Cargando…

Visualization of endogenous p27 and Ki67 reveals the importance of a c-Myc-driven metabolic switch in promoting survival of quiescent cancer cells

Rationale: Recurrent and metastatic cancers often undergo a period of dormancy, which is closely associated with cellular quiescence, a state whereby cells exit the cell cycle and are reversibly arrested in G0 phase. Curative cancer treatment thus requires therapies that either sustain the dormant s...

Descripción completa

Detalles Bibliográficos
Autores principales: La, Ting, Chen, Song, Guo, Tao, Zhao, Xiao Hong, Teng, Liu, Li, Dandan, Carnell, Michael, Zhang, Yuan Yuan, Feng, Yu Chen, Cole, Nicole, Brown, Alexandra C., Zhang, Didi, Dong, Qihan, Wang, Jenny Y., Cao, Huixia, Liu, Tao, Thorne, Rick F., Shao, Feng-Min, Zhang, Xu Dong, Jin, Lei
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Ivyspring International Publisher 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8490506/
https://www.ncbi.nlm.nih.gov/pubmed/34646389
http://dx.doi.org/10.7150/thno.63763
_version_ 1784578538788093952
author La, Ting
Chen, Song
Guo, Tao
Zhao, Xiao Hong
Teng, Liu
Li, Dandan
Carnell, Michael
Zhang, Yuan Yuan
Feng, Yu Chen
Cole, Nicole
Brown, Alexandra C.
Zhang, Didi
Dong, Qihan
Wang, Jenny Y.
Cao, Huixia
Liu, Tao
Thorne, Rick F.
Shao, Feng-Min
Zhang, Xu Dong
Jin, Lei
author_facet La, Ting
Chen, Song
Guo, Tao
Zhao, Xiao Hong
Teng, Liu
Li, Dandan
Carnell, Michael
Zhang, Yuan Yuan
Feng, Yu Chen
Cole, Nicole
Brown, Alexandra C.
Zhang, Didi
Dong, Qihan
Wang, Jenny Y.
Cao, Huixia
Liu, Tao
Thorne, Rick F.
Shao, Feng-Min
Zhang, Xu Dong
Jin, Lei
author_sort La, Ting
collection PubMed
description Rationale: Recurrent and metastatic cancers often undergo a period of dormancy, which is closely associated with cellular quiescence, a state whereby cells exit the cell cycle and are reversibly arrested in G0 phase. Curative cancer treatment thus requires therapies that either sustain the dormant state of quiescent cancer cells, or preferentially, eliminate them. However, the mechanisms responsible for the survival of quiescent cancer cells remain obscure. Methods: Dual genome-editing was carried out using a CRISPR/Cas9-based system to label endogenous p27 and Ki67 with the green and red fluorescent proteins EGFP and mCherry, respectively, in melanoma cells. Analysis of transcriptomes of isolated EGFP-p27(high)mCherry-Ki67(low) quiescent cells was conducted at bulk and single cell levels using RNA-sequencing. The extracellular acidification rate and oxygen consumption rate were measured to define metabolic phenotypes. SiRNA and inducible shRNA knockdown, chromatin immunoprecipitation and luciferase reporter assays were employed to elucidate mechanisms of the metabolic switch in quiescent cells. Results: Dual labelling of endogenous p27 and Ki67 with differentiable fluorescent probes allowed for visualization, isolation, and analysis of viable p27(high)Ki67(low) quiescent cells. Paradoxically, the proto-oncoprotein c-Myc, which commonly drives malignant cell cycle progression, was expressed at relatively high levels in p27(high)Ki67(low) quiescent cells and supported their survival through promoting mitochondrial oxidative phosphorylation (OXPHOS). In this context, c-Myc selectively transactivated genes encoding OXPHOS enzymes, including subunits of isocitric dehydrogenase 3 (IDH3), whereas its binding to cell cycle progression gene promoters was decreased in quiescent cells. Silencing of c-Myc or the catalytic subunit of IDH3, IDH3α, preferentially killed quiescent cells, recapitulating the effect of treatment with OXPHOS inhibitors. Conclusion: These results establish a rigorous experimental system for investigating cellular quiescence, uncover the high selectivity of c-Myc in activating OXPHOS genes in quiescent cells, and propose OXPHOS targeting as a potential therapeutic avenue to counter cancer cells in quiescence.
