女性生殖道和肠道微生物在卵巢癌中的研究进展
030001 太原,山西医科大学第二医院妇产科
Research Progress of Female Reproductive Tract and Gut Microbiome in Ovarian Cancer
Department of Obstetrics and Gynecology, Second Hospital of Shanxi Medical University, Taiyuan 030001, China
Corresponding authors: PING Yi, E-mail:pingyi7110@126.com
Received: 2021-08-3
卵巢癌致死率极高,但发病机制尚不清楚。近年随着高通量测序技术的发展,女性生殖道和肠道微生物在卵巢癌方面的研究已成为热点。研究发现,在卵巢癌中,上、下生殖道优势菌群厚壁杆菌丰度降低,拟杆菌数量增多,肠道菌群频繁失调。其可能通过自身成分和代谢产物引起致癌反应、升高循环中的雌激素水平、促进卵巢癌发生,介导炎症反应影响免疫调节,在卵巢癌的发生发展中起重要作用。此外,女性生殖道和肠道微生物群还参与卵巢癌的治疗过程,与耐药和疗效密切相关。其中,粪菌移植、益生菌补充及抗生素的应用已得到初步探索。综述女性生殖道和肠道微生物群结构、与卵巢癌的相关性以及临床治疗研究,旨在为卵巢癌的筛查、治疗及预防提供新的方向和策略。
关键词:
The mortality rate of ovarian cancer is very high, but the pathogenesis is still unclear. In recent years, with the development of high-throughput sequencing technologies, the study of the female reproductive tract and gut microbiome in ovarian cancer has become a hot spot. It was found that in ovarian cancer, the abundance of firmicutes in the upper and lower reproductive tract decreased, the number of bacteroides increased and the gut microbiome was often dysbiosis. It may play an important role in the development of ovarian cancer by causing carcinogenic reactions through its components and metabolites, increasing the ratio of estrogen to promote ovarian carcinogenesis, and mediating inflammatory responses that affect immune regulation. In addition, the female reproductive tract and gut microbiome are involved in the therapeutic process of ovarian cancer and are closely associated with drug resistance and efficacy. Among these, fecal transplantation, probiotic supplementation, and antibiotic application have been initially explored. This article focuses on the structure of the female reproductive tract and gut microbiome, its relevance to ovarian cancer, and clinical treatment studies, to provide new directions and strategies for the screening, treatment, and prevention of ovarian cancer.
Keywords:
本文引用格式
郭文迪, 平毅.
GUO Wen-di, PING Yi.
人类微生物组与宿主以互利共生的关系存在。微生物组动态变化影响黏膜稳态、代谢、炎症及宿主免疫,其失调是多种肿瘤发生的重要潜在机制。2020年全球卵巢癌新发病例313 959例,死亡人数207 252例,在妇科肿瘤中致死率持续居首位[1],5年生存率在30%~40%。子宫内膜异位症恶变及输卵管起源是Ⅰ、Ⅱ型卵巢上皮性肿瘤发病的主流学说,但缺乏对病因的进一步探索。有效的筛查治疗手段和抗化疗耐药复发方面仍面临严峻挑战。近年研究相继发现女性生殖道及肠道微生物参与卵巢癌的进展并影响预后[2]。现对卵巢癌患者生殖道及肠道微生物群的结构变化,及其可能诱发卵巢癌及影响治疗的潜在机制进行综述,为寻求癌症标志物、优化目前治疗及预防卵巢癌提供新的方向和依据。
1 卵巢癌相关微生物群概况
1.1 下生殖道微生物组成
高通量测序将阴道微生物群分为5种群落类型(community state types,CST)。健康女性阴道微生物多样性低,通常由厚壁杆菌组成,富含不同种类的乳杆菌,主要是卷曲杆菌(Lactobacillus crispatus,CST-Ⅰ)、加氏乳杆菌(Lactobacillus gasseri,CST-Ⅱ)、惰性乳杆菌(Lactobacillus iners,CST-Ⅲ)和詹氏乳杆菌(Lactobacillus jensenii,CST-Ⅴ)。这些乳杆菌通过产生乳酸、细菌素和过氧化氢(hydrogen peroxide,H2O2)与宿主互利,减少病原菌的定植。
临床常见的阴道微生物失调为细菌性阴道病,以乳杆菌丰度降低、厌氧菌丰度增加为特征。病原微生物包括加德纳菌、普雷沃菌、阿托波菌、支原体和衣原体等。在上皮性卵巢癌患者中,超过60%的样本检测到布鲁氏菌、衣原体及支原体,10%的样本检测到人乳头瘤病毒,这些微生物感染均是卵巢癌患病的风险因素,有助于卵巢非典型上皮细胞的进展[3]。一项由176例卵巢癌患者、109例乳腺癌易感基因1(breast cancer susceptibility gene 1,BRCA1)突变携带者和295例健康对照者组成的多中心研究发现,罹患卵巢癌(OR=2.80,P=0.020)、BRCA1突变(OR=2.79,P=0.012)与宫颈阴道微生物乳杆菌占比小于50%相关[4],这表明宫颈阴道微生物群失调可能对卵巢癌的发生起重要作用,且阴道乳杆菌是卵巢癌的保护物种。
1.2 上生殖道微生物组成
微生物存在于健康人群的上生殖道(子宫、输卵管和卵巢)中,健康的输卵管、卵巢微生物是卵巢癌的抑制因素[5]。同一个体,与下生殖道相比,上生殖道乳杆菌的丰度降低,不动杆菌、假单胞菌的占比升高。当致病菌通过宫颈或淋巴途径进入子宫、输卵管、卵巢造成上生殖道微生物失调时,将导致盆腔炎。女性盆腔炎患者发生上皮性卵巢癌的风险增加[6]。衣原体作为盆腔炎中最常见的致病微生物通过激活旁分泌Wnt通路,阻断线粒体半胱氨酸天冬氨酸蛋白酶3(caspase-3)和细胞色素C的释放,逃避CD8+T细胞的攻击抑制凋亡,导致输卵管上皮细胞结构和功能的破坏。衣原体质粒蛋白抗体3(chlamydial plasmid-encoded protein 3,Pgp3)阳性会增加卵巢癌患病风险[7]。
在卵巢癌患者中,微生物结构及其占比常发生特异性改变。近年研究者分别在卵巢癌患者阴道、宫颈阴道部、肿瘤组织、腹膜、血清及粪便中鉴定出微生物,发现上生殖道、肿瘤组织及腹膜中微生物α多样性下降。Wang等[8]研究显示,与癌旁组织相比,癌灶内拟杆菌数量增多,不动杆菌、乳球菌缺乏,变形杆菌丰度增加,变形杆菌/厚壁菌比值增大,这将有助于卵巢癌的诊断。
1.3 肠道微生物组成
正常肠道微生物群是动态复杂的,受宿主特征(种族、性别、年龄、激素水平、免疫反应)和环境影响(饮食、吸烟、药物)而改变。健康的肠道微生物群主要由5种细菌门类组成:革兰阳性厚壁菌门、革兰阴性拟杆菌门、放线菌门、变形菌门和梭杆菌门,约占肠道总微生物群的90%。
动物模型研究表明,使用抗生素耗尽人卵巢腺癌细胞SKOV-3细胞荷瘤裸鼠的肠道菌群后,卵巢癌肿瘤生长速度明显加快,证实肠道微生物群的失调促进卵巢癌进展[9]。此外,Miao等[10]对10例卵巢癌患者和20例良性卵巢肿瘤患者的腹腔液进行16S核糖体核糖核酸(16S ribosomal ribonucleic acid,16S rRNA)高通量测序,发现卵巢癌患者的腹腔积液富含肠道来源的革兰阴性菌,并鉴定出18种微生物可作为卵巢癌新标志物。其联合血清标志物糖类抗原125(carbohydrate antigen 125,CA125)、人附睾蛋白4(human epididymis protein 4,HE4)可提高卵巢癌诊断的准确性。
2 卵巢癌相关微生物群的功能
2.1 自身成分及代谢产物
脂多糖(lipopolysaccharides, LPS)、溶血磷脂、色氨酸及其衍生物在卵巢癌中有明确作用,其他代谢产物如短链脂肪酸、生物胺及次级胆汁酸在卵巢癌中的作用尚未统一。卵巢癌患者肿瘤组织中革兰阴性菌的比例增加,自身促炎成分上调[8]。LPS可以刺激Toll样受体2(Toll-like receptor 2,TLR2)、TLR4和TLR5,激活磷脂酰肌醇3激酶(phosphatidylinositol 3-kinase,PI3K)信号传导、基质金属蛋白酶(matrix metalloproteinases,MMP)相关家族表达,激活肿瘤相关巨噬细胞并诱导上皮-间质转化(epithelial-mesenchymal transition,EMT)[11]。溶血磷脂酸及溶血磷脂酰丝氨酸被证实能够诱导蛋白激酶B(protein kinase B,Akt)、丝裂原活化蛋白激酶(mitogen-activated protein kinase,MAPK)和Ca2+信号传导,上调血管生成因子表达,诱导卵巢癌细胞增殖、迁移及侵袭。肠道中小营养分子也具有重要作用,Xie等[12]发现卵巢癌患者血清色氨酸和吲哚丙酸减少,且水平越低预后越差。色氨酸作为能量来源支持乳杆菌的生长,抑制病原菌扩张,通过激活芳香烃受体(aryl hydrocarbon receptor,AHR)、孕烷X受体(pregnane X receptor,PXR)调节黏膜免疫[13]。此外,部分细菌还可以穿过上皮屏障直接将毒素插入宿主细胞,如大肠杆菌素直接损伤DNA引起致癌反应[14]。
2.2 影响雌激素调控
高雌激素水平是卵巢癌的风险因素,一项巢式病例对照研究表明,高水平循环雌二醇(estradiol,E2)的女性患非浆液性卵巢癌的风险是低水平E2妇女的3倍[OR(五分位数5:五分位数1)=3.01,95%CI:1.17~7.73,P-trend=0.03;P-het<0.01][15]。在围绝经期使用激素替代疗法的人群,卵巢癌患病风险至少在5年内增加。高雌激素促癌主要通过直接与受体结合促进基因转录,间接激活其他转录因子参与炎症反应,控制细胞增殖、分化及凋亡。
2.3 介导炎症及免疫调节
