentrate prominin-1 (CD133), a somatic stem cell marker and defining constituent of a specific plasma membrane microdomain. Moreover, these apical midbodies are released, as a whole or in part, into the extracellular space, yielding the prominin-1-enriched membrane particles found in the neural tube fluid. The primary cilium of neuroepithelial cells also concentrates prominin-1 and appears to be a second source of the prominin-1-bearing extracellular membrane particles. Our data reveal novel origins of extracellular membrane traffic that enable neural stem and progenitor cells to avoid the asymmetric inheritance of the midbody observed for other cells and, by releasing a stem cell membrane microdomain, to potentially influence the balance of their proliferation versus differentiation.


Subject(s)
Antigens, CD/metabolism , Cell Membrane/metabolism , Cilia/metabolism , Epithelial Cells/metabolism , Glycoproteins/metabolism , Neurons/metabolism , Peptides/metabolism , Stem Cells/metabolism , AC133 Antigen , Animals , Biomarkers/metabolism , Cell Differentiation/physiology , Cell Division/physiology , Cell Membrane/ultrastructure , Cell Proliferation , Cells, Cultured , Chick Embryo , Cilia/ultrastructure , Cytokinesis/physiology , Epithelial Cells/ultrastructure , Extracellular Space/metabolism , Membrane Microdomains/metabolism , Membrane Microdomains/ultrastructure , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neurons/ultrastructure , Protein Transport/physiology , Stem Cells/ultrastructure
20.
Proc Natl Acad Sci U S A ; 106(38): 16487-92, 2009 Sep 22.
Article in English | MEDLINE | ID: mdl-19805325

ABSTRACT

Interkinetic nuclear migration (INM) is a hallmark of the polarized stem and progenitor cells in the ventricular zone (VZ) of the developing vertebrate CNS. INM is responsible for the pseudostratification of the VZ, a crucial aspect of brain evolution. The nuclear migration toward the apical centrosomes in G2 is thought to be a dynein-microtubule-based process. By contrast, the cytoskeletal machinery involved in the basally directed nuclear translocation away from the centrosome in G1 has been enigmatic. Studying the latter aspect of INM requires manipulation of the cytoskeleton without impairing mitosis and cytokinesis. To this end, we have established a culture system of mouse embryonic telencephalon that reproduces cortical development, and have applied it to explore a role of actomyosin in INM. Using the nonmuscle myosin II inhibitor blebbistatin at a low concentration at which neither cell cycle progression nor cytokinesis is impaired, we show that myosin II is required for the apical-to-basal (ap-->bl), ab-centrosomal INM. Myosin II activity is also necessary for the nuclear translocation during delamination of subventricular zone (SVZ) cells, a second, telencephalon-specific type of neural progenitor. Moreover, the inhibition of ab-centrosomal INM changes the balance between VZ and SVZ progenitor cell fate. Our data suggest a unifying concept in which the actomyosin contraction underlying ab-centrosomal INM sets the stage for the evolutionary increase in VZ pseudostratification and for SVZ progenitor delamination, a key process in cortical expansion.


Subject(s)
Cell Nucleus/physiology , Myosin Type II/metabolism , Neurons/physiology , Stem Cells/physiology , Animals , Bromodeoxyuridine/metabolism , Cell Culture Techniques , Cell Cycle , Cell Nucleus/ultrastructure , Cell Proliferation , Cells, Cultured , Centrosome/metabolism , Cerebral Ventricles/cytology , Cerebral Ventricles/embryology , Fluorescent Antibody Technique , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Heterocyclic Compounds, 4 or More Rings/pharmacology , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Myosin Type II/antagonists & inhibitors , Neurons/cytology , Neurons/metabolism , Neurons/ultrastructure , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Stem Cells/metabolism , Stem Cells/ultrastructure , Time Factors , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL