73 results on '"Ben-Sahra I"'
Search Results
2. The mitochondrial NADH shuttle system is a targetable vulnerability for Group 3 medulloblastoma in a hypoxic microenvironment
- Author
-
Contenti, J., Guo, Y., Mazzu, A., Irondelle, M., Rouleau, M., Lago, C., Leva, G., Tiberi, L., Ben-Sahra, I., Bost, F., and Mazure, N. M.
- Published
- 2023
- Full Text
- View/download PDF
3. Eif5a hypusination shapes metabolic platicity in prostate cancer
- Author
-
Kahi, M., primary, Mazzu, A., additional, Tiroille, V., additional, Vincent, A., additional, Irondelle, M., additional, Lacas-Gervais, S., additional, Camoin, L., additional, Auderbert, S., additional, Diaz, J-J., additional, Chen, Y., additional, Ben-Sahra, I., additional, Peraldi, P., additional, Mazure, N., additional, and Bost, F., additional
- Published
- 2023
- Full Text
- View/download PDF
4. Oncogenic PI3K and K-Ras stimulate de novo lipid synthesis through mTORC1 and SREBP
- Author
-
Ricoult, S J H, Yecies, J L, Ben-Sahra, I, and Manning, B D
- Published
- 2016
- Full Text
- View/download PDF
5. LB06 - Eif5a hypusination shapes metabolic platicity in prostate cancer
- Author
-
Kahi, M., Mazzu, A., Tiroille, V., Vincent, A., Irondelle, M., Lacas-Gervais, S., Camoin, L., Auderbert, S., Diaz, J-J., Chen, Y., Ben-Sahra, I., Peraldi, P., Mazure, N., and Bost, F.
- Published
- 2023
- Full Text
- View/download PDF
6. OS6.1 Targeting Purine Metabolism to Overcome Therapeutic Resistance in Glioblastoma
- Author
-
Ahmed, A U, primary, Shireman, J, additional, Atashi, F, additional, Saathoff, M, additional, Ali, E, additional, Lee, G, additional, Park, C, additional, Savchuk, S, additional, Rayyan, J, additional, James, C, additional, and Ben-Sahra, I, additional
- Published
- 2019
- Full Text
- View/download PDF
7. Oncogenic PI3K and K-Ras stimulate de novo lipid synthesis through mTORC1 and SREBP
- Author
-
Ricoult, S J H, primary, Yecies, J L, additional, Ben-Sahra, I, additional, and Manning, B D, additional
- Published
- 2015
- Full Text
- View/download PDF
8. Low-dose radiation exposure induces a HIF-1-mediated adaptive and protective metabolic response
- Author
-
Lall, R, primary, Ganapathy, S, additional, Yang, M, additional, Xiao, S, additional, Xu, T, additional, Su, H, additional, Shadfan, M, additional, Asara, J M, additional, Ha, C S, additional, Ben-Sahra, I, additional, Manning, B D, additional, Little, J B, additional, and Yuan, Z-M, additional
- Published
- 2014
- Full Text
- View/download PDF
9. Sestrin2 integrates Akt and mTOR signaling to protect cells against energetic stress-induced death
- Author
-
Ben-Sahra, I, primary, Dirat, B, additional, Laurent, K, additional, Puissant, A, additional, Auberger, P, additional, Budanov, A, additional, Tanti, J-F, additional, and Bost, F, additional
- Published
- 2012
- Full Text
- View/download PDF
10. Oncogenic PI3K and K-Ras stimulate de novolipid synthesis through mTORC1 and SREBP
- Author
-
Ricoult, S J H, Yecies, J L, Ben-Sahra, I, and Manning, B D
- Abstract
An enhanced capacity for de novolipid synthesis is a metabolic feature of most cancer cells that distinguishes them from their cells of origin. However, the mechanisms through which oncogenes alter lipid metabolism are poorly understood. We find that expression of oncogenic PI3K (H1047R) or K-Ras (G12V) in breast epithelial cells is sufficient to induce de novolipogenesis, and this occurs through the convergent activation of the mechanistic target of rapamycin complex 1 (mTORC1) downstream of these common oncogenes. Oncogenic stimulation of mTORC1 signaling in this isogenic setting or a panel of eight breast cancer cell lines leads to activation of the sterol regulatory element-binding proteins (SREBP1 and SREBP2) that are required for oncogene-induced lipid synthesis. The SREBPs are also required for the growth factor-independent growth and proliferation of oncogene-expressing cells. Finally, we find that elevated mTORC1 signaling is associated with increased mRNA and protein levels of canonical SREBP targets in primary human breast cancer samples. These data suggest that the mTORC1/SREBP pathway is a major mechanism through which common oncogenic signaling events induce de novolipid synthesis to promote aberrant growth and proliferation of cancer cells.
- Published
- 2016
- Full Text
- View/download PDF
11. Sestrin2 integrates Akt and mTOR signaling to protect cells against energetic stress-induced death.
- Author
-
Ben-Sahra, I, Dirat, B, Laurent, K, Puissant, A, Auberger, P, Budanov, A, Tanti, J-F, and Bost, F
- Subjects
- *
MTOR protein , *PHOSPHOINOSITIDES , *CELL metabolism , *CANCER cell proliferation , *APOPTOSIS - Abstract
The phosphoinositide-3 kinase/Akt (PI3K/Akt) pathway has a central role in cancer cell metabolism and proliferation. More importantly, it is one of the cardinal pro-survival pathways mediating resistance to apoptosis. The role of Akt in response to an energetic stress is presently unclear. Here, we show that Sestrin2 (Sesn2), also known as Hi95, a p53 target gene that protects cells against oxidative and genotoxic stresses, participates in the protective role of Akt in response to an energetic stress induced by 2-deoxyglucose (2-DG). Sesn2 is upregulated in response to an energetic stress such as 2-DG and metformin, and mediates the inhibition of mammalian target of rapamycin (mTOR), the major cellular regulator of energy metabolism. The increase of Sesn2 is independent of p53 but requires the anti-apoptotic pathway, PI3K/Akt. Inhibition of Akt, as well as loss of Sesn2, sensitizes cells to 2-DG-induced apoptosis. In addition, the rescue of Sesn2 partially reverses the pro-apoptotic effects of 2-DG. In conclusion, we identify Sesn2 as a new energetic stress sensor, which appears to be protective against energetic stress-induced apoptosis that integrates the pro-survival function of Akt and the negative regulation of mTOR. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
12. Brain-penetrating molecule might offer a route to treat glioblastoma tumours.
- Author
-
Nengroo MA and Ben-Sahra I
- Abstract
Competing Interests: The authors declare no competing interests.
- Published
- 2024
- Full Text
- View/download PDF
13. ASCT2 is a major contributor to serine uptake in cancer cells.
- Author
-
Conger KO, Chidley C, Ozgurses ME, Zhao H, Kim Y, Semina SE, Burns P, Rawat V, Lietuvninkas L, Sheldon R, Ben-Sahra I, Frasor J, Sorger PK, DeNicola GM, and Coloff JL
- Subjects
- Humans, Glutamine metabolism, Cell Line, Tumor, Estrogen Receptor alpha metabolism, Neoplasms metabolism, Neoplasms pathology, Neoplasms genetics, Animals, Biological Transport, Female, MCF-7 Cells, Amino Acid Transport System ASC metabolism, Amino Acid Transport System ASC genetics, Serine metabolism, Minor Histocompatibility Antigens metabolism, Minor Histocompatibility Antigens genetics
- Abstract
The non-essential amino acid serine is a critical nutrient for cancer cells due to its diverse biosynthetic functions. While some tumors can synthesize serine de novo, others are auxotrophic and therefore reliant on serine uptake. Importantly, despite several transporters being known to be capable of transporting serine, the transporters that mediate serine uptake in cancer cells are not known. Here, we characterize the amino acid transporter ASCT2 (SLC1A5) as a major contributor to serine uptake in cancer cells. ASCT2 is well known as a glutamine transporter in cancer, and our work demonstrates that serine and glutamine compete for uptake through ASCT2. We further show that ASCT2-mediated serine uptake is essential for purine nucleotide biosynthesis and that estrogen receptor α (ERα) promotes serine uptake by directly activating SLC1A5 transcription. Collectively, our work defines an additional important role for ASCT2 as a serine transporter in cancer and evaluates ASCT2 as a potential therapeutic target., Competing Interests: Declaration of interests P.K.S. is a member of the SAB or BOD for Applied Biomath, RareCyte. Nanostring, Glencoe Software, and Montai; he is consultant for Merck. None of these activities impact the content of this manuscript., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
14. Longitudinal trajectories of branched chain amino acids through young adulthood and diabetes in later life.
- Author
-
Sawicki KT, Ning H, Allen NB, Carnethon MR, Wallia A, Otvos JD, Ben-Sahra I, McNally EM, Snell-Bergeon JK, and Wilkins JT
- Subjects
- Humans, Male, Female, Adult, Longitudinal Studies, Young Adult, Adolescent, Diabetes Mellitus epidemiology, Middle Aged, Amino Acids, Branched-Chain metabolism
- Published
- 2024
- Full Text
- View/download PDF
15. Pro-905, a Novel Purine Antimetabolite, Combines with Glutamine Amidotransferase Inhibition to Suppress Growth of Malignant Peripheral Nerve Sheath Tumor.
- Author
-
Lemberg KM, Ali ES, Krecmerova M, Aguilar JMH, Alt J, Peters DE, Zhao L, Wu Y, Nuha N, Asara JM, Staedtke V, Pratilas CA, Majer P, Rais R, Ben-Sahra I, and Slusher BS
- Subjects
- Humans, Animals, Mice, Glutamine, Cell Line, Tumor, Antimetabolites therapeutic use, Neurofibrosarcoma, Nerve Sheath Neoplasms drug therapy
- Abstract
Malignant peripheral nerve sheath tumors (MPNST) are highly aggressive soft-tissue sarcomas that arise from neural tissues and carry a poor prognosis. Previously, we found that the glutamine amidotransferase inhibitor JHU395 partially impeded tumor growth in preclinical models of MPNST. JHU395 inhibits de novo purine synthesis in human MPNST cells and murine tumors with partial decreases in purine monophosphates. On the basis of prior studies showing enhanced efficacy when glutamine amidotransferase inhibition was combined with the antimetabolite 6-mercaptopurine (6-MP), we hypothesized that such a combination would be efficacious in MPNST. Given the known toxicity associated with 6-MP, we set out to develop a more efficient and well-tolerated drug that targets the purine salvage pathway. Here, we report the discovery of Pro-905, a phosphoramidate protide that delivered the active nucleotide antimetabolite thioguanosine monophosphate (TGMP) to tumors over 2.5 times better than equimolar 6-MP. Pro-905 effectively prevented the incorporation of purine salvage substrates into nucleic acids and inhibited colony formation of human MPNST cells in a dose-dependent manner. In addition, Pro-905 inhibited MPNST growth and was well-tolerated in both human patient-derived xenograft (PDX) and murine flank MPNST models. When combined with JHU395, Pro-905 enhanced the colony formation inhibitory potency of JHU395 in human MPNST cells and augmented the antitumor efficacy of JHU395 in mice. In summary, the dual inhibition of the de novo and purine salvage pathways in preclinical models may safely be used to enhance therapeutic efficacy against MPNST., (©2023 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
16. HIF-1 inactivation empowers HIF-2 to drive hypoxia adaptation in aggressive forms of medulloblastoma.
- Author
-
Contenti J, Guo Y, Larcher M, Mirabal-Ortega L, Rouleau M, Irondelle M, Tiroille V, Mazzu A, Duranton-Tanneur V, Pedeutour F, Ben-Sahra I, Lago C, Leva G, Tiberi L, Robert G, Pouponnot C, Bost F, and Mazure NM
- Abstract
Medulloblastoma (MB) is the most prevalent brain cancer in children. Four subgroups of MB have been identified; of these, Group 3 is the most metastatic. Its genetics and biology remain less clear than the other groups, and it has a poor prognosis and few effective treatments available. Tumor hypoxia and the resulting metabolism are known to be important in the growth and survival of tumors but, to date, have been only minimally explored in MB. Here we show that Group 3 MB tumors do not depend on the canonical transcription factor hypoxia-inducible factor-1α (HIF-1α) to mount an adaptive response to hypoxia. We discovered that HIF-1α is rendered inactive either through post-translational methylation, preventing its nuclear localization specifically in Group 3 MB, or by a low expression that prevents modulation of HIF-target genes. Strikingly, we found that HIF-2 takes over the role of HIF-1 in the nucleus and promotes the activation of hypoxia-dependent anabolic pathways. The exclusion of HIF-1 from the nucleus in Group 3 MB cells enhances the reliance on HIF-2's transcriptional role, making it a viable target for potential anticancer strategies. By combining pharmacological inhibition of HIF-2α with the use of metformin, a mitochondrial complex I inhibitor to block respiration, we effectively induced Group 3 MB cell death, surpassing the effectiveness observed in Non-Group 3 MB cells. Overall, the unique dependence of MB cells, but not normal cells, on HIF-2-mediated anabolic metabolism presents an appealing therapeutic opportunity for treating Group 3 MB patients with minimal toxicity.
- Published
- 2023
- Full Text
- View/download PDF
17. ASCT2 is the primary serine transporter in cancer cells.
- Author
-
Conger KO, Chidley C, Ozgurses ME, Zhao H, Kim Y, Semina SE, Burns P, Rawat V, Sheldon R, Ben-Sahra I, Frasor J, Sorger PK, DeNicola GM, and Coloff JL
- Abstract
The non-essential amino acid serine is a critical nutrient for cancer cells due to its diverse biosynthetic functions. While some tumors can synthesize serine de novo , others are auxotrophic for serine and therefore reliant on the uptake of exogenous serine. Importantly, however, the transporter(s) that mediate serine uptake in cancer cells are not known. Here, we characterize the amino acid transporter ASCT2 (coded for by the gene SLC1A5 ) as the primary serine transporter in cancer cells. ASCT2 is well-known as a glutamine transporter in cancer, and our work demonstrates that serine and glutamine compete for uptake through ASCT2. We further show that ASCT2-mediated serine uptake is essential for purine nucleotide biosynthesis and that ERα promotes serine uptake by directly activating SLC1A5 transcription. Together, our work defines an additional important role for ASCT2 as a serine transporter in cancer and evaluates ASCT2 as a potential therapeutic target in serine metabolism., Competing Interests: Declaration of competing interests PKS is a member of the SAB or BOD for Applied Biomath, RareCyte. Nanostring, Glencoe Software and Montai; he is consultant for Merck. None of these activities impact the content of this manuscript. The other authors declare no competing interests.
- Published
- 2023
- Full Text
- View/download PDF
18. Mechano-dependent sorbitol accumulation supports biomolecular condensate.
- Author
-
Torrino S, Oldham WM, Tejedor AR, Burgos IS, Rachedi N, Fraissard K, Chauvet C, Sbai C, O'Hara BP, Abélanet S, Brau F, Clavel S, Collepardo-Guevara R, Espinosa JR, Ben-Sahra I, and Bertero T
- Abstract
Biomolecular condensates regulate a wide range of cellular functions from signaling to RNA metabolism
1, 2 , yet, the physiologic conditions regulating their formation remain largely unexplored. Biomolecular condensate assembly is tightly regulated by the intracellular environment. Changes in the chemical or physical conditions inside cells can stimulate or inhibit condensate formation3-5 . However, whether and how the external environment of cells can also regulate biomolecular condensation remain poorly understood. Increasing our understanding of these mechanisms is paramount as failure to control condensate formation and dynamics can lead to many diseases6, 7 . Here, we provide evidence that matrix stiffening promotes biomolecular condensation in vivo . We demonstrate that the extracellular matrix links mechanical cues with the control of glucose metabolism to sorbitol. In turn, sorbitol acts as a natural crowding agent to promote biomolecular condensation. Using in silico simulations and in vitro assays, we establish that variations in the physiological range of sorbitol, but not glucose, concentrations, are sufficient to regulate biomolecular condensates. Accordingly, pharmacologic and genetic manipulation of intracellular sorbitol concentration modulates biomolecular condensates in breast cancer - a mechano-dependent disease. We propose that sorbitol is a mechanosensitive metabolite enabling protein condensation to control mechano-regulated cellular functions. Altogether, we uncover molecular driving forces underlying protein phase transition and provide critical insights to understand the biological function and dysfunction of protein phase separation.- Published
- 2023
- Full Text
- View/download PDF
19. Therapeutic targeting of metabolic vulnerabilities in cancers with MLL3/4-COMPASS epigenetic regulator mutations.
- Author
-
Zhao Z, Cao K, Watanabe J, Philips CN, Zeidner JM, Ishi Y, Wang Q, Gold SR, Junkins K, Bartom ET, Yue F, Chandel NS, Hashizume R, Ben-Sahra I, and Shilatifard A
- Subjects
- Humans, Animals, Mice, Histone-Lysine N-Methyltransferase genetics, Mutation, Epigenesis, Genetic, Proteomics, Neoplasms genetics
- Abstract
Epigenetic status-altering mutations in chromatin-modifying enzymes are a feature of human diseases, including many cancers. However, the functional outcomes and cellular dependencies arising from these mutations remain unresolved. In this study, we investigated cellular dependencies, or vulnerabilities, that arise when enhancer function is compromised by loss of the frequently mutated COMPASS family members MLL3 and MLL4. CRISPR dropout screens in MLL3/4-depleted mouse embryonic stem cells (mESCs) revealed synthetic lethality upon suppression of purine and pyrimidine nucleotide synthesis pathways. Consistently, we observed a shift in metabolic activity toward increased purine synthesis in MLL3/4-KO mESCs. These cells also exhibited enhanced sensitivity to the purine synthesis inhibitor lometrexol, which induced a unique gene expression signature. RNA-Seq identified the top MLL3/4 target genes coinciding with suppression of purine metabolism, and tandem mass tag proteomic profiling further confirmed upregulation of purine synthesis in MLL3/4-KO cells. Mechanistically, we demonstrated that compensation by MLL1/COMPASS was underlying these effects. Finally, we demonstrated that tumors with MLL3 and/or MLL4 mutations were highly sensitive to lometrexol in vitro and in vivo, both in culture and in animal models of cancer. Our results depicted a targetable metabolic dependency arising from epigenetic factor deficiency, providing molecular insight to inform therapy for cancers with epigenetic alterations secondary to MLL3/4 COMPASS dysfunction.
- Published
- 2023
- Full Text
- View/download PDF
20. Ribonucleotide reductase regulatory subunit M2 drives glioblastoma TMZ resistance through modulation of dNTP production.
- Author
-
Perrault EN, Shireman JM, Ali ES, Lin P, Preddy I, Park C, Budhiraja S, Baisiwala S, Dixit K, James CD, Heiland DH, Ben-Sahra I, Pott S, Basu A, Miska J, and Ahmed AU
- Subjects
- Humans, Cell Line, Tumor, Temozolomide pharmacology, Temozolomide therapeutic use, Brain Neoplasms drug therapy, Brain Neoplasms genetics, Brain Neoplasms metabolism, Glioblastoma drug therapy, Glioblastoma genetics, Glioblastoma metabolism, Ribonucleotide Reductases genetics, Ribonucleotide Reductases therapeutic use, Drug Resistance, Neoplasm genetics
- Abstract
During therapy, adaptations driven by cellular plasticity are partly responsible for driving the inevitable recurrence of glioblastoma (GBM). To investigate plasticity-induced adaptation during standard-of-care chemotherapy temozolomide (TMZ), we performed in vivo single-cell RNA sequencing in patient-derived xenograft (PDX) tumors of GBM before, during, and after therapy. Comparing single-cell transcriptomic patterns identified distinct cellular populations present during TMZ therapy. Of interest was the increased expression of ribonucleotide reductase regulatory subunit M2 ( RRM2 ), which we found to regulate dGTP and dCTP production vital for DNA damage response during TMZ therapy. Furthermore, multidimensional modeling of spatially resolved transcriptomic and metabolomic analysis in patients' tissues revealed strong correlations between RRM2 and dGTP. This supports our data that RRM2 regulates the demand for specific dNTPs during therapy. In addition, treatment with the RRM2 inhibitor 3-AP (Triapine) enhances the efficacy of TMZ therapy in PDX models. We present a previously unidentified understanding of chemoresistance through critical RRM2-mediated nucleotide production.
- Published
- 2023
- Full Text
- View/download PDF
21. Longitudinal trajectories of branched chain amino acids through young adulthood and diabetes in later life.
- Author
-
Sawicki KT, Ning H, Allen NB, Carnethon MR, Wallia A, Otvos JD, Ben-Sahra I, McNally EM, Snell-Bergeon JK, and Wilkins JT
- Subjects
- Young Adult, Male, Humans, Adult, Risk Factors, Prospective Studies, Amino Acids, Branched-Chain metabolism, Diabetes Mellitus, Type 2 metabolism
- Abstract
BACKGROUNDElevated circulating branched chain amino acids (BCAAs), measured at a single time point in middle life, are strongly associated with an increased risk of developing type 2 diabetes mellitus (DM). However, the longitudinal patterns of change in BCAAs through young adulthood and their association with DM in later life are unknown.METHODSWe serially measured BCAAs over 28 years in the Coronary Artery Risk Development in Young Adults (CARDIA) study, a prospective cohort of apparently healthy Black and White young adults at baseline. Trajectories of circulating BCAA concentrations from years 2-30 (for prevalent DM) or years 2-20 (for incident DM) were determined by latent class modeling.RESULTSAmong 3,081 apparently healthy young adults, trajectory analysis from years 2-30 revealed 3 distinct BCAA trajectory groups: low-stable (n = 1,427), moderate-stable (n = 1,384), and high-increasing (n = 270) groups. Male sex, higher body mass index, and higher atherogenic lipid fractions were more common in the moderate-stable and high-increasing groups. Higher risk of prevalent DM was associated with the moderate-stable (OR = 2.59, 95% CI: 1.90-3.55) and high-increasing (OR = 6.03, 95% CI: 3.86-9.43) BCAA trajectory groups in adjusted models. A separate trajectory group analysis from years 2-20 for incident DM after year 20 showed that moderate-stable and high-increasing trajectory groups were also significantly associated with higher risk of incident DM, after adjustment for clinical variables and glucose levels.CONCLUSIONBCAA levels track over a 28-year span in most young adults, but serial clinical metabolomic measurements identify subpopulations with rising levels associated with high risk of DM in later life.FUNDINGThis research was supported by the NIH, under grants R01 HL146844 (JTW) and T32 HL069771 (MRC). The CARDIA study is conducted and supported by the NIH National Heart, Lung, and Blood Institute in collaboration with the University of Alabama at Birmingham (HHSN268201800005I and HHSN268201800007I), Northwestern University (HHSN268201800003I), the University of Minnesota (HHSN268201800006I), and Kaiser Foundation Research Institute (HHSN268201800004I).
- Published
- 2023
- Full Text
- View/download PDF
22. Purine synthesis suppression reduces the development and progression of pulmonary hypertension in rodent models.
- Author
-
Ma Q, Yang Q, Xu J, Sellers HG, Brown ZL, Liu Z, Bordan Z, Shi X, Zhao D, Cai Y, Pareek V, Zhang C, Wu G, Dong Z, Verin AD, Gan L, Du Q, Benkovic SJ, Xu S, Asara JM, Ben-Sahra I, Barman S, Su Y, Fulton DJR, and Huo Y
- Subjects
- Mice, Animals, Rodentia metabolism, Vascular Remodeling physiology, Pulmonary Artery, Purines metabolism, Cells, Cultured, Hypoxia metabolism, RNA, Messenger metabolism, Platelet-Derived Growth Factor metabolism, Cell Proliferation, Myocytes, Smooth Muscle metabolism, Hypertension, Pulmonary, Pulmonary Arterial Hypertension
- Abstract
Aims: Proliferation of vascular smooth muscle cells (VSMCs) is a hallmark of pulmonary hypertension (PH). Proliferative cells utilize purine bases from the de novo purine synthesis (DNPS) pathways for nucleotide synthesis; however, it is unclear whether DNPS plays a critical role in VSMC proliferation during development of PH. The last two steps of DNPS are catalysed by the enzyme 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase/inosine monophosphate cyclohydrolase (ATIC). This study investigated whether ATIC-driven DNPS affects the proliferation of pulmonary artery smooth muscle cells (PASMCs) and the development of PH., Methods and Results: Metabolites of DNPS in proliferative PASMCs were measured by liquid chromatography-tandem mass spectrometry. ATIC expression was assessed in platelet-derived growth factor-treated PASMCs and in the lungs of PH rodents and patients with pulmonary arterial hypertension. Mice with global and VSMC-specific knockout of Atic were utilized to investigate the role of ATIC in both hypoxia- and lung interleukin-6/hypoxia-induced murine PH. ATIC-mediated DNPS at the mRNA, protein, and enzymatic activity levels were increased in platelet-derived growth factor-treated PASMCs or PASMCs from PH rodents and patients with pulmonary arterial hypertension. In cultured PASMCs, ATIC knockdown decreased DNPS and nucleic acid DNA/RNA synthesis, and reduced cell proliferation. Global or VSMC-specific knockout of Atic attenuated vascular remodelling and inhibited the development and progression of both hypoxia- and lung IL-6/hypoxia-induced PH in mice., Conclusion: Targeting ATIC-mediated DNPS compromises the availability of purine nucleotides for incorporation into DNA/RNA, reducing PASMC proliferation and pulmonary vascular remodelling and ameliorating the development and progression of PH., Competing Interests: Conflict of interest statement All authors declare no conflict of interest for this contribution., (© The Author(s) 2023. Published by Oxford University Press on behalf of the European Society of Cardiology. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2023
- Full Text
- View/download PDF
23. GATOR2 rings GATOR1 to speak to mTORC1.
- Author
-
Sahu U and Ben-Sahra I
- Subjects
- Mechanistic Target of Rapamycin Complex 1 genetics, Leucine, Lysosomes, TOR Serine-Threonine Kinases genetics, Multiprotein Complexes genetics
- Abstract
The mechanistic target of rapamycin complex 1 (mTORC1) senses cellular leucine levels through the GATOR1/2-Rag axis. Jiang et al. show that the Ring domains of GATOR2 subunits maintain the integrity of the complex and promote ubiquitination and inhibition of GATOR1, thereby leading to mTORC1 activation., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2022 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
24. Oncogenic deubiquitination controls tyrosine kinase signaling and therapy response in acute lymphoblastic leukemia.
- Author
-
Jin Q, Gutierrez Diaz B, Pieters T, Zhou Y, Narang S, Fijalkwoski I, Borin C, Van Laere J, Payton M, Cho BK, Han C, Sun L, Serafin V, Yacu G, Von Loocke W, Basso G, Veltri G, Dreveny I, Ben-Sahra I, Goo YA, Safgren SL, Tsai YC, Bornhauser B, Suraneni PK, Gaspar-Maia A, Kandela I, Van Vlierberghe P, Crispino JD, Tsirigos A, and Ntziachristos P
- Subjects
- Humans, Cell Line, Tumor, Glucocorticoids pharmacology, Glucocorticoids therapeutic use, Lymphocyte Specific Protein Tyrosine Kinase p56(lck) metabolism, Receptors, Glucocorticoid metabolism, Signal Transduction, Thiolester Hydrolases metabolism, Thiolester Hydrolases therapeutic use, Ubiquitin-Specific Peptidase 7 metabolism, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics
- Abstract
Dysregulation of kinase signaling pathways favors tumor cell survival and therapy resistance in cancer. Here, we reveal a posttranslational regulation of kinase signaling and nuclear receptor activity via deubiquitination in T cell acute lymphoblastic leukemia (T-ALL). We observed that the ubiquitin-specific protease 11 (USP11) is highly expressed and associates with poor prognosis in T-ALL. USP11 ablation inhibits leukemia progression in vivo, sparing normal hematopoiesis. USP11 forms a complex with USP7 to deubiquitinate the oncogenic lymphocyte cell-specific protein-tyrosine kinase (LCK) and enhance its activity. Impairment of LCK activity leads to increased glucocorticoid receptor (GR) expression and glucocorticoids sensitivity. Genetic knockout of USP7 improved the antileukemic efficacy of glucocorticoids in vivo. The transcriptional activation of GR target genes is orchestrated by the deubiquitinase activity and mediated via an increase in enhancer-promoter interaction intensity. Our data unveil how dysregulated deubiquitination controls leukemia survival and drug resistance, suggesting previously unidentified therapeutic combinations toward targeting leukemia.
- Published
- 2022
- Full Text
- View/download PDF
25. ATIC-Associated De Novo Purine Synthesis Is Critically Involved in Proliferative Arterial Disease.
- Author
-
Ma Q, Yang Q, Xu J, Zhang X, Kim D, Liu Z, Da Q, Mao X, Zhou Y, Cai Y, Pareek V, Kim HW, Wu G, Dong Z, Song WL, Gan L, Zhang C, Hong M, Benkovic SJ, Weintraub NL, Fulton D Jr, Asara JM, Ben-Sahra I, and Huo Y
- Subjects
- Humans, Mice, Animals, Neointima, Purines, Cell Proliferation, Myocytes, Smooth Muscle, Hydroxymethyl and Formyl Transferases, Atherosclerosis genetics
- Abstract
Background: Proliferation of vascular smooth muscle cells (VSMCs) is a hallmark of arterial diseases, especially in arterial restenosis after angioplasty or stent placement. VSMCs reprogram their metabolism to meet the increased requirements of lipids, proteins, and nucleotides for their proliferation. De novo purine synthesis is one of critical pathways for nucleotide synthesis. However, its role in proliferation of VSMCs in these arterial diseases has not been defined., Methods: De novo purine synthesis in proliferative VSMCs was evaluated by liquid chromatography-tandem mass spectrometry. The expression of ATIC (5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase/inosine monophosphate cyclohydrolase), the critical bifunctional enzyme in the last 2 steps of the de novo purine synthesis pathway, was assessed in VSMCs of proliferative arterial neointima. Global and VSMC-specific knockout of Atic mice were generated and used for examining the role of ATIC-associated purine metabolism in the formation of arterial neointima and atherosclerotic lesions., Results: In this study, we found that de novo purine synthesis was increased in proliferative VSMCs. Upregulated purine synthesis genes, including ATIC, were observed in the neointima of the injured vessels and atherosclerotic lesions both in mice and humans. Global or specific knockout of Atic in VSMCs inhibited cell proliferation, attenuating the arterial neointima in models of mouse atherosclerosis and arterial restenosis., Conclusions: These results reveal that de novo purine synthesis plays an important role in VSMC proliferation in arterial disease. These findings suggest that targeting ATIC is a promising therapeutic approach to combat arterial diseases.
- Published
- 2022
- Full Text
- View/download PDF
26. The mTORC1-SLC4A7 axis stimulates bicarbonate import to enhance de novo nucleotide synthesis.
- Author
-
Ali ES, Lipońska A, O'Hara BP, Amici DR, Torno MD, Gao P, Asara JM, Yap MF, Mendillo ML, and Ben-Sahra I
- Subjects
- Humans, Phosphorylation, Bicarbonates metabolism, Mechanistic Target of Rapamycin Complex 1 genetics, Mechanistic Target of Rapamycin Complex 1 metabolism, Nucleotides biosynthesis, Sodium-Bicarbonate Symporters genetics, Sodium-Bicarbonate Symporters metabolism
- Abstract
Bicarbonate (HCO
3 - ) ions maintain pH homeostasis in eukaryotic cells and serve as a carbonyl donor to support cellular metabolism. However, whether the abundance of HCO3 - is regulated or harnessed to promote cell growth is unknown. The mechanistic target of rapamycin complex 1 (mTORC1) adjusts cellular metabolism to support biomass production and cell growth. We find that mTORC1 stimulates the intracellular transport of HCO3 - to promote nucleotide synthesis through the selective translational regulation of the sodium bicarbonate cotransporter SLC4A7. Downstream of mTORC1, SLC4A7 mRNA translation required the S6K-dependent phosphorylation of the translation factor eIF4B. In mTORC1-driven cells, loss of SLC4A7 resulted in reduced cell and tumor growth and decreased flux through de novo purine and pyrimidine synthesis in human cells and tumors without altering the intracellular pH. Thus, mTORC1 signaling, through the control of SLC4A7 expression, harnesses environmental bicarbonate to promote anabolic metabolism, cell biomass, and growth., Competing Interests: Declaration of interests The authors declare no conflict of interests., (Copyright © 2022 Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
27. The hexokinase "HKDC1" interaction with the mitochondria is essential for liver cancer progression.
- Author
-
Khan MW, Terry AR, Priyadarshini M, Ilievski V, Farooq Z, Guzman G, Cordoba-Chacon J, Ben-Sahra I, Wicksteed B, and Layden BT
- Subjects
- Glucose metabolism, Humans, Mitochondria metabolism, Hexokinase genetics, Hexokinase metabolism, Liver Neoplasms genetics
- Abstract
Liver cancer (LC) is the fourth leading cause of death from cancer malignancies. Recently, a putative fifth hexokinase, hexokinase domain containing 1 (HKDC1), was shown to have significant overexpression in LC compared to healthy liver tissue. Using a combination of in vitro and in vivo tools, we examined the role of HKDC1 in LC development and progression. Importantly, HKDC1 ablation stops LC development and progression via its action at the mitochondria by promoting metabolic reprogramming and a shift of glucose flux away from the TCA cycle. HKDC1 ablation leads to mitochondrial dysfunction resulting in less cellular energy, which cannot be compensated by enhanced glucose uptake. Moreover, we show that the interaction of HKDC1 with the mitochondria is essential for its role in LC progression, and without this interaction, mitochondrial dysfunction occurs. As HKDC1 is highly expressed in LC cells, but only to a minimal degree in hepatocytes under normal conditions, targeting HKDC1, specifically its interaction with the mitochondria, may represent a highly selective approach to target cancer cells in LC., (© 2022. This is a U.S. Government work and not under copyright protection in the US; foreign copyright protection may apply.)
- Published
- 2022
- Full Text
- View/download PDF
28. C16orf72/HAPSTR1 is a molecular rheostat in an integrated network of stress response pathways.
- Author
-
Amici DR, Ansel DJ, Metz KA, Smith RS, Phoumyvong CM, Gayatri S, Chamera T, Edwards SL, O'Hara BP, Srivastava S, Brockway S, Takagishi SR, Cho BK, Goo YA, Kelleher NL, Ben-Sahra I, Foltz DR, Li J, and Mendillo ML
- Subjects
- Amino Acid Motifs, Animals, Cell Line, Tumor, Conserved Sequence, Humans, Protein Domains, Signal Transduction genetics, Tumor Suppressor Proteins metabolism, Ubiquitin-Protein Ligases metabolism, DNA Damage genetics, Genetic Fitness, Nuclear Proteins chemistry, Nuclear Proteins genetics, Nuclear Proteins metabolism, Stress, Physiological genetics
- Abstract
All cells contain specialized signaling pathways that enable adaptation to specific molecular stressors. Yet, whether these pathways are centrally regulated in complex physiological stress states remains unclear. Using genome-scale fitness screening data, we quantified the stress phenotype of 739 cancer cell lines, each representing a unique combination of intrinsic tumor stresses. Integrating dependency and stress perturbation transcriptomic data, we illuminated a network of genes with vital functions spanning diverse stress contexts. Analyses for central regulators of this network nominated C16orf72/HAPSTR1, an evolutionarily ancient gene critical for the fitness of cells reliant on multiple stress response pathways. We found that HAPSTR1 plays a pleiotropic role in cellular stress signaling, functioning to titrate various specialized cell-autonomous and paracrine stress response programs. This function, while dispensable to unstressed cells and nematodes, is essential for resilience in the presence of stressors ranging from DNA damage to starvation and proteotoxicity. Mechanistically, diverse stresses induce HAPSTR1, which encodes a protein expressed as two equally abundant isoforms. Perfectly conserved residues in a domain shared between HAPSTR1 isoforms mediate oligomerization and binding to the ubiquitin ligase HUWE1. We show that HUWE1 is a required cofactor for HAPSTR1 to control stress signaling and that, in turn, HUWE1 feeds back to ubiquitinate and destabilize HAPSTR1. Altogether, we propose that HAPSTR1 is a central rheostat in a network of pathways responsible for cellular adaptability, the modulation of which may have broad utility in human disease.
- Published
- 2022
- Full Text
- View/download PDF
29. mTORC1 functional assay reveals SZT2 loss-of-function variants and a founder in-frame deletion.
- Author
-
Calhoun JD, Aziz MC, Happ HC, Gunti J, Gleason C, Mohamed N, Zeng K, Hiller M, Bryant E, Mithal DS, Bellinski I, Kinsley L, Grimmel M, Schwaibold EMC, Smith-Hicks C, Chassevent A, Scala M, Accogli A, Torella A, Striano P, Capra V, Bird LM, Ben-Sahra I, Ekhilevich N, Hershkovitz T, Weiss K, Millichap J, Gerard EE, and Carvill GL
- Subjects
- Humans, Mechanistic Target of Rapamycin Complex 1 genetics, Nerve Tissue Proteins genetics, Tumor Suppressor Proteins genetics, Epilepsies, Partial, Epilepsy genetics, Megalencephaly genetics
- Abstract
Biallelic pathogenic variants in SZT2 result in a neurodevelopmental disorder with shared features, including early-onset epilepsy, developmental delay, macrocephaly, and corpus callosum abnormalities. SZT2 is as a critical scaffolding protein in the amino acid sensing arm of the mTORC1 signalling pathway. Due to its large size (3432 amino acids), lack of crystal structure, and absence of functional domains, it is difficult to determine the pathogenicity of SZT2 missense and in-frame deletions, but these variants are increasingly detected and reported by clinical genetic testing in individuals with epilepsy. To exemplify this latter point, here we describe a cohort of 12 individuals with biallelic SZT2 variants and phenotypic overlap with SZT2-related neurodevelopmental disorders. However, the majority of individuals carried one or more SZT2 variants of uncertain significance (VUS), highlighting the need for functional characterization to determine, which, if any, of these VUS were pathogenic. Thus, we developed a novel individualized platform to identify SZT2 loss-of-function variants in the context of mTORC1 signalling and reclassify VUS. Using this platform, we identified a recurrent in-frame deletion (SZT2 p.Val1984del) which was determined to be a loss-of-function variant and therefore likely pathogenic. Haplotype analysis revealed that this single in-frame deletion is a founder variant in those of Ashkenazi Jewish ancestry. Moreover, this approach allowed us to tentatively reclassify all of the VUS in our cohort of 12 individuals, identifying five individuals with biallelic pathogenic or likely pathogenic variants. Clinical features of these five individuals consisted of early-onset seizures (median 24 months), focal seizures, developmental delay and macrocephaly similar to previous reports. However, we also show a widening of the phenotypic spectrum, as none of the five individuals had corpus callosum abnormalities, in contrast to previous reports. Overall, we present a rapid assay to resolve VUS in SZT2, identify a founder variant in individuals of Ashkenazi Jewish ancestry, and demonstrate that corpus callosum abnormalities is not a hallmark feature of this condition. Our approach is widely applicable to other mTORopathies including the most common causes of the focal genetic epilepsies, DEPDC5, TSC1/2, MTOR and NPRL2/3., (© The Author(s) 2022. Published by Oxford University Press on behalf of the Guarantors of Brain. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2022
- Full Text
- View/download PDF
30. ZFP36L2 suppresses mTORc1 through a P53-dependent pathway to prevent peripartum cardiomyopathy in mice.
- Author
-
Kouzu H, Tatekoshi Y, Chang HC, Shapiro JS, McGee WA, De Jesus A, Ben-Sahra I, Arany Z, Leor J, Chen C, Blackshear PJ, and Ardehali H
- Subjects
- Animals, Female, Mice, Myocytes, Cardiac metabolism, Peripartum Period, Peroxidases genetics, Peroxidases metabolism, Pregnancy, RNA, Messenger metabolism, Tristetraprolin metabolism, Cardiomyopathies genetics, Cardiomyopathies pathology, Mechanistic Target of Rapamycin Complex 1 genetics, Mechanistic Target of Rapamycin Complex 1 metabolism, Nuclear Proteins metabolism, Pregnancy Complications, Cardiovascular metabolism, Pregnancy Complications, Cardiovascular therapy, Transcription Factors metabolism, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism
- Abstract
Pregnancy is associated with substantial physiological changes of the heart, and disruptions in these processes can lead to peripartum cardiomyopathy (PPCM). The molecular processes that cause physiological and pathological changes in the heart during pregnancy are not well characterized. Here, we show that mTORc1 was activated in pregnancy to facilitate cardiac enlargement that was reversed after delivery in mice. mTORc1 activation in pregnancy was negatively regulated by the mRNA-destabilizing protein ZFP36L2 through its degradation of Mdm2 mRNA and P53 stabilization, leading to increased SESN2 and REDD1 expression. This pathway impeded uncontrolled cardiomyocyte hypertrophy during pregnancy, and mice with cardiac-specific Zfp36l2 deletion developed rapid cardiac dysfunction after delivery, while prenatal treatment of these mice with rapamycin improved postpartum cardiac function. Collectively, these data provide what we believe to be a novel pathway for the regulation of mTORc1 through mRNA stabilization of a P53 ubiquitin ligase. This pathway was critical for normal cardiac growth during pregnancy, and its reduction led to PPCM-like adverse remodeling in mice.
- Published
- 2022
- Full Text
- View/download PDF
31. Purine nucleotide depletion prompts cell migration by stimulating the serine synthesis pathway.
- Author
-
Soflaee MH, Kesavan R, Sahu U, Tasdogan A, Villa E, Djabari Z, Cai F, Tran DH, Vu HS, Ali ES, Rion H, O'Hara BP, Kelekar S, Hallett JH, Martin M, Mathews TP, Gao P, Asara JM, Manning BD, Ben-Sahra I, and Hoxhaj G
- Subjects
- Carbon, Cell Movement, Purines, Purine Nucleotides, Serine metabolism
- Abstract
Purine nucleotides are necessary for various biological processes related to cell proliferation. Despite their importance in DNA and RNA synthesis, cellular signaling, and energy-dependent reactions, the impact of changes in cellular purine levels on cell physiology remains poorly understood. Here, we find that purine depletion stimulates cell migration, despite effective reduction in cell proliferation. Blocking purine synthesis triggers a shunt of glycolytic carbon into the serine synthesis pathway, which is required for the induction of cell migration upon purine depletion. The stimulation of cell migration upon a reduction in intracellular purines required one-carbon metabolism downstream of de novo serine synthesis. Decreased purine abundance and the subsequent increase in serine synthesis triggers an epithelial-mesenchymal transition (EMT) and, in cancer models, promotes metastatic colonization. Thus, reducing the available pool of intracellular purines re-routes metabolic flux from glycolysis into de novo serine synthesis, a metabolic change that stimulates a program of cell migration., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
32. Hexokinase 1 cellular localization regulates the metabolic fate of glucose.
- Author
-
De Jesus A, Keyhani-Nejad F, Pusec CM, Goodman L, Geier JA, Stoolman JS, Stanczyk PJ, Nguyen T, Xu K, Suresh KV, Chen Y, Rodriguez AE, Shapiro JS, Chang HC, Chen C, Shah KP, Ben-Sahra I, Layden BT, Chandel NS, Weinberg SE, and Ardehali H
- Subjects
- Animals, Glycolysis, Hexokinase genetics, Mice, Mitochondria metabolism, Pentose Phosphate Pathway, Glucose metabolism, Hexokinase metabolism
- Abstract
The product of hexokinase (HK) enzymes, glucose-6-phosphate, can be metabolized through glycolysis or directed to alternative metabolic routes, such as the pentose phosphate pathway (PPP) to generate anabolic intermediates. HK1 contains an N-terminal mitochondrial binding domain (MBD), but its physiologic significance remains unclear. To elucidate the effect of HK1 mitochondrial dissociation on cellular metabolism, we generated mice lacking the HK1 MBD (ΔE1HK1). These mice produced a hyper-inflammatory response when challenged with lipopolysaccharide. Additionally, there was decreased glucose flux below the level of GAPDH and increased upstream flux through the PPP. The glycolytic block below GAPDH is mediated by the binding of cytosolic HK1 with S100A8/A9, resulting in GAPDH nitrosylation through iNOS. Additionally, human and mouse macrophages from conditions of low-grade inflammation, such as aging and diabetes, displayed increased cytosolic HK1 and reduced GAPDH activity. Our data indicate that HK1 mitochondrial binding alters glucose metabolism through regulation of GAPDH., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2022 Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
33. Hepatic mTORC1 signaling activates ATF4 as part of its metabolic response to feeding and insulin.
- Author
-
Byles V, Cormerais Y, Kalafut K, Barrera V, Hughes Hallett JE, Sui SH, Asara JM, Adams CM, Hoxhaj G, Ben-Sahra I, and Manning BD
- Subjects
- Activating Transcription Factor 4 deficiency, Animal Feed, Animals, Feeding Behavior, Insulin metabolism, Mice, Mice, Inbred C57BL, Mice, Knockout, Mice, Transgenic, Signal Transduction, Activating Transcription Factor 4 metabolism, Liver metabolism, Mechanistic Target of Rapamycin Complex 1 metabolism
- Abstract
Objective: The mechanistic target of rapamycin complex 1 (mTORC1) is dynamically regulated by fasting and feeding cycles in the liver to promote protein and lipid synthesis while suppressing autophagy. However, beyond these functions, the metabolic response of the liver to feeding and insulin signaling orchestrated by mTORC1 remains poorly defined. Here, we determine whether ATF4, a stress responsive transcription factor recently found to be independently regulated by mTORC1 signaling in proliferating cells, is responsive to hepatic mTORC1 signaling to alter hepatocyte metabolism., Methods: ATF4 protein levels and expression of canonical gene targets were analyzed in the liver following fasting and physiological feeding in the presence or absence of the mTORC1 inhibitor, rapamycin. Primary hepatocytes from wild-type or liver-specific Atf4 knockout (LAtf4
KO ) mice were used to characterize the effects of insulin-stimulated mTORC1-ATF4 function on hepatocyte gene expression and metabolism. Both unbiased steady-state metabolomics and stable-isotope tracing methods were employed to define mTORC1 and ATF4-dependent metabolic changes. RNA-sequencing was used to determine global changes in feeding-induced transcripts in the livers of wild-type versus LAtf4KO mice., Results: We demonstrate that ATF4 and its metabolic gene targets are stimulated by mTORC1 signaling in the liver, in a hepatocyte-intrinsic manner by insulin in response to feeding. While we demonstrate that de novo purine and pyrimidine synthesis is stimulated by insulin through mTORC1 signaling in primary hepatocytes, this regulation was independent of ATF4. Metabolomics and metabolite tracing studies revealed that insulin-mTORC1-ATF4 signaling stimulates pathways of nonessential amino acid synthesis in primary hepatocytes, including those of alanine, aspartate, methionine, and cysteine, but not serine., Conclusions: The results demonstrate that ATF4 is a novel metabolic effector of mTORC1 in the liver, extending the molecular consequences of feeding and insulin-induced mTORC1 signaling in this key metabolic tissue to the control of amino acid metabolism., (Copyright © 2021 The Author(s). Published by Elsevier GmbH.. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
34. Dual Covalent Inhibition of PKM and IMPDH Targets Metabolism in Cutaneous Metastatic Melanoma.
- Author
-
Zerhouni M, Martin AR, Furstoss N, Gutierrez VS, Jaune E, Tekaya N, Beranger GE, Abbe P, Regazzetti C, Amdouni H, Driowya M, Dubreuil P, Luciano F, Jacquel A, Tulic MK, Cluzeau T, O'Hara BP, Ben-Sahra I, Passeron T, Benhida R, Robert G, Auberger P, and Rocchi S
- Subjects
- Aged, Aminoimidazole Carboxamide pharmacology, Animals, Cell Line, Tumor, Female, HEK293 Cells, Humans, Melanoma enzymology, Melanoma pathology, Mice, Mice, Nude, Random Allocation, Skin Neoplasms enzymology, Skin Neoplasms pathology, Thyroid Hormones, Xenograft Model Antitumor Assays, Thyroid Hormone-Binding Proteins, Melanoma, Cutaneous Malignant, Aminoimidazole Carboxamide analogs & derivatives, Carrier Proteins antagonists & inhibitors, IMP Dehydrogenase antagonists & inhibitors, Melanoma drug therapy, Membrane Proteins antagonists & inhibitors, Ribonucleotides pharmacology, Skin Neoplasms drug therapy
- Abstract
Overcoming acquired drug resistance is a primary challenge in cancer treatment. Notably, more than 50% of patients with BRAF
V600E cutaneous metastatic melanoma (CMM) eventually develop resistance to BRAF inhibitors. Resistant cells undergo metabolic reprogramming that profoundly influences therapeutic response and promotes tumor progression. Uncovering metabolic vulnerabilities could help suppress CMM tumor growth and overcome drug resistance. Here we identified a drug, HA344, that concomitantly targets two distinct metabolic hubs in cancer cells. HA344 inhibited the final and rate-limiting step of glycolysis through its covalent binding to the pyruvate kinase M2 (PKM2) enzyme, and it concurrently blocked the activity of inosine monophosphate dehydrogenase, the rate-limiting enzyme of de novo guanylate synthesis. As a consequence, HA344 efficiently targeted vemurafenib-sensitive and vemurafenib-resistant CMM cells and impaired CMM xenograft tumor growth in mice. In addition, HA344 acted synergistically with BRAF inhibitors on CMM cell lines in vitro . Thus, the mechanism of action of HA344 provides potential therapeutic avenues for patients with CMM and a broad range of different cancers. SIGNIFICANCE: Glycolytic and purine synthesis pathways are often deregulated in therapy-resistant tumors and can be targeted by the covalent inhibitor described in this study, suggesting its broad application for overcoming resistance in cancer., (©2021 American Association for Cancer Research.)- Published
- 2021
- Full Text
- View/download PDF
35. SGLT2 Inhibition on Cardiac Mitochondrial Function: Searching for a Sweet Spot.
- Author
-
Sawicki KT, Ben-Sahra I, and McNally EM
- Subjects
- Humans, Mitochondria, Sodium-Glucose Transporter 2, Diabetic Cardiomyopathies
- Published
- 2021
- Full Text
- View/download PDF
36. mTORC1 stimulates cell growth through SAM synthesis and m 6 A mRNA-dependent control of protein synthesis.
- Author
-
Villa E, Sahu U, O'Hara BP, Ali ES, Helmin KA, Asara JM, Gao P, Singer BD, and Ben-Sahra I
- Subjects
- Adenosine metabolism, Animals, Base Sequence, Breast Neoplasms metabolism, Breast Neoplasms pathology, Cell Cycle Proteins metabolism, Cell Proliferation, Female, HEK293 Cells, HeLa Cells, Humans, Methionine Adenosyltransferase genetics, Methionine Adenosyltransferase metabolism, Methylation, Mice, Nude, Proto-Oncogene Proteins c-myc metabolism, RNA Splicing Factors metabolism, RNA, Messenger genetics, RNA, Messenger metabolism, Signal Transduction, Transcription, Genetic, Mice, Adenosine analogs & derivatives, Mechanistic Target of Rapamycin Complex 1 metabolism, Protein Biosynthesis, S-Adenosylmethionine metabolism
- Abstract
The mechanistic target of rapamycin complex 1 (mTORC1) regulates metabolism and cell growth in response to nutrient, growth, and oncogenic signals. We found that mTORC1 stimulates the synthesis of the major methyl donor, S-adenosylmethionine (SAM), through the control of methionine adenosyltransferase 2 alpha (MAT2A) expression. The transcription factor c-MYC, downstream of mTORC1, directly binds to intron 1 of MAT2A and promotes its expression. Furthermore, mTORC1 increases the protein abundance of Wilms' tumor 1-associating protein (WTAP), the positive regulatory subunit of the human N
6 -methyladenosine (m6 A) RNA methyltransferase complex. Through the control of MAT2A and WTAP levels, mTORC1 signaling stimulates m6 A RNA modification to promote protein synthesis and cell growth. A decline in intracellular SAM levels upon MAT2A inhibition decreases m6 A RNA modification, protein synthesis rate, and tumor growth. Thus, mTORC1 adjusts m6 A RNA modification through the control of SAM and WTAP levels to prime the translation machinery for anabolic cell growth., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2021 Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
37. De novo purine biosynthesis is a major driver of chemoresistance in glioblastoma.
- Author
-
Shireman JM, Atashi F, Lee G, Ali ES, Saathoff MR, Park CH, Savchuk S, Baisiwala S, Miska J, Lesniak MS, James CD, Stupp R, Kumthekar P, Horbinski CM, Ben-Sahra I, and Ahmed AU
- Subjects
- Animals, Antineoplastic Agents, Alkylating pharmacology, Brain Neoplasms pathology, Drug Resistance, Neoplasm drug effects, Enzyme Inhibitors pharmacology, Gene Expression Regulation, Neoplastic drug effects, Glioblastoma pathology, Heterografts, Humans, Mice, Mice, Nude, Mycophenolic Acid pharmacology, Temozolomide pharmacology, Tumor Cells, Cultured, Brain Neoplasms metabolism, Drug Resistance, Neoplasm physiology, Gene Expression Regulation, Neoplastic physiology, Glioblastoma metabolism, Purines biosynthesis
- Abstract
Glioblastoma is a primary brain cancer with a near 100% recurrence rate. Upon recurrence, the tumour is resistant to all conventional therapies, and because of this, 5-year survival is dismal. One of the major drivers of this high recurrence rate is the ability of glioblastoma cells to adapt to complex changes within the tumour microenvironment. To elucidate this adaptation's molecular mechanisms, specifically during temozolomide chemotherapy, we used chromatin immunoprecipitation followed by sequencing and gene expression analysis. We identified a molecular circuit in which the expression of ciliary protein ADP-ribosylation factor-like protein 13B (ARL13B) is epigenetically regulated to promote adaptation to chemotherapy. Immuno-precipitation combined with liquid chromatography-mass spectrometry binding partner analysis revealed that that ARL13B interacts with the purine biosynthetic enzyme inosine-5'-monophosphate dehydrogenase 2 (IMPDH2). Further, radioisotope tracing revealed that this interaction functions as a negative regulator for purine salvaging. Inhibition of the ARL13B-IMPDH2 interaction enhances temozolomide-induced DNA damage by forcing glioblastoma cells to rely on the purine salvage pathway. Targeting the ARLI3B-IMPDH2 circuit can be achieved using the Food and Drug Administration-approved drug, mycophenolate mofetil, which can block IMPDH2 activity and enhance the therapeutic efficacy of temozolomide. Our results suggest and support clinical evaluation of MMF in combination with temozolomide treatment in glioma patients., (© The Author(s) (2021). Published by Oxford University Press on behalf of the Guarantors of Brain. All rights reserved. For permissions, please email: journals.permissions@oup.com.)
- Published
- 2021
- Full Text
- View/download PDF
38. NOTCH1-driven UBR7 stimulates nucleotide biosynthesis to promote T cell acute lymphoblastic leukemia.
- Author
-
Srivastava S, Sahu U, Zhou Y, Hogan AK, Sathyan KM, Bodner J, Huang J, Wong KA, Khalatyan N, Savas JN, Ntziachristos P, Ben-Sahra I, and Foltz DR
- Subjects
- Humans, Proteomics, T-Lymphocytes pathology, Ubiquitination, Nucleotides biosynthesis, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma metabolism, Receptor, Notch1 genetics, Receptor, Notch1 metabolism, Ubiquitin-Protein Ligases genetics, Ubiquitin-Protein Ligases metabolism
- Abstract
Ubiquitin protein ligase E3 component N-recognin 7 (UBR7) is the most divergent member of UBR box-containing E3 ubiquitin ligases/recognins that mediate the proteasomal degradation of its substrates through the N-end rule. Here, we used a proteomic approach and found phospho r ibosyl pyrophosphate synthetases (PRPSs), the essential enzymes for nucleotide biosynthesis, as strong interacting partners of UBR7. UBR7 stabilizes PRPS catalytic subunits by mediating the polyubiquitination-directed degradation of PRPS-associated protein (PRPSAP), the negative regulator of PRPS. Loss of UBR7 leads to nucleotide biosynthesis defects. We define UBR7 as a transcriptional target of NOTCH1 and show that UBR7 is overexpressed in NOTCH1-driven T cell acute lymphoblastic leukemia (T-ALL). Impaired nucleotide biosynthesis caused by UBR7 depletion was concomitant with the attenuated cell proliferation and oncogenic potential of T-ALL. Collectively, these results establish UBR7 as a critical regulator of nucleotide metabolism through the regulation of the PRPS enzyme complex and uncover a metabolic vulnerability in NOTCH1-driven T-ALL., (Copyright © 2021 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works. Distributed under a Creative Commons Attribution License 4.0 (CC BY).)
- Published
- 2021
- Full Text
- View/download PDF
39. ERK2 Phosphorylates PFAS to Mediate Posttranslational Control of De Novo Purine Synthesis.
- Author
-
Ali ES, Sahu U, Villa E, O'Hara BP, Gao P, Beaudet C, Wood AW, Asara JM, and Ben-Sahra I
- Subjects
- A549 Cells, Animals, Carbon-Nitrogen Ligases with Glutamine as Amide-N-Donor genetics, Cell Cycle physiology, Cell Line, Tumor, Cell Proliferation genetics, Extracellular Signal-Regulated MAP Kinases metabolism, HeLa Cells, Humans, MAP Kinase Signaling System physiology, Phosphorylation, Purines metabolism, Signal Transduction physiology, ras Proteins metabolism, Carbon-Nitrogen Ligases with Glutamine as Amide-N-Donor metabolism, Mitogen-Activated Protein Kinase 1 metabolism, Purines biosynthesis
- Abstract
The RAS-ERK/MAPK (RAS-extracellular signal-regulated kinase/mitogen-activated protein kinase) pathway integrates growth-promoting signals to stimulate cell growth and proliferation, at least in part, through alterations in metabolic gene expression. However, examples of direct and rapid regulation of the metabolic pathways by the RAS-ERK pathway remain elusive. We find that physiological and oncogenic ERK signaling activation leads to acute metabolic flux stimulation through the de novo purine synthesis pathway, thereby increasing building block availability for RNA and DNA synthesis, which is required for cell growth and proliferation. We demonstrate that ERK2, but not ERK1, phosphorylates the purine synthesis enzyme PFAS (phosphoribosylformylglycinamidine synthase) at T619 in cells to stimulate de novo purine synthesis. The expression of nonphosphorylatable PFAS (T619A) decreases purine synthesis, RAS-dependent cancer cell-colony formation, and tumor growth. Thus, ERK2-mediated PFAS phosphorylation facilitates the increase in nucleic acid synthesis required for anabolic cell growth and proliferation., Competing Interests: Declaration of Interests The authors declare no competing interests., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
40. PGC1α Inhibits Polyamine Synthesis to Suppress Prostate Cancer Aggressiveness.
- Author
-
Kaminski L, Torrino S, Dufies M, Djabari Z, Haider R, Roustan FR, Jaune E, Laurent K, Nottet N, Michiels JF, Gesson M, Rocchi S, Mazure NM, Durand M, Tanti JF, Ambrosetti D, Clavel S, Ben-Sahra I, and Bost F
- Subjects
- Aged, Aged, 80 and over, Animals, Apoptosis, Biomarkers, Tumor genetics, Cell Proliferation, Dicarboxylic Acid Transporters genetics, Follow-Up Studies, Humans, Male, Mice, Mice, Nude, Middle Aged, Mitochondria metabolism, Mitochondria pathology, Mitochondrial Membrane Transport Proteins genetics, Neoplasm Metastasis, Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha genetics, Prognosis, Prostatic Neoplasms genetics, Prostatic Neoplasms metabolism, Proto-Oncogene Proteins c-myc genetics, Signal Transduction, Survival Rate, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Biomarkers, Tumor metabolism, Dicarboxylic Acid Transporters metabolism, Gene Expression Regulation, Neoplastic, Mitochondrial Membrane Transport Proteins metabolism, Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha metabolism, Polyamines metabolism, Prostatic Neoplasms pathology, Proto-Oncogene Proteins c-myc metabolism
- Abstract
Although tumorigenesis is dependent on the reprogramming of cellular metabolism, the metabolic pathways engaged in the formation of metastases remain largely unknown. The transcriptional coactivator peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α) plays a pleiotropic role in the control of cancer cell metabolism and has been associated with a good prognosis in prostate cancer. Here, we show that PGC1α represses the metastatic properties of prostate cancer cells via modulation of the polyamine biosynthesis pathway. Mechanistically, PGC1α inhibits the expression of c-MYC and ornithine decarboxylase 1 (ODC1), the rate-limiting enzyme for polyamine synthesis. Analysis of in vivo metastases and clinical data from patients with prostate cancer support the proposition that the PGC1α/c-MYC/ODC1 axis regulates polyamine biosynthesis and prostate cancer aggressiveness. In conclusion, downregulation of PGC1α renders prostate cancer cells dependent on polyamine to promote metastasis. SIGNIFICANCE: These findings show that a major regulator of mitochondrial metabolism controls polyamine synthesis and prostate cancer aggressiveness, with potential applications in therapy and identification of new biomarkers., (©2019 American Association for Cancer Research.)
