beta-Arrestin 1

β - 抑制素 1
  • 文章类型: Journal Article
    非典型趋化因子受体3(ACKR3)属于G蛋白偶联受体家族,但不通过G蛋白发出信号。控制ACKR3活化的功能选择性和构象动力学的结构特性知之甚少。这里,我们结合了氢/氘交换质谱,定点诱变,和分子动力学模拟,以检查不同功效的ACKR3配体的结合模式和作用机理。我们的结果表明,ACKR3的激活或抑制受螺旋6,胞内环2和螺旋7的细胞内构象变化控制,而DRY基序在这两个过程中都受到保护。此外,我们确定了β-抑制蛋白1结合时ACKR3的结合位点和变构调节。总之,这项研究强调了小配体的结构-功能关系,β-抑制蛋白1的结合模式,激活动力学,以及ACKR3中的非典型动态特征可能导致其无法激活G蛋白。
    Atypical Chemokine Receptor 3 (ACKR3) belongs to the G protein-coupled receptor family but it does not signal through G proteins. The structural properties that govern the functional selectivity and the conformational dynamics of ACKR3 activation are poorly understood. Here, we combined hydrogen/deuterium exchange mass spectrometry, site-directed mutagenesis, and molecular dynamics simulations to examine the binding mode and mechanism of action of ACKR3 ligands of different efficacies. Our results show that activation or inhibition of ACKR3 is governed by intracellular conformational changes of helix 6, intracellular loop 2, and helix 7, while the DRY motif becomes protected during both processes. Moreover, we identified the binding sites and the allosteric modulation of ACKR3 upon β-arrestin 1 binding. In summary, this study highlights the structure-function relationship of small ligands, the binding mode of β-arrestin 1, the activation dynamics, and the atypical dynamic features in ACKR3 that may contribute to its inability to activate G proteins.
    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
    将成纤维细胞募集到肿瘤中并将其激活为癌症相关成纤维细胞(CAF)是肿瘤细胞用于指导细胞外基质(ECM)重塑的策略。入侵,和转移,强调需要研究驱动CAF功能的分子机制。内皮素-1(ET-1)调节癌症与基质之间的联系,并促进浆液性卵巢癌(SOC)的进展。通过与内皮素A(ETA)和B(ETB)受体结合,ET-1能够募集β-arrestin1(β-arr1)并形成协调肿瘤进展的信号复合物。然而,ET-1受体如何“教育”人类卵巢成纤维细胞(HOFs)产生改变的ECM并促进转移仍有待阐明。这项研究确定ET-1是能够蛋白水解ECM重塑的CAF活化和含有具有转移倾向的癌细胞的异型球体产生的关键因素。自分泌/旁分泌ET-1/ETA/BR/β-arr1环增强HOF增殖,上调CAF标记表达,分泌促炎细胞因子,并增加胶原蛋白的收缩力,和细胞运动。此外,ET-1通过促进侵袭小体的裂解活性和整合素β1的激活来促进ECM重塑。此外,ET-1信号传导支持异型HOF/SOC球状体的形成,具有增强的迁移通过间皮单层的能力,入侵,代表转移单位。ETA/BR或β-arr1沉默的阻断可防止CAF活化,invadosome函数,间皮间隙,异型球状体的侵袭能力。在体内,使用波生坦(BOS)的ETA/BR的治疗性抑制显着降低了组合HOFs/SOC细胞的转移潜力,与增强对肿瘤细胞和基质成分的凋亡作用有关。这些发现支持一个模型,其中ET-1/β-arr1通过CAF激活增强肿瘤/基质相互作用,并促进SOC细胞的存活和转移特性,这可以被ETA/BR拮抗剂抵消。
    Recruitment of fibroblasts to tumors and their activation into cancer-associated fibroblasts (CAFs) is a strategy used by tumor cells to direct extracellular matrix (ECM) remodeling, invasion, and metastasis, highlighting the need to investigate the molecular mechanisms driving CAF function. Endothelin-1 (ET-1) regulates the communication between cancer and stroma and facilitates the progression of serous ovarian cancer (SOC). By binding to Endothelin A (ETA) and B (ETB) receptors, ET-1 enables the recruitment of β-arrestin1 (β-arr1) and the formation of signaling complexes that coordinate tumor progression. However, how ET-1 receptors might \"educate\" human ovarian fibroblasts (HOFs) to produce altered ECM and promote metastasis remains to be elucidated. This study identifies ET-1 as a pivotal factor in the activation of CAFs capable of proteolytic ECM remodeling and the generation of heterotypic spheroids containing cancer cells with a propensity to metastasize. An autocrine/paracrine ET-1/ETA/BR/β-arr1 loop enhances HOF proliferation, upregulates CAF marker expression, secretes pro-inflammatory cytokines, and increases collagen contractility, and cell motility. Furthermore, ET-1 facilitates ECM remodeling by promoting the lytic activity