KRAS Activation and over-expression of SIRT1/BCL6 Contributes to the Pathogenesis of Endometriosis and Progesterone Resistance

Jung-Yoon Yoo, Tae Hoon Kim, Asgerally T Fazleabas, Wilder A Palomino, Soo Hyun Ahn, Chandrakant Tayade, David P Schammel, Steven L Young, Jae-Wook Jeong, Bruce A Lessey, Jung-Yoon Yoo, Tae Hoon Kim, Asgerally T Fazleabas, Wilder A Palomino, Soo Hyun Ahn, Chandrakant Tayade, David P Schammel, Steven L Young, Jae-Wook Jeong, Bruce A Lessey

Abstract

Endometriosis is an inflammatory condition that is associated with progesterone resistance and cell proliferation, resulting in pain, infertility and pregnancy loss. We previously demonstrated phosphorylation of STAT3 in eutopic endometrium of infertile women with this disorder leading to over-expression of the oncogene BCL6 and stabilization of hypoxia-induced factor 1 alpha (HIF-1α). Here we report coordinated activation of KRAS and over-expression of Sirtuin 1 (SIRT1), a histone deacetylase and gene silencer, in the eutopic endometrium from women with endometriosis throughout the menstrual cycle. The mice with conditional activation of KRAS in the PGR positive cells reveal an increase of SIRT1 expression in the endometrium compared to control mice. The expression of progesterone receptor target genes including the Indian Hedgehog pathway genes are significantly down-regulated in the mutant mice. SIRT1 co-localizes with BCL6 in the nuclei of affected individuals and both proteins bind to and suppress the promoter of GLI1, a critical mediator of progesterone action in the Indian Hedgehog pathway, by ChIP analysis. In eutopic endometrium, GLI1 expression is reduced in women with endometriosis. Together, these data suggest that KRAS, SIRT1 and BCL6 are coordinately over-expressed in eutopic endometrium of women with endometriosis and likely participate in the pathogenesis of endometriosis.

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Figure 1
Figure 1
Endometriosis is associated with elevated serum inflammatory cytokines including (a) IL-1a, (b) IL-6 and (c) IL-17, compared to normal controls.
Figure 2
Figure 2
Correlation of between KRAS and SIRT1 in human endometrium with endometriosis. (a) Western blot analysis of SIRT1 and BCL6 proteins in proliferative and secretory phases of human endometrium with endometriosis. β-actin was used as sample-loading control. Representative blots have been cropped to reduce unnecessary area. Full-length blots are presented in Supplementary Fig. S2. (b) Densitometric analysis of KRAS and SIRT1 protein levels by Western blot analysis in eutopic endometrium from proliferative and secretory phase in women with and without endometriosis. (c) Correlation between SIRT1 and KRAS in women with endometriosis throughout the menstrual cycle phases based on Western blot analysis (correlation coefficient = 0.4641, p = 0.0009). (d and e) H-score of KRAS (d) and SIRT1 (e) expression in endometrium from women with and without endometriosis and representative photomicrograph of immunohistochemical staining of KRAS in endometrium from women without and with endometriosis. The results represent the mean ± SEM. ***p < 0.001.
Figure 3
Figure 3
Correlation of between SIRT1 and BCL6. (a) Correlation analysis between SIRT1 and BCL6 in human endometrium with endometriosis. (b) Immunoprecipitation (IP) analysis between SIRT1 and BCL6 in Ishikawa cells and Human endometrium with endometriosis. Representative blots have been cropped to reduce unnecessary area. Full-length blots are presented in Supplementary Fig. S4. (c) Co-localization of SIRT1 and BCL6 in the human endometrium without and with endometriosis by immunofluorescence analysis.
Figure 4
Figure 4
Levels of SIRT1 and BCL6 proteins during progression of endometriosis in a baboon model. (a and b) H-score of SIRT1 (a) and BCL6 (b) expression in endometriosis baboon model induced by intraperitoneal inoculation of menstrual endometrium during progression of endometriosis. The results represent the mean ± SEM. *p < 0.05 and ***p < 0.001. (c and d) Representative photomicrograph of immunohistochemical staining of SIRT1 (c) and BCL6 (d) in the baboon endometrium of pre-inoculation and 3, 9 and 15 months post-inoculation during endometriosis progression.
Figure 5
Figure 5
Levels of SIRT1 in the KRAS activation mouse model. (a) Representative photomicrograph of immunohistochemical staining of SIRT1 in the control and KRAS activation mouse. (b) The mRNA expression level of P4 target genes in the uterus from control and KRAS activation mice (n = 9). The results represent the mean ± SEM. *p < 0.05 and ***p < 0.001.
Figure 6
Figure 6
Regulation of GLI1 gene expression by SIRT1 and BCL6 proteins. (a) Western blot analysis of BCL6 and SIRT1 in Ishikawa cells treated with E2 + MPA for 0, 30 min, 6, 12, and 24 hours. β-actin was used as sample-loading control. Representative blots have been cropped to reduce unnecessary area. Full-length blots are presented in Supplementary Fig. S5. (b) Quantitative real time PCR analysis of GLI1 gene expression in Ishikawa cells treated with E2 + MPA for 0, 6, 12, and 24 hours. (c) Map of BCL6 binding site on the GLI1 promoter (Gray boxes). Negative control (N.C.) region on the GLI1 gene was used as negative control of ChIP assay. Primers used in ChIP assay are presented by arrows. (d) ChIP assay using anti-SIRT1 antibody on GLI1 promoter in Ishikawa cells treated with or without E2 + MPA for 24 hours. The results represent the mean ± SEM. *p < 0.05, **p < 0.01, and ***p < 0.001.
Figure 7
Figure 7
Levels of GLI1 in endometrium from women with and without endometriosis. (a) H-score of GLI1 expression in endometrium from women with and without endometriosis. The results represent the mean ± SEM. *p < 0.05. (b) Representative photomicrograph of immunohistochemical staining of GLI1 in endometrium from women without and with endometriosis.

References

    1. Simoens S, et al. The burden of endometriosis: costs and quality of life of women with endometriosis and treated in referral centres. Hum Reprod. 2012;27:1292–1299. doi: 10.1093/humrep/des073.
    1. Fourquet J, Baez L, Figueroa M, Iriarte RI, Flores I. Quantification of the impact of endometriosis symptoms on health-related quality of life and work productivity. Fertil Steril. 2011;96:107–112. doi: 10.1016/j.fertnstert.2011.04.095.
    1. Bulun SE. Endometriosis. The New England journal of medicine. 2009;360:268–279. doi: 10.1056/NEJMra0804690.
    1. de Ziegler D, Borghese B, Chapron C. Endometriosis and infertility: pathophysiology and management. Lancet. 2010;376:730–738. doi: 10.1016/S0140-6736(10)60490-4.
    1. Bulun SE. Endometriosis. N Engl J Med. 2009;360:268–279. doi: 10.1056/NEJMra0804690.
    1. Wang C, Mavrogianis PA, Fazleabas AT. Endometriosis is associated with progesterone resistance in the baboon (Papio anubis) oviduct: evidence based on the localization of oviductal glycoprotein 1 (OVGP1) Biology of reproduction. 2009;80:272–278. doi: 10.1095/biolreprod.108.072496.
    1. Lessey BA, Palomino WA, Apparao KB, Young SL, Lininger RA. Estrogen receptor-alpha (ER-alpha) and defects in uterine receptivity in women. Reproductive biology and endocrinology: RB&E. 2006;4(Suppl 1):S9. doi: 10.1186/1477-7827-4-S1-S9.
    1. Dinulescu DM, et al. Role of K-ras and Pten in the development of mouse models of endometriosis and endometrioid ovarian cancer. Nature medicine. 2005;11:63–70. doi: 10.1038/nm1173.
    1. Cheng CW, et al. Activation of mutated K-ras in donor endometrial epithelium and stroma promotes lesion growth in an intact immunocompetent murine model of endometriosis. The Journal of pathology. 2011;224:261–269. doi: 10.1002/path.2852.
    1. Luong HT, et al. No evidence for genetic association with the let-7 microRNA-binding site or other common KRAS variants in risk of endometriosis. Hum Reprod. 2012;27:3616–3621. doi: 10.1093/humrep/des329.
    1. Santamaria X, Taylor H. MicroRNA and gynecological reproductive diseases. Fertility and sterility. 2014;101:1545–1551. doi: 10.1016/j.fertnstert.2014.04.044.
