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

l-Carnitine improves endocrine function and folliculogenesis by reducing inflammation, oxidative stress and apoptosis in mice following induction of polycystic ovary syndrome

Zahra Kalhori A , Malek Soleimani Mehranjani orcid.org/0000-0003-0899-003X A C , Mehri Azadbakht B and Mohammad Ali Shariatzadeh A
+ Author Affiliations
- Author Affiliations

A Department of Biology, Faculty of Science, Arak University, PO Box 381-5688138, Arak, Iran.

B Department of Biology, Faculty of Science, Razi University, PO Box 6714967346, Kermanshah, Iran.

C Corresponding author. Email: m-Soleimani@araku.ac.ir

Reproduction, Fertility and Development 31(2) 282-293 https://doi.org/10.1071/RD18131
Submitted: 29 July 2017  Accepted: 29 June 2018   Published: 17 August 2018

Abstract

Polycystic ovary syndrome (PCOS) is related to low levels of serum l-carnitine, which has antioxidant, anti-inflammatory and antiapoptotic properties. The aim of this study was to investigate the effect of l-carnitine on folliculogenesis in mice following induction of PCOS. PCOS was induced by daily injections of testosterone enanthate (1 mg per 100 g, s.c., for 35 days). NMRI mice (21 days old) were divided into four groups (n = 6 per group): Control, Control + l-carnitine, PCOS and PCOS + l-carnitine. Mice were treated with 500 mg kg−1, i.p., l-carnitine every second day for 28 days. Ovaries were studied stereologically and serum concentrations of FSH, LH, testosterone, interleukin (IL)-6 and tumour necrosis factor (TNF)-α were determined using ELISA kits. Serum concentrations of malondialdehyde (MDA) and the ferric ion reducing antioxidant power (FRAP) were also analysed. Apoptosis of follicles was evaluated by terminal deoxyribonucleotidyl transferase-mediated dUTP–digoxigenin nick end-labelling (TUNEL). CD31 was assessed immunohistochemically. Data were analysed using one-way analysis of variance (ANOVA) and Tukey’s test, differences considered significant at P < 0.05.The total volume of the ovary, cortex volume, oocyte volume, zona pellucida thickness and the number of antral follicles increased significantly, whereas the number of primary and preantral follicles decreased significantly, in the PCOS + l-carnitine versus PCOS group. In the PCOS + l-carnitine group, serum concentrations of FSH and FRAP increased significantly, whereas there were significant decreases in serum concentrations of testosterone, LH, MDA, IL-6 and TNF-α, as well as in the percentage of TUNEL-positive apoptotic cells, compared with the PCOS group. l-Carnitine improves folliculogenesis and is therefore suggested as a therapeutic supplement in the treatment of PCOS.

Additional keywords: androgen, oocyte, ovulation.


References

Ali, H. I., Elsadawy, M. E., and Khater, N. H. (2016). Ultrasound assessment of polycystic ovaries: ovarian volume and morphology; which is more accurate in making the diagnosis?! Egyptian Journal of Radiology and Nuclear Medicine 47, 347–350.
Ultrasound assessment of polycystic ovaries: ovarian volume and morphology; which is more accurate in making the diagnosis?!Crossref | GoogleScholarGoogle Scholar |

Allahbadia, G. N., and Merchant, R. (2011). Polycystic ovary syndrome and impact on health. Middle East Fertil. Soc. J. 16, 19–37.
Polycystic ovary syndrome and impact on health.Crossref | GoogleScholarGoogle Scholar |

Amato, G., Conte, M., Mazziotti, G., Lalli, E., Vitolo, G., Tucker, A. T., Bellastella, A., Carella, C., and Izzo, A. (2003). Serum and follicular fluid cytokines in polycystic ovary syndrome during stimulated cycles. Obstet. Gynecol. 101, 1177–1182.

Bavari, M., Tabandeh, M. R., Najafzadeh Varzi, H., and Bahramzadeh, S. (2016). Neuroprotective, antiapoptotic and antioxidant effects of l-carnitine against caffeine-induced neurotoxicity in SH-SY5Y neuroblastoma cell line. Drug Chem. Toxicol. 39, 157–166.
Neuroprotective, antiapoptotic and antioxidant effects of l-carnitine against caffeine-induced neurotoxicity in SH-SY5Y neuroblastoma cell line.Crossref | GoogleScholarGoogle Scholar |

Beloosesky, R., Gold, R., Almog, B., Sasson, R., Dantes, A., Land-Bracha, A., Hirshi, L., Itskovitz-Eldor, J., Lessing, J. B., Homburg, R., and Amsterdam, A. (2004). Induction of polycystic ovary by testosterone in immature female rate: modulation of apoptosis and attenuation of glucose/insulin ratio. Int. J. Mol. Med. 14, 207–215.

Benzie, I. F., and Strain, J. J. (1996). The ferric reducing ability of plasma (FRAP) as a measure of ‘antioxidant power’ the FRAP assay. Anal. Biochem. 239, 70–76.
The ferric reducing ability of plasma (FRAP) as a measure of ‘antioxidant power’ the FRAP assay.Crossref | GoogleScholarGoogle Scholar |

Buege, J. A., and Aust, S. D. (1978). Microsomal lipid peroxidation. Methods Enzymol. 52, 302–310.
Microsomal lipid peroxidation.Crossref | GoogleScholarGoogle Scholar |

Calado, A. M., Rocha, E., Colaco, A., and Sousa, M. (2003). Stereological characterization of bovine (Bos taurus) cumulus–oocyte complexes aspirated from small antral follicles during the metestrus and proestrus phases. Theriogenology 60, 429–443.
Stereological characterization of bovine (Bos taurus) cumulus–oocyte complexes aspirated from small antral follicles during the metestrus and proestrus phases.Crossref | GoogleScholarGoogle Scholar |

Cheraghi, E., Soleimani Mehranjani, M., Shariatzadeh, M. A., Nasr Esfahani, M. H., and Ebrahimi, Z. (2016). N-Acetylcysteine improves oocyte and embryo quality in polycystic ovary syndrome patients undergoing intracytoplasmic sperm injection: an alternative to metformin. Reprod. Fertil. Dev. 28, 723–731.
N-Acetylcysteine improves oocyte and embryo quality in polycystic ovary syndrome patients undergoing intracytoplasmic sperm injection: an alternative to metformin.Crossref | GoogleScholarGoogle Scholar |

Diamanti-Kandarakis, E. (2008). Polycystic ovarian syndrome: pathophysiology, molecular aspects and clinical implications. Expert Rev. Mol. Med. 10, e3.
Polycystic ovarian syndrome: pathophysiology, molecular aspects and clinical implications.Crossref | GoogleScholarGoogle Scholar |

Duranay, M., Akay, H., Yilmaz, F. M., Senes, M., Tekeli, N., and Yücel, D. (2006). Effects of l-carnitine infusions on inflammatory and nutritional markers in haemodialysis patients. Nephrol. Dial. Transplant. 21, 3211–3214.
Effects of l-carnitine infusions on inflammatory and nutritional markers in haemodialysis patients.Crossref | GoogleScholarGoogle Scholar |

Eagleson, C. A., Gingrich, M. B., Pastor, C. L., Arora, T. K., Burt, C. M., Evans, W. S., and Marshall, J. C. (2000). Polycystic ovary syndrome: evidence that flutamide restores sensitivity of the gonadotropin-releasing hormone pulse generator to inhibition by estradiol and progesterone. J. Clin. Endocrinol. Metab. 85, 4047–4052.

Fenkci, S. M., Fenkci, V., Oztekinb, O., Rota, S., and Karagenc, N. (2008). Serum total l-carnitine levels in non-obese women with polycystic ovary syndrome. Hum. Reprod. 23, 1602–1606.
Serum total l-carnitine levels in non-obese women with polycystic ovary syndrome.Crossref | GoogleScholarGoogle Scholar |

Ferrando, R. E., Nyengaard, J. R., Hays, S. R., Fahy, J. V., and Woodruff, P. G. (2003). Applying stereology to measure thickness of the basement membrane zone in bronchial biopsy specimens. J. Allergy Clin. Immunol. 112, 1243–1245.
Applying stereology to measure thickness of the basement membrane zone in bronchial biopsy specimens.Crossref | GoogleScholarGoogle Scholar |

Franks, S., Mason, H., and Willis, D. (2000). Follicular dynamics in the polycystic ovary syndrome. Mol. Cell. Endocrinol. 163, 49–52.
Follicular dynamics in the polycystic ovary syndrome.Crossref | GoogleScholarGoogle Scholar |

González, F. (2012). Inflammation in polycystic ovary syndrome: underpinning of insulin resistance and ovarian dysfunction. Steroids 77, 300–305.
Inflammation in polycystic ovary syndrome: underpinning of insulin resistance and ovarian dysfunction.Crossref | GoogleScholarGoogle Scholar |

Gülçin, L. (2006). Antioxidant and antiradical activities of l-carnitine. Life. Sci. 78, 803–811.
Antioxidant and antiradical activities of l-carnitine.Crossref | GoogleScholarGoogle Scholar |

Hubscher, C. H., Brooks, D. L., and Johnson, J. R. (2005). A quantitative method for assessing stages of the rat estrous cycle. Biotech. Histochem. 80, 79–87.
A quantitative method for assessing stages of the rat estrous cycle.Crossref | GoogleScholarGoogle Scholar |

Ikeda, K., Baba, T., Morishita, M., Honnma, H., Endo, T., Kiya, T., and Saito, T. (2014). Long-term treatment with dehydroepiandrosterone may lead to follicular atresia through interaction with anti-Mullerian hormone. J. Ovarian Res. 7, 46.

Ismail, A. M., Hamed, A. H., Saso, S., and Thabet, H. H. (2014). Adding l-carnitine to clomiphene resistant PCOS women improves the quality of ovulation and the pregnancy rate. A randomized clinical trial. Eur. J. Obstet. Gynecol. Reprod. Biol. 180, 148–152.
Adding l-carnitine to clomiphene resistant PCOS women improves the quality of ovulation and the pregnancy rate. A randomized clinical trial.Crossref | GoogleScholarGoogle Scholar |

Jonard, S., and Dewailly, D. (2004). The follicular excess in polycystic ovaries, due to intraovarian hyperandrogenism, may be the main culprit for the follicular arrest. Hum. Reprod. Update 10, 107–117.
The follicular excess in polycystic ovaries, due to intraovarian hyperandrogenism, may be the main culprit for the follicular arrest.Crossref | GoogleScholarGoogle Scholar |

Kalhori, Z., and Azadbakht, M. (2015). Hydrostatic pressure improves maturation of oocyte in cultures of follicles derived from mouse polycystic ovary. Acta. Endocrinol. (Buc) 11, 26–31.
Hydrostatic pressure improves maturation of oocyte in cultures of follicles derived from mouse polycystic ovary.Crossref | GoogleScholarGoogle Scholar |

Kanter, M., Topcu-Tarladacalisir, Y., and Parlar, S. (2010). Antiapoptotic effect of l-carnitine on testicular irradiation in rats. J. Mol. Histol. 41, 121–128.
Antiapoptotic effect of l-carnitine on testicular irradiation in rats.Crossref | GoogleScholarGoogle Scholar |

Karbalay-Doust, S., and Noorafshan, A. (2012). Stereological estimation of ovarian oocyte volume, surface area and number: application on mice treated with nandrolone decanoate. Folia Histochem. Cytobiol. 50, 275–279.
Stereological estimation of ovarian oocyte volume, surface area and number: application on mice treated with nandrolone decanoate.Crossref | GoogleScholarGoogle Scholar |

Lee, B. J., Lin, J. S., Lin, Y. C., and Lin, P. T. (2015). Anti-inflammation effects of l-carnitine supplementation (1000 mg/d) in coronary artery disease patients. Nutrition 31, 475–479.
Anti-inflammation effects of l-carnitine supplementation (1000 mg/d) in coronary artery disease patients.Crossref | GoogleScholarGoogle Scholar |

Li, J. L., Wang, Q. Y., Luan, H. Y., Kang, Z. C., and Wang, C. B. (2012). Effects of l-carnitine against oxidative stress in human hepatocytes: involvement of peroxisome proliferator-activated receptor alpha. J. Biomed. Sci. 19, 32.
Effects of l-carnitine against oxidative stress in human hepatocytes: involvement of peroxisome proliferator-activated receptor alpha.Crossref | GoogleScholarGoogle Scholar |

Mahmoodi, M., Soleimani Mehranjani, M., Shariatzadeh, S. M. A., Eimani, H., and Shahverdi, A. H. (2014). Effects of erythropoietin on ischemia, follicular survival, and ovarian function in ovarian grafts. Reproduction 147, 733–741.
Effects of erythropoietin on ischemia, follicular survival, and ovarian function in ovarian grafts.Crossref | GoogleScholarGoogle Scholar |

Mahmoodi, M., Soleimani Mehranjani, M., Shariatzadeh, S. M. A., Eimani, H., and Shahverdi, A. H. (2015). N-Acetylcysteine improves function and follicular survival in mice ovarian grafts through inhibition of oxidative stress. Reprod. Biomed. Online 30, 101–110.
N-Acetylcysteine improves function and follicular survival in mice ovarian grafts through inhibition of oxidative stress.Crossref | GoogleScholarGoogle Scholar |

Marquard, K. L., Stephens, S. M., Jungheim, E. S., Ratts, V. S., Odem, R. R., Lanzendorf, S., and Moley, K. H. (2011). Polycystic ovary syndrome and maternal obesity affect oocyte size in in vitro fertilization/intracytoplasmic sperm injection cycles. Fertil. Steril. 95, 2146–2149.
Polycystic ovary syndrome and maternal obesity affect oocyte size in in vitro fertilization/intracytoplasmic sperm injection cycles.Crossref | GoogleScholarGoogle Scholar |

Murri, M., Luque-Ramı’rez, M., Insenser, M., Ojeda-Ojeda, M., and Escobar-Morreale, H. F. (2013). Circulating markers of oxidative stress and polycystic ovary syndrome (PCOS): a systematic review and meta-analysis. Hum. Reprod. Update 19, 268–288.
Circulating markers of oxidative stress and polycystic ovary syndrome (PCOS): a systematic review and meta-analysis.Crossref | GoogleScholarGoogle Scholar |

Myers, M., Britt, K. L., Wreford, N. G., Ebling, F. J., and Kerr, J. B. (2004). Methods for quantifying follicular numbers within the mouse ovary. Reproduction 127, 569–580.
Methods for quantifying follicular numbers within the mouse ovary.Crossref | GoogleScholarGoogle Scholar |

Noorafshan, A., Ahmadi, M., Mesbah, S. F., and Karbalay-Doust, S. (2013). Stereological study of the effects of letrozole and estradiol valerate treatment on the ovary of rats. Clin. Exp. Reprod. Med. 40, 115–121.
Stereological study of the effects of letrozole and estradiol valerate treatment on the ovary of rats.Crossref | GoogleScholarGoogle Scholar |

Ota, H., Fukushima, M., and Maki, M. (1983). Endocrinological and histological aspects of the process of polycystic ovary formation in the rat treated with testosterone propionate. Tohoku. J. Exp. Med. 140, 121–131.

Ouladsahebmadarek, E., and Khaki, A. (2014). Ultra-structural study by transmission electron microscopy: effect of omega-3 on ovary cell organelles after experimental induced poly cystic ovary syndrome. Int. J. Women’s Health Reprod. Sci. 2, 186–194.
Ultra-structural study by transmission electron microscopy: effect of omega-3 on ovary cell organelles after experimental induced poly cystic ovary syndrome.Crossref | GoogleScholarGoogle Scholar |

Pellatt, L., Hanna, L., Brincat, M., Galea, R., Brain, H., Whitehead, S., and Mason, H. (2007). Granulosa cell production of antimullerian hormone is increased in polycystic ovaries. J. Clin. Endocrinol. Metab. 92, 240–245.
Granulosa cell production of antimullerian hormone is increased in polycystic ovaries.Crossref | GoogleScholarGoogle Scholar |

Qiu, S., Wu, C., Lin, F., Chen, L., Huang, Z., and Jiang, Z. (2009). Exercise training improved insulin sensitivity and ovarian morphology in rats with polycystic ovary syndrome. Horm. Metab. Res. 41, 880–885.
Exercise training improved insulin sensitivity and ovarian morphology in rats with polycystic ovary syndrome.Crossref | GoogleScholarGoogle Scholar |

Rice, S., Ojha, K., Whitehead, S., and Mason, H. (2007). Stage-specific expression of androgen receptor, follicle-stimulating hormone receptor, and antimullerian hormone type II receptor in single, isolated, human preantral follicles: relevance to polycystic ovaries. J. Clin. Endocrinol. Metab. 92, 1034–1040.
Stage-specific expression of androgen receptor, follicle-stimulating hormone receptor, and antimullerian hormone type II receptor in single, isolated, human preantral follicles: relevance to polycystic ovaries.Crossref | GoogleScholarGoogle Scholar |

