鞘氨醇激酶 2 和 p62 的调控是肝细胞癌性别二态性的决定因素

IF 7 2区 医学 Q1 ENDOCRINOLOGY & METABOLISM Molecular Metabolism Pub Date : 2024-06-24 DOI:10.1016/j.molmet.2024.101971
Christopher D. Green , Ryan D.R. Brown , Baasanjav Uranbileg , Cynthia Weigel , Sumit Saha , Makoto Kurano , Yutaka Yatomi , Sarah Spiegel
{"title":"鞘氨醇激酶 2 和 p62 的调控是肝细胞癌性别二态性的决定因素","authors":"Christopher D. Green ,&nbsp;Ryan D.R. Brown ,&nbsp;Baasanjav Uranbileg ,&nbsp;Cynthia Weigel ,&nbsp;Sumit Saha ,&nbsp;Makoto Kurano ,&nbsp;Yutaka Yatomi ,&nbsp;Sarah Spiegel","doi":"10.1016/j.molmet.2024.101971","DOIUrl":null,"url":null,"abstract":"<div><h3>Objective</h3><p>Hepatocellular carcinoma (HCC) is the third leading cause of cancer mortality, and its incidence is increasing due to endemic obesity. HCC is sexually dimorphic in both humans and rodents with higher incidence in males, although the mechanisms contributing to these correlations remain unclear. Here, we examined the role of sphingosine kinase 2 (SphK2), the enzyme that regulates the balance of bioactive sphingolipid metabolites, sphingosine-1-phosphate (S1P) and ceramide, in gender specific MASH-driven HCC.</p></div><div><h3>Methods</h3><p>Male and female mice were fed a high fat diet with sugar water, a clinically relevant model that recapitulates MASH-driven HCC in humans followed by physiological, biochemical cellular and molecular analyses. In addition, correlations with increased risk of HCC recurrence were determined in patients.</p></div><div><h3>Results</h3><p>Here, we report that deletion of SphK2 protects both male and female mice from Western diet-induced weight gain and metabolic dysfunction without affecting hepatic lipid accumulation or fibrosis. However, SphK2 deficiency decreases chronic diet-induced hepatocyte proliferation in males but increases it in females. Remarkably, SphK2 deficiency reverses the sexual dimorphism of HCC, as SphK2<sup>−/−</sup> male mice are protected whereas the females develop liver cancer. Only in male mice, chronic western diet induced accumulation of the autophagy receptor p62 and its downstream mediators, the antioxidant response target NQO1, and the oncogene c-Myc. SphK2 deletion repressed these known drivers of HCC development. Moreover, high p62 expression correlates with poor survival in male HCC patients but not in females. In hepatocytes, lipotoxicity-induced p62 accumulation is regulated by sex hormones and prevented by SphK2 deletion. Importantly, high SphK2 expression in male but not female HCC patients is associated with a more aggressive HCC differentiation status and increased risk of cancer recurrence.</p></div><div><h3>Conclusions</h3><p>This work identifies SphK2 as a potential regulator of HCC sexual dimorphism and suggests SphK2 inhibitors now in clinical trials could have opposing, gender-specific effects in patients.</p></div>","PeriodicalId":18765,"journal":{"name":"Molecular Metabolism","volume":null,"pages":null},"PeriodicalIF":7.0000,"publicationDate":"2024-06-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2212877824001029/pdfft?md5=2a1ec55c7d5849dc7d2af6fd26836662&pid=1-s2.0-S2212877824001029-main.pdf","citationCount":"0","resultStr":"{\"title\":\"Sphingosine kinase 2 and p62 regulation are determinants of sexual dimorphism in hepatocellular carcinoma\",\"authors\":\"Christopher D. Green ,&nbsp;Ryan D.R. Brown ,&nbsp;Baasanjav Uranbileg ,&nbsp;Cynthia Weigel ,&nbsp;Sumit Saha ,&nbsp;Makoto Kurano ,&nbsp;Yutaka Yatomi ,&nbsp;Sarah Spiegel\",\"doi\":\"10.1016/j.molmet.2024.101971\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<div><h3>Objective</h3><p>Hepatocellular carcinoma (HCC) is the third leading cause of cancer mortality, and its incidence is increasing due to endemic obesity. HCC is sexually dimorphic in both humans and rodents with higher incidence in males, although the mechanisms contributing to these correlations remain unclear. Here, we examined the role of sphingosine kinase 2 (SphK2), the enzyme that regulates the balance of bioactive sphingolipid metabolites, sphingosine-1-phosphate (S1P) and ceramide, in gender specific MASH-driven HCC.</p></div><div><h3>Methods</h3><p>Male and female mice were fed a high fat diet with sugar water, a clinically relevant model that recapitulates MASH-driven HCC in humans followed by physiological, biochemical cellular and molecular analyses. In addition, correlations with increased risk of HCC recurrence were determined in patients.</p></div><div><h3>Results</h3><p>Here, we report that deletion of SphK2 protects both male and female mice from Western diet-induced weight gain and metabolic dysfunction without affecting hepatic lipid accumulation or fibrosis. However, SphK2 deficiency decreases chronic diet-induced hepatocyte proliferation in males but increases it in females. Remarkably, SphK2 deficiency reverses the sexual dimorphism of HCC, as SphK2<sup>−/−</sup> male mice are protected whereas the females develop liver cancer. Only in male mice, chronic western diet induced accumulation of the autophagy receptor p62 and its downstream mediators, the antioxidant response target NQO1, and the oncogene c-Myc. SphK2 deletion repressed these known drivers of HCC development. Moreover, high p62 expression correlates with poor survival in male HCC patients but not in females. In hepatocytes, lipotoxicity-induced p62 accumulation is regulated by sex hormones and prevented by SphK2 deletion. Importantly, high SphK2 expression in male but not female HCC patients is associated with a more aggressive HCC differentiation status and increased risk of cancer recurrence.</p></div><div><h3>Conclusions</h3><p>This work identifies SphK2 as a potential regulator of HCC sexual dimorphism and suggests SphK2 inhibitors now in clinical trials could have opposing, gender-specific effects in patients.</p></div>\",\"PeriodicalId\":18765,\"journal\":{\"name\":\"Molecular Metabolism\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":7.0000,\"publicationDate\":\"2024-06-24\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.sciencedirect.com/science/article/pii/S2212877824001029/pdfft?md5=2a1ec55c7d5849dc7d2af6fd26836662&pid=1-s2.0-S2212877824001029-main.pdf\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Molecular Metabolism\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://www.sciencedirect.com/science/article/pii/S2212877824001029\",\"RegionNum\":2,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q1\",\"JCRName\":\"ENDOCRINOLOGY & METABOLISM\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Molecular Metabolism","FirstCategoryId":"3","ListUrlMain":"https://www.sciencedirect.com/science/article/pii/S2212877824001029","RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"ENDOCRINOLOGY & METABOLISM","Score":null,"Total":0}
引用次数: 0

摘要

目的:肝细胞癌(HCC)是导致癌症死亡的第三大原因,其发病率因地方性肥胖而不断上升。在人类和啮齿类动物中,HCC 具有性别二形性,雄性的发病率更高,但导致这些相关性的机制仍不清楚。在此,我们研究了鞘氨醇激酶 2(SphK2)--一种调节生物活性鞘脂代谢物(鞘氨醇-1-磷酸(S1P)和神经酰胺)平衡的酶--在性别特异性 MASH 驱动的 HCC 中的作用:雄性和雌性小鼠均以高脂肪饮食加糖水喂养,这种临床相关模型再现了人类 MASH 驱动的 HCC,随后进行了生理、生化细胞和分子分析。此外,还确定了患者 HCC 复发风险增加的相关性:我们在此报告,SphK2 的缺失可保护雄性和雌性小鼠免受西方饮食引起的体重增加和代谢功能障碍的影响,而不会影响肝脏脂质积累或纤维化。然而,SphK2 的缺乏会减少慢性饮食诱导的雄性肝细胞增殖,但会增加雌性肝细胞增殖。值得注意的是,SphK2 的缺乏会逆转 HCC 的性别二态性,因为 SphK2-/-雄性小鼠受到保护,而雌性小鼠则会发展成肝癌。只有在雄性小鼠中,长期西式饮食才会诱导自噬受体p62及其下游介质、抗氧化反应靶标NQO1和癌基因c-Myc的积累。SphK2的缺失抑制了这些已知的HCC发展驱动因素。此外,p62的高表达与男性HCC患者的存活率低有关,而与女性患者的存活率无关。在肝细胞中,脂肪毒性诱导的 p62 积累受性激素调节,而 SphK2 缺失则可阻止这种积累。重要的是,SphK2在男性而非女性HCC患者中的高表达与更具侵袭性的HCC分化状态和癌症复发风险增加有关:结论:这项研究发现 SphK2 是 HCC 性双态性的潜在调节因子,并表明目前正在进行临床试验的 SphK2 抑制剂可能会对患者产生对立的、有性别特异性的影响。
本文章由计算机程序翻译,如有差异,请以英文原文为准。

摘要图片

查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
Sphingosine kinase 2 and p62 regulation are determinants of sexual dimorphism in hepatocellular carcinoma

Objective

Hepatocellular carcinoma (HCC) is the third leading cause of cancer mortality, and its incidence is increasing due to endemic obesity. HCC is sexually dimorphic in both humans and rodents with higher incidence in males, although the mechanisms contributing to these correlations remain unclear. Here, we examined the role of sphingosine kinase 2 (SphK2), the enzyme that regulates the balance of bioactive sphingolipid metabolites, sphingosine-1-phosphate (S1P) and ceramide, in gender specific MASH-driven HCC.

Methods

Male and female mice were fed a high fat diet with sugar water, a clinically relevant model that recapitulates MASH-driven HCC in humans followed by physiological, biochemical cellular and molecular analyses. In addition, correlations with increased risk of HCC recurrence were determined in patients.

Results

Here, we report that deletion of SphK2 protects both male and female mice from Western diet-induced weight gain and metabolic dysfunction without affecting hepatic lipid accumulation or fibrosis. However, SphK2 deficiency decreases chronic diet-induced hepatocyte proliferation in males but increases it in females. Remarkably, SphK2 deficiency reverses the sexual dimorphism of HCC, as SphK2−/− male mice are protected whereas the females develop liver cancer. Only in male mice, chronic western diet induced accumulation of the autophagy receptor p62 and its downstream mediators, the antioxidant response target NQO1, and the oncogene c-Myc. SphK2 deletion repressed these known drivers of HCC development. Moreover, high p62 expression correlates with poor survival in male HCC patients but not in females. In hepatocytes, lipotoxicity-induced p62 accumulation is regulated by sex hormones and prevented by SphK2 deletion. Importantly, high SphK2 expression in male but not female HCC patients is associated with a more aggressive HCC differentiation status and increased risk of cancer recurrence.

Conclusions

This work identifies SphK2 as a potential regulator of HCC sexual dimorphism and suggests SphK2 inhibitors now in clinical trials could have opposing, gender-specific effects in patients.

求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
Molecular Metabolism
Molecular Metabolism ENDOCRINOLOGY & METABOLISM-
CiteScore
14.50
自引率
2.50%
发文量
219
审稿时长
43 days
期刊介绍: Molecular Metabolism is a leading journal dedicated to sharing groundbreaking discoveries in the field of energy homeostasis and the underlying factors of metabolic disorders. These disorders include obesity, diabetes, cardiovascular disease, and cancer. Our journal focuses on publishing research driven by hypotheses and conducted to the highest standards, aiming to provide a mechanistic understanding of energy homeostasis-related behavior, physiology, and dysfunction. We promote interdisciplinary science, covering a broad range of approaches from molecules to humans throughout the lifespan. Our goal is to contribute to transformative research in metabolism, which has the potential to revolutionize the field. By enabling progress in the prognosis, prevention, and ultimately the cure of metabolic disorders and their long-term complications, our journal seeks to better the future of health and well-being.
期刊最新文献
A novel long non-coding RNA connects obesity to impaired adipocyte function. Hepatic Retinol Dehydrogenase 11 Dampens Stress Associated with the Maintenance of Cellular Cholesterol Levels. The metabolic sensor AMPK: Twelve enzymes in one. miR-10a regulates cell death and inflammation in adipose tissue of male mice with diet-induced obesity. Identifying targetable metabolic dependencies across colorectal cancer progression.
×
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
现在去查看 取消
×
提示
确定
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 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 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1