cardiac neural crest

心脏神经嵴
  • 文章类型: Journal Article
    背景:在胚胎发生过程中,心脏神经c衍生细胞(NC)迁移到咽弓并产生咽弓动脉(PAA)的血管平滑肌细胞(vSMC)。vSMC对于将PAA重塑为最终的成人配置至关重要,引起主动脉弓及其动脉(AAAs)。
    结果:我们使用谱系特异性诱导型小鼠品系研究了SMAD4在NC到vSMC分化中的作用。我们发现SMAD4在NC中的表达对于调节心脏NC的存活是不可磨灭的。尽管在NC谱系中E9.5处消融SMAD4导致咽弓中几乎完全没有NC,PAA投资于来自补偿性来源的vSMC。在E16.5对AAA发育的分析表明,替代的vSMC来源弥补了NC衍生的vSMC的缺乏,并挽救了AAA形态发生。
    结论:我们的研究揭示了SMAD4在NC对咽弓间质的贡献中的必要作用。我们发现,在没有SMAD4+NC的情况下,PAA周围的vSMC来自不同的祖先来源,拯救AAA形态发生。这些发现揭示了控制AAA发展的发展机制的显着可塑性。
    BACKGROUND: During embryogenesis, cardiac neural crest-derived cells (NCs) migrate into the pharyngeal arches and give rise to the vascular smooth muscle cells (vSMCs) of the pharyngeal arch arteries (PAAs). vSMCs are critical for the remodeling of the PAAs into their final adult configuration, giving rise to the aortic arch and its arteries (AAAs).
    RESULTS: We investigated the role of SMAD4 in NC-to-vSMC differentiation using lineage-specific inducible mouse strains. We found that the expression of SMAD4 in the NC is indelible for regulating the survival of cardiac NCs. Although the ablation of SMAD4 at E9.5 in the NC lineage led to a near-complete absence of NCs in the pharyngeal arches, PAAs became invested with vSMCs derived from a compensatory source. Analysis of AAA development at E16.5 showed that the alternative vSMC source compensated for the lack of NC-derived vSMCs and rescued AAA morphogenesis.
    CONCLUSIONS: Our studies uncovered the requisite role of SMAD4 in the contribution of the NC to the pharyngeal arch mesenchyme. We found that in the absence of SMAD4+ NCs, vSMCs around the PAAs arose from a different progenitor source, rescuing AAA morphogenesis. These findings shed light on the remarkable plasticity of developmental mechanisms governing AAA development.
    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
    神经c(NC)是脊椎动物胚胎发生过程中源于背侧神经管的多能且暂时迁移的细胞群。心脏神经c细胞(NCC),NC的指定子群体,对正常的心血管发育至关重要,因为它们对咽弓动脉有很大的贡献,发育中的心脏流出道(OFT),心脏瓣膜,和室间隔.各种信号通路被证明可以协调适当的迁移,压实,和心脏NCCs在心血管发育过程中的分化。心脏NCCs信号通路的任何丢失或失调都可能导致胚胎发生过程中的异常心血管发育。导致异常分类为先天性心脏缺陷(CHD)。本文综述了心脏NCCs对心血管形成的贡献。讨论了由各种调节因素的破坏引起的心脏缺陷,并总结了多种信号通路在胚胎发育过程中的作用。对心脏NC及其庞大的调节网络的更好理解将提供对相关异常机制的更深入的了解。导致潜在的治疗进步。
    The neural crest (NC) is a multipotent and temporarily migratory cell population stemming from the dorsal neural tube during vertebrate embryogenesis. Cardiac neural crest cells (NCCs), a specified subpopulation of the NC, are vital for normal cardiovascular development, as they significantly contribute to the pharyngeal arch arteries, the developing cardiac outflow tract (OFT), cardiac valves, and interventricular septum. Various signaling pathways are shown to orchestrate the proper migration, compaction, and differentiation of cardiac NCCs during cardiovascular development. Any loss or dysregulation of signaling pathways in cardiac NCCs can lead to abnormal cardiovascular development during embryogenesis, resulting in abnormalities categorized as congenital heart defects (CHDs). This review focuses on the contributions of cardiac NCCs to cardiovascular formation, discusses cardiac defects caused by a disruption of various regulatory factors, and summarizes the role of multiple signaling pathways during embryonic development. A better understanding of the cardiac NC and its vast regulatory network will provide a deeper insight into the mechanisms of the associated abnormalities, leading to potential therapeutic advancements.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    心脏神经c细胞出现在尾后脑,然后通过咽弓迁移到心脏。这些细胞有助于心脏的形成,包括流出道,并且是这个神经脊群体所独有的。MafB是在早期迁移的心脏神经c细胞以及菱形(r)5和6中特异性表达的转录因子。这里,我们确定了鸡基因组中控制内源性MafB转录物表达的调控区,并在报告基因测定中使用这些必需元件在心脏神经c中表达MafB.由这个调控区驱动的报告子被用来追踪这些细胞迁移到咽弓中。该调控区在心脏神经c中表现出转录活性,但在其他神经c细胞亚群中却没有。例如颅骨和躯干细胞。这项研究提供了对神经c细胞群体中指定心脏神经c细胞的基因调控机制的见解。
    Cardiac neural crest cells arise in the caudal hindbrain and then migrate to the heart through the pharyngeal arches. These cells contribute to the formation of the heart, including the outflow tract, and are unique to this neural crest population. MafB is a transcription factor expressed specifically in early migrating cardiac neural crest cells as well as in rhombomeres (r) 5 and 6. Here, we identified the regulatory region in the chicken genome controlling the expression of endogenous MafB transcripts and used these essential elements to express MafB in the cardiac neural crest in reporter assays. A reporter driven by this regulatory region was employed to trace the migration of these cells into the pharyngeal arches. This regulatory region demonstrated transcriptional activity in the cardiac neural crest but not in other neural crest cell subpopulations, such as the cranial and trunk cells. This study provides insights into the gene regulatory mechanisms that specify cardiac neural crest cells among neural crest cell populations.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    背景:神经c是早期胚胎发生中存在的一种瞬时结构。头颅神经c细胞迁移到咽弓和额鼻突中,成为前额和中面部结构。它们还有助于形成威利斯环及其分支的动脉介质。心脏神经峰在升主动脉中产生血管平滑肌细胞,心脏隔膜和冠状动脉。
    方法:在这篇综述中,我们从神经c细胞分布的角度评估了神经c在烟雾病中的作用以及烟雾病和心血管疾病并发的病理意义。
    结果:中线颅面和中枢神经系统异常伴眼睛异常,烟雾病患者的牵牛花椎间盘异常都可以解释为头神经病变的亚型。Further,烟雾病与心脏表现(先天性心脏缺陷和冠状动脉疾病)之间的关联也有报道.头部神经c和心脏神经c都有助于这些并发动脉疾病,作为心头神经病。
    结论:头/心-头神经病的概念为理解潜在的病因关联和开发未来合并烟雾病和心血管疾病的治疗方法提供了新的视角。
    BACKGROUND: The neural crest is a transient structure present in early embryogenesis. Cephalic neural crest cells migrate into the pharyngeal arches and the frontonasal process that becomes the forehead and midfacial structures. They also contribute to forming the media of the arteries of the circle of Willis and their branches. The cardiac neural crest produces vascular smooth muscle cells in the ascending aorta, cardiac septum and coronary arteries.
    METHODS: In this review, we evaluate the role of the neural crest in moyamoya disease and the pathological implications from the concurrence of moyamoya disease and cardiovascular diseases from the point of view of neural crest cell distributions.
    RESULTS: Midline craniofacial and central nervous system anomalies with eye anomalies, morning glory disc anomaly in patients with moyamoya disease can both be explained as a subtype of cephalic neurocristopathy. Further, the association between moyamoya disease and cardiac manifestations (congenital cardiac defects and coronary artery disease) have also been reported. Both the cephalic neural crest and cardiac neural crest contribute to these concurrent arterial diseases, as cardio-cephalic neurocristopathy.
    CONCLUSIONS: The concept of cephalic/cardio-cephalic neurocristopathy provides a new perspective to understanding the underlying aetiological associations and to developing future therapeutic approaches for concomitant moyamoya disease and cardiovascular diseases.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    影响心脏流出道(OFT)的先天性心脏缺陷(CHD)是发病率和死亡率的重要原因。OFT从包括心脏神经c细胞(cNCC)和次级心脏视野(SHF)衍生的心肌和心内膜的迁移细胞群发展而来。相关转录因子HAND1和HAND2与涉及OFT的人类CHD有关。尽管Hand1在OFT中表示,Hand1NCC特异性条件性敲除小鼠(H1CKOs)是可行的。在这里,我们显示这些H1CKO呈现OFT表型的低外显率,而SHF特异性Hand1消融不显示任何心脏表型。Further,HAND1和HAND2在cNCC中出现功能冗余,由于Hand2在NCC特异性H1CKO背景上的减少/消融导致明显的OFT缺陷。双条件Hand1和Hand2NCC敲除表现出持续的动脉干(PTA),外显率为100%。NCC谱系追踪和Sema3c原位mRNA表达表明,Sema3c表达细胞定位错误,导致S1CKO;H2CKO胚胎的OFTs内出现畸形的间隔桥。有趣的是,Hand1和Hand2也在SHF内发生遗传相互作用,由于Hand2杂合背景上的SHFH1CKOs表现出室间隔缺损(VSD),外显率不完全。以前,我们确定了一个BMP,HAND2和GATA依赖性Hand1OFT增强子足以驱动新生OFT和主动脉内的报告基因表达。使用这些转录输入作为探针,我们确定了一种新的Hand2OFT增强子,提示保守的BMP-GATA依赖性机制在转录上调节两种HAND因子。这些发现支持HAND因子解释cNCC内的BMP信号以协同协调OFT形态发生的假设。
    Congenital heart defects (CHDs) affecting the cardiac outflow tract (OFT) constitute a significant cause of morbidity and mortality. The OFT develops from migratory cell populations which include the cardiac neural crest cells (cNCCs) and secondary heart field (SHF) derived myocardium and endocardium. The related transcription factors HAND1 and HAND2 have been implicated in human CHDs involving the OFT. Although Hand1 is expressed within the OFT, Hand1 NCC-specific conditional knockout mice (H1CKOs) are viable. Here we show that these H1CKOs present a low penetrance of OFT phenotypes, whereas SHF-specific Hand1 ablation does not reveal any cardiac phenotypes. Further, HAND1 and HAND2 appear functionally redundant within the cNCCs, as a reduction/ablation of Hand2 on an NCC-specific H1CKO background causes pronounced OFT defects. Double conditional Hand1 and Hand2 NCC knockouts exhibit persistent truncus arteriosus (PTA) with 100% penetrance. NCC lineage-tracing and Sema3c in situ mRNA expression reveal that Sema3c-expressing cells are mis-localized, resulting in a malformed septal bridge within the OFTs of H1CKO;H2CKO embryos. Interestingly, Hand1 and Hand2 also genetically interact within the SHF, as SHF H1CKOs on a heterozygous Hand2 background exhibit Ventricular Septal Defects (VSDs) with incomplete penetrance. Previously, we identified a BMP, HAND2, and GATA-dependent Hand1 OFT enhancer sufficient to drive reporter gene expression within the nascent OFT and aorta. Using these transcription inputs as a probe, we identify a novel Hand2 OFT enhancer, suggesting that a conserved BMP-GATA dependent mechanism transcriptionally regulates both HAND factors. These findings support the hypothesis that HAND factors interpret BMP signaling within the cNCCs to cooperatively coordinate OFT morphogenesis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    双叶主动脉瓣(BAV)是最常见的先天性心脏畸形,常伴有主动脉病变和瓣膜病变。怀疑BAV的先天性起源会影响成年生活中疾病的发展。在过去的十年里,大量有关先天性心脏病胚胎发育的研究显著提高了我们对BAV病因学的认识.他们描述了发育缺陷,在分子上,细胞和形态学水平,导致先天性心脏畸形,包括BAV,在动物模型中。这些模型由自发仓鼠和几种小鼠模型组成,这些小鼠模型在属于多种途径的基因中具有不同的遗传操作。在这篇综述论文中,我们的目标是收集这些动物模型中导致BAV形成的发育缺陷的信息,为了初步解释表征人类BAV的瓣膜形态谱的形态发生起源。BAV可能是小鼠基因操作导致的唯一缺陷,但通常表现为一系列畸形中较不严重的缺陷,最常影响心脏流出道。导致BAV改变的基因属于不同的遗传途径,但其中许多与NOTCH途径直接或间接相关。这些分子改变影响心脏发育过程中的三种基本细胞机制,即,心内膜到间质转化,心脏神经峰(CNC)细胞行为和瓣膜垫间充质细胞分化。有缺陷的细胞功能影响三种可能的形态发生机制,即,流出道心内膜垫形成,流出道分隔和阀垫开挖。虽然心内膜垫异常通常会导致侧外侧BAV和前后BAV的间隔缺损,垫层开挖的改变可能会导致两种BAV类型。原始缺损的严重程度很可能决定了特定的主动脉瓣表型,其中包括连合融合和融合。根据目前对心脏流出道发育机制的了解,我们提出了BAV形成的统一假设,基于CNC细胞在BAV发育的三种机制中的诱导作用。在三个可能的替代关键瓣膜生成过程中,CNC细胞行为的改变可能导致BAV的整个范围。
