You are on page 1of 17

Regulatory Toxicology and Pharmacology 99 (2018) 142–158

Contents lists available at ScienceDirect

Regulatory Toxicology and Pharmacology


journal homepage: www.elsevier.com/locate/yrtph

Distinguishing between endocrine disruption and non-specific effects on T


endocrine systems
M. Sue Martya,∗, Chris Borgertb, Katie Coadya, Richard Greenc, Steven L. Levined, Ellen Mihaiche,
Lisa Ortegof, James R. Wheelerc, Kun Don Yig, Leah M. Zorrillaf
a
The Dow Chemical Company, Toxicology & Environmental Research and Consulting, 1803 Building, Midland, MI, 48674, USA
b
Applied Pharmacology and Toxicology, Inc., C.E.H.T. Dept. Physiological Sciences, University of FL College of Veterinary Medicine, 2250 NW 24th Avenue, Gainesville,
FL, 32605, USA
c
Dow AgroSciences, 3b Park Square, Milton Park, Abingdon, Oxfordshire, OX14 4RN, United Kingdom
d
Monsanto Company, Global Regulatory Science, 700 Chesterfield Parkway W, Chesterfield, MO, 63017, USA
e
Environmental and Regulatory Resources, LLC, 6807 Lipscomb Drive, Durham, NC, 27712, USA
f
Bayer CropScience, 2 TW Alexander Dr, Research Triangle Park, NC, 27709, USA
g
Syngenta Crop Protection, LLC, 410 S Wing Rd, Greensboro, NC, 27409, USA

A R T I C LE I N FO A B S T R A C T

Keywords: The endocrine system is responsible for growth, development, maintaining homeostasis and for the control of
Endocrine disruption many physiological processes. Due to the integral nature of its signaling pathways, it can be difficult to dis-
Endocrine system tinguish endocrine-mediated adverse effects from transient fluctuations, adaptive/compensatory responses, or
Systemic toxicity adverse effects on the endocrine system that are caused by mechanisms outside the endocrine system. This is
Stress
particularly true in toxicological studies that require generation of effects through the use of Maximum Tolerated
Weight of evidence
Endocrine screening
Doses (or Concentrations). Endocrine-mediated adverse effects are those that occur as a consequence of the
Maximum tolerated dose interaction of a chemical with a specific molecular component of the endocrine system, for example, a hormone
receptor. Non-endocrine-mediated adverse effects on the endocrine system are those that occur by other me-
chanisms. For example, systemic toxicity, which perturbs homeostasis and affects the general well-being of an
organism, can affect endocrine signaling. Some organs/tissues can be affected by both endocrine and non-en-
docrine signals, which must be distinguished. This paper examines in vitro and in vivo endocrine endpoints that
can be altered by non-endocrine processes. It recommends an evaluation of these issues in the assessment of
effects for the determination of endocrine disrupting properties of chemicals. This underscores the importance of
using a formal weight of evidence (WoE) process to evaluate potential endocrine activity.

1. Introduction molecules (hormones) with their conjugate receptors operating in lar-


gely the same set of hormonally responsive tissues. This simple fact
Biological functions are regulated in higher organisms by complex makes it difficult to design sensitive, hormone-specific eco/tox-
signaling networks within the neuroendocrine system. The endocrine icological assays that clearly distinguish permanent changes from
branch of this system is critically important in two broad classes of transient fluctuations, or adverse alterations from adaptive (and com-
biological functions – 1) regulation of growth and development, and 2) pensatory) responses (Keller et al., 2012). The difficulty is increased by
maintenance of internal homeostasis and normal physiological pro- the fact that all organs/tissues can be affected by physiological stimuli,
cesses (reproduction, temperature regulation, metabolism, etc). Some xenobiotic chemicals or natural substances, which may lead to tox-
of these functions are critical at a single point in the life cycle of an icological processes and events outside the control of the endocrine
organism, while others must be repeated continually throughout life. system, but which in turn, indirectly affect or trigger an endocrine re-
Although often studied separately, the endocrine system serves sponse.
these two major physiological roles using the same set of signaling This paper will identify and explain some of the non-endocrine


Corresponding author.
E-mail addresses: msmarty@dow.com (M.S. Marty), cjborgert@apt-pharmatox.com (C. Borgert), kcoady@dow.com (K. Coady),
Richard_Green@vrtx.com (R. Green), steven.l.levine@monsanto.com (S.L. Levine), emihaich@nc.rr.com (E. Mihaich), lisa.ortego@bayer.com (L. Ortego),
jrwheeler@dow.com (J.R. Wheeler), sue.yi@syngenta.com (K.D. Yi), leah.zorrilla@bayer.com (L.M. Zorrilla).

https://doi.org/10.1016/j.yrtph.2018.09.002
Received 13 July 2018; Accepted 4 September 2018
Available online 12 September 2018
0273-2300/ © 2018 The Authors. Published by Elsevier Inc. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/BY-NC-ND/4.0/).
M.S. Marty et al. Regulatory Toxicology and Pharmacology 99 (2018) 142–158

pathways and processes that can affect endpoint measurements and 2016).
interpretation of endocrine-mediated responses in endocrine screening Consideration of dose and realistic exposures is important for un-
assays and eco/toxicological studies. Factors such as cytotoxicity, derstanding toxicity responses that have an endocrine component. The
general systemic toxicity (including overt toxicity), stress, and experi- likelihood of altered homeostasis affecting endocrine-related endpoints
mental conditions can influence endocrine responses, which may ap- is increased in toxicological studies requiring the use of a maximum
pear as either reduced or enhanced endocrine signaling. This paper tolerated dose or concentration (MTD or MTC), which aims to produce
describes influential variables that can result in altered estrogen, an- an adverse effect in the test system. Even with endocrine-mediated ef-
drogen or thyroid signaling in vitro and in vivo. This information is fects, it is important to consider whether effective dose levels are close
useful to consider when conducting weight of evidence (WoE) evalua- to realistic exposure levels or whether there are large margins of ex-
tions and can reduce the degree of uncertainty in the overall assessment posure.
or in the interpretation of various endpoints. This may also assist with Addressing these and other challenges has been central to devel-
dose/concentration level selection and/or the inclusion of additional opment and continued refinement of the US EPA's EDSP and the de-
clarifying endpoints when designing studies to evaluate endocrine- velopment of the OECD's Conceptual Framework for the testing and
sensitive endpoints. assessment of endocrine disrupters (OECD, 2012a). During a review of
the EDSP, the FIFRA Scientific Advisory Panel (SAP) was asked to
2. Criteria for distinguishing endocrine-mediated from NON- comment on the validity of including treatments that demonstrated
endocrine-mediated effects on estrogen, androgen or thyroid overt systemic toxicity to assess effects of potential endocrine interac-
signaling tion. On the issue of overt systemic toxicity, the SAP in May of 2013
stated: “In summary, the Panel agreed that little, if any, weight should be
Endocrine-mediated effects are those that occur as a consequence of placed on signs of endocrine disruption in the presence of overt systemic
the interaction of a chemical with a specific molecular component of toxicity. All effects in endocrine sensitive tissues should be evaluated in terms
the endocrine system, such as a hormone receptor or transporter, a of primary interactions with the endocrine system vs. secondary effects re-
transcriptional enhancer or repressor of steroid-responsive genes, an lated to toxicity in non-endocrine organs or overall disruptions in home-
enzyme specific to the endocrine system, etc. The use of the potent ostasis”. The US EPA (2009a, b) recommended that an approximate ten
pharmaceutical estrogen, ethinylestradiol in oral contraceptives is a percent decrease in terminal body weight (with a statistically sig-
clear example of intentional and safe use of an endocrine-mediated nificant decrease in body weight gain) should be used as a general
adverse effect (i.e., endocrine disruption) to achieve control of a spe- guideline for mammalian assays because body weight decreases greater
cific reproductive process. Adverse effects on male reproductive per- than ten percent have been shown to confound the interpretation of
formance and secondary sex characteristics in response to potent reproductive system-related endpoints in the pubertal assays (Laws
pharmaceutical estrogens is also endocrine disruptive. Both occur as a et al., 2007). Furthermore, a 6% decrease in terminal body weight can
consequence of interaction with estrogen receptors (ERs) in target tis- affect pubertal assay thyroid endpoints. Similarly, clinical pathology
sues and in areas of the brain that mediate feedback control of hormone alterations like increased blood urea nitrogen and creatinine in the
production. absence of body weight changes indicate that the MTD has been ex-
Non-endocrine-mediated adverse effects on the endocrine system ceeded. Physiological changes associated with doses that produce sys-
are those that occur by mechanisms other than direct interaction of a temic or overt toxicity are often the consequence of activation of the
chemical with a specific component of the endocrine system. Such non- hypothalamic-pituitary-adrenal axis, which in turn affects all endocrine
endocrine-mediated modes of actions (MoAs) could include toxicity to a systems including those that are the focus of the EDSP. This thinking
tissue or organ of the endocrine system or to physiological processes supports the regulatory approach in the US, but acceptance at the OECD
that either control or are controlled by the endocrine system. For ex- level is less clear. Greater emphasis should be placed on using these
ample, multiple changes in hormone levels occur and contribute to concepts in the design and interpretation of eco/toxicological assays.
parturition; however, this process also requires muscle contraction. If a
toxicant blocks muscle contraction, this is not an endocrine MoA. A 3. In vitro endocrine screening assays – responses confounded by
second example is the reduction of testosterone levels caused by abuse description of normal cellular function and cytotoxicity
of ethyl alcohol, which is due to altered metabolism of steroid hor-
mones that arises due to chronic liver toxicity rather than to a direct New toxicity testing strategies increasingly incorporate in vitro as-
interaction of ethanol with steroidogenesis. says, either to replace traditional animal studies or to aid in identifying
Endocrine- versus non-endocrine-mediated adverse effects can often the key biochemical perturbations that produce adverse effects in those
be distinguished using the lead toxic effect approach (Bars et al., 2011, studies (NRC, 2007; Judson et al., 2010). In vitro assays can be useful
2012), where the adverse effect that occurs at the lowest dose/con- screening tools and provide mechanistic information on initiating or
centration is considered the lead toxic effect. This approach describes intermediate events in adverse outcome pathways (AOPs) (Judson
the most sensitive effect, which may assist in the determination of et al., 2013; Ankley et al., 2010). However, to reliably use results from
whether the apparent endocrine effect is mediated by an interaction in vitro endocrine assays, assessors must be able to distinguish between
with a component specific to the endocrine system or is instead pro- endocrine-mediated and non-endocrine-mediated effects. The analog in
duced as a result of the lead toxic effect itself. This concept is critically in vitro assays for overt chemical toxicity observed in whole animal
important as it can help determine the necessity for conducting addi- assays is cell stress and cytotoxicity-mediated processes that disrupt
tional tests to describe the hazard of a substance. For instance, in the specific biomolecular functions. Cell-disruptive processes include pro-
EPA's Endocrine Disruptor Screening Program (EDSP), a true endocrine tein, DNA or lipid reactivity, physico-chemical disruption of proteins or
disruptive effect may require additional higher tier testing to fully de- membranes (e.g., by surfactants), or processes such as apoptosis, oxi-
scribe the dose/concentration relationship before risk assessment can dative stress response, mitochondrial disruption, endoplasmic re-
be performed. On the contrary, if the effect is due to a non-endocrine ticulum stress, microtubule disruption, or a heat shock response among
MoA, the existing data package may already fully describe the tox- others. There are also confounding factors that can alter assay stability
icological response for the lead effect, making additional testing re- in cell free systems that can include changes in buffer pH, receptor
dundant. Further, in some regions it may be extremely important to denaturation, protein-protein and protein-membrane interactions, dis-
separate endocrine-mediated from non-endocrine-mediated toxicities as ruption/enhancement of binding kinetics, test substance precipitation,
some legal frameworks will regulate these differently (e.g. the endo- aggregation of individual molecules and potentially other mechanisms.
crine hazard-based cut-off criterion in the EU) (Wheeler and Coady, Recently, some limitations of in vitro screening assays in ToxCast

143
M.S. Marty et al. Regulatory Toxicology and Pharmacology 99 (2018) 142–158

were identified by examining a diverse chemical and assay space comprehensive assessment of non-specific factors that could potentially
(Judson et al., 2016). ToxCast is a large battery of assays with over 800 confound assay results and interpretation and it is incumbent on the
in vitro assay endpoints from seven high-throughput platforms. Ap- investigators to find the maximum allowable in vitro concentration for
proximately 15% of the assays in ToxCast assess endocrine endpoints these diverse biological systems. The National Toxicology Program In-
related to the estrogen, androgen and thyroid pathways. To aid in the teragency Center for the Evaluation of Alternative Toxicological
interpretation of results, ToxCast includes numerous cell stress and Methods (NICEATM) and the Interagency Coordinating Committee on
cytotoxicity assays but not all assays are evaluated for cytotoxicity or the Validation of Alternative Methods (ICCVAM) recommended for cell
impacts to cell-free systems. Cytotoxicity has been defined within line assays that only concentrations that do not cause greater than 10%
ToxCast as a positive call in two or more cytotoxicity assays or cyto- cytotoxicity should be included in an analysis (NICEATM and ICCVAM,
static effects in a proliferation assay. The threshold of two was selected 2011). Allowing for high levels of cytotoxicity (i.e., > 10%), without a
based on the observation that most chemicals with activity in two or correction for cytotoxicity, greatly increases the likelihood of producing
more of these cytotoxicity assays displayed a concentration response. a false-positive or false-negative response depending on the test system.
Not surprisingly, cell-free assays in ToxCast showed a rapid increase in Although the ER binding assay has been used as a research tool for
the frequency of responses at concentrations where cell stress/cyto- decades, several factors may affect assay performance and interpret-
toxicity responses were observed in the cell-based assays. The observed ability (Laws et al., 2006). In an assessment of the performance of the
activity could be divided into two categories: (1) specific biomolecular ER binding assay (OCSPP 890.1250), seven out of 50 chemicals that
interactions against one or more target concentrations below the covered a broad range of structural and physiochemical diversity re-
threshold for overt cell stress/cytotoxicity, and (2) non-specific bio- quired diagnostic secondary Ki experiments to demonstrate that these
molecular interactions associated with cell stress or cytotoxicity re- chemicals were not true competitive inhibitors of ER binding (Laws
sulting from triggering specific cell stress pathways, disruption of pro- et al., 2006). From these secondary plots, it was shown that the seven
teins or membranes, or broad low-affinity non-covalent interactions. chemicals altered the stability of the assay by either changing buffer
Several trends were identified in the ToxCast analysis (Judson et al., pH, denaturing ER and changing its conformation, or disrupting the ER
2016). Chemicals showing a greater number of specific biomolecular binding kinetics. If one of these factors is at play, it is not uncommon to
interactions were generally designed to be bioactive (e.g., pharmaceu- see a large change in ER binding (i.e., steep slope) over a single log unit
ticals) whereas intentional food-use chemicals or pesticides targeting (e.g., 0% inhibition to 90% inhibition). Therefore, if a response is ob-
systems that are phylogenetically distant from humans tended to show served in the ER binding assays, it is important to evaluate whether the
the fewest specific interactions (e.g., herbicides). For many chemicals, a response represents a competitive or non-competitive interaction and is
disproportionately large number of positive assay responses was ob- not merely an artifact of one of the above-mentioned factors. The an-
served in the concentration range where cytotoxicity was observed. drogen receptor (AR) binding assay, like the ER binding assay, has also
This phenomenon, where there was a steep increase in cytotoxicity was been used as a research tool for decades and the EPA test guideline for
termed the “burst region”. A metric termed the Z-score was developed the AR binding assay outlined in OCSPP 890.1150 is based on the es-
to assess specificity by indicating whether true activity or cytotoxicity tablished methodology of Kelce et al. (1995). The same factors dis-
has occurred, with a high Z-score representing high specificity and a cussed above that impact the ER binding assay apply to the perfor-
low Z-score representing low specificity. A class of compounds that mance and interpretability of the AR binding assay.
consistently showed non-specific responses was surfactants and this In addition to the ER and AR binding assays, the Tier 1 EDSP battery
non-specific activity reflects the ability of surfactants to disrupt cell also required screening for potential estrogen agonist activity with a
membrane structure and function and protein–protein interactions. cell-based ER transcriptional activation (ERTA) assay (OCSPP
Graphical analyses comparing the concentration where cytotoxicity 890.1300). However, since the time of the initial compounds going
occurs versus responses for endocrine and non-endocrine screening through the EDSP, the ERTA has been validated to also assess antag-
assays in ToxCast is presented in Mihaich et al. (2017a, b) for propi- onism (OECD, 2012b) and OECD has recently developed a guideline for
conazole and triclosan. an ARTA assay that can assess agonism and antagonism (OECD 455;
Recently, high-content imaging (HCI), which applies automated OECD, 2016). For both assays, cytotoxicity is evaluated with a con-
image analysis to capture multiple cytological features, has been eval- stitutive luciferase reporter construct and cannot exceed 20%. An as-
uated within ToxCast to determine a “tipping point” or point of de- sessment of cytotoxicity is arguably more important for the assessment
parture for adverse impacts in cell line-based assays (Shah et al., 2016). of antagonism because cytotoxicity can impact reporter activity by
The aim of this work was to integrate an analysis of impacts on normal disturbing normal cellular function. A positive response for antagonism
cellular function to identify transition points between adaptive changes is achieved with ≥30% inhibition of induced ER or AR reporter ac-
and adverse effects that will allow for the appropriate interpretation of tivity. Therefore, it is recommended that when cytotoxicity is ap-
ToxCast data. The results of Shah et al. (2016) underscore re- proaching a significant level (e.g., ≥10%), ER and AR reporter activity
commendations by Judson et al. (2016) regarding the importance of should be corrected for the level of observed cytotoxicity (i.e., signal
characterizing concentration- and time-dependent effects on cytotoxi- adjusted for the proportion of viable cells remaining).
city and MoA endpoints to accurately characterize the sequence of ef- The aromatase assay (OCSPP 890.1200) measures the ability of a
fects and to support or negate a possible MoA for an adverse endocrine test substance to inhibit conversion of testosterone to estrogen. The EPA
outcome. More development is required to implement HCI metho- test guideline recommends conducting the assay using commercially-
dology into in vitro platforms to understand the role of adaptive cellular available human recombinant microsomes containing CYP19 plus re-
responses and pathways that lead to adverse in vivo outcomes ductase, and as such, may be less susceptible to inconsistency compared
(Boekelheide and Andersen, 2010). to cytosolic preparations from animal tissue. Overall, the method has
The EDSP battery used to evaluate List 1 compounds included five in been shown to be sensitive and specific. However, like the ER and AR
vitro assays that screened estrogen and androgen endpoints (Table 1) competitive binding assays, chemicals that denature or affect the
and these assays are included in Level 2 of the OECD Conceptual Fra- structure of the enzyme in vitro could produce a false positive result.
mework (OECD, 2012a). The androgen receptor transcriptional acti- This in vitro assay can be especially susceptible to factors that denature
vation assay (ARTA; OECD 458; OECD, 2016, Table 2) was not vali- or affect the structure of the enzyme because the total protein con-
dated at the time List 1 chemicals were screened but if it was available, centration in the assay is extremely low. For this reason, cytosolic
it likely would have been included in the in vitro battery. These in vitro preparations could be less sensitive to chemicals that denature or affect
assays screen for potential endocrine modulation through a specific the structure of the enzyme. Evidence of this assay's potential sus-
endocrine mechanism. Therefore, it is critical to include a ceptibility to confounding effects from test chemicals is foreshadowed

144
M.S. Marty et al. Regulatory Toxicology and Pharmacology 99 (2018) 142–158

Table 1
EPA's EDSP Tier I in vitro Screening Battery.
Assay Type Assay Description

ER binding assay An ER binding assay that utilizes rat uterine cytosol to examine the ability of a test chemical to bind with estrogen receptors. Assesses ER
agonism and antagonism.
ER transcriptional activation assay An ER binding assay that uses a human cell line to examine the ability of a test chemical to bind with one of the estrogen receptors. Assesses
ER agonism but can also assess and differentiate antagonism.
AR binding assay An AR binding assay that utilizes rat prostate cytosol to examine the ability of a test chemical to bind with androgen receptors. Assesses AR
agonism and antagonism.
Aromatase assay Aromatase is an enzyme responsible for estrogen biosynthesis that converts androgens into estrogens, estradiol, and estrone. The aromatase
assay uses a human recombinant form of the protein and focuses on this portion of the steroidogenic pathway to detect substances that
inhibit aromatase activity.
H295R Steroidogenesis assay The Steroidogenesis assay utilizes the H295R human adrenocortical carcinoma cell line to detect interference and induction with the body's
production of male and female steroid sex hormones (estrogen and testosterone).

Table 2 measured in the assay are concentrations of testosterone and 17β-es-


Referenced test guideline (TG) studies to evaluate potential endocrine activity. tradiol in the cell culture media after 48 h of exposure. This assay re-
Test Guideline U.S. EPA TG No. OECD TG
quires an evaluation of cytotoxicity that is preferably run parallel and
No. cytotoxicity should not exceed 20%. The test guideline recommends
performing either a visual categorical examination of the cells, per-
In Vitro Assays forming the Live/Dead Assay® or performing the MTT assay to assess
ER Binding Assay (Rat uterine cytosol) OCSPP 890.1250 OECD 493
cytotoxicity. On mechanistic grounds, the MTT assay is probably the
ERα Transcriptional Activation Assay OCSPP 890.1300 OECD 455 most appropriate assay because it largely measures mitochondrial
AR Binding Assay OCSPP 890.1150 function (Berridge et al., 2005). A reliable evaluation of mitochondrial
AR Transcriptional Activation Assay OECD 458 function is critical because the mitochondrial electrochemical gradient
Steroidogenesis Assay (H295R cells) OCSPP 890.1550 OECD 456
is required for the steroidogenic acute regulatory protein to transfer
Aromatase Assay OCSPP 890.1200
cholesterol to the inner membrane of mitochondria and initiate ster-
In Vivo Assays oidogenesis (King and Stocco, 1996). Therefore, to properly assess
mitochondrial function and potential impacts to steroidogenesis, a
Uterotrophic Assay (Rat) OCSPP 890.1600 OECD 440
more sensitive indicator of mitochondrial function than the MTT assay
Hershberger Assay (Rat) OCSPP 890.1400 OECD 441
Pubertal Female Assay (Rat) OCSPP 890.1450
may be required and the MitoTracker® Red CMXRos assay that directly
Pubertal Male Assay (Rat) OCSPP 890.1500 assesses mitochondrial potential is a consideration for ToxCast assays
Reproduction/Developmental Toxicity OCSPP OECD 421; (Levine et al., 2007; Shah et al., 2016). A positive call in the test
Screening Test (RDTS) (Rat); Combined 870.3550; 422 guideline is detecting a statistically significant difference from the
Repeated Dose Toxicity Study with RDTS 870.3650
control, which could be less than a 20% mean difference from control
Prenatal Developmental Toxicity Study (Rat, OCSPP 870.3700 OECD 414
Rabbit) levels. Consequently, it is recommended that when cytotoxicity is ap-
Two-Generation Reproductive Toxicity Study OCSPP 870.3800 OECD 416 proaching a significant level, but still less than 20%, reporter activity
(Rat) should be corrected for this cytotoxicity.
Extended One-Generation Reproductive OECD 443 Recently, the H295R steroidogenesis assay has become one of the
Toxicity Study (Rat)
Fish Short-term Reproduction Assay OCSPP 890.1350 OECD 229
high-throughput assays included in ToxCast and these data could be
Amphibian Metamorphosis Assay (Frog) OCSPP 890.1100 OECD 231 used to prioritize testing under the EDSP (Karmaus et al., 2016; US EPA,
Medaka Extended One Generation OCSPP 890.2200 2017a). Modifications to the OECD 456 test guideline by ToxCast in-
Reproduction Test clude raising the level of basal steroidogenesis with a 48-h pre-induc-
tion with forskolin. Measurement of 13 steroidogenic hormones from
Test guidelines available at:
four hormone classes (glucocorticoids, progesterones, androgens, es-
OCSPP: https://www.epa.gov/test-guidelines-pesticides-and-toxic-substances/
series-890-endocrine-disruptor-screening-program and https://www.epa.gov/ trogens) is performed using a novel high throughput HPLC-MS/MS
test-guidelines-pesticides-and-toxic-substances/series-870-health-effects-test- (high-performance liquid chromatography followed by tandem mass
guidelines. spectrometry) method. The permissible level of cytotoxicity using the
OECD: https://www.oecd-ilibrary.org/environment/oecd-guidelines-for-the- MTT assay, before a concentration is rejected from analysis, is currently
testing-of-chemicals-section-4-health-effects_20745788. ≥30%. A concern with using this criterion to set a maximally tolerated
concentration is that a ≥30% effect on mitochondrial function could
in the guideline, which cautions that the presence of any detergent artificially produce an unacceptably large number of false positive re-
residue on glassware can impact activity. A chemical that has detergent sults. In addition, the MitoTracker assay discussed above could be a
like properties can denature proteins or impact protein-protein inter- replacement for the MTT assay to improve sensitivity and specificity.
actions (i.e., CYP19 interactions with its associated reductase), affecting Therefore, either a correction for cytotoxicity or lowering the criterion
assay results and would not be representative of potential effects in vivo. for allowable toxicity may be required to minimize false positives.
This cautionary statement is consistent with the findings of Judson et al.
(2016), which pointed out low Z-scores for surfactants in ToxCast. 4. In vivo endocrine screening assays: non-endocrine target organ
The steroidogenesis assay (OECD 456; OECD, 2011), measures the effects from chemicals
ability of a chemical to stimulate or inhibit the production of steroid
hormones with a unique steroid hormone-producing human cell line. In intact animals, the endocrine system integrates activity across
The EDSP initially intended to use a minced testes assay, but that multiple organ systems to maintain homeostasis and interpret whether
method was found to be unsuitable because it could not adequately external environmental conditions are suitable for reproduction.
distinguish between cytotoxicity and steroid synthesis inhibition However, not only does the endocrine system affect the functioning of
(Powlin et al., 1998). The EPA, in coordination with OECD, subse- multiple organ systems, multiple organ systems also can affect endo-
quently developed and validated the H295R assay. The endpoints crine system function. Consequently, when a substance is evaluated for

145
M.S. Marty et al. Regulatory Toxicology and Pharmacology 99 (2018) 142–158

potential endocrine activity, careful characterization of target organ upon long term exposure” (EFSA, 2016).
toxicity should be included. These data can provide information on Conversely, hepatic enzyme induction is often seen in toxicity stu-
potential non-endocrine-mediated effects on endocrine endpoints and dies with rodents. Some test substances may induce liver enzymes with
provide some regulatory context to interpret changes in endocrine- enhanced testosterone metabolism and clearance (e.g., Freyberger and
sensitive tissues. If endocrine endpoints are affected at treatment levels Schladt, 2009; Marty et al., 2015). In general, metabolism markedly
higher than those required to produce target organ toxicity, regulatory decreases or inactivates testosterone's androgenic activity with the ex-
exposure limits are likely to be protective against potential effects on ception of dihydrotestosterone (DHT). Prepubertal animals may be
the endocrine system. particularly sensitive to this MoA, because, unlike the negative feed-
Many aspects of the endocrine system are conserved across species, back loop in adult male rats, where higher testosterone levels have an
including, in some cases, the response to systemic (general) toxicity. inhibitory effect on GnRH/LH release, in prepubescent male rats, in-
While the endocrine system is still being characterized, some interesting creasing testosterone levels are stimulatory, potentiating the pituitary's
parallels between mammalian and non-mammalian responses to sys- response to GnRH, resulting in maximum LH release between 35 and 45
temic toxicity have come to light in recent years. In both mammalian days of age (Stoker et al., 2000b). The increase in LH further increases
and non-mammalian species, systemic (e.g., stress) or target organ testosterone secretion, resulting in puberty onset and increased acces-
toxicity (e.g., toxicity in the liver, kidneys, nervous system) can alter sory sex tissue (AST) size. With hepatic enzyme induction prior to
endocrine endpoints as described below. These examples highlight the puberty, testosterone may be metabolized at a greater rate, delaying
importance of a WoE approach to differentiate specific vs. non-specific maximum LH release and attainment of sufficient testosterone levels to
endocrine findings. drive puberty onset and/or AST weight increases, which are marked
between 40 and 55 days of age; (Monosson et al., 1999; Ashby and
4.1. Liver effects Lefevre, 2000). There is some evidence for this MoA in a recent series of
studies with pronamide (Marty et al., 2015; Rasoulpour et al., 2015).
4.1.1. Mammals Liver effects in rats also can lead to activation of the hypothalamic-
While not the primary source of estrogen, androgen or thyroid pituitary-thyroid (HPT) axis and secondary alterations in thyroid end-
hormone production, the liver can affect serum levels of these hor- points, including serum hormone levels, thyroid weight and histo-
mones. The liver plays an important role in synthesizing transport pathology. Enzyme induction in the liver may enhance thyroid hor-
proteins for hormone binding, as well as hormone metabolism and mone (thyroxine, T4) conjugation and clearance, particularly in rats
excretion (DePeyster and Mihaich, 2014; Khan et al., 1981, 2002). that, as a species, are sensitive to this MoA (e.g., McClain, 1995; Jahnke
Thus, liver effects, which are a relatively common response in MTD/ et al., 2004). Liver enzyme induction is typically accompanied by in-
MTC toxicity studies, can cause secondary endocrine effects in mam- creases in liver weights, as well as changes in liver histopathology. This
mals through: (a) Direct damage or degenerative changes to the liver decrease in serum T4 will activate the HPT axis and subsequently can
leading to reduced functional capacity; or (b) Enlargement of the liver affect thyroid-related endpoints. However, humans are quantitatively
accompanied by induction of biotransformation enzymes leading to less sensitive to this MoA (Dellarco et al., 2006), because: 1) transient
increased hormone clearance, resulting in secondary effects on circu- decreases in T4 do not readily stimulate TSH increases in humans; and
lating hormone levels. Regulations that protect against liver toxicity 2) humans have thyroid binding globulin (TBG) with a higher binding
may be sufficient to protect against indirect effects on the endocrine affinity for thyroid hormone transport in the blood, making it less likely
system. to release T4 for hepatic metabolism and clearance than albumin, the
In human cases of marked liver dysfunction, altered steroid hor- primary transporter in adult rats (Choksi et al., 2003).
mone levels have been reported, possibly due to alterations in binding In contrast to rats, humans are likely to be less sensitive to hormone
proteins and/or enhanced hormone catabolism. Sex hormone altera- metabolism secondary to enzyme induction due to the presence of a
tions have been demonstrated in males and females with chronic liver high-affinity steroid hormone transport protein, SHBG and TBG (Dunn
disease. Men with liver disease are often hypogonadal with gyneco- et al., 1981). In humans, most testosterone/estrogen and thyroid hor-
mastia, testicular atrophy, decreased testosterone and androstenedione mones travel tightly bound to SHBG or TBG, respectively, with only a
levels and increased dehydroepiandrostenedione sulphate levels small fraction (1–2%) unbound as the biologically active fraction. These
(Bannister et al., 1987; Gluud, 1988). Both men and women were re- proteins regulate access of hormones into target cells (Hammond et al.,
ported to have increased levels of estrogens and sex hormone binding 2012; Siiteri et al., 1982). Conversely, peripubescent and adult rodents
globulin (SHBG), which can affect hormone signaling. While total rely heavily on albumin to transport hormones in blood, which has a
hormone levels may appear increased for a period with increased much lower dissociation constant (Kd) for steroids (10−6 to 10−4 M)
SHBG, free hormone levels may be decreased when liver dysfunction is than SHBG (10−10 to 10−8 M). A similar difference is seen with thyroid
coupled with elevated SHBG. In women, liver disease can alter sex hormone, where the T4 association constant (Ka) is 1010 for TBG vs.
hormone metabolism, resulting in decreased serum levels of steroid 2 × 108 for transthyretin and 1.5 × 106 for human serum albumin
sulphates and 5-alpha-dihydrotestosterone (Becker, 1993). Chronic (Refetoff, 2015). Free serum hormones are more readily metabolized
liver disease may lead to early menopause and has been associated with and cleared from the body; thus, the concentration and affinity of
lower levels of hepatic ER in females (Becker, 1993; Shimizu, 2003). hormones for transport proteins has been directly related to metabolic
Burra (2013) has reviewed the relationship between liver abnormalities clearance rates (Siiteri et al., 1982).
and endocrine diseases, including altered thyroid hormone levels in These data raise the question as to whether liver toxicity with en-
some patients with liver disease. zyme induction, increased liver weight and centrilobular hypertrophy is
Liver dysfunction may result in decreased hepatic enzyme activity, considered systemic toxicity when endocrine changes occur secondary
which can have secondary effects on the endocrine system. A good to these effects (Maronpot et al., 2010). EPA Guidance (US EPA, 2003)
example of this is inhibition of hepatic hydroxyphenolpyruvate dehy- provides that a WoE for hepatic centrilobular hypertrophy may be
drogenase (HPPD), an enzyme involved in tyrosine catabolism. “considered adverse if associated with biologically relevant increases in
Inhibition of HPPD results in accumulation of tyrosine (tyrosinaemia) clinical chemistry parameters, particularly those indicative of effects on
which has been shown to result in non-endocrine-mediated effects on hepatocyte function, hepatocellular membrane leakage, …” For ex-
the thyroid in the rat. For example, in their recent review of mesotrione ample, a quantitative associated clinical chemistry indicator for hepa-
(an HPPD-inhibiting herbicide), the European Food Safety Authority tocellular membrane injury in the liver of rats and mice is at least a 2-
(EFSA) stated that: “Increased incidence of thyroid follicular adenomas was fold increase in alanine aminotransferase (ALT) enzyme levels as
also considered a secondary effect of increased levels of tyrosinaemia in rats compared to controls. Thus, liver hypertrophy and a subsequent

146
M.S. Marty et al. Regulatory Toxicology and Pharmacology 99 (2018) 142–158

increase in hormone clearance could occur without meeting a definition vitamin D levels as a result of impaired kidney function trigger a
of systemic toxicity. This “non-adverse” designation is consistent with a compensatory increase in parathyroid hormone synthesis and para-
variety of natural dietary constituents that induce phase I and phase II thyroid hyperplasia (Lewin et al., 2005).
enzymes (e.g., cruciferous vegetables, coffee/tea, herbs/spices like The role of the kidney in health maintenance and its ability to im-
garlic, etc.; Manson et al., 1997) and could increase hormone catabo- pact the endocrine system is acknowledged by the US EPA by including
lism without inducing systemic toxicity. renal function as part of the MTD criteria for the male and female
pubertal assays (OCSPP 890.1450 and 890.1500). These test guidelines
4.1.2. Fish state, “… abnormal blood chemistry values at termination (particularly
Similarly to mammals, fish undergo alterations in some endocrine- creatinine and blood urea nitrogen (BUN)) may indicate that MTD was
sensitive endpoints in response to alterations in liver function. One exceeded, even in the absence of a reduction in terminal body weight
primary endocrine endpoint monitored in fish, vitellogenin (Vtg), is compared to controls”. However, creatinine and BUN are insensitive
sensitive to modulation by both endocrine effects and altered liver biomarkers of kidney damage; and some kidney injury can progress
function. Vtg is the primary egg yolk protein in fish. Vtg is under es- undetected until damage is advanced. One option when testing com-
trogenic control via free estradiol (E2) interaction with the ER in the pounds that target the kidney is to better characterize kidney toxicity
liver leading to increased hepatic Vtg synthesis (Wheeler et al., 2005; during the range-finding study by using newer urinary biomarkers (e.g.,
Hutchinson et al., 2006). Decreased circulating Vtg in female fish α-glutathione-S-transferase (α-GST), clusterin, and/or renal papillary
plasma may indicate lower E2 production due to steroidogenesis in- antigen-1 (RPA-1)) in conjunction with BUN and creatinine (Harpur
hibition (Ankley and Gray, 2013) or a direct anti-estrogenic interaction et al., 2011; Fuchs and Hewitt, 2011). These new biomarkers, which
with ER (Smeets et al., 1999; Panter et al., 2002). However, other have been accepted by the US Food and Drug Administration and the
factors that may influence the expression of Vtg also should be con- European Medicines Agency, are more sensitive and specific for the
sidered as they could lead to a false positive conclusion for endocrine detection of early stage renal damage and may localize the site of da-
activity from fish endocrine assays (e.g., Fish Short-Term Reproduction mage. These data may be useful to document functional kidney
Assay [FSTRA]; OCSPP 890.1350) (Wheeler and Coady, 2016). changes, coupled to renal pathology, in selecting an MTD based on the
A number of specific, but non-endocrine mechanisms can lead to kidney effects for assays with endocrine-sensitive endpoints (e.g.,
reduced Vtg in female fish (Wheeler and Coady, 2016). Liver effects are pubertal assays).
a relatively common response (particularly for pesticidal active sub-
stances). Similar to mammals, both mechanisms (liver dysfunction and
enzyme induction) could potentially occur in fish, leading to a reduc- 4.2.2. Fish
tion in circulating female Vtg. Direct toxicity to the liver could reduce The kidneys in fish also play important roles in ion and water bal-
its capacity to produce Vtg or non-endocrine-mediated liver effects ance as well as the excretion of endogenous and exogenous solutes.
could reduce circulating levels of E2 (via induction of biotransforma- While kidney structure and function varies among different fish species,
tion enzymes) available to bind the ER and so transcribe Vtg mRNA. For most fish kidneys are composed of an encapsulated capillary network
example, the triazole fungicide, tebuconazole, has been shown to cause called the glomerulus and a tubule lined with epithelial cells.
reduced yolk accumulation in oocytes (histopathologically) of fathead Freshwater fish, since they are hyperosmotic relative to their environ-
minnow exposed over sexual development (EU DAR, 2012). However, ment, have a higher glomular filtration rate as compared to saltwater
this was only concomitant with liver cell degenerative changes in both fish, which are hypoosmotic. In fish, up to 80% of the cardiac output
sexes, indicating it did not have a primary endocrine mechanism but drains (via the caudal vein) to the kidneys, compared to ∼20% of
was secondary to liver toxicity. Additionally, in a tebuconazole FSTRA, cardiac output flowing to the kidneys in mammals. Thus, along with the
blood plasma Vtg was reduced at the high treatment level, and again gills, the kidneys are important for the elimination of exogenous com-
histopathological changes in the liver (necrosis) were concurrent at the pounds, and fish kidneys receive a large amount of blood flow, making
reduced Vtg and next lower concentration (US EPA, 2015a), further them targets of chemical exposure (Kleinow et al., 2008).
supporting that endocrine-related changes may be secondary to liver Fish kidney histopathology has been evaluated in toxicity studies,
toxicity. particularly in response to metal exposure (Lemly, 2002; Mishra and
Mohanty, 2008). Renal toxicity has reportedly been the result of in-
4.2. Kidney effects terference with ion exchange activity at lower concentrations, and as
the result of lethal cell damage at higher concentrations (Bonga and
4.2.1. Mammals Lock, 2008). Histopathology of the kidney of the medaka fish at 15
The kidneys play a critical role in fluid and electrolyte balance, weeks post fertilization is included in the OECD and USEPA test
detoxification and excretion in mammals, and alterations in kidney guidelines for the Medaka Extended One Generation Reproduction Test
function can have secondary effects that can be misinterpreted as direct (MEOGRT; US EPA, 2015b; OECD, 2015). Renal impairment can be the
effects on steroidogenesis. For example, kidney alterations in electro- result of exposure to estrogenic chemicals resulting in increased pro-
lytes or blood volume could result in secondary effects on the adrenal duction of Vtg which can damage the kidney via protein overload. This
gland through the renin-angiotensin system, which can result in in- is expected to be more pronounced in male fish with estrogenically
creased adrenal weight and adrenal histopathology (Haschek et al., induced high levels of Vtg protein which have no outlet via deposition
2010). A change in adrenal weight is a potential indicator of ster- in developing oocytes. However, histopathological findings in the
oidogenesis inhibition; however, it is important to distinguish this kidney of exposed fish may also reflect the influence of systemic toxi-
kidney-adrenal response from primary effects on steroidogenesis or city. Limited guidance on differentiation of kidney histopathology that
effects on the hypothalamic-pituitary-adrenal axis, which could be results from systemic toxicity versus exposure to estrogenic substances
mediated by stress. In cases of electrolyte or blood volume changes, the is provided in Appendix 12 of the US EPA MEOGRT test guideline
pituitary may not exhibit histopathological changes, because the hy- (OCSPP 890.2200; US EPA, 2015b). As experience progresses with the
pothalamic-pituitary-adrenal axis is not involved in these adrenal glo- evaluation of kidney toxicity in fish, it will be important to summarize
merulosa-kidney interactions. Under other circumstances, renin can histopathological findings and their connection to either endocrine
affect the gonadal hormone pathways (see below). Another example of MoA or in response to more general, systemic toxicity that can interfere
how kidney toxicity can result in secondary effects on the endocrine with ion transport or result in kidney cell damage and/or death.
system is renal osteodystrophy (ROD) resulting in secondary hy-
perparathyroidism. In ROD, imbalances in calcium, phosphorous and

147
M.S. Marty et al. Regulatory Toxicology and Pharmacology 99 (2018) 142–158

Table 3
High dose levels for EDSP tier 1 assays compared with points of departure for neurotoxicity or biomarkers.
Chemical Uterotrophic High Dose Hershberger High Dose Male Pubertal High Female Pubertal High POD NOAEL (mg/kg/day)- POD NOAEL (mg/kg/day)-
Level (mg/kg/day) Level (mg/kg/day) Dose Level (mg/kg/ Dose Level (mg/kg/ Chronic (EPA Risk Subchronic (EPA Risk
day) day) Assessment) Assessment)

Permethrin NA 150 120 150 25 11


Bifenthrin 15 10 10 10 3.1 3.1
Cyfluthrin 20 40 40 40 2.4
Chlorpyrifos 4 12 2 2 ≥0.78a
Imidacloprid 100 80 100 100 8 8

Abbreviations: POD - Point of Departure; NOAEL - No Observed Adverse Effect Level.


a
Benchmark Dose (BMD) is lower based on red blood cell cholinesterase inhibition; BMD10 ≥ 0.15 mg/kg/day.

4.3. Nervous system effects endpoints that may be altered; 2) the magnitude of brain AChE in-
hibition that is occurring; and 3) whether regulating these compounds
4.3.1. Mammals for protection against inhibition of brain AChE (or the more sensitive
Toxicants may target the nervous system, resulting in primary endpoint, RBC cholinesterase) will be protective for endocrine-related
neurotoxicity, which at higher dose levels can progress to secondary effects.
alterations in endocrine signaling pathways. One example of target The pyrethroid insecticides are another class of chemicals that act
organ toxicity in the nervous system, which has been associated with on the nervous system, but also have been reported to alter endocrine
potential endocrine activity, is acetylcholinesterase (AChE) inhibition function in some studies. The pyrethroids are synthetic derivatives of
by organophosphates and carbamates. Chlorpyrifos, an organopho- the natural pyrethrins, which are designed to be more photostable than
sphate, can be used as one case study. In one published study with the pyrethrins, and therefore suited for crop protection and other
chlorpyrifos, Joshi et al. (2007) reported that male Wistar rats exposed commercial uses. Pyrethroids act on voltage-sensitive sodium channels
to 7.5, 12.5 or 17.5 mg/kg bw/day chlorpyrifos by oral gavage had in the insect and in mammals to produce transient manifestations of
decreased testis weights, reduced spermatid/sperm counts, decreased acute neurotoxicity (Soderlund et al., 2002). The effects observed fol-
serum testosterone levels, degenerative changes in the seminiferous lowing oral administration of pyrethroids, the preferred route in the
tubules of the testis, and decreased fertility. Rats in all dose groups EDSP studies, are well defined following administration in rodents, and
exhibited significant clinical signs, which were likely due to substantial the two subclasses are based upon chemical structure and the produc-
inhibition of brain AChE. Thus, these doses were confounded by overt tion of either tremors (T-syndrome) with Type I pyrethroids or chor-
toxicity as defined by the EDSP test guidelines (e.g., US EPA, 2009a, b). eoathetosis with salivation (CS-syndrome) with Type II pyrethroids.
A second study by De Angelis et al. (2009) reported decreased serum T4 One of the criteria used for dose selection for the mammalian EDSP tests
and altered thyroid histopathology (exfoliated necrotic follicular cells) is evidence of acute neurotoxicity. With cyfluthrin, dose levels for the
at postnatal day (PND) 150 in CD1 mice exposed to ≥ 3 mg/kg bw/kg Uterotrophic, Hershberger and Male and Female Pubertal Assays were
chlorpyrifos (0, 3 or 6 mg/kg bw/day on gestation day 15–18 by oral largely based upon transient neurotoxic signs.
gavage and 0, 1 and 3 mg/kg/day on PND 11–14 by subcutaneous ex- The dose levels chosen for the Uterotrophic Assay (5, 10 and 20 mg/
posure). Neither the Joshi et al. (2007) nor De Angelis et al. (2009) kg/day; OCSPP 890.1600) and for the Male and Female Pubertal Assays
studies included concurrent measurements of AChE activity in various and the Hershberger Assays (10 and 20 mg/kg/day; OCSPP 890.1400)
tissues, although both studies were clearly within dose ranges where with cyfluthrin are approximately 10–20 times higher than the point of
significant brain AChE inhibition would be expected (Marty et al., departure dose for chronic exposures (2.4 mg/kg/day, Table 3), which
2012). is based on acute neurotoxic signs in the one-year dog study. Thus,
Chlorpyrifos was included on the US EPA's Priority List 1 for en- these data indicate that the nervous system is more sensitive to per-
docrine screening using the eleven assays in the EDSP Tier 1 battery. turbation by pyrethroids than the endocrine system; and protection
Dose ranges used for in vitro assays were based on test guideline re- against neurotoxicity would therefore protect against any endocrine-
quirements, whereas in vivo assays were conducted at dose levels related effects due to systemic toxicity, as the EPA concluded in the List
causing significant AChE inhibition. The US EPA (US EPA, 2017b) 1 EDSP WoE assessments. Therefore, it may be reasonable to evaluate
concluded that cholinesterase inhibition was relevant for chlorpyrifos. other scientifically relevant information to determine if the existing
The conclusion of the subsequent WoE analysis, which included Tier 1 data and points of departure are protective for potential endocrine ef-
assay results, regulatory guideline studies and published literature, was fects prior to initiating any additional screening studies on new com-
that chlorpyrifos had no interactions with the estrogen, androgen or pounds.
thyroid pathways at doses below the dose levels that cause significant
brain AChE inhibition (Juberg et al., 2013; US EPA, 2015a). Regulatory
4.3.2. Fish and frogs
exposure limits are sufficient to protect against potential endocrine
Similar effects also can be seen in other species when neurotoxicity
activity as exposure limits are based on red blood cell cholinesterase
is the lead toxic effect. AChE inhibition can cause secondary effects on
inhibition, which is more sensitive (affected more quickly and at lower
endocrine function in wildlife species. In fish, fecundity decreases in
concentrations than brain cholinesterase inhibition).
response to brain AChE inhibition have been reported (Beauvais et al.,
With sufficient brain AChE inhibition, alteration of other endocrine
2001; Beyers and Sikoski, 1994; Zinkl et al., 1987a; b). For example,
pathways may occur, including effects on anterior pituitary hormone
when chlorpyrifos was tested in the FSTRA, concentrations resulting in
secretion and the hypothalamic-pituitary-adrenal (HPA) axis, which
0.251–3.02 μg/L did not affect wet weight, length, gonado-somatic
can be stimulated by central cholinergic signaling (Rhodes et al., 2001).
index, tubercle score, plasma concentrations of Vtg, 17β-estradiol,
Brain cholinesterase inhibition also has been associated with decreased
testosterone, or testicular or ovarian histopathology; however, brain
serum levels of TSH, prolactin (PRL), luteinizing hormone (LH), growth
AChE was affected at all concentrations in female fish (male brain AChE
hormone (GH) and thyroid hormone in rats (Smallridge et al., 1991;
affected at the mid- and high concentrations) and fecundity was re-
Rocksén et al., 2008). Therefore, when assessing potential endocrine
duced (Juberg et al., 2013). Thus, it was concluded that brain AChE
effects from AChE inhibitors, it is useful to document: 1) the endocrine
inhibition was a more sensitive marker of chlorpyrifos exposure in the

148
M.S. Marty et al. Regulatory Toxicology and Pharmacology 99 (2018) 142–158

FSTRA than endocrine-sensitive endpoints. SER can alter the activity of cytochrome P450s that are embedded in it
In the AMA, decreased tadpole length and weight, as well as delayed and responsible for carrying out the final transformations of steroid
development and decreased normalized hind limb length, were ob- biosynthesis leading to vacuolization and potentially necrosis, cell lysis
served at 13.6 μg/L chlorpyrifos (Juberg et al., 2013). These effects or apoptosis (King and Stocco, 1996; Rosol et al., 2001). Therefore, a
were seen in the presence of cholinesterase inhibition in tadpole tissues, visual assessment of mitochondrial and SER ultrastructure can be used
which occurred at concentrations ≥3.68 μg/L chlorpyrifos. Thyroid as a sensitive and specific indicator of mitochondrial and SER function
histopathology was unaffected with chlorpyrifos exposure. Once again, and can be used to differentiate between direct and indirect non-spe-
cholinesterase inhibition was a more sensitive target than endocrine cific cytotoxic effects on steroidogenesis (Ribelin, 1984; Levine et al.,
effects. To characterize the relative sensitivity of these endpoints, 2007).
cholinesterase activity could be included as an endpoint in these assays
when evaluating compounds that operate via this MoA. 5.2. Thyroid

4.4. Altered muscle contractility 5.2.1. Mammals


Serum thyroid hormones undergo endogenous fluctuations and
4.4.1. Mammals there are numerous factors that can alter thyroid hormone levels, in-
Some endpoints that are commonly associated with endocrine cluding decreased growth rate, stress and study-specific parameters. For
function can be mediated by other factors as well. For example, par- example, serum T4 levels can be decreased secondary to effects on body
turition is a time of altered endocrine signaling, resulting in cervical weight gain in growing animals. In the male pubertal assay, doses
ripening and uterine contractility for successful delivery of offspring. causing decreases of approximately 6% or 9% resulted in a 14% or 23%
Conversely, dystocia, a state of difficult/protracted delivery, may be decrease in serum T4 levels (Laws et al., 2007). Thus, a careful WoE
based on alterations in the initiation or progression of parturition. approach and/or additional studies may be necessary to properly in-
While dystocia may be related to altered endocrine signaling, labor is a terpret thyroid endpoints in the presence of decreased growth rate.
complex process with many physiological pathways involved. For ex- Stress also can influence thyroid hormone status. This may be due to
ample, calcium channel blockers (e.g., nifedipine, verapamil, diltiazem) housing (i.e., isolation), generalized stress (e.g., cage transport, hand-
have been used to stop delivery of fetuses in a rat model of preterm ling, nutrition) or stress at study termination related to the method of
labor (i.e., pregnant rats that were ovariectomized on gestation day 16) blood collection, euthanasia and/or anesthesia (Dӧhler et al., 1979).
(Sharma and Gupta, 1994). Similar to the therapeutic role of these Liver effects that result in altered transporter protein synthesis or in-
compounds (i.e., reducing impulse conduction in cardiac muscle or creased hormone catabolism also can affect thyroid endpoints, as dis-
relaxing the smooth muscle in blood vessels), calcium channel blockers cussed in the section on Liver.
decrease uterine contractility. Thus, the origins of dystocia also may Age also can influence thyroid hormone signaling. For example,
include myogenic factors from agents like calcium channel blockers during the change from a light to dark period, serum TSH exhibited a
that alter myometrial contractility by affecting calcium entry short duration, rapid increase in adult male rats, a longer duration and
(Blackburn, 2014). slower increase in pubertal male rats and no change in prepubertal male
rats (Dӧhler et al., 1979; Wong and Dӧhler, 1978). T4 levels followed a
5. In vivo assays: non-specific effects on endocrine organs similar pattern. Pregnancy status also can affect thyroid functioning as
dams must make sufficient T4 to support fetal neurodevelopment for
5.1. Adrenal nearly all of gestation (Perez-Castillo et al., 1985; Grijota-Martinez
et al., 2011). Lastly, the stage of the estrous cycle also can affect thyroid
The adrenal gland is one of the most common target organs and signaling (Dӧhler et al., 1979), which may make these data more dif-
endocrine organs for chemical toxicity (Rosol et al., 2001). Observing ficult to interpret in the female pubertal assay.
non-specific cytotoxic adrenal effects by a hydrophobic chemical at
high dose levels in the adrenal cortex is not uncommon or surprising 5.2.2. Frogs
(Ribelin, 1984; Frith et al., 2000). The adrenal cortex is divided into As stated in the section on Stress, amphibian metamorphosis is not
three layers, the zona glomerulosa that synthesizes mineralocorticoids, solely dependent on thyroid hormone. Instead, there is a second sig-
the zona fasciculata that synthesizes glucocorticoids and the zona re- naling pathway involving hypothalamic corticotropin releasing factor
ticularis that synthesizes glucocorticoids/androgens (Rosol et al., (CRF) that can accelerate metamorphosis in response to changing en-
2001). The adrenal gland is vulnerable to toxicity because of its high vironmental conditions, such as decreased water levels or high popu-
perfusion rates, unique anatomical features of the adrenal blood supply, lation density (Denver, 1997a; b; Okada et al., 2007; Kühn et al., 1998;
and its ability to accumulate hydrophobic chemicals in its lipid-rich Rose, 2005; Coady et al., 2010). Furthermore, developmental/growth
cells (Inomata and Sasano, 2015; Rosol et al., 2001). This accumulation delays can hinder metamorphosis. Given the potential contribution of
can disrupt normal cellular function of adrenal cortical cells leading to multiple factors, it is important to integrate all available information
degeneration, reduction in cell volume, and subsequently atrophy when determining whether effects on metamorphosis are specific to
(Ribelin, 1984; Rosol et al., 2001). The zona fasciculata and zona re- altered thyroid function. Thyroid histopathology provides a more spe-
ticularis are the most frequently affected adrenal cortical zones by cific endpoint to assess thyroid involvement.
toxicants (Ribelin, 1984, 2010; Suttie and Sutcliffe, 2018). Effects to the
zona fasciculata/reticularis are characterized as degeneration and/or 6. Stressors
reduction in cell volume and vacuolization (Rosol et al., 2001). In the
zona fasciculata/reticularis, degeneration leading to atrophy is gen- Stress has been known to impact steroidogenesis in both mammals
erally initiated with cytotoxic effects to the mitochondria and the and fish (e.g., DePeyster et al., 2014; Marić et al., 1996; Almeida et al.,
smooth endoplasmic reticulum (SER) and is followed by vacuolization 1998). Thus, stress responses should be considered when evaluating the
of neutral lipids and cholesterol. As previously discussed, the initial and potential endocrine activity of test substances, particularly when toxi-
rate limiting steps of steroidogenesis take place in the mitochondria and city tests are conducted at MTD/MTC levels. Sometimes stress re-
subsequent steps in the steroidogenic pathway are performed by cyto- sponses, which may include increases in adrenal weights and histo-
chrome P450s anchored in the SER (Stocco, 2001; Rosol et al., 2001). pathology, may be difficult to discern from broad spectrum
Disruption of the mitochondrial electrochemical gradient by cytotoxi- steroidogenesis inhibitors (e.g., ketoconazole), which can alter these
city blocks the rate limiting steps in steroidogenesis and stress to the same endpoints.

149
M.S. Marty et al. Regulatory Toxicology and Pharmacology 99 (2018) 142–158

6.1. Generalized stress response corticosterone to return to baseline is considerably slower in pre-
pubescent rats, requiring twice as long as adults. Pubertal rats transition
6.1.1. Mammals rapidly to adult responsiveness between 50 and 60 days of age (Klein
Excellent review papers on stress responses and their interpretation and Romeo, 2013). Thus, changes in stress-responsiveness overlap with
in toxicology studies has been published (e.g., Everds et al., 2013; changes in the production and secretion of sex steroid from the hy-
Witorsch, 2016). In Everds et al. (2013), the term “stress” is used to pothalamic-pituitary-gonadal (HPG) axis. While testosterone generally
encompass physiological responses to perturbations of homeostasis. reduces stress responsiveness in adult males (i.e., male rats castrated
Stress commonly affects functioning of the immune, reproductive and/ after puberty show slightly higher corticosterone responses to stress)
or endocrine systems. Common effects that can occur secondary to (Handa et al., 1994; McCormick et al., 2002; Viau and Meaney, 1996),
stress involve multiple systems and include changes in serum hormone prepubertal male rats continued to show a protracted stress response
levels (e.g., increased corticosterone and progesterone, LH suppression, even when given adult testosterone levels (Romeo et al., 2004a). In
decreased testosterone and androstenedione), decreases in body prepubescent male rats, stress can decrease levels of testosterone even
weight/gain, food consumption, activity, estrous cyclicity, organ at the stage when only low baseline levels of testosterone are present
weights (thymus, spleen and reproductive organs, including weights of (Foilb et al., 2011). In adult females, estradiol levels increase stress
the epididymides, seminal vesicles, prostate, ovaries and uterus), lym- responsiveness; however, regardless of estrogen status, prepubertal fe-
phocyte depletion in the thymus and spleen, leukocyte counts and al- male rats showed enhanced ACTH/corticosterone responses to stress
tered reproductive functions (Witorsch, 2016; Everds et al., 2013). (Klein and Romeo, 2013). Thus, while testosterone and estrogen can
Adrenal weights are generally increased and may be accompanied by influence adult programmed stress response, these hormones do not
hypertrophy/hyperplasia in the cortex. Histopathologically, decreased mediate enhanced prepubertal stress responses, which appear to be age
thymic and splenic cellularity may be seen, as well as lipid depletion of dependent.
the zona fasciculata of the adrenal gland and atrophy or inactivity in These examples illustrate that stress responses are complex. The
the epididymides, prostate, seminal vesicles, ovary, uterus and vagina. endpoints that are altered and the magnitude of the response will de-
Testicular effects may include disorganized seminiferous tubular epi- pend on the type, duration and magnitude of the stress, as well as
thelium and enlargement of the interstitial spaces (Everds et al., 2013; species, strain, gender, age, physiological state and individual animal
DePeyster and Mihaich, 2014; Pellegrini et al., 1998). variability. Thus, discerning specific endocrine-mediated effects from
Everds et al. (2013) reported that stress responses should not be secondary, stress-induced changes in endocrine-sensitive endpoints can
interpreted as primary endocrine-mediated effects, but rather as non- be challenging.
endocrine-mediated effects of toxicant exposure. Stress responses re-
flect activation of the sympathetic nervous system or the hypothalamic- 6.1.2. Fish
pituitary-adrenal axis in an effort to restore normal physiological con- Stress responses are important to understanding the specificity of
ditions. Rats also can reflexively induce physiological adaptations that any observed reduced Vtg response. At the most basic level, stress can
include decreased metabolic demand (e.g., decreased cardiac output, lead to a trade-off between energy allocation for survival (e.g. detxifi-
blood pressure/placental perfusion, acid-base status, hemoglobin con- cation processes) and non-essential activities (i.e., reproduction and
centrations and oxygen saturation) and hypothermia. The presence of somatic growth) (i.e., the energy allocation principle) (Schreck et al.,
stress responses in toxicological studies is not surprising and may ap- 2001). Indeed, non-specific decreases in Vtg have been observed in the
pear as toxicant-induced adverse effects. For example, decreased feto- validation studies for presumed non-endocrine active materials for the
placental oxygenation, secondary to a maternal stress response, was FSTRA (US EPA, 2007). The test concentration setting guidance for the
considered to be a likely cause of decreased fetal and placental growth FSTRA (US EPA, 2009c) requires that the high treatment should be
and developmental delays in a cyfluthrin inhalation developmental approximately one third of the fish acute 96-h LC50; therefore, it is quite
toxicity study (Pauluhn, 2018; OECD 414). In this study, cyfluthrin likely systemic stress will be encountered (Wheeler et al., 2013). Thus,
exposures were above the chemosensory threshold and triggered a decreases in Vtg from systemic stress may be common, at least, in the
stress response in pregnant dams. Thus, a WoE approach is needed to high treatment levels of regulatory studies.
differentiate stress-mediated effects from direct toxicity. The evaluation Another example is the impact of stress on the hypothalamus-pi-
of clinical pathology parameters, body temperature, adrenal function tuitary-inter-renal axis (HPI) of fish (essentially equivalent to HPA in
and/or immunotoxicity may be needed to determine the contribution of mammals). Stress can lead to increased synthesis of cortisol, and cor-
stress to the results of toxicity studies. tisol can act as an inhibitory hormone of both male and female fish
As noted in Everds et al. (2013), the response to stress can be reproductive physiology over the whole reproductive cycle (Milla et al.,
gender- and age-dependent, a point that is especially relevant for the 2010). Cortisol can lower sex steroid levels, leading to decreased go-
male and female pubertal assays. In adults, chronic stress has been nado-somatic index (GSI), decreased Vtg levels and decreased sec-
shown to decrease circulating gonadal hormones and may reduce fer- ondary sex characteristics (tubercle score) (Foo and Lam, 1993;
tility (Cameron, 1997; Kalantaridou et al., 2004). However, in some Carragher and Sumpter, 1990; Aluru and Vijayan, 2009; Milla et al.,
cases, repeated stress may result in a decreased stress response due to 2010; Pankhurst and Van der Kraak, 2000; Haddy and Pankhurst, 1999;
negative feedback by glucocorticoids. Stress-related parameters can Wu et al., 2003; Lethimonier et al., 2000). These results have been
return to baseline levels (habituation) even in the continued presence of confirmed using in vivo studies of fish exposed to confinement stress
stressors. Female rats exhibit a greater peak corticosterone response to (Pankhurst and Van der Kraak, 2000) or implanted with cortisol (Foo
stress than males, but females return to baseline more quickly (Romeo and Lam, 1993) leading to reduced sex hormone levels.
et al., 2004a; b). In addition, stress mediated through the glucocorticoid pathway can
Age differences include increased responsiveness of the HPA axis to delay fish gonadal development and lower gamete quality (Aluru and
stress during puberty and adolescence which has been demonstrated in Vijayan, 2009). The effects of cortisol on sex steroids (Schreck et al.,
several studies (reviewed in Klein and Romeo, 2013). This includes a 2001) can potentially impact gonadal histopathological evaluations.
greater release of adrenocorticotropic hormone (ACTH) from the For instance oocyte size, atresia and reabsorption have been shown to
anterior pituitary in prepubescent rats (Klein and Romeo, 2013). Be- respond to actual or simulated stress (cortisol treatment) (Schreck et al.,
tween 30 and 40 days of age, prepubertal male and female rats begin 2001; Clearwater and Pankhurst, 1997).
showing a protracted hormone response to acute and heterotypic Therefore, it is clear that stress can influence the very measures or
stresses (Foilb et al., 2011; Lui et al., 2012), releasing more corticos- endpoints included in fish endocrine screening assays to detect endo-
terone in response to stress than adults. In addition, the rate for crine activity. It is, therefore, very important to distinguish responses in

150
M.S. Marty et al. Regulatory Toxicology and Pharmacology 99 (2018) 142–158

these parameters from general stress and specific endocrine interac- et al., 2014). There is evidence indicating that obesity in girls can ac-
tions. Further regulatory guidance is required to develop this assess- celerate pubertal signs and alter progression, possibly due to altered
ment and interpretation of effects. For example, there is limited gui- hormone levels; however, the evidence in boys is inconsistent
dance for the reporting of clinical signs in ecotoxicology assays and it is (Marcovecchio and Chiarelli, 2013; Soliman et al., 2014). Under-nu-
likely there is great variation amongst performing laboratories. This trition or a restricted nutrition has been shown to impair growth that
will undoubtedly limit evaluators' ability to contextualize endocrine can lead to a delay and/or prolongation of puberty (Plant, 1994). In
and stress responses in cases where anything less than overt toxicity is humans, it has been demonstrated that food availability/consumption,
observed. the balance between energy intake and expenditure, is a major en-
vironmental factor modulating the timing of sexual maturation
6.1.3. Frogs (Villamor and Jansen, 2016; Ellis, 2004; Soliman et al., 2014). The
Generalized stress, for example the presence of predators (Laurila tremendous metabolic demand placed by intense athletic training like
et al., 1998), can delay growth in tadpoles, affecting weight and length long distance running, gymnastics, and ballet are often associated with
measurements in the amphibian metamorphosis assay (Wilbur and delayed pubertal development or interrupted menstrual cycles
Collins, 1973; OECD, 2007). Delays in growth also can result in delays (Georgopoulos et al., 2010). These effects are normally reversible
in the progression of metamorphosis (Wilbur and Collins, 1973; OECD, within months after return to “normal”. In toto, experimental data
2007). Chemical stressors can result in delayed development including suggests that effects on food intake (thus on body weight gain) are
reductions in wet weight, snout-vent length and hind limb length in the expected to have a major impact on sexual maturation.
absence of specific thyroid activity. In an evaluation of amphibian Rats show similar effects on puberty onset as humans in response to
metamorphosis test results by the US EPA, myclobutanil resulted in over- and under-nutrition. Cumulative dietary intake has been shown to
significant decreases in all of these morphological parameters at test determine the onset of puberty in female rats following guideline pro-
levels in which amphibians exhibited transient abnormal behaviors tocols for pubertal assays whereby higher metabolizable energy intake
(floating on the surface, lying on the bottom of the tank, inverted or was associated with earlier puberty onset. In fact, energy intake of 2300
irregular swimming, lack of surfacing activity, and being nonresponsive kJ/rat beginning at weaning was identified as the trigger for puberty
to stimulus) but no increase in mortality. There was also no notable onset as measured by vaginal opening (Odum et al., 2004). In addition,
thyroid pathology. In the same review, isophorone also caused de- Odum et al. (2004) demonstrated that guideline uterotrophic assays
creases in wet weight and snout-vent length in the absence of delayed (i.e., uterine weight) using immature rats also were affected by cumu-
developmental stage or treatment-related thyroid histopathological lative dietary intake, because uterine weights are more closely related
findings (US EPA, 2009b). No signs of generalized toxicity were noted, to body weight in weanling rats than adult ovariectomized rats (OECD,
so it is important to consider that either the generalized toxicity was of 2003).
a subclinical level or these endpoints are susceptible to spurious out- In contrast, decreased growth rates have been associated with de-
comes, both scenarios potentially confounding interpretation. In the layed puberty onset in both male and female rats (e.g., Engelbregt et al.,
cases of the chemical stressors, it becomes critical to correlate mor- 2000; Ashby and Lefevre, 2000; Stump et al., 2014). Several peer-re-
phological findings with thyroid histopathogy (stage-matched samples) viewed publications report studies that have been conducted utilizing
in order to maximize an accurate interpretation of potential thyroid feed restriction in rats, resulting in varying degrees of decreased weight
effects. gain, to determine the influence of body weight reduction on puberty
While stress generally delays responses in mammals, stress also can onset. In most toxicity studies, depressed body weight gain is a common
advance developmental stage in amphibians, since increased stress can sign of systemic toxicity. Thus, effects observed after chemical treat-
cause premature metamorphosis via increased CRF from the hypotha- ment and unrelated to the endocrine system can confound the inter-
lamus (Denver, 1997a; b; Okada et al., 2007; Kühn et al., 1998; Rose, pretation of results of endpoints evaluated in standard reproductive
2005; Coady et al., 2010). An acceleration of metamorphosis was ob- toxicology studies.
served as a result of water-level manipulation in Rana temporaria The effects of decreased growth rate on puberty onset and other
(Loman, 1999; Laurila and Kujasalo, 1999) independent of tempera- endocrine-related endpoints has been examined for decades; however,
ture. The proposed mechanism involves increased CRF from the hy- newer studies have examined the effects of decreased growth rates
pothalamus that reduces appetite and foraging behavior and stimulates using similar study designs and body weight decreases of relevant
the hormonal system responsible for metamorphosis (Denver, 1997a; b; magnitude for regulatory toxicity studies (e.g., OECD 416, OCSPP
Okada et al., 2007; Kühn et al., 1998; Rose, 2005; Coady et al., 2010). 890.1450, 890.1500; Carney et al., 2004; Chapin et al., 1993; Laws
This phenomenon was also observed for other species (Hyla pseudo- et al., 2000, 2007; Marty et al., 2003; Stoker et al., 2000a). Feed-re-
puma, Crump, 1989; Scaphiopus couchii, Newman, 1988, 1989; Sca- striction studies using study designs similar to the Tier 1 male and fe-
phipus multiplicatus, Pfennig, 1990; Bufo calamita, Tejedo and Reques, male pubertal assays were summarized by Stump et al. (2014). For the
1994; Scaphiopus hammondi, Denver and Denver, 1995). Development female pubertal assay, there was no apparent effect of feed restriction
rate can also be increased by competition density (Wilbur, 1976, 1977; on age at puberty onset with terminal body weight effects ranging from
Dash and Hota, 1980; Morin, 1986), probably due to reduced food re- 2.1 to 18.9% less than ad libitum-fed controls on PND 42. The greatest
sources. Of course, what this represents is a very plastic approach to difference was a 1.6-day delay in age at vaginal opening with an 8.6%
metamorphosis that may be affected by different stressors. This makes change in terminal body weight; this level of feed restriction also af-
it critical that changes in the endpoint are interpreted using all avail- fected the number of 4–5 day estrous cycles and pituitary weight. Pi-
able information and in the context of the exposure conditions and tuitary, adrenal and ovarian weights were affected at higher feed re-
general toxic response that may result from the chemical exposure. striction levels. For the male pubertal assay, Ashby and Lefevre (2000)
demonstrated a relationship between initial body weight on PND 22
6.2. Nutrition/energy status and altered rate of growth and age/body weight at preputial separation and also concluded that
there appears to be a continuum between body weight and age at
6.2.1. Mammals preputial separation in pubertal male rats. Whereas one study reported
Numerous factors can influence the onset of puberty in mammalian no effects on preputial separation with feed restriction resulting in
species, including humans. Nutritional status in childhood (postnatal terminal body weights ranging from 1.8 to 19.2% less than ad libitum-
period) can have significant effects on pubertal development, such as fed controls (Laws et al., 2007), two other studies reported a 1.8-day
timing of the onset of puberty. Both over-nutrition and under-nutrition and 2.1-day delay in age at preputial separation with feed restriction
have been associated with altered puberty onset in humans (Soliman resulting in an 11.3% and 15.0% decrease in terminal body weight,

