Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

NRF2 mediates melanoma addiction to GCDH by modulating apoptotic signalling

Abstract

Tumour dependency on specific metabolic signals has been demonstrated and often guided numerous therapeutic approaches. We identify melanoma addiction to the mitochondrial protein glutaryl-CoA dehydrogenase (GCDH), which functions in lysine metabolism and controls protein glutarylation. GCDH knockdown induced cell death programmes in melanoma cells, an activity blocked by inhibition of the upstream lysine catabolism enzyme DHTKD1. The transcription factor NRF2 mediates GCDH-dependent melanoma cell death programmes. Mechanistically, GCDH knockdown induces NRF2 glutarylation, increasing its stability and DNA binding activity, with a concomitant transcriptional upregulation of ATF4, ATF3, DDIT3 and CHAC1, resulting in cell death. In vivo, inducible inactivation of GCDH effectively inhibited melanoma tumour growth. Correspondingly, reduced GCDH expression correlated with improved survival of patients with melanoma. These findings identify melanoma cell addiction to GCDH, limiting apoptotic signalling by controlling NRF2 glutarylation. Inhibiting the GCDH pathway could thus represent a therapeutic approach to treat melanoma.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: GCDH is required for melanoma cell survival.
Fig. 2: GCDH inhibition promotes DHTKD1-dependent cell death.
Fig. 3: GCDH loss increases NRF2 levels and enhances cell death.
Fig. 4: Cell death following GCDH inhibition is NRF2 dependent.
Fig. 5: Lysine glutarylation attenuates KEAP1 binding increasing NRF2 stability.
Fig. 6: Inducible GCDH KD decreases melanoma proliferation and tumourigenesis.

Similar content being viewed by others

Data availability

RNA-seq data that support the findings of this study have been deposited in Gene Expression Omnibus under accession code GSE200424. The pan-cancer data were derived from the TCGA Research Network: http://cancergenome.nih.gov/. The dataset derived from this resource that supports the findings of this study is available in cBioPortal (https://www.cbioportal.org/). Databases/datasets used in the study for human genome and gene annotations: human genome hg38, http://ftp.ensembl.org/pub/release-84/fasta/homo_sapiens/dna/; Ensembl human gene annotations version 84, http://ftp.ensembl.org/pub/release-84/gtf/homo_sapiens/. Source data are provided with this paper. All other data supporting the findings of this study are available from the corresponding author on reasonable request.

References

  1. Palm, W. & Thompson, C. B. Nutrient acquisition strategies of mammalian cells. Nature 546, 234–242 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Zhu, J. & Thompson, C. B. Metabolic regulation of cell growth and proliferation. Nat. Rev. Mol. Cell Biol. 20, 436–450 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Vander Heiden, M. G. & DeBerardinis, R. J. Understanding the intersections between metabolism and cancer biology. Cell 168, 657–669 (2017).

    Article  PubMed Central  CAS  Google Scholar 

  4. Lieu, E. L., Nguyen, T., Rhyne, S. & Kim, J. Amino acids in cancer. Exp. Mol. Med 52, 15–30 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Hirschey, M. D. & Zhao, Y. Metabolic regulation by lysine malonylation, succinylation, and glutarylation. Mol. Cell Proteom. 14, 2308–2315 (2015).

    Article  CAS  Google Scholar 

  6. Altman, B. J., Stine, Z. E. & Dang, C. V. From Krebs to clinic: glutamine metabolism to cancer therapy. Nat. Rev. Cancer 16, 619–634 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Butler, M., van der Meer, L. T. & van Leeuwen, F. N. Amino acid depletion therapies: starving cancer cells to death. Trends Endocrinol. Metab. 32, 367–381 (2021).

    Article  CAS  PubMed  Google Scholar 

  8. Lukey, M. J., Katt, W. P. & Cerione, R. A. Targeting amino acid metabolism for cancer therapy. Drug Discov. Today 22, 796–804 (2017).

    Article  CAS  PubMed  Google Scholar 

  9. Chaturvedi, S., Hoffman, R. M. & Bertino, J. R. Exploiting methionine restriction for cancer treatment. Biochem. Pharmacol. 154, 170–173 (2018).

    Article  CAS  PubMed  Google Scholar 

  10. Knott, S. R. V. et al. Asparagine bioavailability governs metastasis in a model of breast cancer. Nature 554, 378–381 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Maddocks, O. D. K. et al. Modulating the therapeutic response of tumours to dietary serine and glycine starvation. Nature 544, 372–376 (2017).

    Article  CAS  PubMed  Google Scholar 

  12. Chang, C. H. et al. Metabolic competition in the tumor microenvironment is a driver of cancer progression. Cell 162, 1229–1241 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Nakaya, M. et al. Inflammatory T cell responses rely on amino acid transporter ASCT2 facilitation of glutamine uptake and mTORC1 kinase activation. Immunity 40, 692–705 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Cruzat, V., Macedo Rogero, M., Noel Keane, K., Curi, R. & Newsholme, P. Glutamine: metabolism and immune function, supplementation and clinical translation. Nutrients 10, 1564 (2018).

    Article  PubMed Central  CAS  Google Scholar 

  15. O’Neill, L. A., Kishton, R. J. & Rathmell, J. A guide to immunometabolism for immunologists. Nat. Rev. Immunol. 16, 553–565 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Ananieva, E. Targeting amino acid metabolism in cancer growth and anti-tumor immune response. World J. Biol. Chem. 6, 281–289 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  17. Pathria, G. & Ronai, Z. A. Harnessing the co-vulnerabilities of amino acid-restricted cancers. Cell Metab. 33, 9–20 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Pathria, G. et al. Translational reprogramming marks adaptation to asparagine restriction in cancer. Nat. Cell Biol. 21, 1590–1603 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Kim, H. et al. PRMT5 control of cGAS/STING and NLRC5 pathways defines melanoma response to antitumor immunity. Sci. Transl. Med. 12, eaaz5683 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Leandro, J. & Houten, S. M. The lysine degradation pathway: subcellular compartmentalization and enzyme deficiencies. Mol. Genet. Metab. 131, 14–22 (2020).

    Article  CAS  PubMed  Google Scholar 

  21. Fallarino, F. et al. Modulation of tryptophan catabolism by regulatory T cells. Nat. Immunol. 4, 1206–1212 (2003).

    Article  CAS  PubMed  Google Scholar 

  22. Schmiesing, J. et al. Disease-linked glutarylation impairs function and interactions of mitochondrial proteins and contributes to mitochondrial heterogeneity. Cell Rep. 24, 2946–2956 (2018).

    Article  CAS  PubMed  Google Scholar 

  23. Tan, M. et al. Lysine glutarylation is a protein posttranslational modification regulated by SIRT5. Cell Metab. 19, 605–617 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Biagosch, C. et al. Elevated glutaric acid levels in Dhtkd1-/Gcdh-double knockout mice challenge our current understanding of lysine metabolism. Biochim. Biophys. Acta Mol. Basis Dis. 1863, 2220–2228 (2017).

    Article  CAS  PubMed  Google Scholar 

  25. Wajner, M., Amaral, A. U., Leipnitz, G. & Seminotti, B. Pathogenesis of brain damage in glutaric acidemia type I: lessons from the genetic mice model. Int J. Dev. Neurosci. 78, 215–221 (2019).

    Article  CAS  PubMed  Google Scholar 

  26. Zinnanti, W. J. et al. A diet-induced mouse model for glutaric aciduria type I. Brain 129, 899–910 (2006).

    Article  PubMed  Google Scholar 

  27. Seminotti, B. et al. Oxidative stress, disrupted energy metabolism, and altered signaling pathways in glutaryl-CoA dehydrogenase knockout mice: potential implications of quinolinic acid toxicity in the neuropathology of glutaric acidemia type I. Mol. Neurobiol. 53, 6459–6475 (2016).

    Article  CAS  PubMed  Google Scholar 

  28. Rojo de la Vega, M., Chapman, E. & Zhang, D. D. NRF2 and the hallmarks of cancer. Cancer Cell 34, 21–43 (2018).

    Article  CAS  PubMed  Google Scholar 

  29. Malhotra, D. et al. Global mapping of binding sites for Nrf2 identifies novel targets in cell survival response through ChIP–seq profiling and network analysis. Nucleic Acids Res. 38, 5718–5734 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Hoetzenecker, W. et al. ROS-induced ATF3 causes susceptibility to secondary infections during sepsis-associated immunosuppression. Nat. Med. 18, 128–134 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Romero, R. et al. Keap1 mutation renders lung adenocarcinomas dependent on Slc33a1. Nat. Cancer 1, 589–602 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Teske, N. et al. Chemical hypoxia-induced integrated stress response activation in oligodendrocytes is mediated by the transcription factor nuclear factor (erythroid-derived 2)-like 2 (NRF2). J. Neurochem. 144, 285–301 (2018).

    Article  CAS  PubMed  Google Scholar 

  33. Wakabayashi, N. et al. Keap1-null mutation leads to postnatal lethality due to constitutive Nrf2 activation. Nat. Genet. 35, 238–245 (2003).

    Article  CAS  PubMed  Google Scholar 

  34. Menegon, S., Columbano, A. & Giordano, S. The dual roles of NRF2 in cancer. Trends Mol. Med. 22, 578–593 (2016).

    Article  CAS  PubMed  Google Scholar 

  35. Sporn, M. B. & Liby, K. T. NRF2 and cancer: the good, the bad and the importance of context. Nat. Rev. Cancer 12, 564–571 (2012).

    Article  CAS  PubMed  Google Scholar 

  36. Wu, Z. et al. TPO-induced metabolic reprogramming drives liver metastasis of colorectal cancer CD110+ tumor-initiating cells. Cell Stem Cell 17, 47–59 (2015).

    Article  CAS  PubMed  Google Scholar 

  37. Cantor, J. R. et al. Physiologic medium rewires cellular metabolism and reveals uric acid as an endogenous inhibitor of UMP synthase. Cell 169, 258–272 e217 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Mungrue, I. N., Pagnon, J., Kohannim, O., Gargalovic, P. S. & Lusis, A. J. CHAC1/MGC4504 is a novel proapoptotic component of the unfolded protein response, downstream of the ATF4–ATF3–CHOP cascade. J. Immunol. 182, 466–476 (2009).

    Article  CAS  PubMed  Google Scholar 

  39. Cazzalini, O., Scovassi, A. I., Savio, M., Stivala, L. A. & Prosperi, E. Multiple roles of the cell cycle inhibitor p21(CDKN1A) in the DNA damage response. Mutat. Res. 704, 12–20 (2010).

    Article  CAS  PubMed  Google Scholar 

  40. Afonyushkin, T. et al. Oxidized phospholipids regulate expression of ATF4 and VEGF in endothelial cells via NRF2-dependent mechanism: novel point of convergence between electrophilic and unfolded protein stress pathways. Arterioscler. Thromb. Vasc. Biol. 30, 1007–1013 (2010).

    Article  CAS  PubMed  Google Scholar 

  41. Kim, K. H., Jeong, J. Y., Surh, Y. J. & Kim, K. W. Expression of stress-response ATF3 is mediated by Nrf2 in astrocytes. Nucleic Acids Res. 38, 48–59 (2010).

    Article  CAS  PubMed  Google Scholar 

  42. Romero, R. et al. Keap1 loss promotes Kras-driven lung cancer and results in dependence on glutaminolysis. Nat. Med. 23, 1362–1368 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Cullinan, S. B. et al. Nrf2 is a direct PERK substrate and effector of PERK-dependent cell survival. Mol. Cell. Biol. 23, 7198–7209 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Kobayashi, A. et al. Oxidative stress sensor Keap1 functions as an adaptor for Cul3-based E3 ligase to regulate proteasomal degradation of Nrf2. Mol. Cell. Biol. 24, 7130–7139 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. He, F. et al. NRF2 activates growth factor genes and downstream AKT signaling to induce mouse and human hepatomegaly. J. Hepatol. 72, 1182–1195 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Sanghvi, V. R. et al. The oncogenic action of NRF2 depends on de-glycation by fructosamine-3-kinase. Cell 178, 807–819 e821 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Sun, Z., Chin, Y. E. & Zhang, D. D. Acetylation of Nrf2 by p300/CBP augments promoter-specific DNA binding of Nrf2 during the antioxidant response. Mol. Cell. Biol. 29, 2658–2672 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. He, C. H. et al. Identification of activating transcription factor 4 (ATF4) as an Nrf2-interacting protein. Implication for heme oxygenase-1 gene regulation. J. Biol. Chem. 276, 20858–20865 (2001).

    Article  CAS  PubMed  Google Scholar 

  49. Fujita, K., Maeda, D., Xiao, Q. & Srinivasula, S. M. Nrf2-mediated induction of p62 controls Toll-like receptor-4-driven aggresome-like induced structure formation and autophagic degradation. Proc. Natl Acad. Sci. USA 108, 1427–1432 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Ratnikov, B. et al. Glutamate and asparagine cataplerosis underlie glutamine addiction in melanoma. Oncotarget 6, 7379–7389 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  51. Verma, S., Ali, A., Arora, S. & Banerjea, A. C. Inhibition of β-TrcP-dependent ubiquitination of p53 by HIV-1 Vpu promotes p53-mediated apoptosis in human T cells. Blood 117, 6600–6607 (2011).

    Article  CAS  PubMed  Google Scholar 

  52. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  53. Li, B. & Dewey, C. N. RSEM: accurate transcript quantification from RNA-seq data with or without a reference genome. BMC Bioinformatics 12, 323 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Ewels, P., Magnusson, M., Lundin, S. & Kaller, M. MultiQC: summarize analysis results for multiple tools and samples in a single report. Bioinformatics 32, 3047–3048 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  56. Gu, Z., Eils, R. & Schlesner, M. Complex heatmaps reveal patterns and correlations in multidimensional genomic data. Bioinformatics 32, 2847–2849 (2016).

    Article  CAS  PubMed  Google Scholar 

  57. Hoadley, K. A. et al. Cell-of-origin patterns dominate the molecular classification of 10,000 tumors from 33 types of cancer. Cell 173, 291–304 e296 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Cerami, E. et al. The cBio Cancer Genomics Portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2, 401–404 (2012).

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

We thank D. D. Zhang (University of Arizona) and V. R. Sanghvi (University of Miami) for providing us with the NRF2 mutants (HA-NRF2 K6R, K12R and K18R) and H.-G. Wendel for providing us with NRF2-glycation mutant (NRF2E79V-A6), Y. Altman for help with flow cytometry analysis, members of the Prebys Center for Chemical Genomics for help with the screen for GCDH inhibitors and members of the Ronai lab for discussions. National Cancer Insitute (NCI) support (R35CA197465) to Z.R. and SBP support for translational initiatives (to E.S. and Z.A.R.) is gratefully acknowledged. Sanford Burnham Prebys Shared Resources are supported by an NCI Cancer Center Support Grant (P30 CA030199). The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

S.V., G.P., A.K., D.S. and Y.F. performed experiments; D.C., E.R. and R.M. performed bioinformatic analyses; Z.A.R. and S.V. designed the studies; E.S., C.-T.M., S.H.O. and M.J. designed GCDH targeting studies, and Z.A.R. and S.V. wrote the manuscript.

Corresponding author

Correspondence to Ze’ev A. Ronai.

Ethics declarations

Competing interests

Z.A.R. and E.R. (fully divested) are founders and scientific advisors for Pangea Therapeutics. All other authors declare no conflicts of interest.

Peer review

Peer review information

Nature Cell Biology thanks Afshin Samali, Thales Papagiannakopoulos and the other, anonymous, reviewer for their contribution to the peer review of this work. Peer reviewer reports are available.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 GCDH inhibition promotes apoptosis of melanoma cells.

(A) UACC903 and 1205LU melanoma cells were transduced with siRNAs targeting AASS, AADAT, DHTKD1, GCDH, or ECHS1 or with a control sequence using Jetprime for 96 hr, followed by an analysis of cell viability. (B) Various melanoma cells were transfected with siRNAs targeting AASS, AADAT, DHTKD1, GCDH, or ECHS1 or with a control sequence for 72 hr, and then lysates were subjected to western analysis with indicated antibodies. (C) Cell growth after GCDH KD with two independent siRNAs for 0–96 hr in UACC903 or 1205LU cells. Proliferation was analyzed by counting cells at indicated time points. (D) UACC903 or 1205LU cells were transfected with GCDH siRNA for 72 hr, and then lysates were subjected to western analysis with indicated antibodies. (E) Viability assay of control or GCDH KD A375 cells treated with the caspase inhibitor Z-VAD(OH)-FMK (10µM for 48 hr). (F) A375 cells transfected with GCDH siRNA were grown in the indicated growth medium, and then viability was determined 96 hr-post transfections. (G) Measurement of basal, maximum oxygen consumption rate (OCR), and spare respiratory capacity using Agilent Seahorse XF Analyzers. A375 cells were transfected with GCDH siRNA for 48 hours before analysis. (H) Viability assay of immortalized H3A cells, 96 hr after transfection with indicated constructs. Viability was measured by quantifying crystal violet staining. (I) Immortalized H3A cells were transfected with siRNAs targeting AASS, AADAT, DHTKD1, GCDH, or ECHS1 or with a control sequence for 72 hours, then lysates were subjected to western analysis with indicated antibodies. Data are representative of three experiments for (B, D, and I) and presented as the mean values ± SD of n = 3 (A, C, and H), n= 4 (E and F) independent experiments and mean values ± SD of n = 6 technical replicates for (G). Each p value is adjusted to account for multiple comparisons. Statistical significance (indicated p-value or ns- not significant w.r.t control) was calculated using One-way ANOVA for (B, E, F and H) and two-way ANOVA for (C). Statistics source data for Extended Data Fig. 1 can be found in Source data files - Extended Data Fig. 1 and uncropped scans of all blots and gels for Extended Data Fig. 1 can be found in Source data files for blots - Extended Fig. 1.

Source data

Extended Data Fig. 2 GCDH expression correlates with melanoma patients outcome.

(A) Survival correlation analysis of GCDH expression in various cancer subtypes (prostate adenocarcinoma (PRAD), breast cancer (BRCA), diffuse large B-cell lymphoma (DLBC), glioblastoma (GBM), acute myeloid leukemia (LAML), liver hepatocellular carcinoma (LIHC), lung adenocarcinoma (LUAD), and lung squamous cell carcinoma (LUSC), using TCGA. (B) Comparison of patient survival based on analysis of NRF2 expression in melanoma patients using TCGA. Total number (n) of patients = 428; n (NRF2 high) =362 and n (NRF2 low) =66. (C) Scatterplot depicting a comparison of GCDH and NRF2 expression levels in TCGA-SKCM (melanoma) patient cohort. GCDH and NRF2 expressions are anti-correlated (Spearman correlation coefficient = -0.41). (D) Boxplots depicting GCDH expression in NRF2 high (n=362) and NRF2 low (n=66) patient samples stratified according to survival analysis of TCGA-SKCM patient cohort based on NRF2 expression (refer to NRF2 survival analysis in (B). The difference in expression of GCDH between NRF2 high and low groups is statistically significant (p-value = 8.653e-14, calculated using two-sided Wilcoxon Rank Sum test). NRF2 low group minimum = 242.2, maximum = 1907, median = 815.6, 1st quartile (25th percentile) = 633 and 3rd quartile (75th percentile) = 1142.5. NRF2 high group minimum = 132.8, maximum =1242.4, median =556.4, 1st quartile (25th percentile) = 446 and 3rd quartile (75th percentile) = 686.6. (E) Various cancer cells were transfected with siRNAs targeting GCDH or with a control sequence for 72 hr, and then lysates were subjected to confirm GCDH-KD. (F) GCDH expression was compared among various cancer cells and immortalized H3A cells. HSP90 was used as a loading control. Data are representative of three experiments. Uncropped scans of all blots and gels for Extended Data Fig. 2 can be found in Source data files for blots - Extended figure 2.

Source data

Extended Data Fig. 3 DHTKD1 KD rescues GCDH KD- induced phenotypes.

(A) Selected enriched canonical pathways from Ingenuity Pathway Analysis (IPA) of siGCDH vs. siControl differentially expressed genes. Shown pathways were selected from the top 20 canonical pathways based on enrichment p-value (p-value < 5.14e-3). (B) RT-qPCR analysis of UACC903 cells for relative expression of indicated transcripts following GCDH-KD or DHTKD1-KD alone or GCDH/DHTKD1 double KD. (C) Western blot analysis with indicated antibodies of lysates from A375 cells transfected with indicated siRNA for 96 hr. (D) RT-qPCR analysis of A375 cells for relative expression of indicated transcripts following GCDH-KD. (E) Western blot analysis on A375 cells after GCDH KD for 24–96 hr in A375 cells. Cells were transfected with siRNA targeting GCDH, and cell lysates were prepared at indicated time points. (F) SubG0 DNA content analysis by flow cytometry to evaluate A375 cell apoptosis. A375 cells were transfected for 72 hr with indicated siRNAs, harvested and fixed in ethanol, and stained with propidium iodide (PI). (G) Western blot analysis of indicated proteins in A375 cells following GCDH-KD or DHTKD1-KD alone or GCDH/DHTKD1 double KD. (H) Rescue of cell death seen in GCDH-KD 1205LU by DHTKD1-KD. Viability was measured by quantifying crystal violet staining. (I) Western blot analysis of indicated proteins in A375 lines. Data are representative of three experiments (C, E, G, and I), n= 1 experiment for F and presented as the mean values ± SD of n = 3 independent experiments for B, D and H. Statistical significance (indicated p-value or ns- not significant w.r.t control) was calculated using one-way ANOVA. Each p value is adjusted to account for multiple comparisons. Statistics source data for Extended Data Fig. 3 can be found in Source data files - Extended Data Fig. 3 and uncropped scans of all blots and gels for Extended Data Fig. 3 can be found in Source data files for blots - Extended figure 3.

Source data

Extended Data Fig. 4 GCDH antagonizes NRF2-mediated activation of ATF3/4-dependent apoptotic signaling.

(A) GC-MS analysis of glutarate concentrations in A375 cells. (B) Western blot analysis of indicated proteins in UACC903 cells, 72 hr after transfection with siRNAs targeting GCDH, NRF2, or DHTKD1 or combinations thereof. (C) RT-qPCR analysis of NRF2 transcript levels in A375 and UACC903 cells, 72 hr following transfection with GCDH siRNAs. (D) RT-qPCR analysis of ATF3, ATF4, DDIT3, and CHAC1 transcript levels in UACC903 cells, 72 hr following transfection with indicated siRNAs. (E) Western blot analysis of indicated proteins in UACC903 cells 72 hr following transfection with siRNAs targeting GCDH, ATF3, or DHTKD1 or combinations thereof. After quantification, signals obtained in panels B and E for 3 independent experiments were used to calculate the ratio of the indicated protein relative to HSP90 levels, described as fold change (FD) +/-SD for indicated proteins. (F) RT-qPCR analysis of ATF3, ATF4, DDIT3, and CHAC1 expression levels in UACC903 following transfection with indicated siRNAs. Data are representative of three experiments for (B and E), presented as the mean values ± SD of n = 3 independent experiments for C, D and F and mean values ± SD of n = 3 technical replicates for A. Statistical significance (indicated p-value or ns- not significant w.r.t control) was calculated using one-way ANOVA. Each p value is adjusted to account for multiple comparisons. Statistics source data for Extended Data Fig. 4 can be found in Source data files - Extended Data Fig. 4 and uncropped scans of all blots and gels for Extended Data Fig. 4 can be found in Source data files for blots - Extended figure 4.

Source data

Extended Data Fig. 5 Cell death upon GCDH KD in melanoma is ATF3/4-dependent.

(A) Selected upstream regulators and their activation z-scores obtained from Ingenuity Pathway Analysis (IPA) of differentially expressed (DE) genes in siGCDH vs. siControl comparisons in A375 (melanoma), SK-Hep1 (liver cancer), and SKBR3 (breast cancer). (B) Heatmap of log2 fold-changes of selected targets of ATF4-ATF3 computed from siGCDH vs. siControl comparisons in melanoma (A375), liver cancer (SK-Hep1), and breast cancer (SKBR3). Rows (genes) were hierarchically clustered. Missing values correspond to genes that were not expressed in the given samples. (C) RTqPCR analysis of ATF3, ATF4, DDIT3 and CHAC1 expression in various cancer cells transfected with indicated siRNAs. (D) Western blot analysis of indicated proteins in indicated cells 72 hr following transfection with control or GCDH siRNAs. Data are representative of three experiments for (D) and presented as the mean values +/- SD of n = 3 independent experiments for (C). Statistical significance (indicated p-value or ns- not significant w.r.t control) was calculated using one-way ANOVA. Each p value is adjusted to account for multiple comparisons. Statistics source data for Extended Data Fig. 5 can be found in Source data files - Extended Data Fig. 5 and uncropped scans of all blots and gels for Extended Data Fig. 5 can be found in Source data files for blots - Extended figure 5.

Source data

Extended Data Fig. 6 Lysine glutarylation antagonizes KEAP1 binding, increasing NRF2 stability.

(A) Cycloheximide (CHX) chase analysis of HA-NRF2 stability in control and GCDH KD HEK-293T cells ectopically expressing HA-NRF2. HEK293T cells were transfected for 72 hr with indicated constructs and treated with 50 µM Cycloheximide (CHX) for different time periods, followed by western blotting with indicated antibodies. After quantification, signals obtained were used to calculate HA-NRF2/HSP90 ratios and described as fold change relative to CHX treatment time = 0. (B) Immunoprecipitation and Western blot analysis of A375 transfected with indicated constructs. Cells were treated with the proteasomal inhibitor MG132 for 4 hr, followed by IP/Western blotting analysis with antibodies to detect K-Glu PTM, KEAP1, and NRF2. (C) Immunoprecipitation and Western blot analysis of UACC903 cells transfected with indicated constructs as described in (B). (D) HEK293T cells were transfected with HA-NRF2 for 72 hr, and NRF2 glutarylation was measured in HA-NRF2 pull-downs fractions using HA-binding beads. (E) Enrichment of NRF2 glutarylation in nuclear fractions was measured in HA-NRF2 pull-downs from HEK293T cells transfected with HA-NRF2 after an initial cell fractionation step to isolate membrane (MF), cytoplasmic (CF), and nuclear (NF) fractions. Successful fractionation was confirmed by immunoblotting with indicated markers: MF (E-cadherin), CF (GAPDH), and NF (Histone H3). (F) HEK293T cells transfected with indicated constructs and IP of indicated HA-NRF2 mutants using HA-affinity beads were performed, followed by Western blot analysis using K-Glu and HA-tag antibodies. (G) HEK293T cells were transfected with indicated constructs and IP of various HA-NRF2 mutants using HA-affinity beads, followed by Western blot analysis using K-Glu and HA-tag antibodies. After quantification, signals obtained were used to calculate K-Glu/HA-NRF2 ratios and described as fold change relative to HA-NRF2 control. After quantification, signals obtained in panels C for 3 independent experiments were used to calculate the ratio of the indicated protein relative to NRF2 levels in IP samples, described as fold change (FD) +/-SD for indicated proteins. Data are representative of three experiments. Statistics source data for Extended Data Fig. 6 can be found in Source data files - Extended Data Fig. 6 and uncropped scans of all blots and gels for Extended Data Fig. 6 can be found in Source data files for blots - Extended figure 6.

Source data

Extended Data Fig. 7 Melanoma Addiction to GCDH is mediated by NRF2.

The model shows GCDH control of NRF2 glutarylation and the implications for ATF4-ATF3-DDIT3-CHAC1 signaling culminating in apoptosis of melanoma cells subjected to GCDH inhibition.

Supplementary information

Supplementary Fig. 1

Gating strategy for Extended Data Fig. 3f.

Reporting Summary

Peer Review File

Supplementary Table

Supplementary Table 1. Patient survival based on GCDH expression in TCGA of melanoma specimens (relate to Fig. 1i). Supplementary Table 2. Differential gene expression in melanoma A375 cells subjected to siGCDH compared with siControl (batch 1). Supplementary Tables 3–5. Differential gene expression in breast cancer SKBR3 (Table 3), liver cancer SK-HEP1 (Table 4) and melanoma A375 cells (Table 5, batch 2), based on RNA-seq analyses of siGCDH compared with siControl-treated cells. Supplementary Table 6. Sequence of the primers used in this study. Supplementary Table 7. Complete list of antibodies used in this study.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 1

Unprocessed western blots.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 2

Unprocessed western blots.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 3

Unprocessed western blots.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 4

Unprocessed western blots.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 5

Unprocessed western blots.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 6

Unprocessed western blots.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 1

Unprocessed western blots.

Source Data Extended Data Fig. 2

Unprocessed western blots.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 3

Unprocessed western blots.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 4

Unprocessed western blots.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 5

Unprocessed western blots.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 6

Unprocessed western blots.

Rights and permissions

Springer Nature or its licensor holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Verma, S., Crawford, D., Khateb, A. et al. NRF2 mediates melanoma addiction to GCDH by modulating apoptotic signalling. Nat Cell Biol 24, 1422–1432 (2022). https://doi.org/10.1038/s41556-022-00985-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-022-00985-x

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer