MetaboList – November 2017

———————————————-
Methods/Global approaches
———————————————-

Noninvasive liquid diet delivery of stable isotopes into mouse models for deep metabolic network tracing
Ramon C. Sun, Teresa W.-M. Fan, Pan Deng,  Richard M. Higashi, Andrew N. Lane, Anh-Thu Le, Timothy L. Scott, Qiushi Sun, Marc O. Warmoes & Ye Yang
https://www.nature.com/articles/s41467-017-01518-z

Exploring Metabolic Configurations of Single Cells within Complex Tissue Microenvironments
Anne Miller,  Csörsz Nagy, Bernhard Knapp, Johannes Laengle, Elisabeth Ponweiser, Marion Groeger, Philipp Starkl, Michael Bergmann, Oswald Wagner, Arvand Haschem
http://www.cell.com/cell-metabolism/fulltext/S1550-4131(17)30499-0

Metabolic Profiling of healthy and cancerous tissues in 2D and 3D
Shonagh Russell, Jonathan Wojtkowiak, Andy Neilson & Robert J. Gillies
https://www.nature.com/articles/s41598-017-15325-5

High-throughput metabolic profiling based on small amount of hepatic cells
Zhou, Lina; Yin, Peiyuan; Luo, Ping; Tang, Ling; Wang, Zhichao; Gao, Peng; Piao, Hailong; Lu, Xin; Xu, Guowang
http://onlinelibrary.wiley.com/doi/10.1002/elps.201600539/full

A Highly Sensitive FRET Biosensor for AMPK Exhibits Heterogeneous AMPK Responses among Cells and Organs
Yumi Konagaya, Kenta Terai, Yusuke Hirao, Kanako Takakura, Masamichi Imajo, Yuji Kamioka, Norio Sasaoka, Akira Kakizuka, Kenta Sumiyama, Tomoichiro Asano, Michiyuki Matsuda
http://www.cell.com/cell-reports/fulltext/S2211-1247(17)31593-0

———————————————–
Amino acids and ammonia
———————————————–

Lysosomal metabolomics reveals V-ATPase- and mTOR-dependent regulation of amino acid efflux from lysosomes
Monther Abu-Remaileh, Gregory A. Wyant, Choah Kim, Nouf N. Laqtom, Maria Abbasi, Sze Ham Chan, Elizaveta Freinkman, David M. Sabatini
http://science.sciencemag.org/content/358/6364/807

Metabolic recycling of ammonia via glutamate dehydrogenase supports breast cancer biomass
Jessica B. Spinelli, Haejin Yoon, Alison E. Ringel, Sarah Jeanfavre, Clary B. Clish, Marcia C. Haigis
http://science.sciencemag.org/content/358/6365/941

BCAT1 restricts αKG levels in AML stem cells leading to IDHmut-like DNA hypermethylation
Simon Raffel, Mattia Falcone […] Andreas Trumpp
https://www.nature.com/articles/nature24294

Glutamine Addiction in Kidney Cancer Suppresses Oxidative Stress and Can Be Exploited for Real-Time Imaging
Omran Abu Aboud, Samy L. Habib, Josephine Trott, Benjamin Stewart, Sitai Liang, Abhijit J. Chaudhari, Julie Sutcliffe and Robert H. Weiss
http://cancerres.aacrjournals.org/content/77/23/6746

Keap1 loss promotes Kras-driven lung cancer and results in dependence on glutaminolysis
Rodrigo Romero, Volkan I Sayin[…],Thales Papagiannakopoulos
http://www.nature.com/nm/journal/v23/n11/full/nm.4407.html

PHGDH as a Key Enzyme for Serine Biosynthesis in HIF2α-Targeting Therapy for Renal Cell Carcinoma
Hirofumi Yoshino, Nijiro Nohata, Kazutaka Miyamoto, Masaya Yonemori, Takashi Sakaguchi, Satoshi Sugita, Toshihiko Itesako, Satoshi Kofuji, Masayuki Nakagawa, Rajvir Dahiya and Hideki Enokida
http://cancerres.aacrjournals.org/content/77/22/6321

Glutamine deficiency induces DNA alkylation damage and sensitizes cancer cells to alkylating agents through inhibition of ALKBH enzymes
Thai Q. Tran, Mari B. Ishak Gabra, Xazmin H. Lowman, Ying Yang, Michael A. Reid, Min Pan, Timothy R. O’Connor, Mei Kong
http://journals.plos.org/plosbiology/article?id=10.1371/journal.pbio.2002810

ENDOGENOUS GLUTAMINE DECREASE IS ASSOCIATED WITH PANCREATIC CANCER PROGRESSION
Cecilia Roux, Chiara Riganti, Sammy Ferri Borgogno, Roberta Curto, Claudia Curcio, Valeria Catanzaro, Giuseppe Digilio, Sergio Padovan, Maria Paola Puccinelli, Monica Isabello, Silvio Aime, Paola Cappello,  and  Francesco Novelli
http://www.impactjournals.com/oncotarget/index.php?journal=oncotarget&page=article&op=view&path%5B%5D=20545&path%5B%5D=65492

The arginine metabolome in Acute Lymphoblastic Leukemia can be targeted by the PEG-recombinant Arginase I BCT-100
Carmela De Santo, Sarah Booth, Ashley Vardon, Antony Cousins, Vanessa Tubb, Tracey Perry, Boris Noyvert, Andrew Beggs, Margaret Ng, Christina Halsey, Pamela Kearns, Paul Cheng, Francis Mussai
http://onlinelibrary.wiley.com/doi/10.1002/ijc.31170/full/

LC-MS-BASED METABOLOMICS REVEALED SLC25A22 AS AN ESSENTIAL REGULATOR OF ASPARTATE-DERIVED AMINO ACIDS AND POLYAMINES IN KRAS-MUTANT COLORECTAL CANCER
Xiaona Li, Arthur C.K. Chung, Shangfu Li, Lilan Wu, Jiaying Xu, Jun Yu, Chichun Wong,  and  Zongwei Cai
http://www.impactjournals.com/oncotarget/index.php?journal=oncotarget&page=article&op=view&path%5B%5D=21093&path%5B%5D=67162

———————————
Glucose and lactate
———————————

Regulation of the Hippo-YAP Pathway by Glucose Sensor O-GlcNAcylation
Changmin Peng, Yue Zhu, Wanjun Zhang, Qinchao Liao, Yali Chen, Xinyuan Zhao, Qiang Guo, Pan Shen, Bei Zhen, Xiaohong Qian, Dong Yang, Jin-San Zhang, Dongguang Xiao, Weijie Qin, Huadong Pei
http://www.cell.com/molecular-cell/fulltext/S1097-2765(17)30759-1

CDK8 Kinase Activity Promotes Glycolysis
Matthew D. Galbraith, Zdenek Andrysik, Ahwan Pandey, Maria Hoh, Elizabeth A. Bonner, Amanda A. Hill, Kelly D. Sullivan, Joaquín M. Espinosa
http://www.cell.com/cell-reports/fulltext/S2211-1247(17)31505-X

Pyruvate dehydrogenase kinase 4 exhibits a novel role in the activation of mutant KRAS, regulating cell growth in lung and colorectal tumour cells
A G Trinidad, N Whalley, R Rowlinson, O Delpuech, P Dudley, C Rooney and S E Critchlow
http://www.nature.com/onc/journal/v36/n44/abs/onc2017224a.html

Disrupting glucose-6-phosphate isomerase fully suppresses the “Warburg effect” and activates OXPHOS with minimal impact on tumor growth except in hypoxia
Monique Cunha de Padua, Giulia Delodi, Milica Vučetić, Jérôme Durivault, Valérie Vial, Pascale Bayer, Guilhermina Rodrigues Noleto, Nathalie M. Mazure, Maša Ždralević and Jacques Pouysségur
http://www.impactjournals.com/oncotarget/index.php?journal=oncotarget&page=article&op=view&path%5B0%5D=21007&path%5B1%5D=66903

MDA-MB-231 breast cancer cells fuel osteoclast metabolism and activity: A new rationale for the pathogenesis of osteolytic bone metastases
SilviaLemma, GemmaDi Pompo, Paolo E. Porporato, Martina Sboarina, Shonagh Russell, Robert J.Gillies, Nicola Baldini, Pierre Sonveaux, Sofia Avnet
http://www.sciencedirect.com/science/article/pii/S0925443917303125?via%3Dihub

LncRNA wires up Hippo and Hedgehog signaling to reprogramme glucose metabolism
Xin Zheng, Han Han, Guang‐Ping Liu, Yan‐Xiu Ma, Ruo‐Lang Pan, Ling‐Jie Sang, Rui‐Hua Li, Luo‐Jia Yang, Jeffrey R Marks, Wenqi Wang and Aifu Lin
http://EMBOJ.embopress.org/content/36/22/3325?etoc

SALL4 promotes glycolysis and chromatin remodeling via modulating HP1α-Glut1 pathway
J Kim, S Xu, L Xiong, L Yu, X Fu and Y Xu
https://www.nature.com/articles/onc2017265

————————————
P53 and metabolism
————————————

The p53‐inducible long noncoding RNA TRINGS protects cancer cells from necrosis under glucose starvation
Muhammad Riaz Khan, Shaoxun Xiang, Zhiyin Song and Mian Wu
http://emboj.embopress.org/content/36/23/3483?etoc

Effect of Mutant p53 Proteins on Glycolysis and Mitochondrial Metabolism
Matilda Eriksson, Gorbatchev Ambroise, Amanda Tomie Ouchida, Andre Lima Queiroz, Dominique Smith, Alfredo Gimenez-Cassina, Marcin P. Iwanicki, Patricia A. Muller, Erik Norberg and Helin Vakifahmetoglu-Norberg
http://mcb.asm.org/content/37/24/e00328-17.abstract?etoc

Cytoplasmic p53 couples oncogene-driven glucose metabolism to apoptosis and is a therapeutic target in glioblastoma
Wilson X Mai, Laura Gosa, Veerle W Daniels, Lisa Ta, Jonathan E Tsang, Brian Higgins, W Blake Gilmore, Nicholas A Bayley, Mitra Dehghan Harati, Jason T Lee, William H Yong, Harley I Kornblum, Steven J Bensinger, Paul S Mischel, P Nagesh Rao, Peter M Clark, Timothy F Cloughesy, Anthony Letai & David A Nathanson
https://www.nature.com/articles/nm.4418

———–
Lipids
———–

HIF drives lipid deposition and cancer in ccRCC via repression of fatty acid metabolism
Weinan Du, Luchang Zhang, Adina Brett-Morris, Brittany Aguila, Janos Kerner, Charles L. Hoppel, Michelle Puchowicz, Dolors Serra, Laura Herrero, Brian I. Rini, Steven Campbell & Scott M. Welford
https://www.nature.com/articles/s41467-017-01965-8

Elevated tumor LDLR expression accelerates LDL cholesterol-mediated breast cancer growth in mouse models of hyperlipidemia
E J Gallagher, Z Zelenko, B A Neel, I M Antoniou, L Rajan, N Kase and D LeRoith
https://www.nature.com/articles/onc2017247

Metabolomics guided pathway analysis reveals link between cancer metastasis, cholesterol sulfate, and phospholipids
Caroline H. Johnson, Antonio F. Santidrian, Sarah E. LeBoeuf, Michael E. Kurczy, Nicholas J. W. Rattray, Zahra Rattray, Benedikt Warth, Melissa Ritland, Linh T. Hoang, Celine Loriot, Jason Higa, James E. Hansen, Brunhilde H. Felding and Gary Siuzdak
https://cancerandmetabolism.biomedcentral.com/articles/10.1186/s40170-017-0171-2

A cytoplasmic COMPASS is necessary for cell survival and triple-negative breast cancer pathogenesis by regulating metabolism
Lu Wang, Clayton K. Collings, Zibo Zhao, Kira Alia Cozzolino, Quanhong Ma, Kaiwei Liang, Stacy A. Marshall, Christie C. Sze, Rintaro Hashizume, Jeffrey Nicholas Savas and Ali Shilatifard
http://genesdev.cshlp.org/content/31/20/2056.abstract?etoc

————————–
Mitochondria
————————–

Pyruvate dehydrogenase kinase 4 exhibits a novel role in the activation of mutant KRAS, regulating cell growth in lung and colorectal tumour cells
A G Trinidad, N Whalley, R Rowlinson, O Delpuech, P Dudley, C Rooney and S E Critchlow
https://www.nature.com/articles/onc2017224

Metformin regulates global DNA methylation via mitochondrial one-carbon metabolism
E Cuyàs, S Fernández-Arroyo, S Verdura, R Á-F García, J Stursa, L Werner, E Blanco-González, M Montes-Bayón, J Joven, B Viollet, J Neuzil & J A Menendez
https://www.nature.com/articles/onc2017367

PGC-1α Promotes Breast Cancer Metastasis and Confers Bioenergetic Flexibility against Metabolic Drugs
Sylvia Andrzejewski, Eva Klimcakova, Radia M. Johnson, Sébastien Tabariès, Matthew G. Annis, Shawn McGuirk, Jason J. Northey, Valérie Chénard, Urshila Sriram, David J. Papadopoli, Peter M. Siegel, Julie St-Pierre
http://www.cell.com/cell-metabolism/fulltext/S1550-4131(17)30557-0

Genetic Dissociation of Glycolysis and the TCA Cycle Affects Neither Normal nor Neoplastic Proliferation
Laura E. Jackson, Sucheta Kulkarni, Huabo Wang, Jie Lu, James M. Dolezal, Sivakama S. Bharathi, Sarangarajan Ranganathan, Mulchand S. Patel, Rahul Deshpande, Frances Alencastro, Stacy G. Wendell, Eric S. Goetzman, Andrew W. Duncan and Edward V. Prochownik
http://cancerres.aacrjournals.org/content/77/21/5795

A conserved mammalian mitochondrial isoform of acetyl-CoA carboxylase
ACC1 provides the malonyl-CoA essential for mitochondrial biogenesis in tandem with ACSF3
Geoffray Monteuuis, Fumi Suomi, Juha M. Kerätär, Ali J. Masud and Alexander J. Kastaniotis
http://www.biochemj.org/content/474/22/3783?etoc

SDHC promoter methylation, a novel pathogenic mechanism in parasympathetic paragangliomas
Cristóbal Bernardo-Castiñeira, Nuria Valdés, Marta I Sierra, Inés Sáenz-de-Santa-María, Gustavo F Bayón, Raúl F Perez, Agustín F Fernández, Mario F Fraga, Aurora Astudillo, Rafael Menéndez, Belén Fernández, Maribel del Olmo Carlos Suarez, María-Dolores Chiara
https://academic.oup.com/jcem/article-abstract/doi/10.1210/jc.2017-01702/4590228

Post-translational regulation of metabolism in fumarate hydratase deficient cancer cells
Emanuel Gonçalves, Marco Sciacovelli, Ana S.H. Costa, Maxine Gia Binh Tran, Timothy Isaac Johnson, Daniel Machado, Christian Frezza, Julio Saez-Rodriguez
http://www.sciencedirect.com/science/article/pii/S1096717617303270

MITOCHONDRIAL PHOSPHOENOLPYRUVATE CARBOXYKINASE (PEPCK-M) REGULATES THE CELL METABOLISM OF PANCREATIC NEUROENDOCRINE TUMORS (PNET) AND DE-SENSITIZES PNET TO MTOR INHIBITORS
Pei-Yi Chu, Shih Sheng Jiang, Yan-Shen Shan, Wen-Chun Hung, Ming-Huang Chen, Hui-You Lin, Yu-Lin Chen,Hui-Jen Tsai,  and  Li-Tzong Chen
http://www.impactjournals.com/oncotarget/index.php?journal=oncotarget&page=article&op=view&path%5B%5D=21665&path%5B%5D=68794

——————————————–
Metabolic stress / sensing
——————————————–

Sensing and Transmitting Intracellular Amino Acid Signals through Reversible Lysine Aminoacylations
Xia-Di He, Wei Gong, Jia-Nong Zhang, Ji Nie, Cui-Fang Yao, Fu-Shen Guo, Yan Lin, Xiao-Hui Wu, Feng Li, Jie Li, Wei-Cheng Sun, En-Duo Wang, Yan-Peng An, Hui-Ru Tang, Guo-Quan Yan, Peng-Yuan Yang, Yun Wei, Yun-Zi Mao, Peng-Cheng Lin, Jian-Yuan Zhao, Yanhui Xu, Wei Xu, Shi-Min Zhao
http://www.cell.com/cell-metabolism/fulltext/S1550-4131(17)30629-0

SAMTOR is an S-adenosylmethionine sensor for the mTORC1 pathway
Xin Gu, Jose M. Orozco, Robert A. Saxton, Kendall J. Condon, Grace Y. Liu, Patrycja A. Krawczyk, Sonia M. Scaria, J. Wade Harper, Steven P. Gygi, David M. Sabatini
http://science.sciencemag.org/content/358/6364/813

Oncogenic p95HER2/611CTF primes human breast epithelial cells for metabolic stress-induced down-regulation of FLIP and activation of TRAIL-R/Caspase-8-dependent apoptosis
Rosa Martín-Pérez, Rosario Yerbes, Rocío Mora-Molina, Ana Cano-González, Joaquín Arribas, Massimiliano Mazzone, Abelardo López-Rivas and Carmen Palacios
http://oncotarget.com/oncotarget/index.php?journal=oncotarget&page=article&op=view&path%5B%5D=21458

MTORC1 Regulates both General Autophagy and Mitophagy Induction after Oxidative Phosphorylation Uncoupling
Alberto Bartolomé, Ana García-Aguilar, Shun-Ichiro Asaharad, Yoshiaki Kido, Carlos Guillén, Utpal B. Pajvani and Manuel Benito
http://mcb.asm.org/content/37/23/e00441-17.abstract?etoc

Metabolic reprogramming ensures cancer cell survival despite oncogenic signaling blockade
Hui-wen Lue, Jennifer Podolak, Kevin Kolahi, Larry Cheng, Soumya Rao, Devin Garg, Chang-Hui Xue, Juha K. Rantala, Jeffrey W. Tyner, Kent L. Thornburg, Ann Martinez-Acevedo, Jen-Jane Liu, Christopher L. Amling, Charles Truillet, Sharon M. Louie, Kimberly E. Anderson, Michael J. Evans, Valerie B. O’Donnell, Daniel K. Nomura, Justin M. Drake, Anna Ritz and George V. Thomas
http://genesdev.cshlp.org/content/31/20/2067.abstract?etoc

———————————-
Hypoxia / hyperoxia
———————————-

NFS1 undergoes positive selection in lung tumours and protects cells from ferroptosis
Samantha W. Alvarez, Vladislav O. Sviderskiy, Erdem M. Terzi, Thales Papagiannakopoulos, Andre L. Moreira, Sylvia Adams, David M. Sabatini, Kıvanç Birsoy & Richard Possemato
https://www.nature.com/articles/nature24637

The interactome of metabolic enzyme carbonic anhydrase IX reveals novel roles in tumor cell migration and invadopodia/MMP14-mediated invasion
M Swayampakula, P C McDonald, M Vallejo, E Coyaud, S C Chafe, A Westerback, G Venkateswaran, J Shankar, G Gao, E M N Laurent, Y Lou, K L Bennewith, C T Supuran, I R Nabi, B Raught and S Dedhar
https://www.nature.com/articles/onc2017219

A circadian clock gene, PER2, activates HIF-1 as an effector molecule for recruitment of HIF-1α to promoter regions of its downstream genes
Minoru Kobayashi, Akiyo Morinibu, Sho Koyasu, Yoko Goto, Masahiro Hiraoka and Hiroshi Harada
http://onlinelibrary.wiley.com/doi/10.1111/febs.14280/abstract

Hypoxia and hypoxia-inducible factor (HIF) downregulate antigen-presenting MHC class I molecules limiting tumor cell recognition by T cells
Shalini Sethumadhavan, Murillo Silva, Phaethon Philbrook, Thao Nguyen, Stephen M. Hatfield, Akio Ohta, Michail V. Sitkovsky
http://journals.plos.org/plosone/article?id=10.1371/journal.pone.0187314

Parkin targets HIF-1α for ubiquitination and degradation to inhibit breast tumor progression
Juan Liu, Cen Zhang, Yuhan Zhao, Xuetian Yue, Hao Wu, Shan Huang, James Chen, Kyle Tomsky, Haiyang Xie, Christen A. Khella, Michael L. Gatza, Dajing Xia, Jimin Gao, Eileen White, Bruce G. Haffty, Wenwei Hu & Zhaohui Feng
https://www.nature.com/articles/s41467-017-01947-w

————————-
Miscellaneous
————————-

Caloric Restriction Prevents Carcinogen-Initiated Liver Tumorigenesis in Mice
Jonathan M. Ploeger, Juan C. Manivel, Lauren N. Boatner and Douglas G. Mashek
http://cancerpreventionresearch.aacrjournals.org/content/10/11/660

Branched‐chain ketoacids secreted by glioblastoma cells via MCT1 modulate macrophage phenotype
Lidia Santos Silva, Gernot Poschet, Yannic Nonnenmacher, Holger M Becker, Sean Sapcariu, Ann‐Christin Gaupel, Magdalena Schlotter, Yonghe Wu, Niclas Kneisel, Martina Seiffert, Rüdiger Hell, Karsten Hiller, Peter Lichter and Bernhard Radlwimmer
http://EMBOr.embopress.org/content/18/12/2172?etoc

Vps34 PI 3-kinase inactivation enhances insulin sensitivity through reprogramming of mitochondrial metabolism
Benoit Bilanges, Samira Alliouachene, […] Bart Vanhaesebroeck
https://www.nature.com/articles/s41467-017-01969-4

RANK rewires energy homeostasis in lung cancer cells and drives primary lung cancer
Shuan Rao, Verena Sigl, Reiner Alois Wimmer, Maria Novatchkova, Alexander Jais, Gabriel Wagner, Stephan Handschuh, Iris Uribesalgo, Astrid Hagelkruys, Ivona Kozieradzki, Luigi Tortola, Roberto Nitsch, Shane J. Cronin, Michael Orthofer, Daniel Branstetter, Jude Canon, John Rossi, Manolo D’Arcangelo, Johan Botling, Patrick Micke, Linnea La Fleur, Karolina Edlund, Michael Bergqvist, Simon Ekman, Thomas Lendl, Helmut Popper, Hiroshi Takayanagi, Lukas Kenner, Fred R. Hirsch, William Dougall and Josef M. Penninger
http://genesdev.cshlp.org/content/31/20/2099.abstract?etoc

——————
Reviews
——————

Focus: Metabolism: MYC and tumor metabolism: chicken and egg
Francesca R Dejure and Martin Eilers
http://EMBOJ.embopress.org/content/36/23/3409?etoc

The Glycogen Shunt Maintains Glycolytic Homeostasis and the Warburg Effect in Cancer
Robert G. Shulman, Douglas L. Rothman
http://www.cell.com/trends/cancer/fulltext/S2405-8033(17)30193-0

Mitochondrial OXPHOS Induced by RB1 Deficiency in Breast Cancer: Implications for Anabolic Metabolism, Stemness, and Metastasis
Eldad Zacksenhaus, Mariusz Shrestha, Jeff C. Liu, Ioulia Vorobieva, Philip E.D. Chung, YoungJun Ju, Uri Nir, Zhe Jiang
http://www.cell.com/trends/cancer/fulltext/S2405-8033(17)30176-0

Surviving Stress: Modulation of ATF4-Mediated Stress Responses in Normal and Malignant Cells
Inge M.N. Wortel, Laurens T. van der Meer, Michael S. Kilberg, Frank N. van Leeuwen
http://www.cell.com/trends/endocrinology-metabolism/abstract/S1043-2760(17)30100-5

Isocitrate dehydrogenases in physiology and cancer: biochemical and molecular insight
Hamoud Al-Khallaf
https://cellandbioscience.biomedcentral.com/articles/10.1186/s13578-017-0165-3

SREBP-regulated lipid metabolism: convergent physiology — divergent pathophysiology
Hitoshi Shimano & Ryuichiro Sato
https://www.nature.com/articles/nrendo.2017.91

Surviving Stress: Modulation of ATF4-Mediated Stress Responses in Normal and Malignant Cells
Inge M.N. Wortel , Laurens T. van der Meer , Michael S. Kilberg, Frank N. van Leeuwen
http://www.cell.com/trends/endocrinology-metabolism/fulltext/S1043-2760(17)30100-5

Understanding Mitochondrial Polymorphisms in Cancer
Karen M. Bussard and Linda D. Siracusa
http://cancerres.aacrjournals.org/content/77/22/6051

METABOLOMIC MAPPING OF CANCER STEM CELLS FOR REDUCING AND EXPLOITING TUMOR HETEROGENEITY
Elisabet Cuyàs, Sara Verdura, Salvador Fernández-Arroyo, Joaquim Bosch-Barrera, Begoña Martin-Castillo, Jorge Joven,  and  Javier A. Menendez
http://www.impactjournals.com/oncotarget/index.php?journal=oncotarget&page=article&op=view&path%5B%5D=21834&path%5B%5D=69257

ATM Modulation of Carbon Metabolism in Cancer
Erika S. Dahl and Katherine M. Aird
https://www.frontiersin.org/articles/10.3389/fonc.2017.00291/abstract

Non-Canonical Mechanisms Regulating Hypoxia-Inducible Factor 1 Alpha in Cancer
Luisa Iommarini, Anna Maria Porcelli, Giuseppe Gasparre and Ivana Kurelac
https://www.frontiersin.org/articles/10.3389/fonc.2017.00286/full

E2F1, a Novel Regulator of Metabolism
Pierre-Damien Denechaud, Lluis Fajas and Albert Giralt
https://www.frontiersin.org/articles/10.3389/fendo.2017.00311/full

Mitochondria, Bioenergetics and Apoptosis in Cancer
Peter J. Burke
http://www.cell.com/trends/cancer/fulltext/S2405-8033(17)30200-5

Mitochondrial Transfer in the Leukemia Microenvironment
Emmanuel Griessinger, Ruxanda Moschoi, Giulia Biondani, Jean-François Peyron
http://www.cell.com/trends/cancer/fulltext/S2405-8033(17)30197-8

Drosophila as a Model to Study the Link between Metabolism and Cancer
Héctor Herranz  and Stephen M. Cohen
http://www.mdpi.com/2221-3759/5/4/15

————————————–
Comments / Previews
————————————–

Resistance Is Futile: Targeting Mitochondrial Energetics and Metabolism to Overcome Drug Resistance in Cancer Treatment
Claudie Bosc, Mary A. Selak, Jean-Emmanuel Sarry
http://www.cell.com/cell-metabolism/fulltext/S1550-4131(17)30627-7

AMPK: keeping the (power)house in order?
Claire Thornton
http://www.neuronalsignaling.org/content/1/2/NS20160020

The folate cycle is a new metabolic weakness of cancer
Derek Lee & Carmen Chak-Lui Wong
http://www.tandfonline.com/doi/full/10.1080/23723556.2017.1327004

Sugar industry sponsorship of germ-free rodent studies linking sucrose to hyperlipidemia and cancer: An historical analysis of internal documents
Cristin E. Kearns, Dorie Apollonio, Stanton A. Glantz
http://journals.plos.org/plosbiology/article?id=10.1371/journal.pbio.2003460

Metabolism: More lactate, please
https://www.nature.com/articles/nrc.2017.101

Serving cancer its last meal
http://stm.sciencemag.org/content/9/415/eaaq1233.full

Fatty-Acid Catabolism Promotes T-cell Revitalization in Melanoma
http://cancerdiscovery.aacrjournals.org/content/7/11/1213.2