of invadosome and activation of integrin β1. In addition, ET-1 signaling supports the formation of heterotypic HOF/SOC spheroids with enhanced ability to migrate through the mesothelial monolayer, and invade, representing metastatic units. The blockade of ETA/BR or β-arr1 silencing prevents CAF activation, invadosome function, mesothelial clearance, and the invasive ability of heterotypic spheroids. In vivo, therapeutic inhibition of ETA/BR using bosentan (BOS) significantly reduces the metastatic potential of combined HOFs/SOC cells, associated with enhanced apoptotic effects on tumor cells and stromal components. These findings support a model in which ET-1/β-arr1 reinforces tumor/stroma interaction through CAF activation and fosters the survival and metastatic properties of SOC cells, which could be counteracted by ETA/BR antagonists.
    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
    µ-阿片受体(µOR)是疼痛管理的重要靶标1,对µOR药物作用的分子理解将有助于开发更好的治疗方法。在这里我们展示,利用双电子-电子共振和单分子荧光共振能量转移,μOR的配体特异性构象变化如何在换能器水平转化为广泛的内在效力。我们确定了受体的细胞质表面的几种构象,这些构象在不同的时间尺度上相互转换,包括能够结合G蛋白的预激活构象,和完全激活的构象显着降低三元复合物内的GDP亲和力。β-抑制蛋白-1与μOR核心结合位点的相互作用似乎特异性较低,并且与Gi的结合相比亲和力低得多。
    The µ-opioid receptor (µOR) is an important target for pain management1 and molecular understanding of drug action on µOR will facilitate the development of better therapeutics. Here we show, using double electron-electron resonance and single-molecule fluorescence resonance energy transfer, how ligand-specific conformational changes of µOR translate into a broad range of intrinsic efficacies at the transducer level. We identify several conformations of the cytoplasmic face of the receptor that interconvert on different timescales, including a pre-activated conformation that is capable of G-protein binding, and a fully activated conformation that markedly reduces GDP affinity within the ternary complex. Interaction of β-arrestin-1 with the μOR core binding site appears less specific and occurs with much lower affinity than binding of Gi.
    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
    肺腺癌(LUAD)是对公众健康的严重威胁,并伴随着全球发病率和死亡率的增加。神经元PAS结构域蛋白2(NPAS2)已被证实为LUAD的癌基因;然而,对其分子机制知之甚少。这里,在LUAD细胞系和16HBE细胞中检测到NPAS2的表达水平。进行了功能增益和功能丧失实验。细胞计数试剂盒-8,集落形成,流式细胞术,进行伤口愈合和Transwell测定以评估细胞增殖,凋亡,移民和入侵,分别。通过耗氧率(OCR)评估葡萄糖代谢的重编程,复杂的活动,乳酸生产和葡萄糖消耗。通过蛋白质印迹检查关键蛋白的表达。我们证明了LUAD细胞系中NPAS2和β-抑制素-1(ARRB1)的异常上调。发现ARRB1是NPAS2的关键转录因子,在NPAS2的启动子区域内具有结合位点,从而引起其转录激活。功能实验表明,NPAS2耗竭通过抑制细胞增殖显著抑制A549细胞的恶性行为,迁移,侵袭和上皮间质转化及促进细胞凋亡。同时,NPAS2耗竭增加了OCR和复合物的活性(I,II,III和V),并减少了A549细胞中的乳酸产生和葡萄糖摄取,表明NPAS2消耗抑制有氧糖酵解,伴有糖酵解酶表达降低。然而,ARRB1过表达部分恢复了NPAS2敲低引起的变化。总之,我们的研究表明,ARRB1可以转录激活NPAS2,促进恶性活动和糖酵解,并最终促进LUAD的发展,证明了治疗LUAD的新治疗策略。
    Lung adenocarcinoma (LUAD) is a serious threat to public health and is accompanied by increased morbidity and mortality worldwide. Neuronal PAS domain protein2 (NPAS2) has been confirmed as an oncogene in LUAD; however, little is known about its molecular mechanism. Here, the expression level of NPAS2 was detected in LUAD cell lines and 16HBE cells. Gain- and loss-of-function experiments were performed. Cell Counting Kit-8, colony formation, flow cytometry, wound-healing and Transwell assays were conducted to assess cell proliferation, apoptosis, migration and invasion, respectively. Reprogramming of glucose metabolism was evaluated via oxygen consumption rate (OCR), complexes activities, lactic production and glucose consumption. The expression