bims-glucam Biomed News
on Glutamine cancer metabolism
Issue of 2021‒07‒04
eleven papers selected by
Sreeparna Banerjee
Middle East Technical University


  1. Cell Mol Biol (Noisy-le-grand). 2020 Dec 31. 66(8): 33-40
      This experiment was performed to investigate the effect of paeonol on the proliferation, apoptosis, migration, invasion and glutamine of gastric cancer HGC-27 cells and its possible mechanism. For this purpose, the MTT method was used to detect cell viability; Flow cytometry experiment was used to detect cell apoptosis; Transwell chamber experiment was used to detect cell migration and invasion; Western blotting was used to detect the expression levels of MMP2 and MMP9 protein; The decomposition of glutamine was evaluated by detecting the expression levels of glutamine, glutamic acid and α-ketoglutarate (α-KG). This study used RT-PCR to detect the expression of circSFMBT2 and miR-665. The targeting relationship between circSFMBT2 and miR-665 was verified by the dual-luciferase report experiment and RIP experiment. Results showed that different concentrations of Paeonol could significantly inhibit the proliferation, migration, invasion and glutamine decomposition of HGC-27 cells, and induce cell apoptosis in a dose-dependent manner. In gastric cancer tissues and cells, the expression of circSFMBT2 was up-regulated, and the expression of miR-665 was down-regulated. Over-expression of circSFMBT2 could partially restore the effects of paeonol on the proliferation, apoptosis, migration, invasion and glutamine of HGC-27 cells. CircSFMBT2 could target and negatively regulate the expression of miR-665. Overexpression of miR-665 could partially restore the effects of Pae and circSFMBT2 on the proliferation, apoptosis, migration, invasion and glutamine of HGC-27 cells. It was concluded that paeonol can inhibit the proliferation, migration, invasion and glutamine decomposition of gastric cancer HGC-27 cells via circSFMBT2/miR-665 axis, and also induce cell apoptosis.
    Keywords:  Apoptosis; Gastric cancer; Glutamine.; Invasion; Migration; Paeonol; Proliferation
  2. Elife. 2021 Jun 28. pii: e65150. [Epub ahead of print]10
      Chemotherapy resistance is a critical barrier in cancer treatment. Metabolic adaptations have been shown to fuel therapy resistance; however, little is known regarding the generality of these changes and whether specific therapies elicit unique metabolic alterations. Using a combination of metabolomics, transcriptomics, and functional genomics, we show that two anthracyclines, doxorubicin and epirubicin, elicit distinct primary metabolic vulnerabilities in human breast cancer cells. Doxorubicin-resistant cells rely on glutamine to drive oxidative phosphorylation and de novo glutathione synthesis, while epirubicin-resistant cells display markedly increased bioenergetic capacity and mitochondrial ATP production. The dependence on these distinct metabolic adaptations is revealed by the increased sensitivity of doxorubicin-resistant cells and tumor xenografts to buthionine sulfoximine (BSO), a drug that interferes with glutathione synthesis, compared with epirubicin-resistant counterparts that are more sensitive to the biguanide phenformin. Overall, our work reveals that metabolic adaptations can vary with therapeutics and that these metabolic dependencies can be exploited as a targeted approach to treat chemotherapy-resistant breast cancer.
    Keywords:  PGC-1; anthracyclines; bioenergetics; breast cancer; cancer biology; human; metabolomics; mouse; therapeutic resistance
    DOI:  https://doi.org/10.7554/eLife.65150
  3. Cancers (Basel). 2021 Jun 19. pii: 3067. [Epub ahead of print]13(12):
      Altered cell metabolism is a hallmark of cancer cell biology, and the adaptive metabolic strategies of cancer cells have been of recent interest to many groups. Metabolic reprogramming has been identified as a critical step in glial cell transformation, and the use of antimetabolites against glioblastoma has been investigated. One-carbon (1-C) metabolism and its associated biosynthetic pathways, particularly purine nucleotide synthesis, are critical for rapid proliferation and are altered in many cancers. Purine metabolism has also been identified as essential for glioma tumourigenesis. Additionally, alterations of 1-C-mediated purine synthesis have been identified as commonly present in brain tumour initiating cells (BTICs) and could serve as a phenotypic marker of cells responsible for tumour recurrence. Further research is required to elucidate mechanisms through which metabolic vulnerabilities may arise in BTICs and potential ways to therapeutically target these metabolic processes. This review aims to summarize the role of 1-C metabolism-associated vulnerabilities in glioblastoma tumourigenesis and progression and investigate the therapeutic potential of targeting this pathway in conjunction with other treatment strategies.
    Keywords:  de novo purine synthesis; glioblastoma; glioma; metabolic reprogramming; metabolic treatment; one-carbon metabolism
    DOI:  https://doi.org/10.3390/cancers13123067
  4. Cancers (Basel). 2021 Jun 28. pii: 3230. [Epub ahead of print]13(13):
      Metabolic reprogramming is a hallmark of cancer that enables cancer cells to grow, proliferate and survive. This metabolic rewiring is intrinsically regulated by mutations in oncogenes and tumor suppressors, but also extrinsically by tumor microenvironment factors (nutrient and oxygen availability, cell-to-cell interactions, cytokines, hormones, etc.). Intriguingly, only a few cancers are driven by mutations in metabolic genes, which lead metabolites with oncogenic properties (i.e., oncometabolites) to accumulate. In the last decade, there has been rekindled interest in understanding how dysregulated metabolism and its crosstalk with various cell types in the tumor microenvironment not only sustains biosynthesis and energy production for cancer cells, but also contributes to immune escape. An assessment of dysregulated intratumor metabolism has long since been exploited for cancer diagnosis, monitoring and therapy, as exemplified by 18F-2-deoxyglucose positron emission tomography imaging. However, the efficient delivery of precision medicine demands less invasive, cheaper and faster technologies to precisely predict and monitor therapy response. The metabolomic analysis of tumor and/or microenvironment-derived metabolites in readily accessible biological samples is likely to play an important role in this sense. Here, we review altered cancer metabolism and its crosstalk with the tumor microenvironment to focus on energy and biomass sources, oncometabolites and the production of immunosuppressive metabolites. We provide an overview of current pharmacological approaches targeting such dysregulated metabolic landscapes and noninvasive approaches to characterize cancer metabolism for diagnosis, therapy and efficacy assessment.
    Keywords:  LC-MS; MRI; MRS; biomarkers; cancer metabolism; cancer therapy; imaging; metabolic inhibitors; metabolomics; oncometabolites
    DOI:  https://doi.org/10.3390/cancers13133230
  5. Int J Mol Sci. 2021 Jun 10. pii: 6262. [Epub ahead of print]22(12):
      Colorectal carcinoma (CRC) is one of the most frequently diagnosed carcinomas and one of the leading causes of cancer-related death worldwide. Metabolic reprogramming, a hallmark of cancer, is closely related to the initiation and progression of carcinomas, including CRC. Accumulating evidence shows that activation of oncogenic pathways and loss of tumor suppressor genes regulate the metabolic reprogramming that is mainly involved in glycolysis, glutaminolysis, one-carbon metabolism and lipid metabolism. The abnormal metabolic program provides tumor cells with abundant energy, nutrients and redox requirements to support their malignant growth and metastasis, which is accompanied by impaired metabolic flexibility in the tumor microenvironment (TME) and dysbiosis of the gut microbiota. The metabolic crosstalk between the tumor cells, the components of the TME and the intestinal microbiota further facilitates CRC cell proliferation, invasion and metastasis and leads to therapy resistance. Hence, to target the dysregulated tumor metabolism, the TME and the gut microbiota, novel preventive and therapeutic applications are required. In this review, the dysregulation of metabolic programs, molecular pathways, the TME and the intestinal microbiota in CRC is addressed. Possible therapeutic strategies, including metabolic inhibition and immune therapy in CRC, as well as modulation of the aberrant intestinal microbiota, are discussed.
