Multi-omics analysis of disulfidptosis regulators and therapeutic potential reveals glycogen synthase 1 as a disulfidptosis triggering target for triple-negative breast cancer

Jindong Xie1, Xinpei Deng1, Yi Xie1, Hongbo Zhu2, Peng Liu1, Wei Deng1, Li Ning1, Yuhui Tang1, Yuying Sun1, Hailin Tang1, Manbo Cai2(), Xiaoming Xie1(), Yutian Zou1()

PDF
MedComm ›› 2024, Vol. 5 ›› Issue (3) : e502. DOI: 10.1002/mco2.502
ORIGINAL ARTICLE

Multi-omics analysis of disulfidptosis regulators and therapeutic potential reveals glycogen synthase 1 as a disulfidptosis triggering target for triple-negative breast cancer

  • Jindong Xie1, Xinpei Deng1, Yi Xie1, Hongbo Zhu2, Peng Liu1, Wei Deng1, Li Ning1, Yuhui Tang1, Yuying Sun1, Hailin Tang1, Manbo Cai2(), Xiaoming Xie1(), Yutian Zou1()
Author information +
History +

Abstract

Disruption of disulfide homeostasis during biological processes can have fatal consequences. Excess disulfides induce cell death in a novel manner, termed as “disulfidptosis.” However, the specific mechanism of disulfidptosis has not yet been elucidated. To determine the cancer types sensitive to disulfidptosis and outline the corresponding treatment strategies, we firstly investigated the crucial functions of disulfidptosis regulators pan-cancer at multi-omics levels. We found that different tumor types expressed dysregulated levels of disulfidptosis regulators, most of which had an impact on tumor prognosis. Moreover, we calculated the disulfidptosis activity score in tumors and validated it using multiple independent datasets. Additionally, we found that disulfidptosis activity was correlated with classic biological processes and pathways in various cancers. Disulfidptosis activity was also associated with tumor immune characteristics and could predict immunotherapy outcomes. Notably, the disulfidptosis regulator, glycogen synthase 1 (GYS1), was identified as a promising target for triple-negative breast cancer and validated via in vitro and in vivo experiments. In conclusion, our study elucidated the complex molecular phenotypes and clinicopathological correlations of disulfidptosis regulators in tumors, laying a solid foundation for the development of disulfidptosis-targeting strategies for cancer treatment.

Keywords

disulfidptosis / pan-cancer / prognosis / single-cell RNA-seq / tumor microenvironment

Cite this article

Download citation ▾
Jindong Xie, Xinpei Deng, Yi Xie, Hongbo Zhu, Peng Liu, Wei Deng, Li Ning, Yuhui Tang, Yuying Sun, Hailin Tang, Manbo Cai, Xiaoming Xie, Yutian Zou. Multi-omics analysis of disulfidptosis regulators and therapeutic potential reveals glycogen synthase 1 as a disulfidptosis triggering target for triple-negative breast cancer. MedComm, 2024, 5(3): e502 https://doi.org/10.1002/mco2.502

References

1 X Liu, L Nie, Y Zhang, et al. Actin cytoskeleton vulnerability to disulfide stress mediates disulfidptosis. Nat Cell Biol. 2023;25(3):404-414.
2 Y Zou, J Xie, S Zheng, et al. Leveraging diverse cell-death patterns to predict the prognosis and drug sensitivity of triple-negative breast cancer patients after surgery. Int J Surg. 2022;107:106936.
3 DL Tang, R Kang, T Vanden Berghe, P Vandenabeele, G Kroemer. The molecular machinery of regulated cell death. Cell Res. 2019;29(5):347-364.
4 EB Daly, T Wind, XM Jiang, L Sun, PJ Hogg. Secretion of phosphoglycerate kinase from tumour cells is controlled by oxygen-sensing hydroxylases. Biochim Biophys Acta. 2004;1691(1):17-22.
5 PJ Hogg. Biological regulation through protein disulfide bond cleavage. Redox Rep. 2002;7(2):71-77.
6 R Xie, P Zhang, L Cai, et al. Tumor-specific cyclic amplification of oxidative stress by disulfide-loaded fluoropolymer nanogels. Eur J Pharm Biopharm. 2022;180:212-223.
7 X Liu, L Zhuang, B Gan. Disulfidptosis: disulfide stress-induced cell death. Trends Cell Biol. 2023.
8 C Chen, M Shen, H Liao, et al. A paclitaxel and microRNA-124 coloaded stepped cleavable nanosystem against triple negative breast cancer. J Nanobiotechnol. 2021;19(1):55.
9 Y Wang, Y Jiang, D Wei, et al. Nanoparticle-mediated convection-enhanced delivery of a DNA intercalator to gliomas circumvents temozolomide resistance. Nat Biomed Eng. 2021;5(9):1048-1058.
10 Q Zong, K Wang, X Xiao, et al. Amplification of tumor oxidative stresses by poly(disulfide acetal) for multidrug resistance reversal. Biomaterials. 2021;276:121005.
11 X Wang, L Ren, Z Diao, et al. Robust spontaneous Raman flow cytometry for single-cell metabolic phenome profiling via pDEP-DLD-RFC. Adv Sci (Weinh). 2023;10(16):e2207497.
12 X Liu, K Olszewski, Y Zhang, et al. Cystine transporter regulation of pentose phosphate pathway dependency and disulfide stress exposes a targetable metabolic vulnerability in cancer. Nat Cell Biol. 2020;22(4):476-486.
13 C Wu, J Tan, X Wang, et al. Pan-cancer analyses reveal molecular and clinical characteristics of cuproptosis regulators. iMeta. 2022;2(1):Portico.
14 Z Liu, Q Zhao, ZX Zuo, et al. Systematic analysis of the aberrances and functional implications of ferroptosis in cancer. iScience. 2020;23(7):101302.
15 J Huang, J Zhang, F Zhang, et al. Identification of a disulfidptosis-related genes signature for prognostic implication in lung adenocarcinoma. Comput Biol Med. 2023;165:107402.
16 H Chen, W Yang, Y Li, L Ma, Z Ji. Leveraging a disulfidptosis-based signature to improve the survival and drug sensitivity of bladder cancer patients. Front Immunol. 2023;14:1198878.
17 M Lehto, M Stoffel, L Groop, R Espinosa, MM Le Beau 3rd, GI Bell. Assignment of the gene encoding glycogen synthase (GYS) to human chromosome 19, band q13.3. Genomics. 1993;15(2):460-461.
18 JC Ferrer, C Favre, RR Gomis, et al. Control of glycogen deposition. FEBS Lett. 2003;546(1):127-132.
19 JN Nielsen, EA Richter. Regulation of glycogen synthase in skeletal muscle during exercise. Acta Physiol Scand. 2003;178(4):309-319.
20 JC Lopez-Ramos, J Duran, A Gruart, JJ Guinovart, JM Delgado-Garcia. Role of brain glycogen in the response to hypoxia and in susceptibility to epilepsy. Front Cell Neurosci. 2015;9:431.
21 JM Cameron, V Levandovskiy, N MacKay, et al. Identification of a novel mutation in GYS1 (muscle-specific glycogen synthase) resulting in sudden cardiac death, that is diagnosable from skin fibroblasts. Mol Genet Metab. 2009;98(4):378-382.
22 BA Pederson, H Chen, JM Schroeder, W Shou, AA DePaoli-Roach, PJ Roach. Abnormal cardiac development in the absence of heart glycogen. Mol Cell Biol. 2004;24(16):7179-7187.
23 S Ahonen, S Nitschke, TR Grossman, et al. Gys1 antisense therapy rescues neuropathological bases of murine Lafora disease. Brain. 2021;144(10):2985-2993.
24 KJ Donohue, B Fitzsimmons, RC Bruntz, et al. Gys1 antisense therapy prevents disease-driving aggregates and epileptiform discharges in a Lafora disease mouse model. Neurotherapeutics. 2023;20(6):1808-1819.
25 E Gumusgoz, DR Guisso, S Kasiri, et al. Targeting Gys1 with AAV-SaCas9 decreases pathogenic polyglucosan bodies and neuroinflammation in adult polyglucosan body and lafora disease mouse models. Neurotherapeutics. 2021;18(2):1414-1425.
26 SL Chen, QS Huang, YH Huang, et al. GYS1 induces glycogen accumulation and promotes tumor progression via the NF-kappaB pathway in clear cell renal carcinoma. Theranostics. 2020;10(20):9186-9199.
27 H Bhanot, MM Reddy, A Nonami, et al. Pathological glycogenesis through glycogen synthase 1 and suppression of excessive AMP kinase activity in myeloid leukemia cells. Leukemia. 2015;29(7):1555-1563.
28 H Xie, J Song, J Godfrey, et al. Glycogen metabolism is dispensable for tumour progression in clear cell renal cell carcinoma. Nat Metab. 2021;3(3):327-336.
29 LN Johnson. Glycogen phosphorylase: control by phosphorylation and allosteric effectors. FASEB J. 1992;6(6):2274-2282.
30 J Xie, J Zhang, W Tian, et al. The pan-cancer multi-omics landscape of FOXO family relevant to clinical outcome and drug resistance. Int J Mol Sci. 2022;23(24):15647.
31 S Saghafinia, M Mina, N Riggi, D Hanahan, G Ciriello. Pan-cancer landscape of aberrant DNA methylation across human tumors. Cell Rep. 2018;25(4):1066-1080.e1068.
32 DD Lee, M Komosa, NM Nunes, U Tabori. DNA methylation of the TERT promoter and its impact on human cancer. Curr Opin Genet Dev. 2020;60:17-24.
33 K Yoshihara, M Shahmoradgoli, E Martinez, et al. Inferring tumour purity and stromal and immune cell admixture from expression data. Nat Commun. 2013;4:2612.
34 Z Gu. Complex heatmap visualization. iMeta. 2022;1(3):e43.
35 DM Pardoll. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12(4):252-264.
36 J Xie, X Luo, X Deng, et al. Advances in artificial intelligence to predict cancer immunotherapy efficacy. Front Immunol. 2022;13:1076883.
37 A Ribas, JD Wolchok. Cancer immunotherapy using checkpoint blockade. Science. 2018;359(6382):1350-1355.
38 B Hwang, JH Lee, D Bang. Single-cell RNA sequencing technologies and bioinformatics pipelines. Exp Mol Med. 2018;50(8):1-14.
39 J Xie, W Deng, X Deng, et al. Single-cell histone chaperones patterns guide intercellular communication of tumor microenvironment that contribute to breast cancer metastases. Cancer Cell Int. 2023;23(1):311.
40 T Zheng, Q Liu, F Xing, C Zeng, W Wang. Disulfidptosis: a new form of programmed cell death. J Exp Clin Cancer Res. 2023;42(1):137.
41 P Zheng, C Zhou, Y Ding, S Duan. Disulfidptosis: a new target for metabolic cancer therapy. J Exp Clin Cancer Res. 2023;42(1):103.
42 MH Bailey, C Tokheim, E Porta-Pardo, et al. Comprehensive characterization of cancer driver genes and mutations. Cell. 2018;173(2):371-385.e318.
43 Y Teng, H Qin, A Bahassan, NG Bendzunas, EJ Kennedy, JK Cowell. The WASF3-NCKAP1-CYFIP1 complex is essential for breast cancer metastasis. Cancer Res. 2016;76(17):5133-5142.
44 F Xu, JB Addis, JM Cameron, BH Robinson. LRPPRC mutation suppresses cytochrome oxidase activity by altering mitochondrial RNA transcript stability in a mouse model. Biochem J. 2012;441(1):275-283.
45 M Olahova, SA Hardy, J Hall, et al. LRPPRC mutations cause early-onset multisystem mitochondrial disease outside of the French-Canadian population. Brain. 2015;138(12):3503-3519.
46 EC de Heer, CE Zois, E Bridges, et al. Glycogen synthase 1 targeting reveals a metabolic vulnerability in triple-negative breast cancer. J Exp Clin Cancer Res. 2023;42(1):143.
47 M Rousset, A Zweibaum, J Fogh. Presence of glycogen and growth-related variations in 58 cultured human tumor cell lines of various tissue origins. Cancer Res. 1981;41(3):1165-1170.
48 Z Gu, L Gu, R Eils, M Schlesner, B Brors. circlize Implements and enhances circular visualization in R. Bioinformatics. 2014;30(19):2811-2812.
49 DS Chen, I Mellman. Oncology meets immunology: the cancer-immunity cycle. Immunity. 2013;39(1):1-10.
50 V Thorsson, DL Gibbs, SD Brown, et al. The immune landscape of cancer. Immunity. 2018;48(4):812-830.e814.
51 D Zeng, Z Ye, R Shen, et al. IOBR: multi-omics immuno-oncology biological research to decode tumor microenvironment and signatures. Front Immunol. 2021;12:687975.
52 I Caffa, V D'Agostino, P Damonte, et al. Fasting potentiates the anticancer activity of tyrosine kinase inhibitors by strengthening MAPK signaling inhibition. Oncotarget. 2015;6(14):11820-11832.