151
M.S. Marty et al. Regulatory Toxicology and Pharmacology 99 (2018) 142–158

respectively (Marty et al., 2003; Stoker et al., 2000b). Other endpoints acquisition and/or increasing energy demand, in response to a toxicant,
were more sensitive to growth rate changes than age at preputial se- will result in a decreased ability to maintain non-essential processes
paration as pituitary weights were affected when terminal body weights such as expression of vitellogenin”. Thus, effects on the well-being/
were decreased by ≥ 5.9% and seminal vesicle, ventral prostate and energy status of fish can confound endocrine endpoints too. As in-
epididymal weights affected with a ≥11.3% decrease in terminal body dicated above, assessing such effects in fish can be problematic due to
weights (Stump et al., 2014). Testes weights were not altered. These an inability to accurately measure food consumption and assess growth
results are similar to the study by Rehm et al. (2008), where Sprague- parameters in screening studies such as the FSTRA. This further un-
Dawley rats were feed restricted by 20% from six to eight weeks of age derlines the importance of using all available data where other tests not
(14.6% lower terminal body weight than ad libitum-fed control rats). specifically designed to assess endocrine effects may provide better
These animals exhibited decreased absolute epididymal and relative information on the general health status and growth effects of the or-
seminal vesicle weights, a 72% decrease in plasma testosterone and ganism under similar exposure levels (e.g. the fish early life stage test).
‘minimal’ grade pachytene spermatocyte degeneration in Stage VII of
the spermatogenic cycle. 7. Other factors (non-test article variables)
The potential effect of growth rate/body weight on pubertal assay
endpoints has been recognized in the male and female pubertal assay When conducting toxicological studies in general, or the EDSP
test guidelines (OCSPP 890.1450, 890.1500; US EPA, 2009a, b), where screening assays in particular, some aspects of study design, animal
a difference in terminal body weight of > 10% is considered to exceed welfare/husbandry, etc., can affect study outcome. Some factors to
the MTD (although the male pubertal test guideline also states that a consider include:
6% decrease in body weight gain at termination can complicate assay
interpretation). Therefore, careful interpretation of age at puberty 7.1. Dose Administration
onset, estrous cyclicity, and endocrine-sensitive tissue weights is
needed when body weight gain is decreased relative to control animals. Oral gavage (bolus) dosing of a test material at MTD dose levels is a
Maternal under-nutrition during pregnancy and/or lactation has significant source of stress in rodent toxicity studies. While daily dose
been associated with reduced primordial, secondary and antral follicle administration via gavage of animals is a common and accepted method
numbers in rat offspring (Bernal et al., 2010); notably, the timing and in toxicology studies, it still causes stress even if done properly (Brown
magnitude at which under-nutrition occurs impacts outcome. In addi- et al., 2000; Gonzales et al., 2014). Oral gavage can be difficult de-
tion, maternal toxicity/under-nutrition in lactating rats has been shown pending upon the experience and skill of the technician, and there is a
to result in an increased period of lactational diestrous when nursing potential for increased morbidity or stress due to repeated incorrect
their litters (Woodside, 1991). In these rats, ovarian pathology, follicle dose administrations (Atcha et al., 2010; Gonzales et al., 2014). In-
numbers and corpora lutea of dosed animals experiencing toxicity may correct intubations in which the gavage tube enters the trachea instead
appear different from those of control animals as they have not returned of the esophagus or causes reflux due to rapid dose administration can
to normal cycling. lead to significant stress, abnormal clinical observations, or death.
Alterations in nutritional status also may affect steroidogenesis. For Gavage errors are not always distinguished from chemically caused
example, hypercholesterolemia has been associated with decreased deaths in study reports. Proper scruffing of the animal is also important
testosterone levels in humans, rats and mice (Tanaka et al., 2001; to ensure it cannot move its head during gavage or alternatively be-
Martínez-Martos et al., 2011). Cholesterol serves as a precursor for come distressed from being too tightly scruffed (Atcha et al., 2010).
steroid hormone synthesis. Diet-induced hypercholesterolemia in male Vehicle-treated animals are also gavaged to control for stress, but some
mice resulted in decreased circulating testosterone levels, hypotheti- test compounds have a very pungent taste and/or odor and could irri-
cally through inhibition of testosterone synthesis by the renin-angio- tate the stomach or produce damage due to gavage-related reflux.
tensin system in the testis (Martínez-Martos et al., 2011; Khanum and Therefore, one can imagine an experimental animal gavaged with some
Dufau, 1988). The renin-angiotensin system also may play a role in test materials, regardless of the dose, might find that experience more
progesterone synthesis in the rat ovary (De la Chica et al., 2007). stressful than being gavaged only with vehicle (e.g., DePeyster and
Thus, in instances where systemic toxicity affects body weight/rate Mihaich, 2014).
of growth, a WoE analysis of endpoints within and across studies may Damsch et al. (2011) reviewed the limited literature database on
be needed to determine the specificity of effects on the endocrine gavage-related reflux in regards to risk factors, interpretation of histo-
system. logical changes, and toxicological relevance. Gavage-related reflux can
occur and cause aspiration of irritant material, resulting in respiratory
6.2.2. Fish effects as well as irritation in the larynx, pharynx, sinuses, middle ear,
Similar to mammals, effects on body weight/size can affect endo- and nose (Damsch et al., 2011). Many factors can cause gavage-related
crine-sensitive endpoints in fish. For example, male fat pad weight, and reflux including feeding status, high volume administered (> 10 ml/
in both sexes gonad and liver weights were found to be proportional to kg/day), increased gastric acid production, delayed gastric emptying,
body weight (Watanabe et al., 2007). Thus, if there are systemic effects and decreased muscular tone or dysfunction of the esophagus. In ad-
on body weight (reduced feeding, etc.) then other tissue weights would dition, high viscosity formulations, or those with irritant properties can
be impacted also, affecting study metrics such as Gonadal Somatic cause technical issues as discussed above due to increased reactions of
Index (GSI). Tubercle score also has been correlated with fish size the animals. Gonzales et al. (2014) evaluated plasma corticosterone
(Watanabe et al., 2007). Hutchinson et al. (2009) reported that de- concentrations after oral administration of a drug formulation using a
creases in the expression of male secondary sex characteristics may not peanut butter pellet compared to oral gavage in mice. As expected, the
be endocrine specific. Therefore, changes in GSI or gonad stage/histo- plasma levels of corticosterone in animals which were treated with drug
pathology should not be regarded as solely or specifically indicative of by oral gavage were significantly increased above those given the drug
exposure to an endocrine disruptor in the FSTRA. by pellet, or animals not treated by any method. Furthermore, the an-
Energy status in the presence of systemic toxicity also impacts en- imals treated for 2 days by oral gavage had corticosterone levels as high
docrine endpoints in environmental species. As stated in Wheeler et al. as those animals which were restrained (and not administered drug).
(2013): “The response of these (endocrine) endpoints to systemic
toxicity is not surprising, since the amount of energy that an individual 7.2. Terminal estrous stage and female reproductive endpoints
can invest in the processes supporting maintenance, growth and re-
production is limited (Sibly and Calow, 1986). Reductions in energy The stage of terminal estrous influences hormone levels and

152
M.S. Marty et al. Regulatory Toxicology and Pharmacology 99 (2018) 142–158

reproductive organ weights at necropsy in cycling female animals (e.g., however, genetic factors as well as nutritional factors (i.e. deficiencies
the uterus weighs more during proestrus than diestrus). In most assays, in the amino acid, tryptophan; phospholipid deficiencies) have been
there is no requirement to necropsy animals at the same stage of es- discussed as potential causes for bent tail in tadpole and/or fish larvae
trous; therefore, stage of terminal estrous is an uncontrolled variable. (Droin, 1992; Cahu et al., 2003). Since length measurements are made
Females, which were undergoing normal estrous cycles, could be in from snout-to-vent in the AMA, the presence of bent tails is not ex-
proestrus, estrus, metestrus or diestrus at the time of necropsy. Thus, it pected to hamper data collection in this particular assay, especially if
is important to evaluate terminal estrous stage across treatment groups the incidences of bent tail are relatively minor and do not translate into
to provide context for organ weight and hormone data. In studies where spinal deformities in subsequent metamorphs.
(anti)estrogenicity or steroidogenesis inhibition is suggested, a more
thorough evaluation of estrous cycle length and estrous cycle pattern 8. Discussion/conclusion
may be useful. As stated in the pubertal assay Standard Evaluation
Procedures document (US EPA, 2011), regularity of the estrous cycle While not exhaustive, this paper provides some examples of how
should be given more weight in the evaluation of potential endocrine specific target organ toxicity, systemic toxicity and/or altered home-
effects, whereas ovarian and uterine weights should be interpreted with ostasis can affect endocrine-sensitive endpoints by non-endocrine
caution due to normal variability in cycling animals. Pathologists MoAs. This illustrates some of the challenges of distinguishing endo-
should determine whether uterine and ovarian histopathology appears crine from non-endocrine MoAs, which can have critical impacts on
normal for various stages of the estrous cycle. It is important to note regulatory decisions. To support accurate determination of the endo-
that aside from estrogenicity/antiestrogenicity and altered ster- crine-disrupting potential of substances under evaluation, it is im-
oidogenesis, stress also can alter the regularity of estrous cycles; portant to consider several factors:
therefore, a WoE is needed to interpret differences in female estrous
cyclicity and reproductive organ weights relative to other endpoints 1) The endocrine system maintains various aspects of dynamic
and other available data. homeostasis; therefore, environmental conditions, animal
well-being, and perturbations of other organ systems affect
7.3. Testicular histopathology in juvenile dogs endocrine function. These effects can be observed across species
and can be related to lifestyle factors (e.g., high intensity exercise,
Rehm (2000) and Goedken et al. (2008) evaluated spontaneous nutritional status) or environment (e.g., accelerated development in
testicular and epididymal histopathological findings in control beagle frogs due to habitat constriction or increased population density).
dogs. These researchers reported that young dogs (e.g., < 8 months) Alterations in endocrine system function may not necessarily in-
routinely have numerous testicular observations, including lower tes- dicate an adverse effect, but may instead indicate an adaptive re-
ticular weights, hypospermatogenesis, atrophy/hypoplasia in semi- sponse to different perturbations. Because the endocrine system has
niferous tubules and incomplete filling of epididymal tails with sperm. a primary role in maintaining normal physiology to manage stress,
These findings, which can mimic those seen in studies with testicular the apparent endocrine-disrupting effects that may be observed in
toxicants, were attributed to sexual immaturity. Thus, male beagles toxicity studies may be difficult to discern from effects triggered by
should not be necropsied at less than ten months or older if sperma- alterations in another target organ.
togenesis evaluations are needed. Some observations (e.g., atrophy/ 2) The lead toxic effect drives the hazard profile and can be pro-
hypoplasia of seminiferous tubules) were elevated in dogs under 12 tective of endocrine-related effects using established risk as-
months of age. sessment procedures. The lead toxic effect approach (Bars et al.,
2012) involves considering all available toxicity data for a substance
7.4. Parasites and identifying the adverse effect that occurs at the lowest dose.
This adverse effect is considered the lead toxic effect and describes
7.4.1. Fish the most sensitive toxicological response and drives the hazard
Parasitic infections (microsporidia, mycobacteria, tapeworm) in the profile of a substance. Any risk management measures based on the
FSTRA can affect gonadal histopathology and inhibit reproduction lead toxic effect is likely to be protective of other toxic effects (in-
(Coady et al., 2014). Histopathological changes in fish ovaries caused cluding endocrine effects) occurring at higher treatment levels.
by parasites commonly found in aquarium fish such as Pleistophora Therefore, endocrine disruption is only of high regulatory concern
infestation in fathead minnows, Pimephales promelas (Rafinesque), have for a substance if it is the lead toxic effect.
been reported (Ruehl-Fehlert et al., 2005). Microsporidia also can cause 3) Dose/concentration selection is important for lower and higher
the degeneration of ovarian tissue and the appearance of testicular tier screening/testing. Treatment level (dose or concentration)
tissue in infected fish (Wiklund et al., 1996). In addition, the tapeworm, selection takes on enormous importance in studies screening or
Ligula intestinalis, has been shown to inhibit reproduction by interfering testing for potential endocrine activity/disruption. These assays are
with the pituitary–gonadal axis of its fish host (Arme, 1997; Williams not simply testing for an adverse effect but rather an adverse effect
et al., 1998). through an endocrine mechanism. The hypotheses that are being
tested in screening assays are whether there is the potential for
7.4.2. Frogs endocrine modulation through a specific endocrine mechanism
Bent tails (i.e. scoliosis) have been commonly observed in laboratory (e.g., estrogen, androgen, thyroid or steroidogenic pathway). The
populations of African-clawed frog tadpoles (Xenopus laevis); an in- purpose of higher tier testing is to further examine a potential en-
cidence of ∼20% bent tail across both control and treated tadpoles was docrine effect observed in screening assays and then put the findings
not an uncommon observation in some tadpole cohorts during testing in the context of a human or ecological risk assessment (or definitive
efforts for the AMA (OCSPP 890.1100; Coady et al., 2014). Since, in this hazard assessment for endocrine disrupting properties for EU hazard
case, bent tails among X. laevis tadpoles were reported to occur at the based cut-off). Therefore, results of endocrine screens and tests
same rate in control and test material-exposed populations, the cause of should include a careful assessment of systemic toxicity to not
the bent tails was not attributable to specific chemical exposures confound the interpretation of potential impacts on endocrine me-
(Coady et al., 2014). However, it should be noted that effects on tadpole chanisms. As discussed, the use of the MTD and MTC approaches
axial skeleton development has been noted in response to some che- means that this evaluation and separation of endocrine-mediated
mical stressors (Dumpert and Zietz, 1984; Bacchetta et al., 2008). The from non-endocrine-mediated effects is extremely important in
cause of bent tail amongst X. laevis control populations is not known; regulatory application.

153
M.S. Marty et al. Regulatory Toxicology and Pharmacology 99 (2018) 142–158

In mammalian screening assays, the MTD approach has been of endocrine effects (e.g., Vtg in male fish as an indicator of estro-
adopted to set the highest dose level. The MTD is not a biological genicity, tubercles in female fish as an indicator of androgenicity).
constant, therefore, the MTD can vary between range-finding assays Borgert et al. (2014) designated these endpoints as Rank 1 and ap-
and definitive assays. For example, a response to a given dose level plied greater weight to these effects in the WoE process. Conversely,
in the range-finding assay may only cause a 5% bodyweight loss but less specific endpoints (e.g., estrous cyclicity in female pubertal rats,
in the definitive study, the effect could result in a 10% loss. fecundity in fish) were assigned Rank 3 and were used to provide
Consequently, there is a likelihood that the MTD based on range- supporting data for a potential endocrine MoA; Rank 3 endpoints
finding assays can result in systemic toxicity and confound study alone were considered ‘insufficient’ evidence of endocrine activity.
interpretation due to toxicity to non-endocrine targets and/or acti- Rank 2 endpoints are intermediate in their specificity and support
vation of the hypothalamic-pituitary-adrenal axis, which in turn for an endocrine MoA. In this way, relevance weighting can be
affects all endocrine systems (US EPA, 2013). considered in a WoE assessment.
For aquatic testing, the approach is termed the MTC which is cal- Across the EDSP Tier 1 assays, there is sufficient redundancy (i.e.,
culated as a function of median lethal concentrations. The current endpoints sensitive to estrogen, androgen or thyroid perturbations)
MTC guidance in the test guideline is to set the highest tested con- to allow for the detection of endocrine activity in multiple assays
centration for the 21-day AMA and 21-day FSTRA at 1/3rd of the (US EPA, 2013); however, this pattern of effects may be complicated
96-h LC50 value. This approach may be acceptable for a small group by route of exposure, treatment level and species differences. For
of compounds that exhibit very steep concentration-effect relation- example, a compound that shows ER binding and transactivation
ships. However, for classes of compounds that do not have steep may be negative in a uterotrophic assay via the oral route, but may
concentration-effect relationships, this approach will likely produce be detected in the Fish Short Term Reproduction test. Furthermore,
overt toxicity in a 21-day continuous exposure. Therefore, flexibility compounds may be detected as bioactive in in vitro assays at doses
in the concentration setting approach is required and concentration that are either not achieved systemically in animal models or that
setting needs to be handled on a substance-by-substance basis. One result in marked systemic toxicity. In these cases, in vitro-to-in vivo
recommendation is to perform 14–21-day range-finding assays that pharmacokinetic models may provide some context for these dose
include sensitive indicators of sub-lethal toxicity and provide a levels. Lastly, differences in mammalian and non-mammalian re-
predictive assessment of systemic toxicity. Examples of sub-lethal sponses may suggest a species difference in response or sensitivity to
endpoints that could be included in these extended range-finding the endocrine activity being tested (Ankley and Gray, 2013).
assays include gill, liver, and kidney structure and/or function,
presence of edema, as well as other indicators of systemic toxicity. 5) To distinguish direct from non-endocrine-mediated effects on
A toxicokinetic (TK) approach could also be employed to set doses endocrine-sensitive endpoints, there may be additional end-
for endocrine screening and testing. The goal of this approach is to points or other data that can be useful to better characterize
identify the highest dose level that does not exceed linear pharma- stress and systemic toxicity. These suggested endpoints should be
cokinetics in plasma. The use of excessively high doses or con- considered in light of the particular substance being tested and its
centrations can create TK processes that result in a systemic ex- known toxicity.In vitro
posure to either parent compound or metabolites that is not dose
proportional (i.e., no longer increases with increasing dose, or a • Consider whether the class of chemistry is amenable to in vitro
systemic exposure that hugely increases with a small increase in screening or whether false positive results are possible (e.g., sur-
dose). If there is a sufficient margin of exposure, exceedingly high factants, denaturing agents, etc.).
doses and associated non-linear TK will likely produce results that • For cell systems, consider whether effects are specific or potentially
are not relevant for hazard or risk assessments (Saghir et al., 2012). non-specific. Include assays to characterize cell cytotoxicity and
Therefore, the driver for adoption of the TK approach for dose set- stress, particularly for “loss of signal assays” where a decrease in
ting in mammalian toxicology studies has been to improve testing signal indicates potential endocrine activity. Note that more than
and assessment methodologies and not confound the interpretation one assay may be needed to adequately characterize cell status.In
for human relevance (Boobis et al., 2008). Creton et al. (2012) vivo
discuss practical examples that underscore the role TK can play in
improving study design and interpretation through selection of ap- • Clinical signs and frequent body weight measurements can provide
propriate high doses (e.g., use of OECD 421 to establish kinetically useful information on overall well-being of test animals. However in
derived maximum dose levels for an OECD 443; Saghir et al., 2013; fish and frogs, descriptors and evaluations are not well standardized
Marty et al., 2013). Application of TK approaches for dose setting in and there is variability across laboratories as to what is recorded and
ecotoxicology is rare. One of the barriers to the application of TK in how it is interpreted. A better understanding of body weight and
ecotoxicology would be development of improved sampling length changes may be useful in fish and frog endocrine screens, but
methods and analytical capabilities, but this would be a highly often these data are difficult to collect in a meaningful manner (see
productive area of research for the aquatic and wildlife toxicology Wheeler et al., 2014) and interpret in relation to screening assay
community in general. outcomes; further work to standardize assessments is encouraged.
Clinical signs in rodents and ecological species may be particularly
4) In an effort to distinguish endocrine-mediated from non-endo- important for neuroactive compounds or systemic toxicity. Food
crine-mediated effects on the endocrine system, it is useful to and/or water consumption also provide useful information.
adopt a WoE approach when evaluating endocrine data. In this • Characterize the toxicity of known target organs or biochemical
paper, there are numerous examples that different types of systemic markers in eco/toxicology studies designed to examine potential
toxicity and/or stress can affect endocrine-sensitive endpoints. endocrine effects. As indicated above, significant target organ toxi-
Ideally, eco/toxicological data across studies should be examined in city can affect endocrine endpoints. Target organ evaluation should
a systematic manner for evidence of potential endocrine activity or include a more routine evaluation of liver tissue from female fish
altered endpoints indicative of other types of toxicity. For the EDSP showing decreased Vtg production (Wolf and Wheeler, 2018).
Tier 1 assays, the US EPA proposed a WoE approach (US EPA, 2013). • Include clinical chemistry parameters in mammalian studies to
A more formalized, structured WoE approach also has been pro- evaluate physiological state, paying particular attention to target-
posed by Borgert et al. (2014). In this approach, it is recognized that organ-related parameters. Body temperature also may provide
some EDSP Tier 1 endpoints can be specific and sensitive indicators useful information as rodents can induce a hypothermic response

154
M.S. Marty et al. Regulatory Toxicology and Pharmacology 99 (2018) 142–158

(decreased core temperature and heart rate) to toxicant exposure Atcha, Z., Rourke, C., Neo, A.H., Goh, C.W., Lim, J.S., Aw, C.C., Browne, E.R., Pemberton,
(e.g., Watkinson et al., 2003). D.J., 2010. Alternative method of oral dosing for rats. J. Am. Assoc. Lab. Anim. Sci.


