Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
BMC Neurol ; 23(1): 338, 2023 Sep 26.
Article in English | MEDLINE | ID: mdl-37749503

ABSTRACT

BACKGROUND: Focal cortical dysplasia (FCD) is a malformation of cortical development that causes medical refractory seizures, and one of the main treatments may be surgical resection of the affected area of the brain. People affected by FCD may present with seizures of variable severity since childhood. Despite many medical treatments available, only surgery can offer cure. The pathophysiology of the disease is not yet understood; however, it is known that several gene alterations may play a role. The WNT/ß-catenin pathway is closely related to the control and balance of cell proliferation and differentiation in the central nervous system. The aim of this study was to explore genes related to the WNT/ß-catenin pathway in lesional and perilesional brain tissue in patients with FCD type II. METHODS: Dysplastic and perilesional tissue from the primary dysplastic lesion of patients with FCD type IIa were obtained from two patients who underwent surgical treatment. The analysis of the relative expression of genes was performed by a qRT-PCR array (super array) containing 84 genes related to the WNT pathway. RESULTS: Our results suggest the existence of molecular alteration in some genes of the WNT pathway in tissue with dysplastic lesions and of perilesional tissue. We call this tissue of normal-appearing adjacent cortex (NAAC). Of all genes analyzed, a large number of genes show similar behavior between injured, perilesional and control tissues. However, some genes have similar characteristics between the perilesional and lesional tissue and are different from the control brain tissue, presenting the perilesional tissue as a molecularly altered material. CONCLUSION: Our results suggest that the perilesional area after surgical resection of tissue with cortical dysplasia presents molecular changes that may play a role in the recurrence of seizures in these patients. The perilesional tissue should receive expanded attention beyond the somatic mutations described and associated with FCD, such as mTOR, for example, to new signaling pathways that may play a crucial role in seizure recurrence.


Subject(s)
Drug Resistant Epilepsy , Focal Cortical Dysplasia , Humans , Child , Drug Resistant Epilepsy/genetics , Drug Resistant Epilepsy/surgery , Wnt Signaling Pathway/genetics , beta Catenin , Seizures
2.
Cells ; 11(21)2022 10 27.
Article in English | MEDLINE | ID: mdl-36359798

ABSTRACT

Lysosomes are highly dynamic organelles involved in the breakdown and recycling of macromolecules, cell cycle, cell differentiation, and cell death, among many other functions in eukaryotic cells. Recently, lysosomes have been identified as cellular hubs for the modulation of intracellular signaling pathways, such as the Wnt/beta-catenin pathway. Here we analyzed morphological and functional characteristics of lysosomes in muscle and non-muscle cells during chick myogenesis, as well as their modulation by the Wnt/beta-catenin pathway. Our results show that (i) muscle and non-muscle cells show differences in lysosomal size and its distribution, (ii) lysosomes are found in spherical structures in myoblasts and fibroblasts and tubular structures in myotubes, (iii) lysosomes are found close to the plasma membrane in fibroblasts and close to the nucleus in myoblasts and myotubes, (iv) lysosomal distribution and size are dependent on the integrity of microtubules and microfilaments in myogenic cells, (v) alterations in lysosomal function, in the expression of LAMP2, and in Wnt/beta-catenin pathway affect the distribution and size of lysosomes in myogenic cells, (vi) the effects of the knockdown of LAMP2 on myogenesis can be rescued by the activation of the Wnt/beta-catenin pathway, and (vii) the chloroquine Lys05 is a potent inhibitor of both the Wnt/beta-catenin pathway and lysosomal function. Our data highlight the involvement of the Wnt/beta-catenin pathway in the regulation of the positioning, size, and function of lysosomes during chick myogenesis.


Subject(s)
Muscle Development , beta Catenin , beta Catenin/metabolism , Muscle Development/physiology , Wnt Signaling Pathway , Muscle Fibers, Skeletal/metabolism , Cytoskeleton/metabolism , Lysosomes/metabolism
3.
Cancer Chemother Pharmacol ; 87(4): 567-578, 2021 04.
Article in English | MEDLINE | ID: mdl-33471160

ABSTRACT

PURPOSE: 5-Fluorouracil (5-FU), an anti-cancer drug, has been used for hepatoblastoma (HB) chemotherapy in children, who may have impaired  ovarian follicle pool reserve with lasting effects to reproduction. Therefore, this study aimed to investigate 5-FU effects on survival, growth, and morphology of ovarian preantral follicles from C57BL6J young mice. METHODS: Experiments were carried-out both in vivo and in vitro. Mice were treated with 5-FU injection (450 mg/kg i.p) or saline and sacrificed 3 days after to obtain ovaries for histology and molecular biology. Ovaries for in vitro studies were obtained from unchallenged mice and cultured under basic culture medium (BCM) or BCM plus 5-FU (9.2, 46.1, 92.2 mM). Preantral follicles were classified according to developmental stages, and as normal or degenerated. To assess cell viability, caspase-3 immunostaining was performed. Transcriptional levels for apoptosis (Bax, Bcl2, p53, Bax/Bcl2) and Wnt pathway genes (Wnt2 and Wnt4) were also analyzed. Ultrastructural analyses were carried-out on non-cultured ovaries. In addition, ß-catenin immunofluorescence was assessed in mouse ovaries. RESULTS: The percentage of all-types normal follicles was significantly lower after 5-FU challenge. A total loss of secondary normal follicles was found in the 5-FU group. The highest 5-FU concentrations reduced the percentage of cultured normal primordial follicles. Large vacuoles were seen in granulosa cells and ooplasm of preantral follicles by electron microscopy. A significantly higher gene expression for Bax and Bax/Bcl2 ratio was seen after 5-FU treatment. A marked reduction in ß-catenin immunolabeling was seen in 5-FU-challenged preantral follicles. In the in vitro experiments, apoptotic and Wnt gene transcriptions were significantly altered. CONCLUSION: Altogether, our findings suggest that 5-FU can deleteriously affect the ovarian follicle reserve by reducing preantral follicles survival.


Subject(s)
Fluorouracil/toxicity , Ovarian Follicle/drug effects , Animals , Caspase 3/analysis , Female , Immunohistochemistry , Mice , Mice, Inbred C57BL , Ovarian Follicle/pathology , Ovarian Follicle/ultrastructure
4.
Dev Comp Immunol ; 114: 103840, 2021 01.
Article in English | MEDLINE | ID: mdl-32858087

ABSTRACT

In the present work we have focused on the Histone Deacetylase (HDAC) control of myeloid cells behavior during Xenopus tail regeneration. Here we show that myeloid differentiation is crucial to modulate the regenerative ability of Xenopus tadpoles in a HDAC activity-dependent fashion. HDAC activity inhibition during the first wave of myeloid differentiation disrupted myeloid cells dynamics in the regenerative bud as well the mRNA expression pattern of myeloid markers, such as LURP, MPOX, Spib and mmp7. We also functionally bridge the spatial and temporal dynamics of lipid droplets, the main platform of lipid mediators synthesis in myeloid cells during the inflammatory response, and the regenerative ability of Xenopus tadpoles. In addition, we showed that 15-LOX activity is necessary during tail regeneration. Taken together our results support a role for the epigenetic control of myeloid behavior during tissue and organ regeneration, which may positively impact translational approaches for regenerative medicine.


Subject(s)
Histone Deacetylases/metabolism , Myeloid Cells/metabolism , Xenopus laevis/physiology , Animals , Biomarkers/metabolism , Cell Differentiation , Cells, Cultured , Epigenesis, Genetic , Gene Expression Regulation, Developmental , Humans , Organogenesis , Regeneration , Regenerative Medicine
5.
Elife ; 92020 09 14.
Article in English | MEDLINE | ID: mdl-32924931

ABSTRACT

Wnt signaling through the Frizzled (FZD) family of serpentine receptors is essential for embryogenesis and homeostasis, and stringent control of the FZD protein level is critical for stem cell regulation. Through CRISPR/Cas9 genome-wide screening in human cells, we identified TMEM79/MATTRIN, an orphan multi-span transmembrane protein, as a specific inhibitor of Wnt/FZD signaling. TMEM79 interacts with FZD during biogenesis and promotes FZD degradation independent of ZNRF3/RNF43 ubiquitin ligases (R-spondin receptors). TMEM79 interacts with ubiquitin-specific protease 8 (USP8), whose activating mutations underlie human tumorigenesis. TMEM79 specifically inhibits USP8 deubiquitination of FZD, thereby governing USP8 substrate specificity and promoting FZD degradation. Tmem79 and Usp8 genes have a pre-bilaterian origin, and Tmem79 inhibition of Usp8 and Wnt signaling is required for anterior neural development and gastrulation in Xenopus embryos. TMEM79 is a predisposition gene for Atopic dermatitis, suggesting deregulation of Wnt/FZD signaling a possible cause for this most common yet enigmatic inflammatory skin disease.


Subject(s)
Embryonic Development/physiology , Frizzled Receptors/metabolism , Membrane Proteins/metabolism , Xenopus laevis/embryology , Animals , Embryonic Development/genetics , Frizzled Receptors/genetics , HEK293 Cells , Humans , Membrane Proteins/genetics , Wnt Signaling Pathway/genetics , Xenopus laevis/genetics
6.
Article in English | MEDLINE | ID: mdl-31828046

ABSTRACT

Toxoplasma gondii is the causative agent of toxoplasmosis, a parasitic disease with a wide global prevalence. The parasite forms cysts in skeletal muscle cells and neurons, although no evident association with inflammatory infiltrates has been typically found. We studied the impact of T. gondii infection on the myogenic program of mouse skeletal muscle cells (SkMC). The C2C12 murine myoblast cell line was infected with T. gondii tachyzoites (ME49 strain) for 24 h followed by myogenic differentiation induction. T. gondii infection caused a general decrease in myotube differentiation, fusion and maturation, along with decreased expression of myosin heavy chain. The expression of Myogenic Regulatory Factors Myf5, MyoD, Mrf4 and myogenin was modulated by the infection. Infected cultures presented increased proliferation rates, as assessed by Ki67 immunostaining, whereas neither host cell lysis nor apoptosis were significantly augmented in infected dishes. Cytokine Bead Array indicated that IL-6 and MCP-1 were highly increased in the medium from infected cultures, whereas TGF-ß1 was consistently decreased. Inhibition of the IL-6 receptor or supplementation with recombinant TGF-ß failed to reverse the deleterious effects caused by the infection. However, conditioned medium from infected cultures inhibited myogenesis in C2C12 cells. Activation of the Wnt/ß-catenin pathway was impaired in T. gondii-infected cultures. Our data indicate that T. gondii leads SkMCs to a pro-inflammatory phenotype, leaving cells unresponsive to ß-catenin activation, and inhibition of the myogenic differentiation program. Such deregulation may suggest muscle atrophy and molecular mechanisms similar to those involved in myositis observed in human patients.


Subject(s)
Host-Pathogen Interactions , Muscle Development , Myogenic Regulatory Factors/metabolism , Toxoplasma/physiology , Toxoplasmosis/metabolism , Animals , Biomarkers , Cell Differentiation , Cell Line , Cell Proliferation , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Fluorescent Antibody Technique , Gene Expression , Genes, Reporter , Mice , Myoblasts, Skeletal/metabolism , Myoblasts, Skeletal/parasitology , Myogenic Regulatory Factors/genetics , Toxoplasmosis/parasitology , Wnt Signaling Pathway
7.
Mol Neurobiol ; 56(2): 1517-1530, 2019 Feb.
Article in English | MEDLINE | ID: mdl-29948952

ABSTRACT

Glioblastoma is an extremely aggressive and deadly brain tumor known for its striking cellular heterogeneity and capability to communicate with microenvironment components, such as microglia. Microglia-glioblastoma interaction contributes to an increase in tumor invasiveness, and Wnt signaling pathway is one of the main cascades related to tumor progression through changes in cell migration and invasion. However, very little is known about the role of canonical Wnt signaling during microglia-glioblastoma crosstalk. Here, we show for the first time that Wnt3a is one of the factors that regulate interactions between microglia and glioblastoma cells. Wnt3a activates the Wnt/ß-catenin signaling of both glioblastoma and microglial cells. Glioblastoma-conditioned medium not only induces nuclear translocation of microglial ß-catenin but also increases microglia viability and proliferation as well as Wnt3a, cyclin-D1, and c-myc expression. Moreover, glioblastoma-derived Wnt3a increases microglial ARG-1 and STI1 expression, followed by an upregulation of IL-10 mRNA levels, and a decrease in IL1ß gene expression. The presence of Wnt3a in microglia-glioblastoma co-cultures increases the formation of membrane nanotubes accompanied by changes in migration capability. In vivo, tumors formed from Wnt3a-stimulated glioblastoma cells presented greater microglial infiltration and more aggressive characteristics such as growth rate than untreated tumors. Thus, we propose that Wnt3a belongs to the arsenal of factors capable of stimulating the induction of M2-like phenotype on microglial cells, which contributes to the poor prognostic of glioblastoma, reinforcing that Wnt/ß-catenin pathway can be a potential therapeutic target to attenuate glioblastoma progression.


Subject(s)
Microglia/metabolism , Wnt Signaling Pathway/physiology , Wnt3A Protein/metabolism , beta Catenin/metabolism , Cell Movement/physiology , Cell Proliferation/physiology , Glioblastoma/genetics , Humans , Phenotype
8.
Int J Mol Sci ; 19(9)2018 Sep 14.
Article in English | MEDLINE | ID: mdl-30223494

ABSTRACT

Bufadienolides are cardiotonic steroids (CTS) identified in mammals. Besides Na⁺/K⁺-ATPase inhibition, they activate signal transduction via protein⁻protein interactions. Diversity of endogenous bufadienolides and mechanisms of action may indicate the presence of functional selectivity and unique cellular outcomes. We evaluated whether the bufadienolides telocinobufagin and marinobufagin induce changes in proliferation or viability of pig kidney (LLC-PK1) cells and the mechanisms involved in these changes. In some experiments, ouabain was used as a positive control. CTS exhibited an inhibitory IC50 of 0.20 (telocinobufagin), 0.14 (ouabain), and 3.40 µM (marinobufagin) for pig kidney Na⁺/K⁺-ATPase activity and concentrations that barely inhibited it were tested in LLC-PK1 cells. CTS induced rapid ERK1/2 phosphorylation, but corresponding proliferative response was observed for marinobufagin and ouabain instead of telocinobufagin. Telocinobufagin increased Bax:Bcl-2 expression ratio, sub-G0 cell cycle phase and pyknotic nuclei, indicating apoptosis. Src and MEK1/2 inhibitors blunted marinobufagin but not telocinobufagin effect, which was also not mediated by p38, JNK1/2, and PI3K. However, BIO, a GSK-3ß inhibitor, reduced proliferation and, as telocinobufagin, phosphorylated GSK-3ß at inhibitory Ser9. Combination of both drugs resulted in synergistic antiproliferative effect. Wnt reporter activity assay showed that telocinobufagin impaired Wnt/ß-catenin pathway by acting upstream to ß-catenin stabilization. Our findings support that mammalian endogenous bufadienolides may exhibit functional selectivity.


Subject(s)
Bufanolides/pharmacology , LLC-PK1 Cells/drug effects , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Glycogen Synthase Kinase 3 beta/metabolism , MAP Kinase Signaling System/drug effects , Sodium-Potassium-Exchanging ATPase/metabolism , Swine , Wnt Signaling Pathway/drug effects
9.
Cell Signal ; 40: 30-43, 2017 12.
Article in English | MEDLINE | ID: mdl-28844868

ABSTRACT

Wnt signaling refers to a conserved signaling pathway, widely studied due to its roles in cellular communication, cell fate decisions, development and cancer. However, the exact mechanism underlying inhibition of the GSK phosphorylation towards ß-catenin and activation of the pathway after biding of Wnt ligand to its cognate receptors at the plasma membrane remains unclear. Wnt target genes are widely spread over several animal phyla. They participate in a plethora of functions during the development of an organism, from axial specification, gastrulation and organogenesis all the way to regeneration and repair in adults. Temporal and spatial oncogenetic re-activation of Wnt signaling almost certainly leads to cancer. Wnt signaling components have been extensively studied as possible targets in anti-cancer therapies. In this review we will discuss one of the most intriguing questions in this field, that is how ß-catenin, a major component in this pathway, escapes the destruction complex, gets stabilized in the cytosol and it is translocated to the nucleus where it acts as a co-transcription factor. Four major models have evolved during the past 20years. We dissected each of them along with current views and future perspectives on this pathway. This review will focus on the molecular mechanisms by which Wnt proteins modulate ß-catenin cytoplasmic levels and the relevance of this pathway for the development and cancer.


Subject(s)
Transcriptional Activation/genetics , Wnt Proteins/genetics , Wnt Signaling Pathway/genetics , beta Catenin/genetics , Cell Nucleus/genetics , Cytosol/metabolism , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , beta Catenin/chemistry
10.
Mech Dev ; 142: 30-39, 2016 11.
Article in English | MEDLINE | ID: mdl-27687541

ABSTRACT

Wnt/ß-catenin has been described as crucial for dorsal-ventral and antero-posterior patterning, playing multiple roles at different stages of development. Cholesterol-rich membrane microdomains (CRMMs), cholesterol- and sphingolipid-enriched domains of the plasma membrane, are known as platforms for signaling pathways. Although we have demonstrated the importance of the CRMMs for head development, how they participate in prechordal plate formation and embryo axis patterning remains an open question. Moreover, the participation of the CRMMs in the Wnt/ß-catenin signaling pathway activity in vivo is unclear, particularly during embryonic development. In this study, we demonstrated that CRMMs disruption by methyl-beta-cyclodextrin (MßCD) potentiates the activation of the Wnt/ß-catenin signaling pathway in vitro and in vivo during embryonic development, causing head defects by expanding the Wnt expression domain. Furthermore, we also found that the action of CRMMs depends on the microenvironmental context because it also works in conjunction with dkk1, when dkk1 is overexpressed. Thus, we propose CRMMs as a further mechanism of prechordal plate protection against the Wnt signals secreted by posterolateral cells, complementing the action of secreted antagonists.


Subject(s)
Body Patterning/genetics , Membrane Microdomains/genetics , Wnt Proteins/genetics , beta Catenin/genetics , Animals , Cholesterol/metabolism , Gene Expression Regulation, Developmental/drug effects , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway/drug effects , Xenopus laevis/genetics , Xenopus laevis/growth & development , beta Catenin/metabolism , beta-Cyclodextrins/pharmacology
11.
Dev Cell ; 32(6): 719-30, 2015 Mar 23.
Article in English | MEDLINE | ID: mdl-25771893

ABSTRACT

Secreted Wnt morphogens are essential for embryogenesis and homeostasis and require a lipid/palmitoleoylate modification for receptor binding and activity. Notum is a secreted Wnt antagonist that belongs to the α/ß hydrolase superfamily, but its mechanism of action and roles in vertebrate embryogenesis are not fully understood. Here, we report that Notum hydrolyzes the Wnt palmitoleoylate adduct extracellularly, resulting in inactivated Wnt proteins that form oxidized oligomers incapable of receptor binding. Thus, Notum is a Wnt deacylase, and palmitoleoylation is obligatory for the Wnt structure that maintains its active monomeric conformation. Notum is expressed in naive ectoderm and neural plate in Xenopus and is required for neural and head induction. These findings suggest that Notum is a prerequisite for the "default" neural fate and that distinct mechanisms of Wnt inactivation by the Tiki protease in the Organizer and the Notum deacylase in presumptive neuroectoderm orchestrate vertebrate brain development.


Subject(s)
Esterases/genetics , Head/embryology , Neurogenesis/physiology , Wnt Proteins/metabolism , Xenopus Proteins/metabolism , Animals , Body Patterning/genetics , Ectoderm/metabolism , Embryo, Nonmammalian , Gene Expression Regulation, Developmental , Gene Knockout Techniques , Gene Silencing , HEK293 Cells , Humans , Mice , Molecular Sequence Data , Morpholinos , Neural Plate/metabolism , Oxidation-Reduction , Palmitic Acid/chemistry , Protein Binding , Protein Conformation , Wnt Proteins/antagonists & inhibitors , Wnt Proteins/chemistry , Xenopus Proteins/genetics , Xenopus laevis
12.
Cancer Cell Int ; 14: 61, 2014.
Article in English | MEDLINE | ID: mdl-25120383

ABSTRACT

Connective tissue growth factor (CTGF)/CCN family member 2 (CCN2) is a CCN family member of matricellular signaling modulators. It has been shown that CCN2/CTGF mediates cell adhesion, aggregation and migration in a large variety of cell types, including vascular endothelial cells, fibroblasts, epithelial cells, aortic smooth muscle and also pluripotent stem cells. Others matricellular proteins are capable of interacting with CCN2/CTGF to mediate its function. Cell migration is a key feature for tumor cell invasion and metastasis. CCN2/CTGF seems to be a prognostic marker for cancer. In addition, here we intend to discuss recent discoveries and a new strategy to develop therapies against CCN2/CTGF, in order to treat cancer metastasis.

13.
PLoS One ; 8(1): e55605, 2013.
Article in English | MEDLINE | ID: mdl-23383241

ABSTRACT

Connective-tissue growth factor (CTGF/CCN2) is a matricellular-secreted protein involved in complex processes such as wound healing, angiogenesis, fibrosis and metastasis, in the regulation of cell proliferation, migration and extracellular matrix remodeling. Glioblastoma (GBM) is the major malignant primary brain tumor and its adaptation to the central nervous system microenvironment requires the production and remodeling of the extracellular matrix. Previously, we published an in vitro approach to test if neurons can influence the expression of the GBM extracellular matrix. We demonstrated that neurons remodeled glioma cell laminin. The present study shows that neurons are also able to modulate CTGF expression in GBM. CTGF immnoreactivity and mRNA levels in GBM cells are dramatically decreased when these cells are co-cultured with neonatal neurons. As proof of particular neuron effects, neonatal neurons co-cultured onto GBM cells also inhibit the reporter luciferase activity under control of the CTGF promoter, suggesting inhibition at the transcription level. This inhibition seems to be contact-mediated, since conditioned media from embryonic or neonatal neurons do not affect CTGF expression in GBM cells. Furthermore, the inhibition of CTGF expression in GBM/neuronal co-cultures seems to affect the two main signaling pathways related to CTGF. We observed inhibition of TGFß luciferase reporter assay; however phopho-SMAD2 levels did not change in these co-cultures. In addition levels of phospho-p44/42 MAPK were decreased in co-cultured GBM cells. Finally, in transwell migration assay, CTGF siRNA transfected GBM cells or GBM cells co-cultured with neurons showed a decrease in the migration rate compared to controls. Previous data regarding laminin and these results demonstrating that CTGF is down-regulated in GBM cells co-cultured with neonatal neurons points out an interesting view in the understanding of the tumor and cerebral microenvironment interactions and could open up new strategies as well as suggest a new target in GBM control.


Subject(s)
Cell Communication , Connective Tissue Growth Factor/metabolism , Glioblastoma/metabolism , Neurons/metabolism , Animals , Cell Line, Tumor , Cell Movement , Coculture Techniques , Connective Tissue Growth Factor/genetics , Gene Expression , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Primary Cell Culture , Promoter Regions, Genetic , Rats , Signal Transduction , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transcriptional Activation , Transforming Growth Factor beta/metabolism
14.
Cell ; 151(6): 1200-13, 2012 Dec 07.
Article in English | MEDLINE | ID: mdl-23217707

ABSTRACT

Ten-Eleven Translocation (Tet) family of dioxygenases dynamically regulates DNA methylation and has been implicated in cell lineage differentiation and oncogenesis. Yet their functions and mechanisms of action in gene regulation and embryonic development are largely unknown. Here, we report that Xenopus Tet3 plays an essential role in early eye and neural development by directly regulating a set of key developmental genes. Tet3 is an active 5mC hydroxylase regulating the 5mC/5hmC status at target gene promoters. Biochemical and structural studies further demonstrate that the Tet3 CXXC domain is critical for specific Tet3 targeting. Finally, we show that the enzymatic activity and CXXC domain are both crucial for Tet3's biological function. Together, these findings define Tet3 as a transcription regulator and reveal a molecular mechanism by which the 5mC hydroxylase and DNA binding activities of Tet3 cooperate to control target gene expression and embryonic development.


Subject(s)
Dioxygenases/chemistry , Dioxygenases/metabolism , Eye/embryology , Neurogenesis , Xenopus Proteins/chemistry , Xenopus Proteins/metabolism , Xenopus laevis/embryology , Animals , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Dioxygenases/genetics , Molecular Sequence Data , Promoter Regions, Genetic , Protein Structure, Tertiary , Xenopus Proteins/genetics , Xenopus laevis/metabolism
15.
Cell ; 149(7): 1565-77, 2012 Jun 22.
Article in English | MEDLINE | ID: mdl-22726442

ABSTRACT

Secreted Wnt morphogens are signaling molecules essential for embryogenesis, pathogenesis, and regeneration and require distinct modifications for secretion, gradient formation, and activity. Whether Wnt proteins can be posttranslationally inactivated during development and homeostasis is unknown. Here we identify, through functional cDNA screening, a transmembrane protein Tiki1 that is expressed specifically in the dorsal Spemann-Mangold Organizer and is required for anterior development during Xenopus embryogenesis. Tiki1 antagonizes Wnt function in embryos and human cells via a TIKI homology domain that is conserved from bacteria to mammals and acts likely as a protease to cleave eight amino-terminal residues of a Wnt protein, resulting in oxidized Wnt oligomers that exhibit normal secretion but minimized receptor-binding capability. Our findings identify a Wnt-specific protease that controls head formation, reveal a mechanism for morphogen inactivation through proteolysis-induced oxidation-oligomerization, and suggest a role of the Wnt amino terminus in evasion of oxidizing inactivation. TIKI proteins may represent potential therapeutic targets.


Subject(s)
Body Patterning , Head/embryology , Membrane Proteins/metabolism , Metalloproteases/metabolism , Wnt Signaling Pathway , Xenopus Proteins/metabolism , Xenopus/embryology , Amino Acid Sequence , Animals , Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental , HEK293 Cells , HeLa Cells , Humans , Membrane Proteins/genetics , Metalloproteases/genetics , Molecular Sequence Data , Organizers, Embryonic/metabolism , Sequence Alignment , Xenopus/metabolism , Xenopus Proteins/genetics
17.
Toxicol Pathol ; 37(2): 175-82, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19332661

ABSTRACT

Cigarette smoke has been associated with poor healing in several studies, but the precise mechanisms involving this impairment are still not elucidated. The aim of this work was to investigate cigarette smoke exposure effects on initial phases of cutaneous healing in mice, focusing mainly on gene expression of two molecules involved in wound repair (Ccn2/Ctgf and Tgfb1) and to study if these effects are strain dependent. Mice were exposed to the smoke of nine cigarettes per day, three times per day, for ten days. In the eleventh day an excisional wound was made. The control group was sham-exposed. The cigarette smoke exposure protocol was performed until euthanasia, seven days after wounding. Wound contraction was evaluated. Sections were stained with hematoxylin-eosin, Sirius red, and toluidine blue, and also immunostained for alpha-smooth muscle actin. Gene expression of Ccn2/Ctgf and Tgfb1 was evaluated by semiquantitative reverse transcriptase polymerase chain reaction (RT-PCR). Smoke-exposed animals presented delay in wound contraction; fibroblastic, inflammatory, and mast cell recruitment; re-epithelialization; myofibroblastic differentiation; and Ccn2/Ctgf and Tgfb1 gene expression. Those alterations were strain dependent. This work confirmed the deleterious effects of cigarette smoke exposure on mouse cutaneous healing depending on mouse strain and links these effects to an overexpression of Ccn2/Ctgf.


Subject(s)
Connective Tissue Growth Factor/metabolism , Nicotiana/toxicity , Smoke/adverse effects , Wound Healing/genetics , Animals , Connective Tissue Growth Factor/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Skin/chemistry , Species Specificity
18.
Differentiation ; 75(3): 184-92, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17359297

ABSTRACT

Myogenic differentiation is a multistep process that begins with the commitment of mononucleated precursors that withdraw from cell cycle. These myoblasts elongate while aligning to each other, guided by the recognition between their membranes. This step is followed by cell fusion and the formation of long and striated multinucleated myotubes. We have recently shown that cholesterol depletion by methyl-beta-cyclodextrin (MbetaCD) induces myogenic differentiation by enhancing myoblast recognition and fusion. Here, we further studied the signaling pathways responsible for early steps of myogenesis. As it is known that Wnt plays a role in muscle differentiation, we used the chemical MbetaCD to deplete membrane cholesterol and investigate the involvement of the Wnt/beta-catenin pathway during myogenesis. We show that cholesterol depletion promoted a significant increase in expression of beta-catenin, its nuclear translocation and activation of the Wnt pathway. Moreover, we show that the activation of the Wnt pathway after cholesterol depletion can be inhibited by the soluble protein Frzb-1. Our data suggest that membrane cholesterol is involved in Wnt/beta-catenin signaling in the early steps of myogenic differentiation.


Subject(s)
Cholesterol/metabolism , Muscle Fibers, Skeletal/physiology , Wnt Proteins/metabolism , beta Catenin/metabolism , Animals , Cell Differentiation , Cell Membrane/metabolism , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Chick Embryo/metabolism , Frizzled Receptors/metabolism , Humans , Models, Biological , Muscle Fibers, Skeletal/cytology , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Transfection , Wnt Proteins/antagonists & inhibitors , beta Catenin/antagonists & inhibitors , beta-Cyclodextrins/pharmacology
19.
Differentiation ; 74(9-10): 562-72, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17177853

ABSTRACT

The harmonious development of the central nervous system depends on the interactions of the neuronal and glial cells. Extracellular matrix elements play important roles in these interactions, especially laminin produced by astrocytes, which has been shown to be a good substrate for neuron growth and axonal guidance. Glioblastomas are the most common subtypes of primary brain tumors and may be astrocytes in origin. As normal laminin-producing glial cells are the preferential substrate for neurons, and glial tumors have been shown to produce laminin, we questioned whether glioblastoma retained the same normal glial-neuron interactive properties with respect to neuronal growth and differentiation. Then, rat neurons were co-cultured onto rat normal astrocytes or onto three human glioblastoma cell lines obtained from neurosurgery. The co-culture confirmed that human glioblastoma cells as well as astrocytes maintained the ability to support neuritogenesis, but non-neural normal or tumoral cells failed to do so. However, glioblastoma cells did not distinguish embryonic from post-natal neurons in relation to neurite pattern in the co-cultures, as normal astrocytes did. Further, the laminin organization on both normal and tumoral glial cells was altered from a filamentous arrangement to a mixed punctuate/filamentous pattern when in co-culture with neurons. Together, these results suggest that glioblastoma cells could identify neuronal cells as partners, to support their growth and induce complex neurites, but they lost the normal glia property to distinguish neuronal age. In addition, our results show for the first time that neurons modulate the organization of astrocytes and glioblastoma laminin on the extracellular matrix.


Subject(s)
Astrocytes/chemistry , Brain Neoplasms/physiopathology , Brain/cytology , Glioblastoma/physiopathology , Laminin/analysis , Neurites/ultrastructure , Neurons/physiology , Animals , Astrocytes/cytology , Astrocytes/physiology , Brain Neoplasms/chemistry , Brain Neoplasms/pathology , Cell Differentiation , Cells, Cultured , Glioblastoma/chemistry , Glioblastoma/pathology , Humans , Neurites/metabolism , Neurons/metabolism , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...