Cargando…

Targeting CPT1A-mediated fatty acid oxidation sensitizes nasopharyngeal carcinoma to radiation therapy

Nasopharyngeal carcinoma (NPC) has a particularly high prevalence in southern China, southeastern Asia and northern Africa. Radiation resistance remains a serious obstacle to successful treatment in NPC. This study aimed to explore the metabolic feature of radiation-resistant NPC cells and identify...

Descripción completa

Detalles Bibliográficos
Autores principales: Tan, Zheqiong, Xiao, Lanbo, Tang, Min, Bai, Fang, Li, Jiangjiang, Li, Liling, Shi, Feng, Li, Namei, Li, Yueshuo, Du, Qianqian, Lu, Jingchen, Weng, Xinxian, Yi, Wei, Zhang, Hanwen, Fan, Jia, Zhou, Jian, Gao, Qiang, Onuchic, José N., Bode, Ann M., Luo, Xiangjian, Cao, Ya
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Ivyspring International Publisher 2018
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5928893/
https://www.ncbi.nlm.nih.gov/pubmed/29721083
http://dx.doi.org/10.7150/thno.21451
_version_ 1783319316110770176
author Tan, Zheqiong
Xiao, Lanbo
Tang, Min
Bai, Fang
Li, Jiangjiang
Li, Liling
Shi, Feng
Li, Namei
Li, Yueshuo
Du, Qianqian
Lu, Jingchen
Weng, Xinxian
Yi, Wei
Zhang, Hanwen
Fan, Jia
Zhou, Jian
Gao, Qiang
Onuchic, José N.
Bode, Ann M.
Luo, Xiangjian
Cao, Ya
author_facet Tan, Zheqiong
Xiao, Lanbo
Tang, Min
Bai, Fang
Li, Jiangjiang
Li, Liling
Shi, Feng
Li, Namei
Li, Yueshuo
Du, Qianqian
Lu, Jingchen
Weng, Xinxian
Yi, Wei
Zhang, Hanwen
Fan, Jia
Zhou, Jian
Gao, Qiang
Onuchic, José N.
Bode, Ann M.
Luo, Xiangjian
Cao, Ya
author_sort Tan, Zheqiong
collection PubMed
description Nasopharyngeal carcinoma (NPC) has a particularly high prevalence in southern China, southeastern Asia and northern Africa. Radiation resistance remains a serious obstacle to successful treatment in NPC. This study aimed to explore the metabolic feature of radiation-resistant NPC cells and identify new molecular-targeted agents to improve the therapeutic effects of radiotherapy in NPC. Methods: Radiation-responsive and radiation-resistant NPC cells were used as the model system in vitro and in vivo. Metabolomics approach was used to illustrate the global metabolic changes. (13)C isotopomer tracing experiment and Seahorse XF analysis were undertaken to determine the activity of fatty acid oxidation (FAO). qRT-PCR was performed to evaluate the expression of essential FAO genes including CPT1A. NPC tumor tissue microarray was used to investigate the prognostic role of CPT1A. Either RNA interference or pharmacological blockade by Etomoxir were used to inhibit CPT1A. Radiation resistance was evaluated by colony formation assay. Mitochondrial membrane potential, apoptosis and neutral lipid content were measured by flow cytometry analysis using JC-1, Annexin V and LipidTOX Red probe respectively. Molecular markers of mitochondrial apoptosis were detected by western blot. Xenografts were treated with Etomoxir, radiation, or a combination of Etomoxir and radiation. Mitochondrial apoptosis and lipid droplets content of tumor tissues were detected by cleaved caspase 9 and Oil Red O staining respectively. Liquid chromatography coupled with tandem mass spectrometry approach was used to identify CPT1A-binding proteins. The interaction of CPT1A and Rab14 were detected by immunoprecipitation, immunofluorescence and in situ proximity ligation analysis. Fragment docking and direct coupling combined computational protein-protein interaction prediction method were used to predict the binding interface. Fatty acid trafficking was measured by pulse-chase assay using BODIPY C16 and MitoTracker Red probe. Results: FAO was active in radiation-resistant NPC cells, and the rate-limiting enzyme of FAO, carnitine palmitoyl transferase 1 A (CPT1A), was consistently up-regulated in these cells. The protein level of CPT1A was significantly associated with poor overall survival of NPC patients following radiotherapy. Inhibition of CPT1A re-sensitized NPC cells to radiation therapy by activating mitochondrial apoptosis both in vitro and in vivo. In addition, we identified Rab14 as a novel CPT1A binding protein. The CPT1A-Rab14 interaction facilitated fatty acid trafficking from lipid droplets to mitochondria, which decreased radiation-induced lipid accumulation and maximized ATP production. Knockdown of Rab14 attenuated CPT1A-mediated fatty acid trafficking and radiation resistance. Conclusion: An active FAO is a vital signature of NPC radiation resistance. Targeting CPT1A could be a beneficial regimen to improve the therapeutic effects of radiotherapy in NPC patients. Importantly, the CPT1A-Rab14 interaction plays roles in CPT1A-mediated radiation resistance by facilitating fatty acid trafficking. This interaction could be an attractive interface for the discovery of novel CPT1A inhibitors.
