open access

Vol 73, No 5 (2022)
Original paper
Submitted: 2022-02-28
Accepted: 2022-05-09
Published online: 2022-07-27
Get Citation

Low miR-222 expression in human visceral adipose tissue is associated with insulin resistance and PTEN and p53 mRNA levels

Paulina Estrella-Ibarra12, Andrés Cruz-Hernández3, Ana Karen Torres-Arreola12, Francisco Campos-Maldonado12, Adriana Jheny Rodríguez-Mendez2, Olga Patricia García-Obregón4, Juan Carlos Solís-Sáinz2, María del Carmen Aburto-Fernández5, Ricardo M. Lerma-Alvarado5, Pablo García-Solís2
·
Pubmed: 35971931
·
Endokrynol Pol 2022;73(5):846-855.
Affiliations
  1. Biological Sciences PhD Program, School of Natural Sciences, Autonomous University of Queretaro, Queretaro, Mexico
  2. Center of Advanced Biomedical Research, School of Medicine, Autonomous University of Queretaro, Queretaro, Mexico
  3. Agronomy School, De La Salle Bajio University, Guanajuato, Mexico
  4. Department of Human Nutrition, School of Natural Sciences, Autonomous University of Queretaro, Queretaro, Mexico
  5. Department of General Surgery, General Hospital of Queretaro, Queretaro, Mexico

open access

Vol 73, No 5 (2022)
Original Paper
Submitted: 2022-02-28
Accepted: 2022-05-09
Published online: 2022-07-27

Abstract

Introduction: The aim of this is study was to analyse the expression of miR-193b, miR-378, miR-Let7-d, and miR-222 in human visceral adipose tissue (VAT), as well as their association with obesity, insulin resistance (IR), and their role in the regulation of genes controlling adipose tissue homeostasis, including adipocytokines, the phosphatase and tension homologue (PTEN), and tumour protein 53 (p53).

Material and methods: VAT was obtained from normal-weight (NW), overweight, and obese (OW/OB) subjects with and without IR. Stem-loop RT-qPCR was used to evaluate miRNA expression levels. miRTarBase 4.0, miRWalk, and DIANA-TarBase v8 were used for prediction of validated target gene of the miRNA analysed. A qPCR was used to evaluate PTEN, p53, leptin (LEP), and adiponectin (ADIPOQ) mRNA.

Results: miR-222 was lower in IR subjects, and miR-222 and miR-378 negatively correlated with HOMA-IR. PTEN and p53 are miR-222 direct targets according to databases. mRNA expression of PTEN and p53 was lower in OW/OB subjects with and without IR, compared to NW group and its levels positively associated with miR-222. Additionally, p53 and PTEN are positively associated with serum leptin levels. On the other hand, miR-193b and miR-378 negatively correlated with serum leptin but not with mRNA levels. Moreover, miR-Let-7d negatively correlated with serum adiponectin but not with adiponectin mRNA levels.

Conclusions: Lower miR-222 levels are associated with IR, and PTEN and p53 expression; the implication of these genes in adipose tissue homeostasis needs more research.

Abstract

Introduction: The aim of this is study was to analyse the expression of miR-193b, miR-378, miR-Let7-d, and miR-222 in human visceral adipose tissue (VAT), as well as their association with obesity, insulin resistance (IR), and their role in the regulation of genes controlling adipose tissue homeostasis, including adipocytokines, the phosphatase and tension homologue (PTEN), and tumour protein 53 (p53).

Material and methods: VAT was obtained from normal-weight (NW), overweight, and obese (OW/OB) subjects with and without IR. Stem-loop RT-qPCR was used to evaluate miRNA expression levels. miRTarBase 4.0, miRWalk, and DIANA-TarBase v8 were used for prediction of validated target gene of the miRNA analysed. A qPCR was used to evaluate PTEN, p53, leptin (LEP), and adiponectin (ADIPOQ) mRNA.

Results: miR-222 was lower in IR subjects, and miR-222 and miR-378 negatively correlated with HOMA-IR. PTEN and p53 are miR-222 direct targets according to databases. mRNA expression of PTEN and p53 was lower in OW/OB subjects with and without IR, compared to NW group and its levels positively associated with miR-222. Additionally, p53 and PTEN are positively associated with serum leptin levels. On the other hand, miR-193b and miR-378 negatively correlated with serum leptin but not with mRNA levels. Moreover, miR-Let-7d negatively correlated with serum adiponectin but not with adiponectin mRNA levels.

Conclusions: Lower miR-222 levels are associated with IR, and PTEN and p53 expression; the implication of these genes in adipose tissue homeostasis needs more research.

Get Citation

Keywords

microRNA (miRNA); visceral adipose tissue; obesity; insulin resistance; adipocytokines

About this article
Title

Low miR-222 expression in human visceral adipose tissue is associated with insulin resistance and PTEN and p53 mRNA levels

Journal

Endokrynologia Polska

Issue

Vol 73, No 5 (2022)

Article type

Original paper

Pages

846-855

Published online

2022-07-27

Page views

4363

Article views/downloads

425

DOI

10.5603/EP.a2022.0057

Pubmed

35971931

Bibliographic record

Endokrynol Pol 2022;73(5):846-855.

Keywords

microRNA (miRNA)
visceral adipose tissue
obesity
insulin resistance
adipocytokines

Authors

Paulina Estrella-Ibarra
Andrés Cruz-Hernández
Ana Karen Torres-Arreola
Francisco Campos-Maldonado
Adriana Jheny Rodríguez-Mendez
Olga Patricia García-Obregón
Juan Carlos Solís-Sáinz
María del Carmen Aburto-Fernández
Ricardo M. Lerma-Alvarado
Pablo García-Solís

References (44)
  1. Saini V. Molecular mechanisms of insulin resistance in type 2 diabetes mellitus. World J Diabetes. 2010; 1(3): 68–75.
  2. Goossens GH. The role of adipose tissue dysfunction in the pathogenesis of obesity-related insulin resistance. Physiol Behav. 2008; 94(2): 206–218.
  3. Castoldi A, Naffah de Souza C, Câmara NO, et al. The Macrophage Switch in Obesity Development. Front Immunol. 2015; 6: 637.
  4. Lihn AS, Bruun JM, He G, et al. Lower expression of adiponectin mRNA in visceral adipose tissue in lean and obese subjects. Mol Cell Endocrinol. 2004; 219(1-2): 9–15.
  5. Sáinz N, Barrenetxe J, Moreno-Aliaga MJ, et al. Leptin resistance and diet-induced obesity: central and peripheral actions of leptin. Metabolism. 2015; 64(1): 35–46.
  6. Huang W, Queen NJ, McMurphy TB, et al. Adipose PTEN regulates adult adipose tissue homeostasis and redistribution via a PTEN-leptin-sympathetic loop. Mol Metab. 2019; 30: 48–60.
  7. Lee YK, Chung Y, Lee JiH, et al. The Intricate Role of p53 in Adipocyte Differentiation and Function. Cells. 2020; 9(12).
  8. Stambolic V, MacPherson D, Sas D, et al. Regulation of PTEN transcription by p53. Mol Cell. 2001; 8(2): 317–325.
  9. Venniyoor A, Venniyoor A. PTEN: A Thrifty Gene That Causes Disease in Times of Plenty? Front Nutr. 2020; 7: 81.
  10. Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004; 116(2): 281–297.
  11. Lewis BP, Burge CB, Bartel DP. Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell. 2005; 120(1): 15–20.
  12. Park JH, Kim SH, Lee MS, et al. Epigenetic modification by dietary factors: Implications in metabolic syndrome. Mol Aspects Med. 2017; 54: 58–70.
  13. Li Z, Rana TM. Therapeutic targeting of microRNAs: current status and future challenges. Nat Rev Drug Discov. 2014; 13(8): 622–638.
  14. Lavebratt C, Almgren M, Ekström TJ. Epigenetic regulation in obesity. Int J Obes (Lond). 2012; 36(6): 757–765.
  15. Kim VN. MicroRNA biogenesis: coordinated cropping and dicing. Nat Rev Mol Cell Biol. 2005; 6(5): 376–385.
  16. Ortega FJ, Moreno-Navarrete JM, Pardo G, et al. MiRNA expression profile of human subcutaneous adipose and during adipocyte differentiation. PLoS One. 2010; 5(2): e9022.
  17. Arner E, Mejhert N, Kulyté A, et al. Adipose tissue microRNAs as regulators of CCL2 production in human obesity. Diabetes. 2012; 61(8): 1986–1993.
  18. Meerson A, Traurig M, Ossowski V, et al. Human adipose microRNA-221 is upregulated in obesity and affects fat metabolism downstream of leptin and TNF-α. Diabetologia. 2013; 56(9): 1971–1979.
  19. Belarbi Y, Mejhert N, Lorente-Cebrián S, et al. MicroRNA-193b Controls Adiponectin Production in Human White Adipose Tissue. J Clin Endocrinol Metab. 2015; 100(8): E1084–E1088.
  20. Ishida M, Shimabukuro M, Yagi S, et al. MicroRNA-378 regulates adiponectin expression in adipose tissue: a new plausible mechanism. PLoS One. 2014; 9(11): e111537.
  21. Brennan E, Wang Bo, McClelland A, et al. Protective Effect of let-7 miRNA Family in Regulating Inflammation in Diabetes-Associated Atherosclerosis. Diabetes. 2017; 66(8): 2266–2277.
  22. World Health Organization, “Obesity and overweight,” 2020. https://www.who.int/news-room/fact-sheets/detail/obesity-and-overweight (Dec. 29, 2020).
  23. Ascaso J, Pardo S, Real J, et al. Diagnosing Insulin Resistance by Simple Quantitative Methods in Subjects With Normal Glucose Metabolism. Diabetes Care. 2003; 26(12): 3320–3325.
  24. Chen C, Ridzon DA, Broomer AJ, et al. Real-time quantification of microRNAs by stem-loop RT-PCR. Nucleic Acids Res. 2005; 33(20): e179.
  25. Czimmerer Z, Hulvely J, Simandi Z, et al. A versatile method to design stem-loop primer-based quantitative PCR assays for detecting small regulatory RNA molecules. PLoS One. 2013; 8(1): e55168.
  26. Heneghan HM, Miller N, McAnena OJ, et al. Differential miRNA expression in omental adipose tissue and in the circulation of obese patients identifies novel metabolic biomarkers. J Clin Endocrinol Metab. 2011; 96(5): E846–E850.
  27. Jones A, Danielson KM, Benton MC, et al. miRNA Signatures of Insulin Resistance in Obesity. Obesity (Silver Spring). 2017; 25(10): 1734–1744.
  28. Yamauchi T, Nio Y, Maki T, et al. Targeted disruption of AdipoR1 and AdipoR2 causes abrogation of adiponectin binding and metabolic actions. Nat Med. 2007; 13(3): 332–339.
  29. Blüher M, Williams CJ, Klöting N, et al. Gene expression of adiponectin receptors in human visceral and subcutaneous adipose tissue is related to insulin resistance and metabolic parameters and is altered in response to physical training. Diabetes Care. 2007; 30(12): 3110–3115.
  30. Bauche IB, Ait El Mkadem S, Rezsohazy R, et al. Adiponectin downregulates its own production and the expression of its AdipoR2 receptor in transgenic mice. Biochem Biophys Res Commun. 2006; 345(4): 1414–1424.
  31. Brennan E, Wang Bo, McClelland A, et al. Protective Effect of let-7 miRNA Family in Regulating Inflammation in Diabetes-Associated Atherosclerosis. Diabetes. 2017; 66(8): 2266–2277.
  32. Rydén M, Andersson DP, Bergström IB, et al. Adipose tissue and metabolic alterations: regional differences in fat cell size and number matter, but differently: a cross-sectional study. J Clin Endocrinol Metab. 2014; 99(10): E1870–E1876.
  33. Fang L, Guo F, Zhou L, et al. The cell size and distribution of adipocytes from subcutaneous and visceral fat is associated with type 2 diabetes mellitus in humans. Adipocyte. 2015; 4(4): 273–279.
  34. Li D, Song H, Shuo L, et al. Gonadal white adipose tissue-derived exosomal MiR-222 promotes obesity-associated insulin resistance. Aging (Albany NY). 2020; 12(22): 22719–22743.
  35. Al-Rawaf HA. Circulating microRNAs and adipokines as markers of metabolic syndrome in adolescents with obesity. Clin Nutr. 2019; 38(5): 2231–2238.
  36. Prats-Puig A, Ortega FJ, Mercader JM, et al. Changes in circulating microRNAs are associated with childhood obesity. J Clin Endocrinol Metab. 2013; 98(10): E1655–E1660.
  37. Thomou T, Mori MA, Dreyfuss JM, et al. Adipose-derived circulating miRNAs regulate gene expression in other tissues. Nature. 2017; 542(7642): 450–455.
  38. Kokkinopoulou I, Maratou E, Mitrou P, et al. Decreased expression of microRNAs targeting type-2 diabetes susceptibility genes in peripheral blood of patients and predisposed individuals. Endocrine. 2019; 66(2): 226–239.
  39. Shu J, Silva BV, Gao T, et al. Dynamic and Modularized MicroRNA Regulation and Its Implication in Human Cancers. Sci Rep. 2017; 7(1): 13356.
  40. Kadkhoda G, Zarkesh M, Saidpour A, et al. Association of dietary intake of fruit and green vegetables with PTEN and P53 mRNA gene expression in visceral and subcutaneous adipose tissues of obese and non-obese adults. Gene. 2020; 733: 144353.
  41. Morley TS, Xia JY, Scherer PE. Selective enhancement of insulin sensitivity in the mature adipocyte is sufficient for systemic metabolic improvements. Nat Commun. 2015; 6: 7906.
  42. Ortega FJ, Moreno-Navarrete JM, Mayas D, et al. Inflammation and insulin resistance exert dual effects on adipose tissue tumor protein 53 expression. Int J Obes (Lond). 2014; 38(5): 737–745.
  43. Huang Q, Liu M, Du X, et al. Role of p53 in preadipocyte differentiation. Cell Biol Int. 2014; 38(12): 1384–1393.
  44. Liu Y, Beyer A, Aebersold R. On the Dependency of ular Protein Levels on mRNA Abundance. Cell. 2016; 38(12): 1384–93.

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