of critical proteins was examined by western blot. We demonstrated aberrant upregulation of NPAS2 and β-arrestin-1 (ARRB1) in LUAD cell lines. ARRB1 was found to be a critical transcription factor of NPAS2 with binding sites within the promoter region of NPAS2, thereby causing its transcriptional activation. Functional experiments revealed that NPAS2 depletion significantly inhibited the malignant behaviours of A549 cells by suppressing cell proliferation, migration, invasion and epithelial-mesenchymal transition and promoting cell apoptosis. Meanwhile, NPAS2 depletion increased OCR and activities of complexes (I, II, III and V), and reduced lactic acid production and glucose uptake in A549 cells, indicating that NPAS2 depletion inhibited aerobic glycolysis, accompanied by reduced expression of glycolytic enzymes. However, the changes caused by NPAS2 knockdown were partly restored by ARRB1 overexpression. In conclusion, our study suggests that ARRB1 could transcriptionally activate NPAS2, facilitating malignant activities and glycolysis, and ultimately promoting the progression of LUAD, proving a novel therapeutic strategy for the treatment of LUAD.
    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
    胰高血糖素样肽1受体(GLP-1R)是治疗2型糖尿病和肥胖症的有效临床靶标。与大多数G蛋白偶联受体(GPCRs)不同,GLP-1R经历不需要β-抑制素的非典型内在化模式.虽然已观察到临床使用的GLP-1R激动剂之间的GLP-1R运输和β-抑制素募集的差异,G蛋白偶联受体激酶(GRKs)在影响这些途径中的作用尚未得到全面评估.在这项研究中,我们量化了GRKs对激动剂介导的GLP-1R内化和β-抑制素募集谱的贡献,使用内源性β-抑制素,或者使用CRISPR/Cas9基因组编辑敲除非视觉GRK。我们的结果证实了先前建立的GLP-1R内化的非典型β-抑制蛋白非依赖性模式,并揭示了GLP-1R内化依赖于GRK的表达。有趣的是,激动剂介导的GLP-1Rβ-抑制素1和β-抑制素2募集受到内源性GRK敲除的不同影响,β-抑制素1募集对GRK敲除比β-抑制素2募集更敏感。此外,在新产生的GRK2/3/4/5/6HEK293细胞中个别过表达GRK2、GRK3、GRK5或GRK6,拯救的激动剂介导的β-抑制素1募集和内化谱达到相似水平,这表明没有特定的GRK同工型驱动这些途径。这项研究推进了对激动剂介导的GLP-1R内化的机制理解,并提供了GRKs如何微调GLP-1R信号的新见解。
    The glucagon-like peptide 1 receptor (GLP-1R) is a validated clinical target for the treatment of type 2 diabetes and obesity. Unlike most G protein-coupled receptors (GPCRs), the GLP-1R undergoes an atypical mode of internalisation that does not require β-arrestins. While differences in GLP-1R trafficking and β-arrestin recruitment have been observed between clinically used GLP-1R agonists, the role of G protein-coupled receptor kinases (GRKs) in affecting these pathways has not been comprehensively assessed. In this study, we quantified the contribution of GRKs to agonist-mediated GLP-1R internalisation and β-arrestin recruitment profiles using cells where endogenous β-arrestins, or non-visual GRKs were knocked out using CRISPR/Cas9 genome editing. Our results confirm the previously established atypical β-arrestin-independent mode of GLP-1R internalisation and revealed that GLP-1R internalisation is dependent on the expression of GRKs. Interestingly, agonist-mediated GLP-1R β-arrestin 1 and β-arrestin 2 recruitment were differentially affected by endogenous GRK knockout with β-arrestin 1 recruitment more sensitive to GRK knockout than β-arrestin 2 recruitment. Moreover, individual overexpression of GRK2, GRK3, GRK5 or GRK6 in a newly generated GRK2/3/4/5/6 HEK293 cells, rescued agonist-mediated β-arrestin 1 recruitment and internalisation profiles to similar levels, suggesting that there is no specific GRK isoform that drives these pathways. This study advances mechanistic understanding of agonist-mediated GLP-1R internalisation and provides novel insights into how GRKs may fine-tune GLP-1R signalling.
