info@biomedres.us   +1 (502) 904-2126   One Westbrook Corporate Center, Suite 300, Westchester, IL 60154, USA   Site Map
ISSN: 2574 -1241

Impact Factor : 0.548

  Submit Manuscript

Mini ReviewOpen Access

Non-Coding RNAS and Steroidogenesis Volume 10- Issue 2

Kenji Ohe*, Yoshihiro Harada, Hiroyoshi Harada, Hiroki Terai, Masayoshi Mori, Yusuke Murata and Munechika Enjoji

  • Department of Pharmacotherapeutics, Fukuoka University, Japan

Received: October 06, 2018;   Published: October 17, 2018

*Corresponding author: Kenji Ohe, Department of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan

DOI: 10.26717/BJSTR.2018.10.001910

Abstract PDF

Also view in:

Abstract

Non-coding RNAs have retained a prominent role in regulating gene expression. They are RNA species that are not transcribed but functional participate in almost every aspect of cellular function. One of the triggers of their discovery was the human genome project. The number of genes found in the human genome fluctuated during the analyses due to the abundant non-coding RNAs that were difficult to be judged as a gene or not. These non-coding RNAs are consisted of: microRNAs (mi-RNAs) that were found as RNAs transcribed form ultra-conserved regions; and other evolutionary conserved ones such as circular RNAs (circ-RNAs) and long non-coding RNAs (lnc-RNAs), as well as PIWI-interacting RNAs, small nucleolar RNAs, transcribed ultra conserved regions, and large intergenic non-coding RNAs. The amount of these functional non-coding RNAs have been increasing in a nearly exponential manner since their discoveries and have been listed up to more than half of the transcribed RNAs in the human cell. This review will focus on non-coding RNAs related with genes important for the function of the adrenal cortex.

Introduction

Adrenal Cortex and Non-Coding RNAs

In the human adrenal cortex, many intermediate steroids are metabolized from cholesterol by steroidogenic enzymes as shown in the "steroid map" of the adrenal cortex. The H295R cell line originates from adrenocortical cancer (ACC), and is commonly used for studying steroidogenesis from its steroid-producing features. From 2009, many studies searched for miRNAs in the adrenal gland as biomarkers to distinguish between adrenal cancer, adrenal adenomas, and normal adrenal cortex tissue. The discovery of insulin-like growth factor 2 (IGF2) overexpressed in ACCs compared to adenomas makes it a useful biomarker [1]. A miRNA that is expressed from the intronic region of IGF2 also correlated with ACCs [2]. The mi-RNA that targets IGF2 in ACCs is mi-RNA-100[3], which is also known to target other cancers [4-7]. Mi-RNA-195 down-regulation and mi-RNA-483-5p up-regulation were found to be associated with poor prognosis in ACCs [8]. In later studies, these two mi-RNAs were found to be detectable in the serum of patients with high recurrence risk of ACCs [9]. Especially, mi-RNA- 483-5p predicts recurrence and was shown to correlate with the amount of circulating tumor cells ACCs [10]. Other studies show mi- RNA-675, miRNA-139-3p, and micro-RNA-335 to be upregulated in ACCs compared to adrenal adenomas [11]. Combined with pathway analysis, miR-184 and mi-R-503 show higher and mi-R- 511 and miR-214 show lower expression in ACCs involving the G2/M checkpoint [12]. In adrenocortical tumors microRNA profiles tend to differ from normal adrenal cortex. miRNA-24 was down- regulated in aldosterone-producing adenomas [13]. A bilateral adrenal hyperplasia form of adrenal adenoma called primary pigmented nodular adrenocortical disease (PPNAD) is known for its disorders of protein kinase A (PKA) regulatory subunit type 1A (PRKARIA) and leads to Cushing's syndrome in young people [14].

Let-7a was found to be upregulated in these patient's adrenal adenomas [15]. miR-200b and miR-203 were found to be down- regulated while miR-210 and miR-484 were up-regulated in a different study [16]. PPNADs were found to have up-regulated miRNA-449 expression, the miRNA that has been recently implicated in treatment of various cancers [17]. Though PPNAD rarely lead to ACC, there are some cases that do [18]. To this end, an extensive analysis of 45 ACCs by exome sequencing and SNP analysis revealed numerous mutations and methylation in the ACC group of poor prognosis and miRNA clusters in the ACC group with good prognosis. ZNRF3, encoding a cell surface E3 ubiquitin ligase was the most frequently altered in the study [19]. A genome wide DNA copy number report shows that whole genome duplication triggers massive DNA loss [20]. It is interesting that plants may have a defense mechanism relying on alternative splicing when this whole genome duplication occurs [21]. This would mean that if ACC cells survive whole genome duplication through alternative splicing, they would be prone to splicing inhibitors.

The recent discovery of circular RNAs (circRNAs) has given a large impact on human disease as well as RNA biology [22]. They are developed from back-splicing [23-25] and some function as micro-RNA sponges [26]. A recent report searching for circ-RNA by analyzing 123 r-RNA depleted RNA-seq data sets found a total of more than 300,000 of them, the majority of which are derived from coding regions of the gene, implying their role in translation or as backup RNAs. The tissue with the most circRNAs was the brain with 11.9% of circ-RNAs to be tissue-specific that may have a role in tissue development or differentiation. The circ-RNAs had many binding-sites for micro-RNA and RNA-binding protein. These tissue-specific circ-RNAs can be found as a database online (http:// gb.whu.edu.cn/TSCD) [27]. Another report found nearly half of circ-RNAs expressed in a tissue-specific manner with more found in gland tissues: mammary gland has nearly 10,000; adrenal gland has more than 2,000; and the average of normal tissue is around 1,200 [28]. The characterization of these circRNAs have just started and functional relation with steroidogenesis has yet to be deciphered.

Steroidogenesis and Non-Coding RNAs

For functional studies targeting adrenocortical hormone production, there is scarce information of involving non-coding RNAs. Bioinformatic analyses of the 3' UTR of CYP11B1(11p- hydroxylase) and CYP11B2 (aldosterone synthase) found mi- RNA-24 to be the common regulator that eventually regulates cortisol and aldosterone production, respectively [13]. The mi- RNA, let-7b was found to expressed in a negative correlated manner with cortisol levels of PPNAD patients [15]. Mi-RNA-21 was found to be upregulated in an angiotensin E -stimulated screen of more than 200 mi-RNAs in H295R cells. Mi-RNA-21 per se induced aldosterone secretion as well as H295R cell proliferation [29]. All the steroidogenic enzymes are regulated by NR5A1, previously known as steroidogenic factor-1 (SF-1) [30] or Ad4-binding protein (Ad4BP) [31]. NR5A1 knockout mice lack the adrenal gland [32], which shows its importance in adrenal development as well as steroidogenic function. miRNAs targeting NR5A1 can be found in the ovary where miRNA-107 is involved in ovary development [33] . NR5A1 is also known for other targets in the adrenal gland implicating its multi-functional role in adrenocortical homeostasis [34] .

Perspectives

The known CRH-ACTH-cortisol axis is well-defined and will probably be found to be more fine-tuned by functional analyses of related non-coding RNAs. Moreover, its interplay with neurosteroid production and maybe with their non-coding RNAs may unveil pathogenesis of diseases of unknown etiology such as schizophrenic states or mood- or neurotic-disorders.

References

  1. Giordano TJ, Kuick R, Gauger PG, Vinco M, Bauersfeld J, et al. (2009) Molecular classification and prognostication of adrenocortical tumors by transcriptome profiling. Clin Cancer Res 15(2): 668-676.
  2. Patterson EE, Holloway AK, Weng J, Fojo T, Kebebew E (2011) Micro- RNA profiling of adrenocortical tumors reveals miR-483 as a marker of malignancy. Cancer 117: 1630-1639.
  3. Doghman M, El Wakil A, Cardinaud B, Thomas E, Wang J, et al. (2010) Regulation of insulin-like growth factor-mammalian target of rapamycin signaling by microRNA in childhood adrenocortical tumors. Cancer research 70(11): 4666-4675.
  4. Tovar V, Alsinet C, Villanueva A, Hoshida Y, Sole M, et al. (2010) IGF activation in a molecular subclass of hepatocellular carcinoma and pre- clinical efficacy of IGF-1R blockage. Journal of hepatology 52(4): 550559.
  5. Nagaraja AK, Creighton CJ, Yu Z, Zhu H, Gunaratne PH, et al. (2010) A link between mir-100 and FRAP1/mTOR in clear cell ovarian cancer. Molecular Endocrinology 24(2): 447-463.
  6. Shi W, Alajez NM, Bastianutto C, Hui AB, Mocanu JD, et al. (2010) Significance of Plk1 regulation by miR-100 in human nasopharyngeal cancer. International Journal of Cancer 126(9): 2036-2048.
  7. Gebeshuber CA, Martinez J (2013) miR-100 suppresses IGF2 and inhibits breast tumorigenesis by interfering with proliferation and survival signaling. Oncogene 32(27): 3306-3310.
  8. Soon PS, Tacon LJ, Gill AJ, Bambach CP, Sywak MS, et al. (2009) miR- 195 and miR-483-5p Identified as Predictors of Poor Prognosis in Adrenocortical Cancer. Clin Cancer Res 15(24): 7684-7692.
  9. Chabre O, Libe R, Assie G, Barreau O, Bertherat J, et al. (2013) Serum miR- 483-5p and miR-195 are predictive of recurrence risk in adrenocortical cancer patients. Endocr Relat Cancer 20(4): 579-594.
  10. Salvianti F, Canu L, Poli G, Armignacco R, Scatena C, et al. (2017) New insights in the clinical and translational relevance of miR483-5p in adrenocortical cancer. Oncotarget 8(39): 65525-65533.
  11. Schmitz K, Helwig J, Bertram S, Sheu S, Suttorp A, et al. (2011) Differential expression of microRNA-675, microRNA-139-3p and microRNA-335 in benign and malignant adrenocortical tumors. J Clin Pathol 64(6): 529.
  12. Tombol Z, Szabo PM, Molnar V, Wiener Z, Tolgyesi G, et al. (2009) Integrative molecular bioinformatics study of human adrenocortical tumors: microRNA, tissue-specific target prediction, and pathway analysis. Endocr Relat Cancer 16(3): 895-906.
  13. Robertson S, MacKenzie SM, Alvarez-Madrazo S, Diver LA, Lin J, et al. (2013) MicroRNA-24 is a novel regulator of aldosterone and cortisol production in the human adrenal cortex. Hypertension 62(3): 572-578.
  14. Stratakis CA (2008) Cushing syndrome caused by adrenocortical tumors and hyperplasias (corticotropin- independent Cushing syndrome). Endocr Dev 13: 117-132.
  15. Iliopoulos D, Bimpaki EI, Nesterova M, Stratakis CA. (2009) MicroRNA signature of primary pigmented nodular adrenocortical disease: clinical correlations and regulation of Wnt signaling. Cancer Res 69(8): 32783282.
  16. Bimpaki EI, Iliopoulos D, Moraitis A, Stratakis CA (2010) MicroRNA signature in massive macronodular adrenocortical disease and implications for adrenocortical tumorigenesis. Clinical endocrinology 72(6): 744-751.
  17. Yong Ming H, Ai Jun J, Xiao Yue X, Jian Wei L, Chen Y, et al. (2017) miR- 449a: a potential therapeutic agent for cancer. Anticancer Drugs 28(10): 1067-1078.
  18. Anselmo J, Medeiros S, Carneiro V, Greene E, Levy I, et al. (2012) A large family with Carney complex caused by the S147G PRKAR1A mutation shows a unique spectrum of disease including adrenocortical cancer. J Clin Endocrinol Metab 97(2): 351-359.
  19. Assie G, Letouze E, Fassnacht M, Jouinot A, Luscap W, et al. (2014) Integrated genomic characterization of adrenocortical carcinoma. Nature genetics 46(6): 607-612.
  20. Zheng S, Cherniack AD, Dewal N, Moffitt RA, Danilova L, et al. (2016) Comprehensive pan-genomic characterization of adrenocortical carcinoma. Cancer cell 29(5): 723-736.
  21. Zhang PG, Huang SZ, Pin AL, Adams KL (2010) Extensive divergence in alternative splicing patterns after gene and genome duplication during the evolutionary history of Arabidopsis. Molecular biology and evolution 27(7): 1686-1697.
  22. Salzman J, Gawad C, Wang PL, Lacayo N, Brown PO (2012) Circular RNAs are the predominant transcript isoform from hundreds of human genes in diverse cell types 7: e30733.
  23. Ashwal Fluss R, Meyer M, Pamudurti NR, Ivanov A, Bartok O, et al. (2014) circ RNA biogenesis competes with pre-mRNA splicing. Mol Cell 56(1): 55-66.
  24. Zhang XO, Wang HB, Zhang Y, Lu X, Chen LL, et al. (2014) Complementary sequence-mediated exon circularization. Cell 159(2): 134-147.
  25. Starke S, Jost I, Rossbach O, Schneider T, Schreiner S, Hung LH, et al. (2015) Exon circularization requires canonical splice signals. Cell Rep 10(1): 103-111.
  26. Hansen TB, Jensen TI, Clausen BH, Bramsen JB, Finsen B, et al. (2013) Natural RNA circles function as efficient microRNA sponges. Nature 495: 384-388.
  27. Xia S, Feng J, Lei L, Hu J, Xia L, et al. (2017) Comprehensive characterization of tissue-specific circular RNAs in the human and mouse genomes. Brief Bio inform 18(6): 984-992.
  28. Xu T, Wu J, Han P, Zhao Z, Song, X (2017) Circular RNA expression profiles and features in human tissues: a study using RNA-seq data supp6: 680.
  29. Romero DG, Plonczynski MW, Carvajal CA, Gomez Sanchez EP, Gomez Sanchez CE (2008) Microribonucleic acid-21 increases aldosterone secretion and proliferation in H295R human adrenocortical cells. Endocrinology 149(5): 2477-2483.
  30. Lala DS, Rice DA, Parker KL (1992) Steroidogenic factor I, a key regulator of steroidogenic enzyme expression, is the mouse homolog of fushi tarazu-factor I. Mol Endocrinol 6(8): 1249-1258.
  31. Honda S, Morohashi K, Nomura M, Takeya H, Kitajima M (1993) Ad4BP regulating steroidogenic P-450 gene is a member of steroid hormone receptor superfamily. J Biol Chem 268(10): 7494-7502.
  32. Luo X, Ikeda YA, Parker KL (1994) A cell-specific nuclear receptor is essential for adrenal and gonadal development and sexual differentiation. Cell 77(4): 481-490.
  33. Miao N, Wang X, Hou Y, Feng Y, Gong, Y (2016) Identification of male- biased microRNA-107 as a direct regulator for nuclear receptor subfamily 5 group A member 1 based on sexually dimorphic microRNA expression profiling from chicken embryonic gonads. Mol Cell Endocrinol 429: 2940.
  34. Lalli E, Doghman M, De Late PL, El Wakil A, Mus Veteau I (2013) Beyond steroidogenesis: novel target genes for SF-1 discovered by genomics. Mol Cell Endocrinol 371(1-2): 154-159.