Register      Login
Reproduction, Fertility and Development Reproduction, Fertility and Development Society
Vertebrate reproductive science and technology
REVIEW

The impact of leptin on sperm

Fayez A. Almabhouh https://orcid.org/0000-0002-8119-3531 A and Harbindar Jeet Singh A B *
+ Author Affiliations
- Author Affiliations

A Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh Campus, Sungai Buloh, Selangor, Malaysia.

B I-PerFForm, Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh Campus, Sungai Buloh, Selangor, Malaysia.

* Correspondence to: hjsingh@uitm.edu.my

Handling Editor: Jessica Dunleavy

Reproduction, Fertility and Development 35(8) 459-468 https://doi.org/10.1071/RD22222
Published online: 18 May 2023

© 2023 The Author(s) (or their employer(s)). Published by CSIRO Publishing

Abstract

Despite its important role in numerous physiological functions, including regulation of appetite and body weight, immune function and normal sexual maturation, raised leptin levels could result in significant damaging effects on sperm. The adverse effects of leptin on the male reproductive system result from its direct actions on the reproductive organs and cells instead of the hypothalamus–pituitary–gonadal axis. Binding of leptin to the receptors in the seminiferous tubular cells of the testes increases free radical production and decreases the gene expression and activity of endogenous enzymatic antioxidants. These effects are mediated via the PI3K pathway. The resultant oxidative stress causes significant damage to the seminiferous tubular cells, germ cells and sperm DNA leading to apoptosis, increased sperm DNA fragmentation, decreased sperm count, increased fraction of sperm with abnormal morphology, and decreased seminiferous tubular height and diameter. This review summarises the evidence in the literature on the adverse effects of leptin on sperm, which could underlie the often-reported sperm abnormalities in obese hyperleptinaemic infertile males. Although leptin is necessary for normal reproductive function, its raised levels could be pathologic. There is, therefore, a need to identify the cut-off level in the serum and seminal fluid above which leptin becomes pathological for better management of leptin associated adverse effects on male reproductive function.

Keywords: infertility, leptin, male fertility, oxidative stress, PI3K pathway, sperm, sperm count, sperm morphology.


References

Abbasihormozi, S, Shahverdi, A, Kouhkan, A, Cheraghi, J, Akhlaghi, AA, and Kheimeh, A (2013). Relationship of leptin administration with production of reactive oxygen species, sperm DNA fragmentation, sperm parameters and hormone profile in the adult rat. Archives of Gynecology and Obstetrics 287, 1241–1249.
Relationship of leptin administration with production of reactive oxygen species, sperm DNA fragmentation, sperm parameters and hormone profile in the adult rat.Crossref | GoogleScholarGoogle Scholar |

Abir, R, Ao, A, Jin, S, Barnett, M, Raanani, H, Ben-Haroush, A, and Fisch, B (2005). Leptin and its receptors in human fetal and adult ovaries. Fertility and Sterility 84, 1779–1782.
Leptin and its receptors in human fetal and adult ovaries.Crossref | GoogleScholarGoogle Scholar |

Agarwal, A, and Saleh, RA (2002). Role of oxidants in male infertility: rationale, significance, and treatment. Urologic Clinics of North America 29, 817–827.
Role of oxidants in male infertility: rationale, significance, and treatment.Crossref | GoogleScholarGoogle Scholar |

Agarwal, A, Ikemoto, I, and Loughlin, KR (1994). Relationship of sperm parameters with levels of reactive oxygen species in semen specimens. Journal of Urology 152, 107–110.
Relationship of sperm parameters with levels of reactive oxygen species in semen specimens.Crossref | GoogleScholarGoogle Scholar |

Agarwal, A, Prabakaran, SA, and Said, TM (2005). Prevention of oxidative stress injury to sperm. Journal of Andrology 26, 654–660.
Prevention of oxidative stress injury to sperm.Crossref | GoogleScholarGoogle Scholar |

Ahima, RS, Dushay, J, Flier, SN, Prabakaran, D, and Flier, JS (1997). Leptin accelerates the onset of puberty in normal female mice. Journal of Clinical Investigation 99, 391–395.
Leptin accelerates the onset of puberty in normal female mice.Crossref | GoogleScholarGoogle Scholar |

Al-Sultan, AI, and Al-Elq, AH (2006). Leptin levels in normal weight and obese Saudi adults. Journal of Family & Community Medicine 13, 97–102.

Almabhouh, FA, and Singh, HJ (2018). Adverse effects of leptin on histone-to-protamine transition during spermatogenesis are prevented by melatonin in Sprague–Dawley rats. Andrologia 50, e12814.
Adverse effects of leptin on histone-to-protamine transition during spermatogenesis are prevented by melatonin in Sprague–Dawley rats.Crossref | GoogleScholarGoogle Scholar |

Almabhouh, FA, and Singh, HJ (2022). Systematic review of literature documenting the link between BMI, sperm parameters, and leptin. Fertility & Reproduction 04, 1–10.
Systematic review of literature documenting the link between BMI, sperm parameters, and leptin.Crossref | GoogleScholarGoogle Scholar |

Almabhouh, FA, Osman, K, Siti Fatimah, I, Sergey, G, Gnanou, J, and Singh, HJ (2015). Effects of leptin on sperm count and morphology in Sprague–Dawley rats and their reversibility following a 6-week recovery period. Andrologia 47, 751–758.
Effects of leptin on sperm count and morphology in Sprague–Dawley rats and their reversibility following a 6-week recovery period.Crossref | GoogleScholarGoogle Scholar |

Almabhouh, FA, Osman, K, Ibrahim, SF, Gupalo, S, Gnanou, J, Ibrahim, E, and Singh, HJ (2017). Melatonin ameliorates the adverse effects of leptin on sperm. Asian Journal of Andrology 19, 647–654.
Melatonin ameliorates the adverse effects of leptin on sperm.Crossref | GoogleScholarGoogle Scholar |

Almabhouh, FA, Ahmad Muhammad, FI, Ibrahim, H, and Singh, HJ (2019). Leptin: a pleitropic factor in physiology. Journal of Clinical and Health Sciences 4, 31–57.
Leptin: a pleitropic factor in physiology.Crossref | GoogleScholarGoogle Scholar |

Angelopoulou, R, and Kyriazoglou, M (2005). Sperm oxidative damage and the role of reactive oxygen species in male infertility. Archives of Hellenic Medicine 22, 433–446.

Arora, H, Qureshi, R, Khodamoradi, K, Seetharam, D, Parmar, M, Van Booven, DJ, Issa, IC, Sackstein, D, Lamb, D, Hare, JM, and Ramasamy, R (2022). Leptin secreted from testicular microenvironment modulates hedgehog signaling to augment the endogenous function of Leydig cells. Cell Death & Disease 13, 208.
Leptin secreted from testicular microenvironment modulates hedgehog signaling to augment the endogenous function of Leydig cells.Crossref | GoogleScholarGoogle Scholar |

Aziz, N, Saleh, RA, Sharma, RK, Lewis-Jones, I, Esfandiari, N, Thomas, AJ, and Agarwal, A (2004). Novel association between sperm reactive oxygen species production, sperm morphological defects, and the sperm deformity index. Fertility and Sterility 81, 349–354.
Novel association between sperm reactive oxygen species production, sperm morphological defects, and the sperm deformity index.Crossref | GoogleScholarGoogle Scholar |

Barbagallo, F, Condorelli, RA, Mongioì, LM, Cannarella, R, Cimino, L, Magagnini, MC, Crafa, A, La Vignera, S, and Calogero, AE (2021). Molecular mechanisms underlying the relationship between obesity and male infertility. Metabolites 11, 840.
Molecular mechanisms underlying the relationship between obesity and male infertility.Crossref | GoogleScholarGoogle Scholar |

Barrios-Correa, AA, Estrada, JA, and Contreras, I (2018). Leptin signaling in the control of metabolism and appetite: lessons from animal models. Journal of Molecular Neuroscience 66, 390–402.
Leptin signaling in the control of metabolism and appetite: lessons from animal models.Crossref | GoogleScholarGoogle Scholar |

Batra, A, Okur, B, Glauben, R, Erben, U, Ihbe, J, Stroh, T, Fedke, I, Chang, H-D, Zeitz, M, and Siegmund, B (2010). Leptin: a critical regulator of CD4+ T-cell polarization in vitro and in vivo. Endocrinology 151, 56–62.
Leptin: a critical regulator of CD4+ T-cell polarization in vitro and in vivo.Crossref | GoogleScholarGoogle Scholar |

Blanca, AJ, Ruiz-Armenta, MV, Zambrano, S, Salsoso, R, Miguel-Carrasco, JL, Fortuño, A, Revilla, E, Mate, A, and Vázquez, CM (2016). Leptin induces oxidative stress through activation of NADPH oxidase in renal tubular cells: antioxidant effect of L-carnitine. Journal of Cellular Biochemistry 117, 2281–2288.
Leptin induces oxidative stress through activation of NADPH oxidase in renal tubular cells: antioxidant effect of L-carnitine.Crossref | GoogleScholarGoogle Scholar |

Broker, LE, Kruyt, FAE, and Giaccone, G (2005). Cell death independent of caspases: a review. Clinical Cancer Research 11, 3155–3162.
Cell death independent of caspases: a review.Crossref | GoogleScholarGoogle Scholar |

Casabiell, X, Pineiro, V, Peino, R, Lage, M, Camina, J, Gallego, R, Vallejo, LG, Dieguez, C, and Casanueva, FF (1998). Gender differences in both spontaneous and stimulated leptin secretion by human omental adipose tissue in vitro: dexamethasone and estradiol stimulate leptin release in women, but not in men. The Journal of Clinical Endocrinology & Metabolism 83, 2149–2155.
Gender differences in both spontaneous and stimulated leptin secretion by human omental adipose tissue in vitro: dexamethasone and estradiol stimulate leptin release in women, but not in men.Crossref | GoogleScholarGoogle Scholar |

Cheng, CY, and Mruk, DD (2012). The blood-testis barrier and its implications for male contraception. Pharmacological Reviews 64, 16–64.
The blood-testis barrier and its implications for male contraception.Crossref | GoogleScholarGoogle Scholar |

Clarke, IJ, and Henry, BA (1999). Leptin and reproduction. Reviews of Reproduction 4, 48–55.
Leptin and reproduction.Crossref | GoogleScholarGoogle Scholar |

Cottrell EC, Mercer JG (2012) Leptin receptors. In ‘Appetite control. Handbook of Experimental Pharmacology. Vol. 209’. (Ed. HG Joost) pp. 3–21. (Springer-Verlag: Berlin, Heidelberg, Germany) https://doi.org/10.1007/978-3-642-24716-3_1

Coussens, M, Maresh, JG, Yanagimachi, R, Maeda, G, and Allsopp, R (2008). Sirt1 deficiency attenuates spermatogenesis and germ cell function. PLoS ONE 3, e1571.
Sirt1 deficiency attenuates spermatogenesis and germ cell function.Crossref | GoogleScholarGoogle Scholar |

Cowley, MA, Smart, JL, Rubinstein, M, Cerdán, MG, Diano, S, Horvath, TL, Cone, RD, and Low, MJ (2001). Leptin activates anorexigenic POMC neurons through a neural network in the arcuate nucleus. Nature 411, 480–484.
Leptin activates anorexigenic POMC neurons through a neural network in the arcuate nucleus.Crossref | GoogleScholarGoogle Scholar |

Dardeno, TA, Chou, SH, Moon, H-S, Chamberland, JP, Fiorenza, CG, and Mantzoros, CS (2010). Leptin in human physiology and therapeutics. Frontiers in Neuroendocrinology 31, 377–393.
Leptin in human physiology and therapeutics.Crossref | GoogleScholarGoogle Scholar |

de Lamirande, E, and Gagnon, C (1995). Impact of reactive oxygen species on spermatozoa: a balancing act between beneficial and detrimental effects. Human Reproduction 10, 15–21.
Impact of reactive oxygen species on spermatozoa: a balancing act between beneficial and detrimental effects.Crossref | GoogleScholarGoogle Scholar |

Deng, C-Y, Lv, M, Luo, B-H, Zhao, S-Z, Mo, Z-C, and Xie, Y-J (2021). The role of the PI3K/AKT/mTOR signalling pathway in male reproduction. Current Molecular Medicine 21, 539–548.
The role of the PI3K/AKT/mTOR signalling pathway in male reproduction.Crossref | GoogleScholarGoogle Scholar |

Fortuno, A, Bidegain, J, Baltanas, A, Moreno, MU, Montero, L, Landecho, MF, Beloqui, O, Díez, J, and Zalba, G (2010). Is leptin involved in phagocytic NADPH oxidase overactivity in obesity? Potential clinical implications. Journal of Hypertension 28, 1944–1950.
Is leptin involved in phagocytic NADPH oxidase overactivity in obesity? Potential clinical implications.Crossref | GoogleScholarGoogle Scholar |

Francisco, V, Pino, J, Campos-Cabaleiro, V, Ruiz-Fernández, C, Mera, A, Gonzalez-Gay, MA, Gómez, R, and Gualillo, O (2018). Obesity, fat mass and immune system: role for leptin. Frontiers in Physiology 9, 640.
Obesity, fat mass and immune system: role for leptin.Crossref | GoogleScholarGoogle Scholar |

Fried, SK, Ricci, MR, Russell, CD, and Laferrère, B (2000). Regulation of leptin production in humans. The Journal of Nutrition 130, 3127S–3131S.
Regulation of leptin production in humans.Crossref | GoogleScholarGoogle Scholar |

Fruhbeck, G (2006). Intracellular signalling pathways activated by leptin. Biochemical Journal 393, 7–20.
Intracellular signalling pathways activated by leptin.Crossref | GoogleScholarGoogle Scholar |

Funahashi, H, Yada, T, Suzuki, R, and Shioda, S (2003). Distribution, function, and properties of leptin receptors in the brain. International Review of Cytology 224, 1–27.
Distribution, function, and properties of leptin receptors in the brain.Crossref | GoogleScholarGoogle Scholar |

Galluzzi, L, Vitale, I, Abrams, JM, Alnemri, ES, Baehrecke, EH, Blagosklonny, MV, Dawson, TM, Dawson, VL, El-Deiry, WS, Fulda, S, Gottlieb, E, Green, DR, Hengartner, MO, Kepp, O, Knight, RA, Kumar, S, Lipton, SA, Lu, X, Madeo, F, Malorni, W, Mehlen, P, Nuñez, G, Peter, ME, Piacentini, M, Rubinsztein, DC, Shi, Y, Simon, H-U, Vandenabeele, P, White, E, Yuan, J, Zhivotovsky, B, Melino, G, and Kroemer, G (2012). Molecular definitions of cell death subroutines: recommendations of the Nomenclature Committee on Cell Death. Cell Death & Differentiation 19, 107–120.
Molecular definitions of cell death subroutines: recommendations of the Nomenclature Committee on Cell Death.Crossref | GoogleScholarGoogle Scholar |

Gao, Y, Zhao, G, Song, Y, Haire, A, Yang, A, Zhao, X, and Wusiman, A (2022). Presence of leptin and its receptor in the ram reproductive system and in vitro effect of leptin on sperm quality. PeerJ 10, e13982.
Presence of leptin and its receptor in the ram reproductive system and in vitro effect of leptin on sperm quality.Crossref | GoogleScholarGoogle Scholar |

Garcia-Mayor, RV, Andrade, MA, Rios, M, Lage, M, Dieguez, C, and Casanueva, FF (1997). Serum leptin levels in normal children: relationship to age, gender, body mass index, pituitary-gonadal hormones, and pubertal stage. Journal of Clinical Endocrinology & Metabolism 82, 2849–2855.
Serum leptin levels in normal children: relationship to age, gender, body mass index, pituitary-gonadal hormones, and pubertal stage.Crossref | GoogleScholarGoogle Scholar |

Garcia-Peiro, A, Martinez-Heredia, J, Oliver-Bonet, M, Abad, C, Amengual, MJ, Navarro, J, Jones, C, Coward, K, Gosálvez, J, and Benet, J (2011). Protamine 1 to protamine 2 ratio correlates with dynamic aspects of DNA fragmentation in human sperm. Fertility and Sterility 95, 105–109.
Protamine 1 to protamine 2 ratio correlates with dynamic aspects of DNA fragmentation in human sperm.Crossref | GoogleScholarGoogle Scholar |

Gorska, E, Popko, K, Stelmaszczyk-Emmel, A, Ciepiela, O, Kucharska, A, and Wasik, M (2010). Leptin receptors. European Journal of Medical Research 15, 50.
Leptin receptors.Crossref | GoogleScholarGoogle Scholar |

Grinspoon, SK, Askari, H, Landt, ML, Nathan, DM, Schoenfeld, DA, Hayden, DL, Laposata, M, Hubbard, J, and Klibanski, A (1997). Effects of fasting and glucose infusion on basal and overnight leptin concentrations in normal-weight women. The American Journal of Clinical Nutrition 66, 1352–1356.
Effects of fasting and glucose infusion on basal and overnight leptin concentrations in normal-weight women.Crossref | GoogleScholarGoogle Scholar |

Guerra, B, Santana, A, Fuentes, T, Delgado-Guerra, S, Cabrera-Socorro, A, Dorado, C, and Calbet, JAL (2007). Leptin receptors in human skeletal muscle. Journal of Applied Physiology 102, 1786–1792.
Leptin receptors in human skeletal muscle.Crossref | GoogleScholarGoogle Scholar |

Haron, MN, D’Souza, UJA, Jaafar, H, Zakaria, R, and Singh, HJ (2010). Exogenous leptin administration decreases sperm count and increases the fraction of abnormal sperm in adult rats. Fertility and Sterility 93, 322–324.
Exogenous leptin administration decreases sperm count and increases the fraction of abnormal sperm in adult rats.Crossref | GoogleScholarGoogle Scholar |

Hatami-Baroogh, L, Razavi, S, Zarkesh-Esfahani, H, Tavalaee, M, Tanhaei, S, Ghaedi, K, Deemeh, MR, Rabiee, F, and Nasr-Esfahani, MH (2010). Evaluation of the leptin receptor in human spermatozoa. Reproductive Biology and Endocrinology 8, 17.
Evaluation of the leptin receptor in human spermatozoa.Crossref | GoogleScholarGoogle Scholar |

Houseknecht, KL, and Portocarrero, CP (1998). Leptin and its receptors: regulators of whole-body energy homeostasis. Domestic Animal Endocrinology 15, 457–475.
Leptin and its receptors: regulators of whole-body energy homeostasis.Crossref | GoogleScholarGoogle Scholar |

Hussain, Z, and Khan, JA (2017). Food intake regulation by leptin: mechanisms mediating gluconeogenesis and energy expenditure. Asian Pacific Journal of Tropical Medicine 10, 940–944.
Food intake regulation by leptin: mechanisms mediating gluconeogenesis and energy expenditure.Crossref | GoogleScholarGoogle Scholar |

Jope, T, Lammert, A, Kratzsch, J, Paasch, U, and Glander, H-J (2003). Leptin and leptin receptor in human seminal plasma and in human spermatozoa. International Journal of Andrology 26, 335–341.
Leptin and leptin receptor in human seminal plasma and in human spermatozoa.Crossref | GoogleScholarGoogle Scholar |

Kazmi, A, Sattar, A, Hashim, R, Khan, SP, Younus, M, and Khan, FA (2013). Serum leptin values in the healthy obese and non-obese subjects of Rawalpindi. Journal of Pakistan Medical Association 63, 245–248.

Kemal Duru, N, Morshedi, M, and Oehninger, S (2000). Effects of hydrogen peroxide on DNA and plasma membrane integrity of human spermatozoa. Fertility and Sterility 74, 1200–1207.
Effects of hydrogen peroxide on DNA and plasma membrane integrity of human spermatozoa.Crossref | GoogleScholarGoogle Scholar |

Khawar, MB, Sohail, AM, and Li, W (2022). SIRT1: a key player in male reproduction. Life 12, 318.
SIRT1: a key player in male reproduction.Crossref | GoogleScholarGoogle Scholar |

Kieffer, TJ, Heller, RS, and Habener, JF (1996). Leptin receptors expressed on pancreatic β-cells. Biochemical and Biophysical Research Communications 224, 522–527.
Leptin receptors expressed on pancreatic β-cells.Crossref | GoogleScholarGoogle Scholar |

Kiess, W, Blum, WF, and Aubert, ML (1998). Leptin, puberty and reproductive function: lessons from animal studies and observations in humans. European Journal of Endocrinology 138, 26–29.
Leptin, puberty and reproductive function: lessons from animal studies and observations in humans.Crossref | GoogleScholarGoogle Scholar |

Kolaczynski, JW, Ohannesian, JP, Considine, RV, Marco, CC, and Caro, JF (1996). Response of leptin to short-term and prolonged overfeeding in humans. The Journal of Clinical Endocrinology & Metabolism 81, 4162–4165.
Response of leptin to short-term and prolonged overfeeding in humans.Crossref | GoogleScholarGoogle Scholar |

Kwon, O, Kim, KW, and Kim, M-S (2016). Leptin signalling pathways in hypothalamic neurons. Cellular and Molecular Life Sciences 73, 1457–1477.
Leptin signalling pathways in hypothalamic neurons.Crossref | GoogleScholarGoogle Scholar |

Li, HWR, Chiu, PCN, Cheung, MPL, Yeung, WSB, and O, WS (2009). Effect of leptin on motility, capacitation and acrosome reaction of human spermatozoa. International Journal of Andrology 32, 687–694.
Effect of leptin on motility, capacitation and acrosome reaction of human spermatozoa.Crossref | GoogleScholarGoogle Scholar |

Mahbouli, S, Der Vartanian, A, Ortega, S, Rougé, S, Vasson, M-P, and Rossary, A (2017). Leptin induces ROS via NOX5 in healthy and neoplastic mammary epithelial cells. Oncology Reports 38, 3254–3264.
Leptin induces ROS via NOX5 in healthy and neoplastic mammary epithelial cells.Crossref | GoogleScholarGoogle Scholar |

Mahfouz, RZ, du Plessis, SS, Aziz, N, Sharma, R, Sabanegh, E, and Agarwal, A (2010). Sperm viability, apoptosis, and intracellular reactive oxygen species levels in human spermatozoa before and after induction of oxidative stress. Fertility and Sterility 93, 814–821.
Sperm viability, apoptosis, and intracellular reactive oxygen species levels in human spermatozoa before and after induction of oxidative stress.Crossref | GoogleScholarGoogle Scholar |

Malik, IA, Durairajanayagam, D, and Singh, HJ (2019). Leptin and its actions on reproduction in males. Asian Journal of Andrology 21, 296–299.
Leptin and its actions on reproduction in males.Crossref | GoogleScholarGoogle Scholar |

Masuzaki, H, Ogawa, Y, Isse, N, Satoh, N, Okazaki, T, Shigemoto, M, Mori, K, Tamura, N, Hosoda, K, Yoshimasa, Y, Jingami, H, Kawada, T, and Nakao, K (1995). Human obese gene expression: adipocyte-specific expression and regional differences in the adipose tissue. Diabetes 44, 855–858.
Human obese gene expression: adipocyte-specific expression and regional differences in the adipose tissue.Crossref | GoogleScholarGoogle Scholar |

Md Mokhtar, AH, Malik, IA, Abd Aziz, NAA, Almabhouh, FA, Durairajanayagam, D, and Singh, HJ (2019). LY294002, a PI3K pathway inhibitor, prevents leptin-induced adverse effects on spermatozoa in Sprague-Dawley rats. Andrologia 51, e13196.
LY294002, a PI3K pathway inhibitor, prevents leptin-induced adverse effects on spermatozoa in Sprague-Dawley rats.Crossref | GoogleScholarGoogle Scholar |

Park, H-K, and Ahima, RS (2015). Physiology of leptin: energy homeostasis, neuroendocrine function and metabolism. Metabolism 64, 24–34.
Physiology of leptin: energy homeostasis, neuroendocrine function and metabolism.Crossref | GoogleScholarGoogle Scholar |

Petrescu, AD, Grant, S, Williams, E, An, SY, Seth, N, Shell, M, Amundsen, T, Tan, C, Nadeem, Y, Tjahja, M, Weld, L, Chu, CS, Venter, J, Frampton, G, McMillin, M, and DeMorrow, S (2022). Leptin enhances hepatic fibrosis and inflammation in a mouse model of cholestasis. The American Journal of Pathology 192, 484–502.
Leptin enhances hepatic fibrosis and inflammation in a mouse model of cholestasis.Crossref | GoogleScholarGoogle Scholar |

Ravagnan, L, Gurbuxani, S, Susin, SA, Maisse, C, Daugas, E, Zamzami, N, Mak, T, Jäättelä, M, Penninger, JM, Garrido, C, and Kroemer, G (2001). Heat-shock protein 70 antagonizes apoptosis-inducing factor. Nature Cell Biology 3, 839–843.
Heat-shock protein 70 antagonizes apoptosis-inducing factor.Crossref | GoogleScholarGoogle Scholar |

Savitskaya, MA, and Onishchenko, GE (2015). Mechanisms of apoptosis. Biochemistry (Moscow) 80, 1393–1405.
Mechanisms of apoptosis.Crossref | GoogleScholarGoogle Scholar |

Schrauwen, P, van Marken Lichtenbelt, WD, Westerterp, KR, and Saris, WHM (1997). Effect of diet composition on leptin concentration in lean subjects. Metabolism Clinical and Experimental 46, 420–424.
Effect of diet composition on leptin concentration in lean subjects.Crossref | GoogleScholarGoogle Scholar |

Schroeter, MR, Leifheit-Nestler, M, Hubert, A, Schumann, B, Gluckermann, R, Eschholz, N, Krüger, N, Lutz, S, Hasenfuss, G, Konstantinides, S, and Schäfer, K (2013). Leptin promotes neointima formation and smooth muscle cell proliferation via NADPH oxidase activation and signalling in caveolin-rich microdomains. Cardiovascular Research 99, 555–565.
Leptin promotes neointima formation and smooth muscle cell proliferation via NADPH oxidase activation and signalling in caveolin-rich microdomains.Crossref | GoogleScholarGoogle Scholar |

Seetharam, D, Seetharam, D, Qureshi, R, Khodamoradi, K, Parmar, M, Van Booven, D, Vendeko, A, Farber, N, Arora, H, and Ramasamy, R (2022). 52 Leptin effects on leydig stem cell differentiation are specific to patients BMI. The Journal of Sexual Medicine 19, S27–S28.
52 Leptin effects on leydig stem cell differentiation are specific to patients BMI.Crossref | GoogleScholarGoogle Scholar |

Sharma, K, and Considine, RV (1998). The Ob protein (leptin) and the kidney. Kidney International 53, 1483–1487.
The Ob protein (leptin) and the kidney.Crossref | GoogleScholarGoogle Scholar |

Shimizu, H, Shimomura, Y, Nakanishi, Y, Futawatari, T, Ohtani, K, Sato, N, and Mori, M (1997). Estrogen increases in vivo leptin production in rats and human subjects. Journal of Endocrinology 154, 285–292.
Estrogen increases in vivo leptin production in rats and human subjects.Crossref | GoogleScholarGoogle Scholar |

Shiraishi, K, and Naito, K (2007). Effects of 4-hydroxy-2-nonenal, a marker of oxidative stress, on spermatogenesis and expression of p53 protein in male infertility. Journal of Urology 178, 1012–1017.
Effects of 4-hydroxy-2-nonenal, a marker of oxidative stress, on spermatogenesis and expression of p53 protein in male infertility.Crossref | GoogleScholarGoogle Scholar |

Simon, L, Castillo, J, Oliva, R, and Lewis, SEM (2011). Relationships between human sperm protamines, DNA damage and assisted reproduction outcomes. Reproductive BioMedicine Online 23, 724–734.
Relationships between human sperm protamines, DNA damage and assisted reproduction outcomes.Crossref | GoogleScholarGoogle Scholar |

Singh, HJ, Hryciw, D, Durairajanayagam, D, and Abdul Aziz, NAA (2022). Recognising the role of leptin in disease. Journal of Clinical and Health Sciences 7, 1–4.
Recognising the role of leptin in disease.Crossref | GoogleScholarGoogle Scholar |

Sinha, MK, Ohannesian, JP, Heiman, ML, Kriauciunas, A, Stephens, TW, Magosin, S, Marco, C, and Caro, JF (1996). Nocturnal rise of leptin in lean, obese, and non-insulin-dependent diabetes mellitus subjects. The Journal of Clinical Investigation 97, 1344–1347.
Nocturnal rise of leptin in lean, obese, and non-insulin-dependent diabetes mellitus subjects.Crossref | GoogleScholarGoogle Scholar |

