MetaboList – February 2023


Nucleotides


Allosteric regulation of CAD modulates de novo pyrimidine synthesis during the cell cycle – Jong Shin, Hannan Mir, Maaz A. Khurram, Kenji M. Fujihara, Brian D. Dynlacht, Timothy J. Cardozo and Richard Possemato. https://www.nature.com/articles/s42255-023-00735-9

Nucleotide metabolism is linked to cysteine availability during ferroptosis – Annamarie E. Allen, Yudong Sun, Fangchao Wei, Michael A. Reid, Jason W. Locasale. https://www.jbc.org/article/S0021-9258(23)00171-0

p53 promotes peroxisomal fatty acid β-oxidation to repress purine biosynthesis and mediate tumor suppression – Jianhong Zhao, Xiaojun Zhou, Baoxiang Chen, Mingzhu Lu, Genxin Wang, Nagarajan Elumalai, Chenhui Tian, Jinmiao Zhang, Yanliang Liu, Zhiqiang Chen, Xinyi Zhou, Mingzhi Wu, Mengjiao Li, Edward V. Prochownik, Ali Tavassoli, Congqing Jiang and Youjun Li. https://www.nature.com/articles/s41419-023-05625-

NADK-mediated de novo NADP(H) synthesis is a metabolic adaptation essential for breast cancer metastasis – Didem Ilter, Stanislav Drapela, Tanya Schild, Nathan P. Ward, Emma Adhikari, Vivien Low, John Asara, Thordur Oskarsson, Eric K. Lau, Gina M. DeNicola, Melanie R. McReynolds, Ana P. Gomes. https://www.sciencedirect.com/science/article/pii/S2213231723000289


Mitochondria / Hypoxia


Mitochondrial respiration reduces exposure of the nucleus to oxygen – Mateus Prates Mori, Rozhin Penjweini, Jin Ma, Greg Alspaugh, Alessio Andreoni, Young-Chae Kim, Ping-Yuan Wang, Jay R. Knutson, Paul M. Hwang. https://www.jbc.org/article/S0021-9258(23)00150-3

Long noncoding RNA GATA2-AS1 augments endothelial Hypoxia Inducible Factor 1-α induction and regulates hypoxic signaling – H.S. Jeffrey Man, Noeline Subramaniam, Tiana Downs, Aravin N. Sukumar, Aninda D. Saha, Ranju Nair, Lucy Chen, Daniel Teitelbaum, Paul J. Turgeon, Kyung Ha Ku, Eileen Tran, Marc de Perrot, Philip A. Marsden. https://www.jbc.org/article/S0021-9258(23)00161-8

Mitochondrial SENP2 regulates the assembly of SDH complex under metabolic stress – Ying Liu, Kejia Liu, Rick F. Thorne, Ronghua Shi, Qingyuan Zhang, Mian Wu, Lianxin Liu. https://www.cell.com/cell-reports/fulltext/S2211-1247(23)00052-9

Mitochondrial uncoupling induces epigenome remodeling and promotes differentiation in neuroblastoma – Haowen Jiang, Rachel Greathouse, Sarah J. Tiche, Man Zhao, Bo He, Yang Li, Albert M. Li, Balint Forgo, Michaela Yip, Allison Li, Moriah Shih, Selene Banuelos, Meng-Ning Zhou, Joshua J. Gruber, Erinn B. Rankin, Zhen Hu, Hiroyuki Shimada, Bill Chiu, Jiangbin Ye.  https://aacrjournals.org/cancerres/article/83/2/181/715072


Amino acid / Glucose metabolism


Enhanced Glutaminolysis Drives Hypoxia-Induced Chemoresistance in Pancreatic Cancer – Seung Joon Park, Hee Chan Yoo, Eunyong Ahn, Enzhi Luo, Yeabeen Kim, Yulseung Sung, Ya Chun Yu, Kibum Kim, Do Sik Min, Hee Seung Lee, Geum-Sook Hwang, TaeJin Ahn, Junjeong Choi, Seungmin Bang, Jung Min Han. https://aacrjournals.org/cancerres/article-abstract/83/5/735/716680

PHGDH arginine methylation by PRMT1 promotes serine synthesis and represents a therapeutic vulnerability in hepatocellular carcinoma – Kui Wang, Li Luo, Shuyue Fu, Mao Wang, Zihao Wang, Lixia Dong, Xingyun Wu, Lunzhi Dai, Yong Peng, Guobo Shen, Hai-Ning Chen, Edouard Collins Nice, Xiawei Wei and Canhua Huang. https://www.nature.com/articles/s41467-023-36708-5

Elevated Methionine Flux Drives Pyroptosis Evasion in Persister Cancer Cells – Asmaa El-Kenawi, Anders Berglund, Veronica Estrella, Yonghong Zhang, Min Liu, Ryan M. Putney, Sean J. Yoder, Joseph Johnson, Joel Brown, Robert Gatenby. https://aacrjournals.org/cancerres/article/83/5/720/716681

Breast cancer cells that preferentially metastasize to lung or bone are more glycolytic, synthesize serine at greater rates, and consume less ATP and NADPH than parent MDA-MB-231 cells – Mika B. Jekabsons, Mollie Merrell, Anna G. Skubiz, Noah Thornton, Sandra Milasta, Douglas Green, Taosheng Chen, Yan-Hong Wang, Bharathi Avula, Ikhlas A. Khan and Yu-Dong Zhou. https://cancerandmetabolism.biomedcentral.com/articles/10.1186/s40170-023-00303-5


Starvation / mTOR


Cancer cells co-opt nociceptive nerves to thrive in nutrient-poor environments and upon nutrient-starvation therapies – Yu Zhang, Chengzhong Lin, Zheqi Liu, Yiting Sun, Mingtao Chen Yibo Guo, Wei Liu, Chenping Zhang, Wantao Chen, Jian Sun, Ronghui Xia, Yuhua Hu, Xi Yang, Jiang Li, Zhiyuan Zhang, Wei Cao, Shuyang Sun, Xu Wang, Tong Ji. https://www.sciencedirect.com/science/article/pii/S1550413122004909

Snf1/AMPK fine-tunes TORC1 signaling in response to glucose starvation – Marco Caligaris, Raffaele Nicastro Is a corresponding author, Zehan Hu, Farida Tripodi, Johannes Erwin Hummel, Benjamin Pillet, Marie-Anne Deprez, Joris Winderickx, Sabine Rospert, Paola Coccetti, Jörn Dengjel, Claudio De Virgilio. https://elifesciences.org/articles/84319

Monitoring the 5′UTR landscape reveals isoform switches to drive translational efficiencies in cancer – Ramona Weber, Umesh Ghoshdastider, Daniel Spies, Clara Duré, Fabiola Valdivia-Francia, Merima Forny, Mark Ormiston, Peter F. Renz, David Taborsky, Merve Yigit, Martino Bernasconi, Homare Yamahachi and Ataman Sendoel. https://www.nature.com/articles/s41388-022-02578-2

TSC2 regulates tumor susceptibility to TRAIL‐mediated T‐cell killing by orchestrating mTOR signaling – Chun-Pu Lin, Joleen J H Traets, David W Vredevoogd, Nils L Visser, Daniel S Peeper. https://www.embopress.org/doi/abs/10.15252/embj.2022111614


IDH / Lipids


Dysregulated Lipid Synthesis by Oncogenic IDH1 Mutation Is a Targetable Synthetic Lethal Vulnerability – Daniel Thomas, Manhong Wu, Yusuke Nakauchi, Ming Zheng, Chloe A.L. Thompson-Peach, Kelly Lim, Niklas Landberg, Thomas Köhnke, Nirmal Robinson, Satinder Kaur, Monika Kutyna, Melissa Stafford, Devendra Hiwase, Andreas Reinisch, Gary Peltz, Ravindra Majeti. https://aacrjournals.org/cancerdiscovery/article/13/2/496/716313

IDH2 stabilizes HIF‐1α‐induced metabolic reprogramming and promotes chemoresistance in urothelial cancer – Keisuke Shigeta, Masanori Hasegawa, Takako Hishiki, Yoshiko Naito, Yuto Baba, Shuji Mikami, Kazuhiro Matsumoto, Ryuichi Mizuno, Akira Miyajima, Eiji Kikuchi, Hideyuki Saya, Takeo Kosaka, Mototsugu Oya. https://www.embopress.org/doi/full/10.15252/embj.2022110620

Cytoskeletal association of ATP citrate lyase controls the mechanodynamics of macropinocytosis – Joseph Puccini, Jia Wei, Liang Tong and Dafna Bar-Sagi. https://www.pnas.org/doi/10.1073/pnas.2213272120


Cancer Immunometabolism


Extracellular acidosis restricts one-carbon metabolism and preserves T cell stemness – Hongcheng Cheng, Yajing Qiu, Yue Xu, Li Chen, Kaili Ma, Mengyuan Tao, Luke Frankiw, Hongli Yin, Ermei Xie, Xiaoli Pan, Jing Du, Zhe Wang, Wenjie Zhu, Lu Chen, Lianjun Zhang and Guideng Li. https://www.nature.com/articles/s42255-022-00730-6

Metabolic adaptation supports enhanced macrophage efferocytosis in limited-oxygen environments – Ya-Ting Wang, Alissa J. Trzeciak, Waleska Saitz Rojas, Pedro Saavedra, Yan-Ting Chen, Rachel Chirayil, Jon Iker Etchegaray, Christopher D. Lucas, Daniel J. Puleston, Kayvan R. Keshari, Justin S.A. Perry. https://www.cell.com/cell-metabolism/fulltext/S1550-4131(22)00542-3


Miscellaneous


Blockage of autophagosome-lysosome fusion through SNAP29 O-GlcNAcylation promotes apoptosis via ROS production – Francesca Romana Pellegrini, Sara De Martino, Giulia Fianco, Irene Ventura, Davide Valente, Mario Fiore, Daniela Trisciuoglio and Francesca Degrassi. https://www.tandfonline.com/doi/full/10.1080/15548627.2023.2170962

CRISPR metabolic screen identifies ATM and KEAP1 as targetable genetic vulnerabilities in solid tumors – Haojian Li, Yue Liu, Yunjie Xiao, Crystal N. Wilson, Hui Jen Bai, Maxwell D. Jones, Shihchun Wang, Jennie E. DeVore, Esther Y. Maier, Stephen T. Durant, Myriem Boufraqech and Urbain Weyemi. https://www.pnas.org/doi/10.1073/pnas.2212072120

Temporal segregation of biosynthetic processes is responsible for metabolic oscillations during the budding yeast cell cycle – Vakil Takhaveev, Serdar Özsezen, Edward N. Smith, Andre Zylstra, Marten L. Chaillet, Haoqi Chen, Alexandros Papagiannakis, Andreas Milias-Argeitis and Matthias Heinemann. https://www.nature.com/articles/s42255-023-00741-x


Reviews


Acetyl-CoA metabolism in cancer – David A. Guertin and Kathryn E. Wellen. https://www.nature.com/articles/s41568-022-00543-5

Metabolism and epigenetics at the heart of T cell function – Leticia Soriano-Baguet, Dirk Brenner. https://www.cell.com/trends/immunology/fulltext/S1471-4906(23)00002-9

Control of immune cell function by the unfolded protein response – Giusy Di Conza, Ping-Chih Ho, Juan R. Cubillos-Ruiz and Stanley Ching-Cheng Huang. https://www.nature.com/articles/s41577-023-00838-0

The magic bullet: Niclosamide – Haowen Jiang, Albert M. Li, Jiangbin Ye. https://www.frontiersin.org/articles/10.3389/fonc.2022.1004978/full

Fasting and fasting mimicking diets in cancer prevention and therapy – Olga Blaževitš, Maira Di Tano, Valter D. Longo. https://www.cell.com/trends/cancer/fulltext/S2405-8033(22)00268-0

Unfolded protein response and angiogenesis in malignancies – Amin Izadpanah, Kurtis Willingham, Bysani Chandrasekar, Eckhard U. Alt, Reza Izadpanah. https://www.sciencedirect.com/science/article/abs/pii/S0304419X22001640

Effect of metabolism on the immune microenvironment of breast cancer – Yingming Zhou, Hu Wang, Yi Luo, Biguang Tuo, Xuemei Liu, Taolang Li. https://www.sciencedirect.com/science/article/pii/S0304419X23000100

Nutrient transporters: connecting cancer metabolism to therapeutic opportunities – Zeribe Chike Nwosu, Mun Gu Song, Marina Pasca di Magliano, Costas A. Lyssiotis and Sung Eun Kim. https://www.nature.com/articles/s41388-023-02593-x


Comments


Fatty acids prime the lung as a site for tumour spread – Laura V. Pinheiro, Kathryn E. Wellen. https://www.nature.com/articles/d41586-023-00538-8

Oncogenic IDH1 Mutation Imparts Therapeutically Targetable Metabolic Dysfunction in Multiple Tumor Types – Troy M. Robinson, Ross L. Levine. https://aacrjournals.org/cancerdiscovery/article-abstract/13/2/266/716305

MetaboList – January 2023


Warburg metabolism


Slow TCA flux and ATP production in primary solid tumours but not metastases – Caroline R. Bartman, Daniel R. Weilandt, Yihui Shen, Won Dong Lee, Yujiao Han, Tara TeSlaa, Connor S. R. Jankowski, Laith Samarah, Noel R. Park, Victoria da Silva-Diz, Maya Aleksandrova, Yetis Gultekin, Argit Marishta, Lin Wang, Lifeng Yang, Asael Roichman, Vrushank Bhatt, Taijin Lan, Zhixian Hu, Xi Xing, Wenyun Lu, Shawn Davidson, Martin Wühr, Matthew G. Vander Heiden, Daniel Herranz, Jessie Yanxiang Guo, Yibin Kang and Joshua D. Rabinowitz. https://www.nature.com/articles/s41586-022-05661-6

Lactylome analysis suggests lactylation-dependent mechanisms of metabolic adaptation in hepatocellular carcinoma – Zijian Yang, Cong Yan, Jiaqiang Ma, Panpan Peng, Xuelian Ren, Shangli Cai, Xia Shen, Yingcheng Wu, Shu Zhang, Xiaoying Wang, Shuangjian Qiu, Jian Zhou, Jia Fan, He Huang and Qiang Gao. https://www.nature.com/articles/s42255-022-00710-w


Amino acid metabolism


Arginase-1 inhibition reduces migration ability and metastatic colonization of colon cancer cells – Xiangdong Wang, Huihui Xiang, Yujiro Toyoshima, Weidong Shen, Shunsuke Shichi, Hiroki Nakamoto, Saori Kimura, Ko Sugiyama, Shigenori Homma, Yohei Miyagi, Akinobu Taketomi and Hidemitsu Kitamura. https://cancerandmetabolism.biomedcentral.com/articles/10.1186/s40170-022-00301-z

Autophagy loss impedes cancer-associated fibroblast activation via downregulating proline biosynthesis – Jingru Bai, Tong Liu, Bo Tu, Meng Yuan, Zhaoqi Shu, Minghe Fan, Sihan Huo, Yuyao Guo, Lina Wang, Hua Wang and Ying Zhao. https://www.tandfonline.com/doi/abs/10.1080/15548627.2022.2093026


Lipid metabolism


A palmitate-rich metastatic niche enables metastasis growth via p65 acetylation resulting in pro-metastatic NF-κB signaling – Patricia Altea-Manzano, Ginevra Doglioni, Yawen Liu, Alejandro M. Cuadros, Emma Nolan, Juan Fernández-García, Qi Wu, Mélanie Planque, Kathrin Julia Laue, Florencia Cidre-Aranaz, Xiao-Zheng Liu, Oskar Marin-Bejar, Joke Van Elsen, Ines Vermeire, Dorien Broekaert, Sofie Demeyer, Xander Spotbeen, Jakub Idkowiak, Aurélie Montagne, Margherita Demicco, H. Furkan Alkan, Nick Rabas, Carla Riera-Domingo, François Richard, Tatjana Geukens, Maxim De Schepper, Sophia Leduc, Sigrid Hatse, Yentl Lambrechts, Emily Jane Kay, Sergio Lilla, Alisa Alekseenko, Vincent Geldhof, Bram Boeckx, Celia de la Calle Arregui, Giuseppe Floris, Johannes V. Swinnen, Jean-Christophe Marine, Diether Lambrechts, Vicent Pelechano, Massimiliano Mazzone, Sara Zanivan, Jan Cools, Hans Wildiers, Véronique Baud, Thomas G. P. Grünewald, Uri Ben-David, Christine Desmedt, Ilaria Malanchi and Sarah-Maria Fendt. https://www.nature.com/articles/s43018-023-00513-2

Stearate‐derived very long‐chain fatty acids are indispensable to tumor growth – Qiaoyun Chu, Ping Liu, Yihan Song, Ronghui Yang, Jing An, Xuewei Zhai, Jing Niu, Chuanzhen Yang, Binghui Li. https://www.embopress.org/doi/abs/10.15252/embj.2022111268


Mitochondria and TCA


Complex I inhibitor of oxidative phosphorylation in advanced solid tumors and acute myeloid leukemia: phase I trials – Timothy A. Yap, Naval Daver, Mikhila Mahendra, Jixiang Zhang, Carlos Kamiya-Matsuoka, Funda Meric-Bernstam, Hagop M. Kantarjian, Farhad Ravandi, Meghan E. Collins, Maria Emilia Di Francesco, Ecaterina E. Dumbrava, Siqing Fu, Sisi Gao, Jason P. Gay, Sonal Gera, Jing Han, David S. Hong, Elias J. Jabbour, Zhenlin Ju, Daniel D. Karp, Alessia Lodi, Jennifer R. Molina, Natalia Baran, Aung Naing, Maro Ohanian, Shubham Pant, Naveen Pemmaraju, Prithviraj Bose, Sarina A. Piha-Paul, Jordi Rodon, Carolina Salguero, Koji Sasaki, Anand K. Singh, Vivek Subbiah, Apostolia M. Tsimberidou, Quanyun A. Xu, Musa Yilmaz, Qi Zhang, Yuan Li, Christopher A. Bristow, Meenakshi B. Bhattacharjee, Stefano Tiziani, Timothy P. Heffernan, Christopher P. Vellano, Philip Jones, Cobi J. Heijnen, Annemieke Kavelaars, Joseph R. Marszalek and Marina Konopleva. https://www.nature.com/articles/s41591-022-02103-8

Gene signature predicting recurrence in oral squamous cell carcinoma is characterized by increased oxidative phosphorylation – Joo Kyung Noh, Seon Rang Woo, Moonkyoo Kong, Min Kyeong Lee, Jung Woo Lee, Young Chan Lee, Seong-Gyu Ko, Young-Gyu Eun. https://febs.onlinelibrary.wiley.com/doi/full/10.1002/1878-0261.13328

Structural basis of mammalian respiratory complex I inhibition by medicinal biguanides – Hannah R. Bridges, James N. Blaza, Zhan Yin, Injae Chung, Michael N. Pollak, Judy Hirst. https://www.science.org/doi/abs/10.1126/science.ade3332


Starvation


The antitumour effects of caloric restriction are mediated by the gut microbiome – Yu-Qin Mao, Jia-Ting Huang, Shi-Long Zhang, Chao Kong, Zhan-Ming Li, Hui Jing, Hui-Ling Chen, Chao-Yue Kong, Sheng-Hui Huang, Pei-Ran Cai, Bing Han and Li-Shun Wang. https://www.nature.com/articles/s42255-022-00716-4

Phosphorylation of EIF2S1 (eukaryotic translation initiation factor 2 subunit alpha) is indispensable for nuclear translocation of TFEB and TFE3 during ER stress – Thao Thi Dang, Mi-Jeong Kim, Yoon Young Lee, Hien Thi Le, Kook Hwan Kim, Somi Nam, Seung Hwa Hyun, Hong Lim Kim, Su Wol Chung, Hun Taeg Chung, Eek-Hoon Jho, Hiderou Yoshida, Kyoungmi Kim, Chan Young Park, Myung-Shik Lee and Sung Hoon Back. https://www.tandfonline.com/doi/abs/10.1080/15548627.2023.2173900


mTOR


AMPK-dependent phosphorylation of the GATOR2 component WDR24 suppresses glucose-mediated mTORC1 activation – Xiaoming Dai, Cong Jiang, Qiwei Jiang, Lan Fang, Haihong Yu, Jinhe Guo, Peiqiang Yan, Fangtao Chi, Tao Zhang, Hiroyuki Inuzuka, John M. Asara, Ping Wang, Jianping Guo and Wenyi Wei. https://www.nature.com/articles/s42255-022-00732-4

Ring domains are essential for GATOR2-dependent mTORC1 activation – Cong Jiang, Xiaoming Dai, Shaohui He, Hongfei Zhou, Lan Fang, Jianping Guo, Songlei Liu, Tao Zhang, Weijuan Pan, Haihong Yu, Tianmin Fu, Dali Li, Hiroyuki Inuzuka, Ping Wang, Jianru Xiao, Wenyi Wei. https://www.cell.com/molecular-cell/fulltext/S1097-2765(22)01134-0

Dynamic metabolome profiling uncovers potential TOR signaling genes – Stella Reichling, Peter F Doubleday, Tomas Germade, Ariane Bergmann, Robbie Loewith, Uwe Sauer, Duncan Holbrook-Smith. https://elifesciences.org/articles/84295


IDH


Natural and Synthetic 2-Oxogluturate Derivatives are Substrates for Oncogenic Variants of Human Isocitrate Dehydrogenase 1 and 2 – Xiao Liu, Raphael Reinbold, Shuang Liu, Ryan A. Herold, Patrick Rabe, Stéphanie Duclos, Rahul B. Yadav, Martine I. Abboud, Sandrine Thieffine, Fraser A. Armstrong, Lennart Brewitz, Christopher J. Schofield. https://www.jbc.org/article/S0021-9258(23)00005-4/fulltext


Cancer Immunometabolism


Oxidative phosphorylation selectively orchestrates tissue macrophage homeostasis – Stefanie K. Wculek, Ignacio Heras-Murillo, Annalaura Mastrangelo, Diego Mañanes, Miguel Galán, Verónica Miguel, Andrea Curtabbi, Coral Barbas, Navdeep S. Chandel, José Antonio Enríquez, Santiago Lamas, David Sancho. https://www.cell.com/immunity/pdfExtended/S1074-7613(23)00021-3


Miscellaneous


Cell-cell metabolite exchange creates a pro-survival metabolic environment that extends lifespan – Clara Correia-Melo, Stephan Kamrad, Roland Tengölics, Christoph B. Messner, Pauline Trebulle, StJohn Townsend, Sreejith Jayasree Varma, Anja Freiwald, Benjamin M. Heineike, Kate Campbell, Lucía Herrera-Dominguez, Simran Kaur Aulakh, Lukasz Szyrwiel, Jason S.L. Yu, Aleksej Zelezniak, Vadim Demichev, Michael Mülleder, Balázs Papp, Mohammad Tauqeer Alam, Markus Ralser. https://www.cell.com/cell/fulltext/S0092-8674(22)01520-3

Metabolic profiling reveals metabolic features of consolidation therapy in pediatric acute lymphoblastic leukemia – Jinqiu Fu, Aijun Zhang, Qinqin Liu, Dong Li, Xiaoming Wang and Libo Si. https://cancerandmetabolism.biomedcentral.com/articles/10.1186/s40170-023-00302-6

Insulin-regulated serine and lipid metabolism drive peripheral neuropathy – Michal K. Handzlik, Jivani M. Gengatharan, Katie E. Frizzi, Grace H. McGregor, Cameron Martino, Gibraan Rahman, Antonio Gonzalez, Ana M. Moreno, Courtney R. Green, Lucie S. Guernsey, Terry Lin, Patrick Tseng, Yoichiro Ideguchi, Regis J. Fallon, Amandine Chaix, Satchidananda Panda, Prashant Mali, Martina Wallace, Rob Knight, Marin L. Gantner, Nigel A. Calcutt and Christian M. Metallo. https://www.nature.com/articles/s41586-022-05637-6

Progressive development of melanoma-induced cachexia differentially impacts organ systems in mice – Flavia A. Graca, Anna Stephan, Yong-Dong Wang, Abbas Shirinifard, Jianqin Jiao, Peter Vogel, Myriam Labelle, Fabio Demontis. https://www.cell.com/cell-reports/fulltext/S2211-1247(22)01835-6


Reviews


Metabolites as signalling molecules – Steven Andrew Baker & Jared Rutter. https://www.nature.com/articles/s41580-022-00572-w

Diverse effects of obesity on antitumor immunity and immunotherapy – Lydia Dyck, Lydia Lynch. https://www.cell.com/trends/molecular-medicine/fulltext/S1471-4914(22)00294-5


Comments


Time to hit pause on mitochondria-targeting cancer therapies – Xue Zhang & Chi V. Dang. https://www.nature.com/articles/s41591-022-02129-y

AMPK knocks at the gate of GATOR – Nerea Deleyto-Seldas & Alejo Efeyan. https://www.nature.com/articles/s42255-022-00729-z

GATOR2 rings GATOR1 to speak to mTORC1 – Umakant Sahu, Issam Ben-Sahra. https://www.cell.com/molecular-cell/fulltext/S1097-2765(22)01168-6

Thrifty energy metabolism in solid tumours – https://www.nature.com/articles/d41586-023-00151-9

MetaboList – December 2022


Amino acid metabolism


Dynamic partitioning of branched-chain amino acids-derived nitrogen supports renal cancer progression – Marco Sciacovelli, Aurelien Dugourd, Lorea Valcarcel Jimenez, Ming Yang, Efterpi Nikitopoulou, Ana S. H. Costa, Laura Tronci, Veronica Caraffini, Paulo Rodrigues, Christina Schmidt, Dylan Gerard Ryan, Timothy Young, Vincent R. Zecchini, Sabrina H. Rossi, Charlie Massie, Caroline Lohoff, Maria Masid, Vassily Hatzimanikatis, Christoph Kuppe, Alex Von Kriegsheim, Rafael Kramann, Vincent Gnanapragasam, Anne Y. Warren, Grant D. Stewart, Ayelet Erez, Sakari Vanharanta, Julio Saez-Rodriguez and Christian Frezza. https://www.nature.com/articles/s41467-022-35036-4

In vivo characterization of glutamine metabolism identifies therapeutic targets in clear cell renal cell carcinoma – Akash K. Kaushik, Amy Tarangelo, Lindsey K. Boroughs, Mukundan Ragavan, Yuanyuan Zhang, Cheng-Yang Wu, Xiangyi Li, Kristen Ahumada, Jui-Chung Chiang, Vanina T. Tcheuyap, Faeze Saatchi, Quyen N. Do, Cissy Yong, Tracy Rosales, Christina Stevens, Aparna D. Rao, Brandon Faubert, Panayotis Pachnis, Lauren G. Zacharias, Hieu Vu, Feng Cai, Thomas P. Mathews, Giannicola Genovese, Barbara S. Slusher, Payal Kapur, Xiankai Sun, Matthew Merritt, James Brugarolas, Ralph J. DeBerardinis. https://www.science.org/doi/full/10.1126/sciadv.abp8293

The m6A reader IGF2BP2 regulates glutamine metabolism and represents a therapeutic target in acute myeloid leukemia – Hengyou Weng, Feng Huang, Zhaojin Yu, Zhenhua Chen, Emily Prince, Yalin Kang, Keren Zhou, Wei Li, Jiacheng Hu, Chen Fu, Tursunjan Aziz, Hongzhi Li, Jingwen Li, Ying Yang, Li Han, Subo Zhang, Yuelong Ma, Mingli Sun, Huizhe Wu, Zheng Zhang, Mark Wunderlich, Sean Robinson, Daniel Braas, Johanna ten Hoeve, Bin Zhang, Guido Marcucci, James C. Mulloy, Keda Zhou, Hong-Fang Tao, Xiaolan Deng, David Horne, Minjie Wei, Huilin Huang, Jianjun Chen. https://www.cell.com/cancer-cell/fulltext/S1535-6108(22)00495-0

Glutamine synthetase limits β-catenin–mutated liver cancer growth by maintaining nitrogen homeostasis and suppressing mTORC1 – Weiwei Dai, Jianliang Shen, Junrong Yan, Alex J. Bott, Sara Maimouni, Heineken Q. Daguplo, Yujue Wang, Khoosheh Khayati, Jessie Yanxiang Guo, Lanjing Zhang, Yongbo Wang, Alexander Valvezan, Wen-Xing Ding, Xin Chen, Xiaoyang Su, Shenglan Gao and Wei-Xing Zong. https://www.jci.org/articles/view/161408

Gut flora disequilibrium promotes the initiation of liver cancer by modulating tryptophan metabolism and up-regulating SREBP2 – Wen Chen, Liang Wen, Yingying Bao, Zengwei Tang, Jianhui Zhao, Xiaozhen Zhang, Tao Wei, Jian Zhang, Tao Ma, Qi Zhang, Xiao Zhi, Jin Li, Cheng Zhang, Lei Ni, Muchun Li and Tingbo Liang. https://www.pnas.org/doi/10.1073/pnas.2203894119


Lipid/Acetyl CoA metabolism


mTORC1 regulates a lysosome-dependent adaptive shift in intracellular lipid species – Aaron M. Hosios, Meghan E. Wilkinson, Molly C. McNamara, Krystle C. Kalafut, Margaret E. Torrence, John M. Asara and Brendan D. Manning. https://www.nature.com/articles/s42255-022-00706-6

Acetyl-CoA-mediated autoacetylation of fatty acid synthase as a metabolic switch of de novo lipogenesis in Drosophila – Ting Miao, Jinoh Kim, Ping Kang, Hideji Fujiwara, Fong-Fu Hsu and Hua Bai. https://www.pnas.org/doi/10.1073/pnas.2212220119

Alterations in the omics profiles in mevalonate pathway-inhibited cancer cells – Tomoko Warita, Nanami Irie, Yaxuan Zhou, Jiro Tashiro, Akihiro Sugiura, Zoltán N Oltvai, Katsuhiko Warita.. https://www.sciencedirect.com/science/article/pii/S0024320522009493

Monounsaturated and Diunsaturated Fatty Acids Sensitize Cervical Cancer to Radiation Therapy – Naoshad Muhammad, Fiona Ruiz, Jennifer Stanley, Ramachandran Rashmi, Kevin Cho, Kay Jayachandran, Michael C. Zahner, Yi Huang, Jin Zhang, Stephanie Markovina, Gary J. Patti, Julie K. Schwarz. https://aacrjournals.org/cancerres/article-abstract/82/24/4515/711531


Moonlighting functions of metabolic proteins


Metabolic enzyme LDHA activates Rac1 GTPase as a noncanonical mechanism to promote cancer – Juan Liu, Cen Zhang, Tianliang Zhang, Chun-Yuan Chang, Jianming Wang, Ludvinna Bazile, Lanjing Zhang, Bruce G. Haffty, Wenwei Hu and Zhaohui Feng. https://www.nature.com/articles/s42255-022-00708-4

Non‐canonical phosphoglycerate dehydrogenase activity promotes liver cancer growth via mitochondrial translation and respiratory metabolism – Ying Shu, Yijie Hao, Junru Feng, Haiying Liu, Shi-ting Li, Jiaqian Feng, Zetan Jiang, Ling Ye, Yingli Zhou, Yuchen Sun, Zilong Zhou, Haoran Wei, Ping Gao, Huafeng Zhang, Linchong Sun. https://www.embopress.org/doi/abs/10.15252/embj.2022111550

Pyruvate kinase M2 mediates IL-17 signaling in keratinocytes driving psoriatic skin inflammation – Flávio P. Veras, Gabriel A. Publio, Bruno M. Melo, Douglas S. Prado, Thainá Norbiato, Nerry T. Cecilio, Carlos Hiroki, Luis Eduardo A. Damasceno, Rebecca Jung, Juliana E. Toller-Kawahisa, Timna V. Martins, Stella F. Assunção, Diogenes Lima, Marcia G. Alves, Gabriel V. Vieira, Lucas A. Tavares, Ana L.R. Alves-Rezende, Susanne H. Karbach, Helder I. Nakaya, Thiago M. Cunha, Cacilda S. Souza, Fernando Q. Cunha, Katiuchia U. Sales, Ari Waisman, José C. Alves-Filho. https://www.cell.com/cell-reports/fulltext/S2211-1247(22)01796-X

Hypoxic activation of PFKFB4 in breast tumor microenvironment shapes metabolic and cellular plasticity to accentuate metastatic competence – Tao Dai, Spencer R. Rosario, Eriko Katsuta, Abhisha Sawant Dessai, Emily J. Paterson, Aaron T. Novickis, Eduardo Cortes Gomez, Bokai Zhu, Song Liu, Hai Wang, Scott I. Abrams, Mukund Seshadri, Wiam Bshara, Subhamoy Dasgupta. https://www.cell.com/cell-reports/fulltext/S2211-1247(22)01639-4


Glucose/lactate/nucleotide metabolism


Lactate dehydrogenases promote glioblastoma growth and invasion via a metabolic symbiosis – Joris Guyon, Ignacio Fernandez-Moncada, Claire M Larrieu, Cyrielle L Bouchez, Antonio C Pagano Zottola, Johanna Galvis, Tiffanie Chouleur, Audrey Burban, Kevin Joseph, Vidhya M Ravi, Heidi Espedal, Gro Vatne Røsland, Boutaina Daher, Aurélien Barre, Benjamin Dartigues, Slim Karkar, Justine Rudewicz, Irati Romero-Garmendia, Barbara Klink, Konrad Grützmann, Marie-Alix Derieppe, Thibaut Molinié, Nina Obad, Céline Léon, Giorgio Seano, Hrvoje Miletic, Dieter Henrik Heiland, Giovanni Marsicano, Macha Nikolski, Rolf Bjerkvig, Andreas Bikfalvi, Thomas Daubon. https://www.embopress.org/doi/full/10.15252/emmm.202115343

Blocking lncRNA-SNHG16 sensitizes gastric cancer cells to 5-Fu through targeting the miR-506-3p-PTBP1-mediated glucose metabolism – Yan Ding, Sujie Gao, Jiabin Zheng and Xuebo Chen. https://cancerandmetabolism.biomedcentral.com/articles/10.1186/s40170-022-00293-w

Cancer-selective metabolic vulnerabilities in MYC-amplified medulloblastoma – William D. Gwynne, Yujin Suk, Stefan Custers, Nicholas Mikolajewicz, Jeremy K. Chan, Zsolt Zador, Shawn C. Chafe, Kui Zhai, Laura Escudero, Cunjie Zhang, Olga Zaslaver, Chirayu Chokshi, Muhammad Vaseem Shaikh, David Bakhshinyan, Ian Burns, Iqra Chaudhry, Omri Nachmani, Daniel Mobilio, William T. Maich, Patricia Mero, Kevin R. Brown, Andrew T. Quaile, Chitra Venugopal, Jason Moffat, J. Rafael Montenegro-Burke, Sheila K. Singh. https://www.cell.com/cancer-cell/fulltext/S1535-6108(22)00500-1


Mitochondria / Hypoxia


Endosomal lipid signaling reshapes the endoplasmic reticulum to control mitochondrial function – Wonyul Jang, Dmytro Puchkov, Paula Samsó, YongTian Liang, Michal Nadler-Holly, Stephan J. Sigrist, Ulrich Kintscher, Fan Liu, Kamel Mamchaoui, Vincent Mouly, Volker Haucke. https://www.science.org/doi/abs/10.1126/science.abq5209

Pyruvate dehydrogenase phosphatase 1 (PDP1) stimulates HIF activity by supporting histone acetylation under hypoxia – Angeliki Karagiota, Amalia Kanoura, Efrosyni Paraskeva, George Simos, Georgia Chachami. https://febs.onlinelibrary.wiley.com/doi/10.1111/febs.16694

Mitochondria directly sense osmotic stress to trigger rapid metabolic remodeling via regulation of pyruvate dehydrogenase (PDH) phosphorylation – Takeshi Ikizawa, Kazutaka Ikeda, Makoto Arita, Shojiro Kitajima, Tomoyoshi Soga, Hidenori Ichijo, Isao Naguro. https://www.jbc.org/article/S0021-9258(22)01280-7

A Phase II Randomized Trial of Chemoradiation with or without Metformin in Locally Advanced Cervical Cancer – Kathy Han, Anthony Fyles, Tina Shek, Jennifer Croke, Neesha Dhani, David D’Souza, Ting-Yim Lee, Naz Chaudary, Jeffrey Bruce, Melania Pintilie, Rob Cairns, Douglass Vines, Sara Pakbaz, David Jaffray, Ur Metser, Marjan Rouzbahman, Michael Milosevic, Marianne Koritzinsky. https://aacrjournals.org/clincancerres/article-abstract/28/24/5263/711455

Lactate-dependent chaperone-mediated autophagy induces oscillatory HIF-1α activity promoting proliferation of hypoxic cells – Kshitiz, Junaid Afzal. Yasir Suhail, Hao Chang, Maimon E. Hubbi, Archer Hamidzadeh, Ruchi Goyal, Yamin Liu, Peng Sun, Stefania Nicoli, Chi V. Dang, Andre Levchenko. https://www.cell.com/cell-systems/fulltext/S2405-4712(22)00462-8

Exploratory meta-analysis of hypoxic transcriptomes using a precise transcript reference sequence set – Yoko Ono and Hidemasa Bono. https://www.life-science-alliance.org/content/6/1/e202201518.abstract


Starvation / AMPK


Energy sensor AMPK gamma regulates translation via phosphatase PPP6C independent of AMPK alpha – Qi Zhou, Bingbing Hao, Xiaolei Cao, Lin Gao, Zhenyue Yu, Yang Zhao, Mingrui Zhu, Guoxuan Zhong, Fangtao Chi, Xiaoming Dai, Jizhong Mao, Yibing Zhu, Ping Rong, Liang Chen, Xueli Bai, Cunqi Ye, Shuai Chen, Tingbo Liang, Li Li, Xin-Hua Feng, Minjia Tan, Bin Zhao. https://www.cell.com/molecular-cell/fulltext/S1097-2765(22)01058-9

A Subset of VEGFR-TKIs Activates AMPK in LKB1-mutant Lung Cancer – Lujie Yang, Qin Zhang, Yanli Xiong, Zhaoqian Dang, He Xiao, Qian Chen, Xiaoyan Dai, Lei Zhang, Jianwu Zhu, Dong Wang, Mengxia Li. https://onlinelibrary.wiley.com/doi/10.1111/cas.15677

Blocking AMPK β1 myristoylation enhances AMPK activity and protects mice from high-fat diet-induced obesity and hepatic steatosis – Katyayanee Neopane, Natalie Kozlov, Florentina Negoita, Lisa Murray-Segal, Robert Brink, Ashfaqul Hoque, Ashley J. Ovens, Gavin Tjin, Luke M. McAloon, Dingyi Yu, Naomi X.Y. Ling, Matthew J. Sanders, Jonathan S. Oakhill, John W. Scott, Gregory R. Steinberg, Kim Loh, Bruce E. Kemp, Kei Sakamoto, Sandra Galic. https://www.cell.com/cell-reports/fulltext/S2211-1247(22)01758-2

Mitochondrial genome recovery by ATFS-1 is essential for development after starvation – Nandhitha Uma Naresh, Sookyung Kim, Tomer Shpilka, Qiyuan Yang, Yunguang Du, Cole M. Haynes. https://www.cell.com/cell-reports/fulltext/S2211-1247(22)01771-5

4EBP1 senses extracellular glucose deprivation and initiates cell death signaling in lung cancer – Yanan Wang, Jiapeng Lei, Song Zhang, Xiaomei Wang, Jiangbo Jin, Yufeng Liu, Mingxi Gan, Yi Yuan, Longhua Sun, Xiaolei Li, Tianyu Han and Jian-Bin Wang. https://www.nature.com/articles/s41419-022-05466-5


mTOR


Dihydroxyacetone suppresses mTOR nutrient signaling and induces mitochondrial stress in liver cells – Arlet Hernandez, Manoj Sonavane, Kelly R Smith, Jensyn Seiger, Marie E Migaud, Natalie R Gassman. https://journals.plos.org/plosone/article?id=10.1371/journal.pone.0278516

Cell Type-Specific Metabolic Response to Amino Acid Starvation Dictates the Role of Sestrin2 in Regulation of mTORC1 – Biljana Blagojevic, Fadi Almouhanna, Gernot Poschet, Stefan Wölfl. https://pubmed.ncbi.nlm.nih.gov/36497120/

Wdr59 promotes or inhibits TORC1 activity depending on cellular context – Yingbiao Zhang, Chun-Yuan Ting, Shu Yang, John Reich, Karenne Fru and Mary A. Lilly. https://www.pnas.org/doi/10.1073/pnas.2212330120

V-ATPase/TORC1-mediated ATFS-1 translation directs mitochondrial UPR activation in C. elegans – Terytty Yang Li, Arwen W. Gao, Xiaoxu Li, Hao Li, Yasmine J. Liu, Amelia Lalou, Nagammal Neelagandan, Felix Naef, Kristina Schoonjans, Johan Auwerx. https://rupress.org/jcb/article/222/1/e202205045/213623


Cancer Immunometabolism


Targeting the bicarbonate transporter SLC4A4 overcomes immunosuppression and immunotherapy resistance in pancreatic cancer – Federica Cappellesso, Marie-Pauline Orban, Niranjan Shirgaonkar, Emanuele Berardi, Jens Serneels, Marie-Aline Neveu, Daria Di Molfetta, Francesca Piccapane, Rosa Caroppo, Lucantonio Debellis, Tessa Ostyn, Nicolas Joudiou, Lionel Mignion, Elena Richiardone, Bénédicte F. Jordan, Bernard Gallez, Cyril Corbet, Tania Roskams, Ramanuj DasGupta, Sabine Tejpar, Mario Di Matteo, Daniela Taverna, Stephan J. Reshkin, Baki Topal, Federico Virga and Massimiliano Mazzone. https://www.nature.com/articles/s43018-022-00470-2

Immunoediting instructs tumor metabolic reprogramming to support immune evasion – Chin-Hsien Tsai, Yu-Ming Chuang, Xiaoyun Li, Yi-Ru Yu, Sheue-Fen Tzeng, Shao Thing Teoh, Katherine E. Lindblad, Mario Di Matteo, Wan-Chen Cheng, Pei-Chun Hsueh, Kung-Chi Kao, Hana Imrichova, Likun Duan, Hector Gallart-Ayala, Pei-Wen Hsiao, Massimiliano Mazzone, Julijana Ivanesevic, Xiaojing Liu, Karin E. de Visser, Amaia Lujambio, Sophia Y. Lunt, Susan M. Kaech, Ping-Chih Ho. https://www.cell.com/cell-metabolism/fulltext/S1550-4131(22)00540-X

IL-2 is inactivated by the acidic pH environment of tumors enabling engineering of a pH-selective mutein – Silvia Gaggero, Jonathan Martinez-Fabregas, Adeline Cozzani, Paul K. Fyfe, Malo Leprohon, Jie Yang, F. Emil Thomasen, Hauke Winkelmann, Romain Magnez, Alberto G. Conti, Stephan Wilmes, Elizabeth Pohler, Manuel van Gijsel Bonnello, Xavier Thuru, Bruno Quesnel, Fabrice Soncin, Jacob Piehler, Kresten Lindorff-Larsen, Rahul Roychoudhuri, Ignacio Moraga, Suman Mitra. https://www.science.org/doi/full/10.1126/sciimmunol.ade5686

Inhibition of Glucose Uptake Blocks Proliferation but Not Cytotoxic Activity of NK Cells – Lea Katharina Picard, Elisabeth Littwitz-Salomon, Herbert Waldmann, Carsten Watzl. https://www.mdpi.com/2073-4409/11/21/3489

Myeloid-derived itaconate suppresses cytotoxic CD8+ T cells and promotes tumour growth – Hongyun Zhao, Da Teng, Lifeng Yang, Xincheng Xu, Jiajia Chen, Tengjia Jiang, Austin Y. Feng, Yaqing Zhang, Dennie T. Frederick, Lei Gu, Li Cai, John M. Asara, Marina Pasca di Magliano, Genevieve M. Boland, Keith T. Flaherty, Kenneth D. Swanson, David Liu, Joshua D. Rabinowitz and Bin Zheng. https://www.nature.com/articles/s42255-022-00676-9

Metabolic adaptation supports enhanced macrophage efferocytosis in limited-oxygen environments – Ya-Ting Wang, Alissa J. Trzeciak, Waleska Saitz Rojas, Pedro Saavedra, Yan-Ting Chen, Rachel Chirayil, Jon Iker Etchegaray, Christopher D. Lucas, Daniel J. Puleston, Kayvan R. Keshari, Justin S.A. Perry. https://www.cell.com/cell-reports/fulltext/S2211-1247(22)01639-4

Isotope tracing reveals distinct substrate preference in murine melanoma subtypes with differing anti-tumor immunity – Xinyi Zhang, Alexandra A. Halberstam, Wanling Zhu, Brooks P. Leitner, Durga Thakral, Marcus W. Bosenberg and Rachel J. Perry. https://cancerandmetabolism.biomedcentral.com/articles/10.1186/s40170-022-00296-7

Lactate induces metabolic and epigenetic reprogramming of pro‐inflammatory Th17 cells – Aleksandra Lopez Krol, Hannah P Nehring, Felix F Krause, Anne Wempe, Hartmann Raifer, Andrea Nist, Thorsten Stiewe, Wilhelm Bertrams, Bernd Schmeck, Maik Luu, Hanna Leister, Ho-Ryun Chung, Uta-Maria Bauer, Till Adhikary, Alexander Visekruna. https://www.embopress.org/doi/abs/10.15252/embr.202254685

Increased tumor glycolysis is associated with decreased immune infiltration across human solid tumors – Ivan J Cohen, Fresia Pareja, Nicholas D Socci, Ronglai Shen, Ashley S Doane, Jazmin Schwartz, Raya Khanin, Elizabeth A Morris, Elizabeth J Sutton, Ronald G Blasberg. https://www.frontiersin.org/articles/10.3389/fimmu.2022.880959/full

Microenvironmental ammonia enhances T cell exhaustion in colorectal cancer – Hannah N. Bell, Amanda K. Huber, Rashi Singhal, Navyateja Korimerla, Ryan J. Rebernick, Roshan Kumar, Marwa O. El-derany, Peter Sajjakulnukit, Nupur K. Das, Samuel A. Kerk, Sumeet Solanki, Jadyn G. James, Donghwan Kim, Li Zhang, Brandon Chen, Rohit Mehra, Timothy L. Frankel, Balázs Győrffy, Eric R. Fearon, Marina Pasca di Magliano, Frank J. Gonzalez, Ruma Banerjee, Daniel R. Wahl, Costas A. Lyssiotis, Michael Green, Yatrik M. Shah. https://www.cell.com/cell-metabolism/fulltext/S1550-4131(22)00504-6


Miscellaneous


Coupling-dependent metabolic ultradian rhythms in confluent cells – Shuzhang Yang, Shin Yamazaki, Kimberly H. Cox, Yi-Lin Huang, Evan W. Miller and Joseph S. Takahashi. https://www.pnas.org/doi/abs/10.1073/pnas.2211142119

Disrupting metformin adaptation of liver cancer cells by targeting the TOMM34/ATP5B axis – Ping Jin, Jingwen Jiang, Li Zhou, Zhao Huang, Siyuan Qin, Hai-Ning Chen, Liyuan Peng, Zhe Zhang, Bowen Li, Maochao Luo, Tingting Zhang, Hui Ming, Ning Ding, Lei Li, Na Xie, Wei Gao, Wei Zhang, Edouard C Nice, Yuquan Wei, Canhua Huang. https://www.embopress.org/doi/full/10.15252/emmm.202216082

Mouse tissue harvest-induced hypoxia rapidly alters the in vivo metabolome, between-genotype metabolite level differences, and 13C-tracing enrichments – Adam J. Rauckhorst, Nicholas Borcherding, Daniel J. Pape, Alora S. Kraus, Diego A. Scerbo, Eric B. Taylor. https://www.sciencedirect.com/science/article/pii/S221287782200165X

Serum metabolic traits reveal therapeutic toxicities and responses of neoadjuvant chemoradiotherapy in patients with rectal cancer – Hongmiao Wang, Huixun Jia, Yang Gao, Haosong Zhang, Jin Fan, Lijie Zhang, Fandong Ren, Yandong Yin, Yuping Cai, Ji Zhu and Zheng-Jiang Zhu. https://www.nature.com/articles/s41467-022-35511-y

Effects of cancer-associated point mutations on the structure, function, and stability of isocitrate dehydrogenase 2 – Xiang Chen, Peipei Yang, Yue Qiao, Fei Ye, Zhipeng Wang, Mengting Xu, Xiaowang Han, Li Song, Yuehong Wu, Wen-Bin Ou. https://www.nature.com/articles/s41598-022-23659-y

ATM inhibition drives metabolic adaptation via induction of macropinocytosis – Zhentai Huang, Chi-Wei Chen, Raquel Buj, Naveen Kumar Tangudu, Richard S. Fang, Kelly E. Leon, Erika S. Dahl, Erika L. Varner, Eliana von Krusenstiern, Aidan R. Cole, Nathaniel W. Snyder, Katherine M. Aird. https://rupress.org/jcb/article/222/1/e202007026/213697


Reviews


Regulation and function of the mammalian tricarboxylic acid cycle – Paige K. Arnold, Lydia W.S. Finley. https://www.jbc.org/article/S0021-9258(22)01281-9/fulltext

Targeting fatty acid metabolism in glioblastoma – Jason Miska and Navdeep S. Chandel. https://www.jci.org/articles/view/163448

Monitoring and modelling the dynamics of the cellular glycolysis pathway: A review and future perspectives – Nitin Patil, Orla Howe, Paul Cahill, Hugh J. Byrne. https://www.sciencedirect.com/science/article/pii/S2212877822002046

Cytosolic lipolysis in non‐adipose tissues: energy provision and beyond – Siyu Chen, Xun Huang. https://febs.onlinelibrary.wiley.com/doi/full/10.1111/febs.16161

Iron and liver cancer: an inseparable connection – Keisuke Hino, Izumi Yanatori, Yuichi Hara, Sohji Nishina. https://febs.onlinelibrary.wiley.com/doi/full/10.1111/febs.16208

Emerging roles of ferroptosis in infectious diseases – Salimata Bagayoko, Etienne Meunier. https://febs.onlinelibrary.wiley.com/doi/full/10.1111/febs.16244

Obesity Programs Macrophages to Support Cancer Progression – Abhijeet Singh, Shyamananda Singh Mayengbam, Himanshi Yaduvanshi, Mohan R. Wani, Manoj Kumar Bhat. https://aacrjournals.org/cancerres/article-abstract/82/23/4303/710689

mTORC1 beyond anabolic metabolism: Regulation of cell death – Jiajun Zhu, Hua Wang, Xuejun Jiang. https://rupress.org/jcb/article/221/12/e202208103/213609

Tryptophan and its metabolites in normal physiology and cancer etiology – Lizbeth Perez-Castro, Roy Garcia, Niranjan Venkateswaran, Spencer Barnes, Maralice Conacci-Sorrell. https://febs.onlinelibrary.wiley.com/doi/full/10.1111/febs.16245


Comments


Bicarbonate transport as a vulnerability in pancreatic cancer – Li Qiang and Stephanie K. Dougan. https://www.nature.com/articles/s43018-022-00492-w

Revealing de novo pyrimidine synthesis as a key vulnerability in brain tumors – Tanya Schild, Kayvan R. Keshari. https://www.cell.com/cancer-cell/fulltext/S1535-6108(22)00520-7

Fat Fuels the Fire in Cervical Cancer – Kassidy M. Jungles, Michael D. Green. https://aacrjournals.org/cancerres/article-abstract/82/24/4513/711516

mTORC1 takes control of lysosomal lipid breakdown – Laura Tribouillard and Mathieu Laplante. https://www.nature.com/articles/s42255-022-00702-w

Beyond Warburg: LDHA activates RAC for tumour growth – Natsuski Osaka and Atsuo T. Sasaki. https://www.nature.com/articles/s42255-022-00709-3

Early immune pressure makes tumors metabolically stronger – Sujing Yuan, Hongbo Chi. https://www.cell.com/cell-metabolism/fulltext/S1550-4131(22)00546-0

Illuminating lactate in cells, mice, and patient samples – Zefan Li, Hui-wang Ai. https://www.cell.com/cell-metabolism/fulltext/S1550-4131(22)00544-7

Suppression of CD8+ T cells by the metabolite itaconate – https://www.nature.com/articles/s42255-022-00694-7

Metabolist – November 2022


AMPK


AMPK directly phosphorylates TBK1 to integrate glucose sensing into innate immunity – Qian Zhang, Shengduo Liu, Chen-Song Zhang, Qirou Wu, Xinyuan Yu, Ruyuan Zhou, Fansen Meng, Ailian Wang, Fei Zhang, Shasha Chen, Xiaojian Wang, Lei Li, Jun Huang, Yao-Wei Huang, Jian Zou, Jun Qin, Tingbo Liang, Xin-Hua Feng, Sheng-Cai Lin, Pinglong Xu. https://www.cell.com/molecular-cell/fulltext/S1097-2765(22)01054-1

circPRKAA1 activates a Ku80/Ku70/SREBP-1 axis driving de novo fatty acid synthesis in cancer cells – Qidong Li, Hanhui Yao, Yichun Wang, Yang Wu, Rick F. Thorne, Youming Zhu, Mian Wu, Lianxin Liu. https://www.cell.com/cell-reports/fulltext/S2211-1247(22)01581-9

AMPK induces degradation of the transcriptional repressor PROX1 impairing branched amino acid metabolism and tumourigenesis – Yanan Wang, Mengjun Luo, Fan Wang, Yu Tong, Linfeng Li, Yu Shu, Ke Qiao, Lei Zhang, Guoquan Yan, Jing Liu, Hongbin Ji, Youhua Xie, Yonglong Zhang, Wei-Qiang Gao and Yanfeng Liu. https://www.nature.com/articles/s41467-022-34747-y

AMPK-dependent phosphorylation of MTFR1L regulates mitochondrial morphology – Lisa Tilokani, Fiona M. Russell, Stevie Hamilton, Daniel M. Virga, Mayuko Segawa, Vincent Paupe, Anja V. Gruszczyk, Margherita Protasoni, Luis-Carlos Tabara, Mark Johnson, Hanish Anand, Michael P. Murphy, D. Grahame Hardie, Franck Polleux, Julien Prudent. https://www.science.org/doi/full/10.1126/sciadv.abo7956

The phosphorylation of AMPKβ1 is critical for increasing autophagy and maintaining mitochondrial homeostasis in response to fatty acids – Eric M. Desjardins, Brennan K. Smith, Emily A. Day, Serge Ducommun, Matthew J. Sanders, Joshua P. Nederveen, Rebecca J. Ford, Stephen L. Pinkosky, Logan K. Townsend, Robert M. Gutgesell, Rachel Lu, Kei Sakamoto and Gregory R. Steinberg. https://www.pnas.org/doi/10.1073/pnas.2119824119


mTOR


Reciprocal effects of mTOR inhibitors on pro-survival proteins dictate therapeutic responses in tuberous sclerosis complex – McNamara MC, Hosios AM, Torrence ME, Zhao T, Fraser C, Wilkinson M, Kwiatkowski DJ, Henske EP, Wu CL, Sarosiek KA, Valvezan AJ, Manning BD. https://www.cell.com/iscience/fulltext/S2589-0042(22)01730-8

An mTORC1-mediated negative feedback loop constrains amino acid-induced FLCN-Rag activation in renal cells with TSC2 loss – Kaushal Asrani, Juhyung Woo, Adrianna A. Mendes, Ethan Schaffer, Thiago Vidotto, Clarence Rachel Villanueva, Kewen Feng, Lia Oliveira, Sanjana Murali, Hans B. Liu, Daniela C. Salles, Brandon Lam, Pedram Argani and Tamara L. Lotan. https://www.nature.com/articles/s41467-022-34617-7

mTOR inhibition attenuates chemosensitivity through the induction of chemotherapy resistant persisters – Yuanhui Liu, Nancy G. Azizian, Delaney K. Sullivan and Yulin Li. https://www.nature.com/articles/s41467-022-34890-6

An mTORC1 to HRI signaling axis promotes cytotoxicity of proteasome inhibitors in multiple myeloma – Odai Darawshi, Barbara Muz, Shiri Gershon Naamat, Bellam Praveen, Mohamed Mahameed, Karin Goldberg, Priya Dipta, Miriam Shmuel, Francesca Forno, Shatha Boukeileh, Hadas Pahima, Julia Hermann, Marc S. Raab, Alexandra M. Poos, Niels Weinhold, Chaggai Rosenbluh, Moshe E. Gatt, Wilhelm Palm, Abdel Kareem Azab and Boaz Tirosh. https://www.nature.com/articles/s41419-022-05421-4


Starvation/metabolic stress


Identification of purine biosynthesis as an NADH-sensing pathway to mediate energy stress – Ronghui Yang, Chuanzhen Yang, Lingdi Ma, Yiliang Zhao, Zihao Guo, Jing Niu, Qiaoyun Chu, Yingmin Ma and Binghui Li. https://www.nature.com/articles/s41467-022-34850-0

Phosphoproteomics revealed cellular signals immediately responding to disruption of cancer amino acid homeostasis induced by inhibition of L-type amino acid transporter 1 – Hiroki Okanishi, Ryuichi Ohgaki, Minhui Xu, Hitoshi Endou and Yoshikatsu Kanai. https://cancerandmetabolism.biomedcentral.com/articles/10.1186/s40170-022-00295-8

Carbon starvation, senescence and specific mitochondrial stresses, but not nitrogen starvation and general stresses, are major triggers for mitophagy in Arabidopsis – Sylwia M. Kacprzak and Olivier Van Aken. https://www.tandfonline.com/doi/full/10.1080/15548627.2022.2054039

COX7A1 enhances the sensitivity of human NSCLC cells to cystine deprivation-induced ferroptosis via regulating mitochondrial metabolism – Yetong Feng, Jiayi Xu, Mengjiao Shi, Rongrong Liu, Lei Zhao, Xin Chen, Miaomiao Li, Yaping Zhao, Jiahui Chen, Wenjing Du and Pengfei Liu. https://www.nature.com/articles/s41419-022-05430-3


Glucose metabolism


Fructose-1,6-bisphosphatase 1 functions as a protein phosphatase to dephosphorylate histone H3 and suppresses PPARα-regulated gene transcription and tumour growth – Zheng Wang, Min Li, Hongfei Jiang, Shudi Luo, Fei Shao, Yan Xia, Mengke Yang, Xiangle Ren, Tong Liu, Meisi Yan, Xu Qian, Haiyan He, Dong Guo, Yuran Duan, Ke Wu, Lei Wang, Guimei Ji, Yuli Shen, Lin Li, Peixiang Zheng, Bofei Dong, Jing Fang, Min Zheng, Tingbo Liang, Haitao Li, Rilei Yu, Daqian Xu and Zhimin Lu. https://www.nature.com/articles/s41556-022-01009-4

BRAF activation by metabolic stress promotes glycolysis sensitizing NRASQ61-mutated melanomas to targeted therapy – Kimberley McGrail, Paula Granado-Martínez, Rosaura Esteve-Puig, Sara García-Ortega, Yuxin Ding, Sara Sánchez-Redondo, Berta Ferrer, Javier Hernandez-Losa, Francesc Canals, Anna Manzano, Aura Navarro-Sabaté, Ramón Bartrons, Oscar Yanes, Mileidys Pérez-Alea, Eva Muñoz-Couselo, Vicenç Garcia-Patos and Juan A. Recio

https://www.nature.com/articles/s41467-022-34907-0

ALKBH5-Mediated m6A Demethylation of GLUT4 mRNA Promotes Glycolysis and Resistance to HER2-Targeted Therapy in Breast Cancer – Hao Liu, Hui Lyu, Guanmin Jiang, Danyang Chen, Sanbao Ruan, Shuang Liu, Lukun Zhou, Minqiang Yang, Shanshan Zeng, Zhimin He, Hongsheng Wang, Hongsheng Li, Guopei Zheng, Bolin Liu. https://aacrjournals.org/cancerres/article-abstract/82/21/3974/709951

GLUT1 is a Highly Efficient L-Fucose Transporter – Bobby G. Ng, Paulina Sosicka, Zhijie Xia, Hudson H. Freeze. https://www.jbc.org/article/S0021-9258(22)01181-4

Mutual regulation between phosphofructokinase 1 platelet isoform and VEGF promotes glioblastoma tumor growth – Je Sun Lim, YuJie Shi, Su Hwan Park, So Mi Jeon, Chuanbao Zhang, Yun-Yong Park, Rui Liu, Jing Li, Wan-Seob Cho, Linyong Du and Jong-Ho Lee. https://www.nature.com/articles/s41419-022-05449-6

Targeting cancer glycosylation repolarizes tumor-associated macrophages allowing effective immune checkpoint blockade – Michal A. Stanczak, Natalia Rodrigues Mantuano, Nicole Kirchhammer, David E. Sanin, Francis Jacob, Ricardo Coelho, Arun V. Everest-Dass, Jinyu Wang, Marcel P. Trefny, Gianni Monaco, Anne Bärenwaldt, Melissa A. Gray, Adam Petrone, Abhishek S. Kashyap, Katharina Glatz, Benjamin Kasenda, Karl Normington, James Broderick, Li Peng, Oliver M.T. Pearce, Erika L. Pearce, Carolyn R. Bertozzi, Alfred Zippelius, Heinz Läubli. https://www.science.org/doi/abs/10.1126/scitranslmed.abj1270


Amino acid metabolism


Cancer cell survival depends on collagen uptake into tumor-associated stroma – Kuo-Sheng Hsu, James M. Dunleavey, Christopher Szot, Liping Yang, Mary Beth Hilton, Karen Morris, Steven Seaman, Yang Feng, Emily M. Lutz, Robert Koogle, Francesco Tomassoni-Ardori, Saurabh Saha, Xiaoyan M. Zhang, Enrique Zudaire, Pradip Bajgain, Joshua Rose, Zhongyu Zhu, Dimiter S. Dimitrov, Frank Cuttitta, Nancy J. Emenaker, Lino Tessarollo and Brad St. Croix. https://www.nature.com/articles/s41467-022-34643-5

Branched-chain amino acid catabolism breaks glutamine addiction to sustain hepatocellular carcinoma progression – Dongdong Yang, Haiying Liu, Yongping Cai, Kangyang Lu, Xiuying Zhong, Songge Xing, Wei Song, Yaping Zhang, Ling Ye, Xia Zhu, Ting Wang, Pinggen Zhang, Shi-Ting Li, Jiaqian Feng, Weidong Jia, Huafeng Zhang, Ping Gao. https://www.sciencedirect.com/science/article/pii/S2211124

Differential integrated stress response and asparagine production drive symbiosis and therapy resistance of pancreatic adenocarcinoma cells – Christopher J. Halbrook, Galloway Thurston, Seth Boyer, Cecily Anaraki, Jennifer A. Jiménez, Amy McCarthy, Nina G. Steele, Samuel A. Kerk, Hanna S. Hong, Lin Lin, Fiona V. Law, Catherine Felton, Lorenzo Scipioni, Peter Sajjakulnukit, Anthony Andren, Alica K. Beutel, Rima Singh, Barbara S. Nelson, Fran Van Den Bergh, Abigail S. Krall, Peter J. Mullen, Li Zhang, Sandeep Batra, Jennifer P. Morton, Ben Z. Stanger, Heather R. Christofk, Michelle A. Digman, Daniel A. Beard, Andrea Viale, Ji Zhang, Howard C. Crawford, Marina Pasca di Magliano, Claus Jorgensen and Costas A. Lyssiotis. https://www.nature.com/articles/s43018-022-00463-1

Inhibition of glutaminolysis restores mitochondrial function in senescent stem cells – Debanik Choudhury, Na Rong, Izuagie Ikhapoh, Nika Rajabian, Georgios Tseropoulos, Yulun Wu, Pihu Mehrotra, Ramkumar Thiyagarajan, Aref Shahini, Kenneth L. Seldeen, Bruce R. Troen, Pedro Lei, Stelios T. Andreadis. https://www.cell.com/cell-reports/fulltext/S2211-1247(22)01622-9


Lipid / propionate /acetate metabolism


Tumor-produced and aging-associated oncometabolite methylmalonic acid promotes cancer-associated fibroblast activation to drive metastatic progression – Zhongchi Li, Vivien Low, Valbona Luga, Janet Sun, Ethan Earlie, Bobak Parang, Kripa Shobana Ganesh, Sungyun Cho, Jennifer Endress, Tanya Schild, Mengying Hu, David Lyden, Wenbing Jin, Chunjun Guo, Noah Dephoure, Lewis C. Cantley, Ashley M. Laughney and John Blenis. https://www.nature.com/articles/s41467-022-33862-0

Lipid Metabolic Reprogramming Extends beyond Histologic Tumor Demarcations in Operable Human Pancreatic Cancer – Juho Pirhonen, Ábel Szkalisity, Jaana Hagström, Yonghyo Kim, Ede Migh, Mária Kovács, Maarit Hölttä, Johan Peränen, Hanna Seppänen, Caj Haglund, Jeovanis Gil, Melinda Rezeli, Johan Malm, Peter Horvath, György Markó-Varga, Pauli Puolakkainen, Elina Ikonen. https://aacrjournals.org/cancerres/article-abstract/82/21/3932/709964

Acetyl-CoA carboxylase 1 depletion suppresses de novo fatty acid synthesis and mitochondrial β-oxidation in castration-resistant prostate cancer cells – Shaoyou Liu, Jiarun Lai, Yuanfa Feng, Yangjia Zhuo, Hui Zhang, Yupeng Chen, Jinchuang Li, Xinyue Mei, Yanting Zeng, Jiaming Su, Yulin Deng, Funeng Jiang, Shengbang Yang, Huijing Tan, Chi Tin Hon, Sun Wei, Zhaodong Han, Fen Wang, Weide Zhong. https://www.jbc.org/article/S0021-9258(22)01163-2/fulltext

Transcription factors TEAD2 and E2A globally repress acetyl-CoA synthesis to promote tumorigenesis – Sujin Park, Dirk Mossmann, Qian Chen, Xueya Wang, Eva Dazert, Marco Colombi, Alexander Schmidt, Brendan Ryback, Charlotte K.Y. Ng, Luigi M. Terracciano, Markus H. Heim, Michael N. Hall. https://www.cell.com/molecular-cell/fulltext/S1097-2765(22)01055-3


Mitochondrial metabolism and impairment


Reversal of mitochondrial malate dehydrogenase 2 enables anaplerosis via redox rescue in respiration-deficient cells – Patricia Altea-Manzano, Anke Vandekeere, Joy Edwards-Hicks, Mar Roldan,  Emily Abraham, Xhordi Lleshi, Ania Naila Guerrieri, Domenica Berardi, Jimi Wills, Jair Marques Junior, Asimina Pantazi, Juan Carlos Acosta, Rosario M. Sanchez-Martin, Sarah-Maria Fendt, Miguel Martin-Hernandez, Andrew J. Finch. https://www.cell.com/molecular-cell/fulltext/S1097-2765(22)00962-5

Mitochondrial dysfunction triggers actin polymerization necessary for rapid glycolytic activation – Rajarshi Chakrabarti, Tak Shun Fung, Taewook Kang, Pieti W. Elonkirjo, Anu Suomalainen, Edward J. Usherwood, Henry N. Higgs. https://rupress.org/jcb/article/221/11/e202201160/213462

Therapeutic resistance in acute myeloid leukemia cells is mediated by a novel ATM/mTOR pathway regulating oxidative phosphorylation – Hae J Park, Mark A Gregory, Vadym Zaberezhnyy, Andrew Goodspeed, Craig T Jordan, Jeffrey S Kieft, James DeGregori. https://elifesciences.org/articles/79940


Cancer Immunometabolism


GAB functions as a bioenergetic and signalling gatekeeper to control T cell inflammation – Siwen Kang, Lingling Liu, Tingting Wang, Matthew Cannon, Penghui Lin, Teresa W-M Fan, David A Scott, Hsin-Jung Joyce Wu, Andrew N Lane, Ruoning Wang.. https://www.nature.com/articles/s42255-022-00638-1

A novel gene signature unveils three distinct immune-metabolic rewiring patterns conserved across diverse tumor types and associated with outcomes – Leire Pedrosa, Carles Foguet, Helena Oliveres, Iván Archilla, Marta García de Herreros, Adela Rodríguez, Antonio Postigo, Daniel Benítez-Ribas, Jordi Camps, Miriam Cuatrecasas, Antoni Castells, Aleix Prat, Timothy M Thomson, Joan Maurel, Marta Cascante. https://www.frontiersin.org/articles/10.3389/fimmu.2022.926304/full

CD8+ T cell metabolic rewiring defined by scRNA-seq identifies a critical role of ASNS expression dynamics in T cell differentiation – Juan Fernández-García, Fabien Franco, Sweta Parik, Patricia Altea-Manzano, Antonino Alejandro Pane, Dorien Broekaert, Joke van Elsen, Ines Vermeire, Tessa Schalley, Mélanie Planque, Thomas van Brussel, Rogier Schepers, Elodie Modave, Tobias K. Karakach, Peter Carmeliet, Diether Lambrechts, Ping-Chih Ho, Sarah-Maria Fendt. https://www.cell.com/cell-reports/fulltext/S2211-1247(22)01510-8

OXPHOS promotes apoptotic resistance and cellular persistence in TH17 cells in the periphery and tumor microenvironment – Hanna S. Hong, Nneka E. Mbah, Mengrou Shan, Kristen Loesel, Lin Lin, Peter Sajjakulnukit, Luis O. Correa, Anthony Andren, Jason Lin, Atsushi Hayashi, Brian Magnuson, Judy Chen, Zhaoheng Li, Yuying Xie, Li Zhang, Daniel R. Goldstein, Shannon A. Carty, Yu Leo Lei, Anthony W. Opipari, Rafael J. Argüello, Ilona Kryczek, Nobuhiko Kamada, Weiping Zou, Luigi Franchi, Costas A. Lyssiotis. https://www.science.org/doi/full/10.1126/sciimmunol.abm8182


Miscellaneous


Anti-diabetic drug metformin suppresses tumorigenesis through inhibition of mevalonate pathway enzyme HMGCS1 – Yiyan Chen, Min Li, Yanying Yang, Yan Lu, Xiaoying Li. https://www.jbc.org/article/S0021-9258(22)01121-8

Computed tomography-measured body composition and survival in rectal cancer patients: a Swedish cohort study – Angeliki Kotti, Annica Holmqvist, Mischa Woisetschläger and Xiao-Feng Sun. https://cancerandmetabolism.biomedcentral.com/articles/10.1186/s40170-022-00297-6

An Exercise-Induced Metabolic Shield in Distant Organs Blocks Cancer Progression and Metastatic Dissemination – Danna Sheinboim, Shivang Parikh, Paulee Manich, Irit Markus, Sapir Dahan, Roma Parikh, Elisa Stubbs, Gali Cohen, Valentina Zemser-Werner, Rachel E. Bell, Sara Arciniegas Ruiz, Ruth Percik, Ronen Brenner, Stav Leibou, Hananya Vaknine, Gali Arad, Yariv Gerber, Lital Keinan-Boker, Tal Shimony, Lior Bikovski, Nir Goldstein, Keren Constantini, Sapir Labes, Shimonov Mordechai, Hila Doron, Ariel Lonescu, Tamar Ziv, Eran Nizri, Guy Choshen, Hagit Eldar-Finkelman, Yuval Tabach, Aharon Helman, Shamgar Ben-Eliyahu, Neta Erez, Eran Perlson, Tamar Geiger, Danny Ben-Zvi, Mehdi Khaled, Yftach Gepner, Carmit Levy. https://aacrjournals.org/cancerres/article/82/22/4164/710131

Pan-cancer analysis of tissue and single-cell HIF-pathway activation using a conserved gene signature – Olivia Lombardi, Ran Li, Silvia Halim, Hani Choudhry, Peter J. Ratcliffe, David R. Mole. https://www.cell.com/cell-reports/fulltext/S2211-1247(22)01523-6


Reviews


Metabolic determinants of tumour initiation – Julia S. Brunner and Lydia W. S. Finley. https://www.nature.com/articles/s41574-022-00773-5

Metabolite activation of tumorigenic signaling pathways in the tumor microenvironment – Vivien Low, Zhongchi Li, John Blenis. https://www.science.org/doi/abs/10.1126/scisignal.abj4220

Metabolic communication in the tumour–immune microenvironment – Kung-Chi Kao, Stefania Vilbois, Chin-Hsien Tsai and Ping-Chih Ho. https://www.nature.com/articles/s41556-022-01002-x

Targeting the DNA damage response and repair in cancer through nucleotide metabolism – Thomas Helleday,Sean G. Rudd. https://febs.onlinelibrary.wiley.com/doi/full/10.1002/1878-0261.13227

Challenges and Emerging Opportunities for Targeting mTOR in Cancer – Kris C. Wood; J. Silvio Gutkind. https://aacrjournals.org/cancerres/article-abstract/82/21/3884/709952

Emerging metabolomic tools to study cancer metastasis – Luiza Martins Nascentes Melo, Nicholas P. Lesner, Marie Sabatier, Jessalyn M. Ubellacker, Alpaslan Tasdogan. https://www.cell.com/trends/cancer/fulltext/S2405-8033(22)00156-X

Lactic acid and lactate: revisiting the physiological roles in the tumor microenvironment – Petya Apostolova, Erika L. Pearce. https://www.sciencedirect.com/science/article/abs/pii/S1471490622002137

Src: coordinating metabolism in cancer – Sara G. Pelaz and Arantxa Tabernero. https://www.nature.com/articles/s41388-022-02487-4

Glycan quality control in and out of the endoplasmic reticulum of mammalian cells – Yoichiro Harada, Yuki Ohkawa, Kento Maeda, Naoyuki Taniguchi. https://febs.onlinelibrary.wiley.com/doi/full/10.1111/febs.16185

Targeting adaptive cellular responses to mitochondrial bioenergetic deficiencies in human disease – Christopher F. Bennett, Conor T. Ronayne, Pere Puigserver. https://febs.onlinelibrary.wiley.com/doi/full/10.1111/febs.16195


Comments / other


Metabolic phosphatase moonlights for proteins – Scott A. Gerber & Arminja N. Kettenbach. https://www.nature.com/articles/s41556-022-00993-x

Acetyl-CoA, protein acetylation, and liver cancer – Zhengming Wu, Kun-Liang Guan. https://www.cell.com/molecular-cell/fulltext/S1097-2765(22)01015-2

Tumour-derived d-2HG blocks T cell cytotoxicity – Joseph Willson. https://www.nature.com/articles/s41568-022-00534-6

D-2HG comes out of its shell: Metabolic effects on the immune environment – Juan Manuel Schvartzman, Andrew M. Intlekofer. https://www.cell.com/molecular-cell/fulltext/S1097-2765(22)01067-X

Aerobic Glycolysis Promotes an NF-κB–Mediated Increase in PD-L1 Expression. https://aacrjournals.org/cancerdiscovery/article-abstract/12/11/OF5/709972

Cell scientist to watch – Lena Pernas. https://journals.biologists.com/jcs/article/135/21/jcs260704/278594

MetaboList – October 2022


Amino acids and one-carbon


Metabolic regulation by p53 prevents R-loop-associated genomic instability – Emanuele Panatta, Alessio Butera, Eleonora Mammarella, Consuelo Pitolli, Alessandro Mauriello, Marcel Leist, Richard A. Knight, Gerry Melino, Ivano Amelio. https://www.cell.com/cell-reports/fulltext/S2211-1247(22)01429-2

BCAT1 redox function maintains mitotic fidelity – Liliana Francois, Pavle Boskovic, Julian Knerr, Wei He, Gianluca Sigismondo, Carsten Schwan, Tushar H. More, Magdalena Schlotter, Jeroen Krijgsveld, Karsten Hiller, Robert Grosse, Peter Lichter, Bernhard Radlwimmer. https://www.cell.com/cell-reports/fulltext/S2211-1247(22)01379-1

Asparagine bioavailability regulates the translation of MYC oncogene – Sankalp Srivastava, Jie Jiang, Jagannath Misra, Gretchen Seim, Kirk A. Staschke, Minghua Zhong, Leonardo Zhou, Yu Liu, Chong Chen, Utpal Davé, Reuben Kapur, Sandeep Batra, Chi Zhang, Jiehao Zhou, Jing Fan, Ronald C. Wek and Ji Zhang. https://www.nature.com/articles/s41388-022-02474-9

Lysosomal enzyme trafficking factor LYSET enables nutritional usage of extracellular proteins – Catarina Pechincha, Sven Groessl, Robert Kalis, Melanie de Almeida, Andrea Zanotti, Marten Wittmann, Martin Schneider, Rafael P. de Campos, Sarah Rieser, Marlene Brandstetter, Alexander Schleiffer, Karin Müller-Decker, Dominic Helm, Sabrina Jabs, David Haselbach, Marius K. Lemberg, Johannes Zuber, Wilhelm Palm. https://www.science.org/doi/10.1126/science.abn5637

Glutamine synthetase limits b-catenin-mutated liver cancer growth by maintaining nitrogen homeostasis and suppressing mTORC1 – Weiwei Dai, Jianliang Shen, Junrong Yan, Alex J. Bott, Sara Maimouni, Heineken Q. Daguplo, Yujue Wang, Khoosheh Khayati, Jessie Yanxiang Guo, Lanjing Zhang, Yongbo Wang, Alexander Valvezan, Wen-Xing Ding, Xin Chen, Xiaoyang Su, Shenglan Gao, Wei-Xing Zong. https://www.jci.org/articles/view/161408


Starvation / mTOR / UPR-ISR


An unusual mode of baseline translation adjusts cellular protein synthesis capacity to metabolic needs – Cornelius Schneider, Florian Erhard, Beyenech Binotti, Alexander Buchberger, Jörg Vogel, Utz Fischer. https://www.cell.com/cell-reports/fulltext/S2211-1247(22)01317-1

A cell-based chemical-genetic screen for amino acid stress response inhibitors reveals torins reverse stress kinase GCN2 signaling – Johanna B. Brüggenthies, Alessandra Fiore, Marion Russier, Christina Bitsina, Julian Brötzmann, Susanne Kordes, Sascha Menninger, Alexander Wolf, Elena Conti, Jan E. Eickhoff, Peter J. Murray. https://www.jbc.org/article/S0021-9258(22)01072-9/fulltext

Limiting glutamine utilization activates a GCN2/TRAIL-R2/Caspase-8 apoptotic pathway in glutamine-addicted tumor cells – Rosario Yerbes, Rocío Mora-Molina, F. Javier Fernández-Farrán, Laura Hiraldo, Abelardo López-Rivas and Carmen Palacios. https://www.nature.com/articles/s41419-022-05346-y

Non-genomic activation of the AKT-mTOR pathway by the mitochondrial stress response in thyroid cancer – Woo Kyung Lee Doolittle, Sunmi Park, Seul Gi Lee, Seonhyang Jeong, Gibbeum Lee, Dongryeol Ryu, Kristina Schoonjans, Johan Auwerx, Jandee Lee and Young Suk Jo. https://www.nature.com/articles/s41388-022-02484-7

Termination of the unfolded protein response is guided by ER stress-induced HAC1 mRNA nuclear retention – Laura Matabishi-Bibi, Drice Challal, Mara Barucco, Domenico Libri and Anna Babour. https://www.nature.com/articles/s41467-022-34133-8

Inhibition of mTOR signaling protects human glioma cells from hypoxia-induced cell death in an autophagy-independent manner – Iris Divé, Kevin Klann, Jonas B. Michaelis, Dennis Heinzen, Joachim P. Steinbach, Christian Münch and Michael W. Ronellenfitsch. https://www.nature.com/articles/s41420-022-01195-y


Glucose metabolism


ATF4-dependent fructolysis fuels growth of glioblastoma multiforme – Chao Chen, Zhenxing Zhang, Caiyun Liu, Bin Wang, Ping Liu, Shu Fang, Fan Yang, Yongping You and Xinjian Li. https://www.nature.com/articles/s41467-022-33859-9

The hypoxia response pathway promotes PEP carboxykinase and gluconeogenesis in C. elegans – Mehul Vora, Stephanie M. Pyonteck, Tatiana Popovitchenko, Tarmie L. Matlack, Aparna Prashar, Nanci S. Kane, John Favate, Premal Shah and Christopher Rongo. https://www.nature.com/articles/s41467-022-33849-x

HK1 from hepatic stellate cell–derived extracellular vesicles promotes progression of hepatocellular carcinoma – Qi-tao Chen, Zhi-yuan Zhang, Qiao-ling Huang, Hang-zi Chen, Wen-bin Hong, Tianwei Lin, Wen-xiu Zhao, Xiao-min Wang, Cui-yu Ju, Liu-zheng Wu, Ya-ying Huang, Pei-pei Hou, Wei-jia Wang, Dawang Zhou, Xianming Deng and Qiao Wu. https://www.nature.com/articles/s42255-022-00642-5

Glucose-6-phosphate dehydrogenase exerts anti-stress effects independently of its enzymatic activity – Xiaohan Jin, Xuexue Li, Lifang Li, Benfu Zhong, Yang Hong, Jing Niu, Binghui Li. https://www.jbc.org/article/S0021-9258(22)01030-4/fulltext

Anti‐tumour effects of a macrolide analog F806 in oesophageal squamous cell carcinoma cells by targeting and promoting GLUT1 autolysosomal degradation – Xiang Li, Liyan Li, Xiaodong Wu, Bing Wen, Wan Lin, Yufei Cao, Lei Xie, Hefeng Zhang, Geng Dong, Enmin Li, Liyan Xu, Yinwei Cheng. https://febs.onlinelibrary.wiley.com/doi/abs/10.1111/febs.16545

Structural mechanism of SGLT1 inhibitors – Yange Niu, Wenhao Cui, Rui Liu, Sanshan Wang, Han Ke, Xiaoguang Lei and Lei Chen. https://www.nature.com/articles/s41467-022-33421-7


Glucose deprivation and sensing


The aldolase inhibitor aldometanib mimics glucose starvation to activate lysosomal AMPK – Chen-Song Zhang, Mengqi Li, Yu Wang, Xiaoyang Li, Yue Zong, Shating Long, Mingliang Zhang, Jin-Wei Feng, Xiaoyan Wei, Yan-Hui Liu, Baoding Zhang, Jianfeng Wu, Cixiong Zhang, Wenhua Lian, Teng Ma, Xiao Tian, Qi Qu, Yaxin Yu, Jinye Xiong, Dong-Tai Liu, Zhenhua Wu, Mingxia Zhu, Changchuan Xie, Yaying Wu, Zheni Xu, Chunyan Yang, Junjie Chen, Guohong Huang, Qingxia He, Xi Huang, Lei Zhang, Xiufeng Sun, Qingfeng Liu, Abdul Ghafoor, Fu Gui, Kaili Zheng, Wen Wang, Zhi-Chao Wang, Yong Yu, Qingliang Zhao, Shu-Yong Lin, Zhi-Xin Wang, Hai-Long Piao, Xianming Deng and Sheng-Cai Lin. https://www.nature.com/articles/s42255-022-00640-7

Rvb1/Rvb2 proteins couple transcription and translation during glucose starvation – Yang S Chen, Wanfu Hou, Sharon Tracy, Alex T Harvey, Vince Harjono, Fan Xu, James J Moresco, John R Yates III, Brian M Zid. https://elifesciences.org/articles/76965

TSC22D4 interacts with Akt1 to regulate glucose metabolism – Sevgican Demir, Gretchen Wolff, Annika Wieder, Adriano Maida, Lea Bühler, Maik Brune, Oksana Hautzinger, Annette Feuchtinger, Tanja Poth, Julia Szendroedi, Stephan Herzig, Bilgen Ekim Üstünel. https://www.science.org/doi/full/10.1126/sciadv.abo5555


Lipid metabolism


Lipid droplet turnover at the lysosome inhibits growth of hepatocellular carcinoma in a BNIP3-dependent manner – Damian E. Berardi, Althea Bock-Hughes, Alexander R. Terry, Lauren E. Drake, Grazyna Bozek, Kay F. Macleod. https://www.science.org/doi/full/10.1126/sciadv.abo2510

ACSL3 regulates lipid droplet biogenesis and ferroptosis sensitivity in clear cell renal cell carcinoma – Timothy D. Klasson, Edward L. LaGory, Hongjuan Zhao, Star K. Huynh, Ioanna Papandreou, Eui Jung Moon and Amato J. Giaccia. https://cancerandmetabolism.biomedcentral.com/articles/10.1186/s40170-022-00290-z

Inhibiting glutamine utilization creates a synthetic lethality for suppression of ATP citrate lyase in KRas-driven cancer cells – Ahmet Hatipoglu, Deepak Menon, Talia Levy, Maria A Frias, David A Foster. https://journals.plos.org/plosone/article?id=10.1371/journal.pone.0276579

Ovarian cancer cell fate regulation by the dynamics between saturated and unsaturated fatty acids – Guangyuan Zhao, Yuying Tan, Horacio Cardenas, David Vayngart, Yinu Wang, Hao Huang, Russell Keathley, Jian-Jun Wei, Christina R. Ferreira, Sandra Orsulic, Ji-Xin Cheng and Daniela Matei. https://www.pnas.org/doi/10.1073/pnas.2203480119


Cancer Immunometabolism


A Cancer Cell–Intrinsic GOT2–PPARδ Axis Suppresses Antitumor Immunity – Jaime Abrego, Hannah Sanford-Crane, Chet Oon, Xu Xiao, Courtney B. Bett, Duanchen Sun, Shanthi Nagarajan, Luis Diaz, Holly Sandborg, Sohinee Bhattacharyya, Zheng Xia, Lisa M. Coussens, Peter Tontonoz, Mara H. Sherman. https://aacrjournals.org/cancerdiscovery/article/12/10/2414/709426

Spermidine activates mitochondrial trifunctional protein and improves antitumor immunity in mice – Muna Al-Habsi, Kenji Chamoto, Ken Matsumoto, Norimichi Nomura, Baihao Zhang, Yuki Sugiura, Kazuhiro Sonomura, Aprilia Maharani, Yuka Nakajima, Yibo Wu, Yayoi Nomura, Rosemary Menzies, Masaki Tajima, Koji Kitaoka, Yasuharu Haku, Sara Delghandi, Keiko Yurimoto, Fumihiko Matsuda, So Iwata, Toshihiko Ogura, Sidonia Fagarasan, Tasuku Honjo. https://www.science.org/doi/abs/10.1126/science.abj3510

NKT cells adopt a glutamine-addicted phenotype to regulate their homeostasis and function – Ajay Kumar, Emily L. Yarosz, Anthony Andren, Li Zhang, Costas A. Lyssiotis, Cheong-Hee Chang. https://www.cell.com/cell-reports/fulltext/S2211-1247(22)01366-3

Altered acetyl-CoA metabolism presents a new potential immunotherapy target in the obese lung microenvironment – Spencer R. Rosario, Randall J. Smith Jr, Santosh K. Patnaik, Song Liu, Joseph Barbi and Sai Yendamuri. https://cancerandmetabolism.biomedcentral.com/articles/10.1186/s40170-022-00292-x

Increased glucose metabolism in TAMs fuels O-GlcNAcylation of lysosomal Cathepsin B to promote cancer metastasis and chemoresistance – Qingzhu Shi, Qicong Shen, Yanfang Liu, Yang Shi, Wenwen Huang, Xi Wang, Zhiqing Li, Yangyang Chai, Hao Wang, Xiangjia Hu, Nan Li, Qian Zhang, Xuetao Cao. https://www.cell.com/cancer-cell/fulltext/S1535-6108(22)00376-2

Metabolic control of CD47 expression through LAT2-mediated amino acid uptake promotes tumor immune evasion – Zenan Wang, Binghao Li, Shan Li, Wenlong Lin, Zhan Wang, Shengdong Wang, Weida Chen, Wei Shi, Tao Chen, Hao Zhou, Eloy Yinwang, Wenkan Zhang, Haochen Mou, Xupeng Chai, Jiahao Zhang, Zhimin Lu and Zhaoming Ye. https://www.nature.com/articles/s41467-022-34064-4


Mitochondria / nucleotides


Two independent respiratory chains adapt OXPHOS performance to glycolytic switch – Erika Fernández-Vizarra, Sandra López-Calcerrada, Ana Sierra-Magro, Rafael Pérez-Pérez, Luke E. Formosa, Daniella H. Hock, María Illescas, Ana Peñas, Michele Brischigliaro,Shujing Ding, Ian M. Fearnley, Charalampos Tzoulis, Robert D.S. Pitceathly, Joaquín Arenas, Miguel A. Martín, David A. Stroud, Massimo Zeviani, Michael T. Ryan, Cristina Ugalde. https://www.cell.com/cell-metabolism/fulltext/S1550-4131(22)00395-3

HIRA loss transforms FH-deficient cells – Lorea Valcarcel-Jimenez, Connor Rogerson, Cissy Yong, Christina Schmidt, Ming Yang, Monica Cremades-Rodelgo, Victoria Harle, Victoria Offord, Kim Wong, Ariane Mora, Alyson Speed, Veronica Caraffini, Maxine Gia Binh Tran, Eamonn R. Maher, Grant D. Stewart, Sakari Vanharanta, David J. Adams, Christian Frezza. https://www.science.org/doi/full/10.1126/sciadv.abq8297

Therapeutic resistance in acute myeloid leukemia cells is mediated by a novel ATM/mTOR pathway regulating oxidative phosphorylation – Hae J Park, Mark A Gregory, Vadym Zaberezhnyy, Andrew Goodspeed, Craig T Jordan, Jeffrey S Kieft, James DeGregori. https://elifesciences.org/articles/79940

Mitochondrial dysfunction and impaired growth of glioblastoma cell lines caused by antimicrobial agents inducing ferroptosis under glucose starvation – Kenji Miki, Mikako Yagi, Koji Yoshimoto, Dongchon Kang and Takeshi Uchiumi. https://www.nature.com/articles/s41389-022-00437-z

Choroid plexus-CSF-targeted antioxidant therapy protects the brain from toxicity of cancer chemotherapy [methotrexate] – Ahram Jang, Boryana Petrova, Taek-Chin Cheong, Miriam E. Zawadzki, Jill K. Jones, Andrew J. Culhane, Frederick B. Shipley, Roberto Chiarle, Eric T. Wong, Naama Kanarek, Maria K. Lehtinen. https://www.cell.com/neuron/fulltext/S0896-6273(22)00741-3


Miscellaneous


Scaffold-mediated switching of lymphoma metabolism in culture – Rachana Bhatt, Dashnamoorthy Ravi, Andrew M. Evens and Biju Parekkadan. https://cancerandmetabolism.biomedcentral.com/articles/10.1186/s40170-022-00291-y

Regulatory chromatin rewiring promotes metabolic switching during adaptation to oncogenic receptor tyrosine kinase inhibition – Samuel Ogden, Kashmala Carys, Ibrahim Ahmed, Jason Bruce and Andrew D. Sharrocks. https://www.nature.com/articles/s41388-022-02465-w

Data-driven identification of plasma metabolite clusters and metabolites of interest for potential detection of early-stage non-small cell lung cancer cases versus cancer-free controls – Julian O. Kim, Robert Balshaw, Connel Trevena, Shantanu Banerji, Leigh Murphy, David Dawe, Lawrence Tan, Sadeesh Srinathan, Gordon Buduhan, Biniam Kidane, Gefei Qing, Michael Domaratzki and Michel Aliani. https://cancerandmetabolism.biomedcentral.com/articles/10.1186/s40170-022-00294-9


Reviews


The role of lipids in cancer progression and metastasis – Miguel Martin-Perez, Uxue Urdiroz-Urricelqui, Claudia Bigas, Salvador Aznar Benitah. https://www.cell.com/cell-metabolism/fulltext/S1550-4131(22)00447-8

Amino acid metabolism in primary bone sarcomas – Jennifer A. Jiménez, Elizabeth R. Lawlor and Costas A. Lyssiotis. https://www.frontiersin.org/articles/10.3389/fonc.2022.1001318/full

Rerouting the drug response: Overcoming metabolic adaptation in KRAS-mutant cancers – Deborah Y. Moss, Christopher McCann, Emma M. Kerr. https://www.science.org/doi/abs/10.1126/scisignal.abj3490

GOT2 consider the tumor microenvironment – Brian T. Do, Matthew G. Vander Heiden. https://www.cell.com/trends/cancer/fulltext/S2405-8033(22)00195-9

Targeting PDAC metabolism: Environment determines what has GOT2 give – Oliver J. Newsom, Lucas B. Sullivan

https://www.cell.com/cell-metabolism/fulltext/S1550-4131(22)00451-X

Nutrient availability as an arbiter of cell size – Douglas R. Kellogg, Petra Anne Levin. https://www.cell.com/trends/cell-biology/fulltext/S0962-8924(22)00147-7

Non-canonical mTORC1 signaling at the lysosome – Gennaro Napolitano, Chiara Di Malta, Andrea Ballabio. https://www.cell.com/trends/cell-biology/fulltext/S0962-8924(22)00117-9

Principles and functions of metabolic compartmentalization – Liron Bar-Peled and Nora Kory. https://www.nature.com/articles/s42255-022-00645-2

Metabolic guidance and stress in tumors modulate antigen-presenting cells – Jaeoh Park, Limei Wang and Ping-Chih Ho. https://www.nature.com/articles/s41389-022-00438-y

Science-Driven Nutritional Interventions for the Prevention and Treatment of Cancer – Léa Montégut, Rafael de Cabo, Laurence Zitvogel, Guido Kroemer. https://aacrjournals.org/cancerdiscovery/article-abstract/12/10/2258/709430


Comments


Nuclear TCA cycle reactions – Melina Casadio. https://www.nature.com/articles/s41556-022-01006-7

Starving cancer into submission by activating BAT – Mohammed K. Hankir, Annett Hoffmann, Florian Seyfried. https://www.cell.com/cell-metabolism/fulltext/S1550-4131(22)00399-0

Gliomas lean on pyrimidines – Joseph Willson. https://www.nature.com/articles/s41568-022-00515-9

Liver cancer metabolism: a hexokinase from the stars – Aveline Filliol and Robert F. Schwabe. https://www.nature.com/articles/s42255-022-00659-w

Glutamine, MTOR and autophagy: a multiconnection relationship – Clément Bodineau, Mercedes Tomé, Piedad del Socorro Murdoch and Raúl V. Durán. https://www.tandfonline.com/doi/full/10.1080/15548627.2022.2062875

Cold exposure as anti-cancer therapy – Mariia Yuneva. https://www.cell.com/cancer-cell/fulltext/S1535-6108(22)00437-8

Transient Systemic Autophagy Ablation Irreversibly Inhibits Lung Tumor Cell Metabolism and Promotes T-Cell Mediated Tumor Killing – Kyle Nunn and Jessie Yanxiang Guo. https://www.tandfonline.com/doi/abs/10.1080/15548627.2022.2141534

MetaboList – September 2022


Lipid and acetate metabolism


Fatty acids homeostasis during fasting predicts protection from chemotherapy toxicity – Marta Barradas, Adrián Plaza, Gonzalo Colmenarejo, Iolanda Lázaro, Luis Filipe Costa-Machado, Roberto Martín-Hernández, Victor Micó, José Luis López-Aceituno, Jesús Herranz, Cristina Pantoja, Hector Tejero, Alberto Diaz-Ruiz, Fatima Al-Shahrour, Lidia Daimiel, Viviana Loria-Kohen, Ana Ramirez de Molina, Alejo Efeyan, Manuel Serrano, Oscar J. Pozo, Aleix Sala-Vila and Pablo J. Fernandez-Marcos. https://www.nature.com/articles/s41467-022-33352-3

Crosstalk between AML and stromal cells triggers acetate secretion through the metabolic rewiring of stromal cells – Nuria Vilaplana-Lopera, Vincent Cuminetti, Ruba Almaghrabi, Grigorios Papatzikas, Ashok Kumar Rout, Mark Jeeves, Elena González, Yara Alyahyawi, Alan Cunningham, Ayşegül Erdem, Frank Schnütgen, Manoj Raghavan, Sandeep Potluri, Jean-Baptiste Cazier, Jan Jacob Schuringa, Michelle AC Reed, Lorena Arranz, Ulrich L Günther, Paloma Garcia. https://elifesciences.org/articles/75908

KIF11 manipulates SREBP2‐dependent mevalonate cross talk to promote tumor progression in pancreatic ductal adenocarcinoma – Xiang Gu, Qunshan Zhu, Guangyu Tian, Wenbo Song, Tao Wang, Ali Wang, Xiaojun Chen, Songbing Qin. https://onlinelibrary.wiley.com/doi/full/10.1002/cam4.4683

Activation of Drp1 promotes fatty acids-induced metabolic reprograming to potentiate Wnt signaling in colon cancer – Xiaopeng Xiong, Sumati Hasani, Lyndsay E. A. Young, Dylan R. Rivas, Ashley T. Skaggs, Rebecca Martinez, Chi Wang, Heidi L. Weiss, Matthew S. Gentry, Ramon C. Sun and Tianyan Gao. https://www.nature.com/articles/s41418-022-00974-5


Nucleotides


A druggable addiction to de novo pyrimidine biosynthesis in diffuse midline glioma – Sharmistha Pal, Jakub P. Kaplan, Huy Nguyen, Sylwia A. Stopka, Milan R. Savani, Michael S. Regan, Quang-De Nguyen, Kristen L. Jones, Lisa A. Moreau, Jingyu Peng, Marina G. Dipiazza, Andrew J. Perciaccante, Xiaoting Zhu, Bradley R. Hunsel, Kevin X. Liu, Sanda Alexandrescu, Rachid Drissi, Mariella G. Filbin, Samuel K. McBrayer, Nathalie Y.R. Agar, Dipanjan Chowdhury, Daphne A. Haas-Kogan. https://www.cell.com/cancer-cell/fulltext/S1535-6108(22)00325-7

Metabolic collateral lethal target identification reveals MTHFD2 paralogue dependency in ovarian cancer – Abhinav Achreja, Tao Yu, Anjali Mittal, Srinadh Choppara, Olamide Animasahun, Minal Nenwani, Fulei Wuchu, Noah Meurs, Aradhana Mohan, Jin Heon Jeon, Itisam Sarangi, Anusha Jayaraman, Sarah Owen, Reva Kulkarni, Michele Cusato, Frank Weinberg, Hye Kyong Kweon, Chitra Subramanian, Max S. Wicha, Sofia D. Merajver, Sunitha Nagrath, Kathleen R. Cho, Analisa DiFeo, Xiongbin Lu and Deepak Nagrath. https://www.nature.com/articles/s42255-022-00636-


IDH


Oncometabolite d-2HG alters T cell metabolism to impair CD8+ T cell function – Giulia Notarangelo, Jessica B. Spinelli, Elizabeth M. Perez, Gregory J. Baker, Kiran Kurmi, Ilaria Elia, Sylwia A. Stopka, Gerard Baquer, Jia-Ren Lin, Alexandra J. Golby, Shakchhi Joshi, Heide F. Baron, Jefte M. Drijvers, Peter Georgiev, Alison E. Ringel, Elma Zaganjor, Samuel K. McBrayer, Peter K. Sorger, Arlene H. Sharpe, Kai W. Wucherpfennig, Sandro Santagata, Nathalie Y. R. Agar, Mario L. Suvà, Marcia C. Haigis. https://www.science.org/doi/abs/10.1126/science.abj5104

De novo pyrimidine synthesis is a targetable vulnerability in IDH mutant glioma – Diana D. Shi, Milan R. Savani, Michael M. Levitt, Adam C. Wang, Jennifer E. Endress, Cylaina E. Bird, Joseph Buehler, Sylwia A. Stopka, Michael S. Regan, Yu-Fen Lin, Vinesh T. Puliyappadamba, Wenhua Gao, Januka Khanal, Laura Evans, Joyce H. Lee, Lei Guo, Yi Xiao, Min Xu, Bofu Huang, Rebecca B. Jennings, Dennis M. Bonal, Misty S. Martin-Sandoval, Tammie Dang, Lauren C. Gattie, Amy B. Cameron, Sungwoo Lee, John M. Asara, Harley I. Kornblum, Tak W. Mak, Ryan E. Looper, Quang-De Nguyen, Sabina Signoretti, Stefan Gradl, Andreas Sutter, Michael Jeffers, Andreas Janzer, Mark A. Lehrman, Lauren G. Zacharias, Thomas P. Mathews, Julie-Aurore Losman, Timothy E. Richardson, Daniel P. Cahill, Ralph J. DeBerardinis, Keith L. Ligon, Lin Xu, Peter Ly, Nathalie Y.R. Agar, Kalil G. Abdullah, Isaac S. Harris, William G. Kaelin Jr., Samuel K. McBrayer. https://www.cell.com/cancer-cell/fulltext/S1535-6108(22)00324-5


Cancer Immunometabolism


Carbon source availability drives nutrient utilization in CD8+ T cells – Irem Kaymak, Katarzyna M. Luda, Lauren R. Duimstra, Eric H. Ma, Joseph Longo, Michael S. Dahabieh, Brandon Faubert, Brandon M. Oswald, McLane J. Watson, Susan M. Kitchen-Goosen, Lisa M. DeCamp, Shelby E. Compton, Zhen Fu, Ralph J. DeBerardinis, Kelsey S. Williams, Ryan D. Sheldon, Russell G. Jones. https://www.cell.com/cell-metabolism/fulltext/S1550-4131(22)00311-4

Lactate increases stemness of CD8 + T cells to augment anti-tumor immunity – Qiang Feng, Zhida Liu, Xuexin Yu, Tongyi Huang, Jiahui Chen, Jian Wang, Jonathan Wilhelm, Suxin Li, Jiwon Song, Wei Li, Zhichen Sun, Baran D. Sumer, Bo Li, Yang-Xin Fu and Jinming Gao. https://www.nature.com/articles/s41467-022-32521-8

CD36-mediated metabolic crosstalk between tumor cells and macrophages affects liver metastasis – Ping Yang, Hong Qin, Yiyu Li, Anhua Xiao, Enze Zheng, Han Zeng, Chunxiao Su, Xiaoqing Luo, Qiannan Lu, Meng Liao, Lei Zhao, Li Wei, Zac Varghese, John F. Moorhead, Yaxi Chen and Xiong Z. Ruan. https://www.nature.com/articles/s41467-022-33349-y

USP14 promotes tryptophan metabolism and immune suppression by stabilizing IDO1 in colorectal cancer – Dongni Shi, Xianqiu Wu, Yunting Jian, Junye Wang, Chengmei Huang, Shuang Mo, Yue Li, Fengtian Li, Chao Zhang, Dongsheng Zhang, Huizhong Zhang, Huilin Huang, Xin Chen, Y. Alan Wang, Chuyong Lin, Guozhen Liu, Libing Song and Wenting Liao. https://www.nature.com/articles/s41467-022-33285-x

Aerobic glycolysis promotes tumor immune evasion by hexokinase2-mediated phosphorylation of IκBα – Dong Guo, Yingying Tong, Xiaoming Jiang, Ying Meng, Hongfei Jiang, Linyong Du, Qingang Wu, Shan Li, Shudi Luo, Min Li, Liwei Xiao, Haiyan He, Xuxiao He, Qiujing Yu, Jing Fang, Zhimin Lu. https://www.cell.com/cell-metabolism/fulltext/S1550-4131(22)00345-X


Mitochondria


Mitochondrial pyruvate supports lymphoma proliferation by fueling a glutamate pyruvate transaminase 2-dependent glutaminolysis pathway – Peng Wei, Alex J. Bott, Ahmad A. Cluntun, Jeffrey T. Morgan, Corey N. Cunningham, John C. Schell, Yeyun Ouyang, Scott B. Ficarro, Jarrod A. Marto, Nika N. Danial, Ralph J. DeBerardinis, Jared Rutter. https://www.science.org/doi/full/10.1126/sciadv.abq0117

Targeting OXPHOS de novo purine synthesis as the nexus of FLT3 inhibitor-mediated synergistic antileukemic actions – Pu Zhang, Lindsey T. Brinton, Mehdi Gharghabi, Steven Sher, Katie Williams, Matthew Cannon, Janek S. Walker, Daniel Canfield, Larry Beaver, Casey B. Cempre, Hannah Phillips, Xuyong Chen, Pearlly Yan, Amy Lehman, Peggy Scherle, Min Wang, Kris Vaddi, Robert Baiocchi, Ruoning Wang, Deepa Sampath, Lapo Alinari, James S. Blachly, Rosa Lapalombella. https://www.science.org/doi/full/10.1126/sciadv.abp9005?et_rid=33946976

Intrinsic adaptations in OXPHOS power output and reduced tumorigenicity characterize doxorubicin resistant ovarian cancer cells – James T. Hagen, McLane M. Montgomery, Ericka M. Biagioni, Polina Krassovskaia, Filip Jevtovic, Daniel Shookster, Uma Sharma, Kang Tung, Nickolas T. Broskey, Linda May, Hu Huang, Jeffrey J. Brault, P. Darrell Neufer, Myles C. Cabot, Kelsey H. Fisher-Wellman. https://www.sciencedirect.com/science/article/abs/pii/S0005272822003851


Starvation


Sensitisation of cancer cells to radiotherapy by serine and glycine starvation – Mattia Falcone, Alejandro Huerta Uribe, Vasileios Papalazarou, Alice C. Newman, Dimitris Athineos, Katrina Stevenson, Charles-Etienne Gabriel Sauvé, Yajing Gao, Jin K. Kim, Michael Del Latto, Maria Kierstead, Chao Wu, J. Joshua Smith, Paul B. Romesser, Anthony J. Chalmers, Karen Blyth and Oliver D. K. Maddocks. https://www.nature.com/articles/s41416-022-01965-6

Mitochondrial and metabolic remodelling in human skin fibroblasts in response to glucose availability – Sónia A. Pinho, Cláudio F. Costa, Cláudia M. Deus, Sonia L. C. Pinho, Inês Miranda-Santos, Gonçalo Afonso, Olivia Bagshaw, Jeffrey A. Stuart, Paulo J. Oliveira, Teresa Cunha-Oliveira. https://febs.onlinelibrary.wiley.com/doi/abs/10.1111/febs.16413

cAMP-Protein Kinase A and Stress-Activated MAP Kinase signaling mediate transcriptional control of autophagy in fission yeast during glucose limitation or starvation – Armando Jesús Pérez-Díaz, Beatriz Vázquez-Marín, Jero Vicente-Soler, Francisco Prieto-Ruiz, Teresa Soto, Alejandro Franco, José Cansado and Marisa Madrid. https://www.tandfonline.com/doi/abs/10.1080/15548627.2022.2125204

AMPK promotes Arf6 activation in a kinase-independent manner upon glucose starvation [In collection: Metabolism] – Kuan-Jung Chen, Jia-Wei Hsu, Fang-Jen S. Lee. https://journals.biologists.com/jcs/article/135/18/jcs259609/276628


mTOR


A Rag GTPase dimer code defines the regulation of mTORC1 by amino acids – Peter Gollwitzer, Nina Grützmacher, Sabine Wilhelm, Daniel Kümmel and Constantinos Demetriades. https://www.nature.com/articles/s41556-022-00976-y

Brain-enriched RagB isoforms regulate the dynamics of mTORC1 activity through GATOR1 inhibition – Gianluca Figlia, Sandra Müller, Anna M. Hagenston, Susanne Kleber, Mykola Roiuk, Jan-Philipp Quast, Nora ten Bosch, Damian Carvajal Ibañez, Daniela Mauceri, Ana Martin-Villalba and Aurelio A. Teleman. https://www.nature.com/articles/s41556-022-00977-x

Brain-restricted mTOR inhibition with binary pharmacology – Ziyang Zhang, Qiwen Fan, Xujun Luo, Kevin Lou, William A. Weiss and Kevan M. Shokat. https://www.nature.com/articles/s41586-022-05213-y

Invading Bacterial Pathogens Activate Transcription Factor EB in Epithelial Cells through the Amino Acid Starvation Pathway of mTORC1 Inhibition – Liliane Cabral-Fernandes, Shawn Goyal, Armin Farahvash, Jessica Tsalikis, Dana J. Philpott, Stephen E. Girardin. https://journals.asm.org/doi/abs/10.1128/mcb.00241-22

Tissue-restricted inhibition of mTOR using chemical genetics – Douglas R. Wassarman, Kondalarao Bankapalli, Leo J. Pallanck, and Kevan M. Shokat. https://www.pnas.org/doi/10.1073/pnas.2204083119

Glucose-driven TOR–FIE–PRC2 signalling controls plant development – Ruiqiang Ye, Meiyue Wang, Hao Du, Shweta Chhajed, Jin Koh, Kun-hsiang Liu, Jinwoo Shin, Yue Wu, Lin Shi, Lin Xu, Sixue Chen, Yijing Zhang and Jen Sheen. https://www.nature.com/articles/s41586-022-05171-5

Oncogenic RAS commandeers amino acid sensing machinery to aberrantly activate mTORC1 in multiple myeloma – Yandan Yang, Arnold Bolomsky, Thomas Oellerich, Ping Chen, Michele Ceribelli, Björn Häupl, George W. Wright, James D. Phelan, Da Wei Huang, James W. Lord, Callie K. Van Winkle, Xin Yu, Jan Wisniewski, James Q. Wang, Frances A. Tosto, Erin Beck, Kelli Wilson, Crystal McKnight, Jameson Travers, Carleen Klumpp-Thomas, Grace A. Smith, Stefania Pittaluga, Irina Maric, Dickran Kazandjian, Craig J. Thomas and Ryan M. Young. https://www.nature.com/articles/s41467-022-33142-x


Hypoxia


FOXA1 inhibits hypoxia programs through transcriptional repression of HIF1A – Xiaohai Wang, Lourdes Brea, Xiaodong Lu, Galina Gritsina, Su H. Park, Wanqing Xie, Jonathan C. Zhao and Jindan Yu. https://www.nature.com/articles/s41388-022-02423-6

Intermittent hypoxia enhances the expression of hypoxia inducible factor HIF1A through histone demethylation – Chloe-Anne Martinez, Yannasittha Jiramongkol, Neha Bal, Imala Alwis, Polina E. Nedoboy, Melissa MJ. Farnham, Mark D. White, Peter A. Cistulli, Kristina M. Cook. https://www.jbc.org/article/S0021-9258(22)00979-6


Reviews


Stressed to death: Mitochondrial stress responses connect respiration and apoptosis in cancer – Jacob M. Winter, Tarun Yadav, Jared Rutter. https://www.cell.com/molecular-cell/fulltext/S1097-2765(22)00708-0

Glutaminase Inhibitors—Do They Have a Role in the Treatment of Metastatic Clear-Cell Renal Cell Carcinoma? – Janet E. Brown. https://jamanetwork.com/journals/jamaoncology/article-abstract/2795983

Metabolic reprogramming: a bridge between aging and tumorigenesis – Stanislav Drapela, Didem Ilter, Ana P. Gomes. https://febs.onlinelibrary.wiley.com/doi/full/10.1002/1878-0261.13261

Genetic approaches to understand cellular responses to oxygen availability – Brian M. Ortmann, James A. Nathan. https://febs.onlinelibrary.wiley.com/doi/full/10.1111/febs.16072

Regulation of redox signaling in HIF‐1‐dependent tumor angiogenesis – Valeria Manuelli, Chiara Pecorari, Giuseppe Filomeni, Ester Zito. https://febs.onlinelibrary.wiley.com/doi/full/10.1111/febs.16110

Metabolic Reprogramming in Hematologic Malignancies: Advances and Clinical Perspectives – Zhuoya Yu, Xiangxiang Zhou, Xin Wang. https://aacrjournals.org/cancerres/article/82/17/2955/708282

The Central Role of mTORC1 in Amino Acid Sensing – Shusheng Yue, Guanya Li, Shanping He, Tingting Li. https://aacrjournals.org/cancerres/article-abstract/82/17/2964/708284

AMPK: An odyssey of a metabolic regulator, a tumor suppressor, and now a contextual oncogene – Vasudevarao Penugurti, Yasaswi Gayatri Mishra, Bramanandam Manavathi. https://www.sciencedirect.com/science/article/abs/pii/S0304419X2200110X


Comments


Pumping Iron: Ferritinophagy Promotes Survival and Therapy Resistance in Pancreatic Cancer – Vaibhav Jain, Ravi K. Amaravadi. https://aacrjournals.org/cancerdiscovery/article-abstract/12/9/2023/708772

Plasma L-Arginine Predicts Clinical Response to Immune Checkpoint Inhibitors – https://aacrjournals.org/cancerdiscovery/article-abstract/12/9/OF6/708788

Metabolite-driven antitumor immunity [D-2HG] – https://www.science.org/doi/10.1126/science.ade3697


Glucose metabolism


Glycolytic oscillations in HeLa cervical cancer cell spheroids – Takashi Amemiya, Kenichi Shibata, Junpei Takahashi, Masatoshi Watanabe, Satoshi Nakata, Kazuyuki Nakamura, Tomohiko Yamaguchi. https://febs.onlinelibrary.wiley.com/doi/abs/10.1111/febs.16454

ALK fusion promotes metabolic reprogramming of cancer cells by transcriptionally upregulating PFKFB3 – Mengnan Hu, Ruoxuan Bao, Miao Lin, Xiao-Ran Han, Ying-Jie Ai, Yun Gao, Kun-Liang Guan, Yue Xiong and Hai-Xin Yuan. https://www.nature.com/articles/s41388-022-02453-0

PFKFB4 interacts with FBXO28 to promote HIF-1α signaling in glioblastoma – Emma Phillips, Jörg Balss, Frederic Bethke, Stefan Pusch, Stefan Christen, Thomas Hielscher, Martina Schnölzer, Michael N. C. Fletcher, Antje Habel, Claudia Tessmer, Lisa-Marie Brenner, Mona Göttmann, David Capper, Christel Herold-Mende, Andreas von Deimling, Sarah-Maria Fendt and Violaine Goidts. https://www.nature.com/articles/s41389-022-00433-3

Aurora A-mediated pyruvate kinase M2 phosphorylation promotes biosynthesis with glycolytic metabolites and tumor cell cycle progression – Ya Jiang, Ting Wang, Dandan Sheng, Chaoqiang Han, Tian Xu, Peng Zhang, Weiyi You, Weiwei Fan, Zhiyong Zhang, Tengchuan Jin, Xiaotao Duan, Xiao Yuan, Xing Liu, Kaiguang Zhang, Ke Ruan, Jue Shi, Jing Guo, Aoxing Cheng, Zhenye Yang. https://www.jbc.org/article/S0021-9258(22)01005-5/fulltext


Amino acid metabolism


GLS2 Is a Tumor Suppressor and a Regulator of Ferroptosis in Hepatocellular Carcinoma – Sawako Suzuki, Divya Venkatesh, Hiroaki Kanda, Akitoshi Nakayama, Hiroyuki Hosokawa, Eunyoung Lee, Takashi Miki, Brent R. Stockwell, Koutaro Yokote, Tomoaki Tanaka, Carol Prives. https://aacrjournals.org/cancerres/article-abstract/82/18/3209/709027

GOT2 Silencing Promotes Reprogramming of Glutamine Metabolism and Sensitizes Hepatocellular Carcinoma to Glutaminase Inhibitors – Yunzheng Li, Binghua Li, Yanchao Xu, Liyuan Qian, Tiancheng Xu, Gang Meng, Huan Li, Ye Wang, Laizhu Zhang, Xiang Jiang, Qi Liu, Yuanyuan Xie, Chunxiao Cheng, Beicheng Sun, Decai Yu. https://aacrjournals.org/cancerres/article-abstract/82/18/3223/709031

NRF2 mediates melanoma addiction to GCDH by modulating apoptotic signalling – Sachin Verma, David Crawford, Ali Khateb, Yongmei Feng, Eduard Sergienko, Gaurav Pathria, Chen-Ting Ma, Steven H. Olson, David Scott, Rabi Murad, Eytan Ruppin, Michael Jackson and Ze’ev A. Ronai. https://www.nature.com/articles/s41556-022-00985-x

GCN2 eIF2 kinase promotes prostate cancer by maintaining amino acid homeostasis – Ricardo A Cordova, Jagannath Misra, Parth H Amin, Anglea J Klunk, Nur P Damayanti, Kenneth R Carlson, Andrew J Elmendorf, Hyeong-Geug Kim, Emily T Mirek, Bennet D Elzey, Marcus J Miller, X Charlie Dong, Liang Cheng, Tracy G Anthony, Robero Pili Is a corresponding author, Ronald C Wek Is a corresponding author, Kirk A Staschke. https://elifesciences.org/articles/81083

Efficacy and Safety of Telaglenastat Plus Cabozantinib vs Placebo Plus Cabozantinib in Patients With Advanced Renal Cell Carcinoma. The CANTATA Randomized Clinical Trial – Nizar M. Tannir, Neeraj Agarwal, Camillo Porta, Nicola J. Lawrence, Robert Motzer, Bradley McGregor, Richard J. Lee, Rohit K. Jain, Nancy Davis, Leonard J. Appleman, Oscar Goodman Jr, Walter M. Stadler, Sunil Gandhi, Daniel M. Geynisman, Roberto Iacovelli, Begoña Mellado, Juan Manuel Sepúlveda Sánchez, Robert Figlin, Thomas Powles, Lalith Akella, Keith Orford, Bernard Escudier. https://jamanetwork.com/journals/jamaoncology/article-abstract/2795979

MetaboList – August 2022


Glucose/Warburg metabolism


Saturation of the mitochondrial NADH shuttles drives aerobic glycolysis in proliferating cells – Yahui Wang, Ethan Stancliffe, Ronald Fowle-Grider, Rencheng Wang, Cheng Wang, Michaela Schwaiger-Haber, Leah P. Shriver, Gary J. Patti. https://www.cell.com/molecular-cell/fulltext/S1097-2765(22)00703-1 Methionine oxidation activates pyruvate kinase M2 to promote pancreatic cancer metastasis – Dan He, Huijin Feng, Belen Sundberg, Jiaxing Yang, Justin Powers, Alec H. Christian, John E. Wilkinson, Cian Monnin, Daina Avizonis, Craig J. Thomas, Richard A. Friedman, Michael D. Kluger, Michael A. Hollingsworth, Paul M. Grandgenett, Kelsey A. Klute, F. Dean Toste, Christopher J. Chang, Iok In Christine Chio. https://www.cell.com/molecular-cell/fulltext/S1097-2765(22)00541-X RAB21 controls autophagy and cellular energy homeostasis by regulating retromer-mediated recycling of SLC2A1/GLUT1 – Yifei Pei, Shuning Lv, Yong Shi, Jingwen Jia, Mengru Ma, Hailong Han, Rongying Zhang, Jieqiong Tan and Xinjun Zhang. https://www.tandfonline.com/doi/abs/10.1080/15548627.2022.2114271 An Integrated Proteomic and Glycoproteomic Investigation Reveals Alterations in the N-Glycoproteomic Network Induced by 2-Deoxy-D-Glucose in Colorectal Cancer Cells – Cheng Ma, Hong-Yuan Tsai, Qi Zhang, Lakmini Senavirathna, Lian Li, Lih-Shen Chin, Ru Chen and Sheng Pan. https://www.mdpi.com/1422-0067/23/15/8251/htm#app1-ijms-23-08251 PGAM1 regulation of ASS1 contributes to the progression of breast cancer through the cAMP/AMPK/CEBPB pathway – Min Liu, Runmei Li, Min Wang, Ting Liu, Qiuru Zhou, Dong Zhang, Jian Wang, Meng Shen, Xiubao Ren, Qian Sun. https://febs.onlinelibrary.wiley.com/doi/full/10.1002/1878-0261.13259 Monitoring glycolytic dynamics in single cells using a fluorescent biosensor for fructose 1,6-bisphosphate – John N. Koberstein, Melissa L. Stewart, Chadwick B. Smith, Andrei I. Tarasov, Frances M. Ashcroft, Philip J. S. Stork and Richard H. Goodman. https://www.pnas.org/doi/10.1073/pnas.2204407119

Lipid/NADPH sensing and metabolism


Lysosomal GPCR-like protein LYCHOS signals cholesterol sufficiency to mTORC1 – Hijai R. Shin, Y. Rose Citron, Lei Wang, Laura Tribouillard, Claire S. Goul, Robin Stipp, Yusuke Sugasawa, Aakriti Jain, Nolwenn Samson4, Chun-Yan Lim, Oliver B. Davis, David Castaneda-Carpio, Mingxing Qian, Daniel K. Nomura, Rushika M. Perera, Eunyong Park, Douglas F. Covey, Mathieu Laplante, Alex S. Evers, Roberto Zoncu. https://www.science.org/doi/10.1126/science.abg6621 Long non-coding RNA SNHG6 couples cholesterol sensing with mTORC1 activation in hepatocellular carcinoma – Fangzhou Liu, Tian Tian, Zhen Zhang, Shanshan Xie, Jiecheng Yang, Linyu Zhu, Wen Wang, Chengyu Shi, Lingjie Sang, Kaiqiang Guo, Zuozhen Yang, Lei Qu, Xiangrui Liu, Jian Liu, Qingfeng Yan, Huai-qiang Ju, Wenqi Wang, Hai-long Piao, Jianzhong Shao, Tianhua Zhou and Aifu Lin. https://www.nature.com/articles/s42255-022-00616-7 Metabolic reprogramming from glycolysis to fatty acid uptake and beta-oxidation in platinum-resistant cancer cells – Yuying Tan, Junjie Li, Guangyuan Zhao, Kai-Chih Huang, Horacio Cardenas, Yinu Wang, Daniela Matei and Ji-Xin Cheng. https://www.nature.com/articles/s41467-022-32101-w The MARCHF6 E3 ubiquitin ligase acts as an NADPH sensor for the regulation of ferroptosis – Kha The Nguyen, Sang-Hyeon Mun, Jihye Yang, Jongeun Lee, Ok-Hee Seok, Eunjeong Kim, Dasom Kim, So Young An, Dong-Young Seo, Jeong-Yong Suh, Yoontae Lee and Cheol-Sang Hwang. https://www.nature.com/articles/s41556-022-00973-1

Nutrient sensing/mTOR/AMPK


Direct control of lysosomal catabolic activity by mTORC1 through regulation of V-ATPase assembly – Edoardo Ratto, S. Roy Chowdhury, Nora S. Siefert, Martin Schneider, Marten Wittmann, Dominic Helm and Wilhelm Palm. https://www.nature.com/articles/s41467-022-32515-6 Midkine expression by stem-like tumor cells drives persistence to mTOR inhibition and an immune-suppressive microenvironment – Yan Tang, David J. Kwiatkowski and Elizabeth P. Henske. https://www.nature.com/articles/s41467-022-32673-7 The AMPK-HOXB9-KRAS axis regulates lung adenocarcinoma growth in response to cellular energy alterations – Tianzhuo Wang, Huiying Guo, Qianchen Li, Weijie Wu, Miao Yu, Lei Zhang, Cuicui Li, Jiagui Song, Zhenbin Wang, Jing Zhang, Yan Tang, Lei Kang, Hongquan Zhang, Jun Zhan https://www.cell.com/cell-reports/fulltext/S2211-1247(22)01074-9 Redox-dependent AMPK inactivation disrupts metabolic adaptation to glucose starvation in xCT-overexpressing cancer cells – Younghwan Lee, Yoko Itahana, Choon Chen Ong, Koji Itahana. https://journals.biologists.com/jcs/article-abstract/135/15/jcs259090/276257 Manganese is a physiologically relevant TORC1 activator in yeast and mammals – Raffaele Nicastro, Hélène Gaillard, Laura Zarzuela, Marie-Pierre Péli-Gulli, Elisabet Fernández-García, Mercedes Tomé, Néstor García-Rodríguez, Raúl V Durán, Claudio De Virgilio Is a corresponding author, Ralf Erik Wellinger. https://elifesciences.org/articles/80497 DEPDC5-dependent mTORC1 signaling mechanisms are critical for the anti-seizure effects of acute fasting – Christopher J. Yuskaitis, Jinita B. Modasia, Sandra Schrötter, Leigh-Ana Rossitto, Karenna J. Groff, Christopher Morici, Divakar S. Mithal, Ram P. Chakrabarty, Navdeep S. Chandel, Brendan D. Manning, Mustafa Sahin. https://www.cell.com/cell-reports/fulltext/S2211-1247(22)01098-1 PTEN inhibits AMPK to control collective migration – Florent Peglion, Lavinia Capuana, Isabelle Perfettini, Laurent Boucontet, Ben Braithwaite, Emma Colucci-Guyon, Emie Quissac, Karin Forsberg-Nilsson, Flora Llense and Sandrine Etienne-Manneville. https://www.nature.com/articles/s41467-022-31842-y

Nucleotide metabolism and sensing


Nucleotide imbalance decouples cell growth from cell proliferation – Frances F. Diehl, Teemu P. Miettinen, Ryan Elbashir, Christopher S. Nabel, Alicia M. Darnell, Brian T. Do, Scott R. Manalis, Caroline A. Lewis and Matthew G. Vander Heiden. https://www.nature.com/articles/s41556-022-00965-1 The mTORC1-SLC4A7 axis stimulates bicarbonate import to enhance de novo nucleotide synthesis – Eunus S. Ali, Anna Lipońska, Brendan P. O’Hara, David R. Amici, Michael D. Torno, Peng Gao, John M. Asara, Mee-Ngan F. Yap, Marc L. Mendillo, Issam Ben-Sahra. https://www.cell.com/molecular-cell/fulltext/S1097-2765(22)00544-5 SHMT2-mediated mitochondrial serine metabolism drives 5-FU resistance by fueling nucleotide biosynthesis – Erica Pranzini, Elisa Pardella, Livio Muccillo, Angela Leo, Ilaria Nesi, Alice Santi, Matteo Parri, Tong Zhang, Alejandro Huerta Uribe, Tiziano Lottini, Lina Sabatino, Anna Caselli, Annarosa Arcangeli, Giovanni Raugei, Vittorio Colantuoni, Paolo Cirri, Paola Chiarugi, Oliver D.K. Maddocks, Paolo Paoli, Maria Letizia Taddei. https://www.cell.com/cell-reports/fulltext/S2211-1247(22)01050-6 Cancer-associated fibroblasts employ NUFIP1-dependent autophagy to secrete nucleosides and support pancreatic tumor growth – Meng Yuan, Bo Tu, Hengchao Li, Huanhuan Pang, Nan Zhang, Minghe Fan, Jingru Bai, Wei Wang, Zhaoqi Shu, Christopher C. DuFort, Sihan Huo, Jie Zhai, Ke Yao, Lina Wang, Haoqiang Ying, Wei-Guo Zhu, Deliang Fu, Zeping Hu and Ying Zhao. https://www.nature.com/articles/s43018-022-00426-

Amino acid metabolism


Serine metabolism remodeling after platinum-based chemotherapy identifies vulnerabilities in a subgroup of resistant ovarian cancers – Tom Van Nyen, Mélanie Planque, Lilian van Wagensveld, Joao A. G. Duarte, Esther A. Zaal, Ali Talebi, Matteo Rossi, Pierre-René Körner, Lara Rizzotto, Stijn Moens, Wout De Wispelaere, Regina E. M. Baiden-Amissah, Gabe S. Sonke, Hugo M. Horlings, Guy Eelen, Emanuele Berardi, Johannes V. Swinnen, Celia R. Berkers, Peter Carmeliet, Diether Lambrechts, Ben Davidson, Reuven Agami, Sarah-Maria Fendt, Daniela Annibali and Frédéric Amant. https://www.nature.com/articles/s41467-022-32272-6 Glutamine addiction promotes glucose oxidation in triple-negative breast cancer – Lake-Ee Quek, Michelle van Geldermalsen, Yi Fang Guan, Kanu Wahi, Chelsea Mayoh, Seher Balaban, Angel Pang, Qian Wang, Mark J. Cowley, Kristin K. Brown, Nigel Turner, Andrew J. Hoy and Jeff Holst. https://www.nature.com/articles/s41388-022-02408-5 Metabolic requirement for GOT2 in pancreatic cancer depends on environmental context – Samuel A Kerk, Lin Lin, Amy L Myers, Damien J Sutton, Anthony Andren, Peter Sajjakulnukit, Li Zhang, Yaqing Zhang, Jennifer A Jiménez, Barbara S Nelson, Brandon Chen, Anthony Robinson, Galloway Thurston, Samantha B Kemp, Nina G Steele, Megan T Hoffman, Hui-Ju Wen, Daniel Long, Sarah E Ackenhusen, Johanna Ramos, Xiaohua Gao, Zeribe C Nwosu, Stefanie Galban, Christopher J Halbrook, David B Lombard, David R Piwnica-Worms, Haoqiang Ying, Marina Pasca di Magliano, Howard C Crawford, Yatrik M Shah, Costas A Lyssiotis. https://elifesciences.org/articles/73245 I also recommend the selection: “Glutamine cancer metabolism” by Sreeparna Banerjee – Sign up at http://biomed.news/reports

Mitochondria / heme / hypoxia


Operation of a TCA cycle subnetwork in the mammalian nucleus – Eleni Kafkia, Amparo Andres-Pons, Kerstin Ganter, Markus Seiler, Tom S. Smith, Anna Andrejeva, Paula Jouhten, Filipa Pereira, Catarina Franco, Anna Kuroshchenkova, Sergio Leone, Ritwick Sawarkar, Rebecca Boston, James Thaventhiran, Judith B. Zaugg1, Kathryn S. Lilley, Christophe Lancrin, Martin Beck, Kiran Raosaheb Patil. https://www.science.org/doi/10.1126/sciadv.abq5206 In vivo isotope tracing reveals a requirement for the electron transport chain in glucose and glutamine metabolism by tumors – Panayotis Pachnis, Zheng Wu, Brandon Faubert, Alpaslan Tasdogan, Wen Gu, Spencer Shelton, Ashley Solmonson, Aparna D. Rao, Akash K. Kaushik,  Thomas J. Rogers, Jessalyn M. Ubellacker, Collette A. LaVigne, Chendong Yang1, Bookyung Ko1, Vijayashree Ramesh, Jessica Sudderth, Lauren G. Zacharias,  Misty S. Martin-Sandoval1, Duyen Do1, Thomas P. Mathews1, Zhiyu Zhao, Prashant Mishra, Sean J. Morrison, Ralph J. DeBerardinis. https://www.science.org/doi/10.1126/sciadv.abn9550 Pyruvate and uridine rescue the metabolic profile of OXPHOS dysfunction – Isabelle Adant, Matthew Bird, Bram Decru, Petra Windmolders, Marie Wallays, Peter de Witte, Daisy Rymen, Peter Witters, Pieter Vermeersch, David Cassiman, Bart Ghesquière. https://www.sciencedirect.com/science/article/pii/S2212877822001065 Glutamine metabolism mediates sensitivity to respiratory complex II inhibition in acute myeloid leukemia – Alessia Roma,  Matthew Tcheng; Nawaz Ahmed; Sarah Walker; Preethi Jayanth; Mark D. Minden; Kristin Hope; Praveen P. Nekkar Rao; Jessica Luc; Andrew C. Doxey; Julie A. Reisz; Rachel Culp-Hill; Angelo D’Alessandro; Paul A. Spagnuolo. https://aacrjournals.org/mcr/article-abstract/doi/10.1158/1541-7786.MCR-21-1032/708293 Hypoxia‐induced ELF3 promotes tumor angiogenesis through IGF1/IGF1R – Seung Hee Seo, Soo-Yeon Hwang, Seohui Hwang, Sunjung Han, Hyojin Park, Yun-Sil Lee, Seung Bae Rho, Youngjoo Kwon https://www.embopress.org/doi/full/10.15252/embr.202152977 Metabolic-scale gene activation screens identify SLCO2B1 as a heme transporter that enhances cellular iron availability – Gokhan Unlu, Benjamin Prizer, Ranya Erdal, Hsi-Wen Yeh, Erol C. Bayraktar, Kıvanç Birsoy. https://www.cell.com/molecular-cell/fulltext/S1097-2765(22)00491-9 I also recommend the selection: “Mitochondrial metabolism in cancer” by Christian Frezza. Sign up at http://biomed.news/reports

Starvation/the UPR/celll death


Sublethal cytochrome c release generates drug-tolerant persister cells – Halime Kalkavan, Mark J. Chen, Jeremy C. Crawford, Giovanni Quarato, Patrick Fitzgerald, Stephen W.G. Tait, Colin R. Goding, Douglas R. Green. https://www.cell.com/cell/fulltext/S0092-8674(22)00978-3 Glycosylation defects, offset by PEPCK-M, drive entosis in breast carcinoma cells – Petra Hyroššová, Marc Aragó, Cristina Muñoz-Pinedo, Francesc Viñals, Pablo M. García-Rovés, Carmen Escolano, Andrés Méndez-Lucas and Jose C. Perales. https://www.nature.com/articles/s41419-022-05177-x GCN2 inhibition sensitizes arginine-deprived hepatocellular carcinoma cells to senolytic treatment – Rindert Missiaen, Nicole M. Anderson, Laura C. Kim, Bailey Nance, Michelle Burrows, Nicolas Skuli, Madeleine Carens, Romain Riscal, An Steensels, Fuming Li, M. Celeste Simon. https://www.cell.com/cell-metabolism/fulltext/S1550-4131(22)00230-3 Elucidation of an mTORC2-PKC-NRF2 pathway that sustains the ATF4 stress response and identification of Sirt5 as a key ATF4 effector – Ruizhi Li, Kristin F. Wilson and Richard A. Cerione. https://www.nature.com/articles/s41420-022-01156-5 Activating transcription factor 4-dependent hsa-miR-663a transcription mediates mTORC1/p70S6K1 signaling under leucine deprivation – Junki Yamamura, Sihui Ma, Huijuan Jia and Hisanori Kato. https://www.frontiersin.org/articles/10.3389/fnut.2022.965771/full The amino acid sensor GCN2 controls red blood cell clearance and iron metabolism through regulation of liver macrophages – Phoenix Toboz, Mehdi Amiri, Negar Tabatabaei, Catherine R. Dufour, Seung Hyeon Kim, Carine Fillebeen, Charles E. Ayemoba, Arkady Khoutorsky, Manfred Nairz, Lijian Shao, Kostandin V. Pajcini, Ki-Wook Kim, Vincent Giguère, Regiana L. Oliveira, Marco Constante, Manuela M. Santos, Carlos R. Morales, Kostas Pantopoulos, Nahum Sonenberg, Sandra Pinho and Soroush Tahmasebi. https://www.pnas.org/doi/10.1073/pnas.2121251119 Supramolecular assembly of GSK3α as a cellular response to amino acid starvation – Laura Hinze, Sabine Schreek, Andre Zeug, Nurul Khalida Ibrahim, Beate Fehlhaber, Lorent Loxha, Buesra Cinar, Evgeni Ponimaskin, James Degar, Connor McGuckin, Gabriela Chiosis, Cornelia Eckert, Gunnar Cario, Beat Bornhauser, Jean-Pierre Bourquin, Martin Stanulla, Alejandro Gutierrez. https://www.cell.com/molecular-cell/fulltext/S1097-2765(22)00492-0 FOXA1 prevents nutrients deprivation induced autophagic cell death through inducing loss of imprinting of IGF2 in lung adenocarcinoma – Junjun Li, Yongchang Zhang, Li Wang, Min Li, Jianbo Yang, Pan Chen, Jie Zhu, Xiayu Li, Zhaoyang Zeng, Guiyuan Li, Wei Xiong, James B. McCarthy, Bo Xiang and Mei Yi. https://www.nature.com/articles/s41419-022-05150-8 I also recommend the selection: “Unfolded protein response” by Susan Logue. Sign up at http://biomed.news/reports

Cancer Immunometabolism


Tumor cells dictate anti-tumor immune responses by altering pyruvate utilization and succinate signaling in CD8+ T cells – Ilaria Elia, Jared H. Rowe, Sheila Johnson, Shakchhi Joshi, Giulia Notarangelo, Kiran Kurmi, Sarah Weiss, Gordon J. Freeman, Arlene H. Sharpe, Marcia C. Haigis. https://www.cell.com/cell-metabolism/fulltext/S1550-4131(22)00228-5 TCR activation directly stimulates PYGB-dependent glycogenolysis to fuel the early recall response in CD8+ memory T cells – Huafeng Zhang, Jincheng Liu, Zhuoshun Yang, Liping Zeng, Keke Wei, Liyan Zhu, Liang Tang, Dianheng Wang, Yabo Zhou, Jiadi Lv, Nannan Zhou, Ke Tang, Jingwei Ma, Bo Huang. https://www.cell.com/molecular-cell/fulltext/S1097-2765(22)00538-X Fasting renders immunotherapy effective against low-immunogenic breast cancer while reducing side effects [in mice] – Salvatore Cortellino, Alessandro Raveane, Claudia Chiodoni, Gloria Delfanti, Federica Pisati, Vanessa Spagnolo, Euplio Visco, Giuseppe Fragale, Federica Ferrante, Serena Magni, Fabio Iannelli, Federica Zanardi, Giulia Casorati, Francesco Bertolini, Paolo Dellabona, Mario P. Colombo, Claudio Tripodo, Valter D. Longo. https://www.cell.com/cell-reports/fulltext/S2211-1247(22)01074-9 Glutathione peroxidase 2 is a metabolic driver of the tumor immune microenvironment and immune checkpoint inhibitor response – Kazi Mokim Ahmed, Ratna Veeramachaneni, Defeng Deng, Nagireddy Putluri, Vasanta Putluri, Maria F Cardenas, David A Wheeler, William K Decker, Andy I Frederick, Sawad Kazi, Andrew G Sikora, Vlad C Sandulache and Mitchell J Frederick. https://jitc.bmj.com/content/10/8/e004752 I also recommend the selection: “Immunometabolism of infection, cancer and immune-mediated disease” by Dylan Ryan. Sign up at http://biomed.news/reports

IDH


Inhibition of mutant IDH1 promotes cycling of acute myeloid leukemia stem cells – Emily Gruber, Joan So, Alexander C. Lewis, Rheana Franich, Rachel Cole, Luciano G. Martelotto, Amy J. Rogers, Eva Vidacs, Peter Fraser, Kym Stanley, Lisa Jones, Anna Trigos, Niko Thio, Jason Li, Brandon Nicolay, Scott Daigle, Adriana E. Tron, Marc L. Hyer, Jake Shortt, Ricky W. Johnstone, Lev M. Kats. https://www.cell.com/cell-reports/fulltext/S2211-1247(22)00995-0 Resistance to the isocitrate dehydrogenase 1 mutant inhibitor ivosidenib can be overcome by alternative dimer-interface binding inhibitors – Raphael Reinbold, Ingvild C. Hvinden, Patrick Rabe, Ryan A. Herold, Alina Finch, James Wood, Melissa Morgan, Maximillian Staudt, Ian J. Clifton, Fraser A. Armstrong, James S. O. McCullagh, Jo Redmond, Chiara Bardella, Martine I. Abboud and Christopher J. Schofield. https://www.nature.com/articles/s41467-022-32436-4
Miscellaneous
MYC sensitises cells to apoptosis by driving energetic demand – Joy Edwards-Hicks, Huizhong Su, Maurizio Mangolini, Kubra K. Yoneten, Jimi Wills, Giovanny Rodriguez-Blanco, Christine Young, Kevin Cho, Heather Barker, Morwenna Muir, Ania Naila Guerrieri, Xue-Feng Li, Rachel White, Piotr Manasterski, Elena Mandrou, Karen Wills, Jingyu Chen, Emily Abraham, Kianoosh Sateri, Bin-Zhi Qian, Peter Bankhead, Mark Arends, Noor Gammoh, Alex von Kriegsheim, Gary J. Patti, Andrew H. Sims, Juan Carlos Acosta, Valerie Brunton, Kamil R. Kranc, Maria Christophorou, Erika L. Pearce, Ingo Ringshausen and Andrew J. Finch Show fewer authors. https://www.nature.com/articles/s41467-022-32368-z Metastatic triple negative breast cancer adapts its metabolism to destination tissues while retaining key metabolic signatures – Fariba Roshanzamir, Jonathan L. Robinson, Daniel Cook, Mohammad Hossein Karimi-Jafari and Jens Nielsen. https://www.pnas.org/doi/10.1073/pnas.2205456119 Integrative analysis of plasma metabolomics and proteomics reveals the metabolic landscape of breast cancer – Rui An, Haitao Yu, Yanzhong Wang, Jie Lu, Yuzhen Gao, Xinyou Xie and Jun Zhang. https://cancerandmetabolism.biomedcentral.com/articles/10.1186/s40170-022-00289-6 Other recommended reports at http://biomed.news/reportsMetabolism and paracrine crosstalk between cancer and the organism” by Cristina Muñoz Pinedo “Metabolism of non-small cell lung carcinoma“, by the Muñoz Pinedo/Nadal lab “Metabolic-dependent epigenetic reprogramming in differentiation and disease“, by Alessandro Carrer.