卵巢癌组织中微生物的含量与促炎因子上调、免疫抑制途径下调一致。树突状细胞识别微生物后,激活TLR产生不同的细胞因子,调控T细胞分化以实现细菌耐受性和免疫功能的平衡。如果持续微生物失调,相关信号将募集免疫细胞,刺激炎症因子释放,进而导致DNA损伤和突变累积[18]。
其中,TLR4和TLR5在致癌过程中具有重要作用。有研究发现,乳杆菌通过调控微小RNA-21(microRNA-21,miR-21)和miR-200b,调节人上皮性卵巢腺癌细胞CAOV-4中TLR4的反应性,可达到治疗卵巢癌的目的[19]。TLR4和LPS结合后,募集髓样分化因子88(myeloid differentiation factor 88,MyD88)和MyD88配体样(MyD88 adapter-like,MAL)蛋白,启动炎症反应,激活核因子κB(nuclear factor kappa-B,NF-κB)、白细胞介素6(interleukin-6,IL-6)和血管内皮生长因子(vascular endothelial growth factor,VEGF)等因子,促进癌细胞生长并获得肿瘤耐药性[20]。卵巢癌中,IL-6是重要的致癌因子,正反馈增强Janus激酶(Janus kinase,JAK)/信号转导及转录激活因子3(signal transducer and activator of transcription,STAT3)通路,增加MMP表达,促进细胞增殖及EMT,诱导肿瘤耐药[21]。同样,TLR5与菌群介导的免疫抑制、肿瘤炎症及转移密切相关。其与细菌鞭毛蛋白结合后激活MyD88/肿瘤坏死因子受体相关分子6(tumor necrosis factor receptor associated factor 6,TRAF6)/NF-κB通路,上调IL-6等因子,加速肿瘤生长[22]。这种促癌作用依赖肠道菌群,去除肠道细菌后其促瘤生长的影响消失[23]。
3 微生物群与卵巢癌治疗
3.1 参与耐药及影响疗效
3.2 微生物相关治疗
抗生素治疗卵巢癌可能具有潜力,但相关研究的结论尚不一致。氯霉素[33]、盐霉素[34]与顺铂联合使用抑制肿瘤生长。与之相反,氨苄青霉素、万古霉素、新霉素和甲硝唑联合治疗促进裸鼠移植瘤的生长与侵袭[9]。Cheng等[5]研究表明抗生素治疗会影响肠道微生物群,从而调节免疫系统的极化,并通过炎症促进卵巢癌进展。在一项对402例接受手术和铂类化疗的Ⅲ/Ⅳ期上皮性卵巢癌患者的回顾性研究中,化疗期间使用抗生素治疗超过48 h的患者与未使用抗生素治疗的患者相比,无进展生存期(17.4个月 vs. 23.1个月,HR=1.50,P<0.001)和总生存期(45.6个月 vs. 62.4个月,HR=1.50,P<0.001)均缩短,使用抗革兰阳性菌抗生素治疗的患者无进展生存期(16.5个月 vs. 23.1个月,HR=1.85,P<0.001)和总生存期(35.0个月 vs. 62.4个月,HR=2.12,P<0.001)缩短更显著[35]。因此需进一步研究证实抗生素治疗卵巢癌的利弊。
鉴于免疫相关因素在卵巢癌中的重要作用,过度应激及反应不足均会导致卵巢癌进展。减弱卵巢癌的炎症风暴、靶向免疫相关受体及细胞可能成为一种有效的癌症治疗策略。Curiel等[36]证实阻断调节性T细胞的迁移或功能同样有助于卵巢癌的治疗。同时,抑制TRL4可以降低MMP相关酶活性,并阻断EMT过程,起到抗瘤作用[26]。此外,免疫系统激活疗法的有效性取决于微生物的组成和免疫原性。在细胞过继免疫治疗(adoptive T cell transfer,ACT)的新方案中,嵌合抗原受体T细胞免疫疗法(chimeric antigen receptor T-cell immunotherapy,CAR-T)[37]及树突状细胞疫苗[38]旨在增强免疫检查点阻断,加强机体免疫功能,重新杀死癌细胞,目前已注册Ⅲ期临床试验,但个性化免疫治疗卵巢癌仍面临挑战。
4 微生物群与卵巢癌的预防
饮食搭配可以调节女性生殖道及肠道微生物群的组成,并通过遗传及表观遗传改变基因表达、影响疾病易感性。生酮饮食、地中海饮食及素食均已证实有利于卵巢癌预后。经典的生酮饮食脂肪比例高、碳水化合物及蛋白质比例低。患者接受生酮饮食后,从传统的葡萄糖代谢转变为酮体代谢,此种代谢模式改变可减少炎症及肿瘤血管生成,增强细胞凋亡[39]。研究表明大量摄入动物脂肪、饱和脂肪酸和反式脂肪酸会增加卵巢癌患病风险,摄入蔬菜、水果、纤维、维生素C和多不饱和脂肪酸为其保护因素[40]。地中海饮食恰以水果、蔬菜、豆类、全谷物和坚果为特色,控制红肉摄入,脂肪来源于初榨橄榄油。与地中海饮食相似,素食富含纤维及植物雌激素,可增加肠道微生物群的多样性及丰度,具有抗癌作用[40]。
5 结语
在卵巢癌中,微生物群可以通过宿主环境、生活方式及免疫系统,改变其组成结构及代谢产物,进而影响癌症发生、进展及治疗效果。卵巢癌患者肿瘤组织、生殖道、肠道和腹膜中独特的微生物群及其代谢产物可作为早期诊断的生物学标志物,为筛查、治疗分层及预后提供新的思路。目前卵巢癌与微生物间引起复杂相互作用的因果关系尚未完全阐明,特定的微生物未具体明确,其临床价值和可行性暂未得到充分肯定,微生物相关治疗方式的适用性及可靠性仍需探索。今后应进一步结合宿主遗传、表观遗传、微生物群、生活方式和其他环境因素综合挖掘卵巢癌的发生机制,深入探讨利用微生物组进行预防、筛查及治疗干预的价值。
参考文献
Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries
[J].DOI:10.3322/caac.21660 URL [本文引用: 1]
Shifts in gut and vaginal microbiomes are associated with cancer recurrence time in women with ovarian cancer
[J].
DOI:10.7717/peerj.11574
PMID:34178459
[本文引用: 1]
Many studies investigating the human microbiome-cancer interface have focused on the gut microbiome and gastrointestinal cancers. Outside of human papillomavirus driving cervical cancer, little is known about the relationship between the vaginal microbiome and other gynecological cancers, such as ovarian cancer. In this retrospective study, we investigated the relationship between ovarian cancer, platinum-free interval (PFI) length, and vaginal and gut microbiomes. We observed that -dominated vaginal communities were less common in women with ovarian cancer, as compared to existing datasets of similarly aged women without cancer. Primary platinum-resistance (PPR) disease is strongly associated with survivability under one year, and we found over one-third of patients with PPR (PFI < 6 months, = 17) to have a vaginal microbiome dominated by (>20% relative abundance), while only one platinum super-sensitive (PFI > 24 months, = 23) patient had an -dominated microbiome. Additionally, was associated with little, or no, gross residual disease, while other species were dominant in women with >1 cm gross residual disease. In the gut microbiome, we found patients with PPR disease to have lower phylogenetic diversity than platinum-sensitive patients. The trends we observe in women with ovarian cancer and PPR disease, such as the absence of and presence of in the vaginal microbiome as well as low gut microbiome phylogenetic diversity have all been linked to other diseases and/or pro-inflammatory states, including bacterial vaginosis and autoimmune disorders. Future prospective studies are necessary to explore the translational potential and underlying mechanisms driving these associations.© 2021 Jacobson et al.
Serologic markers of Chlamydia trachomatis and other sexually transmitted infections and subsequent ovarian cancer risk: Results from the EPIC cohort
[J].DOI:10.1002/ijc.32999 URL [本文引用: 1]
Association between the cervicovaginal microbiome, BRCA1 mutation status, and risk of ovarian cancer: a case-control study
[J].DOI:10.1016/S1470-2045(19)30340-7 URL [本文引用: 1]
Opportunities and Challenges of the Human Microbiome in Ovarian Cancer
[J].DOI:10.3389/fonc.2020.00163 URL [本文引用: 2]
Risk of ovarian cancer in women with pelvic inflammatory disease: a population-based study
[J].DOI:10.1016/S1470-2045(11)70165-6 URL [本文引用: 1]
Antibodies Against Chlamydia trachomatis and Ovarian Cancer Risk in Two Independent Populations
[J].DOI:10.1093/jnci/djy084 URL [本文引用: 1]
The differential distribution of bacteria between cancerous and noncancerous ovarian tissues in situ
[J].
DOI:10.1186/s13048-019-0603-4
PMID:31954395
[本文引用: 2]
With the improvement of bacterial detection, the theory of the sterile female upper reproductive tract has been frequently challenged in recent years. However, thus far, no researchers have used ovaries as study targets.Six women who were diagnosed with ovarian cancer were included in the cancer group, and ten women who were diagnosed with a noncancerous ovarian condition (including three patients with uterine myoma and seven patients with uterine adenomyosis) were included in the control group. Immunohistochemistry staining using an antibacterial lipopolysaccharide (LPS) antibody was used to confirm the presence of bacteria in the ovarian tissues. In addition, 16S rRNA sequencing was used to compare the differences in the bacteria between ovarian cancer tissues and noncancerous ovarian tissues. BugBase and Phylogenetic Investigation of Communities by Reconstruction of Unobserved States (PICRUSt) were used to predict the functional composition of the bacteria.Bacterial LPS was present in ovarian cancer tissue and noncancerous ovarian tissue, which implied the presence of bacteria in ovarian tissue. When compared to the noncancerous ovarian bacteria at the phylum level, the cancerous ovarian bacteria were composed of increased Aquificae and Planctomycetes and decreased Crenarchaeota. When predicting metagenomes, gene functions associated with the potentially pathogenic and the oxidative stress-tolerant phenotype were enriched in the ovaries of the cancer group. Forty-six significantly different KEGG pathways existed in the ovarian bacteria of the cancer group compared to that of the control group.Different bacteria compositions were present in cancerous and noncancerous ovarian tissues.Chines Clinical Trail Registry, CHiCTR1800020018, Registered 11 September 2018, http://www.chictr.org.cn/.
Intestinal dysbiosis promotes epithelial-mesenchymal transition by activating tumor-associated macrophages in ovarian cancer
[J].DOI:10.1093/femspd/ftz019 [本文引用: 2]
Assessment of peritoneal microbial features and tumor marker levels as potential diagnostic tools for ovarian cancer
[J].DOI:10.1371/journal.pone.0227707 URL [本文引用: 1]
Induction of galectin-1 by TLR-dependent PI3K activation enhances epithelial-mesenchymal transition of metastatic ovarian cancer cells
[J].
DOI:10.3892/or.2017.5533
PMID:28350104
[本文引用: 1]
The expression of different toll-like receptors (TLRs) on tumor cells has been associated with disease aggressiveness, treatment resistance, and poor prognosis. The phosphatidylinositol 3-kinase (PI3K)/AKT pathway is considered critical for cancer cell survival and proliferation. Thus, we investigated the effect of TLR-stimulated PI3K activation on the epithelial-to-mesenchymal transition (EMT) of primary (Caov-3) and metastatic (SK‑OV‑3) epithelial ovarian cancer cell lines in this study. TLR engagement with various ligands promoted the expression of class IA PI3K (p110α, p110β, and p110δ) and increased the expression of mesenchymal markers (N-cadherin, Slug, Vimentin, Snail, α-SMA, and TCF) in SK‑OV‑3 cells. The migratory activity and secretion of EMT-related cytokines of SK‑OV‑3 were significantly higher compared to those of Caov-3 after activation with TLR agonist. Although the invasive capacity and production of EMT-related cytokines of LPS-stimulated SK‑OV‑3 cells were significantly suppressed by all pharmacological inhibitors of the p110 isoform, the Syk/Src-dependent p110β isoform prominently attenuated migration activity. In contrast, the production of IL-10 and galectin-1 was mainly affected by the p110δ isoform. Gene silencing of TLR4 and galectin-1 with siRNA decreased the expression of matrix metalloproteinase-2 (MMP2) and MMP9 and reduced mesenchymal markers in LPS-treated SK‑OV‑3 cells. This study demonstrated that TLR-mediated PI3K activation modulated the invasion and metastasis of ovarian cancer through the production of galectin-1, suggesting that inhibition of the p110 isoform is a promising therapeutic approach against metastatic ovarian cancer.
Metabolic profiling and novel plasma biomarkers for predicting survival in epithelial ovarian cancer
[J].DOI:10.18632/oncotarget.16739 URL [本文引用: 1]
Dietary tryptophan links encephalogenicity of autoreactive T cells with gut microbial ecology
[J].
DOI:10.1038/s41467-019-12776-4
PMID:31653831
[本文引用: 1]
The interaction between the mammalian host and its resident gut microbiota is known to license adaptive immune responses. Nutritional constituents strongly influence composition and functional properties of the intestinal microbial communities. Here, we report that omission of a single essential amino acid - tryptophan - from the diet abrogates CNS autoimmunity in a mouse model of multiple sclerosis. Dietary tryptophan restriction results in impaired encephalitogenic T cell responses and is accompanied by a mild intestinal inflammatory response and a profound phenotypic shift of gut microbiota. Protective effects of dietary tryptophan restriction are abrogated in germ-free mice, but are independent of canonical host sensors of intracellular tryptophan metabolites. We conclude that dietary tryptophan restriction alters metabolic properties of gut microbiota, which in turn have an impact on encephalitogenic T cell responses. This link between gut microbiota, dietary tryptophan and adaptive immunity may help to develop therapeutic strategies for protection from autoimmune neuroinflammation.
Microbiota in cancer development and treatment
[J].DOI:10.1007/s00432-018-2816-0 URL [本文引用: 1]
Circulating estrogens and postmenopausal ovarian and endometrial cancer risk among current hormone users in the Women′s Health Initiative Observational Study
[J].DOI:10.1007/s10552-019-01233-8 URL [本文引用: 1]
Associations of the fecal microbiome with urinary estrogens and estrogen metabolites in postmenopausal women
[J].DOI:10.1210/jc.2014-2222 URL [本文引用: 1]
The Microbiome-Estrogen Connection and Breast Cancer Risk
[J].DOI:10.3390/cells8121642 URL [本文引用: 1]
Nuclear Factor Erythroid 2-Related Factor 2 in Regulating Cancer Metabolism
[J].DOI:10.1089/ars.2020.8024 URL [本文引用: 1]
Modulatory Role of Vaginal-Isolated Lactococcus lactis on the Expression of miR-21, miR-200b, and TLR-4 in CAOV-4 Cells and In Silico Revalidation
[J].
DOI:10.1007/s12602-019-09596-9
PMID:31797280
[本文引用: 1]
Ovarian cancer (OC) is a leading cause of death among women worldwide. Various evidences suggest that oncomiRs and Toll-like receptor 4 (TLR-4) signaling pathways appear to be key players in the initiation and progression of OC. It seems there exists a continuous intercommunication between cancer cells and normal microbiota of the vagina. The biological impacts of vaginal isolated lactococcus lactis on CAOV-4 cells were investigated using several molecular biology experiments, including flow cytometry, DAPI staining, DNA ladder, and scratch assay. The expression of microRNAs (miRNAs/miRs) 21, 200b, and TLR-4 in the CAOV-4 cells was also evaluated by the real-time RT-PCR assay. Furthermore, an integrative in silico analysis was conducted using normalized web-available microarray data (GSE14407) to revalidate the experimental findings and identify potential biomarkers in ovarian cancer. Protein-protein interactions (PPIs) network was studied by means of the STRING database using Cytoscape v3.6.1. The miRNA target genes were identified using the dbDEMC v2.0, miRTarBase, and miRDB databases. Our data demonstrated that L. lactis probiotic candidate downregulates TLR-4, miR-21, and miR-200b expression levels, which correlates with induction of apoptosis as confirmed by DAPI staining, DNA ladder assay, annexin V/PI staining, and inhibition of migration validated by scratch assay. By in silico analysis, several targets (miR-17-5p-BCL2, miR-21-5p-MKNK2, miR-129-5p-CDK6) were identified, while BCL2, CCNB1, and VEGFA were found as the hub proteins in the miRNA-target and PPI networks. Further, downregulation of the TLR-4, miR-21, and miR-200b was partially validated by the in silico analysis. Based on our findings, the vaginal isolated probiotic strain presents great potential to control the ovarian cancer which may provide beneficial impact on the clinical management of ovarian cancer.
TLR4 signaling induced by lipopolysaccharide or paclitaxel regulates tumor survival and chemoresistance in ovarian cancer
[J].
DOI:10.1038/onc.2009.289
PMID:19826413
[本文引用: 1]
Toll-like receptors (TLRs) expressed on immune cells trigger inflammatory responses. TLRs are also expressed on ovarian cancer (OvCa) cells, but the consequences of signaling by the TLR4/MyD88 pathway in these cells are unclear. Here, TLR4 and MyD88 expression in OvCa tissues (n=20) and cell lines (OVCAR3, SKOV3, AD10, A2780 and CP70) was evaluated by reverse transcriptase-PCR, western blots and immunohistochemistry. Cell growth, apoptosis, nuclear factor-kappaB (NF-kappaB) translocation, IRAK4 and TRIF expression and cJun phosphorylation were measured following tumor cell exposure to the TLR4 ligands, lipopolysaccharide (LPS) or paclitaxel (PTX). Culture supernatants were tested for cytokine levels. TLR4 was expressed in all tumors, tumor cell lines and normal epithelium. MyD88 was detectable in tumor tissues and in 3/5 OvCa lines but not in normal cells. In MyD88(+) SCOV3 cells, LPS or PTX binding to TLR4 induced IRAK4 activation and cJun phosphorylation, activated the NF-kappaB pathway and promoted interleukin (IL)-8, IL-6, vascular endothelial growth factor and monocyte chemotactic protein-1 production and resistance to drug-induced apoptosis. Silencing of TLR4 in SCOV3 cells with small interference RNA resulted in phosphorylated-cJun (p-cJun) downregulation and a loss of PTX resistance. In PTX-sensitive, MyD88(neg) A2780 cells, TLR4 stimulation upregulated TRIF, and TLR4 silencing eliminated this effect. Thus, TLR4/MyD88 signaling supports OvCa progression and chemoresistance, promoting immune escape.
IL-6 and ovarian cancer: inflammatory cytokines in promotion of metastasis
[J].
DOI:10.2147/CMAR.S179189
PMID:30584363
[本文引用: 1]
Ovarian cancer is the most fatal gynecological cancer in the USA and the fifth most common cancer-related cause of death in women. Inflammation has been shown to play many roles in ovarian cancer tumor growth, with the proinflammatory cytokine interleukin-6 (IL-6) having been established as a key immunoregulatory cytokine. Ovarian cancer cells continuously secrete cytokines that promote tumorigenicity in both autocrine and paracrine fashions while also receiving signals from the tumor microenvironment (TME). The TME contains many cells including leukocytes and fibroblasts, which respond to proinflammatory cytokines and secrete their own cytokines, which can produce many effects including promotion of chemoresistance, resistance to apoptosis, invasion, angiogenesis by way of overexpression of vascular endothelial growth factor, and promotion of metastatic growth at distant sites. IL-6 and its proinflammatory family members, including oncostatin M, have been found to directly stimulate enhanced invasion of cancer cells through basement membrane degradation caused by the overexpression of matrix metalloproteinases, stimulate promotion of cell cycle, enhance resistance to chemotherapy, and cause epithelial-to-mesenchymal transition (EMT). IL-6 has been shown to activate signaling pathways that lead to tumor proliferation, the most studied of which being the Janus kinase (JAK) and STAT3 pathway. IL-6-induced JAK/STAT activation leads to constitutive activation of STAT3, which has been correlated with enhanced tumor cell growth and resistance to chemotherapy. IL-6 has also been shown to act as a trigger of the EMT, the hypothesized first step in the metastatic cascade. Understanding the important role of IL-6 and its family members' effects on the pathogenesis of ovarian cancer tumor growth and metastasis may lead to more novel treatments, detection methods, and improvement of overall clinical outcomes.
TLR5/7-mediated PI3K activation triggers epithelial-mesenchymal transition of ovarian cancer cells through WAVE3-dependent mesothelin or OCT4/SOX2 expression
[J].
DOI:10.3892/or.2017.5941
PMID:28901470
[本文引用: 1]
Toll-like receptor (TLR)-mediated signaling induces cell migration or invasion in several tumors and various stages of cancer. Interactions of mesothelin, a 40-kDa cell surface glycoprotein, with cancer antigen 125 (CA125) is associated with drug resistance, metastasis, and poor clinical outcome of ovarian cancer patients. In this study, we examined the role of TLR5 and TLR7 in the metastasis of ovarian cancer through the induction of mesothelin/CA125 expression and investigated its underlying mechanism. TLR5 agonist (flagellin) and TLR7 agonist (imiquimod) upregulated mesenchymal phenotypes and produced epithelial-mesenchymal transition (EMT)-related cytokines in the SKOV3 cells; however, TLR7 expressing CaOV3 cells had no response to the specific ligand, imiquimod, for enhancing its EMT processes. Stimulation of the SKOV3 cells with flagellin or imiquimod activated Wiskott-Aldrich syndrome protein verprolin-homologous 3 (WAVE3) and mesothelin/CA125, whereas it suppressed the expression of TAp63. Moreover, knockdown of TLR5 or TLR7 in SKOV3 cells profoundly impaired the TLR5- or TLR7-intiated downstream signaling pathway. Loss of WAVE3 in SKOV3 cells led to the inhibition of invasion, suppression of mesenchymal characteristics, prevention of OCT4/SOX2 secretion, and attenuation of mesothelin/CA125 expression after stimulation with flagellin or imiquimod. Although the disruption of mesothelin decreased the migratory activity of the TLR5/7-activated SKOV3 cells, knockdown of mesothelin failed to reduce the expression of mesenchymal markers, OCT4, and SOX2. In addition, targeting OCT4 or SOX2 with siRNA had no effect on the expression of mesothelin and the suppression of transcriptionally active p63 (TAp63) in the TLR5/7-stimulated SKOV3 cells. Our results suggest that TLR5/7-mediated WAVE3 activation not only controls the mesothelin-related EMT processes but also modulates OCT4/SOX2-mediated mesenchymal marker expression. Taken together, both TLR5 and TLR7 expression are critical for the TLR5/7-induced metastasis of ovarian cancer and the inhibition of WAVE3 might be a new therapeutic target to control ovarian cancer metastasis.
Microbially driven TLR5-dependent signaling governs distal malignant progression through tumor-promoting inflammation
[J].
DOI:10.1016/j.ccell.2014.11.009
PMID:25533336
[本文引用: 1]
The dominant TLR5(R392X) polymorphism abrogates flagellin responses in >7% of humans. We report that TLR5-dependent commensal bacteria drive malignant progression at extramucosal locations by increasing systemic IL-6, which drives mobilization of myeloid-derived suppressor cells (MDSCs). Mechanistically, expanded granulocytic MDSCs cause γδ lymphocytes in TLR5-responsive tumors to secrete galectin-1, dampening antitumor immunity and accelerating malignant progression. In contrast, IL-17 is consistently upregulated in TLR5-unresponsive tumor-bearing mice but only accelerates malignant progression in IL-6-unresponsive tumors. Importantly, depletion of commensal bacteria abrogates TLR5-dependent differences in tumor growth. Contrasting differences in inflammatory cytokines and malignant evolution are recapitulated in TLR5-responsive/unresponsive ovarian and breast cancer patients. Therefore, inflammation, antitumor immunity, and the clinical outcome of cancer patients are influenced by a common TLR5 polymorphism. Copyright © 2015 Elsevier Inc. All rights reserved.
Epigenetic therapy for ovarian cancer: promise and progress
[J].
DOI:10.1186/s13148-018-0602-0
PMID:30646939
[本文引用: 1]
Ovarian cancer is the deadliest gynecologic malignancy, with a 5-year survival rate of approximately 47%, a number that has remained constant over the past two decades. Early diagnosis improves survival, but unfortunately only 15% of ovarian cancers are diagnosed at an early or localized stage. Most ovarian cancers are epithelial in origin and treatment prioritizes surgery and cytoreduction followed by cytotoxic platinum and taxane chemotherapy. While most tumors will initially respond to this treatment, recurrence is likely to occur within a median of 16 months for patients who present with advanced stage disease. New treatment options separate from traditional chemotherapy that take advantage of advances in understanding of the pathophysiology of ovarian cancer are needed to improve outcomes. Recent work has shown that mutations in genes encoding epigenetic regulators are mutated in ovarian cancer, driving tumorigenesis and resistance to treatment. Several of these epigenetic modifiers have emerged as promising drug targets for ovarian cancer therapy. In this article, we delineate epigenetic abnormalities in ovarian cancer, discuss key scientific advances using epigenetic therapies in preclinical ovarian cancer models, and review ongoing clinical trials utilizing epigenetic therapies in ovarian cancer.
Engagement of the mannose receptor by tumoral mucins activates an immune suppressive phenotype in human tumor-associated macrophages
[J].DOI:10.1155/2010/547179 [本文引用: 1]
TLR4 blockade using TAK-242 suppresses ovarian and breast cancer cells invasion through the inhibition of extracellular matrix degradation and epithelial-mesenchymal transition
[J].DOI:10.1016/j.ejphar.2019.03.046 URL [本文引用: 2]
Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment
[J].DOI:10.1126/science.1240527 URL [本文引用: 1]
Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors
[J].DOI:10.1126/science.aan3706 URL [本文引用: 2]
Frontline Science: Microbiota reconstitution restores intestinal integrity after cisplatin therapy
[J].DOI:10.1002/JLB.5HI1117-446RR URL [本文引用: 1]
The administration of Escherichia coli Nissle 1917 ameliorates irinotecan-induced intestinal barrier dysfunction and gut microbial dysbiosis in mice
[J].DOI:10.1016/j.lfs.2019.06.004 URL [本文引用: 1]
Efficacy of antineoplastic treatment is associated with the use of antibiotics that modulate intestinal microbiota
[J].DOI:10.1080/2162402X.2016.1150399 URL [本文引用: 1]
Fecal microbiota transplant overcomes resistance to anti-PD-1 therapy in melanoma patients
[J].DOI:10.1126/science.abf3363 URL [本文引用: 1]
Clarithromycin synergizes with cisplatin to inhibit ovarian cancer growth in vitro and in vivo
[J].
DOI:10.1186/s13048-019-0570-9
PMID:31703731
[本文引用: 1]
Cisplatin-based chemotherapy is the first-line treatment for ovarian cancer. However, acquired resistance to cisplatin treatment often occurs in epithelial ovarian cancer, and effective and practical methods for overcoming this obstacle are urgently needed. The study aimed to demonstrate the synergistic effect of clarithromycin (CAM) with cisplatin to inhibit ovarian carcinoma cells growth in vitro and in vivo.We performed CCK-8 assay to detect apoptosis rates in response to CAM alone or in combination with cisplatin, which were further confirmed by Annexin V and PI staining methods and western blotting. Mechanistically, CAM could reduce endogenous antioxidant enzyme expression and increase the levels of reactive oxygen species (ROS) to augment the cytotoxic effect of cisplatin. Meanwhile, a tumor xenograft model in athymic BALB/c-nude mice demonstrated that CAM combined with cisplatin resulted in reduced tumor growth and weight compared with cisplatin alone.Collectively, our results indicate that CAM works synergistically with cisplatin to inhibit ovarian cancer cell growth, which may be manipulated by a ROS-mediated mechanism that enhances cisplatin therapy, and offers a novel strategy for overcoming cisplatin therapy resistance.
Overcoming Resistance to Platinum-Based Drugs in Ovarian Cancer by Salinomycin and Its Derivatives-An In Vitro Study
[J].DOI:10.3390/molecules25030537 URL [本文引用: 1]
Impact of antibiotic treatment during platinum chemotherapy on survival and recurrence in women with advanced epithelial ovarian cancer
[J].
DOI:10.1016/j.ygyno.2020.09.010
PMID:32950250
[本文引用: 1]
To determine whether antibiotic treatment (ABX) during platinum chemotherapy (PC) for epithelial ovarian cancer (EOC) impacts progression-free survival (PFS) and overall survival (OS).Retrospective single institution cohort study in women with newly diagnosed stage III/IV EOC (n = 424) who underwent cytoreductive surgery (CRS) and PC from 2009 to 2015. ABX for >48 h, including ABX against gram-positive (anti-G + ABX) bacteria were recorded. The impact of ABX on PFS and OS was assessed using univariate and multivariable Cox regression models.Of 424 eligible women, 34.7% (n = 147) received ABX, with 11.3% (n = 48) treated with anti-G + ABX. ABX decreased PFS (17.4 vs. 23.1 months, HR 1.50, 95% CI 1.20-1.88, p < 0.001) and OS (45.6 vs. 62.4 months, HR 1.63, 95% CI 1.27-2.08, p < 0.001) compared to no ABX. Similarly, anti-G + ABX worsened PFS (16.5 vs. 23.1 months; HR 1.85, 95% CI 1.33-2.55) and OS (35.0 vs. 62.4 months; HR 2.12, 95% CI 1.50-3.0, p < 0.001). On multivariable analysis, all ABX and anti-G + ABX significantly worsened PFS (HR 1.31, 95% CI 1.04-1.65, p = 0.02), (HR 1.50, 95% CI 1.07-2.10, p = 0.02) and OS (HR 1.52, 95% CI 1.18-1.96, p = 0.001), (HR 1.83, 95% CI 1.27-2.62, p = 0.001) respectively. Increased Clavien Dindo score was associated with worsened PFS (1-2 - HR 1.52, 95% CI 1.14-2.03, p = 0.004; 3-4 - HR 1.86, 95% CI 1.27-2.72, p = 0.001) but not OS (1/2 - HR 1.35, 95% CI 0.97-1.88, p = 0.08; 3/4 - HR 1.53, 95% CI 1.00-2.34, p = 0.05); residual disease (p < 0.05) and neoadjuvant chemotherapy (p < 0.001) were associated with worse PFS and OS.In this retrospective cohort study of women with advanced EOC undergoing PC, ABX treatment was associated with decreased PFS and OS. Mechanistic studies are needed to investigate the negative impact of ABX upon PC response in EOC.Copyright © 2020 Elsevier Inc. All rights reserved.
Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival
[J].
DOI:10.1038/nm1093
PMID:15322536
[本文引用: 1]
Regulatory T (T(reg)) cells mediate homeostatic peripheral tolerance by suppressing autoreactive T cells. Failure of host antitumor immunity may be caused by exaggerated suppression of tumor-associated antigen-reactive lymphocytes mediated by T(reg) cells; however, definitive evidence that T(reg) cells have an immunopathological role in human cancer is lacking. Here we show, in detailed studies of CD4(+)CD25(+)FOXP3(+) T(reg) cells in 104 individuals affected with ovarian carcinoma, that human tumor T(reg) cells suppress tumor-specific T cell immunity and contribute to growth of human tumors in vivo. We also show that tumor T(reg) cells are associated with a high death hazard and reduced survival. Human T(reg) cells preferentially move to and accumulate in tumors and ascites, but rarely enter draining lymph nodes in later cancer stages. Tumor cells and microenvironmental macrophages produce the chemokine CCL22, which mediates trafficking of T(reg) cells to the tumor. This specific recruitment of T(reg) cells represents a mechanism by which tumors may foster immune privilege. Thus, blocking T(reg) cell migration or function may help to defeat human cancer.
Immunotherapy and Its Development for Gynecological (Ovarian, Endometrial and Cervical) Tumors: From Immune Checkpoint Inhibitors to Chimeric Antigen Receptor (CAR)-T Cell Therapy
[J].DOI:10.3390/cancers13040840 [本文引用: 1]
Dendritic Cell Vaccines in Ovarian Cancer
[J].
DOI:10.3389/fimmu.2020.613773
PMID:33584699
[本文引用: 1]
Ovarian cancer (OC) is one of the most lethal malignant gynecologic tumors, characterized by an uncertain presentation and poor outcomes. With or without neoadjuvant chemotherapy, surgery followed by platinum-based chemotherapy and maintenance therapy are the basis for the treatment of ovarian cancer patients, but the outcome is still highly restricted by their advanced stage when diagnosed and high recurrence rate after chemotherapy. To enhance the anti-tumor effect and postpone recurrence, anti-VEGF agents and PARP inhibitors are suggested as maintenance therapy, but the population that can benefit from these treatments is small. Based on the interactions of immune cells in the tumor microenvironment, immunotherapies are being explored for ovarian cancer treatment. Disappointingly, the immune checkpoint inhibitors show relatively low responses in ovarian cancer. As shown in several studies that have uncovered a relationship between DC infiltration and outcome in ovarian cancer patients, dendritic cell (DC)-based treatments might have a potential effect on ovarian cancer. In this review, we summarize the functions of dendritic cells (DCs) in the tumor microenvironment, as well as the responses and drawbacks of existing clinical studies to draw a comprehensive picture of DC vaccine treatment in ovarian cancer and to discuss the promising future of immune biomarkers.Copyright © 2021 Zhang, He, Li, Chen, Liu, Wu and Guo.
Personalized Nutrition in Disrupting Cancer - Proceedings From the 2017 American College of Nutrition Annual Meeting
[J].
DOI:10.1080/07315724.2018.1500499
PMID:30511901
[本文引用: 1]
Cancer is a major public health problem and is the second leading cause of death in the United States and worldwide; nearly one in six deaths are attributable to cancer. Approximately 20% of all cancers diagnosed in the United States are attributable to unhealthy diet, excessive alcohol consumption, physical inactivity, and body fatness. Individual cancers are distinct disease states that are multifactorial in their causation, making them exceedingly cumbersome to study from a nutrition standpoint. Genetic influences are a major piece of the puzzle and personalized nutrition is likely to be most effective in disrupting cancer during all stages. Increasing evidence shows that after a cancer diagnosis, continuing standard dietary recommendations may not be appropriate. This is because powerful dietary interventions such as short-term fasting and carbohydrate restriction can disrupt tumor metabolism, synergizing with standard therapies such as radiation and drug therapy to improve efficacy and ultimately, cancer survival. The importance of identifying dietary interventions cannot be overstated, and the American College of Nutrition's commitment to advancing knowledge and research is evidenced by dedication of the 2017 ACN Annual Meeting to "Disrupting Cancer: The Role of Personalized Nutrition" and this resulting proceedings manuscript, which summarizes the meeting's findings.
Pre-diagnosis diet and survival after a diagnosis of ovarian cancer
[J].DOI:10.1038/bjc.2017.120 URL [本文引用: 2]
/
〈 |
|
〉 |
