Difference between revisions of "Aop:23"

From AOP-Wiki
Jump to: navigation, search
(unknown aop user updated status section from widget page.)
(unknown aop user updated summary tables from widget page.)
Line 88: Line 88:
 
|-
 
|-
  
|[[Event:309|Vitellogenin uptake into oocytes and oocyte growth/development, Reduction]]||[[Aop:23#Essentiality of the Key Events|Weak]]
+
|[[Event:309|Vitellogenin accumulation into oocytes and oocyte growth/development, Reduction, Reduction]]||[[Aop:23#Essentiality of the Key Events|Weak]]
  
 
|-
 
|-
Line 128: Line 128:
 
|-
 
|-
  
|[[Event:221|Plasma vitellogenin concentrations, Reduction]]||[[Relationship:255|Directly Leads to]]||[[Event:309|Vitellogenin uptake into oocytes and oocyte growth/development, Reduction]]||[[Relationship:255#Weight of Evidence|Moderate]]||[[Relationship:255#Quantitative Understanding of the Linkage|Weak]]
+
|[[Event:221|Plasma vitellogenin concentrations, Reduction]]||[[Relationship:255|Directly Leads to]]||[[Event:309|Vitellogenin accumulation into oocytes and oocyte growth/development, Reduction, Reduction]]||[[Relationship:255#Weight of Evidence|Moderate]]||[[Relationship:255#Quantitative Understanding of the Linkage|Weak]]
  
 
|-
 
|-
Line 148: Line 148:
 
|-
 
|-
  
|[[Event:309|Vitellogenin uptake into oocytes and oocyte growth/development, Reduction]]||[[Relationship:337|Directly Leads to]]||[[Event:78|Cumulative fecundity and spawning, Reduction]]||[[Relationship:337#Weight of Evidence|Moderate]]||[[Relationship:337#Quantitative Understanding of the Linkage|Moderate]]
+
|[[Event:309|Vitellogenin accumulation into oocytes and oocyte growth/development, Reduction, Reduction]]||[[Relationship:337|Directly Leads to]]||[[Event:78|Cumulative fecundity and spawning, Reduction]]||[[Relationship:337#Weight of Evidence|Moderate]]||[[Relationship:337#Quantitative Understanding of the Linkage|Moderate]]
  
 
|-
 
|-

Revision as of 17:12, 6 February 2016

AOP Title

Androgen receptor agonism leading to reproductive dysfunction
Short name: Androgen receptor agonism leading to reproductive dysfunction

Authors

Dan Villeneuve, US EPA Mid-Continent Ecology Division (villeneuve.dan@epa.gov)

Status

Please follow the link to snapshots page to view and create Snapshots of this AOP.

Open for comment

OECD Project 1.12: The Adverse outcome pathways linking aromatase inhibition, androgen receptor agonism, estrogen receptor antagonism, or steroidogenesis inhibition, to impaired reproduction in small repeat-spawning fish species

This AOP page was last modified on 2/6/2016.

Click here to show/hide revision dates for related pages

Page Revision Date/Time

Abstract

This adverse outcome pathway details the linkage between binding and activation of androgen receptor as a nuclear transcription factor in females and reproductive dysfunction as evidenced through reductions cumulative fecundity and spawning in repeat-spawning fish species. Cumulative fecundity is the most apical endpoint considered in the OECD 229 Fish Short Term Reproduction Assay. The OECD 229 assay serves as screening assay for endocrine disruption and associated reproductive impairment (OECD 2012). Cumulative fecundity is one of several variables known to be of demographic significance in forecasting fish population trends. Therefore, this AOP has utility in supporting the application of measures of androgen receptor binding and activation as a nuclear transcription factor as a means to identify chemicals with known potential to adversely affect fish populations.

Summary of the AOP

Please follow link to widget page to edit this section.

If you manually enter text in this section, it will get automatically altered or deleted in subsequent edits using the widgets.

Molecular Initiating Event

Molecular Initiating Event Support for Essentiality
Androgen receptor, Agonism Strong

Key Events

Event Support for Essentiality
Testosterone synthesis by ovarian theca cells, Reduction Moderate
17beta-estradiol synthesis by ovarian granulosa cells, Reduction Moderate
Plasma 17beta-estradiol concentrations, Reduction Strong
Transcription and translation of vitellogenin in liver, Reduction Moderate
Cumulative fecundity and spawning, Reduction Moderate
Plasma vitellogenin concentrations, Reduction Strong
Vitellogenin accumulation into oocytes and oocyte growth/development, Reduction, Reduction Weak
Gonadotropins, circulating concentrations, Reduction Moderate

Adverse Outcome

Adverse Outcome
Population trajectory, Decrease

Relationships Among Key Events and the Adverse Outcome

Event Description Triggers Weight of Evidence Quantitative Understanding
Androgen receptor, Agonism Directly Leads to Gonadotropins, circulating concentrations, Reduction Weak Weak
Plasma vitellogenin concentrations, Reduction Directly Leads to Vitellogenin accumulation into oocytes and oocyte growth/development, Reduction, Reduction Moderate Weak
Testosterone synthesis by ovarian theca cells, Reduction Directly Leads to 17beta-estradiol synthesis by ovarian granulosa cells, Reduction Strong Weak
17beta-estradiol synthesis by ovarian granulosa cells, Reduction Directly Leads to Plasma 17beta-estradiol concentrations, Reduction Strong Weak
Plasma 17beta-estradiol concentrations, Reduction Directly Leads to Transcription and translation of vitellogenin in liver, Reduction Strong Moderate
Transcription and translation of vitellogenin in liver, Reduction Directly Leads to Plasma vitellogenin concentrations, Reduction Strong Moderate
Vitellogenin accumulation into oocytes and oocyte growth/development, Reduction, Reduction Directly Leads to Cumulative fecundity and spawning, Reduction Moderate Moderate
Cumulative fecundity and spawning, Reduction Directly Leads to Population trajectory, Decrease Moderate Moderate
Gonadotropins, circulating concentrations, Reduction Directly Leads to Testosterone synthesis by ovarian theca cells, Reduction Strong Weak
Androgen receptor, Agonism Indirectly Leads to Testosterone synthesis by ovarian theca cells, Reduction Strong Weak

Network View

   Cytoscape Web will replace the contents of this div with your graph.

Click nodes or edges.

Life Stage Applicability

Life Stage Evidence Links
Adult, reproductively mature

Taxonomic Applicability

Name Scientific Name Evidence Links
Pimephales promelas Pimephales promelas NCBI

Sex Applicability

Sex Evidence Links
Female

Graphical Representation

Click to upload graphical representation

Overall Assessment of the AOP

Weight of Evidence Summary

Summary Table
Provide an overall summary of the weight of evidence based on the evaluations of the individual linkages from the Key Event Relationship pages.

Biological Plausibility
  • The biochemistry of steroidogenesis and the predominant role of the gonad in synthesis of the sex steroids are well established.
  • Similarly, the role of E2 as the major regulator of hepatic vitellogenin production is widely documented in the literature.
  • The direct link between reduced VTG concentrations in the plasma and reduced uptake into oocytes is highly plausible, as the plasma is the primary source of the VTG.
  • The direct connection between reduced VTG uptake and impaired spawning/reduced cumulative fecundity is more tentative. It is not clear, for instance whether impaired VTG uptake limits oocyte growth and failure to reach a critical size in turn impairs physical or inter-cellular signaling processes that promote release of the oocyte from the surrounding follicles. In at least one experiment, oocytes with similar size to vitellogenic oocytes, but lacking histological staining characteristic of vitellogenic oocytes was observed (R. Johnson, personal communication). At present, the link between reductions in circulating VTG concentrations and reduced cumulative fecundity are best supported by the correlation between those endpoints across multiple experiments, including those that impact VTG via other molecular initiating events (Miller et al. 2007).
  • At present, negative feedback is the most biologically plausible explanation for the reductions in ex vivo T and E2 production following exposure to 17β-trenbolone. In vitro exposure of fathead minnow ovary tissue to 17β-trenbolone or spironolactone does not cause reductions in T or E2 synthesis at concentrations comparable to those that produce significant responses in vivo (i.e., at non-cytotoxic concentrations; D.L. Villeneuve, unpublished data; C.A. LaLone unpublished data), nor are there any known reports of 17β-trenbolone directly inhibiting steroid biosynthesis. When tested in an in vitro steroidogenesis assay using H295R adrenal carcinoma cells, trenbolone caused a concentration-dependent increase in estradiol production, as opposed to any reductions in steroid hormone concentrations, an effect that was concurrent with increased transcription of CYP19 (aromatase) in the cell line (Gracia et al. 2007). Given the lack of any established direct effect on steroidogenic enzyme activity, negative feedback is currently the most likely explanation for the consistent effects observed in vivo. That said, many uncertainties regarding the exact mechanisms through which an exogenous, non-aromatizable, AR agonist elicits negative feedback remain.

Concordance of dose-response relationships: There are a limited number of studies in which multiple key events were considered in the same study following exposure to known, non-aromatizable, AR agonists. These were considered the most useful for evaluating the concordance of dose-response relationships. In general, effects on downstream key events occurred at concentrations equal to or greater than those at which upstream events occurred Concordance Table. For exposures to 17b-trenbolone, key events related to steroid production and circulating estradiol and vitellogenin concentrations were impacted at the same dose at which effects on cumulative fecundity were observed. Effects on vitellogenin transcription were only observed at greater concentrations, but data for comparable species and dose ranges were unavailable at present. For two other AR agonists tested in fish, available studies examined a single time-point only. Consequently, it was unclear whether lower effect concentrations for certain downstream KEs, relative to upstream were due to a lack of dose-response concordance, or due to decreased sensitivity of the upstream later in the exposure time-course.

While not directly addressing dose-response concordance, the dependence of the key events on the concentration of the androgen agonist has been established for all key events starting at and down-stream of reduced T synthesis. However, to date we are not aware of any studies that have established a concentration-response relationship between exposure to non-endogenous AR agonists (e.g., xenobiotics, pharmaceuticals) and circulating gonadotropin concentrations in fish or other vertebrates.

  • Exposure of female fathead minnows to the AR agonist 17β-trenbolone for 21 d caused concentration-dependent reductions in circulating T, E2, and VTG concentrations over a range from 0.005 to 0.5 μg/L. The concentration response for all three variables had a “U”-shaped concentration response curve which may indicate concentration-dependent differences in the feedback response and/or compensatory processes. Histological evidence of reduced VTG uptake and reduced gonad stage were evident, although the concentration-response of histological effects was not determined. Despite the “U”-shaped concentration-response at the biochemical level, concentration-dependent reductions in cumulative fecundity were observed (Ankley et al. 2003). Effective concentrations were consistent with those causing phenotypic masculinization in female fish.
  • Jensen et al. (2006) also demonstrated concentration-dependent reductions in circulating T, E2, and VTG following 21 d of in vivo exposure to 17α-trenbolone (Jensen et al. 2006).
  • In a time-course experiment in which female fathead minnows were exposed to to 33 or 472 ng 17β-trenbolone/L ex vivo T, ex vivo E2, plasma E2, and plasma VTG all showed concentration-dependent reductions that were consistent with the AOP (Ekman et al. 2011).
  • Exposure of female fathead minnows to spironolactone, a pharmaceutical that binds the fathead minnow AR, for 21 d caused concentration-dependent reductions in cumulative fecundity, plasma VTG and VTG mRNA expression, and plasma E2 concentrations. The frequency and severity of females with decreased yolk accumulation, and increased oocyte atresia was concentration-dependent. The chemical also induced phenotypic masculinization in female fish. (Lalone et al. 2013).
  • Exposure of female medaka to spironolactone caused concentration-dependent reductions in cumulative fecundity and VTG mRNA expression (impacts on steroid hormone concentrations were not measured). Spironolactone also caused phenotypic masculinization of female medaka (Lalone et al. 2013).
Temporal concordance among the key events and adverse effect: Temporal concordance between activation of the AR as a nuclear transcription factor and onset of a negative feedback response resulting in decreased gonadotropin secretion has not been established. Temporal concordance of the key events starting with reduced T biosynthesis and proceeding through reductions in plasma vitellogenin has been established Concordance table. Temporal concordance beyond the key event of reductions in plasma vitellogenin has not been established, in large part due to disconnect in the time-scales over which the events can be measured. For example, most small fish used in reproductive toxicity testing can spawn anywhere from once daily to several days per week. Given the variability in daily spawning rates, it is neither practical nor effective to evaluate cumulative fecundity at a time scale shorter than roughly a week. Since the impacts at lower levels of biological organization can be detected within hours of exposure, lack of impact on cumulative fecundity before the other key events are impacted cannot be effectively measured. Overall, among those key events whose temporal concordance can reasonably be evaluated based on currently available data, the temporal profile observed is consistent with the AOP.
Consistency: We are aware of no cases where the pattern of key events described was observed without also observing a significant impact on cumulative fecundity. Due to variability in the cumulative fecundity endpoint and potential compensatory responses ((Villeneuve et al. 2009; Villeneuve et al. 2013; Ankley et al. 2009b; Zhang et al. 2008; Ekman et al. 2012), the cumulative fecundity endpoint can be less sensitive than key events measured at lower levels of biological organization. Nonetheless, the occurrence of the final adverse outcome when the other key events are observed is very consistent. The final adverse effect is not specific to this AOP. Many of the key events included in this AOP overlap with AOPs linking other molecular initiating events to reproductive dysfunction in small fish.
  • In general, there is a consistent body of evidence linking exposure to an AR agonist to decreased T synthesis, E2 synthesis, circulating E2 and VTG concentrations, and cumulative fecundity in female fish. For example, the association between 17β-trenbolone exposure and reduced vitellogenin concentrations in females has been replicated in over a dozen independent experiments (Ekman et al. 2011; Ankley et al. 2003; Jensen et al. 2006; Ankley et al. 2010; Hemmer et al. 2008; Seki et al. 2006; Brockmeier et al. 2013). However, relatively few exogenous, non-aromatizable, AR agonists have been tested. Other than recent work with spironolactone (Lalone et al. 2013), we are not aware of the profile of responses being demonstrated for other AR agonists.
Uncertainties, inconsistencies, and data gaps: There are two major areas of uncertainty and data gaps in the current AOP. First, there remains considerable uncertainty as to the specific mechanism(s) through which AR agonism elicits a negative feedback response at the level of the hypothalamus and/or pituitary. There is also a substantial data gap relative to establishing that exposure to an AR agonist like 17β-trenbolone causes concentration-dependent reductions in circulating gonadotropins. The second major uncertainty in this AOP relates to whether there is a direct biological linkage between impaired VTG uptake into oocytes and impaired spawning/reduced cumulative fecundity. Plausible biological connections have been hypothesized, but have not yet been tested experimentally.

Essentiality of the Key Events

Molecular Initiating Event Summary, Key Event Summary

  • In general, few studies have directly addressed the essentiality of the proposed sequence of key events.
  • Ekman et al. 2011 provide evidence that in fathead minnow, cessation of trenbolone exposure resulted in recovery of plasma E2 and VTG concentrations which were depressed by continuous exposure to 17beta trenbolone. This provides some support for the essentiality of these two key events.
  • Essentiality of the proposed negative feedback key event is supported by experimental work that evaluated the ability of AR agonists to reduce T or E2 production in vitro. In vitro exposure of fathead minnow ovary tissue to 17β-trenbolone or spironolactone does not cause reductions in T or E2 synthesis at concentrations comparable to those that produce significant responses in vivo (i.e., at non-cytotoxic concentrations; D.L. Villeneuve, unpublished data; C.A. LaLone unpublished data), nor are there any known reports of 17β-trenbolone directly inhibiting steroid biosynthesis. When tested in an in vitro steroidogenesis assay using H295R adrenal carcinoma cells, trenbolone caused a concentration-dependent increase in estradiol production, as opposed to any reductions in steroid hormone concentrations, an effect that was concurrent with increased transcription of CYP19 (aromatase) in the cell line (Gracia et al. 2007).

Quantitative Considerations

Summary Table

Assessment of quantitative understanding of the AOP: At present, the quantitative understanding of the AOP is insufficient to directly link a measure of chemical potency as an AR agonist (e.g., as measured in a transcriptional activation assay) to a predicted effect concentration at the level of cumulative fecundity. However, a number of mechanistic and statistical models are sufficiently developed to facilitate predictions of cumulative outcomes based on intermediate key event measurements such as circulating vitellogenin concentrations. Because the current models were developed based on a fairly limited range of model compounds and species, the general applicability and degree of accuracy and precision in the model-derived predictions remains uncertain.

Applicability of the AOP

Life Stage Applicability, Taxonomic Applicability, Sex Applicability

Domain(s) of Applicability

Chemical: This AOP applies to non-aromatizable androgens. Compounds which can bind the AR in vitro, but are converted to high potency estrogens in vivo through aromatization do not produce the profile of effects described in the present AOP (e.g., methyltestosterone [Ankley et al. 2001; Pawlowski et al. 2004]; androstenedione [OECD 2007]).
Sex: The AOP applies to females only.
Life stages: The relevant life stages for this AOP are reproductively mature adults. This AOP does not apply to adult stages that lack a sexually mature ovary, for example as a result of seasonal or environmentally-induced gonadal senescence (i.e., through control of temperature, photo-period, etc. in a laboratory setting).
Taxonomic: At present, the assumed taxonomic applicability domain of this AOP is iteroparous teleost fish species. However, to date the majority of toxicological data on which this AOP is based has been limited to two small fish species, fathead minnow (Pimephales promelas) and Japanese medaka (Oryzias latipes). Given that species dependent differences in endocrine feedback responses, likely associated with different reproductive strategies, have been reported the applicability domain may prove more restricted than currently assumed.
  • Reductions in plasma VTG concentrations and/or hepatic VTG mRNA abundance in females following exposure to 17β-trenbolone has been observed in Pimephales promelas, Oryzias latipes, Danio rerio, (Seki et al. 2006), Cyprinodon variegatus (Hemmer et al. 2008), Gambusia holbrooki and Gambusia affinis (Brockmeier et al. 2013)
  • European eel may be an exception to the generalizability of the negative feedback response to a non-aromatizable xenoandrogen (Huang et al. 1997).

Considerations for Potential Applications of the AOP (optional)

References

  • 1. Amano M, Iigo M, Ikuta K, Kitamura S, Yamada H, Yamamori K. 2000. Roles of melatonin in gonadal maturation of underyearling precocious male masu salmon. General and comparative endocrinology 120(2): 190-197.
  • 2. Ankley GT, Bencic D, Cavallin JE, Jensen KM, Kahl MD, Makynen EA, et al. 2009b. Dynamic nature of alterations in the endocrine system of fathead minnows exposed to the fungicide prochloraz. Toxicol Sci 112(2): 344-353.
  • 3. Ankley GT, Bencic DC, Cavallin JE, Jensen KM, Kahl MD, Makynen EA, et al. 2009a. Dynamic nature of alterations in the endocrine system of fathead minnows exposed to the fungicide prochloraz. Toxicological sciences : an official journal of the Society of Toxicology 112(2): 344-353.
  • 4. Ankley GT, Cavallin JE, Durhan EJ, Jensen KM, Kahl MD, Makynen EA, et al. 2012. A time-course analysis of effects of the steroidogenesis inhibitor ketoconazole on components of the hypothalamic-pituitary-gonadal axis of fathead minnows. Aquatic toxicology 114-115: 88-95.
  • 5. Ankley GT, Jensen KM, Durhan EJ, Makynen EA, Butterworth BC, Kahl MD, et al. 2005. Effects of two fungicides with multiple modes of action on reproductive endocrine function in the fathead minnow (Pimephales promelas). Toxicol Sci 86(2): 300-308.
  • 6. Ankley GT, Jensen KM, Kahl MD, Durhan EJ, Makynen EA, Cavallin JE, et al. 2010. Use of chemical mixtures to differentiate mechanisms of endocrine action in a small fish model. Aquatic toxicology 99(3): 389-396.
  • 7. Ankley GT, Jensen KM, Kahl MD, Makynen EA, Blake LS, Greene KJ, et al. 2007. Ketoconazole in the fathead minnow (Pimephales promelas): reproductive toxicity and biological compensation. Environ Toxicol Chem 26(6): 1214-1223.
  • 8. Ankley GT, Jensen KM, Makynen EA, Kahl MD, Korte JJ, Hornung MW, et al. 2003. Effects of the androgenic growth promoter 17--trenbolone on fecundity and reproductive endocrinology of the fathead minnow. Environmental Toxicology and Chemistry 22(6): 1350-1360.
  • 9. Ankley GT, Kahl MD, Jensen KM, Hornung MW, Korte JJ, Makynen EA, et al. 2002. Evaluation of the aromatase inhibitor fadrozole in a short-term reproduction assay with the fathead minnow (Pimephales promelas). Toxicological Sciences 67: 121-130.
  • 10. Ankley GT, Miller DH, Jensen KM, Villeneuve DL, Martinovic D. 2008. Relationship of plasma sex steroid concentrations in female fathead minnows to reproductive success and population status. Aquatic toxicology 88(1): 69-74.
  • 11. Araki N, Ohno K, Nakai M, Takeyoshi M, Iida M. 2005. Screening for androgen receptor activities in 253 industrial chemicals by in vitro reporter gene assays using AR-EcoScreen cells. Toxicology in vitro : an international journal published in association with BIBRA 19(6): 831-842.
  • 12. Arukwe A, Goksøyr A. 2003. Eggshell and egg yolk proteins in fish: hepatic proteins for the next generation: oogenetic, population, and evolutionary implications of endocrine disruption. Comparative Hepatology 2(4): 1-21.
  • 13. Baker ME. 1997. Steroid receptor phylogeny and vertebrate origins. Molecular and cellular endocrinology 135(2): 101-107.
  • 14. Baker ME. 2011. Origin and diversification of steroids: co-evolution of enzymes and nuclear receptors. Molecular and cellular endocrinology 334(1-2): 14-20.
  • 15. Benninghoff AD, Thomas P. 2006. Gonadotropin regulation of testosterone production by primary cultured theca and granulosa cells of Atlantic croaker: I. Novel role of CaMKs and interactions between calcium- and adenylyl cyclase-dependent pathways. General and comparative endocrinology 147(3): 276-287.
  • 16. Biales AD, Bencic DC, Lazorchak JL, Lattier DL. 2007. A quantitative real-time polymerase chain reaction method for the analysis of vitellogenin transcripts in model and nonmodel fish species. Environ Toxicol Chem 26(12): 2679-2686.
  • 17. Bohl CE, Chang C, Mohler ML, Chen J, Miller DD, Swaan PW, et al. 2004. A ligand-based approach to identify quantitative structure-activity relationships for the androgen receptor. Journal of medicinal chemistry 47(15): 3765-3776.
  • 18. Bowman CJ, Kroll KJ, Hemmer MJ, Folmar LC, Denslow ND. 2000. Estrogen-induced vitellogenin mRNA and protein in sheepshead minnow (Cyprinodon variegatus). General and comparative endocrinology 120(3): 300-313.
  • 19. Breen MS, Villeneuve DL, Breen M, Ankley GT, Conolly RB. 2007. Mechanistic computational model of ovarian steroidogenesis to predict biochemical responses to endocrine active compounds. Annals of biomedical engineering 35(6): 970-981.
  • 20. Brockmeier EK, Ogino Y, Iguchi T, Barber DS, Denslow ND. 2013. Effects of 17beta-trenbolone on Eastern and Western mosquitofish (Gambusia holbrooki and G. affinis) anal fin growth and gene expression patterns. Aquatic toxicology 128-129: 163-170.
  • 21. Campbell BK, Baird DT, Webb R. 1998. Effects of dose of LH on androgen production and luteinization of ovine theca cells cultured in a serum-free system. Journal of reproduction and fertility 112(1): 69-77.
  • 22. Centenera MM, Harris JM, Tilley WD, Butler LM. 2008. The contribution of different androgen receptor domains to receptor dimerization and signaling. Molecular endocrinology 22(11): 2373-2382.
  • 23. Cheng GF, Yuen CW, Ge W. 2007. Evidence for the existence of a local activin follistatin negative feedback loop in the goldfish pituitary and its regulation by activin and gonadal steroids. The Journal of endocrinology 195(3): 373-384.
  • 24. Claessens F, Denayer S, Van Tilborgh N, Kerkhofs S, Helsen C, Haelens A. 2008. Diverse roles of androgen receptor (AR) domains in AR-mediated signaling. Nuclear receptor signaling 6: e008.
  • 25. Cutress ML, Whitaker HC, Mills IG, Stewart M, Neal DE. 2008. Structural basis for the nuclear import of the human androgen receptor. Journal of cell science 121(Pt 7): 957-968.
  • 26. Eick GN, Thornton JW. 2011. Evolution of steroid receptors from an estrogen-sensitive ancestral receptor. Molecular and cellular endocrinology 334(1-2): 31-38.
  • 27. Ekman DR, Hartig PC, Cardon M, Skelton DM, Teng Q, Durhan EJ, et al. 2012. Metabolite profiling and a transcriptional activation assay provide direct evidence of androgen receptor antagonism by bisphenol A in fish. Environmental science & technology 46(17): 9673-9680.
  • 28. Ekman DR, Villeneuve DL, Teng Q, Ralston-Hooper KJ, Martinovic-Weigelt D, Kahl MD, et al. 2011. Use of gene expression, biochemical and metabolite profiles to enhance exposure and effects assessment of the model androgen 17beta-trenbolone in fish. Environmental toxicology and chemistry / SETAC 30(2): 319-329.
  • 29. Eppig JJ. 1994. Further reflections on culture systems for the growth of oocytes in vitro. Human reproduction 9(6): 974-976.
  • 30. Genovese G, Regueira M, Piazza Y, Towle DW, Maggese MC, Lo Nostro F. 2012. Time-course recovery of estrogen-responsive genes of a cichlid fish exposed to waterborne octylphenol. Aquatic toxicology 114-115: 1-13.
  • 31. Gopurappilly R, Ogawa S, Parhar IS. 2013. Functional significance of GnRH and kisspeptin, and their cognate receptors in teleost reproduction. Frontiers in endocrinology 4: 24.
  • 32. Govoroun M, Chyb J, Breton B. 1998. Immunological cross-reactivity between rainbow trout GTH I and GTH II and their alpha and beta subunits: application to the development of specific radioimmunoassays. General and comparative endocrinology 111(1): 28-37.
  • 33. Gracia T, Hilscherova K, Jones PD, Newsted JL, Higley EB, Zhang X, et al. 2007. Modulation of steroidogenic gene expression and hormone production of H295R cells by pharmaceuticals and other environmentally active compounds. Toxicology and applied pharmacology 225(2): 142-153.
  • 34. Habibi HR, Huggard DL. 1998. Testosterone regulation of gonadotropin production in goldfish. Comparative biochemistry and physiology Part C, Pharmacology, toxicology & endocrinology 119(3): 339-344.
  • 35. Havelock JC, Rainey WE, Carr BR. 2004. Ovarian granulosa cell lines. Molecular and cellular endocrinology 228(1-2): 67-78.
  • 36. Heemers HV, Tindall DJ. 2007. Androgen receptor (AR) coregulators: a diversity of functions converging on and regulating the AR transcriptional complex. Endocrine reviews 28(7): 778-808.
  • 37. Hemmer MJ, Cripe GM, Hemmer BL, Goodman LR, Salinas KA, Fournie JW, et al. 2008. Comparison of estrogen-responsive plasma protein biomarkers and reproductive endpoints in sheepshead minnows exposed to 17beta-trenbolone. Aquatic toxicology 88(2): 128-136.
  • 38. Hong H, Fang H, Xie Q, Perkins R, Sheehan DM, Tong W. 2003. Comparative molecular field analysis (CoMFA) model using a large diverse set of natural, synthetic and environmental chemicals for binding to the androgen receptor. SAR and QSAR in environmental research 14(5-6): 373-388.
  • 39. Iguchi T, Irie F, Urushitani H, Tooi O, Kawashima Y, Roberts M, et al. 2006. Availability of in vitro vitellogenin assay for screening of estrogenic and anti-estrogenic activities of environmental chemicals. Environ Sci 13(3): 161-183.
  • 40. Jamnongjit M, Hammes SR. 2005. Oocyte maturation: the coming of age of a germ cell. Seminars in reproductive medicine 23(3): 234-241.
  • 41. Jensen K, Korte J, Kahl M, Pasha M, Ankley G. 2001. Aspects of basic reproductive biology and endocrinology in the fathead minnow (Pimephales promelas). Comparative Biochemistry and Physiology Part C 128: 127-141.
  • 42. Jensen KM, Makynen EA, Kahl MD, Ankley GT. 2006. Effects of the feedlot contaminant 17alpha-trenbolone on reproductive endocrinology of the fathead minnow. Environmental science & technology 40(9): 3112-3117.
  • 43. Jolly C, Katsiadaki I, Le Belle N, Mayer I, Dufour S. 2006. Development of a stickleback kidney cell culture assay for the screening of androgenic and anti-androgenic endocrine disrupters. Aquatic toxicology 79(2): 158-166.
  • 44. Kah O, Pontet A, Nunez Rodriguez J, Calas A, Breton B. 1989. Development of an enzyme-linked immunosorbent assay for goldfish gonadotropin. Biology of reproduction 41(1): 68-73.
  • 45. Kim TS, Yoon CY, Jung KK, Kim SS, Kang IH, Baek JH, et al. 2010. In vitro study of Organization for Economic Co-operation and Development (OECD) endocrine disruptor screening and testing methods- establishment of a recombinant rat androgen receptor (rrAR) binding assay. The Journal of toxicological sciences 35(2): 239-243.
  • 46. Korte JJ, Kahl MD, Jensen KM, Mumtaz SP, Parks LG, LeBlanc GA, et al. 2000. Fathead minnow vitellogenin: complementary DNA sequence and messenger RNA and protein expression after 17B-estradiol treatment. Environmental Toxicology and Chemistry 19(4): 972-981.
  • 47. Lalone CA, Villeneuve DL, Cavallin JE, Kahl MD, Durhan EJ, Makynen EA, et al. 2013. Cross species sensitivity to a novel androgen receptor agonist of environmental concern, spironolactone. Environmental toxicology and chemistry / SETAC (in press).
  • 48. Leino R, Jensen K, Ankley G. 2005. Gonadal histology and characteristic histopathology associated with endocrine disruption in the adult fathead minnow. Environmental Toxicology and Pharmacology 19: 85-98.
  • 49. Levavi-Sivan B, Bogerd J, Mananos EL, Gomez A, Lareyre JJ. 2010. Perspectives on fish gonadotropins and their receptors. General and comparative endocrinology 165(3): 412-437.
  • 50. Li Z, Kroll KJ, Jensen KM, Villeneuve DL, Ankley GT, Brian JV, et al. 2011a. A computational model of the hypothalamic: pituitary: gonadal axis in female fathead minnows (Pimephales promelas) exposed to 17alpha-ethynylestradiol and 17beta-trenbolone. BMC systems biology 5: 63.
  • 51. Li Z, Villeneuve DL, Jensen KM, Ankley GT, Watanabe KH. 2011b. A computational model for asynchronous oocyte growth dynamics in a batch-spawning fish. Can J Fish Aquat Sci 68: 1528-1538.
  • 52. Magoffin DA. 2005. Ovarian theca cell. The international journal of biochemistry & cell biology 37(7): 1344-1349.
  • 53. Mak P, Cruz FD, Chen S. 1999. A yeast screen system for aromatase inhibitors and ligands for androgen receptor: yeast cells transformed with aromatase and androgen receptor. Environmental health perspectives 107(11): 855-860.
  • 54. Markov GV, Laudet V. 2011. Origin and evolution of the ligand-binding ability of nuclear receptors. Molecular and cellular endocrinology 334(1-2): 21-30.
  • 55. McMaster ME MK, Jardine JJ, Robinson RD, Van Der Kraak GJ. 1995. Protocol for measuring in vitro steroid production by fish gonadal tissue. Canadian Technical Report of Fisheries and Aquatic Sciences 1961 1961: 1-78.
  • 56. Miller DH, Ankley GT. 2004. Modeling impacts on populations: fathead minnow (Pimephales promelas) exposure to the endocrine disruptor 17trenbolone as a case study. Ecotoxicology and Environmental Safety 59: 1-9.
  • 57. Miller DH, Jensen KM, Villeneuve DL, Kahl MD, Makynen EA, Durhan EJ, et al. 2007. Linkage of biochemical responses to population-level effects: a case study with vitellogenin in the fathead minnow (Pimephales promelas). Environ Toxicol Chem 26(3): 521-527.
  • 58. Miller DH, Tietge JE, McMaster ME, Munkittrick KR, Xia X, Ankley GT. 2013. Assessment of Status of White Sucker (Catostomus Commersoni) Populations Exposed to Bleached Kraft Pulp Mill Effluent. Environmental toxicology and chemistry / SETAC.
  • 59. Miller WL, Strauss JF, 3rd. 1999. Molecular pathology and mechanism of action of the steroidogenic acute regulatory protein, StAR. The Journal of steroid biochemistry and molecular biology 69(1-6): 131-141.
  • 60. Miller WL. 1988. Molecular biology of steroid hormone synthesis. Endocrine reviews 9(3): 295-318.
  • 61. Murphy CA, Rose KA, Rahman MS, Thomas P. 2009. Testing and applying a fish vitellogenesis model to evaluate laboratory and field biomarkers of endocrine disruption in Atlantic croaker (Micropogonias undulatus) exposed to hypoxia. Environmental toxicology and chemistry / SETAC 28(6): 1288-1303.
  • 62. Murphy CA, Rose KA, Thomas P. 2005. Modeling vitellogenesis in female fish exposed to environmental stressors: predicting the effects of endocrine disturbance due to exposure to a PCB mixture and cadmium. Reproductive toxicology 19(3): 395-409.
  • 63. Nagahama Y, Yoshikumi M, Yamashita M, Sakai N, Tanaka M. 1993. Molecular endocrinology of oocyte growth and maturation in fish. Fish Physiology and Biochemistry 11: 3-14.
  • 64. Navas JM, Segner H. 2006. Vitellogenin synthesis in primary cultures of fish liver cells as endpoint for in vitro screening of the (anti)estrogenic activity of chemical substances. Aquat Toxicol 80(1): 1-22.
  • 65. Norris DO. 2007. Vertebrate Endocrinology. Fourth ed. New York: Academic Press.
  • 66. Norris JD, Joseph JD, Sherk AB, Juzumiene D, Turnbull PS, Rafferty SW, et al. 2009. Differential presentation of protein interaction surfaces on the androgen receptor defines the pharmacological actions of bound ligands. Chemistry & biology 16(4): 452-460.
  • 67. Oakley AE, Clifton DK, Steiner RA. 2009. Kisspeptin signaling in the brain. Endocrine reviews 30(6): 713-743.
  • 68. OECD. 2012. Test No. 229: Fish Short Term Reproduction Assay. Paris, France:Organization for Economic Cooperation and Development.
  • 69. Olsson P-E, Berg A, von Hofsten J, Grahn B, Hellqvist A, Larsson A, et al. 2005. Molecular cloning and characterization of a nuclear androgen receptor activated by 11-ketotestosterone. Reproductive Biology and Endocrinology 3: 1-17.
  • 70. Payne AH, Hales DB. 2004. Overview of steroidogenic enzymes in the pathway from cholesterol to active steroid hormones. Endocrine reviews 25(6): 947-970.
  • 71. Prat F, Sumpter JP, Tyler CR. 1996. Validation of radioimmunoassays for two salmon gonadotropins (GTH I and GTH II) and their plasma concentrations throughout the reproductivecycle in male and female rainbow trout (Oncorhynchus mykiss). Biology of reproduction 54(6): 1375-1382.
  • 72. Prescott J, Coetzee GA. 2006. Molecular chaperones throughout the life cycle of the androgen receptor. Cancer letters 231(1): 12-19.
  • 73. Quignot N, Bois FY. 2013. A computational model to predict rat ovarian steroid secretion from in vitro experiments with endocrine disruptors. PloS one 8(1): e53891.
  • 74. Schmid T, Gonzalez-Valero J, Rufli H, Dietrich DR. 2002. Determination of vitellogenin kinetics in male fathead minnows (Pimephales promelas). Toxicol Lett 131(1-2): 65-74.
  • 75. Schmieder P, Tapper M, Linnum A, Denny J, Kolanczyk R, Johnson R. 2000. Optimization of a precision-cut trout liver tissue slice assay as a screen for vitellogenin induction: comparison of slice incubation techniques. Aquat Toxicol 49(4): 251-268.
  • 76. Schultz IR, Orner G, Merdink JL, Skillman A. 2001. Dose-response relationships and pharmacokinetics of vitellogenin in rainbow trout after intravascular administration of 17alpha-ethynylestradiol. Aquatic toxicology 51(3): 305-318.
  • 77. Seki M, Fujishima S, Nozaka T, Maeda M, Kobayashi K. 2006. Comparison of response to 17 beta-estradiol and 17 beta-trenbolone among three small fish species. Environmental toxicology and chemistry / SETAC 25(10): 2742-2752.
  • 78. Serafimova R, Walker J, Mekenyan O. 2002. Androgen receptor binding affinity of pesticide "active" formulation ingredients. QSAR evaluation by COREPA method. SAR and QSAR in environmental research 13(1): 127-134.
  • 79. Shoemaker JE, Gayen K, Garcia-Reyero N, Perkins EJ, Villeneuve DL, Liu L, et al. 2010. Fathead minnow steroidogenesis: in silico analyses reveals tradeoffs between nominal target efficacy and robustness to cross-talk. BMC systems biology 4: 89.
  • 80. Skolness SY, Blanksma CA, Cavallin JE, Churchill JJ, Durhan EJ, Jensen KM, et al. 2013. Propiconazole Inhibits Steroidogenesis and Reproduction in the Fathead Minnow (Pimephales promelas). Toxicological sciences : an official journal of the Society of Toxicology 132(2): 284-297.
  • 81. Skolness SY, Durhan EJ, Garcia-Reyero N, Jensen KM, Kahl MD, Makynen EA, et al. 2011. Effects of a short-term exposure to the fungicide prochloraz on endocrine function and gene expression in female fathead minnows (Pimephales promelas). Aquat Toxicol 103(3-4): 170-178.
  • 82. Sower SA, Freamat M, Kavanaugh SI. 2009. The origins of the vertebrate hypothalamic-pituitary-gonadal (HPG) and hypothalamic-pituitary-thyroid (HPT) endocrine systems: new insights from lampreys. General and comparative endocrinology 161(1): 20-29.
  • 83. Sperry TS, Thomas P. 1999. Identification of two nuclear androgen receptors in kelp bass (Paralabrax clathratus) and their binding affinities for xenobiotics: comparison with Atlantic croaker (Micropogonias undulatus) androgen receptors. Biology of reproduction 61(4): 1152-1161.
  • 84. Sun L, Wen L, Shao X, Qian H, Jin Y, Liu W, et al. 2010. Screening of chemicals with anti-estrogenic activity using in vitro and in vivo vitellogenin induction responses in zebrafish (Danio rerio). Chemosphere 78(7): 793-799.
  • 85. Sun L, Zha J, Spear PA, Wang Z. 2007. Toxicity of the aromatase inhibitor letrozole to Japanese medaka (Oryzias latipes) eggs, larvae and breeding adults. Comp Biochem Physiol C Toxicol Pharmacol 145(4): 533-541.
  • 86. Thornton JW. 2001. Evolution of vertebrate steroid receptors from an ancestral estrogen receptor by ligand exploitation and serial genome expansions. Proceedings of the National Academy of Sciences of the United States of America 98(10): 5671-5676.
  • 87. Tilley WD, Marcelli M, Wilson JD, McPhaul MJ. 1989. Characterization and expression of a cDNA encoding the human androgen receptor. Proceedings of the National Academy of Sciences of the United States of America 86(1): 327-331.
  • 88. Todorov M, Mombelli E, Ait-Aissa S, Mekenyan O. 2011. Androgen receptor binding affinity: a QSAR evaluation. SAR and QSAR in environmental research 22(3): 265-291.
  • 89. Trudeau VL, Spanswick D, Fraser EJ, Lariviére K, Crump D, Chiu S, et al. 2000. The role of amino acid neurotransmitters in the regulation of pituitary gonadotropin release in fish. Biochemistry and Cell Biology 78: 241-259.
  • 90. Trudeau VL. 1997. Neuroendocrine regulation of gonadotropin II release and gonadal growth in the goldfish, Carassius auratus. Reviews of Reproduction 2: 55-68.
  • 91. Tyler C, Sumpter J. 1996. Oocyte growth and development in teleosts. Reviews in Fish Biology and Fisheries 6: 287-318.
  • 92. Tyler C, van der Eerden B, Jobling S, Panter G, Sumpter J. 1996. Measurement of vitellogenin, a biomarker for exposure to oestrogenic chemicals, in a wide variety of cyprinid fish. Journal of Comparative Physiology and Biology 166: 418-426.
  • 93. van der Burg B, Winter R, Man HY, Vangenechten C, Berckmans P, Weimer M, et al. 2010. Optimization and prevalidation of the in vitro AR CALUX method to test androgenic and antiandrogenic activity of compounds. Reproductive toxicology 30(1): 18-24.
  • 94. Villeneuve DL, Ankley GT, Makynen EA, Blake LS, Greene KJ, Higley EB, et al. 2007. Comparison of fathead minnow ovary explant and H295R cell-based steroidogenesis assays for identifying endocrine-active chemicals. Ecotoxicol Environ Saf 68(1): 20-32.
  • 95. Villeneuve DL, Breen M, Bencic DC, Cavallin JE, Jensen KM, Makynen EA, et al. 2013. Developing Predictive Approaches to Characterize Adaptive Responses of the Reproductive Endocrine Axis to Aromatase Inhibition: I. Data Generation in a Small Fish Model. Toxicological sciences : an official journal of the Society of Toxicology.
  • 96. Villeneuve DL, Mueller ND, Martinovic D, Makynen EA, Kahl MD, Jensen KM, et al. 2009. Direct effects, compensation, and recovery in female fathead minnows exposed to a model aromatase inhibitor. Environ Health Perspect 117(4): 624-631.
  • 97. Waller CL, Juma BW, Gray EJ, Kelce WR. 1996. Three-dimensional quantitative structure-activity relationships for androgen receptor ligands. Toxicology and Applied Pharmacolgy 137: 219-227.
  • 98. Wilson VS, Bobseine K, Lambright CR, Gray LE. 2002. A novel cell line, MDA-kb2, that stably expresses an androgen- and glucocorticoid-responsive reporter for the detection of hormone receptor agonists and antagonists. Toxicological Sciences 66: 69-81.
  • 99. Wilson VS, Cardon MC, Gray LE, Jr., Hartig PC. 2007. Competitive binding comparison of endocrine-disrupting compounds to recombinant androgen receptor from fathead minnow, rainbow trout, and human. Environmental toxicology and chemistry / SETAC 26(9): 1793-1802.
  • 100. Wolf JC, Dietrich DR, Friederich U, Caunter J, Brown AR. 2004. Qualitative and quantitative histomorphologic assessment of fathead minnow Pimephales promelas gonads as an endpoint for evaluating endocrine-active compounds: a pilot methodology study. Toxicol Pathol 32(5): 600-612.
  • 101. Yaron Z. 1995. Endocrine control of gametogenesis and spawning induction in the carp. Aquaculture 129: 49-73.
  • 102. Yin D, He Y, Perera MA, Hong SS, Marhefka C, Stourman N, et al. 2003. Key structural features of nonsteroidal ligands for binding and activation of the androgen receptor. Molecular pharmacology 63(1): 211-223.
  • 103. Young JM, McNeilly AS. 2010. Theca: the forgotten cell of the ovarian follicle. Reproduction 140(4): 489-504.
  • 104. Zhang X, Hecker M, Tompsett AR, Park JW, Jones PD, Newsted J, et al. 2008. Responses of the medaka HPG axis PCR array and reproduction to prochloraz and ketoconazole. Environ Sci Technol 42(17): 6762-6769.
  • 105. Organization for Economic Cooperation and Development. 2007. Report of Eight 21-day Fish Endocrine Screening Assays With Additional Test Substances for Phase-3 of the OECD Validation Program: Studies with Octylphenol in the Fathead Minnow (Pimephales promelas) and Zebrafish (Danio rerio) and with Sodium Pentachlorophenol and Androstenedione in the Fathead Minnow (Pimephales promelas). Phase-3 OECD 21-day Fish Screening Assay Validation Report Additional Test Substances Studies. Paris, France.
  • 106. Pawlowski S, Sauer A, Shears JA, Tyler CR, Braunbeck T. 2004. Androgenic and estrogenic effects of the synthetic androgen 17α-methyltestosterone on sexual development and reproductive performance in the fathead minnow (Pimephales promelas) determined using the gonadal recrudescence assay. Aquat Toxicol 68:277-291.
  • 107. Ankley GT, Jensen KM, Kahl MD, Korte JJ, Makynen EA. 2001. Description and evaluation of a short-term reproduction test with the fathead minnow (Pimephales promelas). Environ Toxicol Chem 20:1276-1290.