Reviews


A practical guide for the analysis, standardization and interpretation of oxygen consumption measurements – Ajit S. Divakaruni and Martin Jastroch. https://www.nature.com/articles/s42255-022-00619-4 Lipids in cancer: a global view of the contribution of lipid pathways to metastatic formation and treatment resistance – Sophie Vasseur and Fabienne Guillaumond. https://www.nature.com/articles/s41389-022-00420-8 Regulation of gene expression by glycolytic and gluconeogenic enzymes – Xueli Bian, Hongfei Jiang, Ying Meng, Ying-ping Li, Jing Fang, Zhimin Lu. https://www.cell.com/trends/cell-biology/fulltext/S0962-8924(22)00036- Transsulfuration, minor player or crucial for cysteine homeostasis in cancer – Hai-Feng Zhang, Ramon I. Klein Geltink, Seth J. Parker, Poul H. Sorensen. https://www.cell.com/trends/cell-biology/fulltext/S0962-8924(22)00036-8 Hypoxia-responsive circRNAs: A novel but important participant in non-coding RNAs ushered toward tumor hypoxia – Benzheng Jiao, Shanshan Liu, Hongguang Zhao, Yuying Zhuang, Shumei Ma, Chenghe Lin, Jifan Hu and Xiaodong Liu. https://www.nature.com/articles/s41419-022-05114-y

Comments/other


The Warburg effect: Saturation of mitochondrial NADH shuttles triggers aerobic lactate fermentation – Hyllana C.D. Medeiros, Sophia Y. Lunt. https://www.cell.com/molecular-cell/fulltext/S1097-2765(22)00757-2 PYGBacking on glycogen metabolism to fuel early memory T cell recall responses – Joseph Longo, McLane J. Watson, Matthew J. Vos, Kelsey S. Williams, Russell G. Jones. https://www.cell.com/molecular-cell/fulltext/S1097-2765(22)00712-2 SLC4A7 and mTORC1 raise nucleotide synthesis with bicarbonate – Jessica C. Koe, Keeley G. Hewton, Seth J. Parker. https://www.cell.com/molecular-cell/fulltext/S1097-2765(22)00763-8 Leucine Dieting Inhibits Immunosuppressive B Cells in Colorectal Cancer. https://aacrjournals.org/cancerdiscovery/article-abstract/12/8/OF15/707242 NUFIP1+ stroma keeps PDAC fed – Holly Sandborg and Mara H. Sherman. https://www.nature.com/articles/s43018-022-00421-x Conference: EACR “Cancer Metabolism“. Bilbao, Spain : 11 – 13 October 2022. Organizers: Arkaitz Carracedo, Christian Frezza, Sarah-Maria Fendt, Sara Zanivan. https://www.eacr.org/conference/cancermetabolism2022/scientific-programme Conference: “Problems in the Powerhouse: Mitochondrial Dysfunction in Cancer“. Oct. 24/25th. Organizers: Payam Gammage, Stephen Tait , Tom MacVicar. Glasgow, UK. https://www.beatson.gla.ac.uk/Beatson-Workshop-October-2022/programme.html

MetaboList – July 2022


Nutrient sensing/starvation


Sestrin mediates detection of and adaptation to low-leucine diets in Drosophila – Xin Gu, Patrick Jouandin, Pranav V. Lalgudi, Rich Binari, Max L. Valenstein, Michael A. Reid, Annamarie E. Allen, Nolan Kamitaki, Jason W. Locasale, Norbert Perrimon and David M. Sabatini. https://www.nature.com/articles/s41586-022-04960-2

Sensing of the non-essential amino acid tyrosine governs the response to protein restriction in Drosophila – Hina Kosakamoto, Naoki Okamoto, Hide Aikawa, Yuki Sugiura, Makoto Suematsu, Ryusuke Niwa, Masayuki Miura and Fumiaki Obata. https://www.nature.com/articles/s42255-022-00608-7

Structure of the nutrient-sensing hub GATOR2 – Max L. Valenstein, Kacper B. Rogala, Pranav V. Lalgudi, Edward J. Brignole, Xin Gu, Robert A. Saxton, Lynne Chantranupong, Jonas Kolibius, Jan-Philipp Quast and David M. Sabatini. https://www.nature.com/articles/s41586-022-04939-z

DRAM‐4 and DRAM‐5 are compensatory regulators of autophagy and cell survival in nutrient‐deprived conditions – Valentin J. A. Barthet, Michaela Mrschtik, Elzbieta Kania, David G. McEwan, Dan Croft, James O’Prey, Jaclyn S. Long, Kevin M. Ryan. https://febs.onlinelibrary.wiley.com/doi/full/10.1111/febs.16365

Spatial regulation of AMPK signaling revealed by a sensitive kinase activity reporter – Danielle L. Schmitt, Stephanie D. Curtis, Anne C. Lyons, Jin-fan Zhang, Mingyuan Chen, Catherine Y. He, Sohum Mehta, Reuben J. Shaw and Jin Zhang. https://www.nature.com/articles/s41467-022-31190-x


Starvation – UPR


The amino acid sensor GCN2 suppresses Terminal Oligopyrimidine (TOP) mRNA translation via La-related Protein 1 (LARP1) – Zeenat Farooq, Fedho Kusuma, Phillip Burke, Catherine R. Dufour, Duckgue Lee, Negar Tabatabaei, Phoenix Toboz, Ernest Radovani, Jack Greenblatt, Jalees Rehman, Jacob Class, Arkady Khoutorsky, Bruno D. Fonseca, Justin M. Richner, Eloi Mercier, Guillaume Bourque, Vincent Giguère, Arvind R. Subramaniam, Jaeseok Han, Soroush Tahmasebi. https://www.jbc.org/article/S0021-9258(22)00719-0/fulltext

Activation of the canonical ER Stress IRE1-XBP1 Pathway by Insulin Regulates Glucose and Lipid Metabolism – Jinghua Peng, Caolitao Qin, Balamurugan Ramatchandirin, Alexia Pearah, Shaodong Guo, Mehboob Hussain, Liqing Yu, Fredric E. Wondisford, Ling He. https://www.jbc.org/article/S0021-9258(22)00725-6

FKBP11 rewires UPR signaling to promote glucose homeostasis in type 2 diabetes and obesity – Hilde Herrema, Dongxian Guan, Jae Won Choi, Xudong Feng, Mario Andres Salazar Hernandez, Farhana Faruk, Thomas Auen, Eliza Boudett, Rongya Tao, Hyonho Chun, Umut Ozcan. https://www.cell.com/cell-metabolism/fulltext/S1550-4131(22)00227-3

Activated amino acid response pathway generates apatinib resistance by reprograming glutamine metabolism in non-small-cell lung cancer – Xiaoshu Zhou, Rui Zhou, Xinrui Rao, Jiaxin Hong, Qianwen Li, Xiaohua Jie, Jian Wang, Yingzhuo Xu, Kuikui Zhu, Zhenyu Li and Gang Wu. https://www.nature.com/articles/s41419-022-05079-y


mTOR


Localization of a TORC1-eIF4F translation complex during CD8+ T cell activation drives divergent cell fate – Swantje Liedmann, Xueyan Liu, Clifford S. Guy, Jeremy Chase Crawford, Diego A. Rodriguez, Duygu Kuzuoğlu-Öztürk, Ao Guo, Katherine C. Verbist, Jamshid Temirov, Mark J. Chen, Davide Ruggero, Hui Zhang, Paul G. Thomas, Douglas R. Green. https://www.cell.com/molecular-cell/fulltext/S1097-2765(22)00327-6

Stress-induced perturbations in intracellular amino acids reprogram mRNA translation in osmoadaptation independently of the ISR – Dawid Krokowski, Raul Jobava, Krzysztof J. Szkop, Chien-Wen Chen, Xu Fu, Sarah Venus, Bo-Jhih Guan, Jing Wu, Zhaofeng Gao, Wioleta Banaszuk, Marek Tchorzewski, Tingwei Mu, Phil Ropelewski, William C. Merrick, Yuanhui Mao, Aksoylu Inci Sevval, Helen Miranda, Shu-Bing Qian, Maria Manifava, Nicholas T. Ktistakis, Anastasios Vourekas, Eckhard Jankowsky, Ivan Topisirovic, Ola Larsson, Maria Hatzoglou. https://www.cell.com/cell-reports/fulltext/S2211-1247(22)00894-4


Hypoxia


Hypoxia-mediated stabilization of HIF1A in prostatic intraepithelial neoplasia promotes cell plasticity and malignant progression – Mohamed A. Abu el Maaty, Julie Terzic, Céline Keime, Daniela Rovito, Régis Lutzing, Darya Yanushko, Maxime Parisotto, Elise Grelet, Izzie Jacques Namer, Véronique Lindner, Gilles Laverny, Daniel Metzger. https://www.science.org/doi/full/10.1126/sciadv.abo229

Hypoxia induces HIF1α-dependent epigenetic vulnerability in triple negative breast cancer to confer immune effector dysfunction and resistance to anti-PD-1 immunotherapy – Shijun Ma, Yue Zhao, Wee Chyan Lee, Li-Teng Ong, Puay Leng Lee, Zemin Jiang, Gokce Oguz, Zhitong Niu, Min Liu, Jian Yuan Goh, Wenyu Wang, Matias A. Bustos, Sidse Ehmsen, Adaikalavan Ramasamy, Dave S. B. Hoon, Henrik J. Ditzel, Ern Yu Tan, Qingfeng Chen and Qiang Yu. https://www.nature.com/articles/s41467-022-31764-9


IDH


Limited nutrient availability in the tumor microenvironment renders pancreatic tumors sensitive to allosteric IDH1 inhibitors – Ali Vaziri-Gohar, Joel Cassel, Farheen S. Mohammed, Mehrdad Zarei, Jonathan J. Hue, Omid Hajihassani, Hallie J. Graor, Yellamelli V. V. Srikanth, Saadia A. Karim, Ata Abbas, Erin Prendergast, Vanessa Chen, Erryk S. Katayama, Katerina Dukleska, Imran Khokhar, Anthony Andren, Li Zhang, Chunying Wu, Bernadette Erokwu, Chris A. Flask, Mahsa Zarei, Rui Wang, Luke D. Rothermel, Andrea M. P. Romani, Jessica Bowers, Robert Getts, Curtis Tatsuoka, Jennifer P. Morton, Ilya Bederman, Henri Brunengraber, Costas A. Lyssiotis, Joseph M. Salvino, Jonathan R. Brody and Jordan M. Winter. https://www.nature.com/articles/s43018-022-00393-y

Accumulation of oncometabolite D-2-Hydroxyglutarate by SLC25A1 inhibition: A metabolic strategy for induction of HR-ness and radiosensitivity – Kexu Xiang, Christian Kalthoff, Corinna Münch, Verena Jendrossek and Johann Matschke. https://www.nature.com/articles/s41419-022-05098-9


Glucose/lactate metabolism


Pyruvate Kinase M1 Suppresses Development and Progression of Prostate Adenocarcinoma – Shawn M. Davidson; Daniel R. Schmidt; Julia E. Heyman; James P. O’Brien; Amy C. Liu; William J. Israelsen; Talya L. Dayton; Raghav Sehgal; Roderick T. Bronson; Elizaveta Freinkman; Howard H. Mak; Giuseppe Nicolò Fanelli; Scott Malstrom; Gary Bellinger; Arkaitz Carracedo; Pier Paolo Pandolfi; Kevin D. Courtney; Abhishek Jha; Ronald A. DePinho; James W. Horner; Craig J. Thomas; Lewis C. Cantley; Massimo Loda; Matthew G. Vander Heiden. https://aacrjournals.org/cancerres/article/82/13/2403/705036/

Riboregulation of Enolase 1 activity controls glycolysis and embryonic stem cell differentiation – Ina Huppertz, Joel I. Perez-Perri, Panagiotis Mantas, Thileepan Sekaran, Thomas Schwarzl, Francesco Russo, Dunja Ferring-Appel, Zuzana Koskova, Lyudmila Dimitrova-Paternoga, Eleni Kafkia, Janosch Hennig, Pierre A. Neveu, Kiran Patil, Matthias W. Hentze. https://www.cell.com/molecular-cell/fulltext/S1097-2765(22)00486-5

G6PD-mediated increase in de novo NADP+ biosynthesis promotes antioxidant defense and tumor metastasis – Yang Zhang, Yi Xu, Wenyun Lu, Jinyang Li, Sixiang Yu, Eric J. Brown, Ben Z. Stanger, Joshua D. Rabinowitz, Xiaolu Yang. https://www.science.org/doi/full/10.1126/sciadv.abo040

The hexokinase “HKDC1” interaction with the mitochondria is essential for liver cancer progression – Md. Wasim Khan, Alexander R. Terry, Medha Priyadarshini, Vladimir Ilievski, Zeenat Farooq, Grace Guzman, Jose Cordoba-Chacon, Issam Ben-Sahra, Barton Wicksteed and Brian T. Layden. https://www.nature.com/articles/s41419-022-04999-z

Lactate promotes metastasis of normoxic colorectal cancer stem cells through PGC-1α-mediated oxidative phosphorylation – Shuang Liu, Hui Zhao, Yibing Hu, Chang Yan, Yulong Mi, Xiaolan Li, Deding Tao and Jichao Qin. https://www.nature.com/articles/s41419-022-05111-1


Amino acid metabolism


Tumoral microenvironment prevents de novo asparagine biosynthesis in B cell lymphoma, regardless of ASNS expression – Manuel Grima-Reyes, Ashaina Vandenberghe, Ivan Nemazanyy, Pauline Meola, Rachel Paul, Julie Reverso-Meinietti, Adriana Martinez-Turtos, Nicolas Nottet, Wai-Kin Chan, Philip L. Lorenzi, Sandrine Marchetti, Jean-Ehrland Ricci, Johanna Chiche. https://www.science.org/doi/full/10.1126/sciadv.abn6491

Targeting BCAT1 Combined with α-Ketoglutarate Triggers Metabolic Synthetic Lethality in Glioblastoma – Bo Zhang, Hui Peng, Mi Zhou, Lei Bao, Chenliang Wang, Feng Cai, Hongxia Zhang, Jennifer E. Wang, Yanling Niu, Yan Chen, Yijie Wang, Kimmo J. Hatanpaa, John A. Copland, Ralph J. DeBerardinis, Yingfei Wang, Weibo Luo. https://aacrjournals.org/cancerres/article-abstract/82/13/2388/705041

Cyclocreatine Suppresses Creatine Metabolism and Impairs Prostate Cancer Progression – Rachana Patel, Catriona A. Ford, Lisa Rodgers, Linda K. Rushworth, Janis Fleming, Ernest Mui, Tong Zhang, David Watson, Victoria Lynch, Gillian Mackay, David Sumpton, Owen J. Sansom, Johan Vande Voorde, Hing Y. Leung. https://aacrjournals.org/cancerres/article/82/14/2565/706947

Synthetic Essentiality of Tryptophan 2,3-Dioxygenase 2 in APC-Mutated Colorectal Cancer – Rumi Lee, Jiexi Li, Jun Li, Chang-Jiun Wu, Shan Jiang, Wen-Hao Hsu, Deepavali Chakravarti, Peiwen Chen, Kyle A. LaBella, Jing Li, Denise J. Spring, Di Zhao, Y. Alan Wang, Ronald A. DePinho. https://aacrjournals.org/cancerdiscovery/article-abstract/12/7/1702/705207

Autophagy loss impedes cancer-associated fibroblast activation via downregulating proline biosynthesis – Jingru Bai, Tong Liu, Bo Tu, Meng Yuan, Zhaoqi Shu, Minghe Fan, Sihan Huo, Yuyao Guo, Lina Wang, Hua Wang and Ying Zhao. https://www.tandfonline.com/doi/abs/10.1080/15548627.2022.2093026

Blocking glycine utilization inhibits multiple myeloma progression by disrupting glutathione balance – Jiliang Xia, Jingyu Zhang, Xuan Wu, Wanqing Du, Yinghong Zhu, Xing Liu, Zhenhao Liu, Bin Meng, Jiaojiao Guo, Qin Yang, Yihui Wang, Qinglin Wang, Xiangling Feng, Guoxiang Xie, Yi Shen, Yanjuan He, Juanjuan Xiang, Minghua Wu, Gang An, Lugui Qiu, Wei Jia and Wen Zhou. https://www.nature.com/articles/s41467-022-31248-w


Lipid metabolism


PI3K drives the de novo synthesis of coenzyme A from vitamin B5 – Christian C. Dibble, Samuel A. Barritt, Grace E. Perry, Evan C. Lien, Renee C. Geck, Sarah E. DuBois-Coyne, David Bartee, Thomas T. Zengeya, Emily B. Cohen, Min Yuan, Benjamin D. Hopkins, Jordan L. Meier, John G. Clohessy, John M. Asara, Lewis C. Cantley and Alex Toker. https://www.nature.com/articles/s41586-022-04984-8

Lipidomic profiling of exosomes from colorectal cancer cells and patients reveals potential biomarkers – Mohammed I. Y. Elmallah, Pablo Ortega-Deballon, Laure Hermite, Jean-Paul Pais-De-Barros, Jessica Gobbo, Carmen Garrido. https://febs.onlinelibrary.wiley.com/doi/full/10.1002/1878-0261.13223

Targeting de novo lipogenesis and the Lands cycle induces ferroptosis in KRAS-mutant lung cancer – Caterina Bartolacci, Cristina Andreani, Gonçalo Vale, Stefano Berto, Margherita Melegari, Anna Colleen Crouch, Dodge L. Baluya, George Kemble, Kurt Hodges, Jacqueline Starrett, Katerina Politi, Sandra L. Starnes, Daniele Lorenzini, Maria Gabriela Raso, Luisa M. Solis Soto, Carmen Behrens, Humam Kadara, Boning Gao, Ignacio I. Wistuba, John D. Minna, Jeffrey G. McDonald and Pier Paolo Scaglioni. https://www.nature.com/articles/s41467-022-31963-4


Nucleotides


Glutamine deficiency in solid tumor cells confers resistance to ribosomal RNA synthesis inhibitors – Melvin Pan, Christiane Zorbas, Maki Sugaya, Kensuke Ishiguro, Miki Kato 1, Miyuki Nishida, Hai-Feng Zhang, Marco M Candeias, Akimitsu Okamoto, Takamasa Ishikawa, Tomoyoshi Soga, Hiroyuki Aburatani, Juro Sakai, Yoshihiro Matsumura, Tsutomu Suzuki, Christopher G Proud, Denis L J Lafontaine, Tsuyoshi Osawa. https://www.nature.com/articles/s41467-022-31418-w

Inhibition of pyrimidine biosynthesis targets protein translation in acute myeloid leukemia – Joan So, Alexander C Lewis, Lorey K Smith, Kym Stanley, Rheana Franich, David Yoannidis, Lizzy Pijpers, Pilar Dominguez, Simon J Hogg, Stephin J Vervoort, Fiona C Brown, Ricky W Johnstone, Gabrielle McDonald, Danielle B Ulanet, Josh Murtie, Emily Gruber, Lev M Kats. https://www.embopress.org/doi/full/10.15252/emmm.202115203

Dihydroorotate dehydrogenase inhibition reveals metabolic vulnerability in chronic myeloid leukemia – Mohammad Houshmand, Nicoletta Vitale, Francesca Orso, Alessandro Cignetti, Ivan Molineris, Valentina Gaidano, Stefano Sainas, Marta Giorgis, Donatella Boschi, Carmen Fava, Alice Passoni, Marta Gai, Massimo Geuna, Federica Sora, Alessandra Iurlo, Elisabetta Abruzzese, Massimo Breccia, Olga Mulas, Giovanni Caocci, Fausto Castagnetti, Daniela Taverna, Salvatore Oliviero, Fabrizio Pane, Marco Lucio Lolli, Paola Circosta and Giuseppe Saglio. https://www.nature.com/articles/s41419-022-05028-9


Mitochondria and TCA


13C tracer analysis suggests extensive recycling of endogenous CO2 in vivo – Likun Duan, Daniel E. Cooper, Grace Scheidemantle, Jason W. Locasale, David G. Kirsch and Xiaojing Liu. https://cancerandmetabolism.biomedcentral.com/articles/10.1186/s40170-022-00287-8

Metabolic requirement for GOT2 in pancreatic cancer depends on environmental context – Samuel Kerk, Lin Lin, Amy L Myers, Damien J Sutton, Anthony Andren, Peter Sajjakulnukit, Li Zhang, Yaqing Zhang, Jennifer A Jiménez, Barbara S Nelson, Brandon Chen, Anthony Robinson, Galloway Thurston, Samantha B Kemp, Nina G Steele, Megan T Hoffman, Hui-Ju Wen, Daniel Long, Sarah E Ackenhusen, Johanna Ramos, Xiaohua Gao, Zeribe C Nwosu, Stefanie Galban, Christopher J Halbrook, David B Lombard, David R Piwnica-Worms, Haoqiang Ying, Marina Pasca di Magliano, Howard C Crawford , Yatrik M Shah, Costas A Lyssiotis. https://elifesciences.org/articles/73245

The mitochondrial enzyme FAHD1 regulates complex II activity in breast cancer cells and is indispensable for basal BT-20 cells in vitro – Max Holzknecht, Lena Guerrero-Navarro, Michele Petit, Eva Albertini, Elisabeth Damisch, Anna Simonini, Fernando Schmitt, Walther Parson, Heidelinde Fiegl, Alexander Weiss, Pidder Jansen-Duerr. https://febs.onlinelibrary.wiley.com/doi/10.1002/1873-3468.14462


Miscellaneous


Remission of obesity and insulin resistance is not sufficient to restore mitochondrial homeostasis in visceral adipose tissue – Alba Gonzalez-Franquesa, Pau Gama-Perez, Marta Kulis, Karolina Szczepanowska, Norma Dahdah, Sonia Moreno-Gomez, Ana Latorre-Pellicer, Rebeca Fernández-Ruiz, Antoni Aguilar-Mogas, Anne Hoffman, Erika Monelli, Sara Samino, Joan Miró-Blanch, GregorOemer, Xavier Duran, Estrella Sanchez-Rebordelo, Marc Schneebergerg, Merce Obac, Joel Montane, Giancarlo Castellano, Vicente Chapaprieta, Wenfei Sun, Lourdes Navarro, Ignacio Prieto, Carlos Castaño, Anna Novials, Ramon Gomis, Maria Monsalve, Marc Claret, Mariona Graupera, Guadalupe Soria, Christian Wolfrum, Joan Vendrell, Sonia Fernández-Veledo, Jose Antonio Enríque, Angel Carracedo, José Carlos Perales, Rubén Nogueiras, Laura Herrero, Aleksandra Trifunovi, Markus A.Keller, Oscar Yanes, Marta Sales-Pardo, Roger Guimerà, Matthias Blüher, José Ignacio Martín-Subero, Pablo M.Garcia-Roves. https://doi.org/10.1016/j.redox.2022.102353

Ketogenic diets slow melanoma growth in vivo regardless of tumor genetics and metabolic plasticity – Daniela D. Weber, Sepideh Aminzadeh-Gohari, Maheshwor Thapa, Anna-Sophia Redtenbacher, Luca Catalano, Tânia Capelôa, Thibaut Vazeille, Michael Emberger, Thomas K. Felder, René G. Feichtinger, Peter Koelblinger, Guido Dallmann, Pierre Sonveaux, Roland Lang and Barbara Kofler. https://cancerandmetabolism.biomedcentral.com/articles/10.1186/s40170-022-00288-7

The onset of PI3K-related vascular malformations occurs during angiogenesis and is prevented by the AKT inhibitor miransertib – Piotr Kobialka, Helena Sabata, Odena Vilalta, Leonor Gouveia, Ana Angulo-Urarte, Laia Muixí, Jasmina Zanoncello, Oscar Muñoz-Aznar, Nagore G Olaciregui, Lucia Fanlo, Anna Esteve-Codina, Cinzia Lavarino, Biola M Javierre, Veronica Celis, Carlota Rovira, Susana López-Fernández, Eulàlia Baselga, Jaume Mora, Sandra D Castillo, Mariona Graupera. https://www.embopress.org/doi/full/10.15252/emmm.202115619

FKBP11 rewires UPR signaling to promote glucose homeostasis in type 2 diabetes and obesity – Hilde Herrema, Dongxian Guan, Jae Won Choi, Xudong Feng, Mario Andres Salazar Hernandez, Farhana Faruk, Thomas Auen, Eliza Boudett, Rongya Tao, Hyonho Chun, Umut Ozcan. https://www.cell.com/cell-metabolism/fulltext/S1550-4131(22)00187-5


Reviews


Targeting mitochondrial metabolism for precision medicine in cancer – Lourdes Sainero-Alcolado, Judit Liaño-Pons, María Victoria Ruiz-Pérez and Marie Arsenian-Henriksson. https://www.nature.com/articles/s41418-022-01022-y

Targeting the Leukemic stem cell protein machinery by inhibition of mitochondrial pyrimidine synthesis – Elisa Donato, Andreas Trumpp. https://www.embopress.org/doi/full/10.15252/emmm.202216171

Polyamines in cancer: integrating organismal metabolism and antitumour immunity – Cassandra E. Holbert, Michael T. Cullen, Robert A. Casero Jr and Tracy Murray Stewart. https://www.nature.com/articles/s41568-022-00473-2

Scaling of biosynthesis and metabolism with cell size – Clotilde Cadart and Rebecca Heald. https://www.molbiolcell.org/doi/abs/10.1091/mbc.E21-12-0627

Therapeutic Targeting of Tumor Cells and Tumor Immune Microenvironment Vulnerabilities – Balaraman Kalyanaraman, Gang Cheng and Micael Hardy. https://www.frontiersin.org/articles/10.3389/fonc.2022.816504/full

The Promise of Targeting Hypoxia to Improve Cancer Immunotherapy: Mirage or Reality? – Bassam Janji, Salem Chouaib. https://www.frontiersin.org/articles/10.3389/fimmu.2022.880810/full

Beyond controlling cell size: functional analyses of S6K in tumorigenesis – Xueji Wu, Wei Xie, Wenxuan Xie, Wenyi Wei and Jianping Guo. https://www.nature.com/articles/s41419-022-05081-4


Comments


Driving with Both Feet: Supplementing AKG While Inhibiting BCAT1 Leads to Synthetic Lethality in GBM – Noah Meurs, Deepak Nagrath. https://aacrjournals.org/cancerres/article-abstract/82/13/2354/705023

IFNα Potentiates Immune-Checkpoint Blockade by Rewiring Metabolic Cross-talk – Kung-Chi Kao, Alison Jaccard, Ping-Chih Ho. https://aacrjournals.org/cancerdiscovery/article-abstract/12/7/1615/705216

Akt protein boosts cancer metabolism through a two-pronged attack – Philipp Poeller and Almut Schulze. https://www.nature.com/articles/d41586-022-01603-4

Low Phosphoglycerate Dehydrogenase Levels Drive Metastatic Disseminationhttps://aacrjournals.org/cancerdiscovery/article-abstract/12/7/1611/705198

Colorectal Cancer Growth Is Reduced by the Ketone Body β-Hydroxybutyratehttps://aacrjournals.org/cancerdiscovery/article-abstract/12/7/OF15/705202

Ceramide Accumulation Induces the Integrated Stress Response in AMLhttps://aacrjournals.org/cancerdiscovery/article-abstract/12/7/OF14/705203

MetaboList – June 2022


Nutrient sensing – mTOR


Zonated leucine sensing by Sestrin-mTORC1 in the liver controls the response to dietary leucine – Andrew L. Cangelosi, Anna M. Puszynska, Justin M. Roberts, Andrea Armani, Thao P. Nguyen, Jessica B. Spinelli, Tenzin Kunchok, Brianna Wang, Sze Ham Chan, Caroline A. Lewis, William C. Comb, George W. Bell, Aharon Helman, David M. Sabatini. https://www.science.org/doi/abs/10.1126/science.abi9547

Prolonged deprivation of arginine or leucine induces PI3K/Akt-dependent reactivation of mTORC1 – Gwen R. Buel, Huy Q. Dang, John M. Asara, John Blenis, Anders P. Mutvei. https://www.jbc.org/article/S0021-9258(22)00470-7/fulltext

Vps33B controls Treg cell suppressive function through inhibiting lysosomal nutrient sensing complex-mediated mTORC1 activation – Hongrui Xiang, Yuexiao Tao, Zhenyan Jiang, Xian Huang, Huizi Wang, Wei Cao, Jia Li, Rui Ding, Mingyi Shen, Ru Feng, Linsen Li, Chenyang Guan, Jiamin Liu, Jun Ni, Lei Chen, Zhengting Wang, Youqiong Ye, Qing Zhong, Junling Liu, Qiang Zou, Xuefeng Wu. https://www.cell.com/cell-reports/fulltext/S2211-1247(22)00725-2


Nutrient sensing – ISR


A stromal Integrated Stress Response activates perivascular cancer-associated fibroblasts to drive angiogenesis and tumour progression – Ioannis I. Verginadis, Harris Avgousti, James Monslow, Giorgos Skoufos, Frank Chinga, Kyle Kim, Nektaria Maria Leli, Ilias V. Karagounis, Brett I. Bell, Anastasia Velalopoulou, Carlo Salas Salinas, Victoria S. Wu, Yang Li, Jiangbin Ye, David A. Scott, Andrei L. Osterman, Arjun Sengupta, Aalim Weljie, Menggui Huang, Duo Zhang, Yi Fan, Enrico Radaelli, John W. Tobias, Florian Rambow, Panagiotis Karras, Jean-Christophe Marine, Xiaowei Xu, Artemis G. Hatzigeorgiou, Sandra Ryeom, J. Alan Diehl, Serge Y. Fuchs, Ellen Puré and Constantinos Koumenis. https://www.nature.com/articles/s41556-022-00918-8

Activation and execution of the hepatic integrated stress response by dietary essential amino acid deprivation is amino acid specific – Jonsson WO, Mirek ET, Wek RC, Anthony TG. https://faseb.onlinelibrary.wiley.com/doi/10.1096/fj.202200204RR

Cellular responses to halofuginone reveal a vulnerability of the GCN2 branch of the integrated stress response – Aleksandra P Pitera, Maria Szaruga, Sew-Yeu Peak-Chew, Steven W Wingett, Anne Bertolotti. https://www.embopress.org/doi/full/10.15252/embj.2021109985


Nutrient sensing: AMPK


A novel AMPK activator shows therapeutic potential in hepatocellular carcinoma by suppressing HIF1α‐mediated aerobic glycolysis – Hsing-I Tseng, Yi-Siang Zeng, Ying-Chung Jimmy Lin, Jui-Wen Huang, Chih-Lung Lin, Meng-Hsuan Lee, Fan-Wei Yang, Te-Ping Fang, Ai-Chung Mar, Jung-Chen Su. https://febs.onlinelibrary.wiley.com/doi/full/10.1002/1878-0261.13211

Structure-function analysis of the AMPK activator SC4 and identification of a potent pan AMPK activator – Ashley J. Ovens, Yi Sing Gee, Naomi X.Y. Ling, Dingyi Yu, Justin P. Hardee, Jin D. Chung; Kevin R.W. Ngoei, Nicholas J. Waters, Nolan J. Hoffman, John W. Scott, Kim Loh, Katrin Spengler, Regine Heller, Michael W. Parker, Gordon S. Lynch, Fei Huang, Sandra Galic, Bruce E. Kemp, Jonathan B. Baell, Jonathan S. Oakhill, Christopher G. Langendorf. https://portlandpress.com/biochemj/article/479/11/1181/231304

Translation initiation factor eIF3a regulates glucose metabolism and cell proliferation via promoting small GTPase Rheb synthesis and AMPK activation – Shijie Ma, Zizheng Dong, Yanfei Huang, Jing-Yuan Liu, Jian-Ting Zhang. https://www.jbc.org/article/S0021-9258(22)00484-7/fulltext


Nutrient sensing – miscellaneous


The polyol pathway is an evolutionarily conserved system for sensing glucose uptake – Hiroko Sano, Akira Nakamura, Mariko Yamane, Hitoshi Niwa, Takashi Nishimura, Kimi Araki, Kazumasa Takemoto, Kei-ichiro Ishiguro, Hiroki Aoki, Yuzuru Kato, Masayasu Kojima. https://www.nature.com/articles/s42255-022-00568-y

Ammonia stimulates SCAP/Insig dissociation and SREBP-1 activation to promote lipogenesis and tumour growth – Chunming Cheng, Feng Geng, Zoe Li, Yaogang Zhong, Huabao Wang, Xiang Cheng, Yue Zhao, Xiaokui Mo, Craig Horbinski, Wenrui Duan, Arnab Chakravarti, Xiaolin Cheng and Deliang Guo. https://www.nature.com/articles/s42255-022-00568-y

Glutamine deficiency in solid tumor cells confers resistance to ribosomal RNA synthesis inhibitors – Melvin Pan, Christiane Zorbas and Tsuyoshi Osawa. https://www.nature.com/articles/s41467-022-31418-w

A YAP/TAZ-TEAD signalling module links endothelial nutrient acquisition to angiogenic growth – Yu Ting Ong, Jorge Andrade, Max Armbruster, Chenyue Shi, Marco Castro, Ana S. H. Costa, Toshiya Sugino, Guy Eelen, Barbara Zimmermann, Kerstin Wilhelm, Joseph Lim, Shuichi Watanabe, Stefan Guenther, Andre Schneider, Francesca Zanconato, Manuel Kaulich, Duojia Pan, Thomas Braun, Holger Gerhardt, Alejo Efeyan, Peter Carmeliet, Stefano Piccolo, Ana Rita Grosso and Michael Potente. https://www.nature.com/articles/s42255-022-00584-y


Hypoxia


Adaptive stimulation of macropinocytosis overcomes aspartate limitation in cancer cells under hypoxia – Javier Garcia-Bermudez, Michael A. Badgley, Sheela Prasad, Lou Baudrier, Yuyang Liu, Konnor La, Mariluz Soula, Robert T. Williams, Norihiro Yamaguchi, Rosa F. Hwang, Laura J. Taylor, Elisa de Stanchina, Bety Rostandy, Hanan Alwaseem, Henrik Molina, Dafna Bar-Sagi and Kıvanç Birsoy. https://www.nature.com/articles/s42255-022-00583-z

Impaired oxygen-sensitive regulation of mitochondrial biogenesis within the von Hippel–Lindau syndrome – Shuijie Li, Wenyu Li, Juan Yuan, Petra Bullova, Jieyu Wu, Xuepei Zhang, Yong Liu, Monika Plescher, Javier Rodriguez, Oscar C. Bedoya-Reina, Paulo R. Jannig, Paula Valente-Silva, Meng Yu, Marie Arsenian Henriksson, Roman A. Zubarev, Anna Smed-Sörensen, Carolyn K. Suzuki, Jorge L. Ruas, Johan Holmberg, Catharina Larsson, C. Christofer Juhlin, Alex von Kriegsheim, Yihai Cao and Susanne Schlisio. https://www.nature.com/articles/s42255-022-00593-x

Hypoxia-inducible factor-1α and poly [ADP ribose] polymerase 1 cooperatively regulate Notch3 expression under hypoxia via a non-canonical mechanism – Hideaki Nakamura, Hiroki Sekine, Hiroyuki Kato, Hisao Masai, Katarina Gradin, Lorenz Poellinger. https://www.jbc.org/article/S0021-9258(22)00579-8/fulltext


Glucose metabolism


G6PD inhibition sensitizes ovarian cancer cells to oxidative stress in the metastatic omental microenvironment – Shree Bose, Qiang Huang, Yunhan Ma, Lihua Wang, Grecia O. Rivera, Yunxin Ouyang, Regina Whitaker, Rebecca A. Gibson, Christopher D. Kontos, Andrew Berchuck, Rebecca A. Previs, Xiling Shen. https://www.cell.com/cell-reports/fulltext/S2211-1247(22)00801-4

A Paradigm of Cancer Immunotherapy Based on 2-[18F]FDG and Anti–PD-L1 mAb Combination to Enhance the Antitumor Effect – Xuejun Wen; Changrong Shi;, Xinying Zeng, Liang Zhao, Lanlin Yao, Zhida Liu, Lixia Feng, Deliang Zhang, Jinxiong Huang, Yesen Li, Qin Lin, Haojun Chen, Rongqiang Zhuang, Xiaoyuan Chen,  Xianzhong Zhang, Zhide Guo. https://aacrjournals.org/clincancerres/article-abstract/28/13/2923/704989

The metabolic enzyme hexokinase 2 localizes to the nucleus in AML and normal haematopoietic stem and progenitor cells to maintain stemness – Geethu Emily Thomas, Grace Egan, Laura García-Prat, Aaron Botham, Veronique Voisin, Parasvi S. Patel, Fieke W. Hoff, Jordan Chin, Boaz Nachmias, Kerstin B. Kaufmann, Dilshad H. Khan, Rose Hurren, Xiaoming Wang, Marcela Gronda, Neil MacLean, Cristiana O’Brien, Rashim P. Singh, Courtney L. Jones, Shane M. Harding, Brian Raught, Andrea Arruda, Mark D. Minden, Gary D. Bader, Razq Hakem, Steve Kornblau, John E. Dick and Aaron D. Schimmer. https://www.nature.com/articles/s41556-022-00925-9


Amino acid metabolism


Cancer-associated fibroblasts require proline synthesis by PYCR1 for the deposition of pro-tumorigenic extracellular matrix – Emily J. Kay, Karla Paterson, Carla Riero-Domingo, David Sumpton, J. Henry M. Däbritz, Saverio Tardito, Claudia Boldrini, Juan R. Hernandez-Fernaud, Dimitris Athineos, Sandeep Dhayade, Ekaterina Stepanova, Enio Gjerga, Lisa J. Neilson, Sergio Lilla, Ann Hedley, Grigorios Koulouras, Grace McGregor, Craig Jamieson, Radia Marie Johnson, Morag Park, Kristina Kirschner, Crispin Miller, Jurre J. Kamphorst, Fabricio Loayza-Puch, Julio Saez-Rodriguez, Massimiliano Mazzone, Karen Blyth, Michele Zagnoni and Sara Zanivan. https://www.nature.com/articles/s42255-022-00582-0

Mitochondrial Calcium Uniporter Drives Metastasis and Confers a Targetable Cystine Dependency in Pancreatic Cancer – Xiuchao Wang, Yunzhan Li, Zekun Li, Shengchen Lin, Hongwei Wang, Jianwei Sun, Chungen Lan, Liangliang Wu, Dongxiao Sun, Chongbiao Huang, Pankaj K. Singh, Nadine Hempel, Mohamed Trebak, Gina M. DeNicola, Jihui Hao, Shengyu Yang. https://aacrjournals.org/cancerres/article-abstract/82/12/2254/699357

Characterizing unexpected interactions of a glutamine transporter inhibitor with members of the SLC1A transporter family – Natasha Freidman, Chelsea Briot, Renae Ryan. https://www.jbc.org/article/S0021-9258(22)00620-2

mTORC1-independent translation control in mammalian cells by methionine adenosyltransferase 2A and S-adenosylmethionine – Mahabub Alam, Hiroki Shima, Yoshitaka Matsuo, Nguyen Chi Long, Mitsuyo Matsumoto, Yusho Ishii, Nichika Sato, Takato Sugiyama, Risa Nobuta, Satoshi Hashimoto, Liang Liu, Mika K. Kaneko, Yukinari Kato, Toshifumi Inada, Kazuhiko Igarashi. https://www.jbc.org/article/S0021-9258(22)00525-7/fulltext

Loss of PRMT7 reprograms glycine metabolism to selectively eradicate leukemia stem cells in CML – Chang Liu, Waiyi Zou, Danian Nie, Shuyi Li, Chen Duan, Min Zhou, Peilong Lai, Shengyong Yang, Sen Ji, Yangqiu Li, Mei Mei, Shilai Bao, Yanli Jin, Jingxuan Pan. https://www.cell.com/cell-metabolism/fulltext/S1550-4131(22)00131-0

Asparagine synthetase regulates lung-cancer metastasis by stabilizing the β-catenin complex and modulating mitochondrial response – Dong-Jing Cai, Zi-Yu Zhang, Yue Bu, Li Li, Yue-Zhen Deng, Lun-Quan Sun, Cheng-Ping Hu and Min Li. https://www.nature.com/articles/s41419-022-05015-0


Lipid metabolism


Cancer cells depend on environmental lipids for proliferation when electron acceptors are limited – Zhaoqi Li, Brian W. Ji, Purushottam D. Dixit, Konstantine Tchourine, Evan C. Lien, Aaron M. Hosios, Keene L. Abbott, Justine C. Rutter, Anna M. Westermark, Elizabeth F. Gorodetsky, Lucas B. Sullivan, Matthew G. Vander Heiden and Dennis Vitkup. https://www.nature.com/articles/s42255-022-00588-8

The short-chain fatty acid acetate modulates epithelial-to-mesenchymal transition – Junfang Lyu, Mehdi Pirooznia, Yuesheng Li, and Jianhua Xiong. https://www.molbiolcell.org/doi/10.1091/mbc.E22-02-0066?ai=s5&ui=2oy6&af=T

Ct-OATP1B3 promotes high-grade serous ovarian cancer metastasis by regulation of fatty acid beta-oxidation and oxidative phosphorylation – Yutang Huang, Yan Du, Yujie Zheng, Chunjie Wen, Hecun Zou, Jiafeng Huang, Honghao Zhou, Hongbo Zhao and Lanxiang Wu. https://www.nature.com/articles/s41419-022-05014-1

MYCN-driven fatty acid uptake is a metabolic vulnerability in neuroblastoma – Ling Tao, Mahmoud A. Mohammad and Eveline Barbieri. https://www.nature.com/articles/s41467-022-31331-2


Mitochondria and iron


NCOA4-mediated ferritinophagy is a pancreatic cancer dependency via maintenance of iron bioavailability for iron-sulfur cluster proteins – Naiara Santana-Codina, Maria Quiles del Rey, Kevin S. Kapner, Huan Zhang, Ajami Gikandi, Callum Malcolm, Clara Poupault, Miljan Kuljanin, Kristen M. John, Douglas E. Biancur, Brandon Chen, Nupur K. Das, Kristen E. Lowder, Connor J. Hennessey, Wesley Huang, Annan Yang, Yatrik M. Shah, Jonathan A. Nowak, Andrew J. Aguirre, Joseph D. Mancias. https://aacrjournals.org/cancerdiscovery/article/doi/10.1158/2159-8290.CD-22-0043/704854

Coordinated transcriptional and catabolic programs support iron dependent adaptation to RAS-MAPK pathway inhibition in pancreatic cancer – Mirunalini Ravichandran, Jingjie Hu, Charles Cai, Nathan P. Ward, Anthony Venida, Callum Foakes, Miljan Kuljanin, Annan Yang, Connor J. Hennessey, Yang Yang, Brandon R. Desousa, Gilles Rademaker, Annelot A.L. Staes, Zeynep Cakir, Isha H. Jain, Andrew J. Aguirre, Joseph D. Mancias, Yin Shen, Gina M. DeNicola, Rushika M. Perera. https://aacrjournals.org/cancerdiscovery/article-abstract/doi/10.1158/2159-8290.CD-22-0044/706733

Macrophage mitochondrial bioenergetics and tissue invasion are boosted by an Atossa‐Porthos axis in Drosophila – Shamsi Emtenani, Elliot T Martin, Attila Gyoergy, Julia Bicher, Jakob-Wendelin Genger, Thomas Köcher, Maria Akhmanova, Mariana Guarda, Marko Roblek, Andreas Bergthaler, Thomas R Hurd, Prashanth Rangan, Daria E Siekhaus. https://www.embopress.org/doi/full/10.15252/embj.2021109049


Cancer Immunometabolism


Non-oxidative pentose phosphate pathway controls regulatory T cell function by integrating metabolism and epigenetics – Qi Liu, Fangming Zhu, Xinnan Liu, Ying Lu, Ke Yao, Na Tian, Lingfeng Tong, David A. Figge, Xiuwen Wang, Yichao Han, Yakui Li, Yemin Zhu, Lei Hu, Yingning Ji, Nannan Xu, Dan Li, Xiaochuan Gu, Rui Liang, Guifang Gan, Lifang Wu, Ping Zhang, Tianle Xu, Hui Hu, Zeping Hu, Huji Xu, Dan Ye, Hui Yang, Bin Li and Xuemei Tong. https://www.nature.com/articles/s42255-022-00575-z

Leucine-tRNA-synthase-2-expressing B cells contribute to colorectal cancer immunoevasion – Zhiqiang Wang, Zhou Lu, Shengli Lin, Jie Xia, Ziwen Zhong, Zhangjuan Xie, Yun Xing, Jingbo Qie, Mengxia Jiao, Yifan Li, Haoyu Wen, Pengyuan Zhao, Dan Zhang, Pinghong Zhou, Jiawen Qian, Feifei Luo, Luman Wang, Hongxiu Yu, Jie Liu, Jie Gu, Ronghua Liu, Yiwei Chu. https://www.cell.com/immunity/fulltext/S1074-7613(22)00188-1

Endogenous glutamine is rate-limiting for anti-CD3 and anti-CD28 induced CD4+ T-cell proliferation and glycolytic activity under hypoxia and normoxia – Jonas A. Wik, Azazul Chowdhury, Shrikant Kolan, Nasser E. Bastani, Gaoyang Li, Kazi Alam, Franco Grimolizzi, Bjørn S. Skålhegg. https://portlandpress.com/biochemj/article/479/11/1221/231349

Tumor metabolite lactate promotes tumorigenesis by modulating MOESIN lactylation and enhancing TGF-β signaling in regulatory T cells – Jian Gu, Jinren Zhou, Qiuyang Chen, Xiaozhang Xu, Ji Gao, Xiangyu Li, Qing Shao, Bo Zhou, Haoming Zhou, Song Wei, Qi Wang, Yuan Liang, Ling Lu. https://www.cell.com/cell-reports/fulltext/S2211-1247(22)00772-0


Miscellaneous


Metformin-induced reductions in tumor growth involves modulation of the gut microbiome – Lindsay A. Broadfield, Amna Saigal, Jake C. Szamosi, Joanne A. Hammill, Ksenia Bezverbnaya, Dongdong Wang, Jaya Gautam, Evangelia E. Tsakiridis, Fiorella Di Pastena, Jamie McNicol, Jianhan Wu, Saad Syed, James S.V. Lally, Amogelang R. Raphenya, Marie-Jose Blouin, Michael Pollak, Andrea Sacconi, Giovanni Blandino, Andrew G. McArthur, Jonathan D. Schertzer. https://www.sciencedirect.com/science/article/pii/S2212877822000679

An exercise-inducible metabolite that suppresses feeding and obesity – Veronica L. Li, Yang He, Kévin Contrepois, Hailan Liu, Joon T. Kim, Amanda L. Wiggenhorn, Julia T. Tanzo, Alan Sheng-Hwa Tung, Xuchao Lyu, Peter-James H. Zushin, Robert S. Jansen, Basil Michael, Kang Yong Loh, Andrew C. Yang, Christian S. Carl, Christian T. Voldstedlund, Wei Wei, Stephanie M. Terrell, Benjamin C. Moeller, Rick M. Arthur, Gareth A. Wallis, Koen van de Wetering, Andreas Stahl, Bente Kiens, Erik A. Richter, Steven M. Banik, Michael P. Snyder, Yong Xu and Jonathan Z. Long. https://www.nature.com/articles/s41586-022-04828-5


Reviews


Ribosome biogenesis and the cellular energy economy – David Shore, Benjamin Albert. https://www.cell.com/current-biology/fulltext/S0960-9822(22)00719-9

Ferroptosis at the intersection of lipid metabolism and cellular signaling – Deguang Liang, Alexander M. Minikes, Xuejun Jiang. https://www.cell.com/molecular-cell/fulltext/S1097-2765(22)00260-X

Shaping fuel utilization by mitochondria – Lukas Alan, Luca Scorrano. https://www.cell.com/current-biology/fulltext/S0960-9822(22)00765-5

Therapeutic Targeting of Tumor Cells and Tumor Immune Microenvironment Vulnerabilities – Balaraman Kalyanaraman, Gang Cheng, Micael Hardy. https://www.frontiersin.org/articles/10.3389/fonc.2022.816504/full

Fatty acid synthase: a druggable driver of breast cancer brain metastasis – Javier A. Menendez and Ruth Lupu. https://www.tandfonline.com/doi/full/10.1080/14728222.2022.2077189


Comments


NAD+ regeneration drives cancer cell proliferation – Xu Han and M. Celeste Simon. https://www.nature.com/articles/s42255-022-00579-9

YAP/TAZ–TEAD link angiogenesis to nutrients – Roxana E. Oberkersch and Massimo M. Santoro. https://www.nature.com/articles/s42255-022-00579-9

TCA cycle off, ATF4 on for metabolic homeostasis – Durga Mahor, Rishikesh Pandey, Vinay Bulusu. https://www.cell.com/trends/biochemical-sciences/fulltext/S0968-0004(22)00065-2

Research is a process – Sarah-Maria Fendt. https://www.nature.com/articles/s41556-022-00954-4

MetaboList – May 2022


Nucleotides / one carbon


Purine nucleotide depletion prompts cell migration by stimulating the serine synthesis pathway – Mona Hoseini Soflaee, Rushendhiran Kesavan, Umakant Sahu, Alpaslan Tasdogan, Elodie Villa, Zied Djabari, Feng Cai, Diem H. Tran, Hieu S. Vu, Eunus S. Ali, Halie Rion, Brendan P. O’Hara, Sherwin Kelekar, James Hughes Hallett, Misty Martin, Thomas P. Mathews, Peng Gao, John M. Asara, Brendan D. Manning, Issam Ben-Sahra and Gerta Hoxhaj. https://www.nature.com/articles/s41467-022-30362-z

Suppression of nuclear GSK3 signaling promotes serine/one-carbon metabolism and confers metabolic vulnerability in lung cancer cells – Long He, Jennifer Endress, Sungyun Cho, Zhongchi Li, Yuxiang Zheng, John M. Asara, John Blenis. https://www.science.org/doi/full/10.1126/sciadv.abm8786

Mitochondria preserve an autarkic one-carbon cycle to confer growth-independent cancer cell migration and metastasis – Nicole Kiweler, Catherine Delbrouck, Vitaly I. Pozdeev, Laura Neises, Leticia Soriano-Baguet, Kim Eiden, Feng Xian, Mohaned Benzarti, Lara Haase, Eric Koncina, Maryse Schmoetten, Christian Jaeger, Muhammad Zaeem Noman, Alexei Vazquez, Bassam Janji, Gunnar Dittmar, Dirk Brenner, Elisabeth Letellier and Johannes Meiser. https://www.nature.com/articles/s41467-022-30363-y

CDK12 promotes tumorigenesis but induces vulnerability to therapies inhibiting folate one-carbon metabolism in breast cancer – G. Filippone, D. Gaglio, R. Bonfanti, F. A. Tucci, E. Ceccacci, R. Pennisi, M. Bonanomi, G. Jodice, M. Tillhon, F. Montani, G. Bertalot, S. Freddi, M. Vecchi, A. Taglialatela, M. Romanenghi, F. Romeo, N. Bianco, E. Munzone, F. Sanguedolce, G. Vago, G. Viale, P. P. Di Fiore, S. Minucci, L. Alberghina, M. Colleoni, P. Veronesi, D. Tosoni and S. Pece. https://www.nature.com/articles/s41467-022-30375-8


Mitochondria / IDH


Germline mutations in mitochondrial complex I reveal genetic and targetable vulnerability in IDH1-mutant acute myeloid leukaemia – Mahmoud A. Bassal, Saumya E. Samaraweera, Kelly Lim, Brooks A. Bernard, Sheree Bailey, Satinder Kaur, Paul Leo, John Toubia, Chloe Thompson-Peach, Tran Nguyen, Kyaw Ze Ya Maung, Debora A. Casolari, Diana G. Iarossi, Ilaria S. Pagani, Jason Powell, Stuart Pitson, Siria Natera, Ute Roessner, Ian D. Lewis, Anna L. Brown, Daniel G. Tenen, Nirmal Robinson, David M. Ross, Ravindra Majeti, Thomas J. Gonda, Daniel Thomas and Richard J. D’Andrea. https://www.nature.com/articles/s41467-022-30223-9

IDH2-mediated regulation of the biogenesis of the oxidative phosphorylation system – Anjaneyulu Murari, Naga S. V. Goparaju, Shauna-Kay Rhooms, Kaniz F. B. Hossain, Felix G. Liang, Christian J. Garcia, Cindy Osei, Tong Liu, Hong Li, Richard N. Kitsis, Rajesh Patel, Edward Owusu-Ansah. https://www.science.org/doi/full/10.1126/sciadv.abl8716

Inducing respiratory complex I impairment elicits an increase in PGC1α in ovarian cancer – Monica De Luise, Manuela Sollazzo, Eleonora Lama, Camelia Alexandra Coadă, Licia Bressi, Maria Iorio, Beatrice Cavina, Luigi D’Angelo, Sara Milioni, Lorena Marchio, Stefano Miglietta, Sara Coluccelli, Greta Tedesco, Anna Ghelli, Silvia Lemma, Anna Myriam Perrone, Ivana Kurelac, Luisa Iommarini, Anna Maria Porcelli and Giuseppe Gasparre. https://www.nature.com/articles/s41598-022-11620-y

Mitochondrial calcium uniporter stabilization preserves energetic homeostasis during Complex I impairment – Enrique Balderas, David R. Eberhardt, Sandra Lee, John M. Pleinis, Salah Sommakia, Anthony M. Balynas, Xue Yin, Mitchell C. Parker, Colin T. Maguire, Scott Cho, Marta W. Szulik, Anna Bakhtina, Ryan D. Bia, Marisa W. Friederich, Timothy M. Locke, Johan L. K. Van Hove, Stavros G. Drakos, Yasemin Sancak, Martin Tristani-Firouzi, Sarah Franklin, Aylin R. Rodan and Dipayan Chaudhuri. https://www.nature.com/articles/s41467-022-30236-4

OXPHOS deficiencies affect peroxisome proliferation by downregulating genes controlled by the SNF1 signaling pathway [yeast] – Jean-Claude Farre , Krypton Carolino, Lou Devanneaux, Suresh Subramani. https://elifesciences.org/articles/75143

Analysis of the metabolic proteome of lung adenocarcinomas by reverse-phase protein arrays (RPPA) emphasizes mitochondria as targets for therapy – Laura Torresano, Fulvio Santacatterina, Sonia Domínguez-Zorita, Cristina Nuevo-Tapioles, Alfonso Núñez-Salgado, Pau B. Esparza-Moltó, Lucía González-Llorente, Inés Romero-Carramiñana, Cristina Núñez de Arenas, Brenda Sánchez-Garrido, Laura Nájera, Clara Salas, Mariano Provencio and José M. Cuezva. https://www.nature.com/articles/s41389-022-00400-y

Inhibition of mitochondrial complex I reverses NOTCH1-driven metabolic reprogramming in T-cell acute lymphoblastic leukemia – Natalia Baran, Alessia Lodi, Yogesh Dhungana, Shelley Herbrich, Meghan Collins, Shannon Sweeney, Renu Pandey, Anna Skwarska, Shraddha Patel, Mathieu Tremblay, Vinitha Mary Kuruvilla, Antonio Cavazos, Mecit Kaplan, Marc O. Warmoes, Diogo Troggian Veiga, Ken Furudate, Shanti Rojas-Sutterin, Andre Haman, Yves Gareau, Anne Marinier, Helen Ma, Karine Harutyunyan, May Daher, Luciana Melo Garcia, Gheath Al-Atrash, Sujan Piya, Vivian Ruvolo, Wentao Yang, Sriram Saravanan Shanmugavelandy, Ningping Feng, Jason Gay, Di Du, Jun J. Yang, Fieke W. Hoff, Marcin Kaminski, Katarzyna Tomczak, R. Eric Davis, Daniel Herranz, Adolfo Ferrando, Elias J. Jabbour, M. Emilia Di Francesco, David T. Teachey, Terzah M. Horton, Steven Kornblau, Katayoun Rezvani, Guy Sauvageau, Mihai Gagea, Michael Andreeff, Koichi Takahashi, Joseph R. Marszalek, Philip L. Lorenzi, Jiyang Yu, Stefano Tiziani, Trang Hoang and Marina Konopleva. https://www.nature.com/articles/s41467-022-30396-3


Glucose metabolism


PHGDH heterogeneity potentiates cancer cell dissemination and metastasis – Matteo Rossi, Patricia Altea-Manzano, Margherita Demicco, Ginevra Doglioni, Laura Bornes, Marina Fukano, Anke Vandekeere, Alejandro M. Cuadros, Juan Fernández-García, Carla Riera-Domingo, Cristina Jauset, Mélanie Planque, H. Furkan Alkan, David Nittner, Dongmei Zuo, Lindsay A. Broadfield, Sweta Parik, Antonino Alejandro Pane, Francesca Rizzollo, Gianmarco Rinaldi, Tao Zhang, Shao Thing Teoh, Arin B. Aurora, Panagiotis Karras, Ines Vermeire, Dorien Broekaert, Joke Van Elsen, Maximilian M. L. Knott, Martin F. Orth, Sofie Demeyer, Guy Eelen, Lacey E. Dobrolecki, Ayse Bassez, Thomas Van Brussel, Karl Sotlar, Michael T. Lewis, Harald Bartsch, Manfred Wuhrer, Peter van Veelen, Peter Carmeliet, Jan Cools, Sean J. Morrison, Jean-Christophe Marine, Diether Lambrechts, Massimiliano Mazzone, Gregory J. Hannon, Sophia Y. Lunt, Thomas G. P. Grünewald, Morag Park, Jacco van Rheenen and Sarah-Maria Fendt. https://www.nature.com/articles/s41586-022-04758-2

The hexosamine pathway and coat complex II promote malignant adaptation to nutrient scarcity – Helena Dragic, Audrey Barthelaix, VCédric Duret, Simon Le Goupil, Hadrien Laprade, Sophie Martin, Sabine Brugière, Yohann Couté, Christelle Machon, Jerome Guitton, Justine Rudewicz, Paul Hofman, Serge Lebecque, Cedric Chaveroux, Carole Ferraro-Peyret, Toufic Renno, Verge N Manié. https://www.life-science-alliance.org/content/5/7/e202101334

TKTL1 Knockdown Impairs Hypoxia-Induced Glucose-6-phosphate Dehydrogenase and Glyceraldehyde-3-phosphate Dehydrogenase Overexpression – Inês Baptista, Effrosyni Karakitsou, Jean-Baptiste Cazier, Ulrich L Günther, Silvia Marin, Marta Cascante. https://www.mdpi.com/1422-0067/23/7/3574

The lactate-NAD+ axis activates cancer-associated fibroblasts by downregulating p62 – Juan F. Linares, Tania Cid-Diaz, Angeles Duran, Marta Osrodek, Anxo Martinez-Ordoñez, Miguel Reina-Campos, Hui-Hsuan Kuo, Olivier Elemento, M. Laura Martin, Thekla Cordes, Timothy C. Thompson, Christian M. Metallo, Jorge Moscat, Maria T. Diaz-Meco. https://www.cell.com/cell-reports/fulltext/S2211-1247(22)00559-9

Metabolic flux analysis of 3D spheroids reveals significant differences in glucose metabolism from matched 2D cultures of colorectal cancer and pancreatic ductal adenocarcinoma cell lines – Tia R. Tidwell, Gro V. Røsland, Karl Johan Tronstad, Kjetil Søreide and Hanne R. Hagland. https://cancerandmetabolism.biomedcentral.com/articles/10.1186/s40170-022-00285-w

Subcellular regulation of glucose metabolism through multienzyme glucosome assemblies by EGF–ERK1/2 signaling pathways – Miji Jeon, Krishna M. Chauhan, Gregory L. Szeto, Minjoung Kyoung, Songon An. https://www.jbc.org/article/S0021-9258(22)00115-6/fulltext

Molecular basis for inhibiting human glucose transporters by exofacial inhibitors – Nan Wang, Shuo Zhang, Yafei Yuan, Hanwen Xu, Elisabeth Defossa, Hans Matter, Melissa Besenius, Volker Derdau, Matthias Dreyer, Nis Halland, Kaihui Hu He, Stefan Petry, Michael Podeschwa, Norbert Tennagels, Xin Jiang and Nieng Yan. https://www.nature.com/articles/s41467-022-30326-3


Amino acids


Cysteine is a limiting factor for glioma proliferation and survival – Victor Ruiz-Rodado, Tyrone Dowdy, Adrian Lita, Tamalee Kramp, Meili Zhang, Jinkyu Jung, Ana Dios-Esponera, Lumin Zhang, Christel C. Herold-Mende, Kevin Camphausen, Mark R. Gilbert, Mioara Larion. https://febs.onlinelibrary.wiley.com/doi/full/10.1002/1878-0261.13148

Loss of MAT2A compromises methionine metabolism and represents a vulnerability in H3K27M mutant glioma by modulating the epigenome – Brian J. Golbourn, Matthew E. Halbert, Katharine Halligan, Srinidhi Varadharajan, Brian Krug, Nneka E. Mbah, Nisha Kabir, Ann-Catherine J. Stanton, Abigail L. Locke, Stephanie M. Casillo, Yanhua Zhao, Lauren M. Sanders, Allison Cheney, Steven J. Mullett, Apeng Chen, Michelle Wassell, Anthony Andren, Jennifer Perez, Esther P. Jane, Daniel R. David Premkumar, Robert F. Koncar, Shideh Mirhadi, Lauren H. McCarl, Yue-Fang Chang, Yijen L. Wu, Taylor A. Gatesman, Andrea F. Cruz, Michal Zapotocky, Baoli Hu, Gary Kohanbash, Xiuxing Wang, Alenoush Vartanian, Michael F. Moran, Frank Lieberman, Nduka M. Amankulor, Stacy G. Wendell, Olena M. Vaske, Ashok Panigrahy, James Felker, Kelsey C. Bertrand, Claudia L. Kleinman, Jeremy N. Rich, Robert M. Friedlander, Alberto Broniscer, Costas Lyssiotis, Nada Jabado, Ian F. Pollack, Stephen C. Mack and Sameer Agnihotri. https://www.nature.com/articles/s43018-022-00348-3

Filamentous GLS1 promotes ROS-induced apoptosis upon glutamine deprivation via insufficient asparagine synthesis – Bin Jiang, Jia Zhang, Guohui Zhao, Mengjue Liu, Jielu Hu, Furong Lin, Jinyang Wang, Wentao Zhao, Huanhuan Ma, Cixiong Zhang, Caiming Wu, Luming Yao, Qingfeng Liu, Xin Chen, Yating Cao, Yi Zheng, Chensong Zhang, Aidong Han, Donghai Lin, Qinxi Li. https://www.sciencedirect.com/science/article/abs/pii/S1097276522002544?via%3Dihub

Clinical significance of plasma-free amino acids and tryptophan metabolites in patients with non-small cell lung cancer receiving PD-1 inhibitor: a pilot cohort study for developing a prognostic multivariate model – Koichi Azuma, Huihui Xiang, Tomoyuki Tagami et al. https://jitc.bmj.com/content/10/5/e004420

Cytosolic aspartate aminotransferase moonlights as a ribosome binding modulator of Gcn2 activity during oxidative stress – Robert A Crawford, Mark Peter Ashe, Simon J Hubbard, Graham D Pavitt. https://elifesciences.org/articles/73466


Lipid metabolism


Therapeutic targeting of the mevalonate–geranylgeranyl diphosphate pathway with statins overcomes chemotherapy resistance in small cell lung cancer – Chenchen Guo, Ruijie Wan, Yayi He, Shu-Hai Lin, Jiayu Cao, Ying Qiu, Tengfei Zhang, Qiqi Zhao, Yujia Niu, Yujuan Jin, Hsin-Yi Huang, Xue Wang, Li Tan, Roman K. Thomas, Hua Zhang, Luonan Chen, Kwok-Kin Wong, Liang Hu and Hongbin Ji. https://www.nature.com/articles/s43018-022-00358-1

27-hydroxycholesterol linked high cholesterol diet to lung adenocarcinoma metastasis – Xingkai Li, Hengchi Chen, Lizhen Zhang, Li Chen, Wei Wei, Shugeng Gao, Qi Xue, Yue Li, Bing Wang, Jiagen Li, Yushun Gao and Yanliang Lin. https://www.nature.com/articles/s41388-022-02285-y

Lipogenesis mediated by OGR1 regulates metabolic adaptation to acid stress in cancer cells via autophagy – Smitha Pillai, Iqbal Mahmud, Rohit Mahar, Crystal Griffith, Michael Langsen, Jonathan Nguyen, Jonathan W. Wojtkowiak, Pawel Swietach, Robert A. Gatenby, Marilyn M. Bui, Matthew E. Merritt, Patricia McDonald, Timothy J. Garrett, Robert J. Gillies. https://www.cell.com/cell-reports/fulltext/S2211-1247(22)00569-1

FASN promotes lymph node metastasis in cervical cancer via cholesterol reprogramming and lymphangiogenesis – Qiqiao Du, Pan Liu, Chunyu Zhang, Tianyu Liu, Wei Wang, Chunliang Shang, Jieyu Wu, Yuandong Liao, Yili Chen, Jiaming Huang, Hao Tan, Yunhe Zhao, Meng Xia, Junxiu Liu and Shuzhong Yao. https://www.nature.com/articles/s41419-022-04926-2

Fatty acid oxidation protects cancer cells from apoptosis by increasing mitochondrial membrane lipids – Yi-Jia Li, Johannes Francois Fahrmann, Maryam Aftabizadeh, Qianqian Zhao, Satyendra C. Tripathi, Chunyan Zhang, Yuan Yuan, David Ann, Samir Hanash, Hua Yu. https://www.cell.com/cell-reports/fulltext/S2211-1247(22)00645-3

PI(18:1/18:1) is a SCD1-derived lipokine that limits stress signaling – Maria Thürmer, André Gollowitzer, Helmut Pein, Konstantin Neukirch, Elif Gelmez, Lorenz Waltl, Natalie Wielsch, René Winkler, Konstantin Löser, Julia Grander, Madlen Hotze, Sönke Harder, Annika Döding, Martina Meßner, Fabiana Troisi, Maximilian Ardelt, Hartmut Schlüter, Johanna Pachmayr, Óscar Gutiérrez-Gutiérrez, Karl Lenhard Rudolph, Kathrin Thedieck, Ulrike Schulze-Späte, Cristina González-Estévez, Christian Kosan, Aleš Svatoš, Marcel Kwiatkowski and Andreas Koeberle. https://www.nature.com/articles/s41467-022-30374-9

Fatty acid oxidation enzyme Δ3, Δ2-enoyl-CoA isomerase 1 (ECI1) drives aggressive tumor phenotype and predicts poor clinical outcome in prostate cancer patients – Yogesh M. Bramhecha, Karl-Philippe Guérard, Étienne Audet-Walsh, Shaghayegh Rouzbeh, Ola Kassem, Erwan Pernet, Eleonora Scarlata, Lucie Hamel, Fadi Brimo, Maziar Divangahi, Armen G. Aprikian, Simone Chevalier, Vincent Giguère and Jacques Lapointe. https://www.nature.com/articles/s41388-022-02276-z


Starvation, nutrient sensing


O-GlcNAc modification of leucyl-tRNA synthetase 1 integrates leucine and glucose availability to regulate mTORC1 and the metabolic fate of leucine – Kibum Kim, Hee Chan Yoo, Byung Gyu Kim, Sulhee Kim, Yulseung Sung, Ina Yoon, Ya Chun Yu, Seung Joon Park, Jong Hyun Kim, Kyungjae Myung, Kwang Yeon Hwang, Sunghoon Kim and Jung Min Han. https://www.nature.com/articles/s41467-022-30696-8

Dysfunction of the energy sensor NFE2L1 triggers uncontrollable AMPK signaling and glucose metabolism reprogramming – Lu Qiu, Qiufang Yang, Wenshan Zhao, Yadi Xing, Peng Li, Xiaowen Zhou, Haoming Ning, Ranran Shi, Shanshan Gou, Yalan Chen, Wenjie Zhai, Yahong Wu, Guodong Li, Zhenzhen Chen, Yonggang Ren, Yanfeng Gao, Yiguo Zhang and Yuanming Qi. https://www.nature.com/articles/s41419-022-04917-3

Bcl-xL enforces a slow-cycling state necessary for survival in the nutrient-deprived microenvironment of pancreatic cancer – Yogev Sela, Jinyang Li, Shivahamy Maheswaran, Robert J Norgard, Salina Yuan, Maimon E Hubbi, Miriam Doepner, Jimmy P Xu, Elaine S Ho, Clementina Mesaros, Colin Sheehan, Grace Croley, Alexander Muir, Ian A Blair, Ophir Shalem, Chi V Dang, Ben Z Stanger. https://aacrjournals.org/cancerres/article-abstract/doi/10.1158/0008-5472.CAN-22-0431/682341

Inhibitor of glutamine metabolism V9302 promotes ROS-induced autophagic degradation of B7H3 to enhance antitumor immunity – Qian Li, Xiaofang Zhong, Weicheng Yao, Junli Yu, Chao Wang, Zongyan Li, Shengqing Lai, Fanli Qu, Xiaoyan Fu, Xiaojia Huang, Dawei Zhang, Yujie Liu, Haiyan Li. https://www.jbc.org/article/S0021-9258(22)00193-4/fulltext


mTOR / AMPK


PET-Based Radiogenomics Supports mTOR Pathway Targeting for Hepatocellular Carcinoma – Jihyun An, Minyoung Oh, Seog-Young Kim, Yoo-Jin Oh, Bora Oh, Ji-Hye Oh, Wonkyung Kim, Jin Hwa Jung, Ha Il Kim, Jae-Seung Kim, Chang Ohk Sung, Ju Hyun Shim. https://aacrjournals.org/clincancerres/article-abstract/28/9/1821/694445

The non-essential TSC complex component TBC1D7 restricts tissue mTORC1 signaling and brain and neuron growth – Sandra Schrötter, Christopher J. Yuskaitis, Michael R. MacArthur, Sarah J. Mitchell, Aaron M. Hosios, Maria Osipovich, Margaret E. Torrence, James R. Mitchell, Gerta Hoxhaj, Mustafa Sahin, Brendan D. Manning. https://www.cell.com/cell-reports/fulltext/S2211-1247(22)00595-2

Turnover of the mTOR inhibitor, DEPTOR, and downstream AKT phosphorylation in multiple myeloma cells, is dependent on ERK1-mediated phosphorylation – Mario Vega, Yu Chen, Yijiang Shi, Joseph Gera, Alan Lichtenstein. https://www.jbc.org/article/S0021-9258(22)00190-9/fulltext

Biochemical purification uncovers mammalian sterile 3 (MST3) as a new protein kinase for multifunctional protein kinases AMPK and SIK3 – Yuxiang Liu, Tao V. Wang, Yunfeng Cui, Shengxian Gao, Yi Rao. https://www.jbc.org/article/S0021-9258(22)00369-6/fulltext


Metabolism and angiogenesis


Aspartate metabolism in endothelial cells activates the mTORC1 pathway to initiate translation during angiogenesis – Roxana E. Oberkersch, Giovanna Pontarin, Matteo Astone, Marianna Spizzotin, Liaisan Arslanbaeva, Giovanni Tosi, Emiliano Panieri, Sara Ricciardi, Maria Francesca Allega, Alessia Brossa, Paolo Grumati, Benedetta Bussolati, Stefano Biffo, Saverio Tardito, Massimo M. Santoro. https://www.cell.com/developmental-cell/fulltext/S1534-5807(22)00286-6

The requirement of phosphoenolpyruvate carboxykinase 1 for angiogenesis in vitro and in vivo – Jin Yao, Xin-yuan Wu, Qing Yu, Shuo-fei Yang, Jin Yuan, Zhi-qing Zhang, Jin-song Xue, Qin Jiang, Min-bin Chen, Guan-hua Xue, Cong Cao. https://www.science.org/doi/full/10.1126/sciadv.abn6928

SLC1A1-mediated cellular and mitochondrial influx of R-2-hydroxyglutarate in vascular endothelial cells promotes tumor angiogenesis in IDH1-mutant solid tumors – Xiaomin Wang, Ziqi Chen, Jun Xu, Shuai Tang, Nan An, Lei Jiang, Yixiang Zhang, Shaoying Zhang, Qingli Zhang, Yanyan Shen, Shijie Chen, Xiaojing Lan, Ting Wang, Linhui Zhai, Siyuwei Cao, Siqi Guo, Yingluo Liu, Aiwei Bi, Yuehong Chen, Xiameng Gai, Yichen Duan, Ying Zheng, Yixian Fu, Yize Li, Liang Yuan, Linjiang Tong, Kun Mo, Mingcheng Wang, Shu-Hai Lin, Minjia Tan, Cheng Luo, Yi Chen, Jia Liu, Qiansen Zhang, Leping Li, Min Huang.. https://www.nature.com/articles/s41422-022-00650-w


Cancer Immunometabolism


Obesity modulates the immune macroenvironment associated with breast cancer development – Aleida Núñez-Ruiz, Flor Sánchez-Brena, Cynthia López-Pacheco, Naray A Acevedo-Domínguez, Gloria Soldevila.. https://journals.plos.org/plosone/article?id=10.1371/journal.pone.0266827


Miscellaneous


A pan-cancer metabolic atlas of the tumor microenvironment – Neha Rohatgi, Umesh Ghoshdastider, Probhonjon Baruah, Tanmay Kulshrestha, Anders Jacobsen Skanderup. https://www.cell.com/cell-reports/fulltext/S2211-1247(22)00567-8

Effect of Metformin vs Placebo on Invasive Disease–Free Survival in Patients With Breast Cancer, The MA.32 Randomized Clinical Trial – Pamela J. Goodwin, Bingshu E. Chen, Karen A. Gelmon, Timothy J. Whelan, Marguerite Ennis, Julie Lemieux, Jennifer A. Ligibel, Dawn L. Hershman, Ingrid A. Mayer, MD9, Timothy J. Hobday, Judith M. Bliss, Priya Rastogi, Manuela Rabaglio-Poretti, Som D. Mukherjee, John R. Mackey, Vandana G. Abramson, Conrad Oja, Robert Wesolowski, Alastair M. Thompson, Daniel W. Rea, Paul M. Stos, Lois E. Shepherd, Vuk Stambolic, Wendy R. Parulekar. https://jamanetwork.com/journals/jama/fullarticle/2792615

Targeting SLC7A11 improves efferocytosis by dendritic cells and wound healing in diabetes – Sophia Maschalidi, Parul Mehrotra, Burcu N. Keçeli, Hannah K. L. De Cleene, Kim Lecomte, Renée Van der Cruyssen, Pauline Janssen, Jonathan Pinney, Geert van Loo, Dirk Elewaut, Ann Massie, Esther Hoste and Kodi S. Ravichandran. https://www.nature.com/articles/s41586-022-04754-6

Comprehensive Analysis of Metabolic Isozyme Targets in Cancer – Michal Marczyk, Vignesh Gunasekharan, David Casadevall, Tao Qing, Julia Foldi, Raghav Sehgal, Naing Lin Shan, Kim R.M. Blenman, Tess A. O’Meara, Sheila Umlauf, Yulia V. Surovtseva, Viswanathan Muthusamy, Jesse Rinehart, Rachel J. Perry, Richard Kibbey, Christos Hatzis, Lajos Pusztai. https://aacrjournals.org/cancerres/article-abstract/82/9/1698/694711


Reviews


Transcriptional control of energy metabolism by nuclear receptors – Charlotte Scholtes and Vincent Giguère. https://www.nature.com/articles/s41580-022-00486-7

Metabolic profile of the Warburg effect as a tool for molecular prognosis and diagnosis of cancer – Gerardo M. Nava and Luis Alberto Madrigal-Perez. https://www.tandfonline.com/doi/abs/10.1080/14737159.2022.2065196?journalCode=iero20

Targeting oncometabolism to maximize immunotherapy in malignant brain tumors – Joshua D. Bernstock, Kyung-Don Kang, Neil V. Klinger, Hannah E. Olsen, Sam Gary, Stacie K. Totsch, Gelare Ghajar-Rahimi, David Segar, Eric M. Thompson, Victor Darley-Usmar, Bryan T. Mott, Luca Peruzzotti-Jametti and Gregory K. Friedman. https://www.nature.com/articles/s41388-022-02312-y

Metabolic Regulation of Epigenetic Modifications and Cell Differentiation in Cancer – Pasquale Saggese, Assunta Sellitto, Cesar A. Martinez, Giorgio Giurato, Giovanni Nassa, Francesca Rizzo, Roberta Tarallo and Claudio Scafoglio. https://www.mdpi.com/2072-6694/12/12/3788

Targeting Stearoyl-CoA Desaturase in Solid Tumors – Casie S. Kubota; Peter J. Espenshade. https://aacrjournals.org/cancerres/article-abstract/82/9/1682/694698

Metabolic tricks of cancer cells – Katerina Hönigova, Jiri Navratil, Barbora Peltanova, Hana Holcova Polanska, Martina Raudenska, Michal Masarik. https://www.sciencedirect.com/science/article/abs/pii/S0304419X22000300

The role of eIF2 phosphorylation in cell and organismal physiology: new roles for well-known actors – Xuemin Wang; Christopher G. Proud. https://portlandpress.com/biochemj/article-abstract/479/10/1059/231335

mTOR substrate phosphorylation in growth control – Stefania Battaglioni, Don Benjamin, Matthias Wälchli, Timm Maier, Michael N. Hall. https://www.cell.com/cell/fulltext/S0092-8674(22)00460-3

KEAP1-Mutant NSCLC: The Catastrophic Failure of a Cell-Protecting Hub – Stefano Scalera, Marco Mazzotta, Clelia Cortile, Eriseld Krasniqi, Ruggero De Maria, Federico Cappuzzo, Gennaro Ciliberto, Marcello Maugeri-Saccà. https://www.jto.org/article/S1556-0864(22)00166-6


Comments


Metabolic diversity drives cancer cell invasion – Sanjeethan C. Baksh and Lydia W. S. Finley. https://www.nature.com/articles/d41586-022-01301-1

Who Will Benefit From Metformin? – Joanne E. Mortimer; Victoria Seewaldt. https://jamanetwork.com/journals/jamaoncology/fullarticle/2792811

Formate for tumor progression – Annalisa M. VanHook. https://www.science.org/doi/full/10.1126/scisignal.add1844

Upcoming conferences:

PIVAC – Torino, Italy. September 22.. Conference about immunotherapy, with focus on microenvironment and metabolism. https://www.pivac22.it/

CIMUS symposium, Santiago, July 22. Organized by Rubén Nogueiras, Carlos Diéguez, Miguel López, Jose L. Labandeira, Fernando Dominguez, Diana Guallar, Ricardo Villa-Bellosta, includes several sessions about metabolism and disease https://www.cimussymposium.com/en/index.php