49 (3), 335–343.
Evaluate hepatic enzyme induction, particularly in cases where de- Bacchetta, R., Mantecca, P., Andrioletti, M., Vismara, C., Vailati, G., 2008. Axial-skeletal
creased endocrine responses are seen in the presence of liver weight defects caused by Carbaryl in Xenopus laevis embryos. Sci. Total Environ. 392,
increases and/or liver hypertrophy or hyperplasia. 110–118.

• If kidney and adrenal changes are seen in rodents, examine clinical


Bannister, P., Oadkes, J., Sheridan, P., Losowsky, M.S., 1987. Sex hormone changes in
chronic liver disease: a matched study of alcoholic versus non-alcoholic liver disease.
pathology parameters related to electrolytes, BUN, creatinine or Q. J. Med. 63 (240), 305–313.
other more sensitive biomarkers (see Kidney section above). If re- Bars, R., Broeckaert, F., Fegert, I., Gross, M., Hallmark, N., Kedwards, T., Lewis, D.,
O'Hagan, S., Panter, G., Weltje, L., Weyers, A., Wheeler, J., Galay-Burgos, M., 2011.
levant clinical pathology parameters are affected, consider including Science based guidance for the assessment of endocrine disrupting properties of
an evaluation of the pitutiary to determine whether there is evi- chemicals. Regul. Toxicol. Pharmacol. 59, 37–46.
dence of hypothalamic-pituitary activation. Kidney histopathology Bars, R., Fegert, I., Gross, M., Lewis, D., Weltje, L., Weyers, A., Wheeler, J., Galay-Burgos,
M., 2012. Risk assessment of endocrine disrupting chemicals. Regul. Toxicol.
may be a relevant endpoint to characterize systemic toxicity in fish
Pharmacol. 64, 143–154.
studies so further research should be encouraged. Becker, U., 1993. The influence of ethanol and liver disease on sex hormones and hepatic
• Consider whether corticosterone measurements in rodents or cor- oestrogen receptors in women. Dan. Med. Bull. 40 (4), 447–459.
Beauvais, S.L., Jones, S.B., Parris, J.T., Brewer, S.K., Little, E.E., 2001. Cholinergic and
tisol measurements in fish would be useful, particularly in cases
behavioral neurotoxicity of carbaryl and cadmium to larval rainbow trout
where adrenal weights are increased and/or the adrenal gland ex- (Oncorhynchus mykiss). Ecotoxicol. Environ. Saf. 49 (1), 84–90.
hibits hypertrophy or hyperplasia. Bernal, A.B., Vickers, M.H., Hampton, M.B., Poynton, R.A., Sloboda, D.M., 2010. Maternal

• Consider whether the route of exposure also might contribute to undernutrition significantly impacts ovarian follicle number and increases oxidative
stress in adult rat offspring. PLoS One 5 (12), e15558. www.plosone.org.
stress (e.g., gavage). Include an evaluation of lung and trachea as Berridge, M.V., Herst, P.M., Tan, A.S., 2005. Tetrazolium dyes as tools in cell biology: new
part of these evaluations to rule out problems with dose adminis- insights into their cellular reduction. Biotechnol. Annu. Rev. 11, 127–152.
tration. Beyers, D.W., Sikoski, P.J., 1994. Acetylcholinesterase inhibition in federally endangered

• Evaluate TK to determine if a kinetically derived maximum dose


Colorado squawfish exposed to carbaryl and malathion. Environ. Toxicol. Chem. 13
(6), 935–939.
(KMD) level may be used instead of MTD/MTC (e.g., Saghir et al., Blackburn, S., 2014. Maternal, Fetal and Neonatal Physiology – E-book. Unit 1:
2012). Reproductive and Developmental Processes. Elsevier Health Sciences, New York, pp.

• Include histopathology of affected endocrine pathways (e.g., 136. https://books.google.com/books?id=RNLsAwAAQBAJ&pg=PA136&lpg=


PA136&dq=nifedipine+and+dystocia&source=bl&ots=G9kjLh0I9U&sig=
thyroid, ovary, testis, pituitary, etc.) as this may establish a pattern 1JvYWBe5PL-ckCLIbFie8OYgE9c&hl=en&sa=X&ved=0ahUKEwi20q_
of endocrine changes. When evaluating potential thyroid changes in fnszXAhXCOSYKHSF9CcIQ6AEINTAD#v=onepage&q=nifedipine%20and
%20dystocia&f=false, Accessed date: 19 September 2017.
female rats, stage of estrous cycle also should be monitored.
Boekelheide, K., Andersen, M.E., 2010. A mechanistic redefinition of adverse effects – a
• Include estrous stage when evaluating female rodent reproductive key step in the toxicity testing paradigm shift. ALTEX 27, 243–252.
organ weights. Bonga, S.W.E., Lock, R.A.C., 2008. The osmoregulatory system. Chapter 8. In: DiGiulio,

• Fish should be screened for parasitic infections before use in endo- R.T., Hinton, D.E. (Eds.), The Toxicology of Fishes. CRC Press Taylor & Francis Group,
Boca Raton, FL, USA.
crine tests. If observed in test animals, caution should be taken with Boobis, A.R., Doe, J.E., Heinrich-Hirsch, B., Meek, M.E., Muss, S., Ruchirawat, M.,
data interpretation. Schlatter, J., Seed, J., Vickers, C., 2008. IPCS framework for analyzing the relevance
of a noncancer mode of action for humans. Crit. Rev. Toxicol. 38, 87–96.
Borgert, C.J., Stuchal, L.D., Mihaich, E.M., Becker, R.A., Bentley, K.S., Brausch, J.M.,
Most importantly, a WOE approach across all relevant studies is needed Coady, K., Geter, D.R., Gordon, E., Geter, D.R., Gordon, E., Guiney, P.D., Hess, F.,
to place potentially endocrine-related effects in the context of other Holmes, C.M., LeBaron, M.J., Levine, S., Marty, S., Mukhi, S., Neal, B.H., Ortego, L.S.,
toxicity produced by the chemical in question. Saltmiras, D.A., Snajdr, S., Staveley, J., Tobia, A., 2014. Relevance weighting of Tier
1 endocrine screening endpoints by rank order. Birth Defects Res (Part B) 101,
90–113.
Acknowledgements Brown, A.P., Dinger, N., Levine, B.S., 2000. Stress produced by gavage administration in
the rat. Contemp. Top. Lab. Anim. Sci. 9, 17–21.
Burra, P., 2013. Liver abnormalities and endocrine diseases. Best Pract. Res. Clin.
There was no specific grant associated with this work. This paper
Gastroenterol. 27 (4), 553–563.
was supported by each authors' employer and The Endocrine Policy Cahu, C., Infane, J.Z., Takeuchi, T., 2003. Nutritional components affecting skeletal de-
Forum. velopment in fish larvae. Aquaculture 227, 245–258.
Cameron, J.L., 1997. Stress and behaviorally induced reproductive dysfunction in pri-
mates. Semin. Reprod. Endocrinol. 15 (1), 37–45.
Transparency document Carney, E.W., Zablotny, C.L., Marty, M.S., Crissman, J.W., Anderson, P., Woolhiser, M.,
Holsapple, M., 2004. The effects of feed restriction during in utero and postnatal
Transparency document related to this article can be found online at development in rats. Toxicol. Sci. 82 (1), 237–249.
Carragher, J.F., Sumpter, J.P., 1990. The effect of cortisol on the secretion of sex steroids
https://doi.org/10.1016/j.yrtph.2018.09.002. from cultured ovarian follicles of rainbow trout. Gen. Comp. Endocrinol. 77,
403–407.
References Chapin, R.E., Gulati, D.K., Barnes, L.H., Teague, J.L., 1993. The effects of feed restriction
on reproductive function in Sprague- Dawley rats. Fund. Appl. Toxicol. 20, 23–29.
Choksi, N.Y., Jahnke, G.D., St Hilaire, C., Shelby, M., 2003. Role of thyroid hormones in
Almeida, S.A., Petenusci, S.O., Anselmo-Franci, J.A., Rosa-e-Silva, A.A.M., Lamano- human and laboratory animal reproductive health. Birth Defects Res (Part B) 68,
Carvalho, T.L., 1998. Decreased spermatogenic and androgenic testicular functions in 479–491.
adult rats submitted to immobilization-induced stress from prepuberty. Braz. J. Med. Clearwater, S.J., Pankhurst, N.W., 1997. The response to capture and confinement stress
Biol. Res. 31 (11), 1443–1448. of plasma cortisol, plasma sex steroids and vitellogenic oocytes in the marine teleost,
Aluru, N., Vijayan, M., 2009. Stress transcriptomics in fish: a role for genomic cortisol red gurnard. J. Fish. Biol. 50, 429–441.
signaling. Gen. Comp. Endocrinol. 164, 142–150. Coady, K., Marino, T., Thomas, J., Currie, R., Hancock, G., Crofoot, J., McNalley, L.,
Ankley, G.T., Bennett, R.S., Erickson, R.J., Hoff, D.J., Hornung, M.W., Johnson, R.D., McFadden, L., Geter, D., Klecka, G., 2010. Evaluation of the amphibian metamor-
Mount, D.R., Nichols, J.W., Russom, C.L., Schmieder, P.K., Serrano, J.A., Tietge, J.E., phosis assay: exposure to the goitrogen methimazole and the endogenous thyroid
Villeneuve, D.L., 2010. Adverse outcome pathways: a conceptual framework to hormone L-thyroxine. Environ. Toxicol. Chem. 29, 869–880.
support ecotoxicology research and risk assessment. Environ. Toxicol. Chem. 29, Coady, K.K., Lehman, C.M., Currie, R.J., Marino, T.A., 2014. Challenges and approaches
730–741. to conducting and interpreting the Amphibian metamorphosis assay and the fish
Ankley, G.T., Gray, L.E., 2013. Cross-Species conservation of endocrine pathways: a cri- short-term reproduction assay. Birth Defects Research (Part B) 101, 80–89.
tical analysis of Tier 1 fish and rat screening assays with 12 model chemicals. Creton, S., Saghir, S.A., Bartels, M.J., Billington, R., Bus, J.S., Davies, W., Dent, M.P.,
Environ. Toxicol. Chem. 32, 1084–1087. Hawksworth, G.M., Parry, S., Travis, K.Z., 2012. Use of toxicokinetics to support
Arme, C., 1997. Ligula intestinalis: interactions with the pituitary-gonadal axis of its fish chemical evaluation: informing high dose selection and study interpretation. Regul.
host. J. Helminthol. 71, 83–84. Toxicol. Pharmacol. 62 (2), 241–247.
Ashby, J., Lefevre, P.A., 2000. The peripubertal male rat assay as an alternative to the Crump, M.L., 1989. Effect of habitat drying on developmental time and size at meta-
Hershberger castrated male rat assay for the detection of anti-androgens, oestrogens morphosis in Hyla pseudopuma. Copeia 1989 (3), 794–797.
and metabolic modulators. J. Appl. Toxicol. 20 (1), 35–47. Damsch, S., Eichenbaum, G., Tonelli, A., Lammens, L., Van Den Bulck, K., Feyen, B.,

155
M.S. Marty et al. Regulatory Toxicology and Pharmacology 99 (2018) 142–158

Vandenberghe, J., Megens, A., Knight, E., Kelley, M., 2011. Gavage-related reflux in receptors and sex differences in the hypothalamo-pituitary-adrenal axis. Horm.
rats: identification pathogenesis and toxicological implications (review). Toxicol. Behav. 28 (4), 464–476.
Pathol. 39, 348–360. Harpur, E., Ennulat, D., Hoffman, D., Betton, G., Gautire, J.-C., Riefke, B., Bounous, D.,
Dash, M.C., Hota, A.K., 1980. Density effects on the survival growth rate and metamor- Schuster, K., Beushausen, S., Guffroy, M., Shaw, M., Lock, E., Pettit, S., 2011.
phosis of Rana tigrina tadpoles. Ecology 61, 1025–1028. Biological qualification of biomarkers of chemical-induced renal toxicity in two
De Angelis, S., Tassinari, R., Maranghi, F., Eusepi, A., Di Virgilio, A., Chiarotti, F., Ricceri, strains of male rat. Toxicol. Sci. 122, 235–252.
L., Venerosi Pesciolini, A., Gilardi, E., Moracci, G., Calamandrei, G., Olivieri, A., Haschek, W.M., Rousseaux, C.G., Wallig, M.A., 2010. Fundamentals of Toxicologic
Montovani, A., 2009. Developmental exposure to chlorpyrifos induces alterations in Pathology. Kidney and Lower Urinary Tract, second ed. Academic Press, San Diego,
thyroid and thyroid hormone levels without other toxicity signs in CD1 mice. Toxicol. CA, pp. 261–318.
Sci. 108, 311–319. Hutchinson, T., Ankley, G., Segner, H., Tyler, C., 2006. Screening and testing for endo-
De la Chica, S., Cortes-Denia, P., Ramirez-Exposito, M.J., Arias de Saavedra, J.M., crine disruption in fish – biomarkers as signposts not traffic lights in risk assessment.
Sanchez-Agesta, R., Perez, M.C., Martinez-Martos, J.M., 2007. Doxazosin blockade of Environ. Health Perspect. 114, 106–114.
alpha 1-adrenergic receptors increases rat serum progesterone levels: a putative role Hutchinson, T., Bögi, C., Winter, M., Owens, J., 2009. Benefits of the maximum tolerated
of ovarian angiotensin III in steroidogenesis. Fertil. Steril. 88, 1171–1175. dose (MTD) and maximum tolerated concentration (MTC) concept in aquatic tox-
Dellarco, V.L., McGregor, D., Berry, C Sir, Cohen, S.M., Boobis, A.R., 2006. Thiazopyr and icology. Aquat. Toxicol. 91, 197–202.
thyroid disruption: case study within the context of the 2006 IPCS human relevance Inomata, A., Sasano, H., 2015. Practical approaches for evaluating adrenal toxicity in
framework for analysis of a cancer mode of action. Crit. Rev. Toxicol. 36, 793–801. nonclinical safety assessment. J. Toxicol. Pathol. 28, 125–132.
Denver, R.J., 1997a. Proximate mechanisms of phenotypic plasticity in amphibian me- Jahnke, G.D., Choksi, N.Y., Moore, J.A., Shelby, M.D., 2004. Thyroid toxicants: assessing
tamorphosis. Am. Zool. 37, 172–184. reproductive health effects. Environ. Health Perspect. 112, 363–368.
Denver, R., 1997b. Environmental stress as a developmental cue: corticotropin releasing Joshi, S.C., Mathur, R., Gulati, N., 2007. Testicular toxicity of chlorpyrifos (an organo-
hormone is a proximate mediator of adaptive phenotypic plasticity in amphibian phosphate insecticide) in albino rat. Toxicol. Ind. Health 23, 439–444.
metamorphosis. Horm. Behav. 31, 169–179. Juberg, D.R., Gehen, S.C., Coady, K.K., LeBaron, M.J., Kramer, V.J., Lu, H., Marty, M.S.,
Denver, P., Denver, R.J., 1995. Plasticity of the developmental response to habitat de- 2013. Chlorpyrifos: weight of evidence evaluation of potential interaction with the
siccation in tadpoles of the western spadefoot toad. Am. Zool. 35, 67A. estrogen, androgen, or thyroid pathway. Regul. Toxicol. Pharmacol. 66, 249–263.
DePeyster, A., Mihaich, E., 2014. Hypothesis-driven weight of evidence analysis to de- Judson, R.S., Houck, K.A., Kavlock, R.J., Knudsen, T.B., Martin, M.T., Mortensen, H.M.,
termine potential endocrine activity of MTBE. Regul. Toxicol. Pharmacol. 69, Reif, D.M., Rotroff, D.M., Shah, I., Richard, A.M., Dix, D.J., 2010. Predictive in vitro
348–370. screening of environmental chemicals – the ToxCast project. Environ. Health
DePeyster, A., Mihaich, E., Kim, D.H., Elyea, W.A., Nemec, M.J., Hirakawa, B.P., Leggieri, Perspect. 118, 485–492.
S.E., 2014. Responses of the steroidogenic pathway from exposure to methyl-tert- Judson, R., Kavlock, R., Martin, M., Reif, D., Houck, K., Knudsen, T., Richard, A., Tice,
butyl ether and tert-butanol. Toxicology 319, 23–37. R.R., Whelan, M., Xia, M., Huang, R., Austin, C., Daston, G., Hartung, T., Fowle J.R.
Dӧhler, K.-D., Wong, C.C., Von zur Muhlen, A., 1979. The rat as model for the study of 3rd, , Wooge, W., Tong, W., Dix, D., 2013. Perspectives on validation of high-
drug effects on thyroid function: consideration of methodological problems. throughput assays supporting 21st century toxicity testing. ALTEX 30 (1), 51–56.
Pharmacol. Therapeut. 5, 305–318. Judson, R., Houck, K., Martin, M., Richard, A.M., Knudsen, T.B., Shah, I., Little, S.,
Droin, A., 1992. The developmental mutants of Xenopus. Int Dev Biol 36, 455–464. Wambaugh, J., Setzer, R.W., Kothiya, P., Phuong, J., Filer, D., Smith, D., Reif, D.,
Dumpert, K., Zietz, E., 1984. Plantanna (Xenopus laevis) as a test organism for determining Rotroff, D., Kleinstreuer, N., Sipes, N., Xia, M., Huang, R., Crofton, K., Thomas, R.S.,
the embryotoxic effects of environmental chemicals. Ecotoxicol. Environ. Saf. 8, 2016. Analysis of the effects of cell stress and cytotoxicity on in vitro assay activity
55–74. across a diverse chemical and assay space. Toxicol. Sci. 152 (2) 323-39 and Erratum:
Dunn, J.F., Nisula, B.C., Rodbard, D., 1981. Transport of steroid hormones: binding of 21 Toxicol Sci 153(2):409.
endogenous steroids to both testosterone-binding globulin and corticosteroid-binding Kalantaridou, S.N., Makrigiannakis, A., Zoumakis, E., Chrousos, G.P., 2004. Stress and the
globulin in human plasma. J. Clin. Endocrinol. Metab. 53 (1), 58–68. female reproductive system. J. Reprod. Immunol. 62 (1–2), 61–68.
EFSA, 2016. European Food Safety Authority. Peer review of the pesticide risk assessment Karmaus, A.L., Toole, C.M., Filer, D.L., Lewis, K.C., Martin, M.T., 2016. High-throughput
of the active substance mesotrione. EFSA J. 14 (3), 4419. screening of chemical effects on steroidogenesis using H295R human adrenocortical
Ellis, B.J., 2004. Timing of pubertal maturation in girls: an integrated life history ap- carcinoma cells. Toxicol. Sci. 150, 323–332. https://doi.org/10.1093/toxsci/kfw002.
proach. Psychol. Bull. 130 (6), 920–958. Kelce, W.R., Stone, C.R., Laws, S.C., Gray, L.E., Kemppainen, J.A., Wilson, E.M., 1995.
Engelbregt, M.J., Houdijk, M.E., Popp-Snijders, C., Delemarre-van de Waal, H.A., 2000. Persistent DDT metabolite p,p'-DDE is a potent androgen receptor antagonist. Nature
The effects of intra-uterine growth retardation and postnatal undernutrition on onset 375 (6532), 581–585.
of puberty in male and female rats. Pediatr. Res. 48 (6), 803–807. Keller, D.A., Juberg, D.R., Catlin, N., Farland, W.H., Hess, F.G., Wolf, D.C., Doerrer, N.G.,
Everds, N.E., Snyder, P.W., Bailey, K.L., Bolon, B., Creasy, D.M., Foley, G.L., Rosol, T.J., 2012. Identification and characterization of adverse effects in 21st century toxicology.
Sellers, T., 2013. Interpreting stress responses during routine toxicity studies: a re- Toxicol. Sci. 126 (2), 291–297.
view of the biology, impact and assessment. Toxicol. Pathol. 41, 560–614. Khan, M.S., Knowles, B.B., Aden, D.P., Rosner, W., 1981. Secretion of testosterone-es-
EU, D.A.R., 2012. European Union Draft Assessment Report on Tebuconazole - Compiled tradiol-binding globulin by a human hepatoma-derived cell line. J. Clin. Endocrinol.
Addendum Draft Assessment Report B.9 Ecotoxicology. Rapporteur Member State: Metab. 53 (2), 448–449.
Denmark. Khan, K.K., He, Y.Q., Correia, M.A., Halpert, J.R., 2002. Differential oxidation of mife-
Foilb, A.R., Lui, P., Romeo, R.D., 2011. The transformation of hormonal stress responses pristone by cytochromes P450 3A4 and 3A5: selective inactivation of P450 3A4. Drug
throughout puberty and adolescence. J. Endocrinol. 210 (3), 391–398. Metab. Dispos. 30 (9), 985–990.
Foo, J.T.W., Lam, T.J., 1993. Retardation of ovarian growth and depression of serum Khanum, A., Dufau, M.L., 1988. Angiotensin II receptors and inhibitory actions in Leydig
steroid-levels in the Tilalpia, Oreochromis mossambicus, by cortisol implantation. cells. J. Biol. Chem. 263, 5070–5074.
Aquaculture 115, 133–143. King, S.R., Stocco, D.M., 1996. ATP and a mitochondrial electrochemical gradient are
Freyberger, A., Schladt, L., 2009. Evaluation of the rodent Hershberger bioassay on intact required for functional activity of the steroidogenic acute regulatory (StAR) protein
juvenile males—testing of coded chemicals and supplementary biochemical in- in isolated mitochondria. Endocr. Res. 22 (4), 505–514.
vestigations. Toxicology 262, 114–120. Klein, Z.A., Romeo, R.D., 2013. Changes in hypothalamic-pituitary-adrenal stress re-
Frith, C.H., Botts, S., Jokinen, M.P., Hilary, J.R., Morgan, S.F., Chandra, M., 2000. Non- sponsiveness before and after puberty in rats. Horm. Behav. 64, 357–363.
proliferative lesions of the endocrine system of rats. In: Guides for Toxicologic Kleinow, K.M., Nichols, J.W., Hayton, W.L., McKim, J.M., Barron, M.G., 2008. In:
Pathology, (STP/ARP/AFIP, Washington DC). DiGiulio, R.T., Hinton, D.E. (Eds.), Toxicokinetics in Fishes. The Toxicology of Fishes.
Fuchs, T.C., Hewitt, P., 2011. Biomarkers for drug-induced renal damage and ne- CRC Press, Boca Raton, FL, pp. 55–152.
phrotoxicity – an overview for applied toxicology. AAPS J. 13, 615–631. Kühn, E.R., Geris, K.L., van der Geyten, S., Mol, K.A., Darras, V.M., 1998. Inhibition and
Georgopoulos, N.A., Roupas, N.D., Theodoropoulou, A., Tsekouras, A., Vagenakis, A.G., activation of the thyroidal axis by the adrenal axis in vertebrates. Comp. Biochem.
Markou, K.B., 2010. The influence of intensive physical training on growth and Physiol. Mol. Integr. Physiol. 120 (1), 169–174.
pubertal development in athletes. Ann. NY Acad. Sci. 1205, 39–44. Laurila, A., Kujasalo, J., Ranta, E., 1998. Predator-induced changes in life history in two
Gluud, C., 1988. Testosterone and alcoholic cirrhosis. Epidemiologic, pathophysiologic anuran tadpoles: effects of predator diet. Oikos 83, 307–317.
and therapeutic studies in men. Dan. Med. Bull. 35 (6), 564–575. Laurila, A., Kujasalo, J., 1999. Habitat duration, predation risk and phenotypic plasticity
Goedken, M.J., Kerlin, R.L., Morton, D., 2008. Spontaneous and age-related testicular in common frog (Rana temporaria) tadpoles. J. Anim. Ecol. 68, 1123–1132.
findings in beagle dogs. Toxicol. Pathol. 36 (3), 465–471. Laws, S.C., Ferrell, J.M., Stoker, T.E., Schmid, J., Cooper, R.L., 2000. The effects of
Gonzales, C., Zaleska, M.M., Riddell, D.R., Atchison, K.P., Robshaw, A., Zhou, H., Sukoff atrazine on female Wistar rats: an evaluation of the protocol for assessing pubertal
Rizzo, S.J., 2014. Alternative method of oral administration by peanut butter pellet development and thyroid function. Toxicol. Sci. 58 (2), 366–376.
formulation results in target engagement of BACE1 and attenuation of gavage-in- Laws, S.C., Yavanhxay, S., Cooper, R.L., Eldridge, J.C., 2006. Nature of the binding in-
duced stress responses in mice. Pharmacol. Biochem. Behav. 126, 28–35. teraction for 50 structurally diverse chemicals with rat estrogen receptors. Toxicol.
Grijota-Martinez, C., Diez, D., Morreale de Escobar, G., Bernal, J., Morte, B., 2011. Lack of Sci. 94 (1), 46–56.
action of exogenously administered T3 on the fetal rat brain despite expression of the Laws, S.C., Stoker, T.E., Ferrell, J.M., Hotchkiss, M.G., Cooper, R.L., 2007. Effects of al-
monocarboxylate transporter 8. Endocrinology 152, 1713–1721. tered food intake during pubertal development in male and female Wistar rats.
Haddy, J., Pankhurst, N., 1999. Stress-induced changes in concentrations of plasma sex Toxicol. Sci. 100 (1), 194–202.
steroids in black bream. J. Fish. Biol. 55, 1304–1316. Lemly, D.A., 2002. Symptoms and implications of selenium toxicity in fish: the Belews
Hammond, G.L., Wu, T.S., Simard, M., 2012. Evolving utility of sex hormone-binding Lake case example. Aquat. Toxicol. 57, 39–49.
globulin measurements in clinical medicine. Curr. Opin. Endocrinol. Diabetes Obes. Lethimonier, C., Flouriot, G., Valotaire, Y., Kah, O., Ducouret, B., 2000. Transcriptional
19 (3), 183–189. interference between glucocorticoid receptor and estradiol receptor mediates the
Handa, R.J., Burgess, L.H., Kerr, J.E., O'Keefe, J.A., 1994. Gonadal steroid hormone inhibitory effect of cortisol on fish vitellogenesis. Biol. Reprod. 62, 1763–1771.

156
M.S. Marty et al. Regulatory Toxicology and Pharmacology 99 (2018) 142–158

Levine, S.L., Han, Z., Liu, J., Farmer, D.R., Papadopoulos, V., 2007. Disrupting mi- Guidelines Programme on the Follow-up of This Report. OECD Series on Testing and
tochondrial function with surfactants inhibits MA-10 Leydig cell steroidogenesis. Cell Assessment. Number 92. ENV/JM/MONO(2008)19 Environment Directorate.
Biol. Toxicol. 23, 385–400. Organization for Economic Co-operation and Development, Paris, France Version 24/
Lewin, E., Huan, J., Olgaard, K., 2005. Renal osteodystrophy with special emphasis on 7/2008.
secondary hyperparathyroidism. Rev. Port. Nefrol. Hipert. 19 (1), 9–18. OECD, 2011. H295R Steroidogenesis Assay. Test Guideline 456. Organization for
Loman, J., 1999. Early metamorphosis in common frog Rana temporaria tadpoles at risk of Economic Co-operation and Development, Paris, France. https://www.oecd-ilibrary.
drying: an experimental demonstration. Amphibia-Reptilia 20, 421–430. org/search?value1=steroidogenesis&option1=quicksearch&facetOptions=51&
Lui, P., Padow, V.A., Franco, D., Hall, B.S., Park, B., Klein, Z.A., Romeo, R.D., 2012. facetNames=pub_igoId_facet&operator51=AND&option51=pub_igoId_facet&
Divergent stress-induced neuroendocrine and behavioral responses prior to puberty. value51=%27igo%2Foecd%27, Accessed date: 15 April 2016.
Physiol. Behav. 107 (1), 104–111. OECD, 2012a. Guidance Document on Standardized Test Guidelines for Evaluating
Manson, M.M., Ball, H.W.L., Barrett, M.C., Clark, H.L., Judah, D.J., Williamson, G., Neal, Chemicals for Endocrine Disruption. Series on Testing and Assessment No. 150. vol.
G.E., 1997. Mechanism of action of dietary chemoprotective agents in rat liver: in- 22 Organization for Economic Co-operation and Development, Paris, France ENV/
duction of phase I and II drug metabolizing enzymes and aflatoxin B1 metabolism. JM/MONO(2012).
Carcinogenesis 18 (9), 1729–1738. OECD, 2012b. BG1Luc Estrogen Receptor Transactivation Test Method for Identifying
Marcovecchio, M.L., Chiarelli, F., 2013. Obesity and growth during childhood and pub- Estrogen Receptor Agonists and Antagonists. Test guideline 457. Organization for
erty. World Rev. Nutr. Diet. 106, 135–141. Economic Co-operation and Development, Paris, France. http://www.oecd-ilibrary.
Marić, D., Kostić, T., Kovacević, R., 1996. Effects of acute and chronic immobilization org/environment/test-no-457-bg1luc-estrogen-receptor-transactivation-test-method-
stress on rat Leydig cell steroidogenesis. J. Steroid Biochem. Mol. Biol. 58 (3), for-identifying-estrogen-receptor-agonists-and-antagonists_9789264185395-
351–355. en;jsessionid=7r9189tr9c6g2.x-oecd-live-02, Accessed date: 26 December 2017.
Maronpot, R.R., Yoshizawa, K., Nyska, A., Harada, T., Flake, G., Mueller, G., Singh, B., OECD, 2015. Guideline for the Testing of Chemicals: Medaka Extended One Generation
Ward, J.M., 2010. Hepatic enzyme induction: histopathology. Toxicol. Pathol. 38 (5), Reproductive Toxicity Study (MEOGRT). Test guideline 240. Organization for
776–795. Economic Co-operation and Development, Paris, France. https://www.oecd-ilibrary.
Martínez-Martos, J.M., Arrazola, M., Mayas, M.D., Carrera-González, M.P., García, M.J., org/environment/test-no-240-medaka-extended-one-generation-reproduction-test-
Ramírez-Expósito, M.J., 2011. Diet-induced hypercholesterolemia impaired testicular meogrt_9789264242258-en, Accessed date: 15 April 2018.
steroidogenesis in mice through the renin-angiotensin system. Gen. Comp. OECD, 2016. Stably Transfected Human Androgen Receptor Transcriptional Activation
Endocrinol. 173 (1), 15–19. Assay for Detection of Androgenic Agonist and Antagonist Activity of Chemicals.
Marty, M.S., Johnson, K.A., Carney, E.W., 2003. Effect of feed restriction on Hershberger Test guideline 458. Organization for Economic Co-operation and Development, Paris,
and pubertal male assay endpoints. Birth Defects Res (Part B) 68 (4), 363–374. France. http://www.oecd-ilibrary.org/content/book/9789264264366-en, Accessed
Marty, M.S., Andrus, A.K., Bell, M.P., Passage, J.K., Perala, A.W., Brzak, K.A., Bartels, date: 26 December 2017.
M.J., Beck, M.J., Juberg, D.R., 2012. Cholinesterase inhibition and toxicokinetics in Okada, R., Miller, M., Yamamoto, K., Groef, B.D., Denver, R., Kikuyama, S., 2007.
immature and adult rats after acute or repeated exposures to chlorpyrifos or chlor- Involvement of the corticotropin-releasing factor (CRF) type 2 receptor in CRF-in-
pyrifos-oxon. Regul. Toxicol. Pharmacol. 63, 209–224. duced thyrotropin release by the amphibian pituitary gland. Gen. Comp. Endocrinol.
Marty, M.S., Neal, B.H., Zablotny, C.L., Yano, B.L., Andrus, A.K., Woolhiser, M.R., 150, 437–444.
Boverhof, D.R., Saghir, S.A., Perala, A.W., Passage, J.K., Lawson, M.A., Bus, J.S., Pankhurst, N.W., Van der Kraak, G., 2000. Evidence that acute stress inhibits ovarian
Lamb IV, J.C., Hammond, L., 2013. An F1-extended one generation reproductive steroidogenesis in rainbow trout in vivo, through the action of cortisol. Gen. Comp.
toxicity study (EOGRTS) in CRL:CD(SD) rats with 2,4-dichlorophenoxyacetic acid Endocrinol. 117, 225–237.
(2,4-D). Toxicol. Sci. 136, 527–547. Panter, G., Hutchinson, T., Lange, R., Lye, C., Sumpter, J., Zerulla, M., Tyler, C., 2002.
Marty, M.S., Papineni, S., Coady, K.K., Rasoulpour, R.J., Pottenger, L.H., Eisenbrandt, Utility of a juvenile fathead minnow screening assay for detecting (anti-)estrogenic
D.L., 2015. Pronamide: weight of evidence for potential estrogen, androgen and substances. Environ. Toxicol. Chem. 21 (2), 319–326.
thyroid effects. Regul. Toxicol. Pharmacol. 72 (2), 405–422. Pauluhn, J., 2018. Upper respiratory tract nociceptor stimulation and stress response
McClain, R.M., 1995. Mechanistic considerations for the relevance of animal data on following acute and repeated Cyfluthrin inhalation in normal and pregnant rats:
thyroid neoplasia to human risk assessment. Mutat. Res. 333 (1–2), 131–142. physiological rat-specific adaptations can easily be misunderstood as adversities.
McCormick, C.M., Linkroum, W., Sallinen, B.J., Miller, N.W., 2002. Peripheral and central Toxicol. Lett. 282, 8–24.
sex steroids have differential effects on the HPA axis of male and female rats. Stress 5 Pellegrini, A., Greico, M., Materazzi, G., Gesi, M., Ricciardi, M.P., 1998. Stress-induced
(4), 235–247. morphohistochemical and functional changes in rat adrenal cortex, testis and major
Mihaich, E.M., Schäfers, C., Dreier, D.A., Hecker, M., Ortego, L., Kawashima, Y., Dang, salivary glands. Histochem. J. 30 (10), 695–701.
Z.C., Solomon, K., 2017a. Challenges in assigning endocrine-specific modes of action: Perez-Castillo, A., Bernal, J., Ferreiro, B., Pans, T., 1985. The early ontogenesis of thyroid
recommendations for researchers and regulators. Integrated Environ. Assess. Manag. hormone receptor in the rat fetus. Endocrinology 117, 2457–2461.
13, 280–292. Pfennig, D., 1990. The adaptive significance of an environmentally-cued developmental
Mihaich, E., Capdevielle, M., Urbach-Ross, D., Slezak, B., 2017b. Hypothesis-driven switch in an anuran tadpole. Oecologia 85, 101–107.
weight-of-evidence analysis of endocrine disruption potential: a case study with tri- Plant, T.M., 1994. Puberty in primates. In: In: Knobil, E., Neill, J.D. (Eds.), Physiology of
closan. Crit. Rev. Toxicol. 47, 263–285. Reproduction, vol. 2. Raven Press Limited, New York, pp. 453–485.
Milla, S., Mathieu, C., Wang, N., Lambert, S., Nadzialek, S., Massart, S., Henrotte, E., Powlin, S.S., Cook, J.C., Novak, S., O'Connor, J.C., 1998. Ex vivo and in vitro testis and
Douxfils, J., Melard, C., Mandiki, S.N., Kestemont, P., 2010. Spleen immune status is ovary explants: utility for identifying steroid biosynthesis inhibitors and comparison
affected after acute handling stress but not regulated by cortisol in Eurasian perch, to a Tier I screening battery. Toxicol. Sci. 46, 61–74.
Perca Fluviatilis. Fish Shellfish Immunol. 28 (5–6), 931–941. Rasoulpour, R.J., Andrus, A.K., Marty, M.S., Zhang, F., Thomas, J., LeBaron, M.J.,
Mishra, A.K., Mohanty, B., 2008. Acute toxicity impacts of hexavalent chromium on Papineni, S., Pottenger, L.H., Eisenbrandt, D.L., 2015. Pronamide: human relevance
behavior and histopathology of gill, kidney and liver of the freshwater fish, Channa of liver-mediated rat Leydig cell tumors. Regul. Toxicol. Pharmacol. 72, 394–404.
punctatus (Bloch). Environ. Toxicol. Pharmacol. 26, 136–141. Refetoff, S., 2015. Thyroid hormone serum transport proteins. In: De Groot, L.J.,
Monosson, E., Kelce, W.R., Lambright, C., Ostby, J., Gray Jr., L.E., 1999. Peripubertal Chrousos, G., Dungan, K., Feingold, K.R., Grossman, A., Hershman, J.M., Koch, C.,
exposure to the antiandrogen fungicide, vinclozolin, delays puberty, inhibits the Korbonits, M., McLachlan, R., New, M., Purnell, J., Rebar, R., Singer, F., Vinik, A.
development of androgen-dependent tissues, and alters androgen receptor function in (Eds.), Endotext [Internet]. MDText.com, Inc, South Dartmouth, MA. https://www.
the male rat. Toxicol. Ind. Health 15, 65–79. ncbi.nlm.nih.gov/books/NBK285566/, Accessed date: 21 December 2017.
Morin, P.J., 1986. Interactions between intraspecific competition and predation in an Rehm, S., White, T.E., Zahalka, E.A., Stanislaus, D.J., Boyce, R.W., Wier, P.J., 2008.
amphibian predator-prey system. Ecology 67, 713–720. Effects of food restriction on testis and accessory sex glands in maturing rats. Toxicol.
Newman, R.A., 1988. Adaptive plasticity in development of Scaphiopus couchii tadpoles in Pathol. 36, 687–694.
desert ponds. Evolution 42, 774–783. Rehm, S., 2000. Spontaneous testicular lesions in purpose-bred beagle dogs. Toxicol.
Newman, R.A., 1989. Developmental plasticity of Scaphiopus couchii tadpoles in an un- Pathol. 28 (6), 782–787.
predictable environment. Ecology 70, 1775–1787. Rhodes, M.E., O'Toole, S.M., Wright, S.L., Czambel, R.K., Rubin, R.T., 2001. Sexual
NICEATM and ICCVAM, 2011. National Toxicology Program (NTP) Interagency Center diergism in rat hypothalamic-pituitary-adrenal axis responses to cholinergic stimu-
for the Evaluation of Alternative Toxicological Methods (NICEATM) and the lation and antagonism. Brain Res. Bull. 54 (1), 101–113.
Interagency Coordinating Committee on the Validation of Alternative Methods Ribelin, W.E., 1984. The effects of drugs and chemicals upon the structure of the adrenal
(ICCVAM). Independent Scientific Peer Review Panel Report; Evaluation of the Lumi- gland. Fund. Appl. Toxicol. 4, 105–119.
cell ER (BG1Luc ER TA) Test Method. May 2011. http://iccvam.niehs.nih.gov/docs/ Rocksén, D., Elfsmark, D., Heldestad, V., Wallgren, K., Cassel, G., Gӧransson Nyberg, A.,
endo_docs/EDPRPRept2011.pdf. 2008. An animal model to study health effects during continuous low-dose exposure
NRC, 2007. Toxicity Testing in the 21st Century: a Vision and a Strategy. National to the nerve agent VX. Toxicology 250 (1), 32–38.
Academies Press, Washington D.C. Romeo, R.D., Lee, S.J., Chhua, N., McPherson, C.R., McEwen, B.S., 2004a. Testosterone
Odum, J., Tinwell, H., Tobin, G., Ashby, J., 2004. Cumulative dietary energy intake de- cannot activate an adult-like stress response in prepubertal male tats.
termines the onset of puberty in female rats. Environ. Health Perspect. 112, Neuroendocrinology 79, 125–132.
1472–1480. Romeo, R.D., Lee, S.J., McEwen, B.S., 2004b. Differential stress reactivity in intact and
OECD, 2003. Detailed Background Review of the Uterotrophic Bioassay. OECD Series on ovariectomized prepubertal and adult female rats. Neuroendocrinology 80, 387–393.
Testing and Assessment. Number 38. ENV/JM/MONO(2003)1 Environment Rose, C., 2005. Integrating ecology and developmental biology to explain the timing of
Directorate. Organization for Economic Co-operation and Development, Paris, France frog metamorphosis. Trends Ecol. Evol. 20, 129–135.
27-Mar-2003. Rosol, T.J., Yarrington, J.T., Latendresse, J., Capen, C.C., 2001. Adrenal gland: structure,
OECD, 2007. Report of the Validation Peer Review for the Amphibian Metamorphosis function, and mechanisms of toxicity. Toxicol. Pathol. 29, 41–48.
Assay and Agreement of the Working Group of the National Coordinators of the Test Ruehl-Fehlert, C., Bomke, C., Dorgerloh, M., Palazzi, X., Rosenbruch, M., 2005.

157
M.S. Marty et al. Regulatory Toxicology and Pharmacology 99 (2018) 142–158

Pleistophora infestation in fathead minnows, Pimephales promelas (Rafinesque). J. US EPA, 2011. OCSPP Guideline 890.1450. Endocrine Disruptor Screening Program Test
Fish. Dis. 28, 629–637. Guidelines - OPPTS 890.1450: Pubertal Development and Thyroid Function in Intact
Saghir, S.A., Bartels, M.J., Rick, D.L., McCoy, A.T., Rasoulpour, R.J., Ellis-Hutchings, R.G., Juvenile/Peripubertal Female Rats Standard Evaluation Procedure. August 2011.
Marty, M.S., Terry, C., Bailey, J.P., Billington, R., Bus, J.S., 2012. Assessment of US EPA, 2013. SAP Review of Weight of Evidence: Evaluating Results of EDSP Tier 1
diurnal systemic dose (toxicokinetics) of agrochemicals in regulatory toxicity testing - Screening. http://www.regulations.gov/#!documentDetail;D=EPA-HQ-OPP-2013-
an integrated approach without additional animal use. Regul. Toxicol. Pharmacol. 0230-0004.
63, 321–332. US EPA, 2015a. EDSP: Weight of Evidence Analysis of Potential Interaction with the
Saghir, S.A., Marty, M.S., Zablotny, C., Passage, J., Perala, A., Neal, B.H., Hammond, L., Estrogen, Androgen or Thyroid Pathways. Chemicals: Chlorpyrifos, Ethoprop,
Bus, J., 2013. Life-stage-, sex- and dose-dependent dietary toxicokinetics and re- Glyphosate, Isophorone, Myclobutanil, Tebuconazole. https://www.epa.gov/
lationship to toxicity of 2,4-dichlorophenoxyacetic acid (2,4-D) in rats: implications endocrine-disruption/endocrine-disruptor-screening-program-tier-1-screening-
for toxicity test dose selection, design and interpretation. Toxicol. Sci. 136, 294–307. determinations-and, Accessed date: 19 November 2017.
Schreck, C.B., Contreras-Sanchez, W., Fitzpatrick, M.S., 2001. Effects of stress on fish US EPA, 2015b. Endocrine Disruptor Screening Program Test Guidelines 890.2200:
reproduction, gamete quality, and progeny. In: Cheng-Sheng, L., Donaldson, E.M. Medaka Extended One Generation Reproduction Test (MEOGRT). EPA No. 740-C-15-
(Eds.), Reproductive Biotechnology in Finfish Aquaculture. Elsevier, Amsterdam, pp. 002. https://www.regulations.gov/document?D=EPA-HQ-OPPT-2009-0576-0019,
3–24 ISBN 9780444509130. Accessed date: 26 December 2017.
Shah, I., Setzer, R.W., Jack, J., Houck, K.A., Judson, R.S., Knudsen, T.B., Liu, J., Martin, US EPA, 2017a. Continuing Development of Alternative High-throughput Screens to
M.T., Reif, D.M., Richard, A.M., Thomas, R.S., Crofton, K.M., Dix, D.J., Kavlock, R.J., Determine Endocrine Disruption, Focusing on Androgen Receptor, Steroidogenesis,
2016. Using ToxCast™ data to reconstruct dynamic cell state trajectories and estimate and Thyroid Pathways. Endocrine Disruptor Screening Program. FIFRA SAP
toxicological points of departure. Environ. Health Perspect. 124, 910–919. https:// November 28-30, 2017. https://cfpub.epa.gov/si/si_public_record_Report.cfm?
doi.org/10.1289/ehp.1409029. dirEntryId=338571, Accessed date: 15 April 2016.
Sharma, S., Gupta, U., 1994. Delay of experimentally induced preterm labour in rats with US EPA, 2017b. Pesticide Tolerances: Chlorpyrifos. Federal Register Number:2017-
calcium channel blockers and salbutamol. Indian J. Exp. Biol. 32 (2), 109–112. 06777. EPA-HQ-OPP-2007-1005-0100. Posted April 5, 2017. https://www.
Shimizu, I., 2003. Impact of oestrogens on the progression of liver disease. Liver Int. 23 regulations.gov/document?D=EPA-HQ-OPP-2007-1005-0100, Accessed date: 15
(1), 63–69. April 2017.
Sibly, R., Calow, P., 1986. Physiological Ecology of Animals: an Evolutionary Approach. Viau, V., Meaney, M.J., 1996. The inhibitory effect of testosterone on hypothalamic-pi-
Blackwell Scientific Publications, Oxford. tuitary-adrenal responses to stress is mediated by the medial preoptic area. J.
Siiteri, P.K., Murai, J.T., Hammond, G.L., Nisker, J.A., Raymoure, W.J., Kuhn, R.W., 1982. Neurosci. 16 (5), 1866–1876.
The serum transport of steroid hormones. Recent Prog. Horm. Res. 38, 457–510. Villamor, E., Jansen, E.C., 2016. Nutritional determinants of the timing of puberty. Annu.
Smallridge, R.C., Carr, F.E., Fein, H.G., 1991. Diisopropylfluorophosphate (DFP) reduces Rev. Publ. Health 37, 33–46.
serum prolactin, thyrotropin, luteinizing hormone, and growth hormone and in- Watanabe, K.H., Jensen, K.M., Orlando, E.F., Ankley, G.T., 2007. What is normal? A
creases adrenocorticotropin and corticosterone in rats: involvement of dopaminergic characterization of the values and variability in reproductive endpoints in the fathead
and somatostatinergic as well as cholinergic pathways. Toxicol. Appl. Pharmacol. minnow. Comp. Biochem. Physiol., C 146, 348–356.
108, 284–295. Watkinson, W.P., Campen, M.J., Wichers, L.B., Nolan, J.P., Costa, D.L., 2003. Cardiac and
Smeets, J., Rankouhi, T., Nichols, K., Komen, H., Kaminski, N., Giesy, J., Berg, M.V.D., thermoregulatory responses to inhaled pollutants in healthy and compromised ro-
1999. In vitro vitellogenin production by carp (Cyprinus carpio) hepatocytes as a dents: modulation via interaction with environmental factors. Environ. Res. 92,
screening method for determining (anti)estrogenic activity of xenobiotics. Toxicol. 35–47.
Appl. Pharmacol. 157 (1), 68–76. Wheeler, J.R., Coady, K., 2016. Are all chemicals endocrine disruptors? Integrated
Soderlund, D.M., Clark, J.M., Sheets, L.P., Mullin, L.S., Piccirillo, V.J., Sargent, D., Environ. Assess. Manag. 12 (2), 402–403.
Stevens, J.T., Weiner, M.L., 2002. Mechanisms of pyrethroid neurotoxicity: im- Wheeler, J.R., Gimeno, S., Crane, M., Lopez-Juez, E., Morritt, D., 2005. Vitellogenin: a
plications for cumulative risk assessment. Toxicology 171 (1), 3–59. review of analytical methods to detect (anti) estrogenic activity in fish. Toxicol.
Soliman, A., De Sanctis, V., Elalaily, R., 2014. Nutrition and pubertal development. Indian Mech. Meth. 15, 293–306.
J Endocrinol. Metab. 18 (1), S39–S47. Wheeler, J.R., Panter, G., Weltje, L., Thorpe, K.L., 2013. Test concentration setting for fish
Stocco, D.M., 2001. StAR protein and the regulation of steroid hormone biosynthesis. in vivo endocrine screening assays. Chemosphere 92, 1067–1076.
Annu. Rev. Physiol. 63, 193–213. Wheeler, J.R., Weltje, L., Green, R.M., 2014. Mind the gap: concerns using endpoints from
Stoker, T.E., Laws, S.C., Guidici, D.L., Cooper, R.L., 2000a. The effect of atrazine on endocrine screening assays in risk assessment. Regul. Toxicol. Pharmacol. 69,
puberty in male Wistar rats: an evaluation in the protocol for the assessment of 289–295.
pubertal development and thyroid function. Toxicol. Sci. 58 (1), 50–59. Wiklund, T., Lounasheimo, L., Lom, J., Bylund, G., 1996. Gonad impairment in roach
Stoker, T.E., Parks, L.G., Gray, L.E., Cooper, R.L., 2000b. Endocrine-disrupting chemicals: Rutilus rutilus from Finnish coastal areas of the northern Baltic sea. Dis. Aquat. Org.
prepubertal exposures and effects on sexual maturation and thyroid function in the 26, 163–171.
male rat. A focus on the EDSTAC recommendations. Crit. Rev. Toxicol. 30 (2), Wilbur, H.M., Collins, J.P., 1973. Ecological aspects of amphibian metamorphosis.
197–252. Science 182, 1305–1314.
Stump, D.G., O'Connor, J.C., Lewis, J.M., Marty, M.S., 2014. Key lessons from perfor- Wilbur, H.M., 1976. Density-dependent aspects of metamorphosis in Ambystoma and
mance of the U.S.EPA Endocrine Disruptor Screening Program (EDSP) tier 1 male and Rana sylvatica. Ecology 57, 1289–1296.
female pubertal assays. Birth Defects Res (Part B) 101, 43–62. Wilbur, H.M., 1977. Density dependent aspects of growth and metamorphosis in Bufo
Suttie, A.W., Sutcliffe, C., 2018. Adrenal gland. Chapter 32. Pages 649-667. In: Boorman's americanus. Ecology 58, 196–200.
Pathology of the Rat. Elsevier. https://doi.org/10.1016/13978-0-12-391448-4. Williams, M.A., Penlington, M.C., King, J.A., Hoole, D., Arme, C., 1998. Ligula intestinalis
00033-2. (Cestoda) infections of roach (Rutilus rutilus) (Cyprinidae): immunocytochemical
Tanaka, M., Nakaya, S., Kumai, T., Watanabe, M., Matsumoto, N., Kobayashi, S., 2001. investigations into the salmon- and chicken-II type gonadotrophin-releasing hormone
Impaired testicular function in rats with diet-induced hypercholesterolemia and/or (GnRH) systems in host brains. Acta Parasitol. 43, 232–235.
streptozotocin-induced diabetes mellitus. Endocr. Res. 27, 109–117. Witorsch, R.J., 2016. Effect of stress or elevated glucocorticoids on reproduction and
Tejedo, M., Reques, R., 1994. Plasticity in metamorphic traits of natterjack tadpoles: the development: relevance to the assessment of potential endocrine disruptors. Crit.
interactive effects of density and pond duration. Oikos 71, 295–304. Rev. Toxicol. 46, 420–436.
US EPA, 2003. Health Effects Division (HED) Interim Guidance Document # G2003.02. Wolf, J.C., Wheeler, J.R., 2018. A critical review of histopathological findings associated
Rodent Carcinogenicity Studies: Dose Selection and Evaluation. Washington, DC. with endocrine and non-endocrine hepatic toxicity in fish models. Aquat. Toxicol.
US EPA, 2007. Validation of the Fish Short-term Reproduction Assay: Integrated 197, 60–78.
Summary Report. Endocrine Disruptor Screening Program. Washington, DC. version Wong, C.C., Dӧhler, K.-D., 1978. Effects of light-dark changes on serum levels of pro-
12/15/2007. lactin. TSH. T3, and T4 in male rats of different ages. Acta Endocrinol. 87 (215),
US EPA, 2009a. Endocrine Disruptor Screening Program Test Guidelines - OPPTS 83–84.
890.1500: Pubertal Development and Thyroid Function in Intact Juvenile/ Woodside, B., 1991. Effects of food restriction on the length of lactational diestrus in rats.
Peripubertal Male Rats. EPA 740-C-09-012. https://www.regulations.gov/ Horm. Behav. 25 (1), 70–83.
document?D=EPA-HQ-OPPT-2009-0576-0010, Accessed date: 26 December 2017. Wu, R., Zhou, B., Randall, D., Woo, N., Lam, P., 2003. Aquatic hypoxia is an endocrine
US EPA, 2009b. Endocrine Disruptor Screening Program Test Guidelines - OPPTS disruptor and impairs fish reproduction. Environ. Sci. Technol. 37, 1137–1141.
890.1450: Pubertal Development and Thyroid Function in Intact Juvenile/ Zinkl, J.G., Shea, P.J., Nakamoto, R.J., Callman, J., 1987a. Effects on cholinesterases of
Peripubertal Female Rats. EPA 740-C-09-009. https://www.regulations.gov/ rainbow trout exposed to acephate and methamidophos. Bull. Environ. Contam.
document?D=EPA-HQ-OPPT-2009-0576-0009, Accessed date: 26 December 2017. Toxicol. 38 (1), 22–28.
US EPA, 2009c. Endocrine Disruptor Screening Program Test Guidelines OPPTS Zinkl, J.G., Shea, P.J., Nakamoto, R.J., Callman, J., 1987b. Brain cholinesterase activity of
890.1350: Fish Short-term Reproduction Assay. https://www.regulations.gov/ rainbow trout poisoned by carbaryl. Bull. Environ. Contam. Toxicol. 38 (1), 29–35.
document?D=EPA-HQ-OPPT-2009-0576-0007, Accessed date: 26 December 2017.

158

You might also like