    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
    β-抑制蛋白-1已被证明参与几种疾病的炎症反应的调节。因此,本研究旨在探讨巨噬细胞β-抑制素-1在溃疡性结肠炎(UC)发生发展中的作用。建立髓样β-抑制蛋白-1条件性敲除小鼠模型以探索巨噬细胞β-抑制蛋白-1的作用。采用DSS建立溃疡性结肠炎小鼠模型,使用TNF-α作为体外炎症应激源。通过Westernblot和免疫荧光法检测β-arrestin-1的表达水平,在DSS诱导的结肠炎模型中,通过临床评分和H&E染色评估疾病严重程度。在体外实验中,使用实时PCR检测炎性细胞因子的水平。通过双荧光素酶报告系统检测NF-κB的激活,westernblot,和电泳迁移率变动分析(EMSA)。BAY11-7082用于抑制NF-κB活化。我们的结果表明,在DSS诱导的结肠炎小鼠或TNF-α攻击下,单核细胞/巨噬细胞中β-抑制蛋白-1的水平升高。此外,有条件地敲除髓样β-抑制素-1的表达减轻了疾病的严重程度,而敲除β-arrestin-1的表达降低了炎性细胞因子的水平。此外,NF-κB被确定为β-抑制蛋白-1启动子的中心调控元件,在TNF-α攻击下,使用BAY11-7082抑制NF-κB激活可降低β-抑制素-1的水平。β-抑制蛋白-1通过在TNF-α攻击下增强与IκBα和IKK的结合而导致NF-κB信号通路的激活。一起来看,我们的研究结果表明,巨噬细胞β-抑制蛋白-1通过与NF-κB信号的相互作用促进DSS诱导的结肠炎的恶化,从而突出了UC治疗的新靶点。
    β-arrestin-1 has been demonstrated to participate in the regulation of inflammatory reactions in several diseases. Thus, this study aimed to investigate the role of macrophage β-arrestin-1 in the pathogenesis and progression of ulcerative colitis (UC). A myeloid β-arrestin-1 conditional knockout mouse model was generated to explore the role of macrophage β-arrestin-1. DSS was employed for the establishment of an ulcerative colitis mouse model, using TNF-α as an inflammatory stressor in vitro. The expression level of β-arrestin-1 was detected via western blot and immunofluorescence assays, whilst disease severity was evaluated by clinical score and H&E staining in the DSS-induced colitis model. In the in vitro experiments, the levels of inflammatory cytokines were examined using real-time PCR. NF-κB activation was detected through the double luciferase reporter system, western blot, and electrophoretic mobility shift assay (EMSA). BAY11-7082 was used to inhibit NF-κB activation. Our results exposed that the level of β-arrestin-1 was increased in monocytes/macrophages derived from DSS-induced colitis mice or under the TNF-α challenge. Moreover, conditionally knocking out the expression of myeloid β-arrestin-1 alleviated disease severity, while knocking out the expression of β-arrestin-1 decreased the levels of inflammatory cytokines. Additionally, NF-κB was identified as a central regulatory element of β-arrestin-1 promoter, and using BAY11-7082 to inhibit NF-κB activation lowered the level of β-arrestin-1 under TNF-α challenge. β-arrestin-1 led to the activation of the NF-κB signaling pathway by enhancing binding to IκBα and IKK under the TNF-α challenge. Taken together, our findings demonstrated macrophage β-arrestin-1 contributes to the deterioration of DSS-induced colitis through the interaction with NF-κB signaling, thus highlighting a novel target for the treatment of UC.