    1. Grechukhina O, et al. A polymorphism in a let-7 microRNA binding site of KRAS in women with endometriosis. EMBO molecular medicine. 2012;4:206–217. doi: 10.1002/emmm.201100200.
    1. Burney, R. O. et al. MicroRNA expression profiling of eutopic secretory endometrium in women with versus without endometriosis. Molecular human reproduction15, 625–631, doi:gap068 [pii] 10.1093/molehr/gap068 (2009).
    1. Kasinski AL, Slack FJ. miRNA-34 prevents cancer initiation and progression in a therapeutically resistant K-ras and p53-induced mouse model of lung adenocarcinoma. Cancer research. 2012;72:5576–5587. doi: 10.1158/0008-5472.CAN-12-2001.
    1. Stahlhut C, Slack FJ. Combinatorial Action of MicroRNAs let-7 and miR-34 Effectively Synergizes with Erlotinib to Suppress Non-small Cell Lung Cancer Cell Proliferation. Cell Cycle. 2015;14:2171–2180. doi: 10.1080/15384101.2014.1003008.
    1. Okada N, et al. A positive feedback between p53 and miR-34 miRNAs mediates tumor suppression. Genes & development. 2014;28:438–450. doi: 10.1101/gad.233585.113.
    1. Basso K, Dalla-Favera R. Roles of BCL6 in normal and transformed germinal center B cells. Immunol Rev. 2012;247:172–183. doi: 10.1111/j.1600-065X.2012.01112.x.
    1. Okabe S, et al. BAZF, a novel Bcl6 homolog, functions as a transcriptional repressor. Mol Cell Biol. 1998;18:4235–4244. doi: 10.1128/MCB.18.7.4235.
    1. Phan, R. T. & Dalla-Favera, R. The BCL6 proto-oncogene suppresses p53 expression in germinal-centre B cells. Nature432, 635–639, doi:nature03147 [pii] 10.1038/nature03147 (2004).
    1. Harris MB, et al. Transcriptional repression of Stat6-dependent interleukin-4-induced genes by BCL-6: specific regulation of iepsilon transcription and immunoglobulin E switching. Mol Cell Biol. 1999;19:7264–7275. doi: 10.1128/MCB.19.10.7264.
    1. Arguni E, et al. JunD/AP-1 and STAT3 are the major enhancer molecules for high Bcl6 expression in germinal center B cells. International immunology. 2006;18:1079–1089. doi: 10.1093/intimm/dxl041.
    1. Kim BG, et al. Aberrant activation of signal transducer and activator of transcription-3 (STAT3) signaling in endometriosis. Human reproduction. 2015;30:1069–1078. doi: 10.1093/humrep/dev050.
    1. Evans-Hoeker E, et al. Endometrial BCL6 Overexpression in Eutopic Endometrium of Women With Endometriosis. Reprod Sci. 2016;23:1234–1241. doi: 10.1177/1933719116649711.
    1. Frye RA. Characterization of five human cDNAs with homology to the yeast SIR2 gene: Sir2-like proteins (sirtuins) metabolize NAD and may have protein ADP-ribosyltransferase activity. Biochem Biophys Res Commun. 1999;260:273–279. doi: 10.1006/bbrc.1999.0897.
    1. Luo, J. et al. Negative control of p53 by Sir2alpha promotes cell survival under stress. Cell107, 137–148, doi:S0092-8674(01)00524-4 [pii] (2001).
    1. Poulose N, Raju R. Sirtuin regulation in aging and injury. Biochim Biophys Acta. 2015;1852:2442–2455. doi: 10.1016/j.bbadis.2015.08.017.
    1. Song NY, Surh YJ. Janus-faced role of SIRT1 in tumorigenesis. Ann N Y Acad Sci. 2012;1271:10–19. doi: 10.1111/j.1749-6632.2012.06762.x.
    1. Lin L, et al. SIRT1 promotes endometrial tumor growth by targeting SREBP1 and lipogenesis. Oncol Rep. 2014;32:2831–2835.
    1. Taguchi A, et al. Resveratrol suppresses inflammatory responses in endometrial stromal cells derived from endometriosis: a possible role of the sirtuin 1 pathway. J Obstet Gynaecol Res. 2014;40:770–778. doi: 10.1111/jog.12252.
    1. Mvunta, D. H. et al. Overexpression of SIRT1 is Associated With Poor Outcomes in Patients With Ovarian Carcinoma. Applied immunohistochemistry & molecular morphology: AIMM/official publication of the Society for Applied Immunohistochemistry, doi:10.1097/PAI.0000000000000316 (2016).
    1. Kriegl L, Vieth M, Kirchner T, Menssen A. Up-regulation of c-MYC and SIRT1 expression correlates with malignant transformation in the serrated route to colorectal cancer. Oncotarget. 2012;3:1182–1193. doi: 10.18632/oncotarget.628.
    1. Barnoud, T., Wilkey, D. W., Merchant, M. L., Clark, J. A. & Donninger, H. Proteomics Analysis Reveals Novel RASSF2 Interaction Partners. Cancers8, doi:10.3390/cancers8030037 (2016).
    1. Tiberi L, et al. A BCL6/BCOR/SIRT1 complex triggers neurogenesis and suppresses medulloblastoma by repressing Sonic Hedgehog signaling. Cancer Cell. 2014;26:797–812. doi: 10.1016/j.ccell.2014.10.021.
    1. Braundmeier AG, Fazleabas AT. The non-human primate model of endometriosis: research and implications for fecundity. Molecular human reproduction. 2009;15:577–586. doi: 10.1093/molehr/gap057.
    1. Johnson L, et al. Somatic activation of the K-ras oncogene causes early onset lung cancer in mice. Nature. 2001;410:1111–1116. doi: 10.1038/35074129.
    1. Soyal SM, et al. Cre-mediated recombination in cell lineages that express the progesterone receptor. Genesis. 2005;41:58–66. doi: 10.1002/gene.20098.
    1. Kim TH, et al. The Synergistic Effect of Conditional Pten Loss and Oncogenic K-ras Mutation on Endometrial Cancer Development Occurs via Decreased Progesterone Receptor Action. Journal of oncology. 2010;2010:139087.
    1. Martin L, Das RM, Finn CA. The inhibition by progesterone of uterine epithelial proliferation in the mouse. J Endocrinol. 1973;57:549–554. doi: 10.1677/joe.0.0570549.
    1. Martin L, Finn CA, Trinder G. Hypertrophy and hyperplasia in the mouse uterus after oestrogen treatment: an autoradiographic study. J Endocrinol. 1973;56:133–144. doi: 10.1677/joe.0.0560133.
    1. Bokhman JV. Two pathogenetic types of endometrial carcinoma. Gynecol Oncol. 1983;15:10–17. doi: 10.1016/0090-8258(83)90111-7.
    1. Sherman ME, et al. Risk factors and hormone levels in patients with serous and endometrioid uterine carcinomas. Mod Pathol. 1997;10:963–968.
    1. Deligdisch L, Holinka CF. Endometrial carcinoma: two diseases? Cancer Detect Prev. 1987;10:237–246.
    1. Kurman RJ, Kaminski PF, Norris HJ. The behavior of endometrial hyperplasia. A long-term study of “untreated” hyperplasia in 170 patients. Cancer. 1985;56:403–412. doi: 10.1002/1097-0142(19850715)56:2<403::AID-CNCR2820560233>;2-X.
    1. Al-Sabbagh M, Lam EW, Brosens JJ. Mechanisms of endometrial progesterone resistance. Molecular and cellular endocrinology. 2012;358:208–215. doi: 10.1016/j.mce.2011.10.035.
    1. Burney RO, et al. Gene expression analysis of endometrium reveals progesterone resistance and candidate susceptibility genes in women with endometriosis. Endocrinology. 2007;148:3814–3826. doi: 10.1210/en.2006-1692.
    1. Attia GR, et al. Progesterone receptor isoform A but not B is expressed in endometriosis. The Journal of clinical endocrinology and metabolism. 2000;85:2897–2902.
    1. Zannoni GF, et al. Mutational status of KRAS, NRAS, and BRAF in primary clear cell ovarian carcinoma. Virchows Archiv: an international journal of pathology. 2014;465:193–198. doi: 10.1007/s00428-014-1599-1.
    1. Bruner-Tran KL, Herington JL, Duleba AJ, Taylor HS, Osteen KG. Medical management of endometriosis: emerging evidence linking inflammation to disease pathophysiology. Minerva ginecologica. 2013;65:199–213.
    1. Pillarisetti S. A review of Sirt1 and Sirt1 modulators in cardiovascular and metabolic diseases. Recent Pat Cardiovasc Drug Discov. 2008;3:156–164. doi: 10.2174/157489008786263989.
    1. Walker SR, et al. STAT5 outcompetes STAT3 to regulate the expression of the oncogenic transcriptional modulator BCL6. Mol Cell Biol. 2013;33:2879–2890. doi: 10.1128/MCB.01620-12.
    1. Rahman S, Islam R. Mammalian Sirt1: insights on its biological functions. Cell communication and signaling: CCS. 2011;9:11. doi: 10.1186/1478-811X-9-11.
    1. Basso K, Dalla-Favera R. BCL6: master regulator of the germinal center reaction and key oncogene in B cell lymphomagenesis. Advances in immunology. 2010;105:193–210. doi: 10.1016/S0065-2776(10)05007-8.
    1. Evans-Hoeker, E. et al. Endometrial BCL6 Overexpression in Eutopic Endometrium of Women with Endometriosis. Reprod Sciences in press (2016).
    1. Huynh KD, Fischle W, Verdin E, Bardwell VJ. BCoR, a novel corepressor involved in BCL-6 repression. Genes Dev. 2000;14:1810–1823.
    1. Young SL, Lessey BA. Progesterone function in human endometrium: clinical perspectives. Seminars in reproductive medicine. 2010;28:5–16. doi: 10.1055/s-0029-1242988.
    1. Lessey BA, Young SL. Homeostasis imbalance in the endometrium of women with implantation defects: the role of estrogen and progesterone. Seminars in reproductive medicine. 2014;32:365–375. doi: 10.1055/s-0034-1376355.
    1. Lessey BA, Palomino WA, Apparao KB, Young SL, Lininger RA. Estrogen receptor-alpha (ER-alpha) and defects in uterine receptivity in women. Reproductive biology and endocrinology: RB&E. 2006;4(Suppl. 1):S9. doi: 10.1186/1477-7827-4-S1-S9.
    1. Bulun SE, et al. Progesterone resistance in endometriosis: link to failure to metabolize estradiol. Molecular and cellular endocrinology. 2006;248:94–103. doi: 10.1016/j.mce.2005.11.041.
    1. Pavone ME, et al. Altered retinoid uptake and action contributes to cell survival in endometriosis. The Journal of clinical endocrinology and metabolism. 2010;95:E300–309. doi: 10.1210/jc.2010-0459.
    1. Hardy DB, Janowski BA, Chen CC, Mendelson CR. Progesterone receptor inhibits aromatase and inflammatory response pathways in breast cancer cells via ligand-dependent and ligand-independent mechanisms. Mol Endocrinol. 2008;22:1812–1824. doi: 10.1210/me.2007-0443.
    1. Bulun, S. E. et al. Estrogen production and metabolism in endometriosis. Annals of the New York Academy of Sciences955, 75–85, discussion 86–78, 396–406 (2002).
    1. Li X, et al. COUP-TFII Regulates Human Endometrial Stromal Genes Involved in Inflammation. Mol Endocrinol. 2013;27:2041–2054. doi: 10.1210/me.2013-1191.
    1. Jackson, K. S. et al. The altered distribution of the steroid hormone receptors and the chaperone immunophilin FKBP52 in a baboon model of endometriosis is associated with progesterone resistance during the window of uterine receptivity. Reprod Sci14, 137–150, doi:14/2/137 [pii] 10.1177/1933719106298409 (2007).
    1. Heard ME, Simmons CD, Simmen FA, Simmen RC. Kruppel-like factor 9 deficiency in uterine endometrial cells promotes ectopic lesion establishment associated with activated notch and hedgehog signaling in a mouse model of endometriosis. Endocrinology. 2014;155:1532–1546. doi: 10.1210/en.2013-1947.
    1. Jeong JW, et al. Mig-6 modulates uterine steroid hormone responsiveness and exhibits altered expression in endometrial disease. Proceedings of the National Academy of Sciences of the United States of America. 2009;106:8677–8682. doi: 10.1073/pnas.0903632106.
    1. Aghajanova, L., Velarde, M. C. & Giudice, L. C. The progesterone receptor coactivator Hic-5 is involved in the pathophysiology of endometriosis. Endocrinology150, 3863–3870, doi:en.2009-0008 [pii] 10.1210/en.2009-0008 (2009).
    1. Igarashi TM, et al. Reduced expression of progesterone receptor-B in the endometrium of women with endometriosis and in cocultures of endometrial cells exposed to 2,3,7,8-tetrachlorodibenzo-p-dioxin. Fertility and sterility. 2005;84:67–74. doi: 10.1016/j.fertnstert.2005.01.113.
    1. Large MJ, DeMayo FJ. The regulation of embryo implantation and endometrial decidualization by progesterone receptor signaling. Molecular and cellular endocrinology. 2012;358:155–165. doi: 10.1016/j.mce.2011.07.027.
    1. Wei Q, Levens ED, Stefansson L, Nieman LK. Indian Hedgehog and its targets in human endometrium: menstrual cycle expression and response to CDB-2914. The Journal of clinical endocrinology and metabolism. 2010;95:5330–5337. doi: 10.1210/jc.2010-0637.
    1. Diehl SA, Schmidlin H, Nagasawa M, Blom B, Spits H. IL-6 triggers IL-21 production by human CD4+ T cells to drive STAT3-dependent plasma cell differentiation in B cells. Immunology and cell biology. 2012;90:802–811. doi: 10.1038/icb.2012.17.
    1. Walker SR, et al. Reciprocal effects of STAT5 and STAT3 in breast cancer. Molecular cancer research: MCR. 2009;7:966–976. doi: 10.1158/1541-7786.MCR-08-0238.
    1. Maruyama T, Yoshimura Y. Molecular and cellular mechanisms for differentiation and regeneration of the uterine endometrium. Endocrine journal. 2008;55:795–810. doi: 10.1507/endocrj.K08E-067.
    1. Shen T, et al. SIRT1 Functions as an Important Regulator of Estrogen-Mediated Cardiomyocyte Protection in Angiotensin II-Induced Heart Hypertrophy. Oxidative medicine and cellular longevity. 2014;2014:713894. doi: 10.1155/2014/713894.
    1. Yamakuchi M. MicroRNA Regulation of SIRT1. Frontiers in physiology. 2012;3:68. doi: 10.3389/fphys.2012.00068.
    1. Yamakuchi M, Lowenstein CJ. MiR-34, SIRT1 and p53: the feedback loop. Cell Cycle. 2009;8:712–715. doi: 10.4161/cc.8.5.7753.
    1. Olivo-Marston SE, et al. Effects of calorie restriction and diet-induced obesity on murine colon carcinogenesis, growth and inflammatory factors, and microRNA expression. PloS one. 2014;9:e94765. doi: 10.1371/journal.pone.0094765.
    1. Hirota Y, Cha J, Dey SK. Revisiting reproduction: Prematurity and the puzzle of progesterone resistance. Nature medicine. 2010;16:529–531. doi: 10.1038/nm0510-529.
    1. Fox C, Morin S, Jeong JW, Scott RT, Jr., Lessey BA. Local and systemic factors and implantation: what is the evidence? Fertility and sterility. 2016;105:873–884. doi: 10.1016/j.fertnstert.2016.02.018.
    1. Yoo JY, et al. CRISPLD2 is a target of progesterone receptor and its expression is decreased in women with endometriosis. PLoS One. 2014;9:e100481. doi: 10.1371/journal.pone.0100481.
    1. Kim TH, et al. ARID1A Is Essential for Endometrial Function during Early Pregnancy. PLoS Genet. 2015;11:e1005537. doi: 10.1371/journal.pgen.1005537.
    1. Afshar Y, et al. Changes in eutopic endometrial gene expression during the progression of experimental endometriosis in the baboon, Papio anubis. Biology of reproduction. 2013;88:44. doi: 10.1095/biolreprod.112.104497.
    1. Kim TH, et al. Mig-6 suppresses endometrial cancer associated with Pten deficiency and ERK activation. Cancer research. 2014;74:7371–7382. doi: 10.1158/0008-5472.CAN-14-0794.
    1. Ishibashi H, et al. Sex steroid hormone receptors in human thymoma. J Clin Endocrinol Metab. 2003;88:2309–2317. doi: 10.1210/jc.2002-021353.

Source: PubMed

3
Iratkozz fel