ADAM10 Protein

ADAM10 蛋白
  • 文章类型: Journal Article
    去整合素和金属蛋白酶结构域10(ADAM10),ADAM家族的一员,是具有潜在粘附和蛋白酶/转化酶功能的细胞表面蛋白。天然产物[腺苷(AD)及其类似物在癌症中的表达调控,虫草素(CD),和N6,N6-二甲基腺苷(m62A)],和免疫调节还不清楚。作为结果,AD,CD,m62A抑制ADAM10在各种癌细胞系中的表达,表明它们在抗癌药物中的作用。与ADAM10蛋白进一步的分子对接发现,所有小分子的所有对接基团的结合能均<-7kcal/mol(AD,CD和m62A),提出了非常好的绑定活动。此外,对肿瘤免疫调节作用的分析表明,ADAM10与免疫调节基因如CCL27、CCL14、CCL25、CXCR5、HLA-B、HLA-DOB1,LAG3,TNFRSF18和TNFRSF4在膀胱尿路上皮癌中,胸腺瘤,乳腺浸润性癌,TGCT,肾乳头状细胞癌,SKCM与甲状腺癌,表明ADAM10的免疫促进作用。体内AD和CD均降低了LAG3mRNA水平。ADAM10还与肿瘤免疫抑制呈负相关,并与肿瘤的免疫浸润相关。总的来说,本研究确定了AD的ADAM10表达,CD和m62A,在癌症中的AD或CD/ADAM10/LAG3信号传导中,并提出了一种使用小分子AD靶向ADAM10的癌症免疫治疗的潜在方法,CD和m62A。
    A disintegrin and metalloproteinase domain 10 (ADAM10), a member of the ADAM family, is a cellular surface protein with potential adhesion and protease/convertase functions. The expression regulations in cancers by natural products [adenosine (AD) and its analogs, cordycepin (CD), and N6, N6-dimethyladenosine (m6 2A)], and immune regulation are unclear. As results, AD, CD, and m6 2A inhibited ADAM10 expression in various cancer cell lines, indicating their roles in anti-cancer agents. Further molecular docking with ADAM10 protein found the binding energies of all docking groups were <-7 kcal/mol for all small-molecules (AD, CD and m6 2A), suggesting very good binding activities. In addition, analysis of the immunomodulatory roles in cancer showed that ADAM10 was negatively correlated with immunomodulatory genes such as CCL27, CCL14, CCL25, CXCR5, HLA-B, HLA-DOB1, LAG3, TNFRSF18, and TNFRSF4 in bladder urothelial carcinoma, thymoma, breast invasive carcinoma, TGCT, kidney renal papillary cell carcinoma, SKCM and thyroid carcinoma, indicating the immune-promoting roles for ADAM10. LAG3 mRNA levels were reduced by both AD and CD in vivo. ADAM10 is also negatively associated with tumor immunosuppression and interrelated with the immune infiltration of tumors. Overall, the present study determined ADAM10 expression by AD, CD and m6 2A, and in AD or CD/ADAM10/LAG3 signaling in cancers, and suggested a potential method for immunotherapy of cancers by targeting ADAM10 using the small molecules AD, CD and m6 2A.
    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
    肿瘤免疫治疗对PD-1阻断的耐药性极大地限制了其临床应用。T细胞免疫球蛋白和含粘蛋白结构域-3(Tim-3),一个有前途的免疫检查点目标,被ADAM10/17切割以在人类中产生其可溶形式(sTim-3),可能参与抗PD-1耐药。在这里,在非小细胞肺癌(NSCLC)和各种消化道肿瘤中观察到血清sTim-3上调。值得注意的是,在接受抗PD-1治疗的NSCLC患者和抗PD-1耐药的胆管癌患者中,血清sTim-3进一步上调。此外,sTim-3过表达促进肿瘤进展并在多种肿瘤小鼠模型中赋予抗PD-1抗性。机械上,sTim-3通过癌胚抗原相关细胞粘附分子1(CEACAM-1)诱导末端T细胞衰竭并减弱CD8T细胞对PD-1阻断的反应。此外,ADAM10抑制剂GI254023X,这阻碍了sTim-3的生产,减少Tim-3人源化小鼠的肿瘤进展并逆转人肿瘤浸润淋巴细胞(TIL)的抗PD-1抗性。总的来说,人类sTim-3在肿瘤免疫治疗中具有巨大的预测和治疗潜力。
    Resistance to PD-1 blockade in onco-immunotherapy greatly limits its clinical application. T cell immunoglobulin and mucin domain containing-3 (Tim-3), a promising immune checkpoint target, is cleaved by ADAM10/17 to produce its soluble form (sTim-3) in humans, potentially becoming involved in anti-PD-1 resistance. Herein, serum sTim-3 upregulation was observed in non-small cell lung cancer (NSCLC) and various digestive tumors. Notably, serum sTim-3 is further upregulated in non-responding patients undergoing anti-PD-1 therapy for NSCLC and anti-PD-1-resistant cholangiocarcinoma patients. Furthermore, sTim-3 overexpression facilitates tumor progression and confers anti-PD-1 resistance in multiple tumor mouse models. Mechanistically, sTim-3 induces terminal T cell exhaustion and attenuates CD8+ T cell response to PD-1 blockade through carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM-1). Moreover, the ADAM10 inhibitor GI254023X, which blocks sTim-3 production, reduces tumor progression in Tim-3 humanized mice and reverses anti-PD-1 resistance in human tumor-infiltrating lymphocytes (TILs). Overall, human sTim-3 holds great predictive and therapeutic potential in onco-immunotherapy.
    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
    慢性炎症是导致阿尔茨海默病(AD)进展的重要致病因素,和专门的促分解脂质介质(SPM)在AD发病过程中调节炎症反应中起关键作用。Maresin1(MaR1)是最新发现的SPM,并且发现MaR1通过调节神经营养通路来保护AD突触并减少Aβ的产生,从而改善AD的认知障碍,这使得MaR1成为AD治疗的候选药物。不幸的是,潜在的机制仍然是众所周知的。在这项研究中,对AD小鼠和细胞模型进行MaR1治疗,我们发现MaR1减少Aβ的产生以改善AD相关症状,并增加ADAM10/17,sAPPα和sAPPβ的表达水平以发挥其抗炎作用。此外,正如通过西方印迹分析确定的那样,我们观察到MaR1可以影响神经保护信号通路。具体来说,MaR1下调p57NTR,上调TrkA激活p75NTR/TrkA信号通路,它可以增加p-PI3K和p-Akt的表达水平,并下调p-mTOR激活PI3K/AKT/ERK/mTOR通路。最后,我们验证了ADAM10/17在调节AD进展中的作用,我们发现ADAM10/17的沉默使上述神经保护信号通路失活,从而加重AD的发病机制。总之,通过消除Aβ的产生,MaR1被证实是AD的潜在治疗剂,上调ADAM10/17、sAPPα和sAPPβ,并激活神经保护性p75NTR/TrkA通路和PI3K/AKT/ERK/mTOR通路。
    Chronic inflammation is an important pathogenetic factor that leads to the progression of Alzheimer\'s disease (AD), and specialized pro-resolving lipid mediators (SPMs) play critical role in regulating inflammatory responses during AD pathogenesis. Maresin1 (MaR1) is the latest discovered SPMs, and it is found that MaR1 improves AD cognitive impairment by regulating neurotrophic pathways to protect AD synapses and reduce Aβ production, which made MaR1 as candidate agent for AD treatment. Unfortunately, the underlying mechanisms are still largely known. In this study, the AD mice and cellular models were subjected to MaR1 treatment, and we found that MaR1 reduced Aβ production to ameliorate AD-related symptoms and increased the expression levels of ADAM10/17, sAPPα and sAPPβ to exert its anti-inflammatory role. In addition, as it was determined by Western Blot analysis, we observed that MaR1 could affected the neuroprotective signal pathways. Specifically, MaR1 downregulated p57NTR and upregulated TrkA to activate the p75NTR/TrkA signal pathway, and it could increase the expression levels of p-PI3K and p-Akt, and downregulated p-mTOR to activate the PI3K/AKT/ERK/mTOR pathway. Finally, we verified the role of ADAM10/17 in regulating AD progression, and we found that silencing of ADAM10/17 inactivated the above neuroprotective signal pathways to aggravate AD pathogenesis. In conclusion, MaR1 is verified as potential therapeutic agent for AD by eliminating Aβ production, upregulating ADAM10/17, sAPPα and sAPPβ, and activating the neuroprotective p75NTR/TrkA pathway and the PI3K/AKT/ERK/mTOR 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
    背景:α-硫辛酸(ALA)对神经退行性疾病具有神经保护作用。在诊所里,ALA可以改善阿尔茨海默病(AD)和其他痴呆患者的认知功能损害。动物研究证实了ALA的抗淀粉样变性作用,但其潜在机制仍不清楚。特别是,ALA在淀粉样β前体蛋白(APP)代谢中的作用尚未完全阐明。
    目的:研究ALA在AD转基因小鼠模型中是否能降低APP的淀粉样生成作用,并研究这种效应的潜在机制。
    方法:对2月龄的APP23/PS45转基因小鼠连续4个月输入ALA,然后评估其认知功能和AD样病理。将ALA药物浓度梯度应用于体外20E2细胞,以评估其对APP蛋白水解酶和代谢产物表达的影响。使用GI254023X研究了ALA影响APP加工的机制,解整合素和金属蛋白酶10(ADAM10)的抑制剂,以及线粒体毒性药物羰基氰化物间氯苯腙(CCCP)。
    结果:施用ALA改善了APP23/PS45小鼠脑组织中的淀粉样蛋白斑神经病理学,并减少了学习和记忆障碍。ALA还增加了ADAM10在20E2细胞中的表达和APP的非淀粉样蛋白生成加工以产生83个氨基酸的C末端片段(C83)。除了激活自噬,ALA还显著促进线粒体自噬。BNIP3L敲低降低了ADAM10的mat/pro比。通过使用CCCP,发现ALA调节BNIP3L介导的线粒体自噬,从而促进APP的α-裂解。
    结论:ADAM10增强APP的α-分泌酶裂解是ALA改善APP23/PS45转基因小鼠认知缺陷的主要机制。BNIP3L介导的线粒体自噬通过促进ADAM10的成熟而有助于ALA的抗淀粉样蛋白特性。本研究为ALA治疗AD提供了新的实验证据。
    BACKGROUND: Alpha-lipoic acid (ALA) has a neuroprotective effect on neurodegenerative diseases. In the clinic, ALA can improve cognitive impairments in patients with Alzheimer\'s disease (AD) and other dementias. Animal studies have confirmed the anti-amyloidosis effect of ALA, but its underlying mechanism remains unclear. In particular, the role of ALA in amyloid-β precursor protein (APP) metabolism has not been fully elucidated.
    OBJECTIVE: To investigate whether ALA can reduce the amyloidogenic effect of APP in a transgenic mouse model of AD, and to study the mechanism underlying this effect.
    METHODS: ALA was infused into 2-month-old APP23/PS45 transgenic mice for 4 consecutive months and their cognitive function and AD-like pathology were then evaluated. An ALA drug concentration gradient was applied to 20E2 cells in vitro to evaluate its effect on the expression of APP proteolytic enzymes and metabolites. The mechanism by which ALA affects APP processing was studied using GI254023X, an inhibitor of A Disintegrin and Metalloproteinase 10 (ADAM10), as well as the mitochondrial toxic drug carbonyl cyanide m-chlorophenylhydrazone (CCCP).
    RESULTS: Administration of ALA ameliorated amyloid plaque neuropathology in the brain tissue of APP23/PS45 mice and reduced learning and memory impairment. ALA also increased the expression of ADAM10 in 20E2 cells and the non-amyloidogenic processing of APP to produce the 83 amino acid C-terminal fragment (C83). In addition to activating autophagy, ALA also significantly promoted mitophagy. BNIP3L-knockdown reduced the mat/pro ratio of ADAM10. By using CCCP, ALA was found to regulate BNIP3L-mediated mitophagy, thereby promoting the α-cleavage of APP.
    CONCLUSIONS: The enhanced α-secretase cleavage of APP by ADAM10 is the primary mechanism through which ALA ameliorates the cognitive deficits in APP23/PS45 transgenic mice. BNIP3L-mediated mitophagy contributes to the anti-amyloid properties of ALA by facilitating the maturation of ADAM10. This study provides novel experimental evidence for the treatment of AD with ALA.
    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:淀粉样蛋白β的沉积,它是由β-和γ-分泌酶对APP的淀粉样裂解产生的,是AD病理学的主要标志之一。APP也可以通过α-和γ-分泌酶依次处理,为了产生sAPPα,通过促进神经突生长和神经元存活,等。方法:使用miRNA-seq分析来自11名AD患者以及14名年龄和性别匹配的认知正常志愿者的血浆样品中miRNA的整体表达谱。然后,在体内和体外都过表达miR-140和miR-122,并在体外敲低miR-140和miR-122的内源性表达。使用了多种技术的组合,包括分子生物学,免疫组织化学,检测miRNA对AD病理的影响。
    结果:在这项研究中,我们确定了两个miRNAs,miR-140-3p和miR-122-5p,都靶向ADAM10,中枢神经系统的主要α-分泌酶,在AD患者的血浆中上调。这两种miRNA在小鼠脑中的过表达在野生型C57BL/6J小鼠中诱导认知下降以及在APP/PS1小鼠中加剧认知功能障碍。尽管未检测到APP和总Aβ的显着变化,显著下调ADAM10及其非淀粉样蛋白产物,sAPPα,在过表达miR-140/miR-122的小鼠脑中观察到。免疫组织学分析显示神经突营养不良增加,这与这些小鼠海马中的小胶质细胞趋化性减少有关,独立于参与调节小胶质细胞免疫活性的其他两种ADAM10底物(神经元CX3CL1和小胶质细胞TREM2)。进一步的体外分析表明,过表达miR-140/miR-122的小鼠胚胎神经元细胞的神经炎性生长减少和与过表达miR-140/miR-122的HT22细胞共培养的小胶质细胞中的Aβ吞噬作用减少都可以通过过表达miR-140/miR-122TuD的特异性抑制序列以及通过添加sAPα来挽救。使这些miRNA成为潜在的治疗靶标。
    结论:我们的结果表明,神经保护性sAPPα是miR-140和miR-122表达失调诱导的神经病理进展的关键角色。靶向这些miRNA可能作为AD治疗中的有希望的治疗策略。
    BACKGROUND: Deposition of amyloid β, which is produced by amyloidogenic cleavage of APP by β- and γ-secretase, is one of the primary hallmarks of AD pathology. APP can also be processed by α- and γ-secretase sequentially, to generate sAPPα, which has been shown to be neuroprotective by promoting neurite outgrowth and neuronal survival, etc. METHODS: The global expression profiles of miRNA in blood plasma samples taken from 11 AD patients as well as from 14 age and sex matched cognitively normal volunteers were analyzed using miRNA-seq. Then, overexpressed miR-140 and miR-122 both in vivo and in vitro, and knock-down of the endogenous expression of miR-140 and miR-122 in vitro. Used a combination of techniques, including molecular biology, immunohistochemistry, to detect the impact of miRNAs on AD pathology.
    RESULTS: In this study, we identified that two miRNAs, miR-140-3p and miR-122-5p, both targeting ADAM10, the main α-secretase in CNS, were upregulated in the blood plasma of AD patients. Overexpression of these two miRNAs in mouse brains induced cognitive decline in wild type C57BL/6J mice as well as exacerbated dyscognition in APP/PS1 mice. Although significant changes in APP and total Aβ were not detected, significantly downregulated ADAM10 and its non-amyloidogenic product, sAPPα, were observed in the mouse brains overexpressing miR-140/miR-122. Immunohistology analysis revealed increased neurite dystrophy that correlated with the reduced microglial chemotaxis in the hippocampi of these mice, independent of the other two ADAM10 substrates (neuronal CX3CL1 and microglial TREM2) that were involved in regulating the microglial immunoactivity. Further in vitro analysis demonstrated that both the reduced neuritic outgrowth of mouse embryonic neuronal cells overexpressing miR-140/miR-122 and the reduced Aβ phagocytosis in microglia cells co-cultured with HT22 cells overexpressing miR-140/miR-122 could be rescued by overexpressing the specific inhibitory sequence of miR-140/miR-122 TuD as well as by addition of sAPPα, rendering these miRNAs as potential therapeutic targets.
    CONCLUSIONS: Our results suggested that neuroprotective sAPPα was a key player in the neuropathological progression induced by dysregulated expression of miR-140 and miR-122. Targeting these miRNAs might serve as a promising therapeutic strategy in AD treatment.
    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
    皮肤黑素瘤(SKCM)是死亡率最高的皮肤恶性肿瘤,其发病率在全球范围内呈上升趋势。吸烟是黑色素瘤预后不良的独立标志。α5-烟碱乙酰胆碱受体(α5-nAChR),尼古丁的受体之一,参与了扩散,SKCM细胞的迁移和侵袭。据报道,尼古丁可促进整合素和金属蛋白酶10(ADAM10)的表达,这是黑素瘤进展的关键基因。这里,我们探讨了尼古丁相关皮肤黑色素瘤中α5-nAChR和ADAM10之间的联系。SKCM中α5-nAChR的表达与ADAM10的表达和较低的生存率相关。α5-nAChR通过STAT3介导尼古丁诱导的ADAM10表达。α5-nAChR/ADAM10信号轴参与SKCM细胞的干性和迁移。此外,α5-nAChR表达与ADAM10表达相关,尼古丁相关小鼠同源移植组织中的EMT标记表达和干性标记表达。这些结果表明α5-nAChR/ADAM10信号通路在尼古丁诱导的黑色素瘤进展中的作用。
    Skin cutaneous melanoma (SKCM) is the skin malignancy with the highest mortality rate, and its morbidity rate is on the rise worldwide. Smoking is an independent marker of poor prognosis in melanoma. The α5-nicotinic acetylcholine receptor (α5-nAChR), one of the receptors for nicotine, is involved in the proliferation, migration and invasion of SKCM cells. Nicotine has been reported to promote the expression of a disintegrin and metalloproteinase 10 (ADAM10), which is the key gene involved in melanoma progression. Here, we explored the link between α5-nAChR and ADAM10 in nicotine-associated cutaneous melanoma. α5-nAChR expression was correlated with ADAM10 expression and lower survival in SKCM. α5-nAChR mediated nicotine-induced ADAM10 expression via STAT3. The α5-nAChR/ADAM10 signaling axis was involved in the stemness and migration of SKCM cells. Furthermore, α5-nAChR expression was associated with ADAM10 expression, EMT marker expression and stemness marker expression in nicotine-related mice homograft tissues. These results suggest the role of the α5-nAChR/ADAM10 signaling pathway in nicotine-induced melanoma progression.
    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
    在诊断为糖尿病(DM)的患者中,血管病变经常作为并发症出现。目前,经皮冠状动脉介入治疗(PCI)和抗血栓治疗是主要治疗方法.然而,PCI术后支架内再狭窄仍然是一个具有挑战性的临床问题,缺乏持续有效的治疗.linarin(LN)具有多种药理活性,被认为是治疗各种疾病的潜在药物。包括DM。但其在DM患者血管损伤后再狭窄中的具体作用尚不清楚。建立了糖尿病相关再狭窄大鼠模型,以评估LN在新生内膜增生中的作用。高糖刺激的血管平滑肌细胞(VSMC)(HG,30mM)进行LN处理。此外,构建解整合素和金属蛋白酶(ADAM10)的过表达质粒以转染VSMC。我们雇佣了CCK-8Brdu,伤口愈合划痕,和transwell迁移试验来评估VSMC的增殖和迁移。此外,采用westernblot和免疫荧光法检测ADAM10和下游Notch信号通路在体内和体外模型中的表达。LN可显着减轻DM大鼠血管损伤后的内膜增生,并降低ADAM10的蛋白表达,以及其下游Notch1信号通路相关蛋白(Notch1,NICD和Hes1)在大鼠颈动脉组织中的表达。LN能有效抑制HG诱导的VSMCs的增殖和迁移,下调ADAM10、Notch1、NICD和Hes1的蛋白表达。此外,我们的研究结果表明,ADAM10过表达显著逆转LN对增殖的影响,迁移,和Notch1信号通路相关蛋白在HG处理的VSMCs中的表达。LN在解决糖尿病相关血管损伤后的再狭窄方面具有潜在的治疗功效,ADAM10介导的Notch信号通路起着举足轻重的作用。
    Vascular lesions frequently arise as complication in patients diagnosed with diabetes mellitus (DM). Presently, percutaneous coronary intervention (PCI) and antithrombotic therapy serve as primary treatments. However, in-stent restenosis persists as a challenging clinical issue following PCI, lacking sustained and effective treatment. Linarin (LN) exhibits diverse pharmacological activities and is regarded as a potential drug for treating various diseases, including DM. But its specific role in restenosis after vascular injury in DM patients remains unclear. A rat model of diabetes-related restenosis was established to evaluate the role of LN on neointimal hyperplasia. Vascular smooth muscle cells (VSMCs) stimulated by high glucose (HG, 30 mM) underwent LN treatment. Additionally, an overexpression plasmid of A disintegrin and metalloproteinases (ADAM10) was constructed to transfect VSMCs. We employed CCK-8, Brdu, wound-healing scratch, and transwell migration assays to evaluate the proliferation and migration of VSMCs. Furthermore, western blot and immunofluorescence assays were utilized to investigate the expressions of ADAM10 and the downstream Notch signaling pathway in vivo and in vitro models. LN notably alleviated intimal hyperplasia after vascular injury in DM rats and reduced the protein expression of ADAM10, alongside its downstream Notch1 signaling pathway-related proteins (Notch1, NICD and Hes1) in rat carotid artery tissues. LN effectively suppressed the proliferation and migration of VSMCs induced by HG, downregulating the protein expression of ADAM10, Notch1, NICD and Hes1. Moreover, our findings indicated that ADAM10 overexpression significantly reversed LN\'s effects on proliferation, migration, and the expression of Notch1 signaling pathway-related proteins in HG-treated VSMCs. LN demonstrates potential therapeutic efficacy in addressing restenosis after diabetic-related vascular injury, with the ADAM10 mediated Notch signaling pathway playing a pivotal role.
    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
    转移是肝癌相关死亡的主要原因,也是肝癌预后不良的主要原因。尽管血小板已被证明可以加速肿瘤细胞的转移,确切的机制仍有待充分理解。这里,我们发现高血小板计数和肿瘤组织ADAM10表达增加提示HCC患者预后不良。同时,血小板计数与肿瘤组织ADAM10表达呈正相关。体外,我们发现血小板通过TLR4/NF-κB信号通路增加肿瘤细胞ADAM10的表达。ADAM10催化与CX3CR1受体结合的CX3CL1的脱落,然后诱导上皮细胞向间充质转化并激活癌细胞中的RhoA信号传导。此外,敲低肝癌细胞TLR4(Tlr4)或抑制ADAM10可阻止血小板增加的肿瘤细胞迁移,侵袭和内皮通透性。在体内,我们在小鼠肺转移模型中进一步验证了血小板通过癌细胞TLR4/ADAM10/CX3CL1轴加速肿瘤转移。总的来说,我们的研究为血小板诱导HCC转移的潜在机制提供了新的见解.因此,在癌细胞中靶向TLR4/ADAM10/CX3CL1轴有望抑制血小板促进的HCC肺转移。
    Metastasis is the main culprit of cancer-related death and account for the poor prognosis of hepatocellular carcinoma. Although platelets have been shown to accelerate tumor cell metastasis, the exact mechanism remained to be fully understood. Here, we found that high blood platelet counts and increased tumor tissue ADAM10 expression indicated the poor prognosis of HCC patients. Meanwhile, blood platelet count has positive correlation with tumor tissue ADAM10 expression. In vitro, we revealed that platelet increased ADAM10 expression in tumor cell through TLR4/NF-κB signaling pathway. ADAM10 catalyzed the shedding of CX3CL1 which bound to CX3CR1 receptor, followed by inducing epithelial to mesenchymal transition and activating RhoA signaling in cancer cells. Moreover, knockdown HCC cell TLR4 (Tlr4) or inhibition of ADAM10 prevented platelet-increased tumor cell migration, invasion and endothelial permeability. In vivo, we further verified in mice lung metastatic model that platelet accelerated tumor metastasis via cancer cell TLR4/ADAM10/CX3CL1 axis. Overall, our study provides new insights into the underlying mechanism of platelet-induced HCC metastasis. Therefore, targeting the TLR4/ADAM10/CX3CL1 axis in cancer cells hold promise for the inhibition of platelet-promoted lung metastasis of HCC.
    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
    APP,在阿尔茨海默病的发展中进行了充分的研究,最近已被确定为与TSCC相关的关键基因。这里,我们研究了APP及其通过ADAM10的蛋白水解裂解在TSCC发病机制中的功能。共纳入63例TSCC患者和30例健康对照,IHC检测结果显示,与配对的癌旁组织相比,ADAM10和APP在TSCC组织中的高表达。有趣的是,TSCC患者中APP的表达与ADAM10的表达相关,它们的联合表达与不良患者的生存有关。我们发现APP在TSCC细胞中α裂解形成sAPPα,TSCC患者血清sAPPα而非sAPPβ水平高于健康对照组。全长APP和sAPPα的过表达均可促进TSCC细胞增殖,移民和入侵。siRNA下调APP或ADAM10可减少sAPPα的产生,抑制ERK1/2和p38通路的活性,从而减少TSCC细胞增殖,移民和入侵。用ERK1/2或p38激动剂或sAPPα过表达的治疗逆转了APP或ADAM10敲低的作用。总之,我们的数据证明了ADAM10裂解的APP在TSCC细胞中激活ERK1/2和p38通路的致病作用.ADAM10和APP的高表达均与预后不良有关。靶向ADAM10裂解的APP可能是TSCC治疗的潜在策略。
    APP, well-studied in the development of Alzheimer\'s disease, has been recently identified as the key gene correlated with TSCC. Here, we investigate the function of APP and its proteolytic cleavage by ADAM10 in the pathogenesis of TSCC. A total of 63 TSCC patients and 30 healthy controls were included and the results of IHC assay showed high expressions of ADAM10 and APP in TSCC tissues compared to paired para-carcinoma tissues. Interestingly, APP expression in TSCC patients was correlated with ADAM10 expression and their combined expression was related to the poor patients\' survival. We found that APP was ɑ-cleaved in TSCC cells to form sAPPα, and the serum level of sAPPα but not sAPPβ in TSCC patients was higher than healthy controls. Both overexpression with full-length APP and sAPPα promoted TSCC cell proliferation, migration and invasion. Downregulation of APP or ADAM10 by siRNA decreased the generation of sAPPα and inhibited the activity of ERK1/2 and p38 pathways, thereby reducing TSCC cell proliferation, migration and invasion. Treatment with ERK1/2 or p38 agonist or sAPPα overexpression reversed the effects of APP or ADAM10 knockdown. In conclusion, our data demonstrated the pathogenic roles of APP cleaved by ADAM10 to activate ERK1/2 and p38 pathways in TSCC cells. Both high expressions of ADAM10 and APP were related to poor prognosis. Targeting APP cleaved by ADAM10 might be a potential strategy in TSCC treatment.
    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
    目的:过敏性鼻炎(AR)是一种常见的吸入性过敏性炎症性疾病,发病率较高。环状RNA(circularRNAs,circRNAs)已被揭示参与AR的发病机制。本研究探讨了cirMIRLET7BHG(hsa_circ_0008668)在AR进展中的生物学功能。
    方法:采用卵清蛋白(OVA)暴露的人鼻上皮细胞系(HNepC)和小鼠作为AR的体外和体内模型。免疫荧光染色用于确定上皮紧密连接蛋白的表达。通过RT-qPCR和Western印迹评估靶分子水平。通过RNA-FISH和免疫荧光观察到cirMIRLET7BHG和IGF2BP1的定位。通过跨上皮电阻和异硫氰酸荧光素-葡聚糖(FD4)通透性确定上皮屏障的损伤。血清IgE浓度,sIgE,IFN-γ,通过ELISA检测IL-4和IL-5。细胞凋亡,病理变化,通过TUNEL评估鼻粘膜组织中的嗜酸性粒细胞浸润,H&E,和天狼星红染色,分别。通过RNA下拉法分析了分子机制,RIP,和MeRIP测定。
    结果:在AR患者和实验模型中发现cirmMIRLET7BHG的表达增加。cirMIRLET7BHG的下调通过减轻上皮厚度来减轻OVA诱导的过敏症状,嗜酸性粒细胞浸润,凋亡,和小鼠的炎症反应。随后,circIRLET7BHG缺乏通过降低上皮通透性来预防OVA诱导的上皮屏障功能障碍,抑制紧密连接蛋白。机械上,甲基转移酶样3(METTL3)通过m6A甲基化增强cirmMIRLET7BHG表达,通过与IGF2BP1相互作用增强ADAM10mRNA的稳定性。
    结论:METTL3介导的m6A修饰增加了circirmMIRLET7BHG表达,因此通过与IGF2BP1相互作用提高了ADAM10mRNA的稳定性,从而通过诱导上皮屏障功能障碍促进AR。
    OBJECTIVE: Allergic rhinitis (AR) remains a frequent aspiratory allergic inflammatory disorder with a high incidence. Circular RNAs (circRNAs) have been revealed to participate in the pathogenesis of AR. This study investigated the biological function of circMIRLET7BHG (hsa_circ_0008668) in AR progression.
    METHODS: Ovalbumin (OVA)-exposed human nasal epithelial cell line (HNEpC) and mice were adopted as the in vitro and in vivo models of AR. Immunofluorescence staining was used to determine epithelial tight junction protein expression. Target molecule levels were assessed by RT-qPCR and Western blotting. Localization of circMIRLET7BHG and IGF2BP1 was observed by RNA-FISH and immunofluorescence. Epithelial barrier damage was determined by transepithelial electrical resistance and fluorescein isothiocyanate-dextran (FD4) permeability. Serum concentrations of IgE, sIgE, IFN-γ, IL-4, and IL-5 were detected by ELISA. Apoptosis, pathological changes, and eosinophil infiltration in nasal mucosa tissues were evaluated by TUNEL, H&E, and Sirius red staining, respectively. Molecular mechanism was analyzed by RNA pull-down, RIP, and MeRIP assays.
    RESULTS: An increased expression of circMIRLET7BHG was found in AR patients and experimental models. Down-regulation of circMIRLET7BHG attenuated OVA-induced allergic symptoms via relieving epithelial thicknesses, eosinophil infiltration, apoptosis, and inflammatory response in mice. Subsequently, circMIRLET7BHG deficiency prevented OVA-induced epithelial barrier dysfunction by reducing epithelial permeability, and inhibiting tight junction proteins. Mechanistically, methyltransferase-like 3 (METTL3) enhanced circMIRLET7BHG expression via m6A methylation, which enhanced ADAM10 mRNA stability via interaction with IGF2BP1.
    CONCLUSIONS: METTL3-mediated m6A modification increased circMIRLET7BHG expression that consequently raised ADAM10 mRNA stability via interplay with IGF2BP1, thereby promoting AR by inducing epithelial barrier dysfunction.
    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