Skip to main content
Erschienen in: Journal of Cancer Research and Clinical Oncology 8/2023

21.10.2022 | Research

LDHA is a prognostic biomarker on the immune response in pancreatic adenocarcinoma and associated with m6A modification

verfasst von: Zhen Tan, Jiang Liu, Jin Xu, Bo Zhang, Xianjun Yu, Wei Wang, Chen Liang

Erschienen in: Journal of Cancer Research and Clinical Oncology | Ausgabe 8/2023

Einloggen, um Zugang zu erhalten

Abstract

Purpose

N6-methyladenosine (m6A) is tightly associated with the progression of pancreatic ductal adenocarcinoma (PDAC). Another prominent feature of PDAC is metabolic reprogramming, which provides sufficient nutrients to support rapid cell growth via the tumor microenvironment. However, the underlying influences of m6A-associated metabolic genes on the PDAC microenvironment remain poorly understood. Therefore, we sought to construct a survival prediction model using m6A-related genes to clarify the molecular characteristics of PDAC.

Methods

In the present study, m6A-related metabolic genes were obtained from The Cancer Genome Atlas (TCGA) pancreatic adenocarcinoma dataset and subjected to coexpression analysis. Consensus clustering recognized two distinct subgroups with different immune cell infiltration patterns according to the expression of m6A-associated metabolic genes. Multivariate Cox regression analyses and least absolute shrinkage and selection operator (LASSO) analysis were adopted to create an m6A-related metabolism model. A nomogram including clinical features and the risk score based on the expression of m6A-related metabolism regulators was constructed.

Results

A four-gene signature comprising ATP8B2, GMPS, LDHA and SDR39U1 was built to predict the overall survival (OS) of PDAC patients. This signature also robustly predicted survival in two independent validation cohorts from the International Cancer Genome Consortium (ICGC) and ArrayExpress (E-MTAB-6134). The four-gene signature divided patients into high- and low-risk groups with distinct OS values as verified by the log-rank test. Among the four genes, LDHA was upregulated in both PDAC tissues and cell lines.

Conclusions

Collectively, we analyzed the immune microenvironment, predicted drug sensitivity and assessed the implications of the mutation landscape based on the crosstalk between m6A regulators and metabolic rewiring, and we also constructed a novel signature based on m6A-associated metabolic genes to predict PDAC prognosis.
Anhänge
Nur mit Berechtigung zugänglich
Literatur
Zurück zum Zitat Cheng CS, Tan HY, Wang N et al (2021) Functional inhibition of lactate dehydrogenase suppresses pancreatic adenocarcinoma progression. Clin Transl Med 11:e467CrossRefPubMedPubMedCentral Cheng CS, Tan HY, Wang N et al (2021) Functional inhibition of lactate dehydrogenase suppresses pancreatic adenocarcinoma progression. Clin Transl Med 11:e467CrossRefPubMedPubMedCentral
Zurück zum Zitat DeBerardinis RJ, Lum JJ, Hatzivassiliou G et al (2008) The biology of cancer: metabolic reprogramming fuels cell growth and proliferation. Cell Metab 7:11–20CrossRefPubMed DeBerardinis RJ, Lum JJ, Hatzivassiliou G et al (2008) The biology of cancer: metabolic reprogramming fuels cell growth and proliferation. Cell Metab 7:11–20CrossRefPubMed
Zurück zum Zitat Garcia-Canaveras JC, Chen L, Rabinowitz JD (2019) The tumor metabolic microenvironment: lessons from lactate. Can Res 79:3155–3162CrossRef Garcia-Canaveras JC, Chen L, Rabinowitz JD (2019) The tumor metabolic microenvironment: lessons from lactate. Can Res 79:3155–3162CrossRef
Zurück zum Zitat Gong Y, Ji P, Yang YS et al (2021) Metabolic-pathway-based subtyping of triple-negative breast cancer reveals potential therapeutic targets. Cell Metab 33(51–64):e59 Gong Y, Ji P, Yang YS et al (2021) Metabolic-pathway-based subtyping of triple-negative breast cancer reveals potential therapeutic targets. Cell Metab 33(51–64):e59
Zurück zum Zitat Hanzelmann S, Castelo R, Guinney J (2013) GSVA: gene set variation analysis for microarray and RNA-seq data. BMC Bioinform 14:7CrossRef Hanzelmann S, Castelo R, Guinney J (2013) GSVA: gene set variation analysis for microarray and RNA-seq data. BMC Bioinform 14:7CrossRef
Zurück zum Zitat Hosein AN, Beg MS (2018) Pancreatic cancer metabolism: molecular mechanisms and clinical applications. Curr Oncol Rep 20:56CrossRefPubMed Hosein AN, Beg MS (2018) Pancreatic cancer metabolism: molecular mechanisms and clinical applications. Curr Oncol Rep 20:56CrossRefPubMed
Zurück zum Zitat Huang H, Weng H, Chen J (2020) m(6)A modification in coding and non-coding RNAs: roles and therapeutic implications in cancer. Cancer Cell 37:270–288CrossRefPubMedPubMedCentral Huang H, Weng H, Chen J (2020) m(6)A modification in coding and non-coding RNAs: roles and therapeutic implications in cancer. Cancer Cell 37:270–288CrossRefPubMedPubMedCentral
Zurück zum Zitat Jiang F, Ma S, Xue Y et al (2016) LDH-A promotes malignant progression via activation of epithelial-to-mesenchymal transition and conferring stemness in muscle-invasive bladder cancer. Biochem Biophys Res Commun 469:985–992CrossRefPubMed Jiang F, Ma S, Xue Y et al (2016) LDH-A promotes malignant progression via activation of epithelial-to-mesenchymal transition and conferring stemness in muscle-invasive bladder cancer. Biochem Biophys Res Commun 469:985–992CrossRefPubMed
Zurück zum Zitat Lan Q, Liu PY, Haase J et al (2019) The critical role of RNA m(6)A methylation in cancer. Can Res 79:1285–1292CrossRef Lan Q, Liu PY, Haase J et al (2019) The critical role of RNA m(6)A methylation in cancer. Can Res 79:1285–1292CrossRef
Zurück zum Zitat Lan Q, Liu PY, Bell JL et al (2021) The emerging roles of RNA m(6)A methylation and demethylation as critical regulators of tumorigenesis, drug sensitivity, and resistance. Can Res 81:3431–3440CrossRef Lan Q, Liu PY, Bell JL et al (2021) The emerging roles of RNA m(6)A methylation and demethylation as critical regulators of tumorigenesis, drug sensitivity, and resistance. Can Res 81:3431–3440CrossRef
Zurück zum Zitat Liang C, Shi S, Qin Y et al (2020) Localisation of PGK1 determines metabolic phenotype to balance metastasis and proliferation in patients with SMAD4-negative pancreatic cancer. Gut 69:888–900CrossRefPubMed Liang C, Shi S, Qin Y et al (2020) Localisation of PGK1 determines metabolic phenotype to balance metastasis and proliferation in patients with SMAD4-negative pancreatic cancer. Gut 69:888–900CrossRefPubMed
Zurück zum Zitat Liu Y, Liang G, Xu H et al (2021) Tumors exploit FTO-mediated regulation of glycolytic metabolism to evade immune surveillance. Cell Metab 33(1221–1233):e1211 Liu Y, Liang G, Xu H et al (2021) Tumors exploit FTO-mediated regulation of glycolytic metabolism to evade immune surveillance. Cell Metab 33(1221–1233):e1211
Zurück zum Zitat Markert CL, Shaklee JB, Whitt GS (1975) Evolution of a gene. Multiple genes for LDH isozymes provide a model of the evolution of gene structure, function and regulation. Science 189:102–114CrossRefPubMed Markert CL, Shaklee JB, Whitt GS (1975) Evolution of a gene. Multiple genes for LDH isozymes provide a model of the evolution of gene structure, function and regulation. Science 189:102–114CrossRefPubMed
Zurück zum Zitat Meng Q, Shi S, Liang C et al (2018a) Abrogation of glutathione peroxidase-1 drives EMT and chemoresistance in pancreatic cancer by activating ROS-mediated Akt/GSK3beta/Snail signaling. Oncogene 37:5843–5857CrossRefPubMed Meng Q, Shi S, Liang C et al (2018a) Abrogation of glutathione peroxidase-1 drives EMT and chemoresistance in pancreatic cancer by activating ROS-mediated Akt/GSK3beta/Snail signaling. Oncogene 37:5843–5857CrossRefPubMed
Zurück zum Zitat Meng Q, Xu J, Liang C et al (2018b) GPx1 is involved in the induction of protective autophagy in pancreatic cancer cells in response to glucose deprivation. Cell Death Dis 9:1187CrossRefPubMedPubMedCentral Meng Q, Xu J, Liang C et al (2018b) GPx1 is involved in the induction of protective autophagy in pancreatic cancer cells in response to glucose deprivation. Cell Death Dis 9:1187CrossRefPubMedPubMedCentral
Zurück zum Zitat Nombela P, Miguel-Lopez B, Blanco S (2021) The role of m(6)A, m(5)C and Psi RNA modifications in cancer: novel therapeutic opportunities. Mol Cancer 20:18CrossRefPubMedPubMedCentral Nombela P, Miguel-Lopez B, Blanco S (2021) The role of m(6)A, m(5)C and Psi RNA modifications in cancer: novel therapeutic opportunities. Mol Cancer 20:18CrossRefPubMedPubMedCentral
Zurück zum Zitat Shi M, Cui J, Du J et al (2014) A novel KLF4/LDHA signaling pathway regulates aerobic glycolysis in and progression of pancreatic cancer. Clin Cancer Res 20:4370–4380CrossRefPubMedPubMedCentral Shi M, Cui J, Du J et al (2014) A novel KLF4/LDHA signaling pathway regulates aerobic glycolysis in and progression of pancreatic cancer. Clin Cancer Res 20:4370–4380CrossRefPubMedPubMedCentral
Zurück zum Zitat Son J, Lyssiotis CA, Ying H et al (2013) Glutamine supports pancreatic cancer growth through a KRAS-regulated metabolic pathway. Nature 496:101–105CrossRefPubMedPubMedCentral Son J, Lyssiotis CA, Ying H et al (2013) Glutamine supports pancreatic cancer growth through a KRAS-regulated metabolic pathway. Nature 496:101–105CrossRefPubMedPubMedCentral
Zurück zum Zitat Sun H, Zhou Y, Skaro MF et al (2020) Metabolic reprogramming in cancer is induced to increase proton production. Can Res 80:1143–1155CrossRef Sun H, Zhou Y, Skaro MF et al (2020) Metabolic reprogramming in cancer is induced to increase proton production. Can Res 80:1143–1155CrossRef
Zurück zum Zitat Taketo K, Konno M, Asai A et al (2018) The epitranscriptome m6A writer METTL3 promotes chemo- and radioresistance in pancreatic cancer cells. Int J Oncol 52:621–629PubMed Taketo K, Konno M, Asai A et al (2018) The epitranscriptome m6A writer METTL3 promotes chemo- and radioresistance in pancreatic cancer cells. Int J Oncol 52:621–629PubMed
Zurück zum Zitat Tang B, Yang Y, Kang M et al (2020) m(6)A demethylase ALKBH5 inhibits pancreatic cancer tumorigenesis by decreasing WIF-1 RNA methylation and mediating Wnt signaling. Mol Cancer 19:3CrossRefPubMedPubMedCentral Tang B, Yang Y, Kang M et al (2020) m(6)A demethylase ALKBH5 inhibits pancreatic cancer tumorigenesis by decreasing WIF-1 RNA methylation and mediating Wnt signaling. Mol Cancer 19:3CrossRefPubMedPubMedCentral
Zurück zum Zitat Uddin MB, Wang Z, Yang C (2021) The m(6)A RNA methylation regulates oncogenic signaling pathways driving cell malignant transformation and carcinogenesis. Mol Cancer 20:61CrossRefPubMedPubMedCentral Uddin MB, Wang Z, Yang C (2021) The m(6)A RNA methylation regulates oncogenic signaling pathways driving cell malignant transformation and carcinogenesis. Mol Cancer 20:61CrossRefPubMedPubMedCentral
Zurück zum Zitat Valvona CJ, Fillmore HL, Nunn PB et al (2016) The regulation and function of lactate dehydrogenase A: therapeutic potential in brain tumor. Brain Pathol 26:3–17CrossRefPubMed Valvona CJ, Fillmore HL, Nunn PB et al (2016) The regulation and function of lactate dehydrogenase A: therapeutic potential in brain tumor. Brain Pathol 26:3–17CrossRefPubMed
Zurück zum Zitat Xu F, Huang X, Li Y et al (2021) m(6)A-related lncRNAs are potential biomarkers for predicting prognoses and immune responses in patients with LUAD. Mol Ther Nucleic Acids 24:780–791CrossRefPubMedPubMedCentral Xu F, Huang X, Li Y et al (2021) m(6)A-related lncRNAs are potential biomarkers for predicting prognoses and immune responses in patients with LUAD. Mol Ther Nucleic Acids 24:780–791CrossRefPubMedPubMedCentral
Zurück zum Zitat Zdralevic M, Brand A, Di Ianni L et al (2018) Double genetic disruption of lactate dehydrogenases A and B is required to ablate the “Warburg effect” restricting tumor growth to oxidative metabolism. J Biol Chem 293:15947–15961CrossRefPubMedPubMedCentral Zdralevic M, Brand A, Di Ianni L et al (2018) Double genetic disruption of lactate dehydrogenases A and B is required to ablate the “Warburg effect” restricting tumor growth to oxidative metabolism. J Biol Chem 293:15947–15961CrossRefPubMedPubMedCentral
Metadaten
Titel
LDHA is a prognostic biomarker on the immune response in pancreatic adenocarcinoma and associated with m6A modification
verfasst von
Zhen Tan
Jiang Liu
Jin Xu
Bo Zhang
Xianjun Yu
Wei Wang
Chen Liang
Publikationsdatum
21.10.2022
Verlag
Springer Berlin Heidelberg
Erschienen in
Journal of Cancer Research and Clinical Oncology / Ausgabe 8/2023
Print ISSN: 0171-5216
Elektronische ISSN: 1432-1335
DOI
https://doi.org/10.1007/s00432-022-04400-8

Weitere Artikel der Ausgabe 8/2023

Journal of Cancer Research and Clinical Oncology 8/2023 Zur Ausgabe

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Erhöhtes Risiko fürs Herz unter Checkpointhemmer-Therapie

28.05.2024 Nebenwirkungen der Krebstherapie Nachrichten

Kardiotoxische Nebenwirkungen einer Therapie mit Immuncheckpointhemmern mögen selten sein – wenn sie aber auftreten, wird es für Patienten oft lebensgefährlich. Voruntersuchung und Monitoring sind daher obligat.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.