全氟辛烷磺酸等环境污染物染毒哺乳期大鼠对成年期Leydig细胞影响的实验研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
全氟有机化合物(PFOCs)和邻苯二甲酸盐作为人造材料在工业上的应用已有几十年的历史,其中以全氟辛烷磺酸盐(PFOCs)全氟辛酸(PFOA)和邻苯二甲酸盐应用最为广泛。PFOS和:PFOA同时具备疏脂、疏水等特性,被广泛用于日常生活近千种产品。通常条件下PFOS和PFOA极难分解,而且在有机生物体内有聚积效应。邻苯二甲酸盐这一类化合物,主要用于聚氯乙烯材料,被普遍应用于日常生活数百种产品中,在人体和动物体内发挥着类似雌性激素的作用,可干扰生殖内分泌系统功能,是造成男子生殖问题的“罪魁祸首”。在大部分动物体内,邻苯二甲酸盐会快速分解成邻苯二甲酸单体,如DMP可分解成MMP单体,DEHP分解成MEHP。并且,这些单体的毒性远远大于其前体。
     本研究采用多种实验手段观察到,PFOA显著抑制大鼠睾丸微粒体和Leydig细胞中的3β-羟基脱氢酶(3p-HSD) HSD和17β羟基脱氢酶3(17β-HSD3)活性,并抑制Leydig细胞睾丸酮的产生。PFOA抑制这两种酶活性,进而抑制睾丸酮的生成。
     PFOS和PFOSK抑制大鼠3β-HSD,而PFHxSK和PFBSK无抑制能力。同时,5种不同的全氟烃基物质对人3β-HSD均无抑制作用。PFOS及PFOSK对人睾丸中17β羟基脱氢酶有显著的抑制作用,而且存在结构和种属的特异性。
     PFOS高剂量组大鼠体重及睾丸重量显著性降低。高、低剂量组血清睾丸酮显著性降低。Real-time PCR检测Cypllal表达显著降低。酶学检测P450c17和17βHSD活性显著降低。在高、低剂量组Leydig细胞数目显著降低。PFOS染毒哺乳期大鼠,成年期生殖内分泌功能发生明显改变。
     DPrP和DBP明显抑制人和大鼠11βHSD2。DCHP明显抑制大鼠11βHSD2。单体MEHP明显抑制人和大鼠11βHSD2。邻苯二甲酸类化合物对11β-HSD2有明显的抑制作用,且这种抑制作用与邻苯二甲酸类化合物的空间结构有一定的相关性。
     本研究结果为政府环保部门制定相应的环保政策、法规提供科学依据,对我国保护环境、提高人口素质具有重要的科学意义。
The term' testicular dysgenesis syndrome'(TDS) refers to a spectrum of reproductive disorders that originate in male fetal life. TDS includes cryptorchidism (undescended testes) and hypospadias (abnormal formation of the urethral meatus) in newborn boys and testicular cancer and reduced fertility in adult males. Environmental endocrine disruptors are suspected contributors to the increase in TDS, perfluorooctane sulfonate (PFOS) is wildly used, and their reproductive toxicity and reproductive endocrine toxicity begun to attract the attention of scientists. The present study was to test the effects of PFOS on male reproductive in Long-Evans rats after exposure to PFOS in lactational. The aim of this study was to test the foetus body weight and testis weight; measure testosterone with radioimmunoassay, enzyme assay and biomarker of Leydig cells with realtime PCR, and evaluate the spectrum of protein of testis with proteinomics. Our objective was to study the effects of PFOS on male reproductive endocrine toxicity and discover the mechanism of affect of PFOS on Leydig stem cells.
     1 Inhibition of 3β-and 17β-hydroxysteroid dehydrogenase activities in rat Leydig cells by perfluorooctane acid
     Aim:PFOA interferes with Leydig cell steroidogenic enzymes by measuring its effect on 3β-hydroxysteroid dehydrogenase (3β-HSD) and 17(3-hydroxysteroid dehydrogenase 3 (17β-HSD3) activities in rat testis microsomes and Leydig cells.
     Method:Testicular microsome and Leydig cells from SD rats were prepared. The experiment was divided into control and positive groups. We incubated DMEM and PFOA with testicular microsome and Leydig cells, enzyme assay for 3β-hydroxysteroid dehydrogenase (3β-HSD) and 17β-hydroxysteroid dehydrogenase 3 (17β-HSD3) activities, and tritium-based radioimmunoassay to test testosterone concentration.
     Results:PFOA inhibited microsomal 3β-HSD activity with an IC50 of 53.2±25.9μmol/L and 17β-HSD3 activity with an IC50 17.7±6.8μmol/L. PFOA inhibited intact Leydig cell 3p-HSD activity with an IC50 of 146.1±0.9μmol/L and 17β-HSD3 activity with an IC50 of 194.8±1.0μmol/L. The inhibitions of 3P-HSD and 17p-HSD3 activities by PFOA were competitive for the substrates.
     Conclusion:PFOA inhibits 3β-HSD and 17β-HSD3 activities in rat Leydig cells.
