HACE1

HACE1
  • 文章类型: Case Reports
    有或没有癫痫发作(SPPRS)的痉挛性截瘫和精神运动发育迟缓是一种罕见的神经发育障碍,与HACE1基因的常染色体隐性突变有关。该病例报告介绍了一名11个月大女孩和她的妹妹患有SPPRS的临床特征和遗传分析,使其成为中东第三起报告的病例,沙特阿拉伯第二起病例。病人表现出张力减退,全球发育迟缓,说话延迟,吞咽困难,和反复呼吸道感染。HACE1基因中的纯合致病性变体(p。R664*)是通过遗传分析鉴定的,确认SPPRS的诊断。本病例报告强调了考虑临床表现变化的重要性,特别是在罕见疾病中,只有少数病例报告。需要进一步的研究和案例研究来更好地了解SPPRS的完整表型谱及其并发症。
    Spastic paraplegia and psychomotor retardation with or without seizures (SPPRS) is a rare neurodevelopmental disorder associated with autosomal recessive mutations in the HACE1 gene. This case report presents the clinical features and genetic analysis of an 11-month-old girl and her sister with SPPRS, making it the third reported case in the Middle East and the second in Saudi Arabia. The patient exhibited hypotonia, global developmental delay, speech delay, swallowing difficulties, and recurrent respiratory infections. A homozygous pathogenic variant in the HACE1 gene (p.R664*) was identified through genetic analysis, confirming the diagnosis of SPPRS. This case report emphasizes the importance of considering variations in clinical presentation, especially in rare disorders where only a few cases are reported. Further research and case studies are needed to better understand the complete phenotypic spectrum of SPPRS and its complications.
    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
    目的:本研究探讨HECT结构域和含有E3泛素蛋白连接酶1(HACE1)的锚蛋白重复序列在食管癌(ESCA)中的意义及其通过诱导RAC1泛素化和降解调节ESCA的潜在机制。方法:进行了ESCA临床组织和细胞系中HACE1的表征研究。接下来,通过沉默和过表达HACE1检测HACE1对ESCA细胞生物学行为的影响。使用来自String网站的数据分析涉及HACE1的蛋白质-蛋白质相互作用(PPIs)。使用蛋白酶体抑制剂MG132验证了HACE1在RAC1蛋白泛素化中的功能。通过在荷瘤裸鼠模型中应用RAC1抑制剂EHop-016来阐明HACE1通过RAC1对ESCA细胞的作用。为了建立HACE1和TRIP12之间的关系,进行了救援实验,主要评价TRIP12沉默对HACE1介导的RAC1调节的体内外作用。HACE1和TRIP12之间的PPI及其亚细胞定位通过免疫共沉淀和免疫荧光染色进一步表征,分别。结果:ESCA细胞中HACE1蛋白表达明显减少,而正常组织中HACE1蛋白表达上调。HACE1过表达抑制ESCA细胞的恶性生物学行为,导致小鼠肿瘤生长受限。这种效应与促进RAC1蛋白泛素化和随后的降解有关。相反,沉默HACE1表现出对比结果。PPI存在于HACE1和TRIP12之间,并因其相似的亚细胞定位而复合。有趣的是,TRIP12抑制阻断HACE1驱动的RAC1泛素化,减轻HACE1对ESCA细胞的抑制作用,减轻荷瘤裸鼠模型中的肿瘤生长。结论:ESCA细胞中HACE1表达下调,提示它通过TRIP12介导的泛素化诱导RAC1蛋白降解来抑制ESCA进展。
    Objective: This study investigated the significance of HECT domain and ankyrin repeat containing E3 ubiquitin protein ligase 1 (HACE1) in esophageal cancer (ESCA) and its underlying mechanism in ESCA regulation through the induction of RAC1 ubiquitination and degradation. Methods: Characterization studies of HACE1 in ESCA clinical tissues and cell lines were performed. Next, the effects of HACE1 on the biological behavior of ESCA cells were examined by silencing and overexpressing HACE1. Protein-protein interactions (PPIs) involving HACE1 were analyzed using data from the String website. The function of HACE1 in RAC1 protein ubiquitination was validated using the proteasome inhibitor MG132. The effects of HACE1 on ESCA cells through RAC1 were elucidated by applying the RAC1 inhibitor EHop-016 in a tumor-bearing nude mouse model. To establish the relationship between HACE1 and TRIP12, rescue experiments were conducted, mainly to evaluate the effect of TRIP12 silencing on HACE1-mediated RAC1 regulation in vitro and in vivo. The PPI between HACE1 and TRIP12 and their subcellular localization were further characterized through co-immunoprecipitation and immunofluorescence staining assays, respectively. Results: HACE1 protein expression was notably diminished in ESCA cells but upregulated in normal tissues. HACE1 overexpression inhibited the malignant biological behavior of ESCA cells, leading to restrained tumor growth in mice. This effect was coupled with the promotion of RAC1 protein ubiquitination and subsequent degradation. Conversely, silencing HACE1 exhibited contrasting results. PPI existed between HACE1 and TRIP12, compounded by their similar subcellular localization. Intriguingly, TRIP12 inhibition blocked HACE1-driven RAC1 ubiquitination and mitigated the inhibitory effects of HACE1 on ESCA cells, alleviating tumor growth in the tumor-bearing nude mouse model. Conclusion: HACE1 expression was downregulated in ESCA cells, suggesting that it curbs ESCA progression by inducing RAC1 protein degradation through TRIP12-mediated ubiquitination.
