Fam3a-mediated prohormone convertase switch in α-cells regulates pancreatic GLP-1 production in an Nr4a2-Foxa2-dependent manner.

IF 10.8 1区 医学 Q1 ENDOCRINOLOGY & METABOLISM Metabolism: clinical and experimental Pub Date : 2024-10-01 DOI:10.1016/j.metabol.2024.156042
Dandan Wang, Tianjiao Wei, Xiaona Cui, Li Xia, Yafei Jiang, Deshan Yin, Xinyue Liao, Fei Li, Jian Li, Qi Wu, Xiafang Lin, Shan Lang, Yunyi Le, Jichun Yang, Jin Yang, Rui Wei, Tianpei Hong
{"title":"Fam3a-mediated prohormone convertase switch in α-cells regulates pancreatic GLP-1 production in an Nr4a2-Foxa2-dependent manner.","authors":"Dandan Wang, Tianjiao Wei, Xiaona Cui, Li Xia, Yafei Jiang, Deshan Yin, Xinyue Liao, Fei Li, Jian Li, Qi Wu, Xiafang Lin, Shan Lang, Yunyi Le, Jichun Yang, Jin Yang, Rui Wei, Tianpei Hong","doi":"10.1016/j.metabol.2024.156042","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>Fam3a has been demonstrated to regulate pancreatic β-cell function and glucose homeostasis. However, the role and mechanism of Fam3a in regulating α-cell function remain unexplored.</p><p><strong>Methods: </strong>Glucagon and glucagon-like peptide-1 (GLP-1) levels in pancreas and plasma were measured in global Fam3a knockout (Fam3a<sup>-/-</sup>) mice. Human islet single-cell RNA sequencing (scRNA-seq) datasets were utilized to analyze gene expression correlations between FAM3A and PCSK1 (encoding PC1/3, which processes proglucagon into GLP-1). Mouse pancreatic α-cell line αTC1.9 cells were transfected with Fam3a siRNA or plasmid for Fam3a knockdown or overexpression to explore the effects of Fam3a on PC1/3 expression and GLP-1 production. The downstream mediator (including Nr4a2) was identified by transcriptomic analysis, and its role was confirmed by Fam3a knockdown or overexpression in αTC1.9 cells. Based on the interacted protein of Nr4a2 and the direct binding to Pcsk1 promoter, the transcription factor Foxa2 was selected for further verification. Nuclear translocation assay and dual-luciferase reporter assay were used to clarify the involvement of Fam3a-Nr4a2-Foxa2 pathway in PC1/3 expression and GLP-1 production. Moreover, α-cell-specific Fam3a knockout (Fam3a<sup>α-/-</sup>) mice were constructed to evaluate the metabolic variables and hormone levels under normoglycemic, high-fat diet (HFD)-fed and streptozotocin (STZ)-induced diabetic conditions. Exendin 9-39 (Ex9), a GLP-1 receptor antagonist, was used to investigate GLP-1 paracrine effects in Fam3a<sup>α-/-</sup> mice and in their primary islets.</p><p><strong>Results: </strong>Compared with wild-type mice, pancreatic and plasma active GLP-1 levels were increased in Fam3a<sup>-/-</sup> mice. Analysis of human islet scRNA-seq datasets showed a significant negative correction between FAM3A and PCSK1 in α-cells. Fam3a knockdown upregulated PC1/3 expression and GLP-1 production in αTC1.9 cells, while Fam3a overexpression displayed inverse effects. Transcriptomic analysis identified Nr4a2 as a key downstream mediator of Fam3a, and Nr4a2 expression in αTC1.9 cells was downregulated and upregulated by Fam3a knockdown and overexpression, respectively. Nr4a2 silencing increased PC1/3 expression, albeit Nr4a2 did not directly bind to Pcsk1 promoter. Instead, Nr4a2 formed a complex with Foxa2 to facilitate Fam3a-mediated Foxa2 nuclear translocation. Foxa2 negatively regulated PC1/3 expression and GLP-1 production. Besides, Foxa2 inhibited the transcriptional activity of Pcsk1 promoter at specific binding sites 10 and 6, and this inhibition was intensified by Nr4a2 in αTC1.9 cells. Compared with Flox/cre littermates, improved glucose tolerance, increased active GLP-1 level in pancreas and plasma, upregulated plasma insulin level in response to glucose, and decreased plasma glucagon level were observed in Fam3a<sup>α-/-</sup> mice. Primary islets isolated from Fam3a<sup>α-/-</sup> mice also showed an increase in active GLP-1 and insulin release. In addition, the insulinotropic effect of intra-islet GLP-1 was blocked by Ex9 in Fam3a<sup>α-/-</sup> mice and in their primary islets. Similarly, HFD-fed Fam3a<sup>α-/-</sup> mice also exhibited an improved glucose tolerance. Both HFD-fed and STZ-induced diabetic Fam3a<sup>α-/-</sup> mice showed an increased pancreatic active GLP-1 level, an elevated plasma insulin level and a reduced plasma glucagon level.</p><p><strong>Conclusions: </strong>Fam3a deficiency in α-cells enhances pancreatic GLP-1 production to improve β-cell function via paracrine signaling in an Nr4a2-Foxa2-PC1/3-dependent manner. Our study unveils a novel strategy for reprogramming α-cell proglucagon processing output from glucagon to GLP-1 and deepen the understanding of crosstalk between α-cells and β-cells.</p>","PeriodicalId":18694,"journal":{"name":"Metabolism: clinical and experimental","volume":" ","pages":"156042"},"PeriodicalIF":10.8000,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Metabolism: clinical and experimental","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1016/j.metabol.2024.156042","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"ENDOCRINOLOGY & METABOLISM","Score":null,"Total":0}
引用次数: 0

Abstract

Background: Fam3a has been demonstrated to regulate pancreatic β-cell function and glucose homeostasis. However, the role and mechanism of Fam3a in regulating α-cell function remain unexplored.

Methods: Glucagon and glucagon-like peptide-1 (GLP-1) levels in pancreas and plasma were measured in global Fam3a knockout (Fam3a-/-) mice. Human islet single-cell RNA sequencing (scRNA-seq) datasets were utilized to analyze gene expression correlations between FAM3A and PCSK1 (encoding PC1/3, which processes proglucagon into GLP-1). Mouse pancreatic α-cell line αTC1.9 cells were transfected with Fam3a siRNA or plasmid for Fam3a knockdown or overexpression to explore the effects of Fam3a on PC1/3 expression and GLP-1 production. The downstream mediator (including Nr4a2) was identified by transcriptomic analysis, and its role was confirmed by Fam3a knockdown or overexpression in αTC1.9 cells. Based on the interacted protein of Nr4a2 and the direct binding to Pcsk1 promoter, the transcription factor Foxa2 was selected for further verification. Nuclear translocation assay and dual-luciferase reporter assay were used to clarify the involvement of Fam3a-Nr4a2-Foxa2 pathway in PC1/3 expression and GLP-1 production. Moreover, α-cell-specific Fam3a knockout (Fam3aα-/-) mice were constructed to evaluate the metabolic variables and hormone levels under normoglycemic, high-fat diet (HFD)-fed and streptozotocin (STZ)-induced diabetic conditions. Exendin 9-39 (Ex9), a GLP-1 receptor antagonist, was used to investigate GLP-1 paracrine effects in Fam3aα-/- mice and in their primary islets.

Results: Compared with wild-type mice, pancreatic and plasma active GLP-1 levels were increased in Fam3a-/- mice. Analysis of human islet scRNA-seq datasets showed a significant negative correction between FAM3A and PCSK1 in α-cells. Fam3a knockdown upregulated PC1/3 expression and GLP-1 production in αTC1.9 cells, while Fam3a overexpression displayed inverse effects. Transcriptomic analysis identified Nr4a2 as a key downstream mediator of Fam3a, and Nr4a2 expression in αTC1.9 cells was downregulated and upregulated by Fam3a knockdown and overexpression, respectively. Nr4a2 silencing increased PC1/3 expression, albeit Nr4a2 did not directly bind to Pcsk1 promoter. Instead, Nr4a2 formed a complex with Foxa2 to facilitate Fam3a-mediated Foxa2 nuclear translocation. Foxa2 negatively regulated PC1/3 expression and GLP-1 production. Besides, Foxa2 inhibited the transcriptional activity of Pcsk1 promoter at specific binding sites 10 and 6, and this inhibition was intensified by Nr4a2 in αTC1.9 cells. Compared with Flox/cre littermates, improved glucose tolerance, increased active GLP-1 level in pancreas and plasma, upregulated plasma insulin level in response to glucose, and decreased plasma glucagon level were observed in Fam3aα-/- mice. Primary islets isolated from Fam3aα-/- mice also showed an increase in active GLP-1 and insulin release. In addition, the insulinotropic effect of intra-islet GLP-1 was blocked by Ex9 in Fam3aα-/- mice and in their primary islets. Similarly, HFD-fed Fam3aα-/- mice also exhibited an improved glucose tolerance. Both HFD-fed and STZ-induced diabetic Fam3aα-/- mice showed an increased pancreatic active GLP-1 level, an elevated plasma insulin level and a reduced plasma glucagon level.

Conclusions: Fam3a deficiency in α-cells enhances pancreatic GLP-1 production to improve β-cell function via paracrine signaling in an Nr4a2-Foxa2-PC1/3-dependent manner. Our study unveils a novel strategy for reprogramming α-cell proglucagon processing output from glucagon to GLP-1 and deepen the understanding of crosstalk between α-cells and β-cells.

查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
α细胞中 Fam3a 介导的原激素转换酶转换以 Nr4a2-Foxa2 依赖性方式调节胰腺 GLP-1 的产生。
背景:Fam3a已被证实能调节胰岛β细胞功能和葡萄糖稳态。然而,Fam3a在调节α细胞功能方面的作用和机制仍有待探索:方法:测定了全基因Fam3a基因敲除(Fam3a-/-)小鼠胰腺和血浆中的胰高血糖素和胰高血糖素样肽-1(GLP-1)水平。利用人体胰岛单细胞 RNA 测序(scRNA-seq)数据集分析 FAM3A 和 PCSK1(编码 PC1/3,将胰高血糖素转化为 GLP-1)之间的基因表达相关性。用Fam3a siRNA或质粒转染小鼠胰腺α细胞系αTC1.9细胞,以敲除或过表达Fam3a,从而探讨Fam3a对PC1/3表达和GLP-1生成的影响。通过转录组分析确定了下游介质(包括Nr4a2),并通过Fam3a在αTC1.9细胞中的敲除或过表达证实了其作用。根据 Nr4a2 的相互作用蛋白和与 Pcsk1 启动子的直接结合,转录因子 Foxa2 被选中进行进一步验证。核转位实验和双荧光素酶报告实验明确了Fam3a-Nr4a2-Foxa2通路参与了PC1/3的表达和GLP-1的产生。此外,还构建了α细胞特异性Fam3a基因敲除(Fam3aα-/-)小鼠,以评估正常血糖、高脂饮食(HFD)和链脲佐菌素(STZ)诱导糖尿病条件下的代谢变量和激素水平。GLP-1受体拮抗剂Exendin 9-39(Ex9)被用来研究GLP-1在Fam3aα-/-小鼠及其原代胰岛中的旁分泌效应:结果:与野生型小鼠相比,Fam3a-/-小鼠的胰腺和血浆活性GLP-1水平均有所增加。对人类胰岛 scRNA-seq 数据集的分析表明,α 细胞中的 FAM3A 和 PCSK1 之间存在显著的负校正。Fam3a敲除会上调αTC1.9细胞中PC1/3的表达和GLP-1的产生,而Fam3a过表达则会产生反作用。转录组分析发现,Nr4a2是Fam3a的一个关键下游介质,Fam3a敲除和过表达分别下调和上调了Nr4a2在αTC1.9细胞中的表达。Nr4a2 沉默会增加 PC1/3 的表达,尽管 Nr4a2 并不直接与 Pcsk1 启动子结合。相反,Nr4a2与Foxa2形成复合物,促进Fam3a介导的Foxa2核转位。Foxa2 负向调节 PC1/3 的表达和 GLP-1 的产生。此外,在αTC1.9细胞中,Foxa2在特定结合位点10和6抑制了Pcsk1启动子的转录活性,Nr4a2加强了这种抑制作用。与Flox/cre同窝小鼠相比,Fam3aα-/-小鼠的糖耐量得到改善,胰腺和血浆中活性GLP-1水平升高,血浆胰岛素水平对葡萄糖的反应上调,血浆胰高血糖素水平降低。从 Fam3aα-/- 小鼠体内分离的原代胰岛也显示出活性 GLP-1 和胰岛素释放的增加。此外,在Fam3aα-/-小鼠及其原代胰岛中,胰岛内GLP-1的促胰岛素作用被Ex9阻断。同样,HFD喂养的Fam3aα-/-小鼠的糖耐量也有所改善。HFD喂养和STZ诱导的糖尿病Fam3aα-/-小鼠均表现出胰腺活性GLP-1水平升高、血浆胰岛素水平升高和血浆胰高血糖素水平降低:结论:α细胞中 Fam3a 的缺乏可通过 Nr4a2-Foxa2-PC1/3 依赖性旁分泌信号增强胰腺 GLP-1 的产生,从而改善β细胞的功能。我们的研究揭示了一种将α细胞胰高血糖素加工输出从胰高血糖素重编程为GLP-1的新策略,并加深了对α细胞和β细胞之间串联的理解。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 去求助
来源期刊
Metabolism: clinical and experimental
Metabolism: clinical and experimental 医学-内分泌学与代谢
CiteScore
18.90
自引率
3.10%
发文量
310
审稿时长
16 days
期刊介绍: Metabolism upholds research excellence by disseminating high-quality original research, reviews, editorials, and commentaries covering all facets of human metabolism. Consideration for publication in Metabolism extends to studies in humans, animal, and cellular models, with a particular emphasis on work demonstrating strong translational potential. The journal addresses a range of topics, including: - Energy Expenditure and Obesity - Metabolic Syndrome, Prediabetes, and Diabetes - Nutrition, Exercise, and the Environment - Genetics and Genomics, Proteomics, and Metabolomics - Carbohydrate, Lipid, and Protein Metabolism - Endocrinology and Hypertension - Mineral and Bone Metabolism - Cardiovascular Diseases and Malignancies - Inflammation in metabolism and immunometabolism
期刊最新文献
Targeted metabolomics identified novel metabolites, predominantly phosphatidylcholines and docosahexaenoic acid-containing lipids, predictive of incident chronic kidney disease in middle-to-elderly-aged Chinese adults. The role of IL-1 family cytokines in diabetic cardiomyopathy. Bilirubin bioconversion to urobilin in the gut-liver-kidney axis: A biomarker for insulin resistance in the Cardiovascular-Kidney-Metabolic (CKM) Syndrome Reducing the global prevalence of cardiometabolic risk factors: a bet worth winning An updated overview on hepatocellular carcinoma in patients with Metabolic dysfunction-Associated Steatotic Liver Disease: Trends, pathophysiology and risk-based surveillance
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1