Endocrinol Metab.  2021 Dec;36(6):1287-1297. 10.3803/EnM.2021.1217.

Clinical and Molecular Characteristics of PRKACA L206R Mutant Cortisol-Producing Adenomas in Korean Patients

Affiliations
  • 1Translational Research Institute, Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
  • 2Department of Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
  • 3Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
  • 4Division of Surgery, Thyroid Center, Seoul National University Cancer Hospital, Seoul, Korea
  • 5Department of Surgery, Chung-Ang University Hospital, Seoul, Korea
  • 6Department of Laboratory Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
  • 7Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea

Abstract

Background
An activating mutation (c.617A>C/p.Lys206Arg, L206R) in protein kinase cAMP-activated catalytic subunit alpha (PRKACA) has been reported in 35% to 65% of cases of cortisol-producing adenomas (CPAs). We aimed to compare the clinical characteristics and transcriptome analysis between PRKACA L206R mutants and wild-type CPAs in Korea.
Methods
We included 57 subjects with CPAs who underwent adrenalectomy at Seoul National University Hospital. Sanger sequencing for PRKACA was conducted in 57 CPA tumor tissues. RNA sequencing was performed in 13 fresh-frozen tumor tissues.
Results
The prevalence of the PRKACA L206R mutation was 51% (29/57). The mean age of the study subjects was 42±12 years, and 87.7% (50/57) of the patients were female. Subjects with PRKACA L206R mutant CPAs showed smaller adenoma size (3.3±0.7 cm vs. 3.8±1.2 cm, P=0.059) and lower dehydroepiandrosterone sulfate levels (218±180 ng/mL vs. 1,511±3,307 ng/mL, P=0.001) than those with PRKACA wild-type CPAs. Transcriptome profiling identified 244 differentially expressed genes (DEGs) between PRKACA L206R mutant (n=8) and wild-type CPAs (n=5), including five upregulated and 239 downregulated genes in PRKACA L206R mutant CPAs (|fold change| ≥2, P<0.05). Among the upstream regulators of DEGs, CTNNB1 was the most significant transcription regulator. In several pathway analyses, the Wnt signaling pathway was downregulated and the steroid biosynthesis pathway was upregulated in PRKACA mutants. Protein-protein interaction analysis also showed that PRKACA downregulates Wnt signaling and upregulates steroid biosynthesis.
Conclusion
The PRKACA L206R mutation in CPAs causes high hormonal activity with a limited proliferative capacity, as supported by transcriptome profiling.

Keyword

Adrenocortical adenomas; Cushing syndrome; Gene expression profiling; PRKACA mutation; Wnt signaling pathway

Figure

  • Fig. 1 Analysis of differentially expressed genes (DEGs) between protein kinase cAMP-activated catalytic subunit alpha (PRKACA) L206R mutants and wild-type cortisol-producing adenomas (CPAs). (A) Volcano plot of DEGs, showing the magnitude (x-axis) and the statistical significance (y-axis) of differences in gene expression between PRKACA L206R mutants and wild-type CPAs. (B) A t-distributed stochastic neighbor embedding (tSNE) plot. FC, fold change.

  • Fig. 2 Catenin beta 1 (CTNNB1) as an upstream regulator of downregulated genes such as bone morphogenetic protein 4 (BMP4), cyclin D1 (CCND1), lymphoid enhancer binding factor 1 (LEF1), plastin 3 (PLS3), patched 1 (PTCH1), transducer of ERBB2, 2 (TOB2), TSC22 domain family member 1 (TSC22D1), UDP-glucose ceramide glucosyltransferase (UGCG), and versican (VCAN) in protein kinase cAMP-activated catalytic subunit alpha (PRKACA) L206R mutants. Green, downregulation in PRKACA L206R mutants.

  • Fig. 3 Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis of Cushing syndrome (hsa04934). Red, upregulation in protein kinase cAMP-activated catalytic subunit alpha (PRKACA) L206R mutants; green, downregulation in PRKACA L206R mutants. Steroid hormone biosynthesis, including STAR, CYP11A1, CYP17A1, HSD3B2, and CYP21A, was upregulated, but Wnt signaling-related genes, including Wnt, Frizzled, DVL, glycogen synthase kinase 3 beta (GSK3B), Axin, and TCF/LEF, were downregulated.


Reference

1. Lacroix A, Feelders RA, Stratakis CA, Nieman LK. Cushing’s syndrome. Lancet. 2015; 386:913–27.
Article
2. Lodish M, Stratakis CA. A genetic and molecular update on adrenocortical causes of Cushing syndrome. Nat Rev Endocrinol. 2016; 12:255–62.
Article
3. Kirschner LS, Carney JA, Pack SD, Taymans SE, Giatzakis C, Cho YS, et al. Mutations of the gene encoding the protein kinase A type I-alpha regulatory subunit in patients with the Carney complex. Nat Genet. 2000; 26:89–92.
Article
4. Libe R, Horvath A, Vezzosi D, Fratticci A, Coste J, Perlemoine K, et al. Frequent phosphodiesterase 11A gene (PDE11A) defects in patients with Carney complex (CNC) caused by PRKAR1A mutations: PDE11A may contribute to adrenal and testicular tumors in CNC as a modifier of the phenotype. J Clin Endocrinol Metab. 2011; 96:E208–14.
5. Weinstein LS, Shenker A, Gejman PV, Merino MJ, Friedman E, Spiegel AM. Activating mutations of the stimulatory G protein in the McCune-Albright syndrome. N Engl J Med. 1991; 325:1688–95.
Article
6. Beuschlein F, Fassnacht M, Assie G, Calebiro D, Stratakis CA, Osswald A, et al. Constitutive activation of PKA catalytic subunit in adrenal Cushing’s syndrome. N Engl J Med. 2014; 370:1019–28.
Article
7. Sato Y, Maekawa S, Ishii R, Sanada M, Morikawa T, Shiraishi Y, et al. Recurrent somatic mutations underlie corticotropin-independent Cushing’s syndrome. Science. 2014; 344:917–20.
Article
8. Cao Y, He M, Gao Z, Peng Y, Li Y, Li L, et al. Activating hotspot L205R mutation in PRKACA and adrenal Cushing’s syndrome. Science. 2014; 344:913–7.
Article
9. Goh G, Scholl UI, Healy JM, Choi M, Prasad ML, Nelson-Williams C, et al. Recurrent activating mutation in PRKACA in cortisol-producing adrenal tumors. Nat Genet. 2014; 46:613–7.
Article
10. Dalmazi GD, Beuschlein F. PRKACA mutations in adrenal adenomas: genotype/phenotype correlations. Horm Metab Res. 2017; 49:301–6.
Article
11. Calebiro D, Hannawacker A, Lyga S, Bathon K, Zabel U, Ronchi C, et al. PKA catalytic subunit mutations in adrenocortical Cushing’s adenoma impair association with the regulatory subunit. Nat Commun. 2014; 5:5680.
Article
12. Thiel A, Reis AC, Haase M, Goh G, Schott M, Willenberg HS, et al. PRKACA mutations in cortisol-producing adenomas and adrenal hyperplasia: a single-center study of 60 cases. Eur J Endocrinol. 2015; 172:677–85.
Article
13. Zennaro MC, Boulkroun S, Fernandes-Rosa F. Genetic causes of functional adrenocortical adenomas. Endocr Rev. 2017; 38:516–37.
Article
14. Nakajima Y, Okamura T, Gohko T, Satoh T, Hashimoto K, Shibusawa N, et al. Somatic mutations of the catalytic subunit of cyclic AMP-dependent protein kinase (PRKACA) gene in Japanese patients with several adrenal adenomas secreting cortisol [Rapid Communication]. Endocr J. 2014; 61:825–32.
Article
15. Bolger AM, Lohse M, Usadel B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics. 2014; 30:2114–20.
Article
16. Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013; 29:15–21.
Article
17. Li B, Dewey CN. RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinformatics. 2011; 12:323.
Article
18. Kuleshov MV, Jones MR, Rouillard AD, Fernandez NF, Duan Q, Wang Z, et al. Enrichr: a comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res. 2016; 44:W90–7.
Article
19. Slenter DN, Kutmon M, Hanspers K, Riutta A, Windsor J, Nunes N, et al. WikiPathways: a multifaceted pathway database bridging metabolomics to other omics research. Nucleic Acids Res. 2018; 46:D661–7.
Article
20. Kramer A, Green J, Pollard J Jr, Tugendreich S. Causal analysis approaches in Ingenuity Pathway Analysis. Bioinformatics. 2014; 30:523–30.
Article
21. Hong AR, Kim JH, Hong ES, Kim IK, Park KS, Ahn CH, et al. Limited diagnostic utility of plasma adrenocorticotropic hormone for differentiation between adrenal Cushing syndrome and Cushing disease. Endocrinol Metab (Seoul). 2015; 30:297–304.
Article
22. Dennedy MC, Annamalai AK, Prankerd-Smith O, Freeman N, Vengopal K, Graggaber J, et al. Low DHEAS: a sensitive and specific test for the detection of subclinical hypercortisolism in adrenal incidentalomas. J Clin Endocrinol Metab. 2017; 102:786–92.
23. Sugawara T, Saito M, Fujimoto S. Sp1 and SF-1 interact and cooperate in the regulation of human steroidogenic acute regulatory protein gene expression. Endocrinology. 2000; 141:2895–903.
Article
24. Zhou W, Wu L, Xie J, Su T, Jiang L, Jiang Y, et al. Steroidogenic acute regulatory protein overexpression correlates with protein kinase A activation in adrenocortical adenoma. PLoS One. 2016; 11:e0162606.
Article
25. Di Dalmazi G, Kisker C, Calebiro D, Mannelli M, Canu L, Arnaldi G, et al. Novel somatic mutations in the catalytic subunit of the protein kinase A as a cause of adrenal Cushing’s syndrome: a European multicentric study. J Clin Endocrinol Metab. 2014; 99:E2093–100.
Article
26. Almeida MQ, Harran M, Bimpaki EI, Hsiao HP, Horvath A, Cheadle C, et al. Integrated genomic analysis of nodular tissue in macronodular adrenocortical hyperplasia: progression of tumorigenesis in a disorder associated with multiple benign lesions. J Clin Endocrinol Metab. 2011; 96:E728–38.
Article
27. Hino S, Tanji C, Nakayama KI, Kikuchi A. Phosphorylation of beta-catenin by cyclic AMP-dependent protein kinase stabilizes beta-catenin through inhibition of its ubiquitination. Mol Cell Biol. 2005; 25:9063–72.
Article
28. Taurin S, Sandbo N, Qin Y, Browning D, Dulin NO. Phosphorylation of beta-catenin by cyclic AMP-dependent protein kinase. J Biol Chem. 2006; 281:9971–6.
29. Almeida MQ, Muchow M, Boikos S, Bauer AJ, Griffin KJ, Tsang KM, et al. Mouse Prkar1a haploinsufficiency leads to an increase in tumors in the Trp53+/− or Rb1+/− backgrounds and chemically induced skin papillomas by dysregulation of the cell cycle and Wnt signaling. Hum Mol Genet. 2010; 19:1387–98.
Article
30. Gaujoux S, Tissier F, Groussin L, Libe R, Ragazzon B, Launay P, et al. Wnt/beta-catenin and 3′,5′-cyclic adenosine 5′-monophosphate/protein kinase A signaling pathways alterations and somatic beta-catenin gene mutations in the progression of adrenocortical tumors. J Clin Endocrinol Metab. 2008; 93:4135–40.
31. Drelon C, Berthon A, Sahut-Barnola I, Mathieu M, Dumontet T, Rodriguez S, et al. PKA inhibits WNT signalling in adrenal cortex zonation and prevents malignant tumour development. Nat Commun. 2016; 7:12751.
Article
Full Text Links
  • ENM
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr