A JAGN1-associated severe congenital neutropenia zebrafish model revealed an altered G-CSFR signaling and UPR activation.

IF 7.4 1区 医学 Q1 HEMATOLOGY Blood advances Pub Date : 2024-08-13 DOI:10.1182/bloodadvances.2023011656
Larissa Doll, Karl Welte, Julia Skokowa, Baubak Bajoghli
{"title":"A JAGN1-associated severe congenital neutropenia zebrafish model revealed an altered G-CSFR signaling and UPR activation.","authors":"Larissa Doll, Karl Welte, Julia Skokowa, Baubak Bajoghli","doi":"10.1182/bloodadvances.2023011656","DOIUrl":null,"url":null,"abstract":"<p><strong>Abstract: </strong>A variety of autosomal recessive mutations in the JAGN1 gene cause severe congenital neutropenia (CN). However, the underlying pathomechanism remains poorly understood, mainly because of the limited availability of primary hematopoietic stem cells from JAGN1-CN patients and the absence of animal models. In this study, we aimed to address these limitations by establishing a zebrafish model of JAGN1-CN. We found 2 paralogs of the human JAGN1 gene, namely jagn1a and jagn1b, which play distinct roles during zebrafish hematopoiesis. Using various approaches such as morpholino-based knockdown, CRISPR/Cas9-based gene editing, and misexpression of a jagn1b harboring a specific human mutation, we successfully developed neutropenia while leaving other hematopoietic lineages unaffected. Further analysis of our model revealed significant upregulation of apoptosis and genes involved in the unfolded protein response (UPR). However, neither UPR nor apoptosis is the primary mechanism that leads to neutropenia in zebrafish. Instead, Jagn1b has a critical role in granulocyte colony-stimulating factor receptor signaling and steady-state granulopoiesis, shedding light on the pathogenesis of neutropenia associated with JAGN1 mutations. The establishment of a zebrafish model for JAGN1-CN represents a significant advancement in understanding the specific pathologic pathways underlying the disease. This model provides a valuable in vivo tool for further investigation and exploration of potential therapeutic strategies.</p>","PeriodicalId":9228,"journal":{"name":"Blood advances","volume":null,"pages":null},"PeriodicalIF":7.4000,"publicationDate":"2024-08-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11342096/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Blood advances","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1182/bloodadvances.2023011656","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"HEMATOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

Abstract: A variety of autosomal recessive mutations in the JAGN1 gene cause severe congenital neutropenia (CN). However, the underlying pathomechanism remains poorly understood, mainly because of the limited availability of primary hematopoietic stem cells from JAGN1-CN patients and the absence of animal models. In this study, we aimed to address these limitations by establishing a zebrafish model of JAGN1-CN. We found 2 paralogs of the human JAGN1 gene, namely jagn1a and jagn1b, which play distinct roles during zebrafish hematopoiesis. Using various approaches such as morpholino-based knockdown, CRISPR/Cas9-based gene editing, and misexpression of a jagn1b harboring a specific human mutation, we successfully developed neutropenia while leaving other hematopoietic lineages unaffected. Further analysis of our model revealed significant upregulation of apoptosis and genes involved in the unfolded protein response (UPR). However, neither UPR nor apoptosis is the primary mechanism that leads to neutropenia in zebrafish. Instead, Jagn1b has a critical role in granulocyte colony-stimulating factor receptor signaling and steady-state granulopoiesis, shedding light on the pathogenesis of neutropenia associated with JAGN1 mutations. The establishment of a zebrafish model for JAGN1-CN represents a significant advancement in understanding the specific pathologic pathways underlying the disease. This model provides a valuable in vivo tool for further investigation and exploration of potential therapeutic strategies.

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
与 JAGN1 相关的严重先天性中性粒细胞减少症斑马鱼模型显示,G-CSFR 信号传导和 UPR 激活发生了改变。
JAGN1基因的多种常染色体隐性突变会导致严重的先天性中性粒细胞减少症(CN)。然而,由于从 JAGN1-CN 患者身上获得的原始造血干细胞有限以及缺乏动物模型,人们对其潜在的病理机制仍然知之甚少。在本研究中,我们旨在通过建立 JAGN1-CN 的斑马鱼模型来解决这些局限性。我们发现了人类 JAGN1 基因的两个旁系亲属:jagn1a 和 jagn1b,它们在斑马鱼造血过程中发挥着不同的作用。我们采用了多种方法,如基于吗啉基的基因敲除、基于 CRISPR/Cas9 的基因编辑以及误表达携带特定人类突变的 jagn1b,成功地培养出了中性粒细胞减少症,而其他造血系则不受影响。我们对模型的进一步分析表明,细胞凋亡和参与未折叠蛋白反应(UPR)的基因明显上调。然而,UPR 和细胞凋亡都不是导致斑马鱼中性粒细胞减少症的主要机制。相反,Jagn1b 在 G-CSFR 信号转导和稳态粒细胞生成中起着关键作用,这揭示了与 JAGN1 基因突变相关的中性粒细胞减少症的发病机制。JAGN1-CN斑马鱼模型的建立是了解该疾病特定病理途径的重大进展。该模型为进一步研究和探索潜在的治疗策略提供了宝贵的体内工具。
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
Blood advances
Blood advances Medicine-Hematology
CiteScore
12.70
自引率
2.70%
发文量
840
期刊介绍: Blood Advances, a semimonthly medical journal published by the American Society of Hematology, marks the first addition to the Blood family in 70 years. This peer-reviewed, online-only, open-access journal was launched under the leadership of founding editor-in-chief Robert Negrin, MD, from Stanford University Medical Center in Stanford, CA, with its inaugural issue released on November 29, 2016. Blood Advances serves as an international platform for original articles detailing basic laboratory, translational, and clinical investigations in hematology. The journal comprehensively covers all aspects of hematology, including disorders of leukocytes (both benign and malignant), erythrocytes, platelets, hemostatic mechanisms, vascular biology, immunology, and hematologic oncology. Each article undergoes a rigorous peer-review process, with selection based on the originality of the findings, the high quality of the work presented, and the clarity of the presentation.
期刊最新文献
Acalabrutinib plus venetoclax and rituximab in treatment-naive mantle cell lymphoma: 2-year safety and efficacy analysis. Air pollutant impact on disease characteristics and outcomes in patients with acute myeloid leukemia. Safety and efficacy of luspatercept for the treatment of anemia in patients with myelofibrosis. Comparison of treatment response measures in cutaneous sclerosis after allogeneic hematopoietic cell transplantation. CMV reactivation during pretransplantation evaluation: a novel risk factor for posttransplantation CMV reactivation.
×
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