Impaired steroidogenesis and implantation failure in Bmal1-/- mice

Christine K Ratajczak, Katie L Boehle, Louis J Muglia, Christine K Ratajczak, Katie L Boehle, Louis J Muglia

Abstract

Evidence in humans and rodents suggests that normal circadian rhythmicity is important for supporting reproductive function. A molecular clock underlies circadian rhythmicity. Impaired fertility is observed in some genetically altered mice with deficiencies in genes of the molecular clock, suggesting a critical role for these genes in reproduction. Here we systematically characterize the reproductive phenotype of females deficient in the clock gene Bmal1. Bmal1(-/-) females are infertile. They exhibit progression through the estrous cycle, although these cycles are prolonged. Normal follicular development occurs in Bmal1(-/-) females, and healthy embryos of the expected developmental stage are found in the reproductive tract of Bmal1(-/-) females 3.5 d after mating to wild-type males. However, serum progesterone levels are significantly lower in Bmal1(-/-) vs. Bmal1(+/+/-) females on d 3.5 of gestation. Low progesterone levels in Bmal1(-/-) females are accompanied by decreased expression of steroidogenic acute regulatory protein in corpora lutea of Bmal1(-/-) vs. Bmal1(+/+/-) females. Whereas implantation of embryos is not observed in untreated or vehicle-treated Bmal1(-/-) females, exogenous administration of progesterone to Bmal1(-/-) females is able to reinstitute implantation. These data suggest that implantation failure due to impaired steroidogenesis causes infertility of Bmal1(-/-) females.

Figures

F ig . 1.
Fig. 1.
Prolonged estrous cycles in Bmal1−/− females. Estrous cycle was assessed by histological analysis of vaginal smears. Progression through the stages of the estrous cycle in representative Bmal1+/+ (A) and Bmal1−/− (B) females is shown. Mean cycle length (C) and proportion of time spent in each estrous stage (D) (n = 5 for each group) is also shown. Values are means ± sem. *, P < 0.001.
F ig . 2.
Fig. 2.
Normal follicular development in Bmal1−/− females. Follicles were counted on sections of hematoxylin and eosin-stained ovary. A, Normal preantral follicles (indicated by arrows), antral follicles (indicated by arrows), and corpora lutea inBmal1+/± and Bmal1−/− ovaries. Black bars, 50 μm. B, Abundance of follicles in different stages of development. Values are means ± sem (n = 4,Bmal1+/±; n = 3, Bmal1−/−). PF, Primordial and primary follicles; PrF, preantral follicles; APrF, atretic preantral follicles; AnF, antral follicles; AAnF, atretic antral follicles; CL, corpora lutea.
F ig . 3.
Fig. 3.
Healthy preimplantation embryos but low serum progesterone levels inBmal1−/− females at d 3.5 of gestation. Oocytes and embryos were flushed from the reproductive tract at d 3.5 of gestation and analyzed under a light microscope. Serum progesterone and estradiol at d 3.5 of gestation were assayed by solid-phase 125I-RIA. A, Average number of oocytes/embryos isolated from each gravid female. Values are means ± sem. Numbers in bars indicate the number of females with oocytes or embryos/total number of females analyzed. B, Percentage of embryos isolated identified as healthy, unfertilized, and degenerating. C, Percentage of healthy embryos isolated in the blastocyst or morula stage. D, Serum progesterone at ZT 4 on d 3.5 (n = 14,Bmal1+/±; n = 15, Bmal1−/−). Values are means ± sem. *, P < 0.0001. E, Circadian serum progesterone throughout d 3.5 (n = 3, Bmal1+/±; n = 3–4,Bmal1−/− per time point). Values are means ± sem (P < 0.0001 with respect to genotype). F, Serum estradiol (n = 12, Bmal1+/±; n = 15,Bmal1−/−). Values are means ± sem.
F ig . 4.
Fig. 4.
Bmal1 and StAR are expressed in corpora lutea of d 3.5 Bmal1+/± females. Ovary sections were subjected toin situ hybridization with radiolabeled antisenseBmal1 or StAR probes and then hematoxylin and eosin stained for histological analysis. A, Autoradiograph from in situ hybridization for Bmal1 at ZT 4 d 3.5. B, Hematoxylin and eosin staining of the same section. Arrows point to corpora lutea. Black bar, 200 μm. C and E, Representative autoradiographs from in situ hybridization for StAR at ZT 4 d 3.5. D and F, Hematoxylin and eosin staining of the same sections. Arrowspoint to corpora lutea. Black bars, 200 μm (n = 3,Bmal1+/±; n = 3, Bmal1−/−). G, Circadian StAR expression throughout d 3.5. Values are means ± sem (n = 3, Bmal1+/±; n = 3–7,Bmal1−/− per time point). +, No corpora lutea observed in any mice examined at this time point (P < 0.0001 with respect to genotype).
F ig . 5.
Fig. 5.
Implantation sites found in progesterone-supplementedBmal1−/− females are small but histologically normal. Paraffin sections of implantation sites were hematoxylin and eosin stained. Diameter of whole implantation sites was measured under a light microscope. Representative implantation sites from untreated Bmal1+/± females (A), vehicle-treated Bmal1+/± females (B), progesterone-treatedBmal1+/± females (C), and progesterone-treatedBmal1−/− females (D) are shown. Black bar, 1 mm. Em, Embryo; Pl, placenta; FM, fetal membranes; Ut, uterus. E, Implantation site diameter (n = 3 implantation sites from each of three to four females per group). *, P < 0.001 compared with each of the other groups. P4, Progesterone.

