Loss of the tumor suppressor LKB1 promotes metabolic reprogramming of cancer cells via HIF-1α

Significance Liver kinase B1 (LKB1) is a serine/threonine kinase often inactivated in human cancer. We demonstrate here that loss of LKB1 expression in cancer cells promotes a progrowth metabolic profile that enables increased cell growth and proliferation. Loss of LKB1 promotes increased tumor cell metabolism through mammalian target of rapamycin complex 1- and reactive oxygen species-dependent increases in hypoxia-inducible factor-1α (HIF-1α). LKB1-null cells are dependent on HIF-1α to maintain cellular ATP and viability under poor nutrient conditions, raising the possibility of targeting HIF-1α for synthetic lethality in LKB1-deficient tumors. Together, our data reveal that regulation of cellular metabolism is a key function of LKB1 that may contribute to its tumor-suppressor function in human cancer. One of the major metabolic changes associated with cellular transformation is enhanced nutrient utilization, which supports tumor progression by fueling both energy production and providing biosynthetic intermediates for growth. The liver kinase B1 (LKB1) is a serine/threonine kinase and tumor suppressor that couples bioenergetics to cell-growth control through regulation of mammalian target of rapamycin (mTOR) activity; however, the influence of LKB1 on tumor metabolism is not well defined. Here, we show that loss of LKB1 induces a progrowth metabolic program in proliferating cells. Cells lacking LKB1 display increased glucose and glutamine uptake and utilization, which support both cellular ATP levels and increased macromolecular biosynthesis. This LKB1-dependent reprogramming of cell metabolism is dependent on the hypoxia-inducible factor-1α (HIF-1α), which accumulates under normoxia in LKB1-deficient cells and is antagonized by inhibition of mTOR complex I signaling. Silencing HIF-1α reverses the metabolic advantages conferred by reduced LKB1 signaling and impairs the growth and survival of LKB1-deficient tumor cells under low-nutrient conditions. Together, our data implicate the tumor suppressor LKB1 as a central regulator of tumor metabolism and growth control through the regulation of HIF-1α–dependent metabolic reprogramming.

[1]  D. Hardie,et al.  LKB1 and AMPK and the cancer-metabolism link - ten years after , 2013, BMC Biology.

[2]  Liu Wei,et al.  LKB1 inactivation dictates therapeutic response of non-small cell lung cancer to the metabolism drug phenformin. , 2013, Cancer cell.

[3]  Takla Griss,et al.  AMPK is a negative regulator of the Warburg effect and suppresses tumor growth in vivo. , 2013, Cell metabolism.

[4]  Gerald C. Chu,et al.  Oncogenic Kras Maintains Pancreatic Tumors through Regulation of Anabolic Glucose Metabolism , 2012, Cell.

[5]  Brian Keith,et al.  HIF1α and HIF2α: sibling rivalry in hypoxic tumour growth and progression , 2011, Nature Reviews Cancer.

[6]  Christian M. Metallo,et al.  Reductive glutamine metabolism by IDH1 mediates lipogenesis under hypoxia , 2011, Nature.

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

[8]  Jesse M. Platt,et al.  Hypoxia promotes isocitrate dehydrogenase-dependent carboxylation of α-ketoglutarate to citrate to support cell growth and viability , 2011, Proceedings of the National Academy of Sciences.

[9]  G. Semenza Oxygen sensing, homeostasis, and disease. , 2011, The New England journal of medicine.

[10]  W. H. Schaefer,et al.  Metabolic profiles show specific mitochondrial toxicities in vitro in myotube cells , 2011, Journal of biomolecular NMR.

[11]  G. Semenza Regulation of metabolism by hypoxia-inducible factor 1. , 2011, Cold Spring Harbor symposia on quantitative biology.

[12]  G. Semenza mechanisms of disease Oxygen Sensing , Homeostasis , and Disease , 2011 .

[13]  J. Yokota,et al.  Involvement of LKB1 in epithelial-mesenchymal transition (EMT) of human lung cancer cells. , 2010, Lung cancer.

[14]  David Carling,et al.  Signaling Kinase AMPK Activates Stress-Promoted Transcription via Histone H2B Phosphorylation , 2010, Science.

[15]  D. Sgroi,et al.  Negative feedback control of HIF-1 through REDD1-regulated ROS suppresses tumorigenesis , 2010, Proceedings of the National Academy of Sciences.

[16]  D. Nicholls,et al.  Neuronal Glutamate and Gabaa Receptor Function in Health and Disease Spare Respiratory Capacity, Oxidative Stress and Excitotoxicity Glutamate as an Excitotoxin the Role of Oxidative Stress , 2022 .

[17]  R. Shaw,et al.  The LKB1–AMPK pathway: metabolism and growth control in tumour suppression , 2009, Nature Reviews Cancer.

[18]  Chin-Lee Wu,et al.  mTOR and HIF-1α-mediated tumor metabolism in an LKB1 mouse model of Peutz-Jeghers syndrome , 2009, Proceedings of the National Academy of Sciences.

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

[20]  Kwok-Kin Wong,et al.  Somatic LKB1 Mutations Promote Cervical Cancer Progression , 2009, PloS one.

[21]  Russell G. Jones,et al.  Tumor suppressors and cell metabolism: a recipe for cancer growth. , 2009, Genes & development.

[22]  R. Shaw LKB1: cancer, polarity, metabolism, and now fertility. , 2008, The Biochemical journal.

[23]  D. Hayes,et al.  Role of LKB1 in lung cancer development , 2008, British Journal of Cancer.

[24]  Ralph J Deberardinis,et al.  Brick by brick: metabolism and tumor cell growth. , 2008, Current opinion in genetics & development.

[25]  Kwok-Kin Wong,et al.  Loss of Lkb1 provokes highly invasive endometrial adenocarcinomas. , 2008, Cancer research.

[26]  R. Deberardinis,et al.  The biology of cancer: metabolic reprogramming fuels cell growth and proliferation. , 2008, Cell metabolism.

[27]  D. Neil Hayes,et al.  LKB1 modulates lung cancer differentiation and metastasis , 2007, Nature.

[28]  R. Deberardinis,et al.  The transcription factor HIF-1alpha plays a critical role in the growth factor-dependent regulation of both aerobic and anaerobic glycolysis. , 2007, Genes & development.

[29]  T. Mak,et al.  Metabolic Targeting as an Anticancer Strategy: Dawn of a New Era? , 2007, Science's STKE.

[30]  S. Gruber,et al.  Frequency and Spectrum of Cancers in the Peutz-Jeghers Syndrome , 2006, Clinical Cancer Research.

[31]  N. Denko,et al.  HIF-1 mediates adaptation to hypoxia by actively downregulating mitochondrial oxygen consumption. , 2006, Cell metabolism.

[32]  G. Semenza,et al.  HIF-1-mediated expression of pyruvate dehydrogenase kinase: a metabolic switch required for cellular adaptation to hypoxia. , 2006, Cell metabolism.

[33]  Daniel E Bauer,et al.  ATP citrate lyase inhibition can suppress tumor cell growth. , 2005, Cancer cell.

[34]  P. Schumacker,et al.  Mitochondrial dysfunction resulting from loss of cytochrome c impairs cellular oxygen sensing and hypoxic HIF-alpha activation. , 2005, Cell metabolism.

[35]  P. Schumacker,et al.  Mitochondrial complex III is required for hypoxia-induced ROS production and cellular oxygen sensing. , 2005, Cell metabolism.

[36]  Massimo Zeviani,et al.  Oxygen sensing requires mitochondrial ROS but not oxidative phosphorylation. , 2005, Cell metabolism.

[37]  Russell G. Jones,et al.  AMP-activated protein kinase induces a p53-dependent metabolic checkpoint. , 2005, Molecular cell.

[38]  R. DePinho,et al.  The LKB1 tumor suppressor negatively regulates mTOR signaling. , 2004, Cancer cell.

[39]  R. DePinho,et al.  Regulation of the TSC pathway by LKB1: evidence of a molecular link between tuberous sclerosis complex and Peutz-Jeghers syndrome. , 2004, Genes & development.

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

[41]  G. Sapkota,et al.  LKB1, a protein kinase regulating cell proliferation and polarity , 2003, FEBS letters.

[42]  W. Sellers,et al.  TSC 2 regulates VEGF through mTOR-dependent and-independent pathways , 2003 .

[43]  David Sidransky,et al.  Inactivation of LKB1/STK11 is a common event in adenocarcinomas of the lung. , 2002, Cancer research.

[44]  A. Harris,et al.  Relation of hypoxia inducible factor 1α and 2α in operable non-small cell lung cancer to angiogenic/molecular profile of tumours and survival , 2001, British Journal of Cancer.

[45]  L. Aaltonen,et al.  LKB1 somatic mutations in sporadic tumors. , 1999, The American journal of pathology.

[46]  M. Stratton,et al.  A serine/threonine kinase gene defective in Peutz–Jeghers syndrome , 1998, Nature.

[47]  J. Folch,et al.  Preparation of lipide extracts from brain tissue. , 1951, The Journal of biological chemistry.