MetaboList – November 2023


Mitochondria, TCA


Vitamin B5 supports MYC oncogenic metabolism and tumor progression in breast cancer – Peter Kreuzaler, Paolo Inglese, Avinash Ghanate, Ersa Gjelaj, Vincen Wu, Yulia Panina, Andres Mendez-Lucas, Catherine MacLachlan, Neill Patani, Catherine B. Hubert, Helen Huang, Gina Greenidge, Oscar M. Rueda, Adam J. Taylor, Evdoxia Karali, Emine Kazanc, Amy Spicer, Alex Dexter, Wei Lin, Daria Thompson, Mariana Silva Dos Santos, Enrica Calvani, Nathalie Legrave, James K. Ellis, Wendy Greenwood, Mary Green, Emma Nye, Emma Still, CRUK Rosetta Grand Challenge Consortium, Simon Barry, Richard J. A. Goodwin, Alejandra Bruna, Carlos Caldas, James MacRae, Luiz Pedro Sório de Carvalho, George Poulogiannis, Greg McMahon, Zoltan Takats, Josephine Bunch and Mariia Yuneva. https://www.nature.com/articles/s42255-023-00915-7

Mitochondrial temperature homeostasis resists external metabolic stresses – Mügen Terzioglu, Kristo Veeroja, Toni Montonen, Teemu O. Ihalainen, Tiina S. Salminen, Paule Bénit, Pierre Rustin, Young-Tae Chang, Takeharu Nagai, Howard T. Jacobs. https://elifesciences.org/reviewed-preprints/89232

Inhibiting NR5A2 targets stemness in pancreatic cancer by disrupting SOX2/MYC signaling and restoring chemosensitivity – Quan Zheng, Jiajia Tang, Alexandra Aicher, Tony Bou Kheir, Berina Sabanovic, Preeta Ananthanarayanan, Chiara Reina, Minchun Chen, Jian-Min Gu, Bin He, Sonia Alcala, Diana Behrens, Rita T. Lawlo, Aldo Scarpa, Manuel Hidalgo, Bruno Sainz Jr., Patricia Sancho and Christopher Heeschen. https://link.springer.com/article/10.1186/s13046-023-02883-y


Glucose/lactate metabolism


Tumor-resident Lactobacillus iners confer chemoradiation resistance through lactate-induced metabolic rewiring – Lauren E. Colbert, Molly B. El Alam, Rui Wang, Tatiana Karpinets, David Lo, Erica J. Lynn, Timothy A. Harris, Jacob H. Elnaggar, Kyoko Yoshida-Court, Katarina Tomasic, Julianna K. Bronk, Julie Sammouri, Ananta V. Yanamandra, Adilene V. Olvera, Lily G. Carlin, Travis Sims, Andrea Y. Delgado Medrano, Tatiana Cisneros Napravnik, Madison O’Hara, Daniel Lin, Chike O. Abana, Hannah X. Li, Patricia J. Eifel, Anuja Jhingran, Melissa Joyner, Lilie Lin, Lois M. Ramondetta, Andrew M. Futreal, Kathleen M. Schmeler, Geena Mathew, Stephanie Dorta-Estremera, Jianhua Zhang, Xiaogang Wu, Nadim J. Ajami, Matthew Wong, Cullen Taniguchi, Joseph F. Petrosino, K. Jagannadha Sastry, Pablo C. Okhuysen, Sara A. Martinez, Lin Tan, Iqbal Mahmud, Philip L. Lorenzi, Jennifer A. Wargo, Ann H. Klopp. https://www.cell.com/cancer-cell/fulltext/S1535-6108(23)00328-8

PDK4-dependent hypercatabolism and lactate production of senescent cells promotes cancer malignancy – Xuefeng Dou, Qiang Fu, Qilai Long, Shuning Liu, Yejun Zou, Da Fu, Qixia Xu, Zhirui Jiang, Xiaohui Ren, Guilong Zhang, Xiaoling Wei, Qingfeng Li, Judith Campisi, Yuzheng Zhao and Yu Sun. https://www.nature.com/articles/s42255-023-00912-w

D-mannose induces TFE3-dependent lysosomal degradation of EGFR and inhibits the progression of NSCLC – Xue Sun, Yue Dai, Jing He, Hongchen Li, Xuhui Yang, Wenjing Dong, Xiao Xie, Mingsong Wang, Yanping Xu and Lei Lv. https://www.nature.com/articles/s41388-023-02856-7

ATM inhibition blocks glucose metabolism and amplifies the sensitivity of resistant lung cancer cell lines to oncogene driver inhibitors – Cristina Terlizzi, Viviana De Rosa, Francesca Iommelli, Antonio Pezone, Giovanna G. Altobelli, Maurizio Maddalena, Jelena Dimitrov, Caterina De Rosa, Carminia Maria Della Corte, Vittorio Enrico Avvedimento and Silvana Del Vecchio. https://cancerandmetabolism.biomedcentral.com/articles/10.1186/s40170-023-00320-4

p53-responsive CMBL reprograms glucose metabolism and suppresses cancer development by destabilizing phosphofructokinase PFKP – Yingdan Huang, Chen Xiong, Chunmeng Wang, Jun Deng, Zhixiang Zuo, Huijing Wu, Jianping Xiong, Xiaohua Wu, Hua Lu, Qian Hao, Xiang Zhou. https://www.cell.com/cell-reports/fulltext/S2211-1247(23)01438-9


Cystine / Glutathione


Autoregulatory control of mitochondrial glutathione homeostasis – Yuyang Liu, Shanshan Liu, Anju Tomar, Frederick S Yen, Gokhan Unlu, Nathalie Ropek, Ross A Weber, Ying Wang, Artem Khan, Mark Gad, Junhui Peng, Erdem Terzi, Hanan Alwaseem, Alexandra E Pagano, Søren Heissel, Henrik Molina, Benjamin Allwein, Timothy C Kenny, Richard L Possemato, Li Zhao, Richard K Hite, Ekaterina V Vinogradova, Sheref S Mansy, Kıvanç Birsoy. https://www.science.org/doi/10.1126/science.adf4154


Amino acids


Arginine reprograms metabolism in liver cancer via RBM39 – Dirk Mossmann, Christoph Müller, Sujin Park, Uwe Sauer, Markus H. Heim, Michael N. Hall. https://www.cell.com/cell/fulltext/S0092-8674(23)01032-2

Tyrosine catabolism enhances genotoxic chemotherapy by suppressing translesion DNA synthesis in epithelial ovarian cancer – Jie Li, Cuimiao Zheng, Qiuwen Mai, Xi Huang, Wenfeng Pan, Jingyi Lu, Zhengfan Chen, Suman Zhang, Chunyu Zhang, Hua Huang, Yangyang Chen, Hongbo Guo, Zhenyin Wu, Chunnuan Deng, Yiting Jiang, Bo Li, Junxiu Liu, Shuzhong Yao, Chaoyun Pan. https://www.cell.com/cell-metabolism/fulltext/S1550-4131(23)00371-6

Inhibition of the proline metabolism rate-limiting enzyme P5CS allows proliferation of glutamine-restricted cancer cells – Samantha J. Linder, Tiziano Bernasocchi, Bárbara Martínez-Pastor, Kelly D. Sullivan, Matthew D. Galbraith, Caroline A. Lewis, Christina M. Ferrer, Ruben Boon, Giorgia G. Silveira, Hyo Min Cho, Charles Vidoudez, Stuti Shroff, Joao P. Oliveira-Costa, Kenneth N. Ross, Rami Massri, Yusuke Matoba, Eugene Kim, Bo R. Rueda, Shannon L. Stott, Eyal Gottlieb, Joaquin M. Espinosa and Raul Mostoslavsky


Lipid metabolism


Oncogenic KRASG12D Reprograms Lipid Metabolism by Upregulating SLC25A1 to Drive Pancreatic Tumorigenesis – Ruowen Zhang, Xiaogang Peng, James Xianxing Du, Rebecca Boohaker, Igor L. Estevao, Brian I. Grajeda, Marc B. Cox, Igor C. Almeida, Weiqin Lu. https://aacrjournals.org/cancerres/article-abstract/83/22/3739/729993

CD36 maintains lipid homeostasis via selective uptake of monounsaturated fatty acids during matrix detachment and tumor progression – Alexander R. Terry, Veronique Nogueira, Hyunsoo Rho, Gopalakrishnan Ramakrishnan, Jing Li, Soeun Kang, Koralege C. Pathmasiri, Sameer Ahmed Bhat, Liping Jiang, Shafi Kuchay, Stephanie M. Cologna, Nissim Hay. https://www.cell.com/cell-metabolism/fulltext/S1550-4131(23)00368-6

Inhibiting the endoplasmic reticulum stress response enhances the effect of doxorubicin by altering the lipid metabolism of liver cancer cells – Maria Kopsida, Ada Lerma Clavero, Jaafar Khaled, David Balgoma, Clara Luna-Marco, Azazul Chowdhury, Sofi Sennefelt Nyman, Fredrik Rorsman, CharlotteEbeling Barbier, Peter Bergsten, Hans Lennernäs, Mikael Hedeland, Femke Heindryck. https://www.sciencedirect.com/science/article/pii/S2212877823001801


Nucleotides


IL-6/JAK2-dependent G6PD phosphorylation promotes nucleotide synthesis and supports tumor growth – Xuemei Qiu, Hongping Ye, Xiaofei Li, Dan Li, Lu Jiang, Rui Liu, Zhe Zhao, Dan He. https://www.sciencedirect.com/science/article/pii/S2212877823001709


mTOR/nutrient sensing


Nutrient-regulated control of lysosome function by signaling lipid conversion – Michael Ebner, Dmytro Puchkov, Orestes López-Ortega, Pathma Muthukottiappan,Yanwei Su, Christopher Schmied, Silke Zillmann, Iryna Nikonenko, Jochen Koddebusch, Gillian L. Dornan, Max T. Lucht, Vonda Koka, Wonyul Jang, Philipp Alexander Koch, Alexander Wallroth, Martin Lehmann, Britta Brügger, Mario Pende, Dominic Winter, Volker Haucke. https://www.cell.com/cell/fulltext/S0092-8674(23)01081-4

PRMT1 orchestrates with SAMTOR to govern mTORC1 methionine sensing via Arg-methylation of NPRL2 – Cong Jiang, Jing Liu, Shaohui He, Wei Xu, Runzhi Huang, Weijuan Pan, Xiaolong Li, Xiaoming Dai, Jianping Guo, Tao Zhang, Hiroyuki Inuzuka, Ping Wang, John M. Asara, Jianru Xiao, Wenyi Wei. https://www.sciencedirect.com/science/article/abs/pii/S1550413123004114

PI3K-C2β limits mTORC1 signaling and angiogenic growth – Piotr Kobialka, Judith Llena, Nerea Deleyto-Seldas, Margalida Munar-Gelabert, Jose A Dengra, Pilar Villacampa, Alba Albinyà-Pedrós, Laia Muixi, Jorge Andrade, Hielke van Splunder, Ana Angulo-Urarte, Michael Potente, Joaquim Grego-Bessa, Sandra D Castillo, Bart Vanhaesebroeck, Alejo Efeyan, Mariona Graupera. https://www.science.org/doi/10.1126/scisignal.adg1913


Hypoxia


Hypoxia activates SREBP2 through Golgi disassembly in bone marrow‐derived monocytes for enhanced tumor growth – Ryuichi Nakahara, Sho Aki, Maki Sugaya, Haruka Hirose, Miki Kato, Keisuke Maeda, Daichi M Sakamoto, Yasuhiro Kojima, Miyuki Nishida, Ritsuko Ando, Masashi Muramatsu, Melvin Pan, Rika Tsuchida, Yoshihiro Matsumura, Hideyuki Yanai, Hiroshi Takano, Ryoji Yao, Shinsuke Sando, Masabumi Shibuya, Juro Sakai, Tatsuhiko Kodama, Hiroyasu Kidoya, Teppei Shimamura, Tsuyoshi Osawa. https://www.embopress.org/doi/full/10.15252/embj.2023114032

Thiol reductive stress activates the hypoxia response pathway – Ravi, Ajay Kumar, Shalmoli Bhattacharyya, Jogender Singh. https://www.embopress.org/doi/abs/10.15252/embj.2023114093


Cancer Immunometabolism


A Metabolic Gene Survey Pinpoints Fucosylation as a Key Pathway Underlying the Suppressive Function of Regulatory T Cells in Cancer – Sotiria Pinioti, Himal Sharma, Nina C. Flerin, Qian Yu, Amalia Tzoumpa, Sarah Trusso Cafarello, Elien De Bousser, Nico Callewaert, Guillaume Oldenhove, Susan Schlenner, Bernard Thienpont, Abhishek D. Garg, Mario Di Matteo, Massimiliano Mazzone. https://aacrjournals.org/cancerimmunolres/article-abstract/doi/10.1158/2326-6066.CIR-22-0606/

T-cell dysfunction by pseudohypoxia and autocrine purinergic signaling in chronic lymphocytic leukemia – Chiara Montironi, Chaja F. Jacobs, Gaspard Cretenet, Fleur S. Peters, Bauke V. Schomakers, Michel van Weeghel, Arnon P. Kater, Helga Simon-Molas, Eric Eldering. https://ashpublications.org/bloodadvances/article/7/21/6540/497366/T-cell-dysfunction-by-pseudohypoxia-and-autocrine

Label-free single-cell live imaging reveals fast metabolic switch in T lymphocytes – Noémie Paillon, Thi Phuong Lien Ung, Stéphanie Dogniaux, Chiara Stringari, and Claire Hivroz. https://www.molbiolcell.org/doi/abs/10.1091/mbc.E23-01-0009

Inflammatory macrophages reprogram to immunosuppression by reducing mitochondrial translation – Marlies Cortés, Agnese Brischetto, M. C. Martinez-Campanario, Chiara Ninfali, Verónica Domínguez, Sara Fernández, Raquel Celis, Anna Esteve-Codina, Juan J. Lozano, Julia Sidorova, Gloria Garrabou, Anna-Maria Siegert, Carlos Enrich, Belén Pintado, Manuel Morales-Ruiz, Pedro Castro, Juan D. Cañete and Antonio Postigo. https://www.nature.com/articles/s41467-023-42277-4

Dietary fructose-mediated adipocyte metabolism drives antitumor CD8+ T cell responses – Yuerong Zhang,  Xiaoyan Yu, Rujuan Bao, Haiyan Huang,  Chuanjia Gu,  Qianming Lv, Qiaoqiao Han, Xian Du, Xu-Yun Zhao, Youqiong Ye, Ren Zhao, Jiayuan Sun, Qiang Zou. https://www.cell.com/cell-metabolism/fulltext/S1550-4131(23)00367-4

The glucose transporter 2 regulates CD8+ T cell function via environment sensing – Hongmei Fu, Juho Vuononvirta, Silvia Fanti, Fabrizia Bonacina, Antonio D’Amati, Guosu Wang, Thanushiyan Poobalasingam, Maria Fankhaenel, Davide Lucchesi, Rachel Coleby, David Tarussio, Bernard Thorens, Robert J. Hearnden, M. Paula Longhi, Paul Grevitt, Madeeha H. Sheikh, Egle Solito, Susana A. Godinho, Michele Bombardieri, David M. Smith, Dianne Cooper, Asif J. Iqbal, Jeffrey C. Rathmell, Samuel Schaefer, Valle Morales, Katiuscia Bianchi, Giuseppe Danilo Norata and Federica M. Marelli-Berg. https://www.nature.com/articles/s42255-023-00913-9


Diet, systemic metabolism and cachexia


High dietary fructose promotes hepatocellular carcinoma progression by enhancing O-GlcNAcylation via microbiota-derived acetate – Peng Zhou, Wen-yi Chang, De-ao Gong, Jie Xia, Wei Chen, Lu-yi Huang, Rui Liu, Yi Liu, Chang Chen, Kai Wang, Ni Tang, Ai-long Huang. https://www.cell.com/cell-metabolism/fulltext/S1550-4131(23)00340-6

Endothelial Notch1 signaling in white adipose tissue promotes cancer cachexia – Jacqueline Taylor, Leonie Uhl, Iris Moll, Sana Safatul Hasan, Lena Wiedmann, Jakob Morgenstern, Benedetto Daniele Giaimo, Tobias Friedrich, Elisenda Alsina-Sanchis, Francesca De Angelis Rigotti, Ronja Mülfarth, Sarah Kaltenbach, Darius Schenk, Felix Nickel, Thomas Fleming, David Sprinzak, Carolin Mogler, Thomas Korff, Adrian T. Billeter, Beat P. Müller-Stich, Mauricio Berriel Diaz, Tilman Borggrefe, Stephan Herzig, Maria Rohm, Juan Rodriguez-Vita and Andreas Fischer. https://www.nature.com/articles/s43018-023-00622-y

The effects of chemotherapy on resting energy expenditure, body composition, and cancer-related fatigue in women with breast cancer: a prospective cohort study – Timia Van Soom, Wiebren Tjalma, Konstantinos Papadimitriou, Nick Gebruers and Eric van Breda. https://cancerandmetabolism.biomedcentral.com/articles/10.1186/s40170-023-00322-2


Miscellaneous


The adaptive antioxidant response during fasting-induced muscle atrophy is oppositely regulated by ZEB1 and ZEB2 – Chiara Ninfali, Marlies Cortés, M. C. Martínez-Campanario, Verónica Domínguez, Lu Han, Ester Tobías, Anna Esteve-Codina, Carlos Enrich, Belén Pintado, Gloria Garrabou, and Antonio Postigo. https://www.pnas.org/doi/10.1073/pnas.2301120120

Reducing branched-chain amino acids improves cardiac stress response in mice by decreasing histone H3K23 propionylation – Zhi Yang, Minzhen He, Julianne Austin, Danish Sayed, and Maha Abdellatif. https://www.jci.org/articles/view/169399

Autofluorescence imaging of endogenous metabolic cofactors in response to cytokine stimulation of classically activated macrophages – Shelby N. Bess, Matthew J. Igoe, Abby C. Denison and Timothy J. Muldoon. https://cancerandmetabolism.biomedcentral.com/articles/10.1186/s40170-023-00325-z

Metabolic dependencies of metastasis-initiating cells in female breast cancer – Megan Young, Laurent Beziaud, Pierre Dessen, Angela Madurga Alonso, Albert Santamaria-Martínez and Joerg Huelsken. https://www.nature.com/articles/s41467-023-42748-8


Reviews


Metabolic pathway analysis using stable isotopes in patients with cancer – Caroline R. Bartman, Brandon Faubert, Joshua D. Rabinowitz and Ralph J. DeBerardinis. https://www.nature.com/articles/s41568-023-00632-z

The molecular basis of nutrient sensing and signalling by mTORC1 in metabolism regulation and disease – Claire Goul, Roberta Peruzzo and Roberto Zoncu. https://www.nature.com/articles/s41580-023-00641-8

Metabolic control of cancer metastasis: role of amino acids at secondary organ sites – Breelyn Karno, Deanna N. Edwards and Jin Chen. https://www.nature.com/articles/s41388-023-02868-3

Regulatory mechanisms of one-carbon metabolism enzymes – Boryana Petrova, Adam G. Maynard, Peng Wang, Naama Kanarek. https://www.jbc.org/article/S0021-9258(23)02485-7

The immunometabolic ecosystem in cancer – Glenn R. Bantug and Christoph Hess. https://www.nature.com/articles/s41590-023-01675-y


Comments/other


Lysosomal cystine: an unexpected alarm bell for cysteine scarcity – Fabio Hecht, Isaac S. Harris. https://www.cell.com/trends/cell-biology/fulltext/S0962-8924(23)00213-1

mTOR inhibits starvation-induced nuclear export of the proteasome – Kim Baumann. https://www.nature.com/articles/s41580-023-00678-9

Endothelium directs fat remodeling in cachexia – Brittany R. Counts and Teresa A. Zimmers. https://www.nature.com/articles/s43018-023-00644-6

The Warburg effect: a signature of mitochondrial overload – Yahui Wang, Gary J. Patti. https://www.cell.com/trends/cell-biology/fulltext/S0962-8924(23)00070-3

Saving ribosomal proteins for later – Joshua J. Black & Rachel Green. https://www.nature.com/articles/s41556-023-01251-4

O-GlcNAc regulates YTHDF1 and YTHDF3 activity – Mary W. N. Burns and Jennifer J. Kohler. https://www.nature.com/articles/s41556-023-01275-w

Mitochondrial control of antigen presentation in cancer cells – Ruth Soler-Agesta, Alberto Anel, Lorenzo Galluzzi. https://www.cell.com/cancer-cell/fulltext/S1535-6108(23)00357-4

A metabolic signaling role for arginine in liver cancer – David Sokolov and Lucas B. Sullivan. https://academic.oup.com/lifemeta/advance-article/doi/10.1093

MOF moves into mitochondria – Natalie Niemi. https://www.nature.com/articles/s42255-023-00892-x

A century of the Warburg effect – Craig B. Thompson, Karen H. Vousden, Randall S. Johnson, Willem H. Koppenol, Helmut Sies, Zhimin Lu, Lydia W. S. Finley, Christian Frezza, Jiyeon Kim, Zeping Hu and Caroline R. Bartman. https://www.nature.com/articles/s42255-023-00927-3

Leave a comment