Insulin signaling

胰岛素信号
  • 文章类型: Journal Article
    1型糖尿病(T1DM)的发病机制涉及氧化应激和炎症反应。姜黄素,一种在姜黄中发现的天然多酚化合物,已知具有抗氧化和抗炎特性,化学稳定性差,低生物利用度,和快速的新陈代谢。具有β-二酮结构缺失和增强的稳定性和生物利用度的姜黄素的单羰基类似物(MAC)提出了与使用姜黄素相关的挑战的潜在解决方案。本研究旨在评估两种MAC的效果,C66和B2BrBC,关于氧化应激标志物,抗氧化酶活性,糖尿病相关基因的表达,和信号通路蛋白在T1DM的背景下。链脲佐菌素(STZ)诱导的雄性Wistar大鼠或大鼠胰腺RIN-m细胞用于体内和体外实验,分别。在STZ治疗之前或之后给予C66或B2BrBC。在各种组织中确定了氧化应激标记和抗氧化酶活性。使用RT-qPCR评估糖尿病相关基因的表达,通过Westernblot分析确定胰腺中信号通路蛋白的活性。用C66和B2BrBC处理可显着降低氧化应激标志物,并对抗氧化酶活性产生积极影响。此外,两种化合物均抑制胰腺中的JNK活性,同时增强对β细胞存活以及葡萄糖和氧化还原稳态至关重要的基因的表达。研究结果强调了C66和B2BrBC在改善氧化应激方面的多方面潜力,影响与糖尿病相关的基因表达模式,调节胰腺中的关键信号通路。研究结果表明,这些化合物可以潜在地解决糖尿病相关的病理过程。
    The pathogenesis of type 1 diabetes mellitus (T1DM) involves oxidative stress and inflammation. Curcumin, a natural polyphenolic compound found in turmeric, known to exhibit antioxidative and anti-inflammatory properties, is characterized by poor chemical stability, low bioavailability, and rapid metabolism. Monocarbonyl analogs of curcumin (MACs) with a structural absence of β-diketone and enhanced stability and bioavailability present a potential solution to the challenges associated with the use of curcumin. This study aimed to evaluate the effect of two MACs, C66 and B2BrBC, on oxidative stress markers, antioxidant enzyme activity, expression of diabetes-associated genes, and signaling pathway proteins in the context of T1DM. Streptozotocin (STZ)-induced male Wistar rats or rat pancreatic RIN-m cells were used for in vivo and in vitro experiments, respectively. C66 or B2BrBC were given either before or after STZ treatment. Oxidative stress markers and antioxidant enzyme activities were determined in various tissues. Expression of diabetes-associated genes was assessed using RT-qPCR, and the activity of signaling pathway proteins in the pancreas was determined through Western blot analysis. Treatment with C66 and B2BrBC significantly reduced oxidative stress markers and positively influenced antioxidant enzyme activities. Moreover, both compounds inhibited JNK activity in the pancreas while enhancing the expression of genes crucial for β-cell survival and glucose and redox homeostasis. The findings highlight the multifaceted potential of C66 and B2BrBC in ameliorating oxidative stress, influencing gene expression patterns linked to diabetes, and modulating key signaling pathways in the pancreas. The findings suggest that these compounds can potentially address diabetes-related pathological processes.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    生酮饮食(KD)的脂肪含量很高,碳水化合物含量很低。有证据支持KD改善肥胖和/或胰岛素抵抗的人类和啮齿动物的葡萄糖代谢。相反,健康啮齿动物的研究结果表明,KD可能损害葡萄糖稳态.此外,大多数实验KD由饱和和单不饱和脂肪酸组成,几乎没有omega-3长链多不饱和脂肪酸(n-3LCPUFA)。证据支持n-3LCPUFA在代谢挑战背景下对葡萄糖稳态的有益作用。据我们所知,没有研究检测n-3LCPUFA的纳入是否会影响KD对葡萄糖稳态的影响.这项研究的目的是检查KD对大鼠全身葡萄糖耐量和骨骼肌胰岛素反应的影响,并确定用薄荷油增加KD中n-3LCPUFA的含量是否可以改善代谢结果。雄性SpragueDawley大鼠配对喂养一种低脂饮食,高脂肪饮食,KD,或补充了薄荷油(KDn-3)的KD,持续8周。全身葡萄糖耐量无显著差异,骨骼肌胰岛素信号,或者在饮食组间检测到骨骼肌胰岛素刺激的葡萄糖摄取。我们的研究结果表明,KD喂养,有或没有补充n-3LCPUFA,在配对喂养条件下不影响全身葡萄糖稳态或骨骼肌胰岛素反应。
    Ketogenic diets (KDs) are very high in fat and low in carbohydrates. Evidence supports that KDs improve glucose metabolism in humans and rodents that are obese and/or insulin resistant. Conversely, findings in healthy rodents suggest that KDs may impair glucose homeostasis. Additionally, most experimental KDs are composed of saturated and monounsaturated fatty acids, with almost no omega-3 long-chain polyunsaturated fatty acids (n-3 LCPUFA). Evidence supports a beneficial role for n-3 LCPUFA on glucose homeostasis in the context of a metabolic challenge. To our knowledge, no study has examined whether the inclusion of n-3 LCPUFA affects the impact of a KD on glucose homeostasis. The objective of this study was to examine the impact of a KD on whole-body glucose tolerance and skeletal muscle insulin response in rats, and to determine if increasing the n-3 LCPUFA content in a KD with menhaden oil could improve metabolic outcomes. Male Sprague Dawley rats were pair-fed one of a low-fat diet, high-fat diet, KD, or a KD supplemented with menhaden oil (KDn-3) for 8 weeks. No significant differences in whole-body glucose tolerance, skeletal muscle insulin signaling, or skeletal muscle insulin-stimulated glucose uptake were detected between the dietary groups. Our findings suggest that KD feeding, with or without supplementation of n-3 LCPUFA, does not affect whole-body glucose homeostasis or skeletal muscle insulin response under pair-feeding conditions.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    胰岛素信号对调节细胞代谢至关重要,增长,和生存途径,特别是在脂肪等组织中,骨骼肌,肝脏,和大脑。它在心脏中的作用,然而,不太深入的探索。心脏,需要大量的ATP来推动其收缩机制,依靠胰岛素信号传导来管理心肌底物供应并直接影响心肌代谢。这篇综述调查了胰岛素-心脏轴,关注胰岛素对心脏功能的多方面影响,从代谢调节到生理性心肌肥大的发展。本综述的中心主题是胰岛素抵抗的病理生理学及其对心脏健康的深远影响。我们讨论了胰岛素信号调节心肌细胞葡萄糖和脂肪酸代谢的复杂分子机制。强调其在维持心脏能量稳态方面的关键作用。胰岛素抵抗破坏了这些过程,导致严重的心脏代谢紊乱,自主神经功能障碍,亚细胞信号异常,和激活肾素-血管紧张素-醛固酮系统。这些因素共同促进糖尿病性心肌病和其他心血管疾病的进展。胰岛素抵抗与肥大有关,纤维化,舒张功能障碍,收缩性心力衰竭,增加冠状动脉疾病和心力衰竭的风险。了解胰岛素-心脏轴对于制定治疗策略以减轻与胰岛素抵抗和糖尿病相关的心血管并发症至关重要。
    Insulin signaling is vital for regulating cellular metabolism, growth, and survival pathways, particularly in tissues such as adipose, skeletal muscle, liver, and brain. Its role in the heart, however, is less well-explored. The heart, requiring significant ATP to fuel its contractile machinery, relies on insulin signaling to manage myocardial substrate supply and directly affect cardiac muscle metabolism. This review investigates the insulin-heart axis, focusing on insulin\'s multifaceted influence on cardiac function, from metabolic regulation to the development of physiological cardiac hypertrophy. A central theme of this review is the pathophysiology of insulin resistance and its profound implications for cardiac health. We discuss the intricate molecular mechanisms by which insulin signaling modulates glucose and fatty acid metabolism in cardiomyocytes, emphasizing its pivotal role in maintaining cardiac energy homeostasis. Insulin resistance disrupts these processes, leading to significant cardiac metabolic disturbances, autonomic dysfunction, subcellular signaling abnormalities, and activation of the renin-angiotensin-aldosterone system. These factors collectively contribute to the progression of diabetic cardiomyopathy and other cardiovascular diseases. Insulin resistance is linked to hypertrophy, fibrosis, diastolic dysfunction, and systolic heart failure, exacerbating the risk of coronary artery disease and heart failure. Understanding the insulin-heart axis is crucial for developing therapeutic strategies to mitigate the cardiovascular complications associated with insulin resistance and diabetes.