Serum Response Factor

血清反应因子
  • 文章类型: Journal Article
    血清反应因子(SRF)控制血管平滑肌细胞(VSMC)中的基因转录,并调节VSMC从收缩状态到合成状态的表型转换,在心血管疾病(CVD)的发病机制中起着关键作用。不知道翻译后SUMO化如何调节CVD中的SRF活性。在这里,我们显示,在VSMCSenp1缺乏增加SUMOylatedSRF和SRF-ELK复合物,导致小鼠血管重塑和新内膜形成增强。机械上,VSMC中的SENP1缺乏会增加赖氨酸143处的SRFSUMO化,从而减少SRF溶酶体定位,同时增加核积累,并将收缩表型响应性SRF-myocardin复合物转换为合成表型响应性SRF-ELK1复合物。来自CVD患者冠状动脉的VSMC中SUMO化SRF和磷酸-ELK1增加。重要的是,ELK抑制剂AZD6244可防止SRF-myocardin转变为SRF-ELK复合物,在Senp1缺陷小鼠中减弱VSMC合成表型和新内膜形成。因此,靶向SRF复合物可能具有治疗CVD的治疗潜力.
    Serum response factor (SRF) controls gene transcription in vascular smooth muscle cells (VSMCs) and regulates VSMC phenotypic switch from a contractile to a synthetic state, which plays a key role in the pathogenesis of cardiovascular diseases (CVD). It is not known how post-translational SUMOylation regulates the SRF activity in CVD. Here we show that Senp1 deficiency in VSMCs increased SUMOylated SRF and the SRF-ELK complex, leading to augmented vascular remodeling and neointimal formation in mice. Mechanistically, SENP1 deficiency in VSMCs increases SRF SUMOylation at lysine 143, reducing SRF lysosomal localization concomitant with increased nuclear accumulation and switching a contractile phenotype-responsive SRF-myocardin complex to a synthetic phenotype-responsive SRF-ELK1 complex. SUMOylated SRF and phospho-ELK1 are increased in VSMCs from coronary arteries of CVD patients. Importantly, ELK inhibitor AZD6244 prevents the shift from SRF-myocardin to SRF-ELK complex, attenuating VSMC synthetic phenotypes and neointimal formation in Senp1-deficient mice. Therefore, targeting the SRF complex may have a therapeutic potential for the treatment of CVD.
    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
    原理:心肌细胞(CMs)在出生后成熟时经历了巨大的结构和功能变化;然而,监管机制仍然非常不清楚。Cypher/Z带选择性剪接的PDZ基序蛋白(ZASP)是维持Z盘稳定性的必需肌节成分。小鼠Cypher的缺失和人ZASP中的突变导致扩张型心肌病(DCM)。尚未回答Cypher/ZASP是否参与CM成熟并因此影响心脏功能。方法:免疫荧光,透射电子显微镜,实时定量PCR,并利用Westernblot鉴定Cypher在CM成熟中的作用。随后,RNA测序和生物信息学分析预测血清反应因子(SRF)是关键调节因子。使用编码SRF的腺病毒或腺相关病毒进行抢救实验,在体外和体内。通过G-肌动蛋白/F-肌动蛋白分级分离阐明了分子机制,核-细胞质提取,肌动蛋白分解分析,和共沉淀测定。结果:Cypher缺失导致线粒体肌节同工型开关受损和形态异常,横小管,和插层光盘。RNA测序分析揭示了与肌节组装相关的关键基因的显著失调,线粒体代谢,和没有Cypher的电生理学。此外,预测SRF是介导转录差异的关键转录因子。随后的抢救实验表明,在出生后的关键时期,SRF的重新表达有效地纠正了Cypher耗竭小鼠的CM成熟缺陷,并显着改善了心脏功能。机械上,Cypher缺乏导致F-肌动蛋白的不稳定和G-肌动蛋白水平的显着增加,从而阻碍肌钙蛋白相关转录因子A(MRTFA)的核定位,并随后启动SRF转录。结论:Cypher/ZASP通过肌动蛋白介导的MRTFA-SRF信号在CM成熟中起着至关重要的作用。提示了CM成熟异常与DCM迟发之间的联系,提供对DCM发病机制和潜在治疗策略的进一步见解。
    Rationale: Cardiomyocytes (CMs) undergo dramatic structural and functional changes in postnatal maturation; however, the regulatory mechanisms remain greatly unclear. Cypher/Z-band alternatively spliced PDZ-motif protein (ZASP) is an essential sarcomere component maintaining Z-disc stability. Deletion of mouse Cypher and mutation in human ZASP result in dilated cardiomyopathy (DCM). Whether Cypher/ZASP participates in CM maturation and thereby affects cardiac function has not been answered. Methods: Immunofluorescence, transmission electron microscopy, real-time quantitative PCR, and Western blot were utilized to identify the role of