Animal Reproduction (AR)
https://animal-reproduction.org/journal/animreprod/article/doi/10.1590/1984-3143-AR2023-0007
Animal Reproduction (AR)
THEMATIC SECTION: IX INTERNATIONAL SYMPOSIUM ON ANIMAL BIOLOGY OF REPRODUCTION (ISABR 2022)

Maternal obesity and ovarian failure: is leptin the culprit?

Yashaswi Sharma; António Miguel Galvão

Downloads: 0
Views: 328

Abstract

At the time of its discovery and characterization in 1994, leptin was mostly considered a metabolic hormone able to regulate body weight and energy homeostasis. However, in recent years, a great deal of literature has revealed leptin’s pleiotropic nature, through its involvement in numerous physiological contexts including the regulation of the female reproductive tract and ovarian function. Obesity has been largely associated with infertility, and leptin signalling is known to be dysregulated in the ovaries of obese females. Hence, the disruption of ovarian leptin signalling was shown to contribute to the pathophysiology of ovarian failure in obese females, affecting transcriptional programmes in the gamete and somatic cells. This review attempts to uncover the underlying mechanisms contributing to female infertility associated with obesity, as well as to shed light on the role of leptin in the metabolic dysregulation within the follicle, the effects on the oocyte epigenome, and the potential long-term consequence to embryo programming.

Keywords

maternal obesity, leptin, oocyte quality, epigenetics, ovarian leptin resistance

References

Al Darwich A, Perreau C, Petit MH, Papillier P, Dupont J, Guillaume D, Mermillod P, Guignot F. Effect of PUFA on embryo cryoresistance, gene expression and AMPKalpha phosphorylation in IVF-derived bovine embryos. Prostaglandins Other Lipid Mediat. 2010;93(1-2):30-6. http://dx.doi.org/10.1016/j.prostaglandins.2010.06.002. PMid:20601073.

Allen JF, Paula WBM. Mitochondrial genome function and maternal inheritance. Biochem Soc Trans. 2013;41(5):1298-304. http://dx.doi.org/10.1042/BST20130106. PMid:24059523.

Almabhouh F, Osman K, Ibrahim S, Gupalo S, Gnanou J, Ibrahim E, Singh HJ. Melatonin ameliorates the adverse effects of leptin on sperm. Asian J Androl. 2017;19(6):647-54. http://dx.doi.org/10.4103/1008-682X.183379. PMid:27748315.

Baltz JM, Tartia AP. Cell volume regulation in oocytes and early embryos: connecting physiology to successful culture media. Hum Reprod Update. 2010;16(2):166-76. http://dx.doi.org/10.1093/humupd/dmp045. PMid:19825850.

Banks AS, Davis SM, Bates SH, Myers MG Jr. Activation of downstream signals by the long form of the leptin receptor. J Biol Chem. 2000;275(19):14563-72. http://dx.doi.org/10.1074/jbc.275.19.14563. PMid:10799542.

Bellver J, Melo MAB, Bosch E, Serra V, Remohí J, Pellicer A. Obesity and poor reproductive outcome: the potential role of the endometrium. Fertil Steril. 2007;88(2):446-51. http://dx.doi.org/10.1016/j.fertnstert.2006.11.162. PMid:17418840.

Bence KK, Delibegovic M, Xue B, Gorgun CZ, Hotamisligil GS, Neel BG, Kahn BB. Neuronal PTP1B regulates body weight, adiposity and leptin action. Nat Med. 2006;12(8):917-24. http://dx.doi.org/10.1038/nm1435. PMid:16845389.

Benkhalifa M, Ferreira YJ, Chahine H, Louanjli N, Miron P, Merviel P, Copin H. Mitochondria: participation to infertility as source of energy and cause of senescence. Int J Biochem Cell Biol. 2014;55:60-4. http://dx.doi.org/10.1016/j.biocel.2014.08.011. PMid:25150832.

Bjørbæk C, Buchholz RM, Davis SM, Bates SH, Pierroz DD, Gu H, Neel BG, Myers MG Jr, Flier JS. Divergent roles of SHP-2 in ERK activation by leptin receptors. J Biol Chem. 2001;276(7):4747-55. http://dx.doi.org/10.1074/jbc.M007439200. PMid:11085989.

Bjørbaek C, Kahn BB. Leptin signaling in the central nervous system and the periphery. Recent Prog Horm Res. 2004;59(1):305-31. http://dx.doi.org/10.1210/rp.59.1.305. PMid:14749508.

Bjørbæk C, Lavery HJ, Bates SH, Olson RK, Davis SM, Flier JS, Myers MG Jr. SOCS3 mediates feedback inhibition of the leptin receptor via Tyr985. J Biol Chem. 2000;275(51):40649-57. http://dx.doi.org/10.1074/jbc.M007577200. PMid:11018044.

Bjørbæk C, Uotani S, Silva B, Flier JS. Divergent signaling capacities of the long and short isoforms of the leptin receptor. J Biol Chem. 1997;272(51):32686-95. http://dx.doi.org/10.1074/jbc.272.51.32686. PMid:9405487.

Blanquer-Rossellõ MM, Santandreu FM, Oliver J, Roca P, Valle A. Leptin modulates mitochondrial function, dynamics and biogenesis in MCF-7 cells. J Cell Biochem. 2015;116(9):2039-48. http://dx.doi.org/10.1002/jcb.25158. PMid:25752935.

Bouret SG, Simerly RB. Developmental programming of hypothalamic feeding circuits. Clin Genet. 2006;70(4):295-301. http://dx.doi.org/10.1111/j.1399-0004.2006.00684.x. PMid:16965320.

Brabant G, Müller G, Horn R, Anderwald C, Roden M, Nave H. Hepatic leptin signaling in obesity. FASEB J. 2005;19(8):1048-50. http://dx.doi.org/10.1096/fj.04-2846fje. PMid:15788447.

Brannian JD, Hansen KA. Leptin and ovarian folliculogenesis: implications for ovulation induction and ART outcomes. Semin Reprod Med. 2002;20(2):103-12. http://dx.doi.org/10.1055/s-2002-32501. PMid:12087495.

Buchbinder A, Lang U, Baker RS, Khoury JC, Mershon J, Clark KE. Leptin in the ovine fetus correlates with fetal and placental size. Am J Obstet Gynecol. 2001;185(4):786-91. http://dx.doi.org/10.1067/mob.2001.117313. PMid:11641652.

Calbet JAL, MacLean DA. Plasma glucagon and insulin responses depend on the rate of appearance of amino acids after ingestion of different protein solutions in humans. J Nutr. 2002;132(8):2174-82. http://dx.doi.org/10.1093/jn/132.8.2174. PMid:12163658.

Cardozo ER, Karmon AE, Gold J, Petrozza JC, Styer AK. Reproductive outcomes in oocyte donation cycles are associated with donor BMI. Hum Reprod. 2016;31(2):385-92. http://dx.doi.org/10.1093/humrep/dev298. PMid:26677960.

Castracane V, Henson MC. (2003). Leptin and reproduction. New York: Springer. http://dx.doi.org/10.1007/978-1-4615-0157-2.

Cervero A, Horcajadas JA, Martín J, Pellicer A, Simón C. The leptin system during human endometrial receptivity and preimplantation development. J Clin Endocrinol Metab. 2004;89(5):2442-51. http://dx.doi.org/10.1210/jc.2003-032127. PMid:15126576.

Chen Q, Yan W, Duan E. Epigenetic inheritance of acquired traits through sperm RNAs and sperm RNA modifications. Nat Rev Genet. 2016;17(12):733-43. http://dx.doi.org/10.1038/nrg.2016.106. PMid:27694809.

Cheng A, Uetani N, Simoncic PD, Chaubey VP, Lee-Loy A, McGlade CJ, Kennedy BP, Tremblay ML. Attenuation of leptin action and regulation of obesity by protein tyrosine phosphatase 1B. Dev Cell. 2002;2(4):497-503. http://dx.doi.org/10.1016/S1534-5807(02)00149-1. PMid:11970899.

Childs G, Odle AK, MacNicol MC, MacNicol AM. The importance of leptin to reproduction. Endocrinology. 2021;162(2):1-18. http://dx.doi.org/10.1210/endocr/bqaa204. PMid:33165520.

Chou SH, Mantzoros C. 20 years of leptin: role of leptin in human reproductive disorders. J Endocrinol. 2014;223(1):T49-62. http://dx.doi.org/10.1530/JOE-14-0245. PMid:25056118.

Considine R, Sinha MK, Heiman ML, Kriauciunas A, Stephens TW, Nyce MR, Ohannesian JP, Marco CC, McKee LJ, Bauer TL, Caro JF. Serum immunoreactive-leptin concentrations in normal-weight and obese humans. N Engl J Med. 1996;334(5):292-5. http://dx.doi.org/10.1056/NEJM199602013340503. PMid:8532024.

Cui Y, Miyoshi K, Claudio E, Siebenlist UK, Gonzalez FJ, Flaws J, Wagner KU, Hennighausen L. Loss of the peroxisome proliferation-activated receptor gamma (PPARgamma) does not affect mammary development and propensity for tumor formation but leads to reduced fertility. J Biol Chem. 2002;277(20):17830-5. http://dx.doi.org/10.1074/jbc.M200186200. PMid:11884400.

D’souza AM, Neumann UH, Glavas MM, Kieffer TJ. The glucoregulatory actions of leptin. Mol Metab. 2017;6(9):1052-65. http://dx.doi.org/10.1016/j.molmet.2017.04.011. PMid:28951828.

Dağ ZÖ, Dilbaz B. Impact of obesity on infertility in women. J Turk Ger Gynecol Assoc. 2015;16(2):111-7. http://dx.doi.org/10.5152/jtgga.2015.15232. PMid:26097395.

Dalamaga M, Chou SH, Shields K, Papageorgiou P, Polyzos SA, Mantzoros CS. Leptin at the intersection of neuroendocrinology and metabolism: current evidence and therapeutic perspectives. Cell Metab. 2013;18(1):29-42. http://dx.doi.org/10.1016/j.cmet.2013.05.010. PMid:23770129.

Daxinger L, Whitelaw E. Understanding transgenerational epigenetic inheritance via the gametes in mammals. Nat Rev Genet. 2012;13(3):153-62. http://dx.doi.org/10.1038/nrg3188. PMid:22290458.

Derghal A, Djelloul M, Airault C, Pierre C, Dallaporta M, Troadec JD, Tillement V, Tardivel C, Bariohay B, Trouslard J, Mounien L. Leptin is required for hypothalamic regulation of miRNA stargeting POMC 3′UTR. Front Cell Neurosci. 2015;9:172. http://dx.doi.org/10.3389/FNCEL.2015.00172/ABSTRACT. PMid:25999818.

Downs SM, Hudson ED. Energy substrates and the completion of spontaneous meiotic maturation. Zygote. 2000;8(4):339-51. http://dx.doi.org/10.1017/S0967199400001131. PMid:11108555.

Downs SM, Mosey JL, Klinger J. Fatty acid oxidation and meiotic resumption in mouse oocytes. Mol Reprod Dev. 2009;76(9):844-53. http://dx.doi.org/10.1002/mrd.21047. PMid:19455666.

Dunkerton S, Mrcog M, Aiken C, Bchir M, Mrcp M. Impact of the intrauterine environment on future reproductive and metabolic health. Obstet Gynaecol. 2022;24(2):93-100. http://dx.doi.org/10.1111/tog.12797.

Dunning KR, Akison LK, Russell DL, Norman RJ, Robker RL. Increased beta-oxidation and improved oocyte developmental competence in response to L-Carnitine during ovarian in vitro follicle development in mice. Biol Reprod. 2011;85(3):548-55. http://dx.doi.org/10.1095/biolreprod.110.090415. PMid:21613630.

Dunning KR, Russell DL, Robker RL. Lipids and oocyte developmental competence: the role of fatty acids and β-oxidation. Reproduction. 2014;148(1):R15-27. http://dx.doi.org/10.1530/REP-13-0251. PMid:24760880.

Enriori PJ, Evans AE, Sinnayah P, Cowley MA. Leptin resistance and obesity. Obesity. 2006;14(Suppl 5):254S-8S. http://dx.doi.org/10.1038/oby.2006.319. PMid:17021377.

Enriori PJ, Evans AE, Sinnayah P, Jobst EE, Tonelli-Lemos L, Billes SK, Glavas MM, Grayson BE, Perello M, Nillni EA, Grove KL, Cowley MA. Diet-induced obesity causes severe but reversible leptin resistance in arcuate melanocortin neurons. Cell Metab. 2007;5(3):181-94. http://dx.doi.org/10.1016/j.cmet.2007.02.004. PMid:17339026.

Eppig JJ, Marin-Bivens C, Viveiros MM, de la Fuente R. Regulation of mammalian oocyte maturation. In: Leung PCK, Adashi EY, editors. The ovary. 2nd ed. London: Academic Press; 2004. p. 113-29. http://dx.doi.org/10.1016/B978-012444562-8/50008-2.

Farooqi IS, O’Rahilly S. Leptin: a pivotal regulator of human energy homeostasis. Am J Clin Nutr. 2009;89(3):980S-4S. http://dx.doi.org/10.3945/ajcn.2008.26788C. PMid:19211814.

Ferguson EM, Leese HJ. A potential role for triglyceride as an energy source during bovine oocyte maturation and early embryo development. Mol Reprod Dev. 2006;73(9):1195-201. http://dx.doi.org/10.1002/mrd.20494. PMid:16804881.

Fiedor E, Zajda K, Gregoraszczuk EL. Leptin receptor antagonists’ action on HDAC expression eliminating the negative effects of leptin in ovarian cancer. Cancer Genomics Proteomics. 2018;15(4):329-36. http://dx.doi.org/10.21873/cgp.20091. PMid:29976638.

Flaus A, Owen-Hughes T. Mechanisms for ATP-dependent chromatin remodelling: the means to the end. FEBS J. 2011;278(19):3579-95. http://dx.doi.org/10.1111/j.1742-4658.2011.08281.x. PMid:21810178.

Forhead AJ, Fowden AL. The hungry fetus? Role of leptin as a nutritional signal before birth. J Physiol. 2009;587(Pt 6):1145-52. http://dx.doi.org/10.1113/jphysiol.2008.167072. PMid:19188249.

Forhead AJ, Lamb CA, Franko KL, O’Connor DM, Wooding FBP, Cripps RL, Ozanne S, Blache D, Shen QW, Du M, Fowden AL. Role of leptin in the regulation of growth and carbohydrate metabolism in the ovine fetus during late gestation. J Physiol. 2008;586(Pt 9):2393-403. http://dx.doi.org/10.1113/jphysiol.2007.149237. PMid:18325979.

Friedman JM, Halaas JL. Leptin and the regulation of body weight in mammals. Nature. 1998;395(6704):763-70. http://dx.doi.org/10.1038/27376. PMid:9796811.

Frühbeck G, Catalán V, Rodríguez A, Ramírez B, Becerril S, Salvador J, Colina I, Gómez-Ambrosi J. Adiponectin-leptin ratio is a functional biomarker of adipose tissue inflammation. Nutrients. 2019;11(2):454. http://dx.doi.org/10.3390/nu11020454. PMid:30813240.

Galvão A, Henriques S, Pestka D, Lukasik K, Skarzynski D, Mateus LM, Ferreira-Dias GML. Equine luteal function regulation may depend on the interaction between cytokines and vascular endothelial growth factor: an in vitro study. Biol Reprod. 2012;86(6):187. http://dx.doi.org/10.1095/biolreprod.111.097147. PMid:22492973.

Ge H, Tollner TL, Hu Z, Dai M, Li X, Guan H, Shan D, Zhang X, Lv J, Huang C, Dong Q. The importance of mitochondrial metabolic activity and mitochondrial DNA replication during oocyte maturation in vitro on oocyte quality and subsequent embryo developmental competence. Mol Reprod Dev. 2012;79(6):392-401. http://dx.doi.org/10.1002/mrd.22042. PMid:22467220.

Ge ZJ, Luo SM, Lin F, Liang QX, Huang L, Wei YC, Hou Y, Han ZM, Schatten H, Sun QY. DNA methylation in oocytes and liver of female mice and their offspring: effects of high-fat-diet-induced obesity. Environ Health Perspect. 2014;122(2):159-64. http://dx.doi.org/10.1289/ehp.1307047. PMid:24316659.

Ge ZJ, Schatten H, Zhang CL, Sun QY. Oocyte ageing and epigenetics. Reproduction. 2015;149(3):R103-14. http://dx.doi.org/10.1530/REP-14-0242. PMid:25391845.

Gilbody S, Lewis S, Lightfoot T. Methylenetetrahydrofolate reductase (MTHFR) genetic polymorphisms and psychiatric disorders: a HuGE review. Am J Epidemiol. 2007;165(1):1-13. http://dx.doi.org/10.1093/aje/kwj347. PMid:17074966.

Grewal S, Gubbi S, Fosam A, Sedmak C, Sikder S, Talluru H, Brown RJ, Muniyappa R. Metabolomic analysis of the effects of leptin replacement therapy in patients with lipodystrophy. J Endocr Soc. 2020;4(1):bvz022. http://dx.doi.org/10.1210/jendso/bvz022. PMid:32010873.

Gu L, Liu H, Gu X, Boots C, Moley KH, Wang Q. Metabolic control of oocyte development: linking maternal nutrition and reproductive outcomes. Cell Mol Life Sci. 2015;72(2):251-71. http://dx.doi.org/10.1007/s00018-014-1739-4. PMid:25280482.

Guelinckx I, Devlieger R, Beckers K, Vansant G. Maternal obesity: pregnancy complications, gestational weight gain and nutrition. Obes Rev. 2008;9(2):140-50. http://dx.doi.org/10.1111/j.1467-789X.2007.00464.x. PMid:18221480.

Harris SE, Adriaens I, Leese HJ, Gosden RG, Picton HM. Carbohydrate metabolism by murine ovarian follicles and oocytes grown in vitro. Reproduction. 2007;134(3):415-24. http://dx.doi.org/10.1530/REP-07-0061. PMid:17709560.

Hassink SG, de Lancey E, Sheslow D, Smith-Kirwin SM, O’Connor DM, Considine R, Opentanova I, Dostal K, Spear ML, Leef K, Ash M, Spitzer AR, Funanage VL. Placental leptin: an important new growth factor in intrauterine and neonatal development? Pediatrics. 1997;100(1):E1. http://dx.doi.org/10.1542/peds.100.1.e1. PMid:9200375.

Hausman GJ, Barb CR, Lents CA. Leptin and reproductive function. Biochimie. 2012;94(10):2075-81. http://dx.doi.org/10.1016/j.biochi.2012.02.022. PMid:22980196.

He M, Zhang T, Yang Y, Wang C. Mechanisms of oocyte maturation and related epigenetic regulation. Front Cell Dev Biol. 2021;9:654028. http://dx.doi.org/10.3389/fcell.2021.654028. PMid:33842483.

Hegyi K, Fülöp K, Kovács K, Tóth S, Falus A. Leptin-induced signal transduction pathways. Cell Biol Int. 2004;28(3):159-69. http://dx.doi.org/10.1016/j.cellbi.2003.12.003. PMid:14984741.

Henry BA, Andrews ZB, Rao A, Clarke IJ. Central leptin activates mitochondrial function and increases heat production in skeletal muscle. Endocrinology. 2011;152(7):2609-18. http://dx.doi.org/10.1210/en.2011-0143. PMid:21558317.

Hong J, Lee E. Intrafollicular amino acid concentration and the effect of amino acids in a defined maturation medium on porcine oocyte maturation, fertilization, and preimplantation development. Theriogenology. 2007;68(5):728-35. http://dx.doi.org/10.1016/j.theriogenology.2007.06.002. PMid:17658593.

Hruby A, Hu FB. The Epidemiology of Obesity: A Big Picture. PharmacoEconomics. 2015;33(7):673-89. http://dx.doi.org/10.1007/s40273-014-0243-x. PMid:25471927.

Hunt PA, Hassold TJ. Human female meiosis: what makes a good egg go bad? Trends Genet. 2008;24(2):86-93. http://dx.doi.org/10.1016/j.tig.2007.11.010. PMid:18192063.

Huypens P, Sass S, Wu M, Dyckhoff D, Tschöp M, Theis F, Marschall S, de Angelis MH, Beckers J. Epigenetic germline inheritance of diet-induced obesity and insulin resistance. Nat Genet. 2016;48(5):497-9. http://dx.doi.org/10.1038/ng.3527. PMid:26974008.

Igosheva N, Abramov AY, Poston L, Eckert JJ, Fleming TP, Duchen MR, McConnell J. Maternal diet-induced obesity alters mitochondrial activity and redox status in mouse oocytes and zygotes. PLoS One. 2010;5(4):e10074. http://dx.doi.org/10.1371/journal.pone.0010074. PMid:20404917.

Islam MS, Sjöholm Å, Emilsson V. Fetal pancreatic islets express functional leptin receptors and leptin stimulates proliferation of fetal islet cells. Int J Obes. 2000;24(10):1246-53. http://dx.doi.org/10.1038/sj.ijo.0801370. PMid:11093284.

Jansson N, Greenwood SL, Johansson BR, Powell TL, Jansson T. Leptin stimulates the activity of the system A amino acid transporter in human placental villous fragments. J Clin Endocrinol Metab. 2003;88(3):1205-11. http://dx.doi.org/10.1210/jc.2002-021332. PMid:12629107.

Javaid MK, Godfrey KM, Taylor P, Robinson SM, Crozier SR, Dennison EM, Robinson JS, Breier BR, Arden NK, Cooper C. Umbilical cord leptin predicts neonatal bone mass. Calcif Tissue Int. 2005;76(5):341-7. http://dx.doi.org/10.1007/s00223-004-1128-3. PMid:15864467.

Johnson MT, Freeman EA, Gardner DK, Hunt PA. Oxidative metabolism of pyruvate is required for meiotic maturation of murine oocytes in vivo. Biol Reprod. 2007;77(1):2-8. http://dx.doi.org/10.1095/biolreprod.106.059899. PMid:17314311.

Kalive M, Faust JJ, Koeneman BA, Capco DG. Involvement of the PKC family in regulation of early development. Mol Reprod Dev. 2010;77(2):95-104. http://dx.doi.org/10.1002/mrd.21112. PMid:19777543.

Kamei Y, Suganami T, Ehara T, Kanai S, Hayashi K, Yamamoto Y, Miura S, Ezaki O, Okano M, Ogawa Y. Increased expression of DNA methyltransferase 3a in obese adipose tissue: studies with transgenic mice. Obesity. 2010;18(2):314-21. http://dx.doi.org/10.1038/oby.2009.246. PMid:19680236.

Karakosta P, Georgiou V, Fthenou E, Papadopoulou E, Roumeliotaki T, Margioris A, Castanas E, Kampa M, Kogevinas M, Chatzi L. Maternal weight status, cord blood leptin and fetal growth: a prospective mother-child cohort study (Rhea study). Paediatr Perinat Epidemiol. 2013;27(5):461-71. http://dx.doi.org/10.1111/ppe.12074. PMid:23930782.

Khan R, Jiang X, Hameed U, Shi Q. Role of lipid metabolism and signaling in mammalian oocyte maturation, quality, and acquisition of competence. Front Cell Dev Biol. 2021;9:639704. http://dx.doi.org/10.3389/fcell.2021.639704. PMid:33748128.

Kirchberg FF, Brandt S, Moß A, Peissner W, Koenig W, Rothenbacher D, Brenner H, Koletzko B, Hellmuth C, Wabitsch M. Metabolomics reveals an entanglement of fasting leptin concentrations with fatty acid oxidation and gluconeogenesis in healthy children. PLoS One. 2017;12(8):e0183185. http://dx.doi.org/10.1371/journal.pone.0183185. PMid:28817652.

Kirillova A, Smitz JEJ, Sukhikh GT, Mazunin I. The role of mitochondria in oocyte maturation. Cells. 2021;10(9):2484. http://dx.doi.org/10.3390/cells10092484. PMid:34572133.

Koch C, Augustine RA, Steger J, Ganjam GK, Benzler J, Pracht C, Lowe C, Schwartz MW, Shepherd PR, Anderson GM, Grattan DR, Tups A. Leptin rapidly improves glucose homeostasis in obese mice by increasing hypothalamic insulin sensitivity. J Neurosci. 2010;30(48):16180-7. http://dx.doi.org/10.1523/JNEUROSCI.3202-10.2010. PMid:21123564.

Kooistra SM, Helin K. Molecular mechanisms and potential functions of histone demethylases. Nat Rev Mol Cell Biol. 2012;13(5):297-311. http://dx.doi.org/10.1038/nrm3327. PMid:22473470.

Kristensen J, Vestergaard M, Wisborg K, Kesmodel U, Secher NJ. Pre-pregnancy weight and the risk of stillbirth and neonatal death. BJOG. 2005;112(4):403-8. http://dx.doi.org/10.1111/j.1471-0528.2005.00437.x. PMid:15777435.

Kurus M, Karakaya C, Karalok MH, To G, Johnson J. The control of oocyte survival by intrinsic and extrinsic factors. Adv Exp Med Biol. 2013;761:7-18. http://dx.doi.org/10.1007/978-1-4614-8214-7_2. PMid:24097378.

Kyrou I, Randeva HS, Tsigos C, Kaltsas G, Weickert MO. Clinical problems caused by obesity. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, Herder WW, Dhatariya K, Hofland J, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrère B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, editors. Endotext [Internet]. South Dartmouth: MDText.com; 2018 [cited 2023 Jan 10]. Available from: https://www.ncbi.nlm.nih.gov/books/NBK278973/

Lawler K, Huang-Doran I, Sonoyama T, Collet TH, Keogh JM, Henning E, O’Rahilly S, Bottolo L, Farooqi IS. Leptin-mediated changes in the human metabolome. J Clin Endocrinol Metab. 2020;105(8):2541-52. http://dx.doi.org/10.1210/clinem/dgaa251. PMid:32392278.

Leong I. Link between maternal obesity and offspring is STELLA. Nat Rev Endocrinol. 2018;14(4):189. http://dx.doi.org/10.1038/nrendo.2018.26. PMid:29498374.

Li L, Zhu S, Shu W, Guo Y, Guan Y, Zeng J, Wang H, Han L, Zhang J, Liu X, Li C, Hou X, Gao M, Ge J, Ren C, Zhang H, Schedl T, Guo X, Chen M, Wang Q. Characterization of metabolic patterns in mouse oocytes during meiotic maturation. Mol Cell. 2020;80(3):525-540.e9. http://dx.doi.org/10.1016/j.molcel.2020.09.022. PMid:33068521.

Lin J, Barb CR, Matteri RL, Kraeling RR, Chen X, Meinersmann RJ, Rampacek GB. Long form leptin receptor mRNA expression in the brain, pituitary, and other tissues in the pig. Domest Anim Endocrinol. 2000;19(1):53-61. http://dx.doi.org/10.1016/S0739-7240(00)00064-3. PMid:10962198.

Liu J, Yang X, Yu S, Zheng R. The leptin resistance. Adv Exp Med Biol. 2018;1090:145-63. http://dx.doi.org/10.1007/978-981-13-1286-1_8. PMid:30390289.

Liu T, Qu J, Tian M, Yang R, Song X, Li R, Yan J, Qiao J. Lipid metabolic process involved in oocyte maturation during folliculogenesis. Front Cell Dev Biol. 2022;10:806890. http://dx.doi.org/10.3389/fcell.2022.806890. PMid:35433675.

Luo GF, Yu TY, Wen XH, Li Y, Yang GS. Alteration of mitochondrial oxidative capacity during porcine preadipocyte differentiation and in response to leptin. Mol Cell Biochem. 2008;307(1-2):83-91. http://dx.doi.org/10.1007/s11010-007-9587-2. PMid:17909948.

Luzzo KM, Wang Q, Purcell SH, Chi M, Jimenez PT, Grindler N, Schedl T, Moley KH. High fat diet induced developmental defects in the mouse: oocyte meiotic aneuploidy and fetal growth retardation/brain defects. PLoS One. 2012;7(11):e49217. http://dx.doi.org/10.1371/journal.pone.0049217. PMid:23152876.

Maffei M, Halaas J, Ravussin E, Pratley RE, Lee GH, Zhang Y, Fei H, Kim S, Lallone R, Ranganathan S, Kern PA, Friedman JM. Leptin levels in human and rodent: measurement of plasma leptin and ob RNA in obese and weight-reduced subjects. Nat Med. 1995;1(11):1155-61. http://dx.doi.org/10.1038/nm1195-1155. PMid:7584987.

Magariños MP, Sánchez-Margalet V, Kotler M, Calvo JC, Varone CL. Leptin promotes cell proliferation and survival of trophoblastic cells. Biol Reprod. 2007;76(2):203-10. http://dx.doi.org/10.1095/biolreprod.106.051391. PMid:17021346.

Mantzoros CS, Cramer DW, Liberman RF, Barbieri RL. Predictive value of serum and follicular fluid leptin concentrations during assisted reproductive cycles in normal women and in women with the polycystic ovarian syndrome. Hum Reprod. 2000;15(3):539-44. http://dx.doi.org/10.1093/humrep/15.3.539. PMid:10686193.

Mark AL, Correia MLG, Rahmouni K, Haynes WG. Selective leptin resistance: a new concept in leptin physiology with cardiovascular implications. J Hypertens. 2002;20(7):1245-50. http://dx.doi.org/10.1097/00004872-200207000-00001. PMid:12131511.

Matarese G, Procaccini C, De Rosa V. Leptin and immune function, inflammation and angiognenesis. In: Castracane VD, Henson MC, editors. Leptin. Boston: Springer; 2006. (Endocrine Updates; no. 25). http://dx.doi.org/10.1007/978-0-387-31416-7_7.

Matilainen O, Quirós PM, Auwerx J. Mitochondria and epigenetics: crosstalk in homeostasis and stress. Trends Cell Biol. 2017;27(6):453-63. http://dx.doi.org/10.1016/j.tcb.2017.02.004. PMid:28274652.

McKeegan PJ, Sturmey RG. The role of fatty acids in oocyte and early embryo development. Reprod Fertil Dev. 2011;24(1):59-67. http://dx.doi.org/10.1071/RD11907. PMid:22394718.

McMillen IC, Adam CL, Mühlhäusler BS. Early origins of obesity: programming the appetite regulatory system. J Physiol. 2005;565(Pt 1):9-17. http://dx.doi.org/10.1113/jphysiol.2004.081992. PMid:15705647.

McPherson NO, Bell VG, Zander-Fox DL, Fullston T, Wu LL, Robker RL, Lane M. When two obese parents are worse than one! Impacts on embryo and fetal development. Am J Physiol Endocrinol Metab. 2015;309(6):E568-81. http://dx.doi.org/10.1152/ajpendo.00230.2015. PMid:26199280.

Menzies KJ, Zhang H, Katsyuba E, Auwerx J. Protein acetylation in metabolism-metabolites and cofactors. Nat Rev Endocrinol. 2016;12(1):43-60. http://dx.doi.org/10.1038/nrendo.2015.181. PMid:26503676.

Miller KK, Parulekar MS, Schoenfeld E, Anderson E, Hubbard J, Klibanski A, Grinspoon SK. Decreased leptin levels in normal weight women with hypothalamic amenorrhea: the effects of body composition and nutritional intake. J Clin Endocrinol Metab. 1998;83(7):2309-12. http://dx.doi.org/10.1210/jc.83.7.2309. PMid:9661600.

Minokoshi Y, Kim YB, Peroni OD, Fryer LGD, Müller C, Carling D, Kahn BB. Leptin stimulates fatty-acid oxidation by activating AMP-activated protein kinase. Nature. 2002;415(6869):339-43. http://dx.doi.org/10.1038/415339a. PMid:11797013.

Minokoshi Y, Toda C, Okamoto S. Regulatory role of leptin in glucose and lipid metabolism in skeletal muscle. Indian J Endocrinol Metab. 2012;16(9, Suppl 3):S562. http://dx.doi.org/10.4103/2230-8210.105573. PMid:23565491.

Moley KH, Chi MMY, Knudson CM, Korsmeyer SJ, Mueckler MM. Hyperglycemia induces apoptosis in pre-implantation embryos through cell death effector pathways. Nat Med. 1998;4(12):1421-4. http://dx.doi.org/10.1038/4013. PMid:9846581.

Montgomery DC, Sorum AW, Guasch L, Nicklaus MC, Meier JL. Metabolic regulation of histone acetyltransferases by endogenous Acyl-CoA cofactors. Chem Biol. 2015;22(8):1030-9. http://dx.doi.org/10.1016/j.chembiol.2015.06.015. PMid:26190825.

Moore LL, Singer MR, Bradlee ML, Rothman KJ, Milunsky A. A prospective study of the risk of congenital defects associated with maternal obesity and diabetes mellitus. Epidemiology. 2000;11(6):689-94. http://dx.doi.org/10.1097/00001648-200011000-00013. PMid:11055631.

Morgan DA, Thedens DR, Weiss R, Rahmouni K. Mechanisms mediating renal sympathetic activation to leptin in obesity. Am J Physiol Regul Integr Comp Physiol. 2008;295(6):R1730-6. http://dx.doi.org/10.1152/ajpregu.90324.2008. PMid:18815209.

Morrish F, Noonan J, Perez-Olsen C, Gafken PR, Fitzgibbon M, Kelleher J, VanGilst M, Hockenbery D. Myc-dependent mitochondrial generation of acetyl-CoA contributes to fatty acid biosynthesis and histone acetylation during cell cycle entry. J Biol Chem. 2010;285(47):36267-74. http://dx.doi.org/10.1074/jbc.M110.141606. PMid:20813845.

Moslehi N, Shab-Bidar S, Tehrani FR, Mirmiran P, Azizi F. Is ovarian reserve associated with body mass index and obesity in reproductive aged women? A meta-analysis. Menopause. 2018;25(9):1046-55. http://dx.doi.org/10.1097/GME.0000000000001116. PMid:29738413.

Mühlhäusler BS, Roberts CT, McFarlane JR, Kauter KG, McMillen IC. Fetal leptin is a signal of fat mass independent of maternal nutrition in ewes fed at or above maintenance energy requirements. Biol Reprod. 2002;67(2):493-9. http://dx.doi.org/10.1095/biolreprod67.2.493. PMid:12135887.

Münzberg H, Flier JS, Bjørbæk C. Region-specific leptin resistance within the hypothalamus of diet-induced obese mice. Endocrinology. 2004;145(11):4880-9. http://dx.doi.org/10.1210/en.2004-0726. PMid:15271881.

Myers MG Jr, Leibel RL, Seeley RJ, Schwartz MW. Obesity and leptin resistance: distinguishing cause from effect. trends endocrinol metab. 2010;21(11):643-51. http://dx.doi.org/10.1016/j.tem.2010.08.002. PMid:20846876.

Myers MG Jr. Leptin receptor signaling and the regulation of mammalian physiology. Recent Prog Horm Res. 2004;59(1):287-304. http://dx.doi.org/10.1210/rp.59.1.287. PMid:14749507.

Myers MP, Andersen JN, Cheng A, Tremblay ML, Horvath CM, Parisien JP, Salmeen A, Barford D, Tonks NK. TYK2 and JAK2 are substrates of protein-tyrosine phosphatase 1B. J Biol Chem. 2001;276(51):47771-4. http://dx.doi.org/10.1074/jbc.C100583200. PMid:11694501.

Nakanishi N, Nakagawa Y, Tokushige N, Aoki N, Matsuzaka T, Ishii K, Yahagi N, Kobayashi K, Yatoh S, Takahashi A, Suzuki H, Urayama O, Yamada N, Shimano H. The up-regulation of microRNA-335 is associated with lipid metabolism in liver and white adipose tissue of genetically obese mice. Biochem Biophys Res Commun. 2009;385(4):492-6. http://dx.doi.org/10.1016/j.bbrc.2009.05.058. PMid:19460359.

Orsi NM, Gopichandran N, Leese HJ, Picton HM, Harris SE. Fluctuations in bovine ovarian follicular fluid composition throughout the oestrous cycle. Reproduction. 2005;129(2):219-28. http://dx.doi.org/10.1530/rep.1.00460. PMid:15695616.

Ozcan L, Ergin AS, Lu A, Chung J, Sarkar S, Nie D, Myers MG Jr, Ozcan U. Endoplasmic reticulum stress plays a central role in development of leptin resistance. Cell Metab. 2009;9(1):35-51. http://dx.doi.org/10.1016/j.cmet.2008.12.004. PMid:19117545.

Palou M, Picó C, McKay JA, Sánchez J, Priego T, Mathers JC, Palou A. Protective effects of leptin during the suckling period against later obesity may be associated with changes in promoter methylation of the hypothalamic pro-opiomelanocortin gene. Br J Nutr. 2011;106(5):769-78. http://dx.doi.org/10.1017/S0007114511000973. PMid:21554805.

Palou M, Picó C, Palou A. Leptin as a breast milk component for the prevention of obesity. Nutr Rev. 2018;76(12):875-92. http://dx.doi.org/10.1093/nutrit/nuy046. PMid:30285146.

Park J, Kusminski CM, Chua SC, Scherer PE. Leptin receptor signaling supports cancer cell metabolism through suppression of mitochondrial respiration in vivo. Am J Pathol. 2010;177(6):3133-44. http://dx.doi.org/10.2353/ajpath.2010.100595. PMid:21056997.

Park JH, Stoffers DA, Nicholls RD, Simmons RA. Development of type 2 diabetes following intrauterine growth retardation in rats is associated with progressive epigenetic silencing of Pdx1. J Clin Invest. 2008;118(6):2316-24. http://dx.doi.org/10.1172/JCI33655. PMid:18464933.

Paula-Lopes FF, Boelhauve M, Habermann FA, Sinowatz F, Wolf E. Leptin promotes meiotic progression and developmental capacity of bovine oocytes via cumulus cell-independent and-dependent mechanisms. Biol Reprod. 2007;76(3):532-41. http://dx.doi.org/10.1095/biolreprod.106.054551. PMid:17093200.

Pelland AMD, Corbett HE, Baltz JM. Amino acid transport mechanisms in mouse oocytes during growth and meiotic maturation. Biol Reprod. 2009;81(6):1041-54. http://dx.doi.org/10.1095/biolreprod.109.079046. PMid:19605782.

Pereira S, Cline DL, Glavas MM, Covey SD, Kieffer TJ. Tissue-specific effects of leptin on glucose and lipid metabolism. Endocr Rev. 2021;42(1):1-28. http://dx.doi.org/10.1210/endrev/bnaa027. PMid:33150398.

Pérez-Pérez A, Sánchez-Jiménez F, Maymó J, Dueñas JL, Varone C, Sánchez-Margalet V. Role of leptin in female reproduction. Clin Chem Lab Med. 2015;53(1):15-28. http://dx.doi.org/10.1515/cclm-2014-0387. PMid:25014521.

Pinney SE, Jaeckle Santos LJ, Han Y, Stoffers DA, Simmons RA. Exendin-4 increases histone acetylase activity and reverses epigenetic modifications that silence Pdx1 in the intrauterine growth retarded rat. Diabetologia. 2011;54(10):2606-14. http://dx.doi.org/10.1007/s00125-011-2250-1. PMid:21779870.

Pinney SE, Simmons RA. Epigenetic mechanisms in the development of type 2 diabetes. Trends Endocrinol Metab. 2010;21(4):223-9. http://dx.doi.org/10.1016/j.tem.2009.10.002. PMid:19864158.

Pirola L, Balcerczyk A, Okabe J, El-Osta A. Epigenetic phenomena linked to diabetic complications. Nat Rev Endocrinol. 2010;6(12):665-75. http://dx.doi.org/10.1038/nrendo.2010.188.

Poretsky L, Cataldo NA, Rosenwaks Z, Giudice LC. The insulin-related ovarian regulatory system in health and disease. Endocr Rev. 1999;20(4):535-82. http://dx.doi.org/10.1210/edrv.20.4.0374. PMid:10453357.

Qi L, Chen X, Wang J, Lv B, Zhang J, Ni B, Xue Z. Mitochondria: the panacea to improve oocyte quality? Ann Transl Med. 2019;7(23):789. http://dx.doi.org/10.21037/atm.2019.12.02. PMid:32042805.

Rabe K, Lehrke M, Parhofer KG, Broedl UC. Adipokines and insulin resistance. Mol Med. 2008;14(11-12):741-51. http://dx.doi.org/10.2119/2008-00058.Rabe. PMid:19009016.

Rando OJ, Simmons RA. I’m eating for two: parental dietary effects on offspring metabolism. Cell. 2015;161(1):93-105. http://dx.doi.org/10.1016/j.cell.2015.02.021. PMid:25815988.

Reidy SP, Weber JM. Leptin: an essential regulator of lipid metabolism. Comp Biochem Physiol A Mol Integr Physiol. 2000;125(3):285-98. http://dx.doi.org/10.1016/S1095-6433(00)00159-8. PMid:10794958.

Reina-Campos M, Diaz-Meco MT, Moscat J. The complexity of the serine glycine one-carbon pathway in cancer. J Cell Biol. 2020;219(1):e201907022. http://dx.doi.org/10.1083/jcb.201907022. PMid:31690618.

Robker RL, Akison LK, Bennett BD, Thrupp PN, Chura LR, Russell DL, Lane M, Norman RJ. Obese women exhibit differences in ovarian metabolites, hormones, and gene expression compared with moderate-weight women. J Clin Endocrinol Metab. 2009;94(5):1533-40. http://dx.doi.org/10.1210/jc.2008-2648.

Robker RL, Wu LLY, Yang X. Inflammatory pathways linking obesity and ovarian dysfunction. J Reprod Immunol. 2011;88(2):142-8. http://dx.doi.org/10.1016/j.jri.2011.01.008. PMid:21333359.

Rooke JA, Ainslie A, Watt RG, Alink FM, McEvoy TG, Sinclair KD, Garnsworthy PC, Webb R. Dietary carbohydrates and amino acids influence oocyte quality in dairy heifers. Reprod Fertil Dev. 2009;21(3):419-27. http://dx.doi.org/10.1071/RD08193. PMid:19261219.

Ruiz-Cortés ZT, Men T, Palin MF, Downey BR, Lacroix DA, Murphy BD. Porcine leptin receptor: molecular structure and expression in the ovary. Mol Reprod Dev. 2000;56(4):465-74. http://dx.doi.org/10.1002/1098-2795(200008)56:4<465::AID-MRD4>3.0.CO;2-Q. PMid:10911396.

Russell DL, Gilchrist RB, Brown HM, Thompson JG. Bidirectional communication between cumulus cells and the oocyte: old hands and new players? Theriogenology. 2016;86(1):62-8. http://dx.doi.org/10.1016/j.theriogenology.2016.04.019. PMid:27160446.

Ryan NK, Woodhouse CM, van der Hoek KH, Gilchrist RB, Armstrong DT, Norman RJ. Expression of leptin and its receptor in the murine ovary: possible role in the regulation of oocyte maturation. Biol Reprod. 2002;66(5):1548-54. http://dx.doi.org/10.1095/biolreprod66.5.1548. PMid:11967222.

Sáinz N, Barrenetxe J, Moreno-Aliaga MJ, Martínez JA. Leptin resistance and diet-induced obesity: central and peripheral actions of leptin. Metabolism. 2015;64(1):35-46. http://dx.doi.org/10.1016/j.metabol.2014.10.015. PMid:25497342.

Saito T, Hiroi M, Kato T. Development of glucose utilization studied in single oocytes and preimplantation embryos from mice. Biol Reprod. 1994;50(2):266-70. http://dx.doi.org/10.1095/biolreprod50.2.266. PMid:8142545.

Sampath H, Ntambi JM. Polyunsaturated fatty acid regulation of genes of lipid metabolism. Annu Rev Nutr. 2005;25(1):317-40. http://dx.doi.org/10.1146/annurev.nutr.25.051804.101917. PMid:16011470.

Sangiao-Alvarellos S, Pena-Bello L, Manfredi-Lozano M, Tena-Sempere M, Cordido F. Perturbation of hypothalamic microRNA expression patterns in male rats after metabolic distress: impact of obesity and conditions of negative energy balance. Endocrinology. 2014;155(5):1838-50. http://dx.doi.org/10.1210/en.2013-1770. PMid:24517225.

Santangeli L, Sattar N, Huda SS. Impact of maternal obesity on perinatal and childhood outcomes. Best Pract Res Clin Obstet Gynaecol. 2015;29(3):438-48. http://dx.doi.org/10.1016/j.bpobgyn.2014.10.009. PMid:25497183.

Schatten H, Sun Q-Y, Prather R. The impact of mitochondrial function/dysfunction on IVF and new treatment possibilities for infertility. Reprod Biol Endocrinol. 2014;12(1):111. http://dx.doi.org/10.1186/1477-7827-12-111. PMid:25421171.

Shafiei Sheykhani HR, Batavani RA, Najafi GR. Protective effect of leptin on induced apoptosis with trichostatin A on buffalo oocytes. Vet Res Forum. 2016;7(2):99-104. PMid:27482353.

Shen L, Cordero JF, Wang JS, Shen Y, Li S, Liang L, Zou Z, Li C. Association between genetically determined leptin and blood lipids considering alcohol consumption: a Mendelian randomisation study. BMJ Open. 2019;9(11):e026860. http://dx.doi.org/10.1136/bmjopen-2018-026860. PMid:31699712.

Short KR, Chadwick JQ, Teague AM, Tullier MA, Wolbert L, Coleman C, Copeland KC. Effect of obesity and exercise training on plasma amino acids and amino metabolites in american indian adolescents. J Clin Endocrinol Metab. 2019;104(8):3249-61. http://dx.doi.org/10.1210/jc.2018-02698. PMid:31216576.

Silva E, Paczkowski M, Krisher RL. The effect of leptin on maturing porcine oocytes is dependent on glucose concentration. Mol Reprod Dev. 2012;79(4):296-307. http://dx.doi.org/10.1002/mrd.22029. PMid:22368147.

Sinclair KD, Kuran M, Gebbie FE, Webb R, McEvoy TG. Nitrogen metabolism and fertility in cattle: II. Development of oocytes recovered from heifers offered diets differing in their rate of nitrogen release in the rumen. J Anim Sci. 2000;78(10):2670-80. http://dx.doi.org/10.2527/2000.78102670x. PMid:11048933.

Sirard MA. Follicle environment and quality of in vitro matured oocytes. J Assist Reprod Genet. 2011;28(6):483-8. http://dx.doi.org/10.1007/s10815-011-9554-4. PMid:21394521.

Sivan E, Lin WM, Homko CJ, Reece EA, Boden G. Leptin is present in human cord blood. Diabetes. 1997;46(5):917-9. http://dx.doi.org/10.2337/diab.46.5.917. PMid:9133565.

Smith BC, Denu JM. Chemical mechanisms of histone lysine and arginine modifications. Biochim Biophys Acta. 2009;1789(1):45-57. http://dx.doi.org/10.1016/j.bbagrm.2008.06.005. PMid:18603028.

Song NY, Lee YH, Na HK, Baek JH, Surh YJ. Leptin induces SIRT1 expression through activation of NF-E2-related factor 2: implications for obesity-associated colon carcinogenesis. Biochem Pharmacol. 2018;153:282-91. http://dx.doi.org/10.1016/j.bcp.2018.02.001. PMid:29427626.

Songsasen N, Wildt DE. Oocyte biology and challenges in developing in vitro maturation systems in the domestic dog. Anim Reprod Sci. 2007;98(1-2):2-22. http://dx.doi.org/10.1016/j.anireprosci.2006.10.004. PMid:17097840.

Sonne SB, Yadav R, Yin G, Dalgaard MD, Myrmel LS, Gupta R, Wang J, Madsen L, Kajimura S, Kristiansen K. Obesity is associated with depot-specific alterations in adipocyte DNA methylation and gene expression. Adipocyte. 2017;6(2):124-33. http://dx.doi.org/10.1080/21623945.2017.1320002. PMid:28481699.

Sperber H, Mathieu J, Wang Y, Ferreccio A, Hesson J, Xu Z, Fischer KA, Devi A, Detraux D, Gu H, Battle SL, Showalter M, Valensisi C, Bielas JH, Ericson NG, Margaretha L, Robitaille AM, Margineantu D, Fiehn O, Hockenbery D, Blau CA, Raftery D, Margolin AA, Hawkins RD, Moon RT, Ware CB, Ruohola-Baker H. The metabolome regulates the epigenetic landscape during naïve to primed human embryonic stem cell transition. Nat Cell Biol. 2015;17(12):1523-35. http://dx.doi.org/10.1038/ncb3264. PMid:26571212.

Spinelli JB, Haigis MC. The multifaceted contributions of mitochondria to cellular metabolism. Nat Cell Biol. 2018;20(7):745-54. http://dx.doi.org/10.1038/s41556-018-0124-1. PMid:29950572.

Stocker CJ, Cawthorne MA. The influence of leptin on early life programming of obesity. Trends Biotechnol. 2008;26(10):545-51. http://dx.doi.org/10.1016/j.tibtech.2008.06.004. PMid:18706724.

Strum JC, Swenson KI, Turner JE, Bell RM. Ceramide triggers meiotic cell cycle progression in Xenopus oocytes. A potential mediator of progesterone-induced maturation. J Biol Chem. 1995;270(22):13541-7. http://dx.doi.org/10.1074/jbc.270.22.13541. PMid:7768956.

Sturmey RG, Brison DR, Leese HJ. Assessing embryo viability by measurement of amino acid turnover. Reprod Biomed Online. 2008;17(4):486-96. http://dx.doi.org/10.1016/S1472-6483(10)60234-9. PMid:18854101.

Su Y-Q, Sugiura K, Eppig JJ. mouse oocyte control of granulosa cell development and function: paracrine regulation of cumulus cell metabolism. Semin Reprod Med. 2009;27(1):032-042. http://dx.doi.org/10.1055/s-0028-1108008. PMid:19197803.

Sugiura K, Eppig JJ. Control of metabolic cooperativity between oocytes and their companion granulosa cells by mouse oocytes. Reprod Fertil Dev. 2005;17(7):667-74. http://dx.doi.org/10.1071/RD05071. PMid:16364219.

Sutton-McDowall ML, Gilchrist RB, Thompson JG. The pivotal role of glucose metabolism in determining oocyte developmental competence. Reproduction. 2010;139(4):685-95. http://dx.doi.org/10.1530/REP-09-0345. PMid:20089664.

Tartaglia LA, Dembski M, Weng X, Deng N, Culpepper J, Devos R, Richards GJ, Campfield LA, Clark FT, Deeds J, Muir C, Sanker S, Moriarty A, Moore KJ, Smutko JS, Mays GG, Wool EA, Monroe CA, Tepper RI. Identification and expression cloning of a leptin receptor, OB-R. Cell. 1995;83(7):1263-71. http://dx.doi.org/10.1016/0092-8674(95)90151-5. PMid:8548812.

Tchio CM, Harbuzariu A, Harmon T, Beech D, Gonzalez-Perez R. Abstract 1901: leptin modulation of PCSC, HDAC, and microRNA in pancreatic adenocarcinoma. Cancer Res. 2016;76(14, Suppl):1901. http://dx.doi.org/10.1158/1538-7445.AM2016-1901.

Thompson RF, Einstein FH. Epigenetic basis for fetal origins of age-related disease. J Womens Health (Larchmt). 2010;19(3):581-7. http://dx.doi.org/10.1089/jwh.2009.1408. PMid:20136551.

Tong Q, Xu Y. Central Leptin Regulation of Obesity and Fertility. Curr Obes Rep. 2012;1(4):236-44. http://dx.doi.org/10.1007/s13679-012-0025-8. PMid:23243628.

Tups A. Physiological Models of Leptin Resistance. J Neuroendocrinol. 2009;21(11):961-71. http://dx.doi.org/10.1111/j.1365-2826.2009.01916.x. PMid:19732287.

Udagawa J, Hatta T, Hashimoto R, Otani H. Roles of leptin in prenatal and perinatal brain development. Congenit Anom (Kyoto). 2007;47(3):77-83. http://dx.doi.org/10.1111/j.1741-4520.2007.00150.x. PMid:17688465.

Valūnienė M, Verkauskienė R, Boguszewski M, Dahlgren J, Lašienė D, Lašas L, Wikland KA. Leptin levels at birth and in early postnatal life in small- and appropriate-for-gestational-age infants. Medicina. 2007;43(10):784. http://dx.doi.org/10.3390/medicina43100100.

van Winkle LJ. Amino acid transport regulation and early embryo development. Biol Reprod. 2001;64(1):1-12. http://dx.doi.org/10.1095/biolreprod64.1.1. PMid:11133652.

Varvarigou A, Mantzoros CS, Beratis NG. Cord blood leptin concentrations in relation to intrauterine growth. Clin Endocrinol. 1999;50(2):177-83. http://dx.doi.org/10.1046/j.1365-2265.1999.00630.x. PMid:10396359.

Vickers MH, Gluckman PD, Coveny AH, Hofman PL, Cutfield WS, Gertler A, Breier BH, Harris M. Neonatal leptin treatment reverses developmental programming. Endocrinology. 2005;146(10):4211-6. http://dx.doi.org/10.1210/en.2005-0581. PMid:16020474.

Wai T, Ao A, Zhang X, Cyr D, Dufort D, Shoubridge EA. The role of mitochondrial DNA copy number in mammalian fertility. Biol Reprod. 2010;83(1):52-62. http://dx.doi.org/10.1095/biolreprod.109.080887. PMid:20130269.

Watson AJ. Oocyte cytoplasmic maturation: a key mediator of oocyte and embryo developmental competence. J Anim Sci. 2007;85(13, Suppl):E1-3. http://dx.doi.org/10.2527/jas.2006-432. PMid:17322120.

Welt CK, Chan JL, Bullen J, Murphy R, Smith P, DePaoli AM, Karalis A, Mantzoros CS. Recombinant human leptin in women with hypothalamic amenorrhea. N Engl J Med. 2004;351(10):987-97. http://dx.doi.org/10.1056/NEJMoa040388. PMid:15342807.

Whidden L, Martel J, Rahimi S, Chaillet JR, Chan D, Trasler JM. Compromised oocyte quality and assisted reproduction contribute to sex-specific effects on offspring outcomes and epigenetic patterning. Hum Mol Genet. 2016;25(21):4649-60. http://dx.doi.org/10.1093/hmg/ddw293. PMid:28173052.

White V, González E, Capobianco E, Pustovrh C, Martínez N, Higa R, Baier M, Jawerbaum A. Leptin modulates nitric oxide production and lipid metabolism in human placenta. Reprod Fertil Dev. 2006;18(4):425-32. http://dx.doi.org/10.1071/RD05105. PMid:16737635.

Wołodko K, Castillo‐fernandez J, Kelsey G, Galvão A. Revisiting the impact of local leptin signaling in folliculogenesis and oocyte maturation in obese mothers. Int J Mol Sci. 2021;22(8):4270. http://dx.doi.org/10.3390/ijms22084270. PMid:33924072.

Wołodko K, Walewska E, Adamowski M, Castillo-Fernandez J, Kelsey G, Galvão A. Leptin resistance in the ovary of obese mice is associated with profound changes in the transcriptome of cumulus cells. Cell Physiol Biochem. 2020;54(3):417-37. http://dx.doi.org/10.33594/000000228. PMid:32348667.

Wróblewski A, Strycharz J, Świderska E, Drewniak K, Drzewoski J, Szemraj J, Kasznicki J, Śliwińska A. Molecular insight into the interaction between epigenetics and leptin in metabolic disorders. Nutrients. 2019;11(8):1872. http://dx.doi.org/10.3390/nu11081872. PMid:31408957.

Wu LLY, Dunning KR, Yang X, Russell DL, Lane M, Norman RJ, Robker RL. High-fat diet causes lipotoxicity responses in cumulus-oocyte complexes and decreased fertilization rates. Endocrinology. 2010;151(11):5438-45. http://dx.doi.org/10.1210/en.2010-0551. PMid:20861227.

Wu X, Xu S, Weng J. Hyperglycemia-mediated oocyte TET3 insufficiency predisposes offspring to glucose intolerance. J Diabetes Investig. 2022;13(10):1649-51. http://dx.doi.org/10.1111/jdi.13885. PMid:35839128.

Wyman A, Pinto AB, Sheridan R, Moley KH. One-cell zygote transfer from diabetic to nondiabetic mouse results in congenital malformations and growth retardation in offspring. Endocrinology. 2008;149(2):466-9. http://dx.doi.org/10.1210/en.2007-1273. PMid:18039778.

Yamagishi SI, Edelstein D, Du XL, Kaneda Y, Guzmán M, Brownlee M. Leptin induces mitochondrial superoxide production and monocyte chemoattractant protein-1 expression in aortic endothelial cells by increasing fatty acid oxidation via protein kinase A. J Biol Chem. 2001;276(27):25096-100. http://dx.doi.org/10.1074/jbc.M007383200. PMid:11342529.

Yang X, Wu LL, Chura LR, Liang X, Lane M, Norman RJ, Robker RL. Exposure to lipid-rich follicular fluid is associated with endoplasmic reticulum stress and impaired oocyte maturation in cumulus-oocyte complexes. Fertil Steril. 2012;97(6):1438-43. http://dx.doi.org/10.1016/j.fertnstert.2012.02.034. PMid:22440252.

Yau-Qiu ZX, Picó C, Rodríguez AM, Palou A. Leptin distribution in rat foetal and extraembryonic tissues in late gestation: a physiological view of amniotic fluid leptin. Nutrients. 2020;12(9):1-14. http://dx.doi.org/10.3390/nu12092542. PMid:32825787.

Yeung BHY, Griffiths K, Berger L, Paudel O, Shin MK, Rui L, Sham JSK, Polotsky VY, Tang WY. Leptin induces epigenetic regulation of transient receptor potential melastatin 7 in rat adrenal pheochromocytoma cells. Am J Respir Cell Mol Biol. 2021;65(2):214-21. http://dx.doi.org/10.1165/rcmb.2020-0374OC. PMid:33891828.

Yin S, Jiang X, Jiang H, Gao Q, Wang F, Fan S, Khan T, Jabeen N, Khan M, Ali A, Xu P, Pandita TK, Fan H-Y, Zhang Y, Shi Q. Histone acetyltransferase KAT8 is essential for mouse oocyte development by regulating reactive oxygen species levels. Development. 2017;144(12):2165-74. http://dx.doi.org/10.1242/dev.149518. PMid:28506985.

You D, Nilsson E, Tenen DE, Lyubetskaya A, Lo JC, Jiang R, Deng J, Dawes BA, Vaag A, Ling C, Rosen ED, Kang S. Dnmt3a is an epigenetic mediator of adipose insulin resistance. eLife. 2017;6:e30766. http://dx.doi.org/10.7554/eLife.30766. PMid:29091029.

Zhang F, Chen Y, Heiman M, DiMarchi R. Leptin: structure, function and biology. Vitam Horm. 2005;71:345-72. http://dx.doi.org/10.1016/S0083-6729(05)71012-8. PMid:16112274.

Zhang LH, Tan XY, Wu K, Zhuo MQ, Song YF, Chen QL. Regulation and mechanism of leptin on lipid metabolism in ovarian follicle cells from yellow catfish Pelteobagrus fulvidraco. Gen Comp Endocrinol. 2015;222:116-23. http://dx.doi.org/10.1016/j.ygcen.2015.06.008. PMid:26119184.

Zhang Y, Proenca R, Maffei M, Barone M, Leopold L, Friedman JM. Positional cloning of the mouse obese gene and its human homologue. Nature. 1994;372(6505):425-32. http://dx.doi.org/10.1038/372425a0. PMid:7984236.

Zhou Y, Qiu L, Xiao Q, Wang Y, Meng X, Xu R, Wang S, Na R. Obesity and diabetes related plasma amino acid alterations. Clin Biochem. 2013;46(15):1447-52. http://dx.doi.org/10.1016/j.clinbiochem.2013.05.045. PMid:23697717.
 


Submitted date:
01/10/2023

Accepted date:
01/24/2023

63ee0d67a9539545eb03e717 animreprod Articles
Links & Downloads

Anim Reprod

Share this page
Page Sections