تاثیر FSH و eCG بر روی تکامل فولیکول‌ها و بیان کاسپاز-9 و FSHR تخمدان در موش سوری

نوع مقاله : مقاله کامل

نویسندگان

چکیده

هدف مطالعه حاضر ارزیابی اثرات FSH و eCG بر روی تکامل تخمدان و فولیکول، سطوح بیان FSHR و کاسپاز-9 تخمدان‌ها در محیط داخل بدن بود. تعداد 105 موش سوری پیش از بلوغ به گروه‌های FSH-1، FSH-2، FSH-3، eCG-1، eCG-2، eCG-3 و گروه کنترل (CG) اختصاص داده شدند. به موش‌ها در گروه‌های FSH-1، FSH-2 و FSH-3 به صورت داخل عضلانی دو بار (روز 0 و 4) به ترتیب با 5، 10 و 20 IU FSH تزریق شدند. موش‌ها در گروه‌های eCG-1، eCG-2 و eCG-3 در روز 0 و 4 به صورت داخل صفاقی با 10، 20 و 40 IU eCG مورد تزریق قرار گرفتند. به موش‌ها در گروه کنترل 5/0 میلی لیتر نرمال سالین در روز 0 و 4 تزریق شد. تخمدان‌های چپ و راست هر موش به صورت استریل به ترتیب در روزهای 7، 14 و 21 تشریح شدند. نتایج نشان دادند که در روز 14 و 21 اندازه تخمدان‌ها و تعداد فولیکول‌های گروه‌های FSH-3 و eCG-3 بیشتر از CG بودند (P<0.05). FSHR mRNA در گروه‌های FSH-2 و eCG-1 در روزهای 14 و 21 بیشتر از CG بودند (P<0.05). پروتین‌های FSHR گروه FSH-3 در روزهای 14 و 21 بیشتر از CG بودند (P<0.05). mRNA کاسپاز-9 در گروه‌های FSH و eCG کمتر از CG بودند. روابط مثبتی بین تعداد فولیکول‌ها و دوزهای FSH و eCG وجود داشت. بیان پروتئین FSHR ارتباط مثبتی بین وزن و اندازه تخمدان و تعداد فولیکول‌ها در موش‌های درمان شده با FSH داشت (r=0.971, P<0.05). غلظت سرمی FSH در گروه‌های FSH-2، FSH-3، eCG-2 و eCG-3 بیشتر از آن در گروه CG بود. در نتیجه، eCG و FSH تکامل تخمدانی، تشکیل فولیکول، ترشح FSH، بیان mRNA و پروتئین FSHR را در تخمدان‌های موش‌های سوری افزایش دادند. FSH و eCG بیان mRNA کاسپاز-9 تخمدان را مهار کردند.

کلیدواژه‌ها


Alejandro, CI; Manuel, XCV; Gustavo, RLC; Román, EC; Alejandro, CJC; Maximino, MM; Rubén, HC; Abel, VM; de Lourdes, JM; Pedro, SA and Eulogio, GL (2012). Effect of cloprostenol and fluorogestone acetate more PMSG on synchronization and no return to estrus in seasonal anestrus Dorper sheep. J. App. Sci. Res., 8: 1612-1614.
Ali, MS; Khandoker, MAM; Afroz, MA and Bhuiyan, AKFH (2012). Ovarian response to different dose levels of follicle stimulating hormone (FSH) in different genotypes of Bangladeshi cattle. Asian-Australas. J. Anim. Sci., 25: 52-58.
Asahara, S; Sato, A; Aljonaid, AA and Maruo, T (2003). Thyroid hormone synergizes with follicle stimulating hormone to inhibit apoptosis in porcine granulosa cells selectively from small follicles. Kobe J. Med. Sci., 49: 107-116.
Cao, J; Zhou, G; Liu, Y; Liao, G; Zhang, Q; Ye, K; Pan, D and Ou, C (2014). Activation of caspase-9 and its influencing factors in beef during conditioning. Animals. 8: 504-509.
Crawford, JL; Heath, DA; Haydon, LJ; Thomson, BP and Eckery, DC (2009). Gene expression and secretion of LH and FSH in relation to gene expression of GnRH receptors in the brushtail possum (Trichosurus vulpecula) demons-trates highly conserved mechanisms. Reproduction. 137: 129-140.
David, EN; Timothy, JP and Gary, CWE (2009). Veterinary reproduction and obstetrics. 9th Edn., Philadelphia, USA, Elsevier Medicine Press. PP: 127-189.
El-Nefiawy Nagwa, E (2011). Effect of exogenous prostag-landin E2 administration on ovarian follicle growth and angiogenesis in rat with reference to pregnant mare serum gonadotrophin. Egypt. J. Histol., 34: 251-259.
Ene, AC; Park, S; Edelmann, W and Taketo, T (2013). Caspase 9 is constitutively activated in mouse oocytes and plays a key role in oocyte elimination during meiotic prophase progression. Dev. Biol., 377: 213-223.
Erickson, GF and Shimasaki, S (2001). The physiology of folliculogenesis: the role of novel growth factors. Fertil. Steril., 76: 943-949.
Hillier, SG (2001). Gonadotropic control of ovarian follicular growth and development. Mol. Cell Endocrinol., 179: 39-46.
Hu, P; Huang, H; Liu, Y; Zhang, J; Lang, H and Zhang, G (2005). Effect of PMSG on the ovarian and uterine Kunming mouse immature and histology. Heilongjiang J. Anim. Reprod. China. 13: 6-7.
Hunzicker-Dunn, M and Maizels, ET (2006). FSH signaling pathways in immature granulosa cells that regulate target gene expression: branching out from protein kinase A. Cell Signal. 18: 1351-1359.
Layman, LC and McDonough, PG (2000). Mutations of follicle stimulating hormone-β and its receptor in human and mouse: genotype/phenotype. Mol. Cell Endocrinol., 161: 9-17.
Li, P; Yue, W; Pang, Y; Yu, X; Huang, Y; Ren, Y and Lv, L (2013). Effects of FSH and insulin on sheep ovarian follicular granulose cells in vitro culture. Acta Vet. et Zoot. Sin., 44: 1386-1391.
Maritza, PM; Jorg, G; Hermanan, MB; Claudia, G; Eberhard, N and Manuela, S (2000). Ovarian response to follicle-stimulating hormone (FSH) stimulation depends on the FSH receptor genotype. J. Clin. Endocrinol. Metabol., 85: 3365-3369.
McStay, GP; Salvesen, GS and Green, DR (2008). Over-
lapping cleavage motif selectivity of caspases: implications for analysis of apoptotic pathways. Cell Death Differ., 15: 322-331.
Menon, KMJ; Anil, K; Nair, LW and Helle, P (2007). Regulation of luteinizing hormone receptor mRNA expression by a specific RNA binding protein in the ovary. Mol. Cell Endocrinol., 260: 109-116.
Miro, F and Hillier, SG (1996). Modulation of granulosa cell deoxyribonucleic acid synthesis and differentiation by activin. Endocrinology. 137: 464-468.
Roberta, NC; Ana, BG; Duarte, G; Rodrigues, Q; Juliana, JHC; Gerlane, MS and Claudio, AP (2012). Effects of insulin and follicle-simulating hormone (FSH) during in vitro development of ovarian: goat preantral follicles and the relative mRNA expression for insulin and FSH receptors and cytochrome P450 aromatase in cultured follicles. Biol. Reprod., 87: 321-329.
Roy, SK and Albee, L (2000). Requirement for follicle-stimulating hormone action in the formation of primordial follicles during perinatal ovarian development in the hamster. Endocrinology. 114: 4449-4456.
Seekallu, SV; Toosi, BM; Duggavathi, R; Barrett, DMW; Davies, KL; Waldner, C and Rawlings, NC (2010). Ovarian antral follicular dynamics in sheep revisited: comparison among estrous cycles with three or four follicular waves. Theriogenology. 73: 670-680.
Susan, MJ and Roy, S (2000). Effects of exogenous FSH on follicular recruitment in a viviparous lizard Niveoscincus metallicus (Scincidae). Comparative biochemistry and physiology. Part A: Mol. Integrat. Physiol., 127: 487-493.
Thomas, FH and Vanderhyden, BC (2003). Oocyte granulosa cell interactions during mouse follicular development: regulation of kit ligand expression and its role in oocyte growth. Reprod. Biol. Endocrinol., 1: 1-7.
Wei, S; Chen, S; Gong, Z; Ouyang, X; An, L; Xie, K; Dong, J and Wei, M (2013a). Alarelin active immunization influences expression levels of GnRHR, FSHR and LHR proteins in the ovary and enhances follicular development in ewes. Anim. Sci. J., 84: 466-475.
Wei, S; Gong, Z; Ouyang, X; Xie, K and Wei, M (2013b). Modulation of expression, localization of FSHR and uterine development by GnRH agonist active immunization in ewes. J. Anim. Plant Sci., 19: 2888-2900.
Yuan, L; Wang, B; Piao, S; Tan, J and An, T (2008). Effect of PMSG on development of reproductive organs in different day-old immature mice. Acta Lab. Anim. Sci. Sin., 16: 338-341.
Zeleznik, A (2004). The physiology of follicle selection. Reprod. Biol. Endocrinol., 2: 31-34.
Zhang, C; Liu, L; Di, M; Zhao, D; Zhao, L; Xu, P and Rui, R (2007). Effects of the PMSG dosages on superovulation and oocyte quality in immature rats. Chinese J. Comp. Med., 17: 338-344. (in Chinese)
Zhang, C; Xia, G and Tsang, BK (2011). Interactions of thyroid hormone and FSH in the regulation of rat granulosa cell apoptosis. Front Biosci., 3: 1401-1413.
Zhou, J (1995). Research on estrus synchronization and superovulation in KM mice. Chinese J. Exp. Anim. Sci., 5: 139-141.
Zhou, XL; Teng, Y; Cao, R; Fu, H; Xiong, K; Sun, WX; Zhu, CC; Huang, XJ; Xiao, P and Liu, HL (2013). Rescue from dominant follicle atresia by follicle-stimulating hormone in mice. Genet. Mol. Res., 12: 2945-2952.