- Published
- 2019
- Full Text
- View/download PDF
41. Cancer Cells Tune the Signaling Pathways to Empower de Novo Synthesis of Nucleotides.
- Author
-
Villa E, Ali ES, Sahu U, and Ben-Sahra I
- Abstract
Cancer cells exhibit a dynamic metabolic landscape and require a sufficient supply of nucleotides and other macromolecules to grow and proliferate. To meet the metabolic requirements for cell growth, cancer cells must stimulate de novo nucleotide synthesis to obtain adequate nucleotide pools to support nucleic acid and protein synthesis along with energy preservation, signaling activity, glycosylation mechanisms, and cytoskeletal function. Both oncogenes and tumor suppressors have recently been identified as key molecular determinants for de novo nucleotide synthesis that contribute to the maintenance of homeostasis and the proliferation of cancer cells. Inactivation of tumor suppressors such as TP53 and LKB1 and hyperactivation of the mTOR pathway and of oncogenes such as MYC , RAS , and AKT have been shown to fuel nucleotide synthesis in tumor cells. The molecular mechanisms by which these signaling hubs influence metabolism, especially the metabolic pathways for nucleotide synthesis, continue to emerge. Here, we focus on the current understanding of the molecular mechanisms by which oncogenes and tumor suppressors modulate nucleotide synthesis in cancer cells and, based on these insights, discuss potential strategies to target cancer cell proliferation.
- Published
- 2019
- Full Text
- View/download PDF
42. Efferocytosis Fuels Requirements of Fatty Acid Oxidation and the Electron Transport Chain to Polarize Macrophages for Tissue Repair.
- Author
-
Zhang S, Weinberg S, DeBerge M, Gainullina A, Schipma M, Kinchen JM, Ben-Sahra I, Gius DR, Yvan-Charvet L, Chandel NS, Schumacker PT, and Thorp EB
- Subjects
- Animals, Cytophagocytosis, Electron Transport, Humans, Inflammation metabolism, Jurkat Cells, Macrophages cytology, Mice, Mice, Inbred C57BL, Oxidation-Reduction, Wound Healing, Fatty Acids metabolism, Interleukin-10 metabolism, Macrophages metabolism, Mitochondria metabolism, NAD metabolism
- Abstract
During wound injury, efferocytosis fills the macrophage with a metabolite load nearly equal to the phagocyte itself. A timely question pertains to how metabolic phagocytic signaling regulates the signature anti-inflammatory macrophage response. Here we report the metabolome of activated macrophages during efferocytosis to reveal an interleukin-10 (IL-10) cytokine escalation that was independent of glycolysis yet bolstered by apoptotic cell fatty acids and mitochondrial β-oxidation, the electron transport chain, and heightened coenzyme NAD
+ . Loss of IL-10 due to mitochondrial complex III defects was remarkably rescued by adding NAD+ precursors. This activated a SIRTUIN1 signaling cascade, largely independent of ATP, that culminated in activation of IL-10 transcription factor PBX1. Il-10 activation by the respiratory chain was also important in vivo, as efferocyte mitochondrial dysfunction led to cardiac rupture after myocardial injury. These findings highlight a new paradigm whereby macrophages leverage efferocytic metabolites and electron transport for anti-inflammatory reprogramming that culminates in organ repair., (Copyright © 2018 Elsevier Inc. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
43. HER2 Signaling Hijacks the Creatine Shuttle to Fuel Breast Cancer Cell Growth.
- Author
-
Ben-Sahra I and Puissant A
- Subjects
- Creatine, Creatine Kinase, Mitochondrial Form, Humans, Phosphorylation, Tyrosine, Breast Neoplasms, Receptor, ErbB-2
- Abstract
Alteration of cell energy metabolism represents a major determinant of cancer progression; however, our understanding of oncogenic mechanisms underlying this rewiring remains elusive. In this issue of Cell Metabolism, Kurmi et al. (2018) show that HER2 signaling promotes ABL-mediated phosphorylation of the mitochondrial creatine kinase (MtCK1), providing ATP to support breast tumor growth., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
44. mRNA-binding protein tristetraprolin is essential for cardiac response to iron deficiency by regulating mitochondrial function.
- Author
-
Sato T, Chang HC, Bayeva M, Shapiro JS, Ramos-Alonso L, Kouzu H, Jiang X, Liu T, Yar S, Sawicki KT, Chen C, Martínez-Pastor MT, Stumpo DJ, Schumacker PT, Blackshear PJ, Ben-Sahra I, Puig S, and Ardehali H
- Subjects
- Animals, Cell Line, Electron Transport Complex III genetics, Electron Transport Complex III metabolism, Iron-Sulfur Proteins genetics, Mice, Mice, Knockout, Mitochondria, Heart enzymology, NADH Dehydrogenase genetics, Oxidation-Reduction, Tristetraprolin genetics, Iron Deficiencies, Iron-Sulfur Proteins metabolism, Mitochondria, Heart metabolism, Myocardium metabolism, NADH Dehydrogenase metabolism, Tristetraprolin metabolism
- Abstract
Cells respond to iron deficiency by activating iron-regulatory proteins to increase cellular iron uptake and availability. However, it is not clear how cells adapt to conditions when cellular iron uptake does not fully match iron demand. Here, we show that the mRNA-binding protein tristetraprolin (TTP) is induced by iron deficiency and degrades mRNAs of mitochondrial Fe/S-cluster-containing proteins, specifically Ndufs1 in complex I and Uqcrfs1 in complex III, to match the decrease in Fe/S-cluster availability. In the absence of TTP, Uqcrfs1 levels are not decreased in iron deficiency, resulting in nonfunctional complex III, electron leakage, and oxidative damage. Mice with deletion of Ttp display cardiac dysfunction with iron deficiency, demonstrating that TTP is necessary for maintaining cardiac function in the setting of low cellular iron. Altogether, our results describe a pathway that is activated in iron deficiency to regulate mitochondrial function to match the availability of Fe/S clusters., Competing Interests: The authors declare no conflict of interest.
- Published
- 2018
- Full Text
- View/download PDF
45. Amino Acid Restriction Triggers Angiogenesis via GCN2/ATF4 Regulation of VEGF and H 2 S Production.
- Author
-
Longchamp A, Mirabella T, Arduini A, MacArthur MR, Das A, Treviño-Villarreal JH, Hine C, Ben-Sahra I, Knudsen NH, Brace LE, Reynolds J, Mejia P, Tao M, Sharma G, Wang R, Corpataux JM, Haefliger JA, Ahn KH, Lee CH, Manning BD, Sinclair DA, Chen CS, Ozaki CK, and Mitchell JR
- Subjects
- Activating Transcription Factor 4 antagonists & inhibitors, Activating Transcription Factor 4 genetics, Amino Acids, Sulfur metabolism, Animals, Cystathionine gamma-Lyase metabolism, Disease Models, Animal, Female, Human Umbilical Vein Endothelial Cells, Humans, Hypoxia-Inducible Factor 1, alpha Subunit antagonists & inhibitors, Hypoxia-Inducible Factor 1, alpha Subunit genetics, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Ischemia metabolism, Ischemia pathology, Male, Mice, Mice, Inbred C57BL, Neovascularization, Physiologic, Physical Conditioning, Animal, RNA Interference, RNA, Small Interfering metabolism, Vascular Endothelial Growth Factor A genetics, Activating Transcription Factor 4 metabolism, Amino Acids, Sulfur deficiency, Hydrogen Sulfide metabolism, Protein Serine-Threonine Kinases metabolism, Vascular Endothelial Growth Factor A metabolism
- Abstract
Angiogenesis, the formation of new blood vessels by endothelial cells (ECs), is an adaptive response to oxygen/nutrient deprivation orchestrated by vascular endothelial growth factor (VEGF) upon ischemia or exercise. Hypoxia is the best-understood trigger of VEGF expression via the transcription factor HIF1α. Nutrient deprivation is inseparable from hypoxia during ischemia, yet its role in angiogenesis is poorly characterized. Here, we identified sulfur amino acid restriction as a proangiogenic trigger, promoting increased VEGF expression, migration and sprouting in ECs in vitro, and increased capillary density in mouse skeletal muscle in vivo via the GCN2/ATF4 amino acid starvation response pathway independent of hypoxia or HIF1α. We also identified a requirement for cystathionine-γ-lyase in VEGF-dependent angiogenesis via increased hydrogen sulfide (H
2 S) production. H2 S mediated its proangiogenic effects in part by inhibiting mitochondrial electron transport and oxidative phosphorylation, resulting in increased glucose uptake and glycolytic ATP production., (Copyright © 2018 Elsevier Inc. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
