pancreatic islet

胰岛
  • 文章类型: Journal Article
    我们研究了内质网(ER)应激调节的长链非编码RNA(lncRNA)lncMGC在胰岛和1型糖尿病(T1D)病理中的作用。以及基于lncMGC的疗法的潜力。在体内,与野生型STZ相比,lncMGC敲除(KO)-链脲佐菌素(STZ)治疗的糖尿病小鼠的血糖水平(BGL)和HbA1c显著降低.与GapmeR阴性对照(NC)相比,靶向lncMGC的反义寡核苷酸(GapmeR)显著减弱TlDNOD小鼠中的胰岛炎和BGL。与Akita-NC小鼠相比,注射GapmeR的TlDAkita小鼠显示出显著更低的BGL。与NC注射的小鼠相比,hlncMGC-GapmeR在部分人源化的lncMGC(hlncMGC)-STZ小鼠中降低BGL。CHOP(ER应激调节转录因子)和lncMGC在糖尿病小鼠的胰岛中上调,但在lncMGC-KO和GapmeR注射的糖尿病小鼠中没有上调,表明ER压力参与。体外,hlncMGC-GapmeR增加了来自人类供体和hlncMGC小鼠的分离胰岛的活力,并保护它们免受细胞因子诱导的凋亡。抗内质网应激和抗凋亡基因上调,但是促凋亡基因在lncMGCKO小鼠胰岛和GapmeR处理的人胰岛中下调。一起来看,这些结果表明,GapmeR靶向lncMGC可有效改善小鼠的糖尿病,并保留人类和小鼠的胰岛活力,暗示临床翻译潜力。
    We investigated the role of the endoplasmic reticulum (ER) stress-regulated long noncoding RNA (lncRNA) lncMGC in pancreatic islets and the pathology of type 1 diabetes (T1D), as well as the potential of lncMGC-based therapeutics. In vivo, blood glucose levels (BGLs) and HbA1c were significantly lower in lncMGC-knockout (KO)-streptozotocin (STZ)-treated diabetic mice compared to wild-type STZ. Antisense oligonucleotides (GapmeR) targeting lncMGC significantly attenuated insulitis and BGLs in T1D NOD mice compared to GapmeR-negative control (NC). GapmeR-injected T1D Akita mice showed significantly lower BGLs compared to Akita-NC mice. hlncMGC-GapmeR lowered BGLs in partially humanized lncMGC (hlncMGC)-STZ mice compared to NC-injected mice. CHOP (ER stress regulating transcription factor) and lncMGC were upregulated in islets from diabetic mice but not in lncMGC-KO and GapmeR-injected diabetic mice, suggesting ER stress involvement. In vitro, hlncMGC-GapmeR increased the viability of isolated islets from human donors and hlncMGC mice and protected them from cytokine-induced apoptosis. Anti-ER stress and anti-apoptotic genes were upregulated, but pro-apoptotic genes were down-regulated in lncMGC KO mice islets and GapmeR-treated human islets. Taken together, these results show that a GapmeR-targeting lncMGC is effective in ameliorating diabetes in mice and also preserves human and mouse islet viability, implicating clinical translation potential.
    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:本研究旨在评估为分离α,beta,和成年小鼠胰岛的δ细胞,将其应用扩展到新生和老年小鼠的胰岛。此外,它试图检查小鼠胰腺内分泌胰岛细胞在整个出生后发育过程中的转录组动力学,并验证这些细胞群体中与年龄相关的改变。
    方法:我们利用β细胞上CD71和δ细胞上CD24的高表面表达来FACS纯化α,beta,和来自新生儿(1周龄)的三角洲细胞,成人(12周龄),和老(18个月大)小鼠。对这些纯化的细胞群体进行了大量RNA测序,随后的生物信息学分析包括差异基因表达,代表性过高,和交叉分析。
    结果:Alpha,beta,使用与成年小鼠相同的方法成功地FACS纯化了新生和老年小鼠的δ细胞。我们对α的年龄相关转录变化的分析,beta,和δ细胞群显示,在从新生小鼠到成年小鼠的过渡过程中,细胞周期减少,神经元样特征过程增加。从成年小鼠发展到老年小鼠,我们确定了与衰老(炎症)相关的炎症基因特征,包括β-2微球蛋白和主要组织相容性复合体(MHC)I类表达的增加.
    结论:我们的研究证明了我们的细胞分选技术在从不同年龄的小鼠胰岛中纯化内分泌亚群的有效性。我们为更好地了解内分泌胰腺衰老提供了宝贵的资源,并确定了具有β-2微球蛋白和MHCI类表达增加的炎症基因标签作为旧α的常见标志,beta,和三角洲细胞,与免疫反应调节和年龄相关的糖尿病的潜在影响。
    OBJECTIVE: This study aimed to evaluate the efficacy of a purification method developed for isolating alpha, beta, and delta cells from pancreatic islets of adult mice, extending its application to islets from newborn and aged mice. Furthermore, it sought to examine transcriptome dynamics in mouse pancreatic endocrine islet cells throughout postnatal development and to validate age-related alterations within these cell populations.
    METHODS: We leveraged the high surface expression of CD71 on beta cells and CD24 on delta cells to FACS-purify alpha, beta, and delta cells from newborn (1-week-old), adult (12-week-old), and old (18-month-old) mice. Bulk RNA sequencing was conducted on these purified cell populations, and subsequent bioinformatic analyses included differential gene expression, overrepresentation, and intersection analysis.
    RESULTS: Alpha, beta, and delta cells from newborn and aged mice were successfully FACS-purified using the same method employed for adult mice. Our analysis of the age-related transcriptional changes in alpha, beta, and delta cell populations revealed a decrease in cell cycling and an increase in neuron-like features processes during the transition from newborn to adult mice. Progressing from adult to old mice, we identified an inflammatory gene signature related to aging (inflammaging) encompassing an increase in β-2 microglobulin and major histocompatibility complex (MHC) Class I expression.
    CONCLUSIONS: Our study demonstrates the effectiveness of our cell sorting technique in purifying endocrine subsets from mouse islets at different ages. We provide a valuable resource for better understanding endocrine pancreas aging and identified an inflammaging gene signature with increased β-2 microglobulin and MHC Class I expression as a common hallmark of old alpha, beta, and delta cells, with potential implications for immune response regulation and age-related 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
    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
    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)

  • 文章类型: Journal Article
    功能不足的β细胞量是2型糖尿病(T2D)的核心病理标志。尽管有几种有效的糖尿病管理药物,在糖尿病状态下,迫切需要新的药物来保护胰腺β细胞。综合器官交叉交流控制能量平衡和葡萄糖稳态。肝脏和胰岛具有动态的交叉通讯,其中肝脏可以在胰岛素抵抗条件下引发代偿性β细胞团扩增并增强激素分泌。然而,促进β细胞增殖和胰岛素分泌的不可缺少的元素尚未完全确定.外泌体是大多数细胞类型释放的重要的细胞外载体(EV),传递生物信号。包括代谢信使,如miRNA和肽,细胞和器官之间。
    我们研究了β细胞是否可以吸收肝脏来源的外泌体,并检查了它们对β细胞功能基因和胰岛素表达的影响。使用各种方法表征从人肝HepG2细胞分离的外泌体,包括透射电子显微镜(TEM),动态光散射(DLS),和外泌体标记的蛋白质印迹分析。使用CM-Dil染料评估外泌体标记和细胞摄取。肝细胞来源的外泌体对Min6β细胞的影响通过使用qPCR的β细胞标志物和胰岛素的基因表达分析来确定,以及使用蛋白质印迹的Akt信号。
    用胰岛素受体拮抗剂S961处理的人肝脏HepG2细胞中分离的外泌体处理Min6β细胞与从未处理的细胞中分离的外泌体相比显著增加了β细胞标志物Pdx1、NeuroD1和Ins1的表达。与此相符,AKT激酶的活性,胰岛素受体途径的组成部分,胰腺β细胞升高,以AKT下游底物的增加为代表,FoxO1磷酸化。
    这项研究表明,肝脏来源的外泌体可能携带特定的分子货物,可以影响胰腺β细胞中胰岛素的表达,最终影响葡萄糖稳态。
    An insufficient functional beta cell mass is a core pathological hallmark of type 2 diabetes (T2D). Despite the availability of several effective pharmaceuticals for diabetes management, there is an urgent need for novel medications to protect pancreatic beta cells under diabetic conditions. Integrative organ cross-communication controls the energy balance and glucose homeostasis. The liver and pancreatic islets have dynamic cross-communications where the liver can trigger a compensatory beta cell mass expansion and enhanced hormonal secretion in insulin-resistant conditions. However, the indispensable element(s) that foster beta cell proliferation and insulin secretion have yet to be completely identified. Exosomes are important extracellular vehicles (EVs) released by most cell types that transfer biological signal(s), including metabolic messengers such as miRNA and peptides, between cells and organs.
    We investigated whether beta cells can take up liver-derived exosomes and examined their impact on beta cell functional genes and insulin expression. Exosomes isolated from human liver HepG2 cells were characterized using various methods, including Transmission Electron Microscopy (TEM), dynamic light scattering (DLS), and Western blot analysis of exosomal markers. Exosome labeling and cell uptake were assessed using CM-Dil dye. The effect of liver cell-derived exosomes on Min6 beta cells was determined through gene expression analyses of beta cell markers and insulin using qPCR, as well as Akt signaling using Western blotting.
    Treatment of Min6 beta cells with exosomes isolated from human liver HepG2 cells treated with insulin receptor antagonist S961 significantly increased the expression of beta cell markers Pdx1, NeuroD1, and Ins1 compared to the exosomes isolated from untreated cells. In line with this, the activity of AKT kinase, an integral component of the insulin receptor pathway, is elevated in pancreatic beta cells, as represented by an increase in AKT\'s downstream substrate, FoxO1 phosphorylation.
    This study suggests that liver-derived exosomes may carry a specific molecular cargo that can affect insulin expression in pancreatic beta cells, ultimately affecting glucose homeostasis.
    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
    GIP_HUMAN[22-51]是最近发现的肽,其与葡萄糖依赖性促胰岛素多肽(GIP)共享相同的前体分子。在体内,在ApoE-/-小鼠中长期输注GIP_HUMAN[22-51]可促进主动脉粥样硬化病变的发展,并上调炎症和致动脉粥样硬化蛋白。在本研究中,我们评估了GIP_HUMAN[22-51]对胰岛素分泌INS-1E细胞和离体大鼠胰岛中胰岛素mRNA表达和分泌的影响。此外,我们描述了GIP_HUMAN[22-51]对细胞增殖和死亡以及NF-kB核易位的影响。大鼠产生胰岛素的INS-1E细胞和胰岛,从雄性Wistar大鼠中分离出来,在这项研究中使用。使用实时PCR评估基因表达。使用BrdU掺入测定法研究细胞增殖。通过评估组蛋白复合的DNA片段来定量细胞死亡。使用ELISA测试确定胰岛素分泌。使用免疫荧光检测NF-kB核易位。GIP_HUMAN[22-51]抑制INS-1E细胞和胰岛中的胰岛素(Ins1和Ins2)。此外,GIP_HUMAN[22-51]促进NF-κB从细胞质向细胞核的易位。在存在NF-κB的药理学抑制剂的情况下,GIP_HUMAN[22-51]不能抑制Ins2mRNA表达。此外,GIP_HUMAN[22-51]在低(2.8mmol/L)但不高(16.7mmol/L)葡萄糖浓度下下调胰岛素分泌。相比之下,GIP_HUMAN[22-51]未能影响细胞增殖和凋亡。我们得出结论,GIP_HUMAN[22-51]抑制胰腺β细胞中胰岛素的表达和分泌,而不影响β细胞的增殖或凋亡。值得注意的是,GIP_HUMAN[22-51]对胰岛素分泌的影响是葡萄糖依赖性的。
    GIP_HUMAN [22-51] is a recently discovered peptide that shares the same precursor molecule with glucose-dependent insulinotropic polypeptide (GIP). In vivo, chronic infusion of GIP_HUMAN [22-51] in ApoE-/- mice enhanced the development of aortic atherosclerotic lesions and upregulated inflammatory and proatherogenic proteins. In the present study, we evaluate the effects of GIP_HUMAN [22-51] on insulin mRNA expression and secretion in insulin-producing INS-1E cells and isolated rat pancreatic islets. Furthermore, we characterize the influence of GIP_HUMAN [22-51] on cell proliferation and death and on Nf-kB nuclear translocation. Rat insulin-producing INS-1E cells and pancreatic islets, isolated from male Wistar rats, were used in this study. Gene expression was evaluated using real-time PCR. Cell proliferation was studied using a BrdU incorporation assay. Cell death was quantified by evaluating histone-complexed DNA fragments. Insulin secretion was determined using an ELISA test. Nf-kB nuclear translocation was detected using immunofluorescence. GIP_HUMAN [22-51] suppressed insulin (Ins1 and Ins2) in INS-1E cells and pancreatic islets. Moreover, GIP_HUMAN [22-51] promoted the translocation of NF-κB from cytoplasm to the nucleus. In the presence of a pharmacological inhibitor of NF-κB, GIP_HUMAN [22-51] was unable to suppress Ins2 mRNA expression. Moreover, GIP_HUMAN [22-51] downregulated insulin secretion at low (2.8 mmol/L) but not high (16.7 mmol/L) glucose concentration. By contrast, GIP_HUMAN [22-51] failed to affect cell proliferation and apoptosis. We conclude that GIP_HUMAN [22-51] suppresses insulin expression and secretion in pancreatic β cells without affecting β cell proliferation or apoptosis. Notably, the effects of GIP_HUMAN [22-51] on insulin secretion are glucose-dependent.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号