Ovarian cancer metastasis

卵巢癌转移
  • 文章类型: Journal Article
    背景:尽管细胞减灭术后辅助化疗作为早期卵巢癌的标准治疗方法是有效的,大多数卵巢癌病例在晚期被诊断为扩散到腹膜腔,导致预后不良。因此,了解转移的细胞和分子机制以及确定新的治疗靶点至关重要。
    目的:在本研究中,我们旨在阐明在获得转移潜能过程中基因表达改变的潜在机制,并表征卵巢癌细胞中的转移亚群.
    方法:我们对两种人卵巢癌细胞系进行了单细胞RNA测序:SKOV-3和SKOV-3-13,一种高度转移的SKOV-3亚克隆。通过siRNA介导的敲低和CRISPR-Cas9介导的敲除进行NFE2L1表达的抑制。
    结果:聚类和伪时间轨迹分析揭示了这些细胞内的促转移亚群。此外,基因集富集分析和预后分析显示,NFE2L1可能是获得转移潜能的关键转录因子。NFE2L1的抑制显著降低两种细胞的迁移和活力。此外,NFE2L1敲除细胞在小鼠异种移植模型中表现出显著降低的肿瘤生长,计算机模拟和体外结果。
    结论:本研究的结果加深了我们对卵巢癌转移的分子发病机制的理解,最终目标是在转移前开发针对前转移亚克隆的治疗方法。
    Although cytoreductive surgery followed by adjuvant chemotherapy is effective as a standard treatment for early-stage ovarian cancer, the majority of ovarian cancer cases are diagnosed at the advanced stages with dissemination to the peritoneal cavity, leading to a poor prognosis. Therefore, it is crucial to understand the cellular and molecular mechanisms underlying metastasis and identify novel therapeutic targets.
    In this study, we aimed to elucidate the mechanisms underlying gene expression alterations during the acquisition of metastatic potential and characterize the metastatic subpopulations within ovarian cancer cells.
    We conducted single-cell RNA sequencing of two human ovarian cancer cell lines: SKOV-3 and SKOV-3-13, a highly metastatic subclone of SKOV-3. Suppression of NFE2L1 expression was performed through siRNA-mediated knockdown and CRISPR-Cas9-mediated knockout.
    Clustering and pseudotime trajectory analysis revealed pro-metastatic subpopulation within these cells. Furthermore, gene set enrichment analysis and prognosis analysis indicated that NFE2L1 could be a key transcription factor in the acquisition of metastasis potential. Inhibition of NFE2L1 significantly reduced migration and viability of both cells. In addition, NFE2L1 knockout cells exhibited significantly reduced tumor growth in a mouse xenograft model, recapitulating in silico and in vitro results.
    The results presented in this study deepen our understanding of the molecular pathogenesis of ovarian cancer metastasis with the ultimate goal of developing treatments targeting pro-metastatic subclones prior to metastasis.
    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
    广泛的腹膜转移是妇科恶性肿瘤中卵巢癌致死率最高的原因。腹水在卵巢癌转移中起关键作用,但涉及的机制尚不确定。这里,我们进行了腹水的定量蛋白质组学,发现与正常腹膜液相比,上皮性卵巢癌患者的腹水中I型胶原α1(COL1A1)明显升高,并证实COL1A1升高主要来源于成纤维细胞。COL1A1促进卵巢癌细胞的迁移和侵袭,但此类效应被COL1A1抗体部分消除.腹腔注射COL1A1可加速NOD/SCID小鼠卵巢癌移植瘤的腹腔转移。Further,COL1A1通过结合膜表面受体整联蛋白β1(ITGB1)激活下游AKT磷酸化。敲除或阻断ITGB1逆转COL1A1增强卵巢癌细胞的迁移和侵袭。相反,卵巢癌腹水和纤维蛋白原促进成纤维细胞分泌COL1A1。腹水中纤维蛋白原的升高可能与卵巢癌引起的血管通透性增加有关。我们的研究结果表明,肿瘤细胞和基质细胞重塑的微环境促进成纤维细胞分泌COL1A1并促进卵巢癌的转移,这可能为卵巢癌的治疗提供新的途径。
    Wide peritoneal metastasis is the cause of the highest lethality of ovarian cancer in gynecologic malignancies. Ascites play a key role in ovarian cancer metastasis, but involved mechanism is uncertain. Here, we performed a quantitative proteomics of ascites, and found that collagen type I alpha 1 (COL1A1) was notably elevated in ascites from epithelial ovarian cancer patients compared to normal peritoneal fluids, and verified that elevated COL1A1 was mainly originated from fibroblasts. COL1A1 promoted migration and invasion of ovarian cancer cells, but such effects were partially eliminated by COL1A1 antibodies. Intraperitoneally injected COL1A1 accelerated intraperitoneal metastasis of ovarian cancer xenograft in NOD/SCID mice. Further, COL1A1 activated downstream AKT phosphorylation by binding to membrane surface receptor integrin β1 (ITGB1). Knockdown or blockage of ITGB1 reversed COL1A1 enhanced migration and invasion in ovarian cancer cells. Conversely, ovarian cancer ascites and fibrinogen promoted fibroblasts to secrete COL1A1. Elevated fibrinogen in ascites might be associated with increased vascular permeability induced by ovarian cancer. Our findings suggest that microenvironment remodeled by tumor cells and stromal cells promotes fibroblasts to secrete COL1A1 and facilitates the metastasis of ovarian cancer, which may provide a new approach for ovarian cancer therapeutics.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    背景:我们旨在开发可以防止SOS1/EPS8/ABI1三复合物蛋白质相互作用的抑制性短肽,卵巢癌转移的关键成分。
    方法:将含有HA标记的ABI1的各种区域的质粒与Flag标记的SOS1或Myc标记的EPS8共转染到卵巢癌细胞中。使用免疫共沉淀和GST下拉测定来鉴定负责SOS1和EPS8结合的ABI1区域。合成这些结合区的抑制性短肽并用HIV-TAT序列修饰。通过GST-pulldown测定法测定肽在体外和体内对ABI1-SOS1或ABI1-EPS8相互作用的阻断作用。通过Matrigel侵袭测定和腹膜转移定植测定测试了这些短肽抑制卵巢癌细胞侵袭和转移的能力。
    结果:在LPA诱导的卵巢癌细胞侵袭事件中检测到内源性SOS1/EPS8/ABI1三复合物的形成。在三复合体中,ABI1充当将SOS1和EPS8保持在一起的支架蛋白。ABI1的SH3和多聚脯氨酸+PxxDY区负责SOS1和EPS8的结合,分别。抑制性短肽p-8(ppppppppppvdyedee)和SH3-3(ekvvaiydytkdkdkddelsfmegaii)可以在体外阻断ABI1-SOS1和ABI1-EPS8的相互作用。TAT-p+p-8肽可在体内破坏ABI1-EPS8相互作用并抑制卵巢癌细胞的侵袭和转移。
    结论:TAT-p+p-8肽能有效破坏ABI1-EPS8相互作用,三配合物的形成,并阻断卵巢癌细胞的侵袭和转移。
    BACKGROUND: We aimed to develop inhibitory short peptides that can prevent protein interactions of SOS1/EPS8/ABI1 tri-complex, a key component essential for ovarian cancer metastasis.
    METHODS: Plasmids containing various regions of HA-tagged ABI1 were co-transfected into ovarian cancer cells with Flag-tagged SOS1 or Myc-tagged EPS8. Co-immunoprecipitation and GST-pulldown assay were used to identify the regions of ABI1 responsible for SOS1 and EPS8 binding. Inhibitory short peptides of these binding regions were synthesized and modified with HIV-TAT sequence. The blocking effects of the peptides on ABI1-SOS1 or ABI1-EPS8 interactions in vitro and in vivo were determined by GST-pulldown assay. The capability of these short peptides in inhibiting invasion and metastasis of ovarian cancer cell was tested by Matrigel invasion assay and peritoneal metastatic colonization assay.
    RESULTS: The formation of endogenous SOS1/EPS8/ABI1 tri-complex was detected in the event of LPA-induced ovarian cancer cell invasion. In the tri-complex, ABI1 acted as a scaffold protein holding together SOS1 and EPS8. The SH3 and poly-proline+PxxDY regions of ABI1 were responsible for SOS1 and EPS8 binding, respectively. Inhibitory short peptides p + p-8 (ppppppppvdyedee) and SH3-3 (ekvvaiydytkdkddelsfmegaii) could block ABI1-SOS1 and ABI1-EPS8 interaction in vitro. TAT-p + p-8 peptide could disrupt ABI1-EPS8 interaction and suppress the invasion and metastasis of ovarian cancer cells in vivo.
    CONCLUSIONS: TAT-p + p-8 peptide could efficiently disrupt the ABI1-EPS8 interaction, tri-complex formation, and block the invasion and metastasis of ovarian cancer cells.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    卵巢癌是一种极其致命的妇科疾病;高级别浆液亚型主要与低生存率相关。缺乏早期诊断生物标志物和治疗后复发的患病率,在治疗卵巢癌方面存在重大挑战。这些癌症的特征还在于高度异质性和长期转移。进一步复杂的治疗。在卵巢肿瘤微环境中,肿瘤干细胞样细胞和机械刺激是两个未被重视的关键因素,在促进这些结果方面发挥着至关重要的作用.在这篇评论文章中,我们强调了它们在调节卵巢癌转移中的作用。具体来说,我们概述了癌症干细胞样细胞的临床意义,以及与它们的鉴定和表征相关的挑战,并总结它们调节卵巢癌转移的方式。Further,我们回顾了卵巢肿瘤微环境中的机械线索,包括,紧张,剪力,压缩和基体刚度,影响(癌症干细胞样细胞和)卵巢癌转移。最后,我们概述了与探索卵巢癌转移的这些重要调节剂相关的挑战,并为将这些线索纳入关注转移的基础生物学和转化研究提供了建议。我们得出结论,未来对卵巢癌转移的研究将受益于对机械刺激和癌症干细胞的仔细考虑,最终允许开发更有效的疗法。
    Ovarian cancer is an extremely lethal gynecologic disease; with the high-grade serous subtype predominantly associated with poor survival rates. Lack of early diagnostic biomarkers and prevalence of post-treatment recurrence, present substantial challenges in treating ovarian cancers. These cancers are also characterized by a high degree of heterogeneity and protracted metastasis, further complicating treatment. Within the ovarian tumor microenvironment, cancer stem-like cells and mechanical stimuli are two underappreciated key elements that play a crucial role in facilitating these outcomes. In this review article, we highlight their roles in modulating ovarian cancer metastasis. Specifically, we outline the clinical relevance of cancer stem-like cells, and challenges associated with their identification and characterization and summarize the ways in which they modulate ovarian cancer metastasis. Further, we review the mechanical cues in the ovarian tumor microenvironment, including, tension, shear, compression and matrix stiffness, that influence (cancer stem-like cells and) metastasis in ovarian cancers. Lastly, we outline the challenges associated with probing these important modulators of ovarian cancer metastasis and provide suggestions for incorporating these cues in basic biology and translational research focused on metastasis. We conclude that future studies on ovarian cancer metastasis will benefit from the careful consideration of mechanical stimuli and cancer stem cells, ultimately allowing for the development of more effective therapies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    卵巢癌是美国女性癌症死亡的第五大原因,也是最致命的妇科恶性肿瘤。这种致死率主要是由于大多数病例在预后不良时被诊断为疾病的转移阶段。流行病学研究一致表明,产妇患卵巢癌的风险降低,更多的新生儿提供了更大的保护;然而,关于胎次对卵巢癌转移的影响知之甚少。在这里,我们报道了在年龄匹配的同基因鼠同种异体移植模型中,多胎小鼠对卵巢癌转移的敏感性较低。发现干扰素途径在多胎小鼠的健康脂肪组织中上调,提示了多胎相关的抗转移保护作用的可能机制。这种保护作用随着年龄的增长而消失。基于这项工作,未来的研究探索治疗策略,利用这里证明的多产性相关保护作用是有必要的.
    Ovarian cancer is the fifth leading cause of cancer deaths in U.S. women and the deadliest gynecologic malignancy. This lethality is largely due to the fact that most cases are diagnosed at metastatic stages of the disease when the prognosis is poor. Epidemiologic studies consistently demonstrate that parous women have a reduced risk of developing ovarian cancer, with a greater number of births affording greater protection; however little is known about the impact of parity on ovarian cancer metastasis. Here we report that multiparous mice are less susceptible to ovarian cancer metastasis in an age-matched syngeneic murine allograft model. Interferon pathways were found to be upregulated in healthy adipose tissue of multiparous mice, suggesting a possible mechanism for the multiparous-related protective effect against metastasis. This protective effect was found to be lost with age. Based on this work, future studies exploring therapeutic strategies which harness the multiparity-associated protective effect demonstrated here are warranted.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    尽管有化疗和手术切除的选择,卵巢癌经常复发和传播,预后不良。然而,卵巢癌转移的分子机制仍未阐明.肿瘤微环境,由基质细胞(包括成纤维细胞,巨噬细胞,调节性T细胞,骨髓来源的抑制细胞,内皮细胞,周细胞和血小板),细胞外基质成分(EMC)(包括炎性细胞因子,趋化因子,基质金属蛋白酶,整合素,和其他分泌分子)和外来体(载有分子的小细胞外囊泡),建立自分泌-旁分泌通信电路,通过相互信号增强侵袭和癌细胞转移。最近的证据揭示了肿瘤微环境对卵巢癌转移的重要贡献。在这次审查中,我们提供了肿瘤基质和卵巢癌细胞之间相互转移的综合景观,旨在为未来临床实践中卵巢癌新型诊断生物标志物和治疗靶点的开发提供新线索。
    Despite chemotherapy and surgical debulking options, ovarian cancer recurs and disseminates frequently, with poor prognosis. However, the molecular mechanisms underlying ovarian cancer metastasis still remain unelucidated. The tumor microenvironment, consisting of stromal cells (including fibroblasts, macrophages, regulatory T cells, myeloid-derived suppressor cells, endothelial cells, pericytes and platelets), the extracellular matrix component (EMC) (including inflammatory cytokines, chemokines, matrix metalloproteinases, integrins, and other secreted molecules) and exosomes (small extracellular vesicles loaded with molecules), establishes an autocrine-paracrine communication circuit that reinforces invasion and cancer cell metastasis via reciprocal signaling. Recent evidences have unraveled the significant contribution of tumor microenvironment to ovarian cancer metastasis. In this review, we provide a comprehensive landscape of the reciprocity between tumor stroma and ovarian cancer cells upon metastasis, aiming to offer novel clues on the development of novel diagnostic biomarkers and therapeutic targets for ovarian cancer in future clinical practice.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    The current study presents a case of sarcoidosis manifesting as hepatic and splenic nodules, which was difficult to differentiate from ovarian cancer metastases. A 24-year-old female, who was previously diagnosed with right ovarian cancer and underwent surgery at the age of 21, was found to have two nodules in the spleen revealed by contrast-enhanced computed tomography (CT). 18F-fluorodeoxyglucose positron emission tomography/CT revealed two abnormal high uptake lesions in the spleen and one abnormal high uptake lesion in the liver. Under a diagnosis of hepatic and splenic metastases from right ovarian cancer, a laparoscopic splenectomy and partial hepatectomy were performed. Histopathological examination showed that a large number of non-caseating epithelioid cell granulomas formed these nodules, which was compatible with sarcoidosis. This case indicates that it is difficult to distinguish sarcoidosis from metastatic disease even using the latest modalities, and that laparoscopic surgery is a minimally invasive and useful tool for forming a differential diagnosis.
    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
    间皮细胞在卵巢癌腹腔内散布中的感化至今仍难以捉摸。特别是,目前尚不清楚这些细胞是否构成阻止癌细胞进展的被动屏障,或者可能是该过程的主动启动子。在此报告中,我们显示了网膜人腹膜间皮细胞(HPMC)刺激卵巢癌细胞(A2780,OVCAR-3,SKOV-3)的粘附和增殖。后者与HPMC释放到环境中的GRO-1,IL-6和IL-8的旁分泌活性有关。此外,腹膜内注射卵巢癌细胞和HPMC产生的卵巢癌异种移植物的生长动力学明显高于单独植入癌细胞。在每个异种移植模型中,肿瘤块附近始终存在一层腹膜间皮。总之,我们的结果表明,HPMC在卵巢恶性肿瘤的腹膜内侵袭中起着辅助作用,其作用可能归因于它们刺激癌细胞粘附和增殖的能力。
    The role of mesothelial cells in the intraperitoneal spread of ovarian cancer is still elusive. In particular, it is unclear whether these cells constitute a passive barrier preventing cancer cell progression or perhaps act as an active promoter of this process. In this report we show that omental human peritoneal mesothelial cells (HPMCs) stimulate adhesion and proliferation of ovarian cancer cells (A2780, OVCAR-3, SKOV-3). The latter was associated with the paracrine activity of GRO-1, IL-6, and IL-8 released to the environment by HPMCs. Furthermore, the growth dynamics of ovarian cancer xenografts produced in response to i.p. injection of ovarian cancer cells together with HPMCs was remarkably greater than for implantation of cancer cells alone. A layer of peritoneal mesothelium was consistently present in close proximity to the tumor mass in every xenograft model. In conclusion, our results indicate that HPMCs play a supporting role in the intraperitoneal invasiveness of ovarian malignancy, whose effect may be attributed to their ability to stimulate adhesion and proliferation of cancer cells.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    尽管目前局部卵巢癌的治疗非常有效,这种癌症仍然是最致命的妇科恶性肿瘤,主要是由于它通常仅在肿瘤细胞离开原发性肿瘤后才被检测到。长期以来,临床医生一直注意到卵巢癌转移到软网膜的明确倾向。这里,我们表明,这种取向不仅是由于化学信号,而且机械线索。转移性卵巢癌细胞(OCC)优先粘附于软微环境并表现出增强的恶性表型,包括增加的移民,增殖和化学抗性。要了解用于感测底物刚性的细胞-基质相互作用,我们利用牵引力显微镜(TFM)发现,在柔软的基材上,人类OCC增加了牵引力的大小及其极化程度。在软基质上培养后,细胞经历了上皮-间质转化(EMT)的形态伸长特征,分子分析证实了这一点。与机械线索是卵巢癌扩散的关键决定因素的观点一致,在转移性较小的OCC中,观察到的机械敏感性大大降低.最后,我们证明了这种机械取向是通过Rho-ROCK信号通路控制的。
    Although current treatments for localized ovarian cancer are highly effective, this cancer still remains the most lethal gynecological malignancy, largely owing to the fact that it is often detected only after tumor cells leave the primary tumor. Clinicians have long noted a clear predilection for ovarian cancer to metastasize to the soft omentum. Here, we show that this tropism is due not only to chemical signals but also mechanical cues. Metastatic ovarian cancer cells (OCCs) preferentially adhere to soft microenvironments and display an enhanced malignant phenotype, including increased migration, proliferation and chemoresistance. To understand the cell-matrix interactions that are used to sense the substrate rigidity, we utilized traction force microscopy (TFM) and found that, on soft substrates, human OCCs increased both the magnitude of traction forces as well as their degree of polarization. After culture on soft substrates, cells underwent morphological elongation characteristic of epithelial-to-mesenchymal transition (EMT), which was confirmed by molecular analysis. Consistent with the idea that mechanical cues are a key determinant in the spread of ovarian cancer, the observed mechanosensitivity was greatly decreased in less-metastatic OCCs. Finally, we demonstrate that this mechanical tropism is governed through a Rho-ROCK 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: Case Reports
    背景。伴有乳腺转移的原发性卵巢癌是罕见的,目前文献中只有39例报道。卵巢转移到乳腺表现为炎性乳腺癌甚至更罕见,报告只有6例。案例。我们介绍了一名患者,该患者在最初诊断后约1年从IIIC期乳头状浆液性卵巢腺癌发展为乳腺转移性炎性癌。病理分析证实了肿瘤的起源:高级腺癌在形态上与先前诊断的卵巢癌相似。此外,该肿瘤的CA-125免疫组织化学强阳性,与卵巢原发性相同。患者在发现乳腺肿瘤5个月后死于弥漫性转移。结论。虽然卵巢转移到乳腺表现为炎性乳腺癌是罕见的,对于任何有个人卵巢癌病史的患者,应将其纳入鉴别诊断.准确的分化是必要的,因为对于卵巢转移到乳腺的患者,治疗有很大的不同。与原发性炎性乳腺癌患者相比。卵巢转移至乳腺导致不良预后:患者生存期为3至18个月,诊断为乳腺转移后的中位生存期为6个月。
    Background. Primary ovarian carcinoma with metastasis to the breast is rare, with only 39 cases reported in the current literature. Ovarian metastasis to the breast presenting as inflammatory breast carcinoma is even more infrequent, with only 6 cases reported.Case. We present a patient who developed metastatic inflammatory cancer of the breast from a stage IIIC papillary serous ovarian adenocarcinoma approximately 1 year after the original diagnosis. Pathologic analysis confirmed the origin of the tumor: a high-grade adenocarcinoma morphologically similar to the previously diagnosed ovarian cancer. In addition, the tumor was strongly positive on immunohistochemistry for CA-125, identical to the ovarian primary. The patient died of diffuse metastasis 5 months after the breast tumor was noted.Conclusion. Although ovarian metastasis to the breast presenting as inflammatory breast cancer is rare, it should be included in the differential diagnosis for any patient with a personal history of ovarian cancer. Accurate differentiation is necessary because treatment differs significantly for patients with ovarian metastasis to the breast, as compared with patients with primary inflammatory breast cancer. Ovarian metastasis to the breast confers a poor prognosis: patient survival ranged from 3 to 18 months, with a median survival of 6 months after the diagnosis of the breast metastasis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号