Cypher in CM maturation. Subsequently, RNA sequencing and bioinformatics analysis predicted serum response factor (SRF) as the key regulator. Rescue experiments were conducted using adenovirus or adeno-associated viruses encoding SRF, both in vitro and in vivo. The molecular mechanisms were elucidated through G-actin/F-actin fractionation, nuclear-cytoplasmic extraction, actin disassembly assays, and co-sedimentation assays. Results: Cypher deletion led to impaired sarcomere isoform switch and morphological abnormalities in mitochondria, transverse-tubules, and intercalated discs. RNA-sequencing analysis revealed significant dysregulation of crucial genes related to sarcomere assembly, mitochondrial metabolism, and electrophysiology in the absence of Cypher. Furthermore, SRF was predicted as key transcription factor mediating the transcriptional differences. Subsequent rescue experiments showed that SRF re-expression during the critical postnatal period effectively rectified CM maturation defects and notably improved cardiac function in Cypher-depleted mice. Mechanistically, Cypher deficiency resulted in the destabilization of F-actin and a notable increase in G-actin levels, thereby impeding the nuclear localisation of myocardin-related transcription factor A (MRTFA) and subsequently initiating SRF transcription. Conclusion: Cypher/ZASP plays a crucial role in CM maturation through actin-mediated MRTFA-SRF signalling. The linkage between CM maturation abnormalities and the late-onset of DCM is suggested, providing further insights into the pathogenesis of DCM and potential treatment strategies.
    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:卵巢子宫内膜瘤(子宫内膜瘤)中的纤维化,主要由转化生长因子-β(TGF-β)诱导,其特征是肌成纤维细胞过度激活和过度的细胞外基质(ECM)沉积,通过损害卵巢储备和卵母细胞质量导致子宫内膜瘤相关症状,如不孕症。然而,TGF-β诱导的子宫内膜瘤相关纤维化进展的确切分子机制尚不清楚。
    方法:在子宫内膜瘤患者和健康对照的子宫内膜活检中验证赖氨酸乙酰转移酶14(KAT14)的表达水平,通过分析已发表的子宫内膜异位症的单细胞转录组(scRNA-seq)数据集,进一步证实了KAT14的转录水平。我们使用过表达,击倒,在永生化人子宫内膜基质细胞(HESCs)或人原发性异位子宫内膜基质细胞(EcESCs)中采用敲除方法,以确定KAT14在TGF-β诱导的纤维化中的作用。此外,在子宫内膜异位症小鼠模型中使用携带KAT14-shRNA的腺相关病毒(AAV)来评估KAT14在体内的作用.
    结果:KAT14在子宫内膜瘤患者的异位病变中上调,主要在活化成纤维细胞中表达。体外研究表明,KAT14过表达显著促进子宫内膜基质细胞TGF-β诱导的促纤维化反应,而KAT14沉默显示了可以通过KAT14重新增强来挽救的不良反应。在体内,Kat14敲除改善子宫内膜异位症小鼠模型异位病变的纤维化。机械上,我们发现KAT14与血清反应因子(SRF)直接相互作用,通过增加启动子区域的组蛋白H4乙酰化来促进α-平滑肌肌动蛋白(α-SMA)的表达;这对于TGF-β诱导的ECM产生和肌成纤维细胞分化是必需的。此外,在TGF-β治疗下,SRF的敲减或药理学抑制显著减弱了KAT14介导的促纤维化作用。值得注意的是,KAT14/SRF复合物在子宫内膜瘤样本中含量丰富,与α-SMA表达呈正相关,进一步支持KAT14/SRF复合物在子宫内膜瘤相关纤维化进展中的关键作用。
    结论:我们的结果揭示了KAT14作为TGF-β诱导的ECM产生和肌成纤维细胞分化的关键效应因子,通过与SRF协同作用促进组蛋白H4乙酰化,代表子宫内膜瘤相关纤维化的潜在治疗靶点。
    BACKGROUND: Fibrogenesis within ovarian endometrioma (endometrioma), mainly induced by transforming growth factor-β (TGF-β), is characterized by myofibroblast over-activation and excessive extracellular matrix (ECM) deposition, contributing to endometrioma-associated symptoms such as infertility by impairing ovarian reserve and oocyte quality. However, the precise molecular mechanisms that underpin the endometrioma- associated fibrosis progression induced by TGF-β remain poorly understood.
    METHODS: The expression level of lysine acetyltransferase 14 (KAT14) was validated in endometrium biopsies from patients with endometrioma and healthy controls, and the transcription level of KAT14 was further confirmed by analyzing a published single-cell transcriptome (scRNA-seq) dataset of endometriosis. We used overexpression, knockout, and knockdown approaches in immortalized human endometrial stromal cells (HESCs) or human primary ectopic endometrial stromal cells (EcESCs) to determine the role of KAT14 in TGF-β-induced fibrosis. Furthermore, an adeno-associated virus (AAV) carrying KAT14-shRNA was used in an endometriosis mice model to assess the role of KAT14 in vivo.
    RESULTS: KAT14 was upregulated in ectopic lesions from endometrioma patients and predominantly expressed in activated fibroblasts. In vitro studies showed that KAT14 overexpression significantly promoted a TGF-β-induced profibrotic response in endometrial stromal cells, while KAT14 silencing showed adverse effects that could be rescued by KAT14 re-enhancement. In vivo, Kat14 knockdown ameliorated fibrosis in the ectopic lesions of the endometriosis mouse model. Mechanistically, we showed that KAT14 directly interacted with serum response factor (SRF) to promote the expression of α-smooth muscle actin (α-SMA) by increasing histone H4 acetylation at promoter regions; this is necessary for TGF-β-induced ECM production and myofibroblast differentiation. In addition, the knockdown or pharmacological inhibition of SRF significantly attenuated KAT14-mediating profibrotic effects under TGF-β treatment. Notably, the KAT14/SRF complex was abundant in endometrioma samples and positively correlated with α-SMA expression, further supporting the key role of KAT14/SRF complex in the progression of endometrioma-associated fibrogenesis.
    CONCLUSIONS: Our results shed light on KAT14 as a key effector of TGF-β-induced ECM production and myofibroblast differentiation in EcESCs by promoting histone H4 acetylation via co-operating with SRF, representing a potential therapeutic target for endometrioma-associated fibrosis.
    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
    肌肉生长过程直接影响猪肉食品的产量和品质。肌肉纤维是在胚胎阶段产生的,出生后长大,并在成年期再生;这些都被认为是肌肉发育的阶段。多层次的转录网络,转录后,通路水平控制着这个过程。过去已经使用了遗传学和基因组学的集成工具箱以及基因组学技术的使用,以试图在不同的选择过程中了解猪的骨骼肌生长和发育背后的分子过程。一类内源性非编码RNA在肌生成中具有重要的调控功能。但是miRNA-423-5p在肌肉发育中的确切功能和相关的分子途径仍然未知。使用目标预测软件,最初,使用骨骼肌生长和发育的各种选择标准鉴定了广西巴马小型猪品系中miR-423-5p的潜在靶基因。血清反应因子(SRF)是潜在的靶基因之一,两者呈负相关,这表明可能有针对性的互动。除了在猪骨骼肌中强烈表达外,miR-423-5p在C2C12细胞发育期间也上调。此外,real-timePCR分析显示miR-423-5p的过表达显著降低了肌原蛋白和肌原分化抗原的表达(p<0.05)。此外,酶联免疫吸附试验(ELISA)的结果表明,miR-423-5p的过表达导致SRF表达显著降低(p<0.05).此外,miR-423-5p下调携带猪SRF3'UTR的报告载体的荧光素酶活性,证实SRF是miR-423-5p的靶基因。一起来看,miR-423-5p可能通过调节SRF参与骨骼肌分化。
