Centro Andaluz de Biología Molecular y Medicina Regenerativa

Metabolism and Cell Signaling

Research Lines

Glutamine metabolism in cancer cells

Glutamine, the most abundant amino acid in the blood, plays a particularly important role in cell growth and metabolism as a precursor for αKG of the tricarboxylic acid cycle, for nucleotides and for other amino acids. The importance of glutamine as a nutrient is further underscored by the observation that cancer cells are particularly dependent on this amino acid. Most cancer patients develop glutamine depletion which ultimately leads to cachexia. Glutamine is metabolized through glutaminolysis, catalyzed by glutaminase (GLS) and glutamate dehydrogenase (GDH), to produce αKG. GLS is regulated at the expression level by the oncogene c-myc, and GLS activity correlates with tumor growth (Tennant, Duran & Gottlieb, Nature Rev. Cancer 2010). The essential amino acid leucine directly binds and activates GDH to stimulate deamination of glutamate and thereby αKG production. Thus, leucine and glutamine, two of the most important amino acids in energy metabolism and nutrient signaling, cooperate in stimulating glutaminolysis. Our recent findings showed that glutamine in combination with leucine activates mTOR pathway by enhancing glutaminolysis and αKG production (Durán et al., Cell Cycle 2012).

mTOR signaling and glutamine metabolism

mTOR is a serine/threonine kinase highly conserved from yeast to humans. In mammalian cells, mTOR integrates several stimuli to regulate cell growth, metabolism and aging. mTOR forms two functionally and structurally distinct complexes termed mTORC1 and mTORC2. mTORC1 regulates protein synthesis, ribosome biogenesis, nutrient uptake and autophagy in response to growth factors, amino acids, and cellular energy. The Rag GTPases, activate mTORC1 in response to amino acids, especially leucine (Duran & Hall, EMBO Rep. 2012). Addition of amino acids allows the Rag heterodimer to bind and thereby recruit mTORC1 to the lysosome, where mTORC1 is activated. Our results showed that glutaminolysis is a critical step in the Rag-dependent recruitment of mTORC1 to the lysosome. Inhibition of glutaminolysis prevents the Rag-dependent lysosomal translocation and subsequent activation of mTORC1. Conversely, enhanced glutaminolysis or a cell permeable αKG analogue stimulates lysosomal translocation and activation of mTORC1. Finally, cell growth and autophagy, two processes controlled by mTORC1, are regulated by glutaminolysis. Thus, mTORC1 senses and is activated by glutamine and leucine via glutaminolysis and αKG production upstream of Rag (Duran et al., Mol. Cell 2012).

Prolyl hydroxylases as regulators of cell metabolism

Our findings position αKG as an intracellular messenger for nutrients availability and suggest new interesting questions, for instance, how is αKG sensed by the cell? On this lead, our studies also suggest that prolyl hydroxylases (PHD) are the αKG sensors that mediate the activation of mTORC1 by glutaminolysis (Duran et al., Oncogene 2013). PHDs are considered the oxygen sensors of cells. However, under normoxic conditions, the availability of αKG is critical for the activity of PHD (Boulahbel, Duran & Gottlieb, Biochem. Soc. Trans. 2009). Importantly, our work shows that amino acid starvation causes depletion of αKG that leads to PHD inactivation. Importantly, loss of PHD activity during amino acid starvation does not activate HIF, likely due to a lack of HIF expression. In agreement with this result, PHD inhibition blocks the response of mTORC1 to amino acids and induces autophagy independently of HIF (Duran et al., Oncogene 2013). Therefore, PHDs act as general metabolic sensors in the cell, detecting scarcity not only of oxygen, but also of amino acids, and forming a link between glutaminolysis and the mTORC1 pathway (Nguyen & Duran, Int J Biochem Cell Biol 2016) (Figure 1).

Glutamoptosis: a new autophagy-inhibited cell death mechanism during nutritional imbalance

Despite the role of both glutaminolysis and mTORC1 in the promotion of cell growth, we recently observed that the unbalanced activation of the glutaminolysis/mTORC1 pathway in the absence of other amino acids induced apoptotic cell death in human cells. We termed this unusual type of cell death as “glutamoptosis” (Villar & Duran, Autophagy 2017). During glutamoptosis, the long-term production of glutaminolysis-derived αKG in the absence of other amino acids was sufficient to activate mTORC1 and to inhibit autophagy, but concomitantly reduced cell viability and activated apoptosis. Inhibition of mTORC1 or re-activation of autophagy were sufficient to suppress the glutaminolysis-induced apoptosis. We additionally observed that the ability of rapamycin to prevent apoptosis resides in its pro-autophagic potential: re-inhibition of autophagy prevented cell survival in rapamycin-treated cells (Villar et al., Nature Comm 2017) (Figure 2). The surprising role of glutaminolysis as a cell death inducing mechanism during nutrient restriction points at the importance of nutritional imbalance in the control of cell viability and the potential use of this metabolic disequilibrium to identify new metabolic components and targets with potential implication in the treatment of nutrition-related diseases, such as obesity, diabetes, cancer, or cardiovascular diseases. In any case, whether the activation of apoptosis by the unbalanced activation of glutaminolysis and mTORC1 plays an active role in the biochemical basis of those diseases is a question that remains to be elucidated.