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    肥胖症的发病率在世界范围内迅速增加。长期以来,肥胖相关的胰岛素抵抗已被确定为肥胖相关疾病如2型糖尿病和动脉粥样硬化的重要危险因素。胰岛素在全身葡萄糖代谢中起关键作用,肝脏,骨骼肌,和脂肪组织作为主要作用组织。胰岛素受体和下游胰岛素信号相关分子在各种组织中表达,包括血管内皮细胞,血管平滑肌细胞,和单核细胞/巨噬细胞。在肥胖症中,胰岛素作用降低被认为是相关疾病的驱动因素。然而,胰岛素的作用对肥胖相关疾病有正面还是负面的影响取决于它所作用的组织.虽然肝脏中胰岛素信号的增强会增加肝脏脂肪积累并加剧血脂异常,脂肪组织中胰岛素信号的增强通过增加脂肪组织中脂肪积累的能力和抑制异位脂肪积累来防止各种器官的肥胖相关功能障碍。因此,这种“健康的脂肪组织扩张”通过增强脂肪组织中的胰岛素敏感性,但不是在肝脏,可能是肥胖相关疾病的有效治疗策略。为了有效解决肥胖相关的代谢紊乱,必须了解肥胖患者各种组织中胰岛素抵抗的机制,并开发增强胰岛素作用的药物。在这篇文章中,我们回顾了增强胰岛素信号的干预措施作为肥胖相关疾病治疗策略的潜力。关注胰岛素在每个组织中作用的分子机制。
    The incidence of obesity is rapidly increasing worldwide. Obesity-associated insulin resistance has long been established as a significant risk factor for obesity-related disorders such as type 2 diabetes and atherosclerosis. Insulin plays a key role in systemic glucose metabolism, with the liver, skeletal muscle, and adipose tissue as the major acting tissues. Insulin receptors and the downstream insulin signaling-related molecules are expressed in various tissues, including vascular endothelial cells, vascular smooth muscle cells, and monocytes/macrophages. In obesity, decreased insulin action is considered a driver for associated disorders. However, whether insulin action has a positive or negative effect on obesity-related disorders depends on the tissue in which it acts. While an enhancement of insulin signaling in the liver increases hepatic fat accumulation and exacerbates dyslipidemia, enhancement of insulin signaling in adipose tissue protects against obesity-related dysfunction of various organs by increasing the capacity for fat accumulation in the adipose tissue and inhibiting ectopic fat accumulation. Thus, this \"healthy adipose tissue expansion\" by enhancing insulin sensitivity in adipose tissue, but not in the liver, may be an effective therapeutic strategy for obesity-related disorders. To effectively address obesity-related metabolic disorders, the mechanisms of insulin resistance in various tissues of obese patients must be understood and drugs that enhance insulin action must be developed. In this article, we review the potential of interventions that enhance insulin signaling as a therapeutic strategy for obesity-related disorders, focusing on the molecular mechanisms of insulin action in each tissue.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    针对脑胰岛素抵抗(BIR)已成为阿尔茨海默病(AD)传统治疗方法的一种有吸引力的替代方法。肠促胰岛素受体激动剂(IRAs),靶向胰高血糖素样肽1(GLP-1)和葡萄糖依赖性促胰岛素多肽(GIP)受体之一或两者,已被证明可以逆转BIR并改善AD小鼠模型的认知。我们之前展示了很多,但不是全部,IRA可在静脉内(IV)递送后穿过血脑屏障(BBB)。在这里,我们确定是否可以通过使用鼻内(IN)递送绕过BBB来实现IRA的广泛脑摄取,其具有最小化全身递送的IRA的不良胃肠道作用的额外优点。在测试的5个放射性标记的IRA中(艾塞那肽,杜拉鲁肽,塞马鲁肽,DA4-JC,和DA5-CH)在CD-1小鼠中,艾塞那肽,杜拉鲁肽,和DA4-JC在IN递送后成功分布在整个大脑中。我们观察到DA4-JC摄取的显著性别差异。Dulaglutide和DA4-JC表现出海马和多个新皮质区域的高摄取。我们进一步测试并发现与AD相关的Aβ病理的存在对杜拉鲁肽和DA4-JC的摄取影响最小。在5个测试的IRA中,杜拉鲁肽和DA4-JC最有能力在给药后进入AD中最脆弱的大脑区域(新皮质和海马)。需要进行未来的研究以确定在AD或该疾病的动物模型中INIRA递送是否可以减少BIR。