format Online
Article
Text
id pubmed-5928893
institution National Center for Biotechnology Information
language English
publishDate 2018
publisher Ivyspring International Publisher
record_format MEDLINE/PubMed
spelling pubmed-59288932018-05-02 Targeting CPT1A-mediated fatty acid oxidation sensitizes nasopharyngeal carcinoma to radiation therapy Tan, Zheqiong Xiao, Lanbo Tang, Min Bai, Fang Li, Jiangjiang Li, Liling Shi, Feng Li, Namei Li, Yueshuo Du, Qianqian Lu, Jingchen Weng, Xinxian Yi, Wei Zhang, Hanwen Fan, Jia Zhou, Jian Gao, Qiang Onuchic, José N. Bode, Ann M. Luo, Xiangjian Cao, Ya Theranostics Research Paper Nasopharyngeal carcinoma (NPC) has a particularly high prevalence in southern China, southeastern Asia and northern Africa. Radiation resistance remains a serious obstacle to successful treatment in NPC. This study aimed to explore the metabolic feature of radiation-resistant NPC cells and identify new molecular-targeted agents to improve the therapeutic effects of radiotherapy in NPC. Methods: Radiation-responsive and radiation-resistant NPC cells were used as the model system in vitro and in vivo. Metabolomics approach was used to illustrate the global metabolic changes. (13)C isotopomer tracing experiment and Seahorse XF analysis were undertaken to determine the activity of fatty acid oxidation (FAO). qRT-PCR was performed to evaluate the expression of essential FAO genes including CPT1A. NPC tumor tissue microarray was used to investigate the prognostic role of CPT1A. Either RNA interference or pharmacological blockade by Etomoxir were used to inhibit CPT1A. Radiation resistance was evaluated by colony formation assay. Mitochondrial membrane potential, apoptosis and neutral lipid content were measured by flow cytometry analysis using JC-1, Annexin V and LipidTOX Red probe respectively. Molecular markers of mitochondrial apoptosis were detected by western blot. Xenografts were treated with Etomoxir, radiation, or a combination of Etomoxir and radiation. Mitochondrial apoptosis and lipid droplets content of tumor tissues were detected by cleaved caspase 9 and Oil Red O staining respectively. Liquid chromatography coupled with tandem mass spectrometry approach was used to identify CPT1A-binding proteins. The interaction of CPT1A and Rab14 were detected by immunoprecipitation, immunofluorescence and in situ proximity ligation analysis. Fragment docking and direct coupling combined computational protein-protein interaction prediction method were used to predict the binding interface. Fatty acid trafficking was measured by pulse-chase assay using BODIPY C16 and MitoTracker Red probe. Results: FAO was active in radiation-resistant NPC cells, and the rate-limiting enzyme of FAO, carnitine palmitoyl transferase 1 A (CPT1A), was consistently up-regulated in these cells. The protein level of CPT1A was significantly associated with poor overall survival of NPC patients following radiotherapy. Inhibition of CPT1A re-sensitized NPC cells to radiation therapy by activating mitochondrial apoptosis both in vitro and in vivo. In addition, we identified Rab14 as a novel CPT1A binding protein. The CPT1A-Rab14 interaction facilitated fatty acid trafficking from lipid droplets to mitochondria, which decreased radiation-induced lipid accumulation and maximized ATP production. Knockdown of Rab14 attenuated CPT1A-mediated fatty acid trafficking and radiation resistance. Conclusion: An active FAO is a vital signature of NPC radiation resistance. Targeting CPT1A could be a beneficial regimen to improve the therapeutic effects of radiotherapy in NPC patients. Importantly, the CPT1A-Rab14 interaction plays roles in CPT1A-mediated radiation resistance by facilitating fatty acid trafficking. This interaction could be an attractive interface for the discovery of novel CPT1A inhibitors. Ivyspring International Publisher 2018-03-22 /pmc/articles/PMC5928893/ /pubmed/29721083 http://dx.doi.org/10.7150/thno.21451 Text en © Ivyspring International Publisher This is an open access article distributed under the terms of the Creative Commons Attribution (CC BY-NC) license (https://creativecommons.org/licenses/by-nc/4.0/). See http://ivyspring.com/terms for full terms and conditions.
spellingShingle Research Paper
Tan, Zheqiong
Xiao, Lanbo
Tang, Min
Bai, Fang
Li, Jiangjiang
Li, Liling
Shi, Feng
Li, Namei
Li, Yueshuo
Du, Qianqian
Lu, Jingchen
Weng, Xinxian
Yi, Wei
Zhang, Hanwen
Fan, Jia
Zhou, Jian
Gao, Qiang
Onuchic, José N.
