Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Neuron ; 92(2): 435-448, 2016 Oct 19.
Article in English | MEDLINE | ID: mdl-27693257

ABSTRACT

The neocortex undergoes extensive developmental growth, but how its architecture adapts to expansion remains largely unknown. Here, we investigated how early born Cajal-Retzius (CR) neurons, which regulate the assembly of cortical circuits, maintain a dense superficial distribution in the growing neocortex. We found that CR cell density is sustained by an activity-dependent importation of olfactory CR cells, which migrate into the neocortex after they have acted as axonal guidepost cells in the olfactory system. Furthermore, using mouse genetics, we showed that CR cell density severely affects the architecture of layer 1, a key site of input integration for neocortical networks, leading to an excitation/inhibition ratio imbalance. Our study reveals that neurons reenter migration several days after their initial positioning, thereby performing sequential developmental roles in olfactory cortex and neocortex. This atypical process is essential to regulate CR cell density during growth, which in turn ensures the correct wiring of neocortical circuitry. VIDEO ABSTRACT.


Subject(s)
Cell Count , Neocortex/embryology , Neurons/physiology , Olfactory Bulb/embryology , Olfactory Cortex/embryology , Animals , Axons , Cell Movement , Interneurons/physiology , Mice , Olfactory Bulb/cytology
3.
Nat Med ; 21(9): 989-97, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26236989

ABSTRACT

Progressive kidney fibrosis contributes greatly to end-stage renal failure, and no specific treatment is available to preserve organ function. During renal fibrosis, myofibroblasts accumulate in the interstitium of the kidney, leading to massive deposition of extracellular matrix and organ dysfunction. The origin of myofibroblasts is manifold, but the contribution of an epithelial-to-mesenchymal transition (EMT) undergone by renal epithelial cells during kidney fibrosis is still debated. We show that the reactivation of Snai1 (encoding snail family zinc finger 1, known as Snail1) in mouse renal epithelial cells is required for the development of fibrosis in the kidney. Damage-mediated Snail1 reactivation induces a partial EMT in tubular epithelial cells that, without directly contributing to the myofibroblast population, relays signals to the interstitium to promote myofibroblast differentiation and fibrogenesis and to sustain inflammation. We also show that Snail1-induced fibrosis can be reversed in vivo and that obstructive nephropathy can be therapeutically ameliorated in mice by targeting Snail1 expression. These results reconcile conflicting data on the role of the EMT in renal fibrosis and provide avenues for the design of novel anti-fibrotic therapies.


Subject(s)
Epithelial-Mesenchymal Transition , Kidney/pathology , Transcription Factors/physiology , Animals , Fibrosis , Folic Acid/toxicity , Inflammation/etiology , Male , Mice , Mice, Inbred C57BL , Renal Insufficiency, Chronic/etiology , Snail Family Transcription Factors , Ureteral Obstruction/complications
4.
J Cell Biol ; 205(6): 791-9, 2014 Jun 23.
Article in English | MEDLINE | ID: mdl-24958772

ABSTRACT

Mitotic spindle orientation relies on a complex dialog between the spindle microtubules and the cell cortex, in which F-actin has been recently implicated. Here, we report that the membrane-actin linkers ezrin/radixin/moesin (ERMs) are strongly and directly activated by the Ste20-like kinase at mitotic entry in mammalian cells. Using microfabricated adhesive substrates to control the axis of cell division, we found that the activation of ERMs plays a key role in guiding the orientation of the mitotic spindle. Accordingly, impairing ERM activation in apical progenitors of the mouse embryonic neocortex severely disturbed spindle orientation in vivo. At the molecular level, ERM activation promotes the polarized association at the mitotic cortex of leucine-glycine-asparagine repeat protein (LGN) and nuclear mitotic apparatus (NuMA) protein, two essential factors for spindle orientation. We propose that activated ERMs, together with Gαi, are critical for the correct localization of LGN-NuMA force generator complexes and hence for proper spindle orientation.


Subject(s)
Antigens, Nuclear/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Matrix-Associated Proteins/metabolism , Protein Serine-Threonine Kinases/physiology , Spindle Apparatus/metabolism , Antigens, Nuclear/analysis , Cell Cycle Proteins , Cell Division , Cytoskeletal Proteins/metabolism , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/analysis , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Nuclear Matrix-Associated Proteins/analysis , Phosphorylation , Protein Serine-Threonine Kinases/analysis , Protein Serine-Threonine Kinases/metabolism
5.
J Cell Sci ; 123(Pt 20): 3467-77, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20930141

ABSTRACT

Although TGF-ß suppresses early stages of tumour development, it later contributes to tumour progression when cells become resistant to its suppressive effects. In addition to circumventing TGF-ß-induced growth arrest and apoptosis, malignant tumour cells become capable of undergoing epithelial-to-mesenchymal transition (EMT), favouring invasion and metastasis. Therefore, defining the mechanisms that allow cancer cells to escape from the suppressive effects of TGF-ß is fundamental to understand tumour progression and to design specific therapies. Here, we have examined the role of Snail1 as a suppressor of TGF-ß-induced apoptosis in murine non-transformed hepatocytes, rat and human hepatocarcinoma cell lines and transgenic mice. We show that Snail1 confers resistance to TGF-ß-induced cell death and that it is sufficient to induce EMT in adult hepatocytes, cells otherwise refractory to this transition upon exposure to TGF-ß. Furthermore, we show that Snail1 silencing prevents EMT and restores the cell death response induced by TGF-ß. As Snail1 is a known target of TGF-ß signalling, our data indicate that Snail1 might transduce the tumour-promoting effects of TGF-ß, namely the EMT concomitant with the resistance to cell death.


Subject(s)
Apoptosis/drug effects , Epithelial-Mesenchymal Transition/physiology , Hepatocytes/drug effects , Hepatocytes/metabolism , Transcription Factors/metabolism , Transforming Growth Factor beta/pharmacology , Animals , Blotting, Western , Cell Line , Cells, Cultured , Electrophoretic Mobility Shift Assay , Epithelial-Mesenchymal Transition/genetics , Hepatocytes/cytology , Humans , Immunohistochemistry , Mice , Mice, Transgenic , Microscopy, Fluorescence , Polymerase Chain Reaction , Promoter Regions, Genetic , Rats , Snail Family Transcription Factors , Transcription Factors/genetics
6.
Am J Med Genet A ; 152A(1): 245-55, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20034074

ABSTRACT

Achondroplasia (ACH), thanatophoric dysplasia (TD) types I and II, hypochondroplasia (HCH), and severe achondroplasia with developmental delay and acanthosis nigricans (SADDAN) are all due to activating mutations in the fibroblast growth factor receptor 3 (FGFR3) gene. We review the clinical, epidemiological, radiological, molecular aspects, and signaling pathways involved in these conditions. It is known that FGFR3 signaling is essential to regulate bone growth. The signal transducers and activators of transcription (STAT1) pathway is involved in the inhibition of chondrocyte proliferation, and the mitogen-activated protein kinase (MAPK) pathways are involved in chondrocyte differentiation. Hence, FGFR3 signaling is pivotal in chondrocyte differentiation and proliferation through these two different active pathways. Recent studies on the molecular mechanisms involved in chondrocyte differentiation and proliferation, demonstrated that Snail1 participates in the control of longitudinal bone growth and appears to be essential to transduce FGFR3 signaling during chondrogenesis. This result was confirmed in a newborn infant with TD, and suggests new non-surgical therapeutic approaches, that is, Snail1 as a new encouraging therapeutic target.


Subject(s)
Achondroplasia/genetics , Thanatophoric Dysplasia/genetics , Humans , Infant, Newborn
7.
EMBO J ; 28(6): 686-96, 2009 Mar 18.
Article in English | MEDLINE | ID: mdl-19197242

ABSTRACT

Bone undergoes continuous remodelling throughout adult life, and the equilibrium between bone formation by osteoblasts and bone resorption by osteoclasts defines the final bone mass. Here we show that Snail1 regulates this balance by controlling osteoblast differentiation. Snail1 is necessary for the early steps of osteoblast development, and it must be downregulated for their final differentiation. At the molecular level, Snail1 controls bone mass by repressing the transcription of both the osteoblast differentiation factor Runx2 and the vitamin D receptor (VDR) genes in osteoblasts. Sustained activation of Snail1 in transgenic mice provokes deficient osteoblast differentiation, which, together with the loss of vitamin D signalling in the bone, also impairs osteoclastogenesis. Indeed, the mineralisation of the bone matrix is severely affected, leading to hypocalcemia-independent osteomalacia. Our data show that the impact of Snail1 activity on the osteoblast population regulates the course of bone cells differentiation and ensures normal bone remodelling.


Subject(s)
Bone and Bones/metabolism , Cell Differentiation , Core Binding Factor Alpha 1 Subunit/genetics , Gene Expression Regulation , Osteoblasts/cytology , Receptors, Calcitriol/genetics , Transcription Factors/metabolism , Animals , Bone Remodeling , Bone and Bones/pathology , Calcification, Physiologic , Cell Differentiation/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Fluoresceins/metabolism , Mice , Mice, Transgenic , Models, Biological , Organ Size , Osteoblasts/metabolism , Osteoclasts/cytology , Osteoclasts/metabolism , Osteomalacia/genetics , Osteomalacia/physiopathology , Receptors, Calcitriol/metabolism , Snail Family Transcription Factors , Transcription, Genetic
8.
Dev Cell ; 13(6): 872-83, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18061568

ABSTRACT

Achondroplasias are the most common genetic forms of dwarfism in humans. They are associated with activating mutations in FGFR3, which signal through the Stat and MAPK pathways in a ligand-independent manner to impair chondrocyte proliferation and differentiation. Snail1 has been implicated in chondrocyte differentiation as it represses Collagen II and aggrecan transcription in vitro. Here we demonstrate that Snail1 overexpression in the developing bone leads to achondroplasia in mice. Snail1 acts downstream of FGFR3 signaling in chondrocytes, regulating both Stat and MAPK pathways. Moreover, FGFR3 requires Snail1 during bone development and disease as the inhibition of Snail1 abolishes its signaling even through achondroplastic- and thanatophoric-activating FGFR3 forms. Significantly, Snail1 is aberrantly upregulated in thanatophoric versus normal cartilages from stillborns. Thus, Snail activity may likely be considered a target for achondroplasia therapies.


Subject(s)
Achondroplasia/metabolism , Chondrocytes/metabolism , Chondrogenesis/physiology , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Signal Transduction , Transcription Factors/physiology , Animals , Blotting, Western , Cartilage/metabolism , Cell Proliferation , Hindlimb/embryology , Hindlimb/metabolism , Humans , Mice/embryology , Mice, Transgenic , Receptor, Fibroblast Growth Factor, Type 3/genetics , Reverse Transcriptase Polymerase Chain Reaction , Snail Family Transcription Factors , Tamoxifen/pharmacology , Transcription, Genetic
9.
EMBO Rep ; 8(1): 104-9, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17124510

ABSTRACT

Retinoic acid (RA) signalling ensures that vertebrate mesoderm segmentation is bilaterally synchronized, and corrects transient interferences from asymmetric left-right (L-R) signals involved in organ lateralization. Snail genes participate in both these processes and, although they are expressed symmetrically in the presomitic mesoderm (PSM), Snail1 transcripts are asymmetrically distributed in the L-R lateral mesoderm. We show that the alteration of the symmetric Snail expression in the PSM induces asynchronous somite formation. Furthermore, in the absence of RA signalling, normal asymmetric Snail1 expression in the lateral mesoderm is extended to the PSM, desynchronizing somitogenesis. Thus, Snail1 is the first cue corrected by RA in the PSM to ensure synchronized bilateral segmentation.


Subject(s)
Body Patterning/genetics , Embryonic Development/genetics , Somites/metabolism , Transcription Factors/genetics , Animals , Body Patterning/drug effects , Chick Embryo , Gene Expression/drug effects , Mesoderm/chemistry , Mesoderm/metabolism , Mice , Mice, Transgenic , RNA Interference , RNA, Messenger/analysis , RNA, Messenger/metabolism , Snail Family Transcription Factors , Somites/chemistry , Tretinoin/pharmacology
10.
EMBO J ; 25(23): 5603-13, 2006 Nov 29.
Article in English | MEDLINE | ID: mdl-17093497

ABSTRACT

During embryonic development, the kidney epithelium originates from cells that undergo a mesenchymal to epithelial transition (MET). The reverse process, epithelium to mesenchyme transition (EMT), has been implicated in epithelial tumor progression and in the fibrosis that leads to end-stage kidney failure. Snail transcription factors induce both natural and pathological EMT, but their implication in renal development and disease is still unclear. We show that Snail genes are downregulated during the MET that occurs during renal development and that this is correlated with Cadherin-16 expression. Snail suppresses Cadherin-16 via the direct repression of the kidney differentiation factor HNF-1beta, a novel route by which Snail disrupts epithelial homeostasis. Indeed, Snail activation is sufficient to induce EMT and kidney fibrosis in adult transgenic mice. Significantly, Snail is also activated in patients with renal fibrosis. Thus, Snail expression is suppressed during renal development and it must remain silent in the mature kidney where its aberrant activation leads to fibrosis.


Subject(s)
Gene Expression Regulation, Developmental , Kidney Diseases/genetics , Kidney/embryology , Kidney/pathology , Transcription Factors/agonists , Animals , Cadherins/genetics , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Embryonic Development/genetics , Epithelial Cells/cytology , Epithelial Cells/metabolism , Fibrosis , Hepatocyte Nuclear Factor 1-beta/genetics , Homeostasis/genetics , Humans , Kidney/chemistry , Mesoderm/cytology , Mesoderm/metabolism , Mice , Mice, Transgenic , RNA, Messenger/analysis , RNA, Messenger/metabolism , Snail Family Transcription Factors , Transcription Factors/genetics , Transcriptional Activation
11.
J Biol Chem ; 277(41): 38803-9, 2002 Oct 11.
Article in English | MEDLINE | ID: mdl-12151403

ABSTRACT

Snail genes encode zinc finger transcription factors required for the development of vertebrate and invertebrate embryos. They trigger epithelial to mesenchymal transitions (EMTs), thereby allowing epithelial cells to emigrate from their place of origin and form tissues such as the mesoderm and the neural crest. Snail genes are also involved in the EMTs responsible for the acquisition of invasiveness during tumor progression. This aspect of their activity is associated with their ability to directly repress E-cadherin transcription. Here we describe the existence of an active human Snail retrogene, inserted within an intron of a novel evolutionarily conserved gene and expressed in different human tissues and cell lines. Functional analyses in cell culture show that this retrogene maintains the potential to induce EMTs, conferring migratory and invasive properties to epithelial cells. In light of this data, we have renamed it SNAIL-like, a new player that must be considered in both physiological and pathological studies of SNAIL function in humans.


Subject(s)
DNA-Binding Proteins/genetics , Embryo, Mammalian/physiology , Transcription Factors/genetics , Zinc Fingers , Amino Acid Sequence , Animals , Cadherins/genetics , Cadherins/metabolism , Cell Movement/physiology , Cells, Cultured , Chromosomes, Human, Pair 2 , DNA-Binding Proteins/metabolism , Exons/genetics , Genes, Reporter , Humans , Introns/genetics , Mice , Molecular Sequence Data , Open Reading Frames , Promoter Regions, Genetic , Recombinant Fusion Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Sequence Alignment , Snail Family Transcription Factors , Tissue Distribution , Transcription Factors/metabolism , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL