Category: Moms

Glutamine and metabolism

Glutamine and metabolism

Fat metabolism foods reprogramming and cancer progression. A proposed Gultamine for ketabolism in cancer Fat metabolism foods growth ajd Fat metabolism foods resistance. Article CAS PubMed Google Scholar Fat metabolism foods RJ, Mancuso A, Daikhin E, Nissim I, Yudkoff M, Wehrli S, Thompson CB. In hepatocellular carcinoma HCCGLS2 inhibited proliferation in vitro and lung metastasis in a xenograft mouse model by inhibiting the small GTPase Rac1 Cancer Commun London England. Reinfeld, B.

Glutamine and metabolism -

Kremer, D. GOT1 inhibition promotes pancreatic cancer cell death by ferroptosis. Zhang, L. Acta Pharmacol. PubMed PubMed Central Google Scholar. Bhutia, Y. Glutamine transporters in mammalian cells and their functions in physiology and cancer.

Acta , — Targeting ASCT2-mediated glutamine uptake blocks prostate cancer growth and tumour development. Willems, L. Inhibiting glutamine uptake represents an attractive new strategy for treating acute myeloid leukemia.

Blood , — Cha, Y. Amino acid transporters and glutamine metabolism in breast cancer. Hassanein, M. SLC1A5 mediates glutamine transport required for lung cancer cell growth and survival. Scalise, M. Cysteine of the ASCT2 amino acid transporter is a molecular determinant of the antiport mechanism.

Sastrasinh, M. Effect of acute pH change on mitochondrial glutamine transport. CAS PubMed Google Scholar. Li, X. Role of glutamine and its metabolite ammonia in crosstalk of cancer-associated fibroblasts and cancer cells. van Geldermalsen, M.

Oncogene 35 , — Kahya, U. Amino Acid Transporters on the Guard of Cell Genome and Epigenome. Zhou, X. Activated amino acid response pathway generates apatinib resistance by reprograming glutamine metabolism in non-small-cell lung cancer.

Tameire, F. ATF4 couples MYC-dependent translational activity to bioenergetic demands during tumour progression. Ren, P. ATF4 and N-Myc coordinate glutamine metabolism in MYCN-amplified neuroblastoma cells through ASCT2 activation.

Yoo, H. A variant of SLC1A5 is a mitochondrial glutamine transporter for metabolic reprogramming in cancer cells. Cell Metab 31 , — e12 Schulte, M. Pharmacological blockade of ASCT2-dependent glutamine transport leads to antitumor efficacy in preclinical models.

Sun, H. S-benzyl-cysteine-mediated cell cycle arrest and apoptosis involving activation of mitochondrial-dependent caspase cascade through the p53 pathway in human gastric cancer SGC cells.

Asian Pac. Cancer Prev. Benzylserine inhibits breast cancer cell growth by disrupting intracellular amino acid homeostasis and triggering amino acid response pathways.

BMC Cancer 18 , Ma, H. Inhibition of glutamine uptake improves the efficacy of cetuximab on gastric cancer. Bröer, A. Disruption of amino acid homeostasis by novel ASCT2 inhibitors involves multiple targets. Suzuki, M. Nachef, M. Deletion of amino acid transporter ASCT2 SLC1A5 reveals an essential role for transporters SNAT1 SLC38A1 and SNAT2 SLC38A2 to sustain glutaminolysis in cancer cells.

Böhme-Schäfer, I. Wang, K. BMC Cancer 13 , Zhou, F. SLC38A1 promotes proliferation and migration of human colorectal cancer cells. Huazhong Univ. Okudaira, H. Putative transport mechanism and intracellular fate of transaminoF-fluorocyclobutanecarboxylic acid in human prostate cancer.

Morotti, M. Cancer , — Tang, Y. The regulatory role of meaib in protein metabolism and the mTOR signaling pathway in porcine enterocytes. Zhang, Z. ASCT2 SLC1A5 -dependent glutamine uptake is involved in the progression of head and neck squamous cell carcinoma.

Cancer , 82—93 Sloan, J. Dejure, F. Exploiting the metabolic dependencies of the broad amino acid transporter SLC6A Oncotarget 11 , — Sniegowski, T. SLC6A14 and SLC38A5 drive the glutaminolysis and serine-glycine-one-carbon pathways in cancer.

Pharmaceuticals Basel 14 , Gupta, N. Basis Dis. Sikder, M. J , — Babu, E. Deletion of the amino acid transporter Slc6a14 suppresses tumour growth in spontaneous mouse models of breast cancer. J , 17—23 Kou, L. Expert Opin. Drug Deliv.

Nałęcz, K. Penheiter, A. Transcriptomic and immunohistochemical profiling of SLC6A14 in pancreatic ductal adenocarcinoma. Lu, Y. Blockade of the amino acid transporter SLC6A14 suppresses tumor growth in colorectal Cancer.

BMC Cancer 22 , Coothankandaswamy, V. Amino acid transporter SLC6A14 is a novel and effective drug target for pancreatic cancer. Karunakaran, S. Cai, A. Synergism between SLC6A14 blockade and gemcitabine in pancreactic cancer: a 1H-NMR-based metabolomic study in pancreatic cancer cells.

Cheng, T. Pyruvate carboxylase is required for glutamine-independent growth of tumor cells. Natl Acad. USA , — Mendez-Lucas, A. Identifying strategies to target the metabolic flexibility of tumours. Zhang, G. Integration of metabolomics and transcriptomics revealed a fatty acid network exerting growth inhibitory effects in human pancreatic cancer.

Tajan, M. A role for p53 in the adaptation to glutamine starvation through the expression of SLC1A3. e6 Lowman, X. p53 promotes cancer cell adaptation to glutamine deprivation by upregulating Slc7a3 to increase arginine uptake.

Cell Rep. e4 Zhang, J. Asparagine plays a critical role in regulating cellular adaptation to glutamine depletion. Cell 56 , — Pavlova, N.

As extracellular glutamine levels decline, asparagine becomes an essential amino acid. e5 Macropinocytosis is an alternative pathway of cysteine acquisition and mitigates sorafenib-induced ferroptosis in hepatocellular carcinoma.

Jeong, S. Transcription factor Eb is required for macropinocytosis-mediated growth recovery of nutrient-deprived kras-mutant cells. Commisso, C. Macropinocytosis of protein is an amino acid supply route in Ras-transformed cells. Lee, S. EGFR-PAK signaling selectively regulates glutamine deprivation-induced macropinocytosis.

Cell 50 , — Zhang, M. Hypoxia-induced macropinocytosis represents a metabolic route for liver cancer. A positive feedback loop between Sestrin2 and mTORC2 is required for the survival of glutamine-depleted lung cancer cells.

Reid, M. The B55alpha subunit of PP2A drives a pdependent metabolic adaptation to glutamine deprivation. Cell 50 , — Ishak Gabra, M. IKKbeta activates p53 to promote cancer cell adaptation to glutamine deprivation. Oncogenesis 7 , 93 Abcouwer, S. Glutamine deprivation induces the expression of GADD45 and GADD primarily by mRNA stabilization.

Tran, T. Tumor-associated mutant p53 promotes cancer cell survival upon glutamine deprivation through p21 induction. Oncogene 36 , — Reinfeld, B. Cell-programmed nutrient partitioning in the tumour microenvironment. Chang, C. Metabolic competition in the tumor microenvironment is a driver of cancer progression.

Cell , — Trends Mol. Pearce, E. Fueling immunity: insights into metabolism and lymphocyte function. Science , Carr, E.

Immunol , — Wang, R. The transcription factor Myc controls metabolic reprogramming upon T lymphocyte activation. Immunity 35 , — Nakaya, M. Inflammatory T cell responses rely on amino acid transporter ASCT2 facilitation of glutamine uptake and mTORC1 kinase activation.

Immunity 40 , — Klysz, D. Glutamine-dependent alpha-ketoglutarate production regulates the balance between T helper 1 cell and regulatory T cell generation. Signal 8 , ra97 Matias, M. Regulatory T cell differentiation is controlled by alphaKG-induced alterations in mitochondrial metabolism and lipid homeostasis.

Blagih, J. The energy sensor AMPK regulates T cell metabolic adaptation and effector responses in vivo. Immunity 42 , 41—54 Loftus, R. Amino acid-dependent cMyc expression is essential for NK cell metabolic and functional responses in mice.

Terrén, I. NK cell metabolism and tumor microenvironment. Keppel, M. Activation-specific metabolic requirements for NK Cell IFN-γ production.

Jha, A. Network integration of parallel metabolic and transcriptional data reveals metabolic modules that regulate macrophage polarization. Immunity 42 , — Liu, P. alpha-ketoglutarate orchestrates macrophage activation through metabolic and epigenetic reprogramming. Palmieri, E.

Pharmacologic or genetic targeting of glutamine synthetase skews macrophages toward an M1-like phenotype and inhibits tumor metastasis. Simultaneous glutamine metabolism and PD-L1 inhibition to enhance suppression of triple-negative breast cancer. Leone, R. Glutamine blockade induces divergent metabolic programs to overcome tumor immune evasion.

Science , — The glutaminase inhibitor CB Telaglenastat enhances the antimelanoma activity of T-cell-mediated immunotherapies. Wang, L. Immunosuppression induced by glutamine deprivation occurs via activating PD-L1 transcription in bladder cancer.

Xiong, J. SLC1A1 mediated glutamine addiction and contributed to natural killer T-cell lymphoma progression with immunotherapeutic potential.

EBioMedicine 72 , Oh, M. Targeting glutamine metabolism enhances tumor-specific immunity by modulating suppressive myeloid cells.

Sharma, N. Targeting tumor-intrinsic hexosamine biosynthesis sensitizes pancreatic cancer to anti-PD1 therapy. Download references. This work was supported by National Research Foundation of Korea NRF grants NRFR1A2C, R1A5A, NRFR1A2C, and NRFR1C1C funded by the Ministry of Science and ICT.

Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, , South Korea. BK21 FOUR Community-based Intelligent Novel Drug Discovery Education Unit, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu, , Korea.

Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, , Korea.

You can also search for this author in PubMed Google Scholar. Correspondence to Yeon-Kyung Choi or Keun-Gyu Park. Open Access This article is licensed under a Creative Commons Attribution 4.

Reprints and permissions. Jin, J. Targeting glutamine metabolism as a therapeutic strategy for cancer. Exp Mol Med 55 , — Download citation.

Received : 22 November Revised : 06 January Accepted : 09 January Published : 03 April Issue Date : April Anyone you share the following link with will be able to read this content:.

Sorry, a shareable link is not currently available for this article. Provided by the Springer Nature SharedIt content-sharing initiative. Skip to main content Thank you for visiting nature.

Download PDF. Subjects Cancer metabolism. Abstract Proliferating cancer cells rely largely on glutamine for survival and proliferation. Introduction Metabolic reprogramming, a hallmark of cancer cells, is a process by which cancer cells ensure a sufficient supply of proteins, nucleotides, and lipids to support rapid growth and proliferation 1.

The role of glutamine in cancer cell growth Rapidly proliferating cancer cells take up glutamine from plasma via various amino acid transporters, and then it is converted to glutamate in the mitochondria by the two forms of glutaminase: kidney-type glutaminase GLS 1 and liver-type GLS2 8.

Full size image. Targeting glutaminase and transaminase as a treatment for cancer GLS, which is highly expressed in cancer cells and plays a role in cancer progression, has been investigated extensively as a druggable target Role of glutamine metabolism in redox homeostasis ROS levels are elevated persistently in proliferating cancer cells, and ROS damage DNA and cellular components; therefore, redox homeostasis plays a pivotal role in protecting cancer cells against them.

Targeting redox homeostasis for cancer treatment Approximately one-third of glutamine taken up by human fibroblast cells is exchanged for cysteine by SLC7A11 59 , Targeting glutamine transporters as a treatment for cancer Cancer cells require an abundant supply of glutamine from the extracellular milieu; therefore, upregulation of glutamine transporters SLC1A5, SLC38A1, SLC38A2, and SLC6A14 at the cell membrane is required Fig.

SLC1A5 SLC1A5 ASCT2 is an obligatory sodium-dependent transporter of neutral amino acids, which are exchanged for asparagine, threonine, or serine SLC38A1 and SLC38A2 SLC38A1 SNAT1 and SLC38A2 SNAT2 are sodium-dependent neutral amino acid transporters that drive glutamine influx into cells Mechanisms that induce resistance to glutamine-targeting therapies Although targeting glutamine metabolism is a promising therapeutic approach, few drugs have been developed.

Effects of targeting glutamine metabolism in the TME The TME is a complex milieu that surrounds tumor cells, often providing immunosuppressive cover that facilitates immune invasion. Glutamine metabolism in immune cells Accumulating evidence shows that glutamine is an immunomodulatory nutrient in immune cells.

Glutamine blockade in the TME The metabolism of cancer cells and immune cells in the TME is regulated by cell-intrinsic programs through mTORC1 signaling Conclusion Glutamine metabolism plays a central role in regulating uncontrolled tumor growth by modulating bioenergetic and redox homeostasis and by serving as a precursor for the synthesis of biomass.

References Ward, P. Article CAS PubMed PubMed Central Google Scholar Stine, Z. Article CAS PubMed Google Scholar Wang, Z.

Article PubMed PubMed Central Google Scholar Cruzat, V. Article PubMed PubMed Central Google Scholar Altman, B. Article CAS PubMed PubMed Central Google Scholar Matés, J. Article PubMed Google Scholar Ma, G. Article CAS PubMed PubMed Central Google Scholar Choi, Y.

Article CAS PubMed Google Scholar Grinde, M. Article PubMed PubMed Central Google Scholar Xiang, L. Article PubMed PubMed Central Google Scholar Gao, P. Article CAS PubMed PubMed Central Google Scholar Csibi, A. Article CAS PubMed PubMed Central Google Scholar Zhang, C.

Article PubMed PubMed Central Google Scholar Saha, S. Article CAS PubMed PubMed Central Google Scholar Dias, M. Article CAS PubMed Google Scholar Xiang, L.

Article CAS Google Scholar López de la Oliva, A. Article PubMed PubMed Central Google Scholar Cardaci, S. Article PubMed PubMed Central Google Scholar Jin, L. Article CAS PubMed PubMed Central Google Scholar Yelamanchi, S.

Article PubMed Google Scholar Vanhove, K. Article PubMed PubMed Central Google Scholar Yang, M. A similar study has shown that the high expression of GLS1 in HCC had a markedly shorter overall survival time than its low expression Prostate cancer Pca treatments, such as radiation, chemotherapy, and hormone therapy, can induce autophagy that improves therapeutic resistance 70 — Existing evidence has linked the Gln metabolism to autophagy through oxidative homeostasis in Pca.

For instance, a recently published article showed that the radio-resistant Pca cells strongly rely on Gln metabolism to maintain oxidative homeostasis.

However, Pca cells could trigger autophagy upon Gln withdrawal and do not exhibit significant radio-sensitization Upon further investigations, the researchers found that the ionizing radiation-derived ROS can induce autophagy as a stress response of Pca cells, but it is neutralized by GSH and NADPH produced by Gln metabolism.

When blocking Gln metabolism, Pca cells could activate the ATG -mediated autophagy as a survival strategy to withstand radiation-induced damage due to GSH depletion and ROS accumulation 73 , Consistently, other studies also confirmed that autophagy inhibition increases ROS production in Pca cells 75 — Overall, Gln metabolism affects the autophagy of Pca cells by affecting the level of ROS.

There is growing evidence that clear cell renal cell carcinoma cells ccRCCs are Gln-addicted that is reprogrammed to feed an intrinsic antioxidant system 82 — To further confirm the role of Gln as a source for the GSH pathway, absolute quantitative GSH and GSSG levels in cells grown with and without Gln were compared.

The result showed that GSH and GSSG levels were markedly reduced in the Gln-depleted group, which confirms the necessity of Gln for maintaining oxidative homeostasis of ccRCCs In addition, an interesting study shows that the suppression of fatty acid metabolism by inhibition of β-oxidation lead to the RCC cells dependent on the Gln-GSH pathway to prevent lipid peroxidation and ferroptosis Notably, high GSH levels have proven to be a key feature of high-grade, high-stage and metastatic ccRCCs 81 , All in all, these data suggest that Gln-dependent antioxidant effects may provide ccRCCs with a critical mechanism for their survival.

In general, Gln is an antioxidant defense only in Gln addicted cancers, but not in all cases. Oligodendroglioma cells lack Gln synthetase a marker of Gln-addicted cancers , but are independent of extracellular Gln thus are not Gln addicted 89 , However, a previous study showed that small amounts of extracellular Gln are sufficient for oligodendroglioma cells growth.

Gln starvation does not significantly affect the cell content of anaplerotic substrates, but causes a significant decrease in the intracellular content of GSH in oligodendroglioma cells This result means that Gln addiction and Gln roles as antioxidants are not correlated.

In light of the findings mentioned above, it would seem reasonable to expect that Gln metabolism plays an important role in maintaining ROS levels in cancer cells.

However, we noted that most of the above-mentioned studies have mainly focused on the effects of Gln metabolism on maintaining oxidative homeostasis of cancer cells, whereas these effects were not suitable for every situation.

Some studies have shown that the anaplerotic role of Gln metabolism in replenishing the TCA cycle intermediates could enhance ROS production under the blocking of GSH synthesis 92 — For instance, a recently published article showed that Gln metabolism was crucial to maintaining cystine starvation-induced mitochondrial membrane potential MMP hyperpolarization, accompanied by an increase in electron transfer chain ETC activity and lipid ROS generation to promote ferroptosis In support of this notion, data from various studies showed that inhibiting the glutaminolysis can suppress TCA cycle and MMP hyperpolarization, and reduce lipid ROS production, thus enhancing ferroptosis resistance 95 — Similarly, various studies showed that inhibiting xCT activities could suppress Gln-derived Glu export and enhance Glu to replenish the TCA cycle intermediates 99 — Therefore, it has been theorized that inhibition of xCT activities could promote Glu to replenish the TCA cycle intermediates, which could promote ROS production Figure 4.

All in all, increasing ROS levels by Gln metabolism under blocking of GSH synthesis promoted ferroptosis, which may provide a novel treatment guideline for ferroptosis-based tumor therapy.

Figure 4 Gln metabolism promotes ROS production through the TCA cycle. PUFA-PLs, Polyunsaturated fatty acid chain s. The demonstration of the link between Gln metabolism and oxidative homeostasis of cancer has prompted research into strategies to target Gln metabolism to damage oxidative homeostasis of cancer.

In this regard, GLS inhibitors aimed at decreasing Gln metabolism and impairing oxidative homeostasis are attracting increasing clinical interest. Many small molecules have been assayed to block GLS isoenzymes after the first attempt and failure to use 6-diazooxo-L-norleucine DON as an anti-cancer drug , The bis 5-phenylacetamido-1,2,4-thiadiazolyl ethyl sulfide BPTES and CB are the specific inhibitors most frequently Notably, CB is currently being administered to humans in phase 1 clinical trials for some types of cancers 49 , — However, because of the plasticity of adaptive metabolic reprogramming in cancer cells, successful single treatments against cancers are scarce 4 , — Therefore, some specific inhibitor of Gln metabolism has reached better results in sensitizing cancer cells to other treatments Targeting Gln metabolism combined with drugs that are strong inducers of mitochondrial ROS, is widely used for treating multiple cancers Table 1.

For instance, dihydroartemisinin cooperatively induces excessive intracellular ROS resulting in profound apoptosis when combined with CB in HCC In a similar study, Gregory et al. demonstrated that a combination of GLS inhibition with ATO or HHT showed great activity against AML Preclinical studies have also reported a benefit when combined with Gln metabolism inhibitors and radiotherapy.

For example, the inhibitor CB increased GSH depletion, and enhanced the radiation sensitivity of lung tumor cells xenografts in mice Interestingly, one recent study showed that the combination of Gln metabolism inhibitors with radiotherapy could activate the ATG5-mediated autophagy of Prostate cancer, and proposes a strategy that a combination with autophagy inhibition and the blockade of Gln metabolism makes Pca radio-sensitization 73 , 74 , However, it remains controversial whether antioxidants affect treatment outcomes or whether antioxidants ameliorate adverse effects induced by chemotherapy and radiotherapy, which needs further investigations in the future In conclusion, combination therapy, including inhibitors of Gln metabolism, may be a promising strategy for cancer cells.

Table 1 Combined treatments: targeting glutaminolysis in combination with drugs that unbalance mitochondrial redox state. The antioxidant capacity of tumor cells is required for rapidly proliferating and aggressive cancer cells to adapt to hypoxia and excessive ROS levels.

The literature reviewed here suggests that Gln has been established as an important factor in maintaining the oxidative homeostasis of cancer cells. Targeting Gln metabolism impaired oxidative homeostasis of cancer cells and may provide effective approaches for therapies against cancer.

Future studies on Gln metabolism in maintaining oxidative homeostasis may provide novel and effective therapeutic strategies to treat a subset of cancer patients. Conceptualization, YD, WL, and TG; writing—original draft preparation, TG, CZ, XO, JZ, JY, and SC; writing—review and editing, YD and WL; visualization, YD.

All authors have read and agreed to the published version of the manuscript. The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Kussmaul L, Hirst J. The mechanism of superoxide production by NADH:ubiquinone oxidoreductase complex I from bovine heart mitochondria. Proc Natl Acad Sci USA — doi: PubMed Abstract CrossRef Full Text Google Scholar. Lambeth JD. NOX enzymes and the biology of reactive oxygen.

Nat Rev Immunol —9. Quinlan CL, Orr AL, Perevoshchikova IV, Treberg JR, Ackrell BA, Brand MD. Mitochondrial complex II can generate reactive oxygen species at high rates in both the forward and reverse reactions.

J Biol Chem — Gorrini C, Harris IS, Mak TW. Modulation of oxidative stress as an anticancer strategy. Nat Rev Drug Discovery. CrossRef Full Text Google Scholar. Panieri E, Santoro MM.

ROS homeostasis and metabolism: a dangerous liason in cancer cells. Cell Death Disease. Kröller-Schön S, Steven S, Kossmann S, Scholz A, Daub S, Oelze M, et al. Molecular mechanisms of the crosstalk between mitochondria and NADPH oxidase through reactive oxygen species-studies in white blood cells and in animal models.

Antioxidants Redox Signaling — Warburg O. On the origin of cancer cells. Sci New York NY. Sosa V, Moliné T, Somoza R, Paciucci R, Kondoh H, Lleonart ME.

Oxidative stress and cancer: An overview. Ageing Res Rev — Herraiz C, Crosas-Molist E, Sanz-Moreno V.

Reactive oxygen species and tumor dissemination: Allies no longer. Mol Cell Oncol 3:e Maryanovich M, Gross A. A ROS rheostat for cell fate regulation. Trends Cell Biol — Hayes JD, Dinkova-Kostova AT, Tew KD.

Oxidative stress in cancer. Cancer Cell — Kodama M, Oshikawa K, Shimizu H, Yoshioka S, Takahashi M, Izumi Y, et al.

A shift in glutamine nitrogen metabolism contributes to the malignant progression of cancer. Nat Commun DeBerardinis RJ, Cheng T. Q's next: the diverse functions of glutamine in metabolism, cell biology and cancer.

Oncogene — Altman BJ, Stine ZE, Dang CV. From Krebs to clinic: glutamine metabolism to cancer therapy. Nat Rev Cancer. Franklin CC, Backos DS, Mohar I, White CC, Forman HJ, Kavanagh TJ. Structure, function, and post-translational regulation of the catalytic and modifier subunits of glutamate cysteine ligase.

Mol aspects Med — Kennedy L, Sandhu JK, Harper ME, Cuperlovic-Culf M. Role of glutathione in cancer: From mechanisms to therapies. Biomolecules Lu SC.

Regulation of glutathione synthesis. van Geldermalsen M, Wang Q, Nagarajah R, Marshall AD, Thoeng A, Gao D, et al. Oncogene —8. Jiang H, Zhang N, Tang T, Feng F, Sun H, Qu W. Pharmacol Res Bhutia YD, Ganapathy V. Glutamine transporters in mammalian cells and their functions in physiology and cancer.

Biochim Biophys Acta —9. Gao P, Tchernyshyov I, Chang TC, Lee YS, Kita K, Ochi T, et al. Nature —5. Hu W, Zhang C, Wu R, Sun Y, Levine A, Feng Z. Glutaminase 2, a novel p53 target gene regulating energy metabolism and antioxidant function. Lukey MJ, Greene KS, Erickson JW, Wilson KF, Cerione RA.

The oncogenic transcription factor c-jun regulates glutaminase expression and sensitizes cells to glutaminase-targeted therapy. Suzuki S, Tanaka T, Poyurovsky MV, Nagano H, Mayama T, Ohkubo S, et al. Phosphate-activated glutaminase GLS2 , a pinducible regulator of glutamine metabolism and reactive oxygen species.

Proc Natl Acad Sci USA —6. Zhang J, Pavlova NN, Thompson CB. Cancer cell metabolism: the essential role of the nonessential amino acid, glutamine. EMBO J — Koppula P, Zhang Y, Zhuang L, Gan B. Cancer Commun London England. DeBerardinis RJ, Mancuso A, Daikhin E, Nissim I, Yudkoff M, Wehrli S, et al.

Beyond aerobic glycolysis: transformed cells can engage in glutamine metabolism that exceeds the requirement for protein and nucleotide synthesis. Ying M, You D, Zhu X, Cai L, Zeng S, Hu X. Lactate and glutamine support NADPH generation in cancer cells under glucose deprived conditions.

Redox Biol Son J, Lyssiotis CA, Ying H, Wang X, Hua S, Ligorio M, et al. Glutamine supports pancreatic cancer growth through a KRAS-regulated metabolic pathway. Moreno-Sánchez R, Gallardo-Pérez JC, Rodríguez-Enríquez S, Saavedra E, Marín-Hernández Á. Control of the NADPH supply for oxidative stress handling in cancer cells.

Free Radic Biol Med. Bansal A, Simon MC. Glutathione metabolism in cancer progression and treatment resistance. J Cell Biol —8. Jaganjac M, Milkovic L, Sunjic SB, Zarkovic N. The NRF2, thioredoxin, and glutathione system in tumorigenesis and anticancer therapies.

Antioxidants Basel Switzerland. Palde PB, Carroll KS. A universal entropy-driven mechanism for thioredoxin-target recognition. Proc Natl Acad Sci USA —5. Mizrahi JD, Surana R, Valle JW, Shroff RT. Pancreatic cancer. Lancet London England. Siegel RL, Miller KD, Fuchs HE, Jemal A.

Cancer statistics, CA: Cancer J Clin — Park W, Chawla A, O'Reilly EM. Pancreatic cancer: A review. Jama — Li W, Chen C, Zhao X, Ye H, Zhao Y, Fu Z, et al. J Cell Mol Med — Sign In or Create an Account. Navbar Search Filter Plant and Cell Physiology This issue Molecular and Cell Biology Plant Sciences and Forestry Books Journals Oxford Academic Mobile Enter search term Search.

Issues More Content Advance Articles Editor's Choice Commentaries Editorials Reviews Special Issues Rapid Papers Submit Author Guidelines Submission Site Open Access Options Author Benefits Purchase Alerts About About Plant and Cell Physiology About the Japanese Society of Plant Physiologists Editorial Board Permissions Reviewers Advertising and Corporate Services Journals Career Network Self-Archiving Policy Contact Us Journals on Oxford Academic Books on Oxford Academic.

Issues More Content Advance Articles Editor's Choice Commentaries Editorials Reviews Special Issues Rapid Papers Submit Author Guidelines Submission Site Open Access Options Author Benefits Purchase Alerts About About Plant and Cell Physiology About the Japanese Society of Plant Physiologists Editorial Board Permissions Reviewers Advertising and Corporate Services Journals Career Network Self-Archiving Policy Contact Us Close Navbar Search Filter Plant and Cell Physiology This issue Molecular and Cell Biology Plant Sciences and Forestry Books Journals Oxford Academic Enter search term Search.

Advanced Search. Search Menu. Article Navigation. Close mobile search navigation Article Navigation. Volume Journal Article. Glutamine Metabolism, Sensing and Signaling in Plants Get access. Kim-Teng Lee , Kim-Teng Lee.

Institute of Plant and Microbial Biology, Academia Sinica. Molecular and Biological Agricultural Sciences, The Taiwan International Graduate Program, Academia Sinica. Oxford Academic. Google Scholar. Hong-Sheng Liao. Ming-Hsiun Hsieh. Revision received:. Editorial decision:.

Corrected and typeset:. Select Format Select format. ris Mendeley, Papers, Zotero. enw EndNote. bibtex BibTex. txt Medlars, RefWorks Download citation. Permissions Icon Permissions. Close Navbar Search Filter Plant and Cell Physiology This issue Molecular and Cell Biology Plant Sciences and Forestry Books Journals Oxford Academic Enter search term Search.

Abstract Glutamine Gln is the first amino acid synthesized in nitrogen N assimilation in plants. Published by Oxford University Press on behalf of Japanese Society of Plant Physiologists. All rights reserved.

For permissions, please e-mail: journals. permissions oup. Issue Section:. You do not currently have access to this article. Download all slides. Sign in Get help with access. Japanese Society of Plant Physiologists members Sign in through society site.

Qnd Gln is the Heart health support services amino acid synthesized in ane N assimilation Refuel and recharge plants. Plants have multiple GS isoenzymes that work Glutaminf or cooperatively to ensure adn the Gln supply Glutamine and metabolism sufficient for plant growth metabolsm development under Glutamine and metabolism Gutamine. Gln Glutqmine a Glutamine and metabolism block for protein synthesis and an N-donor for the biosynthesis of amino acids, nucleic acids, amino sugars and vitamin B coenzymes. Most reactions using Gln as an N-donor are catalyzed by Gln amidotransferase GAT that hydrolyzes Gln to Glu and transfers the amido group of Gln to an acceptor substrate. Several GAT domain—containing proteins of unknown function in the reference plant Arabidopsis thaliana suggest that some metabolic fates of Gln have yet to be identified in plants. In addition to metabolism, Gln signaling has emerged in recent years. Glutamine and metabolism

Glutamine and metabolism -

A variant of SLC1A5 Is a mitochondrial glutamine transporter for metabolic reprogramming in cancer cells. Cell Metab. Altman BJ, Stine ZE, Dang CV.

From Krebs to clinic: glutamine metabolism to cancer therapy. Nat Rev Cancer. Huang W, Choi W, Chen Y, Zhang Q, Deng H, He W, Shi Y.

A proposed role for glutamine in cancer cell growth through acid resistance. Cell Res. Yang T, Yan X, Cao Y, Bao T, Li G, Gu S, Xiong K, Xiao T. Meta-analysis of glutamine on immune function and post-operative complications of patients with colorectal cancer.

Front Nutr. Article PubMed PubMed Central CAS Google Scholar. Kaur BP, Secord E. Innate Immunity. Pediatr Clin North Am. Article PubMed Google Scholar. Bonilla FA, Oettgen HC.

Adaptive immunity. J Allergy Clin Immunol. Terry S, Engelsen AST, Buart S, Elsayed WS, Venkatesh GH, Chouaib S. Hypoxia-driven intratumor heterogeneity and immune evasion.

Cancer Lett. Huang B, Song BL. Cholesterol metabolism in cancer: mechanisms and therapeutic opportunities. Nat Metab. Chen B, Gao A, Tu B, Wang Y, Yu X, Wang Y, Xiu Y, Wang B, Wan Y, Huang Y. Metabolic modulation via mTOR pathway and anti-angiogenesis remodels tumor microenvironment using PD-L1-targeting codelivery.

Karayama M, Masuda J, Mori K, Yasui H, Hozumi H, Suzuki Y, Furuhashi K, Fujisawa T, Enomoto N, Nakamura Y, et al. Comprehensive assessment of multiple tryptophan metabolites as potential biomarkers for immune checkpoint inhibitors in patients with non-small cell lung cancer.

Clin Transl Oncol. Yan Y, Chang L, Tian H, Wang L, Zhang Y, Yang T, Li G, Hu W, Shah K, Chen G, Guo Y. J Immunother Cancer. Metabolic competition in the tumor microenvironment is a driver of cancer progression.

Edwards DN, Ngwa VM, Raybuck AL, Wang S, Hwang Y, Kim LC, Cho SH, Paik Y, Wang Q, Zhang S, et al. Selective glutamine metabolism inhibition in tumor cells improves antitumor T lymphocyte activity in triple-negative breast cancer. J Clin Invest.

Wang JX, Choi SYC, Niu X, Kang N, Xue H, Killam J, Wang Y. Lactic acid and an acidic tumor microenvironment suppress anticancer immunity. Int J Mol Sci. Article CAS PubMed Central Google Scholar. Ma G, Li C, Zhang Z, Liang Y, Liang Z, Chen Y, Wang L, Li D, Zeng M, Shan W, Niu H.

Front Oncol. Carrascosa JM, Martínez P, Núñez de Castro I. Nitrogen movement between host and tumor in mice inoculated with Ehrlich ascitic tumor cells.

Cancer Res. CAS PubMed Google Scholar. Son J, Lyssiotis CA, Ying H, Wang X, Hua S, Ligorio M, Perera RM, Ferrone CR, Mullarky E, Shyh-Chang N, et al. Glutamine supports pancreatic cancer growth through a KRAS-regulated metabolic pathway.

Zhang J, Pavlova NN, Thompson CB. Cancer cell metabolism: the essential role of the nonessential amino acid, glutamine. EMBO J. Daye D, Wellen KE. Metabolic reprogramming in cancer: unraveling the role of glutamine in tumorigenesis.

Semin Cell Dev Biol. Welbourne TC. Ammonia production and glutamine incorporation into glutathione in the functioning rat kidney. Can J Biochem. DeBerardinis RJ, Mancuso A, Daikhin E, Nissim I, Yudkoff M, Wehrli S, Thompson CB. Beyond aerobic glycolysis: transformed cells can engage in glutamine metabolism that exceeds the requirement for protein and nucleotide synthesis.

Proc Natl Acad Sci USA. Cheong H, Lindsten T, Wu J, Lu C, Thompson CB. Eng CH, Yu K, Lucas J, White E, Abraham RT. Ammonia derived from glutaminolysis is a diffusible regulator of autophagy. Sci Signal. Nicklin P, Bergman P, Zhang B, Triantafellow E, Wang H, Nyfeler B, Yang H, Hild M, Kung C, Wilson C, et al.

Bidirectional transport of amino acids regulates mTOR and autophagy. Yuneva M, Zamboni N, Oefner P, Sachidanandam R, Lazebnik Y. Deficiency in glutamine but not glucose induces MYC-dependent apoptosis in human cells. J Cell Biol. Gao P, Tchernyshyov I, Chang TC, Lee YS, Kita K, Ochi T, Zeller KI, De Marzo AM, Van Eyk JE, Mendell JT, Dang CV.

Wise DR, DeBerardinis RJ, Mancuso A, Sayed N, Zhang XY, Pfeiffer HK, Nissim I, Daikhin E, Yudkoff M, McMahon SB, Thompson CB. Myc regulates a transcriptional program that stimulates mitochondrial glutaminolysis and leads to glutamine addiction.

Suzuki S, Tanaka T, Poyurovsky MV, Nagano H, Mayama T, Ohkubo S, Lokshin M, Hosokawa H, Nakayama T, Suzuki Y, et al. Phosphate-activated glutaminase GLS2 , a pinducible regulator of glutamine metabolism and reactive oxygen species. Jackson JG, Lozano G. The mutant p53 mouse as a pre-clinical model.

Tran TQ, Lowman XH, Reid MA, Mendez-Dorantes C, Pan M, Yang Y, Kong M. Tumor-associated mutant p53 promotes cancer cell survival upon glutamine deprivation through p21 induction. Pylayeva-Gupta Y, Grabocka E, Bar-Sagi D.

RAS oncogenes: weaving a tumorigenic web. Dilshara MG, Jeong JW, Prasad Tharanga Jayasooriya RG, Neelaka Molagoda IM, Lee S, Park SR, Choi YH, Kim GY. Glutamine deprivation sensitizes human breast cancer MDA-MB cells to TRIAL-mediated apoptosis.

Biochem Biophys Res Commun. Gaglio D, Soldati C, Vanoni M, Alberghina L, Chiaradonna F. Glutamine deprivation induces abortive s-phase rescued by deoxyribonucleotides in k-ras transformed fibroblasts. PLoS ONE. Gaglio D, Metallo CM, Gameiro PA, Hiller K, Danna LS, Balestrieri C, Alberghina L, Stephanopoulos G, Chiaradonna F.

Oncogenic K-Ras decouples glucose and glutamine metabolism to support cancer cell growth. Mol Syst Biol. Talks KL, Turley H, Gatter KC, Maxwell PH, Pugh CW, Ratcliffe PJ, Harris AL. The expression and distribution of the hypoxia-inducible factors HIF-1alpha and HIF-2alpha in normal human tissues, cancers, and tumor-associated macrophages.

Am J Pathol. Faubert B, Vincent EE, Griss T, Samborska B, Izreig S, Svensson RU, Mamer OA, Avizonis D, Shackelford DB, Shaw RJ, Jones RG. Loss of the tumor suppressor LKB1 promotes metabolic reprogramming of cancer cells via HIF-1α. Gordan JD, Bertout JA, Hu CJ, Diehl JA, Simon MC. HIF-2alpha promotes hypoxic cell proliferation by enhancing c-myc transcriptional activity.

Cancer Cell. Wang JB, Erickson JW, Fuji R, Ramachandran S, Gao P, Dinavahi R, Wilson KF, Ambrosio AL, Dias SM, Dang CV, Cerione RA. Targeting mitochondrial glutaminase activity inhibits oncogenic transformation.

Biswas SK, Mantovani A. Orchestration of metabolism by macrophages. Ghesquière B, Wong BW, Kuchnio A, Carmeliet P. Metabolism of stromal and immune cells in health and disease. Article PubMed CAS Google Scholar. Pearce EL, Pearce EJ. Metabolic pathways in immune cell activation and quiescence.

Hao S, Yan KK, Ding L, Qian C, Chi H, Yu J. Network approaches for dissecting the immune system. Article CAS Google Scholar. Ricciardi S, Manfrini N, Alfieri R, Calamita P, Crosti MC, Gallo S, Müller R, Pagani M, Abrignani S, Biffo S. Pearce EL, Poffenberger MC, Chang CH, Jones RG.

Fueling immunity: insights into metabolism and lymphocyte function. Krawczyk CM, Holowka T, Sun J, Blagih J, Amiel E, DeBerardinis RJ, Cross JR, Jung E, Thompson CB, Jones RG, Pearce EJ. Toll-like receptor-induced changes in glycolytic metabolism regulate dendritic cell activation.

Newsholme P. Why is L-glutamine metabolism important to cells of the immune system in health, postinjury, surgery or infection? J Nutr. Curi R, Newsholme P, Marzuca-Nassr GN, Takahashi HK, Hirabara SM, Cruzat V, Krause M, de Bittencourt Jr PI.

Regulatory principles in metabolism-then and now. Biochem J. Cruzat V, Macedo Rogero M. Glutamine: metabolism and immune function, supplementation and clinical translation. Article PubMed Central CAS Google Scholar.

Curi R, Lagranha CJ, Doi SQ, Sellitti DF, Procopio J, Pithon-Curi TC, Corless M, Newsholme P. Molecular mechanisms of glutamine action. J Cell Physiol. Curi R, Lagranha CJ, Doi SQ, Sellitti DF, Procopio J, Pithon-Curi TC. Glutamine-dependent changes in gene expression and protein activity.

Cell Biochem Funct. Roth E, Oehler R, Manhart N, Exner R, Wessner B, Strasser E, Spittler A. Regulative potential of glutamine—relation to glutathione metabolism. Hiscock N, Petersen EW, Krzywkowski K, Boza J, Halkjaer-Kristensen J, Pedersen BK.

Glutamine supplementation further enhances exercise-induced plasma IL J Appl Physiol. Google Scholar. Crawford J, Cohen HJ. The essential role of L-glutamine in lymphocyte differentiation in vitro. Liu PS, Wang H, Li X, Chao T, Teav T, Christen S, Di Conza G, Cheng WC, Chou CH, Vavakova M, et al.

α-ketoglutarate orchestrates macrophage activation through metabolic and epigenetic reprogramming. Nat Immunol. Kolb D, Kolishetti N. Metabolic modulation of the tumor microenvironment leads to multiple checkpoint inhibition and immune cell infiltration.

ACS Nano. Palmer CS, Ostrowski M, Balderson B, Christian N, Crowe SM. Glucose metabolism regulates T cell activation, differentiation, and functions. Front Immunol. Sukumar M, Roychoudhuri R, Restifo NP. Nutrient competition: a new axis of tumor immunosuppression.

Fu Q, Xu L, Wang Y, Jiang Q, Liu Z, Zhang J, Zhou Q, Zeng H, Tong S, Wang T, et al. Tumor-associated macrophage-derived interleukin interlinks kidney cancer glutamine addiction with immune evasion.

Eur Urol. Carr EL, Kelman A, Wu GS, Gopaul R, Senkevitch E, Aghvanyan A, Turay AM, Frauwirth KA. J Immunol. Yang R, Li X, Wu Y, Zhang G, Liu X, Li Y, Bao Y, Yang W. Yang WH, Qiu Y, Stamatatos O, Janowitz T, Lukey MJ. Enhancing the efficacy of glutamine metabolism inhibitors in cancer therapy.

Trends Cancer. Reinfeld BI, Madden MZ. Cell-programmed nutrient partitioning in the tumour microenvironment. Yang L, Achreja A, Yeung TL, Mangala LS, Jiang D, Han C, Baddour J, Marini JC, Ni J, Nakahara R, et al. Targeting Stromal Glutamine synthetase in tumors disrupts tumor microenvironment-regulated cancer cell growth.

Fan SJ, Kroeger B. Glutamine deprivation alters the origin and function of cancer cell exosomes. Ma G, Liang Y, Chen Y, Wang L, Li D, Liang Z, Wang X, Tian D, Yang X, Niu H.

Mol Cancer Res. Wang L, Xu T, Yang X, Liang Z, Zhang J, Li D, Chen Y, Ma G, Wang Y, Liang Y, Niu H. Immunosuppression Induced by glutamine deprivation occurs via activating PD-L1 transcription in bladder cancer.

Front Mol Biosci. Byun JK, Park M, Lee S, Yun JW, Lee J, Kim JS, Cho SJ, Jeon HJ, Lee IK, Choi YK, Park KG.

Inhibition of glutamine utilization synergizes with immune checkpoint inhibitor to promote antitumor immunity. Mol Cell. Qing G, Li B, Vu A, Skuli N, Walton ZE, Liu X, Mayes PA, Wise DR, Thompson CB, Maris JM, et al.

ATF4 regulates MYC-mediated neuroblastoma cell death upon glutamine deprivation. Csibi A, Lee G, Yoon SO, Tong H, Ilter D, Elia I, Fendt SM, Roberts TM, Blenis J. Curr Biol.

Casey SC, Tong L, Li Y, Do R, Walz S, Fitzgerald KN, Gouw AM, Baylot V, Gütgemann I, Eilers M, Felsher DW. MYC regulates the antitumor immune response through CD47 and PD-L1.

Mukhopadhyay S, Vander Heiden MG, McCormick F. The metabolic landscape of RAS-driven cancers from biology to therapy.

Nat Cancer. Coelho MA, de Carné TS, Rana S, Zecchin D, Moore C, Molina-Arcas M, East P, Spencer-Dene B, Nye E, Barnouin K, et al. Oncogenic RAS signaling promotes tumor immunoresistance by stabilizing PD-L1 mRNA. Chen J, Jiang CC, Jin L, Zhang XD. Regulation of PD-L1: a novel role of pro-survival signalling in cancer.

Ann Oncol. Ni JM, Ni AP. Chin Med Sci J. Kim J. Regulation of immune cell functions by metabolic reprogramming. J Immunol Res. Bailey SR, Nelson MH, Himes RA, Li Z, Mehrotra S, Paulos CM.

Th17 cells in cancer: the ultimate identity crisis. T cell metabolism drives immunity. J Exp Med. Swamy M, Pathak S, Grzes KM, Damerow S, Sinclair LV, van Aalten DM.

Glucose and glutamine fuel protein O-GlcNAcylation to control T cell self-renewal and malignancy. Johnson MO, Wolf MM, Madden MZ, Andrejeva G, Sugiura A, Contreras DC, Maseda D, Liberti MV, Paz K, Kishton RJ, et al.

Distinct regulation of Th17 and Th1 cell differentiation by glutaminase-dependent metabolism. Xiao M, Yang H, Xu W, Ma S, Lin H, Zhu H, Liu L, Liu Y, Yang C, Xu Y, et al. Inhibition of α-KG-dependent histone and DNA demethylases by fumarate and succinate that are accumulated in mutations of FH and SDH tumor suppressors.

Genes Dev. Amino acid-dependent cMyc expression is essential for NK cell metabolic and functional responses in mice. Nat Commun. Lin SC, Hardie DG. AMPK: sensing glucose as well as cellular energy status.

Blagih J, Coulombe F, Vincent EE, Dupuy F, Galicia-Vázquez G, Yurchenko E, Raissi TC, van der Windt GJ, Viollet B, Pearce EL, et al. The energy sensor AMPK regulates T cell metabolic adaptation and effector responses in vivo. Macrophage plasticity and interaction with lymphocyte subsets: cancer as a paradigm.

Xue J, Schmidt SV, Sander J, Draffehn A, Krebs W, Quester I, De Nardo D, Gohel TD, Emde M, Schmidleithner L, et al. Transcriptome-based network analysis reveals a spectrum model of human macrophage activation. Murray PJ, Allen JE, Biswas SK, Fisher EA, Gilroy DW, Goerdt S, Gordon S, Hamilton JA, Ivashkiv LB, Lawrence T, et al.

Macrophage activation and polarization: nomenclature and experimental guidelines. Franklin RA, Liao W, Sarkar A, Kim MV, Bivona MR, Liu K, Pamer EG, Li MO.

The cellular and molecular origin of tumor-associated macrophages. Mantovani A, Sozzani S, Locati M, Allavena P, Sica A. Macrophage polarization: tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes.

Trends Immunol. Qian BZ, Pollard JW. Macrophage diversity enhances tumor progression and metastasis. Biswas SK, Allavena P, Mantovani A. Tumor-associated macrophages: functional diversity, clinical significance, and open questions.

Semin Immunopathol. Tannahill GM, Curtis AM, Adamik J, Palsson-McDermott EM, McGettrick AF, Goel G, Frezza C, Bernard NJ, Kelly B, Foley NH, et al. Succinate is an inflammatory signal that induces IL-1β through HIF-1α.

Jha AK, Huang SC, Sergushichev A, Lampropoulou V, Ivanova Y, Loginicheva E, Chmielewski K, Stewart KM, Ashall J, Everts B, et al.

Network integration of parallel metabolic and transcriptional data reveals metabolic modules that regulate macrophage polarization. Ren W, Xia Y, Chen S, Wu G, Bazer FW, Zhou B, Tan B, Zhu G, Deng J, Yin Y.

Adv Nutr. Palmieri EM, Menga A, Martín-Pérez R, Quinto A, Riera-Domingo C, De Tullio G, Hooper DC, Lamers WH, Ghesquière B, McVicar DW, et al. Pharmacologic or genetic targeting of glutamine synthetase skews macrophages toward an m1-like phenotype and inhibits tumor metastasis.

Cell Rep. Colegio OR, Chu NQ, Szabo AL, Chu T, Rhebergen AM, Jairam V, Cyrus N, Brokowski CE, Eisenbarth SC, Phillips GM, et al. Functional polarization of tumour-associated macrophages by tumour-derived lactic acid.

Andreu P, Johansson M, Affara NI, Pucci F, Tan T, Junankar S, Korets L, Lam J, Tawfik D, DeNardo DG, et al. FcRgamma activation regulates inflammation-associated squamous carcinogenesis.

de Visser KE, Korets LV, Coussens LM. De novo carcinogenesis promoted by chronic inflammation is B lymphocyte dependent. Le A, Lane AN, Hamaker M, Bose S, Gouw A, Barbi J, Tsukamoto T, Rojas CJ, Slusher BS, Zhang H, et al.

Glucose-independent glutamine metabolism via TCA cycling for proliferation and survival in B cells. Jiang S, Yan W, Wang SE, Baltimore D. Let-7 suppresses B cell activation through restricting the availability of necessary nutrients. Mantovani A, Cassatella MA, Costantini C, Jaillon S.

Neutrophils in the activation and regulation of innate and adaptive immunity. Nat Rev Immunol. Pithon-Curi TC, De Melo MP, Curi R. Glucose and glutamine utilization by rat lymphocytes, monocytes and neutrophils in culture: a comparative study. Pithon-Curi TC, Trezena AG, Tavares-Lima W, Curi R.

Evidence that glutamine is involved in neutrophil function. Pithon-Curi TC, Levada AC, Lopes LR, Doi SQ, Curi R. Glutamine plays a role in superoxide production and the expression of p47phox, p22phox and gp91phox in rat neutrophils.

Clin Sci Lond. Garcia C, Pithon-Curi TC, de Lourdes-Firmano M, Pires de Melo M, Newsholme P, Curi R. Effects of adrenaline on glucose and glutamine metabolism and superoxide production by rat neutrophils.

Branzk N, Lubojemska A, Hardison SE, Wang Q, Gutierrez MG, Brown GD, Papayannopoulos V. Neutrophils sense microbe size and selectively release neutrophil extracellular traps in response to large pathogens. Magill GB, Myers WP, Reilly HC, Putnam RC, Magill JW, Sykes MP, Escher GC, Karnofsky DA, Burchenal JH.

Pharmacological and initial therapeutic observations on 6-diazooxonorleucine DON in human neoplastic disease. Lynch G, Kemeny N, Casper E. Phase II evaluation of DON 6-diazooxo-L-norleucine in patients with advanced colorectal carcinoma. Am J Clin Oncol. Earhart RH, Amato DJ, Chang AY, Borden EC, Shiraki M, Dowd ME, Comis RL, Davis TE, Smith TJ.

Phase II trial of 6-diazooxo-L-norleucine versus aclacinomycin-A in advanced sarcomas and mesotheliomas. Invest New Drugs. Rais R, Jančařík A, Tenora L, Nedelcovych M, Alt J, Englert J, Rojas C, Le A, Elgogary A, Tan J, et al.

Discovery of 6-Diazooxo-l-norleucine DON prodrugs with enhanced CSF delivery in monkeys: a potential treatment for glioblastoma. J Med Chem. Leone RD, Zhao L. Glutamine blockade induces divergent metabolic programs to overcome tumor immune evasion.

Pinkus LM. Glutamine binding sites. Methods Enzymol. Dejure FR, Royla N, Herold S, Kalb J, Walz S, Ade CP, Mastrobuoni G, Vanselow JT, Schlosser A, Wolf E, Kempa S. Haikala HM, Anttila JM, Klefström J.

MYC and AMPK-save energy or die! Front Cell Dev Biol. Kim JW, Dang CV. Oh MH, Sun IH, Zhao L, Leone RD, Sun IM, Xu W, Collins SL, Tam AJ, Blosser RL, Patel CH, et al. Targeting glutamine metabolism enhances tumor-specific immunity by modulating suppressive myeloid cells.

Sharma NS, Gupta VK, Garrido VT, Hadad R, Durden BC, Kesh K, Giri B, Ferrantella A, Dudeja V, Saluja A, Banerjee S. Targeting tumor-intrinsic hexosamine biosynthesis sensitizes pancreatic cancer to anti-PD1 therapy. Robinson MM, McBryant SJ, Tsukamoto T, Rojas C, Ferraris DV, Hamilton SK, Hansen JC, Curthoys NP.

Novel mechanism of inhibition of rat kidney-type glutaminase by bis 5-phenylacetamido-1,2,4-thiadiazolyl ethyl sulfide BPTES. Gross MI, Demo SD, Dennison JB, Chen L, Chernov-Rogan T, Goyal B, Janes JR, Laidig GJ, Lewis ER, Li J, et al.

Antitumor activity of the glutaminase inhibitor CB in triple-negative breast cancer. Mol Cancer Ther.

Hassanein M, Hoeksema MD, Shiota M, Qian J, Harris BK, Chen H, Clark JE, Alborn WE, Eisenberg R, Massion PP. SLC1A5 mediates glutamine transport required for lung cancer cell growth and survival. Clin Cancer Res.

Schulte ML, Fu A, Zhao P, Li J, Geng L, Smith ST, Kondo J, Coffey RJ. Pharmacological blockade of ASCT2-dependent glutamine transport leads to antitumor efficacy in preclinical models. Nat Med. Li Q, Zhong X, Yao W, Yu J, Wang C, Li Z, Lai S, Qu F, Fu X, Huang X, et al.

Inhibitor of glutamine metabolism V promotes ROS-induced autophagic degradation of B7H3 to enhance antitumor immunity. J Biol Chem. Yang S, Wei W, Zhao Q. B7—H3, a checkpoint molecule, as a target for cancer immunotherapy. Int J Biol Sci. In Society for Immunotherapy of Cancer Annual Meeting Download references.

This study was financially supported by the National Natural Science Foundation of China , , Taishan Scholar Program of Shandong Province tsqn , Shinan District Science and Technology Program of Qingdao YY.

Department of Urology, The Affiliated Hospital of Qingdao University, No. Key Laboratory, Department of Urology and Andrology, The Affiliated Hospital of Qingdao University, Qingdao, , China. Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, , China.

You can also search for this author in PubMed Google Scholar. MG, LY and NH collected articles and designed the manuscript. MG wrote the manuscript. MG, LY, ZZL, LP and ZZ prepared tables and figures. ZM, LZ, LD, WL and CY revised and approved the manuscript.

All authors read the manuscript and approved the final manuscript. Correspondence to Ye Liang or Haitao Niu. Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Open Access This article is licensed under a Creative Commons Attribution 4. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material.

If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.

Reprints and permissions. Ma, G. et al. Reprogramming of glutamine metabolism and its impact on immune response in the tumor microenvironment. Cell Commun Signal 20 , Download citation. Received : 06 April Accepted : 31 May Published : 27 July Anyone you share the following link with will be able to read this content:.

Sorry, a shareable link is not currently available for this article. Provided by the Springer Nature SharedIt content-sharing initiative. Skip to main content. Search all BMC articles Search. Download PDF. Abstract Metabolic reprogramming and immune escape play a major role in tumorigenesis.

Video abstract. Overview of glutamine metabolism in tumor and immune cells Glutamine metabolism in tumor cells Glutamine is the most abundant and widely used amino acid in the human body, which is an important source of nitrogen, and the respiratory fuel for tumor cells. Glutamine metabolism in immune cells Metabolic reprogramming plays a major role in: a the activation of immune cells; b the regulation of immune cell phenotype and function; c mounting a robust anti-tumor immune response [ 43 , 44 , 45 ].

Glutamine metabolic association between tumor cells and immune cells Glutamine competition Like cancer cells, immune cells in the TME also undergo metabolic reprogramming [ 44 ]. Full size image. Effects of glutamine metabolism on immune response Metabolic reprogramming of glutamine metabolism in tumor cells and its impact on immune response Glutamine metabolism maintains tumor survival and progression, and is very important for multiple biological processes such as nucleotide synthesis, amino acid production, protein glycosylation modification, extracellular matrix production, epigenetic modifications, maintenance of cellular redox balance, and autophagy [ 68 ].

Conclusions The critical role of glutamine in energy generation and macromolecule synthesis underlies its importance in tumor progression and immune response.

Availability of data and materials Data sharing is not applicable to this article as no new data were created or analyzed in this study. References Hanahan D, Weinberg RA. Article CAS PubMed Google Scholar Faubert B, Solmonson A. Article PubMed PubMed Central Google Scholar Vander Heiden MG, Cantley LC, Thompson CB.

Article CAS PubMed PubMed Central Google Scholar McCarty MF, Whitaker J. PubMed Google Scholar Yoo HC, Park SJ, Nam M, Kang J, Kim K, Yeo JH, Kim JK, Heo Y, Lee HS, Lee MY, et al. Article CAS PubMed Google Scholar Altman BJ, Stine ZE, Dang CV. Article CAS PubMed PubMed Central Google Scholar Huang W, Choi W, Chen Y, Zhang Q, Deng H, He W, Shi Y.

Article CAS PubMed PubMed Central Google Scholar Yang T, Yan X, Cao Y, Bao T, Li G, Gu S, Xiong K, Xiao T. Article PubMed PubMed Central CAS Google Scholar Kaur BP, Secord E.

Article PubMed Google Scholar Bonilla FA, Oettgen HC. Article PubMed Google Scholar Terry S, Engelsen AST, Buart S, Elsayed WS, Venkatesh GH, Chouaib S. Article CAS PubMed Google Scholar Huang B, Song BL. Article PubMed Google Scholar Chen B, Gao A, Tu B, Wang Y, Yu X, Wang Y, Xiu Y, Wang B, Wan Y, Huang Y.

Article CAS PubMed Google Scholar Karayama M, Masuda J, Mori K, Yasui H, Hozumi H, Suzuki Y, Furuhashi K, Fujisawa T, Enomoto N, Nakamura Y, et al. In other projects. Wikimedia Commons. For other uses, see GLN disambiguation. Not to be confused with Glutamic acid or Glutaric acid.

Skeletal formula of L -glutamine. Ball-and-stick model. Space-filling model. L-Glutamine levo glutamide 2,5-Diaminooxopentanoic acid 2-Aminocarbamoylbutanoic acid Endari [1]. CAS Number. Interactive image Zwitterion : Interactive image. CHEBI Y. ChEMBL Y. DB Y. C Y. PubChem CID. CompTox Dashboard EPA.

Chemical formula. Solubility in water. Chiral rotation [α] D. ATC code. Except where otherwise noted, data are given for materials in their standard state at 25 °C [77 °F], kPa.

Infobox references. Chemical compound. US DailyMed : Glutamine. A16AA03 WHO. IUPAC name. D C GLN PDBe , RCSB PDB. Interactive image. This section is missing information about possible mechanism of action, pharmacokinetics in PMID Please expand the section to include this information.

Further details may exist on the talk page. November Food and Drug Administration FDA Press release. Retrieved 10 July This article incorporates text from this source, which is in the public domain.

CRC Handbook of Chemistry and Physics 62nd ed. Boca Raton, FL: CRC Press. ISBN Retrieved 23 April IUPAC-IUB Joint Commission on Biochemical Nomenclature. Archived from the original on 9 October Retrieved 5 March In Otten JJ, Hellwig JP, Meyers LD eds. Dietary Reference Intakes: The Essential Guide to Nutrient Requirements PDF.

Washington, D. Archived from the original PDF on 9 March Nutrition Reviews. doi : PMID The Journal of Nutrition. Corbet C, Feron O eds. Current Opinion in Clinical Nutrition and Metabolic Care. S2CID Textbook of Medical Physiology 11th ed. Louis, Mo: Elsevier Saunders.

The Journal of Cell Biology. PMC Current Opinion in Biotechnology. April Bibcode : Natur. Canadian Journal of Biochemistry. Proceedings of the National Academy of Sciences of the United States of America.

Bibcode : PNAS.. Cell Metabolism. Scientific Reports. Bibcode : NatSR Ullmann's Encyclopedia of Industrial Chemistry. Weinheim: Wiley-VCH. Animal Nutrition.

Retrieved 24 January Regulatory Toxicology and Pharmacology. A critical appraisal of the human data". The American Journal of Clinical Nutrition. Journal of Parenteral and Enteral Nutrition. BioMed Research International. January Frontiers in Bioscience.

The Cochrane Database of Systematic Reviews. McGuire W ed. ISSN Other alimentary tract and metabolism products A Ademetionine Betaine Carglumic acid Glutamine Levocarnitine Mercaptamine Metreleptin.

Glutamine is the most abundant amino acid in blood andd tissues, and Fat metabolism foods most important nutrient except Glutamine and metabolism glucose Gluttamine cancer cells. Over Fat metabolism foods Anti-aging nutrition years, most Glutaminf have focused on the role of Gln metabolism in jetabolism energy metabolism rather than maintaining oxidative Glutamije. Here, Fat metabolism foods mftabolism will review the recent scientific literature about the link between Gln metabolism and oxidative homeostasis, with an emphasis on the potential role of Gln metabolism in different cancers. Given that oxidative homeostasis is of critical importance in cancer, understanding the impacts of a Gln metabolism on oxidative homeostasis, gaining great insights into underlying molecular mechanisms, and developing effective therapeutic strategies are of great importance. The reactive oxygen species ROSwhich mainly comes from the mitochondrial membrane as a byproduct of OXPHOS and nicotinamide adenine dinucleotide oxidases NOXscannot avoid being produced in cellular metabolism 1 — 4.

Author: Zulujas

0 thoughts on “Glutamine and metabolism

Leave a comment

Yours email will be published. Important fields a marked *

Design by ThemesDNA.com