46. The mTORC1 Signaling Network Senses Changes in Cellular Purine Nucleotide Levels.
- Author
-
Hoxhaj G, Hughes-Hallett J, Timson RC, Ilagan E, Yuan M, Asara JM, Ben-Sahra I, and Manning BD
- Subjects
- A549 Cells, AMP-Activated Protein Kinases metabolism, Animals, Cell Line, Dihydroorotate Dehydrogenase, Enzyme Inhibitors pharmacology, Fluorouracil pharmacology, HeLa Cells, Humans, Mercaptopurine pharmacology, Methotrexate pharmacology, Mice, Oxidoreductases Acting on CH-CH Group Donors antagonists & inhibitors, Oxidoreductases Acting on CH-CH Group Donors genetics, Oxidoreductases Acting on CH-CH Group Donors metabolism, Phosphoribosylglycinamide Formyltransferase antagonists & inhibitors, Phosphoribosylglycinamide Formyltransferase genetics, Phosphoribosylglycinamide Formyltransferase metabolism, RNA Interference, Ribosomal Protein S6 Kinases, 70-kDa metabolism, Signal Transduction drug effects, Thymidylate Synthase antagonists & inhibitors, Thymidylate Synthase genetics, Thymidylate Synthase metabolism, Tuberous Sclerosis Complex 2 Protein, Tumor Suppressor Proteins antagonists & inhibitors, Tumor Suppressor Proteins genetics, Tumor Suppressor Proteins metabolism, Mechanistic Target of Rapamycin Complex 1 metabolism, Purine Nucleotides metabolism
- Abstract
Mechanistic (or mammalian) target of rapamycin complex 1 (mTORC1) integrates signals from growth factors and nutrients to control biosynthetic processes, including protein, lipid, and nucleic acid synthesis. We find that the mTORC1 pathway is responsive to changes in purine nucleotides in a manner analogous to its sensing of amino acids. Depletion of cellular purines, but not pyrimidines, inhibits mTORC1, and restoration of intracellular adenine nucleotides via addition of exogenous purine nucleobases or nucleosides acutely reactivates mTORC1. Adenylate sensing by mTORC1 is dependent on the tuberous sclerosis complex (TSC) protein complex and its regulation of Rheb upstream of mTORC1, but independent of energy stress and AMP-activated protein kinase (AMPK). Even though mTORC1 signaling is not acutely sensitive to changes in intracellular guanylates, long-term depletion of guanylates decreases Rheb protein levels. Our findings suggest that nucleotide sensing, like amino acid sensing, enables mTORC1 to tightly coordinate nutrient availability with the synthesis of macromolecules, such as protein and nucleic acids, produced from those nutrients., (Copyright © 2017 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
47. Rapamycin-induced miR-21 promotes mitochondrial homeostasis and adaptation in mTORC1 activated cells.
- Author
-
Lam HC, Liu HJ, Baglini CV, Filippakis H, Alesi N, Nijmeh J, Du H, Lope AL, Cottrill KA, Handen A, Asara JM, Kwiatkowski DJ, Ben-Sahra I, Oldham WM, Chan SY, and Henske EP
- Abstract
mTORC1 hyperactivation drives the multi-organ hamartomatous disease tuberous sclerosis complex (TSC). Rapamycin inhibits mTORC1, inducing partial tumor responses; however, the tumors regrow following treatment cessation. We discovered that the oncogenic miRNA, miR-21, is increased in Tsc2-deficient cells and, surprisingly, further increased by rapamycin. To determine the impact of miR-21 in TSC, we inhibited miR-21 in vitro . miR-21 inhibition significantly repressed the tumorigenic potential of Tsc2-deficient cells and increased apoptosis sensitivity. Tsc2-deficient cells' clonogenic and anchorage independent growth were reduced by ∼50% ( p <0.01) and ∼75% ( p <0.0001), respectively, and combined rapamycin treatment decreased soft agar growth by ∼90% ( p <0.0001). miR-21 inhibition also increased sensitivity to apoptosis. Through a network biology-driven integration of RNAseq data, we discovered that miR-21 promotes mitochondrial adaptation and homeostasis in Tsc2-deficient cells. miR-21 inhibition reduced mitochondrial polarization and function in Tsc2-deficient cells, with and without co-treatment with rapamycin. Importantly, miR-21 inhibition limited Tsc2-deficient tumor growth in vivo , reducing tumor size by approximately 3-fold ( p <0.0001). When combined with rapamcyin, miR-21 inhibition showed even more striking efficacy, both during treatment and after treatment cessation, with a 4-fold increase in median survival following rapamycin cessation ( p =0.0008). We conclude that miR-21 promotes mTORC1-driven tumorigenesis via a mechanism that involves the mitochondria, and that miR-21 is a potential therapeutic target for TSC-associated hamartomas and other mTORC1-driven tumors, with the potential for synergistic efficacy when combined with rapalogs., Competing Interests: CONFLICTS OF INTEREST The authors declare no potential conflicts of interest.
- Published
- 2017
- Full Text
- View/download PDF
48. The energy disruptor metformin targets mitochondrial integrity via modification of calcium flux in cancer cells.
- Author
-
Loubiere C, Clavel S, Gilleron J, Harisseh R, Fauconnier J, Ben-Sahra I, Kaminski L, Laurent K, Herkenne S, Lacas-Gervais S, Ambrosetti D, Alcor D, Rocchi S, Cormont M, Michiels JF, Mari B, Mazure NM, Scorrano L, Lacampagne A, Gharib A, Tanti JF, and Bost F
- Subjects
- Animals, Cell Line, Tumor, Endoplasmic Reticulum drug effects, Endoplasmic Reticulum metabolism, Endoplasmic Reticulum Stress drug effects, Humans, Mice, Mitochondria drug effects, Mitochondria ultrastructure, Mitochondrial Membrane Transport Proteins metabolism, Mitochondrial Permeability Transition Pore, Mitochondrial Swelling drug effects, Models, Biological, Organelle Biogenesis, Calcium metabolism, Energy Metabolism drug effects, Metformin pharmacology, Mitochondria metabolism
- Abstract
Mitochondrial integrity is critical for the regulation of cellular energy and apoptosis. Metformin is an energy disruptor targeting complex I of the respiratory chain. We demonstrate that metformin induces endoplasmic reticulum (ER) stress, calcium release from the ER and subsequent uptake of calcium into the mitochondria, thus leading to mitochondrial swelling. Metformin triggers the disorganization of the cristae and inner mitochondrial membrane in several cancer cells and tumors. Mechanistically, these alterations were found to be due to calcium entry into the mitochondria, because the swelling induced by metformin was reversed by the inhibition of mitochondrial calcium uniporter (MCU). We also demonstrated that metformin inhibits the opening of mPTP and induces mitochondrial biogenesis. Altogether, the inhibition of mPTP and the increase in mitochondrial biogenesis may account for the poor pro-apoptotic effect of metformin in cancer cells.
- Published
- 2017
- Full Text
- View/download PDF
49. The creatine kinase pathway is a metabolic vulnerability in EVI1-positive acute myeloid leukemia.
- Author
-
Fenouille N, Bassil CF, Ben-Sahra I, Benajiba L, Alexe G, Ramos A, Pikman Y, Conway AS, Burgess MR, Li Q, Luciano F, Auberger P, Galinsky I, DeAngelo DJ, Stone RM, Zhang Y, Perkins AS, Shannon K, Hemann MT, Puissant A, and Stegmaier K
- Subjects
- Adult, Aged, Aged, 80 and over, Blotting, Western, Computer Simulation, Core Binding Factor Alpha 2 Subunit metabolism, Creatine Kinase metabolism, Female, Flow Cytometry, Gene Expression Profiling, Genome-Wide Association Study, Humans, Leukemia, Myeloid, Acute metabolism, MDS1 and EVI1 Complex Locus Protein, Male, Metabolic Networks and Pathways, Metabolomics, Middle Aged, Mitochondria, Proto-Oncogene Mas, RNA, Small Interfering, Core Binding Factor Alpha 2 Subunit genetics, Creatine Kinase genetics, DNA-Binding Proteins genetics, Gene Expression Regulation, Neoplastic genetics, Leukemia, Myeloid, Acute genetics, Proto-Oncogenes genetics, Transcription Factors genetics
- Abstract
Expression of the MECOM (also known as EVI1) proto-oncogene is deregulated by chromosomal translocations in some cases of acute myeloid leukemia (AML) and is associated with poor clinical outcome. Here, through transcriptomic and metabolomic profiling of hematopoietic cells, we reveal that EVI1 overexpression alters cellular metabolism. A screen using pooled short hairpin RNAs (shRNAs) identified the ATP-buffering, mitochondrial creatine kinase CKMT1 as necessary for survival of EVI1-expressing cells in subjects with EVI1-positive AML. EVI1 promotes CKMT1 expression by repressing the myeloid differentiation regulator RUNX1. Suppression of arginine-creatine metabolism by CKMT1-directed shRNAs or by the small molecule cyclocreatine selectively decreased the viability, promoted the cell cycle arrest and apoptosis of human EVI1-positive cell lines, and prolonged survival in both orthotopic xenograft models and mouse models of primary AML. CKMT1 inhibition altered mitochondrial respiration and ATP production, an effect that was abrogated by phosphocreatine-mediated reactivation of the arginine-creatine pathway. Targeting CKMT1 is thus a promising therapeutic strategy for this EVI1-driven AML subtype that is highly resistant to current treatment regimens.
- Published
- 2017
- Full Text
- View/download PDF
50. Akt-mTORC1 signaling regulates Acly to integrate metabolic input to control of macrophage activation.
- Author
-
Covarrubias AJ, Aksoylar HI, Yu J, Snyder NW, Worth AJ, Iyer SS, Wang J, Ben-Sahra I, Byles V, Polynne-Stapornkul T, Espinosa EC, Lamming D, Manning BD, Zhang Y, Blair IA, and Horng T
- Subjects
- Acetylation, Animals, Cell Proliferation, Chemokines metabolism, Gene Expression Regulation, Histones metabolism, Interleukin-4 metabolism, Macrophages physiology, Mechanistic Target of Rapamycin Complex 1, Mice, Inbred C57BL, Protein Processing, Post-Translational, ATP Citrate (pro-S)-Lyase metabolism, Macrophage Activation, Macrophages metabolism, Multiprotein Complexes metabolism, Proto-Oncogene Proteins c-akt metabolism, Signal Transduction, TOR Serine-Threonine Kinases metabolism
- Abstract
Macrophage activation/polarization to distinct functional states is critically supported by metabolic shifts. How polarizing signals coordinate metabolic and functional reprogramming, and the potential implications for control of macrophage activation, remains poorly understood. Here we show that IL-4 signaling co-opts the Akt-mTORC1 pathway to regulate Acly, a key enzyme in Ac-CoA synthesis, leading to increased histone acetylation and M2 gene induction. Only a subset of M2 genes is controlled in this way, including those regulating cellular proliferation and chemokine production. Moreover, metabolic signals impinge on the Akt-mTORC1 axis for such control of M2 activation. We propose that Akt-mTORC1 signaling calibrates metabolic state to energetically demanding aspects of M2 activation, which may define a new role for metabolism in supporting macrophage activation.
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.