References

    1. Dunson DB, Baird DD, Colombo B 2004. Increased infertility with age in men and women. Obstet Gynecol 103:51–56
    1. Labyak S, Lava S, Turek F, Zee P 2002. Effects of shiftwork on sleep and menstrual function in nurses. Health Care Women Int 23:703–714
    1. Bisanti L, Olsen J, Basso O, Thonneau P, Karmaus W 1996. Shift work and subfecundity: a European multicenter study. European Study Group on Infertility and Subfecundity. J Occup Environ Med 38:352–358
    1. Knutsson A 2003. Health disorders of shift workers. Occup Med (Lond) 53:103–108
    1. Reppert SM, Weaver DR 2002. Coordination of circadian timing in mammals. Nature 418:935–941
    1. Dolatshad H, Campbell EA, O'Hara L, Maywood ES, Hastings MH, Johnson MH 2006. Developmental and reproductive performance in circadian mutant mice. Hum Reprod 21:68–79
    1. Kennaway DJ, Varcoe TJ, Mau VJ 2003. Rhythmic expression of clock and clock-controlled genes in the rat oviduct. Mol Hum Reprod 9:503–507
    1. Karman BN, Tischkau SA 2006. Circadian clock gene expression in the ovary: effects of luteinizing hormone. Biol Reprod 75:624–632
    1. Chappell PE, White RS, Mellon PL 2003. Circadian gene expression regulates pulsatile gonadotropin-releasing hormone (GnRH) secretory patterns in the hypothalamic GnRH-secreting GT1–7 cell line. J Neurosci 23:11202–11213
    1. Mosko SS, Moore RY 1979. Neonatal suprachiasmatic nucleus lesions: effects on the development of circadian rhythms in the rat. Brain Res 164:17–38
    1. Palm IF, Van Der Beek EM, Wiegant VM, Buijs RM, Kalsbeek A 1999. Vasopressin induces a luteinizing hormone surge in ovariectomized, estradiol-treated rats with lesions of the suprachiasmatic nucleus. Neuroscience 93:659–666
    1. Brown-Grant K, Raisman G 1977. Abnormalities in reproductive function associated with the destruction of the suprachiasmatic nuclei in female rats. Proc R Soc Lond B Biol Sci 198:279–296
    1. Herzog ED, Grace MS, Harrer C, Williamson J, Shinohara K, Block GD 2000. The role of Clock in the developmental expression of neuropeptides in the suprachiasmatic nucleus. J Comp Neurol 424:86–98
    1. Kennaway DJ, Boden MJ, Voultsios A 2005. Reproductive performance in female Clock(Δ19) mutant mice. Reprod Fertil Dev 16:801–810
    1. Miller BH, Olson SL, Turek FW, Levine JE, Horton TH, Takahashi JS 2004. Circadian clock mutation disrupts estrous cyclicity and maintenance of pregnancy. Curr Biol 14:1367–1373
    1. Antoch MP, Song EJ, Chang AM, Vitaterna MH, Zhao Y, Wilsbacher LD, Sangoram AM, King DP, Pinto LH, Takahashi JS 1997. Functional identification of the mouse circadian Clock gene by transgenic BAC rescue. Cell 89:655–667
    1. King DP, Vitaterna MH, Chang AM, Dove WF, Pinto LH, Turek FW, Takahashi JS 1997. The mouse Clock mutation behaves as an antimorph and maps within the W19H deletion, distal of Kit. Genetics 146:1049–1060
    1. King DP, Zhao Y, Sangoram AM, Wilsbacher LD, Tanaka M, Antoch MP, Steeves TD, Vitaterna MH, Kornhauser JM, Lowrey PL, Turek FW, Takahashi JS 1997. Positional cloning of the mouse circadian clock gene. Cell 89:641–653
    1. Vitaterna MH, King DP, Chang AM, Kornhauser JM, Lowrey PL, McDonald JD, Dove WF, Pinto LH, Turek FW, Takahashi JS 1994. Mutagenesis and mapping of a mouse gene, Clock, essential for circadian behavior. Science 264:719–725
    1. Kennaway DJ, Voultsios A, Varcoe TJ, Moyer RW 2003. Melatonin and activity rhythm responses to light pulses in mice with the Clock mutation. Am J Physiol 284:R1231–R1240
    1. Kennaway DJ, Owens JA, Voultsios A, Varcoe TJ 2006. Functional central rhythmicity and light entrainment, but not liver and muscle rhythmicity, are Clock independent. Am J Physiol 291:R1172–R1180
    1. Debruyne JP, Noton E, Lambert CM, Maywood ES, Weaver DR, Reppert SM 2006. A clock shock: mouse CLOCK is not required for circadian oscillator function. Neuron 50:465–477
    1. Bunger MK, Walisser JA, Sullivan R, Manley PA, Moran SM, Kalscheur VL, Colman RJ, Bradfield CA 2005. Progressive arthropathy in mice with a targeted disruption of the Mop3/Bmal-1 locus. Genesis 41:122–132
    1. Bunger MK, Wilsbacher LD, Moran SM, Clendenin C, Radcliffe LA, Hogenesch JB, Simon MC, Takahashi JS, Bradfield CA 2000. Mop3 is an essential component of the master circadian pacemaker in mammals. Cell 103:1009–1017
    1. Kondratov RV, Kondratova AA, Gorbacheva VY, Vykhovanets OV, Antoch MP 2006. Early aging and age-related pathologies in mice deficient in BMAL1, the core component of the circadian clock. Genes Dev 20:1868–1873
    1. Alvarez JD, Hansen A, Ord T, Bebas P, Chappell PE, Giebultowicz JM, Williams C, Moss S, Sehgal A 2008. The circadian clock protein BMAL1 is necessary for fertility and proper testosterone production in mice. J Biol Rhythms 23:26–36
    1. Boden MJ, Kennaway DJ 2006. Circadian rhythms and reproduction. Reproduction 132:379–392
    1. Kennaway DJ 2005. The role of circadian rhythmicity in reproduction. Hum Reprod Update 11:91–101
    1. Lee KY, Jeong JW, Wang J, Ma L, Martin JF, Tsai SY, Lydon JP, DeMayo FJ 2007. Bmp2 is critical for the murine uterine decidual response. Mol Cell Biol 27:5468–5478
    1. Allen E 1922. The oestrus cycle in the mouse. Am J Anat 30:297–348
    1. Pedersen T, Peters H 1968. Proposal for a classification of oocytes and follicles in the mouse ovary. J Reprod Fertil 17:555–557
    1. Pangas SA, Li X, Robertson EJ, Matzuk MM 2006. Premature luteinization and cumulus cell defects in ovarian-specific Smad4 knockout mice. Mol Endocrinol 20:1406–1422
    1. Hogan B, Beddington R, Constantini F, Lacy E 1994. Manipulating the mouse embryo: a laboratory manual. Plainview, NY: Cold Spring Harbor Laboratory Press
    1. Gross GA, Imamura T, Luedke C, Vogt SK, Olson LM, Nelson DM, Sadovsky Y, Muglia LJ 1998. Opposing actions of prostaglandins and oxytocin determine the onset of murine labor. Proc Natl Acad Sci USA 95:11875–11879
    1. Mann R, Keri R, Nilson J 1999. Transgenic mice with chronically elevated luteinizing hormone are infertile due to anovulation, defects in uterine receptivity, and midgestation pregnancy failure. Endocrinology 140:2592–2601
    1. Wang H, Dey SK 2006. Roadmap to embryo implantation: clues from mouse models. Nat Rev Genet 7:185–199
    1. de la Iglesia H, Schwartz W 2006. Minireview: timely ovulation: circadian regulation of the female hypothalamo-pituitary-gonadal axis. Endocrinology 147:1148–1153
    1. Ma L, Benson G, Lim H, Dey S, Maas R 1998. Abdominal B (AbdB) Hoxa genes: regulation in adult uterus by estrogen and progesterone and repression in müllerian duct by the synthetic estrogen diethylstilbestrol (DES). Dev Biol 197:141–154
    1. Hasegawa T, Zhao L, Caron KM, Majdic G, Suzuki T, Shizawa S, Sasano H, Parker KL 2000. Developmental roles of the steroidogenic acute regulatory protein (StAR) as revealed by StAR knockout mice. Mol Endocrinol 14:1462–1471
    1. Nakao N, Yasuo S, Nishimura A, Yamamura T, Watanabe T, Anraku T, Okano T, Fukada Y, Sharp PJ, Ebihara S, Yoshimura T 2007. Circadian clock gene regulation of steroidogenic acute regulatory protein gene expression in preovulatory ovarian follicles. Endocrinology 148:3031–3038

Source: PubMed

3
Abonnieren