    The process of muscle growth directly affects the yield and quality of pork food products. Muscle fibers are created during the embryonic stage, grow following birth, and regenerate during adulthood; these are all considered to be phases of muscle development. A multilevel network of transcriptional, post-transcriptional, and pathway levels controls this process. An integrated toolbox of genetics and genomics as well as the use of genomics techniques has been used in the past to attempt to understand the molecular processes behind skeletal muscle growth and development in pigs under divergent selection processes. A class of endogenous noncoding RNAs have a major regulatory function in myogenesis. But the precise function of miRNA-423-5p in muscle development and the related molecular pathways remain largely unknown. Using target prediction software, initially, the potential target genes of miR-423-5p in the Guangxi Bama miniature pig line were identified using various selection criteria for skeletal muscle growth and development. The serum response factor (SRF) was found to be one of the potential target genes, and the two are negatively correlated, suggesting that there may be targeted interactions. In addition to being strongly expressed in swine skeletal muscle, miR-423-5p was also up-regulated during C2C12 cell development. Furthermore, real-time PCR analysis showed that the overexpression of miR-423-5p significantly reduced the expression of myogenin and the myogenic differentiation antigen (p < 0.05). Moreover, the results of the enzyme-linked immunosorbent assay (ELISA) demonstrated that the overexpression of miR-423-5p led to a significant reduction in SRF expression (p < 0.05). Furthermore, miR-423-5p down-regulated the luciferase activities of report vectors carrying the 3\' UTR of porcine SRF, confirming that SRF is a target gene of miR-423-5p. Taken together, miR-423-5p\'s involvement in skeletal muscle differentiation may be through the regulation of SRF.
    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
    转录因子TRPS1是乳腺癌中的环境依赖性癌基因。在乳腺里,TRPS1活性仅限于腔内人群,在青春期和怀孕期间至关重要。然而,其在静息状态下的功能仍不清楚。为了评估它是否可能成为癌症治疗的目标,我们使用条件普遍存在的耗竭小鼠模型研究了TRPS1在健康成人乳腺中的功能,其中长期耗竭不影响健康.使用转录组学方法,流式细胞术和功能测定,我们显示TRPS1活性对于维持功能性腔祖细胞区室至关重要。这需要压制YAP/TAZ和SRF/MRTF的活动。TRPS1通过调节RhoA活性间接抑制SRF/MRTF活性。我们的工作揭示了TRPS1在管腔祖细胞中的迄今未公开的功能,该功能与乳腺中的机械转导内在联系。它还可以提供对TRPS1致癌功能的新见解,因为腔祖细胞可能是许多乳腺癌的起源细胞。
    The transcription factor TRPS1 is a context-dependent oncogene in breast cancer. In the mammary gland, TRPS1 activity is restricted to the luminal population and is critical during puberty and pregnancy. Its function in the resting state remains however unclear. To evaluate whether it could be a target for cancer therapy, we investigated TRPS1 function in the healthy adult mammary gland using a conditional ubiquitous depletion mouse model where long-term depletion does not affect fitness. Using transcriptomic approaches, flow cytometry and functional assays, we show that TRPS1 activity is essential to maintain a functional luminal progenitor compartment. This requires the repression of both YAP/TAZ and SRF/MRTF activities. TRPS1 represses SRF/MRTF activity indirectly by modulating RhoA activity. Our work uncovers a hitherto undisclosed function of TRPS1 in luminal progenitors intrinsically linked to mechanotransduction in the mammary gland. It may also provide new insights into the oncogenic functions of TRPS1 as luminal progenitors are likely the cells of origin of many breast cancers.
    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
    神经元轴突的髓鞘形成对神经系统发育至关重要。髓鞘形成需要少突胶质细胞中戏剧性的细胞骨架动力学,但是肌动蛋白在髓鞘形成过程中是如何调节的,人们知之甚少。我们最近确定了血清反应因子(SRF)-一种已知在其他细胞类型中调节肌动蛋白和肌动蛋白调节剂表达的转录因子-作为老年大脑髓鞘形成的关键驱动因素。然而,在了解SRF在少突胶质细胞谱系细胞中的机制作用方面仍存在很大差距.这里,我们表明,SRF是少突胶质细胞在发育过程中髓鞘形成所必需的细胞自主功能。将ChIP-seq与RNA-seq组合鉴定少突胶质细胞前体细胞和少突胶质细胞中的SRF靶基因,其包括肌动蛋白和其他关键细胞骨架基因。因此,SRF敲除少突胶质细胞在分化早期表现出显著降低的肌动蛋白丝水平,与其在肌动蛋白依赖性髓鞘起始中的作用一致。令人惊讶的是,少突胶质细胞限制性的SRF损失导致与衰老和神经退行性疾病相关的基因标记的上调。一起,我们的发现将SRF确定为转录调节因子,它控制少突胶质细胞髓鞘形成所需的细胞骨架基因的表达.这项研究确定了调节少突胶质细胞生物学的重要途径,与大脑发育高度相关。老化,和疾病。
    Myelination of neuronal axons is essential for nervous system development. Myelination requires dramatic cytoskeletal dynamics in oligodendrocytes, but how actin is regulated during myelination is poorly understood. We recently identified serum response factor (SRF)-a transcription factor known to regulate expression of actin and actin regulators in other cell types-as a critical driver of myelination in the aged brain. Yet, a major gap remains in understanding the mechanistic role of SRF in oligodendrocyte lineage cells. Here, we show that SRF is required cell autonomously in oligodendrocytes for myelination during development. Combining ChIP-seq with RNA-seq identifies SRF-target genes in oligodendrocyte precursor cells and oligodendrocytes that include actin and other key cytoskeletal genes. Accordingly, SRF knockout oligodendrocytes exhibit dramatically reduced actin filament levels early in differentiation, consistent with its role in actin-dependent myelin sheath initiation. Surprisingly, oligodendrocyte-restricted loss of SRF results in upregulation of gene signatures associated with aging and neurodegenerative diseases. Together, our findings identify SRF as a transcriptional regulator that controls the expression of cytoskeletal genes required in oligodendrocytes for myelination. This study identifies an essential pathway regulating oligodendrocyte biology with high relevance to brain development, aging, and disease.
    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
    RhoA通过血清反应因子(SRF)及其转录辅因子myocardin相关转录因子(MRTF)信号通路调节基因转录,已成为多种疾病药理干预的有希望的治疗靶点。线粒体代谢改变是癌症的主要标志之一,因此,这种上调是Rho/MRTF/SRF抑制剂可靶向的漏洞.最近的进展确定了一系列破坏SRF转录的新型恶二唑-硫醚化合物,然而,这些化合物的直接分子靶标尚不清楚。在这里,我们证明了CCG-203971和CCG-232601在人肺成纤维细胞和小鼠成肌细胞的正常细胞系中的Rho/MRTF/SRF抑制机制。进一步的研究调查了这些分子在靶向线粒体功能中的作用。我们已经表明,这些分子高乙酰化组蛋白H4K12和H4K16,并调节参与线粒体功能和动力学的基因。这些小分子抑制剂通过抑制氧化磷酸化和增加糖酵解来调节线粒体功能作为补偿机制。我们的数据表明,这些CCG分子可有效抑制线粒体电子传递链的所有复合物并进一步诱导氧化应激。因此,我们目前的发现强调了CCG-203971和CCG-232601的治疗潜力,这可能被证明是靶向异常生物能量学的有希望的方法.
    RhoA-regulated gene transcription by serum response factor (SRF) and its transcriptional cofactor myocardin-related transcription factors (MRTFs) signaling pathway has emerged as a promising therapeutic target for pharmacological intervention in multiple diseases. Altered mitochondrial metabolism is one of the major hallmarks of cancer, therefore, this upregulation is a vulnerability that can be targeted with Rho/MRTF/SRF inhibitors. Recent advances identified a novel series of oxadiazole-thioether compounds that disrupt the SRF transcription, however, the direct molecular target of these compounds is unclear. Herein, we demonstrate the Rho/MRTF/SRF inhibition mechanism of CCG-203971 and CCG-232601 in normal cell lines of human lung fibroblasts and mouse myoblasts. Further studies investigated the role of these molecules in targeting mitochondrial function. We have shown that these molecules hyperacetylate histone H4K12 and H4K16 and regulate the genes involved in mitochondrial function and dynamics. These small molecule inhibitors regulate mitochondrial function as a compensatory mechanism by repressing oxidative phosphorylation and increasing glycolysis. Our data suggest that these CCG molecules are effective in inhibiting all the complexes of mitochondrial electron transport chains and further inducing oxidative stress. Therefore, our present findings highlight the therapeutic potential of CCG-203971 and CCG-232601, which may prove to be a promising approach to target aberrant bioenergetics.
    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:血清反应因子(SRF)在肌肉发育中很重要,组织修复,和神经元调节。
    目的:本研究旨在全面检查SRF对脊髓损伤(SCI)的影响及其对神经元轴突恢复和再生的影响。
    方法:研究人员建立大鼠脊髓损伤和脊髓初级神经元划痕损伤模型,观察神经元损伤后相关因子的表达。
    结果:我们发现SRF,拉斯,Raf,伤后cofilin水平升高并逐渐恢复至正常水平。之后,研究人员给予SCI大鼠一种SRF抑制剂(CCG1423),并用核磁共振和透射电子显微镜研究了其作用.SRF抑制剂啮齿动物的脊髓恢复和轴突再生比对照组差。SRF抑制剂组划痕损伤后原代神经元的凋亡明显增高。此外,研究人员利用慢病毒转染改变SRF在神经元中的表达.SRF过表达增加了神经元的迁移,而沉默SRF则减少了神经元的迁移。最后,采用Westernblotting和RT-PCR检测SRF表达改变后相关因子的表达变化。结果显示SRF过表达增加了Ras,Raf,和cofilin表达式。沉默SRF降低了Ras,Raf,和Cofilin表达式。
    结论:根据我们的研究,SRF通过激活“Ras-Raf-Cofilin”信号通路促进轴突再生。
    Serum response factor (SRF) is important in muscle development, tissue repair, and neuronal regulation.
    This research aims to thoroughly examine the effects of SRF on spinal cord injury (SCI) and its ability to significantly impact the recovery and regeneration of neuronal axons.
    The researchers created rat models of SCI and scratch injury to primary spinal cord neurons to observe the expression of relevant factors after neuronal injury.
    We found that the SRF, Ras, Raf, and cofilin levels increased after injury and gradually returned to normal levels. Afterward, researchers gave rats with SCI an SRF inhibitor (CCG1423) and studied the effects with nuclear magnetic resonance and transmission electron microscopy. The SRF inhibitor rodents had worse spinal cord recovery and axon regrowth than the control group. And the apoptosis of primary neurons after scratch injury was significantly higher in the SRF inhibitor group. Additionally, the researchers utilized lentiviral transfection to modify the SRF expression in neurons. SRF overexpression increased neuron migration while silencing SRF decreased it. Finally, Western blotting and RT-PCR were conducted to examine the expression changes of related factors upon altering SRF expression. The results revealed SRF overexpression increased Ras, Raf, and cofilin expression. Silencing SRF decreased Ras, Raf, and Cofilin expression.
    Based on our research, the SRF promotes axonal regeneration by activating the \"Ras-Raf-Cofilin\" signaling pathway.
    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
    在胸膜纤维化的进展过程中,胸膜间皮细胞(PMCs)经历一种称为间皮-间充质转化(MesoMT)的表型转换过程.在MesoMT期间,转化的PMC成为肌成纤维细胞,产生增加的细胞外基质(ECM)蛋白,包括胶原蛋白和纤连蛋白(FN1),这对纤维化的发展至关重要。这里,我们研究了调节源自HPMC的肌成纤维细胞中FN1表达的机制。我们发现Myocardin(Myocd),血清反应因子(SRF)的转录共激活因子和平滑肌和心肌分化的主要调节因子,强控制HPMCFN1基因表达。Myocd基因沉默显著抑制FN1表达。FN1启动子分析显示,Smad3结合元件的缺失降低了FN1启动子的活性,而推定的SRF结合元件的缺失增加了FN1启动子的活性。Smad3基因沉默降低FN1表达,而SRF基因沉默增加FN1表达。此外,SRF与Smad3竞争与Myocd的绑定。这些结果表明Myocd通过Smad3激活FN1表达,而SRF抑制HPMC中的FN1表达。在HPMC中,TGF-β诱导Smad3核定位,TGF-β刺激细胞核后,Myocd和Smad3之间的邻近连接信号显着增加,提示TGF-β促进Smad3的核易位以及Smad3与Myocd之间的相互作用。此外,将Myocd和Smad3共免疫沉淀并分离Myocd和Smad3蛋白彼此直接结合。染色质免疫沉淀分析显示Myocd在Smad3结合共有序列与FN1启动子相互作用。结果表明,Myocd通过相互作用和Smad3转录因子的激活来调节FN1基因的激活。
    During the progression of pleural fibrosis, pleural mesothelial cells (PMCs) undergo a phenotype switching process known as mesothelial-mesenchymal transition (MesoMT). During MesoMT, transformed PMCs become myofibroblasts that produce increased extracellular matrix (ECM) proteins, including collagen and fibronectin (FN1) that is critical to develop fibrosis. Here, we studied the mechanism that regulates FN1 expression in myofibroblasts derived from human pleural mesothelial cells (HPMCs). We found that myocardin (Myocd), a transcriptional coactivator of serum response factor (SRF) and a master regulator of smooth muscle and cardiac muscle differentiation, strongly controls FN1 gene expression. Myocd gene silencing markedly inhibited FN1 expression. FN1 promoter analysis revealed that deletion of the Smad3-binding element diminished FN1 promoter activity, whereas deletion of the putative SRF-binding element increased FN1 promoter activity. Smad3 gene silencing decreased FN1 expression, whereas SRF gene silencing increased FN1 expression. Moreover, SRF competes with Smad3 for binding to Myocd. These results indicate that Myocd activates FN1 expression through Smad3, whereas SRF inhibits FN1 expression in HPMCs. In HPMCs, TGF-β induced Smad3 nuclear localization, and the proximity ligation signal between Myocd and Smad3 was markedly increased after TGF-β stimulation at nucleus, suggesting that TGF-β facilitates nuclear translocation of Smad3 and interaction between Smad3 and Myocd. Moreover, Myocd and Smad3 were coimmunoprecipitated and isolated Myocd and Smad3 proteins directly bound each other. Chromatin immunoprecipitation assays revealed that Myocd interacts with the FN1 promoter at the Smad3-binding consensus sequence. The results indicate that Myocd regulates FN1 gene activation through interaction and activation of the Smad3 transcription factor.NEW & NOTEWORTHY During phenotype switching from mesothelial to mesenchymal, pleural mesothelial cells (PMCs) produce extracellular matrix (ECM) proteins, including collagen and fibronectin (FN1), critical components in the development of fibrosis. Here, we found that myocardin, a transcriptional coactivator of serum response factor (SRF), strongly activates FN1 expression through Smad3, whereas SRF inhibits FN1 expression. This study provides insights about the regulation of FN1 that could lead to the development of novel interventional approaches to prevent pleural fibrosis.
    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
    反应性星形胶质增生是中枢神经系统(CNS)损伤的常见病理标志,感染,和神经变性,反应性星形胶质细胞可以保护或损害正常的大脑功能。目前,调节神经保护性星形胶质细胞的机制和神经保护的程度知之甚少。这里,我们报道,成人星形胶质细胞中血清反应因子(SRF)的条件性缺失导致整个小鼠脑内星形胶质细胞反应性肥大.这些SrfGFAP-ERCKO星形胶质细胞不影响神经元存活,突触数,突触可塑性或学习和记忆。然而,Srf敲除小鼠的大脑对海藻酸诱导的兴奋毒性细胞死亡表现出神经保护作用。与人类神经退行性疾病有关,在帕金森病和阿尔茨海默病小鼠模型中,SrfGFAP-ERCKO星形胶质细胞消除黑质多巴胺能神经元死亡并减少b淀粉样蛋白斑块,分别。一起来看,这些发现将SRF确立为产生具有神经保护功能的反应性星形胶质细胞的关键分子开关,从而在神经退行性疾病的背景下减轻神经元损伤.
    Reactive astrogliosis is a common pathological hallmark of CNS injury, infection, and neurodegeneration, where reactive astrocytes can be protective or detrimental to normal brain functions. Currently, the mechanisms regulating neuroprotective astrocytes and the extent of neuroprotection are poorly understood. Here, we report that conditional deletion of serum response factor (SRF) in adult astrocytes causes reactive-like hypertrophic astrocytes throughout the mouse brain. These SrfGFAP-ERCKO astrocytes do not affect neuron survival, synapse numbers, synaptic plasticity or learning and memory. However, the brains of Srf knockout mice exhibited neuroprotection against kainic-acid induced excitotoxic cell death. Relevant to human neurodegenerative diseases, SrfGFAP-ERCKO astrocytes abrogate nigral dopaminergic neuron death and reduce β-amyloid plaques in mouse models of Parkinson\'s and Alzheimer\'s disease, respectively. Taken together, these findings establish SRF as a key molecular switch for the generation of reactive astrocytes with neuroprotective functions that attenuate neuronal injury in the setting of neurodegenerative diseases.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号