Nonhormonal therapy for endometriosis based on energy metabolism regulation.

Reproduction & Fertility Pub Date : 2021-11-25 eCollection Date: 2021-12-01 DOI:10.1530/RAF-21-0053
Hiroshi Kobayashi, Hiroshi Shigetomi, Shogo Imanaka
{"title":"Nonhormonal therapy for endometriosis based on energy metabolism regulation.","authors":"Hiroshi Kobayashi,&nbsp;Hiroshi Shigetomi,&nbsp;Shogo Imanaka","doi":"10.1530/RAF-21-0053","DOIUrl":null,"url":null,"abstract":"<p><p>Ovarian function suppression is the current pharmacotherapy of endometriosis with limited benefit and adverse effects. New therapeutic strategies other than hormonal therapy are developed based on the molecular mechanisms involved in the hypoxic and oxidative stress environments and metabolism unique to endometriosis. A literature search was performed between January 2000 and March 2021 in the PubMed database using a combination of specific terms. Endometriosis-associated metabolic changes have been organized into four hallmarks: (1) glucose uptake, (2) aerobic glycolysis, (3) lactate production and accumulation, and (4) metabolic conversion from mitochondrial oxidative phosphorylation (OXPHOS) to aerobic glycolysis. Endometriotic cells favor glycolytic metabolism over mitochondrial OXPHOS to produce essential energy for cell survival. Hypoxia, a common feature of the endometriosis environment, is a key player in this metabolic conversion, which may lead to glucose transporter overexpression, pyruvate dehydrogenase kinase 1 (PDK1) and lactate dehydrogenase kinase A (LDHA) activation, and pyruvate dehydrogenase complex inactivation. Evading mitochondrial OXPHOS mitigates excessive generation of reactive oxygen species (ROS) that may trigger cell death. Therefore, the coinactivation of LDHA and PDK1 can induce the accumulation of mitochondrial ROS by converting energy metabolism to mitochondrial OXPHOS, causing endometriotic cell death. Metabolic pattern reconstruction in endometriotic lesions is a critical factor in cell survival and disease progression. One therapeutic strategy that may avoid hormone manipulation is focused on mitigating metabolic changes that have been detected in cells/tissues from women with endometriosis.</p><p><strong>Lay summary: </strong>The most commonly used medical therapies for endometriosis have contraceptives and other side effects associated with hormone suppression and are therefore unsuitable for women desiring pregnancy. One therapeutic strategy that may avoid hormone manipulation is focused on changing metabolic profiles that have been detected in cells/tissues from women with endometriosis. Endometriotic cells favor glycolytic metabolism over mitochondrial oxidative phosphorylation (OXPHOS) to produce essential energy for cell growth. Furthermore, the metabolic conversion from mitochondrial OXPHOS to aerobic glycolysis suppresses cell death through the reduced generation of reactive oxygen species (ROS). This unique metabolic feature of endometriosis is important for cell survival and disease progression. Thus, changing the specific metabolic switch may increase mitochondrial ROS production, causing severe oxidative stress and cell death. This review describes new treatments by changing the metabolic profiles of endometriosis.</p>","PeriodicalId":21128,"journal":{"name":"Reproduction & Fertility","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2021-11-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8788578/pdf/","citationCount":"10","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Reproduction & Fertility","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.1530/RAF-21-0053","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"2021/12/1 0:00:00","PubModel":"eCollection","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 10

Abstract

Ovarian function suppression is the current pharmacotherapy of endometriosis with limited benefit and adverse effects. New therapeutic strategies other than hormonal therapy are developed based on the molecular mechanisms involved in the hypoxic and oxidative stress environments and metabolism unique to endometriosis. A literature search was performed between January 2000 and March 2021 in the PubMed database using a combination of specific terms. Endometriosis-associated metabolic changes have been organized into four hallmarks: (1) glucose uptake, (2) aerobic glycolysis, (3) lactate production and accumulation, and (4) metabolic conversion from mitochondrial oxidative phosphorylation (OXPHOS) to aerobic glycolysis. Endometriotic cells favor glycolytic metabolism over mitochondrial OXPHOS to produce essential energy for cell survival. Hypoxia, a common feature of the endometriosis environment, is a key player in this metabolic conversion, which may lead to glucose transporter overexpression, pyruvate dehydrogenase kinase 1 (PDK1) and lactate dehydrogenase kinase A (LDHA) activation, and pyruvate dehydrogenase complex inactivation. Evading mitochondrial OXPHOS mitigates excessive generation of reactive oxygen species (ROS) that may trigger cell death. Therefore, the coinactivation of LDHA and PDK1 can induce the accumulation of mitochondrial ROS by converting energy metabolism to mitochondrial OXPHOS, causing endometriotic cell death. Metabolic pattern reconstruction in endometriotic lesions is a critical factor in cell survival and disease progression. One therapeutic strategy that may avoid hormone manipulation is focused on mitigating metabolic changes that have been detected in cells/tissues from women with endometriosis.

