p62/SQSTM1 Cooperates with Hyperactive mTORC1 to Regulate Glutathione Production, Maintain Mitochondrial Integrity, and Promote Tumorigenesis.

p62/sequestosome-1 (SQSTM1) is a multifunctional adaptor protein and autophagic substrate that accumulates in cells with hyperactive mTORC1, such as kidney cells with mutations in the tumor suppressor genes tuberous sclerosis complex (TSC)1 or TSC2. Here we report that p62 is a critical mediator of TSC2-driven tumorigenesis, as Tsc2+/- and Tsc2f/f Ksp-CreERT2+ mice crossed to p62-/- mice were protected from renal tumor development. Metabolic profiling revealed that depletion of p62 in Tsc2-null cells decreased intracellular glutamine, glutamate, and glutathione (GSH). p62 positively regulated the glutamine transporter Slc1a5 and increased glutamine uptake in Tsc2-null cells. We also observed p62-dependent changes in Gcl, Gsr, Nqo1, and Srxn1, which were decreased by p62 attenuation and implicated in GSH production and utilization. p62 attenuation altered mitochondrial morphology, reduced mitochondrial membrane polarization and maximal respiration, and increased mitochondrial reactive oxygen species and mitophagy marker PINK1. These mitochondrial phenotypes were rescued by addition of exogenous GSH and overexpression of Sod2, which suppressed indices of mitochondrial damage and promoted growth of Tsc2-null cells. Finally, p62 depletion sensitized Tsc2-null cells to both oxidative stress and direct inhibition of GSH biosynthesis by buthionine sulfoximine. Our findings show how p62 helps maintain intracellular pools of GSH needed to limit mitochondrial dysfunction in tumor cells with elevated mTORC1, highlighting p62 and redox homeostasis as nodal vulnerabilities for therapeutic targeting in these tumors. Cancer Res; 77(12); 3255-67. ©2017 AACR.

[1]  S. Belinsky,et al.  TSC2 Deficiency Unmasks a Novel Necrosis Pathway That Is Suppressed by the RIP1/RIP3/MLKL Signaling Cascade. , 2016, Cancer research.

[2]  J. Rothberg,et al.  Impaired Mitochondrial Dynamics and Mitophagy in Neuronal Models of Tuberous Sclerosis Complex. , 2016, Cell reports.

[3]  Z. Fan,et al.  ASCT2 (SLC1A5) is an EGFR-associated protein that can be co-targeted by cetuximab to sensitize cancer cells to ROS-induced apoptosis. , 2016, Cancer letters.

[4]  W. Gu,et al.  Autophagy Regulates Chromatin Ubiquitination in DNA Damage Response through Elimination of SQSTM1/p62. , 2016, Molecular cell.

[5]  S. Subramaniam,et al.  p62, Upregulated during Preneoplasia, Induces Hepatocellular Carcinogenesis by Maintaining Survival of Stressed HCC-Initiating Cells. , 2016, Cancer cell.

[6]  W. Ritchie,et al.  ASCT2/SLC1A5 controls glutamine uptake and tumour growth in triple-negative basal-like breast cancer , 2015, Oncogene.

[7]  A. Durán,et al.  Amino Acid Activation of mTORC1 by a PB1-Domain-Driven Kinase Complex Cascade. , 2015, Cell reports.

[8]  E. Aronica,et al.  Tuberous sclerosis complex neuropathology requires glutamate-cysteine ligase , 2015, Acta Neuropathologica Communications.

[9]  Terje Johansen,et al.  The selective autophagy receptor p62 forms a flexible filamentous helical scaffold. , 2015, Cell reports.

[10]  J. Blenis,et al.  The mTORC1/S6K1 Pathway Regulates Glutamine Metabolism through the eIF4B-Dependent Control of c-Myc Translation , 2014, Current Biology.

[11]  J. Asara,et al.  High-Throughput Drug Screen Identifies Chelerythrine as a Selective Inducer of Death in a TSC2-null Setting , 2014, Molecular Cancer Research.

[12]  D. Kwiatkowski,et al.  Molecular basis of giant cells in tuberous sclerosis complex. , 2014, The New England journal of medicine.

[13]  K. Haley,et al.  Systems-level regulation of microRNA networks by miR-130/301 promotes pulmonary hypertension. , 2014, The Journal of clinical investigation.

[14]  Christian M. Metallo,et al.  Metabolic reprogramming of stromal fibroblasts through p62-mTORC1 signaling promotes inflammation and tumorigenesis. , 2014, Cancer cell.

[15]  D. Kwiatkowski,et al.  Coordinated regulation of protein synthesis and degradation by mTORC1 , 2014, Nature.

[16]  N. Chandel Mitochondria and cancer , 2014, Cancer & metabolism.

[17]  C. Sander,et al.  SQSTM1 is a pathogenic target of 5q copy number gains in kidney cancer. , 2013, Cancer cell.

[18]  A. Choi,et al.  Autophagy-Dependent Metabolic Reprogramming Sensitizes TSC2-Deficient Cells to the Antimetabolite 6-Aminonicotinamide , 2013, Molecular Cancer Research.

[19]  M. Harper,et al.  Unearthing the secrets of mitochondrial ROS and glutathione in bioenergetics. , 2013, Trends in biochemical sciences.

[20]  D. Klionsky,et al.  Participation of mitochondrial fission during mitophagy , 2013, Cell cycle.

[21]  Ralph J DeBerardinis,et al.  Glutamine and cancer: cell biology, physiology, and clinical opportunities. , 2013, The Journal of clinical investigation.

[22]  Douglas J. Chapski,et al.  The mTORC1 Pathway Stimulates Glutamine Metabolism and Cell Proliferation by Repressing SIRT4 , 2013, Cell.

[23]  J. Moscat,et al.  p62: a versatile multitasker takes on cancer. , 2012, Trends in biochemical sciences.

[24]  J. Asara,et al.  A positive/negative ion–switching, targeted mass spectrometry–based metabolomics platform for bodily fluids, cells, and fresh and fixed tissue , 2012, Nature Protocols.

[25]  D. Sabatini,et al.  mTOR Signaling in Growth Control and Disease , 2012, Cell.

[26]  M. Hung,et al.  KrasG12D-induced IKK2/β/NF-κB activation by IL-1α and p62 feedforward loops is required for development of pancreatic ductal adenocarcinoma. , 2012, Cancer cell.

[27]  E. Henske,et al.  Autophagy: an 'Achilles' heel of tumorigenesis in TSC and LAM. , 2011, Autophagy.

[28]  Peter M. Klein,et al.  Regulable neural progenitor-specific Tsc1 loss yields giant cells with organellar dysfunction in a model of tuberous sclerosis complex , 2011, Proceedings of the National Academy of Sciences.

[29]  W. Marston Linehan,et al.  Reductive carboxylation supports growth in tumor cells with defective mitochondria , 2011, Nature.

[30]  T. H. Sasongko,et al.  Tuberous sclerosis complex , 2020, Cleveland Clinic Journal of Medicine.

[31]  Aleksey A. Porollo,et al.  p62 is a key regulator of nutrient sensing in the mTORC1 pathway. , 2011, Molecular cell.

[32]  M. Komatsu Potential role of p62 in tumor development , 2011, Autophagy.

[33]  D. Kwiatkowski,et al.  Tumorigenesis in tuberous sclerosis complex is autophagy and p62/sequestosome 1 (SQSTM1)-dependent , 2011, Proceedings of the National Academy of Sciences.

[34]  Keiji Tanaka,et al.  Persistent activation of Nrf2 through p62 in hepatocellular carcinoma cells , 2011, The Journal of cell biology.

[35]  R. Youle,et al.  p62/SQSTM1 is required for Parkin-induced mitochondrial clustering but not mitophagy; VDAC1 is dispensable for both , 2010, Autophagy.

[36]  G. Dorn,et al.  Nix Is Critical to Two Distinct Phases of Mitophagy, Reactive Oxygen Species-mediated Autophagy Induction and Parkin-Ubiquitin-p62-mediated Mitochondrial Priming* , 2010, The Journal of Biological Chemistry.

[37]  Sang Gyun Kim,et al.  Glucose addiction of TSC null cells is caused by failed mTORC1-dependent balancing of metabolic demand with supply. , 2010, Molecular cell.

[38]  M. McMahon,et al.  p62/SQSTM1 Is a Target Gene for Transcription Factor NRF2 and Creates a Positive Feedback Loop by Inducing Antioxidant Response Element-driven Gene Transcription* , 2010, The Journal of Biological Chemistry.

[39]  E. White,et al.  A Noncanonical Mechanism of Nrf2 Activation by Autophagy Deficiency: Direct Interaction between Keap1 and p62 , 2010, Molecular and Cellular Biology.

[40]  Mihee M. Kim,et al.  The selective autophagy substrate p62 activates the stress responsive transcription factor Nrf2 through inactivation of Keap1 , 2010, Nature Cell Biology.

[41]  Fabienne C. Fiesel,et al.  PINK1/Parkin-mediated mitophagy is dependent on VDAC1 and p62/SQSTM1 , 2010, Nature Cell Biology.

[42]  R. Deberardinis,et al.  Q's next: the diverse functions of glutamine in metabolism, cell biology and cancer , 2010, Oncogene.

[43]  Davis J. McCarthy,et al.  edgeR: a Bioconductor package for differential expression analysis of digital gene expression data , 2009, Bioinform..

[44]  Hanna Y. Irie,et al.  Antioxidant and oncogene rescue of metabolic defects caused by loss of matrix attachment , 2009, Nature.

[45]  H. Lane,et al.  Equivalent benefit of mTORC1 blockade and combined PI3K-mTOR blockade in a mouse model of tuberous sclerosis , 2009, Molecular Cancer.

[46]  D. Kwiatkowski,et al.  Tuberous Sclerosis Complex Activity Is Required to Control Neuronal Stress Responses in an mTOR-Dependent Manner , 2009, The Journal of Neuroscience.

[47]  D. Sabatini,et al.  mTOR and cancer: many loops in one pathway. , 2009, Current opinion in cell biology.

[48]  J. Flores,et al.  The signaling adaptor p62 is an important NF-kappaB mediator in tumorigenesis. , 2008, Cancer cell.

[49]  V. Schmithorst,et al.  Sirolimus for angiomyolipoma in tuberous sclerosis complex or lymphangioleiomyomatosis. , 2008, The New England journal of medicine.

[50]  Jill P. Mesirov,et al.  GSEA-P: a desktop application for Gene Set Enrichment Analysis , 2007, Bioinform..

[51]  Hongbing Zhang,et al.  Efficacy of a rapamycin analog (CCI‐779) and IFN‐γ in tuberous sclerosis mouse models , 2005 .

[52]  William R Sellers,et al.  TSC2 regulates VEGF through mTOR-dependent and -independent pathways. , 2003, Cancer cell.

[53]  H. Nakano,et al.  The atypical PKC‐interacting protein p62 channels NF‐κB activation by the IL‐1–TRAF6 pathway , 2000, The EMBO journal.

[54]  T. Noda,et al.  Renal carcinogenesis, hepatic hemangiomatosis, and embryonic lethality caused by a germ-line Tsc2 mutation in mice. , 1999, Cancer research.