国际妇产科学杂志, 2024, 51(4): 370-374 doi: 10.12280/gjfckx.20240329

妇科肿瘤研究: 综述

外泌体在卵巢癌血管生成中的研究进展

张静怡, 刘东哲, 陈秀慧,

150000 哈尔滨医科大学附属第二医院妇产科(张静怡,陈秀慧);深圳大学总医院血液肿瘤科(刘东哲)

Research Advances of Exosomes in Angiogenesis of Ovarian Cancer

ZHANG Jing-yi, LIU Dong-zhe, CHEN Xiu-hui,

Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150000, China (ZHANG Jing-yi, CHEN Xiu-hui); Department of Hematology and Oncology, Shenzhen University General Hospital, Shenzhen 518055, Guangdong Province, China (LIU Dong-zhe)

通讯作者: 陈秀慧,E-mail:xchen@hrbmu.edu.cn

审校者

责任编辑: 王昕

收稿日期: 2024-04-15  

基金资助: 国家自然科学基金(81802592)
中国博士后科学基金会基金(2018M641854)

Corresponding authors: CHEN Xiu-hui, E-mail:xchen@hrbmu.edu.cn

Received: 2024-04-15  

摘要

在卵巢癌的治疗过程中,肿瘤细胞往往会对化疗药物产生耐药性,导致治疗效果减弱或消失。因此,抗血管生成治疗作为一种重要的维持治疗手段,通过不同的机制与其他治疗方法协同作用,可以有效减慢疾病进展速度。外泌体是由活细胞分泌的细胞外囊泡,携带着多种遗传物质,在肿瘤微环境中起着细胞通信的重要作用。研究发现,肿瘤细胞及其他细胞来源的外泌体可以通过释放非编码RNA、蛋白质等遗传物质影响血管生成及其他肿瘤相关过程。利用外泌体靶向特定蛋白或作为包裹治疗剂的外源性载体,已经成为卵巢癌抗血管生成治疗的重要策略。综述外泌体对卵巢癌血管生成的调控机制,以及其作为抗血管生成剂的作用潜力,以期为新的治疗方法提供理论依据。

关键词: 外泌体; 卵巢肿瘤; 新生血管化; 治疗; 肿瘤微环境

Abstract

In the treatment of ovarian cancer, tumor cells often become resistant to chemotherapy drugs, leading to weakened or ineffective treatment outcomes. Therefore, anti-angiogenesis therapy serves as an important adjunctive treatment, can effectively slow disease progression by synergizing with other treatment modalities through various mechanisms. Exosomes, extracellular vesicles secreted by living cells, carry a variety of genetic materials and play crucial roles in cell communication within the tumor microenvironment. Studies have shown that exosomes derived from tumor cells and other cell sources can influence angiogenesis and other tumor-related processes by releasing non-coding RNAs, proteins, and other genetic materials. Utilizing exosomes to target specific proteins or as exogenous carriers for encapsulate therapeutic agents has become an important strategy for anti-angiogenesis therapy in ovarian cancer. This review summarized the regulatory mechanism of exosomes on angiogenesis in ovarian cancer and it also explores the potential of exosomes as anti-angiogenic agents, providing a theoretical basis for new treatment methods.

Keywords: Exosomes; Ovarian neoplasms; Neovascularization; Therapy; Tumor microenvironment

PDF (743KB) 元数据 多维度评价 相关文章 导出 EndNote| Ris| Bibtex  收藏本文

本文引用格式

张静怡, 刘东哲, 陈秀慧. 外泌体在卵巢癌血管生成中的研究进展[J]. 国际妇产科学杂志, 2024, 51(4): 370-374 doi:10.12280/gjfckx.20240329

ZHANG Jing-yi, LIU Dong-zhe, CHEN Xiu-hui. Research Advances of Exosomes in Angiogenesis of Ovarian Cancer[J]. Journal of International Obstetrics and Gynecology, 2024, 51(4): 370-374 doi:10.12280/gjfckx.20240329

卵巢癌是常见的女性生殖系统肿瘤,可发生于任何年龄。由于早期卵巢癌症状不典型,且缺乏有效的筛查手段,很难获得早期诊断。大多数患者在初次诊断时已处于疾病晚期,因而其致死率居妇科恶性肿瘤首位。血管生成(angiogenesis)是肿瘤生长和转移的重要过程,肿瘤细胞依赖血管的持续生成来获得增殖所必需的氧气和营养,因此抗血管生成成为治疗卵巢癌的重要途径。近十几年来,多种血管生成抑制剂被推荐用于卵巢癌的治疗,然而包括耐药在内的多种问题限制了其临床疗效,亟需进一步研究肿瘤血管生成的相关调控因子。外泌体是由活细胞分泌的携带核酸、蛋白质等多种遗传物质的细胞外囊泡,具有介导细胞通信的功能,在肿瘤的血管生成、侵袭、转移、化疗耐药中发挥重要的调节作用。现就外泌体在卵巢癌血管生成中的研究进展进行综述。

1 外泌体与肿瘤微环境(tumor microenvironment, TME)

TME即肿瘤细胞所在的微环境,主要由细胞外基质、基质细胞(如成纤维细胞、间充质细胞、周细胞、脂肪细胞、血液和淋巴血管网络)和免疫细胞(包括T淋巴细胞、B淋巴细胞、自然杀伤细胞和肿瘤相关巨噬细胞)等构成[1]。细胞通信是肿瘤进展和转移的关键特征。在TME中,除了肿瘤细胞与未来转移部位的远程信号交流外,肿瘤细胞与各种细胞间存在局部信号传导。研究表明,外泌体是介导细胞通信的重要角色[2]

外泌体是一种由活细胞的内体合成的直径30~100 nm的细胞外囊泡,作为载体携带蛋白质、RNA、DNA片段、细胞因子、趋化因子和其他信号分子,通过释放内容物激活或抑制各种信号通路[3]。越来越多的证据表明,癌细胞主动向周围微环境释放外泌体,他们是细胞外通信以及与TME通信的重要参与者[4]。作为一种新型的细胞间通信媒介,TME中的外泌体在调控肿瘤的上皮-间质转化、免疫逃逸、侵袭转移和化疗抵抗等方面发挥着重要的作用[5]。在卵巢肿瘤中,卵巢癌细胞来源的外泌体还可以增强内皮细胞活力和迁移能力,在促进血管生成的同时,推动转移前微环境的形成[6]

2 外泌体与卵巢癌的血管生成

2.1 卵巢癌的血管生成

血管生成是指从预先存在的内皮细胞中形成新的血管,是对组织修复和发育至关重要的生理过程。在TME中,血管生成是肿瘤细胞获得生长和生存所需氧气和营养的重要过程,贯穿肿瘤生长的始终[7]。肿瘤血管生成主要是由促血管生成因子和抗血管生成因子之间的不平衡引起的[8]。这些因子主要包括血管内皮生长因子(vascular endothelial growth factor,VEGF)、血小板衍生生长因子和成纤维细胞生长因子等[9]。与VEGF相关的细胞内信号传导主要包括Janus激酶/信号转导及转录活化因子(Janus kinase/signal transducer and activator of transcription,JAK/STAT)通路成分、磷脂酰肌醇3激酶( phosphoinositide 3-kinase,PI3K)和丝裂原激活的蛋白激酶(mitogen-activated protein kinase,MAPK)等分子。血小板衍生生长因子是周细胞募集的必需蛋白,是促进血管成熟的关键因素。成纤维细胞生长因子可能与VEGF共同分泌到恶性腹水中,协同促进癌症进展和血管生成[10]

2.2 肿瘤细胞来源外泌体与卵巢癌血管生成

高侵袭性卵巢癌细胞释放的大量外泌体通过扩散在肿瘤细胞和TME之间发挥着重要的中介作用[11]。外泌体高效地富集了血管生成蛋白、微小RNA(microRNA, miRNA)等血管生成相关因子,并诱导其表型,显著提高了上皮细胞的增殖、迁移和出芽能力,从而促进细胞生长和新生血管生成[12]。ExoCarta数据已在外泌体中发现了1 116种脂质、9 796种蛋白质亚群和6 246种mRNA,通过改变受体细胞的功能特征来重新编程,从而调控血管生成[13]。有证据表明,非编码RNA尤其是长链非编码RNA(long non-coding RNA,lncRNA)和miRNA在肿瘤血管生成的调控中起着重要的作用。

2.2.1 肿瘤细胞来源外泌体通过释放非编码RNA调节卵巢癌血管生成

外泌体非编码RNA作为潜在的促血管生成因子在卵巢癌的转移中发挥了一定作用。He等[14]的研究发现,血清外泌体miR-205通过PTEN-蛋白激酶B(protein kinase B,Akt)途径诱导肿瘤发生,与非转移性卵巢癌患者相比,转移性卵巢癌患者的miR-205表达显著上调,且肿瘤血管生成明显增强。由卵巢癌细胞外泌体分泌到TME中的miR-484能够调节血管内皮细胞生长因子受体2(vascular endothelial growth factor receptor 2,VEGFR2)的表达,从而影响内皮细胞形成和维持血管样结构。在体内试验中,miR-484同时抑制了癌细胞和内皮细胞中相应受体VEGFA的表达,内源性抑制血管生成[15]。上皮性卵巢癌释放含有miR-141-3p的外泌体通过激活JAK/STAT3信号通路和诱导VEGFR2表达促进血管生成[16]。亦有研究发现miR-130a在耐药卵巢癌细胞来源的外泌体中表达显著上调,发挥促进血管生成的作用,其具体机制尚需进一步研究明确[17]

lncRNA是一类长度超过200个核苷酸的非编码RNA,其中一些被发现参与了各种关键的病理生物学过程。人肺腺癌转移相关转录本1(metastasis associated lung adenocarcinoma transcript 1,MALAT1)是目前了解较多的lncRNA,在卵巢癌体内实验中观察到敲除MALAT1后,由外泌体介导的肿瘤生长和血管生成被显著抑制。而在体外实验中发现,一些众所周知的促血管生成基因,包括VEGFAVEGFDENA-78、胎盘生长因子(placental growth factor,PlGF)、白细胞介素-8(interleukin-8,IL-8)等,皆为外泌体MALAT1的潜在下游靶点。因此,MALAT1可以借助外泌体从卵巢癌细胞转移到受体人脐静脉内皮细胞(human unbilical vein endothelial cell,HUVEC)从而调节肿瘤血管生成[18]。另有研究发现卵巢癌细胞来源的外泌体lncRNA ATB能够通过调节miR-204-3p/转化生长因子β受体2(transforming growth factor beta recptor 2,TGFβR2)促进卵巢癌的发生和肿瘤的体内生长,影响内皮细胞活性,调节血管生成和迁移[19]。同样的,外泌体lncRNA H19可以由肿瘤干细胞分泌并被转运到靶细胞,通过合成和释放VEGF刺激HUVEC血管生成[20]。与正常卵巢上皮细胞相比,IncRNA NEAT1在卵巢癌细胞中显著高表达,其通过海绵吸附miR-36上调成纤维细胞生长因子影响TME,促进卵巢癌细胞增殖[20]

2.2.2 肿瘤细胞来源外泌体通过释放蛋白质调节卵巢癌血管生成

外泌体蛋白存在于外泌体囊泡内或镶嵌在其表面,随着蛋白质组学分析的发展,外泌体蛋白的分泌和功能逐步被揭示[21]。在研究卵巢癌血管生成的分子水平调控时,发现可溶性上皮型钙黏蛋白(soluble E-cadherin,sE-cad)在卵巢癌患者的恶性腹水中高表达,其定位在外泌体表面,以脂质形式大量释放,是血管生成的关键调节因子[6]。前动力蛋白1(prokineticin 1,PROK1)是促动素的一种,已知其影响包括肾上腺、卵巢、胰腺和子宫内膜等在内的多种组织中的生理性血管生成,并在某些病理情况下参与肿瘤血管生成。前动力蛋白受体1(prokineticin receptor 1,PKR1)是PROK1的G蛋白耦联受体,激活PKR1可诱导血管生成功能,由于血管生成对肿瘤生长和转移至关重要,有研究从卵巢癌细胞系A2780和HO-8910细胞的培养基中分离了外泌体,发现PKR1阳性外泌体促进了内皮细胞的迁移和血管生成。这表明PKR1可以通过外泌体被周围细胞摄取,并通过细胞间的信号通信诱导血管生成效应[22]。在分子水平上,蛋白质组学揭示了卵巢癌外泌体中激活转录因子2(activating transcription factor 2,ATF2)、转移相关蛋白1(metastasis-associated protein 1,MTA1)等也可促进血管生成,但具体机制尚不清楚[23]

另一方面,亦有外泌体蛋白质表现出抑制血管生成的作用。CD151和tspan8是四跨膜蛋白家族的成员,CD151和tspan8阳性的外泌体参与细胞外基质的降解,影响细胞重塑、血管生成和细胞运动。在异种小鼠模型中,tspan8特异性抗体降低了体内细胞的运动性并阻断了肿瘤血管生成,而使用抗CD151抗体也可显著降低卵巢癌的侵袭和转移。四跨膜蛋白家族的其他成员(如CD63、CD9和CD81)在血管生成和血管发生中也发挥重要作用[24]。总之,外泌体蛋白抗体可能为卵巢癌抗血管生成治疗提供新的途径。

2.3 其他细胞来源的外泌体与卵巢癌血管生成

如前所述,肿瘤细胞源性外泌体对卵巢癌的血管生成发挥了重要的作用。而TME中的其他细胞,如肿瘤干细胞、成纤维细胞、免疫细胞等也能够释放具有调节血管生成作用的外泌体,并对肿瘤产生影响[25]。内皮细胞衍生的外泌体所含的蛋白质和mRNA/miRNA能够通过旁分泌或自分泌途径,诱导内皮细胞入侵和毛细血管状结构的形成,从而促进肿瘤血管生成和转移[26]。间充质干细胞是TME的重要组成部分,有研究发现,在顺铂的刺激下来源于卵巢癌肿瘤干细胞的外泌体可以直接提高间充质干细胞诱导内皮细胞血管生成的能力[27]。同时,由肿瘤细胞来源的外泌体重新编程的间充质干细胞能够将基质细胞转化为癌相关成纤维细胞(cancer-associated fibroblasts,CAFs)[28]。CAFs进一步通过调节包括血管生成、免疫抑制和药物获取在内的多个过程重塑肿瘤基质,进而促进肿瘤进展。外泌体还介导了部分肿瘤细胞和CAFs之间的通信[29]。例如CAFs释放的含有TGF-β的外泌体能够触发Smad信号级联,促进细胞迁移[30]。此外,肿瘤相关巨噬细胞来源的外泌体能够靶向miR-146b-5p/肿瘤坏死因子受体相关因子6(TNF receptor associated factor 6,TRAF6)/核因子κB(nuclear factor-κB,NF-κB)/基质金属蛋白酶-2(matrix metalloproteinase- 2,MMP-2)通路抑制内皮细胞迁移,从而影响卵巢癌的血管生成[31]

3 外泌体与卵巢癌抗血管生成治疗

当前,晚期卵巢癌的标准治疗主要是手术联合化学治疗,而抗血管生成治疗则是重要的维持治疗手段之一。抗血管生成药物是首个被批准用于卵巢癌的靶向药,越来越多的创新抗血管生成药物目前正处于临床试验阶段,然而抗血管生成治疗也存在着一定的局限。首先,血管生成和治疗抵抗的确切机制仍不清楚。其次,血供的限制也影响了抗肿瘤药物在肿瘤内部的有效转运,从而削弱了其抗肿瘤作用甚至引发治疗抵抗。生物学标志物对于选择更有可能从抗血管生成治疗中获益的患者至关重要[32]。由于肿瘤细胞来源外泌体携带了母细胞的生物学信息,因此可能成为抗血管生成治疗筛选的潜在非侵入性生物学标志物,同时也可能成为抗血管生成治疗的特异性靶点[12]

3.1 外泌体与靶向治疗

卵巢癌来源的外泌体circNFIX通过miR-518a-3p/TRIM44轴调控JAK/STAT1通路促进血管生成[33];sE-cad阳性外泌体与内皮细胞上的血管内皮钙黏蛋白结合,通过激活β-连环蛋白和NF-κB的信号级联影响血管内皮细胞迁移[6]。这些发现证明了卵巢癌血管生成的外泌体依赖机制,表明外泌体可能是卵巢癌的潜在治疗靶点。此外,De等[34]的研究发现余甘子提取物增加了卵巢癌细胞完全培养基中外泌体中的miR-375的表达,发现其具有抗血管生成功能,因此可能为卵巢癌抗血管生成治疗提供新的路径。

3.2 外泌体与外源性递送

由于外泌体具有无毒性和非免疫原性的特点,同时其逃逸了内涵体-溶酶体途径,外泌体可能成为卵巢癌治疗药物递送的一种功能性手段。通过靶向特定的蛋白质,外泌体包裹的治疗剂及其携带的生物分子能够选择性地作用于癌灶[31,35]。有研究表明,RGD肽含有Arg-Gli-Asp序列,该序列可以特异性结合在血管内皮细胞高度表达的整合素αvβ3上,因此RGD工程外泌体具有靶向血管上皮细胞的能力。而人工合成的miR-92b-3p过表达的工程外泌体可能与含有Arg-Gly-Asp(RGD)序列的肽结合,在裸鼠模型中更为显著地抑制移植肿瘤生长和血管生成,这表明具有过表达miR-92b-3p的工程外泌体可能有潜力用作抗血管生成剂,为卵巢癌提供了一种新的抗血管生成治疗方法[36]。此外,通过RGD修饰的外泌体靶向递送miR-484在诱导血管正常化的同时,也提高了卵巢癌对化疗的敏感性[15]

4 结语与展望

外泌体在卵巢癌的诊断、治疗方面都展现了巨大的研究价值和应用前景。抗血管生成已成为治疗卵巢癌的重要临床策略,而近年来外泌体对肿瘤血管生成的调控作用受到越来越多的关注。尽管已有研究探讨了外泌体通过释放蛋白质、非编码RNA、脂质等生物功能物质对于卵巢癌的血管生成的影响,外泌体调控卵巢癌血管生成确切的通路和作用靶点仍有待进一步探索。未来仍需要更多的深入研究发掘外泌体在卵巢癌抗血管生成治疗中更大的应用价值。

参考文献

Roma-Rodrigues C, Mendes R, Baptista PV, et al.