    Targeting brain insulin resistance (BIR) has become an attractive alternative to traditional therapeutic treatments for Alzheimer\'s disease (AD). Incretin receptor agonists (IRAs), targeting either or both of the glucagon-like peptide 1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) receptors, have proven to reverse BIR and improve cognition in mouse models of AD. We previously showed that many, but not all, IRAs can cross the blood-brain barrier (BBB) after intravenous (IV) delivery. Here we determined if widespread brain uptake of IRAs could be achieved by circumventing the BBB using intranasal (IN) delivery, which has the added advantage of minimizing adverse gastrointestinal effects of systemically delivered IRAs. Of the 5 radiolabeled IRAs tested (exenatide, dulaglutide, semaglutide, DA4-JC, and DA5-CH) in CD-1 mice, exenatide, dulaglutide, and DA4-JC were successfully distributed throughout the brain following IN delivery. We observed significant sex differences in uptake for DA4-JC. Dulaglutide and DA4-JC exhibited high uptake by the hippocampus and multiple neocortical areas. We further tested and found the presence of AD-associated Aβ pathology minimally affected uptake of dulaglutide and DA4-JC. Of the 5 tested IRAs, dulaglutide and DA4-JC are best capable of accessing brain regions most vulnerable in AD (neocortex and hippocampus) after IN administration. Future studies will need to be performed to determine if IN IRA delivery can reduce BIR in AD or animal models of that disorder.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    目的:骨骼是人体最大的器官之一,其中代谢与全身能量代谢整合。然而,骨细胞的生物能量编程,最丰富的骨细胞协调骨代谢,没有明确定义。这里,使用部分穿透骨细胞特异性PPARG缺失的小鼠模型,我们证明PPARG独立于循环硬化素水平控制骨细胞生物能学及其对全身能量代谢的贡献,以前与髓外脂肪库的代谢状态相关。
    方法:骨细胞特异性PPARG缺失的体内和体外模型,即Dmp1CrePparγflfl雄性和雌性小鼠(γOTKO)和MLO-Y4骨细胞样细胞,具有siRNA沉默或CRISPR/Cas9编辑的Pparγ。如适用,分析了模型的能量代谢水平,葡萄糖代谢,和髓外脂肪组织的代谢谱,以及骨细胞转录组,线粒体功能,生物能学,胰岛素信号,和氧化应激。
    结果:γOTKO雄性和雌性小鼠的循环硬化素水平与对照小鼠没有差异。雄性γOTKO小鼠表现出一种以呼吸增加为特征的高能量表型,产热,运动和食物摄入。男性的这种高能量表型与周围脂肪库的“米色”无关。然而,两种性别都表现出脂肪量减少和明显的胰岛素抵抗的趋势,而葡萄糖耐量没有变化,这与通过AKT激活测量的骨细胞对胰岛素的反应性降低相关。从γOTKO雄性分离的骨细胞的转录组表明细胞代谢发生了深刻的变化,燃料运输,线粒体功能障碍,胰岛素信号和氧化应激增加。在MLO-Y4骨细胞中,PPARG缺乏与高活性线粒体相关,增加ATP产量,和活性氧(ROS)的积累。
    结论:男性骨细胞中的PPARG作为线粒体功能的分子断裂,以及对氧化应激和ROS积累的保护。它还调节骨细胞胰岛素信号和燃料使用以产生能量。这些数据提供了对骨细胞生物能学及其对全身能量代谢平衡的性别特异性贡献之间联系的见解。这些发现支持骨骼通过骨细胞代谢控制全身能量消耗的概念。
    OBJECTIVE: The skeleton is one of the largest organs in the body, wherein metabolism is integrated with systemic energy metabolism. However, the bioenergetic programming of osteocytes, the most abundant bone cells coordinating bone metabolism, is not well defined. Here, using a mouse model with partial penetration of an osteocyte-specific PPARG deletion, we demonstrate that PPARG controls osteocyte bioenergetics and their contribution to systemic energy metabolism independently of circulating sclerostin levels, which were previously correlated with metabolic status of extramedullary fat depots.
    METHODS: In vivo and in vitro models of osteocyte-specific PPARG deletion, i.e. Dmp1CrePparγflfl male and female mice (γOTKO) and MLO-Y4 osteocyte-like cells with either siRNA-silenced or CRISPR/Cas9-edited Pparγ. As applicable, the models were analyzed for levels of energy metabolism, glucose metabolism, and metabolic profile of extramedullary adipose tissue, as well as the osteocyte transcriptome, mitochondrial function, bioenergetics, insulin signaling, and oxidative stress.
    RESULTS: Circulating sclerostin levels of γOTKO male and female mice were not different from control mice. Male γOTKO mice exhibited a high energy phenotype characterized by increased respiration, heat production, locomotion and food intake. This high energy phenotype in males did not correlate with \"beiging\" of peripheral adipose depots. However, both sexes showed a trend for reduced fat mass and apparent insulin resistance without changes in glucose tolerance, which correlated with decreased osteocytic responsiveness to insulin measured by AKT activation. The transcriptome of osteocytes isolated from γOTKO males suggested profound changes in cellular metabolism, fuel transport, mitochondria dysfunction, insulin signaling and increased oxidative stress. In MLO-Y4 osteocytes, PPARG deficiency correlated with highly active mitochondria, increased ATP production, and accumulation of reactive oxygen species (ROS).
    CONCLUSIONS: PPARG in male osteocytes acts as a molecular break on mitochondrial function, and protection against oxidative stress and ROS accumulation. It also regulates osteocyte insulin signaling and fuel usage to produce energy. These data provide insight into the connection between osteocyte bioenergetics and their sex-specific contribution to the balance of systemic energy metabolism. These findings support the concept that the skeleton controls systemic energy expenditure via osteocyte metabolism.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Editorial
    暂无摘要。
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:胰岛素信号调节心脏底物的利用,并与心脏的生理适应有关。心脏内信号应答的改变被认为导致病理状况,例如2型糖尿病和心力衰竭。虽然使用磷酸蛋白质组学策略在几个代谢器官中进行了广泛的研究,一般在心脏组织中引起的信号反应,特别是在专门的心肌细胞中,尚未进行相同程度的调查。
    方法:通过静脉内注射将胰岛素或载体给予C57BL6/JRj雄性小鼠。提取心室组织并进行定量磷酸蛋白质组学分析以评估胰岛素信号传导反应。为了描述心肌细胞特异性反应并研究Tbc1d4在胰岛素信号转导中的作用,来自心脏和骨骼肌特异性Tbc1d4基因敲除小鼠心脏的心肌细胞,以及野生同窝,被研究过。磷酸蛋白质组学研究涉及用串联质量标签(TMT)标记的同量异序肽,富集磷酸化肽,通过微流反相液相色谱进行分馏,和高分辨率质谱测量。
    结果:我们定量了来自心室组织的10,399个磷酸化肽和来自分离心肌细胞的12,739个磷酸化肽,定位到3,232和3,128个独特的蛋白质,分别。在心脏组织中,我们确定了84个胰岛素调节的磷酸化事件,包括胰岛素受体(InsrY1351,Y1175,Y1179,Y1180)本身的位点以及胰岛素受体底物蛋白1(Irs1S522,S526)。响应于胰岛素刺激具有增加的活性的预测激酶包括Rps6kb1、Akt1和Mtor。Tbc1d4在心肌细胞中涌现为主要的磷酸化目标。尽管对全球磷酸化格局的影响有限,心肌细胞中的Tbc1d4缺乏减弱胰岛素诱导的Glut4易位和诱导的蛋白质重塑。我们观察到15种蛋白质在敲除Tbc1d4后被显着调节。虽然Glut4由于Tbc1d4缺乏而表现出蛋白质丰度降低,Txnip水平显著增加。用胰岛素刺激野生型心肌细胞导致262个显著的磷酸化事件的调节,预测受Akt1、Mtor、Akt2和Insr。在心肌细胞中,除了调节专门的心肌细胞蛋白外,还引发了典型的胰岛素信号应答,例如Kcnj11Y12和DspS2597。提供了所有磷酸化位点的细节。
    结论:我们首次概述了心脏组织和分离的成年心肌细胞中胰岛素诱导的磷酸化信号反应,详述具有改变的磷酸化丰度的特定残基。我们的研究标志着了解胰岛素信号在与胰岛素抵抗相关的心脏病中的作用的重要一步。
    BACKGROUND: Insulin signaling regulates cardiac substrate utilization and is implicated in physiological adaptations of the heart. Alterations in the signaling response within the heart are believed to contribute to pathological conditions such as type-2 diabetes and heart failure. While extensively investigated in several metabolic organs using phosphoproteomic strategies, the signaling response elicited in cardiac tissue in general, and specifically in the specialized cardiomyocytes, has not yet been investigated to the same extent.
    METHODS: Insulin or vehicle was administered to male C57BL6/JRj mice via intravenous injection into the vena cava. Ventricular tissue was extracted and subjected to quantitative phosphoproteomics analysis to evaluate the insulin signaling response. To delineate the cardiomyocyte-specific response and investigate the role of Tbc1d4 in insulin signal transduction, cardiomyocytes from the hearts of cardiac and skeletal muscle-specific Tbc1d4 knockout mice, as well as from wildtype littermates, were studied. The phosphoproteomic studies involved isobaric peptide labeling with Tandem Mass Tags (TMT), enrichment for phosphorylated peptides, fractionation via micro-flow reversed-phase liquid chromatography, and high-resolution mass spectrometry measurements.
    RESULTS: We quantified 10,399 phosphorylated peptides from ventricular tissue and 12,739 from isolated cardiomyocytes, localizing to 3,232 and 3,128 unique proteins, respectively. In cardiac tissue, we identified 84 insulin-regulated phosphorylation events, including sites on the Insulin Receptor (InsrY1351, Y1175, Y1179, Y1180) itself as well as the Insulin receptor substrate protein 1 (Irs1S522, S526). Predicted kinases with increased activity in response to insulin stimulation included Rps6kb1, Akt1 and Mtor. Tbc1d4 emerged as a major phosphorylation target in cardiomyocytes. Despite limited impact on the global phosphorylation landscape, Tbc1d4 deficiency in cardiomyocytes attenuated insulin-induced Glut4 translocation and induced protein remodeling. We observed 15 proteins significantly regulated upon knockout of Tbc1d4. While Glut4 exhibited decreased protein abundance consequent to Tbc1d4-deficiency, Txnip levels were notably increased. Stimulation of wildtype cardiomyocytes with insulin led to the regulation of 262 significant phosphorylation events, predicted to be regulated by kinases such as Akt1, Mtor, Akt2, and Insr. In cardiomyocytes, the canonical insulin signaling response is elicited in addition to regulation on specialized cardiomyocyte proteins, such as Kcnj11Y12 and DspS2597. Details of all phosphorylation sites are provided.
    CONCLUSIONS: We present a first global outline of the insulin-induced phosphorylation signaling response in heart tissue and in isolated adult cardiomyocytes, detailing the specific residues with changed phosphorylation abundances. Our study marks an important step towards understanding the role of insulin signaling in cardiac diseases linked to insulin resistance.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    妊娠期糖尿病(GDM)在全球范围内提出了重大的健康问题,需要全面了解其代谢复杂性以进行有效管理。MicroRNAs(miRNAs)已成为GDM发病机制中的关键调节因子,影响葡萄糖代谢,胰岛素信号,和怀孕期间的脂质稳态。miRNA表达失调,上调和下调,有助于GDM相关的代谢异常。miRNA表达的种族和时间变化强调了GDM易感性的多面性。本文综述了GDM中miRNAs的失调及其在代谢紊乱中的调控功能。我们讨论了特定miRNA在调节GDM发病机制中涉及的关键通路中的参与,比如葡萄糖代谢,胰岛素信号,和脂质稳态。此外,我们探讨了miRNA在GDM管理中的潜在诊断和治疗意义,强调了基于miRNA的干预措施在减轻GDM对孕产妇和后代健康的不利影响方面的前景。
    Gestational Diabetes Mellitus (GDM) presents a significant health concern globally, necessitating a comprehensive understanding of its metabolic intricacies for effective management. MicroRNAs (miRNAs) have emerged as pivotal regulators in GDM pathogenesis, influencing glucose metabolism, insulin signaling, and lipid homeostasis during pregnancy. Dysregulated miRNA expression, both upregulated and downregulated, contributes to GDM-associated metabolic abnormalities. Ethnic and temporal variations in miRNA expression underscore the multifaceted nature of GDM susceptibility. This review examines the dysregulation of miRNAs in GDM and their regulatory functions in metabolic disorders. We discuss the involvement of specific miRNAs in modulating key pathways implicated in GDM pathogenesis, such as glucose metabolism, insulin signaling, and lipid homeostasis. Furthermore, we explore the potential diagnostic and therapeutic implications of miRNAs in GDM management, highlighting the promise of miRNA-based interventions for mitigating the adverse consequences of GDM on maternal and offspring health.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    组织在小的细胞外囊泡(sEV)中释放微小RNA(miRNA),包括外泌体,可以调节远端细胞的基因表达,从而充当局部和全身代谢的调节剂。这里,我们显示胰岛素调节3T3-L1脂肪细胞分泌到sEV中的miRNA,并且该过程与细胞表达不同.因此,在3T3-L1sEV中,胰岛素上调的53个miRNA和下调的66个miRNA,在细胞中只有12个被平行调节。胰岛素部分通过磷酸化hnRNPA1调节这一过程,使其与miRNAs中富含AU的基序结合,将它们的分泌介导到sEV中。重要的是,43%的胰岛素调节sEV-miRNA与肥胖和胰岛素抵抗有关。这些包括let-7和miR-103,我们显示在AML12肝细胞中调节胰岛素信号。一起,这些发现证明了胰岛素调节脂肪生物学的一个重要层面,并提供了肥胖和其他高胰岛素血症状态的组织串扰机制。
    Tissues release microRNAs (miRNAs) in small extracellular vesicles (sEVs) including exosomes, which can regulate gene expression in distal cells, thus acting as modulators of local and systemic metabolism. Here, we show that insulin regulates miRNA secretion into sEVs from 3T3-L1 adipocytes and that this process is differentially regulated from cellular expression. Thus, of the 53 miRNAs upregulated and 66 miRNAs downregulated by insulin in 3T3-L1 sEVs, only 12 were regulated in parallel in cells. Insulin regulated this process in part by phosphorylating hnRNPA1, causing it to bind to AU-rich motifs in miRNAs, mediating their secretion into sEVs. Importantly, 43% of insulin-regulated sEV-miRNAs are implicated in obesity and insulin resistance. These include let-7 and miR-103, which we show regulate insulin signaling in AML12 hepatocytes. Together, these findings demonstrate an important layer to insulin\'s regulation of adipose biology and provide a mechanism of tissue crosstalk in obesity and other hyperinsulinemic states.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号