    Bicuspid aortic valve (BAV) is the most common congenital cardiac malformation, frequently associated with aortopathies and valvulopathies. The congenital origin of BAV is suspected to impact the development of the disease in the adult life. During the last decade, a number of studies dealing with the embryonic development of congenital heart disease have significantly improved our knowledge on BAV etiology. They describe the developmental defects, at the molecular, cellular and morphological levels, leading to congenital cardiac malformations, including BAV, in animal models. These models consist of a spontaneous hamster and several mouse models with different genetic manipulations in genes belonging to a variety of pathways. In this review paper, we aim to gather information on the developmental defects leading to BAV formation in these animal models, in order to tentatively explain the morphogenetic origin of the spectrum of valve morphologies that characterizes human BAV. BAV may be the only defect resulting from gene manipulation in mice, but usually it appears as the less severe defect of a spectrum of malformations, most frequently affecting the cardiac outflow tract. The genes whose alterations cause BAV belong to different genetic pathways, but many of them are direct or indirectly associated with the NOTCH pathway. These molecular alterations affect three basic cellular mechanisms during heart development, i.e., endocardial-to-mesenchymal transformation, cardiac neural crest (CNC) cell behavior and valve cushion mesenchymal cell differentiation. The defective cellular functions affect three possible morphogenetic mechanisms, i.e., outflow tract endocardial cushion formation, outflow tract septation and valve cushion excavation. While endocardial cushion abnormalities usually lead to latero-lateral BAVs and septation defects to antero-posterior BAVs, alterations in cushion excavation may give rise to both BAV types. The severity of the original defect most probably determines the specific aortic valve phenotype, which includes commissural fusions and raphes. Based on current knowledge on the developmental mechanisms of the cardiac outflow tract, we propose a unified hypothesis of BAV formation, based on the inductive role of CNC cells in the three mechanisms of BAV development. Alterations of CNC cell behavior in three possible alternative key valvulogenic processes may lead to the whole spectrum of BAV.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    心脏神经峰出现在后脑,然后迁移到心脏并有助于关键结构,包括流出道隔膜。小鸡心冠消融导致这种中隔失败,人类心脏缺陷持续性动脉干(PTA)的表型,躯干神经脊未能获救。这里,我们探讨了心脊独特潜力的分子机制。转录谱分析鉴定了心脊特异性转录因子,单细胞RNA测序揭示了惊人的异质性,包括早期迁移种群中的外植体间充质亚群。功能缺失分析揭示了一个转录子电路,由Tgif1,Ets1和Sox8组成,对心脏神经c和心脏发育至关重要。重要的是,该子回路的异位表达足以使主干c具有在心脏c消融后挽救PTA的能力。一起,我们的结果揭示了一个转录程序,足以将心脏电位赋予躯干神经c细胞,因此,新的基因与心血管出生缺陷有关。
    The cardiac neural crest arises in the hindbrain, then migrates to the heart and contributes to critical structures, including the outflow tract septum. Chick cardiac crest ablation results in failure of this septation, phenocopying the human heart defect persistent truncus arteriosus (PTA), which trunk neural crest fails to rescue. Here, we probe the molecular mechanisms underlying the cardiac crest\'s unique potential. Transcriptional profiling identified cardiac-crest-specific transcription factors, with single-cell RNA sequencing revealing surprising heterogeneity, including an ectomesenchymal subpopulation within the early migrating population. Loss-of-function analyses uncovered a transcriptional subcircuit, comprised of Tgif1, Ets1, and Sox8, critical for cardiac neural crest and heart development. Importantly, ectopic expression of this subcircuit was sufficient to imbue trunk crest with the ability to rescue PTA after cardiac crest ablation. Together, our results reveal a transcriptional program sufficient to confer cardiac potential onto trunk neural crest cells, thus implicating new genes in cardiovascular birth defects.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Letter
