bims-meluca Biomed News
on Metabolism of non-small cell lung carcinoma
Issue of 2019‒07‒07
eleven papers selected by
Cristina Muñoz Pinedo
L’Institut d’Investigació Biomèdica de Bellvitge


  1. Cell. 2019 Jun 26. pii: S0092-8674(19)30633-6. [Epub ahead of print]
      For tumors to progress efficiently, cancer cells must overcome barriers of oxidative stress. Although dietary antioxidant supplementation or activation of endogenous antioxidants by NRF2 reduces oxidative stress and promotes early lung tumor progression, little is known about its effect on lung cancer metastasis. Here, we show that long-term supplementation with the antioxidants N-acetylcysteine and vitamin E promotes KRAS-driven lung cancer metastasis. The antioxidants stimulate metastasis by reducing levels of free heme and stabilizing the transcription factor BACH1. BACH1 activates transcription of Hexokinase 2 and Gapdh and increases glucose uptake, glycolysis rates, and lactate secretion, thereby stimulating glycolysis-dependent metastasis of mouse and human lung cancer cells. Targeting BACH1 normalized glycolysis and prevented antioxidant-induced metastasis, while increasing endogenous BACH1 expression stimulated glycolysis and promoted metastasis, also in the absence of antioxidants. We conclude that BACH1 stimulates glycolysis-dependent lung cancer metastasis and that BACH1 is activated under conditions of reduced oxidative stress.
    Keywords:  BACH1; KRAS; antioxidants; heme; lung cancer metastasis; mouse models; oxidative stress
    DOI:  https://doi.org/10.1016/j.cell.2019.06.005
  2. Cell. 2019 Jun 19. pii: S0092-8674(19)30631-2. [Epub ahead of print]
      Approximately 30% of human lung cancers acquire mutations in either Keap1 or Nfe2l2, resulting in the stabilization of Nrf2, the Nfe2l2 gene product, which controls oxidative homeostasis. Here, we show that heme triggers the degradation of Bach1, a pro-metastatic transcription factor, by promoting its interaction with the ubiquitin ligase Fbxo22. Nrf2 accumulation in lung cancers causes the stabilization of Bach1 by inducing Ho1, the enzyme catabolizing heme. In mouse models of lung cancers, loss of Keap1 or Fbxo22 induces metastasis in a Bach1-dependent manner. Pharmacological inhibition of Ho1 suppresses metastasis in a Fbxo22-dependent manner. Human metastatic lung cancer display high levels of Ho1 and Bach1. Bach1 transcriptional signature is associated with poor survival and metastasis in lung cancer patients. We propose that Nrf2 activates a metastatic program by inhibiting the heme- and Fbxo22-mediated degradation of Bach1, and that Ho1 inhibitors represent an effective therapeutic strategy to prevent lung cancer metastasis.
    Keywords:  Bach1; CRL complexes; F-box proteins; Fbxo22; Heme; Ho1 inhibitor; Keap1; Nrf2; cullin-RING ubiquitin ligase; lung cancer; metastasis; ubiquitin
    DOI:  https://doi.org/10.1016/j.cell.2019.06.003
  3. Exp Ther Med. 2019 Jul;18(1): 188-198
      Lung cancer is one of the most prevalent types of cancer, but accurate diagnosis remains a challenge. The aim of the present study was to create a model using amino acids and acylcarnitines for lung cancer screening. Serum samples were obtained from two groups of patients with lung cancer recruited in 2015 (including 40 patients and 100 matched controls) and 2017 (including 17 patients and 30 matched controls). Using a metabolomics method, 21 metabolites (13 types of amino acids and 8 types of acylcarnitines) were measured using liquid chromatography-tandem mass spectrometry. Data (from the 2015 and 2017 data sets) were analysed using a Mann-Whitney U test, Student's t-test, Welch's F test, receiver-operator characteristic curve or logistic regression in order to investigate the potential biomarkers. Six metabolites (glycine, valine, methionine, citrulline, arginine and C16-carnitine) were indicated to be involved in distinguishing patients with lung cancer from healthy controls. The six discriminating metabolites from the 2017 data set were further analysed using Partial least squares-discriminant analysis (PLS-DA). The PLS-DA model was verified using Spearman's correlation analysis and receiver operating characteristic curve analysis. These results demonstrated that the PLS-DA model using the six metabolites (glycine, valine, methionine, citrulline, arginine and C16-carnitine) had a strong ability to identify lung cancer. Therefore, the PLS-DA model using glycine, valine, methionine, citrulline, arginine and C16-carnitine may become a novel screening tool in patients with lung cancer.
    Keywords:  acylcarnitines; amino acids; liquid chromatography-tandem mass spectrometry; lung cancer; metabolomics
    DOI:  https://doi.org/10.3892/etm.2019.7533
  4. Int J Cancer. 2019 Jun 28.
      The role of Fyn-related kinase (FRK) in malignant tumors remains controversial. This study investigated the function of FRK in lung cancer. Immunohistochemistry staining and generating a knockout of FRK by CRISPR/Cas9 in H1299 (FRK-KO-H1299) cells were strategies used to explore the role of FRK. Immunohistochemistry staining indicated that FRK expression was elevated in 223 lung cancer tissues compared with 26 distant normal lung tissues. FRK contributed to poor survival status in lung cancer patients and acted as a predictor for poor prognosis of lung cancer. Knockout of FRK by CRISPR/Cas9 dramatically inhibited proliferation, invasion, colony formation and epithelial mesenchymal transition process in the lung cancer cell line H1299. Further exploration indicated that FRK-KO damaged the stemness phenotype of H1299 by inhibiting CD44 and CD133 expression. Seahorse detection and a U-13 C flux assay revealed that FRK-KO induced metabolism reprogramming by inhibiting the Warburg effect and changing the energy type in H1299 cells. Epidermal growth factor stimulation recovered the expression of FRK and biological functions, metabolic reprogramming, and stemness phenotype of H1299 cells. FRK plays an oncogenic role in lung cancer cells via a novel regulation mechanism of enhancing the stemness of H1299 cells by inducing metabolism reprogramming, which finally promotes EMT (Epithelial-mesenchymal transition) and metastasis. Our study also indicates that FRK could be used as a potential therapeutic target for drug development. This article is protected by copyright. All rights reserved.
    Keywords:  FRK; lung cancer; metabolic reprogramming; poor prognosis; stemness
    DOI:  https://doi.org/10.1002/ijc.32530
  5. Front Genet. 2019 ;10 554
      The nuclear receptors known as peroxisome proliferator activated receptor gamma (PPARG) are lipid-activated transcription factors that have emerged as key regulators of inflammation. PPARG ligands have been shown to have an anti-proliferative effect on a variety of cancers. These ligands can induce apoptosis via TP53 (Tumor protein p53) or ERK1/2 (Extracellular signal-regulated kinases 1/2) (EPHB2) pathways. However, the exact mechanism is not known. PPAR, a type II nuclear hormone receptor deserves attention as a selective target for radiotherapy. Our study examines the potential of selective agonism of PPARG for radiation therapy in non-small cell lung carcinoma (NSCLC). We found that the overexpression of PPARG protein as well as its induction using the agonist, rosiglitazone was able to stimulate radiation-induced cell death in otherwise radio resistant NSCLC A549 cell line. This cell death was apoptotic and was found to be BAX (BCL2 associated X) mediated. The treatment also inhibited radiation-induced AKT (Protein Kinase B) phosphorylation. Interestingly, the ionising radiation (IR) induced apoptosis was found to be inversely related to TP53 levels. A relatively significant increase in the levels of radiation induced apoptosis was observed in H1299 cells (TP53 null) under PPARG overexpression condition further supporting the inverse relationship between apoptosis and TP53 levels. The combination of PPARG agonist and radiation was able to induce apoptosis at a radiation dose at which A549 and H1299 are radioresistant, thus confirming the potential of the combinatorial strategy. Taken together, PPARG agonism was found to invigorate the radiosensitising effect and hence its use in combination with radiotherapy is expected to enhance sensitivity in otherwise resistant cancer types.
    Keywords:  BAX; Hedgehog signaling; NSCLC; PPARG; TP53; radiosensitization
    DOI:  https://doi.org/10.3389/fgene.2019.00554
  6. J Cell Physiol. 2019 Jul 04.
      Chemotherapy is the first-line treatment option for patients with lung cancer. However, therapeutic resistance occurs through an incompletely understood mechanism. Our research wants to investigate the influence of Caveolin-1 (Cav-1) on the therapeutic sensitivity of lung cancer in vitro. Results in this study demonstrated that Cav-1 levels were markedly inhibited in A549 lung cancer cells after exposure to cisplatin. Knockdown of caveolin further enhanced cisplatin-triggered cancer death in A549 cells. The functional investigation demonstrated that Cav-1 inhibition amplified the mitochondrial stress signaling induced by cisplatin, as evidenced by the mitochondrial reactive oxygen species burst, cellular metabolic disruption, mitochondrial membrane potential reduction, and mitochondrial caspase-9-related apoptosis activation. At the molecular level, cav-1 augmented cisplatin-mediated mitochondrial damage by inhibiting Parkin-related mitochondrial autophagy. Mitophagy activation effectively attenuated the promotive impact of Cav-1 knockdown on mitochondrial damage and cell death. Furthermore, our data indicated that Cav-1 affected Parkin-related mitophagy by activating the Rho-associated coiled-coil kinase 1 (ROCK1) pathway; inhibition of the ROCK1 axis prevented cav-1 knockdown-mediated cell death and mitochondrial damage. Taken together, our results provide ample data illuminate the necessary action exerted by Cav-1 on affecting cisplatin-related therapeutic resistance. Silencing of Cav-1 inhibited Parkin-related mitophagy, thus amplifying cisplatin-mediated mitochondrial apoptotic signaling. This finding identifies the Cav-1/ROCK1/Parkin/mitophagy axis as a potential target to overcome cisplatin-related resistance in lung cancer cells.
    Keywords:  Cav-1; ROCK1; apoptosis; lung cancer; mitophagy
    DOI:  https://doi.org/10.1002/jcp.29033
  7. Eur J Nucl Med Mol Imaging. 2019 Jul 01.
      PURPOSE: 18F-fluoroaminosuberic acid (18F-FASu) is a recently developed amino acid tracer for positron emission tomography (PET) of oxidative stress that may offer improved tumour assessment over the conventional tracer 18F-fluorodeoxyglucose (18F-FDG). Our aim was to evaluate and relate dynamic 18F-FASu and 18F-FDG uptake with pharmacokinetic modelling to transporter protein expression levels in a panel of diverse tumour xenograft lines.METHODS: Four different tumour xenograft lines were implanted in female athymic nude mice: MAS98.12 and HBCx3 (breast), TPMX (osteosarcoma) and A549 (lung). Dynamic PET over 60 min was performed on a small animal unit. The time-activity curves (TACs) for 18F-FASu and 18F-FDG in individual tumours were used to extract early (SUVE; 2 min p.i.) and late (SUVL; 55 min p.i.) standardised uptake values. Pharmacokinetic two-tissue compartment models were applied to the TACs to estimate rate constants K1-k4 and blood volume fraction vB. Relative levels of cystine/glutamate antiporter subunit xCT were assessed by western blotting, and expression of GLUT1 and CD31 by immunohistochemistry.
    RESULTS: 18F-FASu showed higher SUVE, whilst 18F-FDG exhibited higher SUVL. Influx rate K1 for 18F-FASu was significantly correlated with xCT levels (p = 0.001) and was significantly higher than K1 for 18F-FDG (p < 0.001). K1 for 18F-FDG was significantly correlated with GLUT1 levels (p = 0.002). vB estimated from 18F-FASu and 18F-FDG TACs was highly consistent and significantly correlated (r = 0.85, p < 0.001). Two qualitatively different 18F-FASu uptake profiles were identified: type α with low xCT expression and low K1 (A549 and HBCx3), and type β with high xCT expression and high K1 (MAS98.12 and TPMX).
    CONCLUSION: The influx rate of 18F-FASu reflects xCT activity in tumour xenografts. Dynamic PET with pharmacokinetic modelling is needed to fully appraise 18F-FASu distribution routes.
    Keywords:  18F-FDG; 18F-fluoroaminosuberic acid; Cancer; Dynamic PET; Mouse model; Oxidative stress; Pharmacokinetic modelling; System XC −; Xenograft; xCT
    DOI:  https://doi.org/10.1007/s00259-019-04375-8
  8. Scand J Immunol. 2019 Jul 03. e12802
      Glucose and nutrient uptake is essential in supporting T cell activation and is increased upon CD3/CD28 stimulation. As T cells from pleural effusions secondary to lung cancer show impaired function, we hypothesized that these cells might have altered expression of nutrient transporters. Here, we analyzed by flow cytometry the expression of the transferrin receptor CD71, amino acid transporter CD98, and glucose transporter Glut1 and glucose uptake in pleural effusion-derived T cells from lung cancer patients, after stimulation via CD3/CD28 under normoxia or hypoxia (2% O2). We compared the response of T cells from pleural effusions secondary to lung cancer with that of T cells from nonmalignant effusions. In memory T cells from both groups, anti-CD3/CD28-stimulation under normoxia upregulated CD98 and CD71 expression (measured as median fluorescence intensity, MFI) in comparison with anti-CD3-stimulation. Costimulation under hypoxia tended to increase CD98 expression compared to CD3-stimulation in memory T cells from both groups. Remarkably, in the cancer group, memory T cells stimulated via CD3/CD28 under hypoxia failed to increase CD71 and Glut1 expression levels compared to the cells receiving anti-CD3 stimulation, a phenomenon that contrasted with the behavior of memory T cells from nonmalignant effusions. Consequently, glucose uptake by memory T cells from the cancer group was not increased after CD3/CD28 stimulation under hypoxia, implying that their glycolytic metabolism is defective. As this process is required for inducing an antitumoral response, our study suggests that memory T cells are rendered dysfunctional under hypoxia and are unable to eliminate lung tumor cells. This article is protected by copyright. All rights reserved.
    Keywords:  Immunometabolism; T cell; amino acid heterodimeric transporter CD98; flow cytometry; glucose transporter-1 Glut1; lung cancer; transferrin receptor CD71
    DOI:  https://doi.org/10.1111/sji.12802
  9. Mol Med Rep. 2019 Aug;20(2): 1925-1932
      The secreted frizzled‑related protein 2 (SFRP2) has been reported to inhibit non‑small cell lung cancer (NSCLC) cell survival and metastasis; however, the underlying mechanisms are yet to be fully determined. The present study focused on mitochondrial fission and the Wnt signaling pathway. The results demonstrated that SFRP2 was downregulated in the NSCLC cell line A549 compared with in a normal pulmonary epithelial cell line using western blotting, reverse transcription‑quantitative PCR and immunofluorescence. Subsequently, it was demonstrated that SFRP2 overexpression promoted the apoptosis, and inhibited the proliferation and metastasis of A549 cells using MTT assays, TUNEL staining and 5‑ethynyl‑2'‑deoxyuridine labeling. At the molecular level, the overexpression of SFRP2 in A549 cells led to the activation of mitochondrial fission by inhibiting the Wnt signal pathway. Excessive mitochondrial fission induced low ATP generation, impaired mitochondrial respiratory function, induced mitochondrial potential depolarization, and increased mitochondrial permeability transition pore opening, and imbalances in pro‑ and antiapoptotic protein expression. Furthermore, mitochondrial fission was involved in the inhibition of A549 cell proliferation and metastasis. Thus, SFRP2 may inhibit the survival and metastasis of NSCLC cells via the Wnt/mitochondrial fission pathway.
    DOI:  https://doi.org/10.3892/mmr.2019.10393
  10. Anticancer Agents Med Chem. 2019 Jul 04.
      BACKGROUND: Ginsenoside Rh2 (Rh2), which is extracted form ginseng, exerts antitumor activity. Here we would like to study the role of Rh2 on hypoxia-induced migration in lung adenocarcinoma.METHODS: Lung adenocarcinoma A549 and H1299 cells were cultured in 1% O2 condition to mimic the hypoxic tumor microenvironment. The migrations of cancer cells were measured by transwell assay and scratch assay.
    RESULTS: Rh2 could inhibit hypoxia -induced A549 and H1299 cell migration via increase of mir-491expression. Further, mir-491 antisense oligonucleotide could repress hypoxia-induced migration and the expression of matrix metalloproteinase (MMP)-9 expression in Rh2-treated A549 cells.
    CONCLUSION: These findings suggest that Rh2 exerts anti-metastasis activity in hypoxic tumor microenvironment in lung adenocarcinoma cells via mir-491.
    Keywords:  Ginsenoside Rh2; antitumor activity; hypoxia; lung adenocarcinoma; migration; mir-491
    DOI:  https://doi.org/10.2174/1871520619666190704165205
  11. Dev Cell. 2019 Jun 26. pii: S1534-5807(19)30485-X. [Epub ahead of print]
      Macropinocytosis has emerged as an important nutrient-scavenging pathway that supports tumor cell fitness. By internalizing extracellular protein and targeting it for lysosomal degradation, this endocytic pathway functions as an amino acid supply route, permitting tumor cell growth and survival despite the nutrient-poor conditions of the tumor microenvironment. Here, we provide evidence that a subset of pancreatic ductal adenocarcinoma (PDAC) tumors are wired to integrate contextual metabolic inputs to regulate macropinocytosis, dialing up or down this uptake pathway depending on nutrient availability. We find that regional depletion of amino acids coincides with increased levels of macropinocytosis and that the scarcity of glutamine uniquely drives this process. Mechanistically, this stimulation of macropinocytosis depends on the nutrient stress-induced potentiation of epidermal growth factor receptor signaling that, through the activation of Pak, controls the extent of macropinocytosis in these cells. These results provide a mechanistic understanding of how nutritional cues can control protein scavenging in PDAC tumors.
    Keywords:  EGFR; Pak; Ras; cancer metabolism; macropinocytosis; pancreatic cancer; protein scavenging
    DOI:  https://doi.org/10.1016/j.devcel.2019.05.043