RFX6的缺失会影响iPSC衍生的胰岛器官发育和存活,但对PDX1+/NKX6.1+祖细胞没有影响。

IF 8.4 1区 医学 Q1 ENDOCRINOLOGY & METABOLISM Diabetologia Pub Date : 2024-07-30 DOI:10.1007/s00125-024-06232-2
Noura Aldous, Ahmed K Elsayed, Bushra Memon, Sadaf Ijaz, Sikander Hayat, Essam M Abdelalim
{"title":"RFX6的缺失会影响iPSC衍生的胰岛器官发育和存活,但对PDX1+/NKX6.1+祖细胞没有影响。","authors":"Noura Aldous, Ahmed K Elsayed, Bushra Memon, Sadaf Ijaz, Sikander Hayat, Essam M Abdelalim","doi":"10.1007/s00125-024-06232-2","DOIUrl":null,"url":null,"abstract":"<p><strong>Aims/hypothesis: </strong>Homozygous mutations in RFX6 lead to neonatal diabetes accompanied by a hypoplastic pancreas, whereas heterozygous mutations cause MODY. Recent studies have also shown RFX6 variants to be linked with type 2 diabetes. Despite RFX6's known function in islet development, its specific role in diabetes pathogenesis remains unclear. Here, we aimed to understand the mechanisms underlying the impairment of pancreatic islet development and subsequent hypoplasia due to loss-of-function mutations in RFX6.</p><p><strong>Methods: </strong>We examined regulatory factor X6 (RFX6) expression during human embryonic stem cell (hESC) differentiation into pancreatic islets and re-analysed a single-cell RNA-seq dataset to identify RFX6-specific cell populations during islet development. Furthermore, induced pluripotent stem cell (iPSC) lines lacking RFX6 were generated using CRISPR/Cas9. Various approaches were then employed to explore the consequences of RFX6 loss across different developmental stages. Subsequently, we evaluated transcriptional changes resulting from RFX6 loss through RNA-seq of pancreatic progenitors (PPs) and endocrine progenitors (EPs).</p><p><strong>Results: </strong>RFX6 expression was detected in PDX1<sup>+</sup> cells in the hESC-derived posterior foregut (PF). However, in the PPs, RFX6 did not co-localise with pancreatic and duodenal homeobox 1 (PDX1) or NK homeobox 1 (NKX6.1) but instead co-localised with neurogenin 3, NK2 homeobox 2 and islet hormones in the EPs and islets. Single-cell analysis revealed high RFX6 expression levels in endocrine clusters across various hESC-derived pancreatic differentiation stages. Upon differentiating iPSCs lacking RFX6 into pancreatic islets, a significant decrease in PDX1 expression at the PF stage was observed, although this did not affect PPs co-expressing PDX1 and NKX6.1. RNA-seq analysis showed the downregulation of essential genes involved in pancreatic endocrine differentiation, insulin secretion and ion transport due to RFX6 deficiency. Furthermore, RFX6 deficiency resulted in the formation of smaller islet organoids due to increased cellular apoptosis, linked to reduced catalase expression, implying a protective role for RFX6. Overexpression of RFX6 reversed defective phenotypes in RFX6-knockout PPs, EPs and islets.</p><p><strong>Conclusions/interpretation: </strong>These findings suggest that pancreatic hypoplasia and reduced islet cell formation associated with RFX6 mutations are not due to alterations in PDX1<sup>+</sup>/NKX6.1<sup>+</sup> PPs but instead result from cellular apoptosis and downregulation of pancreatic endocrine genes.</p><p><strong>Data availability: </strong>RNA-seq datasets have been deposited in the Zenodo repository with accession link (DOI: https://doi.org/10.5281/zenodo.10656891 ).</p>","PeriodicalId":11164,"journal":{"name":"Diabetologia","volume":null,"pages":null},"PeriodicalIF":8.4000,"publicationDate":"2024-07-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":"{\"title\":\"Deletion of RFX6 impairs iPSC-derived islet organoid development and survival, with no impact on PDX1<sup>+</sup>/NKX6.1<sup>+</sup> progenitors.\",\"authors\":\"Noura Aldous, Ahmed K Elsayed, Bushra Memon, Sadaf Ijaz, Sikander Hayat, Essam M Abdelalim\",\"doi\":\"10.1007/s00125-024-06232-2\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p><strong>Aims/hypothesis: </strong>Homozygous mutations in RFX6 lead to neonatal diabetes accompanied by a hypoplastic pancreas, whereas heterozygous mutations cause MODY. Recent studies have also shown RFX6 variants to be linked with type 2 diabetes. Despite RFX6's known function in islet development, its specific role in diabetes pathogenesis remains unclear. Here, we aimed to understand the mechanisms underlying the impairment of pancreatic islet development and subsequent hypoplasia due to loss-of-function mutations in RFX6.</p><p><strong>Methods: </strong>We examined regulatory factor X6 (RFX6) expression during human embryonic stem cell (hESC) differentiation into pancreatic islets and re-analysed a single-cell RNA-seq dataset to identify RFX6-specific cell populations during islet development. Furthermore, induced pluripotent stem cell (iPSC) lines lacking RFX6 were generated using CRISPR/Cas9. Various approaches were then employed to explore the consequences of RFX6 loss across different developmental stages. Subsequently, we evaluated transcriptional changes resulting from RFX6 loss through RNA-seq of pancreatic progenitors (PPs) and endocrine progenitors (EPs).</p><p><strong>Results: </strong>RFX6 expression was detected in PDX1<sup>+</sup> cells in the hESC-derived posterior foregut (PF). However, in the PPs, RFX6 did not co-localise with pancreatic and duodenal homeobox 1 (PDX1) or NK homeobox 1 (NKX6.1) but instead co-localised with neurogenin 3, NK2 homeobox 2 and islet hormones in the EPs and islets. Single-cell analysis revealed high RFX6 expression levels in endocrine clusters across various hESC-derived pancreatic differentiation stages. Upon differentiating iPSCs lacking RFX6 into pancreatic islets, a significant decrease in PDX1 expression at the PF stage was observed, although this did not affect PPs co-expressing PDX1 and NKX6.1. RNA-seq analysis showed the downregulation of essential genes involved in pancreatic endocrine differentiation, insulin secretion and ion transport due to RFX6 deficiency. Furthermore, RFX6 deficiency resulted in the formation of smaller islet organoids due to increased cellular apoptosis, linked to reduced catalase expression, implying a protective role for RFX6. Overexpression of RFX6 reversed defective phenotypes in RFX6-knockout PPs, EPs and islets.</p><p><strong>Conclusions/interpretation: </strong>These findings suggest that pancreatic hypoplasia and reduced islet cell formation associated with RFX6 mutations are not due to alterations in PDX1<sup>+</sup>/NKX6.1<sup>+</sup> PPs but instead result from cellular apoptosis and downregulation of pancreatic endocrine genes.</p><p><strong>Data availability: </strong>RNA-seq datasets have been deposited in the Zenodo repository with accession link (DOI: https://doi.org/10.5281/zenodo.10656891 ).</p>\",\"PeriodicalId\":11164,\"journal\":{\"name\":\"Diabetologia\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":8.4000,\"publicationDate\":\"2024-07-30\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Diabetologia\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://doi.org/10.1007/s00125-024-06232-2\",\"RegionNum\":1,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q1\",\"JCRName\":\"ENDOCRINOLOGY & METABOLISM\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Diabetologia","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1007/s00125-024-06232-2","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

摘要

目的/假设:RFX6 的同基因突变会导致新生儿糖尿病,并伴有胰腺发育不全,而杂基因突变则会导致 MODY。最近的研究还显示 RFX6 变异与 2 型糖尿病有关。尽管 RFX6 在胰岛发育中的功能众所周知,但其在糖尿病发病机制中的具体作用仍不清楚。在此,我们旨在了解 RFX6 功能缺失突变导致的胰岛发育障碍和随后的胰岛发育不良的机制:我们检测了人类胚胎干细胞(hESC)分化成胰岛过程中调节因子X6(RFX6)的表达,并重新分析了单细胞RNA-seq数据集,以确定胰岛发育过程中RFX6特异性细胞群。此外,还利用 CRISPR/Cas9 生成了缺乏 RFX6 的诱导多能干细胞(iPSC)系。然后,我们采用了多种方法来探索 RFX6 缺失在不同发育阶段的后果。随后,我们通过对胰腺祖细胞(PPs)和内分泌祖细胞(EPs)进行RNA-seq分析,评估了RFX6缺失导致的转录变化:结果:在hESC衍生的后前肠(PF)的PDX1+细胞中检测到了RFX6的表达。然而,在后前肠(PPs)中,RFX6 并未与胰腺和十二指肠同工酶 1(PDX1)或 NK 同工酶 1(NKX6.1)共定位,而是与神经原蛋白 3、NK2 同工酶 2 以及 EPs 和胰岛激素共定位。单细胞分析显示,在不同的 hESC 衍生胰腺分化阶段,内分泌簇中的 RFX6 表达水平都很高。将缺乏 RFX6 的 iPSCs 分化成胰岛后,在 PF 阶段观察到 PDX1 表达显著下降,但这并不影响共同表达 PDX1 和 NKX6.1 的 PPs。RNA-seq 分析显示,RFX6 缺乏导致参与胰腺内分泌分化、胰岛素分泌和离子转运的重要基因下调。此外,由于细胞凋亡增加,RFX6 缺乏会导致形成较小的胰岛器官组织,这与过氧化氢酶表达减少有关,意味着 RFX6 具有保护作用。RFX6的过表达逆转了RFX6基因敲除的胰岛细胞、胰岛素细胞和胰岛的缺陷表型:这些研究结果表明,与 RFX6 基因突变相关的胰腺发育不良和胰岛细胞形成减少并非由于 PDX1+/NKX6.1+ PPs 的改变,而是细胞凋亡和胰腺内分泌基因下调的结果:RNA-seq数据集已存入Zenodo资源库,并附有加入链接(DOI: https://doi.org/10.5281/zenodo.10656891 )。
本文章由计算机程序翻译,如有差异,请以英文原文为准。

摘要图片

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
Deletion of RFX6 impairs iPSC-derived islet organoid development and survival, with no impact on PDX1+/NKX6.1+ progenitors.

Aims/hypothesis: Homozygous mutations in RFX6 lead to neonatal diabetes accompanied by a hypoplastic pancreas, whereas heterozygous mutations cause MODY. Recent studies have also shown RFX6 variants to be linked with type 2 diabetes. Despite RFX6's known function in islet development, its specific role in diabetes pathogenesis remains unclear. Here, we aimed to understand the mechanisms underlying the impairment of pancreatic islet development and subsequent hypoplasia due to loss-of-function mutations in RFX6.

Methods: We examined regulatory factor X6 (RFX6) expression during human embryonic stem cell (hESC) differentiation into pancreatic islets and re-analysed a single-cell RNA-seq dataset to identify RFX6-specific cell populations during islet development. Furthermore, induced pluripotent stem cell (iPSC) lines lacking RFX6 were generated using CRISPR/Cas9. Various approaches were then employed to explore the consequences of RFX6 loss across different developmental stages. Subsequently, we evaluated transcriptional changes resulting from RFX6 loss through RNA-seq of pancreatic progenitors (PPs) and endocrine progenitors (EPs).

Results: RFX6 expression was detected in PDX1+ cells in the hESC-derived posterior foregut (PF). However, in the PPs, RFX6 did not co-localise with pancreatic and duodenal homeobox 1 (PDX1) or NK homeobox 1 (NKX6.1) but instead co-localised with neurogenin 3, NK2 homeobox 2 and islet hormones in the EPs and islets. Single-cell analysis revealed high RFX6 expression levels in endocrine clusters across various hESC-derived pancreatic differentiation stages. Upon differentiating iPSCs lacking RFX6 into pancreatic islets, a significant decrease in PDX1 expression at the PF stage was observed, although this did not affect PPs co-expressing PDX1 and NKX6.1. RNA-seq analysis showed the downregulation of essential genes involved in pancreatic endocrine differentiation, insulin secretion and ion transport due to RFX6 deficiency. Furthermore, RFX6 deficiency resulted in the formation of smaller islet organoids due to increased cellular apoptosis, linked to reduced catalase expression, implying a protective role for RFX6. Overexpression of RFX6 reversed defective phenotypes in RFX6-knockout PPs, EPs and islets.

Conclusions/interpretation: These findings suggest that pancreatic hypoplasia and reduced islet cell formation associated with RFX6 mutations are not due to alterations in PDX1+/NKX6.1+ PPs but instead result from cellular apoptosis and downregulation of pancreatic endocrine genes.

Data availability: RNA-seq datasets have been deposited in the Zenodo repository with accession link (DOI: https://doi.org/10.5281/zenodo.10656891 ).

求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
Diabetologia
Diabetologia 医学-内分泌学与代谢
CiteScore
18.10
自引率
2.40%
发文量
193
审稿时长
1 months
期刊介绍: Diabetologia, the authoritative journal dedicated to diabetes research, holds high visibility through society membership, libraries, and social media. As the official journal of the European Association for the Study of Diabetes, it is ranked in the top quartile of the 2019 JCR Impact Factors in the Endocrinology & Metabolism category. The journal boasts dedicated and expert editorial teams committed to supporting authors throughout the peer review process.
期刊最新文献
The DPP-4 inhibitor sitagliptin improves glycaemic control and early-stage diabetic nephropathy in adolescents with type 1 diabetes using the MiniMed 780G advanced hybrid closed-loop system: a randomised controlled trial Genetic evidence for efficacy of targeting IL-2, IL-6 and TYK2 signalling in the prevention of type 1 diabetes: a Mendelian randomisation study Risk factors for Charcot foot development in individuals with diabetes mellitus Metabolic health in people living with type 1 diabetes in Belgium: a repeated cross-sectional study Up Front
×
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