Samimi, M., Jamilian, M., Afshar Ebrahimi, F., Rahimi, M., Tajbakhsh, B., and Asemi, Z. (2016). Oral carnitine supplementation reduces body weight and insulin resistance in women with polycystic ovary syndrome: a randomized, double-blind, placebo-controlled trial. Clin. Endocrinol. (Oxf.) 84, 851–857.
Oral carnitine supplementation reduces body weight and insulin resistance in women with polycystic ovary syndrome: a randomized, double-blind, placebo-controlled trial.Crossref | GoogleScholarGoogle Scholar |

Samoto, T., Maruo, T., Ladines-Llave, C. A., Matsuo, H., Deguchi, J., Barnea, E. R., and Mochizuki, M. (1993). Insulin receptor expression in follicular and stromal compartments of the human ovary over the course of follicular growth, regression and atresia. Endocr. J. 40, 715–726.
Insulin receptor expression in follicular and stromal compartments of the human ovary over the course of follicular growth, regression and atresia.Crossref | GoogleScholarGoogle Scholar |

Skrtic, A., Sokolic, L., Borovecki, A., Rosa, J., and Fenzl, V. (2011). Immunohistochemical localization of CD31, NOTCH1 and JAGGED1 proteins in experimentally induced polycystic ovaries of immature rats. Acta Histochem. 113, 262–269.
Immunohistochemical localization of CD31, NOTCH1 and JAGGED1 proteins in experimentally induced polycystic ovaries of immature rats.Crossref | GoogleScholarGoogle Scholar |

Steiber, A., Kerner, J., and Hoppel, C. L. (2004). Carnitine: a nutritional, biosynthetic, and functional perspective. Mol. Aspects Med. 25, 455–473.
Carnitine: a nutritional, biosynthetic, and functional perspective.Crossref | GoogleScholarGoogle Scholar |

Tessaro, I., Modina, S. C., Franciosi, F., Sivelli, G., Terzaghi, L., Lodde, V., and Luciano, A. M. (2015). Effect of oral administration of low-dose follicle stimulating hormone on hyperandrogenized mice as a model of polycystic ovary syndrome. J. Ovarian Res. 8, 64.
Effect of oral administration of low-dose follicle stimulating hormone on hyperandrogenized mice as a model of polycystic ovary syndrome.Crossref | GoogleScholarGoogle Scholar |

Tyndall, V., Broyde, M., Sharpe, R., Welsh, M., Drake, A. J., and McNeilly, A. S. (2012). Effect of androgen treatment during foetal and/or neonatal life on ovarian function in prepubertal and adult rats. Reproduction 143, 21–33.
Effect of androgen treatment during foetal and/or neonatal life on ovarian function in prepubertal and adult rats.Crossref | GoogleScholarGoogle Scholar |

Usta, U., Inan, M., Erbas, H., Aydogdu, N., Puyan, F. O., and Altaner, S. (2008). Tissue damage in rat ovaries subjected to torsion and detorsion: effects of l-carnitine and N-acetyl cysteine. Pediatr. Surg. Int. 24, 567–573.
Tissue damage in rat ovaries subjected to torsion and detorsion: effects of l-carnitine and N-acetyl cysteine.Crossref | GoogleScholarGoogle Scholar |

Valko, M., Leibfritz, D., Moncol, J., Cronin, M. T., Mazur, M., and Telser, J. (2007). Free radicals and antioxidants in normal physiological functions and human disease. Int. J. Biochem. Cell Biol. 39, 44–84.
Free radicals and antioxidants in normal physiological functions and human disease.Crossref | GoogleScholarGoogle Scholar |

Velez, L. M., Heber, M. F., Ferreira, S. R., Abruzzese, G. A., Reynoso, R. M., and Motta, A. B. (2015). Effect of hyperandrogenism on ovarian function. Reproduction 149, 577–585.
Effect of hyperandrogenism on ovarian function.Crossref | GoogleScholarGoogle Scholar |

Vendola, K., Zhou, J., Wang, J., and Bondy, C. A. (1999). Androgens promote insulin-like growth factor-I and insulin-like growth factor-I receptor gene expression in the primate ovary. Hum. Reprod. 14, 2328–2332.
Androgens promote insulin-like growth factor-I and insulin-like growth factor-I receptor gene expression in the primate ovary.Crossref | GoogleScholarGoogle Scholar |

Vigerust, N. F., Bohov, P., Bjorndal, B., Seifert, R., Nygård, O., Svardal, A., Glintborg, D., Berge, R. K., and Gaster, M. (2012). Free carnitine and acylcarnitines in obese patients with polycystic ovary syndrome and effects of pioglitazone treatment. Fertil. Steril. 98, 1620–1626.
Free carnitine and acylcarnitines in obese patients with polycystic ovary syndrome and effects of pioglitazone treatment.Crossref | GoogleScholarGoogle Scholar |

Wassarman, P. M., and Litscher, E. S. (2012). Influence of the zona pellucida of the mouse egg on folliculogenesis and fertility. Int. J. Dev. Biol. 56, 833–839.
Influence of the zona pellucida of the mouse egg on folliculogenesis and fertility.Crossref | GoogleScholarGoogle Scholar |

Wood, J. R., Dumesic, D. A., Abbott, D. H., and Strauss, J. F. (2007). Molecular abnormalities in oocytes from women with polycystic ovary syndrome revealed by microarray analysis. J. Clin. Endocrinol. Metab. 92, 705–713.
Molecular abnormalities in oocytes from women with polycystic ovary syndrome revealed by microarray analysis.Crossref | GoogleScholarGoogle Scholar |

Xiong, Y. L., Liang, X. Y., Yang, X., Li, Y., and Wei, L. N. (2011). Low-grade chronic inflammation in the peripheral blood and ovaries of women with polycystic ovarian syndrome. Eur. J. Obstet. Gynecol. Reprod. Biol. 159, 148–150.
Low-grade chronic inflammation in the peripheral blood and ovaries of women with polycystic ovarian syndrome.Crossref | GoogleScholarGoogle Scholar |

Yang, H., Lee, H. H., Lee, H. C., Ko, D. S., and Kim, S. S. (2008). Assessment of vascular endothelial growth factor expression and apoptosis in the ovarian graft: can exogenous gonadotropin promote angiogenesis after ovarian transplantation? Fertil. Steril. 90, 1550–1558.
Assessment of vascular endothelial growth factor expression and apoptosis in the ovarian graft: can exogenous gonadotropin promote angiogenesis after ovarian transplantation?Crossref | GoogleScholarGoogle Scholar |

Yang, F., Ruan, Y. C., Yang, Y. J., Wang, K., Liang, S. S., Han, Y. B., Teng, X. M., and Yang, J. Z. (2015). Follicular hyperandrogenism downregulates aromatase in luteinized granulosa cells in polycystic ovary syndrome women. Reproduction 150, 289–296.
Follicular hyperandrogenism downregulates aromatase in luteinized granulosa cells in polycystic ovary syndrome women.Crossref | GoogleScholarGoogle Scholar |

Yeh, J., and Kim, H. H. (1996). Polycystic ovary syndrome (PCOS): the possible roles of apoptosis in human granulosa cells. In ‘Polycystic Ovarian Syndrome’. (Ed. R. J. Chang.) pp. 51–70. (Springer-Verlag: New York.)

Zhai, H. L., Wu, H., Xu, H., Weng, P., Xia, F. Z., Chen, Y., and Lu, Y. L. (2012). Trace glucose and lipid metabolism in high androgen and high-fat diet induced polycystic ovary syndrome rats. Reprod. Biol. Endocrinol. 10, 5.
Trace glucose and lipid metabolism in high androgen and high-fat diet induced polycystic ovary syndrome rats.Crossref | GoogleScholarGoogle Scholar |

Zhang, Q., Wang, S. M., Yao, P. B., Zhang, L., Zhang, Y. J., Chen, R. X., Fu, Y., and Zhang, J. M. (2015). Effects of l-carnitine on follicular survival and graft function following autotransplantation of cryopreserved–thawed ovarian tissues. Cryobiology 71, 135–140.
Effects of l-carnitine on follicular survival and graft function following autotransplantation of cryopreserved–thawed ovarian tissues.Crossref | GoogleScholarGoogle Scholar |