Targeting Tumor Microenvironment for Cancer Therapy

[J]. Int J Mol Sci, 2019, 20(4):840. doi: 10.3390/ijms20040840.

[本文引用: 1]

Akoto T, Saini S.

Role of Exosomes in Prostate Cancer Metastasis

[J]. Int J Mol Sci, 2021, 22(7):3528. doi: 10.3390/ijms22073528.

[本文引用: 1]

Wang S, Ma F, Feng Y, et al.

Role of exosomal miR-21 in the tumor microenvironment and osteosarcoma tumorigenesis and progression (Review)

[J]. Int J Oncol, 2020, 56(5):1055-1063. doi: 10.3892/ijo.2020.4992.

[本文引用: 1]

Yang E, Wang X, Gong Z, et al.

Exosome-mediated metabolic reprogramming: the emerging role in tumor microenvironment remodeling and its influence on cancer progression

[J]. Signal Transduct Target Ther, 2020, 5(1):242. doi: 10.1038/s41392-020-00359-5.

[本文引用: 1]

Mashouri L, Yousefi H, Aref AR, et al.

Exosomes: composition, biogenesis, and mechanisms in cancer metastasis and drug resistance

[J]. Mol Cancer, 2019, 18(1):75. doi: 10.1186/s12943-019-0991-5.

PMID:30940145      [本文引用: 1]

Tumor-derived exosomes (TDEs) participate in formation and progression of different cancer processes, including tumor microenvironment (TME) remodeling, angiogenesis, invasion, metastasis and drug-resistance. Exosomes initiate or suppress various signaling pathways in the recipient cells via transmitting heterogeneous cargoes. In this review we discuss exosome biogenesis, exosome mediated metastasis and chemoresistance. Furthermore, tumor derived exosomes role in tumor microenvironment remodeling, and angiogenesis is reviewed. Also, exosome induction of epithelial mesenchymal transition (EMT) is highlighted. More importantly, we discuss extensively how exosomes regulate drug resistance in several cancers. Thus, understanding exosome biogenesis, their contents and the molecular mechanisms and signaling pathways that are responsible for metastasis and drug-resistance mediated by TDEs may help to devise novel therapeutic approaches for cancer progression particularly to overcome therapy-resistance and preventing metastasis as major factors of cancer mortality.

Feng W, Dean DC, Hornicek FJ, et al.

Exosomes promote pre-metastatic niche formation in ovarian cancer

[J]. Mol Cancer, 2019, 18(1):124. doi: 10.1186/s12943-019-1049-4.

PMID:31409361      [本文引用: 3]

Ovarian cancer is one of the most common gynecological malignancies. Upon initial diagnosis, the majority of patients present with widespread metastatic growth within the peritoneal cavity. This metastatic growth occurs in stages, with the formation of a pre-metastatic niche occurring prior to macroscopic tumor cell invasion. Exosomes released by the primary ovarian tumor are small extracellular vesicles which prepare the distant tumor microenvironment for accelerated metastatic invasion. They regulate intercellular communication between tumor cells and normal stroma, cancer-associated fibroblasts, and local immune cells within the tumor microenvironment. In this review, we highlight the emerging roles of ovarian cancer exosomes as coordinators of pre-metastatic niche formation, biomarkers amenable to liquid biopsy, and targets of chemotherapy.

Lim D, Do Y, Kwon BS, et al.

Angiogenesis and vasculogenic mimicry as therapeutic targets in ovarian cancer

[J]. BMB Rep, 2020, 53(6):291-298. doi: 10.5483/BMBRep.2020.53.6.060.

PMID:32438972      [本文引用: 1]

Tumor angiogenesis is an essential process for growth and metastasis of cancer cells as it supplies tumors with oxygen and nutrients. During tumor angiogenesis, many pro-angiogenic factors are secreted by tumor cells to induce their own vascularization via activation of pre-existing host endothelium. However, accumulating evidence suggests that vasculogenic mimicry (VM) is a key alternative mechanism for tumor vascularization when tumors are faced with insufficient supply of oxygen and nutrients. VM is a tumor vascularization mechanism in which tumors create a blood supply system, in contrast to tumor angiogenesis mechanisms that depend on pre-existing host endothelium. VM is closely associated with tumor progression and poor prognosis in many cancers. Therefore, inhibition of VM may be a promising therapeutic strategy and may overcome the limitations of anti-angiogenesis therapy for cancer patients. In this review, we provide an overview of the current anti-angiogenic therapies for ovarian cancer and the current state of knowledge regarding the links between microRNAs and the VM process, with a focus on the mechanism that regulates associated signaling pathways in ovarian cancer. Moreover, we discuss the potential for VM as a therapeutic strategy against ovarian cancer. [BMB Reports 2020; 53(6): 291-298].

Dhani NC, Oza AM.

Targeting Angiogenesis: Taming the Medusa of Ovarian Cancer

[J]. Hematol Oncol Clin North Am, 2018, 32(6):1041-1055. doi: 10.1016/j.hoc.2018.07.008.

[本文引用: 1]

Wu B, Zhang L, Yu Y, et al.

miR-6086 inhibits ovarian cancer angiogenesis by downregulating the OC2/VEGFA/EGFL6 axis

[J]. Cell Death Dis, 2020, 11(5):345. doi: 10.1038/s41419-020-2501-5.

PMID:32393810      [本文引用: 1]

miRNAs have emerged as a pivotal component of gene regulatory networks, mediating cytokines secretion, cell cycle, and differentiation regulation. However, how miRNAs collaborate with transcription factors and downstream effector proteins that determine the fate of ovarian cancer cells remains to be understood, especially regarding to mechanism of tumor angiogenesis regulation. Based on the qRT-PCR and IHC analysis, we found that miR-6086 was maintained a very low level both in ovarian cancer cell lines and tissues. Further, we identified OC2 and EGFL6 as the direct targets of miR-6086 by luciferase assay and we observed an inverse relationship between the expression of miR-6086 and the OC2/VEGFA/EGFL6 axis. The Western blotting analysis suggested that OC2 could directly upregulate VEGFA and indirectly up-regulate EGFL6 through VEGFA. Moreover, miR-6086 could indirectly downregulate VEGFA through OC2. In addition, miR-6086, siOC2 and siEGFL6 could negatively regulate the tumor growth and angiogenesis of ovarian cancer (Skov3) in the animal studies, with the inhibition rates of 77.07%, 69.89%, and 73.62%, respectively (**p < 0.01). Moreover, the tumor cell proliferation, migration, and invasion of ovarian cancer cell lines (Caov3 and Skov3) and vascular formation (HUVECs) were significantly suppressed in vitro, by decreasing the AKT/MAPK pathways (*p < 0.05). Taken together, our results reveal that miR-6086 can suppress the angiogenesis networks in ovarian cancer by down-regulating the OC2/VEGFA/EGFL6 axis, directly or indirectly, which may provide potential targets for tumor therapeutics.

Gavalas NG, Liontos M, Trachana SP, et al.

Angiogenesis-related pathways in the pathogenesis of ovarian cancer

[J]. Int J Mol Sci, 2013, 14(8):15885-15909. doi: 10.3390/ijms140815885.

PMID:23903048      [本文引用: 1]

Ovarian Cancer represents the most fatal type of gynecological malignancies. A number of processes are involved in the pathogenesis of ovarian cancer, especially within the tumor microenvironment. Angiogenesis represents a hallmark phenomenon in cancer, and it is responsible for tumor spread and metastasis in ovarian cancer, among other tumor types, as it leads to new blood vessel formation. In recent years angiogenesis has been given considerable attention in order to identify targets for developing effective anti-tumor therapies. Growth factors have been identified to play key roles in driving angiogenesis and, thus, the formation of new blood vessels that assist in "feeding" cancer. Such molecules include the vascular endothelial growth factor (VEGF), the platelet derived growth factor (PDGF), the fibroblast growth factor (FGF), and the angiopoietin/Tie2 receptor complex. These proteins are key players in complex molecular pathways within the tumor cell and they have been in the spotlight of the development of anti-angiogenic molecules that may act as stand-alone therapeutics, or in concert with standard treatment regimes such as chemotherapy. The pathways involved in angiogenesis and molecules that have been developed in order to combat angiogenesis are described in this paper.

Fanale D, Corsini LR, Bono M, et al.

Clinical relevance of exosome-derived microRNAs in Ovarian Cancer: Looking for new tumor biological fingerprints

[J]. Crit Rev Oncol Hematol, 2024,193:104220. doi: 10.1016/j.critrevonc.2023.104220.

[本文引用: 1]

Ludwig N, Whiteside TL.

Potential roles of tumor-derived exosomes in angiogenesis

[J]. Expert Opin Ther Targets, 2018, 22(5):409-417. doi: 10.1080/14728222.2018.1464141.

[本文引用: 2]

Olejarz W, Kubiak-Tomaszewska G, Chrzanowska A, et al.

Exosomes in Angiogenesis and Anti-angiogenic Therapy in Cancers

[J]. Int J Mol Sci, 2020, 21(16):5840. doi: 10.3390/ijms21165840.

[本文引用: 1]

He L, Zhu W, Chen Q, et al.

Ovarian cancer cell-secreted exosomal miR-205 promotes metastasis by inducing angiogenesis

[J]. Theranostics, 2019, 9(26):8206-8220. doi: 10.7150/thno.37455.

PMID:31754391      [本文引用: 1]

By providing oxygen, nutrients and metastatic conduits, tumour angiogenesis is essential for cancer metastasis. Cancer cell-secreted microRNAs can be packaged into exosomes and are implicated in different aspects of tumour angiogenesis. However, the underlying mechanisms are incompletely understood. The GEPIA database and hybridization assay were used to analyse expression of miR-205 in ovarian tissues. Immunohistochemistry was performed to examine the relationship between miR-205 and microvessel density. Expression of circulating miR-205 was evaluated by RT-PCR and GEO database analysis. Co-culture and exosome labelling experiments were performed to assess exosomal miR-205 transfer from ovarian cancer (OC) cells to endothelial cells ECs. Exosome uptake assays were employed to define the cellular pathways associated with the endocytic uptake of exosomal miR-205. The role of exosomal miR-205 in angiogenesis was further investigated and. Western blotting and rescue experiments were applied to detect regulation of the PTEN-AKT pathway by exosomal miR-205 in ECs. miR-205 was up-regulated in OC tissues, and high expression of miR-205 was associated with metastatic progression in OC patients. Moreover, miR-205 was highly enriched in cancer-adjacent ECs, and up-regulation of miR-205 correlated positively with high microvessel density in OC patients. Importantly, miR-205 was markedly enriched in the serum of OC patients, and a high level of miR-205 in circulating exosomes was associated with OC metastasis. In addition, OC-derived miR-205 was secreted into the extracellular space and efficiently transferred to adjacent ECs in an exosome-dependent manner, and the lipid raft-associated pathway plays an important role in regulating uptake of exosomal miR-205. Exosomal miR-205 from OC cells significantly promoted angiogenesis and accelerated angiogenesis and tumour growth in a mouse model. Furthermore, we found that exosomal miR-205 induces angiogenesis via the PTEN-AKT pathway. These findings demonstrate an exosome-dependent mechanism by which miR-205 derived from cancer cells regulates tumour angiogenesis and implicate exosomal miR-205 as a potential therapeutic target for OC.© The author(s).

Zhao Z, Shuang T, Gao Y, et al.

Targeted delivery of exosomal miR-484 reprograms tumor vasculature for chemotherapy sensitization

[J]. Cancer Lett, 2022, 530:45-58. doi: 10.1016/j.canlet.2022.01.011.

