invadopodia

Invadopodia
  • 文章类型: Journal Article
    多细胞生物由细胞和细胞外基质(ECM)组成。ECM创造了一个细胞微环境,和细胞根据其细胞活性局部降解ECM。修饰ECM的主要酶属于基质金属蛋白酶(MMPs),在各种病理生理事件中起着重要作用。MMP的ECM降解不会在所有细胞环境中发生,而只是在必要时发生,细胞通过定向分泌这些蛋白水解酶来实现。最近的研究表明,这种酶的分泌是通过沿着微管的靶向囊泡运输来实现的,和几种驱动蛋白超家族蛋白(KIFs)已被鉴定为参与该过程的负责任的运动蛋白。本章讨论了MMPs囊泡运输的最新发现及其作用。
    Multicellular organisms consist of cells and extracellular matrix (ECM). ECM creates a cellular microenvironment, and cells locally degrade the ECM according to their cellular activity. A major group of enzymes that modify ECM belongs to matrix metalloproteinases (MMPs) and play major roles in various pathophysiological events. ECM degradation by MMPs does not occur in all cellular surroundings but only where it is necessary, and cells achieve this by directionally secreting these proteolytic enzymes. Recent studies have indicated that such enzyme secretion is achieved by targeted vesicle transport along the microtubules, and several kinesin superfamily proteins (KIFs) have been identified as responsible motor proteins involved in the processes. This chapter discusses recent findings of the vesicle transport of MMPs and their roles.
    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:我们先前已经确定了GJB3的一个意外作用,表明这种连接蛋白蛋白的缺乏在人和鼠细胞中诱导非整倍性,并在异种移植模型中加速细胞转化以及肿瘤形成。GJB3丢失导致非整倍性和癌症发生和进展的分子机制仍未解决。
    方法:通过RT-qPCR和Western印迹测定GJB3表达水平。通过中期染色体计数评估GJB3敲低对基因组不稳定性的影响,多核细胞,通过微核形成和纺锤体取向的确定。通过免疫沉淀和免疫细胞化学分析GJB3与α-微管蛋白和F-肌动蛋白的相互作用。光漂白实验后,通过活细胞成像和荧光恢复测量GJB3缺乏对微管和肌动蛋白动力学的影响,分别。免疫组织化学用于确定人和鼠膀胱癌组织切片上的GJB3水平。通过BBN处理化学诱导小鼠的膀胱癌。
    结果:我们发现GJB3在输尿管和膀胱上皮中高表达,但它在浸润性膀胱癌细胞系中以及在人和小鼠膀胱癌的肿瘤进展过程中下调。GJB3表达的下调导致核型稳定的尿路上皮细胞的非整倍性和基因组不稳定性,并且GJB3水平的实验调节改变了膀胱癌细胞系的迁移和侵袭能力。重要的是,GJB3与α-微管蛋白和F-肌动蛋白相互作用。这些相互作用的损害改变了这些细胞骨架成分的动力学,并导致纺锤体取向缺陷。
    结论:我们得出结论,微管和肌动蛋白动力学失调对适当的染色体分离和肿瘤细胞侵袭和迁移有影响。因此,这些观察结果表明GJB3在膀胱癌的发病和扩散中可能发挥作用,并证明在肿瘤细胞播散过程中增强的非整倍性和侵袭能力之间存在分子联系.
    BACKGROUND: We have previously identified an unsuspected role for GJB3 showing that the deficiency of this connexin protein induces aneuploidy in human and murine cells and accelerates cell transformation as well as tumor formation in xenograft models. The molecular mechanisms by which loss of GJB3 leads to aneuploidy and cancer initiation and progression remain unsolved.
    METHODS: GJB3 expression levels were determined by RT-qPCR and Western blot. The consequences of GJB3 knockdown on genome instability were assessed by metaphase chromosome counting, multinucleation of cells, by micronuclei formation and by the determination of spindle orientation. Interactions of GJB3 with α-tubulin and F-actin was analyzed by immunoprecipitation and immunocytochemistry. Consequences of GJB3 deficiency on microtubule and actin dynamics were measured by live cell imaging and fluorescence recovery after photobleaching experiments, respectively. Immunohistochemistry was used to determine GJB3 levels on human and murine bladder cancer tissue sections. Bladder cancer in mice was chemically induced by BBN-treatment.
    RESULTS: We find that GJB3 is highly expressed in the ureter and bladder epithelium, but it is downregulated in invasive bladder cancer cell lines and during tumor progression in both human and mouse bladder cancer. Downregulation of GJB3 expression leads to aneuploidy and genomic instability in karyotypically stable urothelial cells and experimental modulation of GJB3 levels alters the migration and invasive capacity of bladder cancer cell lines. Importantly, GJB3 interacts both with α-tubulin and F-actin. The impairment of these interactions alters the dynamics of these cytoskeletal components and leads to defective spindle orientation.
    CONCLUSIONS: We conclude that deregulated microtubule and actin dynamics have an impact on proper chromosome separation and tumor cell invasion and migration. Consequently, these observations indicate a possible role for GJB3 in the onset and spreading of bladder cancer and demonstrate a molecular link between enhanced aneuploidy and invasive capacity cancer cells during tumor cell dissemination.
    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
    乳腺癌是一种异质性疾病,仍然是全球女性中最常见的恶性肿瘤。在乳腺肿瘤的基因组分析中,发现IQGAP3的mRNA水平在三阴性肿瘤中上调。随后发现IQGAP3在一组三阴性乳腺癌(TNBC)细胞系中表达。IQGAP3的表达水平下降,延长细胞,破坏的细胞迁移,并抑制细胞形成特化侵袭性粘附结构的能力,称为invadopodia。发现IQGAP3耗竭诱导的形态变化取决于RhoA。的确,IQGAP3表达降低破坏了RhoA活性和肌动球蛋白收缩性。有趣的是,还发现IQGAP3与p-21活化激酶6(PAK6)相互作用;一种已经与细胞形态调节相关的蛋白质。此外,PAK6耗竭在这些细胞中显现IQGAP3耗竭。而PAK6过表达拯救了IQGAP3耗竭表型。我们的工作指出了一个重要的PAK6-IQGAP3-RhoA途径,该途径驱动乳腺癌细胞的细胞收缩性,促进这些细胞的细胞迁移和粘附侵袭。由于这种表型与转移过程和转移的重新接种有关,PAK6的药物靶向可能导致TNBC患者的临床获益。
    Breast cancer is a heterogeneous disease that remains the most common malignancy among women worldwide. During genomic analysis of breast tumours, mRNA levels of IQGAP3 were found to be upregulated in triple negative tumours. IQGAP3 was subsequently found to be expressed across a panel of triple negative breast cancer (TNBC) cell lines. Depleting expression levels of IQGAP3 delivered elongated cells, disrupted cell migration, and inhibited the ability of cells to form specialised invasive adhesion structures, termed invadopodia. The morphological changes induced by IQGAP3 depletion were found to be dependent on RhoA. Indeed, reduced expression of IQGAP3 disrupted RhoA activity and actomyosin contractility. Interestingly, IQGAP3 was also found to interact with p-21 activated kinase 6 (PAK6); a protein already associated with the regulation of cell morphology. Moreover, PAK6 depletion phenocopied IQGAP3 depletion in these cells. Whereas PAK6 overexpression rescued the IQGAP3 depletion phenotype. Our work points to an important PAK6-IQGAP3-RhoA pathway that drives the cellular contractility of breast cancer cells promoting both cell migration and adhesive invasion of these cells. As this phenotype is relevant to the process of metastasis and re-seeding of metastasis, the pharmacological targeting of PAK6 could lead to clinical benefit in TNBC patients.
    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:胰腺导管腺癌(PDAC)是一种致命的疾病,由于早期转移播散和高化学耐药性而具有高死亡率。所有这些因素都受到其富含细胞外基质(ECM)的微环境的青睐,它也是高度缺氧和酸性的。吉西他滨(GEM)仍然是PDAC的一线疗法。然而,它迅速脱去氨基为其无活性的代谢产物。已经出现了几种GEM前药以改善其细胞毒性。这里,我们分析了酸性/低氧肿瘤微环境(TME)如何影响PDAC细胞死亡和侵袭足病介导的ECM蛋白水解对GEM及其C18前药的反应.
    方法:为此,两个PDAC细胞系,PANC-1和MiaPaCa-2适应pH6.6或不适应1个月,生长为3D器官型培养物,并在存在和不存在酸中毒和缺氧诱导剂的情况下暴露于GEM或C18,去铁胺.
    结果:我们发现,在所有培养条件下,尤其是在酸性和低氧环境中,C18均比GEM具有更高的细胞毒性和抗invadopodia活性。
    结论:我们建议C18作为一种更有效的方法来开发克服PDAC化学耐药的新治疗策略。
    BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) is a deadly disease with high mortality due to early metastatic dissemination and high chemoresistance. All these factors are favored by its extracellular matrix (ECM)-rich microenvironment, which is also highly hypoxic and acidic. Gemcitabine (GEM) is still the first-line therapy in PDAC. However, it is quickly deaminated to its inactive metabolite. Several GEM prodrugs have emerged to improve its cytotoxicity. Here, we analyzed how the acidic/hypoxic tumor microenvironment (TME) affects the response of PDAC cell death and invadopodia-mediated ECM proteolysis to both GEM and its C18 prodrug.
    METHODS: For this, two PDAC cell lines, PANC-1 and Mia PaCa-2 were adapted to pHe 6.6 or not for 1 month, grown as 3D organotypic cultures and exposed to either GEM or C18 in the presence and absence of acidosis and the hypoxia inducer, deferoxamine.
    RESULTS: We found that C18 has higher cytotoxic and anti-invadopodia activity than GEM in all culture conditions and especially in acid and hypoxic environments.
    CONCLUSIONS: We propose C18 as a more effective approach to conventional GEM in developing new therapeutic strategies overcoming PDAC chemoresistance.
    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
    乳酸盐-质子协同转运蛋白MCT4及其伴侣CD147在乳腺癌中上调,与患者生存率下降有关。这里,我们检验了MCT4和CD147通过对细胞外基质(ECM)降解的相互依赖作用而有利于乳腺癌侵袭的假设。在MDA-MB-231乳腺癌细胞中,MCT4和CD147的表达和膜定位强烈地相互依赖。MCT4和/或CD174的敲低(KD)和过表达(OE)-和降低,分别,迁移,入侵,和荧光明胶降解。两种蛋白质的OE增加了明胶降解和基质金属蛋白酶(MMP)产生的胶原蛋白I裂解产物reC1M的出现,提示在ECM降解中的协同作用。在F-肌动蛋白修饰的细胞内囊泡中,MCT4和CD147与质膜上的invadopodia标记物以及MMP14,溶酶体标记物LAMP-1,部分与自噬体标记物LC3共定位。我们得出的结论是,MCT4和CD147相互调节,并相互依赖地支持MDA-MB-231乳腺癌细胞的迁移和侵袭性。机械上,这涉及MCT4-CD147依赖性刺激ECM降解,特别是MMP介导的胶原-I降解.我们建议MCT4-CD147复合物与MMP14共同递送至侵袭足。
    Expression levels of the lactate-H+ cotransporter MCT4 (also known as SLC16A3) and its chaperone CD147 (also known as basigin) are upregulated in breast cancers, correlating with decreased patient survival. Here, we test the hypothesis that MCT4 and CD147 favor breast cancer invasion through interdependent effects on extracellular matrix (ECM) degradation. MCT4 and CD147 expression and membrane localization were found to be strongly reciprocally interdependent in MDA-MB-231 breast cancer cells. Overexpression of MCT4 and/or CD147 increased, and their knockdown decreased, migration, invasion and the degradation of fluorescently labeled gelatin. Overexpression of both proteins led to increases in gelatin degradation and appearance of the matrix metalloproteinase (MMP)-generated collagen-I cleavage product reC1M, and these increases were greater than those observed upon overexpression of each protein alone, suggesting a concerted role in ECM degradation. MCT4 and CD147 colocalized with invadopodia markers at the plasma membrane. They also colocalized with MMP14 and the lysosomal marker LAMP1, as well as partially with the autophagosome marker LC3, in F-actin-decorated intracellular vesicles. We conclude that MCT4 and CD147 reciprocally regulate each other and interdependently support migration and invasiveness of MDA-MB-231 breast cancer cells. Mechanistically, this involves MCT4-CD147-dependent stimulation of ECM degradation and specifically of MMP-mediated collagen-I degradation. We suggest that the MCT4-CD147 complex is co-delivered to invadopodia with MMP14.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Preprint
    头颈部鳞状细胞癌(HNSCC)是一个主要的健康问题,因为它的高死亡率来自不良的治疗反应和局部肿瘤侵入头颈部的生命维持结构。对HNSCC侵袭机制的更深入理解具有告知可能增强患者生存的靶向治疗的潜力。我们先前报道了双皮质素样激酶1(DCLK1)调节HNSCC细胞的侵袭。这里,我们检验了DCLK1调节invadopodia内的蛋白质以促进HNSCC入侵的假设。Invadopodia是分泌降解细胞外基质(ECM)的基质金属蛋白酶的特化亚细胞突起。通过比较DCLK1对照和shDCLK1条件的综合蛋白质组分析,我们的研究结果表明,DCLK1在调节细胞骨架和ECM重塑的蛋白质中起着关键作用,有助于细胞入侵。Further,我们在TCGA数据集中证明DCLK1水平与组织学分级和淋巴结转移增加相关.我们发现DCLK1在HNSCC组织的前缘有较高的表达。在HNSCC中敲除DCLK1减少了invadopodia的数量,细胞粘附和集落形成。使用超分辨率显微镜,我们证明了DCLK1在侵袭足中的定位以及与成熟的侵袭足病标记TKS4,TKS5,cortactin和MT1-MMP的共定位。我们进行了磷酸蛋白质组学,并使用免疫荧光和邻近连接试验进行了验证,DCLK1与运动蛋白KIF16B之间的相互作用。药物抑制或敲低DCLK1减少与KIF16B的相互作用,MMPs的分泌,和细胞入侵。这项研究揭示了DCLK1在invadopodia中的新功能,以调节基质降解货物的贩运。这项工作强调了靶向DCLK1抑制局部区域入侵的影响,HNSCC的威胁生命的属性。
    Head and neck squamous cell carcinoma (HNSCC) is a major health concern due to its high mortality from poor treatment responses and locoregional tumor invasion into life sustaining structures in the head and neck. A deeper comprehension of HNSCC invasion mechanisms holds the potential to inform targeted therapies that may enhance patient survival. We previously reported that doublecortin like kinase 1 (DCLK1) regulates invasion of HNSCC cells. Here, we tested the hypothesis that DCLK1 regulates proteins within invadopodia to facilitate HNSCC invasion. Invadopodia are specialized subcellular protrusions secreting matrix metalloproteinases that degrade the extracellular matrix (ECM). Through a comprehensive proteome analysis comparing DCLK1 control and shDCLK1 conditions, our findings reveal that DCLK1 plays a pivotal role in regulating proteins that orchestrate cytoskeletal and ECM remodeling, contributing to cell invasion. Further, we demonstrate in TCGA datasets that DCLK1 levels correlate with increasing histological grade and lymph node metastasis. We identified higher expression of DCLK1 in the leading edge of HNSCC tissue. Knockdown of DCLK1 in HNSCC reduced the number of invadopodia, cell adhesion and colony formation. Using super resolution microscopy, we demonstrate localization of DCLK1 in invadopodia and colocalization with mature invadopodia markers TKS4, TKS5, cortactin and MT1-MMP. We carried out phosphoproteomics and validated using immunofluorescence and proximity ligation assays, the interaction between DCLK1 and motor protein KIF16B. Pharmacological inhibition or knockdown of DCLK1 reduced interaction with KIF16B, secretion of MMPs, and cell invasion. This research unveils a novel function of DCLK1 within invadopodia to regulate the trafficking of matrix degrading cargo. The work highlights the impact of targeting DCLK1 to inhibit locoregional invasion, a life-threatening attribute of HNSCC.
    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: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)

公众号