β-cell

β 细胞
  • 文章类型: Journal Article
    在1型糖尿病(T1D)中,自身反应性免疫细胞浸润胰腺并分泌促炎细胞因子,从而引发产生胰岛素的胰岛β细胞的细胞死亡。蛋白激酶Cδ(PKCδ)在介导细胞因子诱导的β细胞死亡中起作用;然而,确切的机制还没有很好的理解。为了解决这个问题,我们使用了诱导型β细胞特异性PKCδKO小鼠以及PKCδ的小肽抑制剂。我们确定了PKCδ在介导细胞因子诱导的β细胞死亡中的作用,并表明抑制PKCδ可保护胰腺β细胞免受小鼠和人胰岛中细胞因子诱导的凋亡。我们确定细胞因子诱导了PKCδ的核易位和活性,并且细胞因子介导的胰岛细胞凋亡可能需要caspase-3裂解PKCδ。Further,细胞因子激活的PKCδ增加急性治疗的促凋亡Bax和长期治疗的JNK的活性。总的来说,我们的结果表明,PKCδ通过核转位介导细胞因子诱导的细胞凋亡,caspase-3的裂解,以及胰腺β细胞中促凋亡信号的上调。结合PKCδ抑制与δV1-1的保护作用,这项研究的结果将有助于开发新疗法,以预防或延迟β细胞死亡并保留T1D中的β细胞功能。
    In type 1 diabetes (T1D), autoreactive immune cells infiltrate the pancreas and secrete proinflammatory cytokines that initiate cell death in insulin producing islet β-cells. Protein kinase C δ (PKCδ) plays a role in mediating cytokine-induced β-cell death; however, the exact mechanisms are not well understood. To address this, we used an inducible β-cell specific PKCδ KO mouse as well as a small peptide inhibitor of PKCδ. We identified a role for PKCδ in mediating cytokine-induced β-cell death and have shown that inhibiting PKCδ protects pancreatic β-cells from cytokine-induced apoptosis in both mouse and human islets. We determined that cytokines induced nuclear translocation and activity of PKCδ and that caspase-3 cleavage of PKCδ may be required for cytokine-mediated islet apoptosis. Further, cytokine activated PKCδ increases activity both of proapoptotic Bax with acute treatment and C-Jun N-terminal kinase with prolonged treatment. Overall, our results suggest that PKCδ mediates cytokine-induced apoptosis via nuclear translocation, cleavage by caspase-3, and upregulation of proapoptotic signaling in pancreatic β-cells. Combined with the protective effects of PKCδ inhibition with δV1-1, the results of this study will aid in the development of novel therapies to prevent or delay β-cell death and preserve β-cell function in T1D.
    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
    胰腺再生是在正常和病理条件下观察到的复杂过程。这篇综述的目的是全面了解成年胰腺中功能活跃的分泌胰岛素的β细胞群的出现。β细胞的更新受遗传和表观遗传因素的细胞来源之间的多方面相互作用支配。了解β细胞群的发育和异质性对于功能性β细胞再生至关重要。在妊娠和肥胖等情况下,胰腺β细胞的功能质量会增加。然而,在这些条件下能够增加自我繁殖的成熟β细胞群和出生后胰腺祖细胞的特异性标志物仍有待阐明。通过各种途径再生β细胞群的能力,包括预先存在的β细胞的增殖,β细胞新生,从祖细胞群体中分化出β细胞,非β细胞转分化为β细胞,揭示了识别细胞来源和功能性细胞更新诱导物的关键分子机制。这为确定特定的细胞来源和再生机制提供了机会,可以在临床上应用于治疗各种疾病,包括体外细胞技术,加深对不同生理条件下再生的认识。
    Pancreatic regeneration is a complex process observed in both normal and pathological conditions. The aim of this review is to provide a comprehensive understanding of the emergence of a functionally active population of insulin-secreting β-cells in the adult pancreas. The renewal of β-cells is governed by a multifaceted interaction between cellular sources of genetic and epigenetic factors. Understanding the development and heterogeneity of β-cell populations is crucial for functional β-cell regeneration. The functional mass of pancreatic β-cells increases in situations such as pregnancy and obesity. However, the specific markers of mature β-cell populations and postnatal pancreatic progenitors capable of increasing self-reproduction in these conditions remain to be elucidated. The capacity to regenerate the β-cell population through various pathways, including the proliferation of pre-existing β-cells, β-cell neogenesis, differentiation of β-cells from a population of progenitor cells, and transdifferentiation of non-β-cells into β-cells, reveals crucial molecular mechanisms for identifying cellular sources and inducers of functional cell renewal. This provides an opportunity to identify specific cellular sources and mechanisms of regeneration, which could have clinical applications in treating various pathologies, including in vitro cell-based technologies, and deepen our understanding of regeneration in different physiological conditions.
    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:受体相互作用蛋白激酶1(RIPK1)协调肿瘤坏死因子(TNF)和其他细胞因子在细胞存活和细胞死亡之间的决定。而RIPK1的支架功能对于防止TNF诱导的细胞凋亡和坏死性凋亡至关重要,它的激酶活性是细胞坏死所必需的,部分是细胞凋亡所必需的。尽管TNF是一种与糖尿病中β细胞丢失相关的促炎细胞因子,TNF诱导β细胞死亡的机制尚不清楚。
    方法:这里,我们使用缺乏RIPK1的小鼠(Ripk1β-KO小鼠)或表达激酶死亡版本的RIPK1(Ripk1D138N小鼠),分析了RIPK1支架与激酶功能对β细胞死亡调节的贡献,分别。这些小鼠被链脲佐菌素攻击,自身免疫性糖尿病模型.此外,用高脂肪饮食进一步攻击Ripk1β-KO小鼠以诱导高血糖。对于机械学研究,胰岛接受了各种杀伤剂和致敏剂。
    结果:抑制RIPK1激酶活性(Ripk1D138N小鼠)不影响1型糖尿病模型中高血糖的发作和进展。此外,β细胞中RIPK1表达缺失不影响基础条件下的血糖正常或糖尿病挑战下的高血糖.离体,在没有RIPK1的情况下,原代胰岛对TNF诱导的凋亡和坏死不敏感。有趣的是,我们发现胰岛显示高水平的抗凋亡细胞FLICE抑制蛋白(cFLIP)和低水平的凋亡(Caspase-8)和坏死(RIPK3)成分.环己酰亚胺处理,这导致了cFLIP水平的降低,使原代胰岛对TNF诱导的细胞死亡敏感,而TNF诱导的细胞死亡被胱天蛋白酶抑制完全阻断。
    结论:与许多其他细胞类型不同(例如,上皮,和免疫),在生理条件或糖尿病挑战下,RIPK1对于β细胞中的细胞死亡调节不是必需的。此外,体内和体外证据表明,胰腺β细胞不会发生坏死,而主要是响应TNF的caspase依赖性死亡。最后,我们的结果表明β细胞具有独特的TNF-细胞毒性调节模式,该模式独立于RIPK1,并且可能高度依赖于cFLIP.
    OBJECTIVE: Receptor-interacting protein kinase 1 (RIPK1) orchestrates the decision between cell survival and cell death in response to tumor necrosis factor (TNF) and other cytokines. Whereas the scaffolding function of RIPK1 is crucial to prevent TNF-induced apoptosis and necroptosis, its kinase activity is required for necroptosis and partially for apoptosis. Although TNF is a proinflammatory cytokine associated with β-cell loss in diabetes, the mechanism by which TNF induces β-cell demise remains unclear.
    METHODS: Here, we dissected the contribution of RIPK1 scaffold versus kinase functions to β-cell death regulation using mice lacking RIPK1 specifically in β-cells (Ripk1β-KO mice) or expressing a kinase-dead version of RIPK1 (Ripk1D138N mice), respectively. These mice were challenged with streptozotocin, a model of autoimmune diabetes. Moreover, Ripk1β-KO mice were further challenged with a high-fat diet to induce hyperglycemia. For mechanistic studies, pancreatic islets were subjected to various killing and sensitising agents.
    RESULTS: Inhibition of RIPK1 kinase activity (Ripk1D138N mice) did not affect the onset and progression of hyperglycemia in a type 1 diabetes model. Moreover, the absence of RIPK1 expression in β-cells did not affect normoglycemia under basal conditions or hyperglycemia under diabetic challenges. Ex vivo, primary pancreatic islets are not sensitised to TNF-induced apoptosis and necroptosis in the absence of RIPK1. Intriguingly, we found that pancreatic islets display high levels of the antiapoptotic cellular FLICE-inhibitory protein (cFLIP) and low levels of apoptosis (Caspase-8) and necroptosis (RIPK3) components. Cycloheximide treatment, which led to a reduction in cFLIP levels, rendered primary islets sensitive to TNF-induced cell death which was fully blocked by caspase inhibition.
    CONCLUSIONS: Unlike in many other cell types (e.g., epithelial, and immune), RIPK1 is not required for cell death regulation in β-cells under physiological conditions or diabetic challenges. Moreover, in vivo and in vitro evidence suggest that pancreatic β-cells do not undergo necroptosis but mainly caspase-dependent death in response to TNF. Last, our results show that β-cells have a distinct mode of regulation of TNF-cytotoxicity that is independent of RIPK1 and that may be highly dependent on cFLIP.
    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
    在胰岛素抵抗和β细胞质量降低的情况下,β细胞工作量增加,发生在2型和1型糖尿病中,分别。在糖尿病的发病过程中,胰岛素产生和分泌的长期升高会导致β细胞内质网应激。内质网应激过程中β细胞Ca2+内质网的消耗激活了未折叠的蛋白反应,导致β细胞功能障碍。Ca2+ER参与许多对β细胞功能至关重要的途径,比如蛋白质加工,调节细胞器和胞质Ca2+处理,和调节脂质稳态。促进β细胞内质网应激和耗尽Ca2+内质网储存的突变与糖尿病相关或引起糖尿病(例如,ryanodine受体和胰岛素的突变)。因此,改善β细胞Ca2+ER处理和减少糖尿病条件下的ER应激可以保持β细胞功能并延缓或预防糖尿病的发作。这篇综述着重于控制β细胞Ca2ER的机制在糖尿病的发病过程中如何受到干扰并导致β细胞衰竭。
    The β-cell workload increases in the setting of insulin resistance and reduced β-cell mass, which occurs in type 2 and type 1 diabetes, respectively. The prolonged elevation of insulin production and secretion during the pathogenesis of diabetes results in β-cell ER stress. The depletion of β-cell Ca2+ER during ER stress activates the unfolded protein response, leading to β-cell dysfunction. Ca2+ER is involved in many pathways that are critical to β-cell function, such as protein processing, tuning organelle and cytosolic Ca2+ handling, and modulating lipid homeostasis. Mutations that promote β-cell ER stress and deplete Ca2+ER stores are associated with or cause diabetes (e.g., mutations in ryanodine receptors and insulin). Thus, improving β-cell Ca2+ER handling and reducing ER stress under diabetogenic conditions could preserve β-cell function and delay or prevent the onset of diabetes. This review focuses on how mechanisms that control β-cell Ca2+ER are perturbed during the pathogenesis of diabetes and contribute to β-cell failure.
    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
    2型糖尿病(T2D)是一种多基因代谢疾病,其特征是外周组织中的胰岛素抵抗和胰腺的胰岛素分泌受损。虽然胰岛素产生和分泌的下降以前归因于产生胰岛素的β细胞的凋亡,最近的研究表明糖尿病患者的β细胞凋亡率相对较低。相反,β细胞主要经历去分化,在这个过程中,它们失去了专门的身份,并转变为无功能的内分泌祖细胞样细胞,最终导致β细胞衰竭。由于遗传因素和细胞应激的复杂相互作用,驱动β细胞去分化的潜在机制仍然难以捉摸。了解这些机制有可能为旨在逆转T2D中β细胞去分化的创新治疗方法提供信息。这篇综述探讨了β细胞去分化导致β细胞衰竭的拟议驱动因素,并讨论了能够逆转这一过程的当前干预措施,从而恢复β细胞的身份和功能。
    Type 2 diabetes (T2D) is a polygenic metabolic disorder characterized by insulin resistance in peripheral tissues and impaired insulin secretion by the pancreas. While the decline in insulin production and secretion was previously attributed to apoptosis of insulin-producing β-cells, recent studies indicate that β-cell apoptosis rates are relatively low in diabetes. Instead, β-cells primarily undergo dedifferentiation, a process where they lose their specialized identity and transition into non-functional endocrine progenitor-like cells, ultimately leading to β-cell failure. The underlying mechanisms driving β-cell dedifferentiation remain elusive due to the intricate interplay of genetic factors and cellular stress. Understanding these mechanisms holds the potential to inform innovative therapeutic approaches aimed at reversing β-cell dedifferentiation in T2D. This review explores the proposed drivers of β-cell dedifferentiation leading to β-cell failure, and discusses current interventions capable of reversing this process, thus restoring β-cell identity and function.
    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
    1型糖尿病发病率的2个高峰发生在儿童早期和青春期。
    我们试图更好地理解青春期之间的关系,胰岛自身免疫,1型糖尿病。
    青春期,胰岛自身免疫,在青年糖尿病的环境决定因素(TEDDY)研究中,对儿童的1型糖尿病进展进行了前瞻性调查。青春期的开始是由Tanner阶段的受试者自我评估确定的。青春期进展速度之间的关联,青春期生长,体重增加,胰岛素抵抗的稳态模型评估(HOMA-IR),胰岛自身免疫,并评估了1型糖尿病的进展。使用Cox比例风险比分析个体因素的影响。
    在5677名8岁仍在研究的儿童中,95%报告至少1个Tanner阶段得分并纳入研究。与青春期前儿童(Tanner阶段1)相比,青春期儿童(Tanner阶段≥2)发生自身免疫事件的风险较低(HR0.65,95%CI0.45-0.93;P=.019)。体重指数Z评分的增加与胰岛素自身抗体的发生率较高(HR2.88,95%CI1.61-5.15;P<.001)相关。在患有多种自身抗体的儿童中,HOMA-IR和Tanner第4阶段的进展速度均与1型糖尿病的进展无关.
    青春期体重的快速增加与胰岛自身免疫的发展有关。青春期本身对自身抗体或1型糖尿病的出现没有显着影响。需要进一步的研究来更好地了解潜在的机制。
    UNASSIGNED: The 2 peaks of type 1 diabetes incidence occur during early childhood and puberty.
    UNASSIGNED: We sought to better understand the relationship between puberty, islet autoimmunity, and type 1 diabetes.
    UNASSIGNED: The relationships between puberty, islet autoimmunity, and progression to type 1 diabetes were investigated prospectively in children followed in The Environmental Determinants of Diabetes in the Young (TEDDY) study. Onset of puberty was determined by subject self-assessment of Tanner stages. Associations between speed of pubertal progression, pubertal growth, weight gain, homeostasis model assessment of insulin resistance (HOMA-IR), islet autoimmunity, and progression to type 1 diabetes were assessed. The influence of individual factors was analyzed using Cox proportional hazard ratios.
    UNASSIGNED: Out of 5677 children who were still in the study at age 8 years, 95% reported at least 1 Tanner Stage score and were included in the study. Children at puberty (Tanner Stage ≥2) had a lower risk (HR 0.65, 95% CI 0.45-0.93; P = .019) for incident autoimmunity than prepubertal children (Tanner Stage 1). An increase of body mass index Z-score was associated with a higher risk (HR 2.88, 95% CI 1.61-5.15; P < .001) of incident insulin autoantibodies. In children with multiple autoantibodies, neither HOMA-IR nor rate of progression to Tanner Stage 4 were associated with progression to type 1 diabetes.
    UNASSIGNED: Rapid weight gain during puberty is associated with development of islet autoimmunity. Puberty itself had no significant influence on the appearance of autoantibodies or type 1 diabetes. Further studies are needed to better understand the underlying mechanisms.
    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
    糖尿病的分子发病机制是多因素的,涉及尚未完全了解的遗传易感性和环境因素。然而,胰腺β细胞衰竭仍然是2型糖尿病(T2D)进展的主要原因之一,这使得靶向β细胞功能障碍成为糖尿病治疗的有吸引力的途径.为了确定β细胞功能障碍的遗传因素,我们研究了正常或高脂肪喂养的健康(C57BL/6J)和糖尿病(NZO/HlLtJ)小鼠β细胞中单细胞基因表达的变化,高糖饮食(HFHS)。我们的研究提出了因果网络扰动评估(ssNPA)框架与元细胞转录组分析的创新整合,以探索2型糖尿病(T2D)的遗传基础。通过生成参考因果网络和计算机扰动,我们鉴定了与T2D相关的新基因,并使用敲除小鼠表型分析(KOMP)项目数据库验证了我们的候选基因.
    The molecular pathogenesis of diabetes is multifactorial, involving genetic predisposition and environmental factors that are not yet fully understood. However, pancreatic β-cell failure remains among the primary reasons underlying the progression of type-2 diabetes (T2D) making targeting β-cell dysfunction an attractive pathway for diabetes treatment. To identify genetic contributors to β-cell dysfunction, we investigated single-cell gene expression changes in β-cells from healthy (C57BL/6J) and diabetic (NZO/HlLtJ) mice fed with normal or high-fat, high-sugar diet (HFHS). Our study presents an innovative integration of the causal network perturbation assessment (ssNPA) framework with meta-cell transcriptome analysis to explore the genetic underpinnings of type-2 diabetes (T2D). By generating a reference causal network and in silico perturbation, we identified novel genes implicated in T2D and validated our candidates using the Knockout Mouse Phenotyping (KOMP) Project database.
    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
    胰岛β细胞分泌胰岛素是葡萄糖稳态的重要支柱,在肥胖和衰老的情况下受损。生长激素促分泌素受体(GHSR)是营养敏感激素ghrelin的受体。以前,我们表明β细胞GHSR调节年轻小鼠的葡萄糖刺激的胰岛素分泌(GSIS)。在目前的研究中,我们进一步研究了GHSR对饮食诱导的肥胖(DIO)和链脲佐菌素(STZ)诱导的衰老β细胞损伤的雄性小鼠胰岛素分泌的影响。β细胞特异性Ghsr缺陷型(Ghsr-βKO)小鼠在DIO下没有表现出血糖表型,但在衰老中显示出离体GSIS显着改善。我们还检测到体内和离体衰老过程中胰岛素敏感性降低和胰岛素分泌受损。因此,葡萄糖转运蛋白的表达有年龄相关的改变,胰岛素信号通路,和炎症基因。为了进一步确定GHSR缺乏是否影响β细胞对急性损伤的易感性,年轻,中年,和老年Ghsr-βKO小鼠接受STZ。我们发现中年和老年Ghsr-βKO小鼠受到STZ诱导的高血糖和胰岛素分泌受损的保护,与胰岛中胰岛素信号调节因子表达增加相关,但促炎细胞因子降低。总的来说,我们的发现表明,β细胞GHSR对衰老而不是肥胖的胰岛素分泌有重要影响,和GHSR缺乏保护STZ诱导的衰老中的β细胞损伤。
    Insulin secretion from pancreatic β cells is a key pillar of glucose homeostasis, which is impaired under obesity and aging. Growth hormone secretagogue receptor (GHSR) is the receptor of nutrient-sensing hormone ghrelin. Previously, we showed that β-cell GHSR regulated glucose-stimulated insulin secretion (GSIS) in young mice. In the current study, we further investigated the effects of GHSR on insulin secretion in male mice under diet-induced obesity (DIO) and streptozotocin (STZ)-induced β-cell injury in aging. β-cell-specific-Ghsr-deficient (Ghsr-βKO) mice exhibited no glycemic phenotype under DIO but showed significantly improved ex vivo GSIS in aging. We also detected reduced insulin sensitivity and impaired insulin secretion during aging both in vivo and ex vivo. Accordingly, there were age-related alterations in expression of glucose transporter, insulin signaling pathway, and inflammatory genes. To further determine whether GHSR deficiency affected β-cell susceptibility to acute injury, young, middle-aged, and old Ghsr-βKO mice were subjected to STZ. We found that middle-aged and old Ghsr-βKO mice were protected from STZ-induced hyperglycemia and impaired insulin secretion, correlated with increased expression of insulin signaling regulators but decreased pro-inflammatory cytokines in pancreatic islets. Collectively, our findings indicate that β-cell GHSR has a major impact on insulin secretion in aging but not obesity, and GHSR deficiency protects against STZ-induced β-cell injury in aging.
    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
    谷胱甘肽(GSH),一种强大的内源性抗氧化剂,积极参与蛋白质中半胱氨酸残基氧化还原状态的调节。先前的研究表明,GSH可以预防由慢性振荡葡萄糖(OsG)给药引起的β细胞衰竭和糖尿病前期。然而,保护作用的确切机制尚不清楚。我们目前的研究表明,GSH能够逆转Nrf2水平的降低,以及下游基因Grx1和HO-1,在慢性OsG诱导的大鼠胰岛β细胞中。体外实验进一步证明GSH可以防止β细胞去分化,凋亡,和OsG引起的胰岛素分泌受损。此外,GSH促进Nrf2易位到细胞核,导致Nrf2靶向基因如GCLC的上调,Grx1、HO-1和NQO1。值得注意的是,当使用Nrf2抑制剂ML385时,GSH对OsG处理的β细胞的作用被取消。此外,在OsG处理的β细胞中,GSH在Cys273和Cys288增强Keap1的S-谷胱甘肽酰化,但不增强Cys151,导致Nrf2从Keap1解离并促进Nrf2核易位。总之,GSH对OsG诱导的β细胞衰竭的保护作用可以部分归因于其增强Keap1S-谷胱甘肽酰化的能力,从而激活Nrf2信号通路。这些发现为糖尿病前期/糖尿病背景下β细胞衰竭的预防和治疗提供了新的见解。强调GSH的潜力。
    Glutathione (GSH), a robust endogenous antioxidant, actively participates in the modulation of the redox status of cysteine residues in proteins. Previous studies have indicated that GSH can prevent β-cell failure and prediabetes caused by chronic oscillating glucose (OsG) administration. However, the precise mechanism underlying the protective effect is not well understood. Our current research reveals that GSH is capable of reversing the reduction in Nrf2 levels, as well as downstream genes Grx1 and HO-1, in the islet β-cells of rats induced by chronic OsG. In vitro experiments have further demonstrated that GSH can prevent β-cell dedifferentiation, apoptosis, and impaired insulin secretion caused by OsG. Additionally, GSH facilitates the translocation of Nrf2 into the nucleus, resulting in an upregulation of Nrf2-targeted genes such as GCLC, Grx1, HO-1, and NQO1. Notably, when the Nrf2 inhibitor ML385 is employed, the effects of GSH on OsG-treated β-cells are abrogated. Moreover, GSH enhances the S-glutathionylation of Keap1 at Cys273 and Cys288, but not Cys151, in OsG-treated β-cells, leading to the dissociation of Nrf2 from Keap1 and facilitating Nrf2 nuclear translocation. In conclusion, the protective role of GSH against OsG-induced β-cell failure can be partially attributed to its capacity to enhance Keap1 S-glutathionylation, thereby activating the Nrf2 signaling pathway. These findings provide novel insights into the prevention and treatment of β-cell failure in the context of prediabetes/diabetes, highlighting the potential of GSH.
    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
    诱导型一氧化氮合酶(iNOS)诱导后以低微摩尔水平产生一氧化氮,并负责介导细胞因子对胰岛葡萄糖刺激的胰岛素分泌的抑制作用。它是通过抑制线粒体氧化代谢,特别是乌头酸酶和电子传输链的复合物4,一氧化氮抑制胰岛素分泌.一氧化氮也减弱蛋白质合成,诱导DNA损伤,激活DNA修复途径,并刺激β细胞的应激反应(未折叠蛋白质和热休克)。在这份报告中,研究了一氧化氮对已知参与β细胞对该自由基反应的6个基因表达的时间和浓度依赖性影响。这些基因包括Gadd45α(DNA修复),Puma(凋亡),Hmox1(抗氧化防御),Hsp70(热休克),Chop(UPR),和βPpargc1α(线粒体生物发生)。我们显示一氧化氮在〜0.5-1µM的狭窄浓度范围内刺激β细胞基因表达,或对应于iNOS来源的一氧化氮的水平。浓度大于1µM时,一氧化氮不能刺激β细胞的基因表达,这与线粒体氧化代谢的抑制有关。这种狭窄的反应浓度范围是β细胞选择性的,作为一氧化氮在非β细胞中的作用(α细胞,小鼠胚胎成纤维细胞,和巨噬细胞)是浓度依赖性的。我们的研究结果表明,β细胞对狭窄浓度范围的一氧化氮作出反应,这与iNOS诱导后产生的水平一致。并且这些浓度依赖性作用对含胰岛素的细胞具有选择性。
    Nitric oxide is produced at low micromolar levels following the induction of inducible nitric oxide synthase (iNOS) and is responsible for mediating the inhibitory actions of cytokines on glucose-stimulated insulin secretion by islets of Langerhans. It is through the inhibition of mitochondrial oxidative metabolism, specifically aconitase and complex 4 of the electron transport chain, that nitric oxide inhibits insulin secretion. Nitric oxide also attenuates protein synthesis, induces DNA damage, activates DNA repair pathways, and stimulates stress responses (unfolded protein and heat shock) in β-cells. In this report, the time- and concentration-dependent effects of nitric oxide on the expression of six genes known to participate in the response of β-cells to this free radical were examined. The genes included Gadd45α (DNA repair), Puma (apoptosis), Hmox1 (antioxidant defense), Hsp70 (heat shock), Chop (UPR), and Ppargc1α (mitochondrial biogenesis). We show that nitric oxide stimulates β-cell gene expression in a narrow concentration range of ∼0.5-1 µM or levels corresponding to iNOS-derived nitric oxide. At concentrations greater than 1 µM, nitric oxide fails to stimulate gene expression in β-cells, and this is associated with the inhibition of mitochondrial oxidative metabolism. This narrow concentration range of responses is β-cell selective, as the actions of nitric oxide in non-β-cells (α-cells, mouse embryonic fibroblasts, and macrophages) are concentration dependent. Our findings suggest that β-cells respond to a narrow concentration range of nitric oxide that is consistent with the levels produced following iNOS induction, and that these concentration-dependent actions are selective for insulin-containing cells.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号