PMID:35051533      [本文引用: 2]

The vascular dysfunction of ovarian cancer (OC) contributes to the chemotherapeutic resistance. In this study, we aimed to explore whether exosome-mediated angiogenesis blocking could improve the chemotherapy sensitivity via vascular normalization. Exosomes were armed with RGD on the surface by fusing Lamp2b. Candidate miRNAs related to tumor angiogenesis was detected by qRT-PCR. RGD-modified exosomes were loaded with miRNAs via electroporation. The therapeutic effects of the exosomes on angiogenesis, vascular normalization, and chemotherapy sensitivity were systemically analyzed in the xenograft model. RGD-modified exosomes were relatively enriched in the tumor mass, both the tumor cell and the endothelial cells. Among the miRNA candidates, miR-484 was found down-regulated in both the cancer cells and the angiogenic endothelial cells. In vivo xenograft model experiment revealed that injection of RGD-modified exosomes loaded with miR-484 induced vessel normalization and in turn sensitized the cancer cells to chemotherapy induced apoptosis. Mechanistically, miR-484 simultaneously inhibited the expression of VEGF-A from the cancer cells and the corresponding receptors in the endothelial cells. Targeted delivery of miR-484 via RGD-modified exosomes improves the vascular normalization, sensitizes the cancer to chemotherapy, and prolongs the survival time of tumor-bearing mice after chemotherapy, opening an avenue for the clinical management of chemotherapy resistance.Copyright © 2022 Elsevier B.V. All rights reserved.

Masoumi-Dehghi S, Babashah S, Sadeghizadeh M.

microRNA-141-3p-containing small extracellular vesicles derived from epithelial ovarian cancer cells promote endothelial cell angiogenesis through activating the JAK/STAT3 and NF-κB signaling pathways

[J]. J Cell Commun Signal, 2020, 14(2):233-244. doi: 10.1007/s12079-020-00548-5.

PMID:32034654      [本文引用: 1]

Paracrine signaling between tumor and surrounding stromal cells is critical for the maintenance of tumor microenvironment during ovarian cancer progression. Small extracellular vesicles (sEVs; exosomes in particular) are nano-sized vesicles secreted actively by many cells including tumor cells and are found to have fundamental roles in intercellular communication through shuttling functional RNAs. Although microRNAs (also called miRNAs or miRs), small non-coding RNAs regulating gene expression, are selectively accumulated in tumor sEVs and can mediate intercellular communication, the exact biological mechanisms underlying the functions of exosomal miRNAs in ovarian tumor angiogenesis remain unclear. In this study, sEVs were isolated from conditioned medium of the human ovarian carcinoma cell line SKOV-3 using ExoQuick Exosome Precipitation Solution, and characterized by scanning electron microscopy, dynamic light scattering, and immunoblotting. To elucidate the possible paracrine effects on ovarian tumor cell-derived sEVs (TD-sEVs), we investigated the angiogenesis-related signaling events triggered by TD-sEVs in endothelial cells. Due to the possible role in ovarian tumor pathogenesis, we focused on miR-141-3p which was detected to be enriched in TD-sEVs compared with their corresponding donor cells. We identified that sEV transfer of miR-141-3p considerably reduced the expression levels of cytokine-inducible suppressors of cytokine signaling (SOCS)-5 leading to up-regulated JAK-STAT3 pathway in endothelial cells. We also observed that sEV-shuttled miR-141-3p may up-regulate the expression of VEGFR-2 in endothelial cells which leads to promoting endothelial cell migration and angiogenesis. The putative role of miR-141-3p shuttled by TD-sEVs in regulating VEGFR-2 expression was demonstrated by the ability of anti-miR-141-3p to rescue the promoting effects of TD-sEVs on the expression of VEGFR-2 in endothelial cells. Our results also revealed that TD-sEVs trigger the intracellular reactive oxygen species (ROS)-dependent activation of NF-κB signaling in endothelial cells. Taken together, our findings propose a novel model in which sEV transfer of epithelial ovarian cancer-secreted miR-141-3p plays as a significant mediator of intercellular communication, promoting endothelial cell angiogenesis.

Li Z, Yan-Qing W, Xiao Y, et al.

Exosomes secreted by chemoresistant ovarian cancer cells promote angiogenesis

[J]. J Ovarian Res, 2021, 14(1):7. doi: 10.1186/s13048-020-00758-w.

PMID:33413589      [本文引用: 1]

Ovarian cancer (OC) has the highest mortality rate in gynecologic tumors. Despite decades of continuous efforts, the survival rate of patients has not improved significantly, mostly due to drug resistance. Exosomes are hot topics in recent years. Cells can affect the biological behaviors of other cells by transferring exosomes. So far, numerous researchers have found that tumor cells can secrete exosomes which play a important role in the development of tumors. Solid tumors can promote angiogenesis. When drug resistance occurs, it seems that more blood vessels form. We suppose that exosomes derived from chemoresistant OC cells can also promote angiogenesis.We investigate whether exosomes secreted by chemoresistant SKOV3-DDP cells (SKOV3-DDP-exo) and sensitive SKOV3 cells (SKOV3-exo) influence angiogenesis. After exosomes were extracted, exosomes were co-cultured with HUVECs. We found that SKOV3-DDP-exo and SKOV3-exo are absorbed by endothelial cells and promote the proliferation, migration, invasion and tube formation of endothelial cells. Moreover, SKOV3-DDP-exo is more powerful in angiogenesis, suggesting that parts of the components of SKOV3-DDP-exo are significantly radical. We also found that miR-130a was highly expressed in drug-resistant OC cells. Also, we found that miR-130a in SKOV3-DDP-exo is higher than SKOV3-exo. Therefore, we suggest that miR-130a in exosomes is the main cause of chemoresistant OC cells promoting angiogenesis.

Qiu JJ, Lin XJ, Tang XY, et al.

Exosomal Metastasis-Associated Lung Adenocarcinoma Transcript 1 Promotes Angiogenesis and Predicts Poor Prognosis in Epithelial Ovarian Cancer

[J]. Int J Biol Sci, 2018, 14(14):1960-1973. doi: 10.7150/ijbs.28048.

[本文引用: 1]

Yuan D, Guo T, Zhu D, et al.

Exosomal lncRNA ATB Derived from Ovarian Cancer Cells Promotes Angiogenesis via Regulating miR-204-3p/TGFβR2 Axis

[J]. Cancer Manag Res, 2022, 14:327-337. doi: 10.2147/CMAR.S330368.

PMID:35115831      [本文引用: 1]

Ovarian cancer is a life-threatening disease with a high mortality rate in women. Our previous work presented that long non-coding RNA (lncRNA) activated by transforming growth factor beta (TGF-β) (lncRNA ATB) played a role of oncogene in ovarian cancer. However, whether exosomal lncRNA ATB from ovarian cancer cells could regulate the tumorigenesis of ovarian cancer remains unclear.RT-qPCR assay was performed to evaluate the level of lncRNA ATB in cancer cells (SKOV3 and A2780). In addition, ovarian cancer cells-secreted exosomes were collected with ultracentrifugation. CCK8 assay was performed to detect the viability of ovarian cells and HUVECs. Meanwhile, Western blot was performed to detect the expression of mechanism related protein and tube formation assay was used to observe the angiogenesis of HUVECs. Finally, xenograft mice model was used to verify the role of ovarian cancer cell-derived exosomes in vivo.Ovarian cancer cells-derived exosomes promoted the viability, angiogenesis and migration of HUVECs; however, knockdown of lncRNA ATB in HUVECs reversed these phenomena. In addition, exosomal lncRNA ATB promoted the tumorigenesis of ovarian cancer via regulating miR-204-3p/TGFβR2 axis. Furthermore, ovarian cancer cells-secreted exosomal lncRNA ATB increased tumor growth in vivo.Exosomal lncRNA ATB derived from ovarian cancer cells could improve tumor microenvironment via regulating miR-204-3p/TGFβR2 axis. Thus, this study might provide new knowledge for the treatment of ovarian cancer.© 2022 Yuan et al.

Wang M, Fu L, Xu Y, et al.

A comprehensive overview of exosome lncRNAs: Emerging biomarkers and potential therapeutics in gynecological cancers

[J]. Front Oncol, 2023,13:1138142. doi: 10.3389/fonc.2023.1138142.

[本文引用: 2]

Wang X, Huang J, Chen W, et al.

The updated role of exosomal proteins in the diagnosis, prognosis, and treatment of cancer

[J]. Exp Mol Med, 2022, 54(9):1390-1400. doi: 10.1038/s12276-022-00855-4.

[本文引用: 1]

Zhang X, Sheng Y, Li B, et al.

Ovarian cancer derived PKR1 positive exosomes promote angiogenesis by promoting migration and tube formation in vitro

[J]. Cell Biochem Funct, 2021, 39(2):308-316. doi: 10.1002/cbf.3583.

[本文引用: 1]

Yang C, Kim HS, Song G, et al.

The potential role of exosomes derived from ovarian cancer cells for diagnostic and therapeutic approaches

[J]. J Cell Physiol, 2019, 234(12):21493-21503. doi: 10.1002/jcp.28905.

PMID:31144314      [本文引用: 1]

Most patients with ovarian cancer (OC) are diagnosed at the advanced stages due to the absence of appropriate early diagnostic markers. Thus, OC is a gynecological disease with a low-survival rate. Exosomes are extracellular vesicles that are widely being considered as mediators for the noninvasive diagnosis of OC. Exosomes are expected to aid in the effective diagnosis of OC because they carry components, such as RNAs, proteins, and lipids, the compositions of which vary depending on the pathological characteristics of the patient. In this review, we document the methods that have been developed to detect exosomes and their components in OC. We also assess the potential biomarkers contained in exosomes that could be clinically useful, such as proteins, microRNAs (miRNAs), long noncoding RNAs (lncRNAs), and phospholipids. Moreover, we described the role played by exosomes in the tumor microenvironment and in OC angiogenesis, migration, and tumor growth. Various types of cells in the tumor microenvironment, including macrophages, fibroblasts, and mesenchymal stem cells (MSCs), interact directly with exosomes and promote or inhibit the progression of OC. Therefore, we summarize the studies that have suggested a therapeutic approach to OC using exosomes. Collectively, understanding the mechanism of exosome-based OC progression would broaden our knowledge regarding the diagnosis and therapy of OC.© 2019 Wiley Periodicals, Inc.

Yunusova N, Dzhugashvili E, Yalovaya A, et al.

Comparative Analysis of Tumor-Associated microRNAs and Tetraspanines from Exosomes of Plasma and Ascitic Fluids of Ovarian Cancer Patients

[J]. Int J Mol Sci, 2022, 24(1):464. doi: 10.3390/ijms24010464.

[本文引用: 1]

Ribeiro MF, Zhu H, Millard RW, et al.

Exosomes Function in Pro- and Anti-Angiogenesis

[J]. Curr Angiogenes, 2013, 2(1):54-59. doi: 10.2174/22115528113020020001.

[本文引用: 1]

Salem KZ, Moschetta M, Sacco A, et al.

Exosomes in Tumor Angiogenesis

[J]. Methods Mol Biol, 2016,1464:25-34. doi: 10.1007/978-1-4939-3999-2_3.

[本文引用: 1]

Vera N, Acuña-Gallardo S, Grünenwald F, et al.

Small Extracellular Vesicles Released from Ovarian Cancer Spheroids in Response to Cisplatin Promote the Pro-Tumorigenic Activity of Mesenchymal Stem Cells

[J]. Int J Mol Sci, 2019, 20(20):4972. doi: 10.3390/ijms20204972.

[本文引用: 1]

Whiteside TL.

Exosome and mesenchymal stem cell cross-talk in the tumor microenvironment

[J]. Semin Immunol, 2018, 35:69-79. doi: 10.1016/j.smim.2017.12.003.

PMID:29289420      [本文引用: 1]

Mesenchymal stem cells (MSCs) are a major component of the tumor microenvironment (TME) and play a key role in promoting tumor progression. The tumor uses exosomes to co-opt MSCs and re-program their functional profile from normally trophic to pro-tumorigenic. These tumor-derived small vesicles called "TEX" carry and deliver a cargo rich in proteins and nucleic acids to MSCs. Upon interactions with surface receptors on MSCs and uptake of the exosome cargo by MSCs, molecular, transcriptional and translational changes occur that convert MSCs into producers of factors that are necessary for tumor growth and that also alter functions of non-tumor cells in the TME. The MSCs re-programmed by TEX become avid producers of their own exosomes that carry and deliver mRNA and miRNA species as well as molecular signals not only back to tumor cells, directly enhancing their growth, but also horizontally to fibroblasts, endothelial cells and immune cells in the TME, indirectly enhancing their pro-tumor functions. TEX-driven cross-talk of MSCs with immune cells blocks their anti-tumor activity and/or converts them into suppressor cells. MSCs re-programmed by TEX mediate pro-angiogenic activity and convert stromal cells into cancer-associated fibroblasts (CAFs). Although MSCs have a potential to exert anti-tumor activities, they largely provide service to the tumor using the multidirectional communication system established by exosomes in the TME. Future therapeutic options consider disruption of this complex vicious cycle by either molecular or gene-regulated silencing of pro-tumor effects mediated by MSCs in the TME.Copyright © 2017 Elsevier Ltd. All rights reserved.

Giusti I, Di Francesco M, Poppa G, et al.

Tumor-Derived Extracellular Vesicles Activate Normal Human Fibroblasts to a Cancer-Associated Fibroblast-Like Phenotype, Sustaining a Pro-Tumorigenic Microenvironment

[J]. Front Oncol, 2022,12:839880. doi: 10.3389/fonc.2022.839880.

[本文引用: 1]

Kazemi NY, Gendrot B, Berishvili E, et al.

The Role and Clinical Interest of Extracellular Vesicles in Pregnancy and Ovarian Cancer

[J]. Biomedicines, 2021, 9(9):1257. doi: 10.3390/biomedicines9091257.

[本文引用: 1]

Xu WX, Wang DD, Zhao ZQ, et al.

Exosomal microRNAs shuttling between tumor cells and macrophages: cellular interactions and novel therapeutic strategies

[J]. Cancer Cell Int, 2022, 22(1):190. doi: 10.1186/s12935-022-02594-y.

[本文引用: 2]

Mei C, Gong W, Wang X, et al.

Anti-angiogenic therapy in ovarian cancer: Current understandings and prospects of precision medicine

[J]. Front Pharmacol, 2023,14:1147717. doi: 10.3389/fphar.2023.1147717.

[本文引用: 1]

Ye H, Wang RY, Yu XZ, et al.

Exosomal circNFIX promotes angiogenesis in ovarian cancer via miR-518a-3p/TRIM44 axis

[J]. Kaohsiung J Med Sci, 2023, 39(1):26-39. doi: 10.1002/kjm2.12615.

[本文引用: 1]

De A, Powers B, De A, et al.

Emblica officinalis extract downregulates pro-angiogenic molecules via upregulation of cellular and exosomal miR-375 in human ovarian cancer cells

[J]. Oncotarget, 2016, 7(21):31484-31500. doi: 10.18632/oncotarget.8966.

PMID:27129171      [本文引用: 1]

Ovarian cancer (OC) is highly resistant to current treatment strategies based on a combination of surgery, chemotherapy and radiation therapy. We have recently demonstrated the anti-neoplastic effect of Amla extract (Emblica officinalis, AE) on OC cells in vitro and in vivo. We hypothesized that AE attenuates growth of OC through microRNA (miR)-regulated mechanism(s). The inhibitory effect of AE on proliferation, migration and invasiveness (P≤0.001) of SKOV3 cells and >90% attenuation of tumor growth in a xenograft mouse model suggested multiple targets. RT-qPCR analysis of microRNAs associated with OC showed a >2,000-fold increase in the expression of miR-375 in AE-treated SKOV3 cells that was blocked by an exogenous miR-375 inhibitor (P≤0.001). AE also decreased the gene and protein expression of IGF1R, a target of miR-375 (P≤0.001), and SNAIL1 (P≤0.002), an EMT-associated transcription factor that represses E-cadherin expression (P≤0.003). AE increased E-cadherin expression (P≤0.001). Treatment of SKOV3 cells with AE resulted in increased miR-375 in exosomes in the medium (P≤0.01). Finally, AE significantly decreased the expression of IGF1R and SNAIL1 proteins during attenuation of SKOV3-derived xenograft tumor. Together, these results show that AE modulates cancer cells and the tumor microenvironment via activation of miR-375 and by targeting IGF1R and SNAIL1 in OC cells.

Shimizu A, Sawada K, Kimura T.

Pathophysiological Role and Potential Therapeutic Exploitation of Exosomes in Ovarian Cancer

[J]. Cells, 2020, 9(4):814. doi: 10.3390/cells9040814.

[本文引用: 1]

Wang J, Wang C, Li Y, et al.

Potential of peptide-engineered exosomes with overexpressed miR-92b-3p in anti-angiogenic therapy of ovarian cancer

[J]. Clin Transl Med, 2021, 11(5):e425. doi: 10.1002/ctm2.425.

PMID:34047469      [本文引用: 1]

Exosomal microRNA (miRNA) as a mediator of intercellular communication plays an essential part in tumor-relevant angiogenesis. Therapy against angiogenesis has been demonstrated to have a remarkable antitumor efficacy in various malignancies, but not as expected in ovarian cancer.Exosomes were isolated by ultracentrifugation. Exosomal miRNA sequencing and gene function experiments were used to identify the differential expressed miRNAs in exosomes and their mRNA targets. SKOV3 cell line that stably overexpressed miR-92b-3p was constructed by lentivirus. In vitro, angiogenesis was analyzed by tube formation assay and migration assay. The angiogenic and antitumor effects in vivo were assessed in zebrafish and nude mouse models. Combination index was calculated to assess the synergetic inhibition of angiogenesis between miR-92b-3p and Apatinib. Peptides were conjugated with exosomal membranes to obtain engineered exosomes.Ovarian cancer cell-derived exosomes facilitated the angiogenesis and migration capability of vascular endothelial cells in vitro and in vivo. The expression of miR-92b-3p was much lower in ovarian cancer cell-derived exosomes than that in immortalized ovarian epithelial cell-derived exosomes. The exosomal miR-92b-3p modulated tumor-associated angiogenesis via targeting SOX4. Besides, Peptide-engineered exosomes with overexpressed miR-92b-3p showed the stronger abilities of anti-angiogenesis and antitumor than parental exosomes, whether alone or combined with Apatinib.Our findings demonstrate the effect and mechanism of exosomal miR-92b-3p from ovarian cancer cells on tumor-associated angiogenesis and the potential of artificially generated exosomes with overexpressed miR-92b-3p to be used as anti-angiogenic agent, which may provide a new approach for anti-angiogenic therapy of ovarian cancer.© 2021 The Authors. Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.

/

津ICP备05004273
版权所有 © 天津市医学科学技术信息研究所
地址:天津市和平区贵州路94号 邮编:300070 电话:022-23337521 E-mail:fuchanfence@gjfckx.ac.cn
本系统由北京玛格泰克科技发展有限公司设计开发