新闻动态
/
News

Cancer Res:推动卵巢癌转移的”帮凶“

来源:本站 作者:admin 浏览:914 更新时间:2016-09-10

莱斯大学和得克萨斯大学MD安德森癌症中心一项新的研究显示了卵巢癌如何利用腹部组织来源的一种特定类型的成体干细胞来推动其增长。这项研究在线发表在上周的Cancer Research杂志上,该研究提出了一种新的治疗方式来针对侵袭性卵巢癌,即通过破坏促使癌细胞茁壮成长的代谢过程。

这种特定类型干细胞被称为大网膜脂肪基质细胞或O-ASCs,已经与卵巢肿瘤的增殖,迁移和耐药性有关,但此类干细胞的确切作用是未知的,研究论文首席研究员Rice's Deepak Nagrath说:我们发现,O-ASCs供给癌细胞产生一氧化氮所需要的代谢物,一氧化氮是已知增加血流量的重要信号分子。癌症研究人员在80年前发现癌细胞与正常细胞之间的代谢差异。几十年来,科学家相信“Warburg效应”适用于所有的癌症,但Nagrath实验室研究和其他人已经发现,每一种类型的癌症都有自己的代谢特征。

例如,在五月发表的一项研究中,Nagrath及其同事发现高度侵袭性卵巢癌细胞是谷氨酰胺依赖性的,在实验室中剥夺细胞谷氨酰胺的外部来源,一些实验性药物能有效杀死晚期卵巢癌细胞。

癌细胞利用腹部干细胞来推动增长和转移。最新研究表明,一种新的方式即通过破坏使他们能够茁壮成长的代谢过程,来靶向治疗恶性卵巢癌。在新的研究中,合著者Bahar Salimian进行了一系列实验来研究O-ASCs和卵巢癌细胞之间复杂的相互作用。O-ASCs是一类在网膜中发现的成体干细胞,网膜是小腹中的组织,其是卵巢癌最常见的转移部位之一。

当我们在实验室共同培养两种细胞类型(癌细胞和O-ASCs),我们发现,癌细胞会利用干细胞分泌的精氨酸,并且癌细胞通过燃烧精氨酸,来释放瓜氨酸,这又造成了干细胞产生更多精氨酸。

我们的研究结果表明,O-ASCs通过增加一氧化氮水平,上调糖酵解和降低肿瘤细胞的氧化应激,Nagrath说:值得注意的是,我们还发现在癌细胞中O-ASC介导的耐药可以通过改变癌症所依赖的一氧化氮平衡来逆转。Nagrath说一种破坏干细胞和肿瘤细胞之间信号的鸡尾酒疗法,可能破坏卵巢癌依靠的以推动其转移性增长的代谢平衡。

原文摘要:

Nitric oxide mediates metabolic coupling of omentum-derived adipose to ovarian and endometrial cancer Cells

Bahar Salimian,et al.

Omental adipose stromal cells (O-ASCs) are multipotent population of mesenchymal stem cells contained in the omentum tissue which promote endometrial and ovarian tumor proliferation, migration and drug resistance. The mechanistic underpinnings of O-ASCs role in tumor progression and growth are unclear. Here, we propose a novel nitric oxide (NO) mediated metabolic coupling between O-ASCs and gynecological cancer cells in which O-ASCs support NO homeostasis in malignant cells. NO is synthesized endogenously by the conversion of L-arginine into citrulline through nitric oxide synthase (NOS). Through arginine depletion in the media using L-arginase and NOS inhibition in cancer cells using L-NAME, we demonstrate that patient derived O-ASCs increase NO levels in ovarian and endometrial cancer cells and promote proliferation in these cells. O-ASCs and cancer cell cocultures revealed that cancer cells utilize O-ASCs-secreted arginine and in turn secrete citrulline in the microenvironment. Interestingly, citrulline increased adipogenesis potential of the O-ASCs. Furthermore, we found that O-ASCs increased NO synthesis in cancer cells which led to decrease in mitochondrial respiration in these cells. Our findings suggest that O-ASCs upregulate glycolysis and reduce oxidative stress in cancer cells by increasing NO levels through paracrine metabolite secretion. Significantly, we found that O-ASC mediated chemoresistance in cancer cells can be deregulated by altering NO homeostasis. A combined approach of targeting secreted arginine through L-arginase, along with targeting microenvironment secreted factors induced increased NO synthesis in cancer cells using L-NAME, may be a viable therapeutic approach for targeting ovarian and endometrial cancers.(doi:10.1158/0008-5472.CAN-14-1337)

作者:网络 点击: