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 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-p27highmCherry-Ki67low 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 p27highKi67low quiescent cells. Paradoxically, the proto-oncoprotein c-Myc, which commonly drives malignant cell cycle progression, was expressed at relatively high levels in p27highKi67low 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.

[1]  M. Haniffa,et al.  Human oral mucosa cell atlas reveals a stromal-neutrophil axis regulating tissue immunity , 2021, Cell.

[2]  Yihai Cao,et al.  Molecular identity of human limbal heterogeneity involved in corneal homeostasis and privilege. , 2021, The ocular surface.

[3]  G. Qing,et al.  Regulation of cancer cell metabolism: oncogenic MYC in the driver’s seat , 2020, Signal Transduction and Targeted Therapy.

[4]  Nipha Chaicharoenaudomrung,et al.  Three-dimensional cell culture systems as an in vitro platform for cancer and stem cell modeling , 2019, World journal of stem cells.

[5]  I. Ulitsky,et al.  Regulation of gene expression by cis-acting long non-coding RNAs , 2019, Nature Reviews Genetics.

[6]  R. Scott,et al.  LncRNA REG1CP promotes tumorigenesis through an enhancer complex to recruit FANCJ helicase for REG3A transcription , 2019, Nature Communications.

[7]  Ruoyan Li,et al.  Graded regulation of cellular quiescence depth between proliferation and senescence by a lysosomal dimmer switch , 2019, Proceedings of the National Academy of Sciences.

[8]  Stefan Tümpel,et al.  Quiescence: Good and Bad of Stem Cell Aging. , 2019, Trends in cell biology.

[9]  Zhong Zhou,et al.  CircACC1 Regulates Assembly and Activation of AMPK Complex under Metabolic Stress. , 2019, Cell metabolism.

[10]  Xiaodong Zhao,et al.  The histone chaperone complex FACT promotes proliferative switch of G0 cancer cells , 2019, International journal of cancer.

[11]  J. León,et al.  MYC Oncogene Contributions to Release of Cell Cycle Brakes , 2019, Genes.

[12]  A. Recasens,et al.  Targeting Cancer Cell Dormancy. , 2019, Trends in pharmacological sciences.

[13]  E. Darling,et al.  Considerations for high-yield, high-throughput cell enrichment: fluorescence versus magnetic sorting , 2019, Scientific Reports.

[14]  S. Guo,et al.  A p53-Responsive miRNA Network Promotes Cancer Cell Quiescence. , 2018, Cancer research.

[15]  R. Medzhitov,et al.  Emerging Principles of Gene Expression Programs and Their Regulation. , 2018, Molecular cell.

[16]  P. Carmeliet,et al.  Phenotype molding of stromal cells in the lung tumor microenvironment , 2018, Nature Medicine.

[17]  S. Spencer,et al.  Ki67 is a Graded Rather than a Binary Marker of Proliferation versus Quiescence. , 2018, Cell reports.

[18]  M. Protopopova,et al.  An inhibitor of oxidative phosphorylation exploits cancer vulnerability , 2018, Nature Medicine.

[19]  S. Anant,et al.  Cancer Stem Cell Metabolism and Potential Therapeutic Targets , 2018, Front. Oncol..

[20]  Min Zhang,et al.  lncRNA Epigenetic Landscape Analysis Identifies EPIC1 as an Oncogenic lncRNA that Interacts with MYC and Promotes Cell-Cycle Progression in Cancer. , 2018, Cancer cell.

[21]  Danica Chen,et al.  The mitochondrial unfolded protein response is activated upon hematopoietic stem cell exit from quiescence , 2018, Aging cell.

[22]  G. Qing,et al.  Targeting oncogenic Myc as a strategy for cancer treatment , 2018, Signal Transduction and Targeted Therapy.

[23]  Geoff S. Higgins,et al.  Oxidative Phosphorylation as an Emerging Target in Cancer Therapy , 2018, Clinical Cancer Research.

[24]  Lei Jin,et al.  LncRNA IDH1-AS1 links the functions of c-Myc and HIF1α via IDH1 to regulate the Warburg effect , 2018, Proceedings of the National Academy of Sciences.

[25]  Jason W. Locasale,et al.  Melanoma Therapeutic Strategies that Select against Resistance by Exploiting MYC-Driven Evolutionary Convergence. , 2017, Cell reports.

[26]  Hong Zhou,et al.  c‑Myc promotes cholangiocarcinoma cells to overcome contact inhibition via the mTOR pathway. , 2017, Oncology reports.

[27]  J. S. Kwon,et al.  Controlling Depth of Cellular Quiescence by an Rb-E2F Network Switch. , 2017, Cell reports.

[28]  Hamoud H. Al-Khallaf Isocitrate dehydrogenases in physiology and cancer: biochemical and molecular insight , 2017, Cell & Bioscience.

[29]  M. Eccles,et al.  The Slow Cycling Phenotype: A Growing Problem for Treatment Resistance in Melanoma , 2017, Molecular Cancer Therapeutics.

[30]  C. Yao,et al.  p53 Regulates Progenitor Cell Quiescence and Differentiation in the Airway. , 2016, Cell reports.

[31]  Michael Z. Lin,et al.  Fluorescent indicators for simultaneous reporting of all four cell cycle phases , 2016, Nature Methods.

[32]  N. Waterhouse,et al.  Measuring Cell Death by Propidium Iodide Uptake and Flow Cytometry. , 2016, Cold Spring Harbor protocols.

[33]  S. Ramaswamy,et al.  Mechanisms of Cancer Cell Dormancy--Another Hallmark of Cancer? , 2015, Cancer research.

[34]  J. Holst,et al.  p27(Kip1) signaling: Transcriptional and post-translational regulation. , 2015, The international journal of biochemistry & cell biology.

[35]  E. Giannoni,et al.  5-Fluorouracil resistant colon cancer cells are addicted to OXPHOS to survive and enhance stem-like traits , 2015, Oncotarget.

[36]  M. Kassem,et al.  Concise Review: Quiescence in Adult Stem Cells: Biological Significance and Relevance to Tissue Regeneration , 2015, Stem cells.

[37]  A. Sabò,et al.  MYC: connecting selective transcriptional control to global RNA production , 2015, Nature Reviews Cancer.

[38]  K. Kim,et al.  Assaying Cell Cycle Status Using Flow Cytometry , 2015, Current protocols in molecular biology.

[39]  Javier León,et al.  Myc and cell cycle control. , 2015, Biochimica et biophysica acta.

[40]  Stefan W. Hell,et al.  CRISPR/Cas9-mediated endogenous protein tagging for RESOLFT super-resolution microscopy of living human cells , 2015, Scientific Reports.

[41]  J. Mi,et al.  Metabolic reprogramming of cancer-associated fibroblasts by IDH3α downregulation. , 2015, Cell reports.

[42]  D. Wolf Is reliance on mitochondrial respiration a "chink in the armor" of therapy-resistant cancer? , 2014, Cancer cell.

[43]  Kakajan Komurov,et al.  Inhibition of mTORC1/2 overcomes resistance to MAPK pathway inhibitors mediated by PGC1α and oxidative phosphorylation in melanoma. , 2014, Cancer research.

[44]  Marco J. Morelli,et al.  Selective transcriptional regulation by Myc in cellular growth control and lymphomagenesis , 2014, Nature.

[45]  Guang Yao Modelling mammalian cellular quiescence , 2014, Interface Focus.

[46]  D. Felsher,et al.  MYC activation is a hallmark of cancer initiation and maintenance. , 2014, Cold Spring Harbor perspectives in medicine.

[47]  D. Hockenbery,et al.  MYC and mitochondrial biogenesis. , 2014, Cold Spring Harbor perspectives in medicine.

[48]  H. Aburatani,et al.  A novel cell-cycle-indicator, mVenus-p27K−, identifies quiescent cells and visualizes G0–G1 transition , 2014, Scientific Reports.

[49]  L. Ellis,et al.  Colon Cancer Cells Escape 5FU Chemotherapy-Induced Cell Death by Entering Stemness and Quiescence Associated with the c-Yes/YAP Axis , 2013, Clinical Cancer Research.

[50]  C. Dang MYC, metabolism, cell growth, and tumorigenesis. , 2013, Cold Spring Harbor perspectives in medicine.

[51]  Jun S. Song,et al.  Oncogenic BRAF regulates oxidative metabolism via PGC1α and MITF. , 2013, Cancer cell.

[52]  Nathan C Boles,et al.  Less is more: unveiling the functional core of hematopoietic stem cells through knockout mice. , 2012, Cell stem cell.

[53]  C. Simón,et al.  Efficiency and purity provided by the existing methods for the isolation of luteinized granulosa cells: a comparative study. , 2012, Human reproduction.

[54]  Chi V Dang,et al.  MYC on the Path to Cancer , 2012, Cell.

[55]  J. Viola,et al.  Transcriptional regulation of the c-Myc promoter by NFAT1 involves negative and positive NFAT-responsive elements , 2012, Cell cycle.

[56]  M. Cole,et al.  Myc posttranscriptionally induces HIF1 protein and target gene expression in normal and cancer cells. , 2012, Cancer Research.

[57]  P. O’Farrell Quiescence: early evolutionary origins and universality do not imply uniformity , 2011, Philosophical Transactions of the Royal Society B: Biological Sciences.

[58]  M. Henriksson,et al.  Identification of Cytotoxic Drugs That Selectively Target Tumor Cells with MYC Overexpression , 2011, PloS one.

[59]  Andrei L Osterman,et al.  Comparative Metabolic Flux Profiling of Melanoma Cell Lines , 2011, The Journal of Biological Chemistry.

[60]  Elizabeth L. Johnson,et al.  Quiescent Fibroblasts Exhibit High Metabolic Activity , 2010, PLoS biology.

[61]  J. Sarkaria,et al.  Acquisition of Temozolomide Chemoresistance in Gliomas Leads to Remodeling of Mitochondrial Electron Transport Chain* , 2010, The Journal of Biological Chemistry.

[62]  A. Zambon Use of the Ki67 promoter to label cell cycle entry in living cells , 2010, Cytometry. Part A : the journal of the International Society for Analytical Cytology.

[63]  L. Cantley,et al.  Understanding the Warburg Effect: The Metabolic Requirements of Cell Proliferation , 2009, Science.

[64]  T. L. McGee,et al.  Insights from retinitis pigmentosa into the roles of isocitrate dehydrogenases in the Krebs cycle , 2008, Nature Genetics.

[65]  Atsushi Miyawaki,et al.  Visualizing Spatiotemporal Dynamics of Multicellular Cell-Cycle Progression , 2008, Cell.

[66]  C. Thompson,et al.  HIF and c-Myc: sibling rivals for control of cancer cell metabolism and proliferation. , 2007, Cancer cell.

[67]  M. Blagosklonny Target for cancer therapy: proliferating cells or stem cells , 2006, Leukemia.

[68]  P. Hersey,et al.  Human melanoma cells selected for resistance to apoptosis by prolonged exposure to tumor necrosis factor-related apoptosis-inducing ligand are more vulnerable to necrotic cell death induced by cisplatin. , 2006, Clinical cancer research : an official journal of the American Association for Cancer Research.

[69]  B. Thimmapaya,et al.  Repression of c-Myc and inhibition of G1 exit in cells conditionally overexpressing p300 that is not dependent on its histone acetyltransferase activity , 2003, Proceedings of the National Academy of Sciences of the United States of America.

[70]  A. Bird,et al.  Epigenetic regulation of gene expression: how the genome integrates intrinsic and environmental signals , 2003, Nature Genetics.

[71]  J. Gerdes,et al.  The Ki‐67 protein interacts with members of the heterochromatin protein 1 (HP1) family: a potential role in the regulation of higher‐order chromatin structure , 2002, The Journal of pathology.

[72]  G. Lozano,et al.  Differential roles of p21(Waf1) and p27(Kip1) in modulating chemosensitivity and their possible application in drug discovery studies. , 2001, Molecular pharmacology.

[73]  C. Fang,et al.  Deregulated c-myc expression in quiescent CHO cells induces target gene transcription and subsequent apoptotic phenotype , 1999, Cell Research.

[74]  J. Sedivy,et al.  Mysterious liaisons: the relationship between c-Myc and the cell cycle , 1999, Oncogene.

[75]  R. Johnston,et al.  Quiescence versus apoptosis: Myc abundance determines pathway of exit from the cell cycle , 1998, Oncogene.

[76]  J. Sedivy,et al.  Effects of c-myc expression on proliferation, quiescence, and the G0 to G1 transition in nontransformed cells. , 1993, Cell growth & differentiation : the molecular biology journal of the American Association for Cancer Research.

[77]  B. Whitelaw,et al.  Transcriptional regulation of the human c-myc gene. , 1988, The British journal of cancer. Supplement.

[78]  V. Jordan,et al.  Rethinking Extended Adjuvant Antiestrogen Therapy to Increase Survivorship in Breast Cancer. , 2018, JAMA oncology.

[79]  Baocun Sun,et al.  C-myc overexpression drives melanoma metastasis by promoting vasculogenic mimicry via c-myc/snail/Bax signaling , 2016, Journal of Molecular Medicine.

[80]  F. Di Rosa Two Niches in the Bone Marrow: A Hypothesis on Life-long T Cell Memory. , 2016, Trends in immunology.

[81]  E. Kipreos,et al.  Multiple degradation pathways regulate versatile CIP/KIP CDK inhibitors. , 2012, Trends in cell biology.

[82]  Rudolf Jaenisch,et al.  Analysis of histone 2B-GFP retention reveals slowly cycling hematopoietic stem cells , 2009, Nature Biotechnology.

[83]  Chi V. Dang,et al.  The interplay between MYC and HIF in cancer , 2008, Nature Reviews Cancer.

[84]  J. Aguirre-Ghiso,et al.  ERK(MAPK) activity as a determinant of tumor growth and dormancy; regulation by p38(SAPK). , 2003, Cancer research.