podosomes

Podosomes
  • 文章类型: Journal Article
    肺实质缺氧已成为特发性肺纤维化(IPF)的主要特征。缺氧通过依赖于溶血磷脂酸(LPA)受体的信号促进癌细胞的侵袭和转移,LPA1(LPAR1)。大量数据表明LPA1依赖性信号传导也增强IPF中的肺纤维化发生。我们最近报道,从患有IPF的个体的肺中分离的成纤维细胞具有增加的形成亚细胞基质降解结构的能力,称为invadosome。与肺纤维化程度相关的事件。因此,我们假设缺氧通过LPA1依赖性信号传导促进肺成纤维细胞中的侵袭小体形成。这里,已证明,酪氨酸受体激酶抑制剂nintedanib和LPA1的抑制作用都抑制了晚期IPF患者肺成纤维细胞的嵌入体形成。此外,暴露于缺氧或LPA的正常人肺成纤维细胞增加了它们形成包裹体的能力。机械上,研究发现,低氧诱导的肺成纤维细胞侵入体形成涉及LPA1和PDGFR-Akt信号传导.我们得出结论,缺氧通过LPA1和PDGFR-Akt信号轴增加了肺成纤维细胞中侵入体的形成,这代表了抑制肺纤维化的潜在目标。
    Lung parenchymal hypoxia has emerged as a cardinal feature of idiopathic pulmonary fibrosis (IPF). Hypoxia promotes cancer cell invasion and metastasis through signaling that is dependent upon the lysophosphatidic acid (LPA) receptor, LPA1 (LPAR1). Abundant data indicate that LPA1-dependent signaling also enhances lung fibrogenesis in IPF. We recently reported that fibroblasts isolated from the lungs of individuals with IPF have an increased capacity to form subcellular matrix-degradative structures known as invadosomes, an event that correlates with the degree of lung fibrosis. We therefore hypothesized that hypoxia promotes invadosome formation in lung fibroblasts through LPA1-dependent signaling. Here, it is demonstrated that invadosome formation by fibroblasts from the lungs of individuals with advanced IPF is inhibited by both the tyrosine receptor kinase inhibitor nintedanib and inhibition of LPA1. In addition, exposure of normal human lung fibroblasts to either hypoxia or LPA increased their ability to form invadosomes. Mechanistically, the hypoxia-induced invadosome formation by lung fibroblasts was found to involve LPA1 and PDGFR-Akt signaling. We concluded that hypoxia increases the formation of invadosomes in lung fibroblasts through the LPA1 and PDGFR-Akt signaling axis, which represents a potential target for suppressing lung fibrosis.
    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
    Invadosome is an umbrella term used to describe a family of cellular structures including podosomes and invadopodia. They serve as contact zones between the cell plasma membrane and extracellular matrix, contributing to matrix remodeling by locally enriched proteolytic enzymes. Invadosomes, which are actin-dependent, are implicated in cellular processes promoting adhesion, migration, and invasion. Invadosomes, which exist in various cell types, play crucial roles in physiological phenomena such as vascularization and bone resorption. Invadosomes are also implicated in pathological processes such as matrix tissue remodeling during metastatic tumor cell invasion. This review summarizes basic information and recent advances about mechanisms underlying podosome and invadopodia formation, their organization and function.
    UNASSIGNED: Invadosomes - Entre mobilité et invasion, naviguer dans la dualité des fonctions cellulaires.
    UNASSIGNED: Le terme « invadosome » désigne une famille de structures cellulaires, comprenant les podosomes et les invadopodes, qui constituent des zones de contact entre la membrane plasmique des cellules et la matrice extracellulaire. Ces structures contribuent au remodelage de la matrice grâce à un enrichissement local en enzymes protéolytiques qui dégradent ses constituants fibrillaires. Les invadosomes, présents dans des types cellulaires variés, contribuent à des processus physiologiques, tels que la vascularisation, ou pathologiques, comme l’invasion des tissus par les cellules métastatiques.
    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
    对于包括细胞骨架蛋白肌动蛋白和肌球蛋白在内的许多细胞成分,流动或集体运动是经常观察到的现象。为了研究活细胞中的蛋白质流动,我们和其他人以前使用时空图像相关光谱(STICS)分析荧光显微镜图像时间序列。然而,在细胞中,多个蛋白质流通常在不同的尺度上同时发生,导致叠加的荧光强度波动,这对于使用STICS进行分离是具有挑战性的。这里,我们利用了不同的蛋白质流经常发生在图像系列中存在的不同空间尺度上的特征来解开叠加的蛋白质流动力学。我们采用了一种新开发的和已建立的空间滤波算法,以交替地强调或衰减不同空间尺度上的局部图像强度异质性。随后,我们用STICS分析了空间滤波的时间序列,允许量化图像时间序列内的两个不同的叠加流。作为我们分析方法原理的证明,我们使用了模拟的荧光强度波动以及内皮细胞中的非肌肉肌球蛋白II和树突状细胞中的基于肌动蛋白的足细胞的时间序列,并揭示了在这些系统中同时发生的连续和不连续流动动力学.总之,这项工作扩展了STICS在包括肌动球蛋白细胞骨架在内的复杂生物系统中定量多种蛋白质流动动力学的应用。
    Flow or collective movement is a frequently observed phenomenon for many cellular components including the cytoskeletal proteins actin and myosin. To study protein flow in living cells, we and others have previously used spatiotemporal image correlation spectroscopy (STICS) analysis on fluorescence microscopy image time series. Yet, in cells, multiple protein flows often occur simultaneously on different scales resulting in superimposed fluorescence intensity fluctuations that are challenging to separate using STICS. Here, we exploited the characteristic that distinct protein flows often occur at different spatial scales present in the image series to disentangle superimposed protein flow dynamics. We employed a newly developed and an established spatial filtering algorithm to alternatively accentuate or attenuate local image intensity heterogeneity across different spatial scales. Subsequently, we analysed the spatially filtered time series with STICS, allowing the quantification of two distinct superimposed flows within the image time series. As a proof of principle of our analysis approach, we used simulated fluorescence intensity fluctuations as well as time series of nonmuscle myosin II in endothelial cells and actin-based podosomes in dendritic cells and revealed simultaneously occurring contiguous and noncontiguous flow dynamics in each of these systems. Altogether, this work extends the application of STICS for the quantification of multiple protein flow dynamics in complex biological systems including the actomyosin cytoskeleton.
    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
    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
    绝经后雌激素(17β-雌二醇)缺乏会改变骨稳态,从而破骨细胞的骨吸收超过成骨细胞的骨形成。导致女性骨质疏松症。我们建立了一个体外模型,以检查雌激素戒断(E2-WD)对源自小鼠巨噬细胞RAW264.7细胞系的破骨细胞的影响,并利用它来研究绝经后破骨细胞活性增强的机制。我们发现,与连续暴露于雌激素(E2)的破骨细胞相比,接受E2-WD的破骨细胞群体中含有破骨细胞粘附和吸收骨骼所必需的足体带,并且具有升高的吸收活性。我们的结果表明,与连续接受E2的破骨细胞相比,那些接受E2-WD的微管(MT)生长速度更快,减少RhoA激活,和较短的足足寿命。因此,雌激素戒断引起的podosome和MT动力学改变支持破骨细胞的podosome带组装/稳定性,这可以解释它们增强的骨吸收活性。
    Estrogen (17β-estradiol) deficiency post-menopause alters bone homeostasis whereby bone resorption by osteoclasts exceeds bone formation by osteoblasts, leading to osteoporosis in females. We established an in vitro model to examine the consequences of estrogen withdrawal (E2-WD) on osteoclasts derived from the mouse macrophage RAW 264.7 cell line and utilized it to investigate the mechanism behind the enhanced osteoclast activity post-menopause. We found that a greater population of osteoclasts that underwent E2-WD contained a podosome belt necessary for osteoclasts to adhere and resorb bone and possessed elevated resorptive activity compared to osteoclasts exposed to estrogen (E2) continuously. Our results show that compared to osteoclasts that received E2 continuously, those that underwent E2-WD had a faster rate of microtubule (MT) growth, reduced RhoA activation, and shorter podosome lifespan. Thus, altered podosome and MT dynamics induced by the withdrawal of estrogen supports podosome belt assembly/stability in osteoclasts, which may explain their enhanced bone resorption activity.
    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
    成肌是一个多步骤的过程,需要对细胞事件进行时空调节,最终导致成肌细胞融合到多核肌管中。对融合机制的最主要见解似乎从昆虫到哺乳动物都是保守的,包括形成足体样突起(PLPs),对创始人细胞产生驱动力。然而,控制这一过程的机制仍然知之甚少。在这项研究中,我们证明MTM1是负责生产磷脂酰肌醇5-磷酸的主要酶,反过来又为PI5P4-激酶α提供燃料,以产生少量的功能性磷脂酰肌醇4,5-双磷酸酯池,该池浓缩在含有支架蛋白Tks5,Dynamin-2和融合蛋白Myomaker的PLP中。总的来说,我们的数据揭示了在调节PLP形成过程中PI-磷酸酶和PI-激酶之间的功能性串扰。
    Myogenesis is a multistep process that requires a spatiotemporal regulation of cell events resulting finally in myoblast fusion into multinucleated myotubes. Most major insights into the mechanisms underlying fusion seem to be conserved from insects to mammals and include the formation of podosome-like protrusions (PLPs) that exert a driving force toward the founder cell. However, the machinery that governs this process remains poorly understood. In this study, we demonstrate that MTM1 is the main enzyme responsible for the production of phosphatidylinositol 5-phosphate, which in turn fuels PI5P 4-kinase α to produce a minor and functional pool of phosphatidylinositol 4,5-bisphosphate that concentrates in PLPs containing the scaffolding protein Tks5, Dynamin-2, and the fusogenic protein Myomaker. Collectively, our data reveal a functional crosstalk between a PI-phosphatase and a PI-kinase in the regulation of PLP formation.
    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
    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: Journal Article
    在癌症进展期间,肿瘤细胞需要通过重塑肿瘤细胞外基质来传播。最近的一项研究揭示了caveolae和invadosome之间的复杂合作,这种合作促进了癌细胞的扩散。
    During cancer progression, tumor cells need to disseminate by remodeling the extracellular tumor matrix. A recent study sheds light on the intricate cooperation between caveolae and invadosomes that facilitates the spread of cancer cells.
    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