pancreatic islet

胰岛
  • 文章类型: Journal Article
    脂质纳米颗粒(LNP)最近已被用作核酸药物的药物递送系统中的纳米载体。它们的实际应用目前主要限于肝脏和特定器官。然而,改变磷脂的类型和组成比例改善了它们在肝脏以外的器官中的分布,比如脾和肺。本研究旨在阐明LNP组分和颗粒大小对体内分布的影响,通过体循环到胰岛,以实现更好的胰岛靶向。这是糖尿病的基本治疗目标。使用三种磷脂制备荧光标记的LNP:1,2-二硬脂酰-sn-甘油-3-磷酸胆碱(DSPC),1,2-二油酰基-sn-甘油-3-磷酸胆碱(DOPC),和1,2-二油酰基-sn-甘油基-3-磷酸乙醇胺(DOPE),使用微流体装置的颗粒尺寸为30-160nm(直径)。使用具有调节的流速比和总流速的挡板结构iLiNP装置。在对C57BL/6J小鼠静脉内施用LNP后,每种LNP类型在主要器官中的分布,包括胰腺和胰岛,使用离体荧光成像和胰腺组织切片观察进行比较。DSPC-LNPs和DOPE-LNPs在脾脏和肝脏中分布最高,分别。相比之下,DOPC-LNP在胰腺中分布最高,在肝脏和脾脏中分布最低。此外,较小的颗粒在整个胰腺中表现出更好的分布。对于粒径为160nm的DOPC-LNP,观察到胰岛中最显著的LNP分布。此外,较大的LNP倾向于分布在胰岛中,而较小的LNP倾向于分布在外分泌腺中。DOPC-LNP在所有胆固醇浓度下分布在胰岛中,在>40%胆固醇和>3%PEG时观察到高分布,并且在24小时时的分布高于在4小时时的分布。因此,LNP组成和粒径显著影响胰岛分布特征,表明DOPC-LNP可能是有效靶向胰腺和胰岛的药物递送系统。
    Lipid nanoparticles (LNPs) have recently been used as nanocarriers in drug delivery systems for nucleic acid drugs. Their practical applications are currently primarily limited to the liver and specific organs. However, altering the type and composition ratio of phospholipids improves their distribution in organs other than the liver, such as the spleen and lungs. This study aimed to elucidate the effects of LNP components and particle size on in vivo distribution through systemic circulation to pancreatic islets to achieve better targeting of islets, which are a fundamental therapeutic target for diabetes. Fluorescence-labeled LNPs were prepared using three phospholipids: 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC), 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC), and 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE), with particle sizes of 30-160 nm (diameter) using a microfluidic device. Baffled-structured iLiNP devices with adjusted flow-rate ratios and total flow rates were used. After the intravenous administration of LNPs to C57BL/6 J mice, the distribution of each LNP type to the major organs, including the pancreas and pancreatic islets, was compared using ex vivo fluorescence imaging and observation of pancreatic tissue sections. DSPC-LNPs- and DOPE-LNPs showed the highest distribution in the spleen and liver, respectively. In contrast, the DOPC-LNPs showed the highest distribution in the pancreas and the lowest distribution in the liver and spleen. In addition, smaller particles showed better distribution throughout the pancreas. The most significant LNP distribution in the islets was observed for DOPC-LNPs with a particle size of 160 nm. Furthermore, larger LNPs tended to be distributed in the islets, whereas smaller LNPs tended to be distributed in the exocrine glands. DOPC-LNPs were distributed in the islets at all cholesterol concentrations, with a high distribution observed at >40% cholesterol and > 3% PEG and the distribution was higher at 24 h than at 4 h. Thus, LNP composition and particle size significantly affected islet distribution characteristics, indicating that DOPC-LNPs may be a drug delivery system for effectively targeting the pancreas and islets.
    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
    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 pro-inflammatory 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 utilized 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 pro-apoptotic Bax with acute treatment and JNK with prolonged treatment. Overall, our results suggest that PKCδ mediates cytokine-induced apoptosis via nuclear translocation, cleavage by caspase-3, and upregulation of pro-apoptotic 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

    求助全文

  • 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
    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)

公众号