Copper depletion modulates mitochondrial oxidative phosphorylation to impair triple negative breast cancer metastasis

Divya Ramchandani, Mirela Berisa, Diamile A Tavarez, Zhuoning Li, Matthew Miele, Yang Bai, Sharrell B Lee, Yi Ban, Noah Dephoure, Ronald C Hendrickson, Suzanne M Cloonan, Dingcheng Gao, Justin R Cross, Linda T Vahdat, Vivek Mittal, Divya Ramchandani, Mirela Berisa, Diamile A Tavarez, Zhuoning Li, Matthew Miele, Yang Bai, Sharrell B Lee, Yi Ban, Noah Dephoure, Ronald C Hendrickson, Suzanne M Cloonan, Dingcheng Gao, Justin R Cross, Linda T Vahdat, Vivek Mittal

Abstract

Copper serves as a co-factor for a host of metalloenzymes that contribute to malignant progression. The orally bioavailable copper chelating agent tetrathiomolybdate (TM) has been associated with a significant survival benefit in high-risk triple negative breast cancer (TNBC) patients. Despite these promising data, the mechanisms by which copper depletion impacts metastasis are poorly understood and this remains a major barrier to advancing TM to a randomized phase II trial. Here, using two independent TNBC models, we report a discrete subpopulation of highly metastatic SOX2/OCT4+ cells within primary tumors that exhibit elevated intracellular copper levels and a marked sensitivity to TM. Global proteomic and metabolomic profiling identifies TM-mediated inactivation of Complex IV as the primary metabolic defect in the SOX2/OCT4+ cell population. We also identify AMPK/mTORC1 energy sensor as an important downstream pathway and show that AMPK inhibition rescues TM-mediated loss of invasion. Furthermore, loss of the mitochondria-specific copper chaperone, COX17, restricts copper deficiency to mitochondria and phenocopies TM-mediated alterations. These findings identify a copper-metabolism-metastasis axis with potential to enrich patient populations in next-generation therapeutic trials.

Trial registration: ClinicalTrials.gov NCT00195091.

Conflict of interest statement

The authors declare no competing interests.

© 2021. The Author(s).

Figures

Fig. 1. Copper depletion impacts invasion and…
Fig. 1. Copper depletion impacts invasion and metastasis.
a Patient survival with CTR1 or SLC31A1 expression using METABRIC dataset. The graph depicts survival for patients with low expression (first quartile) and high expression (fourth quartile) of the SLC31A1 gene (n = 425 patients/group). Significance was calculated using Log-rank (Mantel–Cox) test. b Distribution of SLC31A1 expression with tumor stage. Significance was calculated using one-way ANOVA with Tukey post-test for comparing multiple groups. N = 433 for Stage 0-I patients, n = 702 for Stage II patients, n = 111 for Stage III–IV patients. p-value (0-I vs. II) < 0.0001, p-value (0-I vs. III–IV) = 0.0011. c Distribution of SLC31A1 mRNA expression in breast cancer subtypes. Significance was calculated using one-way ANOVA with Tukey post-test for comparing multiple groups. n = 290 for ER−/PR− patients, n = 603 for ER+/HER2− High proliferative patients, n = 619 for ER+/HER2− Low proliferative patients, n = 188 for HER2+ patients. p-value < 0.0001. d Event-free survival (EFS) for Stage 4 NED breast cancer patients from TM clinical (Chan et al. 2017). 