AREG

区域
  • 文章类型: Journal Article
    表皮生长因子受体(EGFR)是一种跨膜酪氨酸激酶,通常通过翻译后糖基化进行修饰。在癌症中,在很大一部分非小细胞肺癌和乳腺腺癌中检测到EGFR扩增和促进增殖的热点突变如L858R。分子动力学模拟表明,天冬酰胺残基361(N361)处的糖基化促进二聚化和配体结合。我们稳定表达糖基化缺陷突变EGFRN361A,有或没有致癌突变L858R。免疫荧光和流式细胞术证明突变体各自在细胞膜上良好表达。相对于野生型EGFR,N361A降低增殖以及对配体的敏感性降低。测量EGFR与其结合配偶体HER2在细胞中的共定位的邻近连接测定揭示N361A突变增加共定位。N361A,位于EGFR抑制剂necitumumab的结合界面附近,表达致癌EGFRL858R的脱敏细胞对基于抗体的抑制。这些发现强调了翻译后修饰对癌基因功能的关键相关性。
    结论:EGFR将生长因子的信号传导到细胞增殖中,并且在肿瘤中经常被过度激活。N361的EGFR糖基化调节EGFR二聚化,增殖信号的生长因子刺激,和对靶向抑制的敏感性。对EGFR糖基化的见解可能会扩大治疗机会,使癌症患者受益。
    Epidermal growth factor receptor (EGFR) is a transmembrane tyrosine kinase that is frequently modified by glycosylation post-translationally. In cancer, EGFR amplifications and hotspot mutations such as L858R that promote proliferation have been detected in a significant fraction of non-small cell lung carcinomas and breast adenocarcinomas. Molecular dynamic simulations suggested that glycosylation at asparagine residue 361 (N361) promotes dimerization and ligand binding. We stably expressed glycosylation-deficient mutant EGFR N361A, with or without the oncogenic mutation L858R. Immunofluorescence and flow cytometry demonstrated that the mutants were each well expressed at the cell membrane. N361A decreased proliferation relative to wild-type EGFR as well as decreased sensitivity to ligands. Proximity ligation assays measuring co-localization of EGFR with its binding partner HER2 in cells revealed that N361A mutations increased co-localization. N361A, located near the binding interface for the EGFR inhibitor necitumumab, desensitized cells expressing the oncogenic EGFR L858R to antibody-based inhibition. These findings underline the critical relevance of post-translational modifications on oncogene function.
    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
    牙龈成纤维细胞是维持牙周稳态和介导与牙周炎和口腔鳞状细胞癌相关的病理事件所需的旁分泌信号的重要来源。在潜在的旁分泌信号中,有stanniocalcin-1(STC1),参与氧化应激和细胞存活;双调蛋白(AREG),一种介导免疫细胞和上皮细胞之间交互的生长因子;11号染色体开放阅读框96(C11orf96),生物学功能不明确;以及炎症相关前列腺素E合酶(PTGES).牙龈成纤维细胞越来越多地表达这些基因,以响应含有受损细胞残余物的同种异体骨移植物。因此,基因表达可能是由损伤细胞产生的损伤相关分子模式的局部释放引起的。因此,这项研究的目的是使用已建立的基因组作为生物测定来测量口腔细胞裂解物的损伤相关活性。为了这个目标,我们已经将牙龈成纤维细胞暴露于由鳞癌细胞系TR146和HSC2,口腔上皮细胞制备的裂解物中,牙龈成纤维细胞.我们在这里报道,所有裂解物都显着增加了整个基因组的转录,在蛋白质水平上支持STC1。用SB431542阻断TGF-β受体1激酶仅部分降低STC1,AREG,C11orf96SB431542甚至增加了PTGES的表达。一起,这些发现表明,来自口腔细胞的损伤信号可以改变牙龈成纤维细胞的旁分泌活性。此外,基因表达组可以用作测试用于口服应用的材料的生物相容性的生物测定。
    Gingival fibroblasts are a significant source of paracrine signals required to maintain periodontal homeostasis and to mediate pathological events linked to periodontitis and oral squamous cell carcinomas. Among the potential paracrine signals are stanniocalcin-1 (STC1), involved in oxidative stress and cellular survival; amphiregulin (AREG), a growth factor that mediates the cross-talk between immune cells and epithelial cells; chromosome 11 open reading frame 96 (C11orf96) with an unclear biologic function; and the inflammation-associated prostaglandin E synthase (PTGES). Gingival fibroblasts increasingly express these genes in response to bone allografts containing remnants of injured cells. Thus, the gene expression might be caused by the local release of damage-associated molecular patterns arising from injured cells. The aim of this study is consequently to use the established gene panel as a bioassay to measure the damage-associated activity of oral cell lysates. To this aim, we have exposed gingival fibroblasts to lysates prepared from the squamous carcinoma cell lines TR146 and HSC2, oral epithelial cells, and gingival fibroblasts. We report here that all lysates significantly increased the transcription of the entire gene panel, supported for STC1 at the protein level. Blocking TGF-β receptor 1 kinase with SB431542 only partially reduced the forced expression of STC1, AREG, and C11orf96. SB431542 even increased the PTGES expression. Together, these findings suggest that the damage signals originating from oral cells can change the paracrine activity of gingival fibroblasts. Moreover, the expression panel of genes can serve as a bioassay for testing the biocompatibility of materials for oral application.
    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
    胎盘绒毛内的间充质细胞在形成分支结构的形态和驱动血管的发育中起着至关重要的作用。然而,胎盘绒毛周细胞(PVP)作为胎盘绒毛间充质细胞的不同亚群的标记和功能,仍然不清楚。因此,在这项研究中,研究了PVP的标记和功能。获得来自妊娠早期胎盘绒毛的单细胞测序数据,并使用Seurat工具鉴定PVP标记。使用DAVID数据库进行特定基因的基因本体论(GO)分析。Cellchat工具用于研究PVP和其他细胞之间的相互作用信号。使用免疫荧光确认PVP标志物的表达。通过透射电子显微镜检查了胎盘绒毛间质和PVP中细胞外囊泡的存在。我们的发现表明,胎盘绒毛中的肾素(REN)和双调蛋白(AREG)阳性成纤维细胞特异性表达了几种经典的周细胞标记。在头三个月,观察到周细胞的某些保守功能,它们表现出组织特异性功能,如整合素介导的信号通路和细胞外泌体.此外,发现胎盘绒毛间充质富含细胞外囊泡。AREG在妊娠早期的PVP中特别转录,然而,其蛋白位于合胞体滋养层中。这些见解有助于全面了解早期胎盘发育,并为胎盘源性妊娠并发症提供新的治疗靶点。
    Mesenchymal cells within theplacental villi play a crucial role in shaping the morphology of branching structures and driving the development of blood vessels. However, the markers and functions of placental villous pericytes (PVPs) as distinct subgroups of placental villous mesenchymal cells, remain unclear. Therefore, in this study, the markers and functions of PVPs were investigated. Single-cell sequencing data from the first-trimester placental villi was obtained and the Seurat tool was used to identify PVP markers. Gene Ontology (GO) analysis of specific genes was performed using the DAVID database. The Cellchat tool was employed to investigate the interaction signals between the PVPs and other cells. Expression of the PVP markers was confirmed using immunofluorescence. Presence of extracellular vesicles in the placental villous mesenchyme and PVPs was examined by transmission electron microscopy. Our findings revealed that renin (REN) and amphiregulin (AREG)-positive fibroblasts in the placental villi specifically expressed several classic pericyte markers. In the first trimester, certain conserved functions of pericytes were observed and they displayed tissue-specific functions such as in the integrin-mediated signaling pathway and extracellular exosomes. Moreover, the placental villous mesenchyme was found to be rich in extracellular vesicles. AREG is specifically transcribed in the first trimester PVPs, however, its protein was located in syncytiotrophoblasts. These insights contribute to a comprehensive understanding of early placental development and offer new therapeutic targets for placenta-derived pregnancy complications.
    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
    外泌体是在内体系统细胞内产生的分泌性囊泡。我们之前已经表明,外泌体不仅在晚期内体中产生,而且在以单体G蛋白Rab11a标记的循环内体中也是如此。这些囊泡,称为Rab11a-外泌体,在营养压力下优先分泌几种癌细胞类型,包括HCT116结直肠癌(CRC)细胞。HCT116Rab11a-外泌体具有特别有效的信号活性,一些由表皮生长因子受体(EGFR)配体介导,两栖蛋白(AREG)。KRAS的突变激活形式,EGFR的下游靶点,常见于高级CRC。缺席时,单克隆抗体,如西妥昔单抗,靶向EGFR并阻断EGFR配体的作用,如AREG,可以管理。患者,然而,不可避免地会对西妥昔单抗产生耐药性,通过获得KRAS突变或通过非遗传微环境变化。在这里,我们表明几种CRC细胞系中的营养应激导致携带AREG的Rab11a-外泌体的释放。我们证明,虽然可溶性AREG没有作用,与西妥昔单抗抗性KRAS突变HCT116细胞的Rab11a-外泌体结合的AREG水平低得多,可以抑制西妥昔单抗对KRAS野生型Caco-2CRC细胞的作用。使用中和抗AREG抗体和细胞内EGFR激酶抑制剂,我们表明这种作用是通过AREG激活EGFR介导的,而不是转移激活的KRAS。因此,AREG在Rab11a-外泌体上的呈递会影响其与西妥昔单抗竞争的能力。我们认为,这种Rab11a-外泌体介导的机制有助于在西妥昔单抗敏感的细胞中建立耐药性,并且可以解释为什么在西妥昔单抗耐药的肿瘤中只有一些细胞携带突变型KRAS。
    Exosomes are secreted vesicles made intracellularly in the endosomal system. We have previously shown that exosomes are not only made in late endosomes, but also in recycling endosomes marked by the monomeric G-protein Rab11a. These vesicles, termed Rab11a-exosomes, are preferentially secreted under nutrient stress from several cancer cell types, including HCT116 colorectal cancer (CRC) cells. HCT116 Rab11a-exosomes have particularly potent signalling activities, some mediated by the epidermal growth factor receptor (EGFR) ligand, amphiregulin (AREG). Mutant activating forms of KRAS, a downstream target of EGFR, are often found in advanced CRC. When absent, monoclonal antibodies, such as cetuximab, which target the EGFR and block the effects of EGFR ligands, such as AREG, can be administered. Patients, however, inevitably develop resistance to cetuximab, either by acquiring KRAS mutations or via non-genetic microenvironmental changes. Here we show that nutrient stress in several CRC cell lines causes the release of AREG-carrying Rab11a-exosomes. We demonstrate that while soluble AREG has no effect, much lower levels of AREG bound to Rab11a-exosomes from cetuximab-resistant KRAS-mutant HCT116 cells, can suppress the effects of cetuximab on KRAS-wild type Caco-2 CRC cells. Using neutralising anti-AREG antibodies and an intracellular EGFR kinase inhibitor, we show that this effect is mediated via AREG activation of EGFR, and not transfer of activated KRAS. Therefore, presentation of AREG on Rab11a-exosomes affects its ability to compete with cetuximab. We propose that this Rab11a-exosome-mediated mechanism contributes to the establishment of resistance in cetuximab-sensitive cells and may explain why in cetuximab-resistant tumours only some cells carry mutant KRAS.
    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
    肌肉浸润性膀胱癌(MIBC)仍然是一种流行的癌症,治疗选择有限,消除对创新疗法的需要。表皮生长因子受体(EGFR)是肿瘤进展的关键,是MIBC的潜在治疗靶点。此外,EGFR配体AREG和EREG已显示与结直肠癌患者抗EGFR治疗应答和无进展生存期改善相关.
    我们研究了EGFR的预后意义,区域,和MIBC中的EREG。通过qRT-PCR对100名在曼海姆大学医院接受根治性膀胱切除术的MIBC患者的组织样本进行基因表达和拷贝数分析(MA;中位年龄72,四分位距[IQR]64-78岁,25%女性)。结果在2017年TCGAMIBC队列的361例患者中得到验证(中位年龄69,IQR60-77岁,27%女性),在Chungbuk和MDACC队列中。使用Mann-Whitney检验,基因表达与临床病理参数相关,Kruskal-Wallis检验和Spearman相关性。对于总生存期(OS),使用Kaplan-Meier和Cox比例风险模型分析癌症特异性生存期(CSS)和无病生存期(DFS)基因表达.
    EGFR基因表达差异显著,区域,EREG可以在所有队列中检测到。在TCGA队列中,EGFR扩增和KRAS野生型患者的EGFR表达显著升高。在MA队列中,高AREG表达独立地预测了更长的OS(HR=0.35,CI0.19-0.63,p=0.0004)和CSS(HR=0.42,CI0.18-0.95,p=0.0378)。在TCGA队列中,高EGFR,区域,EREG表达与较短的OS(AREG:HR=1.57,CI1.12-2.20,p=0.0090)和DFS(EGFR:HR=1.91,CI1.31-2.8,p=0.0008)相关。EGFR扩增也与DFS降低相关。
    高EGFR和EREG表明MIBC患者的生存率较差。AREG的预后作用应进一步研究,准系列。四个队列之间不同的生存结果应谨慎解释,考虑到分析方法和人口统计学的差异。进一步的体外研究对于阐明本研究中观察到的关联的功能机制是必要的。
    UNASSIGNED: Muscle invasive bladder cancer (MIBC) remains a prevalent cancer with limited therapeutic options, obviating the need for innovative therapies. The epidermal growth factor receptor (EGFR) is a linchpin in tumor progression and presents a potential therapeutic target in MIBC. Additionally, the EGFR ligands AREG and EREG have shown associations with response to anti-EGFR therapy and improved progression-free survival in colorectal carcinoma.
    UNASSIGNED: We investigated the prognostic significance of EGFR, AREG, and EREG in MIBC. Gene expression and copy number analyses were performed via qRT-PCR on tissue samples from 100 patients with MIBC who underwent radical cystectomy at the University Hospital Mannheim (MA; median age 72, interquartile range [IQR] 64-78 years, 25% female). Results were validated in 361 patients from the 2017 TCGA MIBC cohort (median age 69, IQR 60-77 years, 27% female), in the Chungbuk and MDACC cohort. Gene expressions were correlated with clinicopathologic parameters using the Mann-Whitney test, Kruskal-Wallis- test and Spearman correlation. For overall survival (OS), cancer-specific survival (CSS) and disease-free survival (DFS) gene expression was analyzed with Kaplan-Meier and Cox-proportional hazard models.
    UNASSIGNED: Significant gene expression differences in EGFR, AREG, and EREG could be detected in all cohorts. In the TCGA cohort, EGFR expression was significantly elevated in patients with EGFR amplification and KRAS wildtype. High AREG expression independently predicted longer OS (HR = 0.35, CI 0.19 - 0.63, p = 0.0004) and CSS (HR = 0.42, CI 0.18 - 0.95, p = 0.0378) in the MA cohort. In the TCGA cohort, high EGFR, AREG, and EREG expression correlated with shorter OS (AREG: HR = 1.57, CI 1.12 - 2.20, p = 0.0090) and DFS (EGFR: HR = 1.91, CI 1.31 - 2.8, p = 0.0008). EGFR amplification was also associated with reduced DFS.
    UNASSIGNED: High EGFR and EREG indicate worse survival in patients with MIBC. The prognostic role of AREG should further be investigated in large, prospective series. Divergent survival outcomes between the four cohorts should be interpreted cautiously, considering differences in analysis methods and demographics. Further in vitro investigations are necessary to elucidate the functional mechanisms underlying the associations observed in this study.
    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
    CD133,肿瘤中的癌症干细胞(CSC)标志物,包括黑色素瘤,与肿瘤复发有关,化学抗性,和转移。用表达CD133的Tet-on载体转导患者来源的黑素瘤细胞系,产生多西环素(Dox)诱导型细胞系。将细胞暴露于Dox24小时以诱导CD133表达,其次是RNA-seq和生物信息学分析,揭示了CD133显著上调或下调的基因和途径。CD133后最显著上调的基因是双调蛋白(AREG),通过qRT-PCR和免疫印迹分析验证。诱导CD133表达显著增加细胞生长,S期细胞的百分比,BrdU掺入新生的DNA,和PCNA水平,表明CD133刺激细胞增殖。CD133诱导还激活EGFR和MAPK途径。使用AREG/EGFR抑制剂或AREGmRNA的siRNA敲低进一步描绘了突出CD133和AREG在黑素瘤CSC中的作用的潜在机制。EGFR抑制剂吉非替尼治疗阻断了CD133诱导的细胞生长增加和MAPK通路激活。重要的是,siRNA敲低AREG逆转CD133对细胞生长的刺激作用,表明AREG介导CD133对细胞增殖的影响,因此,在过表达CD133和AREG的黑色素瘤和癌症中,作为新型组合疗法的有吸引力的靶标。
    CD133, a cancer stem cell (CSC) marker in tumors, including melanoma, is associated with tumor recurrence, chemoresistance, and metastasis. Patient-derived melanoma cell lines were transduced with a Tet-on vector expressing CD133, generating doxycycline (Dox)-inducible cell lines. Cells were exposed to Dox for 24 h to induce CD133 expression, followed by RNA-seq and bioinformatic analyses, revealing genes and pathways that are significantly up- or downregulated by CD133. The most significantly upregulated gene after CD133 was amphiregulin (AREG), validated by qRT-PCR and immunoblot analyses. Induced CD133 expression significantly increased cell growth, percentage of cells in S-phase, BrdU incorporation into nascent DNA, and PCNA levels, indicating that CD133 stimulates cell proliferation. CD133 induction also activated EGFR and the MAPK pathway. Potential mechanisms highlighting the role(s) of CD133 and AREG in melanoma CSC were further delineated using AREG/EGFR inhibitors or siRNA knockdown of AREG mRNA. Treatment with the EGFR inhibitor gefitinib blocked CD133-induced cell growth increase and MAPK pathway activation. Importantly, siRNA knockdown of AREG reversed the stimulatory effects of CD133 on cell growth, indicating that AREG mediates the effects of CD133 on cell proliferation, thus serving as an attractive target for novel combinatorial therapeutics in melanoma and cancers with overexpression of both CD133 and AREG.
    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:胆道闭锁(BA)是一种以导管反应为主要致病特征的新生儿纤维炎性胆管病变,可预测患者的生存率。粘膜相关的不变T(MAIT)细胞在人类肝脏中富集,并在肝脏疾病中发挥多种作用。我们旨在研究MAIT细胞在BA中的功能。
    方法:首先,我们分析了肝脏MAIT细胞与临床参数(存活率,丙氨酸转氨酶,胆红素,组织学炎症和纤维化)在两个公共BA患者队列中(美国和中国)。生存数据和其他临床参数采用Kaplan-Meier生存分析和Spearman相关分析。分别。接下来,我们从BA和对照患者的肝脏样本或外周血中获取大量RNA测序,流式细胞术分析,MAIT细胞的免疫固定和功能实验。最后,我们建立了两个体外共培养系统,一种是恒河猴轮状病毒(RRV)感染的共培养系统,用于模拟人BA的免疫功能障碍,该系统通过单细胞RNA测序进行了验证,另一种是由胆道类器官组成的多细胞系统,LX-2和MAIT细胞评价MAIT细胞对导管反响的感化。
    结果:BA肝脏MAIT细胞与低存活率和导管反应呈正相关。此外,肝脏MAIT细胞被激活,以T细胞受体(TCR)依赖性方式表现出伤口愈合特征和高表达的生长因子双调蛋白(AREG)。AREG的拮抗作用消除了BAMAIT细胞对胆管细胞和胆道类器官的增殖作用。RRV感染的共培养系统,概述了人类BA的免疫功能障碍,揭示了RRV引发的MAIT细胞通过AREG促进胆管细胞增殖,并进一步诱导多细胞系统的炎症和纤维化。
    结论:MAIT细胞表现出依赖于TCR信号的伤口愈合特征,并通过AREG促进导管反应,这与晚期纤维化有关,并预示着BA的低生存率。
    背景:这项工作得到了国家自然科学基金资助(82001589和92168108),国家重点研发计划(2023YFA1801600)和广东省基础与应用基础研究基金(2020A1515110921)。
    BACKGROUND: Biliary atresia (BA) is a neonatal fibro-inflammatory cholangiopathy with ductular reaction as a key pathogenic feature predicting poor survival. Mucosal-associated invariant T (MAIT) cells are enriched in human liver and display multiple roles in liver diseases. We aimed to investigate the function of MAIT cells in BA.
    METHODS: First, we analyzed correlations between liver MAIT cell and clinical parameters (survival, alanine transaminase, bilirubin, histological inflammation and fibrosis) in two public cohorts of patients with BA (US and China). Kaplan-Meier survival analysis and spearman correlation analysis were employed for survival data and other clinical parameters, respectively. Next, we obtained liver samples or peripheral blood from BA and control patients for bulk RNA sequencing, flow cytometry analysis, immunostaning and functional experiments of MAIT cells. Finally, we established two in vitro co-culture systems, one is the rhesus rotavirus (RRV) infected co-culture system to model immune dysfunction of human BA which was validated by single cell RNA sequencing and the other is a multicellular system composed of biliary organoids, LX-2 and MAIT cells to evaluate the role of MAIT cells on ductular reaction.
    RESULTS: Liver MAIT cells in BA were positively associated with low survival and ductular reaction. Moreover, liver MAIT cells were activated, exhibited a wound healing signature and highly expressed growth factor Amphiregulin (AREG) in a T cell receptor (TCR)-dependent manner. Antagonism of AREG abrogated the proliferative effect of BA MAIT cells on both cholangiocytes and biliary organoids. A RRV infected co-culture system, recapitulated immune dysfunction of human BA, disclosed that RRV-primed MAIT cells promoted cholangiocyte proliferation via AREG, and further induced inflammation and fibrosis in the multicellular system.
    CONCLUSIONS: MAIT cells exhibit a wound healing signature depending on TCR signaling and promote ductular reaction via AREG, which is associated with advanced fibrosis and predictive of low survival in BA.
    BACKGROUND: This work was funded by National Natural Science Foundation of China grant (82001589 and 92168108), National Key R&D Program of China (2023YFA1801600) and by Basic and Applied Basic Research Foundation of Guangdong (2020A1515110921).
    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
    双调蛋白(AREG)作为表皮生长因子受体(EGFR)的配体,参与重要的生物学功能,包括炎症反应,组织再生,和免疫系统功能。在与EGFR相互作用时,AREG启动了一系列的信号级联所必需的几个生理活动,比如新陈代谢,细胞周期调节,和细胞增殖。最近的发现为AREG在维持受损组织的稳态平衡和在病毒感染影响肺部的情况下保持上皮细胞结构方面的重要作用提供了证据。对流感病毒感染的抗性的发展取决于1型细胞因子应答的存在。在消灭病原体之后,肺随后被与2型免疫反应相关的几种细胞类型定植。这些细胞有助于修复和解决由感染引起的组织损伤和炎症的过程。流感感染后,AREG的激活促进支气管上皮细胞的再生,增强组织的结构完整性,提高感染小鼠的存活率。以同样的方式,当细菌和病毒感染在同一宿主内同时出现时,患有流感的小鼠由于肺部区域的后续细菌感染而经历快速死亡。已经证明AREG参与细菌感染。AREG基因在宿主细胞内经历增加的转录活性,以响应由病原体如大肠杆菌和淋病奈瑟菌引起的细菌感染。此外,AREG已被广泛研究为上皮细胞层中的促有丝分裂刺激。因此,它被认为是一个潜在的竞争者,可能有助于在蠕虫感染中观察到的上皮细胞反应。与这一发现一致,缺乏AREG基因的小鼠表现出肠道寄生虫Trichurismuris的根除延迟。与对照组中的感染动物相比,观察到的延迟与结肠上皮细胞增殖速率的降低有关。上述发现表明AREG在促进肠组织上皮细胞内防御机制的激活中起关键作用。在这种特定情况下,尚未确定AREG的精确细胞来源。然而,很明显,感染小鼠上皮细胞层的增殖增加依赖于CD4T细胞。这一发现的意义在于它证明了免疫细胞和上皮细胞之间的相互作用在调节AREG-EGFR途径中发挥的关键作用。需要进行更多的研究来深入研究控制AREG合成及其组织保护特性的细胞起源和信号机制,独立于感染。
    Amphiregulin (AREG) serves as a ligand for the epidermal growth factor receptor (EGFR) and is involved in vital biological functions, including inflammatory responses, tissue regeneration, and immune system function. Upon interaction with the EGFR, AREG initiates a series of signaling cascades necessary for several physiological activities, such as metabolism, cell cycle regulation, and cellular proliferation. Recent findings have provided evidence for the substantial role of AREG in maintaining the equilibrium of homeostasis in damaged tissues and preserving epithelial cell structure in the context of viral infections affecting the lungs. The development of resistance to influenza virus infection depends on the presence of type 1 cytokine responses. Following the eradication of the pathogen, the lungs are subsequently colonized by several cell types that are linked with type 2 immune responses. These cells contribute to the process of repairing and resolving the tissue injury and inflammation caused by infections. Following influenza infection, the activation of AREG promotes the regeneration of bronchial epithelial cells, enhancing the tissue\'s structural integrity and increasing the survival rate of infected mice. In the same manner, mice afflicted with influenza experience rapid mortality due to a subsequent bacterial infection in the pulmonary region when both bacterial and viral infections manifest concurrently inside the same host. The involvement of AREG in bacterial infections has been demonstrated. The gene AREG experiences increased transcriptional activity inside host cells in response to bacterial infections caused by pathogens such as Escherichia coli and Neisseria gonorrhea. In addition, AREG has been extensively studied as a mitogenic stimulus in epithelial cell layers. Consequently, it is regarded as a prospective contender that might potentially contribute to the observed epithelial cell reactions in helminth infection. Consistent with this finding, mice that lack the AREG gene exhibit a delay in the eradication of the intestinal parasite Trichuris muris. The observed delay is associated with a reduction in the proliferation rate of colonic epithelial cells compared to the infected animals in the control group. The aforementioned findings indicate that AREG plays a pivotal role in facilitating the activation of defensive mechanisms inside the epithelial cells of the intestinal tissue. The precise cellular sources of AREG in this specific context have not yet been determined. However, it is evident that the increased proliferation of the epithelial cell layer in infected mice is reliant on CD4+ T cells. The significance of this finding lies in its demonstration of the crucial role played by the interaction between immunological and epithelial cells in regulating the AREG-EGFR pathway. Additional research is necessary to delve into the cellular origins and signaling mechanisms that govern the synthesis of AREG and its tissue-protective properties, independent of infection.
    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
    同种异体骨广泛用作骨传导支持以引导骨再生。骨同种异体移植物不仅仅是迁移细胞的支架,因为它们保持了原始骨基质的一些生物活性。然而,目前尚不清楚迁移细胞对同种异体骨移植物的反应。为此,我们已经评估了间充质细胞暴露于同种异体骨(ALBA)的酸裂解物的反应。RNAseq显示ALBA对牙龈成纤维细胞的遗传特征有很强的影响,IL11、AREG、C11orf96,STC1和GK-通过RT-PCR确认,以及通过免疫测定的IL11和STC1。考虑到转化生长因子-β(TGF-β)储存在骨基质中并可能引起表达变化,我们进行了蛋白质组学分析,TGF-β免疫测定,和smad2/3核易位。ALBA既未显示可检测的TGF-β,也未显示能够诱导smad2/3易位的裂解物。然而,TGF-β受体I型激酶抑制剂SB431542显著降低IL11、AREG、和C11orf96,表明除TGF-β以外的其他激动剂负责强大的细胞反应。研究结果表明,IL11、AREG、和C11orf96在间充质细胞中的表达可以作为反映同种异体骨的生物活性的生物测定。
    Bone allografts are widely used as osteoconductive support to guide bone regrowth. Bone allografts are more than a scaffold for the immigrating cells as they maintain some bioactivity of the original bone matrix. Yet, it remains unclear how immigrating cells respond to bone allografts. To this end, we have evaluated the response of mesenchymal cells exposed to acid lysates of bone allografts (ALBA). RNAseq revealed that ALBA has a strong impact on the genetic signature of gingival fibroblasts, indicated by the increased expression of IL11, AREG, C11orf96, STC1, and GK-as confirmed by RT-PCR, and for IL11 and STC1 by immunoassays. Considering that transforming growth factor-β (TGF-β) is stored in the bone matrix and may have caused the expression changes, we performed a proteomics analysis, TGF-β immunoassay, and smad2/3 nuclear translocation. ALBA neither showed detectable TGF-β nor was the lysate able to induce smad2/3 translocation. Nevertheless, the TGF-β receptor type I kinase inhibitor SB431542 significantly decreased the expression of IL11, AREG, and C11orf96, suggesting that other agonists than TGF-β are responsible for the robust cell response. The findings suggest that IL11, AREG, and C11orf96 expression in mesenchymal cells can serve as a bioassay reflecting the bioactivity of the bone allografts.
