Islets of Langerhans

朗格汉斯岛
  • 文章类型: Journal Article
    响应碳水化合物的胰岛素分泌的群体水平变化和机制,蛋白质,脂肪仍然没有特征。我们定义了来自140名尸体供体的胰岛中三种常量营养素的典型胰岛素分泌反应,包括2型糖尿病患者。大多数供体胰岛对葡萄糖表现出最高的胰岛素反应,对氨基酸的适度反应,对脂肪酸的反应最小。然而,9%的供体胰岛有氨基酸反应,8%的脂肪酸反应大于葡萄糖刺激的胰岛素反应。我们利用了这种异质性,并使用了多组学来识别营养反应性的分子相关性,以及2型糖尿病中蛋白质和mRNA的改变。我们还检查了干细胞衍生的胰岛中营养刺激的胰岛素释放,并观察到对脂肪的反应性,而不是碳水化合物或蛋白质-可能是不成熟的标志。了解胰岛素对碳水化合物反应的多样性,蛋白质,脂肪为个性化营养奠定了基础。
    Population-level variation and mechanisms behind insulin secretion in response to carbohydrate, protein, and fat remain uncharacterized. We defined prototypical insulin secretion responses to three macronutrients in islets from 140 cadaveric donors, including those with type 2 diabetes. The majority of donors\' islets exhibited the highest insulin response to glucose, moderate response to amino acid, and minimal response to fatty acid. However, 9% of donors\' islets had amino acid responses, and 8% had fatty acid responses that were larger than their glucose-stimulated insulin responses. We leveraged this heterogeneity and used multi-omics to identify molecular correlates of nutrient responsiveness, as well as proteins and mRNAs altered in type 2 diabetes. We also examined nutrient-stimulated insulin release from stem cell-derived islets and observed responsiveness to fat but not carbohydrate or protein-potentially a hallmark of immaturity. Understanding the diversity of insulin responses to carbohydrate, protein, and fat lays the groundwork for personalized nutrition.
    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
    糖尿病涉及胰腺β细胞的死亡或功能障碍。人样品的批量测序分析和使用体外和体内模型的研究表明内质网和炎症信号在糖尿病进展中起重要作用。为了更好地表征细胞类型特异性应激反应,我们进行多重单细胞RNA测序,以确定暴露于内质网和炎性应激的原代人胰岛细胞的转录特征.通过跨胰腺内分泌和外分泌细胞类型的应激反应的全面成对分析,我们定义了在不同的糖尿病相关应激源下每种细胞类型的基因表达变化.我们发现β-,α-,和导管细胞有最大的转录反应。我们利用干细胞衍生的胰岛通过候选基因CIB1研究胰岛健康,该基因在原代人类胰岛中在应激下上调。我们的发现提供了对糖尿病相关压力的细胞类型特异性反应的见解,并建立了确定糖尿病治疗靶标的资源。
    Diabetes involves the death or dysfunction of pancreatic β-cells. Analysis of bulk sequencing from human samples and studies using in vitro and in vivo models suggest that endoplasmic reticulum and inflammatory signaling play an important role in diabetes progression. To better characterize cell type-specific stress response, we perform multiplexed single-cell RNA sequencing to define the transcriptional signature of primary human islet cells exposed to endoplasmic reticulum and inflammatory stress. Through comprehensive pair-wise analysis of stress responses across pancreatic endocrine and exocrine cell types, we define changes in gene expression for each cell type under different diabetes-associated stressors. We find that β-, α-, and ductal cells have the greatest transcriptional response. We utilize stem cell-derived islets to study islet health through the candidate gene CIB1, which was upregulated under stress in primary human islets. Our findings provide insights into cell type-specific responses to diabetes-associated stress and establish a resource to identify targets for diabetes therapeutics.
    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,一个重大的全球公共卫生挑战,严重影响全球人类健康。类器官,创新的体外三维(3D)培养模型,紧密模仿体内的组织或器官。分泌胰岛素的胰岛类器官,来源于体外诱导的3D结构的干细胞,已经成为胰岛移植的潜在替代方案和反映人体体内环境的可能疾病模型,消除物种差异。该技术因其在糖尿病治疗中的潜力而获得了相当大的关注。尽管取得了进展,干细胞分化为胰岛类器官的过程及其培养表明存在缺陷,促使人们不断努力开发更有效的分化方案和3D仿生材料。目前,构建的胰岛类器官在其组成上表现出局限性,结构,与天然胰岛相比时的功能。因此,进一步的研究是必要的,以实现多组织系统的组成和改善的胰岛素分泌功能在胰岛器官,在解决移植相关安全问题的同时,例如致瘤性,免疫排斥,感染,和血栓形成。这篇综述探讨了构建胰岛类器官的方法和策略,其在糖尿病治疗中的应用,以及类器官研究中的关键科学挑战,为更深入地了解糖尿病的发病机制和治疗干预措施的发展提供了新的视角。
    Diabetes mellitus, a significant global public health challenge, severely impacts human health worldwide. The organoid, an innovative in vitro three-dimensional (3D) culture model, closely mimics tissues or organs in vivo. Insulin-secreting islet organoid, derived from stem cells induced in vitro with 3D structures, has emerged as a potential alternative for islet transplantation and as a possible disease model that mirrors the human body\'s in vivo environment, eliminating species difference. This technology has gained considerable attention for its potential in diabetes treatment. Despite advances, the process of stem cell differentiation into islet organoid and its cultivation demonstrates deficiencies, prompting ongoing efforts to develop more efficient differentiation protocols and 3D biomimetic materials. At present, the constructed islet organoid exhibit limitations in their composition, structure, and functionality when compared to natural islets. Consequently, further research is imperative to achieve a multi-tissue system composition and improved insulin secretion functionality in islet organoid, while addressing transplantation-related safety concerns, such as tumorigenicity, immune rejection, infection, and thrombosis. This review delves into the methodologies and strategies for constructing the islet organoid, its application in diabetes treatment, and the pivotal scientific challenges within organoid research, offering fresh perspectives for a deeper understanding of diabetes pathogenesis and the development of therapeutic interventions.
    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
    普遍的RNA选择性剪接(AS)有助于分子多样性,已在细胞功能调节和疾病发病机理中得到证实。然而,糖尿病进展过程中胰岛中AS的作用尚不清楚.这里,我们重新分析了来自已保存数据库的全长单细胞RNA测序数据,以研究非糖尿病(ND)和2型糖尿病(T2D)个体中人类胰腺内分泌细胞类型之间的AS调节.我们的分析表明转录组AS谱和细胞类型特异性之间存在显著关联,可用于区分主要内分泌细胞类型的聚类。此外,AS谱能够清楚地定义健康对照中β细胞的成熟亚群,在T2D中完全丢失。进一步的分析表明,RNA结合蛋白(RBP),预测异质核核糖核蛋白(hnRNP)和FXR1家族蛋白通过调节AS谱诱导β细胞的功能损害。最后,内分泌细胞的轨迹分析表明,在T2D的进展过程中,β细胞的身份通过β细胞的去分化和转分化而发生改变。一起,我们的研究提供了调节β细胞功能的机制,并表明AS程序在糖尿病发病过程中的重要作用。
    The prevalent RNA alternative splicing (AS) contributes to molecular diversity, which has been demonstrated in cellular function regulation and disease pathogenesis. However, the contribution of AS in pancreatic islets during diabetes progression remains unclear. Here, we reanalyze the full-length single-cell RNA sequencing data from the deposited database to investigate AS regulation across human pancreatic endocrine cell types in non-diabetic (ND) and type 2 diabetic (T2D) individuals. Our analysis demonstrates the significant association between transcriptomic AS profiles and cell-type-specificity, which could be applied to distinguish the clustering of major endocrine cell types. Moreover, AS profiles are enabled to clearly define the mature subset of β-cells in healthy controls, which is completely lost in T2D. Further analysis reveals that RNA-binding proteins (RBPs), heterogeneous nuclear ribonucleoproteins (hnRNPs) and FXR1 family proteins are predicted to induce the functional impairment of β-cells through regulating AS profiles. Finally, trajectory analysis of endocrine cells suggests the β-cell identity shift through dedifferentiation and transdifferentiation of β-cells during the progression of T2D. Together, our study provides a mechanism for regulating β-cell functions and suggests the significant contribution of AS program during diabetes pathogenesis.
    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
    锌缺乏与糖尿病的恶化有关,而锌补充剂已被提议改善糖尿病。这项研究检查了边际锌缺乏(MZD)和锌补充(ZS)对肥胖的影响,血糖控制,胰岛,Zucker糖尿病脂肪(ZDF)大鼠的肝脏脂肪变性和肾功能。给雄性ZDF大鼠喂食MZD,锌控制(ZC)或ZS饮食(4、30和300mgZn/kg饮食,分别),和瘦Zucker大鼠喂食ZC饮食8周。MZD和ZS不会改变ZDF大鼠的体重或全身组成。MZDZDF大鼠股骨和胰腺中的锌浓度降低,与ZCZDF相比,基于1.8倍的葡萄糖增量曲线下面积(AUC),胰岛数量增加,对口服葡萄糖负荷的反应减弱。ZSZDF大鼠血清升高,股骨和胰腺锌浓度,与ZCZDF大鼠相比,胰腺参数不变,胰岛素的AUC降低了50%,提示更高的胰岛素敏感性。饮食锌的摄入不会改变肝脏脂肪变性,肌酐清除率,或有助于胰岛素信号传导的蛋白质水平,附睾脂肪中的炎症或锌转运。与ZCZDF大鼠相比,肝脏铜浓度降低和血清尿素升高提示了ZS的潜在不良反应。总之,ZS改善了胰腺胰岛素反应,但没有改善葡萄糖处理。相比之下,ZDF大鼠的锌状态降低导致葡萄糖耐量降低,胰岛的数量和大小代偿性增加,这可能导致β细胞衰竭。
    Zinc deficiency has been associated with the worsening of diabetes while zinc supplementation has been proposed to ameliorate diabetes. This study examined the effects of marginal zinc deficiency (MZD) and zinc supplementation (ZS) on obesity, glycemic control, pancreatic islets, hepatic steatosis and renal function of Zucker diabetic fatty (ZDF) rats. Male ZDF rats were fed an MZD, zinc control (ZC) or ZS diet (4, 30 and 300 mg Zn/kg diet, respectively), and lean Zucker rats were fed a ZC diet for 8 weeks. MZD and ZS did not alter body weight or whole-body composition in ZDF rats. MZD ZDF rats had reduced zinc concentrations in the femur and pancreas, a greater number of enlarged pancreatic islets and a diminished response to an oral glucose load based on a 1.8-fold greater incremental area-under-the-curve (AUC) for glucose compared to ZC ZDF. ZS ZDF rats had elevated serum, femur and pancreatic zinc concentrations, unchanged pancreatic parameters and a 50% reduction in the AUC for insulin compared to ZC ZDF rats, suggesting greater insulin sensitivity. Dietary zinc intake did not alter hepatic steatosis, creatinine clearance, or levels of proteins that contribute to insulin signaling, inflammation or zinc transport in epididymal fat. Potential adverse effects of ZS were suggested by reduced hepatic copper concentrations and elevated serum urea compared to ZC ZDF rats. In summary, ZS improved the pancreatic insulin response but not the glucose handling. In contrast, reduced zinc status in ZDF rats led to impaired glucose tolerance and a compensatory increase in the number and size of pancreatic islets which could lead to β-cell exhaustion.
    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
    糖尿病给全世界带来了巨大的负担。胰岛移植是糖尿病的替代疗法。然而,他克莫司,器官移植后的一种免疫抑制剂,与移植后糖尿病密切相关。间充质干细胞(MSC)因其缓解糖尿病的潜力而引起了人们的兴趣。体内实验显示,人类经血干细胞(MenSCs)治疗改善了他克莫司诱导的血糖,体重,和小鼠的葡萄糖耐量紊乱。RNA测序用于分析MenSC的潜在治疗靶标。在这项研究中,我们说明了在他克莫司诱导的胰岛功能障碍中,胱抑素β-合成酶(CBS)的作用。使用β细胞系(MIN6,β-TC-6),我们证明了MenSCs在体外改善他克莫司诱导的胰岛功能障碍。此外,MenSC降低了他克莫司诱导的CBS水平升高,并显着增强了活力,抗凋亡能力,葡萄糖刺激的胰岛素分泌(GSIS),和β细胞的糖酵解通量。我们进一步揭示了MenSC通过抑制CBS表达以激活IL6/JAK2/STAT3途径发挥其治疗作用。总之,我们表明,MenSCs可能是改善他克莫司诱导的胰岛功能障碍的潜在策略.
