podosomes

Podosomes
  • 文章类型: Journal Article
    基质硬度在各种生物学环境中有效促进恶性表型。因此,鉴定参与机械力信号传导到下游生化信号传导的基因表达将大大有助于鼻咽癌(NPC)治疗的进展.在本研究中,我们检测到cortactin(CTTN)在基质刚度诱导的细胞迁移中起着不可或缺的作用,入侵,和invadopodia形成。癌症研究的进展突出表明,失调的可变剪接作为致癌驱动因素有助于癌症进展。然而,WT-CTTN或剪接变体(SV1-CTTN或SV2-CTTN)是否调节基质僵硬度诱导的恶性表型尚不清楚.我们证明WT-CTTN表达的改变调节了基质刚度诱导的细胞迁移,入侵,和invadopodia形成。考虑到剪接因素可能通过正反馈循环驱动癌症进展,我们分析并展示了剪接因子PTBP2和TIA1如何调节WT-CTTN的产生。此外,我们确定高刚度激活PTBP2表达。一起来看,我们的发现表明,PTBP2-WT-CTTN水平在硬化后增加,然后促进细胞迁移,入侵,和NPC中的invadadopodia形成。
    Matrix stiffness potently promotes the malignant phenotype in various biological contexts. Therefore, identification of gene expression to participate in mechanical force signals transduced into downstream biochemical signaling will contribute substantially to the advances in nasopharyngeal carcinoma (NPC) treatment. In the present study, we detected that cortactin (CTTN) played an indispensable role in matrix stiffness-induced cell migration, invasion, and invadopodia formation. Advances in cancer research have highlighted that dysregulated alternative splicing contributes to cancer progression as an oncogenic driver. However, whether WT-CTTN or splice variants (SV1-CTTN or SV2-CTTN) regulate matrix stiffness-induced malignant phenotype is largely unknown. We proved that alteration of WT-CTTN expression modulated matrix stiffness-induced cell migration, invasion, and invadopodia formation. Considering that splicing factors might drive cancer progression through positive feedback loops, we analyzed and showed how the splicing factor PTBP2 and TIA1 modulated the production of WT-CTTN. Moreover, we determined that high stiffness activated PTBP2 expression. Taken together, our findings showed that the PTBP2-WT-CTTN level increases upon stiffening and then promotes cell migration, invasion, and invadopodia formation in NPC.
    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
    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
    选择性剪接调节基因表达和功能多样性,并且在人类癌症中经常失调。这里,我们发现长链非编码RNA(lncRNA)MIR99AHG调节可变剪接以改变染色质重塑剂的活性并促进结直肠癌(CRC)的转移行为.MIR99AHG在患者的侵袭性CRC细胞和转移性肿瘤中含量丰富,并促进培养的CRC细胞的运动和侵袭。MIR99AHG结合并稳定了RNA剪接因子PTBP1,并且该复合物增加了编码染色质重塑基因SMARCA1的mRNA中的盒外显子内含物。具体来说,MIR99AHG改变了PTBP1与SMARCA1前mRNA内含子12上剪接位点结合的性质,从而触发从跳跃到包括外显子13的拼接转换以产生长的同工型,SMARCA1-L.SMARCA1,但不是SMARCA1-L,抑制了侵入足形成,细胞迁移,和入侵。对CRC样本的分析显示,MIR99AHG转录本的丰度与SMARCA1-LmRNA和PTBP1蛋白的丰度呈正相关,并且与CRC患者的不良预后有关。此外,来自癌症相关成纤维细胞的TGF-β1分泌增加了CRC细胞中MIR99AHG的表达。我们的发现确定了一种lncRNA,该lncRNA由肿瘤微环境的线索诱导,并与PTBP1相互作用以调节可变剪接,可能为转移性CRC提供治疗靶标和预测性生物标志物。
    Alternative splicing regulates gene expression and functional diversity and is often dysregulated in human cancers. Here, we discovered that the long noncoding RNA (lncRNA) MIR99AHG regulated alternative splicing to alter the activity of a chromatin remodeler and promote metastatic behaviors in colorectal cancer (CRC). MIR99AHG was abundant in invasive CRC cells and metastatic tumors from patients and promoted motility and invasion in cultured CRC cells. MIR99AHG bound to and stabilized the RNA splicing factor PTBP1, and this complex increased cassette exon inclusion in the mRNA encoding the chromatin remodeling gene SMARCA1. Specifically, MIR99AHG altered the nature of PTBP1 binding to the splice sites on intron 12 of SMARCA1 pre-mRNA, thereby triggering a splicing switch from skipping to including exon 13 to produce the long isoform, SMARCA1-L. SMARCA1, but not SMARCA1-L, suppressed invadopodia formation, cell migration, and invasion. Analysis of CRC samples revealed that the abundance of MIR99AHG transcript positively correlated with that of SMARCA1-L mRNA and PTBP1 protein and with poor prognosis in patients with CRC. Furthermore, TGF-β1 secretion from cancer-associated fibroblasts increased MIR99AHG expression in CRC cells. Our findings identify an lncRNA that is induced by cues from the tumor microenvironment and that interacts with PTBP1 to regulate alternative splicing, potentially providing a therapeutic target and predictive biomarker for metastatic CRC.
    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
    免疫细胞,如巨噬细胞和树突状细胞,可以利用podosomes,机械敏感的富含肌动蛋白的突起,为了产生力量,migrate,并巡逻外来抗原。个体podosome通过周期性的突起和收缩周期(高度振荡)探测其微环境,而集群中多个podosome的振荡以波状方式协调。然而,控制个体振荡和集体波状动力学的机制仍不清楚。这里,通过整合肌动蛋白聚合,肌球蛋白收缩性,肌动蛋白扩散,和机械敏感信号,我们开发了一个化学力学模型,用于集群中的podosome动力学。我们的模型表明,当肌动蛋白聚合驱动的突起和信号相关的肌球蛋白收缩以相似的速率发生时,足细胞体显示振荡生长,而肌动蛋白单体的扩散驱动波浪状协调的足体振荡。我们的理论预测通过不同的药理学处理和微环境刚度对化学机械波的影响得到了验证。我们提出的框架可以阐明在伤口愈合和癌症免疫疗法的背景下,囊体在免疫细胞机械传感中的作用。
    Immune cells, such as macrophages and dendritic cells, can utilize podosomes, mechanosensitive actin-rich protrusions, to generate forces, migrate, and patrol for foreign antigens. Individual podosomes probe their microenvironment through periodic protrusion and retraction cycles (height oscillations), while oscillations of multiple podosomes in a cluster are coordinated in a wave-like fashion. However, the mechanisms governing both the individual oscillations and the collective wave-like dynamics remain unclear. Here, by integrating actin polymerization, myosin contractility, actin diffusion, and mechanosensitive signaling, we develop a chemo-mechanical model for podosome dynamics in clusters. Our model reveals that podosomes show oscillatory growth when actin polymerization-driven protrusion and signaling-associated myosin contraction occur at similar rates, while the diffusion of actin monomers drives wave-like coordination of podosome oscillations. Our theoretical predictions are validated by different pharmacological treatments and the impact of microenvironment stiffness on chemo-mechanical waves. Our proposed framework can shed light on the role of podosomes in immune cell mechanosensing within the context of wound healing and cancer immunotherapy.
    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
    吞噬体,人类粒细胞无形体病(HGA)的病因,是一种专性细胞内革兰氏阴性细菌。在感染期间,A.吞噬细胞增强嗜中性粒细胞对感染的内皮细胞的粘附。然而,导致这种现象的细菌因素仍然未知。在这项研究中,我们表征了吞噬细胞菌的IV型分泌系统底物,AFAP(一种与肌动蛋白丝相关的吞噬细胞无性体蛋白),发现它动态地改变了其在细胞中的模式和亚细胞位置,并增强了细胞粘附。串联亲和纯化结合质谱鉴定宿主核仁素为AFAP相互作用蛋白。进一步的研究表明,RNA干扰对核仁素的破坏,和核仁素结合DNA适体AS1411的治疗减弱了AFAP介导的细胞粘附,表明AFAP以核仁素依赖性方式增强细胞粘附。细胞粘附增强AFAP的表征和宿主核仁素作为其相互作用伙伴的鉴定可能有助于理解吞噬细胞促进细胞粘附的潜在机制。有助于阐明HGA的发病机制。
    Anaplasma phagocytophilum, the aetiologic agent of human granulocytic anaplasmosis (HGA), is an obligate intracellular Gram-negative bacterium. During infection, A. phagocytophilum enhances the adhesion of neutrophils to the infected endothelial cells. However, the bacterial factors contributing to this phenomenon remain unknown. In this study, we characterized a type IV secretion system substrate of A. phagocytophilum, AFAP (an actin filament-associated Anaplasma phagocytophilum protein) and found that it dynamically changed its pattern and subcellular location in cells and enhanced cell adhesion. Tandem affinity purification combined with mass spectrometry identified host nucleolin as an AFAP-interacting protein. Further study showed the disruption of nucleolin by RNA interference, and the treatment of a nucleolin-binding DNA aptamer AS1411 attenuated AFAP-mediated cell adhesion, indicating that AFAP enhanced cell adhesion in a nucleolin-dependent manner. The characterization of cell adhesion-enhancing AFAP and the identification of host nucleolin as its interaction partner may help understand the mechanism underlying A. phagocytophilum-promoting cell adhesion, facilitating the elucidation of HGA pathogenesis.
    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)

  • 文章类型: Journal Article
    随着年龄的增长,骨组织在组成上经历了显著的改变,architecture,和代谢功能,可能导致老年性骨质疏松症。骨质疏松是骨疾病的绝大部分,并与骨量减少和骨折风险增加有关。骨丢失是由于成骨细胞诱导的骨形成和破骨细胞诱导的骨吸收的病症。作为一种独特的骨吸收细胞类型,成熟的骨吸收破骨细胞表现出动态的基于肌动蛋白的细胞骨架结构,称为podosome,参与细胞-基质粘附,专门用于矿化骨基质的降解。Podosomes共享许多与粘着斑相同的分子组成,但是它们有独特的结构组织,具有富含F-肌动蛋白的中心核心,并被支架蛋白包围,激酶和整合素。这里,我们总结了我们对podosome的体系结构和功能的了解的最新进展。我们还讨论了破骨细胞囊体中的调节途径,为今后对破骨细胞囊体的研究提供参考,并将囊体作为抑制骨吸收的治疗靶点。
    With increasing age, bone tissue undergoes significant alterations in composition, architecture, and metabolic functions, probably causing senile osteoporosis. Osteoporosis possess the vast majority of bone disease and associates with a reduction in bone mass and increased fracture risk. Bone loss is on account of the disorder in osteoblast-induced bone formation and osteoclast-induced bone resorption. As a unique bone resorptive cell type, mature bone-resorbing osteoclasts exhibit dynamic actin-based cytoskeletal structures called podosomes that participate in cell-matrix adhesions specialized in the degradation of mineralized bone matrix. Podosomes share many of the same molecular constitutions as focal adhesions, but they have a unique structural organization, with a central core abundant in F-actin and encircled by scaffolding proteins, kinases and integrins. Here, we conclude recent advancements in our knowledge of the architecture and the functions of podosomes. We also discuss the regulatory pathways in osteoclast podosomes, providing a reference for future research on the podosomes of osteoclasts and considering podosomes as a therapeutic target for inhibiting bone resorption.
    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
    Invadopodia是富含肌动蛋白的膜突起,在癌症转移过程中消化基质屏障。自从发现invadopodia以来,它们在2D矩阵上的不同类型的癌细胞中被可视化为局部和点状结构。在这项调查爱泼斯坦-巴尔病毒(EBV)相关的鼻咽癌(NPC),经常伴有颈部淋巴结和远端器官转移的高侵袭性癌症,我们揭示了一种具有动员功能的新形式。活细胞成像和分子分析的整合揭示了巨噬细胞释放的TNFα和EBV编码的潜伏膜蛋白1(LMP1)在共同激活EGFR/Src/ERK/cortactin和Cdc42/N-WASP信号传导轴以通过横向运动动员内足。这种现象赋予invadopodia巨大的退化能力,可视化为2D明胶上的焦点点状消化图案向树枝状消化图案的转变。值得注意的是,LMP1或TNFα的单一刺激只能增加普通的点状侵入足病的数量,这表明,当遇到富含TNFα的肿瘤微环境时,EBV感染会使NPC细胞敏感,从而形成动员性侵入足病。这项研究揭示了EBV和基质成分在通过释放invadopodia克服基质障碍来驱动NPC的侵入潜力中的相互作用。©2022作者由JohnWiley&SonsLtd代表英国和爱尔兰病理学会出版的病理学杂志。
    Invadopodia are actin-rich membrane protrusions that digest the matrix barrier during cancer metastasis. Since the discovery of invadopodia, they have been visualized as localized and dot-like structures in different types of cancer cells on top of a 2D matrix. In this investigation of Epstein-Barr virus (EBV)-associated nasopharyngeal carcinoma (NPC), a highly invasive cancer frequently accompanied by neck lymph node and distal organ metastases, we revealed a new form of invadopodium with mobilizing features. Integration of live-cell imaging and molecular assays revealed the interaction of macrophage-released TNFα and EBV-encoded latent membrane protein 1 (LMP1) in co-activating the EGFR/Src/ERK/cortactin and Cdc42/N-WASP signaling axes for mobilizing the invadopodia with lateral movements. This phenomenon endows the invadopodia with massive degradative power, visualized as a shift of focal dot-like digestion patterns on a 2D gelatin to a dendrite-like digestion pattern. Notably, single stimulation of either LMP1 or TNFα could only enhance the number of ordinary dot-like invadopodia, suggesting that the EBV infection sensitizes the NPC cells to form mobilizing invadopodia when encountering a TNFα-rich tumor microenvironment. This study unveils the interplay of EBV and stromal components in driving the invasive potential of NPC via unleashing the propulsion of invadopodia in overcoming matrix hurdles. © 2022 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
    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