     2 Comparison of the potencies of perfluoroalkylated substances inhibiting of 3β-and 17β-hydroxysteroid dehydrogenase 3 activities in human and rat testes
     Aim:The objective of the present study was to compare the potencies of five different PFASs including PFOA, PFOS, potassium perfluorooctylsulfonate (PFOSK), potassium perfluorohexanesulfonate (PFHxSK) and potassium perfluorobutane sulfonate (PFBSK) in the inhibition of 3β-hydroxysteroid dehydrogenase (βP-HSD) and 17p-hydroxysteroid dehydrogenase 3 (17β-HSD3) in human and rat testes. Human or rat microsomal enzyme was subjected to the exposure to various PFASs.
     Method:Microsomal preparations of human and rat testes were done according to the author published before which was treated by DMEM and different perfluoroalkylated substances respectively. The experiment was divided into control and positive groups. The activities of 3β-hydroxysteroid dehydrogenase (3β-HSD) and 17β-hydroxysteroid dehydrogenase 3 (17β-HSD3) in human and rat testes were tested with enzyme assay.
     Result:PFOS and PFOSK inhibited rat 3P-HSD activity with IC50 of 1.35±0.05 and 1.77±0.04μmol/L, respectively, and PFHxSK and PFBSK did not inhibit the enzyme at concentrations up to 250μmol/L. However, all of these chemicals weakly inhibited human 3β-HSD activity with IC50s over 250μmol/L. PFOS, PFOSK and PFOA inhibited human 17P-HSD3 activity with IC50s of 6.02±1.02,4.39±0.46 and 127.60±28.52μmol/L, respectively. The potencies of inhibiting 17β-HSD3 activity were PFOSK> PFOS> PFOA> PFHSK= PFBSK for human 17p-HSD3. There was a species difference in the sensitivity to PFAS-mediated inhibition of 17β-HSD3 activity. For example, the IC50s of inhibiting rat 17β-HSD3 activity for PFOS(K) were over 250μmol/L. PFOS(K) showed competitive inhibition of rat 3β-HSD and human 17p-HSD3.
     Conclusion:In conclusion, the present study shows that PFOS and PFOSK are the potent inhibitors of human 17β-HSD3 and that there are structure-and species-dependent differences for the potency inhibiting 17β-HSD3 activity.
     3 The effects of PFOS on adult Leydig cells in lactational in male Long-Evans rat
     Aim:The aims of the present study were to detect whether PFOS exposure results in toxic effects in the adult male reproductive system and determine the mechanism of the PFOS toxicity in the testis when PFOS exposed in lactation.
     Method:Adult dams were treated from PND1 to PND8 with 0 (control, n= 6),5 (= 6) or 10(n= 6) mg/kg PFOS (Sigma-Aldrich Co. Ltd) in 0.05% ween-20 administered daily by oral gavage. Male foetuses were killed by inhalation of carbon dioxide at birth for ALC analysis PND56. And serum testosterone analysis and realtime-PCR, enzyme assay and 3β-HSD staining were done.
     Result:As compared to control, for animals exposed to dose of 10 mg/kg/day, the body and testis weights reduced significantly(P<0.001). T values were 50% lower in the 5 and 10 mg/kg/day groups. The expression of Cyp11A,oding for P450scc, was markedly reduced at dose of 10 mg/kg/day group (P<0.05). The adult Leydig cell numbers, as compared to control, reduced significantly at dose of 5 and 10 mg/kg groups.
     Conclusion:In conclusion, these data, taken together, point out that there are effects on ALC number in lactational rats exposure to PFOS, and steroidogenic capacity is most important. The results suggest that reducing expressions of Cyp11A, P450c17 and 17βHSD might contribute to the reproductive toxicity of PFOS.
     4 Structure-dependent inhibition of human and rat 11β-hydroxysteroid dehydrogenase 2 activities by phthalates
     Aim:In the present study,12 phthalate diesters and four monoesters for inhibition of human and rat kidney 11 (3-HSD2 were tested. The modes of inhibition and looked for a relationship between potency for inhibition and the chemical structures were examined.
     Method:Microsomal preparations of human and rat testes were done according to the author published before, and divided into control and positive group which was treated by DMEM and different phthalates respectively. The activities of 11β-hydroxysteroid dehydrogenase2 (11β-HSD2) in human and rat testes was tested with enzyme assay.
     Result:Of the phthalate diesters we tested, dipropyl phthalate (DPrP) and di-n-butyl phthalate (DBP) significantly inhibited both human and rat 11 (3-HSD2 activities. The IC50s were 85.6μmol/L for DPrP and 13.7μmol/L for DBP when calculated for rat 11β-HSD2. However, phthalates could be inhibitors with six carbons in the alcohol moiety if the carbons were cyclized, as in dicyclohexyl phthalate (DCHP), which inhibited rat 11β-HSD2 with an IC50 of 32.64μmol/L. We showed that mono (2-ethylhexyl) phthalate (MEHP) significantly inhibited human (IC50= 110.8±10.9) and rat (121.8±8.5μM) 11(3-HSD2 activity even though its parent compound, di(2-ethylhexyl) phthalate (DEHP) did not. MEHP was a competitive inhibitor of 11β-HSD2 enzymatic activity.
     Conclusion:In conclusion, we tested phthalates that varied in size and shape, and found that phthalates that inhibited 11β-HSD2 had alcohol moieties that were intermediate in size to those on the non-inhibitory phthalates. Inhibition was not strictly dependent on the number of carbons in the moieties, but on some aspect of the space-filling conformation of the moiety.
引文
[1]Olsen, G.W., Mair DC, Church TR, et al., Decline in perfluorooctanesulfonate and other polyfluoroalkyl chemicals in American Red Cross adult blood donors, 2000-2006. Environ Sci Technol,2008.42(13):p.4989-95.
    [2]Biegel, L.B., Liu RC, Hurtt ME. et al., Effects of ammonium perfluorooctanoate on Leydig cell function:in vitro, in vivo, and ex vivo studies. Toxicol Appl Pharmacol,1995.134(1):p.18-25.
    [3]Lau, C., Anitole K, Hodes C. et al., Perfluoroalkyl acids:a review of monitoring and toxicologicalfindings. Toxicol Sci,2007.99(2):p.366-94.
    [4]Joensen, U.N., Bossi R, Leffers H, et al., Do perfluoroalkyl compounds impair human semen quality? Environ Health Perspect,2009.117(6):p.923-7.
    [5]Betts, K.S., Perfluoroalkyl acids:what is the evidence telling us? Environ Health Perspect,2007.115(5):p. A250-6.
    [6]Apelberg, B.J., Witter FR, Herbstman JB. et al., Cord serum concentrations of perfluorooctane sulfonate (PFOS) and perfluorooctanoate (PFOA) in relation to weight and size at birth. Environ Health Perspect,2007.115(11):p.1670-6.
    [7]Haider, S.G., Laue D, Schwochau G, et al., Morphological studies on the origin of adult-type Leydig cells in rat testis. Ital J Anat Embryol,1995.100 Suppl 1:p. 535-41.
    [8]Haider, S.G., Cell biology of Leydig cells in the testis. Int Rev Cytol,2004.233:p. 181-241.
    [9]Kerr, J.B. and C.M. Knell, The fate of fetal Leydig cells during the development of the fetal and postnatal rat testis. Development,1988.103(3):p.535-44.
    [10]Kuopio, T., Tapanainen J, Pelliniemi LJ, et al, Developmental stages of fetal-type Leydig cells in prepubertal rats. Development,1989.107(2):p.213-20.
    [11]Byskov, A.G., Differentiation of mammalian embryonic gonad. Physiol Rev, 1986.66(1):p.71-117.
    [12]Upadhyay, S. and L. Zamboni, Preliminary observations on the role of the mesonephros in the development of the adrenal cortex. Anat Rec,1982.202(1):p. 105-111.
    [13]Hatano,O., Takakusu A, Nomura M, et al., Identical origin of adrenal cortex and gonad revealed by expression profiles of Ad4BP/SF-1. Genes Cells,1996.1(7):p. 663-71.
    [14]Luo, X., Y. Ikeda, and K.L. Parker, A cell-specific nuclear receptor is essential for adrenal and gonadal development and sexual differentiation. Cell,1994. 77(4):p.481-90.
    [15]Shinoda, K., Lei H, Yoshii H. et al., Developmental defects of the ventromedial hypothalamic nucleus and pituitary gonadotroph in the Ftz-Fl disrupted mice. Dev Dyn,1995.204(1):p.22-9.
    [16]Davidoff, M.S., Middendorff R, Kofuncu E. et al., Leydig cells of the human testis possess astrocyte and oligodendrocyte marker molecules. Acta Histochem, 2002.104(1):p.39-49.
    [17]Davidoff, M.S., Middendorff R, Koeva Y, et al., Glial cell line-derived neurotrophic factor (GDNF) and its receptors GFRalpha-1 and GFRalpha-2 in the human testis. Ital J Anat Embryol,2001.106(2 Suppl 2):p.173-80.
    [18]Brennan, J., C. Tilmann, and B. Capel, Pdgfr-alpha mediates testis cord organization and fetal Leydig cell development in the XY gonad. Genes Dev, 2003.17(6):p.800-10.
    [19]Karl, J. and B. Capel, Three-dimensional structure of the developing mouse genital ridge. Philos Trans R Soc Lond B Biol Sci,1995.350(1333):p.235-42.
    [20]Jeays-Ward, K., Hoyle C, Brennan J et al., Endothelial and steroidogenic cell migration are regulated by WNT4 in the developing mammalian gonad. Development,2003.130(16):p.3663-70.
    [21]Nishino, K., Yamanouchi K, Naito K. et al., Characterization of mesonephric cells that migrate into the XY gonad during testis differentiation. Exp Cell Res, 2001.267(2):p.225-32.
    [22]Schmahl, J., Eicher EM, Washburn LL, et al., Sry induces cell proliferation in the mouse gonad. Development,2000.127(1):p.65-73.
    [23]Burgoyne, P.S., Buehr M, Koopman P, et al., Cell-autonomous action of the testis-determining gene:Sertoli cells are exclusively XY in XX----XY chimaeric mouse testes. Development,1988.102(2):p.443-50.
    [24]Clark, A.M., K.K. Garland, and L.D. Russell, Desert hedgehog (Dhh) gene is required in the mouse testis for formation of adult-type Leydig cells and normal
    development of peritubular cells and seminiferous tubules. Biol Reprod,2000. 63(6):p.1825-38.
    [25]Yao, H.H., W. Whoriskey, and B. Capel, Desert Hedgehog/Patched 1 signaling specifies fetal Ley dig cell fate in testis organogenesis. Genes Dev,2002.16(11): p.1433-40.
    [26]Canto, P., Soderlund D, Reyes E, et al., Mutations in the desert hedgehog (DHH) gene in patients with 46,XY complete pure gonadal dysgenesis. J Clin Endocrinol Metab,2004.89(9):p.4480-3.
    [27]Baker, J., Hardy MP, Zhou J, et al., Effects of an Igfl gene null mutation on mouse reproduction. Mol Endocrinol,1996.10(7):p.903-18.
    [28]Cui, S., Ross A, Stallings N, et al., Disrupted gonadogenesis and male-to-female sex reversal in Podl knockout mice. Development,2004.131(16):p.4095-105.
    [29]Nielsen, M., Bjornsdottir S, H(?)yer PE, et al., Ontogeny of oestrogen receptor alpha in gonads and sex ducts of fetal and newborn mice. J Reprod Fertil,2000. 118(1):p.195-204.
    [30]Delbes, G, Levacher C, Duquenne C, et al., Endogenous estrogens inhibit mouse fetal Leydig cell development via estrogen receptor alpha. Endocrinology,2005. 146(5):p.2454-61.
    [31]Baker, P.J. and P.J. O'Shaughnessy, Role of gonadotrophins in regulating numbers of Leydig and Sertoli cells during fetal and postnatal development in mice. Reproduction,2001.122(2):p.227-34.
    [32]Schafers, B.A., B.G. Schlutius, and S.G Haider, Ontogenesis of oxidative reaction of 17beta-hydroxysteroid dehydrogenase and 11beta-hydroxysteroid dehydrogenase in rat Leydig cells, a histochemical study. Histochem J,2001. 33(9-10):p.585-95.
    [33]Kumagai, J., Hsu SY, Matsumi H, et al., INSL3/Leydig insulin-like peptide activates the LGR8 receptor important in testis descent. J Biol Chem,2002. 277(35):p.31283-6.
    [34]Asthana, S., Bhasin S, Butler RN, et al., Masculine vitality:pros and cons of testosterone in treating the andropause. J Gerontol A Biol Sci Med Sci,2004. 59(5):p.461-5.
    [35]Habert, R. and P. Brignaschi, Developmental changes in in vitro testosterone production by dispersed Leydig cells during fetal life in rats. Arch Androl,1991. 27(2):p.65-71.
    [36]Crawford, B.A., Spaliviero J, Simpson J, et al., Androgen effects on bioactive and immunoreactive gonadotrophin levels during puberty in male baboons. J Pediatr Endocrinol Metab,1997.10(4):p.401-10.
    [37]Gondo, S., Yanase T, Okabe T,et al., SF-1/Ad4BP transforms primary long-term cultured bone marrow cells into ACTH-responsive steroidogenic cells. Genes Cells,2004.9(12):p.1239-47.
    [38]Mendis-Handagama, S.M. and H.B. Ariyaratne, Differentiation of the adult Leydig cell population in the postnatal testis. Biol Reprod,2001.65(3):p.660-71.
    [39]Hardy, M.P., et al., Stress hormone and male reproductive function. Cell Tissue Res,2005.322(1):p.147-53.
    [40]Codesal, J., Regadera J, Nistal M, et al., Involution of human fetal Leydig cells. An immunohistochemical, ultrastructural and quantitative study. J Anat,1990. 172:p.103-14.
    [41]Shan, L.X. and M.P. Hardy, Developmental changes in levels of luteinizing hormone receptor and androgen receptor in rat Leydig cells. Endocrinology, 1992.131(3):p.1107-14.
    [42]Ge, R.S. and M.P. Hardy, Variation in the end products of androgen biosynthesis and metabolism during postnatal differentiation of rat Leydig cells. Endocrinology,1998.139(9):p.3787-95.
    [43]Peters, M., A.M. Muller, and S. Rose-John, Interleukin-6 and soluble interleukin-6 receptor:direct stimulation of gp130 and hematopoiesis. Blood,1998.92(10): p.3495-504.
    [44]Malaval, L., Liu F, Vernallis AB, et al., GP130/OSMR is the only LIF/IL-6 family receptor complex to promote osteoblast differentiation of calvaria progenitors. J Cell Physiol,2005.204(2):p.585-93.
    [45]Wijdenes, J., Heinrich PC, Muller-Newen G, et al., Interleukin-6 signal transducer gp130 has specific binding sites for different cytokines as determined by antagonistic and agonistic anti-gp130 monoclonal antibodies. Eur J Immunol, 1995.25(12):p.3474-81.
    [46]Piquet-Pellorce, C., Dorval-Coiffec I, Pham MD, et al., Leukemia inhibitory factor expression and regulation within the testis. Endocrinology,2000.141(3):p. 1136-41.
    [47]Bornstein, S.R., H. Rutkowski, and I. Vrezas, Cytokines and steroidogenesis. Mol Cell Endocrinol,2004.215(1-2):p.135-41.
    [48]Afane, M., Dubost JJ, Sauvezie B, et al., Modulation of Ley dig cell testosterone production by secretory products of macrophages. Andrologia,1998.30(2):p. 71-8.
    [49]Hoch, R.V. and P. Soriano, Roles of PDGF in animal development. Development, 2003.130(20):p.4769-84.
    [50]Ge, R.S., Dong Q, Sottas CM, et al., Gene expression in rat leydig cells during development from the progenitor to adult stage:a cluster analysis. Biol Reprod, 2005.72(6):p.1405-15.
    [51]Kissel, H., Timokhina I, Hardy MP, et al., Point mutation in kit receptor tyrosine kinase reveals essential roles for kit signaling in spermatogenesis and oogenesis without affecting other kit responses. EMBO J,2000.19(6):p.1312-26.
    [52]Lei, Z.M., Zou W, Mishra S, et al., Epididymal phenotype in luteinizing hormone receptor knockout animals and its response to testosterone replacement therapy. Biol Reprod,2003.68(3):p.888-95.
    [53]Scott, I.S., Charlton HM, Cox BS, et al., Effect of LH injections on testicular steroidogenesis, cholesterol side-chain cleavage P450 mRNA content and Leydig cell morphology in hypogonadal mice. J Endocrinol,1990.125(1):p.131-8.
    [54]Wing, T.Y., L.L. Ewing, and B.R. Zirkin, Effects of luteinizing hormone withdrawal on Leydig cell smooth endoplasmic reticulum and steroidogenic reactions which convert pregnenolone to testosterone. Endocrinology,1984. 115(6):p.2290-6.
    [55]Khan, S.A., K. Teerds, and J. Dorrington, Steroidogenesis-inducing protein promotes deoxyribonucleic acid synthesis in Leydig cells from immature rats. Endocrinology,1992.130(2):p.599-606.
    [56]Baker, P.J., Pakarinen P, Huhtaniemi IT, et al., Failure of normal Leydig cell development in follicle-stimulating hormone (FSH) receptor-deficient mice, but not FSHbeta-deficient mice:role for constitutive FSH receptor activity. Endocrinology,2003.144(1):p.138-45.
    [57]Moore, A. and I.D. Morris, The involvement of insulin-like growth factor-I in local control of steroidogenesis and DNA synthesis of Leydig and non-Leydig cells in the rat testicular interstitium. J Endocrinol,1993.138(1):p.107-14.
    [58]Closset, J., Gothot A, Sente B, et al., Pituitary hormones dependent expression of insulin-like growth factors I and II in the immature hypophysectomized rat testis. Mol Endocrinol,1989.3(7):p.1125-31.
    [59]Gelber, S.J., Hardy MP, Mendis-Handagama SM, et al., Effects of insulin-like growth factor-I on androgen production by highly purified pubertal and adult rat Leydig cells. J Androl,1992.13(2):p.125-30.
    [60]Shan, L., Hardy DO, Catterall JF, et al., Effects of luteinizing hormone (LH) and androgen on steady state levels of messenger ribonucleic acid for LH receptors, androgen receptors, and steroidogenic enzymes in rat Leydig cell progenitors in vivo. Endocrinology,1995.136(4):p.1686-93.
    [61]Murphy, L., I.A. Jeffcoate, and P.J. O'Shaughnessy, Abnormal Leydig cell development at puberty in the androgen-resistant Tfm mouse. Endocrinology, 1994.135(4):p.1372-7.
    [62]Normington, K. and D.W. Russell, Tissue distribution and kinetic characteristics of rat steroid 5 alpha-reductase isozymes. Evidence for distinct physiological functions. J Biol Chem,1992.267(27):p.19548-54.
    [63]Holdcraft, R.W. and R.E. Braun, Hormonal regulation of spermatogenesis. Int J Androl,2004.27(6):p.335-42.
    [64]Stocco, D.M., Tracking the role of a star in the sky of the new millennium. Mol Endocrinol,2001.15(8):p.1245-54.
    [65]Nelson, D.R., Koymans L, Kamataki T, et al., P450 superfamily:update on new sequences, gene mapping, accession numbers and nomenclature. Pharmacogenetics,1996.6(1):p.1-42.
    [66]Chung, B.C., Matteson KJ, Voutilainen R, et al., Human cholesterol side-chain cleavage enzyme, P450scc:cDNA cloning, assignment of the gene to chromosome 15, and expression in the placenta. Proc Natl Acad Sci U S A,1986. 83(23):p.8962-6.
    [67]Payne, A.H. and D.B. Hales, Overview of steroidogenic enzymes in the pathway from cholesterol to active steroid hormones. Endocr Rev,2004.25(6):p.947-70.
    [68]Brock, B.J. and M.R. Waterman, Biochemical differences between rat and human cytochrome P450c17 support the different steroidogenic needs of these two species. Biochemistry,1999.38(5):p.1598-606.
    [69]Fan, Y.S., Sasi R, Lee C, et al., Localization of the human CYP17 gene (cytochrome P450(17 alpha)) to 10q24.3 by fluorescence in situ hybridization and simultaneous chromosome banding. Genomics,1992.14(4):p.1110-1.
    [70]Chen, S.A., Besman MJ, Sparkes RS, et al., Human aromatase:cDNA cloning, Southern blot analysis, and assignment of the gene to chromosome 15. DNA, 1988.7(1):p.27-38.
    [71]Morissette, J., Rheaume E, Leblanc JF, et al., Genetic linkage mapping of HSD3B1 and HSD3B2 encoding human types I and II 3 beta-hydroxysteroid dehydrogenase/delta 5-delta 4-isomerase close to D1S514 and the centromeric D1Z5 locus. Cytogenet Cell Genet,1995.69(1-2):p.59-62.
    [72]Geissler, W.M., Davis DL, Wu L, et al., Male pseudohermaphroditism caused by mutations of testicular 17 beta-hydroxysteroid dehydrogenase 3. Nat Genet,1994. 7(1):p.34-9.
    [73]Sha, J.A., Dudley K, Rajapaksha WR, et al., Sequence of mouse 17beta-hydroxysteroid dehydrogenase type 3 cDNA and tissue distribution of the type 1 and type 3 isoform mRNAs. J Steroid Biochem Mol Biol,1997.60(1-2):p.19-24.
    [74]Parker, K.L., Rice DA, Lala DS, et al., Steroidogenic factor 1:an essential mediator of endocrine development. Recent Prog Horm Res,2002.57:p.19-36.
    [75]Tremblay, J.J. and R.S. Viger, GATA factors differentially activate multiple gonadal promoters through conserved GATA regulatory elements. Endocrinology, 2001.142(3):p.977-86.
    [76]Tremblay, J.J. and R.S. Viger, Novel roles for GATA transcription factors in the regulation of steroidogenesis. J Steroid Biochem Mol Biol,2003.85(2-5):p. 291-8.
    [77]Viger, R.S., Taniguchi H, Robert NM, et al., Role of the GATA family of transcription factors in andrology. J Androl,2004.25(4):p.441-52.
    [78]Anderson, C.M. and C.R. Mendelson, Regulation of steroidogenesis in rat Leydig cells in culture:effect of human chorionic gonadotropin and dibutyryl cyclic AMP on the synthesis of cholesterol side chain cleavage cytochrome P-450 and adrenodoxin. Arch Biochem Biophys,1985.238(2):p.378-87.
    [79]Youngblood, G.L. and A.H. Payne, Isolation and characterization of the mouse P45017 alpha-hydroxylase/C17-20-lyase gene (Cyp17):transcriptional regulation of the gene by cyclic adenosine 3',5'-monophosphate in MA-10 Leydig cells. Mol Endocrinol,1992.6(6):p.927-34.
    [80]Anakwe, O.O. and A.H. Payne, Noncoordinate regulation of de novo synthesis of cytochrome P-450 cholesterol side-chain cleavage and cytochrome P-45017 alpha-hydroxylase/C17-20 lyase in mouse Leydig cell cultures:relation to steroid production. Mol Endocrinol,1987.1(9):p.595-603.
    [81]Hales, D.B. and A.H. Payne, Glucocorticoid-mediated repression of P450scc mRNA and de novo synthesis in cultured Leydig cells. Endocrinology,1989. 124(5):p.2099-104.
    [82]Peng, L. and A.H. Payne, AP-2 gamma and the homeodomain protein distal-less 3 are required for placental-specific expression of the murine 3 beta-hydroxysteroid dehydrogenase VI gene, Hsd3b6. J Biol Chem,2002.277(10):p. 7945-54.
    [83]Baker, P.J., J.H. Sha, and P.J. O'Shaughnessy, Localisation and regulation of 17beta-hydroxysteroid dehydrogenase type 3 mRNA during development in the mouse testis. Mol Cell Endocrinol,1997.133(2):p.127-33.
    [84]Jensen, A.A. and H. Leffers, Emerging endocrine disrupters:perfluoroalkylated substances. Int J Androl,2008.31(2):p.161-9.
    [85]Giesy, J.P. and K. Kannan, Perfluorochemical surfactants in the environment. Environ Sci Technol,2002.36(7):p.146A-152A.
    [86]Olsen, G.W., Burris JM, Ehresman DJ, et al., Half-life of serum elimination of perfluorooctanesulfonate, perfluorohexanesulfonate, and perfluorooctanoate in retired fluorochemical production workers. Environ Health Perspect,2007. 115(9):p.1298-305.
    [87]Shi, Z., Burris JM, Ehresman DJ, et al., Alterations in gene expression and testosterone synthesis in the testes of male rats exposed to perfluorododecanoic acid. Toxicol Sci,2007.98(1):p.206-15.
    [88]Penning, T.M., Molecular endocrinology of hydroxysteroid dehydrogenases. Endocr Rev,1997.18(3):p.281-305.
    [89]Hu, G.X., Zhou HY, Li XW, et al., The (+)-and (-)-gossypols potently inhibit both 3beta-hydroxysteroid dehydrogenase and 17beta-hydroxysteroid
    dehydrogenase 3 in human and rat testes. J Steroid Biochem Mol Biol,2009. 115(1-2):p.14-9.
    [90]Salva, A., G.R. Klinefelter, and M.P. Hardy, Purification of rat leydig cells: increased yields after unit-gravity sedimentation of collagenase-dispersed interstitial cells. J Androl,2001.22(4):p.665-71.
    [91]Payne, A.H., K.L. Wong, and M.M. Vega, Differential effects of single and repeated administrations of gonadotropins on luteinizing hormone receptors and testosterone synthesis in two populations of Leydig cells. J Biol Chem,1980, 255(15):p.7118-22.
    [92]Seethalakshmi, L., Flores C, Malhotra RK, et al, The mechanism of cyclosporine's action in the inhibition of testosterone biosynthesis by rat Leydig cells in vitro. Transplantation,1992.53(1):p.190-5.
    [93]Huhtaniemi, I. and L.J. Pelliniemi, Fetal Leydig cells:cellular origin, morphology, life span, and special functional features. Proc Soc Exp Biol Med, 1992.201(2):p.125-40.
    [94]Akingbemi, B.T., Youker RT, Sottas CM, et al., Modulation of rat Leydig cell steroidogenic function by di(2-ethylhexyl)phthalate. Biol Reprod,2001.65(4):p. 1252-9.
    [95]Olsen, G.W., Gilliland FD, Burlew MM, et al., An epidemiologic investigation of reproductive hormones in men with occupational exposure to perfluorooctanoic acid. J Occup Environ Med,1998.40(7):p.614-22.
    [96]Chengelis, C.P., Kirkpatrick JB, Myers NR, et al., Comparison of the toxicokinetic behavior of perfluorohexanoic acid (PFHxA) and nonafluorobutane-1-sulfonic acid (PFBS) in cynomolgus monkeys and rats. Reprod Toxicol,2009.27(3-4):p.400-6.
    [97]Zhao, B., Chu Y, Hardy DO, et al, Inhibition of 3beta-and 17beta-hydroxysteroid dehydrogenase activities in rat Leydig cells by perfluorooctane acid. J Steroid Biochem Mol Biol.118(1-2):p.13-7.
    [98]Bjork, J.A. and K.B. Wallace, Structure-activity relationships and human relevance for perfluoroalkyl acid-induced transcriptional activation of peroxisome proliferation in liver cell cultures. Toxicol Sci,2009.111(1):p.89-99.
    [99]Chu, Y, et al., Inhibition of 3βand 17β-hydroxysteroid dehydrogenase activities in rat Leydig cells by PFOA. Asian J. Andrology,2009.142:p.11.
    [100]Benton, L., L.X. Shan, and M.P. Hardy, Differentiation of adult Leydig cells. J Steroid Biochem Mol Biol,1995.53(1-6):p.61-8.
    [101]Hu, G.X., Lian QQ, Ge RS,et al., Phthalate-induced testicular dysgenesis syndrome: Leydig cell influence. Trends Endocrinol Metab,2009.20(3):p.139-45.
    [102]Fan, Y.O., Jin YH, Ma YX,_ et al., Effects of perfluorooctane sulfonate on spermiogenesis function of male rats. Wei Sheng Yan Jiu,2005.34(1):p.37-9.
    [103]Barlow, N.J., Phillips SL, Wallace DG, et al., Quantitative changes in gene expression in fetal rat testes following exposure to di(n-butyl) phthalate. Toxicol Sci,2003.73(2):p.431-41.
    [104]Marsee, K., Woodruff TJ, Axelrad DA, et al., Estimated daily phthalate exposures in a population of mothers of male infants exhibiting reduced anogenital distance. Environ Health Perspect,2006.114(6):p.805-9.
    [105]Akingbemi, B.T. and M.P. Hardy, Oestrogenic and antiandrogenic chemicals in the environment:effects on male reproductive health. Ann Med,2001.33(6):p. 391-403.
    [106]Stahlhut, R.W., van Wijngaarden E, Dye TD, et al., Concentrations of urinary phthalate metabolites are associated with increased waist circumference and insulin resistance in adult U.S. males. Environ Health Perspect,2007.115(6):p. 876-82.
    [107]Albro, P.W., van Wijngaarden E, Dye TD, et al., Mono-2-ethylhexyl phthalate, a metabolite of di-(2-ethylhexyl) phthalate, causally linked to testicular atrophy in rats. Toxicol Appl Pharmacol,1989.100(2):p.193-200.
    [108]Kavlock, R., Boekelheide K, Chapin R, et al., NTP Center for the Evaluation of Risks to Human Reproduction:phthalates expert panel report on the reproductive and developmental toxicity of di-n-octyl phthalate. Reprod Toxicol, 2002.16(5):p.721-34.
    [109]Storr, H.L., Chan LF, Grossman AB, et al., Paediatric Cushing's syndrome: epidemiology, investigation and therapeutic advances. Trends Endocrinol Metab, 2007.18(4):p.167-74.
    [110]Monder, C. and C.H. Shackleton,11 beta-Hydroxysteroid dehydrogenase:fact or fancy? Steroids,1984.44(5):p.383-417.
    [111]Agarwal, A.K., Mune T, Monder C, et al., Cloning of cDNA encoding an NAD(+)-dependent isoform of 11 beta-hydroxysteroid dehydrogenase in sheep kidney. Endocr Res,1995.21(1-2):p.389-97.
    [112]White, P.C., T. Mune, and A.K. Agarwal,11 beta-Hydroxysteroid dehydrogenase and the syndrome of apparent mineralocorticoid excess. Endocr Rev,1997. 18(1):p.135-56.
    [113]Wyrwoll, C.S., P.J. Mark, and B.J. Waddell, Developmental programming of renal glucocorticoid sensitivity and the renin-angiotensin system. Hypertension, 2007.50(3):p.579-84.
    [114]Aufdenblatten, M., Baumann M, Raio L, et al., Prematurity is related to high placental cortisol in preeclampsia. Pediatr Res,2009.65(2):p.198-202.
    [115]Ohshima, M., S. Ohno, and S. Nakajin, Inhibitory effects of some possible endocrine-disrupting chemicals on the isozymes of human 11 beta-hydroxysteroid dehydrogenase and expression of their. mRNA in gonads and adrenal glands. Environ Sci,2005.12(4):p.219-30.
    [116]Shneider, B., Schena J, Truog R, et al., Exposure to di(2-ethylhexyl)phthalate in infants receiving extracorporeal membrane oxygenation. N Engl J Med,1989. 320(23):p.1563.
    [117]Saillenfait, A.M., F. Gallissot, and J.P. Sabate, Differential developmental toxicities of di-n-hexyl phthalate and dicyclohexyl phthalate administered orally to rats. J Appl Toxicol,2009.29(6):p.510-21.
    [118]Howdeshell, K.L., Wilson VS, Furr J, et al., A mixture of five phthalate esters inhibits fetal testicular testosterone production in the sprague-dawley rat in a cumulative, dose-additive manner. Toxicol Sci,2008.105(1):p.153-65.
    [119]Gray, L.E., Jr., Ostby J, Furr J, et al., Perinatal exposure to the phthalates DEHP, BBP, and DINP, but not DEP, DMP, or DOTP, alters sexual differentiation of the male rat. Toxicol Sci,2000.58(2):p.350-65.
    [120]Hines, E.P., Calafat AM, Silva MJ, et al., Concentrations of phthalate metabolites in milk, urine, saliva, and Serum of lactating North Carolina women. Environ Health Perspect,2009.117(1):p.86-92.
    [121]Zhang, Y., Lin L, Cao Y, et al., Phthalate levels and low birth weight:a nested case-control study of Chinese newborns. J Pediatr,2009.155(4):p.500-4.
    [122]Jaquiery, A.L., Oliver MH, Bloomfield FH, et al., Fetal exposure to excess glucocorticoid is unlikely to explain the effects of periconceptional undernutrition in sheep. J Physiol,2006.572(Pt 1):p.109-18.
    [123]Seckl, J.R. and M.J. Meaney, Glucocorticoid "programming" and PTSD risk. Ann N Y Acad Sci,2006.1071:p.351-78.
    [124]Dave-Sharma, S., Wilson RC, Harbison MD, et al., Examination of genotype and phenotype relationships in 14 patients with apparent mineralocorticoid excess. J Clin Endocrinol Metab,1998.83(7):p.2244-54.
    [125]Kajantie, E., Dunkel L, Turpeinen U, et al., Placental 11 beta-hydroxysteroid dehydrogenase-2 and fetal cortisol/cortisone shuttle in small preterm infants. J Clin Endocrinol Metab,2003.88(1):p.493-500.
    [126]Thapa, L., C.M. He, and H.P. Chen, Study on the expression of angiotensin II (ANG Ⅱ) receptor subtype 1 (AT1R) in the placenta of pregnancy-induced hypertension. Placenta,2004.25(7):p.637-41.
    [127]Lanz, C.B., Causevic M, Heiniger C, et al., Fluid Shear Stress Reduces llss-Hydroxysteroid Dehydrogenase Type 2. Hypertension,2001.37(1):p.160-169.
    [128]Goland, R.S., Tropper PJ, Warren WB, et al., Concentrations of corticotrophin-releasing hormone in the umbilical-cord blood of pregnancies complicated by pre-eclampsia. Reprod Fertil Dev,1995.7(5):p.1227-30.
    [129]Benediktsson, R., Calder AA, Edwards CR, et al., Placental 11 beta-hydroxysteroid dehydrogenase:a key regulator of fetal glucocorticoid exposure. Clin Endocrinol (Oxf),1997.46(2):p.161-6.
    [130]Ge, R.S., Chen GR, Tanrikut C, et al., Phthalate ester toxicity in Ley dig cells: developmental timing and dosage considerations. Reprod Toxicol,2007.23(3): p.366-73.
    [131]Sharpe, R.M. and N.E. Skakkebaek, Testicular dysgenesis syndrome: mechanistic insights and potential new downstream effects. Fertil Steril,2008. 89(2 Suppl):p. e33-8.
    [132]Honda, Y., S. Ohno, and S. Nakajin, Leydig cells from neonatal pig testis abundantly express 11 beta-hydroxysteroid dehydrogenase (llbeta-HSD) type 2 and effectively inactivate cortisol to cortisone. J Steroid Biochem Mol Biol, 2008.108(1-2):p.91-101.

© 2004-2018 中国地质图书馆版权所有 京ICP备05064691号 京公网安备11010802017129号

地址:北京市海淀区学院路29号 邮编:100083

电话:办公室:(+86 10)66554848;文献借阅、咨询服务、科技查新:66554700