    Diabetes imposes a huge burden worldwide. Islet transplantation is an alternative therapy for diabetes. However, tacrolimus, a kind of immunosuppressant after organ transplantation, is closely related to post-transplant diabetes mellitus. Mesenchymal stem cells (MSCs) have attracted interest for their potential to alleviate diabetes. In vivo experiments revealed that human menstrual blood-derived stem cells (MenSCs) treatment improved tacrolimus-induced blood glucose, body weight, and glucose tolerance disorders in mice. RNA sequencing was used to analyze the potential therapeutic targets of MenSCs. In this study, we illustrated that cystathionine β-synthase (CBS) contributed to tacrolimus -induced islet dysfunction. Using β-cell lines (MIN6, β-TC-6), we demonstrated that MenSCs ameliorated tacrolimus-induced islet dysfunction in vitro. Moreover, MenSC reduced the tacrolimus-induced elevation of CBS levels and significantly enhanced the viability, anti-apoptotic ability, glucose-stimulated insulin secretion (GSIS), and glycolytic flux of β-cells. We further revealed that MenSCs exerted their therapeutic effects by inhibiting CBS expression to activate the IL6/JAK2/STAT3 pathway. In conclusion, we showed that MenSCs may be a potential strategy to improve tacrolimus-induced 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
    非肥胖糖尿病(NOD)小鼠是一种既定的,自发的1型糖尿病模型,其中糖尿病通过胰岛炎发展。使用下一代测序,加上途径分析,早期胰岛炎的分子指纹图谱在一组4~12周龄的小鼠中绘制.所得的动态时间线显示增殖能力的初始降低,随后在6至8周之间出现炎性特征,其在10至12周之间增加至调节平台。通过激活中枢免疫原性因子如Infg,Il1b,还有Tnfa,和典型炎症信号的激活。对调控景观的分析揭示了转录因子Atf3作为NOD胰岛中炎症信号传导的潜在新型调节剂。此外,Hedgehog信号通路与Atf3调节相关,这表明两者在调节胰岛炎症中起作用;然而,需要进一步的研究来确定这种联系的性质。
    Non-obese diabetes (NOD) mice are an established, spontaneous model of type 1 diabetes in which diabetes develops through insulitis. Using next-generation sequencing, coupled with pathway analysis, the molecular fingerprint of early insulitis was mapped in a cohort of mice ranging from 4 to 12 weeks of age. The resulting dynamic timeline revealed an initial decrease in proliferative capacity followed by the emergence of an inflammatory signature between 6 and 8 weeks that increased to a regulatory plateau between 10 and 12 weeks. The inflammatory signature is identified by the activation of central immunogenic factors such as Infg, Il1b, and Tnfa, and activation of canonical inflammatory signaling. Analysis of the regulatory landscape revealed the transcription factor Atf3 as a potential novel modulator of inflammatory signaling in the NOD islets. Furthermore, the Hedgehog signaling pathway correlated with Atf3 regulation, suggesting that the two play a role in regulating islet inflammation; however, further studies are needed to establish the nature of this connection.
    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)是全球增长最快的非传染性疾病。胰腺β细胞分泌胰岛素受损是T2D的标志,但这一缺陷背后的机制特征不够充分。整合多层生物医学信息,比如不同的组学,可以更准确地理解复杂的疾病,如T2D。我们的目标是探索和使用机器学习来整合多种生物/分子信息源(multiOmics)。在我们的RNA测序中,DNA甲基化,来自具有T2D和非糖尿病对照的胰岛供体的SNP和表型数据。我们利用机器学习来进行DNA甲基化的多组学整合,表达式,SNPs,和来自110个个体的胰岛的表型,约30%为T2D病例。使用Infinium甲基化EPIC阵列分析DNA甲基化,使用RNA测序分析表达,并使用HumanOmniExpress阵列分析SNP。通过基于偏最小二乘(PLS)的DIABLO的有监督的线性多元整合在交叉验证后在测试数据集上实现了T2D预测的91±15%的准确度,曲线下面积为0.96±0.08。通过这种多组学整合确定的生物标志物,包括SACS和TXNIPDNA甲基化,OPRD1和RHOT1表达以及注释为ANO1的SNP提供了对导致T2D的不同生物学机制之间相互作用的新见解。这种来自人类胰岛的多维数据的机器学习方法实现了T2D预测的有希望的准确性,这可能在临床诊断中找到广泛的应用。此外,它为T2D提供了新的候选生物标志物,以及它们在不同组学之间的联系。
    Type 2 diabetes (T2D) is the fastest growing non-infectious disease worldwide. Impaired insulin secretion from pancreatic beta-cells is a hallmark of T2D, but the mechanisms behind this defect are insufficiently characterized. Integrating multiple layers of biomedical information, such as different Omics, may allow more accurate understanding of complex diseases such as T2D. Our aim was to explore and use Machine Learning to integrate multiple sources of biological/molecular information (multiOmics), in our case RNA-sequening, DNA methylation, SNP and phenotypic data from islet donors with T2D and non-diabetic controls. We exploited Machine Learning to perform multiOmics integration of DNA methylation, expression, SNPs, and phenotypes from pancreatic islets of 110 individuals, with ~ 30% being T2D cases. DNA methylation was analyzed using Infinium MethylationEPIC array, expression was analyzed using RNA-sequencing, and SNPs were analyzed using HumanOmniExpress arrays. Supervised linear multiOmics integration via DIABLO based on Partial Least Squares (PLS) achieved an accuracy of 91 ± 15% of T2D prediction with an area under the curve of 0.96 ± 0.08 on the test dataset after cross-validation. Biomarkers identified by this multiOmics integration, including SACS and TXNIP DNA methylation, OPRD1 and RHOT1 expression and a SNP annotated to ANO1, provide novel insights into the interplay between different biological mechanisms contributing to T2D. This Machine Learning approach of multiOmics cross-sectional data from human pancreatic islets achieved a promising accuracy of T2D prediction, which may potentially find broad applications in clinical diagnostics. In addition, it delivered novel candidate biomarkers for T2D and links between them across the different Omics.
    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
    暂无摘要。
    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
    1型糖尿病(T1D)之前是异质的临床前阶段,胰岛自身免疫(IA)。我们的目标是确定pre与IA后血清转换(SV)的DNAm变化在三种IA进展表型中有所不同,那些失去自身抗体(回复器)的人,临床T1D进展(进展),或维持自身抗体水平(维持者)。
    这项全表观基因组关联研究(EWAS)包括开发IA的年轻糖尿病自身免疫研究(DAISY)参与者的血液中纵向DNAm测量(Illumina450K和EPIC),在至少两次连续访问中出现一种或多种胰岛自身抗体。我们比较了回复者——血清回复者,在至少两次连续访视中,所有自身抗体均呈阴性,且未出现T1D(n=41);维持者-自身抗体持续检测呈阳性,但未出现T1D(n=60);进展者-出现临床T1D(n=42).在IA之前(SV访视前)和之后(SV访视后)测量DNAm数据。线性混合模型用于测试三组中DNAm的前SV变化与后SV变化的差异。线性混合模型也用于测试平均DNAm的组差异。细胞比例,年龄,和性别在所有模型中都进行了调整。所有参与者的中位随访时间为15.5年。(四分位数间距(IQR):10.8-18.7)。
    SV前就诊的中位年龄为2.2岁。(IQR:0.8-5.3)在进展中,与6.0年相比。(IQR:1.3-8.4)在变频器中,5.7年。(IQR:1.4-9.7)在维护者。回转机1.4年的访问时间中位数相似。(IQR:1-1.9),维护者1.3年。(IQR:1.0-2.0),和进步1.8年。(IQR:1.0-2.0)。DNAm的变化,前与后SV,在一个地点(CG16066195)和11个地区的不同群体。平均DNAm(SV前后的平均值)在22个地区有所不同。
    差异变化的DNAm区域位于与β细胞功能相关的基因组区域,免疫细胞分化,和免疫细胞功能。
    UNASSIGNED: Type 1 diabetes (T1D) is preceded by a heterogenous pre-clinical phase, islet autoimmunity (IA). We aimed to identify pre vs. post-IA seroconversion (SV) changes in DNAm that differed across three IA progression phenotypes, those who lose autoantibodies (reverters), progress to clinical T1D (progressors), or maintain autoantibody levels (maintainers).
    UNASSIGNED: This epigenome-wide association study (EWAS) included longitudinal DNAm measurements in blood (Illumina 450K and EPIC) from participants in Diabetes Autoimmunity Study in the Young (DAISY) who developed IA, one or more islet autoantibodies on at least two consecutive visits. We compared reverters - individuals who sero-reverted, negative for all autoantibodies on at least two consecutive visits and did not develop T1D (n=41); maintainers - continued to test positive for autoantibodies but did not develop T1D (n=60); progressors - developed clinical T1D (n=42). DNAm data were measured before (pre-SV visit) and after IA (post-SV visit). Linear mixed models were used to test for differences in pre- vs post-SV changes in DNAm across the three groups. Linear mixed models were also used to test for group differences in average DNAm. Cell proportions, age, and sex were adjusted for in all models. Median follow-up across all participants was 15.5 yrs. (interquartile range (IQR): 10.8-18.7).
    UNASSIGNED: The median age at the pre-SV visit was 2.2 yrs. (IQR: 0.8-5.3) in progressors, compared to 6.0 yrs. (IQR: 1.3-8.4) in reverters, and 5.7 yrs. (IQR: 1.4-9.7) in maintainers. Median time between the visits was similar in reverters 1.4 yrs. (IQR: 1-1.9), maintainers 1.3 yrs. (IQR: 1.0-2.0), and progressors 1.8 yrs. (IQR: 1.0-2.0). Changes in DNAm, pre- vs post-SV, differed across the groups at one site (cg16066195) and 11 regions. Average DNAm (mean of pre- and post-SV) differed across 22 regions.
    UNASSIGNED: Differentially changing DNAm regions were located in genomic areas related to beta cell function, immune cell differentiation, and immune cell function.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号