Bode, Ann M.
Luo, Xiangjian
Cao, Ya
Targeting CPT1A-mediated fatty acid oxidation sensitizes nasopharyngeal carcinoma to radiation therapy
title Targeting CPT1A-mediated fatty acid oxidation sensitizes nasopharyngeal carcinoma to radiation therapy
title_full Targeting CPT1A-mediated fatty acid oxidation sensitizes nasopharyngeal carcinoma to radiation therapy
title_fullStr Targeting CPT1A-mediated fatty acid oxidation sensitizes nasopharyngeal carcinoma to radiation therapy
title_full_unstemmed Targeting CPT1A-mediated fatty acid oxidation sensitizes nasopharyngeal carcinoma to radiation therapy
title_short Targeting CPT1A-mediated fatty acid oxidation sensitizes nasopharyngeal carcinoma to radiation therapy
title_sort targeting cpt1a-mediated fatty acid oxidation sensitizes nasopharyngeal carcinoma to radiation therapy
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5928893/
https://www.ncbi.nlm.nih.gov/pubmed/29721083
http://dx.doi.org/10.7150/thno.21451
work_keys_str_mv AT tanzheqiong targetingcpt1amediatedfattyacidoxidationsensitizesnasopharyngealcarcinomatoradiationtherapy
AT xiaolanbo targetingcpt1amediatedfattyacidoxidationsensitizesnasopharyngealcarcinomatoradiationtherapy
AT tangmin targetingcpt1amediatedfattyacidoxidationsensitizesnasopharyngealcarcinomatoradiationtherapy
AT baifang targetingcpt1amediatedfattyacidoxidationsensitizesnasopharyngealcarcinomatoradiationtherapy
AT lijiangjiang targetingcpt1amediatedfattyacidoxidationsensitizesnasopharyngealcarcinomatoradiationtherapy
AT lililing targetingcpt1amediatedfattyacidoxidationsensitizesnasopharyngealcarcinomatoradiationtherapy
AT shifeng targetingcpt1amediatedfattyacidoxidationsensitizesnasopharyngealcarcinomatoradiationtherapy
AT linamei targetingcpt1amediatedfattyacidoxidationsensitizesnasopharyngealcarcinomatoradiationtherapy
AT liyueshuo targetingcpt1amediatedfattyacidoxidationsensitizesnasopharyngealcarcinomatoradiationtherapy
AT duqianqian targetingcpt1amediatedfattyacidoxidationsensitizesnasopharyngealcarcinomatoradiationtherapy
AT lujingchen targetingcpt1amediatedfattyacidoxidationsensitizesnasopharyngealcarcinomatoradiationtherapy
AT wengxinxian targetingcpt1amediatedfattyacidoxidationsensitizesnasopharyngealcarcinomatoradiationtherapy
AT yiwei targetingcpt1amediatedfattyacidoxidationsensitizesnasopharyngealcarcinomatoradiationtherapy
AT zhanghanwen targetingcpt1amediatedfattyacidoxidationsensitizesnasopharyngealcarcinomatoradiationtherapy
AT fanjia targetingcpt1amediatedfattyacidoxidationsensitizesnasopharyngealcarcinomatoradiationtherapy
AT zhoujian targetingcpt1amediatedfattyacidoxidationsensitizesnasopharyngealcarcinomatoradiationtherapy
AT gaoqiang targetingcpt1amediatedfattyacidoxidationsensitizesnasopharyngealcarcinomatoradiationtherapy
AT onuchicjosen targetingcpt1amediatedfattyacidoxidationsensitizesnasopharyngealcarcinomatoradiationtherapy
AT bodeannm targetingcpt1amediatedfattyacidoxidationsensitizesnasopharyngealcarcinomatoradiationtherapy
AT luoxiangjian targetingcpt1amediatedfattyacidoxidationsensitizesnasopharyngealcarcinomatoradiationtherapy
AT caoya targetingcpt1amediatedfattyacidoxidationsensitizesnasopharyngealcarcinomatoradiationtherapy