    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
    食管癌(EC)是消化系统恶性程度最高的恶性肿瘤之一,在全球范围内具有较高的临床发病率。厚朴酚,一种天然化合物,对许多癌症有抗癌作用,包括食管癌,但是潜在的机制尚未完全阐明。这里,我们首先发现厚朴酚抑制食管癌细胞的增殖,并以剂量和时间依赖性的方式增强其自噬活性。这项研究表明,厚朴酚增加LC3II的蛋白质水平,伴随着食管癌细胞和异种移植肿瘤中HACE1蛋白水平的增加。产生HACE1敲除(KO)细胞系,HACE1的消融消除了厚朴酚对食管癌细胞的抗增殖和自噬诱导作用。此外,我们的结果表明厚朴酚主要在转录水平上促进HACE1的表达。因此,本研究表明厚朴酚主要通过激活HACE1-OPTN轴介导的自噬发挥抗肿瘤作用。它可以被认为是一种有前途的食管癌治疗药物。
    Esophagus cancer (EC) is one of the most aggressive malignant digestive system tumors and has a high clinical incidence worldwide. Magnolol, a natural compound, has anticancer effects on many cancers, including esophageal carcinoma, but the underlying mechanism has not been fully elucidated. Here, we first find that magnolol inhibits the proliferation of esophageal carcinoma cells and enhances their autophagy activity in a dose- and time-dependent manner. This study demonstrates that magnolol increases the protein levels of LC3 II, accompanied by increased HACE1 protein levels in both esophageal carcinoma cells and xenograft tumors. HACE1-knockout (KO) cell lines are generated, and the ablation of HACE1 eliminates the anti-proliferative and autophagy-inducing effects of magnolol on esophageal carcinoma cells. Additionally, our results show that magnolol primarily promotes HACE1 expression at the transcriptional level. Therefore, this study shows that magnolol primarily exerts its antitumor effect by activating HACE1-OPTN axis-mediated autophagy. It can be considered a promising therapeutic drug for esophageal carcinoma.
    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
    HACE1是含有HECT结构域的E3连接酶的成员,具有909个氨基酸残基,含有N末端锚蛋白重复序列(ANK)和C末端HECT结构域。以前,研究表明,HACE1在人类肿瘤中无活性,并在肿瘤的启动中起着至关重要的作用,programming,和恶性肿瘤的侵袭。最近的研究表明,HACE1可能与神经退行性疾病如阿尔茨海默病密切相关。帕金森病,和亨廷顿病。HACE1与其底物相互作用,包括与Ras相关的C3肉毒杆菌毒素底物1(Rac1),核因子红系2相关因子2(Nrf2),肿瘤坏死因子受体(TNFR),和视神经磷酸酶(OPTN),通过它参与几个病理生理过程,如氧化应激,自噬和炎症。因此,在这次审查中,我们详细描述了HACE1的基本底物,并阐明了HACE1参与神经退行性疾病的病理生理过程。我们为神经退行性疾病提供了新的分子靶点。
    HACE1 is a member of the HECT domain-containing E3 ligases with 909 amino acid residues, containing N-terminal ankyrin-repeats (ANK) and C-terminal HECT domain. Previously, it was shown that HACE1 is inactive in human tumors and plays a crucial role in the initiation, progression, and invasion of malignant tumors. Recent studies indicated that HACE1 might be closely involved in neurodegenerative diseases such as Alzheimer\'s disease, Parkinson\'s disease, and Huntington\'s disease. HACE1 interacts with its substrates, including Ras-related C3 botulinum toxin substrate 1 (Rac1), nuclear factor erythroid 2-related factor 2 (Nrf2), tumor necrosis factor receptor (TNFR), and optineurin (OPTN), through which participates in several pathophysiological processes, such as oxidative stress, autophagy and inflammation. Therefore, in this review, we elaborately describe the essential substrates of HACE1 and illuminate the pathophysiological processes by which HACE1 is involved in neurodegenerative diseases. We provide a new molecular target for neurodegenerative diseases.