53 I Caffa, V Spagnolo, C Vernieri, et al. Fasting-mimicking diet and hormone therapy induce breast cancer regression. Nature. 2020;583(7817):620-624.
54 M Galhuber, H Michenthaler, C Heininger, et al. Complementary omics strategies to dissect p53 signaling networks under nutrient stress. Cell Mol Life Sci. 2022;79(6):326.
55 X Zhang, L Du, Y Qiao, et al. Ferroptosis is governed by differential regulation of transcription in liver cancer. Redox Biol. 2019;24:101211.
56 S Song, T Christova, S Perusini, et al. Wnt inhibitor screen reveals iron dependence of beta-catenin signaling in cancers. Cancer Res. 2011;71(24):7628-7639.
57 K Sung, A Kurowski, C Lansiquot, et al. Selective inhibitors of autophagy reveal new link between the cell cycle and autophagy and lead to discovery of novel synergistic drug combinations. ACS Chem Biol. 2022;17(12):3290-3297.
58 PY Tsai, MS Lee, U Jadhav, et al. Adaptation of pancreatic cancer cells to nutrient deprivation is reversible and requires glutamine synthetase stabilization by mTORC1. Proc Natl Acad Sci U S A. 2021;118(10).
59 DA Braun, Y Hou, Z Bakouny, et al. Interplay of somatic alterations and immune infiltration modulates response to PD-1 blockade in advanced clear cell renal cell carcinoma. Nat Med. 2020;26(6):909-918.
60 S Mariathasan, SJ Turley, D Nickles, et al. TGFbeta attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature. 2018;554(7693):544-548.
61 N Riaz, JJ Havel, V Makarov, et al. Tumor and microenvironment evolution during immunotherapy with nivolumab. Cell. 2017;171(4):934-949.e916.
62 W Hugo, JM Zaretsky, L Sun, et al. Genomic and transcriptomic features of response to anti-PD-1 therapy in metastatic melanoma. Cell. 2016;165(1):35-44.
63 P Jezequel, D Loussouarn, C Guerin-Charbonnel, et al. Gene-expression molecular subtyping of triple-negative breast cancer tumours: importance of immune response. Breast Cancer Res. 2015;17:43.
64 C Brueffer, J Vallon-Christersson, D Grabau, et al. Clinical value of RNA sequencing-based classifiers for prediction of the five conventional breast cancer biomarkers: a report from the population-based multicenter Sweden Cancerome Analysis Network-Breast Initiative. JCO Precis Oncol. 2018;2.
65 R Sabatier, P Finetti, N Cervera, et al. A gene expression signature identifies two prognostic subgroups of basal breast cancer. Breast Cancer Res Treat. 2011;126(2):407-420.
66 YR Liu, YZ Jiang, XE Xu, X Hu, KD Yu, ZM Shao. Comprehensive transcriptome profiling reveals multigene signatures in triple-negative breast cancer. Clin Cancer Res. 2016;22(7):1653-1662.
67 ME Ritchie, B Phipson, D Wu, et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015;43(7):e47.
68 Q Song, GA Hawkins, L Wudel, et al. Dissecting intratumoral myeloid cell plasticity by single cell RNA-seq. Cancer Med. 2019;8(6):3072-3085.
69 X Zhang, L Peng, Y Luo, et al. Dissecting esophageal squamous-cell carcinoma ecosystem by single-cell transcriptomic analysis. Nat Commun. 2021;12(1):5291.
70 J Qian, S Olbrecht, B Boeckx, et al. A pan-cancer blueprint of the heterogeneous tumor microenvironment revealed by single-cell profiling. Cell Res. 2020;30(9):745-762.
71 SZ Wu, G Al-Eryani, DL Roden, et al. A single-cell and spatially resolved atlas of human breast cancers. Nat Genet. 2021;53(9):1334-1347.
72 Y Zou, F Ye, Y Kong, et al. The single-cell landscape of intratumoral heterogeneity and the immunosuppressive microenvironment in liver and brain metastases of breast cancer. Adv Sci (Weinh). 2023;10(5):e2203699.
73 TQ Gong, YZ Jiang, C Shao, et al. Proteome-centric cross-omics characterization and integrated network analyses of triple-negative breast cancer. Cell Rep. 2022;38(9):110460.
74 S Hanzelmann, R Castelo, J Guinney. GSVA: gene set variation analysis for microarray and RNA-seq data. BMC Bioinformatics. 2013;14:7.
75 A Butler, P Hoffman, P Smibert, E Papalexi, R Satija. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat Biotechnol. 2018;36(5):411-420.
76 S Jin, CF Guerrero-Juarez, L Zhang, et al. Inference and analysis of cell-cell communication using CellChat. Nat Commun. 2021;12(1):1088.
PDF

Accesses

Citations

Detail

Sections
Recommended

/