• Nie Znaleziono Wyników

SHORT COMMUNICATION EFFECT OF SINGLE AND REPEATED IN VITRO EXPOSURE OF OVARIAN FOLLICLES TO o,p’-DDT AND p,p’-DDT AND THEIR METABOLITES

N/A
N/A
Protected

Academic year: 2022

Share "SHORT COMMUNICATION EFFECT OF SINGLE AND REPEATED IN VITRO EXPOSURE OF OVARIAN FOLLICLES TO o,p’-DDT AND p,p’-DDT AND THEIR METABOLITES"

Copied!
8
0
0

Pełen tekst

(1)

SHORT COMMUNICATION

EFFECT OF SINGLE AND REPEATED IN VITRO EXPOSURE OF OVARIAN FOLLICLES TO o,p’-DDT AND p,p’-DDT AND THEIR METABOLITES

Anna K. Wójtowicz

1,#

, Ewa L. Gregoraszczuk

1

, Anna Ptak

1

, Jerzy Falandysz

2

Laboratory of Physiology and Toxicology of Reproduction, Department of Animal Physiology, Institute of Zoology, Jagiellonian University, Ingardena 6, PL 30-060 Kraków; University of Gdañsk, Faculty of Chemistry, Chair of Analytical Chemistry, Sobieskiego 18, PL 80-952 Gdañsk, Poland

Effect of single and repeated in vitro exposure of ovarian follicles to o,p’-DDT and p,p’-DDT and their metabolites. A.K. WÓJTOWICZ, E.L. GREGORASZCZUK, A. PTAK, J. FALANDYSZ. Pol. J. Pharmacol., 2004, 56, 465–472.

The aim of the presented study was to compare the effect of o,p’-DDT [1,1-dichloro-2,2-bis-(p,p’-chlorophenyl)-ethylene] and p,p’-DDT [1,1,1- trichloro-2,2-bis-(p-chlorophenyl)-ethane] and their metabolites DDE and DDD on estradiol secretion by ovarian follicles, the target organs of environ- mental estrogens. Theca interna (Tc) and granulosa cells (Gc) were collected from medium size porcine follicles and cultured as a monolayer. The cells were initially cultured for 24 h to allow attachment to the plates and then me- dia were changed for the new ones and o,p’-DDT and p,p’-DDT and their metabolites: o,p’-DDE, p,p’-DDE and o,p’-DDD were added at doses of 4, 40, 400 ng and 4mg/ml medium to investigate dose-dependent effects. Me- dia were collected after 24 h and frozen for estradiol content determination.

When the effect of single and repeated exposure was investigated, the lowest dose of 4 ng/ml and the highest one of 4mg/ml were chosen on the basis of the results of Experiment 1. o,p’-DDT exerted antiestrogenic action at all doses used while its metabolites and p,p’-DDT and its metabolites decreased estradiol secretion only when present in the medium at a dose of 4 ng/ml.

The highest doses caused the increase in estradiol secretion. Parent o,p’- DDT and its metabolites showed antiestrogenic action after single exposure to 4 ng/ml while parent p,p’-DDT and its metabolites caused estrogenic ac- tion. All investigated compounds, except o,p’-DDT, increased estradiol se- cretion after single exposure to the dose of 4mg/ml. Repeated exposure re- sulted in a massive antiestrogenic action of all investigated chemicals.

In conclusion, our study points to time-dependent effect of DDT and its metabolites on ovarian follicles with the strongest estrogenic properties ob- served after single exposure and antiestrogenic action caused by repeated ex- posure. Given the duration of folliculogenesis, one can imagine many differ- ent potential mechanisms by which DDT could influence steroidogenesis.

Key words: granulosa and theca cells, o,p’-DDT, p,p’-DDT, steroid se- cretion

ISSN 1230-6002

(2)

INTRODUCTION

It was found that the insecticide p,p’-DDT [1,1,1-trichloro-2,2-bis-(p-chlorophenyl)-ethane] and its most stable metabolite p,p’-DDE [1,1-di- chloro-2,2-bis-(p,p’-chlorophenyl)-ethylene] have adverse effects on reproduction [3]. In mammals p,p’-DDT is converted by reductive dechlorination to p,p’-DDD and then by dehydrochlorination to p,p’- DDE [9]. Dietary exposure to DDTs results in their accumulation in the lipid phase of the tissues and cells including human blood, adipose tissue, and ovarian follicular fluid [29]. Hirshfield [19]

showed that those xenobiotics could impair fertility by altering follicular growth and hormone biosyn- thesis.

In mammalian species, dose-response studies of toxic effects of these substances, such as distur- bance of sexual development and reproductive function across species, tissues, and ages are very complicated, expensive and time consuming and, therefore, very limited. In vitro systems are uniquely suited to investigate specific cellular and molecular mechanisms of toxic effects in potential target or- gans such as the ovary and, thus, can improve risk assessment. Original toxicants, their precursors or selective inhibitors can be individually added to isolated cell types to evaluate specific toxicity mechanisms. In our laboratory, we collected fol- licular cells from the porcine ovary excised from animals showing natural estrous cycle to study the direct effect of DDT on ovarian steroidogenesis.

The aim of the present study is to compare dose- and time-dependence of effects of insecticides p,p’-DDT, o,p’-DDT as well as their metabolites (DDD and DDE), on estradiol secretion by follicu- lar cells in primary co-culture of granulosa cells (Gc) and theca cells (Tc) isolated from antral por- cine follicles. The rationale of performing the co- culture experiment was based on an idea that theca-derived androgens will serve as a substrate for estradiol production by co-cultured Gc [7, 34].

MATERIALS and METHODS

Chemicals and reagents

Parker medium M199, trypsin, and calf serum (CS) were purchased from the Laboratory of Sera and Vaccines, Lublin, Poland. Antibiotic, antimy- cotic solution (100x) was obtained from Sigma Chemical Co. St. Louis, MO, USA. DDT conge-

ners (p,p’-DDT, o,p’-DDT, p,p’-DDD, o,p’-DDD, p,p’-DDE and o,p’-DDE) were purchased from Reference Standards, EPA, Research Triangle Park, NC, USA. Stock solutions of these test compounds in DMSO were prepared and added to M199 sup- plemented with 5% CS at appropriate concentra- tions every 24 h starting from 24 h till 72 h of cul- ture. The final concentration of DMSO in the me- dium was always 0.6%.

Cell cultures

Porcine ovaries obtained from a local abattoir were collected into a bottle filled with sterilized ice-cold saline and transported to the laboratory.

Approximately 1.5 h elapsed from slaughter to arri- val at the laboratory. Medium (6–8 mm in diame- ter) follicles were obtained from ovaries collected on day 16 of the estrus cycle as previously de- scribed by Gregoraszczuk and Ska³ka [14]. In each experiment, six ovaries from three animals were se- lected for cell preparation. Since each ovary yielded four to six follicles, the total number of fol- licles for each preparation varied between 24–36.

This procedure was chosen to minimize possible variations existing between follicles and animals.

Gc and Tc were subsequently prepared according to the technique described by Stok³osowa et al. [34].

Gc were scrubbed from the follicular wall with round-tipped ophthalmologic tweezers and rinsed several times with PBS. After isolation, Gc were washed three times in M199, collected and resus- pended in M199 supplemented with 10% CS (M199/CS). The Tc from the same follicles was prepared as previously described in detail by Stok³osowa et al. [33]. Briefly, the theca layers were placed in a drop of saline under a dissecting microscope. The theca interna was manually sepa- rated from the underlying theca externa. The iso- lated theca interna tissue was then washed with PBS, cleaned, cut with scissors and exposed to trypsinization with 6–7 ml of 0.25% trypsin in PBS for 10 min at 37°C. The cells were separated by de- cantation and the procedure was repeated three times. Finally, the cells were centrifuged and resus- pended in M199/CS. For co-culture experiments, the viability of granulosa and theca cells was deter- mined by the Trypan blue exclusion test and subse- quently they were inoculated at a concentration of 1.25 × 105and 0.3 × 105cells/well, respectively, in Nunc 48 well tissue culture plates. Thus, the ratio of the cells was comparable to that observed in vivo

(3)

(Gc : Tc = 4:1) according to Stok³osowa et al. [34].

The cultures were maintained at 37°C in a humidi- fied atmosphere of 5% CO2/95% O2.

(Gc : Tc = 4:1) according to Stok³osowa et al. [34].

The cultures were maintained at 37°C in a humidi- fied atmosphere of 5% CO2/95% O2.

Experimental procedure

Experiment 1 was conducted to examine the dose-dependent effect of DDT and its metabolites on estradiol secretion by follicular cells. Cells were isolated and initially cultured without test com- pounds for 24 h to allow for cell attachment to the wells. After 24 h, the medium was discarded and 0.5 ml of fresh M199 medium supplemented with 5% CS was added at the control culture, while to the experimental culture o,p’-DDT and p,p’-DDT or their metabolites were added at concentrations of 4, 40, 400 ng and 4mg/ml medium. Media were collected after 24 h and frozen for estradiol determi- nation. Every treatment was conducted in 4 wells and each experiment was repeated 3 times.

Experiment 2 was conducted to demonstrate the time-dependent effect of DDT and its metabolites on estradiol secretion by follicular cells. Cells were isolated and cultured without test compounds for 24 h to allow for cell attachment to the wells. After 24 h the medium was discarded and 0.5 ml of fresh

M199/CS alone was added to the control culture, while to the experimental cultures, o,p’-DDT or p,p’-DDT or their metabolites were added at a con- centration of 4 ng/ml or 4mg/ml medium. This con- centration of DDT and its metabolites was chosen based on the results from dose-response curves ob- tained in Experiment 1. Media were collected after single (24 h) or repeated (72 h) exposure, and fro- zen for estradiol determination. Thus, in the experi- ments with repeated exposure, the compounds were added 3 times during the experiment. Each treat- ment was conducted in 4 wells and the experiment was repeated 3 times.

Steroid analysis

17b-estradiol (E2) concentration in the media was determined by radioimmunological method us- ing Spectra kits (Orion, Diagnostic, Finland) sup- plied by Polatom (Œwierk, Poland). The detection limit of the assay was 5 pg. The coefficients of variation between and within assays were 10.3%

and 2.9%, respectively. The mean recoveries were 85.6–108.9%. The cross-reaction with ethinylestra- diol was 1.4%. All other tested steroids (estrone, 0

1 2 3 4 5 6 7 8

C o’,p’-DDT o,p’-DDE o,p’-DDD p,p’-DDT p,p’-DDE

Estradiolpg/ml

0 4 ng 40 ng 400 ng 4 gm

* * * *

**

*

* *

*

* *

*

*

*

*

**

**

Fig. 1. The dose-dependent effect of DDT and its metabolites on estradiol secretion by co-culture of granulosa and theca cells isolated from medium-size follicles (MF). Initially the cells were cultured in serum-containing (10% calf serum) M199 for 24 h to allow for cell attachment to the plates. After 24 h, serum-containing M199 was discarded and cells were cultured for an additional 24 h in M199 supplemented with 5% calf serum; 4, 40, 400 ng/ml or 4mg/ml o,p’- DDT and DDT - p,p’and metabolites: o,p’-DDE, p,p’- DDE and o,p’-DDD were added to experimental groups. After 24 h of culture, the media were collected and frozen (–20°C) for estradiol analysis. Each histogram represents the mean ± SEM derived from three different experiments (n = 3) each in quadruplicate. *p <

0.05; **p < 0.01

(4)

estriol, progesterone, testosterone, and corticoster- one) showed less than 1% cross-reaction.

Statistical analysis

Each treatment was repeated three times in quadruplicates and, thus, the total number of repli- cates was 12. Since the variations between the ex- periments were small, those 12 results were aver- aged and analyzed by analysis of variance followed by Duncan’s new multiple range test. All data (n = 12) are expressed as the means ± SEM.

RESULTS

Dose-dependent action of o,p’-DDT and p,p’- DDT and their metabolites o,p’-DDE, o,p’-DDD, p,p’-DDE on estradiol secretion

In control cultures, estradiol secretion into the medium for 24 h resulted in its final concentration of 2.28 ± 0.9 pg/ml.

Parent o,p’-DDT induced a significant decrease in estradiol secretion at all tested concentrations.

The amount of estradiol in the medium of o,p’- DDT-treated cells was on average 1.08, 1.1, 1.02

Single exposure

0 1 2 3 4 5 6 7 8 9

K o’,p’-DDT o,p’-DDE o,p’-DDD p,p’-DDT p,p’-DDE o,’p’-DDT o,p’-DDE o,p’-DDD p,p’-DDT p,p’-DDE

Estradiolpg/ml

4 ng/ml a

* * *

*

*

*

*

*

*

*

Repeated exposure

0 2 4 6 8 10 12

K o’,p’-DDT o,p’-DDE o,p’-DDD p,p’-DDT p,p’-DDE o’,p’-DDT o,p’-DDE o,p’-DDD p,p’-DDT p,p’-DDE

Estradiolpg/ml 4 ng/ml

b

** ** **

* *

*

*

Fig. 2. The effect of a) single and b) repeated exposure to DDT and its metabolites on estradiol secretion by co-culture of granulosa and theca cells isolated from medium-size follicles (MF). Initially the cells were cultured in serum-containing (10% calf serum) M199 for 24 h to allow for cell attachment to the plates. At 24 h, serum-containing M199 was discarded and cells were cultured for an additional 24 h or 72 h in M199 supplemented with 5% calf serum; 4 ng/ml (minimal concentration) and 4 mg/ml (maximal concentration) o,p’- DDT and DDT - p,p’ and metabolites: o,p’-DDE, p,p’ - DDE and o,p’-DDD were added to experimental groups. Each histogram represents the mean ± SEM derived from three different experiments (n = 3) each in quadruplicate. * p < 0.05; **p < 0.01.

(5)

and 1.39 pg/ml vs. 2.28 pg/ml in the control cul- ture. (Fig. 1), o,p’-DDE at a dose of 4 ng/ml de- creased estradiol secretion (p < 0.01) at doses of 40 and 400 ng/ml was without effect, while at a dose of 4 mg/ml significantly increased estradiol secre- tion (p < 0.01) (Fig. 1).

o,p’-DDE only at a dose of 4 ng/ml was without effect on estradiol secretion. At all other doses used in the experiment, it significantly increased secre- tion of estradiol into the medium (3.73, 3.73 and 4.74 ng/ml, respectively, after 40 ng, 400 ng (p < 0.05) and 4mg/ml (p < 0.01). (Fig. 1).

Both parent p,p’-DDT and its metabolite p,p’- DDE showed estrogenic action at all doses used (Fig. 1).

The effect of single and repeated exposure to o,p’-DDT and p,p’-DDT and their metabolites o,p’-DDE, o,p’-DDD, p,p’-DDE on estradiol se- cretion

After single exposure to o,p’-DDT and both metabolites at a lower dose (4 ng/ml), a decreased estradiol secretion was noted while at a greater concentration (4 mg/ml), they showed estrogenic action (2.4-fold and 2-fold increase in estradiol se- cretion, respectively, under the influence of o,p’- DDE and o,p’-DDD, p < 0.05). Parent p,p’-DDT and its metabolite p,p’ -DDE at both the lowest and the highest doses had estrogenic action (Fig. 2a).

After repeated exposure to both isomers of DDT and their metabolites, a decrease in estradiol secretion was noted (Fig. 2b).

DISCUSSION

Estrogen mimicry is one of the most commonly reported effects of endocrine disrupters [24, 28], so it is important to focus on estrogen sensitive tissues in the female reproductive system. The present ex- periments using co-culture of Tc and Gc whose ra- tio was the same as that existing in vivo in the ovar- ian follicles, showed an increase in estradiol secre- tion after single exposure to all tested agents but o,p’-DDT. Oppositely, a huge antiestrogenic action of all investigated chemicals was noted after re- peated exposure. The strongest estrogenic action was observed after exposure to p,p’-DDE which is a very stable and highly lipophilic metabolite of p,p’-DDD, acting as an estrogen [4], an anti- estrogen [39], or antiandrogen [6] depending on the studied system. Clark et al. [4] investigated the ef-

fect of DDT and p,p’-DDE on the development of amphibian gonaducts and showed that DDT an- tagonized the estrogenic action of the steroid treat- ments, while p,p’-DDE acted as an estrogen on the Müllerian duct of females. Danzo [6] examined binding properties of [3H] physiological ligands (present at a concentration of 7 nM) to the andro- gen and showed that DDT, p,p’-DDT, p,p’-DDE and o,p’- DDT caused a statistically significant in- hibition of specific binding of [3H] 5a-DHT to the androgen receptor. A very important question con- cerns the mechanism(s) of this stimulatory effect.

Based on the current literature on estrogenicity of the investigated compounds, three mechanisms can be envisioned: (i) ability to react with the estrogen receptor thereby changing intracellular signaling and causing a stimulation of estradiol synthesis [1, 20, 36], (ii) induction of the activity of enzymes in- volved in estradiol biosynthesis [5, 38], (iii) simi- larly to PCBs they can inhibit the breakdown of es- tradiol, resulting in an increased bioavailability of this steroid [10, 22, 23].

The follicles of the ovary are major producers of estrogen in the body, a function that is influ- enced by compounds such as dioxin [12, 13, 17, 18, 31] and higher chlorinated PCBs [11, 14–17, 25–28]. Thus, it is possible that lower chlorinated PCBs might also interfere with estrogen produc- tion. Indeed, one major effect of PCBs is the induc- tion of metabolizing enzymes, especially the cyto- chromes P450s. Several cytochrome P450 iso- zymes induced by xenobiotics are active in steroid hormone hydroxylation [2], therefore, xenobiotics that are capable of altering steroid hormone hy- droxylation can perturb endogenous steroid me- tabolism and cause disturbances in endocrine ho- meostasis. DDT could possibly increase estrogen production by increasing the activity of the rate limit- ing enzymes that are involved in estradiol synthesis, for example by activating P450 aromatase, which metabolizes androgens to estrogen, as it was sug- gested by Wójtowicz et al. [40, 41] for PCB action.

Finally, Kester et al. [22, 23] suggested that the estrogenic action of some compounds could be in- direct, by increasing estradiol bioavailability in tar- get tissues through an inhibition of the breakdown and inactivation of estrogen. In this case, DDT it- self does not mimic estrogen, but rather alters the levels of endogenous estrogen by inhibiting the breakdown and inactivation of estrogen by inhibit- ing the formation of inactive E2 sulfate.

(6)

Relatively little is known about the presence of individual cytochromes P450 in the ovarian folli- cles. To understand the differences in action of DDT and its metabolites on ovarian steroidogenesis and to elucidate the mechanisms involved in these effects, further studies on the induction of CYP izo- symes by these compounds are needed.

Taking into consideration various data concern- ing the antiandrogenic action of pesticides [21, 30, 35, 37], it seems that the drastic decrease in estradiol secretion observed in the medium and large follicles after repeated exposure to DDTs could be due to the insufficient testosterone production which is a sub- strate for estrogen production. Andersen et al. [1]

tested twenty four pesticides for interactions with the estrogen receptor (ER) and the androgen recep- tor (AR) in transactivation assays on human placen- tal microsomes and documented that dieldrin, endo- sulfan, methiocarb, and fenarimol, acted both as es- trogen agonists and androgen antagonists. Those observations confirm what was suggested in the present study that in the future studies it would be necessary to examine possible action of DDT as an antiandrogen in particular stage of follicular devel- opment.

DDT may be responsible for inadequate estra- diol secretion by follicles, what accounts for the anti-fertility effects. Estrogen mimicry is one of the most commonly reported effects of exposure to en- docrine disrupters [28]. However, in addition to es- trogenic potency, those xenobiotics may act as anti- androgens and, thus, produce “estrogenic environ- ment” [32] but also they can decrease the amount of a substrate available for estrogen production in the follicles and so act as an antiestrogen [16, 39]. The observed decrease in estradiol secretion after re- peated exposure to DDT and its metabolites could be due to the stimulation of progesterone produc- tion by follicular cells. Progesterone is known in- hibitor of aromatase [8, 11].

In summary, both estrogenic and antiestrogenic action of DDT and its metabolites depending on time and doses suggests complex mechanism of their ac- tion.

Acknowledgments. The authors would like to thank M. Mika, PhD, from the Department of Animal Physiology, Academy of Agriculture, Kraków, Poland, for radioimmuno- logical determinations of steroid hormones. The work was supported by Jagiellonian University grant DS/IZ/Fz/2004.

REFERENCES

1. Andersen HR, Vinggaard AM, Rassmusen TH, Gjer- mandsen IM, Bonefeld-Jorgensen EC: Effects of cur- rently used pesticides in assays for estrogenicity, an- drogenicity and aromatase activity in vitro. Toxicol Appl Pharmacol, 2002, 179, 1–12.

2. Augustowska K, Gregoraszczuk EL, Milewicz T, Krzysiek J, Grochowalski A, Chrz¹szcz R: Effects of dioxin (2,3,7,8-TCDD) and PCDDs/PCDFs congeners mixture on steroidogenesis in human placenta tissue culture Endocr Regul, 2003, 37, 11–19.

3. Chedrese JP, Feyles F: The diverse mechanism of ac- tion of dichlorodiphenyl-dichloroethylene (DDE) and methoxychlor in ovarian cells in vitro Reprod Toxicol, 2001, 15, 693–698.

4. Clark E, Norris D, Jones R: Interactions of gonadal steroids and pesticides (DDT, DDE) on gonaduct growth in larval tiger salamander Ambystoma trig- inum. Gen Comp Endocrinol, 1998, 109, 94–105.

5. Crellin NK, Rodway MR, Swan CL, Gillo-Meina C, Chedrese PJ: Dichlorodiphenyldichloroethylene po- tentiates the effect of protein kinase A pathway activa- tors on progesterone synthesis in cultured granulosa cells. Biol Reprod, 1999, 61, 1099–1103.

6. Danzo BJ: Environmental xenobiotics may disrupt normal endocrine function by interfering with the binding of physiological ligands to steroid receptors and binding proteins. Environ Health Perspect, 1997, 105, 294–301.

7. Fortune JE, Armstrong DT: Hormonal control of 17-b-estradiol biosynthesis in proestrous rat follicles:

estradiol production by isolated theca versus granu- losa. Endocrinology, 1978, 102, 227–235.

8. Fortune JE, Vincent SE: Progesterone inhibits the in- duction of aromatase activity in rat granulosa cells in vitro. Biol Reprod, 1983, 28, 1078–1089.

9. Fox SD, Roman JM, Issaq HJ, Nims RW: Metabolic conversion of 1,1-dichloro-2,2-bis(p-chlorophenyl)ethane (DDD) to 1,1-dichloro-2,2-bis(p-chlorophenyl)ethylene (DDE) in the male F344/NCr rat. Arch Environ Con- tam Toxicol, 1998, 35, 104–108.

10. Garner CE, Jefferson WN, Burka LT, Matthews HB, Newbold RR: In vitro estrogenicity of the catechol metabolites of selected polychlorinated biphenyls.

Toxicol Appl Pharmacol, 1999, 154, 188–197.

11. Gregoraszczuk EL: Is progesterone a modulator of lu- teal steroidogenesis in pig? A tissue culture approach.

Folia Histochem Cytobiol, 1994, 32, 31–33.

12. Gregoraszczuk EL.: Dioxin exposure and porcine re- productive hormonal activity. Cad Saude Publica 2002, 18, 453–462.

13. Gregoraszczuk EL, Grochowalski A, Chrz¹szcz R, Wêgiel M: Congener-specific accumulation of poly- chlorinated biphenyls in ovarian follicular wall fol- lows repeated exposure to PCB 126 and PCB 153.

Comparison of tissue levels of PCB and biological changes. Chemosphere, 2003, 50, 481–488.

(7)

14. Gregoraszczuk EL, Ska³ka M: Thyroid hormone as a regulator of basal and hCG-stimulated steroidogene- sis by cultured porcine theca and granulosa cells iso- lated at different stages of the follicular phase. Reprod Fertil Dev, 1996, 8, 961–967.

15. Gregoraszczuk EL, Sowa M, Kajta M, Ptak A, Wójtowicz A: Effect of PCB 126 and PCB 153 on in- cidence of apoptosis in cultured theca and granulosa cells collected from small, medium and large preovu- latory follicles. Reprod Toxicol, 2003, 17, 465–471.

16. Gregoraszczuk EL, Wójtowicz AK: In vitro exposure of porcine ovarian follicular cells to PCB 153 alters steroid secretion but not their viability. Preliminary study. Scientific World Journal, 2002, 2, 261–267.

17. Gregoraszczuk EL, Wojtowicz AK, Zabielny E, Gro- chowalski A: Dose- and time-dependent effect of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) on pro- gesterone secretion by porcine luteal cells cultured in vitro. J Physiol Pharmacol, 2000, 51, 127–135.

18. Gregoraszczuk EL, Zabielny E, Ochwat D: Aryl hy- drocarbon receptor (AhR)-linked inhibition of luteal cell progesterone secretion in 2,3,7,8-tetrachloro- dibenzo-p-dioxin treated cells. J Physiol Pharmacol, 2001, 52, 303–311.

19. Hirshfield AN: Overview of ovarian follicular devel- opment: considerations for the toxicologist. Environ Mol Mutagen, 1997, 29, 10–15.

20. Jaga K: What are the implications of the interaction between DDT and estrogen receptors in the body?

Med Hypotheses, 2000, 54, 18–25.

21. Kelce WR, Lambright CR, Gray LE Jr, Roberts KP:

Vinclozolin and p,p’-DDE alter androgen-dependent gene expression: in vivo confirmation of an androgen receptor-mediated mechanism. Toxicol Appl Pharma- col, 1997, 142, 192–200.

22. Kester MHA, Bulduk S, Tibboel D, Meinl W, Glatt H, Falany CN, Coughtrie WH et al: Potent inhibitor of es- trogen sulfotransferase by hydroxylated PCB metabo- lites: a novel pathway explaining the estrogenic activity of PCBs. Endocrinology, 2000, 141, 1897–1890.

23. Kester MHA, Bulduk S, van Toor H, Tibboel D, Meinl W, Glatt H, Falany CN et al.: Potent inhibition of es- trogen sulfotransferase by hydroxylated metabolites of polychlorinated aromatic hydrocarbons reveals al- ternative mechanism for estrogenic activity of endo- crine disrupters. J Clin Endocrinol Metab, 2002, 87, 1142–1150.

24. Korach KS, Sarver P, Chae K, McLachlan JA, McKin- ney JD: Estrogen receptor-binding activity of poly- chlorinated hydroxybiphenyls: conformationally re- stricted structural probes. Mol Pharmacol, 1988, 33, 120–126.

25. Layton AC, Sanseverino J, Gregory BW, Easter JP, Say- ler GS, Schultz TW: In vitro estrogen receptor binding of PCBs: measured activity and detection of hydroxy- lated metabolites in a recombinant yeast assay. Toxi- col Appl Pharmacol, 2002, 180, 157–163.

26. Lind PM, Eriksen EF, Sahlin L, Edlund M, Orberg J:

Effects of the antiestrogenic environmental pollutant

3,3’,4,4’,5–pentachlorobiphenyl (PCB#126) in rat bone and uterus: diverging effects in ovariectomized and intact animals. Toxicol Appl Pharmacol, 1999, 154, 236–244.

27. Machala M, Bláha L, Vondráèek J, Lehmler H-J, Ro- bertson LW, Malmberg T: Comparison of relative toxic potencies of hydroxylated PCBs and quinones in hepatic and mammary cell lines. Organohalogen Compounds, 2002, 56, 25–28.

28. McKinney JD, Waller CL: Molecular determinants of hormone mimicry: halogenated aromatic hydrocarbon environmental agents. J Toxicol Environ Health B Crit Rev, 1998, 1, 27–58.

29. Morgan DP, Roan CC: Absorption, storage, and meta- bolic conversion of ingested DDT and DDT metabo- lites in man. Arch Environ Health, 1971, 22, 301–308.

30. Ostby J, Kelce WR, Lambright C, Wolf CJ, Mann P, Gray LE Jr: The fungicide procymidone alters sexual differentiation in the male rat by acting as an andro- gen- receptor antagonist in vivo and in vitro. Toxicol Ind Health, 1999, 15, 80–93.

