Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros












Base de dados
Intervalo de ano de publicação
1.
Artigo em Inglês | MEDLINE | ID: mdl-32113851

RESUMO

Glycogen synthase kinase 3ß (GSK3ß) has gained interest regarding its involvement in psychiatric and neurodegenerative disorders. Recently GSK3 inhibitors were highlighted as promising rescuers of cognitive impairments for a gamut of CNS disorders. Growing evidence supports that fast-spiking parvalbumin (PV) interneurons are critical regulators of cortical computation. Albeit, how excitatory receptors on PV interneurons are regulated and how this affects cognitive function remains unknown. To address these questions, we have generated a novel triple-transgenic conditional mouse with GSK3ß genetically deleted from PV interneurons. PV-GSK3ß-/- resulted in increased excitability and augmented excitatory synaptic strength in prefrontal PV interneurons. More importantly, these synaptic changes are correlated with accelerated learning with no changes in locomotion and sociability. Our study, for the first time, examined how GSK3ß activity affects learning capability via regulation of PV interneurons. This study provides a novel insight into how GSK3ß may contribute to disorders afflicted by cognitive deficits.


Assuntos
Potenciais Pós-Sinápticos Excitadores/fisiologia , Glicogênio Sintase Quinase 3 beta/deficiência , Interneurônios/metabolismo , Aprendizagem/fisiologia , Parvalbuminas/biossíntese , Sinapses/metabolismo , Fatores Etários , Animais , Feminino , Deleção de Genes , Expressão Gênica , Glicogênio Sintase Quinase 3 beta/genética , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Transgênicos , Técnicas de Cultura de Órgãos , Parvalbuminas/genética , Sinapses/genética
2.
J Immunol ; 203(11): 2990-2999, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31619538

RESUMO

The protein tyrosine kinase Src regulates the synthesis of TLR3-mediated IFN-ß via the TBK1-IFN regulatory factor 3 axis. However, the molecular mechanisms regulating Src activity in TLR3 signaling remain unclear. In this study, we report that GSK3ß regulates Src phosphorylation via TNFR-associated factor 2 (TRAF2)-mediated Src ubiquitination. GSK3ß deficiency in mouse embryonic fibroblasts significantly reduces polyinosinic:polycytidylic acid-induced IFN-ß and IFN-stimulated gene expression, which is caused by diminished phosphorylation of Src at tyrosine 416. Src undergoes polyinosinic:polycytidylic acid-dependent lysine 63 chain ubiquitination, and TRAF2 is a direct E3 ligase for Src. Our study reveals novel mechanisms underlying TLR3-mediated antiviral responses mediated via the GSK3ß-TRAF2-Src axis.


Assuntos
Glicogênio Sintase Quinase 3 beta/metabolismo , Fator 2 Associado a Receptor de TNF/metabolismo , Receptor 3 Toll-Like/metabolismo , Quinases da Família src/metabolismo , Animais , Células Cultivadas , Glicogênio Sintase Quinase 3 beta/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação , Células RAW 264.7 , Ubiquitinação
3.
Cells ; 8(9)2019 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-31443508

RESUMO

There are contradictory reports on the role of the serine/threonine kinase isoform glycogen synthase kinase-3ß (GSK3ß) after injury to the central nervous system (CNS). Some report that GSK3 activity promotes axonal growth or myelin disinhibition, whilst others report that GSK3 activity prevents axon regeneration. In this study, we sought to clarify if suppression of GSK3ß alone and in combination with the cellular-stress-induced factor RTP801 (also known as REDD1: regulated in development and DNA damage response protein), using translationally relevant siRNAs, promotes retinal ganglion cell (RGC) survival and neurite outgrowth/axon regeneration. Adult mixed retinal cell cultures, prepared from rats at five days after optic nerve crush (ONC) to activate retinal glia, were treated with siRNA to GSK3ß (siGSK3ß) alone or in combination with siRTP801 and RGC survival and neurite outgrowth were quantified in the presence and absence of Rapamycin or inhibitory Nogo-A peptides. In in vivo experiments, either siGSK3ß alone or in combination with siRTP801 were intravitreally injected every eight days after ONC and RGC survival and axon regeneration was assessed at 24 days. Optimal doses of siGSK3ß alone promoted significant RGC survival, increasing the number of RGC with neurites without affecting neurite length, an effect that was sensitive to Rapamycin. In addition, knockdown of GSK3ß overcame Nogo-A-mediated neurite growth inhibition. Knockdown of GSK3ß after ONC in vivo enhanced RGC survival but not axon number or length, without potentiating glial activation. Knockdown of RTP801 increased both RGC survival and axon regeneration, whilst the combined knockdown of GSK3ß and RTP801 significantly increased RGC survival, neurite outgrowth, and axon regeneration over and above that observed for siGSK3ß or siRTP801 alone. These results suggest that GSK3ß suppression promotes RGC survival and axon initiation whilst, when in combination with RTP801, it also enhanced disinhibited axon elongation.


Assuntos
Axônios/metabolismo , Glicogênio Sintase Quinase 3 beta/deficiência , Glicogênio Sintase Quinase 3 beta/genética , Neuritos/metabolismo , RNA Interferente Pequeno/genética , Células Ganglionares da Retina/citologia , Animais , Masculino , Ratos , Ratos Sprague-Dawley
4.
J Pharmacol Exp Ther ; 371(2): 339-347, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31420527

RESUMO

Previous research has demonstrated that activity of glycogen synthase kinase-3 (GSK3) is necessary for the rewarding effects of cocaine. In the present study, a conditional GSK3ß gene knockdown model was used to determine if GSK3ß activity specifically in the nucleus accumbens is important for cocaine conditioned reward. The roles of accumbal GSK3ß in morphine conditioned reward, trans-(±)-3,4-dichloro-N-methyl-N-[2-(1-pyrrolidinyl)cyclohexyl]benzeneacetamide methanesulfonate salt (U50,488H)-induced conditioned place aversion, and cognitive function were also studied. Adult male and female GSK3ß-floxed or wild-type mice were injected with adeno-associated virus/Cre into the nucleus accumbens to reduce expression of GSK3ß and underwent behavioral testing 4 weeks later. The development of cocaine-induced conditioned place preference was significantly attenuated in mice with reduced levels of GSK3ß in the nucleus accumbens, whereas the development of morphine-induced place preference remained intact. Conditional knockdown of GSK3ß in the accumbens prevented the development of conditioned aversion produced by U50,488H, a κ-opioid receptor agonist. Cognitive memory tests revealed deficits in object location memory, but not novel object recognition in mice with accumbal GSK3ß knockdown. These data demonstrate that GSK3ß in the nucleus accumbens is required for cocaine conditioned place preference and U50,488H conditioned place aversion, as well as spatial memory in object location task, indicating differential roles of GSK3ß in the psychostimulant and opiate reward process, as well as in memory for spatial locations and object identity. SIGNIFICANCE STATEMENT: Knockdown of GSK3ß in the nucleus accumbens attenuated the development of cocaine-induced place preference, as well as conditioned place aversion to U50,488H, a κ-opioid receptor agonist. In contrast, the development of morphine place preference was not altered by GSK3ß knockdown. GSK3ß knockdown in nucleus accumbens impaired performance in the object location task, but not the novel object recognition task. These results elucidate different physiological roles of accumbal GSKß in conditioned reward, aversion, and memory.


Assuntos
(trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida/farmacologia , Cocaína/farmacologia , Condicionamento Psicológico/fisiologia , Glicogênio Sintase Quinase 3 beta/deficiência , Memória/fisiologia , Morfina/farmacologia , Núcleo Accumbens/metabolismo , Analgésicos não Narcóticos/farmacologia , Animais , Aprendizagem da Esquiva/efeitos dos fármacos , Aprendizagem da Esquiva/fisiologia , Condicionamento Psicológico/efeitos dos fármacos , Glicogênio Sintase Quinase 3 beta/genética , Memória/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Núcleo Accumbens/efeitos dos fármacos , Distribuição Aleatória
5.
Cell Biochem Funct ; 37(5): 340-347, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31062382

RESUMO

This study was designed to investigate the molecular mechanism and biological roles of long non-coding RNA (lncRNA) brain-derived neurotrophic factor antisense (BDNF-AS) in colorectal cancer (CRC). The quantitative real-time PCR (qRT-PCR) and western blotting were performed to detect the expressions of lncRNA BDNF-AS and glycogen synthase kinase-3ß (GSK-3ß) in human CRC tissues and cell lines. The cell proliferation, transwell migration, and invasion assays were carried out to evaluate the effect of lncRNA BDNF-AS on the growth of CRC cells. RNA pull-down and RNA immunoprecipitation (RIP) assays were conducted to confirm the interaction between lncRNA BDNF-AS and enhancer of Zeste Homologue 2 (EZH2). Chromatin immunoprecipitation (ChIP) assay was used to verify the enrichment of EZH2 and histone H3 lysine 27 trimethylation (H3K27me3) in the promoter region of GSK-3ß in CRC cells. LncRNA BDNF-AS expression was significantly decreased, while GSK-3ß was highly expressed in human CRC tissues and cell lines. Moreover, lncRNA BDNF-AS induced inhibition of proliferation, migration, and invasion of CRC cells via inhibiting GSK-3ß expression. Mechanistically, BDNF-AS led to GSK-3ß promoter silencing in CRC cells through recruitment of EZH2. In conclusion, lncRNA BDNF-AS functioned as an oncogene in CRC and shed new light on lncRNA-directed therapeutics in CRC. SIGNIFICANCE OF THE STUDY: LncRNA BDNF-AS is recently reported to be remarkably downregulated in a variety of tumours and served as a tumour suppressor. However, the functions and underlying mechanism of lncRNA BDNF-AS in CRC pathogenesis have not been reported yet. Our study is the first to demonstrate the effect of lncRNA BDNF-AS in CRC and revealed that lncRNA BDNF-AS expression is negatively correlated with the aggressive biological behaviour of CRC. Further investigation demonstrated that lncRNA BDNF-AS functioned as a tumour suppressor in CRC progression by suppressing GSK-3ß expression through binding to EZH2 and H3K27me3 with the GSK-3ß promoter, shedding light on the diagnosis and therapy for CRC.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/genética , Movimento Celular/genética , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Epigênese Genética/genética , Regulação Neoplásica da Expressão Gênica , Glicogênio Sintase Quinase 3 beta/genética , RNA Longo não Codificante/genética , Proliferação de Células/genética , Glicogênio Sintase Quinase 3 beta/biossíntese , Glicogênio Sintase Quinase 3 beta/deficiência , Células HCT116 , Humanos , RNA Longo não Codificante/metabolismo
6.
PLoS One ; 14(4): e0215213, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30978208

RESUMO

Type 1 diabetic Akita mice develop severe cardiac parasympathetic dysfunction that we have previously demonstrated is due at least in part to an abnormality in the response of the end organ to parasympathetic stimulation. Specifically, we had shown that hypoinsulinemia in the diabetic heart results in attenuation of the G-protein coupled inward rectifying K channel (GIRK) which mediates the negative chronotropic response to parasympathetic stimulation due at least in part to decreased expression of the GIRK1 and GIRK4 subunits of the channel. We further demonstrated that the expression of GIRK1 and GIRK4 is under the control of the Sterol Regulatory element Binding Protein (SREBP-1), which is also decreased in response to hypoinsulinemia. Finally, given that hyperactivity of Glycogen Synthase Kinase (GSK)3ß, had been demonstrated in the diabetic heart, we demonstrated that treatment of Akita mice with Li+, an inhibitor of GSK3ß, increased parasympathetic responsiveness and SREBP-1 levels consistent with the conclusion that GSK3ß might regulate IKACh via an effect on SREBP-1. However, inhibitor studies were complicated by lack of specificity for GSK3ß. Here we generated an Akita mouse with cardiac specific inducible knockout of GSK3ß. Using this mouse, we demonstrate that attenuation of GSK3ß expression is associated with an increase in parasympathetic responsiveness measured as an increase in the heart rate response to atropine from 17.3 ± 3.5% (n = 8) prior to 41.2 ± 5.4% (n = 8, P = 0.017), an increase in the duration of carbamylcholine mediated bradycardia from 8.43 ± 1.60 min (n = 7) to 12.71 ± 2.26 min (n = 7, P = 0.028) and an increase in HRV as measured by an increase in the high frequency fraction from 40.78 ± 3.86% to 65.04 ± 5.64 (n = 10, P = 0.005). Furthermore, patch clamp measurements demonstrated a 3-fold increase in acetylcholine stimulated peak IKACh in atrial myocytes from GSK3ß deficiency mice compared with control. Finally, western blot analysis of atrial extracts from knockout mice demonstrated increased levels of SREBP-1, GIRK1 and GIRK4 compared with control. Taken together with our prior observations, these data establish a role of increased GSK3ß activity in the pathogenesis of parasympathetic dysfunction in type 1 diabetes via the regulation of IKACh and GIRK1/4 expression.


Assuntos
Diabetes Mellitus Tipo 1/fisiopatologia , Glicogênio Sintase Quinase 3 beta/deficiência , Miócitos Cardíacos/enzimologia , Sistema Nervoso Parassimpático/fisiopatologia , Animais , Diabetes Mellitus Tipo 1/enzimologia , Diabetes Mellitus Tipo 1/genética , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Glicogênio Sintase Quinase 3 beta/genética , Glicogênio Sintase Quinase 3 beta/metabolismo , Átrios do Coração/inervação , Átrios do Coração/fisiopatologia , Frequência Cardíaca/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miócitos Cardíacos/fisiologia , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo
7.
J Affect Disord ; 245: 1079-1088, 2019 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-30699850

RESUMO

BACKGROUND: Genetic and physiological studies have implicated the striatum in bipolar disorder (BD). Although Glycogen synthase kinase 3 beta (GSK3ß) has been suggested to play a role in the pathophysiology of BD since it is inhibited by lithium, it remains unknown how GSK3ß activity might be involved. Therefore we examined the functional roles of GSK3ß and one of its substrates, CRMP2, within the striatum. METHODS: Using CRISPR-Cas9 system, we specifically ablated GSK3ß in the striatal neurons in vivo and in vitro. Sholl analysis was performed for the structural studies of medium spiny neurons (MSNs) and amphetamine-induced hyperlocomotion was measured to investigate the effects of gene ablations on the mania-like symptom of BD. RESULTS: GSK3ß deficiency in cultured neurons and in neurons of adult mouse brain caused opposite patterns of neurite changes. Furthermore, specific knockout of GSK3ß in the MSNs of the indirect pathway significantly suppressed amphetamine-induced hyperlocomotion. We demonstrated that these phenotypes of GSK3ß ablation were mediated by CRMP2, a major substrate of GSK3ß. LIMITATIONS: Amphetamine-induced hyperlocomotion only partially recapitulate the symptoms of BD. It requires further study to examine whether abnormality in GSK3ß or CRMP2 is also involved in depression phase of BD. Additionally, we could not confirm whether the behavioral changes observed in GSK3ß-ablated mice were indeed caused by the cellular structural changes observed in the striatal neurons. CONCLUSION: Our results demonstrate that GSK3ß and its substrate CRMP2 critically regulate the neurite structure of MSNs and their functions specifically within the indirect pathway of the basal ganglia network play a critical role in manifesting mania-like behavior of BD. Moreover, our data also suggest lithium may exert its effect on BD through a GSK3ß-independent mechanism, in addition to the GSK3ß inhibition-mediated mechanism.


Assuntos
Transtorno Bipolar/patologia , Corpo Estriado/patologia , Dendritos/ultraestrutura , Glicogênio Sintase Quinase 3 beta/deficiência , Locomoção/genética , Anfetamina/farmacologia , Animais , Transtorno Bipolar/tratamento farmacológico , Células Cultivadas , Corpo Estriado/metabolismo , Depressão , Modelos Animais de Doenças , Glicogênio Sintase Quinase 3 beta/fisiologia , Humanos , Lítio/farmacologia , Locomoção/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neostriado/patologia , Neurônios/citologia
8.
Biochem Biophys Res Commun ; 503(3): 2068-2074, 2018 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-30119888

RESUMO

Medial vascular calcification is a highly regulated process involving osteo-/chondrogenic transdifferentiation of vascular smooth muscle cells. Both, protein kinase B (PKB) and serum- and glucocorticoid-inducible kinase 1 (SGK1) are involved in the intracellular signaling of vascular calcification and both phosphorylate and inactivate glycogen synthase kinase 3 (GSK-3). The present study explored whether PKB/SGK-dependent phosphorylation of GSK-3α/ß is involved in vascular calcification. Experiments were performed in Gsk-3α/ß double knockin mice lacking functional PKB/SGK phosphorylation sites (gsk-3KI) and corresponding wild-type mice (gsk-3WT) following high-dosed cholecalciferol treatment as well as ex vivo in aortic ring explants from gsk-3KI and gsk-3WT mice treated without and with phosphate. In gsk-3WT mice, high-dosed cholecalciferol induced vascular calcification and aortic osteo-/chondrogenic signaling, shown by increased expression of osteogenic markers Msx2, Cbfa1 and tissue-nonspecific alkaline phosphatase (Alpl). All these effects were suppressed in aortic tissue from gsk-3KI mice. Cholecalciferol decreased aortic Gsk-3α/ß phosphorylation (Ser21/9) in gsk-3WT mice, while no phosphorylation was observed in gsk-3KI mice. Moreover, the mRNA expression of type III sodium-dependent phosphate transporter (Pit1) and plasminogen activator inhibitor 1 (Pai1) was increased following cholecalciferol treatment in aortic tissue of gsk-3WT mice, effects again blunted in gsk-3KI mice. In addition, phosphate treatment induced mineral deposition and osteogenic markers expression in aortic ring explants from gsk-3WT mice, effects reduced in aortic ring explants from gsk-3KI mice. In conclusion, vascular PKB/SGK-dependent phosphorylation of GSK-3α/ß contributes to the osteoinductive signaling leading to vascular calcification.


Assuntos
Colecalciferol/metabolismo , Glicogênio Sintase Quinase 3 beta/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Proteínas Imediatamente Precoces/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Calcificação Vascular/metabolismo , Animais , Colecalciferol/administração & dosagem , Quinase 3 da Glicogênio Sintase/deficiência , Glicogênio Sintase Quinase 3 beta/deficiência , Injeções Subcutâneas , Camundongos , Camundongos Knockout , Fosforilação
9.
Glia ; 66(9): 1999-2012, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29761559

RESUMO

Apoptosis is recognized as the main mechanism of oligodendrocyte loss in Multiple Sclerosis caused either by immune mediated injury (Barnett & Prineas, ) or a direct degenerative process (oligodendrogliapathy; Lucchinetti et al., ). Cuprizone induced demyelination is the result of non-immune mediated apoptosis of oligodendrocytes (OL) and represents a model of oligodendrogliapathy (Simmons, Pierson, Lee, & Goverman, ). Glycogen Synthase Kinase (GSK) 3b has been shown to be pro-apoptotic for cells other than OL. Here, we sought to investigate whether GSK3b plays a role in cuprizone-induced apoptosis of OL by using a novel inducible conditional knockout (cKO) of GSK3b in mature OL. While depletion of GSK3b has no effect on survival of uninjured OL, it increases survival of mature OL exposed to cuprizone. We show that GSK3b-deficient OLs are protected against caspase-dependent, but not against caspase-independent apoptosis. Active GSK3b is present in the nuclei of OL at peak of caspase-dependent apoptosis. Significant preservation of myelinated axons is associated with GSK3b depletion and glial cell activation is markedly reduced. Collectively, the data show that GSK3b is pro-apoptotic for caspase-dependent cell death, likely through activation of nuclear GSK3b and its depletion promotes survival of oligodendrocytes and attenuates myelin loss.


Assuntos
Apoptose/fisiologia , Doenças Desmielinizantes/enzimologia , Glicogênio Sintase Quinase 3 beta/deficiência , Bainha de Mielina/enzimologia , Oligodendroglia/enzimologia , Animais , Astrócitos/enzimologia , Astrócitos/patologia , Caspases/metabolismo , Núcleo Celular/enzimologia , Núcleo Celular/patologia , Proliferação de Células/fisiologia , Sobrevivência Celular/fisiologia , Cuprizona , Doenças Desmielinizantes/patologia , Modelos Animais de Doenças , Feminino , Glicogênio Sintase Quinase 3 beta/genética , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microglia/enzimologia , Microglia/patologia , Bainha de Mielina/patologia , Oligodendroglia/patologia
10.
Nat Commun ; 9(1): 1126, 2018 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-29555900

RESUMO

Neural crest migration is critical to its physiological function. Mechanisms controlling mammalian neural crest migration are comparatively unknown, due to difficulties accessing this cell population in vivo. Here we report requirements of glycogen synthase kinase 3 (GSK3) in regulating the neural crest in Xenopus and mouse models. We demonstrate that GSK3 is tyrosine phosphorylated (pY) in mouse neural crest cells and that loss of GSK3 leads to increased pFAK and misregulation of Rac1 and lamellipodin, key regulators of cell migration. Genetic reduction of GSK3 results in failure of migration. We find that pY-GSK3 phosphorylation depends on anaplastic lymphoma kinase (ALK), a protein associated with neuroblastoma. Consistent with this, neuroblastoma cells with increased ALK activity express high levels of pY-GSK3, and blockade of GSK3 or ALK can affect migration of these cells. Altogether, this work identifies a role for GSK3 in cell migration during neural crest development and cancer.


Assuntos
Quinase 3 da Glicogênio Sintase/metabolismo , Crista Neural/citologia , Crista Neural/enzimologia , Proteínas de Xenopus/química , Quinase do Linfoma Anaplásico/antagonistas & inibidores , Quinase do Linfoma Anaplásico/metabolismo , Animais , Linhagem Celular Tumoral , Linhagem da Célula , Movimento Celular/fisiologia , Feminino , Quinase 3 da Glicogênio Sintase/química , Quinase 3 da Glicogênio Sintase/deficiência , Quinase 3 da Glicogênio Sintase/genética , Glicogênio Sintase Quinase 3 beta/deficiência , Glicogênio Sintase Quinase 3 beta/genética , Glicogênio Sintase Quinase 3 beta/metabolismo , Humanos , Camundongos , Camundongos Knockout , Crista Neural/embriologia , Neuroblastoma/enzimologia , Fosforilação , Gravidez , Proteínas de Xenopus/metabolismo , Xenopus laevis/embriologia , Xenopus laevis/metabolismo
11.
Int J Cardiol ; 259: 145-152, 2018 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-29398139