Selected Publications

  • Bodineau C, Tomé M, Murdoch PDS and Durán RV (2022) Glutamine, mTOR and autophagy: a multiconnection relationship, Autophagy 18, 2749–2750 (IF = 13.4).
  • Gómez-Marín E, Posavec-Marjanović M, Zarzuela L, Basurto-Cayuela L, Guerrero-Martínez J, Arribas G, Yerbes R, Ceballos-Chávez M, Rodriguez-Paredes M, Tomé M, Durán RV, Buschbeck M, Reyes JC (2022) The high mobility group protein HMG20A cooperates with the histone reader PHF14 to modulate TGFβ and Hippo pathways, Nucleic Acids Research 50, 9838-9857 (IF = 19.2).
  • Nicastro R, Gaillard H, Zarzuela L, Fernández-García E, Tomé M, García-Rodríguez N, Péli-Gulli MP, Durán RV, De Virgilio C and Wellinger RE (2022) Manganese is a physiologically relevant TORC1 activator in yeast and mammals, eLIFE 11, e80497 (IF = 8.7).
  • Cabré N, Luciano-Mateo F, Chapski DJ, Baiges-Gaya G, Fernández-Arroyo S, Hernández-Aguilera A, Castañé H, Rodríguez-Tomàs E, París M, Sabench M, Del CastilloD, Del Bas JM, Tomé M, Bodineau C, Sola-García A, López-Miranda J, Martín-Montalvo A, Durán RV, Vondriska TM, Rosa-Garrido M, Camps J, Menéndez JA and Joven J (2022) Laparoscopic sleeve gastrectomy in patients with severe obesity restores adaptive responses leading to nonalcoholic steatohepatitis, Int. J. Mol. Sciences 23, 7830 (IF = 6.2).
  • Sarmiento A, Durán-Díaz I, Fondón I, Tomé M, Bodineau C and Durán RV (2022) A method for unsupervised semi-quantification of immunohistochemical staining with beta-divergences, Entropy 24, 546 (IF = 2.7).
  • Kaminski H, Marseres G, Yared N, Nokin MJ, Hooks K, Pitard V, Tarricone A, Zouine A, Garrigue I, Loizon S, Capone M, Gauthereau X, Mamani-Matsuda M, Durán RV, Pinson B, Pellegrin I, Thiébaut R, Couzi L, Merville P and Déchanet-Merville J (2022) mTOR inhibitors prevent CMV infection through restoration of functional αβ and γδ T cells in kidney transplantation, J. Amer. Soc. Nephrol. 33, 121-137 (IF = 15.0).
  • Bodineau C, Tomé M, Courtois S, Costa ASH, Richard E, Sciacovelli M, Vacher P, Bessede E, Varon C, Rousseau B, Soubeyran P, Frezza C, Murdoch PS, Villar VH and Durán RV (2021) Two parallel pathways connect glutamine metabolism and mTORC1 activity to regulate glutamoptosis, Nature Commun. 12, 4814 (IF = 17.7).
  • Nguyen TL, Nokin MJ, Terés S, Tomé M, Galmar O, Pasquet JM, Rousseau B, van Liempd S, Falcon JM, Richard E, Muzotte E, Rezvani HR, Priault M, Bouchecareilh M, Redonnet-Vernhet I, Fuentes P, Calvo J, Uzan B, Pflumio F, Fuentes P, Toribio ML, Khatib AM, Mudoch PS, Soubeyran P and Durán RV (2021). Downregulation of Glutamine Synthetase, not glutaminolysis, is responsible for glutamine addiction in Notch1-driven acute lymphoblastic leukemia, Mol. Oncol. 15, 1412-1431 (IF = 7.5).
  • Klionsky et al. (2021). Guidelines for the use and interpretation of assays for monitoring autophagy (4th edition), Autophagy 17, 1-382 (IF = 13.4).
  • Courtois S, Haykal M, Bodineau C, Sifré E, Ménard A, Azzi-Martin L, Mégraud F, Lehours P, Durán RV, Varon C, and Bessède E (2021). Autophagy induced by Helicobacter pylori infection is necessary for gastric cancer stem cells emergence, Gastric Cancer 24, 133-144 (IF = 7.7).
  • Soulet F, Bodineau C, Hoocks KB, Descarpentrie J, Alves I, Dubreuil M, Mouchard A, Eugenie M, Hoepffner JL, López J, Rosado JA, Soubeyran I, Tomé M, Durán RV, Nikolski M, Villoutreix BO, Evrard S, Siegfried G, and Khatib AM (2020). ELA/APELA precursor cleaved by furin displays tumor suppressor function in renal cell carcinoma through mTORC1 activation, JCI Insight 5, e129070 (IF = 8.3).
  • Chinoy ZS, Bodineau C, Favre C, Moremen K, Durán RV and Friscourt F (2019). Selective Engineering of Linkage Specific α2,6-N-Linked Sialoproteins using Sydnone-Modified Sialic Acid Bioorthogonal Reporters. Angewandte Chem. Int. Ed. 58, 4281-4285 (IF = 12.3)
  • Nguyen TL and Durán RV (2018). Glutamine metabolism in cancer therapy. Cancer Drug Resist. 1, 126-38.
  • Nguyen TL, Nokin MJ, Egorov M, Tomé M, Bodineau C, Di Primo C, Minder L, Wdzieczak-Bakala J, Garcia-Alvarez MC, Bignon J, Thoison O, Delpech B, Surpateanu G, Frapart YM, Peyrot F, Abbas K, Terés S, Evrard S, Khatib AM, Soubeyran P, Iorga B, Durán RV* and Collin P (2018). mTOR inhibition via displacement of phosphatidic acid 2 induces enhanced cytotoxicity specifically in cancer cells. Cancer Research 78, 5384-5397 (IF = 8.4) (*, corresponding author).
  • Courtois S, Durán RV, Giraud J, Sifré E, Izotte J, Mégraud F, Lehours P, Varon C and Bessède E (2017). Metformin targets gastric cancer stem cells. Eur. J. Cancer 84, 193-201 (IF = 7.2).
  • Villar VH & Durán RV (2017). Glutamoptosis: a new cell death mechanism inhibited by autophagy during nutritional imbalance. Autophagy 13, 1078-1079 (IF = 11.1).
  • Villar VH, Nguyen TL, Terés S, Bodineau C and Durán RV (2017). Escaping mTOR inhibition for cancer therapy: tumor suppressor functions of mTOR. Mol. Cell. Oncol. 4, e1297284.
  • Villar VH, Nguyen TL, Delcroix V, Terés S, Bouchecareilh M, Salin B, Bodineau C, Vacher P, Priault M, Soubeyran P and Durán RV (2017). mTORC1 inhibition in cancer cells protects from glutaminolysis-mediated apoptosis during nutrient limitation. Nature Commun. 8, 14124 (IF = 12.4).
  • Nguyen TL and Durán RV (2016). Prolyl hydroxylase domain enzymes and their role in cell signaling and cancer metabolism. Int. J. Biochem. Cell Biol. 80, 71-80 (IF = 3.51).
  • Klionsky DJ et al. (2016). Guidelines for the use and interpretation of assays for monitoring autophagy. Autophagy 12, 1-222 (IF = 8.6).
  • Villar VH, Mehri F, Djavaheri-Mergny M and Durán RV (2015). Glutaminolysis and autophagy in cancer. Autophagy 11, 1198-1208 (IF = 9.11).

Group leader:
  • Dr. Raúl V. Durán
Senior Researchers:
Postdoctorals:
  • Dr. Macarena Morillo Huesca
  • Dr. Mercedes Tomé Montesinos
PhD students:
  • Ignacio González López-Cepero
  • Ana Reina Bando
  • Laura Zarzuela Moncada
Master Students / Erasmus +:
  • Andrea Ramos Luna