    Cardiac neural crest cells (cNCCs) are required for normal heart development. cNCCs are a multipotent and migratory cell lineage that differentiates into multiple cell types. cNCCs migrate into the developing heart to contribute to the septation of the cardiac outflow tract (OFT). Foxc1 and Foxc2 are closely related members of the FOX (Forkhead box) transcription factor family and are expressed in cNCC during heart development. However, the precise role of Foxc1 and Foxc2 in cNCCs has yet to be fully described. We found that compound NCC-specific Foxc1;Foxc2 mutant embryos exhibited persistent truncus arteriosus (PTA), ventricular septal defects (VSDs), and thinning of the ventricular myocardium. Loss of Foxc1/c2 expression in cNCCs resulted in abnormal patterns of cNCC migration into the OFT without the formation of the aorticopulmonary septum. Further, loss of Foxc1 expression in cNCCs resulted in normal OFT development but abnormal ventricular septal formation. In contrast, loss of Foxc2 expression in NCCs led to no obvious cardiac abnormalities. Together, we provide evidence that Foxc1 and Foxc2 in cNCCs are cooperatively required for proper cNCC migration, the formation of the OFT septation, and the development of the ventricles. Our data also suggests that Foxc1 expression may play a larger role in ventricular development compared to Foxc2.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    Proper development of the great vessels of the heart and septation of the cardiac outflow tract requires cardiac neural crest cells. These cells give rise to the parasympathetic cardiac ganglia, the smooth muscle layer of the great vessels, some cardiomyocytes, and the conotruncal cushions and aorticopulmonary septum of the outflow tract. Ablation of cardiac neural crest cells results in defective patterning of each of these structures. Previous studies have shown that targeted deletion of the forkhead transcription factor C2 (Foxc2), results in cardiac phenotypes similar to that derived from cardiac neural crest cell ablation.
    We report that Foxc2-/- embryos on the 129s6/SvEv inbred genetic background display persistent truncus arteriosus and hypoplastic ventricles before embryonic lethality. Foxc2 loss-of-function resulted in perturbed cardiac neural crest cell migration and their reduced contribution to the outflow tract as evidenced by lineage tracing analyses together with perturbed expression of the neural crest cell markers Sox10 and Crabp1. Foxc2 loss-of-function also resulted in alterations in PlexinD1, Twist1, PECAM1, and Hand1/2 expression in association with vascular and ventricular defects.
    Our data indicate Foxc2 is required for proper migration of cardiac neural crest cells, septation of the outflow tract, and development of the ventricles. Developmental Dynamics 247:1286-1296, 2018. © 2018 Wiley Periodicals, Inc.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    The cardiac neural crest originates in the caudal hindbrain, migrates to the heart, and contributes to septation of the cardiac outflow tract and ventricles, an ability unique to this neural crest subpopulation. Here we have used a FoxD3 neural crest enhancer to isolate a pure population of cardiac neural crest cells for transcriptome analysis. This has led to the identification of transcription factors, signaling receptors/ligands, and cell adhesion molecules upregulated in the early migrating cardiac neural crest. We then functionally tested the role of one of the upregulated transcription factors, MafB, and found that it acts as a regulator of Sox10 expression specifically in the cardiac neural crest. Our results not only reveal the genome-wide profile of early migrating cardiac neural crest cells, but also provide molecular insight into what makes the cardiac neural crest unique.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号