Lay summary: The most commonly used medical therapies for endometriosis have contraceptives and other side effects associated with hormone suppression and are therefore unsuitable for women desiring pregnancy. One therapeutic strategy that may avoid hormone manipulation is focused on changing metabolic profiles that have been detected in cells/tissues from women with endometriosis. Endometriotic cells favor glycolytic metabolism over mitochondrial oxidative phosphorylation (OXPHOS) to produce essential energy for cell growth. Furthermore, the metabolic conversion from mitochondrial OXPHOS to aerobic glycolysis suppresses cell death through the reduced generation of reactive oxygen species (ROS). This unique metabolic feature of endometriosis is important for cell survival and disease progression. Thus, changing the specific metabolic switch may increase mitochondrial ROS production, causing severe oxidative stress and cell death. This review describes new treatments by changing the metabolic profiles of endometriosis.

Abstract Image

Abstract Image

Abstract Image

基于能量代谢调节的子宫内膜异位症非激素治疗。
卵巢功能抑制是目前治疗子宫内膜异位症的药物治疗方法,但疗效有限,副作用大。新的治疗策略,而不是激素治疗基于分子机制参与缺氧和氧化应激环境和代谢独特的子宫内膜异位症。在2000年1月至2021年3月期间,使用特定术语组合在PubMed数据库中进行了文献检索。子宫内膜异位症相关的代谢变化有四个标志:(1)葡萄糖摄取,(2)有氧糖酵解,(3)乳酸的产生和积累,以及(4)线粒体氧化磷酸化(OXPHOS)向有氧糖酵解的代谢转化。子宫内膜异位症细胞倾向于糖酵解代谢,而不是线粒体OXPHOS,以产生细胞生存所必需的能量。缺氧是子宫内膜异位症环境的一个共同特征,是这种代谢转化的关键参与者,它可能导致葡萄糖转运蛋白过度表达,丙酮酸脱氢酶激酶1 (PDK1)和乳酸脱氢酶激酶a (LDHA)激活,丙酮酸脱氢酶复合物失活。避免线粒体OXPHOS可减轻可能引发细胞死亡的活性氧(ROS)的过度产生。因此,LDHA和PDK1的共激活可以通过将能量代谢转化为线粒体OXPHOS,诱导线粒体ROS的积累,导致子宫内膜异位症细胞死亡。子宫内膜异位症病变的代谢模式重建是细胞存活和疾病进展的关键因素。一种可能避免激素操纵的治疗策略是专注于减轻在子宫内膜异位症女性细胞/组织中检测到的代谢变化。总结:最常用的治疗子宫内膜异位症的药物有避孕药和其他与激素抑制相关的副作用,因此不适合想怀孕的女性。一种可能避免激素操纵的治疗策略是专注于改变已在子宫内膜异位症妇女的细胞/组织中检测到的代谢谱。子宫内膜异位症细胞倾向于糖酵解代谢而不是线粒体氧化磷酸化(OXPHOS)来产生细胞生长所需的能量。此外,从线粒体OXPHOS到有氧糖酵解的代谢转化通过减少活性氧(ROS)的产生来抑制细胞死亡。子宫内膜异位症这种独特的代谢特征对细胞存活和疾病进展非常重要。因此,改变特定的代谢开关可能会增加线粒体ROS的产生,导致严重的氧化应激和细胞死亡。本文综述了改变子宫内膜异位症代谢谱的新治疗方法。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 求助全文
来源期刊
自引率
0.00%
发文量
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信