Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Psychiatry ; 2023 Oct 05.
Article in English | MEDLINE | ID: mdl-37798419

ABSTRACT

The Wnt/ß-catenin pathway contains multiple high-confidence risk genes that are linked to neurodevelopmental disorders, including autism spectrum disorder. However, its ubiquitous roles across brain cell types and developmental stages have made it challenging to define its impact on neural circuit development and behavior. Here, we show that TCF7L2, which is a key transcriptional effector of the Wnt/ß-catenin pathway, plays a cell-autonomous role in postnatal astrocyte maturation and impacts adult social behavior. TCF7L2 was the dominant Wnt effector that was expressed in both mouse and human astrocytes, with a peak during astrocyte maturation. The conditional knockout of Tcf7l2 in postnatal astrocytes led to an enlargement of astrocytes with defective tiling and gap junction coupling. These mice also exhibited an increase in the number of cortical excitatory and inhibitory synapses and a marked increase in social interaction by adulthood. These data reveal an astrocytic role for developmental Wnt/ß-catenin signaling in restricting excitatory synapse numbers and regulating adult social behavior.

2.
Front Neuroanat ; 16: 838567, 2022.
Article in English | MEDLINE | ID: mdl-35356436

ABSTRACT

The pretectum has a distinct nuclear arrangement and complex neurochemical anatomy. While previous genoarchitectural studies have described rostrocaudal and dorsoventral progenitor domains and subdomains in different species, the relationship between these early partitions and its later derivatives in the mature anatomy is less understood. The signals and transcription factors that control the establishment of pretectal anatomy are practically unknown. We investigated the possibility that some aspects of the development of pretectal divisions are controlled by Wnt signaling, focusing on the transitional stage between neurogenesis and histogenesis in zebrafish. Using several molecular markers and following the prosomeric model, we identified derivatives from each rostrocaudal pretectal progenitor domain and described the localization of gad1b-positive GABAergic and vglut2.2-positive glutamatergic cell clusters. We also attempted to relate these clusters to pretectal nuclei in the mature brain. Then, we examined the influence of Wnt signaling on the size of neurochemically distinctive pretectal areas, using a chemical inhibitor of the Wnt pathway and the CRISPR/Cas9 approach to knock out genes that encode the Wnt pathway mediators, Lef1 and Tcf7l2. The downregulation of the Wnt pathway led to a decrease in two GABAergic clusters and an expansion of a glutamatergic subregion in the maturing pretectum. This revealed an instructive role of the Wnt signal in the development of the pretectum during neurogenesis. The molecular anatomy presented here improves our understanding of pretectal development during early postmitotic stages and support the hypothesis that Wnt signaling is involved in shaping the neurochemical organization of the pretectum.

3.
Cell Death Dis ; 12(12): 1111, 2021 11 27.
Article in English | MEDLINE | ID: mdl-34839359

ABSTRACT

Chemoresistance constitutes a major challenge in the treatment of triple-negative breast cancer (TNBC). Mixed-Lineage Kinase 4 (MLK4) is frequently amplified or overexpressed in TNBC where it facilitates the aggressive growth and migratory potential of breast cancer cells. However, the functional role of MLK4 in resistance to chemotherapy has not been investigated so far. Here, we demonstrate that MLK4 promotes TNBC chemoresistance by regulating the pro-survival response to DNA-damaging therapies. We observed that MLK4 knock-down or inhibition sensitized TNBC cell lines to chemotherapeutic agents in vitro. Similarly, MLK4-deficient cells displayed enhanced sensitivity towards doxorubicin treatment in vivo. MLK4 silencing induced persistent DNA damage accumulation and apoptosis in TNBC cells upon treatment with chemotherapeutics. Using phosphoproteomic profiling and reporter assays, we demonstrated that loss of MLK4 reduced phosphorylation of key DNA damage response factors, including ATM and CHK2, and compromised DNA repair via non-homologous end-joining pathway. Moreover, our mRNA-seq analysis revealed that MLK4 is required for DNA damage-induced expression of several NF-кB-associated cytokines, which facilitate TNBC cells survival. Lastly, we found that high MLK4 expression is associated with worse overall survival of TNBC patients receiving anthracycline-based neoadjuvant chemotherapy. Collectively, these results identify a novel function of MLK4 in the regulation of DNA damage response signaling and indicate that inhibition of this kinase could be an effective strategy to overcome TNBC chemoresistance.


Subject(s)
DNA Damage/genetics , High-Throughput Nucleotide Sequencing/methods , MAP Kinase Kinase Kinases/genetics , Oncogenes/genetics , Triple Negative Breast Neoplasms/genetics , Animals , Cell Line, Tumor , Female , Humans , Mice , Transfection , Triple Negative Breast Neoplasms/pathology
4.
Neuroscience ; 453: 81-101, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33227236

ABSTRACT

Studies of cortical function-recovery require a comparison between normal and post-stroke conditions that lead to changes in cortical metaplasticity. Focal cortical stroke impairs experience-dependent plasticity in the neighboring somatosensory cortex and usually evokes periinfarct depolarizations (PiDs) - spreading depression-like waves. Experimentally induced spreading depressions (SDs) affect gene expression and some of these changes persist for at least 30 days. In this study we compare the effects of non-stroke depolarizations that impair cortical experience-dependent plasticity to the effects of stroke, by inducing experience-dependent plasticity in rats with SDs or PiDs by a month of contralateral partial whiskers deprivation. We found that whiskers' deprivation after SDs resulted in normal cortical representation enlargement suggesting that SDs and PiDs depolarization have no influence on experience-dependent plasticity cortical map reorganization. PiDs and the MMP-9, -3, -2 or COX-2 proteins, which are assumed to influence metaplasticity in rats after stroke were compared between SDs induced by high osmolarity KCl solution and the PiDs that followed cortical photothrombotic stroke (PtS). We found that none of these factors directly caused cortical post-stroke metaplasticity changes. The only significant difference between stoke and induced SD was a greater imbalance in interhemispheric activity equilibrium after stroke. The interhemispheric interactions that were modified by stroke may therefore be promising targets for future studies of post-stroke experience-dependent plasticity and of recuperation studies.


Subject(s)
Cortical Spreading Depression , Stroke , Animals , Depression , Rats , Somatosensory Cortex , Vibrissae
5.
Development ; 147(16)2020 08 25.
Article in English | MEDLINE | ID: mdl-32675279

ABSTRACT

Neuronal phenotypes are controlled by terminal selector transcription factors in invertebrates, but only a few examples of such regulators have been provided in vertebrates. We hypothesised that TCF7L2 regulates different stages of postmitotic differentiation in the thalamus, and functions as a thalamic terminal selector. To investigate this hypothesis, we used complete and conditional knockouts of Tcf7l2 in mice. The connectivity and clustering of neurons were disrupted in the thalamo-habenular region in Tcf7l2-/- embryos. The expression of subregional thalamic and habenular transcription factors was lost and region-specific cell migration and axon guidance genes were downregulated. In mice with a postnatal Tcf7l2 knockout, the induction of genes that confer thalamic terminal electrophysiological features was impaired. Many of these genes proved to be direct targets of TCF7L2. The role of TCF7L2 in terminal selection was functionally confirmed by impaired firing modes in thalamic neurons in the mutant mice. These data corroborate the existence of master regulators in the vertebrate brain that control stage-specific genetic programmes and regional subroutines, maintain regional transcriptional network during embryonic development, and induce terminal selection postnatally.


Subject(s)
Cell Differentiation , Gene Expression Regulation, Developmental , Mitosis , Synaptic Transmission , Thalamus/embryology , Transcription Factor 4/metabolism , Animals , Mice , Mice, Knockout , Thalamus/cytology , Transcription Factor 4/genetics
6.
FEBS Lett ; 593(13): 1654-1674, 2019 07.
Article in English | MEDLINE | ID: mdl-31218672

ABSTRACT

Canonical Wnt signaling, which is transduced by ß-catenin and lymphoid enhancer factor 1/T cell-specific transcription factors (LEF1/TCFs), regulates many aspects of metazoan development and tissue renewal. Although much evidence has associated canonical Wnt/ß-catenin signaling with mood disorders, the mechanistic links are still unknown. Many components of the canonical Wnt pathway are involved in cellular processes that are unrelated to classical canonical Wnt signaling, thus further blurring the picture. The present review critically evaluates the involvement of classical Wnt/ß-catenin signaling in developmental processes that putatively underlie the pathology of mental illnesses. Particular attention is given to the roles of LEF1/TCFs, which have been discussed surprisingly rarely in this context. Highlighting recent discoveries, we propose that alterations in the activity of LEF1/TCFs, and particularly of transcription factor 7-like 2 (TCF7L2), result in defects previously associated with neuropsychiatric disorders, including imbalances in neurogenesis and oligodendrogenesis, the functional disruption of thalamocortical circuitry and dysfunction of the habenula.


Subject(s)
Brain/cytology , Brain/growth & development , Mental Disorders/metabolism , Mental Disorders/pathology , Transcription Factor 7-Like 2 Protein/metabolism , Wnt Signaling Pathway , Animals , Brain/metabolism , Brain/pathology , Humans , Neurogenesis
7.
Glia ; 65(1): 34-49, 2017 01.
Article in English | MEDLINE | ID: mdl-27534376

ABSTRACT

ST8SIA2 is a polysialyltransferase that attaches polysialic acid to the glycoproteins NCAM1 and CADM1. Polysialylation is involved in brain development and plasticity. ST8SIA2 is a schizophrenia candidate gene, and St8sia2-/- mice exhibit schizophrenia-like behavior. We sought to identify new pathological consequences of ST8SIA2 deficiency. Our proteomic analysis suggested myelin impairment in St8sia2-/- mice. Histological and immune staining together with Western blot revealed that the onset of myelination was not delayed in St8sia2-/- mice, but the content of myelin was lower. Ultrastructure analysis of the corpus callosum showed thinner myelin sheaths, smaller and irregularly shaped axons, and white matter lesions in adult St8sia2-/- mice. Then we evaluated oligodendrocyte differentiation in vivo and in vitro. Fewer OLIG2+ cells in the cortex and corpus callosum, together with the higher percentage of undifferentiated oligodenroglia in St8sia2-/- mice suggested an impairment in oligodendrocyte generation. Experiment on primary cultures of oligodendrocyte precursor cells (OPCs) confirmed a cell-autonomous effect of ST8SIA2 in oligodendroglia, and demonstrated that OPC to oligodendrocyte transition is inhibited in St8sia2-/- mice. Concluding, ST8SIA2-mediated polysialylation influences on oligodendrocyte differentiation, and oligodendrocyte deficits in St8sia2 mice are a possible cause of the demyelination and degeneration of axons, resembling nerve fiber alterations in schizophrenia. GLIA 2016;65:34-49.


Subject(s)
Axons/drug effects , Cell Differentiation/drug effects , Myelin Sheath/drug effects , Oligodendroglia/drug effects , Sialyltransferases/pharmacology , Animals , Axons/metabolism , Brain/metabolism , Brain/pathology , Cell Differentiation/physiology , Mice, Knockout , Myelin Sheath/metabolism , Nerve Tissue Proteins/metabolism , Neurogenesis/drug effects , Oligodendroglia/cytology , Oligodendroglia/metabolism , Stem Cells/drug effects , Stem Cells/metabolism
8.
Neuropharmacology ; 113(Pt A): 490-501, 2017 02.
Article in English | MEDLINE | ID: mdl-27793772

ABSTRACT

The mechanism of lithium's therapeutic action remains obscure, hindering the discovery of safer treatments for bipolar disorder. Lithium can act as an inhibitor of the kinase GSK3α/ß, which in turn negatively regulates ß-catenin, a co-activator of LEF1/TCF transcription factors. However, unclear is whether therapeutic levels of lithium activate ß-catenin in the brain, and whether this activation could have a therapeutic significance. To address this issue we chronically treated mice with lithium. Although the level of non-phospho-ß-catenin increased in all of the brain areas examined, ß-catenin translocated into cellular nuclei only in the thalamus. Similar results were obtained when thalamic and cortical neurons were treated with a therapeutically relevant concentration of lithium in vitro. We tested if TCF7L2, a member of LEF1/TCF family that is highly expressed in the thalamus, facilitated the activation of ß-catenin. Silencing of Tcf7l2 in thalamic neurons prevented ß-catenin from entering the nucleus, even when the cells were treated with lithium. Conversely, when Tcf7l2 was ectopically expressed in cortical neurons, ß-catenin shifted to the nucleus, and lithium augmented this process. Lastly, we silenced tcf7l2 in zebrafish and exposed them to lithium for 3 days, to evaluate whether TCF7L2 is involved in the behavioral response. Lithium decreased the dark-induced activity of control zebrafish, whereas the activity of zebrafish with tcf7l2 knockdown was unaltered. We conclude that therapeutic levels of lithium activate ß-catenin selectively in thalamic neurons. This effect is determined by the presence of TCF7L2, and potentially contributes to the therapeutic response.


Subject(s)
Lithium/administration & dosage , Locomotion/physiology , Models, Animal , Neurons/physiology , Transcription Factor 7-Like 2 Protein/physiology , Animals , Brain/cytology , Brain/drug effects , Brain/physiology , Cells, Cultured , Drug Administration Schedule , Locomotion/drug effects , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Rats , Zebrafish
9.
Toxicol In Vitro ; 26(6): 897-906, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22543296

ABSTRACT

We have examined the effects of nitric oxide donors and acrylamide on mesenchymal progenitor cell (hMPC) viability, programmed cell death (PCD) and differentiation. Acrylamide was examined at 0.5mM and 1.5mM concentrations, NOC-18 at 10µM and SNP at 100µM. Cell viability was assayed with MTS, PCD was determined by phosphatidylserine, caspase-9 and -3/7 and mitochondrial membrane potential assays, and osteogenic cell differentiation was evaluated by alkaline phosphatase activity (ALP) and mRNA levels for collagen type I, bone sialoprotein, ostepontin and osteocalcin. Serum-free hMPC cultures treated with 1.5mM acrylamide and SNP for 72h demonstrated reduced viability. PCD analyses revealed that SNP stimulated cells to necrosis in reactive species-dependent manner. Acrylamide (1.5mM) led to apoptosis independent of reactive species. Acrylamide and SNP reduced ALP activity and collagen type I mRNA levels but mRNA levels for bone sialoprotein and osteopontin increased in SNP treated cells and remained unchanged in acrylamide. Acrylamide had no effect on guanylate cyclase and cGMP osteogenic signaling pathway. The study suggests that acrylamide might impair bone development and remodeling upon acute or prolonged intoxication with this compound of mesenchymal cells.


Subject(s)
Acrylamide/toxicity , Mesenchymal Stem Cells/drug effects , Nitric Oxide Donors/toxicity , Nitroprusside/toxicity , Nitroso Compounds/toxicity , Alkaline Phosphatase/metabolism , Caspases/metabolism , Cell Death/drug effects , Cell Differentiation/drug effects , Cell Line , Cell Survival/drug effects , Collagen Type I/genetics , Humans , Membrane Potential, Mitochondrial/drug effects , Mesenchymal Stem Cells/metabolism , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...