    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:神经上皮转化基因1(NET1)是RhoA亚家族鸟嘌呤核苷酸交换因子,控制着广泛的生物学过程。然而,其在减数分裂卵母细胞中的作用尚不清楚.我们在此证明NET1-HACE1-RAC1途径介导卵母细胞成熟进程中的减数分裂缺陷。
    方法:使用特定的小干扰RNA在小鼠卵母细胞中降低NET1。主轴总成,染色体排列,肌动蛋白帽,和染色体分布通过免疫染色可视化,并在共聚焦显微镜下分析。我们还应用了质谱,和这项调查的蛋白质印迹分析。
    结果:我们的结果显示NET1在GV阶段定位于细胞核,在GVBD之后,NET1定位于细胞质,主要分布在染色体周围,与减数分裂进展相称。NET1位于细胞质中,并在MI和MII阶段明显积累在纺锤体上。耗尽Net1的小鼠卵母细胞表现出异常的第一极体挤压和不对称分裂缺陷。我们还确定,Net1耗竭导致小鼠卵母细胞中RAC1蛋白表达减少,NET1保护RAC1免受HACE1降解,这对于肌动蛋白动力学和减数分裂纺锤体形成至关重要。重要的是,在Net1缺失的卵母细胞中的外源性RAC1表达显著挽救了这些缺陷.
    结论:我们的结果表明,NET1在小鼠卵母细胞减数分裂过程中在纺锤体稳定性和肌动蛋白动力学中表现出多种作用。
    BACKGROUND: Neuroepithelial transforming gene 1 (NET1) is a RhoA subfamily guanine nucleotide exchange factor that governs a wide array of biological processes. However, its roles in meiotic oocyte remain unclear. We herein demonstrated that the NET1-HACE1-RAC1 pathway mediates meiotic defects in the progression of oocyte maturation.
    METHODS: NET1 was reduced using a specific small interfering RNA in mouse oocytes. Spindle assembly, chromosomal alignment, the actin cap, and chromosomal spreads were visualized by immunostaining and analyzed under confocal microscopy. We also applied mass spectroscopy, and western blot analysis for this investigation.
    RESULTS: Our results revealed that NET1 was localized to the nucleus at the GV stage, and that after GVBD, NET1 was localized to the cytoplasm and predominantly distributed around the chromosomes, commensurate with meiotic progression. NET1 resided in the cytoplasm and significantly accumulated on the spindle at the MI and MII stages. Mouse oocytes depleted of Net1 exhibited aberrant first polar body extrusion and asymmetric division defects. We also determined that Net1 depletion resulted in reduced RAC1 protein expression in mouse oocytes, and that NET1 protected RAC1 from degradation by HACE1, and it was essential for actin dynamics and meiotic spindle formation. Importantly, exogenous RAC1 expression in Net1-depleted oocytes significantly rescued these defects.
    CONCLUSIONS: Our results suggest that NET1 exhibits multiple roles in spindle stability and actin dynamics during mouse oocyte meiosis.
    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
    HACE1是HECT家族E3泛素蛋白连接酶,具有广泛但不完全了解的肿瘤抑制活性。这里,我们报道了HACE1和含有哺乳动物雷帕霉素靶蛋白(mTOR)的信号复合物之间以前未发现的联系.HACE1通过以E3连接酶依赖性方式降低mTOR稳定性来阻断mTORC1和mTORC2活性。机械上,当RAC1与mTOR复合物相关时,HACE1结合并泛素化Ras相关的C3肉毒杆菌毒素底物1(RAC1),包括在粘着灶处,导致RAC1的蛋白酶体降解。这进而降低mTOR降低mTORC1和mTORC2活性的稳定性。缺乏HACE1的细胞显示增强的mTORC1/2活性,通过化学或遗传RAC1失活而逆转,但在表达HACE1不敏感突变体的细胞中却没有逆转,RAC1K147R.在体内,Rac1缺失逆转Hace1-/-小鼠的KRasG12D驱动的肺肿瘤中mTOR表达增强。HACE1与mTOR和RAC1共定位,导致mTOR蛋白稳定性的RAC1依赖性丧失。一起,我们的数据表明,HACE1通过靶向mTOR相关复合物内的RAC1来使mTOR不稳定,揭示了一个独特的泛素依赖性过程来控制mTOR信号复合物的活性。
    HACE1 is a HECT family E3 ubiquitin-protein ligase with broad but incompletely understood tumor suppressor activity. Here, we report a previously unrecognized link between HACE1 and signaling complexes containing mammalian target of rapamycin (mTOR). HACE1 blocks mTORC1 and mTORC2 activities by reducing mTOR stability in an E3 ligase-dependent manner. Mechanistically, HACE1 binds to and ubiquitylates Ras-related C3 botulinum toxin substrate 1 (RAC1) when RAC1 is associated with mTOR complexes, including at focal adhesions, leading to proteasomal degradation of RAC1. This in turn decreases the stability of mTOR to reduce mTORC1 and mTORC2 activity. HACE1 deficient cells show enhanced mTORC1/2 activity, which is reversed by chemical or genetic RAC1 inactivation but not in cells expressing the HACE1-insensitive mutant, RAC1K147R . In vivo, Rac1 deletion reverses enhanced mTOR expression in KRasG12D -driven lung tumors of Hace1-/- mice. HACE1 co-localizes with mTOR and RAC1, resulting in RAC1-dependent loss of mTOR protein stability. Together, our data demonstrate that HACE1 destabilizes mTOR by targeting RAC1 within mTOR-associated complexes, revealing a unique ubiquitin-dependent process to control the activity of mTOR signaling complexes.
    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
    HACE1是含有HECT型E3泛素连接酶的锚蛋白重复(AKR),其与多种底物相互作用并使其泛素化。虽然HACE1是一种众所周知的肿瘤抑制剂,其结构和泛素化模式尚不清楚。作者介绍了人HACE1的冷冻EM结构以及体外功能研究,这些研究提供了对HACE1酶活性如何调节的见解。HACE1包含一个N端AKR结构域,中间(MID)域,和C末端HECT结构域。其独特的G形结构作为同二聚体相互作用,与以反平行方式排列的单体。在这种二聚体排列中,HACE1泛素化活性受到阻碍,因为一个单体的N端螺旋限制了对另一个单体的C端结构域的访问。体外泛素化试验,氢-氘交换质谱(HDX-MS)分析,诱变,计算机建模表明,HACE1MID结构域与AKRs在RAC1底物识别中起着至关重要的作用。
    HACE1 is an ankyrin repeat (AKR) containing HECT-type E3 ubiquitin ligase that interacts with and ubiquitinates multiple substrates. While HACE1 is a well-known tumor suppressor, its structure and mode of ubiquitination are not understood. The authors present the cryo-EM structures of human HACE1 along with in vitro functional studies that provide insights into how the enzymatic activity of HACE1 is regulated. HACE1 comprises of an N-terminal AKR domain, a middle (MID) domain, and a C-terminal HECT domain. Its unique G-shaped architecture interacts as a homodimer, with monomers arranged in an antiparallel manner. In this dimeric arrangement, HACE1 ubiquitination activity is hampered, as the N-terminal helix of one monomer restricts access to the C-terminal domain of the other. The in vitro ubiquitination assays, hydrogen-deuterium exchange mass spectrometry (HDX-MS) analysis, mutagenesis, and in silico modeling suggest that the HACE1 MID domain plays a crucial role along with the AKRs in RAC1 substrate recognition.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Case Reports
    痉挛型截瘫和精神运动发育迟缓伴或不伴癫痫发作(SPPRS,OMIM616756)是由HACE1基因的双等位基因致病变异所惹起的罕见遗传病。最初,据报道,这些突变与肿瘤易感性有关.尽管如此,通过2015年的全外显子组测序,HACE1突变被认为是一种新的常染色体隐性遗传神经发育障碍的原因,其特征是痉挛,肌张力减退,智力残疾。迄今为止,已经描述了14个HACE1致病性变体;这些变体具有功能丧失效应,导致具有不同严重度的临床表现。然而,尚未提及HACE1基因中的严重缺失是导致痉挛性截瘫的原因。这里,我们报告了一例2岁男性痉挛的临床病例,主要影响下肢,和发育迟缓。外显子组测序,染色体微阵列分析,和mRNA分析用于鉴定致病基因。我们发现临床发现是由于先前未描述的HACE1双等位基因缺失。我们确定了外显子7:c的缺失。(5341_535-1)_(6171_618-1)del(NM_020771.4)和6q16.3基因座中的总缺失,影响整个HACE1基因:g.105018931_105337494del,(GRCh37)。由于可能存在错误的纯合性结果,因此需要对HACE1中具有原始纯合性突变的患者进行全面的诊断方法。据报道超过80%的所述突变是纯合的。初始半合子很难通过定量方法检测,这可能会对痉挛性截瘫患者的分子诊断鉴定提出挑战。
    Spastic paraplegia and psychomotor retardation with or without seizures (SPPRS, OMIM 616756) is a rare genetic disease caused by biallelic pathogenic variants in the HACE1 gene. Originally, these mutations have been reported to be implicated in tumor predisposition. Nonetheless, via whole exome sequencing in 2015, HACE1 mutations were suggested to be the cause of a new autosomal recessive neurodevelopmental disorder, which is characterized by spasticity, muscular hypotonia, and intellectual disability. To date, 14 HACE1 pathogenic variants have been described; these variants have a loss-of-function effect that leads to clinical presentations with variable severities. However, gross deletions in the HACE1 gene have not yet been mentioned as a cause of spastic paraplegia. Here, we report a clinical case involving a 2-year-old male presenting with spasticity, mainly affecting the lower limbs, and developmental delay. Exome sequencing, chromosomal microarray analysis, and mRNA analysis were used to identify the causative gene. We revealed that the clinical findings were due to previously undescribed HACE1 biallelic deletions. We identified the deletion of exon 7: c.(534+1_535-1)_(617+1_618-1)del (NM_020771.4) and the gross deletion in the 6q16.3 locus, which affected the entire HACE1 gene: g.105018931_105337494del, (GRCh37). A comprehensive diagnostic approach for the patients with originally homozygous mutations in HACE1 is required since false homozygosity results are possible. More than 80% of the described mutations were reported to be homozygous. Initial hemizygosity is hard to detect by quantitative methods, and this may challenge molecular diagnostic identification in patients with spastic paraplegia.
    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
    HECT结构域和含有E3泛素蛋白连接酶1(Hace1)的锚蛋白重复序列是多种病理疾病的关键介质。然而,关于Hace1在心肌缺血/再灌注损伤中的作用的研究很少。这里,我们在体外使用缺氧/复氧(H/R)损伤的心肌细胞研究了Hace1在心肌缺血/再灌注损伤中的功能作用。在暴露于H/R的心脏细胞中观察到Hace1水平降低。Hace1过表达的心肌细胞对H/R损伤具有抗性,凋亡减少,降低氧化应激,和抑制的炎症反应。随后的分析显示,Hace1过表达增强了核因子(红系衍生的2)样2(Nrf2)的核易位激活,并增加了H/R暴露的心脏细胞中Nrf2的转录活性。Kelch样ECH相关蛋白1(Keap1)的敲除降低了Hace1对Nrf2激活的调节作用。此外,抑制Nrf2可逆转Hace1在H/R损伤的心肌细胞中引起的心脏保护作用。总之,这些数据表明Hace1过表达通过通过Keap1增强Nrf2途径减轻心肌H/R损伤.这项工作强调了Hace1在心肌缺血/再灌注损伤中的可能作用,并建议Hace1作为开发心脏保护性治疗的候选靶标。
    HECT domain and ankyrin repeat containing E3 ubiquitin protein ligase 1 (Hace1) is a crucial mediator of multiple pathological disorders. However, there are few studies regarding the role of Hace1 in myocardial ischemia/reperfusion injury. Here, we studied the functional role of Hace1 on myocardial ischemia/reperfusion injury using hypoxia/reoxygenation (H/R)-injured cardiac cells in vitro. Reduced levels of Hace1 were observed in H/R-exposed cardiac cells. Hace1-overexpressed cardiac cells were resistant to H/R injuries with reduced apoptosis, lowered oxidative stress, and a suppressed inflammatory response. Subsequent analysis revealed that Hace1 overexpression enhanced the activation of nuclear translocation of nuclear factor (erythroid-derived 2)-like 2 (Nrf2) and increased the transcriptional activity of Nrf2 in H/R-exposed cardiac cells. The knockout of kelch-like ECH-associated protein 1 (Keap1) diminished the regulatory role of Hace1 on Nrf2 activation. Additionally, inhibiting Nrf2 reversed Hace1-elicited cardioprotective effects in H/R-injured cardiac cells. In short, these data demonstrated that Hace1 overexpression mitigated myocardial H/R injury by enhancing the Nrf2 pathway via Keap1. This work underlines a possible role of Hace1 in myocardial ischemia/reperfusion injury and suggests Hace1 as a candidate target for exploiting cardioprotective therapy.
    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
    暂无摘要。
    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