Mef2

MEF2
  • 文章类型: Journal Article
    肌细胞增强因子-2(MEF2)转录因子在协调肌肉分化中起着至关重要的作用。虽然MEF2不能有效诱导幼稚细胞中的肌生成,它可以有效地加速中胚层细胞的肌生成。这包括果蝇的假想盘成肌细胞,在这些成年肌肉祖细胞中触发过早的肌肉基因表达已成为理解肌源性程序调节的范例。这里,我们调查了MEF2过表达在想象翼盘成肌细胞中的整体后果,通过结合RNA测序与RT-qPCR和免疫荧光。我们观察到包含肌肉和细胞质肌球蛋白的肌节样结构的形成,和肌肉基因表达的显著上调,特别是肌原纤维形成和功能所必需的基因。这些转录物具有功能,因为使用免疫荧光在圆盘中检测到许多肌原纤维蛋白。有趣的是,在这些成年成肌细胞中,表达仅限于成年阶段的肌肉基因未被激活。这些研究证实了响应于MEF2表达的肌源性程序的广泛激活,并表明需要其他调节因子来促进成人肌肉特异性程序。我们的发现有助于理解控制肌肉发育的调节机制,并强调MEF2在协调这一复杂过程中的多方面作用。
    The Myocyte enhancer factor-2 (MEF2) transcription factor plays a vital role in orchestrating muscle differentiation. While MEF2 cannot effectively induce myogenesis in naïve cells, it can potently accelerate myogenesis in mesodermal cells. This includes in Drosophila melanogaster imaginal disc myoblasts, where triggering premature muscle gene expression in these adult muscle progenitors has become a paradigm for understanding the regulation of the myogenic program. Here, we investigated the global consequences of MEF2 overexpression in the imaginal wing disc myoblasts, by combining RNA-sequencing with RT-qPCR and immunofluorescence. We observed the formation of sarcomere-like structures that contained both muscle and cytoplasmic myosin, and significant upregulation of muscle gene expression, especially genes essential for myofibril formation and function. These transcripts were functional since numerous myofibrillar proteins were detected in discs using immunofluorescence. Interestingly, muscle genes whose expression is restricted to the adult stages were not activated in these adult myoblasts. These studies confirm a broad activation of the myogenic program in response to MEF2 expression and suggest that additional regulatory factors are required for promoting the adult muscle-specific program. Our findings contribute to understanding the regulatory mechanisms governing muscle development and highlight the multifaceted role of MEF2 in orchestrating this intricate process.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    心脏发育是一个由复杂的转录网络控制的微调过程,其中转录因子(TF)与其他调节层相互作用。在这一章中,我们介绍核心心脏TFs,包括Gata,手,Nkx2,Mef2,Srf,Tbx这些因子调节彼此的表达,并且还可以组合方式作用于它们的下游靶标。它们的破坏导致小鼠的各种心脏表型,人类的突变与先天性心脏缺陷有关。在本章的第二部分,我们讨论了不同级别的监管,包括顺式监管元素,染色质结构,和microRNAs,可以与转录因子相互作用,调节它们的功能,或者是下游目标。最后,提供了导致人类先天性心脏病的心脏调节网络紊乱的例子。
    Cardiac development is a fine-tuned process governed by complex transcriptional networks, in which transcription factors (TFs) interact with other regulatory layers. In this chapter, we introduce the core cardiac TFs including Gata, Hand, Nkx2, Mef2, Srf, and Tbx. These factors regulate each other\'s expression and can also act in a combinatorial manner on their downstream targets. Their disruption leads to various cardiac phenotypes in mice, and mutations in humans have been associated with congenital heart defects. In the second part of the chapter, we discuss different levels of regulation including cis-regulatory elements, chromatin structure, and microRNAs, which can interact with transcription factors, modulate their function, or are downstream targets. Finally, examples of disturbances of the cardiac regulatory network leading to congenital heart diseases in human are provided.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    肌细胞增强因子(MEF2)家族的转录因子,最初发现它在肌肉发育和功能中的关键作用,已经成为大脑发育和神经元可塑性各个方面的重要调节剂。已知MEF2转录因子调节神经系统中的许多重要基因,包括脑源性神经营养因子(BDNF),一种负责促进生存的小分泌神经营养蛋白,增长,和神经元的分化。Bdnf基因的表达受与其近端和远端调节区结合的各种转录因子的时空控制。虽然先前的研究已经研究了MEF2转录因子与Bdnf之间的联系,MEF2因子在Bdnf转录调控中的内源性功能仍然未知。这里,我们旨在加深对MEF2转录因子及其在大鼠皮质和海马神经元中Bdnf调控中的作用的认识。因此,我们证明,位于Bdnf基因-4.8kb的MEF2转录因子依赖性增强子调节海马神经元中Bdnf的内源性表达。此外,我们证实了体内-4.8kb增强子的神经元活性依赖性激活。最后,我们表明,特定的MEF2家族转录因子在调节Bdnf中具有独特的作用,特定功能根据特定的大脑区域和刺激而变化。总之,我们提出MEF2家族转录因子作为BDNF表达的关键调节因子,通过远端和近端调节区微调Bdnf表达。
    The myocyte enhancer factor (MEF2) family of transcription factors, originally discovered for its pivotal role in muscle development and function, has emerged as an essential regulator in various aspects of brain development and neuronal plasticity. The MEF2 transcription factors are known to regulate numerous important genes in the nervous system, including brain-derived neurotrophic factor (BDNF), a small secreted neurotrophin responsible for promoting the survival, growth, and differentiation of neurons. The expression of the Bdnf gene is spatiotemporally controlled by various transcription factors binding to both its proximal and distal regulatory regions. While previous studies have investigated the connection between MEF2 transcription factors and Bdnf, the endogenous function of MEF2 factors in the transcriptional regulation of Bdnf remains largely unknown. Here, we aimed to deepen the knowledge of MEF2 transcription factors and their role in the regulation of Bdnf comparatively in rat cortical and hippocampal neurons. As a result, we demonstrate that the MEF2 transcription factor-dependent enhancer located at -4.8 kb from the Bdnf gene regulates the endogenous expression of Bdnf in hippocampal neurons. In addition, we confirm neuronal activity-dependent activation of the -4.8 kb enhancer in vivo. Finally, we show that specific MEF2 family transcription factors have unique roles in the regulation of Bdnf, with the specific function varying based on the particular brain region and stimuli. Altogether, we present MEF2 family transcription factors as crucial regulators of Bdnf expression, fine-tuning Bdnf expression through both distal and proximal regulatory regions.
    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
    来自酿酒酵母的β-葡聚糖在血管生成中的重要性尚未得到很好的研究。我们研究了β-葡聚糖是否通过PI3K/Src和ERK1/2信号通路在HUVECs中诱导血管生成。我们确定β-葡聚糖诱导PI3K的磷酸化,Src,Akt,eNOS,和ERK1/2。随后,我们发现这种磷酸化增加了HUVECs的活力.我们还观察到β-葡聚糖的刺激促进了MEF2和MEF2依赖性促血管生成基因的活性,包括EGR2、EGR3、KLF2和KLF4。此外,β-葡聚糖在血管生成中的作用是通过体外和离体实验证实的,包括细胞迁移,毛细管样管形成和小鼠主动脉环测定。为了确定β-葡聚糖对PI3K/Akt/eNOS和ERK1/2信号通路的影响,使用PI3K抑制剂wortmannin和ERK1/2抑制剂SCH772984。通过基质胶塞测定,我们证实β-葡聚糖显著增加体内血管生成。一起来看,我们的研究表明β-葡聚糖通过PI3K和ERK1/2信号通路促进血管生成.
    The importance of β-glucan from S. cerevisiae in angiogenesis has not been well studied. We investigated whether β-glucan induces angiogenesis through PI3K/Src and ERK1/2 signaling pathway in HUVECs. We identified that β-glucan induced phosphorylation of PI3K, Src, Akt, eNOS, and ERK1/2. Subsequently, we found that this phosphorylation increased the viability of HUVECs. We also observed that stimulation of β-glucan promoted the activity of MEF2 and MEF2-dependent pro-angiogenic genes, including EGR2, EGR3, KLF2, and KLF4. Additionally, the role of β-glucan in angiogenesis was confirmed using in vitro and ex vivo experiments including cell migration, capillary-like tube formation and mouse aorta ring assays. To determine the effect of β-glucan on the PI3K/Akt/eNOS and ERK1/2 signaling pathway, PI3K inhibitor wortmannin and ERK1/2 inhibitor SCH772984 were used. Through the Matrigel plug assay, we confirmed that β-glucan significantly increased angiogenesis in vivo. Taken together, our study demonstrates that β-glucan promotes angiogenesis via through PI3K and ERK1/2 signaling pathway.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    转录因子肌细胞增强因子2(MEF2)家族成员与IIa类组蛋白脱乙酰酶(HDAC)的相互作用与多种疾病有关。尽管多年来积累了关于这个主题的大量知识,仍然缺乏对多个IIa类HDAC衍生肽与MEF2D的结合模式的高分辨率和详细分析。为了弥补这个差距,我们在这里报道了与双链DNA和四种不同的IIa类HDAC衍生肽复合的MEF2D的晶体结构,即HDAC4、HDAC5、HDAC7和HDAC9。所有IIa类HDAC衍生的肽形成延伸的两亲性α-螺旋结构,其紧密地配合在MEF2D结构域的疏水凹槽中。IIa类HDAC衍生肽与MEF2D的结合模式非常相似,并且主要通过由高度保守的分支疏水氨基酸介导的非极性相互作用而发生。进一步的研究表明,IIa类HDAC衍生的肽在溶液中是未结构化的,并且似乎仅在与MEF2D结合时采用折叠的α-螺旋结构。我们的肽-蛋白质复合物与先前表征的MEF2结构的比较,该结构与不同的共激活剂和共阻遏物结合,强调了两者的差异和相似之处,并揭示了MEF2在蛋白质-蛋白质相互作用中的适应性。与多个IIa类HDAC衍生肽复合的MEF2D的三维结构的阐明不仅提供了对它们相互作用的分子基础的更好理解,而且对新型拮抗剂的开发也有意义。
    Interaction of transcription factor myocyte enhancer factor-2 (MEF2) family members with class IIa histone deacetylases (HDACs) has been implicated in a wide variety of diseases. Though considerable knowledge on this topic has been accumulated over the years, a high resolution and detailed analysis of the binding mode of multiple class IIa HDAC derived peptides with MEF2D is still lacking. To fulfil this gap, we report here the crystal structure of MEF2D in complex with double strand DNA and four different class IIa HDAC derived peptides, namely HDAC4, HDAC5, HDAC7 and HDAC9. All class IIa HDAC derived peptides form extended amphipathic α-helix structures that fit snugly in the hydrophobic groove of MEF2D domain. Binding mode of class IIa HDAC derived peptides to MEF2D is very similar and occur primarily through nonpolar interactions mediated by highly conserved branched hydrophobic amino acids. Further studies revealed that class IIa HDAC derived peptides are unstructured in solution and appear to adopt a folded α-helix structure only upon binding to MEF2D. Comparison of our peptide-protein complexes with previously characterized structures of MEF2 bound to different co-activators and co-repressors, highlighted both differences and similarities, and revealed the adaptability of MEF2 in protein-protein interactions. The elucidation of the three-dimensional structure of MEF2D in complex with multiple class IIa HDAC derived peptides provide not only a better understanding of the molecular basis of their interactions but also have implications for the development of novel antagonist.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    肌细胞增强因子2(MEF2)蛋白参与多种发育,生理,和脊椎动物的病理过程。蛋白质-蛋白质相互作用是MEF2A影响的过多生物过程的基础,需要MEF2A相互作用组的详细表征。采用基于纳米抗体的亲和纯化/质谱策略来实现该目标。具体来说,使用固定有GBP-纳米抗体的GFP标记的MEF2A蛋白从生肌裂解物中捕获MEF2A蛋白复合物,然后进行LC-MS/MS蛋白质组学分析以鉴定MEF2A相互作用物.经过生物信息学分析,我们进一步表征了MEF2A与转录阻遏物的相互作用,FOXP1。FOXP1与MEF2A共沉淀在分化后减少的增殖生肌细胞中(肌管形成)。FOXP1的异位表达抑制了MEF2A驱动的生肌报告基因(源自肌酸激酶肌肉和肌生成素基因),并延迟了分化过程中内源性肌生成素的诱导。相反,FOXP1耗竭增强了MEF2A反式激活特性和肌原蛋白表达。FoxP1:MEF2A相互作用也保留在心肌细胞中,并且FoxP1耗竭增强心肌细胞肥大。FOXP1通过p38MAPK通路阻止MEF2A磷酸化和活化。总的来说,这些数据暗示FOXP1限制MEF2A功能,以避免肌原祖细胞的过早分化,并可能阻止心肌细胞肥大中胚胎基因表达的重新激活.
