MetaboList – January 2024


Amino acids


The extracellular matrix supports breast cancer cell growth under amino acid starvation by promoting tyrosine catabolism – Mona Nazemi, Bian Yanes, Montserrat Llanses Martinez, Heather J. Walker, Khoa Pham, Mark O. Collins, Frederic Bard, Elena Rainero. https://journals.plos.org/plosbiology/article?id=10.1371/journal.pbio.3002406

Targeting pancreatic cancer metabolic dependencies through glutamine antagonism – Joel Encarnación-Rosado, Albert S. W. Sohn, Douglas E. Biancur, Elaine Y. Lin, Victoria Osorio-Vasquez, Tori Rodrick, Diana González-Baerga, Ende Zhao, Yumi Yokoyama, Diane M. Simeone, Drew R. Jones, Seth J. Parker, Robert Wild and Alec C. Kimmelman. https://www.nature.com/articles/s43018-023-00647-3

Glutamine mimicry suppresses tumor progression through asparagine metabolism in pancreatic ductal adenocarcinoma – Maria Victoria Recouvreux, Shea F. Grenier, Yijuan Zhang, Edgar Esparza, Guillem Lambies, Cheska Marie Galapate, Swetha Maganti, Karen Duong-Polk, Deepika Bhullar, Razia Naeem, David A. Scott, Andrew M. Lowy, Hervé Tiriac and Cosimo Commisso. https://www.nature.com/articles/s43018-023-00649-1

FOXO3a-regulated arginine metabolic plasticity adaptively promotes esophageal cancer proliferation and metastasis – Wenbo Sun, Hengyuan Kou, Yao Fang, Fan Xu, Zhi Xu, Xiumei Wang, Rong Yin, Qin Zhang, Qin Jiang and Yong Xu. https://www.nature.com/articles/s41388-023-02906-0

Blocking methionine catabolism induces senescence and confers vulnerability to GSK3 inhibition in liver cancer – Fuming Li, Pingyu Liu, Wen Mi, Liucheng Li, Nicole M. Anderson, Nicholas P. Lesner, Michelle Burrows, Jacqueline Plesset, Ariana Majer, Guanlin Wang, Jinyang Li, Lingzhi Zhu, Brian Keith and M. Celeste Simon. https://www.nature.com/articles/s43018-023-00671-3

Hypoxia-inducible factor induces cysteine dioxygenase and promotes cysteine homeostasis in Caenorhabditis elegans – Kurt Warnhoff, Sushila Bhattacharya, Jennifer Snoozy, Peter C. Breen, Gary Ruvkun. https://elifesciences.org/reviewed-preprints/89173


Glucose metabolism


An ERK5-PFKFB3 axis regulates glycolysis and represents a therapeutic vulnerability in pediatric diffuse midline glioma – Stephanie M. Casillo, Taylor A. Gatesman, Akanksha Chilukuri, Srinidhi Varadharajan, Brenden J. Johnson, Daniel R. David Premkumar, Esther P. Jane, Tritan J. Plute, Robert F. Koncar, Ann-Catherine J. Stanton, Carlos A.O. Biagi-Junior, Callie S. Barber, Matthew E. Halbert, Brian J. Golbourn, Katharine Halligan, Andrea F. Cruz, Neveen M. Mansi, Allison Cheney, Steven J. Mullett, Clinton Van’t Land, Jennifer L. Perez, Max I. Myers, Nishant Agrawal, Joshua J. Michel, Yue-Fang Chang, Olena M. Vaske, Antony MichaelRaj, Frank S. Lieberman, James Felker, Sruti Shiva, Kelsey C. Bertrand, Nduka Amankulor, Costas G. Hadjipanayis, Kalil G. Abdullah, Pascal O. Zinn, Robert M. Friedlander, Taylor J. Abel, Javad Nazarian, Sriram Venneti, Mariella G. Filbin, Stacy L. Gelhaus, Stephen C. Mack, Ian F. Pollack, Sameer Agnihotri. https://www.cell.com/cell-reports/fulltext/S2211-1247(23)01569-3

Preneoplastic cells switch to Warburg metabolism from their inception exposing multiple vulnerabilities for targeted elimination – Henna Myllymäki, Lisa Kelly, Abigail M. Elliot, Roderick N. Carter, Jeanette Astorga Johansson, Kai Yee Chang, Justyna Cholewa-Waclaw, Nicholas M. Morton and Yi Feng. https://www.nature.com/articles/s41389-024-00507-4

AKT1 phosphorylation of cytoplasmic ME2 induces a metabolic switch to glycolysis for tumorigenesis – Taiqi Chen, Siyi Xie, Jie Cheng, Qiao Zhao, Hong Wu, Peng Jiang and Wenjing Du. https://www.nature.com/articles/s41467-024-44772-8


Lipid metabolism


Metabolomic Rewiring Promotes Endocrine Therapy Resistance in Breast Cancer – Songyeon Ahn, Jun Hyoung Park, Sandra L. Grimm, Danthasinghe Waduge Badrajee Piyarathna, Tagari Samanta, Vasanta Putluri, Dereck Mezquita, Suzanne A.W. Fuqua, Nagireddy Putluri, Cristian Coarfa, Benny Abraham Kaipparettu. https://aacrjournals.org/cancerres/article-abstract/84/2/291/733113


Nucleotides


GTP Signaling Links Metabolism, DNA Repair, and Responses to Genotoxic Stress – Weihua Zhou, Zitong Zhao, Angelica Lin, John Z. Yang, Jie Xu, Kari Wilder-Romans, Annabel Yang, Jing Li, Sumeet Solanki, Jennifer M. Speth, Natalie Walker, Andrew J. Scott, Lu Wang, Bo Wen, Anthony Andren, Li Zhang, Ayesha U. Kothari, Yangyang Yao, Erik R. Peterson, Navyateja Korimerla, Christian K. Werner, Alexander Ullrich, Jessica Liang, Janna Jacobson, Sravya Palavalasa, Alexandra M. O’Brien, Ameer L. Elaimy, Sean P. Ferris, Shuang G. Zhao, Jann N. Sarkaria, Balázs Győrffy, Shuqun Zhang, Wajd N. Al-Holou, Yoshie Umemura, Meredith A. Morgan, Theodore S. Lawrence, Costas A. Lyssiotis, Marc Peters-Golden, Yatrik M. Shah, Daniel R. Wahl. https://aacrjournals.org/cancerdiscovery/article-abstract/14/1/158/732569


Mitochondria


Systems-level analyses dissociate genetic regulators of reactive oxygen species and energy production – Neal K. Bennett, Megan Lee, Adam L. Orr, and Ken Nakamura. https://www.pnas.org/doi/10.1073/pnas.2307904121

Electron transport chain and mTOR inhibition synergistically decrease CD40 signaling and counteract venetoclax resistance in chronic lymphocytic leukemia – Zhenghao Chen, Gaspard Cretenet, Valeria Carnazzo, Helga Simon-Molas, Arnon P. Kater, Gerritje J. W. van der Windt, Eric Eldering. https://haematologica.org/article/view/haematol.2023.282760

SIRT5-mediated ME2 desuccinylation promotes cancer growth by enhancing mitochondrial respiration – Peng Teng, Kaisa Cui, Surui Yao, Bojian Fei, Feng Ling, Chaoqun Li and Zhaohui Huang. https://www.nature.com/articles/s41418-023-01240-y

α-Ketoglutarate supplementation and NAD+ modulation enhance metabolic rewiring and radiosensitization in SLC25A1 inhibited cancer cells – Kexu Xiang, Mikhail Kunin, Safa Larafa, Maike Busch, Nicole Dünker, Verena Jendrossek and Johann Matschke. https://www.nature.com/articles/s41420-024-01805-x

Phosphate starvation signaling increases mitochondrial membrane potential through respiration-independent mechanisms – Yeyun Ouyang, Mi-Young Jeong … Jared Rutter. https://elifesciences.org/articles/84282


Starvation / AMPK


Glucose Deprivation Promotes Pseudohypoxia and Dedifferentiation in Lung Adenocarcinoma – Pasquale Saggese, Aparamita Pandey, Martín Alcaraz Jr, Eileen Fung, Abbie Hall, Jane Yanagawa, Erika F. Rodriguez, Tristan R. Grogan, Giorgio Giurato, Giovanni Nassa, Annamaria Salvati, Orian S. Shirihai, Alessandro Weisz, Steven M. Dubinett, Claudio Scafoglio. https://aacrjournals.org/cancerres/article/84/2/305/733107

Glucose stress causes mRNA retention in nuclear Nab2 condensates [yeast] – Stephanie Heinrich, Maria Hondele, Désirée Marchand, Carina Patrizia Derrer, Mostafa Zedan, Alexandra Oswald, Liliana Malinovska, Federico Uliana, Sarah Khawaja, Roberta Mancini, David Grunwald, Karsten Weis. https://www.cell.com/cell-reports/fulltext/S2211-1247(23)01605-4

A beneficial adaptive role for CHOP in driving cell fate selection during ER stress – Kaihua Liu, Chaoxian Zhao, Reed C Adajar, Diane DeZwaan-McCabe, D Thomas Rutkowski. https://www.embopress.org/doi/full/10.1038/s44319-023-00026-0

The BHLHE40‒PPM1F‒AMPK pathway regulates energy metabolism and is associated with the aggressiveness of endometrial cancer – Kazuo Asanoma, Hiroshi Yagi, Ichiro Onoyama, Lin Cui, Emiko Hori, Minoru Kawakami, Shoji Maenohara, Kazuhisa Hachisuga, Hiroshi Tomonobe, Keisuke Kodama, Masafumi Yasunaga, Tatsuhiro Ohgami, Kaoru Okugawa, Hideaki Yahata, Hiroyuki Kitao, Kiyoko Kato. https://www.jbc.org/article/S0021-9258(24)00071-1/fulltext

The yeast AMP-activated protein kinase Snf1 phosphorylates the inositol polyphosphate kinase Kcs1 – Sham Sunder, Joshua S. Bauman, Stuart J. Decker, Alexandra R. Lifton, Anuj Kumar. https://www.jbc.org/article/S0021-9258(24)00033-4/fulltext


Nutrient sensing / mTOR


Disruption of lysosomal nutrient sensing scaffold hyperactivates mTORC1 contributing to pathogenesis of a fatal neurodegenerative disease – Maria B. Bagh, Abhilash P. Appu, Tamal Sadhukhan, Avisek Mondal, Nisha Plavelil, Mahadevan Raghavankutty, Ajayan M. Supran, Sriparna Sadhukhan, Aiyi Liu, Anil B. Mukherjee. https://www.jbc.org/article/S0021-9258(24)00017-6/fulltext

Independent activation of CREB3L2 by glucose fills a regulatory gap in mouse β-cells by co-ordinating insulin biosynthesis with secretory granule formation – Nancy Sue, Le May Thai, Atsushi Saito, Cierra K. Boyer, Ashleigh M. Fordham, Chenxu Yan, Aimee Davenport, Jiang Tao, Mohammed Bensellam, James Cantley, Yan-Chuan Shi, Samuel B. Stephens, Kazunori Imaizumi, Trevor J. Biden. https://www.sciencedirect.com/science/article/pii/S2212877823001795

Pib2 is a cysteine sensor involved in TORC1 activation in Saccharomyces cerevisiae – Qingzhong Zeng, Yasuhiro Araki, Takeshi Noda. https://www.cell.com/cell-reports/fulltext/S2211-1247(23)01611-X

MORG1 limits mTORC1 signaling by inhibiting Rag GTPases – Yakubu Princely Abudu, Athanasios Kournou. https://www.cell.com/molecular-cell/fulltext/S1097-2765(23)00968-1

TFEB drives mTORC1 hyperactivation and kidney disease in Tuberous Sclerosis Complex – Nicola Alesi, Damir Khabibullin, Dean M. Rosenthal, Elie W. Akl, Pieter M. Cory, Michel Alchoueiry, Samer Salem, Melissa Daou, William F. Gibbons, Jennifer A. Chen, Long Zhang, Harilaos Filippakis, Laura Graciotti, Caterina Miceli, Jlenia Monfregola, Claudia Vilardo, Manrico Morroni, Chiara Di Malta, Gennaro Napolitano, Andrea Ballabio and Elizabeth P. Henske. https://www.nature.com/articles/s41467-023-44229-4


Diet, systemic metabolism and cachexia


Leukemia inhibitory factor suppresses hepatic de novo lipogenesis and induces cachexia in mice – Xue Yang, Jianming Wang, Chun-Yuan Chang, Fan Zhou, Juan Liu, Huiting Xu, Maria Ibrahim, Maria Gomez, Grace L. Guo, Hao Liu, Wei-Xing Zong, Fredric E. Wondisford, Xiaoyang Su, Eileen White, Zhaohui Feng and Wenwei Hu. https://www.nature.com/articles/s41467-024-44924-w