    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
    胰高血糖素样肽-1受体(GLP-1R)是2型糖尿病和肥胖症的主要药物靶标。配体引发的GLP-1R与G蛋白的相互作用已经得到了很好的研究,但不是β-抑制素1/2。因此,生物发光共振能量转移(BRET),使用诱变和操作模型来评估GLP-1R上85个细胞外表面残基在由三种代表性GLP-1R激动剂(GLP-1,exendin-4和胃泌酸调节素)触发的β-抑制素1/2募集中的作用。残基选择性调节不同配体的β-抑制蛋白1/2募集,并鉴定了β-抑制蛋白亚型。跨膜螺旋1(TM1)-胞外域(ECD)接头上残基K130-S136,L142和Y145的突变减少了β-抑制素1的募集,但增加了β-抑制素2的募集。其他胞外环(ECL)突变,包括P137A,Q211A,D222A和M303A选择性影响β-抑制素1募集,而D215A,L217A,Q221A,S223A,Y289A,S301A,F381A和I382A更多地参与配体的β-抑制蛋白2募集。与GLP-1和exendin-4相比,泌酸调节素更广泛地参与GLP-1R细胞外表面以驱动β-抑制素1/2募集;I147,W214和L218参与β-抑制素1募集,而L141、D215、L218、D293和F381在β-抑制素2募集胃泌酸调节素中尤为明显。此外,β-抑制蛋白1/2C结构域上的非保守残基有助于与GLP-1R的相互作用。在有或没有β-抑制素1/2过表达的配体刺激下,GLP-1R稳定表达的细胞系的进一步蛋白质组学分析证明了与同源配体和β-抑制素相关的常见和偏倚调节的蛋白质和途径。我们的研究提供了有关GLP-1R介导的配体诱导的β-抑制素募集以及随之而来的细胞内信号传导事件的有价值的信息。
    Glucagon-like peptide-1 receptor (GLP-1R) is a prime drug target for type 2 diabetes and obesity. The ligand initiated GLP-1R interaction with G protein has been well studied, but not with β-arrestin 1/2. Therefore, bioluminescence resonance energy transfer (BRET), mutagenesis and an operational model were used to evaluate the roles of 85 extracellular surface residues on GLP-1R in β-arrestin 1/2 recruitment triggered by three representative GLP-1R agonists (GLP-1, exendin-4 and oxyntomodulin). Residues selectively regulated β-arrestin 1/2 recruitment for diverse ligands, and β-arrestin isoforms were identified. Mutation of residues K130-S136, L142 and Y145 on the transmembrane helix 1 (TM1)-extracellular domain (ECD) linker decreased β-arrestin 1 recruitment but increased β-arrestin 2 recruitment. Other extracellular loop (ECL) mutations, including P137A, Q211A, D222A and M303A selectively affected β-arrestin 1 recruitment while D215A, L217A, Q221A, S223A, Y289A, S301A, F381A and I382A involved more in β-arrestin 2 recruitment for the ligands. Oxyntomodulin engaged more broadly with GLP-1R extracellular surface to drive β-arrestin 1/2 recruitment than GLP-1 and exendin-4; I147, W214 and L218 involved in β-arrestin 1 recruitment, while L141, D215, L218, D293 and F381 in β-arrestin 2 recruitment for oxyntomodulin particularly. Additionally, the non-conserved residues on β-arrestin 1/2 C-domains contributed to interaction with GLP-1R. Further proteomic profiling of GLP-1R stably expressed cell line upon ligand stimulation with or without β-arrestin 1/2 overexpression demonstrated both commonly and biasedly regulated proteins and pathways associated with cognate ligands and β-arrestins. Our study offers valuable information about ligand induced β-arrestin recruitment mediated by GLP-1R and consequent intracellular signaling events.
    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
    大麻素CB1受体是在中枢神经系统中广泛表达的G蛋白偶联受体(GPCR)。这种典型的G^i/o偶联受体介导Δ9-四氢大麻酚(THC)和合成大麻素受体激动剂(SCRA)的作用。娱乐使用SCRAs与严重的不良健康影响有关,优先考虑这些化合物的药理学研究。一些研究假设信号传导偏差可以解释SCRAs和THC之间的不同毒理学特征。先前的研究集中在通过环磷酸腺苷(cAMP)抑制和β-抑制蛋白易位测量的G蛋白活化之间的偏差。相比之下,本研究表征了G^i/o家族的G^亚型与β-抑制素之间的偏倚;该方法通过评估未经历主要扩增的信号,有助于更准确地评估配体偏倚.我们使用实时BRET报告基因表征了β-抑制蛋白1和2的G蛋白解离和易位。在所测试的G蛋白亚型中,由每个SCRA产生的应答与由参考配体AMB-FUBINACA产生的应答一致。通过应用操作分析来检测配体偏差,以确定Gi/o家族内的偏差,以及G蛋白亚型和β-抑制素之间。总的来说,这些结果证实SCRAs是平衡的,与低功效配体THC相比,高功效配体,只有一个SCRA,4CN-MPP-BUT7IACA,在一条途径比较中显示出统计学上的显着偏差(与G^oA/oB相比时,朝向β-抑制蛋白1)。这表明由SCRA引起的不利影响是由于在CB1处的高效力和功效,而不是偏向的激动作用。
    The cannabinoid CB1 receptor (CB1) is a G protein-coupled receptor (GPCR) with widespread expression in the central nervous system. This canonically G⍺i/o-coupled receptor mediates the effects of Δ9-tetrahydrocannabinol (THC) and synthetic cannabinoid receptor agonists (SCRAs). Recreational use of SCRAs is associated with serious adverse health effects, making pharmacological research into these compounds a priority. Several studies have hypothesised that signalling bias may explain the different toxicological profiles between SCRAs and THC. Previous studies have focused on bias between G protein activation measured by cyclic adenosine monophosphate (cAMP) inhibition and β-arrestin translocation. In contrast, the current study characterises bias between G⍺ subtypes of the G⍺i/o family and β-arrestins; this method facilitates a more accurate assessment of ligand bias by assessing signals that have not undergone major amplification. We have characterised G protein dissociation and translocation of β-arrestin 1 and 2 using real-time BRET reporters. The responses produced by each SCRA across the G protein subtypes tested were consistent with the responses produced by the reference ligand AMB-FUBINACA. Ligand bias was probed by applying the operational analysis to determine biases within the G⍺i/o family, and between G protein subtypes and β-arrestins. Overall, these results confirm SCRAs to be balanced, high-efficacy ligands compared to the low efficacy ligand THC, with only one SCRA, 4CN-MPP-BUT7IACA, demonstrating statistically significant bias in one pathway comparison (towards β-arrestin 1 when compared with G⍺oA/oB). This suggests that the adverse effects caused by SCRAs are due to high potency and efficacy at CB1, rather than biased agonism.
    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
    最近强调与稳态相关的关键皮肤细胞功能的昼夜节律,再生和衰老揭示了PER2生物钟基因作为重要的抗肿瘤基因的重要性。此外,δ阿片受体(DOPrs)已被确定为在皮肤分化中起关键作用,扩散和迁移,这不仅对伤口愈合至关重要,而且有助于癌症的发展。在这项研究中,我们提出了皮肤阿片受体(OPr)活性与昼夜节律之间的显着关联。为了调查这个链接,我们进行了48小时的昼夜节律实验,在这期间每5小时收集RNA样品。我们发现DOPr通过其内源性激动剂Met-脑啡肽在N/TERT-1角质形成细胞中的激活,同步地塞米松,导致核心时钟基因PER2的表达延迟有统计学意义的5.6小时。共聚焦显微镜进一步证实了DOPr-β-抑制蛋白-1复合物的同时核定位。此外,DOPr激活不仅增强了β-抑制蛋白-1与PER2启动子的节律结合,而且还诱导了相移。此外,我们观察到β-抑制蛋白-1调节其靶基因的转录,包括PER2,通过促进组蛋白4乙酰化。通过ChIP分析,我们确定Met-脑啡肽增强PER2启动子中β-抑制蛋白-1与乙酰化H4的结合。总之,我们的研究结果表明,DOPr激活通过β-抑制蛋白-1促进染色质重塑导致PER2表达发生相移.因此,这些结果表明,DOPr,就像它在伤口愈合中的作用一样,也可能通过影响PER2在癌症发展中发挥作用。