31. Piek³o R, Grochowalski A, Gregoraszczuk EL: 2,3,7,8- Tetrachlorodibenzo-p-dioxin alters follicular steroido- genesis in time- and cell-specific manner. Exp Clin Endocrinol Diabetes, 2000, 108, 299–304.

32. Safe SH: Polychlorinated biphenyls (PCBs): environ- mental impact, biochemical and toxic responses, and implications for risk assessment. Crit Rev Toxicol, 1994, 24, 87–149.

33. Stok³osowa S, Bahr J, Gregoraszczuk E: Some mor- phological and functional characteristics of cells of the porcine theca interna in tissue culture. Biol Re- prod, 1978 19, 712–719.

34. Stok³osowa S, Gregoraszczuk EL, Channing CP: Es- trogen and progesterone secretion by isolated cultured porcine thecal and granulosa cells. Biol Reprod, 1982, 26, 943–952.

35. Tamura H, Maness SC, Reischmann K, Dorman DC, Gray LE, Gaido KW: Androgen receptor antagonism by the organophosphate insecticide fenitrothion. Toxi- col Sci, 2001, 62, 56–62.

36. Tiemann U, Schneider F, Tuchscherer A: Effects of or- ganochlorine pesticides on DNA synthesis of cultured oviductal and uterine cells and on estrogen receptor of uterine tissue from heifers. Arch Toxicol, 1996, 70, 490–496.

37. Vinggaard AM, Breinholt V, Larsen JC: Screening of selected pesticides for oestrogen receptor activation in vitro. Food Addit Contam,1999, 16, 533–542.

38. Vinggaard AM, Hnida C, Breinholt V, Larsen JC:

Screening of selected pesticides for inhibition of CYP19 aromatase activity in vitro. Toxicol In vitro, 2000, 14, 227–234.

39. Vonier PM, Crain DA, McLachlan JA, Guillete LJ Jr, Arnold SF: Interaction of environmental chemicals with the estrogen and progesterone receptors from oviduct of the American alligator. Environ Health Per- spect,1996, 104, 1318–1322.

(8)

40. Wójtowicz AK, Gregoraszczuk EL, Lyche JL, Rop- stad E: Time- and cell-specific action of polychlori- nated biphenyls (PCB 153 and PCB 126) on steroid secretion, by porcine theca and granulosa cells in mono- and co-culture. J Physiol Pharmacol, 2000, 51, 555–568.

41. Wójtowicz A, Ropstad E, Gregoraszczuk E: Estrous cycle-dependent changes in steroid secretion by pig ovarian cells in vitro to polychlorinated biphenyl (PCB 153) Endocr Regul, 2001, 35, 223 –228.

Received: January 23, 2004, in revised form: June 2, 2004.

Cytaty

Powiązane dokumenty

The following parameters were considered the best criteria for an evaluation of the remelting process: master heat analysis, the melt history sheets prepared by WSK Rzeszów,

Politicisation of ageing society: a perspective of policy and politics Th e aim of the chapter is to present the process of politicisation of the aging population in Poland

ABSTRACT: The aim of the present study was to examine the effect of number of sprint repetitions on the variation of blood lactate concentration (blood [La]) during

EFFECTS OF PLYOMETRIC TRAINING ON VERTICAL LANDING FORCE, JUMP HEIGHT, RANGE OF KNEE FLEXION, AND LANDING TIME IN REPEATED COUNTERMOVEMENT JUMP (RCMJ).. DATA

Previous studies indicate that the decreased serum con- centration of adiponectin and elevated concentration of leptin are characteristic of obesity and associated with a higher

Napromienianie ca∏ego cia∏a (total body irradiation, TBI) jest ustalonà procedurà przeprowadzanà jako cz´Êç przy- gotowania chorych ze z∏oÊliwymi schorzeniami hematolo-

Our study demonstrated that three months of statin treatment with either atorvastatin or rosuvastatin decreased the P-wave amplitude, the P-wave dispersion on the surface ECG, and

The significance of the 12 C 4.44→gs line shape model lies in the fact that, theoretically, it allows for the reconstruction of the gamma angular distri- bution of the nuclear