    Keywords:  CRC therapy; colorectal cancer; intestinal microbiota; metabolism; the tumor microenvironment
    DOI:  https://doi.org/10.3390/ijms22126262
  6. Front Cell Dev Biol. 2021 ;9 698264
      The cAMP response element binding protein (CREB) family activating transcription factor 1 (ATF1) and cAMP response element binding protein 1 (CREB1) have been reported in a diverse group of tumors, however, the mechanistic basis for this remains unclear. Here we found that CREB1 and ATF1 unexpectedly regulate glutathione (GSH) biosynthesis by suppressing the expression of glutamate-cysteine ligase modifier subunit (GCLM) and glutathione synthase (GSS), two key enzymes of GSH biosynthesis pathway. Mechanistic studies reveal that GCLM and GSS are direct transcriptional targets of CREB1 and ATF1. Through repressing the expression of these two enzymes, CREB1 and ATF1 reduce the GSH biosynthesis and the capability of cells to detoxicate reactive oxygen species (ROS), thereby increasing cellular susceptibility to oxidative stress. Therefore, our findings link CREB1 family to cellular metabolism, and uncover a potential therapeutic approach by targeting GCLM or oxidative stress for the treatment of tumors with relatively high expression of CREB1 family proteins.
    Keywords:  ATF1; CREB1; GSH; ROS; proliferation; survival
    DOI:  https://doi.org/10.3389/fcell.2021.698264
  7. Biomolecules. 2021 Jun 21. pii: 917. [Epub ahead of print]11(6):
      In order to maintain redox homeostasis, non-small-cell lung cancer (NSCLC) increases the activation of many antioxidant systems, including the heme-oxygenase (HO) system. The overexpression of HO-1 has been often associated with chemoresistance and tumor aggressiveness. Our results clearly showed an overexpression of the HO-1 protein in A549 NSCLC cell lines compared to that in non-cancerous cells. Thus, we hypothesized that "off-label" use of tin mesoporphyrin, a well-known HO activity inhibitor clinically used for neonatal hyperbilirubinemia, has potential use as an anti-cancer agent. The pharmacological inhibition of HO activity caused a reduction in cell proliferation and migration of A549. SnMP treatment caused an increase in oxidative stress, as demonstrated by the upregulation of reactive oxygen species (ROS) and the depletion of glutathione (GSH) content. To support these data, Western blot analysis was performed to analyze glucose-6-phosphate dehydrogenase (G6PD), TP53-induced glycolysis and the apoptosis regulator (TIGAR), and the glutamate cysteine ligase catalytic (GCLC) subunit, as they represent the main regulators of the pentose phosphate pathway (PPP) and glutathione synthesis, respectively. NCI-H292, a subtype of the NSCLC cell line, did not respond to SnMP treatment, possibly due to low basal levels of HO-1, suggesting a cellular-dependent antitumorigenic effect. Altogether, our results suggest HO activity inhibition may represent a potential target for selective chemotherapy in lung cancer subtypes.
    Keywords:  cancer; heme-oxygenase; oxidative stress; porphyrins
    DOI:  https://doi.org/10.3390/biom11060917
  8. Int J Mol Sci. 2021 Jun 19. pii: 6587. [Epub ahead of print]22(12):
      Citrate plays a central role in cancer cells' metabolism and regulation. Derived from mitochondrial synthesis and/or carboxylation of α-ketoglutarate, it is cleaved by ATP-citrate lyase into acetyl-CoA and oxaloacetate. The rapid turnover of these molecules in proliferative cancer cells maintains a low-level of citrate, precluding its retro-inhibition on glycolytic enzymes. In cancer cells relying on glycolysis, this regulation helps sustain the Warburg effect. In those relying on an oxidative metabolism, fatty acid β-oxidation sustains a high production of citrate, which is still rapidly converted into acetyl-CoA and oxaloacetate, this latter molecule sustaining nucleotide synthesis and gluconeogenesis. Therefore, citrate levels are rarely high in cancer cells. Resistance of cancer cells to targeted therapies, such as tyrosine kinase inhibitors (TKIs), is frequently sustained by aerobic glycolysis and its key oncogenic drivers, such as Ras and its downstream effectors MAPK/ERK and PI3K/Akt. Remarkably, in preclinical cancer models, the administration of high doses of citrate showed various anti-cancer effects, such as the inhibition of glycolysis, the promotion of cytotoxic drugs sensibility and apoptosis, the neutralization of extracellular acidity, and the inhibition of tumors growth and of key signalling pathways (in particular, the IGF-1R/AKT pathway). Therefore, these preclinical results support the testing of the citrate strategy in clinical trials to counteract key oncogenic drivers sustaining cancer development and resistance to anti-cancer therapies.
    Keywords:  ACLY; Warburg effect; cancer cells; citrate; resistance to therapies
    DOI:  https://doi.org/10.3390/ijms22126587
  9. Cancer Res. 2021 Jun 28. pii: canres.3956.2020. [Epub ahead of print]
      One carbon (1C) metabolism has a key role in metabolic programming with both mitochondrial (m1C) and cytoplasmic (c1C) components. Here we show that Activating Transcription Factor 4 (ATF4) exclusively activates gene expression involved in m1C, but not c1C cycle in prostate cancer (PCa) cells. This includes activation of Methylenetetrahydrofolate dehydrogenase 2(MTHFD2) expression, the central player in the m1C cycle. Consistent with the key role of m1C cycle in PCa, MTHFD2 knockdown inhibited PCa cell growth, prostatosphere formation and growth of patient-derived xenograft (PDX) organoids. In addition, therapeutic silencing of MTHFD2 by systemically administered nanoliposomal siRNA profoundly inhibited tumor growth in a preclinical PCa mouse model. Consistently, MTHFD2 expression is significantly increased in human PCa and a gene expression signature based on the m1C cycle has significant prognostic value. Furthermore, MTHFD2 expression is coordinately regulated by ATF4 and the oncoprotein c-MYC, which has been implicated in PCa. These data suggest that the m1C cycle is essential for PCa progression and may serve as a novel biomarker and therapeutic target.
    DOI:  https://doi.org/10.1158/0008-5472.CAN-20-3956
  10. Sci Rep. 2021 Jul 01. 11(1): 13637
      Recently, there has been a resurgence of interest in metabolic rewiring of tumors to identify clinically relevant genes. However, most of these studies have had either focused on individual tumors, or are too general, providing a broad outlook on overall changes. In this study, we have first curated an extensive list of genes encoding metabolic enzymes and metabolite transporters relevant to carbohydrate, fatty acid and amino acid oxidation and biosynthesis. Next, we have used publicly available transcriptomic data for 20 different tumor types from The Cancer Genome Atlas Network (TCGA) and focused on differential expression of these genes between tumor and adjacent normal tissue. Our study revealed major transcriptional alterations in genes that are involved in central metabolism. Most tumors exhibit upregulation in carbohydrate and amino acid transporters, increased glycolysis and pentose phosphate pathway, and decreased fatty acid and amino acid oxidation. On the other hand, the expression of genes of the tricarboxylic acid cycle, anaplerotic reactions and electron transport chain differed between tumors. Although most transcriptomic alterations were conserved across many tumor types suggesting the initiation of common regulatory programs, expression changes unique to specific tumors were also identified, which can provide gene expression fingerprints as potential biomarkers or drug targets. Our study also emphasizes the value of transcriptomic data in the deeper understanding of metabolic changes in diseases.
    DOI:  https://doi.org/10.1038/s41598-021-93003-3
  11. Front Oncol. 2021 ;11 644197
      Reactive oxygen species (ROS) play an important role in cellular metabolism. Many chemotherapeutic drugs are known to promote apoptosis through the production of ROS. In the present study, the novel curcumin derivative, 1g, was found to inhibit tumor growth in colon cancer cells both in vitro and in vivo. Bioinformatics was used to analyze the differentially expressed mRNAs. The mechanism of this effect was a change in mitochondrial membrane potential caused by 1g that increased its pro-apoptotic activity. In addition, 1g produced ROS, induced G1 checkpoint blockade, and enhanced endoplasmic reticulum (ER)-stress in colon cancer cells. Conversely, pretreatment with the ROS scavenging agent N-acetyl-l-cysteine (NAC) inhibited the mitochondrial dysfunction caused by 1g and reversed ER-stress, cell cycle stagnation, and apoptosis. Additionally, pretreatment with the p-PERK inhibitor GSK2606414 significantly reduced ER-stress and reversed the apoptosis induced by colon cancer cells. In summary, the production of ROS plays an important role in the destruction of colon cancer cells by 1g and demonstrates that targeted strategies based on ROS represent a promising approach to inhibit colon cancer proliferation. These findings reveal that the novel curcumin derivative 1g represents a potential candidate therapeutics for the treatment of colon cancer cells, via apoptosis caused by mitochondrial dysfunction and endoplasmic reticulum stress.
    Keywords:  1g; ER-stress; ROS—reactive oxygen species; apoptosis; colon cancer
    DOI:  https://doi.org/10.3389/fonc.2021.644197