    Myocyte enhancer factor 2 (MEF2) proteins are involved in multiple developmental, physiological, and pathological processes in vertebrates. Protein-protein interactions underlie the plethora of biological processes impacted by MEF2A, necessitating a detailed characterization of the MEF2A interactome. A nanobody based affinity-purification/mass spectrometry strategy was employed to achieve this goal. Specifically, the MEF2A protein complexes were captured from myogenic lysates using a GFP-tagged MEF2A protein immobilized with a GBP-nanobody followed by LC-MS/MS proteomic analysis to identify MEF2A interactors. After bioinformatic analysis, we further characterized the interaction of MEF2A with a transcriptional repressor, FOXP1. FOXP1 coprecipitated with MEF2A in proliferating myogenic cells which diminished upon differentiation (myotube formation). Ectopic expression of FOXP1 inhibited MEF2A driven myogenic reporter genes (derived from the creatine kinase muscle and myogenin genes) and delayed induction of endogenous myogenin during differentiation. Conversely, FOXP1 depletion enhanced MEF2A transactivation properties and myogenin expression. The FoxP1:MEF2A interaction is also preserved in cardiomyocytes and FoxP1 depletion enhanced cardiomyocyte hypertrophy. FOXP1 prevented MEF2A phosphorylation and activation by the p38MAPK pathway. Overall, these data implicate FOXP1 in restricting MEF2A function in order to avoid premature differentiation in myogenic progenitors and also to possibly prevent re-activation of embryonic gene expression in cardiomyocyte hypertrophy.
    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
    心肌细胞特异性增强子结合因子2(MEF2),属于MADS超家族,是一种关键且保守的转录因子,可与E-box基序结合以控制肌肉基因的表达。肌肉生长抑制素(mstn),一种肌肉生长抑制剂,是TGF-β超家族的重要成员。目前,缺乏对MEF2介导的A.latusmstn(Almstn)转录调控在鱼类肌肉发育中的机制的理解。在本研究中,从刺五加中鉴定了两个AlMEF2(AlMEF2A和AlMEF2B)和Almstn2a。AlMEF2A和AlMEF2B具有456和315个氨基酸(aa)残基,分别。两个典型地区,一个MADS盒子,MEF2和转录激活(TAD)域,存在于两个AlMEF2中。两个AlMEF2基因的表达谱相似。AlMEF2基因主要在脑内表达,白色肌肉,还有肝脏,而Almstn2a在脑中的表达高于其他组织。此外,5个群体在饥饿和再摄食后,AlMEF2s和Almstn2a的表达趋势发生了显着变化。此外,截短实验表明,-987到+168和-105到+168是Almstn2a的核心启动子,响应AlMEF2A和AlMEF2B,分别。点突变实验证实Almstn2a转录依赖于AlMEF2A和AlMEF2B调控的突变结合位点1或5(M1/5)和突变结合位点4或5(M4/5),分别。电泳移动移位测定(EMSA)进一步证实M1(-527至-512)是AlMEF2A作用于Almstn2a基因的关键位点。此外,siRNA干扰基因表达实验表明,降低AlMEF2A或AlMEF2B水平可以显着增加Almstn2a转录。这些结果提供了有关AlMEF2s调控Almstn2a转录活性的新信息,并为鱼类肌肉发育的调控机制提供了理论基础。
    Myocyte-specific enhancer binding factor 2 (MEF2), which belongs to the MADS superfamily, is a pivotal and conserved transcription factor that combines with the E-box motif to control the expression of muscle genes. Myostatin (mstn), a muscle growth inhibitor, is a vital member of the TGF-β superfamily. Currently, an understanding of the mechanisms of A. latus mstn (Almstn) transcriptional regulation mediated by MEF2 in fish muscle development is lacking. In the present study, two AlMEF2s (AlMEF2A and AlMEF2B) and Almstn2a were characterized from Acanthopagrus latus. AlMEF2A and AlMEF2B had 456 and 315 amino acid (aa) residues, respectively. Two typical regions, a MADS-box, MEF2, and transcriptionally activated (TAD) domains, are present in both AlMEF2s. The expression profiles of the two AlMEF2 genes were similar. The AlMEF2 genes were mainly expressed in the brain, white muscle, and liver, while Almstn2a expression was higher in the brain than in other tissues. Moreover, the expression trends of AlMEF2s and Almstn2a were significantly changed after starvation and refeeding in the five groups. Additionally, truncation experiments showed that -987 to +168 and -105 to +168 were core promoters of Almstn2a that responded to AlMEF2A and AlMEF2B, respectively. The point mutation experiment confirmed that Almstn2a transcription relies on the mutation binding sites 1 or 5 (M1/5) and mutation binding sites 4 or 5 (M4/5) for AlMEF2A and AlMEF2B regulation, respectively. The electrophoretic mobile shift assay (EMSA) further verified that M1 (-527 to -512) was a pivotal site where AlMEF2A acted on the Almstn2a gene. Furthermore, a siRNA interference gene expression experiment showed that reduced levels of AlMEF2A or AlMEF2B could prominently increase Almstn2a transcription. These results provide new information about the regulation of Almstn2a transcriptional activity by AlMEF2s and a theoretical basis for the regulatory mechanisms involved in muscle development in fish.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    背景:在牙列发育早期和胚带形成之前,后组织增厚形成后坑。在这种增厚的前面形成了一个凹槽,胚胎狭缝,这标志着发育中的胚胎的前后方向。这个狭缝被一些作者认为是胚孔,因此内胚层的起源,而其他人则认为后坑代表了胚孔。这场争论具有进化意义,因为如果狭缝代表胚孔,那么这将支持两栖式假说,该假说表明,双边祖先中的狭缝状胚孔进化为原造孔和氘代造口术。
    结果:在本文中,我们总结了我们目前关于内胚层和中胚层发育的知识,并提供了早期内胚层和中胚层决定标记基因的额外数据,如飞艇,Mox,还有T-box基因.
    结论:我们得出的结论是,在胚胎狭缝发育之前,牙列的内胚层形成,因此,狭缝不是内胚层的主要起源。因此,胚胎狭缝不太可能代表胚孔。相反,我们建议后坑确实代表了胚孔的嘴唇,胚胎狭缝(和周围组织)代表了一个形态上的浅表初肠样结构。我们进一步得出结论,内胚层和中胚层发育都受到保守基因调控网络的控制。在节肢动物中发现的许多特征,包括模型果蝇。
    BACKGROUND: Early during onychophoran development and prior to the formation of the germ band, a posterior tissue thickening forms the posterior pit. Anterior to this thickening forms a groove, the embryonic slit, that marks the anterior-posterior orientation of the developing embryo. This slit is by some authors considered the blastopore, and thus the origin of the endoderm, while others argue that the posterior pit represents the blastopore. This controversy is of evolutionary significance because if the slit represents the blastopore, then this would support the amphistomy hypothesis that suggests that a slit-like blastopore in the bilaterian ancestor evolved into protostomy and deuterostomy.
    RESULTS: In this paper, we summarize our current knowledge about endoderm and mesoderm development in onychophorans and provide additional data on early endoderm- and mesoderm-determining marker genes such as Blimp, Mox, and the T-box genes.
    CONCLUSIONS: We come to the conclusion that the endoderm of onychophorans forms prior to the development of the embryonic slit, and thus that the slit is not the primary origin of the endoderm. It is thus unlikely that the embryonic slit represents the blastopore. We suggest instead that the posterior pit indeed represents the lips of the blastopore, and that the embryonic slit (and surrounding tissue) represents a morphologically superficial archenteron-like structure. We conclude further that both endoderm and mesoderm development are under control of conserved gene regulatory networks, and that many of the features found in arthropods including the model Drosophila melanogaster are likely derived.
    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:组蛋白脱乙酰酶4(HDAC4)的核质穿梭失调与几种神经发育和神经退行性疾病有关。因此,了解细胞核和细胞质HDAC4的作用以及调节核进出的机制是一个共同努力的领域。有效的核进入依赖于转录因子MEF2的结合,因为MEF2结合区中的突变导致HDAC4的细胞质积累。众所周知,细胞核退出和细胞质保留依赖于14-3-3结合,和影响结合的突变被广泛用于诱导HDAC4的核积累。虽然HDAC4穿梭的调节显然很重要,在理解HDAC4的核和细胞质分布如何影响其功能方面存在差距。此外,目前还不清楚蛋白质的其他特征,包括催化位点,MEF2结合区和/或锚蛋白重复结合基序影响HDAC4在神经元中的分布和/或活性。由于HDAC4功能在果蝇中是保守的,HDAC4的核积累增加也导致神经发育受损,我们使用果蝇作为研究HDAC4功能的遗传模型。
    结果:在这里,我们通过对由此产生的亚细胞分布和核聚集的深入检查,产生了一系列用于HDAC4功能解剖的突变体,并将它们与在果蝇蘑菇体和眼的神经元形态发生模型中表达的发育表型相关联。我们发现在蘑菇体内,HDAC4在细胞核或细胞质中的强制隔离导致轴突形态发生缺陷。HDAC4导致发育受损的作用依赖于MEF2结合区,由锚蛋白重复结合基序调制,并且很大程度上独立于完整的催化位点。相比之下,眼发育的中断在很大程度上与MEF2结合无关,但催化位点的突变显着降低了表型,表明HDAC4以神经元亚型特异性方式起作用。
    结论:我们发现,锚蛋白重复结合基序的突变加剧了HDAC4核积累对蘑菇体和眼睛发育的损害,而对MEF2结合位点和完整催化位点有不同的要求。确定HDAC4在核聚集体和这些组织的细胞质中的结合配偶体以进一步理解其作用机制将是重要的。
    Dysregulation of nucleocytoplasmic shuttling of histone deacetylase 4 (HDAC4) is associated with several neurodevelopmental and neurodegenerative disorders. Consequently, understanding the roles of nuclear and cytoplasmic HDAC4 along with the mechanisms that regulate nuclear entry and exit is an area of concerted effort. Efficient nuclear entry is dependent on binding of the transcription factor MEF2, as mutations in the MEF2 binding region result in cytoplasmic accumulation of HDAC4. It is well established that nuclear exit and cytoplasmic retention are dependent on 14-3-3-binding, and mutations that affect binding are widely used to induce nuclear accumulation of HDAC4. While regulation of HDAC4 shuttling is clearly important, there is a gap in understanding of how the nuclear and cytoplasmic distribution of HDAC4 impacts its function. Furthermore, it is unclear whether other features of the protein including the catalytic site, the MEF2-binding region and/or the ankyrin repeat binding motif influence the distribution and/or activity of HDAC4 in neurons. Since HDAC4 functions are conserved in Drosophila, and increased nuclear accumulation of HDAC4 also results in impaired neurodevelopment, we used Drosophila as a genetic model for investigation of HDAC4 function.
    Here we have generated a series of mutants for functional dissection of HDAC4 via in-depth examination of the resulting subcellular distribution and nuclear aggregation, and correlate these with developmental phenotypes resulting from their expression in well-established models of neuronal morphogenesis of the Drosophila mushroom body and eye. We found that in the mushroom body, forced sequestration of HDAC4 in the nucleus or the cytoplasm resulted in defects in axon morphogenesis. The actions of HDAC4 that resulted in impaired development were dependent on the MEF2 binding region, modulated by the ankyrin repeat binding motif, and largely independent of an intact catalytic site. In contrast, disruption to eye development was largely independent of MEF2 binding but mutation of the catalytic site significantly reduced the phenotype, indicating that HDAC4 acts in a neuronal-subtype-specific manner.
    We found that the impairments to mushroom body and eye development resulting from nuclear accumulation of HDAC4 were exacerbated by mutation of the ankyrin repeat binding motif, whereas there was a differing requirement for the MEF2 binding site and an intact catalytic site. It will be of importance to determine the binding partners of HDAC4 in nuclear aggregates and in the cytoplasm of these tissues to further understand its mechanisms of action.
    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
    Mef2c是介导促进软骨内骨化和骨形成的关键细胞行为的转录因子。以前,Mef2c已被证明通过其骨细胞特异性增强子调节Sost转录,ECR5和Col1-Cre或Dmp1-Cre的Mef2cfl/f的条件性缺失产生了与VanBuchem病表型一致的全身性高骨量(HBM)。然而,Sost-/-;Mef2cfl/fl;Dmp1-Cre小鼠产生了显着更高的骨量表型,Sost-/-单独表明Mef2c通过其他机制调节骨量,独立于Sost。为了鉴定在骨代谢中重要的新的Mef2c转录靶标,我们通过单细胞RNA测序分析了从Mef2cfl/fl分离的细胞亚群中的基因表达;Dmp1-Cre和Mef2cfl/fl;Bglap-Cre股骨,两种菌株表现出相似的高骨量表型。然而,我们发现Mef2cfl/fl;Bglap-Cre也显示出生长板缺陷,其特征是几个骨祖细胞亚群的扩张。差异基因表达分析鉴定了Mef2cfl/fl中总共96个上调和2434个下调基因;Mef2cfl/fl中的Bglap-Cre和176个下调基因;Dmpl-Cre骨细胞亚群与野生型小鼠相比。Mef2c缺失影响了几种细胞类型的转录组,包括间充质祖细胞(MP),骨祖细胞(OSP),成骨细胞(OB),和骨细胞(OCY)亚群。几种能量代谢基因,如Uqcrb,Ndufv2,Ndufs3,Ndufa13,Ndufb9,Ndufb5,Cox6a1,Cox5a,Atp5o,Atp5g2,Atp5b,Atp5在Mef2c缺陷型OBs和OCYs中显著下调,在这两种菌株中。差异表达基因启动子区的结合基序分析确定了骨唾液酸蛋白(BSP/Ibsp)中的Mef2c结合,一种已知会导致Ibsp-/-小鼠股骨小梁BV/TV增加的基因。免疫组织化学分析证实OBs和OCYs中不存在Ibsp蛋白。这些发现表明,Sost-/-;Mef2cfl/fl;Dmp1-Cre中的HBM是由调节骨形成的基因中的多种转录变化引起的,其中两个是Sost和Ibsp。
    Mef2c is a transcription factor that mediates key cellular behaviors that promote endochondral ossification and bone formation. Previously, Mef2c has been shown to regulate Sost transcription via its osteocyte-specific enhancer, ECR5, and conditional deletions of Mef2cfl/fl with either Col1-Cre or Dmp1-Cre produced generalized high bone mass (HBM) consistent with Van Buchem Disease phenotypes. However, Sost-/-; Mef2cfl/fl; Dmp1-Cre mice produced a significantly higher bone mass phenotype that Sost-/- alone suggesting that Mef2c modulates bone mass through additional mechanisms, independent of Sost. To identify new Mef2c transcriptional targets important in bone metabolism, we profiled gene expression by single-cell RNA sequencing in subpopulations of cells isolated from Mef2cfl/fl; Dmp1-Cre and Mef2cfl/fl; Bglap-Cre femurs, both strains exhibiting similar high bone mass phenotypes. However, we found Mef2cfl/fl; Bglap-Cre to also display a growth plate defect characterized by an expansion of several osteoprogenitor subpopulations. Differential gene expression analysis identified a total of 96 up- and 2434 down- regulated genes in Mef2cfl/fl; Bglap-Cre and 176 up- and 1041 down- regulated genes in Mef2cfl/fl; Dmp1-Cre bone cell subpopulations compared to wildtype mice. Mef2c deletion affected the transcriptomes across several cell types including mesenchymal progenitors (MP), osteoprogenitors (OSP), osteoblast (OB), and osteocyte (OCY) subpopulations. Several energy metabolism genes such as Uqcrb, Ndufv2, Ndufs3, Ndufa13, Ndufb9, Ndufb5, Cox6a1, Cox5a, Atp5o, Atp5g2, Atp5b, Atp5 were significantly down regulated in Mef2c-deficient OBs and OCYs, in both strains. Binding motif analysis of promoter regions of differentially expressed genes identified Mef2c binding in Bone Sialoprotein (BSP/Ibsp), a gene known to cause increased trabecular BV/TV in the femurs of Ibsp-/- mice. Immunohistochemical analysis confirmed the absence of Ibsp protein in OBs and OCYs. These findings suggests that the HBM in Sost-/-; Mef2cfl/fl; Dmp1-Cre is caused by a multitude of transcriptional changes in genes that regulate bone formation, two of which are Sost and Ibsp.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号