    The recent emphasis on circadian rhythmicity in critical skin cell functions related to homeostasis, regeneration and aging has shed light on the importance of the PER2 circadian clock gene as a vital antitumor gene. Furthermore, delta-opioid receptors (DOPrs) have been identified as playing a crucial role in skin differentiation, proliferation and migration, which are not only essential for wound healing but also contribute to cancer development. In this study, we propose a significant association between cutaneous opioid receptor (OPr) activity and circadian rhythmicity. To investigate this link, we conducted a 48 h circadian rhythm experiment, during which RNA samples were collected every 5 h. We discovered that the activation of DOPr by its endogenous agonist Met-Enkephalin in N/TERT-1 keratinocytes, synchronized by dexamethasone, resulted in a statistically significant 5.6 h delay in the expression of the core clock gene PER2. Confocal microscopy further confirmed the simultaneous nuclear localization of the DOPr-β-arrestin-1 complex. Additionally, DOPr activation not only enhanced but also induced a phase shift in the rhythmic binding of β-arrestin-1 to the PER2 promoter. Furthermore, we observed that β-arrestin-1 regulates the transcription of its target genes, including PER2, by facilitating histone-4 acetylation. Through the ChIP assay, we determined that Met-Enkephalin enhances β-arrestin-1 binding to acetylated H4 in the PER2 promoter. In summary, our findings suggest that DOPr activation leads to a phase shift in PER2 expression via β-arrestin-1-facilitated chromatin remodeling. Consequently, these results indicate that DOPr, much like its role in wound healing, may also play a part in cancer development by influencing PER2.
    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
    蛋白质通过蛋白质-蛋白质相互作用发挥其功能。在果蝇中,G蛋白偶联受体如视紫红质(Rh1)与G蛋白相互作用以激活视觉信号转导,并与抑制素相互作用以终止激活。此外,像Rh1这样的膜蛋白在内质网折叠过程中参与蛋白质-蛋白质相互作用,在它们的囊泡运输过程中以及从细胞表面去除和降解时。这里,我们在果蝇光感受器中表达了Rh1-TurboID融合蛋白(Rh1::TbID),以通过生物素邻近标记鉴定体内Rh1相互作用伴侣。我们证明Rh1::TbID形成功能性视紫红质,在视紫红质2与视紫红质相互作用的条件下介导视紫红质2的生物素化。我们还观察到Rh1::TbID和天然Rh1以及大多数视觉信号转导蛋白的生物素化。这些发现表明横纹肌中的信号成分接近视紫红质,在ca的范围内。10nm。此外,我们已经检测到参与视紫红质成熟的蛋白质和负责膜蛋白运输的元素,类似于RH1的潜在互动伙伴。其中包括内质网的伴侣,参与Clathrin介导的内吞作用的蛋白质以及以前未被注意到的视紫红质运输的贡献者,例如Rab32、Vap33或PIP82。
    Proteins exert their function through protein-protein interactions. In Drosophila, G protein-coupled receptors like rhodopsin (Rh1) interact with a G protein to activate visual signal transduction and with arrestins to terminate activation. Also, membrane proteins like Rh1 engage in protein-protein interactions during folding within the endoplasmic reticulum, during their vesicular transport and upon removal from the cell surface and degradation. Here, we expressed a Rh1-TurboID fusion protein (Rh1::TbID) in Drosophila photoreceptors to identify in vivo Rh1 interaction partners by biotin proximity labeling. We show that Rh1::TbID forms a functional rhodopsin that mediates biotinylation of arrestin 2 in conditions where arrestin 2 interacts with rhodopsin. We also observed biotinylation of Rh1::TbID and native Rh1 as well as of most visual signal transduction proteins. These findings indicate that the signaling components in the rhabdomere approach rhodopsin closely, within a range of ca. 10 nm. Furthermore, we have detected proteins engaged in the maturation of rhodopsin and elements responsible for the trafficking of membrane proteins, resembling potential interaction partners of Rh1. Among these are chaperons of the endoplasmic reticulum, proteins involved in Clathrin-mediated endocytosis as well as previously unnoticed contributors to rhodopsin transportation, such as Rab32, Vap33, or PIP82.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号