open access

Vol 74, No 2 (2023)
Original paper
Submitted: 2022-09-16
Accepted: 2022-11-22
Published online: 2023-03-10
Get Citation

Feasibility analysis of ACTH adenoma model in USP8–/– mice

Jia Li1, Na Wu1, Dimin Zhu2, Yonghong Zhu1
·
Pubmed: 36916541
·
Endokrynol Pol 2023;74(2):181-189.
Affiliations
  1. Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
  2. Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China

open access

Vol 74, No 2 (2023)
Original Paper
Submitted: 2022-09-16
Accepted: 2022-11-22
Published online: 2023-03-10

Abstract

Introduction: Patients with adrenocorticotropic hormone (ACTH)-secreting pituitary tumours (35% to 60%) present with somatic mutations in the USP8 gene. USP8 mutations lead to enhanced deubiquitination of the epidermal growth factor receptor (EGFR) and result in an imbalance in EGFR signalling, accompanied by excessive activation of ACTH production and cell growth. USP8 emerged as a novel and exciting candidate gene for Cushing’s disease.

Material and methods: In this study, USP8 mutant mouse models (USP8+/– and USP8–/–) were established, their phenotypes were analysed and identified, biochemical indexes were detected, pituitary and adrenal tissue specimens were taken for HE staining and immunohistochemical identification of hormones, and the differences between the 2 groups of mutant mice and wild type mice were analysed and compared.

Results: Compared with the control group (wild type), immunofluorescence assay results for USP8+/– mice and USP8–/– mice showed increased pituitary ACTH expression, which was statistically different (p < 0.05), and there were no significant differences in body weight, plasma ACTH, 24-hour urinary free cortisol, and immunohistochemical results. Higher blood glucose in USP8–/– mice than in USP8+/+
mice was observed. The heart rates of USP8–/– mice were higher than those of USP8+/– mice and USP8+/+ mice. HE staining and tissue fibre staining were done, and no significant pathological changes were seen in the 3 groups of pituitary and adrenal tissues.

Conclusion: USP8 knockout mice have the potential to form an animal model of ACTH adenoma.

Abstract

Introduction: Patients with adrenocorticotropic hormone (ACTH)-secreting pituitary tumours (35% to 60%) present with somatic mutations in the USP8 gene. USP8 mutations lead to enhanced deubiquitination of the epidermal growth factor receptor (EGFR) and result in an imbalance in EGFR signalling, accompanied by excessive activation of ACTH production and cell growth. USP8 emerged as a novel and exciting candidate gene for Cushing’s disease.

Material and methods: In this study, USP8 mutant mouse models (USP8+/– and USP8–/–) were established, their phenotypes were analysed and identified, biochemical indexes were detected, pituitary and adrenal tissue specimens were taken for HE staining and immunohistochemical identification of hormones, and the differences between the 2 groups of mutant mice and wild type mice were analysed and compared.

Results: Compared with the control group (wild type), immunofluorescence assay results for USP8+/– mice and USP8–/– mice showed increased pituitary ACTH expression, which was statistically different (p < 0.05), and there were no significant differences in body weight, plasma ACTH, 24-hour urinary free cortisol, and immunohistochemical results. Higher blood glucose in USP8–/– mice than in USP8+/+
mice was observed. The heart rates of USP8–/– mice were higher than those of USP8+/– mice and USP8+/+ mice. HE staining and tissue fibre staining were done, and no significant pathological changes were seen in the 3 groups of pituitary and adrenal tissues.

Conclusion: USP8 knockout mice have the potential to form an animal model of ACTH adenoma.

Get Citation

Keywords

USP8; Cushing’s disease; animal model; ACTH adenoma

About this article
Title

Feasibility analysis of ACTH adenoma model in USP8–/– mice

Journal

Endokrynologia Polska

Issue

Vol 74, No 2 (2023)

Article type

Original paper

Pages

181-189

Published online

2023-03-10

Page views

2223

Article views/downloads

417

DOI

10.5603/EP.a2023.0006

Pubmed

36916541

Bibliographic record

Endokrynol Pol 2023;74(2):181-189.

Keywords

USP8
Cushing’s disease
animal model
ACTH adenoma

Authors

Jia Li
Na Wu
Dimin Zhu
Yonghong Zhu

References (52)
  1. Newell-Price J, Bertagna X, Grossman AB, et al. Cushing's syndrome. Lancet. 2006; 367(9522): 1605–1617.
  2. Pivonello R, De Martino MC, De Leo M, et al. Cushing's Syndrome. Endocrinol Metab Clin North Am. 2008; 37(1): 135–49, ix.
  3. Clayton RN, Raskauskiene D, Reulen RC, et al. Mortality and morbidity in Cushing's disease over 50 years in Stoke-on-Trent, UK: audit and meta-analysis of literature. J Clin Endocrinol Metab. 2011; 96(3): 632–642.
  4. van Haalen FM, Broersen LHA, Jorgensen JO, et al. Management of endocrine disease: Mortality remains increased in Cushing's disease despite biochemical remission: a systematic review and meta-analysis. Eur J Endocrinol. 2015; 172(4): R143–R149.
  5. Farshi P, Deshmukh RR, Nwankwo JO, et al. Deubiquitinases (DUBs) and DUB inhibitors: a patent review. Expert Opin Ther Pat. 2015; 25(10): 1191–1208.
  6. Reyes-Turcu FE, Ventii KH, Wilkinson KD. Regulation and cellular roles of ubiquitin-specific deubiquitinating enzymes. Annu Rev Biochem. 2009; 78: 363–397.
  7. Reincke M, Sbiera S, Hayakawa A, et al. Mutations in the deubiquitinase gene USP8 cause Cushing's disease. Nat Genet. 2015; 47(1): 31–38.
  8. Weigand I, Knobloch L, Flitsch J, et al. Impact of USP8 Gene Mutations on Protein Deregulation in Cushing Disease. J Clin Endocrinol Metab. 2019; 104(7): 2535–2546.
  9. Nieman LK. Cushing's syndrome: update on signs, symptoms and biochemical screening. Eur J Endocrinol. 2015; 173(4): M33–M38.
  10. Sbiera S, Kunz M, Weigand I, et al. The New Genetic Landscape of Cushing's Disease: Deubiquitinases in the Spotlight. Cancers (Basel). 2019; 11(11).
  11. Rämet M, Manfruelli P, Pearson A, et al. Functional genomic analysis of phagocytosis and identification of a Drosophila receptor for E. coli. Nature. 2002; 416(6881): 644–648.
  12. Li M, Brooks CL, Kon N, et al. A dynamic role of HAUSP in the p53-Mdm2 pathway. Mol Cell. 2004; 13(6): 879–886.
  13. Sesta A, Cassarino MF, Terreni M, et al. Ubiquitin-Specific Protease 8 Mutant Corticotrope Adenomas Present Unique Secretory and Molecular Features and Shed Light on the Role of Ubiquitylation on ACTH Processing. Neuroendocrinology. 2020; 110(1-2): 119–129.
  14. Treppiedi D, Barbieri AM, Di Muro G, et al. Genetic Profiling of a Cohort of Italian Patients with ACTH-Secreting Pituitary Tumors and Characterization of a Novel Gene Variant. Cancers (Basel). 2021; 13(16).
  15. Ma ZY, Song ZJ, Chen JH, et al. Recurrent gain-of-function USP8 mutations in Cushing's disease. Cell Res. 2015; 25(3): 306–317.
  16. Fukuoka H, Cooper O, Ben-Shlomo A, et al. EGFR as a therapeutic target for human, canine, and mouse ACTH-secreting pituitary adenomas. J Clin Invest. 2011; 121(12): 4712–4721.
  17. Jian FF, Li YF, Chen YF, et al. Inhibition of Ubiquitin-specific Peptidase 8 Suppresses Adrenocorticotropic Hormone Production and Tumorous Corticotroph Cell Growth in AtT20 Cells. Chin Med J (Engl). 2016; 129(17): 2102–2108.
  18. Kageyama K, Asari Y, Sugimoto Y, et al. Ubiquitin-specific protease 8 inhibitor suppresses adrenocorticotropic hormone production and corticotroph tumor cell proliferation. Endocr J. 2020; 67(2): 177–184.
  19. Asari Y, Kageyama K, Sugiyama A, et al. Lapatinib decreases the ACTH production and proliferation of corticotroph tumor cells. Endocr J. 2019; 66(6): 515–522.
  20. Treppiedi D, Di Muro G, Marra G, et al. USP8 inhibitor RA-9 reduces ACTH release and cell growth in tumor corticotrophs. Endocr Relat Cancer. 2021; 28(8): 573–582.
  21. Tascou S, Trappe R, Nayernia K, et al. TSPY-LTA transgenic mice develop endocrine tumors of the pituitary and adrenal gland. Mol Cell Endocrinol. 2003; 200(1-2): 9–18.
  22. Helseth A, Haug E, Nesland JM, et al. Endocrine and metabolic characteristics of polyoma large T transgenic mice that develop ACTH-producing pituitary tumors. J Neurosurg. 1995; 82(5): 879–885.
  23. Helseth A, Siegal GP, Haug E, et al. Transgenic mice that develop pituitary tumors. A model for Cushing's disease. Am J Pathol. 1992; 140(5): 1071–1080.
  24. Stenzel-Poore MP, Cameron VA, Vaughan J, et al. Development of Cushing's syndrome in corticotropin-releasing factor transgenic mice. Endocrinology. 1992; 130(6): 3378–3386.
  25. Bentley L, Esapa CT, Nesbit MA, et al. An N-ethyl-N-nitrosourea induced corticotropin-releasing hormone promoter mutation provides a mouse model for endogenous glucocorticoid excess. Endocrinology. 2014; 155(3): 908–922.
  26. Liu NA, Jiang H, Ben-Shlomo A, et al. Targeting zebrafish and murine pituitary corticotroph tumors with a cyclin-dependent kinase (CDK) inhibitor. Proc Natl Acad Sci U S A. 2011; 108(20): 8414–8419.
  27. Pivonello R, De Martino MC, De Leo M, et al. Cushing's syndrome: aftermath of the cure. Arq Bras Endocrinol Metabol. 2007; 51(8): 1381–1391.
  28. Pivonello R, Faggiano A, Lombardi G, et al. The metabolic syndrome and cardiovascular risk in Cushing's syndrome. Endocrinol Metab Clin North Am. 2005; 34(2): 327–39, viii.
  29. Bertoia ML, Waring ME, Gupta PS, et al. Implications of new hypertension guidelines in the United States. Hypertension. 2012; 60(3): 639–644.
  30. Bi W, Lan X, Zhang J, et al. USP8 ameliorates cognitive and motor impairments via microglial inhibition in a mouse model of sepsis-associated encephalopathy. Brain Res. 2019; 1719: 40–48.
  31. Baykara M, Yaman M, Buyukberber S. Clinical and prognostic importance of XIAP and USP8 in advanced stages of non-small cell lung cancer. J Buon. 2013; 18(4): 921–927.
  32. Pomp D, Nehrenberg D, Estrada-Smith D. Complex genetics of obesity in mouse models. Annu Rev Nutr. 2008; 28: 331–345.
  33. Pearson G, Chai B, Vozheiko T, et al. Clec16a, Nrdp1, and USP8 Form a Ubiquitin-Dependent Tripartite Complex That Regulates β-Cell Mitophagy. Diabetes. 2018; 67(2): 265–277.
  34. Levine HJ. Rest heart rate and life expectancy. Editorial. J Am Coll Cardiol. 1997; 30(4): 1104–1106.
  35. Stessman J, Jacobs JM, Stessman-Lande I, et al. Aging, resting pulse rate, and longevity. J Am Geriatr Soc. 2013; 61(1): 40–45.
  36. Gent S, Kleinbongard P, Dammann P, et al. Heart rate reduction and longevity in mice. Basic Res Cardiol. 2015; 110(2): 2.
  37. Böhm M, Swedberg K, Komajda M, et al. Heart rate as a risk factor in chronic heart failure (SHIFT): the association between heart rate and outcomes in a randomised placebo-controlled trial. Lancet. 2010; 376(9744): 886–894.
  38. Cooney MT, Vartiainen E, Laatikainen T, et al. Elevated resting heart rate is an independent risk factor for cardiovascular disease in healthy men and women. Am Heart J. 2010; 159(4): 612–619.e3.
  39. Fox K, Borer JS, Camm AJ, et al. Heart Rate Working Group. Resting heart rate in cardiovascular disease. J Am Coll Cardiol. 2007; 50(9): 823–830.
  40. Nanchen D, Leening MJG, Locatelli I, et al. Resting heart rate and the risk of heart failure in healthy adults: the Rotterdam Study. Circ Heart Fail. 2013; 6(3): 403–410.
  41. Sugiyama F, Yagami K, Paigen B. Mouse models of blood pressure regulation and hypertension. Curr Hypertens Rep. 2001; 3(1): 41–48.
  42. Kovacs K, Horvath E, Bayley T, et al. Silent corticotroph cell adenoma with lysosomal accumulation and crinophagy. Am J Med. 1978; 64(3): 492–499.
  43. García-Martínez A, Fuentes-Fayos AC, Fajardo C, et al. Differential Expression of MicroRNAs in Silent and Functioning Corticotroph Tumors. J Clin Med. 2020; 9(6).
  44. Tani Y, Sugiyama T, Izumiyama H, et al. Differential gene expression profiles of POMC-related enzymes, transcription factors and receptors between non-pituitary and pituitary ACTH-secreting tumors. Endocr J. 2011; 58(4): 297–303.
  45. Jahangiri A, Wagner JR, Pekmezci M, et al. A comprehensive long-term retrospective analysis of silent corticotrophic adenomas vs hormone-negative adenomas. Neurosurgery. 2013; 73(1): 8–17; discussion 17.
  46. García-Martínez A, Cano DA, Flores-Martínez A, et al. Why don't corticotroph tumors always produce Cushing's disease? Eur J Endocrinol. 2019; 181(3): 351–361.
  47. Tateno T, Izumiyama H, Doi M, et al. Differential gene expression in ACTH -secreting and non-functioning pituitary tumors. Eur J Endocrinol. 2007; 157(6): 717–724.
  48. Neou M, Villa C, Armignacco R, et al. Pangenomic Classification of Pituitary Neuroendocrine Tumors. Cancer Cell. 2020; 37(1): 123–134.e5.
  49. Ohta S, Nishizawa S, Oki Y, et al. Significance ofAbsent Prohormone Convertase 1/3 in Inducing Clinically Silent Corticotroph Pituitary Adenoma ofSubtype I — Immunohistochemical Study. Pituitary. 2002; 5(4): 221–223.
  50. Chen J, Jian X, Deng S, et al. Identification of recurrent USP48 and BRAF mutations in Cushing's disease. Nat Commun. 2018; 9(1): 3171.
  51. Sbiera S, Perez-Rivas LG, Taranets L, et al. Driver mutations in USP8 wild-type Cushing's disease. Neuro Oncol. 2019; 21(10): 1273–1283.
  52. Srirangam Nadhamuni V, Korbonits M. Novel Insights into Pituitary Tumorigenesis: Genetic and Epigenetic Mechanisms. Endocr Rev. 2020; 41(6): 821–846.

Regulations

Important: This website uses cookies. More >>

The cookies allow us to identify your computer and find out details about your last visit. They remembering whether you've visited the site before, so that you remain logged in - or to help us work out how many new website visitors we get each month. Most internet browsers accept cookies automatically, but you can change the settings of your browser to erase cookies or prevent automatic acceptance if you prefer.

Via MedicaWydawcą jest  VM Media Group sp. z o.o., Grupa Via Medica, ul. Świętokrzyska 73, 80–180 Gdańsk

tel.:+48 58 320 94 94, faks:+48 58 320 94 60, e-mail:  viamedica@viamedica.pl