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

Sulfasalazine exposure during pregnancy and lactation: reproductive outcomes in male rat offspring

Simone Forcato https://orcid.org/0000-0002-2892-7575 A , Ana Beatriz de Oliveira Aquino A , Lorena I. Borges A , Maria Luiza Francisconi Lubanco Thomé A , Júlia O. Bilibio A , Hannah Hamada Mendonça Lens B , Rafaela P. Erthal C , Flávia A. Guarnier B , Glaura Scantamburlo Alves Fernandes https://orcid.org/0000-0002-6043-1131 C and Daniela Cristina Ceccatto Gerardin https://orcid.org/0000-0002-8692-8007 A *
+ Author Affiliations
- Author Affiliations

A Department of Physiological Sciences, Londrina State University, Londrina, PR, Brazil.

B Department of General Pathology, Londrina State University, Londrina, PR, Brazil.

C Department of General Biology, Londrina State University, Londrina, PR, Brazil.

* Correspondence to: dcgerardin@uel.br

Handling Editor: Andrew Pask

Reproduction, Fertility and Development 35(8) 469-479 https://doi.org/10.1071/RD22240
Published online: 21 April 2023

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

Abstract

Context: Sulfasalazine (SAS) is a drug prescribed for pregnant and breastfeeding women with chronic inflammatory bowel diseases. SAS treatment induces transitory infertility in both adult men and male rats. Although SAS crosses the placenta and passes into maternal milk, the consequences of maternal SAS exposure on the reproductive development of male offspring needs further study.

Aims: The current study evaluated whether maternal SAS exposure interferes with the reproductive development of male rat offspring in the neonatal, infant, pubertal and adulthood periods.

Methods: Pregnant Wistar rats (n = 10/group) received 300 mg/kg/day of SAS dissolved in carboxymethyl cellulose (CMC), by gavage, from gestational day 0 to lactation day 21, and 3 mg/kg/day of folic acid during gestation. The control group received CMC.

Key results: During puberty, maternal SAS exposure increased the total length of seminiferous tubules, and round cells were observed in the lumen of caput and cauda epididymis. Moreover, SAS induced oxidative stress-related alterations in the testes of infant and adolescent rats.

Conclusions: Although maternal SAS treatment caused reproductive alterations in infant and adolescent male rats, in adulthood, there were no impairments in sperm parameters that could compromise fertility.

Implications: This study investigated the consequences of maternal exposure to SAS on the reproductive development of male rat offspring from birth to adulthood, employing a human-relevant dose. Thus, this study provides information for better understanding of SAS treatment during critical periods of development.

Keywords: development, gestation, lactation, oxidative stress, pharmacology, reproduction, sperm, testis.


References

Abbasi, M, Mousavi, MJ, Jamalzehi, S, Alimohammadi, R, Bezvan, MH, Mohammadi, H, and Aslani, S (2019). Strategies toward rheumatoid arthritis therapy; the old and the new. Journal of Cellular Physiology 234, 10018–10031.
Strategies toward rheumatoid arthritis therapy; the old and the new.Crossref | GoogleScholarGoogle Scholar |

Alonso, V, Linares, V, Bellés, M, Albina, ML, Sirvent, JJ, Domingo, JL, and Sánchez, DJ (2009). Sulfasalazine induced oxidative stress: a possible mechanism of male infertility. Reproductive Toxicology 27, 35–40.
Sulfasalazine induced oxidative stress: a possible mechanism of male infertility.Crossref | GoogleScholarGoogle Scholar |

Berlin, CM, and Yaffe, SJ (1980). Disposition of Salicylazosulfapyridine (Azulfidine) and metabolites in human breast milk. Developmental Pharmacology and Therapeutics 1, 31–39.
Disposition of Salicylazosulfapyridine (Azulfidine) and metabolites in human breast milk.Crossref | GoogleScholarGoogle Scholar |

Birnie, GG, Mcleod, TI, and Watkinson, G (1981). Incidence of sulphasalazine-induced male infertility. Gut 22, 452–455.
Incidence of sulphasalazine-induced male infertility.Crossref | GoogleScholarGoogle Scholar |

Czeizel, AE, Dudás, I, Vereczkey, A, and Bánhidy, F (2013). Folate deficiency and folic acid supplementation: the prevention of neural-tube defects and congenital heart defects. Nutrients 5, 4760–4775.
Folate deficiency and folic acid supplementation: the prevention of neural-tube defects and congenital heart defects.Crossref | GoogleScholarGoogle Scholar |

Dik, B, Sonmez, G, Faki, HE, and Bahcivan, E (2018). Sulfasalazine treatment can cause a positive effect on LPS-induced endotoxic rats. Experimental Animals 67, 403–412.
Sulfasalazine treatment can cause a positive effect on LPS-induced endotoxic rats.Crossref | GoogleScholarGoogle Scholar |

Ellman, GL (1959). Tissue sulfhydryl groups. Archives of Biochemistry and Biophysics 82, 70–77.
Tissue sulfhydryl groups.Crossref | GoogleScholarGoogle Scholar |

Esbjörner, E, Järnerot, G, and Wranne, L (1987). Sulphasalazine and sulphapyridine serum levels in children to mothers treated with sulphasalazine during pregnancy and lactation. Acta Paediatrica Scandinavica 76, 137–142.
Sulphasalazine and sulphapyridine serum levels in children to mothers treated with sulphasalazine during pregnancy and lactation.Crossref | GoogleScholarGoogle Scholar |

Favareto, APA, Fernandez, CDB, da Silva, DAF, Anselmo-Franci, JA, and Kempinas, WDG (2011a). Persistent impairment of testicular histology and sperm motility in adult rats treated with Cisplatin at Peri-puberty. Basic & Clinical Pharmacology & Toxicology 109, 85–96.
Persistent impairment of testicular histology and sperm motility in adult rats treated with Cisplatin at Peri-puberty.Crossref | GoogleScholarGoogle Scholar |

Favareto, APA, de Toledo, FC, and Kempinas, WDG (2011b). Paternal treatment with cisplatin impairs reproduction of adult male offspring in rats. Reproductive Toxicology 32, 425–433.
Paternal treatment with cisplatin impairs reproduction of adult male offspring in rats.Crossref | GoogleScholarGoogle Scholar |

Fernandes, GSA, Arena, AC, Fernandez, CDB, Mercadante, A, Barbisan, LF, and Kempinas, WG (2007). Reproductive effects in male rats exposed to diuron. Reproductive Toxicology 23, 106–112.
Reproductive effects in male rats exposed to diuron.Crossref | GoogleScholarGoogle Scholar |

Filler R (1993) Methods for evaluation of rats epididymal sperm morphology. In ‘Male reproductive toxicology’. (Eds RE Chaplin, JJ Heindel) pp. 334–343. (Academic Press: San Diego, CA, USA)

Flint, J, Panchal, S, Hurrell, A, Van De, VM, Gayed, M, Schreiber, K, Arthanari, S, Cunningham, J, Flanders, L, Moore, L, Crossley, A, Purushotham, N, Desai, A, Piper, M, Nisar, M, Khamashta, M, Williams, D, Gordon, C, Giles, I, and Standards, B (2016). BSR and BHPR guideline on prescribing drugs in pregnancy and breastfeeding – Part I: standard and biologic disease modifying anti-rheumatic drugs and corticosteroids. Rheumatology 55, 1693–1697.
BSR and BHPR guideline on prescribing drugs in pregnancy and breastfeeding – Part I: standard and biologic disease modifying anti-rheumatic drugs and corticosteroids.Crossref | GoogleScholarGoogle Scholar |

Forcato, S, Novi, DRBdS, Costa, NO, Borges, LI, Góes, MLMd, Ceravolo, GS, and Gerardin, DCC (2017). In utero and lactational exposure to metformin induces reproductive alterations in male rat offspring. Reproductive Toxicology 74, 48–58.
In utero and lactational exposure to metformin induces reproductive alterations in male rat offspring.Crossref | GoogleScholarGoogle Scholar |

Forcato, S, de Oliveira Aquino, AB, de Moura Camparoto, N, Lens, HHM, Guarnier, FA, Kiss, ACI, and Gerardin, DCC (2022). Sulfasalazine exposure during pregnancy and lactation induces alterations in reproductive behavior in adult female rat offspring. Life Sciences 293, 120303.
Sulfasalazine exposure during pregnancy and lactation induces alterations in reproductive behavior in adult female rat offspring.Crossref | GoogleScholarGoogle Scholar |

França, LR, and Godinho, CL (2003). Testis morphometry, seminiferous epithelium cycle length, and daily sperm production in domestic cats (Felis catus). Biology of Reproduction 68, 1554–1561.
Testis morphometry, seminiferous epithelium cycle length, and daily sperm production in domestic cats (Felis catus).Crossref | GoogleScholarGoogle Scholar |

Fukushima, T, Kato, M, Adachi, T, Hamada, Y, Horimoto, M, Komiyama, M, Mori, C, and Horii, I (2005). Effects of sulfasalazine on sperm acrosome reaction and gene expression in the male reproductive organs of rats. Toxicological Sciences 85, 675–682.
Effects of sulfasalazine on sperm acrosome reaction and gene expression in the male reproductive organs of rats.Crossref | GoogleScholarGoogle Scholar |

Fukushima, T, Hamada, Y, Komiyama, M, Matsuno, Y, Mori, C, and Horii, I (2007). Early changes in sperm motility, acrosome reaction, and gene expression of reproductive organs in rats treated with sulfasalazine. Reproductive Toxicology 23, 153–157.
Early changes in sperm motility, acrosome reaction, and gene expression of reproductive organs in rats treated with sulfasalazine.Crossref | GoogleScholarGoogle Scholar |

Gallavan, RH, Holson, JF, Stump, DG, Knapp, JF, and Reynolds, VL (1999). Interpreting the toxicologic significance of alterations in anogenital distance: potential for confounding effects of progeny body weights. Reproductive Toxicology 13, 383–390.
Interpreting the toxicologic significance of alterations in anogenital distance: potential for confounding effects of progeny body weights.Crossref | GoogleScholarGoogle Scholar |

Gan, H-T, Chen, Y-Q, and Ouyang, Q (2005). Sulfasalazine inhibits activation of nuclear factor-κB in patients with ulcerative colitis. Journal of Gastroenterology and Hepatology 20, 1016–1024.
Sulfasalazine inhibits activation of nuclear factor-κB in patients with ulcerative colitis.Crossref | GoogleScholarGoogle Scholar |

Gonzalez Flecha, B, Llesuy, S, and Boveris, A (1991). Hydroperoxide-initiated chemiluminescence: an assay for oxidative stress in biopsies of heart, liver, and muscle. Free Radical Biology and Medicine 10, 93–100.
Hydroperoxide-initiated chemiluminescence: an assay for oxidative stress in biopsies of heart, liver, and muscle.Crossref | GoogleScholarGoogle Scholar |

Goodman L, Gilman A (2011) ‘The pharmacological basis of therapeutics: digital edition’. (Eds LL Brunton, JS Lazo, KL Parker). (McGraw-Hill Companies, Inc.: New York, NY)

Guarnier, FA, Cecchini, AL, Suzukawa, AA, Maragno, ALGC, Simão, AN, Gomes, MD, and Cecchini, R (2010). Time course of skeletal muscle loss and oxidative stress in rats with walker 256 solid tumor. Muscle & Nerve 42, 950–958.
Time course of skeletal muscle loss and oxidative stress in rats with walker 256 solid tumor.Crossref | GoogleScholarGoogle Scholar |

Götestam Skorpen, C, Hoeltzenbein, M, Tincani, A, Fischer-Betz, R, Elefant, E, Chambers, C, da Silva, J, Nelson-Piercy, C, Cetin, I, Costedoat-Chalumeau, N, Dolhain, R, Förger, F, Khamashta, M, Ruiz-Irastorza, G, Zink, A, Vencovsky, J, Cutolo, M, Caeyers, N, Zumbühl, C, and Østensen, M (2016). The EULAR points to consider for use of antirheumatic drugs before pregnancy, and during pregnancy and lactation. Annals of the Rheumatic Diseases 75, 795–810.
The EULAR points to consider for use of antirheumatic drugs before pregnancy, and during pregnancy and lactation.Crossref | GoogleScholarGoogle Scholar |

Hernández-Díaz, S, Werler, MM, Walker, AM, and Mitchell, AA (2000). Folic acid antagonists during pregnancy and the risk of birth defects. New England Journal of Medicine 343, 1608–1614.
Folic acid antagonists during pregnancy and the risk of birth defects.Crossref | GoogleScholarGoogle Scholar |

Higashi, CM, Matsumoto, AK, Gameiro, JG, Moura, KF, Higachi, L, Oliveira, LC, Barbosa, DS, Moreira, EG, and Ceravolo, GS (2016). Does fish oil or folic acid prevent vascular changes in female progeny caused by maternal exposure to fluoxetine? Life Sciences 152, 165–170.
Does fish oil or folic acid prevent vascular changes in female progeny caused by maternal exposure to fluoxetine?Crossref | GoogleScholarGoogle Scholar |

Horimoto, M, Isobe, Y, Isogai, Y, and Tachibana, M (2000). Rat epididymal sperm motion changes induced by ethylene glycol monoethyl ether, sulfasalazine, and 2,5-hexandione. Reproductive Toxicology 14, 55–63.
Rat epididymal sperm motion changes induced by ethylene glycol monoethyl ether, sulfasalazine, and 2,5-hexandione.Crossref | GoogleScholarGoogle Scholar |

Hoyt, JA, Fisher, LF, and Swisher, DK (1995). Short-term male reproductive toxicity study with sulfasalazine in the rat. Reproductive Toxicology 9, 315–326.
Short-term male reproductive toxicity study with sulfasalazine in the rat.Crossref | GoogleScholarGoogle Scholar |

Inomata, T, Eguchi, Y, and Nakamura, T (1985). Development of the external genitalia in rat fetuses. Experimental Animals 34, 439–444.
Development of the external genitalia in rat fetuses.Crossref | GoogleScholarGoogle Scholar |

Jacobson, CD, and Gorski, RA (1981). Neurogenesis of the sexually dimorphic nucleus of the preoptic area in the rat. The Journal of Comparative Neurology 196, 519–529.
Neurogenesis of the sexually dimorphic nucleus of the preoptic area in the rat.Crossref | GoogleScholarGoogle Scholar |

Jansen, G, van der Heijden, J, Oerlemans, R, Lems, WF, Ifergan, I, Scheper, RJ, Assaraf, YG, and Dijkmans, BAC (2004). Sulfasalazine is a potent inhibitor of the reduced folate carrier: implications for combination therapies with methotrexate in rheumatoid arthritis. Arthritis & Rheumatism 50, 2130–2139.
Sulfasalazine is a potent inhibitor of the reduced folate carrier: implications for combination therapies with methotrexate in rheumatoid arthritis.Crossref | GoogleScholarGoogle Scholar |

Järnerot, G, Into-Malmberg, M-B, and Esbjörner, E (1981). Placental transfer of sulphasalazine and sulphapyridine and some of its metabolites. Scandinavian Journal of Gastroenterology 16, 693–697.
Placental transfer of sulphasalazine and sulphapyridine and some of its metabolites.Crossref | GoogleScholarGoogle Scholar |

Kaneko, T, Iuchi, Y, Kobayashi, T, Fujii, T, Saito, H, Kurachi, H, and Fujii, J (2002). The expression of glutathione reductase in the male reproductive system of rats supports the enzymatic basis of glutathione function in spermatogenesis. European Journal of Biochemistry 269, 1570–1578.
The expression of glutathione reductase in the male reproductive system of rats supports the enzymatic basis of glutathione function in spermatogenesis.Crossref | GoogleScholarGoogle Scholar |

Khan, AKA, and Truelove, SC (1979). Placental and mammary transfer of sulphasalazine. British Medical Journal 2, 1553.
Placental and mammary transfer of sulphasalazine.Crossref | GoogleScholarGoogle Scholar |

Lappas, M, Permezel, M, Georgiou, HM, and Rice, GE (2002). Nuclear factor kappa B regulation of proinflammatory cytokines in human gestational tissues in vitro. Biology of Reproduction 67, 668–673.
Nuclear factor kappa B regulation of proinflammatory cytokines in human gestational tissues in vitro.Crossref | GoogleScholarGoogle Scholar |

Leidl, W, Bentley, MI, and Gass, GH (1976). Longitudinal growth of the seminiferous tubules in LH and FSH treated rats. Andrologia 8, 131–136.
Longitudinal growth of the seminiferous tubules in LH and FSH treated rats.Crossref | GoogleScholarGoogle Scholar |

Levi, AJ, Fisher, AM, Hughes, L, and Hendry, WF (1979). Male infertility due to sulphasalazine. The Lancet 314, 276–278.
Male infertility due to sulphasalazine.Crossref | GoogleScholarGoogle Scholar |

Levy, RA, de Jesús, GR, de Jesús, NR, and Klumb, EM (2016). Critical review of the current recommendations for the treatment of systemic inflammatory rheumatic diseases during pregnancy and lactation. Autoimmunity Reviews 15, 955–963.
Critical review of the current recommendations for the treatment of systemic inflammatory rheumatic diseases during pregnancy and lactation.Crossref | GoogleScholarGoogle Scholar |

Linares, V, Sánchez, DJ, Bellés, M, Albina, L, Gómez, M, and Domingo, JL (2007). Pro-oxidant effects in the brain of rats concurrently exposed to uranium and stress. Toxicology 236, 82–91.
Pro-oxidant effects in the brain of rats concurrently exposed to uranium and stress.Crossref | GoogleScholarGoogle Scholar |

Lodowska, J, Gruchlik, A, Wolny, D, Wawszczyk, J, Dzierzewicz, Z, and Węglarz, L (2015). The effect of sulfasalazine and 5-aminosalicylic acid on the secretion of interleukin 8 by human colon myofibroblasts. Acta Poloniae Pharmaceutica - Drug Research 72, 917–921.

Luberda, Z (2005). The role of glutathione in mammalian gametes. Reproductive Biology 5, 5–17.

Miller, RJ, and Killian, GJ (1987). Morphometric analyses of the epididymis from normal and vasectomized rats. Journal of Andrology 8, 279–291.
Morphometric analyses of the epididymis from normal and vasectomized rats.Crossref | GoogleScholarGoogle Scholar |

Nielsen E, Ostergaard G, Larsen J (2008) ‘Toxicological risk assessment of chemicals: a practical guide.’ (Informa Healthcare: New York, USA)

Nørgård, B, Pedersen, L, Christensen, LA, and Sørensen, HT (2007). Therapeutic drug use in women with crohn’s disease and birth outcomes: a Danish nationwide cohort study. The American Journal of Gastroenterology 102, 1406–1413.
Therapeutic drug use in women with crohn’s disease and birth outcomes: a Danish nationwide cohort study.Crossref | GoogleScholarGoogle Scholar |

Ojeda SR, Skinner MK (2006) Puberty in the rat. In ‘Knobil and Neill’s physiology of reproduction’. 3rd edn. (Ed. JD Neill) pp. 2061–2126. (Academic Press: London, UK) https://doi.org/10.1016/B978-012515400-0/50043-9

Oliveira, FJA, and Cecchini, R (2000). Oxidative stress of liver in hamsters infected with Leishmania (L.) Chagasi. Journal of Parasitology 86, 1067–1072.
Oxidative stress of liver in hamsters infected with Leishmania (L.) Chagasi.Crossref | GoogleScholarGoogle Scholar |

Ommati, MM, Heidari, R, Jamshidzadeh, A, Zamiri, MJ, Sun, Z, Sabouri, S, Wang, J, Ahmadi, F, Javanmard, N, Seifi, K, Mousapour, S, and Yeganeh, BS (2018). Dual effects of sulfasalazine on rat sperm characteristics, spermatogenesis, and steroidogenesis in two experimental models. Toxicology Letters 284, 46–55.
Dual effects of sulfasalazine on rat sperm characteristics, spermatogenesis, and steroidogenesis in two experimental models.Crossref | GoogleScholarGoogle Scholar |

Osawe, SO, and Farombi, EO (2018). Quercetin and rutin ameliorates sulphasalazine-induced spermiotoxicity, alterations in reproductive hormones and steroidogenic enzyme imbalance in rats. Andrologia 50, e12981.
Quercetin and rutin ameliorates sulphasalazine-induced spermiotoxicity, alterations in reproductive hormones and steroidogenic enzyme imbalance in rats.Crossref | GoogleScholarGoogle Scholar |

Perobelli, JE, Martinez, MF, da Silva Franchi, CA, Fernandez, CDB, Camargo, JLVd, and Kempinas, WdG (2010). Decreased sperm motility in rats orally exposed to single or mixed pesticides. Journal of Toxicology and Environmental Health, Part A 73, 991–1002.
Decreased sperm motility in rats orally exposed to single or mixed pesticides.Crossref | GoogleScholarGoogle Scholar |

Picut, CA, Ziejewski, MK, and Stanislaus, D (2018). Comparative aspects of pre- and postnatal development of the male reproductive system. Birth Defects Research 110, 190–227.
Comparative aspects of pre- and postnatal development of the male reproductive system.Crossref | GoogleScholarGoogle Scholar |

Piffer, RC, Garcia, PC, Gerardin, DCC, Kempinas, WG, and Pereira, OCM (2009). Semen parameters, fertility and testosterone levels in male rats exposed prenatally to betamethasone. Reproduction, Fertility and Development 21, 634–639.
Semen parameters, fertility and testosterone levels in male rats exposed prenatally to betamethasone.Crossref | GoogleScholarGoogle Scholar |

Ragni, G, Porro, GB, Ruspa, M, Barattini, G, Lombardi, C, and Petrillo, M (1984). Abnormal semen quality and low serum testosterone in men with inflammatory bowel disease treated for a long time with Sulfasalazine. Andrologia 16, 162–167.
Abnormal semen quality and low serum testosterone in men with inflammatory bowel disease treated for a long time with Sulfasalazine.Crossref | GoogleScholarGoogle Scholar |

Robb, GW, Amann, RP, and Killian, GJ (1978). Daily sperm production and epididymal sperm reserves of pubertal and adult rats. Journal of Reproduction and Fertility 54, 103–107.
Daily sperm production and epididymal sperm reserves of pubertal and adult rats.Crossref | GoogleScholarGoogle Scholar |

Romeu, M, Mulero, M, Giralt, M, Folch, J, Nogués, MR, Torres, A, Fortuño, A, Sureda, FX, Cabré, M, Paternáin, JL, and Mallol, J (2002). Parameters related to oxygen free radicals in erythrocytes, plasma and epidermis of the hairless rat. Life Sciences 71, 1739–1749.
Parameters related to oxygen free radicals in erythrocytes, plasma and epidermis of the hairless rat.Crossref | GoogleScholarGoogle Scholar |

Scherle, W (1970). A simple method for volumetry of organs in quantitative stereology. Mikroskopie 26, 57–60.

Seed, J, Chapin, RE, Clegg, ED, Dostal, LA, Foote, RH, Hurtt, ME, Klinefelter, GR, Makris, SL, Perreault, SD, Schrader, S, Seyler, D, Sprando, R, Treinen, KA, Veeramachaneni, DNR, and Wise, LD (1996). Methods for assessing sperm motility, morphology, and counts in the rat, rabbit, and dog: a consensus report. Reproductive Toxicology 10, 237–244.
Methods for assessing sperm motility, morphology, and counts in the rat, rabbit, and dog: a consensus report.Crossref | GoogleScholarGoogle Scholar |

Sharma, GCJS, Khajja, KJBS, and Pareek, H (2007). Histological determination of site of antifertility action of sulfasalazine in male albino rats. Pharmacologyonline 2, 95–101.

Siervo, GEML, Ogo, FM, Staurengo-Ferrari, L, Anselmo-Franci, JA, Cunha, FQ, Cecchini, R, Guarnier, FA, Verri, WA, and Fernandes, GSA (2019). Sleep restriction during peripuberty unbalances sexual hormones and testicular cytokines in rats. Biology of Reproduction 100, 112–122.
Sleep restriction during peripuberty unbalances sexual hormones and testicular cytokines in rats.Crossref | GoogleScholarGoogle Scholar |

Simundic, AM (2012). Practical recommendations for statistical analysis and data presentation in Biochemia Medica journal. Biochemia Medica 22, 15–23.
Practical recommendations for statistical analysis and data presentation in Biochemia Medica journal.Crossref | GoogleScholarGoogle Scholar |

Srivastava, SK, and Beutler, E (1967). Permeability of normal and glucose-6-phosphate dehydrogenase deficient erythrocytes to glutathione. Biochemical and Biophysical Research Communications 28, 659–664.
Permeability of normal and glucose-6-phosphate dehydrogenase deficient erythrocytes to glutathione.Crossref | GoogleScholarGoogle Scholar |

Stoker, TE, Parks, LG, Gray, LE, and Cooper, RL (2000). Endocrine-disrupting chemicals: prepubertal exposures and effects on sexual maturation and thyroid function in the male rat. A focus on the EDSTAC recommendations. Critical Reviews in Toxicology 30, 197–252.
Endocrine-disrupting chemicals: prepubertal exposures and effects on sexual maturation and thyroid function in the male rat. A focus on the EDSTAC recommendations.Crossref | GoogleScholarGoogle Scholar |

Sun, Z, Li, S, Yu, Y, Chen, H, Ommati, MM, Manthari, RK, Niu, R, and Wang, J (2018). Alterations in epididymal proteomics and antioxidant activity of mice exposed to fluoride. Archives of Toxicology 92, 169–180.
Alterations in epididymal proteomics and antioxidant activity of mice exposed to fluoride.Crossref | GoogleScholarGoogle Scholar |

Tietze, F (1969). Enzymic method for quantitative determination of nanogram amounts of total and oxidized glutathione: applications to mammalian blood and other tissues. Analytical Biochemistry 27, 502–522.
Enzymic method for quantitative determination of nanogram amounts of total and oxidized glutathione: applications to mammalian blood and other tissues.Crossref | GoogleScholarGoogle Scholar |

U.S. Environmental Protection Agency (EPA) and Office of the Science Advisor Risk Assessment Forum (2011) Recommended use of body weight 3/4 as the default method in derivation of the oral reference dose. Epa/100/R11/0001 1–39. U.S. Environmental Protection Agency.

Vermeire, S, Carbonnel, F, Coulie, PG, Geenen, V, Hazes, JMW, Masson, PL, De Keyser, F, and Louis, E (2012). Management of inflammatory bowel disease in pregnancy. Journal of Crohn’s and Colitis 6, 811–823.
Management of inflammatory bowel disease in pregnancy.Crossref | GoogleScholarGoogle Scholar |

Viktil, KK, Engeland, A, and Furu, K (2012). Outcomes after anti-rheumatic drug use before and during pregnancy: a cohort study among 150 000 pregnant women and expectant fathers. Scandinavian Journal of Rheumatology 41, 196–201.
Outcomes after anti-rheumatic drug use before and during pregnancy: a cohort study among 150 000 pregnant women and expectant fathers.Crossref | GoogleScholarGoogle Scholar |

Voodward, GE (1935). Glyoxalase: III. Glyoxalase as a reagent for the quantitative microestimation of glutathione. The Journal of Biological Chemistry 109, 1–10.
Glyoxalase: III. Glyoxalase as a reagent for the quantitative microestimation of glutathione.Crossref | GoogleScholarGoogle Scholar |

Wahl, C, Liptay, S, Adler, G, and Schmid, RM (1998). Sulfasalazine: a potent and specific inhibitor of nuclear factor kappa B. The Journal of Clinical Investigation 101, 1163–1174.
Sulfasalazine: a potent and specific inhibitor of nuclear factor kappa B.Crossref | GoogleScholarGoogle Scholar |

Weibel, ER (1963). Principles and methods for the morphometric study of the lung and other organs. Laboratory Investigation 12, 131–155.

Weibel, ER (2013). A retrospective of lung morphometry: from 1963 to present. American Journal of Physiology - Lung Cellular and Molecular Physiology 305, L405–L408.
A retrospective of lung morphometry: from 1963 to present.Crossref | GoogleScholarGoogle Scholar |

Weisz, J, and Ward, IL (1980). Plasma testosterone and progesterone titers of pregnant rats, their male and female fetuses, and neonatal offspring. Endocrinology 106, 306–316.
Plasma testosterone and progesterone titers of pregnant rats, their male and female fetuses, and neonatal offspring.Crossref | GoogleScholarGoogle Scholar |

Welsh, M, Saunders, PTK, Fisken, M, Scott, HM, Hutchison, GR, Smith, LB, and Sharpe, RM (2008). Identification in rats of a programming window for reproductive tract masculinization, disruption of which leads to hypospadias and cryptorchidism. The Journal of Clinical Investigation 118, 1479–1490.
Identification in rats of a programming window for reproductive tract masculinization, disruption of which leads to hypospadias and cryptorchidism.Crossref | GoogleScholarGoogle Scholar |

Welsh, M, MacLeod, DJ, Walker, M, Smith, LB, and Sharpe, RM (2010). Critical androgen-sensitive periods of rat penis and clitoris development. International Journal of Andrology 33, e144–e152.
Critical androgen-sensitive periods of rat penis and clitoris development.Crossref | GoogleScholarGoogle Scholar |

Yu, Y, Han, Y, Niu, R, Wang, J, Manthari, RK, Ommati, MM, and Sun, Z (2018). Ameliorative effect of VE, IGF-I, and hCG on the fluoride-induced testosterone release suppression in mice Leydig cells. Biological Trace Element Research 181, 95–103.
Ameliorative effect of VE, IGF-I, and hCG on the fluoride-induced testosterone release suppression in mice Leydig cells.Crossref | GoogleScholarGoogle Scholar |