    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:随着产妇年龄的增长,卵母细胞减数分裂期间的异常明显增加。非整倍体是衰老卵母细胞质量降低的重要缘由。然而,老年卵母细胞非整倍性的分子机制尚不清楚。据报道,组蛋白乙酰转移酶1(HAT1)对哺乳动物发育和基因组稳定性至关重要。并参与多器官衰老。HAT1是否参与卵巢衰老及其详细机制尚待阐明。
    方法:采用免疫组织化学和免疫印迹法检测老年小鼠卵巢中HAT1的水平。探讨HAT1在小鼠卵母细胞成熟过程中的作用,我们使用Anacardic酸(AA)和小干扰RNA(siRNA)体外培养ICR雌性小鼠的卵丘-卵母细胞复合物(COCs),并收集了生发囊泡分解(GVBD)的统计数据,第一极体挤压(PBE),减数分裂缺陷,非整倍体,2细胞胚胎形成,和囊胚形成率。此外,人颗粒细胞(GC)样细胞系KGN细胞用于研究HAT1在这一过程中的机制。
    结果:HAT1在年轻小鼠的卵巢颗粒细胞(GCs)中高表达,在老年GCs中HAT1的表达显着降低。AA和siRNA介导的GCs中HAT1的抑制降低了PBE率,卵母细胞减数分裂缺陷和非整倍体增加。进一步研究表明HAT1可以乙酰化Forkhead盒转录因子O1(FoxO1),导致FoxO1易位进入细胞核。结果,乙酰化FoxO1的易位增加了GCs中双调蛋白(AREG)的表达,在卵母细胞减数分裂中起着重要作用。
    结论:本研究提示GCs中HAT1表达降低是与卵母细胞年龄相关的减数分裂缺陷相应的潜在原因,并为临床干预减少非整倍体卵母细胞提供了潜在的治疗靶点。
    BACKGROUND: With advanced maternal age, abnormalities during oocyte meiosis increase significantly. Aneuploidy is an important reason for the reduction in the quality of aged oocytes. However, the molecular mechanism of aneuploidy in aged oocytes is far from understood. Histone acetyltransferase 1 (HAT1) has been reported to be essential for mammalian development and genome stability, and involved in multiple organ aging. Whether HAT1 is involved in ovarian aging and the detailed mechanisms remain to be elucidated.
    METHODS: The level of HAT1 in aged mice ovaries was detected by immunohistochemical and immunoblotting. To explore the function of HAT1 in the process of mouse oocyte maturation, we used Anacardic Acid (AA) and small interfering RNAs (siRNA) to culture cumulus-oocyte complexes (COCs) from ICR female mice in vitro and gathered statistics of germinal vesicle breakdown (GVBD), the first polar body extrusion (PBE), meiotic defects, aneuploidy, 2-cell embryos formation, and blastocyst formation rate. Moreover, the human granulosa cell (GC)-like line KGN cells were used to investigate the mechanisms of HAT1 in this progress.
    RESULTS: HAT1 was highly expressed in ovarian granulosa cells (GCs) from young mice and the expression of HAT1 was significantly decreased in aged GCs. AA and siRNAs mediated inhibition of HAT1 in GCs decreased the PBE rate, and increased meiotic defects and aneuploidy in oocytes. Further studies showed that HAT1 could acetylate Forkhead box transcription factor O1 (FoxO1), leading to the translocation of FoxO1 into the nucleus. Resultantly, the translocation of acetylated FoxO1 increased the expression of amphiregulin (AREG) in GCs, which plays a significant role in oocyte meiosis.
    CONCLUSIONS: The present study suggests that decreased expression of HAT1 in GCs is a potential reason corresponding to oocyte age-related meiotic defects and provides a potential therapeutic target for clinical intervention to reduce aneuploid oocytes.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号