RESUMO

BACKGROUND AND RATIONALE: Obesity, an independent risk factor for the development of myocardial diseases is a growing healthcare problem worldwide. It's well established that GSK-3ß is critical to cardiac pathophysiology. However, the role cardiomyocyte (CM) GSK-3ß in diet-induced cardiac dysfunction is unknown. METHODS: CM-specific GSK-3ß knockout (CM-GSK-3ß-KO) and littermate controls (WT) mice were fed either a control diet (CD) or high-fat diet (HFD) for 55weeks. Cardiac function was assessed by transthoracic echocardiography. RESULTS: At baseline, body weights and cardiac function were comparable between the WT and CM-GSK-3ß-KOs. However, HFD-fed CM-GSK-3ß-KO mice developed severe cardiac dysfunction. Consistently, both heart weight/tibia length and lung weight/tibia length were significantly elevated in the HFD-fed CM-GSK-3ß-KO mice. The impaired cardiac function and adverse ventricular remodeling in the CM-GSK-3ß-KOs were independent of body weight or the lean/fat mass composition as HFD-fed CM-GSK-3ß-KO and controls demonstrated comparable body weight and body masses. At the molecular level, on a CD, CM-GSK-3α compensated for the loss of CM-GSK-3ß, as evident by significantly reduced GSK-3αs21 phosphorylation (activation) resulting in a preserved canonical ß-catenin ubiquitination pathway and cardiac function. However, this protective compensatory mechanism is lost with HFD, leading to excessive accumulation of ß-catenin in HFD-fed CM-GSK-3ß-KO hearts, resulting in adverse ventricular remodeling and cardiac dysfunction. CONCLUSION: In summary, these results suggest that cardiac GSK-3ß is crucial to protect against obesity-induced adverse ventricular remodeling and cardiac dysfunction.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Deleção de Genes , Glicogênio Sintase Quinase 3 beta/deficiência , Miócitos Cardíacos/enzimologia , Obesidade/enzimologia , Animais , Glicogênio Sintase Quinase 3 beta/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Miócitos Cardíacos/patologia , Obesidade/genética , Obesidade/patologia
12.
Neuropsychopharmacology ; 43(2): 393-405, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28832021

RESUMO

GSK3ß plays an essential role in promoting cell death and is emerging as a potential target for neurological diseases. Understanding the mechanisms that control neuronal GSK3ß is critical. A ubiquitous mechanism to repress GSK3ß involves Akt-mediated phosphorylation of Ser9. Here we show that phosphorylation of GSK3ß on Ser389 mediated by p38 MAPK specifically inactivates nuclear GSK3ß in the cortex and hippocampus. Using GSK3ß Ser389 to Ala mutant mice, we show that failure to inactivate nuclear GSK3ß by Ser389 phosphorylation causes neuronal cell death in subregions of the hippocampus and cortex. Although this focal neuronal death does not impact anxiety/depression-like behavior or hippocampal-dependent spatial learning, it leads to an amplified and prolonged fear response. This phenotype is consistent with some aspects of post-traumatic stress disorder (PTSD). Our studies indicate that inactivation of nuclear GSK3ß by Ser389 phosphorylation plays a key role in fear response, revealing new potential therapeutic approaches to target PTSD.


Assuntos
Comportamento Animal/fisiologia , Morte Celular/fisiologia , Córtex Cerebral/metabolismo , Medo/fisiologia , Glicogênio Sintase Quinase 3 beta/metabolismo , Neurônios/metabolismo , Fosfosserina/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Córtex Cerebral/fisiopatologia , Feminino , Glicogênio Sintase Quinase 3 beta/deficiência , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Masculino , Camundongos , Fosforilação/fisiologia
13.
J Pathol ; 243(1): 65-77, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28639695

RESUMO

Acinar-to-ductal metaplasia (ADM) is a reversible epithelial transdifferentiation process that occurs in the pancreas in response to acute inflammation. ADM can rapidly progress towards pre-malignant pancreatic intraepithelial neoplasia (PanIN) lesions in the presence of mutant KRas and ultimately pancreatic adenocarcinoma (PDAC). In the present work, we elucidate the role and related mechanism of glycogen synthase kinase-3beta (GSK-3ß) in ADM development using in vitro 3D cultures and genetically engineered mouse models. We show that GSK-3ß promotes TGF-α-induced ADM in 3D cultured primary acinar cells, whereas deletion of GSK-3ß attenuates caerulein-induced ADM formation and PanIN progression in KrasG12D transgenic mice. Furthermore, we demonstrate that GSK-3ß ablation influences ADM formation and PanIN progression by suppressing oncogenic KRas-driven cell proliferation. Mechanistically, we show that GSK-3ß regulates proliferation by increasing the activation of S6 kinase. Taken together, these results indicate that GSK-3ß participates in early pancreatitis-induced ADM and thus could be a target for the treatment of chronic pancreatitis and the prevention of PDAC progression. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Células Acinares/enzimologia , Carcinoma in Situ/prevenção & controle , Transdiferenciação Celular , Glicogênio Sintase Quinase 3 beta/deficiência , Pâncreas Exócrino/enzimologia , Ductos Pancreáticos/enzimologia , Neoplasias Pancreáticas/prevenção & controle , Pancreatite/enzimologia , Células Acinares/efeitos dos fármacos , Células Acinares/patologia , Animais , Carcinoma in Situ/enzimologia , Carcinoma in Situ/genética , Carcinoma in Situ/patologia , Proliferação de Células , Transdiferenciação Celular/efeitos dos fármacos , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Células Cultivadas , Ceruletídeo , Modelos Animais de Doenças , Progressão da Doença , Predisposição Genética para Doença , Glicogênio Sintase Quinase 3 beta/genética , Proteínas de Homeodomínio/genética , Masculino , Metaplasia , Camundongos Knockout , Pâncreas Exócrino/efeitos dos fármacos , Pâncreas Exócrino/patologia , Ductos Pancreáticos/efeitos dos fármacos , Ductos Pancreáticos/patologia , Neoplasias Pancreáticas/enzimologia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Pancreatite/induzido quimicamente , Pancreatite/genética , Pancreatite/patologia , Fenótipo , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Quinases S6 Ribossômicas/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Fatores de Tempo , Transativadores/genética , Fator de Necrose Tumoral alfa/farmacologia
14.
Cell Rep ; 17(1): 165-178, 2016 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-27681429

RESUMO

In cold-blooded vertebrates such as zebrafish, Müller glial cells (MGs) readily proliferate to replenish lost retinal neurons. In mammals, however, MGs lack regenerative capability as they do not spontaneously re-enter the cell cycle unless the retina is injured. Here, we show that gene transfer of ß-catenin in adult mouse retinas activates Wnt signaling and MG proliferation without retinal injury. Upstream of Wnt, deletion of GSK3ß stabilizes ß-catenin and activates MG proliferation. Downstream of Wnt, ß-catenin binds to the Lin28 promoter and activates transcription. Deletion of Lin28 abolishes ß-catenin-mediated effects on MG proliferation, and Lin28 gene transfer stimulates MG proliferation. We further demonstrate that let-7 miRNAs are critically involved in Wnt/Lin28-regulated MG proliferation. Intriguingly, a subset of cell-cycle-reactivated MGs express markers for amacrine cells. Together, these results reveal a key role of Wnt-Lin28-let7 miRNA signaling in regulating proliferation and neurogenic potential of MGs in the adult mammalian retina.


Assuntos
Células Ependimogliais/metabolismo , Regulação da Expressão Gênica , MicroRNAs/genética , Proteínas de Ligação a RNA/genética , Proteínas Wnt/genética , Células Amácrinas/citologia , Células Amácrinas/metabolismo , Animais , Ciclo Celular/genética , Diferenciação Celular , Proliferação de Células , Células Ependimogliais/citologia , Glicogênio Sintase Quinase 3 beta/deficiência , Glicogênio Sintase Quinase 3 beta/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , MicroRNAs/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transporte Proteico , Proteínas de Ligação a RNA/metabolismo , Transdução de Sinais , Transcrição Gênica , Proteínas Wnt/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...