pancreatic islet

胰岛
  • 文章类型: Journal Article
    目的:tRNA在蛋白质合成中发挥重要作用。除了这个规范函数,它们最近被发现产生调节不同细胞活性的非编码RNA片段(tRF)。这项研究的目的是评估tRF在免疫细胞和β细胞之间的串扰中的参与,并研究它们对1型糖尿病发展的贡献。
    方法:通过小RNA-seq对存在于4周龄和8周龄NOD小鼠胰岛中以及由活化的CD4+T淋巴细胞释放的细胞外囊泡中的tRF进行了全局分析。通过定量PCR确认特异性片段水平的变化。使用RNA标记方法评估在胰岛炎期间发生的tRF从免疫细胞向β细胞的转移。在1型糖尿病的初始阶段,tRF在β细胞中增加的功能作用是通过在解离的胰岛细胞中过表达它们并通过确定对基因表达和β细胞凋亡的影响来确定的。
    结果:我们发现,在1型糖尿病的初始阶段,NOD小鼠胰岛中的tRF池发生了改变。这些变化的一部分是由β细胞长时间暴露于促炎细胞因子(IL-1β,TNF-α和IFN-γ),而其他原因是CD4T淋巴细胞浸润胰岛产生的tRF的传递。的确,我们鉴定了几个tRF,这些tRF在CD4+/CD25-T细胞的胞外囊泡中富集,并在NOD中这些免疫细胞过继转移后转移至β细胞.SCID小鼠。在自身免疫反应期间递送至β细胞的tRF触发了影响胰岛素分泌细胞的免疫调节能力并使细胞更易于凋亡的基因表达变化。
    结论:我们的数据表明,tRF是免疫系统和胰岛素分泌细胞之间相互作用的新型参与者,并提示这类新型非编码RNA可能参与1型糖尿病的发病机制。
    方法:序列可从基因表达Omnibus(GEO)获得,登录号为GSE242568和GSE256343。
    OBJECTIVE: tRNAs play a central role in protein synthesis. Besides this canonical function, they were recently found to generate non-coding RNA fragments (tRFs) regulating different cellular activities. The aim of this study was to assess the involvement of tRFs in the crosstalk between immune cells and beta cells and to investigate their contribution to the development of type 1 diabetes.
    METHODS: Global profiling of the tRFs present in pancreatic islets of 4- and 8-week-old NOD mice and in extracellular vesicles released by activated CD4+ T lymphocytes was performed by small RNA-seq. Changes in the level of specific fragments were confirmed by quantitative PCR. The transfer of tRFs from immune cells to beta cells occurring during insulitis was assessed using an RNA-tagging approach. The functional role of tRFs increasing in beta cells during the initial phases of type 1 diabetes was determined by overexpressing them in dissociated islet cells and by determining the impact on gene expression and beta cell apoptosis.
    RESULTS: We found that the tRF pool was altered in the islets of NOD mice during the initial phases of type 1 diabetes. Part of these changes were triggered by prolonged exposure of beta cells to proinflammatory cytokines (IL-1β, TNF-α and IFN-γ) while others resulted from the delivery of tRFs produced by CD4+ T lymphocytes infiltrating the islets. Indeed, we identified several tRFs that were enriched in extracellular vesicles from CD4+/CD25- T cells and were transferred to beta cells upon adoptive transfer of these immune cells in NOD.SCID mice. The tRFs delivered to beta cells during the autoimmune reaction triggered gene expression changes that affected the immune regulatory capacity of insulin-secreting cells and rendered the cells more prone to apoptosis.
    CONCLUSIONS: Our data point to tRFs as novel players in the crosstalk between the immune system and insulin-secreting cells and suggest a potential involvement of this novel class of non-coding RNAs in type 1 diabetes pathogenesis.
    METHODS: Sequences are available from the Gene Expression Omnibus (GEO) with accession numbers GSE242568 and GSE256343.
    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β细胞体积减少是2型糖尿病患者的严重问题。并且需要建立适当的治疗方法。越来越多,钠/葡萄糖协同转运蛋白2(SGLT2)抑制剂,对胰腺β细胞有保护作用,正在被处方治疗糖尿病;然而,潜在的机制还没有很好的理解。我们先前对2型糖尿病小鼠模型施用SGLT2抑制剂达格列净,发现早期治疗组中基因表达发生显著变化,这导致我们假设表观遗传调控是这些变化的可能机制。因此,我们在糖尿病模型小鼠服用达帕格列净后,使用分离的胰岛,通过甲基化DNA免疫沉淀法进行了全面的DNA甲基化分析.因此,我们鉴定出31个由于DNA甲基化改变导致表达改变的基因.免疫染色后,在达格列净治疗组中,发现囊性纤维化跨膜传导调节因子和钙黏着蛋白24在胰岛中上调。这些分子可能有助于胰岛形态和胰岛素分泌能力的维持,表明SGLT2抑制剂对胰腺β细胞的保护作用伴随着DNA甲基化的变化,这种影响是长期的,而不是暂时的。在未来的糖尿病护理中,SGLT2抑制剂有望产生积极的治疗效果,包括胰腺β细胞保护。
    Decreased pancreatic β-cell volume is a serious problem in patients with type 2 diabetes mellitus, and there is a need to establish appropriate treatments. Increasingly, sodium/glucose cotransporter 2 (SGLT2) inhibitors, which have a protective effect on pancreatic β-cells, are being prescribed to treat diabetes; however, the underlying mechanism is not well understood. We previously administered SGLT2 inhibitor dapagliflozin to a mouse model of type 2 diabetes and found significant changes in gene expression in the early-treated group, which led us to hypothesize that epigenetic regulation was a possible mechanism of these changes. Therefore, we performed comprehensive DNA methylation analysis by methylated DNA immunoprecipitation using isolated pancreatic islets after dapagliflozin administration to diabetic model mice. As a result, we identified 31 genes with changes in expression due to DNA methylation changes. Upon immunostaining, cystic fibrosis transmembrane conductance regulator and cadherin 24 were found to be upregulated in islets in the dapagliflozin-treated group. These molecules may contribute to the maintenance of islet morphology and insulin secretory capacity, suggesting that SGLT2 inhibitors\' protective effect on pancreatic β-cells is accompanied by DNA methylation changes, and that the effect is long-term and not temporary. In future diabetes care, SGLT2 inhibitors may be expected to have positive therapeutic effects, including pancreatic β-cell protection.
    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
    糖尿病是一种常见的慢性疾病,传统上需要严重依赖药物治疗。口服药物和外源性胰岛素只能暂时维持血糖水平,不能治愈疾病。大多数患者需要终身注射外源性胰岛素。近年来,胰岛移植的进步显著推进了糖尿病的治疗,允许患者停止外源性胰岛素并避免并发症。最近关于胰岛移植的报道的长期随访结果表明,尽管患者仍然需要免疫疗法,但它们提供了显着的治疗益处。表明未来移植策略的重要性。尽管器官短缺仍然是胰岛移植发展的主要障碍,胰岛细胞的新来源,如干细胞和猪胰岛细胞,已经被提议,并逐步纳入临床研究。进一步研究新的移植部位,例如皮下空间和肠系膜脂肪,可能最终取代传统的门静脉内胰岛细胞输注。此外,胰岛移植中的免疫排斥反应将通过联合应用免疫抑制剂来解决,胰岛封装技术,以及最有前途的间充质干细胞/调节性T细胞和胰岛细胞联合移植细胞治疗。本文综述了胰岛移植的研究进展,并讨论了所面临挑战的研究进展和潜在解决方案。
    Diabetes is a prevalent chronic disease that traditionally requires severe reliance on medication for treatment. Oral medication and exogenous insulin can only temporarily maintain blood glucose levels and do not cure the disease. Most patients need life-long injections of exogenous insulin. In recent years, advances in islet transplantation have significantly advanced the treatment of diabetes, allowing patients to discontinue exogenous insulin and avoid complications.Long-term follow-up results from recent reports on islet transplantation suggest that they provide significant therapeutic benefit although patients still require immunotherapy, suggesting the importance of future transplantation strategies. Although organ shortage remains the primary obstacle for the development of islet transplantation, new sources of islet cells, such as stem cells and porcine islet cells, have been proposed, and are gradually being incorporated into clinical research. Further research on new transplantation sites, such as the subcutaneous space and mesenteric fat, may eventually replace the traditional portal vein intra-islet cell infusion. Additionally, the immunological rejection reaction in islet transplantation will be resolved through the combined application of immunosuppressant agents, islet encapsulation technology, and the most promising mesenchymal stem cells/regulatory T cell and islet cell combined transplantation cell therapy. This review summarizes the progress achieved in islet transplantation, and discusses the research progress and potential solutions to the challenges faced.
    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
    胰岛移植可能是1型糖尿病(T1DM)患者最有效的治疗技术。然而,这种方法的临床应用面临着许多限制,包括孤立的胰岛细胞凋亡,收件人拒绝,和移植血管重建。间充质干细胞(MSCs)具有抗凋亡,免疫调节,和血管生成特性。这里,我们回顾了最近关于胰岛与MSCs共培养和共移植的研究。我们总结了共同移植的制备方法,特别是共同文化的优点,以及共同移植的效果。积累的实验证据表明,胰岛与MSCs共培养促进胰岛存活,增强胰岛分泌功能,并通过各种移植前的准备来促进胰岛的生长。本文旨在为探索MSCs在临床胰岛共移植中的应用提供参考。
    Islet transplantation may be the most efficient therapeutic technique for patients with type 1 diabetes mellitus (T1DM). However, the clinical application of this method is faced with numerous limitations, including isolated islet apoptosis, recipient rejection, and graft vascular reconstruction. Mesenchymal stem cells (MSCs) possess anti-apoptotic, immunomodulatory, and angiogenic properties. Here, we review recent studies on co-culture and co-transplantation of islets with MSCs. We have summarized the methods of preparation of co-transplantation, especially the merits of co-culture, and the effects of co-transplantation. Accumulating experimental evidence shows that co-culture of islets with MSCs promotes islet survival, enhances islet secretory function, and prevascularizes islets through various pretransplant preparations. This review is expected to provide a reference for exploring the use of MSCs for clinical islet co-transplantation.
    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
    初级纤毛上蛋白质定位的明确证明一直是纤毛生物学家的挑战。初级纤毛是孤立的线状突起,具有特殊的蛋白质组成,但是纤毛结构覆盖细胞膜和其他细胞部分,纤毛蛋白的身份很难通过常规成像方法如免疫荧光显微镜来确定。表面扫描电子显微镜与免疫标记(免疫SEM)相结合,通过在纤毛的三维超微结构的背景下明确显示蛋白质表达,绕过了其中一些不确定性。这里,我们应用免疫SEM特异性鉴定小鼠和人胰岛初级纤毛上的蛋白质,包括翻译后修饰的微管蛋白,步内运输(IFT)88,小GTP酶Arl13b,以及轴突动力蛋白的亚基。样品制备中的关键参数,讨论了免疫标记和成像采集,以促进纤毛研究界其他人的类似研究。
    The definitive demonstration of protein localization on primary cilia has been a challenge for cilia biologists. Primary cilia are solitary thread-like projections that have a specialized protein composition, but as the ciliary structure overlays the cell membrane and other cell parts, the identity of ciliary proteins are difficult to ascertain by conventional imaging approaches like immunofluorescence microscopy. Surface scanning electron microscopy combined with immunolabeling (immuno-SEM) bypasses some of these indeterminacies by unambiguously showing protein expression in the context of the three-dimensional ultrastructure of the cilium. Here, we apply immuno-SEM to specifically identify proteins on the primary cilia of mouse and human pancreatic islets, including post-translationally modified tubulin, intraflagellar transport (IFT)88, the small GTPase Arl13b, as well as subunits of axonemal dynein. Key parameters in sample preparation, immunolabeling and imaging acquisition are discussed to facilitate similar studies by others in the cilia research community.
    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
    胰岛被ECM包围,ECM为胰岛β细胞提供生化和机械提示,以调节细胞存活和胰岛素分泌。ECM组成和机械特性的变化导致许多胰腺疾病中的β细胞功能障碍。虽然一些研究已经描述了胰岛胰岛素分泌随底物硬度变化的变化,对机械传导信号驱动改变的胰岛功能,以响应机械提示,知之甚少。我们假设增加基质硬度将通过打开机械敏感性离子通道Piezo1并破坏小鼠和人胰岛中的细胞内Ca2动力学而导致胰岛素分泌功能障碍。为了检验我们的假设,将小鼠和人尸体胰岛封装在仿生反向热凝胶(RTG)支架中,该支架具有可调整的刚度,可以与支架形成胰岛斑粘附并在3D中激活Piezo1。我们的结果表明,增加的支架刚度会导致胰岛素分泌功能障碍,这是通过打开机械敏感性Piezo1通道来增加Ca2内流和改变Ca2动力学而介导的。此外,抑制Piezo1挽救了坚硬支架中胰岛中葡萄糖刺激的胰岛素分泌(GSIS)。总的来说,我们的结果强调了胰岛微环境的机械性能在调节功能中的作用。它还支持进一步研究Piezo1通道活性的调节,以恢复2型糖尿病(T2D)和胰腺癌等疾病中的胰岛功能,其中胰岛周围ECM的纤维化导致组织僵硬和胰岛功能障碍增加。
    The pancreatic islet is surrounded by ECM that provides both biochemical and mechanical cues to the islet β-cell to regulate cell survival and insulin secretion. Changes in ECM composition and mechanical properties drive β-cell dysfunction in many pancreatic diseases. While several studies have characterized changes in islet insulin secretion with changes in substrate stiffness, little is known about the mechanotransduction signaling driving altered islet function in response to mechanical cues. We hypothesized that increasing matrix stiffness will lead to insulin secretion dysfunction by opening the mechanosensitive ion channel Piezo1 and disrupting intracellular Ca2+ dynamics in mouse and human islets. To test our hypothesis, mouse and human cadaveric islets were encapsulated in a biomimetic reverse thermal gel (RTG) scaffold with tailorable stiffness that allows formation of islet focal adhesions with the scaffold and activation of Piezo1 in 3D. Our results indicate that increased scaffold stiffness causes insulin secretion dysfunction mediated by increases in Ca2+ influx and altered Ca2+ dynamics via opening of the mechanosensitive Piezo1 channel. Additionally, inhibition of Piezo1 rescued glucose-stimulated insulin secretion (GSIS) in islets in stiff scaffolds. Overall, our results emphasize the role mechanical properties of the islet microenvironment plays in regulating function. It also supports further investigation into the modulation of Piezo1 channel activity to restore islet function in diseases like type 2 diabetes (T2D) and pancreatic cancer where fibrosis of the peri-islet ECM leads to increased tissue stiffness and islet dysfunction.
    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
    在肥胖相关的2型糖尿病(T2D)中,解决胰岛素分泌过多和胰岛素抵抗的上游是具有挑战性的。这里,我们考虑了糖尿病母亲胎儿胰岛素分泌和作用的动力学。我们认为胎儿胰岛素分泌过多首先发生,胰岛素抵抗是一种适应性保护性反应。
    Disentangling which of insulin hypersecretion and insulin resistance is upstream in obesity-related type 2 diabetes (T2D) is challenging. Here, we consider the dynamics of insulin secretion and action in the fetuses of mothers with diabetes. We argue that fetal insulin hypersecretion occurs first, with insulin resistance being an adaptive protective response.
    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
    通过前列腺素E2EP3受体(EP3)的信号传导积极促进2型糖尿病(T2D)的β细胞功能障碍。在T2D模型中,由于胰腺外组织中EP3缺失导致的饮食过多和严重的胰岛素抵抗,全身EP3敲除小鼠的代谢表型比野生型对照明显更差。掩盖β细胞中EP3损失的任何潜在有益作用。我们假设β细胞特异性EP3基因敲除(EP3βKO)小鼠将被保护免受高脂饮食(HFD)诱导的葡萄糖不耐受,缺乏EP3效应子的表型小鼠,Gαz,其组织分布更为有限。当喂HFD16周时,虽然,EP3βKO小鼠部分,但不完全,免受葡萄糖不耐受。此外,exendin-4是肠促胰岛素激素的类似物,胰高血糖素样肽1,与野生型对照相比,对照饮食和HFD喂养的EP3βKO小鼠胰岛中葡萄糖刺激的胰岛素分泌更强,β细胞特异性EP3损失对胰岛胰岛素含量或复制和存活标志物没有影响。然而,经过26周的饮食喂养,来自对照饮食和HFD喂养的EP3βKO小鼠的胰岛分泌的胰岛素含量百分比显着减少,有或没有exendin-4,与β细胞复制和存活标记无关的总胰岛素含量升高,显示严重的β细胞功能障碍。我们的结果表明,EP3在T2D进展过程中在时间上调节β细胞功能中起关键作用,并且其作用背后存在不依赖Gαz的机制。EP3受体是β细胞功能和复制的强抑制剂,表明它是该疾病的潜在治疗靶点。然而,EP3在胰腺外组织中具有保护作用。为了解决这个问题,我们设计了β细胞特异性EP3基因敲除小鼠,并对其进行高脂饮食喂养以诱导葡萄糖不耐受。全身基因敲除小鼠的负代谢表型被消融,和EP3损失改善葡萄糖耐量,对胰岛胰岛素分泌和含量有相反的影响。
    Signaling through prostaglandin E2 EP3 receptor (EP3) actively contributes to the β-cell dysfunction of type 2 diabetes (T2D). In T2D models, full-body EP3 knockout mice have a significantly worse metabolic phenotype than wild-type controls due to hyperphagia and severe insulin resistance resulting from loss of EP3 in extra-pancreatic tissues, masking any potential beneficial effects of EP3 loss in the β cell. We hypothesized β-cell-specific EP3 knockout (EP3 βKO) mice would be protected from high-fat diet (HFD)-induced glucose intolerance, phenocopying mice lacking the EP3 effector, Gαz, which is much more limited in its tissue distribution. When fed a HFD for 16 wk, though, EP3 βKO mice were partially, but not fully, protected from glucose intolerance. In addition, exendin-4, an analog of the incretin hormone, glucagon-like peptide 1, more strongly potentiated glucose-stimulated insulin secretion in islets from both control diet- and HFD-fed EP3 βKO mice as compared with wild-type controls, with no effect of β-cell-specific EP3 loss on islet insulin content or markers of replication and survival. However, after 26 wk of diet feeding, islets from both control diet- and HFD-fed EP3 βKO mice secreted significantly less insulin as a percent of content in response to stimulatory glucose, with or without exendin-4, with elevated total insulin content unrelated to markers of β-cell replication and survival, revealing severe β-cell dysfunction. Our results suggest that EP3 serves a critical role in temporally regulating β-cell function along the progression to T2D and that there exist Gαz-independent mechanisms behind its effects.NEW & NOTEWORTHY The EP3 receptor is a strong inhibitor of β-cell function and replication, suggesting it as a potential therapeutic target for the disease. Yet, EP3 has protective roles in extrapancreatic tissues. To address this, we designed β-cell-specific EP3 knockout mice and subjected them to high-fat diet feeding to induce glucose intolerance. The negative metabolic phenotype of full-body knockout mice was ablated, and EP3 loss improved glucose tolerance, with converse effects on islet insulin secretion and content.
    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
    糖尿病不仅是一种内分泌疾病,也是一种血管疾病。血管缺陷通常被视为糖尿病的结果。然而,在胰岛的水平,在症状发作之前已经描述了血管改变。重要的是,这些早期血管缺陷的细胞和分子机制尚未确定,这些都不会影响胰岛内分泌细胞的功能。在这次审查中,我们将讨论微脉管系统的壁细胞-称为周细胞-的功能障碍是在症状前阶段在胰岛中观察到的血管缺陷的基础。周细胞对于整个身体的血管稳态至关重要,但是它们在胰岛中的生理和病理生理功能直到最近才开始被探索。先前的一项研究已经引起了对这种疾病的“微血管”方法的兴趣。随着我们对胰岛微脉管系统对葡萄糖稳态的关键作用的理解,在这里,我们将重新审视胰岛功能的血管方面,以及它们的失调如何导致糖尿病的发病机制,特别关注1型糖尿病(T1D)。
    Diabetes is not only an endocrine but also a vascular disease. Vascular defects are usually seen as consequence of diabetes. However, at the level of the pancreatic islet, vascular alterations have been described before symptom onset. Importantly, the cellular and molecular mechanisms underlying these early vascular defects have not been identified, neither how these could impact the function of islet endocrine cells. In this review, we will discuss the possibility that dysfunction of the mural cells of the microvasculature-known as pericytes-underlies vascular defects observed in islets in pre-symptomatic stages. Pericytes are crucial for vascular homeostasis throughout the body, but their physiological and pathophysiological functions in islets have only recently started to be explored. A previous study had already raised interest in the \"microvascular\" approach to this disease. With our increased understanding of the crucial role of the islet microvasculature for glucose homeostasis, here we will revisit the vascular aspects of islet function and how their deregulation could contribute to diabetes pathogenesis, focusing in particular on type 1 diabetes (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
    长链非编码RNA(lncRNA)介导的基因调控的转录后和表观遗传景观与许多人类疾病有关。然而,控制人类β细胞功能和存活的调节机制仍然未知。由于技术和道德方面的限制,研究lncRNAs在人体内β细胞功能和存活中的直接作用是困难的。因此,我们利用人类胰岛的人源化小鼠,使用全转录组鸟枪测序研究lncRNA表达。我们的研究旨在表征可能对人类胰岛细胞功能和存活至关重要的lncRNAs。
    在移植有功能性人胰岛的人源化小鼠中诱导人β细胞死亡。使用这些带有诱导β细胞死亡的人类胰岛的人源化小鼠,我们通过全转录组鸟枪测序研究了lncRNA的表达。此外,我们系统地确定,characterized,并探索了对人类胰岛细胞功能和存活潜在重要的lncRNAs的调控功能。
    在移植有功能性人胰岛的人源化小鼠中诱导人胰岛细胞死亡。分离的人胰岛的RNA测序分析,来自有和没有诱导细胞死亡的人源化小鼠的胰岛移植物,揭示了一组不同的lncRNAs的异常表达,这些lncRNAs与对β细胞功能和存活相关的细胞过程和分子通路重要的去调节的mRNAs相关。总共10种lncRNA同工型(SCYL1-1:22,POLG2-1:1,CTRB1-1:1,SRPK1-1:1,GTF3C5-1:1,PPY-1:1,CTRB1-1:5,CPA5-1:1,BCAR1-2:1和CTRB1-1:4)被鉴定为高度富集的,对人类胰岛这些lncRNAs在来自不同BMI和2型糖尿病(T2D)供体的人类胰岛中失调,以及在具有葡萄糖刺激和细胞因子诱导的诱导细胞死亡的培养的人胰岛中。在来自用于用细胞因子培养胰岛的培养基的外来体中检测到这些lncRNA的异常表达。
    富含胰岛和特异性的人lncRNAs在人胰岛移植物和培养的人胰岛中失调,诱导细胞死亡。这些lncRNAs可能对于人β细胞功能和存活至关重要,并且可能对鉴定β细胞丢失的生物标志物和发现新的治疗靶标以增强β细胞功能和存活产生影响。
    Long noncoding RNA (lncRNA)-mediated posttranscriptional and epigenetic landscapes of gene regulation are associated with numerous human diseases. However, the regulatory mechanisms governing human β-cell function and survival remain unknown. Owing to technical and ethical constraints, studying the direct role of lncRNAs in β-cell function and survival in humans in vivo is difficult. Therefore, we utilized humanized mice with human islets to investigate lncRNA expression using whole transcriptome shotgun sequencing. Our study aimed to characterize lncRNAs that may be crucial for human islet cell function and survival.
    Human β-cell death was induced in humanized mice engrafted with functional human islets. Using these humanized mice harboring human islets with induced β-cell death, we investigated lncRNA expression through whole transcriptome shotgun sequencing. Additionally, we systematically identified, characterized, and explored the regulatory functions of lncRNAs that are potentially important for human pancreatic islet cell function and survival.
    Human islet cell death was induced in humanized mice engrafted with functional human islets. RNA sequencing analysis of isolated human islets, islet grafts from humanized mice with and without induced cell death, revealed aberrant expression of a distinct set of lncRNAs that are associated with the deregulated mRNAs important for cellular processes and molecular pathways related to β-cell function and survival. A total of 10 lncRNA isoforms (SCYL1-1:22, POLG2-1:1, CTRB1-1:1, SRPK1-1:1, GTF3C5-1:1, PPY-1:1, CTRB1-1:5, CPA5-1:1, BCAR1-2:1, and CTRB1-1:4) were identified as highly enriched and specific to human islets. These lncRNAs were deregulated in human islets from donors with different BMIs and with type 2 diabetes (T2D), as well as in cultured human islets with glucose stimulation and induced cell death induced by cytokines. Aberrant expression of these lncRNAs was detected in the exosomes from the medium used to culture islets with cytokines.
    Islet-enriched and specific human lncRNAs are deregulated in human islet grafts and cultured human islets with induced cell death. These lncRNAs may be crucial for human β-cell function and survival and could have an impact on identifying biomarkers for β-cell loss and discovering novel therapeutic targets to enhance β-cell function and survival.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号