Miscellaneous


Metabolic reprogramming by histone deacetylase inhibition preferentially targets NRF2-activated tumors – Dimitris Karagiannis, Warren Wu, Albert Li,  Makiko Hayashi, Xiao Chen, Michaela Yip, Vaibhav Mangipudy, Xinjing Xu,  Francisco J. Sánchez-Rivera, Yadira M. Soto-Feliciano, Jiangbin Ye, Thales Papagiannakopoulos, Chao Lu. https://www.cell.com/cell-reports/fulltext/S2211-1247(23)01640-6


Reviews


The bidirectional relationship between metabolism and cell cycle control – Frances F. Diehl, Kiera M. Sapp, Matthew G. Vander Heiden. https://www.cell.com/trends/cell-biology/fulltext/S0962-8924(23)00110-1

Molecular and cellular mechanisms underlying the failure of mitochondrial metabolism drugs in cancer clinical trials – Karthik Vasan and Navdeep S. Chandel. https://www.jci.org/articles/view/176736

Contrasting views on the role of AMPK in autophagy – Do-Hyung Kim. https://onlinelibrary.wiley.com/doi/10.1002/bies.202300211

Modelling cancer metabolism in vitro: current improvements and future challenges – Helena Dragic, Cedric Chaveroux, Erika Cosset, Serge N. Manie. https://febs.onlinelibrary.wiley.com/doi/10.1111/febs.16704

Glucose metabolism in B cell malignancies: a focus on glycolysis branching pathways – Helga Simon-Molas, Rosita Del Prete, Anna Kabanova. https://febs.onlinelibrary.wiley.com/doi/10.1002/1878-0261.13570

Mechanisms controlling cellular and systemic iron homeostasis – Bruno Galy, Marcus Conrad and Martina Muckenthaler. https://www.nature.com/articles/s41580-023-00648-1

Metabolic heterogeneity in cancer – Margherita Demicco, Xiao-Zheng Liu, Katharina Leithner and Sarah-Maria Fendt. https://www.nature.com/articles/s42255-023-00963-z

Amino acid metabolism, redox balance and epigenetic regulation in cancer – Xiang Li, Hong-Sheng Zhang. https://febs.onlinelibrary.wiley.com/doi/full/10.1111/febs.16803?campaign=woletoc

Hurdle or thruster: Glucose metabolism of T cells in anti-tumour immunity – Sirui Zhang, Xiaozhen Zhang, Hanshen Yang, Tingbo Liang, Xueli Bai. https://www.sciencedirect.com/science/article/pii/S0304419X23001713

Targeting lipid metabolism in cancer metastasis – Gloria Pascual, Blanca Majem, Salvador Aznar Benitah. https://www.sciencedirect.com/science/article/abs/pii/S0304419X23002007

Nutrition and dietary restrictions in cancer prevention – Amrendra Mishra, Giacomo Giuliani, Valter D. Longo. https://www.sciencedirect.com/science/article/abs/pii/S0304419X23002123

Lipids as mediators of cancer progression and metastasis – Felix C. E. Vogel, Adriano B. Chaves-Filho and Almut Schulze. https://www.nature.com/articles/s43018-023-00702-z

Regulation of antioxidants in cancer – Fabio Hecht, Marco Zocchi, Fatemeh Alimohammadi, Isaac S. Harris. https://www.cell.com/molecular-cell/fulltext/S1097-2765(23)00917-6


Comments


Glutamine analogs for pancreatic cancer therapy – Nada Y. Kalaany. https://www.nature.com/articles/s43018-023-00678-w

DON of Hope: Starving Pancreatic Cancer by Glutamine Antagonism – Ray Pillai, Thales Papagiannakopoulous. https://aacrjournals.org/cancerres/article-abstract/84/3/349/733842

Exploring the Role of Hyperinsulinemia in Obesity-Associated Tumor Development – Ericka Vélez-Bonet, Kristyn Gumpper-Fedus, Zobeida Cruz-Monserrate. https://aacrjournals.org/cancerres/article-abstract/84/3/351/733844

Fructose sweetens antitumor responses – Stephanie Houston. https://www.nature.com/articles/s41590-023-01730-8


ISCaM 2024 meeting   –  1th Annual Meeting – July 3-5, 2024. BEL Congress Center, Brussels (BE)

https://iscam2024.org/

Leave a comment