FOXD3

FOXD3
  • 文章类型: Journal Article
    染色体1p32p31缺失综合征是一种连续的基因疾病,具有可变的表型,其特征是有或没有尿路缺陷的脑畸形,除了神经发育迟缓和畸形。基于其他发现提出了扩展的表型,包括以前的一份关于烟雾病患者的报告。
    作者报告了一位患有早期神经发育迟缓的患者,脑积水,肾畸形,和畸形。在出现突然的舞蹈运动障碍后,神经影像学检查显示缺血性中风,烟雾病,双侧不完全海马倒置。染色体微阵列分析显示,在1p31.3p32.2处缺失13.2Mb,与该区域微缺失引起的连续基因综合征相容。
    这是一例发展为烟雾病的患者的第二份报告,也是第一次描述这种微缺失综合征中的双侧海马不完全倒置。
    UNASSIGNED: The chromosome 1p32p31 deletion syndrome is a contiguous gene disorder with a variable phenotype characterized by brain malformations with or without urinary tract defects, besides neurodevelopmental delay and dysmorphisms. An expanded phenotype was proposed based on additional findings, including one previous report of a patient presenting with moyamoya disease.
    UNASSIGNED: The authors report a patient presenting with early neurodevelopmental delay, hydrocephalus, renal malformation, and dysmorphisms. After presenting with a sudden choreic movement disorder, the neuroimaging investigation revealed an ischemic stroke, moyamoya disease, and bilateral incomplete hippocampal inversion. Chromosomal microarray analysis revealed a deletion of 13.2 Mb at 1p31.3p32.2, compatible with the contiguous gene syndrome caused by microdeletions of this region.
    UNASSIGNED: This is the second report of a patient who developed Moyamoya disease and the first to describe bilateral incomplete hippocampal inversion in this microdeletion syndrome.
    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
    胃肠道间质瘤(GIST)是间质瘤,据信起源于Cajal(ICC)的间质细胞,通常由酪氨酸激酶受体(TKR)KIT或PDGFRA的过表达引起。这里,我们提供的证据表明,胚胎干细胞因子FOXD3,首先被确定为“创世纪”和功能在胃肠道和神经c细胞发育,与GIST发病机理有关;在体外,斑马鱼和FOXD3缺乏症的小鼠模型中都研究了其参与。共58例野生型GIST患者的样本用于分子分析,包括桑格序列,CGH和甲基化。使用免疫组织化学和Western印迹评估来获得FOXD3表达。此外,我们在组织样本和转染细胞中进行了体外功能研究,以确认鉴定的遗传变异的致病性。种系部分失活FOXD3序列变体(p。在分离的GIST患者中发现了R54H和p.Ala88_Gly91del)。染色体1p缺失是肿瘤中最常见的染色体异常。体外实验证明了在这些变体存在下FOXD3的损害。动物研究显示GI神经网络的破坏以及ICC中数量和分布的变化。FOXD3抑制人细胞中的KIT表达;其失活导致斑马鱼中ICC的增加,和老鼠一样,为FOXD3缺陷和KIT过表达之间的功能联系提供证据导致GIST形成。
    Gastrointestinal stromal tumors (GISTs) are mesenchymal neoplasms, believed to originate from the interstitial cells of Cajal (ICC), often caused by overexpression of tyrosine kinase receptors (TKR) KIT or PDGFRA. Here, we present evidence that the embryonic stem cell factor FOXD3, first identified as \'Genesis\' and involved in both gastrointestinal and neural crest cell development, is implicated in GIST pathogenesis; its involvement is investigated both in vitro and in zebrafish and a mouse model of FOXD3 deficiency. Samples from a total of 58 patients with wild-type GISTs were used for molecular analyses, including Sanger sequencing, comparative genomic hybridization, and methylation analysis. Immunohistochemistry and western blot evaluation were used to assess FOXD3 expression. Additionally, we conducted in vitro functional studies in tissue samples and in transfected cells to confirm the pathogenicity of the identified genetic variants. Germline partially inactivating FOXD3 sequence variants (p.R54H and p.Ala88_Gly91del) were found in patients with isolated GISTs. Chromosome 1p loss was the most frequent chromosomal abnormality identified in tumors. In vitro experiments demonstrate the impairment of FOXD3 in the presence of those variants. Animal studies showed disruption of the GI neural network and changes in the number and distribution in the ICC. FOXD3 suppresses KIT expression in human cells; its inactivation led to an increase in ICC in zebrafish, as well as mice, providing evidence for a functional link between FOXD3 defects and KIT overexpression leading to GIST formation.
    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
    背景:结直肠癌(CRC)治疗的改善和生存时间的延长改善了骨转移的发生率。叉头盒D3(FOXD3)涉及CRC的开发。然而,FOXD3在CRC骨转移发展中的作用和机制尚不清楚。
    目的:结合生物信息学和细胞学实验分析,本研究旨在探讨FOXD3在结肠癌骨转移中的作用机制,从而有助于结肠癌骨转移的治疗和药物靶向标志物的鉴定。
    方法:首先,从癌症基因组图谱(TCGA)数据库获得结肠癌样本和结肠癌转移灶之间FOXD3基因和差异表达基因(DEGs)表达水平的变化.然后,鉴定了FOXD3基因与DEGs的相关性。接下来,使用细胞计数Kit-8(CCK8)和Transwell细胞迁移试验鉴定FOXD3对结肠癌骨转移细胞增殖和侵袭能力的影响,分别。此外,蛋白质印迹分析用于鉴定与EGFR/Ras/Raf/MEK/ERK(EGFR/ERK)信号通路和上皮-间质转化(EMT)相关的蛋白的表达水平。
    结果:FOXD3在结肠癌中表达下调,并可与结肠癌骨转移中的多个DEGs相互作用。FOXD3基因敲低可增加人结肠癌骨转移细胞的增殖及其侵袭能力。FOXD3基因敲低可以激活EGFR/ERK信号通路相关蛋白的表达,抑制/促进EMT相关蛋白的表达,进而促进结肠癌骨转移瘤LoVo细胞的增殖和转移。
    结论:总体而言,这项研究表明,FOXD3基因的下调可能通过EGFR/ERK通路促进结肠癌骨转移细胞系的增殖,并通过EMT促进其迁移,从而作为一个有希望的治疗目标。
    The improvements in the treatment of colorectal cancer (CRC) and prolongation of survival time have improved the incidence of bone metastasis. Forkhead box D3 (FOXD3) is involved in the development of CRC. However, the role and mechanism of FOXD3 in CRC bone metastases development are unknown.
    Using the combined bioinformatics and cytology experimental analyses, this study aimed to explore the mechanistic role of FOXD3 in the bone metastasis of colon cancer, thereby aiding in the treatment of colon cancer bone metastasis and identification of drug-targeting markers.
    First, the changes in the expression levels of the FOXD3 gene and differentially expressed genes (DEGs) between the colon cancer samples and colon cancer metastases were obtained from The Cancer Genome Atlas (TCGA) database. Then, the correlations of the FOXD3 gene with the DEGs were identified. Next, the effects of the FOXD3 on the proliferation and invasion abilities of colon cancer bone metastatic cells were identified using Cell Counting Kit-8 (CCK8) and Transwell cell migration assays, respectively. In addition, Western blot analysis was used to identify the expression levels of the proteins related to the EGFR/Ras/Raf/MEK/ERK (EGFR/ERK) signaling pathway and epithelial-to-mesenchymal transition (EMT).
    FOXD3 was downregulated in colon cancer and could interact with multiple DEGs in colon cancer bone metastases. FOXD3 gene knockdown could increase the proliferation of human colon cancer bone metastatic cells and their invasive ability. FOXD3 gene knockdown could activate the expression of EGFR/ERK signaling pathway-related proteins and inhibit/promote the expression of EMT-related proteins, which in turn promoted the proliferation and metastasis of LoVo cells from colon cancer bone metastases.
    Overall, this study demonstrated that the downregulation of the FOXD3 gene might promote the proliferation of colon cancer bone metastatic cell lines through the EGFR/ERK pathway and promote their migration through EMT, thereby serving as a promising therapeutic target.
    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
    The mechanisms of neural crest cell induction and specification are highly conserved among vertebrate model organisms, but how similar these mechanisms are in mammalian neural crest cell formation remains open to question. The zinc finger of the cerebellum 1 (ZIC1) transcription factor is considered a core component of the vertebrate gene regulatory network that specifies neural crest fate at the neural plate border. In mouse embryos, however, Zic1 mutation does not cause neural crest defects. Instead, we and others have shown that murine Zic2 and Zic5 mutate to give a neural crest phenotype. Here, we extend this knowledge by demonstrating that murine Zic3 is also required for, and co-operates with, Zic2 and Zic5 during mammalian neural crest specification. At the murine neural plate border (a region of high canonical WNT activity) ZIC2, ZIC3, and ZIC5 function as transcription factors to jointly activate the Foxd3 specifier gene. This function is promoted by SUMOylation of the ZIC proteins at a conserved lysine immediately N-terminal of the ZIC zinc finger domain. In contrast, in the lateral regions of the neurectoderm (a region of low canonical WNT activity) basal ZIC proteins act as co-repressors of WNT/TCF-mediated transcription. Our work provides a mechanism by which mammalian neural crest specification is restricted to the neural plate border. Furthermore, given that WNT signaling and SUMOylation are also features of non-mammalian neural crest specification, it suggests that mammalian neural crest induction shares broad conservation, but altered molecular detail, with chicken, zebrafish, and Xenopus neural crest induction.
    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
    Background and Aim: Some studies have verified that miR-133a played an inhibitory role in several cancers. Whereas, the effect of miRNA-133a in colorectal cancer (CRC) has not been fully elucidated. Our study aims to confirm UBA2 as a direct target gene of miRNA-133a and explore the upstream modulatory molecules of miR-133a. In addition, their impacts on the biological characteristics of CRC cells were assessed. Methods: QRT-PCR analyzed miR-133a expression levels in colorectal cells including HCT116, SW48 cells and human normal colorectal cell line NCM460. A serial biological experiment assessed miR-133a effects on cell proliferation, migration, invasion and apoptosis capacities in HCT116 and SW48 cells. MiRNA targeting gene prediction and a dual luciferase assay were employed to confirm miR-133a-targeted UBA2. Transcription factors (TFs) FOXD3 was identified as an upstream regulator of miR-133a via JASPAR. The influence of miR-133a and FOXD3 on UBA2 expression was analyzed by qRT-PCR or western blot. Results: miR-133a was lowly expressed in CRC cells. High miRNA-133a expression suppressed the proliferation, migration, invasion and enhanced apoptosis capacities of CRC cells. MiR-133a targeted the UBA2 mRNA 3\'UTR area and reduced UBA2 protein expression. We also unveiled that FOXD3 high-expression significantly raised miR-133a expression and diminished UBA2 expression. We also discovered that high miR-133a expression augmented the effects of elevated FOXD3 expression on CRC cell proliferation, migration and invasion, whereas, low miR-133a expression generated the opposite outcomes. Conclusion: FOXD3 induced miRNA-133a directly targeting UBA2 could affect the progression and growth of CRC.
    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
    在收敛分化期间,多个发育谱系产生高度相似或相同的细胞类型。然而,很少有驱动收敛分化的分子参与者是已知的。这里,我们表明,秀丽隐杆线虫Forkhead转录因子UNC-130仅在产生相同神经胶质细胞类型的三个会聚谱系之一中是必需的。UNC-130在祖细胞和新出生的末端细胞中暂时充当阻遏物,以允许通过谱系而不是细胞类型或功能相关的细胞的适当规格。规格缺陷与UNC-130相关:DNA结合,UNC-130可以在功能上被它的人类同源物取代,神经c谱系决定因子FoxD3。我们建议,与激活终末分化细胞中细胞类型特异性转录程序的终末选择相反,UNC-130在一个会聚谱系中早期且特异性地起作用以产生也由其他谱系中的分子不同的祖细胞产生的细胞类型。
    During convergent differentiation, multiple developmental lineages produce a highly similar or identical cell type. However, few molecular players that drive convergent differentiation are known. Here, we show that the C. elegans Forkhead transcription factor UNC-130 is required in only one of three convergent lineages that produce the same glial cell type. UNC-130 acts transiently as a repressor in progenitors and newly-born terminal cells to allow the proper specification of cells related by lineage rather than by cell type or function. Specification defects correlate with UNC-130:DNA binding, and UNC-130 can be functionally replaced by its human homolog, the neural crest lineage determinant FoxD3. We propose that, in contrast to terminal selectors that activate cell type-specific transcriptional programs in terminally differentiating cells, UNC-130 acts early and specifically in one convergent lineage to produce a cell type that also arises from molecularly distinct progenitors in other lineages.
    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
    The neural crest (NC) is a transient multipotent cell population that migrates extensively to produce a remarkable array of vertebrate cell types. NC cell specification progresses in an anterior to posterior fashion, resulting in distinct, axial-restricted subpopulations. The anterior-most, cranial, population of NC is specified as gastrulation concludes and neurulation begins, while more posterior populations become specified as the body elongates. The mechanisms that govern development of the more posterior NC cells remain incompletely understood. Here, we report a key role for zebrafish Cdx4, a homeodomain transcription factor, in the development of posterior NC cells. We demonstrate that cdx4 is expressed in trunk NC cell progenitors, directly binds NC cell-specific enhancers in the NC GRN, and regulates expression of the key NC development gene foxd3 in the posterior body. Moreover, cdx4 mutants show disruptions to the segmental pattern of trunk NC cell migration due to loss of normal leader/follower cell dynamics. Finally, using cell transplantation to generate chimeric specimens, we show that Cdx4 does not function in the paraxial mesoderm-the environment adjacent to which crest migrates-to influence migratory behaviors. We conclude that cdx4 plays a critical, and likely tissue autonomous, role in the establishment of trunk NC migratory behaviors. Together, our results indicate that cdx4 functions as an early NC specifier gene in the posterior body of zebrafish embryos.
    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
    皮肤黑色素瘤是最致命的皮肤癌类型之一。预后在很大程度上取决于疾病阶段,因此,早期发现至关重要。新疗法,包括BRAF和MEK抑制剂和免疫疗法,在过去十年中显著改善了患者的生存率。然而,内在和后天的抵抗仍然是一个挑战。在这次审查中,我们讨论了导致黑色素瘤侵袭性的两个主要方面,即,黑素细胞和黑色素瘤细胞的胚胎起源和细胞可塑性。首先,我们总结了表皮黑素细胞的生理功能及其从神经c(NC)起源的前体细胞的发育。接下来,我们讨论了肿瘤内异质性的概念,细胞可塑性,和表型转换,使黑色素瘤能够适应肿瘤微环境的变化,促进疾病进展和耐药性。最后,我们通过关注转录调节因子MSX1,MITF,SOX10、PAX3和FOXD3。这些因素在NC启动中起着关键作用,NC细胞迁移,和黑素细胞的形成,我们讨论了它们如何促进黑色素瘤的细胞可塑性和耐药性。
    Cutaneous melanoma represents one of the deadliest types of skin cancer. The prognosis strongly depends on the disease stage, thus early detection is crucial. New therapies, including BRAF and MEK inhibitors and immunotherapies, have significantly improved the survival of patients in the last decade. However, intrinsic and acquired resistance is still a challenge. In this review, we discuss two major aspects that contribute to the aggressiveness of melanoma, namely, the embryonic origin of melanocytes and melanoma cells and cellular plasticity. First, we summarize the physiological function of epidermal melanocytes and their development from precursor cells that originate from the neural crest (NC). Next, we discuss the concepts of intratumoral heterogeneity, cellular plasticity, and phenotype switching that enable melanoma to adapt to changes in the tumor microenvironment and promote disease progression and drug resistance. Finally, we further dissect the connection of these two aspects by focusing on the transcriptional regulators MSX1, MITF, SOX10, PAX3, and FOXD3. These factors play a key role in NC initiation, NC cell migration, and melanocyte formation, and we discuss how they contribute to cellular plasticity and drug resistance in melanoma.
    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 neural crest is an important group of cells with pluripotency and migratory ability that is crucially involved in tissue and cell specification during development. Craniofacial shaping, sensory neurons, body asymmetry, and pigmentation are linked to neural crest functionality. Despite its prominent role in embryogenesis, neural crest specification as well as the possible part mitochondria play in such a process remains unclarified. Mitochondria are important organelles not only for respiration, but also for regulation of cell proliferation, differentiation and death. Modulation of mitochondrial fitness and depletion of mitochondrial ATP synthesis has been shown to down-regulate Wnt signaling, both in vitro and in vivo. Since Wnt signaling is one of the crucial players during neural crest induction/specification, we hypothesized a signaling cascade connecting mitochondria to embryonic development and neural crest migration and differentiation. Here, by using pharmacological and genetic modulators of mitochondrial function, we provide evidence that a crosstalk between mitochondrial energy homeostasis and Wnt signaling is important in the development of neural crest-derived tissues. Furthermore, our results highlight the possibility to modulate neural crest cell specification by tuning mitochondrial metabolism via FoxD3, an important transcription factor that is regulated by Wnt. FoxD3 ensures the correct embryonic development and contributes to the maintenance of cell stemness and to the induction of epithelial-to-mesenchymal transition. In summary, our work offers new insights into the molecular mechanism of action of FoxD3 and demonstrates that mitochondrial fitness is linked to the regulation of this important transcription factor via Wnt signaling in the context of neural crest specification.
    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
    以前已经发现FOXD3正向调节miR-26b,一种鼻咽癌(NPC)的肿瘤抑制剂。然而,FOXD3在鼻咽癌中的确切功能和相关作用机制尚未得到研究。在这项研究中,使用RT-qPCR评估FOXD3mRNA和蛋白的表达,西方印迹,和免疫组织化学。蛋白质水平参与磷酸肌醇3-激酶-蛋白激酶B(PI3K-Akt)途径通过蛋白质印迹评估,通过MTT和集落形成测定细胞增殖。此外,通过流式细胞术测定细胞凋亡。最后,使用伤口愈合和Transwell测定法确定NPC细胞的迁移和侵袭能力。我们发现FOXD3水平在NPC组织和细胞中相对较低,而增加引起PI3K-Akt途径的抑制。功能实验发现过表达FOXD3抑制细胞增殖,迁移,NPCC6661细胞的侵袭和增强细胞凋亡。IGF-1,PI3K-Akt途径的激活剂,逆转FOXD3的抑制作用。此外,我们发现PI3K-Akt通路上调,FOXD3对C6661细胞活性的抑制作用上调.总之,FOXD3消极地影响PI3K-Akt通路以克制C6661细胞病理进程。这些发现进一步揭示了FOXD3在NPC中的功能和下游轴,并为NPC治疗显示了一个有希望的新靶点。
    FOXD3 has been found previously to positively regulate miR-26b, a tumor inhibitor of nasopharyngeal carcinoma (NPC). However, FOXD3\'s precise function and associated mechanism of action in NPC have not yet been investigated. In this study, the expression of FOXD3 mRNA and protein was evaluated using RT-qPCR, western blotting, and immunohistochemistry. Protein levels involved in the phosphoinositide 3-kinase - protein kinase B (PI3K-Akt) pathway were assessed by western blot, and cell proliferation was determined by MTT and colony forming assays. Additionally, cell apoptosis was assessed by flow cytometric assay. Finally, the migration and invasion capabilities of the NPC cells were determined using wound healing and Transwell assays. We found that FOXD3 levels were relatively low in NPC tissue and cells, while an increase caused the inhibition of the PI3K-Akt pathway. Functional experiments found that overexpression of FOXD3 suppressed cell proliferation, migration, and invasion and enhanced cell apoptosis in NPC C6661 cells. IGF-1, an activator of the PI3K-Akt pathway, reversed the inhibitory effect of FOXD3. Furthermore, we found upregulation of the PI3K-Akt pathway and upregulation of the inhibitory effects of FOXD3 on C6661 cellular activities. In conclusion, FOXD3 negatively affected the PI3K-Akt pathway to restrain the processes involved in C6661 cell pathology. These findings further exposed the function and downstream axis of FOXD3 in NPC and displayed a promising new target for NPC therapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号