invadopodia

Invadopodia
  • 文章类型: Journal Article
    肿瘤来源的细胞外囊泡(EV)是肿瘤的潜在生物标志物,但它们可靠的分子靶标尚未被确定。先前的研究证实泛素特异性蛋白酶22(USP22)在体内和体外促进肺腺癌(LUAD)转移。此外,USP22调节肿瘤细胞的内吞作用并定位到晚期内体。然而,USP22在肿瘤细胞源性EV分泌中的作用尚不清楚.在这项研究中,这表明USP22增加了肿瘤细胞来源的EV的分泌,并加速了它们的迁移和侵袭,invadopodia形成,和通过EV转移的血管生成。USP22通过上调肌球蛋白IB(MYO1B)增强EV分泌。本研究进一步发现USP22通过上调分子KDEL内质网蛋白滞留受体1(KDELR1)激活SRC信号通路,从而促进LUAD细胞进展。该研究为USP22在LUAD中EV分泌和细胞运动调节中的作用提供了新的见解。
    Tumor-derived extracellular vesicles (EVs) are potential biomarkers for tumors, but their reliable molecular targets have not been identified. The previous study confirms that ubiquitin-specific protease 22 (USP22) promotes lung adenocarcinoma (LUAD) metastasis in vivo and in vitro. Moreover, USP22 regulates endocytosis of tumor cells and localizes to late endosomes. However, the role of USP22 in the secretion of tumor cell-derived EVs remains unknown. In this study, it demonstrates that USP22 increases the secretion of tumor cell-derived EVs and accelerates their migration and invasion, invadopodia formation, and angiogenesis via EV transfer. USP22 enhances EV secretion by upregulating myosin IB (MYO1B). This study further discovers that USP22 activated the SRC signaling pathway by upregulating the molecule KDEL endoplasmic reticulum protein retention receptor 1 (KDELR1), thereby contributing to LUAD cell progression. The study provides novel insights into the role of USP22 in EV secretion and cell motility regulation in 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
    背景:脱落酸受体蓝硫氨酸合成酶C样2(LanCL2)是年轻胶质母细胞瘤患者总体生存的预后因素。然而,LanCL2在胶质母细胞瘤中的作用和潜在机制尚不清楚.
    目的:本研究探讨LanCL2对胶质母细胞瘤体外细胞侵袭和体内肿瘤进展的调控作用及其机制。
    方法:使用定点诱变使LanCL2的酪氨酸198或295残基突变以阻断其磷酸化。使用transwell或3D侵袭试验研究了LanCL2在胶质母细胞瘤中的作用,基质降解试验和颅内异种移植模型。
    结果:这项研究显示了LanCL2的核定位信号,并通过脱落酸或LanCL2的过表达在胶质母细胞瘤细胞中增加了核转运。击倒LanCL2抑制迁移,胶质母细胞瘤细胞的侵袭和侵袭足形成,而野生型LanCL2的过表达增强了它们。LanCL2的Tyr295残基磷酸化的阻断阻碍了其核运输,胶质母细胞瘤细胞运动性和侵袭足形成迟缓,并降低了Cortactin和STAT3的磷酸化。c-Met被鉴定为LanCL2的Tyr295残基的上游酪氨酸激酶,并且c-Met的抑制显著抑制了LanCL2的核转运。此外,过表达野生型LanCL2显著促进胶质母细胞瘤的体内原位肿瘤生长,导致小鼠中位生存时间33.5天,而Tyr295突变拯救了它,中位生存时间为49天.
    结论:我们的研究结果表明,Tyr295的磷酸化对LanCL2的激活和核运输以及GBM的侵袭足形成和肿瘤进展至关重要,提供新的信号轴c-Met/LanCL2/STAT3/Cortactin的证据,并首次观察到Tyr295磷酸化对LanCL2的重要性。
    BACKGROUND: Our previous study showed that the abscisic acid receptor lanthionine synthetase C-like 2 (LanCL2) is a significant prognostic factor for overall survival in young glioblastoma patients. However, the role of LanCL2 in glioblastoma remains unclear yet.
    OBJECTIVE: This study aims to investigate the role of LanCL2 in regulating in-vitro cell invasion and in-vivo tumor progression of glioblastoma and its underlying mechanism.
    METHODS: Tyrosine 198 or 295 residue of LanCL2 was mutated using site-directed mutagenesis to block its phosphorylation. The role of LanCL2 in glioblastoma was investigated using transwell or 3D invasion assay, matrix degradation assay and intracranial xenograft model.
    RESULTS: This study showed that nuclear transport of LanCL2 was enhanced by overexpression of LanCL2 or its ligand abscisic acid in glioblastoma cells. Knockdown of LanCL2 suppressed migration, invasion and invadopodia formation of glioblastoma cells, whereas overexpression of wild-type LanCL2 enhanced them. Blocking of Tyr295 residue phosphorylation of LanCL2 impeded its nuclear transport, retarded glioblastoma cell motility and invadopodia formation, and deceased the phosphorylation of Cortactin and STAT3. c-Met was identified as the upstream tyrosine kinase of Tyr295 residue of LanCL2, and inhibition of c-Met markedly suppressed the nuclear transport of LanCL2. Moreover, overexpression of wild-type LanCL2 significantly promoted orthotopic tumor growth of glioblastoma in vivo and led to poor survival of mice with median survival time of 33.5 days, whereas Tyr295 mutation rescued it with median survival time of 49 days.
    CONCLUSIONS: Our findings suggested that Tyr295 phosphorylation is crucial to the activation and nuclear transport of LanCL2, as well as invadopodia formation and tumor progression of glioblastoma, providing the evidence of a novel signaling axis c-Met/LanCL2/STAT3/Cortactin and the first observation of the importance of Tyr295 phosphorylation to LanCL2.
    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
    肌动蛋白结合蛋白1(Fascin)在多种癌症中高表达,包括食管鳞状细胞癌(ESCC),作为一种重要的致癌蛋白,并通过捆绑F-肌动蛋白促进癌细胞的迁移和侵袭,以促进丝状和invadopodia的形成。然而,目前尚不清楚Fascin的功能如何在癌细胞中被乙酰化调节。这里,在ESCC细胞中,组蛋白乙酰转移酶KAT8催化Fascin赖氨酸41(K41)乙酰化,抑制Fascin介导的F-肌动蛋白成束以及丝足和invadopodia的形成。此外,NAD依赖的蛋白质去乙酰化酶sirtuin(SIRT)7介导的Fascin-K41的去乙酰化增强了丝状伪足和invadopodia的形成,促进ESCC细胞的迁移和侵袭。临床上,对ESCC患者的癌组织和癌旁组织样本的分析表明,有淋巴结转移的患者的癌组织中的Fascin-K41乙酰化程度低于无淋巴结转移的患者,和低水平的Fascin-K41乙酰化与ESCC患者预后较差相关。重要的是,K41乙酰化显著阻断NP-G2-044,目前正在临床评估的Fascin抑制剂之一,表明NP-G2-044可能更适合Fascin-K41乙酰化水平低的患者,但不适用于Fascin-K41乙酰化水平高的患者。©2024英国和爱尔兰病理学会。由JohnWiley&Sons出版,Ltd.
    Fascin actin-bundling protein 1 (Fascin) is highly expressed in a variety of cancers, including esophageal squamous cell carcinoma (ESCC), working as an important oncogenic protein and promoting the migration and invasion of cancer cells by bundling F-actin to facilitate the formation of filopodia and invadopodia. However, it is not clear how exactly the function of Fascin is regulated by acetylation in cancer cells. Here, in ESCC cells, the histone acetyltransferase KAT8 catalyzed Fascin lysine 41 (K41) acetylation, to inhibit Fascin-mediated F-actin bundling and the formation of filopodia and invadopodia. Furthermore, NAD-dependent protein deacetylase sirtuin (SIRT) 7-mediated deacetylation of Fascin-K41 enhances the formation of filopodia and invadopodia, which promotes the migration and invasion of ESCC cells. Clinically, the analysis of cancer and adjacent tissue samples from patients with ESCC showed that Fascin-K41 acetylation was lower in the cancer tissue of patients with lymph node metastasis than in that of patients without lymph node metastasis, and low levels of Fascin-K41 acetylation were associated with a poorer prognosis in patients with ESCC. Importantly, K41 acetylation significantly blocked NP-G2-044, one of the Fascin inhibitors currently being clinically evaluated, suggesting that NP-G2-044 may be more suitable for patients with low levels of Fascin-K41 acetylation, but not suitable for patients with high levels of Fascin-K41 acetylation. © 2024 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
    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
    越来越多的证据表明,增加的基质硬度可以显着加强肝细胞癌(HCC)细胞的恶性特征。然而,增加的基质硬度是否以及如何调节HCC细胞内翻的形成仍然未知。在研究中,我们在体外和体内建立了不同的实验系统,以探讨基质刚度对invadopodia形成的影响及其相关分子机制。我们的结果表明,增加的基质刚度显着增强肝癌细胞的迁移和侵袭能力,上调invadopodia相关基因的表达并增加invadopodia的数量。两种调节途径有助于在HCC细胞中一起形成基质刚度驱动的invadopodia,包括通过激活整合素β1或Piezo1/FAK/Src/Arg/cortactin途径直接触发侵袭足形成,并通过改善EGF的产生来激活EGFR/Src/Arg/cortactin通路,从而间接刺激侵袭足病的形成。Src被鉴定为两个协同调节途径的共同hub分子。同时,整合素β1/RhoA/ROCK1/MLC2和Piezo1/Ca2/MLCK/MLC2途径的激活介导基质刚度增强的细胞迁移。这项研究揭示了机械感觉途径和生化信号途径协同调节HCC细胞内翻足病形成的新机制。
    Growing evidence has suggested that increased matrix stiffness can significantly strengthen the malignant characteristics of hepatocellular carcinoma (HCC) cells. However, whether and how increased matrix stiffness regulates the formation of invadopodia in HCC cells remain largely unknown. In the study, we developed different experimental systems in vitro and in vivo to explore the effects of matrix stiffness on the formation of invadopodia and its relevant molecular mechanism. Our results demonstrated that increased matrix stiffness remarkably augmented the migration and invasion abilities of HCC cells, upregulated the expressions of invadopodia-associated genes and enhanced the number of invadopodia. Two regulatory pathways contribute to matrix stiffness-driven invadopodia formation together in HCC cells, including direct triggering invadopodia formation through activating integrin β1 or Piezo1/ FAK/Src/Arg/cortactin pathway, and indirect stimulating invadopodia formation through improving EGF production to activate EGFR/Src/Arg/cortactin pathway. Src was identified as the common hub molecule of two synergistic regulatory pathways. Simultaneously, activation of integrin β1/RhoA/ROCK1/MLC2 and Piezo1/Ca2+/MLCK/MLC2 pathways mediate matrix stiffness-reinforced cell migration. This study uncovers a new mechanism by which mechanosensory pathway and biochemical signal pathway synergistically regulate the formation of invadopodia in HCC cells.
    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
    在胃癌(GC)中,肝脏是远处转移的常见器官,胃癌肝转移(GCLM)患者一般预后较差。GCLM的机制尚不清楚。Invadopodia是由肿瘤细胞形成的特殊的膜突起,可以降解基底膜和ECM。在这里,我们研究了invadopodia在GCLM中的作用。我们发现,肝转移患者的侵袭足病相关蛋白水平明显高于GCLM患者的原发性肿瘤。此外,GC细胞可通过分泌血小板源性生长因子亚基B(PDGFB)激活肝转移瘤微环境中的肝星状细胞(HSC)。活化的HSC分泌肝细胞生长因子(HGF),激活了MET原癌基因,GC细胞的MET受体,从而通过PI3K/AKT途径促进侵袭性足足的形成,并随后增强GC细胞的侵袭和转移。因此,通过PDGFB/血小板衍生生长因子受体β(PDGFRβ)和HGF/MET轴在GC细胞和HSCs之间的交叉可能代表治疗GCLM的潜在治疗靶点。
    In gastric cancer (GC), the liver is a common organ for distant metastasis, and patients with gastric cancer with liver metastasis (GCLM) generally have poor prognosis. The mechanism of GCLM is unclear. Invadopodia are special membrane protrusions formed by tumor cells that can degrade the basement membrane and ECM. Herein, we investigated the role of invadopodia in GCLM. We found that the levels of invadopodia-associated proteins were significantly higher in liver metastasis than in the primary tumors of patients with GCLM. Furthermore, GC cells could activate hepatic stellate cells (HSCs) within the tumor microenvironment of liver metastases through the secretion of platelet-derived growth factor subunit B (PDGFB). Activated HSCs secreted hepatocyte growth factor (HGF), which activated the MET proto-oncogene, MET receptor of GC cells, thereby promoting invadopodia formation through the PI3K/AKT pathway and subsequently enhancing the invasion and metastasis of GC cells. Therefore, cross-talk between GC cells and HSCs by PDGFB/platelet derived growth factor receptor beta (PDGFRβ) and the HGF/MET axis might represent potential therapeutic targets to treat GCLM.
    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
    我们最近表明,ORP3的缺失导致非整倍性诱导并促进肿瘤形成。然而,ORP3有助于倍性控制以及癌症发生和进展的具体机制尚不清楚.这里,我们报道,ORP3在输尿管和膀胱上皮中高表达,而其表达在浸润性膀胱癌细胞系和肿瘤进展过程中下调。在人类和小鼠膀胱癌中。此外,在组织特异性Orp3基因敲除小鼠中,我们观察到N-丁基-N-(4-羟丁基)-亚硝胺(BBN)诱导的浸润性膀胱癌的发生率增加.实验数据表明,ORP3蛋白在中心体处与γ-微管蛋白相互作用,并与肌动蛋白细胞骨架的成分相互作用。改变ORP3的表达会诱导具有稳定核型的端粒酶永生化尿路上皮细胞的非整倍体和基因组不稳定性,并影响膀胱癌细胞系的迁移和侵袭能力。这些发现证明了ORP3在倍性控制中的关键作用,并表明ORP3是真正的肿瘤抑制蛋白。值得注意的是,提供的数据表明,ORP3通过与细胞骨架成分的相互作用影响细胞侵袭和迁移以及基因组稳定性,提供非整倍性与细胞侵袭和迁移之间的分子联系,转移细胞的两个关键特征。
    We have recently shown that loss of ORP3 leads to aneuploidy induction and promotes tumor formation. However, the specific mechanisms by which ORP3 contributes to ploidy-control and cancer initiation and progression is still unknown. Here, we report that ORP3 is highly expressed in ureter and bladder epithelium while its expression is downregulated in invasive bladder cancer cell lines and during tumor progression, both in human and in mouse bladder cancer. Moreover, we observed an increase in the incidence of N-butyl-N-(4-hydroxybutyl)-nitrosamine (BBN)-induced invasive bladder carcinoma in the tissue-specific Orp3 knockout mice. Experimental data demonstrate that ORP3 protein interacts with γ-tubulin at the centrosomes and with components of actin cytoskeleton. Altering the expression of ORP3 induces aneuploidy and genomic instability in telomerase-immortalized urothelial cells with a stable karyotype and influences the migration and invasive capacity of bladder cancer cell lines. These findings demonstrate a crucial role of ORP3 in ploidy-control and indicate that ORP3 is a bona fide tumor suppressor protein. Of note, the presented data indicate that ORP3 affects both cell invasion and migration as well as genome stability through interactions with cytoskeletal components, providing a molecular link between aneuploidy and cell invasion and migration, two crucial characteristics of metastatic cells.
    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
    原发癌调节骨微环境,在肿瘤细胞中播下休眠和转移的种子,导致多器官转移和死亡。在这项研究中,3D打印和芯片上骨骼(BOC)相结合,开发了一种BOC平台,该平台可以模拟转移前的壁ni(PMN),并有助于阐明在原发癌的影响下,骨骼驻留细胞与转移性肿瘤细胞之间的相互作用。光可交联明胶甲基丙烯酸酯(GelMA)用作3D培养水凝胶来封装细胞,与水凝胶相邻的肿瘤培养基(CM)循环,以验证间充质干细胞(MSCs)和破骨细胞(RAW264.7s)的关键作用。三个生态位:休眠生态位,血管周围的小生境,和“恶性循环”利基,旨在在具有高细胞活力和出色营养交换的一个芯片中概括骨转移。关于肿瘤休眠和再激活,研究了通过cortactin途径与MSCs和RAW264.7通讯的A549肺癌细胞的invadopodia形成。作为概念的证明,通过分析invadopodia的形成和各种细胞的影响,证明了平台的功能性和实用性,动态生态位的建立为理解PMN的形成和相关药物的发现铺平了道路。
    Primary cancer modulates the bone microenvironment to sow the seeds of dormancy and metastasis in tumor cells, leading to multiple organ metastasis and death. In this study, 3D printing and bone-on-a-chip (BOC) are combined to develop a BOC platform that mimics the pre-metastatic niches (PMNs) and facilitates elucidation of the interactions between bone-resident cells and metastatic tumor cells under the influence of primary cancer. Photocrosslinkable gelatin methacrylate (GelMA) is used as a 3D culturing hydrogel to encapsulate cells, and circulate tumor culture medium (CM) adjacent to the hydrogel to verify the critical role of mesenchymal stem cells (MSCs) and osteoclasts (RAW264.7s). Three niches: the dormancy niche, the perivascular niche, and the \"vicious cycle\" niche, are devised to recapitulate bone metastasis in one chip with high cell viability and excellent nutrient exchange. With respect to tumor dormancy and reactivation, the invadopodia formation of A549 lung cancer cells in communication with MSCs and RAW264.7 via the cortactin pathway is researched. As a proof of concept, the functionality and practicality of the platform are demonstrated by analyzing the invadopodia formation and the influence of various cells, and the establishment of the dynamic niches paves the way to understanding PMN formation and related drug discovery.
    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
    作为最具侵袭性的乳腺癌,三阴性乳腺癌(TNBC)仍然无法治愈,并且很容易发生转移。转化生长因子β(TGF-β)诱导的上皮间质转化(EMT)与TNBC的生长和转移至关重要。这项研究报道了一种天然化合物同位素烯丹素(ITSN)通过抑制TGF-β诱导的EMT和invadopodia的形成来减少TNBC转移。ITSN可以直接与TGF-β受体1(TGFβR1)相互作用,并消除TGFβR1的激酶活性,从而阻断TGF-β启动的下游信号通路。此外,在体外TGFβR1过表达的TNBC细胞以及携带TGFβR1过表达的TNBC细胞的小鼠中,ITSN提供的对转移的抑制作用明显消失。此外,发现TGFβR1激酶结构域中的Lys232和Asp351残基对于ITSN与TGFβR1的相互作用至关重要。此外,ITSN还通过抑制肿瘤微环境中的TGF-β介导的EMT来提高程序性细胞死亡1配体1(PD-L1)抗体对TNBC的体内抑制作用。我们的发现不仅突出了TGFβR1在TNBC转移中的关键作用,而且还提供了靶向TGFβR1的先导化合物用于治疗TNBC转移。此外,这项研究还指出了通过联合应用抗PD-L1和TGFβR1抑制剂治疗TNBC的潜在策略。
    As the most aggressive breast cancer, triple-negative breast cancer (TNBC) is still incurable and very prone to metastasis. The transform growth factor β (TGF-β)-induced epithelial-mesenchymal transition (EMT) is crucially involved in the growth and metastasis of TNBC. This study reported that a natural compound isotoosendanin (ITSN) reduced TNBC metastasis by inhibiting TGF-β-induced EMT and the formation of invadopodia. ITSN can directly interact with TGF-β receptor type-1 (TGFβR1) and abrogated the kinase activity of TGFβR1, thereby blocking the TGF-β-initiated downstream signaling pathway. Moreover, the ITSN-provided inhibition on metastasis obviously disappeared in TGFβR1-overexpressed TNBC cells in vitro as well as in mice bearing TNBC cells overexpressed TGFβR1. Furthermore, Lys232 and Asp351 residues in the kinase domain of TGFβR1 were found to be crucial for the interaction of ITSN with TGFβR1. Additionally, ITSN also improved the inhibitory efficacy of programmed cell death 1 ligand 1 (PD-L1) antibody for TNBC in vivo via inhibiting the TGF-β-mediated EMT in the tumor microenvironment. Our findings not only highlight the key role of TGFβR1 in TNBC metastasis, but also provide a leading compound targeting TGFβR1 for the treatment of TNBC metastasis. Moreover, this study also points out a potential strategy for TNBC treatment by using the combined application of anti-PD-L1 with a TGFβR1 inhibitor.
    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
    在之前的研究中,我们的研究组观察到雌激素通过雌激素受体β(ERβ)促进非小细胞肺癌(NSCLC)的转移。Invadopodia是参与肿瘤转移的关键结构。然而,目前尚不清楚ERβ是否通过侵入足来促进NSCLC转移。在我们的研究中,我们使用扫描电子显微镜观察了ERβ过表达和E2处理后invadopodia的形成。使用多种NSCLC细胞系的体外实验表明,ERβ可以增加侵袭足的形成和细胞侵袭。机制研究表明,ERβ可以通过直接与位于ICAM1启动子上的雌激素反应元件(EREs)结合来上调ICAM1的表达,这反过来可以增强Src/cortactin的磷酸化。我们还使用原位肺移植小鼠模型在体内证实了这些发现,验证了体外实验的结果。最后,我们使用免疫组织化学方法检测了ERβ和ICAM1在NSCLC组织和配对转移性淋巴结中的表达。结果证实,ERβ通过ICAM-1/p-Src/p-Cortactin信号通路促进NSCLC细胞内窝形成。
    In a previous study, our research group observed that estrogen promotes the metastasis of non-small cell lung cancer (NSCLC) through the estrogen receptor β (ERβ). Invadopodia are key structures involved in tumor metastasis. However, it is unclear whether ERβ is involved in the promotion of NSCLC metastasis through invadopodia. In our study, we used scanning electron microscopy to observe the formation of invadopodia following the overexpression of ERβ and treatment with E2. In vitro experiments using multiple NSCLC cell lines demonstrated that ERβ can increase the formation of invadopodia and cell invasion. Mechanistic studies revealed that ERβ can upregulate the expression of ICAM1 by directly binding to estrogen-responsive elements (EREs) located on the ICAM1 promoter, which in turn can enhance the phosphorylation of Src/cortactin. We also confirmed these findings in vivo using an orthotopic lung transplantation mouse model, which validated the results obtained from the in vitro experiments. Finally, we examined the expressions of ERβ and ICAM1 using immunohistochemistry in both NSCLC tissue and paired metastatic lymph nodes. The results confirmed that ERβ promotes the formation of invadopodia in NSCLC cells through the ICAM1/p-Src/p-Cortactin signaling 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
    雌激素相关受体α(ERRα)在子宫内膜癌(EC)的进展中起重要作用。然而,ERRα在EC侵袭和转移中的生物学作用尚不清楚。本研究旨在探讨ERRα和3-羟基-3-甲基戊二酰辅酶A合酶1(HMGCS1)在调节细胞内胆固醇代谢以促进EC进展中的作用。通过免疫共沉淀检测ERRα和HMGCS1的相互作用,并通过伤口愈合和transwell小室侵袭试验研究了ERRα/HMGCS1对EC转移的影响。测量细胞胆固醇含量以验证ERRα与细胞胆固醇代谢之间的关系。此外,进行免疫组织化学以确认ERRα和HMGCS1与EC进展相关。此外,该机制通过功能丧失和功能获得试验或辛伐他汀治疗进行了研究.ERRα和HMGCS1的高表达水平促进了细胞内胆固醇代谢,从而形成了invadadopodia。此外,抑制ERRα和HMGCS1的表达在体外和体内显着削弱了EC的恶性进展。我们的功能分析显示ERRα通过HMGCS1介导的细胞内胆固醇代谢途径促进EC的侵袭和转移,依赖于上皮-间质转化途径。我们的研究结果表明,ERRα和HMGCS1是抑制EC进展的潜在靶点。
    Estrogen-related receptor alpha (ERRα) plays an important role in endometrial cancer (EC) progression. However, the biological roles of ERRα in EC invasion and metastasis are not clear. This study aimed to investigate the role of ERRα and 3-hydroxy-3-methylglutaryl-CoA synthase 1 (HMGCS1) in regulating intracellular cholesterol metabolism to promote EC progression. ERRα and HMGCS1 interactions were detected by co-immunoprecipitation, and the effects of ERRα/HMGCS1 on the metastasis of EC were investigated by wound-healing and transwell chamber invasion assays. Cellular cholesterol content was measured to verify the relationship between ERRα and cellular cholesterol metabolism. Additionally, immunohistochemistry was performed to confirm that ERRα and HMGCS1 were related to EC progression. Furthermore, the mechanism was investigated using loss-of-function and gain-of-function assays or treatment with simvastatin. High expression levels of ERRα and HMGCS1 promoted intracellular cholesterol metabolism for invadopodia formation. Moreover, inhibiting ERRα and HMGCS1 expression significantly weakened the malignant progression of EC in vitro and in vivo. Our functional analysis showed that ERRα promoted EC invasion and metastasis through the HMGCS1-mediated intracellular cholesterol metabolism pathway, which was dependent on the epithelial-mesenchymal transition pathway. Our findings suggest that ERRα and HMGCS1 are potential targets to suppress EC progression.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号