1 cycle = 4 weeks. e (Top panel) Development of EO771.ML1 model. (Lower panel) Timeline for metastasis assay with or without TM using EO771.ML1 model. f Ceruloplasmin levels in control vs. TM-treated mice in ML1 model, 2 weeks after TM administration (n = 6/group). Analysis was performed by unpaired two-sided t-test. Center lines of box plots denote median values, top whiskers denote maxima and bottom whiskers minima. p-value = 0.0224. g Primary tumor growth in control and TM cohorts in ML1 model, measured with calipers (n = 10 in the control group, n = 9 in TM group). Analysis was performed by unpaired two-sided t-test. p-value = 0.8365. h BLI measurements of lungs in ML1 model showing metastatic burden (n = 4/group). Analysis was performed by unpaired two-sided t-test. p-value = 0.0015. i Measurement of intracellular copper (Cu) in LM2 cells using graphite furnace atomic absorption spectrometry (y-axis refers to Cu content measured at 324.75 nm wavelength). Significance was calculated using one-way ANOVA with Tukey post-test for comparing multiple groups. p-value (0 vs. 0.1 nM) = 0.0003, p-value (rest) < 0.0001. j MTT assay for viability with increasing TM concentrations, 72 h after treatment of LM2 cells (n = 4/group). Significance was calculated using One-way ANOVA with Tukey post-test for comparing multiple groups. p-value (0 vs. 1 nM) = 0.0162, p-value (0.2 vs. 1 nM) = 0.0041. k Matrigel coated transwell assay showing invasion of LM2 cells treated with TM (0.5 µM) for a total of 72 h (48-h pretreatment, followed by 24 h treatment on Matrigel-coated transwells) and rescue with addition of copper (0.5 µM CuCl2) for the last 24 h in the transwell (along with TM for that cohort). n = 3 replicates/group (7 fields of view/sample). Significance was calculated using one-way ANOVA with Tukey post-test for comparing multiple groups. p-value (Control vs. TM + Cu) = 0.0115. Representative data of at least two independent experiments are depicted. Results are expressed as mean ± SD. *p < 0.05, **p < 0.01, ****p < 0.0001.
Fig. 2. Copper depletion targets SOX2/OCT4+ metastatic…
Fig. 2. Copper depletion targets SOX2/OCT4+ metastatic cells.
a (Top panel) schematic of lentivirus-based SOX2/OCT4-GFP promoter-reporter system. (Bottom panel) Flow-sorted GFP+ and GFP− cells from bulk ML1 cells with CMV control or SOX2/OCT4 reporter system. b Impact of TM (0.5 µM) on the invasion of SOX2/OCT4+ GFP+ and SOX2/OCT4− GFP− cells across a matrix coated transwell in LM2 cells. Significance was calculated using Two-way ANOVA (n = 3 replicates/group, 5 fields of view/sample). c Intracellular copper (Cu) levels in SOX2/OCT4+ GFP+ vs. SOX2/OCT4− GFP− cells measured by atomic absorption spectrometry (y-axis refers to Cu content measured at 324.75 nm wavelength) in ML1 cells. Analysis was performed by unpaired two-sided t-test. p-value = 0.0040. d Tandem mass tags (TMT) isobaric label multiplexed quantitative proteomic analysis of vehicle and TM (0.5 µM) treated ML1 SOX2/OCT4+ GFP+ reporter cells. Proteins with absolute log2 Fold Change (FC) > 1, p < 0.05 and FDR < 0.05 are shown in green and labeled, n = 5/group. Significance was calculated using multiple t-tests, p values were adjusted for multiple comparisons. e Tandem mass tags (TMT) isobaric label multiplexed quantitative proteomic analysis of vehicle and TM (0.5 µM) treated ML1 SOX2/OCT4− GFP− cells. Proteins with log2 > 1 or < −1 and p < 0.05 are shown, n = 5/group. Significance was calculated using multiple t-tests, p values were adjusted for multiple comparisons. f and g Gene ontology (GO) analysis of downregulated proteins (p < 0.05, average log2 < −0.6) after 48 h TM treatment in ML1 SOX2/OCT4+ GFP+ (f) and ML1 SOX2/OCT4− GFP− g cells (log2 fold change from −2.6 to −0.7). log2 fold change is log2 FC of TM 48 h/Control. Count refers to how many genes/proteins showed up in that GO term. GO analysis was performed using the clusterProfiler R package, p values were adjusted using Benjamin–Hochberg method. dsCop destabilized copepod. Representative data of two independent experiments are depicted. Results are expressed as mean ± SD. **p < 0.01 ****p < 0.0001.
Fig. 3. TM treatment impacts mitochondrial Complex…
Fig. 3. TM treatment impacts mitochondrial Complex IV.
a Western blot showing respiratory complexes in LM2 cells following 72-h TM treatment (0, 0.1, 0.2, 0.5 µM). Complexes I–V are referred to as CI (mol. wt. ~19 kDa), CII (mol. wt. ~30 kDa), CIII (mol. wt. ~48 kDa), CIV (mol. wt. ~40 kDa), and CV (mol.wt. ~55 kDa), respectively. NDUFA4 is a subunit of Complex IV. b qPCR analysis showing transcript levels of human Complex IV subunit 4I1 (COX4I1) in control and TM (0.5 µM) treated LM2 cells, treated with TM for 72 h. Analysis was performed by unpaired two-sided t-test. p-value = 0.079. c Protein expression of cytochrome c with and without TM treatment (0.5 µM) (n = 4/group). d Human cytochrome c oxidase activity (Abcam) kinetically measured over 2 h after 72 h of treatment with TM (0.5 µM) in LM2 cells. The graph represents the difference in Complex IV activity at 110th minute during measurement. Analysis was performed by unpaired two-sided t-test (n = 4/group). p-value = 0.0063. Center lines of box plots denote median values, top whiskers denote maxima and bottom whiskers minima. e Mitochondrial DNA content in LM2 cells as determined by qPCR (p = 0.060). Analysis was performed by unpaired two-sided t-test (n = 4/group). f–h MitoTracker deep red analysis in control and TM (0.5 µM) treated LM2 (f), ML1 (g) and MDA-MB-468 (h) cells, treated for 72 h (n = 3/group). i Structure of cristae in the inner mitochondrial membrane as determined by transmission electron microscopy (×50,000), red arrows indicate normal/folded and perturbed cristae morphology in LM2 cells. Scale bar, 500 nm. j Quantification of perturbed cristae per 100 units of mitochondrial area in pixels (n = 30 mitochondria in controls, n = 36 mitochondria for TM group). Analysis was performed using Mann–Whitney two-sided test. Representative data of two independent experiments are depicted. Results are expressed as mean ± SD. **p < 0.01 ****p < 0.0001.
Fig. 4. Copper depletion disrupts mitochondrial respiration…
Fig. 4. Copper depletion disrupts mitochondrial respiration in SOX2/OCT4+ metastatic cells.
a Impact of TM (0.5 µM) on oxygen consumption rate of sorted GFP+ and GFP− ML1 cells as measured by Seahorse assay (n = 8/group). Quantification of basal respiration (b) and ATP-linked respiration (c) in GFP+ vs. GFP- ML1 cells with and without TM (0.5 µM). Significance was calculated using one-way ANOVA with Tukey post-test for comparing multiple groups. Center lines of box plots denote median values, top whiskers denote maxima and bottom whiskers minima. d Ratio of mitochondrial to glycolytic ATP production rate in GFP+ vs. GFP− ML1 cells, measured by Seahorse ATP production rate assay (n = 8/group). Significance was calculated using one-way ANOVA with Tukey post-test for comparing multiple groups. Representative data of two independent experiments are depicted. e Representative IF microscopy images from control and TM-treated (0.7 mg/day) primary LM2 tumors in the mammary gland (n = 5/group). Scale bar, 20 µM. f Flow cytometry analysis showing the percentage of GFP+ cells in primary tumors in the LM2 model (n = 12/group) of total mCherry+ tumor cells. Analysis was performed by unpaired two-sided t-test. p-value = 0.0111. g Disseminated tumor cells in early lungs of control and TM (0.7 mg/day) treated LM2 cohorts. Analysis was performed by unpaired two-sided t-test. p-value = 0.0150. Representative data of two independent experiments are depicted. Results are expressed as mean ± SD. Results are expressed as mean ± SD. **p < 0.01, ****p < 0.0001.
Fig. 5. COX17 knockdown yields TM phenotypes.
Fig. 5. COX17 knockdown yields TM phenotypes.
a qPCR showing shRNA-mediated COX17 knockdown in LM2 cells. Analysis was performed by unpaired t-test (n = 5/group). b Oxygen consumption in shCOX17#1 vs. SCR-sh (n = 8 for SCR-sh, n = 6 for shCOX17#1). c, d Basal and ATP-linked respiration in shCOX17#1 vs. SCR-sh cells using mitostress assay. Analysis was performed by unpaired two-sided t-test. Center lines of box plots denote median values, top whiskers denote maxima and bottom whiskers minima. e Invasion assay through matrix-coated transwell. Analysis was performed by unpaired two-sided t-test (n = 3/group, 5 fields of view/sample). f Representative images of metastatic nodules in lungs using shCOX17#1 and SCR-sh LM2 in vivo model (n = 7/group). g Quantification of lung metastases from panel f. Analysis was performed by unpaired two-sided t-test. p-value = 0.0236. h Oxygen consumption rate in ML1 SCR-sh vs. shCOX20 cells (n = 8/group). Representative data of two independent experiments are depicted. Results are expressed as mean ± SD. *p < 0.05, ****p < 0.0001.
Fig. 6. Copper depletion activates AMPK.
Fig. 6. Copper depletion activates AMPK.
a Glucose consumption and lactate production measured by nutrient analysis after 72 h of TM (0.5 µM) treatment in LM2 cells. Supernatant was replaced 24-h before collection in both groups. Significance was calculated using Two-way ANOVA (n = 3/group). b ECAR for LM2 cells after 72 h of TM (0.5 µM) treatment (n = 8/group). c Levels of nucleotide precursors measured by LC–MS after 72 h of TM (0.5 µM) treatment in LM2 cells. Significance was calculated using Two-way ANOVA, followed by Bonferroni’s multiple comparisons test (n = 5/group). d and e Mitochondrial ATP production rate with TM (0.5 µM) treatment in LM2 (d) and ML1 (e) cells. Significance was calculated using Two-way ANOVA, followed by Tukey’s multiple comparisons test (n = 8/group). f Western blot of p-AMPK (Thr172), p-ACC (Ser79), and Complex IV (CIV) in control and TM (0.5 µM) treated LM2 cells. Complex IV is referred to as CIV. g Mitochondrial ATP production rate in LM2 SCR-sh and shCOX17#1, using Seahorse ATP production rate assay. Significance was calculated using Two-way ANOVA, followed by Tukey’s multiple comparisons test (n = 3 for SCR-sh, n = 8 for shCOX17#1). h Western blot of p-AMPK in shCOX17 and SCR-sh LM2 cells. Complex IV is referred to as CIV. i mTOR pathways (p-Raptor Ser792, p-p70S6K Thr389) downstream of phospho-AMPK in LM2 cells. Representative data of two independent experiments are depicted. Results are expressed as mean ± SD. **p < 0.01, ***p < 0.001, ****p < 0.0001.
Fig. 7. AMPK is a mediator of…
Fig. 7. AMPK is a mediator of downstream TM phenotypes.
a, b Impact of pharmacologic activation of AMPK with AICAR (1.25 mM) and A-769662 (12.5 µM) on the invasion of LM2 cells. Representative data from two independent experiments. Analysis was performed by unpaired two-sided t-test (n = 3/group, 5 fields of view/sample). c Western blot showing activation of AMPK by AICAR (1.25 mM) in LM2 cells. d Western blot showing activation of p-ACC by A-769662 (12.5 µM) in LM2 cells. e Control and 0.5 µM TM-treated cells (after 24 h) were transfected with siRNA against a control (C) sequence or AMPK (A). AMPK expression by western blot in LM2 cells. f Rescue of TM-mediated loss of invasion with siAMPK in LM2 cells. Significance was calculated using Two-way ANOVA, followed by Tukey’s multiple comparisons test (n = 3/group, 5 images/sample). Representative data of two independent experiments are depicted. Results are expressed as mean ± SD. **p < 0.01, ****p < 0.0001.

References

    1. Garrido-Castro AC, Lin NU, Polyak K. Insights into molecular classifications of triple-negative breast cancer: improving patient selection for treatment. Cancer Discov. 2019;9:176–198.
    1. Malhotra MK, Emens LA. The evolving management of metastatic triple negative breast cancer. Semin. Oncol. 2020;47:229–237.
    1. Fouani L, Menezes SV, Paulson M, Richardson DR, Kovacevic Z. Metals and metastasis: exploiting the role of metals in cancer metastasis to develop novel anti-metastatic agents. Pharm. Res. 2017;115:275–287.
    1. Frezza M, et al. Novel metals and metal complexes as platforms for cancer therapy. Curr. Pharm. Des. 2010;16:1813–1825.
    1. Weiss KH, et al. WTX101 in patients newly diagnosed with Wilson disease: final results of a global, prospective phase 2 trial. J. Hepatol. 2017;66:S88.
    1. Safi R, et al. Copper signaling axis as a target for prostate cancer therapeutics. Cancer Res. 2014;74:5819–5831.
    1. Ge, E. J. et al. Connecting copper and cancer: from transition metal signalling to metalloplasia. Nat Rev Cancer 10.1038/s41568-021-00417-2 (2021).
    1. Brewer GJ, et al. Treatment of metastatic cancer with tetrathiomolybdate, an anticopper, antiangiogenic agent: Phase I study. Clin. Cancer Res. 2000;6:1–10.
    1. Brewer GJ, Merajver SD. Cancer therapy with tetrathiomolybdate: antiangiogenesis by lowering body copper—a review. Integr. Cancer Ther. 2002;1:327–337.
    1. Hassouneh B, et al. Tetrathiomolybdate promotes tumor necrosis and prevents distant metastases by suppressing angiogenesis in head and neck cancer. Mol. Cancer Ther. 2007;6:1039–1045.
    1. Pan Q, et al. Copper deficiency induced by tetrathiomolybdate suppresses tumor growth and angiogenesis. Cancer Res. 2002;62:4854–4859.
    1. Blockhuys S, et al. Defining the human copper proteome and analysis of its expression variation in cancers. Metallomics. 2017;9:112–123.
    1. Lowndes S, Harris A. Copper chelation as an antiangiogenic therapy. Oncol. Res. 2004;14:529–539.
    1. Marttila-Ichihara F, Auvinen K, Elima K, Jalkanen S, Salmi M. Vascular adhesion protein-1 enhances tumor growth by supporting recruitment of Gr-1+ CD11b+ myeloid cells into tumors. Cancer Res. 2009;69:7875–7883.
    1. Groleau J, et al. Essential role of copper-zinc superoxide dismutase for ischemia-induced neovascularization via modulation of bone marrow-derived endothelial progenitor cells. Arterioscler. Thromb. Vasc. Biol. 2010;30:2173–2181.
    1. Brady DC, et al. Copper is required for oncogenic BRAF signalling and tumorigenesis. Nature. 2014;509:492–496.
    1. Sammons S, Brady D, Vahdat L, Salama AK. Copper suppression as cancer therapy: the rationale for copper chelating agents in. Melanoma Manag. 2016;3:207–216.
    1. Tsang T, et al. Copper is an essential regulator of the autophagic kinases ULK1/2 to drive lung adenocarcinoma. Nat. Cell Biol. 2020;22:412–424.
    1. Liao Y, et al. Inflammation mobilizes copper metabolism to promote colon tumorigenesis via an IL-17-STEAP4-XIAP axis. Nat. Commun. 2020;11:900.
    1. Voli F, et al. Intratumoral copper modulates PD-L1 expression and influences tumor immune evasion. Cancer Res. 2020;80:4129–4144.
    1. Blockhuys S, Zhang X, Wittung-Stafshede P. Single-cell tracking demonstrates copper chaperone Atox1 to be required for breast cancer cell migration. Proc. Natl Acad. Sci. USA. 2020;117:2014–2019.
    1. Pan Q, Rosenthal DT, Bao L, Kleer CG, Merajver SD. Antiangiogenic tetrathiomolybdate protects against Her2/neu-induced breast carcinoma by hypoplastic remodeling of the mammary gland. Clin. Cancer Res. 2009;15:7441–7446.
    1. Chan N, et al. Influencing the tumor microenvironment: a Phase II Study of copper depletion using tetrathiomolybdate in patients with breast cancer at high risk for recurrence and in preclinical models of lung metastases. Clin. Cancer Res. 2017;23:666–676.
    1. O’Shaughnessy J, et al. Phase III study of iniparib plus gemcitabine and carboplatin versus gemcitabine and carboplatin in patients with metastatic triple-negative breast cancer. J. Clin. Oncol. 2014;32:3840–3847.
    1. Hu XC, et al. Cisplatin plus gemcitabine versus paclitaxel plus gemcitabine as first-line therapy for metastatic triple-negative breast cancer (CBCSG006): a randomised, open-label, multicentre, phase 3 trial. Lancet Oncol. 2015;16:436–446.
    1. Pascual G, Domínguez D, Benitah SA, et al. The contributions of cancer cell metabolism to metastasis. Dis. Model. Mech. 2018;11:dmm032920. doi: 10.1242/dmm.032920.
    1. LeBleu VS, et al. PGC-1α mediates mitochondrial biogenesis and oxidative phosphorylation in cancer cells to promote metastasis. Nat. Cell Biol. 2014;16:1001–1015.
    1. Dupuy F, et al. PDK1-dependent metabolic reprogramming dictates metastatic potential in breast cancer. Cell Metab. 2015;22:577–589.
    1. Warburg O, Wind F, Negelein E. The metabolism of tumors in the body. J. Gen. Physiol. 1927;8:519–530.
    1. Pavlova NN, Thompson CB. The emerging hallmarks of cancer metabolism. Cell Metab. 2016;23:27–47.
    1. Cruz-Bermudez A, et al. PGC-1alpha levels correlate with survival in patients with stage III NSCLC and may define a new biomarker to metabolism-targeted therapy. Sci. Rep. 2017;7:16661.
    1. Liu L, et al. S100A4 alters metabolism and promotes invasion of lung cancer cells by up-regulating mitochondrial complex I protein NDUFS2. J. Biol. Chem. 2019;294:7516–7527.
    1. Ashton TM, McKenna WG, Kunz-Schughart LA, Higgins GS. Oxidative phosphorylation as an emerging target in cancer therapy. Clin. Cancer Res. 2018;24:2482–2490.
    1. Curtis C, et al. The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups. Nature. 2012;486:346–352.
    1. Minn AJ, et al. Genes that mediate breast cancer metastasis to lung. Nature. 2005;436:518–524.
    1. Johnstone CN, et al. Functional and molecular characterisation of EO771.LMB tumours, a new C57BL/6-mouse-derived model of spontaneously metastatic mammary cancer. Dis. Models Mech. 2015;8:237–251.
    1. Yomtoubian S, et al. Inhibition of EZH2 catalytic activity selectively targets a metastatic subpopulation in triple-negative breast cancer. Cell Rep. 2020;30:755–770e756.
    1. Ramchandani D, et al. Nanoparticle delivery of miR-708 mimetic impairs breast cancer metastasis. Mol. Cancer Ther. 2019;18:579–591.
    1. Horn D, Barrientos A. Mitochondrial copper metabolism and delivery to cytochrome c oxidase. IUBMB Life. 2008;60:421–429.
    1. Cogliati S, Enriquez JA, Scorrano L. Mitochondrial cristae: where beauty meets functionality. Trends Biochem. Sci. 2016;41:261–273.
    1. Kim BE, Nevitt T, Thiele DJ. Mechanisms for copper acquisition, distribution and regulation. Nat. Chem. Biol. 2008;4:176–185.
    1. Lutsenko S. Human copper homeostasis: a network of interconnected pathways. Curr. Opin. Chem. Biol. 2010;14:211–217.
    1. Takahashi Y, et al. Mammalian copper chaperone Cox17p has an essential role in activation of cytochrome C oxidase and embryonic development. Mol. Cell. Biol. 2002;22:7614–7621.
    1. Hell K, Tzagoloff A, Neupert W, Stuart RA. Identification of Cox20p, a novel protein involved in the maturation and assembly of cytochrome oxidase subunit 2. J. Biol. Chem. 2000;275:4571–4578.
    1. Spinelli JB, Haigis MC. The multifaceted contributions of mitochondria to cellular metabolism. Nat. Cell Biol. 2018;20:745–754.
    1. Gowans GJ, Hardie DG. AMPK: a cellular energy sensor primarily regulated by AMP. Biochem. Soc. Trans. 2014;42:71–75.
    1. Gwinn DM, et al. AMPK phosphorylation of raptor mediates a metabolic checkpoint. Mol. Cell. 2008;30:214–226.
    1. Canto C, Auwerx J. AMP-activated protein kinase and its downstream transcriptional pathways. Cell. Mol. Life Sci. 2010;67:3407–3423.
    1. Choo AY, Yoon SO, Kim SG, Roux PP, Blenis J. Rapamycin differentially inhibits S6Ks and 4E-BP1 to mediate cell-type-specific repression of mRNA translation. Proc. Natl Acad. Sci. USA. 2008;105:17414–17419.
    1. Qin X, Jiang B, Zhang Y. 4E-BP1, a multifactor regulated multifunctional protein. Cell Cycle. 2016;15:781–786.
    1. Zhou H, Huang S. Role of mTOR signaling in tumor cell motility, invasion and metastasis. Curr. Protein Pept. Sci. 2011;12:30–42.
    1. Chen K, et al. Loss of AMPK activation promotes the invasion and metastasis of pancreatic cancer through an HSF1-dependent pathway. Mol. Oncol. 2017;11:1475–1492.
    1. Cool B, et al. Identification and characterization of a small molecule AMPK activator that treats key components of type 2 diabetes and the metabolic syndrome. Cell Metab. 2006;3:403–416.
    1. Pavithra V, et al. Serum levels of metal ions in female patients with breast cancer. J. Clin. Diagn. Res. 2015;9:BC25–c27.
    1. Kim KI, et al. Detection of increased 64Cu uptake by human copper transporter 1 gene overexpression using PET with 64CuCl2 in human breast cancer xenograft model. J. Nucl. Med. 2014;55:1692–1698.
    1. Ishida S, Andreux P, Poitry-Yamate C, Auwerx J, Hanahan D. Bioavailable copper modulates oxidative phosphorylation and growth of tumors. Proc. Natl Acad. Sci. USA. 2013;110:19507–19512.
    1. Ryumon S, et al. Ammonium tetrathiomolybdate enhances the antitumor effect of cisplatin via the suppression of ATPase copper transporting beta in head and neck squamous cell carcinoma. Oncol. Rep. 2019;42:2611–2621.
    1. Viale A, Corti D, Draetta GF. Tumors and mitochondrial respiration: a neglected connection. Cancer Res. 2015;75:3685–3686.
    1. Yu M. Generation, function and diagnostic value of mitochondrial DNA copy number alterations in human cancers. Life Sci. 2011;89:65–71.
    1. Badet J, et al. Specific binding of angiogenin to calf pulmonary artery endothelial cells. Proc. Natl Acad. Sci. USA. 1989;86:8427–8431.
    1. Erler JT, et al. Lysyl oxidase is essential for hypoxia-induced metastasis. Nature. 2006;440:1222–1226.
    1. Kim KK, et al. Tetrathiomolybdate inhibits mitochondrial complex IV and mediates degradation of hypoxia-inducible factor-1alpha in cancer cells. Sci. Rep. 2015;5:14296.
    1. Glasauer A, Sena LA, Diebold LP, Mazar AP, Chandel NS. Targeting SOD1 reduces experimental non-small-cell lung cancer. J. Clin. Investig. 2014;124:117–128.
    1. Shanbhag V, et al. ATP7A delivers copper to the lysyl oxidase family of enzymes and promotes tumorigenesis and metastasis. Proc. Natl Acad. Sci. USA. 2019;116:6836–6841.
    1. Erler JT, et al. Hypoxia-induced lysyl oxidase is a critical mediator of bone marrow cell recruitment to form the premetastatic niche. Cancer Cell. 2009;15:35–44.
    1. Cox TR, et al. LOX-mediated collagen crosslinking is responsible for fibrosis-enhanced metastasis. Cancer Res. 2013;73:1721–1732.
    1. Berven LA, Willard FS, Crouch MF. Role of the p70(S6K) pathway in regulating the actin cytoskeleton and cell migration. Exp. Cell Res. 2004;296:183–195.
    1. Yi Y, et al. Transcriptional suppression of AMPKalpha1 promotes breast cancer metastasis upon oncogene activation. Proc. Natl Acad. Sci. USA. 2020;117:8013–8021.
    1. Saxena M, et al. AMP-activated protein kinase promotes epithelial–mesenchymal transition in cancer cells through Twist1 upregulation. J. Cell Sci. 2018;131:jcs208314. doi: 10.1242/jcs.208314.
    1. Chou CC, et al. AMPK reverses the mesenchymal phenotype of cancer cells by targeting the Akt–MDM2–Foxo3a signaling axis. Cancer Res. 2014;74:4783–4795.
    1. Liu L, et al. Rapamycin inhibits cell motility by suppression of mTOR-mediated S6K1 and 4E-BP1 pathways. Oncogene. 2006;25:7029–7040.
    1. Mullard A. Cancer metabolism pipeline breaks new ground. Nat. Rev. Drug Discov. 2016;15:735–737.
    1. Goodwin PJ, et al. Effect of metformin vs placebo on and metabolic factors in NCIC CTG MA.32. J. Natl Cancer Inst. 2015;107:djv006. doi: 10.1093/jnci/djv006.
    1. Meric-Bernstam F, et al. CB-839, a glutaminase inhibitor, in combination with cabozantinib in patients with clear cell and papillary metastatic renal cell cancer (mRCC): results of a phase I study. J. Clin. Oncol. 2019;37:549–549.
    1. Covini D, et al. Expanding targets for a metabolic therapy of cancer: l-asparaginase. Recent Pat. Anticancer Drug Discov. 2012;7:4–13.
    1. McAlister GC, et al. MultiNotch MS3 enables accurate, sensitive, and multiplexed detection of differential expression across cancer cell line proteomes. Anal. Chem. 2014;86:7150–7158.
    1. Navarrete-Perea J, Yu Q, Gygi SP, Paulo JA. Streamlined tandem mass tag (SL-TMT) protocol: an efficient strategy for quantitative (phospho)proteome profiling using tandem mass tag-synchronous precursor selection-MS3. J. Proteome Res. 2018;17:2226–2236.
    1. Venegas V, Halberg MC. Measurement of mitochondrial DNA copy number. Methods Mol. Biol. 2012;837:327–335.

Source: PubMed

3
Předplatit