format Online
Article
Text
id pubmed-8490506
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Ivyspring International Publisher
record_format MEDLINE/PubMed
spelling pubmed-84905062021-10-12 Visualization of endogenous p27 and Ki67 reveals the importance of a c-Myc-driven metabolic switch in promoting survival of quiescent cancer cells La, Ting Chen, Song Guo, Tao Zhao, Xiao Hong Teng, Liu Li, Dandan Carnell, Michael Zhang, Yuan Yuan Feng, Yu Chen Cole, Nicole Brown, Alexandra C. Zhang, Didi Dong, Qihan Wang, Jenny Y. Cao, Huixia Liu, Tao Thorne, Rick F. Shao, Feng-Min Zhang, Xu Dong Jin, Lei Theranostics Research Paper Rationale: Recurrent and metastatic cancers often undergo a period of dormancy, which is closely associated with cellular quiescence, a state whereby cells exit the cell cycle and are reversibly arrested in G0 phase. Curative cancer treatment thus requires therapies that either sustain the dormant state of quiescent cancer cells, or preferentially, eliminate them. However, the mechanisms responsible for the survival of quiescent cancer cells remain obscure. Methods: Dual genome-editing was carried out using a CRISPR/Cas9-based system to label endogenous p27 and Ki67 with the green and red fluorescent proteins EGFP and mCherry, respectively, in melanoma cells. Analysis of transcriptomes of isolated EGFP-p27(high)mCherry-Ki67(low) quiescent cells was conducted at bulk and single cell levels using RNA-sequencing. The extracellular acidification rate and oxygen consumption rate were measured to define metabolic phenotypes. SiRNA and inducible shRNA knockdown, chromatin immunoprecipitation and luciferase reporter assays were employed to elucidate mechanisms of the metabolic switch in quiescent cells. Results: Dual labelling of endogenous p27 and Ki67 with differentiable fluorescent probes allowed for visualization, isolation, and analysis of viable p27(high)Ki67(low) quiescent cells. Paradoxically, the proto-oncoprotein c-Myc, which commonly drives malignant cell cycle progression, was expressed at relatively high levels in p27(high)Ki67(low) quiescent cells and supported their survival through promoting mitochondrial oxidative phosphorylation (OXPHOS). In this context, c-Myc selectively transactivated genes encoding OXPHOS enzymes, including subunits of isocitric dehydrogenase 3 (IDH3), whereas its binding to cell cycle progression gene promoters was decreased in quiescent cells. Silencing of c-Myc or the catalytic subunit of IDH3, IDH3α, preferentially killed quiescent cells, recapitulating the effect of treatment with OXPHOS inhibitors. Conclusion: These results establish a rigorous experimental system for investigating cellular quiescence, uncover the high selectivity of c-Myc in activating OXPHOS genes in quiescent cells, and propose OXPHOS targeting as a potential therapeutic avenue to counter cancer cells in quiescence. Ivyspring International Publisher 2021-09-21 /pmc/articles/PMC8490506/ /pubmed/34646389 http://dx.doi.org/10.7150/thno.63763 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
La, Ting
Chen, Song
Guo, Tao
Zhao, Xiao Hong
Teng, Liu
Li, Dandan
Carnell, Michael
Zhang, Yuan Yuan
Feng, Yu Chen
Cole, Nicole
Brown, Alexandra C.
Zhang, Didi
Dong, Qihan
Wang, Jenny Y.
Cao, Huixia
Liu, Tao
Thorne, Rick F.
Shao, Feng-Min
Zhang, Xu Dong
Jin, Lei
Visualization of endogenous p27 and Ki67 reveals the importance of a c-Myc-driven metabolic switch in promoting survival of quiescent cancer cells
title Visualization of endogenous p27 and Ki67 reveals the importance of a c-Myc-driven metabolic switch in promoting survival of quiescent cancer cells
title_full Visualization of endogenous p27 and Ki67 reveals the importance of a c-Myc-driven metabolic switch in promoting survival of quiescent cancer cells
title_fullStr Visualization of endogenous p27 and Ki67 reveals the importance of a c-Myc-driven metabolic switch in promoting survival of quiescent cancer cells
title_full_unstemmed Visualization of endogenous p27 and Ki67 reveals the importance of a c-Myc-driven metabolic switch in promoting survival of quiescent cancer cells
title_short Visualization of endogenous p27 and Ki67 reveals the importance of a c-Myc-driven metabolic switch in promoting survival of quiescent cancer cells
title_sort visualization of endogenous p27 and ki67 reveals the importance of a c-myc-driven metabolic switch in promoting survival of quiescent cancer cells
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8490506/
https://www.ncbi.nlm.nih.gov/pubmed/34646389
http://dx.doi.org/10.7150/thno.63763
work_keys_str_mv AT lating visualizationofendogenousp27andki67revealstheimportanceofacmycdrivenmetabolicswitchinpromotingsurvivalofquiescentcancercells
AT chensong visualizationofendogenousp27andki67revealstheimportanceofacmycdrivenmetabolicswitchinpromotingsurvivalofquiescentcancercells
AT guotao visualizationofendogenousp27andki67revealstheimportanceofacmycdrivenmetabolicswitchinpromotingsurvivalofquiescentcancercells
AT zhaoxiaohong visualizationofendogenousp27andki67revealstheimportanceofacmycdrivenmetabolicswitchinpromotingsurvivalofquiescentcancercells
AT tengliu visualizationofendogenousp27andki67revealstheimportanceofacmycdrivenmetabolicswitchinpromotingsurvivalofquiescentcancercells
AT lidandan visualizationofendogenousp27andki67revealstheimportanceofacmycdrivenmetabolicswitchinpromotingsurvivalofquiescentcancercells
AT carnellmichael visualizationofendogenousp27andki67revealstheimportanceofacmycdrivenmetabolicswitchinpromotingsurvivalofquiescentcancercells
AT zhangyuanyuan visualizationofendogenousp27andki67revealstheimportanceofacmycdrivenmetabolicswitchinpromotingsurvivalofquiescentcancercells
AT fengyuchen visualizationofendogenousp27andki67revealstheimportanceofacmycdrivenmetabolicswitchinpromotingsurvivalofquiescentcancercells
AT colenicole visualizationofendogenousp27andki67revealstheimportanceofacmycdrivenmetabolicswitchinpromotingsurvivalofquiescentcancercells
AT brownalexandrac visualizationofendogenousp27andki67revealstheimportanceofacmycdrivenmetabolicswitchinpromotingsurvivalofquiescentcancercells
AT zhangdidi visualizationofendogenousp27andki67revealstheimportanceofacmycdrivenmetabolicswitchinpromotingsurvivalofquiescentcancercells
AT dongqihan visualizationofendogenousp27andki67revealstheimportanceofacmycdrivenmetabolicswitchinpromotingsurvivalofquiescentcancercells
AT wangjennyy visualizationofendogenousp27andki67revealstheimportanceofacmycdrivenmetabolicswitchinpromotingsurvivalofquiescentcancercells
AT caohuixia visualizationofendogenousp27andki67revealstheimportanceofacmycdrivenmetabolicswitchinpromotingsurvivalofquiescentcancercells
AT liutao visualizationofendogenousp27andki67revealstheimportanceofacmycdrivenmetabolicswitchinpromotingsurvivalofquiescentcancercells
AT thornerickf visualizationofendogenousp27andki67revealstheimportanceofacmycdrivenmetabolicswitchinpromotingsurvivalofquiescentcancercells
AT shaofengmin visualizationofendogenousp27andki67revealstheimportanceofacmycdrivenmetabolicswitchinpromotingsurvivalofquiescentcancercells
AT zhangxudong visualizationofendogenousp27andki67revealstheimportanceofacmycdrivenmetabolicswitchinpromotingsurvivalofquiescentcancercells
AT jinlei visualizationofendogenousp27andki67revealstheimportanceofacmycdrivenmetabolicswitchinpromotingsurvivalofquiescentcancercells