Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Hepatology ; 77(1): 213-229, 2023 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-35363898

RESUMO

BACKGROUND AND AIMS: Metabolism in the liver is dysregulated in obesity, contributing to various health problems including steatosis and insulin resistance. While the pathogenesis of lipid accumulation has been extensively studied, the protective mechanism against lipid challenge in the liver remains unclear. Here, we report that Src homology 3 domain binding kinase 1 (SBK1) is a regulator of hepatic lipid metabolism and systemic insulin sensitivity in response to obesity. APPROACH AND RESULTS: Enhanced Sbk1 expression was found in the liver of high-fat diet (HFD)-induced obese mice and fatty acid (FA)-challenged hepatocytes. SBK1 knockdown in mouse liver cells augmented FA uptake and lipid accumulation. Similarly, liver-specific SBK1 knockout ( Lsko ) mice displayed more severe hepatosteatosis and higher expression of genes in FA uptake and lipogenesis than the Flox/Flox ( Fl/Fl ) control mice when fed the HFD. The HFD-fed Lsko mice also showed symptoms of hyperglycemia, poor systemic glucose tolerance, and lower insulin sensitivity than the Fl/Fl mice. On the other hand, hepatic Sbk1 overexpression alleviated the high-fructose diet-induced hepatosteatosis, hyperlipidemia, and hyperglycemia in mice. White adipose tissue browning was also observed in hepatic SBK1 -overexpressed mice. Moreover, we found that SBK1 was a positive regulator of FGF21 in the liver during energy surplus conditions. Mechanistically, SBK1 phosphorylates the orphan nuclear receptor 4A1 (Nur77) on serine 344 to promote hepatic FGF21 expression and inhibit the transcription of genes involved in lipid anabolism. CONCLUSIONS: Collectively, our data suggest that SBK1 is a regulator of the metabolic adaption against obesity through the Nur77-FGF21 pathway.


Assuntos
Fígado Gorduroso , Resistência à Insulina , Proteínas Quinases , Animais , Camundongos , Dieta Hiperlipídica/efeitos adversos , Fígado Gorduroso/metabolismo , Fígado Gorduroso/patologia , Hiperglicemia/complicações , Hiperglicemia/metabolismo , Hiperglicemia/patologia , Lipídeos , Fígado/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/complicações , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares
2.
J Allergy Clin Immunol ; 151(4): 1110-1122, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36581009

RESUMO

BACKGROUND: Activation of Mas-related G protein-coupled receptor X2 (MRGPRX2) is a crucial non-IgE pathway for mast cell activation associated with allergic reactions and inflammation. Only a few peptides and small compounds targeting MRGPRX2 have been reported, with limited information on their pharmacologic activity. OBJECTIVE: We sought to develop novel small molecule MRGPRX2 antagonists to treat MRGPRX2-mediated allergies and inflammation. METHODS: A computational approach was used to design novel small molecules as MRGPRX2 antagonists. The short-listed molecules were synthesized and characterized by liquid chromatography and mass spectrometry as well as nuclear magnetic resonance. Inhibitory activity on MRGPRX2 signaling was assessed in vitro by using functional bioassays (ß-hexosaminidase, calcium flux, and chemokine synthesis) and receptor activation assays (ß-arrestin recruitment and Western blot analysis) in human LAD-2 mast cells and HTLA cells. In vivo effects of the novel MRGPRX2 antagonists were assessed using a mouse model of acute allergy and systemic anaphylaxis. RESULTS: The novel small molecules demonstrated higher binding affinity with MRGPRX2 in the docking study. The half-maximal inhibitory concentration is in the low micromolar range (5-21 µM). The small molecules inhibited not only the early phase of mast cell activation but also the late phase, associated with chemokine and prostaglandin release. Further, Western blot analysis revealed inhibition of downstream phospholipase C-γ, extracellular signal-regulated protein kinase 1/2, and Akt signaling pathway. Moreover, in the mouse models of allergies, small molecule administration effectively blocks acute, systemic allergic reactions and inflammation and prevents systemic anaphylaxis. CONCLUSION: The small molecules might hold a significant therapeutic promise to treat MRGPRX2-mediated allergies and inflammation.


Assuntos
Anafilaxia , Animais , Camundongos , Humanos , Anafilaxia/patologia , Modelos Animais de Doenças , Receptores Acoplados a Proteínas G/metabolismo , Quimiocinas/metabolismo , Mastócitos/patologia , Inflamação/patologia , Receptores de Neuropeptídeos/metabolismo , Degranulação Celular , Proteínas do Tecido Nervoso/metabolismo
3.
Mol Psychiatry ; 27(8): 3396-3409, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35546632

RESUMO

Diabetes is a risk factor for Alzheimer's disease (AD), which is also called type 3 diabetes with insulin reduction and insulin resistance in AD patient brains. However, the molecular mechanism coupling diabetes to AD onset remains incompletely understood. Here we show that inflammation, associated with obesity and diabetes elicited by high-fat diet (HFD), activates neuronal C/EBPß/AEP signaling that drives AD pathologies and cognitive disorders. HFD stimulates diabetes and insulin resistance in neuronal Thy1-C/EBPß transgenic (Tg) mice, accompanied with prominent mouse Aß accumulation and hyperphosphorylated Tau aggregation in the brain, triggering cognitive deficits. These effects are profoundly diminished when AEP is deleted from C/EBPß Tg mice. Chronic treatment with inflammatory lipopolysaccharide (LPS) facilitates AD pathologies and cognitive disorders in C/EBPß Tg but not in wild-type mice, and these deleterious effects were substantially alleviated in C/EBPß Tg/AEP -/- mice. Remarkably, the anti-inflammatory drug aspirin strongly attenuates HFD-induced diabetes and AD pathologies in neuronal C/EBPß Tg mice. Therefore, our findings demonstrate that inflammation-activated neuronal C/EBPß/AEP signaling couples diabetes to AD.


Assuntos
Doença de Alzheimer , Diabetes Mellitus , Resistência à Insulina , Animais , Camundongos , Doença de Alzheimer/metabolismo , Dieta Hiperlipídica/efeitos adversos , Resistência à Insulina/fisiologia , Camundongos Transgênicos , Inflamação/metabolismo , Encéfalo/metabolismo , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Diabetes Mellitus/metabolismo , Modelos Animais de Doenças
4.
Gen Comp Endocrinol ; 344: 114372, 2023 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-37652166

RESUMO

SH3 domain binding kinase 1 (SBK1) is a serine/threonine kinase that belongs to the new kinase family (NFK) with limited information on its function. Previous studies reported that SBK1 plays a role in memory formation, lipid metabolism, and cancer cell progression. Nevertheless, the regulatory mechanism of Sbk1 expression in various tissues remains unknown. We report here that Sbk1 expression in mouse hepatocytes was downregulated by glucocorticoid, whereas saturated and unsaturated fatty acids were stimulators of Sbk1 expression. The regulatory role of glucocorticoid and fatty acid was further confirmed by the Sbk1 promoter assay, which aligned with the presence of several glucocorticoid-response elements (GRE) and peroxisome proliferator responsive elements (PPRE) in the mouse Sbk1 promoter. The inhibitory effect of glucocorticoids on hepatic Sbk1 expression and protein content could also be demonstrated in vivo after prednisolone injection. Moreover, the expression of SBK1 in goldfish (gfSBK1) was also sensitive to glucocorticoid suppression as their mouse orthologues. In contrast, insulin had a differential action on SBK1 expression that it promoted the expression of all SBK1 isoforms in the goldfish hepatocytes but inhibited Sbk1 expression in the mouse hepatocytes. Together, our findings indicate that SBK1 expression is hormone- and nutrient-sensitive with a species-specific response.


Assuntos
Carpa Dourada , Fatores de Transcrição , Camundongos , Animais , Fatores de Transcrição/metabolismo , Carpa Dourada/genética , Carpa Dourada/metabolismo , Glucocorticoides/metabolismo , Domínios de Homologia de src , Fígado/metabolismo
5.
Genomics ; 114(4): 110440, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35905835

RESUMO

The moth Heortia vitessoides Moore (Lepidoptera: Crambidae) is a major pest of ecologically, commercially and culturally important agarwood-producing trees in the genus Aquilaria. In particular, H. vitessoides is one of the most destructive defoliating pests of the incense tree Aquilaria sinesis, which produces a valuable fragrant wood used as incense and in traditional Chinese medicine [33]. Nevertheless, a genomic resource for H. vitessoides is lacking. Here, we present a chromosomal-level assembly for H. vitessoides, consisting of a 517 megabase (Mb) genome assembly with high physical contiguity (scaffold N50 of 18.2 Mb) and high completeness (97.9% complete BUSCO score). To aid gene annotation, 8 messenger RNA transcriptomes from different developmental stages were generated, and a total of 16,421 gene models were predicted. Expansion of gene families involved in xenobiotic metabolism and development were detected, including duplications of cytosolic sulfotransferase (SULT) genes shared among lepidopterans. In addition, small RNA sequencing of 5 developmental stages of H. vitessoides facilitated the identification of 85 lepidopteran conserved microRNAs, 94 lineage-specific microRNAs, as well as several microRNA clusters. A large proportion of the H. vitessoides genome consists of repeats, with a 29.12% total genomic contribution from transposable elements, of which long interspersed nuclear elements (LINEs) are the dominant component (17.41%). A sharp decrease in the genome-wide percentage of LINEs with lower levels of genetic distance to family consensus sequences suggests that LINE activity has peaked in H. vitessoides. In contrast, opposing patterns suggest a substantial recent increase in DNA and LTR element activity. Together with annotations of essential sesquiterpenoid hormonal pathways, neuropeptides, microRNAs and transposable elements, the high-quality genomic and transcriptomic resources we provide for the economically important moth H. vitessoides provide a platform for the development of genomic approaches to pest management, and contribute to addressing fundamental research questions in Lepidoptera.


Assuntos
Lepidópteros , MicroRNAs , Mariposas , Animais , Elementos de DNA Transponíveis , Lepidópteros/genética , Mariposas/genética , Árvores/genética
6.
Biochem Biophys Res Commun ; 514(2): 407-414, 2019 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-31056256

RESUMO

Skeletal muscle secretes myokines, which are involved in metabolism and muscle function regulation. The role of fasting on myokine expression in skeletal muscle is largely unknown. In this study, we used gastrocnemius skeletal muscle RNA sequencing data from fasting male mice in the Gene Expression Omnibus (GEO) database. Adopted male and female C57BL/6J mice that fasted for 24 h were included to examine the effect of fasting on myokine expression in slow-twitch soleus and fast-twitch tiabialis anterior (TA) skeletal muscle. We found that fasting significantly affected many myokines in muscle. Fasting reduced Fndc5 and Igf1 gene expression in soleus and TA muscles in both male and female mice without muscle phenotype or gender differences, but Il6, Mstn and Erfe expression was influenced by fasting with fibre type- and gender-dependent effects. Fasting also induced muscle atrophy marker genes Murf1 and Fbxo32 and reduced myogenesis factor Mef2 expression without muscle fibre or gender differences. We further found that the expression of transcription factors Pgc1α, Pparα, Pparγ and Pparδ had muscle fibre type-dependent effects, and the expression of Pgc1α and Pparα had gender-dependent effects. The sophisticated expression pattern of myokines would partially explain the complicated cross-talk between skeletal muscle and other organs in different genders and muscles phenotypes, and it is worth further investigation.


Assuntos
Citocinas/genética , Jejum/fisiologia , Regulação da Expressão Gênica , Músculo Esquelético/metabolismo , Caracteres Sexuais , Animais , Citocinas/biossíntese , Feminino , Fibronectinas/genética , Fator de Crescimento Insulin-Like I/genética , Interleucina-6/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fibras Musculares Esqueléticas/classificação , Fibras Musculares Esqueléticas/metabolismo , Proteínas Musculares/genética , Miostatina/genética , Fenótipo , Fatores de Transcrição/genética
7.
Cell Physiol Biochem ; 50(4): 1574-1584, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30359971

RESUMO

BACKGROUND/AIMS: Fibronectin type III domain-containing protein 5 (FNDC5), also known as irisin, is a myokine secreted from muscle in response to exercise. However, the molecular mechanisms that regulate FNDC5 expression and the functional significance of irisn in skeletal muscle remain unknown. In this study, we explored the potential pathways that induce FNDC5 expression and delineated the metabolic effects of irisin on skeletal muscle. METHODS: C2C12 myotubes were treated with drugs at various concentrations and durations. The expression and activation of genes were measured by real-time polymerase chain reaction (qRT-PCR) and Western blotting. Oxidative phosphorylation was quantified by measuring the oxygen consumption rate (OCR). RESULTS: We found that the exercise-mimicking treatment (cAMP, forskolin and isoproterenol) increased Fndc5 expression in C2C12 myotubes. CREB over-expressed C2C12 myotubes displayed higher Fndc5 expression. CREB over-expression also promoted peroxisome proliferator-activated receptor gamma coactivator 1α (PGC-1α) expression. PGC-1α-induced Fndc5 expression was blocked when the dominant negative form of CREB (S133A) was present. PGC-1α mutation (S570A) also decreased Fndc5 expression. Immunoprecipitation showed that overexpressed PGC-1α complexed with CREB in HEK293 cells. C2C12 myotubes treated with forskolin also increased endogenous CREB and PGC-1α binding. Functionally, irisin treatment increased mitochondrial respiration, enhanced ATP production, promoted fatty acid oxidation but decreased glycolysis in myotubes. CONCLUSION: Our observation indicates that cAMP-mediated PGC-1α/CREB interaction triggers Fndc5 expression, which acts as an autocrine/paracrine to shape the metabolic phenotype of myotubes.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Fibronectinas/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Linhagem Celular , Colforsina/farmacologia , AMP Cíclico/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Fibronectinas/genética , Fibronectinas/farmacologia , Células HEK293 , Humanos , Isoproterenol/farmacologia , Camundongos , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/metabolismo , Fosforilação Oxidativa/efeitos dos fármacos , Consumo de Oxigênio/efeitos dos fármacos , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/farmacologia
8.
J Neurosci Res ; 95(1-2): 328-335, 2017 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-27870419

RESUMO

Brain-derived neurotrophic factor (BDNF) is a member of the neurotrophin family that plays a critical role in numerous neuronal activities. Recent studies have indicated that some functions or action mechanisms of BDNF vary in a sex-dependent manner. In particular, BDNF content in some brain parts and the tendency to develop BDNF deficiency-related diseases such as depression are greater in female animals. With the support of relevant studies, it has been suggested that sex hormones or steroids can modulate the activities of BDNF, which may account for its functional discrepancy in different sexes. Indeed, the cross-talk between BDNF and sex steroids has been detected for decades, and some sex steroids, such as estrogen, have a positive regulatory effect on BDNF expression and signaling. Thus, the sex of animal models that are used in studying the functions of BDNF is critical. This Mini-Review summarizes our current findings on the differences in expression, signaling, and functions of BDNF between sexes. We also discuss the potential mechanisms for mediating these differential responses, with a specific emphasis on sex steroids. By presenting and discussing these findings, we seek to encourage researchers to take sex influences into consideration when designing experiments, interpreting results, and drawing conclusions. © 2016 Wiley Periodicals, Inc.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Encéfalo/metabolismo , Caracteres Sexuais , Transdução de Sinais/fisiologia , Animais , Humanos
9.
Proc Natl Acad Sci U S A ; 111(5): 1993-8, 2014 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-24449917

RESUMO

Oligodendrocyte (OL) differentiation and myelin development are complex events regulated by numerous signal transduction factors. Here, we report that phosphoinositide-3 kinase enhancer L (PIKE-L) is required for OL development and myelination. PIKE-L expression is up-regulated when oligodendrocyte progenitor cells commit to differentiation. Conversely, depleting phosphoinositide-3 kinase enhancer (PIKE) expression by shRNA prevents oligodendrocyte progenitor cell differentiation. In both conventional PIKE knockout (PIKE(-/-)) and OL-specific PIKE knockout mice, the number of OLs is reduced in the corpus callosum. PIKE(-/-) OLs also display defects when forming myelin sheath on neuronal axons during neonatal development, which is partially rescued when PTEN is ablated. In addition, Akt/mTOR signaling is impaired in OL-enriched tissues of the PIKE(-/-) mutant, leading to reduced expression of critical proteins for myelin development and hypomyelination. Moreover, myelin repair of lysolecithin-induced lesions is delayed in PIKE(-/-) brain. Thus, PIKE plays pivotal roles to advance OL development and myelinogenesis through Akt/mTOR activation.


Assuntos
Sistema Nervoso Central/metabolismo , GTP Fosfo-Hidrolases/metabolismo , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Oligodendroglia/metabolismo , Animais , Axônios/metabolismo , Encéfalo/crescimento & desenvolvimento , Encéfalo/patologia , Encéfalo/ultraestrutura , Contagem de Células , Células Cultivadas , Sistema Nervoso Central/crescimento & desenvolvimento , Camundongos , Camundongos Knockout , Mutação/genética , Bainha de Mielina/metabolismo , Bainha de Mielina/ultraestrutura , Oligodendroglia/patologia , Oligodendroglia/ultraestrutura , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Regulação para Cima
10.
J Biol Chem ; 289(40): 27571-84, 2014 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-25143381

RESUMO

7,8-dihydroxyflavone (7,8-DHF), a newly identified small molecular TrkB receptor agonist, rapidly activates TrkB in both primary neurons and the rodent brain and mimics the physiological functions of the cognate ligand BDNF. Accumulating evidence supports that 7,8-DHF exerts neurotrophic effects in a TrkB-dependent manner. Nonetheless, the differences between 7,8-DHF and BDNF in activating TrkB remain incompletely understood. Here we show that 7,8-DHF and BDNF exhibit different TrkB activation kinetics in which TrkB maturation may be implicated. Employing two independent biophysical approaches, we confirm that 7,8-DHF interacts robustly with the TrkB extracellular domain, with a Kd of ∼10 nm. Although BDNF transiently activates TrkB, leading to receptor internalization and ubiquitination/degradation, in contrast, 7,8-DHF-triggered TrkB phosphorylation lasts for hours, and the internalized receptors are not degraded. Notably, primary neuronal maturation may be required for 7,8-DHF but not for BDNF to elicit the full spectrum of TrkB signaling cascades. Hence, 7,8-DHF interacts robustly with the TrkB receptor, and its agonistic effect may be mediated by neuronal development and maturation.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/química , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Flavonas/metabolismo , Receptor trkB/metabolismo , Animais , Fenômenos Biofísicos , Fator Neurotrófico Derivado do Encéfalo/genética , Células Cultivadas , Flavonas/química , Humanos , Cinética , Neurônios/química , Neurônios/metabolismo , Ligação Proteica , Ratos , Receptor trkB/agonistas , Receptor trkB/química , Receptor trkB/genética
11.
EMBO J ; 30(20): 4274-86, 2011 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-21847098

RESUMO

AMPAR (α-amino-3-hydroxy-5-methyl-isoxazole-4-propionic acid receptor) is an ion channel involved in the formation of synaptic plasticity. However, the molecular mechanism that couples plasticity stimuli to the trafficking of postsynaptic AMPAR remains poorly understood. Here, we show that PIKE (phosphoinositide 3-kinase enhancer) GTPases regulate neuronal AMPAR activity by promoting GluA2/GRIP1 association. PIKE-L directly interacts with both GluA2 and GRIP1 and forms a tertiary complex upon glycine-induced NMDA receptor activation. PIKE-L is also essential for glycine-induced GluA2-associated PI3K activation. Genetic ablation of PIKE (PIKE(-/-)) in neurons suppresses GluA2-associated PI3K activation, therefore inhibiting the subsequent surface expression of GluA2 and the formation of long-term potentiation. Our findings suggest that PIKE-L is a critical factor in controlling synaptic AMPAR insertion.


Assuntos
GTP Fosfo-Hidrolases/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Receptores de AMPA/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Encéfalo/metabolismo , Proteínas de Transporte/metabolismo , Células Cultivadas , GTP Fosfo-Hidrolases/genética , Proteínas de Ligação ao GTP/genética , Proteínas Ativadoras de GTPase/genética , Glicina/metabolismo , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Potenciação de Longa Duração , Camundongos , Proteínas Monoméricas de Ligação ao GTP/genética , Proteínas do Tecido Nervoso/genética , Neurônios/metabolismo , Ratos , Receptores de N-Metil-D-Aspartato/metabolismo
12.
EMBO J ; 29(5): 956-68, 2010 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-20075866

RESUMO

PI 3-kinase enhancer A (PIKE-A) is critical for the activation of Akt signalling, and has an essential function in promoting cancer cell survival. However, its physiological functions are poorly understood. Here, we show that PIKE-A directly associates with both signal transducer and activator of transcription 5a (STAT5a) and prolactin (PRL) receptor, which is essential for PRL-provoked STAT5a activation and the subsequent gene transcription. Depletion of PIKE-A in HC11 epithelial cells diminished PRL-induced STAT5 activation and cyclin D1 expression, resulting in profoundly impaired cell proliferation in vitro. To confirm the function of PIKE-A in PRL signalling in vivo, we generated PIKE knockout (PIKE-/-) mice. PIKE-/- mice displayed a severe lactation defect that was characterized by enhanced apoptosis and impaired proliferation of mammary epithelial cells. At parturition, STAT5 activation and cyclin D1 expression were substantially reduced in the mammary epithelium of PIKE-/- mice. The defective mammary gland development in PIKE-/- mice was rescued by overexpression of a mammary-specific cyclin D1 transgene. These data establish a critical function for PIKE-A in mediating PRL functions.


Assuntos
GTP Fosfo-Hidrolases/metabolismo , Glândulas Mamárias Animais/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Prolactina/farmacologia , Fator de Transcrição STAT5/metabolismo , Animais , Apoptose/genética , Apoptose/fisiologia , Western Blotting , Linhagem Celular , Proliferação de Células , Ciclina D1/metabolismo , Células Epiteliais/citologia , Células Epiteliais/fisiologia , Feminino , GTP Fosfo-Hidrolases/genética , Expressão Gênica , Genótipo , Imunoprecipitação , Marcação In Situ das Extremidades Cortadas , Lactação/genética , Lactação/fisiologia , Glândulas Mamárias Animais/crescimento & desenvolvimento , Glândulas Mamárias Animais/transplante , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Gravidez , Ligação Proteica/efeitos dos fármacos , Receptores da Prolactina/metabolismo
13.
Front Cell Dev Biol ; 12: 1403463, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38808223

RESUMO

Sporadic inclusion body myositis (sIBM) is a distinct subcategory of Idiopathic Inflammatory Myopathies (IIM), characterized by unique pathological features such as muscle inflammation, rimmed vacuoles, and protein aggregation within the myofibers. Although hyperactivation of the immune system is widely believed as the primary cause of IIM, it is debated whether non-immune tissue dysfunction might contribute to the disease's onset as patients with sIBM are refractory to conventional immunosuppressant treatment. Moreover, the findings that mitochondrial dysfunction can elicit non-apoptotic programmed cell death and the subsequent immune response further support this hypothesis. Notably, abnormal mitochondrial structure and activities are more prominent in the muscle of sIBM than in other types of IIM, suggesting the presence of defective mitochondria might represent an overlooked contributor to the disease onset. The large-scale mitochondrial DNA deletion, aberrant protein aggregation, and slowed organelle turnover have provided mechanistic insights into the genesis of impaired mitochondria in sIBM. This article reviews the disease hallmarks of sIBM, the plausible contributors of mitochondrial damage in the sIBM muscle, and the immunological responses associated with mitochondrial perturbations. Additionally, the potential application of mitochondrial-targeted chemicals as a new treatment strategy to sIBM is explored and discussed.

14.
Sci Signal ; 17(828): eadh2783, 2024 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-38502732

RESUMO

Post-exercise recovery is essential to resolve metabolic perturbations and promote long-term cellular remodeling in response to exercise. Here, we report that muscle-generated brain-derived neurotrophic factor (BDNF) elicits post-exercise recovery and metabolic reprogramming in skeletal muscle. BDNF increased the post-exercise expression of the gene encoding PPARδ (peroxisome proliferator-activated receptor δ), a transcription factor that is a master regulator of lipid metabolism. After exercise, mice with muscle-specific Bdnf knockout (MBKO) exhibited impairments in PPARδ-regulated metabolic gene expression, decreased intramuscular lipid content, reduced ß-oxidation, and dysregulated mitochondrial dynamics. Moreover, MBKO mice required a longer period to recover from a bout of exercise and did not show increases in exercise-induced endurance capacity. Feeding naïve mice with the bioavailable BDNF mimetic 7,8-dihydroxyflavone resulted in effects that mimicked exercise-induced adaptations, including improved exercise capacity. Together, our findings reveal that BDNF is an essential myokine for exercise-induced metabolic recovery and remodeling in skeletal muscle.


Assuntos
PPAR delta , Animais , Camundongos , Fator Neurotrófico Derivado do Encéfalo/genética , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Regulação da Expressão Gênica , Metabolismo dos Lipídeos , Músculo Esquelético/metabolismo , PPAR delta/genética , PPAR delta/metabolismo
15.
J Neurosci ; 32(48): 17262-72, 2012 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-23197718

RESUMO

Serine-arginine protein kinases 2 (SRPK2) is a cell cycle-regulated kinase that phosphorylates serine/arginine domain-containing proteins and mediates pre-mRNA splicing with unclear function in neurons. Here, we show that SRPK2 phosphorylates tau on S214, suppresses tau-dependent microtubule polymerization, and inhibits axonal elongation in neurons. Depletion of SRPK2 in dentate gyrus inhibits tau phosphorylation in APP/PS1 mouse and alleviates the impaired cognitive behaviors. The defective LTP in APP/PS1 mice is also improved after SRPK2 depletion. Moreover, active SRPK2 is increased in the cortex of APP/PS1 mice and the pathological structures of human Alzheimer's disease (AD) brain. Therefore, our study suggests SRPK2 may contribute to the formation of hyperphosphorylated tau and the pathogenesis of AD.


Assuntos
Doença de Alzheimer/metabolismo , Comportamento Animal/fisiologia , Encéfalo/metabolismo , Aprendizagem em Labirinto/fisiologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas tau/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Doença de Alzheimer/psicologia , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Encéfalo/patologia , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Transgênicos , Neuritos/metabolismo , Plasticidade Neuronal/fisiologia , Neurônios/metabolismo , Neurônios/patologia , Fosforilação , Presenilina-1/genética , Presenilina-1/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas tau/genética
16.
J Biol Chem ; 287(9): 6113-27, 2012 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-22215664

RESUMO

Amplification of the epidermal growth factor receptor (EGFR), frequently expressed as a constitutively active deletion mutant (EGFRvIII), occurs commonly in glioblastoma multiformes (GBM). However, blockade of EGFR is therapeutically disappointing for gliomas with PTEN deletion. To search for small molecules treating this aggressive cancer, we have established a cell-based screening and successfully identified acridine yellow G that preferentially blocks cell proliferation of the most malignant U87MG/EGFRvIII cells over the less malignant U87MG/PTEN cells. Oral administration of this compound markedly diminishes the brain tumor volumes in both subcutaneous and intracranial models. It directly inhibits EGFR and PKCs with IC(50) values of ~7.5 and 5 µM, respectively. It dually inhibits EGFR and PKCs, resulting in a blockade of mammalian target of rapamycin signaling and cell cycle arrest in the G(1) phase, which leads to activation of apoptosis in the tumors. Hence, combinatorial inhibition of EGFR and PKCs might provide proof of concept in developing therapeutic agents for treating malignant glioma and other human cancers.


Assuntos
Aminoacridinas/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Receptores ErbB/antagonistas & inibidores , Glioblastoma/tratamento farmacológico , Proteína Quinase C/antagonistas & inibidores , Animais , Antineoplásicos/farmacologia , Neoplasias Encefálicas/enzimologia , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Receptores ErbB/metabolismo , Feminino , Fase G1/efeitos dos fármacos , Glioblastoma/enzimologia , Glioblastoma/patologia , Humanos , Camundongos , Camundongos Nus , Proteína Quinase C/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Rev Neurosci ; 24(4): 401-13, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23828492

RESUMO

Serine-arginine protein kinases (SRPKs) are a group of serine kinases that recognize and phosphorylate protein substrates with serine-arginine dipeptide repeats. They are mainly involved in regulating pre-mRNA splicing via phosphorylating splicing factors, such as ASF/SF2 and SC35. Nevertheless, the functions of SRPKs in the nervous system are sketchy, although the kinases have significant expression in neurons. Our recent studies demonstrate that one of the SRPK members, SRPK2, participates in the neuronal survival, cell cycle progression, and memory determination in Alzheimer's disease. SRPKs are thus a group of unrecognized proteins that may facilitate the pathological progression of disorders caused by neurodegeneration. In this review, we will update our knowledge on SRPKs' functions in various cellular activities and discuss their potential role in neurodegenerative disorders.


Assuntos
Doenças Neurodegenerativas/enzimologia , Doenças Neurodegenerativas/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Processamento Alternativo/genética , Animais , Encéfalo/patologia , Ciclo Celular/genética , Humanos , Mutação/genética , Doenças Neurodegenerativas/patologia , Neurônios/enzimologia
18.
EMBO Rep ; 12(8): 847-54, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21720388

RESUMO

Dysfunction of hepatic insulin receptor tyrosine kinase (IRTK) causes the development of type 2 diabetes. However, the molecular mechanism regulating IRTK activity in the liver remains poorly understood. Here, we show that phosphoinositide 3-kinase enhancer A (PIKE-A) is a new insulin-dependent enhancer of hepatic IRTK. Liver-specific Pike-knockout (LPKO) mice display glucose intolerance with impaired hepatic insulin sensitivity. Specifically, insulin-provoked phosphoinositide 3-kinase/Akt signalling is diminished in the liver of LPKO mice, leading to the failure of insulin-suppressed gluconeogenesis and hyperglycaemia. Thus, hepatic PIKE-A has a key role in mediating insulin signal transduction and regulating glucose homeostasis in the liver.


Assuntos
GTP Fosfo-Hidrolases/metabolismo , Fígado/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Receptor de Insulina/metabolismo , Animais , GTP Fosfo-Hidrolases/genética , Gluconeogênese/fisiologia , Glucose/metabolismo , Intolerância à Glucose/metabolismo , Homeostase/fisiologia , Hiperglicemia/metabolismo , Insulina/metabolismo , Resistência à Insulina/fisiologia , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/fisiologia
19.
Sci Adv ; 9(24): eadg0183, 2023 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-37327338

RESUMO

Junctional folds are unique membrane specializations developed progressively during the postnatal maturation of vertebrate neuromuscular junctions (NMJs), but how they are formed remains elusive. Previous studies suggested that topologically complex acetylcholine receptor (AChR) clusters in muscle cultures undergo a series of transformations, resembling the postnatal maturation of NMJs in vivo. We first demonstrated the presence of membrane infoldings at AChR clusters in cultured muscles. Live-cell super-resolution imaging further revealed that AChRs are gradually redistributed to the crest regions and spatially segregated from acetylcholinesterase along the elongating membrane infoldings over time. Mechanistically, lipid raft disruption or caveolin-3 knockdown not only inhibits membrane infolding formation at aneural AChR clusters and delays agrin-induced AChR clustering in vitro but also affects junctional fold development at NMJs in vivo. Collectively, this study demonstrated the progressive development of membrane infoldings via nerve-independent, caveolin-3-dependent mechanisms and identified their roles in AChR trafficking and redistribution during the structural maturation of NMJs.


Assuntos
Acetilcolinesterase , Caveolina 3 , Caveolina 3/genética , Junção Neuromuscular/fisiologia , Receptores Colinérgicos , Músculos
20.
J Neurosci ; 31(22): 8083-92, 2011 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-21632930

RESUMO

Phosphoinositide 3-kinase enhancer (PIKE) binds and enhances phosphatidylinositol 3-kinase (PI3K)/Akt activities. However, its physiological functions in brain have never been explored. Here we show that PIKE is important in regulating the neuronal survival and development of neocortex. During development, enhanced apoptosis is observed in the ventricular zone of PIKE knock-out (PIKE(-/-)) cortex. Moreover, PIKE(-/-) neurons show reduced dendritic complexity, dendritic branch length, and soma size. These defects are due to the reduced PI3K/Akt activities in PIKE(-/-) neurons, as the impaired dendritic arborization can be rescued when PI3K/Akt cascade is augmented in vitro or in PIKE(-/-)PTEN(-/-) double-knock-out mice. Interestingly, PIKE(-/-) mice display behavioral abnormality in locomotion and spatial navigation. Because of the diminished PI3K/Akt activities, PIKE(-/-) neurons are more vulnerable to glutamate- or stroke-induced neuronal cell death. Together, our data established the critical role of PIKE in regulating neuronal survival and development by substantiating the PI3K/Akt pathway.


Assuntos
Sobrevivência Celular/fisiologia , Dendritos/fisiologia , GTP Fosfo-Hidrolases/fisiologia , Neocórtex/crescimento & desenvolvimento , Neocórtex/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Neurogênese/fisiologia , Neurônios/fisiologia , Animais , Apoptose/genética , Apoptose/fisiologia , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Fator Neurotrófico Derivado do Encéfalo/fisiologia , Técnicas de Cultura de Células , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Dendritos/metabolismo , Dendritos/ultraestrutura , GTP Fosfo-Hidrolases/genética , Ácido Glutâmico/farmacologia , Infarto da Artéria Cerebral Média/metabolismo , Infarto da Artéria Cerebral Média/fisiopatologia , Locomoção/genética , Locomoção/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neocórtex/citologia , Neocórtex/metabolismo , Proteínas do Tecido Nervoso/genética , Neurogênese/genética , Neurônios/metabolismo , Neurônios/ultraestrutura , Tamanho do Órgão , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Comportamento Espacial/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA