Growth hormone ameliorates the age-associated depletion of ovarian reserve and decline of oocyte quality via inhibiting the activation of Fos and Jun signaling

Chuanming Liu, Shiyuan Li, Yifan Li, Jiao Tian, Xiaoling Sun, Tianran Song, Guijun Yan, Lijun Ding, Haixiang Sun, Chuanming Liu, Shiyuan Li, Yifan Li, Jiao Tian, Xiaoling Sun, Tianran Song, Guijun Yan, Lijun Ding, Haixiang Sun

Abstract

Oocyte quality typically begins to decline with aging, which contributes to subfertility and infertility. However, there is still no effective treatment to restore the ovarian reserve and improve aged-oocyte quality. According to the present study, growth hormone (GH) secretion changes with maternal age in female mice. After intraperitoneal injection with GH (1 mg/kg body weight) every two days for two months, the 10-month-old mice showed a better ovarian reserve and oocyte quality than control mice. GH treatment decreased the occurrence rate of aneuploidy caused by spindle/chromosome defects. Additionally, the single oocyte transcriptome analysis indicated that GH decreased the expression of apoptosis-related genes in oocytes. It was also observed that GH treatment reduced the expression of γH2AX and apoptosis of aged oocytes via decreasing the activation of Fos and Jun. Collectively, our results indicate that GH treatment is an effective way to reverse the age-associated depletion of ovarian reserve and the decline of oocyte quality by decreasing apoptosis.

Keywords: aging; apoptosis; growth hormone; meiosis; oocyte quality.

Conflict of interest statement

CONFLICTS OF INTEREST: The authors declare that they have no conflicts of interest.

Figures

Figure 1
Figure 1
The GH levels and oocyte quality were declined in aged mice. (A) The GH levels in the peripheral blood were measured in young (n = 6) and aged (n = 6) mice. (B) Micrographs of young and aged WT mouse ovaries (Scale bar, 1 mm) and HE staining of these ovaries (Scale bar, 100μm, 500 μm). (C) Follicle counts in 6-week (n = 3) and 10-month (n = 3) old WT mice. (D) Chromosomes misalignment and spindle defects (arrowheads) in aged oocytes. The oocytes were stained with α-tubulin (green) and propidium iodide (PI) (red) respectively. Scale bar, 50 μm. (E) Left: The rate of GVBD and Pb1 extrusion were recorded after 4 h and 14 h of culture in M2 medium respectively. Right: Percentages of oocytes with spindle defects in young (n = 97) and aged mice (n = 104). (F) Representative images of MII oocytes collected from young (n = 76) and aged (n = 19) mice and IVF outcomes from these two groups. Black arrowheads point to abnormal oocytes. Scale bar, 100 μm, 400 μm. (G) Quantification of MII oocytes, 2-cell embryo and blastocyst from young and aged mice. Data are presented as mean ± SD. *P < 0.05, **P < 0.01, ****P < 0.0005.
Figure 2
Figure 2
Effects of GH treatment in vitro on the meiotic progress of aged oocytes. (A) Representative images of oocytes after 14 hours cultured in M2 medium (n = 87) and GH treated medium (n = 88). Black arrowheads point to oocytes that fail to extrude a polar body or fail to survive. Scale bar, 100 μm. (B) The rate of GVBD and Pb1 extrusion in these two groups respectively. (C) Left: Spindle morphologies in control and GH group. Oocytes were stained with α-tubulin (green) and hoechst (blue). Scale bar, 50 μm. Right: Percentages of oocytes with spindle defects in control (n = 51) and GH group (n = 68). Data are presented as mean ± SD. **P < 0.01.
Figure 3
Figure 3
Effects of GH administration in vivo on the ovarian reserve. (A) Schematic illustration for the NS and GH-treated mice. (B) The GH levels in the peripheral blood was measured in GH (n = 6) and vehicle (n = 7) group. (C) The changes of body weight and ovary index after GH administration. NS, no significance. (D) Micrographs of NS-treated and GH-treated mouse ovaries. Scale bar, 1 mm. (E) Left: Estrous cycle in representative females. Right: Average numbers of cycles in 8 days in two groups. P, proestrus; E, estrus; M, metestrus; D, diestrus. (F) HE-stained of NS-treated and GH-treated mouse ovaries. Scale bar, 400 μm, 200 μm. (G) Follicle counts and the number of corpus luteum in NS-treated (n = 3) and GH-treated (n = 5) mice. Data are presented as mean ± SD. *P < 0.05, **P < 0.01.
Figure 4
Figure 4
Effects of GH treatment in vivo on the quality and meiotic progress of aged oocytes. (A) Left: Representative images of MII oocytes, morula and blastocysts from NS-treated and GH-treated mice. Black arrows point to fragmented MII oocytes. Scale bar, 200 μm, 100 μm. Right: Number of MII oocytes and percentage of 2-cell embryos and blastocysts in NS-treated and GH-treated mice. (B) After cultured in M2 medium, the rate of GVBD and Pb1 extrusion were recorded. (C) Left: The MII oocytes from NS-treated (n = 37) and GH-treated (n = 42) mice were stained with α-tubulin (green) and propidium iodide (PI) (red). Scale bar, 50 μm. Right: Quantification of NS-treated and GH-treated oocytes with abnormal spindle/chromosomes. Data are presented as mean ± SD. *P < 0.05, **P < 0.01, ***P < 0.001.
Figure 5
Figure 5
ScRNA-seq of NS-treated and GH-treated MII oocytes. (A) Left: Mapping rate of NS-treated oocytes (n = 4) and GH-treated (n = 4) oocytes. Right: The number of detected genes (FPKM > 1) in vehicle and GH group oocytes. NS, no significance. (B) Left: The gene expression heatmap showed the differentially expressed genes (DEGs) in these two groups. Right: Principal components analysis (PCA) of eight samples. (C) The volcano map showed the DEGs between NS-treated and GH-treated mice. (D) The top 30 KEGG pathways involved in the down-regulated genes. The red box encloses the apoptosis pathway. (E) The top 22 KEGG pathways involved in the up-regulated genes. The red box encloses the GnRH signaling, cAMP signaling, calcium signaling and Rap1 signaling pathway. (F) 2 DEGs were selected for QPCR validation (n ≥ 10). Data are presented as mean ± SD. *P < 0.05, **P < 0.01.
Figure 6
Figure 6
GH treatment decreased oocyte apoptosis and DNA damage. (A) Left: Immunostaining for Ki-67 in ovaries from the NS and GH groups. Brown represents positive staining. Scale bar, 400, 100, 50 μm. Right: The mean density levels of Ki-67 in ovarian. (B) The ovaries from NS-treated and GH-treated mice were stained with γH2AX (red) and DAPI (blue). Scale bar, 50 μm. (C) Left: The MII oocytes from NS-treated (n = 7) and GH-treated (n=7) mice were stained with γH2AX (red) and hoechst (blue). Scale bar, 50 μm. Right: Mean density levels of NS-treated and GH-treated oocytes. Data are presented as mean ± SD. *P < 0.05, **P < 0.01.
Figure 7
Figure 7
Effect of GH treatment on the FOS/JUN pathway. (A) The expression of GHR in GH-treated and NS-treated oocytes. (B) The expression of Fos, Fosb, Jun, Junb, Jund, Lmnb2 and Ctsb in GH-treated and NS-treated oocytes according to the sequencing data. (C) Results of single oocyte QPCR in the two groups (n ≥ 10). (D) Immunostaining for Fos, Jun, Fosb and Junb in ovaries from the NS and GH groups. Brown represents positive staining. Data are presented as mean ± SD. *P < 0.05.
Figure 8
Figure 8
Schematic diagram regarding how GH reverses the age-associated depletion of ovarian reserve and declining quality of oocytes. In old mice, the ovarian reserve and the levels of GH are declined. The decreased GH enhances the expression levels of Fos and Jun family which increase the oocyte apoptosis and DNA damage. Administration of GH restores the depletion of ovarian reserve and induces the increment of GHRs. Additionally, GH supplementation exerts an influence on reversing apoptosis via lowering the expression of Fos and Jun which improves the oocyte quality.

References

    1. Cimadomo D, Fabozzi G, Vaiarelli A, Ubaldi N, Ubaldi FM, Rienzi L. Impact of maternal age on oocyte and embryo competence. Front Endocrinol (Lausanne). 2018; 9:327. 10.3389/fendo.2018.00327
    1. Selesniemi K, Lee HJ, Muhlhauser A, Tilly JL. Prevention of maternal aging-associated oocyte aneuploidy and meiotic spindle defects in mice by dietary and genetic strategies. Proc Natl Acad Sci USA. 2011; 108:12319–24. 10.1073/pnas.1018793108
    1. Battaglia DE, Goodwin P, Klein NA, Soules MR. Influence of maternal age on meiotic spindle assembly in oocytes from naturally cycling women. Hum Reprod. 1996; 11:2217–22. 10.1093/oxfordjournals.humrep.a019080
    1. Kalmbach KH, Fontes Antunes DM, Dracxler RC, Knier TW, Seth-Smith ML, Wang F, Liu L, Keefe DL. Telomeres and human reproduction. Fertil Steril. 2013; 99:23–29. 10.1016/j.fertnstert.2012.11.039
    1. Ubaldi FM, Cimadomo D, Vaiarelli A, Fabozzi G, Venturella R, Maggiulli R, Mazzilli R, Ferrero S, Palagiano A, Rienzi L. Advanced maternal age in IVF: still a challenge? the present and the future of its treatment. Front Endocrinol (Lausanne). 2019; 10:94. 10.3389/fendo.2019.00094
    1. Møller N, Jørgensen JO. Effects of growth hormone on glucose, lipid, and protein metabolism in human subjects. Endocr Rev. 2009; 30:152–77. 10.1210/er.2008-0027
    1. Bidlingmaier M, Strasburger CJ. Growth hormone. Handb Exp Pharmacol. 2010; 195:187–200. 10.1007/978-3-540-79088-4_8
    1. Auernhammer CJ, Strasburger CJ. Effects of growth hormone and insulin-like growth factor I on the immune system. Eur J Endocrinol. 1995; 133:635–45. 10.1530/eje.0.1330635
    1. Giustina A, Veldhuis JD. Pathophysiology of the neuroregulation of growth hormone secretion in experimental animals and the human. Endocr Rev. 1998; 19:717–97. 10.1210/edrv.19.6.0353
    1. Ashpole NM, Sanders JE, Hodges EL, Yan H, Sonntag WE. Growth hormone, insulin-like growth factor-1 and the aging brain. Exp Gerontol. 2015; 68:76–81. 10.1016/j.exger.2014.10.002
    1. Takahashi Y. The role of growth hormone and insulin-like growth factor-I in the liver. Int J Mol Sci. 2017; 18:1447. 10.3390/ijms18071447
    1. Chikani V, Ho KK. Action of GH on skeletal muscle function: molecular and metabolic mechanisms. J Mol Endocrinol. 2013; 52:R107–23. 10.1530/JME-13-0208
    1. Forman K, Vara E, García C, Kireev R, Cuesta S, Acuña-Castroviejo D, Tresguerres JA. Influence of aging and growth hormone on different members of the NFkB family and IkB expression in the heart from a murine model of senescence-accelerated aging. Exp Gerontol. 2016; 73:114–20. 10.1016/j.exger.2015.11.005
    1. Abir R, Garor R, Felz C, Nitke S, Krissi H, Fisch B. Growth hormone and its receptor in human ovaries from fetuses and adults. Fertil Steril. 2008; 90:1333–39. 10.1016/j.fertnstert.2007.08.011
    1. Spiliotis BE. Growth hormone insufficiency and its impact on ovarian function. Ann N Y Acad Sci. 2003; 997:77–84. 10.1196/annals.1290.009
    1. Hull KL, Harvey S. Growth hormone and reproduction: a review of endocrine and autocrine/paracrine interactions. Int J Endocrinol. 2014; 2014:234014. 10.1155/2014/234014
    1. Devesa J, Caicedo D. The role of growth hormone on ovarian functioning and ovarian angiogenesis. Front Endocrinol (Lausanne). 2019; 10:450. 10.3389/fendo.2019.00450
    1. Sonntag WE, Carter CS, Ikeno Y, Ekenstedt K, Carlson CS, Loeser RF, Chakrabarty S, Lee S, Bennett C, Ingram R, Moore T, Ramsey M. Adult-onset growth hormone and insulin-like growth factor I deficiency reduces neoplastic disease, modifies age-related pathology, and increases life span. Endocrinology. 2005; 146:2920–32. 10.1210/en.2005-0058
    1. Homburg R, Eshel A, Abdalla HI, Jacobs HS. Growth hormone facilitates ovulation induction by gonadotrophins. Clin Endocrinol (Oxf). 1988; 29:113–17. 10.1111/j.1365-2265.1988.tb00252.x
    1. Xu YM, Hao GM, Gao BL. Application of Growth Hormone in in vitro Fertilization. Front Endocrinol (Lausanne). 2019; 10:502. 10.3389/fendo.2019.00502
    1. Li XL, Wang L, Lv F, Huang XM, Wang LP, Pan Y, Zhang XM. The influence of different growth hormone addition protocols to poor ovarian responders on clinical outcomes in controlled ovary stimulation cycles: a systematic review and meta-analysis. Medicine (Baltimore). 2017; 96:e6443. 10.1097/MD.0000000000006443
    1. Regan SL, Knight PG, Yovich JL, Arfuso F, Dharmarajan A. Growth hormone during in vitro fertilization in older women modulates the density of receptors in granulosa cells, with improved pregnancy outcomes. Fertil Steril. 2018; 110:1298–310. 10.1016/j.fertnstert.2018.08.018
    1. Norman RJ, Alvino H, Hull LM, Mol BW, Hart RJ, Kelly TL, Rombauts L, and LIGHT investigators. Human growth hormone for poor responders: a randomized placebo-controlled trial provides no evidence for improved live birth rate. Reprod Biomed Online. 2019; 38:908–15. 10.1016/j.rbmo.2019.02.003
    1. Magon N, Agrawal S, Malik S, Babu KM. Growth hormone in the management of female infertility. Indian J Endocrinol Metab. 2011. (Suppl 3); 15:S246–47. 10.4103/2230-8210.84876
    1. Wang T, Gao YY, Chen L, Nie ZW, Cheng W, Liu X, Schatten H, Zhang X, Miao YL. Melatonin prevents postovulatory oocyte aging and promotes subsequent embryonic development in the pig. Aging (Albany NY). 2017; 9:1552–64. 10.18632/aging.101252
    1. Zhou J, Xue Z, He HN, Liu X, Yin SY, Wu DY, Zhang X, Schatten H, Miao YL. Resveratrol delays postovulatory aging of mouse oocytes through activating mitophagy. Aging (Albany NY). 2019; 11:11504–19. 10.18632/aging.102551
    1. Li C, He X, Huang Z, Han L, Wu X, Li L, Xin Y, Ge J, Sha J, Yin Z, Wang Q. Melatonin ameliorates the advanced maternal age-associated meiotic defects in oocytes through the SIRT2-dependent H4K16 deacetylation pathway. Aging (Albany NY). 2020; 12:1610–23. 10.18632/aging.102703
    1. Zhang M, Lu Y, Chen Y, Zhang Y, Xiong B. Insufficiency of melatonin in follicular fluid is a reversible cause for advanced maternal age-related aneuploidy in oocytes. Redox Biol. 2020; 28:101327. 10.1016/j.redox.2019.101327
    1. Miao Y, Cui Z, Gao Q, Rui R, Xiong B. Nicotinamide mononucleotide supplementation reverses the declining quality of maternally aged oocytes. Cell Rep. 2020; 32:107987. 10.1016/j.celrep.2020.107987
    1. Bertoldo MJ, Listijono DR, Ho WJ, Riepsamen AH, Goss DM, Richani D, Jin XL, Mahbub S, Campbell JM, Habibalahi A, Loh WN, Youngson NA, Maniam J, et al.. NAD+ repletion rescues female fertility during reproductive aging. Cell Rep. 2020; 30:1670–81.e7. 10.1016/j.celrep.2020.01.058
    1. Hou HY, Wang X, Yu Q, Li HY, Li SJ, Tang RY, Guo ZX, Chen YQ, Hu CX, Yang ZJ, Zhang WK, Qin Y. Evidence that growth hormone can improve mitochondrial function in oocytes from aged mice. Reproduction. 2018; 157:345–58. 10.1530/REP-18-0529
    1. Li Y, Liu H, Yu Q, Liu H, Huang T, Zhao S, Ma J, Zhao H. Growth Hormone Promotes in vitro Maturation of Human Oocytes. Front Endocrinol (Lausanne). 2019; 10:485. 10.3389/fendo.2019.00485
    1. Izadyar F, Hage WJ, Colenbrander B, Bevers MM. The promotory effect of growth hormone on the developmental competence of in vitro matured bovine oocytes is due to improved cytoplasmic maturation. Mol Reprod Dev. 1998; 49:444–53. 10.1002/(SICI)1098-2795(199804)49:4<444::AID-MRD12>;2-U
    1. Apa R, Lanzone A, Miceli F, Mastrandrea M, Caruso A, Mancuso S, Canipari R. Growth hormone induces in vitro maturation of follicle- and cumulus-enclosed rat oocytes. Mol Cell Endocrinol. 1994; 106:207–12. 10.1016/0303-7207(94)90204-6
    1. Shirazi A, Shams-Esfandabadi N, Ahmadi E, Heidari B. Effects of growth hormone on nuclear maturation of ovine oocytes and subsequent embryo development. Reprod Domest Anim. 2010; 45:530–36. 10.1111/j.1439-0531.2008.01290.x
    1. Tesarik J, Hazout A, Mendoza C. Improvement of delivery and live birth rates after ICSI in women aged >40 years by ovarian co-stimulation with growth hormone. Hum Reprod. 2005; 20:2536–41. 10.1093/humrep/dei066
    1. Kolibianakis EM, Venetis CA, Diedrich K, Tarlatzis BC, Griesinger G. Addition of growth hormone to gonadotrophins in ovarian stimulation of poor responders treated by in-vitro fertilization: a systematic review and meta-analysis. Hum Reprod Update. 2009; 15:613–22. 10.1093/humupd/dmp026
    1. Bassiouny YA, Dakhly DM, Bayoumi YA, Hashish NM. Does the addition of growth hormone to the in vitro fertilization/intracytoplasmic sperm injection antagonist protocol improve outcomes in poor responders? A randomized, controlled trial. Fertil Steril. 2016; 105:697–702. 10.1016/j.fertnstert.2015.11.026
    1. Cui N, Li AM, Luo ZY, Zhao ZM, Xu YM, Zhang J, Yang AM, Wang LL, Hao GM, Gao BL. Effects of growth hormone on pregnancy rates of patients with thin endometrium. J Endocrinol Invest. 2019; 42:27–35. 10.1007/s40618-018-0877-1.
    1. van den Eijnden MJ, Strous GJ. Autocrine growth hormone: effects on growth hormone receptor trafficking and signaling. Mol Endocrinol. 2007; 21:2832–46. 10.1210/me.2007-0092
    1. Minoia M, Gentilin E, Molè D, Rossi M, Filieri C, Tagliati F, Baroni A, Ambrosio MR, degli Uberti E, Zatelli MC. Growth hormone receptor blockade inhibits growth hormone-induced chemoresistance by restoring cytotoxic-induced apoptosis in breast cancer cells independently of estrogen receptor expression. J Clin Endocrinol Metab. 2012; 97:E907–16. 10.1210/jc.2011-3340
    1. Weston CR, Davis RJ. The JNK signal transduction pathway. Curr Opin Cell Biol. 2007; 19:142–49. 10.1016/j.ceb.2007.02.001
    1. Angel P, Karin M. The role of Jun, Fos and the AP-1 complex in cell-proliferation and transformation. Biochim Biophys Acta. 1991; 1072:129–57. 10.1016/0304-419x(91)90011-9
    1. Ham J, Eilers A, Whitfield J, Neame SJ, Shah B. c-Jun and the transcriptional control of neuronal apoptosis. Biochem Pharmacol. 2000; 60:1015–21. 10.1016/s0006-2952(00)00372-5
    1. Karin M, Liu Zg, Zandi E. AP-1 function and regulation. Curr Opin Cell Biol. 1997; 9:240–46. 10.1016/s0955-0674(97)80068-3
    1. Colotta F, Polentarutti N, Sironi M, Mantovani A. Expression and involvement of c-fos and c-jun protooncogenes in programmed cell death induced by growth factor deprivation in lymphoid cell lines. J Biol Chem. 1992; 267:18278–83.
    1. Liebermann DA, Gregory B, Hoffman B. AP-1 (Fos/Jun) transcription factors in hematopoietic differentiation and apoptosis. Int J Oncol. 1998; 12:685–700. 10.3892/ijo.12.3.685
    1. Rybak P, Hoang A, Bujnowicz L, Bernas T, Berniak K, Zarębski M, Darzynkiewicz Z, Dobrucki J. Low level phosphorylation of histone H2AX on serine 139 (γH2AX) is not associated with DNA double-strand breaks. Oncotarget. 2016; 7:49574–87. 10.18632/oncotarget.10411
    1. Pospelova TV, Demidenko ZN, Bukreeva EI, Pospelov VA, Gudkov AV, Blagosklonny MV. Pseudo-DNA damage response in senescent cells. Cell Cycle. 2009; 8:4112–18. 10.4161/cc.8.24.10215

Source: PubMed

3
Subscribe