Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Dev Dyn ; 249(5): 646-655, 2020 05.
Article in English | MEDLINE | ID: mdl-31872525

ABSTRACT

BACKGROUND: Dysfunction of GABAergic and glutamatergic neurons in the brain, which establish inhibitory and excitatory networks, respectively, may cause diverse neurological disorders. The mechanism underlying the determination of GABAergic vs. glutamatergic neurotransmitter phenotype in the caudal diencephalon remains largely unknown. RESULTS: In this study, we investigated the consequence of Tcf7l2 (transcription factor 7-like 2) ablation on the neurotransmitter identity of GABAergic and glutamatergic neurons in the caudal diencephalon. We identified positive and negative activity in the control of glutamatergic and GABAergic neuronal gene expression by Tcf7l2. Loss of Tcf7l2 did not alter the initial acquisition of the neurotransmitter identity in thalamic neurons. However, glutamatergic thalamic neurons failed to maintain their excitatory neurotransmitter phenotype in the absence of Tcf7l2. Moreover, a subset of Tcf7l2-deficient thalamic neurons underwent a glutamatergic to GABAergic neurotransmitter identity switch. Our data indicate that Tcf7l2 may promote glutamatergic neuronal differentiation and repress GABAergic neurotransmitter identity in the caudal thalamus. CONCLUSIONS: This study provides evidence for a novel and crucial role of Tcf7l2 in the molecular mechanism by which the neurotransmitter identity of glutamatergic thalamic neurons is established. Our findings exemplify a clear case of neurotransmitter identity regulation that is partitioned into initiation and maintenance phases.


Subject(s)
Thalamus , Transcription Factor 7-Like 2 Protein , Diencephalon , Neurons/metabolism , Neurotransmitter Agents/metabolism , Transcription Factor 7-Like 2 Protein/genetics , Transcription Factor 7-Like 2 Protein/metabolism
2.
Phytother Res ; 32(1): 84-93, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29193419

ABSTRACT

The metabolic syndrome (MetS) is a multicomponent condition with a complex etiology involving genetic and environmental factors. Artichoke leaf extract (ALE) has shown favorable effects on lipid and glucose metabolism. The present study aimed to investigate the effects of ALE supplementation on metabolic parameters in women with MetS, using a nutrigenetics approach. In this double-blind randomized clinical trial, 50 women (aged 20-50 years) with MetS were randomly allocated into the two groups: "ALE group" (received 1,800 mg hydroalcoholic extract of artichoke as four tablets per day) and "placebo group" (received placebo consisted of corn starch, lactose, and avicel as four tablets per day) for 12 weeks. The biochemical and anthropometric parameters were determined before and after the intervention. The FTO-rs9939609 and the TCF7L2-rs7903146 polymorphisms were genotyped by polymerase chain reaction-restriction fragment length polymorphism. In carriers of A allele of the FTO-rs9939609, ALE supplementation resulted in a statistically significant decrease in serum triglyceride level compared with placebo (-19.11% vs. 10.83%; p < .05), with no other significant differences between the two groups. The TCF7L2-rs7903146 polymorphism showed no interaction with response to ALE (p > .05). These findings suggest that ALE supplementation may improve serum triglyceride level in A allele genotype of FTO-rs9939609 polymorphism in women with MetS.


Subject(s)
Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism , Cynara scolymus/chemistry , Metabolic Syndrome/drug therapy , Transcription Factor 7-Like 2 Protein/metabolism , Adult , Double-Blind Method , Female , Genotype , Humans , Male , Metabolic Syndrome/pathology , Middle Aged , Polymorphism, Genetic , Young Adult
3.
Dev Biol ; 424(1): 62-76, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28219675

ABSTRACT

The thalamus acts as a central integrator for processing and relaying sensory and motor information to and from the cerebral cortex, and the habenula plays pivotal roles in emotive decision making by modulating dopaminergic and serotonergic circuits. These neural compartments are derived from a common developmental progenitor domain, called prosomere 2, in the caudal forebrain. Thalamic and habenular neurons exhibit distinct molecular profile, neurochemical identity, and axonal circuitry. However, the mechanisms of how their progenitors in prosomere 2 give rise to these two populations of neurons and contribute to the forebrain circuitry remains unclear. In this study, we discovered a previously unrecognized role for Tcf7l2, a transcription factor known as the canonical Wnt nuclear effector and diabetes risk-conferring gene, in establishing neuronal identity and circuits of the caudal forebrain. Using genetic and chemical axon tracers, we showed that efferent axons of the thalamus, known as the thalamocortical axons (TCAs), failed to elongate normally and strayed from their normal course to inappropriate locations in the absence of Tcf7l2. Further experiments with thalamic explants revealed that the pathfinding defects of Tcf7l2-deficient TCAs were associated at least in part with downregulation of guidance receptors Robo1 and Robo2 expression. Moreover, the fasciculus retroflexus, the main habenular output tract, was missing in embryos lacking Tcf7l2. These axonal defects may result from dysregulation of Nrp2 guidance receptor. Strikingly, loss of Tcf7l2 caused a post-mitotic identity switch between thalamic and habenular neurons. Despite normal acquisition of progenitor identity in prosomere 2, Tcf7l2-deficient thalamic neurons adopted a molecular profile of a neighboring forebrain derivative, the habenula. Conversely, habenular neurons failed to maintain their normal post-mitotic neuronal identity and acquired a subset of thalamic neuronal features in the absence of Tcf7l2. Our findings suggest a unique role for Tcf7l2 in generating distinct neuronal phenotypes from homogeneous progenitor population, and provide a better understanding of the mechanism underlying neuronal specification, differentiation, and connectivity of the developing caudal forebrain.


Subject(s)
Habenula/cytology , Habenula/embryology , Nerve Net/metabolism , Neurons/metabolism , Thalamus/cytology , Thalamus/embryology , Transcription Factor 7-Like 2 Protein/metabolism , Animals , Axon Guidance , Axons/metabolism , Biomarkers/metabolism , Body Patterning , Diencephalon/embryology , Diencephalon/metabolism , Homeodomain Proteins/metabolism , Mice , Mitosis , Mutation/genetics , Protein Binding , Stem Cells/metabolism , Transcription, Genetic
4.
Development ; 144(7): 1211-1220, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28219951

ABSTRACT

The thalamus is a diencephalic structure that plays crucial roles in relaying and modulating sensory and motor information to the neocortex. The thalamus develops in the dorsal part of the neural tube at the level of the caudal forebrain. However, the molecular mechanisms that are essential for thalamic differentiation are still unknown. Here, we have succeeded in generating thalamic neurons from mouse embryonic stem cells (mESCs) by modifying the default method that induces the most-anterior neural type in self-organizing culture. A low concentration of the caudalizing factor insulin and a MAPK/ERK kinase inhibitor enhanced the expression of the caudal forebrain markers Otx2 and Pax6. BMP7 promoted an increase in thalamic precursors such as Tcf7l2+/Gbx2+ and Tcf7l2+/Olig3+ cells. mESC thalamic precursors began to express the glutamate transporter vGlut2 and the axon-specific marker VGF, similar to mature projection neurons. The mESC thalamic neurons extended their axons to cortical layers in both organotypic culture and subcortical transplantation. Thus, we have identified the minimum elements sufficient for in vitro generation of thalamic neurons. These findings expand our knowledge of thalamic development.


Subject(s)
Mouse Embryonic Stem Cells/cytology , Neurons/cytology , Thalamus/cytology , Animals , Bone Morphogenetic Protein 7/pharmacology , Cell Aggregation/drug effects , Cells, Cultured , Embryoid Bodies/cytology , Embryoid Bodies/drug effects , MAP Kinase Signaling System/drug effects , Mice , Mice, Inbred ICR , Mitogen-Activated Protein Kinase Kinases/metabolism , Mouse Embryonic Stem Cells/drug effects , Mouse Embryonic Stem Cells/metabolism , Neurites/drug effects , Neurites/metabolism , Neurons/drug effects , Neurons/metabolism , Neuropeptides/metabolism , Organ Culture Techniques , Protein Kinase Inhibitors/pharmacology , Rats, Sprague-Dawley , Transcription Factor 7-Like 2 Protein/metabolism
5.
Diabetes ; 62(3): 789-800, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22966074

ABSTRACT

The type 2 diabetes risk gene TCF7L2 is the effector of the Wnt signaling pathway. We found previously that in gut endocrine L-cell lines, TCF7L2 controls transcription of the proglucagon gene (gcg), which encodes the incretin hormone glucagon-like peptide-1 (GLP-1). Whereas peripheral GLP-1 stimulates insulin secretion, brain GLP-1 controls energy homeostasis through yet-to-be defined mechanisms. We aim to determine the metabolic effect of a functional knockdown of TCF7L2 by generating transgenic mice that express dominant-negative TCF7L2 (TCF7L2DN) specifically in gcg-expressing cells. The gcg-TCF7L2DN transgenic mice showed reduced gcg expression in their gut and brain, but not in pancreas. Defects in glucose homeostasis were observed in these mice, associated with attenuated plasma insulin levels in response to glucose challenge. The defect in glucose disposal was exacerbated with high-fat diet. Brain Wnt activity and feeding-mediated hypothalamic AMP-activated protein kinase (AMPK) repression in these mice were impaired. Peripheral injection of the cAMP-promoting agent forskolin increased brain ß-cat Ser675 phosphorylation and brain gcg expression and restored feeding-mediated hypothalamic AMPK repression. We conclude that TCF7L2 and Wnt signaling control gut and brain gcg expression and glucose homeostasis and speculate that positive cross-talk between Wnt and GLP-1/cAMP signaling is an underlying mechanism for brain GLP-1 in exerting its metabolic functions.


Subject(s)
Brain/metabolism , Gastrointestinal Tract/metabolism , Gene Expression Regulation , Glucose/metabolism , Proglucagon/metabolism , Transcription Factor 7-Like 2 Protein/metabolism , Wnt Signaling Pathway , AMP-Activated Protein Kinases/metabolism , Animals , Brain/cytology , Brain/drug effects , Cell Line , Colforsin/pharmacology , Cyclic AMP/agonists , Cyclic AMP/metabolism , Gastrointestinal Tract/cytology , Gene Expression Regulation/drug effects , Glucagon-Like Peptide 1/metabolism , Homeostasis/drug effects , Hypothalamus/cytology , Hypothalamus/drug effects , Hypothalamus/metabolism , Male , Mice , Mice, Transgenic , Organ Specificity , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Recombinant Fusion Proteins/metabolism , Transcription Factor 7-Like 2 Protein/genetics , Wnt Signaling Pathway/drug effects
6.
Brain Struct Funct ; 218(6): 1531-49, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23152144

ABSTRACT

ß-Catenin signaling, leading to the activation of lymphoid enhancer-binding factor 1/T cell factor (LEF1/TCF) transcription factors, plays a well-established role in transcription regulation during development and tissue homeostasis. In the adult organism, the activity of this pathway has been found in stem cell niches and postmitotic thalamic neurons. Recently, studies show that mutations in components of ß-catenin signaling networks have been associated with several psychiatric disorders, indicating the involvement of ß-catenin and LEF1/TCF proteins in the proper functioning of the brain. Here, we report a comprehensive analysis of LEF1/TCF protein localization and the expression profile of their isoforms in cortical, thalamic, and midbrain regions in mice. We detected LEF1 and TCF7L2 proteins in neurons of the thalamus and dorsal midbrain, i.e., subcortical regions specialized in the integration of diverse sources of sensory information. These neurons also exhibited nuclear localization of ß-catenin, suggesting the involvement of ß-catenin/LEF1/TCF7L2 in the regulation of gene expression in these regions. Analysis of alternative splicing and promoter usage identified brain-specific TCF7L2 isoforms and revealed a developmentally coordinated transition in the composition of LEF1 and TCF7L2 isoforms. In the case of TCF7L2, the typical brain isoforms lack the so-called C clamp; in addition, the dominant-negative isoforms are predominant in the embryonic thalamus but disappear postnatally. The present study provides a necessary framework to understand the role of LEF1/TCF factors in thalamic and midbrain development until adulthood and predicts that the regulatory role of these proteins in the adult brain is significantly different from their role in the embryonic brain or other non-neural tissues.


Subject(s)
Cerebral Cortex/metabolism , Gene Expression Regulation, Developmental/genetics , Lymphoid Enhancer-Binding Factor 1/metabolism , Mesencephalon/metabolism , Signal Transduction/genetics , Thalamus/metabolism , Transcription Factor 7-Like 2 Protein/metabolism , Animals , DNA Primers/genetics , Fluorescent Antibody Technique , Gene Expression Profiling , HeLa Cells , Humans , Image Processing, Computer-Assisted , Immunoblotting , Lymphoid Enhancer-Binding Factor 1/genetics , Mice , Mice, Inbred C57BL , Plasmids/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factor 7-Like 2 Protein/genetics , beta Catenin/metabolism
7.
J Nutr Biochem ; 23(6): 646-55, 2012 Jun.
Article in English | MEDLINE | ID: mdl-21764279

ABSTRACT

Capsaicin is a pungent ingredient in chili red peppers and has been linked to suppression of growth in various cancer cells. However, the underlying mechanism(s) by which capsaicin induces growth arrest and apoptosis of cancer cells is not completely understood. In the present study, we investigated whether capsaicin alters ß-catenin-dependent signaling in human colorectal cancer cells in vitro. Exposure of SW480, LoVo and HCT-116 cells to capsaicin suppressed cell proliferation. Transient transfection with a ß-catenin/T-cell factor (TCF)-responsive reporter indicated that capsaicin suppressed the transcriptional activity of ß-catenin/TCF. Capsaicin treatment resulted in a decrease of intracellular ß-catenin levels and a reduction of transcripts from the ß-catenin gene (CTNNB1). These results were confirmed by a reduced luciferase reporter activity driven by promoter-reporter construct containing the promoter region of the Catnb gene. In addition, capsaicin destabilized ß-catenin through enhancement of proteosomal-dependent degradation. Western blot and immunoprecipitation studies indicated that capsaicin treatment suppressed TCF-4 expression and disrupted the interaction of TCF-4 and ß-catenin. This study identifies a role for the ß-catenin/TCF-dependent pathway that potentially contributes to the anticancer activity of capsaicin in human colorectal cancer cells.


Subject(s)
Capsaicin/pharmacology , Gene Expression Regulation, Neoplastic , Plant Extracts/pharmacology , beta Catenin/genetics , Apoptosis , Blotting, Western , Capsicum/chemistry , Cell Proliferation/drug effects , Colorectal Neoplasms/metabolism , HCT116 Cells , Humans , Immunoprecipitation , Promoter Regions, Genetic , Signal Transduction , Transcription Factor 7-Like 2 Protein/genetics , Transcription Factor 7-Like 2 Protein/metabolism , Transfection/methods , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL