Cargando…

Hippocalcin-Like 1 blunts liver lipid metabolism to suppress tumorigenesis via directly targeting RUVBL1-mTOR signaling

Rationale: Hepatocellular carcinoma (HCC) is one of the most severe cancers worldwide, with few effective targeted therapies for HCC. Lipid metabolic reprogramming is emerged as a hallmark of cancer metabolism that guides response to antitumoral therapies. Such lipid metabolic alteration in cancers...

Descripción completa

Detalles Bibliográficos
Autores principales: Chen, Tao, Yuan, Zhiqing, Lei, Zhou, Duan, Jinlin, Xue, Junyan, Lu, Ting, Yan, Guouan, Zhang, Lei, Liu, Yanfeng, Li, Qiwei, Zhang, Yonglong
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Ivyspring International Publisher 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9691343/
https://www.ncbi.nlm.nih.gov/pubmed/36438486
http://dx.doi.org/10.7150/thno.75936
_version_ 1784837020451864576
author Chen, Tao
Yuan, Zhiqing
Lei, Zhou
Duan, Jinlin
Xue, Junyan
Lu, Ting
Yan, Guouan
Zhang, Lei
Liu, Yanfeng
Li, Qiwei
Zhang, Yonglong
author_facet Chen, Tao
Yuan, Zhiqing
Lei, Zhou
Duan, Jinlin
Xue, Junyan
Lu, Ting
Yan, Guouan
Zhang, Lei
Liu, Yanfeng
Li, Qiwei
Zhang, Yonglong
author_sort Chen, Tao
collection PubMed
description Rationale: Hepatocellular carcinoma (HCC) is one of the most severe cancers worldwide, with few effective targeted therapies for HCC. Lipid metabolic reprogramming is emerged as a hallmark of cancer metabolism that guides response to antitumoral therapies. Such lipid metabolic alteration in cancers is critically regulated by the mammalian target of rapamycin mTOR, which is considered as a promising therapeutic target. Despite efforts, mTOR inhibitors (mTORi) have produced limited response clinically, partly due to incomplete knowledge of mTORC1 addiction in cancers. Methods: CRISPR-Cas9 system was used to establish Hpcal1 null mice. The liver cancer model in mice was generated using Hpcal1-deficient mice with diethylnitrosamine (DEN) /CCL4 or MYC/Trp53(-/-) via hydrodynamic tail-vein injection. RNA-sequencing (RNA-seq) was used to identify potential signaling pathways. The expression of HPCAL1 and mTOR signaling were determined using quantitative polymerase chain reaction (qPCR), western blot and immunohistochemistry. The role of Hpcal1 in liver tumorigenesis and its response to mTORi was assessed by CCK-8 measurements, colony formation assay and in mouse model. Results: In this study, we identified hippocalcin-like protein 1 (HPCAL1) as an important negative regulator of de novo lipid biosynthesis and mTOR signaling activation, limiting liver tumorigenesis and establishing a metabolic vulnerability of HCC in mice. Genetic loss of HPCAL1 rendered HCC mTORC1-addicted and sensitive to mTORi AZD-8055 in vitro and in vivo. Importantly, HPCAL1 expression was inversely correlated with the levels of mTOR phosphorylation and several critical lipid biosynthesis enzymes in human specimens. Mechanistically, HPCAL1 directly bound to RuvB Like AAA ATPase 1 (RUVBL1), inhibiting the assembly of TEL2-TTI1-TTI2 (TTT)-RUVBL complex and subsequent leading the mTOR signaling suppression. Conclusion: We uncover a metabolic vulnerability and mTOR addiction in HCC with HPCAL1 loss that provides a selective therapeutic window for HCC with mTORC1 hyperactivation using mTORi.
format Online
Article
Text
id pubmed-9691343
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Ivyspring International Publisher
record_format MEDLINE/PubMed
spelling pubmed-96913432022-11-25 Hippocalcin-Like 1 blunts liver lipid metabolism to suppress tumorigenesis via directly targeting RUVBL1-mTOR signaling Chen, Tao Yuan, Zhiqing Lei, Zhou Duan, Jinlin Xue, Junyan Lu, Ting Yan, Guouan Zhang, Lei Liu, Yanfeng Li, Qiwei Zhang, Yonglong Theranostics Research Paper Rationale: Hepatocellular carcinoma (HCC) is one of the most severe cancers worldwide, with few effective targeted therapies for HCC. Lipid metabolic reprogramming is emerged as a hallmark of cancer metabolism that guides response to antitumoral therapies. Such lipid metabolic alteration in cancers is critically regulated by the mammalian target of rapamycin mTOR, which is considered as a promising therapeutic target. Despite efforts, mTOR inhibitors (mTORi) have produced limited response clinically, partly due to incomplete knowledge of mTORC1 addiction in cancers. Methods: CRISPR-Cas9 system was used to establish Hpcal1 null mice. The liver cancer model in mice was generated using Hpcal1-deficient mice with diethylnitrosamine (DEN) /CCL4 or MYC/Trp53(-/-) via hydrodynamic tail-vein injection. RNA-sequencing (RNA-seq) was used to identify potential signaling pathways. The expression of HPCAL1 and mTOR signaling were determined using quantitative polymerase chain reaction (qPCR), western blot and immunohistochemistry. The role of Hpcal1 in liver tumorigenesis and its response to mTORi was assessed by CCK-8 measurements, colony formation assay and in mouse model. Results: In this study, we identified hippocalcin-like protein 1 (HPCAL1) as an important negative regulator of de novo lipid biosynthesis and mTOR signaling activation, limiting liver tumorigenesis and establishing a metabolic vulnerability of HCC in mice. Genetic loss of HPCAL1 rendered HCC mTORC1-addicted and sensitive to mTORi AZD-8055 in vitro and in vivo. Importantly, HPCAL1 expression was inversely correlated with the levels of mTOR phosphorylation and several critical lipid biosynthesis enzymes in human specimens. Mechanistically, HPCAL1 directly bound to RuvB Like AAA ATPase 1 (RUVBL1), inhibiting the assembly of TEL2-TTI1-TTI2 (TTT)-RUVBL complex and subsequent leading the mTOR signaling suppression. Conclusion: We uncover a metabolic vulnerability and mTOR addiction in HCC with HPCAL1 loss that provides a selective therapeutic window for HCC with mTORC1 hyperactivation using mTORi. Ivyspring International Publisher 2022-10-24 /pmc/articles/PMC9691343/ /pubmed/36438486 http://dx.doi.org/10.7150/thno.75936 Text en © The author(s) https://creativecommons.org/licenses/by/4.0/This is an open access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/). See http://ivyspring.com/terms for full terms and conditions.
spellingShingle Research Paper
Chen, Tao
Yuan, Zhiqing
Lei, Zhou
Duan, Jinlin
Xue, Junyan
Lu, Ting
Yan, Guouan
Zhang, Lei
Liu, Yanfeng
Li, Qiwei
Zhang, Yonglong
Hippocalcin-Like 1 blunts liver lipid metabolism to suppress tumorigenesis via directly targeting RUVBL1-mTOR signaling
title Hippocalcin-Like 1 blunts liver lipid metabolism to suppress tumorigenesis via directly targeting RUVBL1-mTOR signaling
title_full Hippocalcin-Like 1 blunts liver lipid metabolism to suppress tumorigenesis via directly targeting RUVBL1-mTOR signaling
title_fullStr Hippocalcin-Like 1 blunts liver lipid metabolism to suppress tumorigenesis via directly targeting RUVBL1-mTOR signaling
title_full_unstemmed Hippocalcin-Like 1 blunts liver lipid metabolism to suppress tumorigenesis via directly targeting RUVBL1-mTOR signaling
title_short Hippocalcin-Like 1 blunts liver lipid metabolism to suppress tumorigenesis via directly targeting RUVBL1-mTOR signaling
title_sort hippocalcin-like 1 blunts liver lipid metabolism to suppress tumorigenesis via directly targeting ruvbl1-mtor signaling
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9691343/
https://www.ncbi.nlm.nih.gov/pubmed/36438486
http://dx.doi.org/10.7150/thno.75936
work_keys_str_mv AT chentao hippocalcinlike1bluntsliverlipidmetabolismtosuppresstumorigenesisviadirectlytargetingruvbl1mtorsignaling
AT yuanzhiqing hippocalcinlike1bluntsliverlipidmetabolismtosuppresstumorigenesisviadirectlytargetingruvbl1mtorsignaling
AT leizhou hippocalcinlike1bluntsliverlipidmetabolismtosuppresstumorigenesisviadirectlytargetingruvbl1mtorsignaling
AT duanjinlin hippocalcinlike1bluntsliverlipidmetabolismtosuppresstumorigenesisviadirectlytargetingruvbl1mtorsignaling
AT xuejunyan hippocalcinlike1bluntsliverlipidmetabolismtosuppresstumorigenesisviadirectlytargetingruvbl1mtorsignaling
AT luting hippocalcinlike1bluntsliverlipidmetabolismtosuppresstumorigenesisviadirectlytargetingruvbl1mtorsignaling
AT yanguouan hippocalcinlike1bluntsliverlipidmetabolismtosuppresstumorigenesisviadirectlytargetingruvbl1mtorsignaling
AT zhanglei hippocalcinlike1bluntsliverlipidmetabolismtosuppresstumorigenesisviadirectlytargetingruvbl1mtorsignaling
AT liuyanfeng hippocalcinlike1bluntsliverlipidmetabolismtosuppresstumorigenesisviadirectlytargetingruvbl1mtorsignaling
AT liqiwei hippocalcinlike1bluntsliverlipidmetabolismtosuppresstumorigenesisviadirectlytargetingruvbl1mtorsignaling
AT zhangyonglong hippocalcinlike1bluntsliverlipidmetabolismtosuppresstumorigenesisviadirectlytargetingruvbl1mtorsignaling