Smith-Kirwin, SM, O’Connor, DM, Johnston, J, de Lancy, E, Hassink, SG, and Funanage, VL (1998). Leptin expression in human mammary epithelial cells and breast milk. The Journal of Clinical Endocrinology & Metabolism 83, 1810–1810.
Leptin expression in human mammary epithelial cells and breast milk.Crossref | GoogleScholarGoogle Scholar |

Song, N-Y, Lee, Y-H, Na, H-K, Baek, J-H, and Surh, Y-J (2018). Leptin induces SIRT1 expression through activation of NF-E2-related factor 2: Implications for obesity-associated colon carcinogenesis. Biochemical Pharmacology 153, 282–291.
Leptin induces SIRT1 expression through activation of NF-E2-related factor 2: Implications for obesity-associated colon carcinogenesis.Crossref | GoogleScholarGoogle Scholar |

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, and Tepper, RI (1995). Identification and expression cloning of a leptin receptor, OB-R. Cell 83, 1263–1271.
Identification and expression cloning of a leptin receptor, OB-R.Crossref | GoogleScholarGoogle Scholar |

Trayhurn, P, and Wood, IS (2005). Signalling role of adipose tissue: adipokines and inflammation in obesity. Biochemical Society Transactions 33, 1078–1081.
Signalling role of adipose tissue: adipokines and inflammation in obesity.Crossref | GoogleScholarGoogle Scholar |

von Schnurbein, J, Moss, A, Nagel, SA, Muehleder, H, Debatin, KM, Farooqi, IS, and Wabitsch, M (2012). Leptin substitution results in the induction of menstrual cycles in an adolescent with leptin deficiency and hypogonadotropic hypogonadism. Hormone Research in Paediatrics 77, 127–133.
Leptin substitution results in the induction of menstrual cycles in an adolescent with leptin deficiency and hypogonadotropic hypogonadism.Crossref | GoogleScholarGoogle Scholar |

Wang, X, Zhang, X, Hu, L, and Li, H (2018). Exogenous leptin affects sperm parameters and impairs blood testis barrier integrity in adult male mice. Reproductive Biology and Endocrinology 16, 55.
Exogenous leptin affects sperm parameters and impairs blood testis barrier integrity in adult male mice.Crossref | GoogleScholarGoogle Scholar |

Weir, CP, and Robaire, B (2007). Spermatozoa have decreased antioxidant enzymatic capacity and increased reactive oxygen species production during aging in the Brown Norway rat. Journal of Andrology 28, 229–240.
Spermatozoa have decreased antioxidant enzymatic capacity and increased reactive oxygen species production during aging in the Brown Norway rat.Crossref | GoogleScholarGoogle Scholar |

Yamamoto, M, and Takahashi, Y (2018). The essential role of SIRT1 in hypothalamic-pituitary axis. Frontiers in Endocrinology 9, 605.
The essential role of SIRT1 in hypothalamic-pituitary axis.Crossref | GoogleScholarGoogle Scholar |

Yan, G, Elbadawi, M, and Efferth, T (2020). Multiple cell death modalities and their key features (review). World Academy of Sciences Journal 2, 39–48.
Multiple cell death modalities and their key features (review).Crossref | GoogleScholarGoogle Scholar |

Yu, S-W, Wang, H, Poitras, MF, Coombs, C, Bowers, WJ, Federoff, HJ, Poirier, GG, Dawson, TM, and Dawson, VL (2002). Mediation of poly(ADP-ribose) polymerase-1-dependent cell death by apoptosis-inducing factor. Science 297, 259–263.
Mediation of poly(ADP-ribose) polymerase-1-dependent cell death by apoptosis-inducing factor.Crossref | GoogleScholarGoogle Scholar |

Zamorano, PL, Mahesh, VB, De Sevilla, LM, Chorich, LP, Bhat, GK, and Brann, DW (1997). Expression and localization of the leptin receptor in endocrine and neuroendocrine tissues of the rat. Neuroendocrinology 65, 223–228.
Expression and localization of the leptin receptor in endocrine and neuroendocrine tissues of the rat.Crossref | GoogleScholarGoogle Scholar |