Mechanisms of action of agrochemicals acting as endocrine disrupting chemicals

Mechanisms of action of agrochemicals acting as endocrine disrupting chemicals

Journal Pre-proof Mechanisms of action of agrochemicals acting as endocrine disrupting chemicals Genoa R. Warner, Vasiliki E. Mourikes, Alison M. Neff...

1MB Sizes 1 Downloads 60 Views

Journal Pre-proof Mechanisms of action of agrochemicals acting as endocrine disrupting chemicals Genoa R. Warner, Vasiliki E. Mourikes, Alison M. Neff, Emily Brehm, Jodi A. Flaws PII:

S0303-7207(19)30382-X

DOI:

https://doi.org/10.1016/j.mce.2019.110680

Reference:

MCE 110680

To appear in:

Molecular and Cellular Endocrinology

Received Date: 1 October 2019 Revised Date:

6 December 2019

Accepted Date: 10 December 2019

Please cite this article as: Warner, G.R., Mourikes, V.E., Neff, A.M., Brehm, E., Flaws, J.A., Mechanisms of action of agrochemicals acting as endocrine disrupting chemicals, Molecular and Cellular Endocrinology (2020), doi: https://doi.org/10.1016/j.mce.2019.110680. This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of record. This version will undergo additional copyediting, typesetting and review before it is published in its final form, but we are providing this version to give early visibility of the article. Please note that, during the production process, errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain. © 2019 Published by Elsevier B.V.

Abstract Submission MCE-D-19-00600 Mechanisms of Action of Agrochemicals Acting as Endocrine Disrupting Chemicals

Agrochemicals represent a significant class of endocrine disrupting chemicals that humans and animals around the world are exposed to constantly. Agrochemicals can act as endocrine disrupting chemicals through a variety of mechanisms.

Recent studies have shown that

several mechanisms of action involve the ability of agrochemicals to mimic the interaction of endogenous hormones with nuclear receptors such as estrogen receptors, androgen receptors, peroxisome proliferator activated receptors, the aryl hydrocarbon receptor, and thyroid hormone receptors. Further, studies indicate that agrochemicals can exert toxicity through non-nuclear receptor-mediated mechanisms of action. Such non-genomic mechanisms of action include interference with peptide, steroid, or amino acid hormone response, synthesis and degradation as well as epigenetic changes (DNA methylation and histone modifications).

This review

summarizes the major mechanisms of action by which agrochemicals target the endocrine system.

1 2 3 4 5

Review

6 7

Mechanisms of Action of Agrochemicals Acting as Endocrine Disrupting Chemicals

8

Special Issue on “Health effects of agrochemicals acting as endocrine disrupters”

9 10

Genoa R. Warner, Vasiliki E. Mourikes, Alison M. Neff, Emily Brehm, Jodi A. Flaws

11 12

Department of Comparative Bioscience, University of Illinois at Urbana-Champaign, Urbana, IL 61802

13 14

Corresponding author

15 16 17 18 19

Jodi A Flaws [email protected] 2001 S. Lincoln Ave Urbana, IL 61802

20 21

22 23

Introduction In Silent Spring in 1962, Rachel Carson recognized that chemicals in the environment,

24

particularly the pesticide dichlorodiphenyltrichloroethane (DDT) and other agrochemicals, were

25

impacting reproduction and development in wildlife and had the potential to harm humans (Carson,

26

1962). Although she did not know it at the time, Rachel Carson was describing the effects of endocrine

27

disrupting chemicals (EDCs), compounds that interfere with the action of hormones in the body (Gore et

28

al., 2015). Over the past half century, scientists have identified the mechanisms through which DDT

29

causes the softening of eggshells and decreases in bird populations described in Silent Spring as well as

30

numerous other mechanisms through which environmental chemicals can interfere with the complex

31

operations of the endocrine system.

32

In this review, we summarize endocrine disruptor mechanisms of action with an emphasis on

33

agrochemicals that act as EDCs. The endocrine system operates through complex and finely tuned

34

hormone signaling pathways. As such, EDCs exhibit numerous modes of action. The most well-known

35

mechanisms of EDC action involve mimicking the interaction of endogenous hormones with nuclear

36

receptors, but evidence supporting non-nuclear receptor-mediated mechanisms of action will also be

37

discussed herein.

38

The Endocrine System

39

The endocrine system controls reproduction, development, growth, metabolism, tissue and brain

40

function, and other physiological functions in the body. Endocrine glands distributed throughout the

41

body, including the brain, thyroid, mammary glands, cardiovascular system, and reproductive organs,

42

produce and release hormones. Understanding the complexities of hormone signaling provides vital

43

context to the wide range of EDC mechanisms.

44 45

The canonical pathway of hormone signaling involves the binding of a hormone to its corresponding nuclear receptor(s). Ligand binding induces structural changes in the receptor that lead to

46

dimerization, exposure of co-factor binding sites, and DNA binding. Genomic binding may occur directly

47

to response elements in the genome (Figure 1A) or through transcription factors (Figure 1B). Hormones

48

may also function through non-genomic rapid signaling via secondary messengers (Figure 1C).

49

Exogenous chemicals, including pharmaceuticals and EDCs, can mimic hormones as ligands via any of

50

these three mechanisms as well as repress transcription by recruiting co-repressors (Figure 1D) (Heldring

51

and Pike, 2007).

52 53

Figure 1: Model of the canonical hormone signaling via nuclear receptors (NR). Hormones (grey

54

triangles) act as ligands for nuclear receptors which can bind directly to DNA (A) or indirectly via

55

transcription factors (TF) (B) to regulate gene expression. Hormones may also signal through non-

56

genomic pathways by interacting with membrane nuclear receptors or cytoplasmic receptors to initiate

57

signaling cascades via secondary messengers (SM) that lead to rapid physiological effects without

58

altering gene expression (C). Hormone mimicking chemicals may operate through any of the mechanisms

59

shown in A-C as well as recruit co-repressors to block gene transcription (D). Adapted from Helding and

60

Pike, 2007.

61

Extremely low concentrations of hormones are present in the body, on the order of pg–ng/mL for

62

estradiol, testosterone, and thyroid hormone (Vandenberg et al., 2012). Furthermore, only small

63

percentages of many circulating hormones, including steroid and thyroid hormones, are bioavailable; the

64

majority are bound to carrier proteins. The endocrine system is tuned to respond to minor changes in

65

bioavailable hormone concentrations. Signaling at these extremely low concentrations is facilitated by the

66

high affinity of hormones for their receptors as well as non-linear relationships between hormone

67

concentration, receptor occupancy, and biological effect (Welshons et al., 2003). Thus, a change in

68

hormone concentrations at low doses has a stronger effect than the same magnitude of change at a higher

69

dose. For a full discussion of this, see Vandenberg et al., 2012. In the context of understanding endocrine

70

disruptor mechanisms, this means that studying low doses of EDCs (below those typically used in

71

toxicological testing) is required to identify mechanisms, and that the mechanisms may change at

72

different doses. This is evident in non-monotonic dose response curves (NMDRCs) observed in

73

toxicological studies of EDCs, in which the sign of the curve changes (i.e. U or inverted U) as the dose

74

changes. NMDRCs are established for hormonal pharmaceuticals such as tamoxifen yet they continue to

75

be challenged in the context of EDCs (Vandenberg et al., 2012).

76

EDC Mechanisms of Action

77

Endocrine disrupting chemicals can bind to receptors to mimic endogenous hormones, but they

78

also act by altering hormone signaling in a variety of other ways. EDCs may interact with multiple

79

receptors, including non-nuclear receptors, as agonists, in which they facilitate genomic interactions, or as

80

antagonists, in which they cause a conformational change to the receptor to block action. They may also

81

trigger non-genomic signaling that is independent of nuclear receptors. Importantly, EDCs can interfere

82

with endogenous hormone synthesis and degradation to alter hormone levels. Recent studies have also

83

identified how EDCs can follow an epigenetic mode of action by altering genomic methylation and

84

histone modifications.

85

Nuclear Receptors

86

Nuclear receptors are ligand dependent transcription factors that bind hormones and exert long

87

term control of their target cell phenotype. This is in contrast to membrane receptors which elicit faster,

88

short term effects on their respective cells. Nuclear receptors play a crucial physiologic role in

89

development, organ homeostasis, metabolism, immune function, and reproduction (Balaguer et al., 2019).

90

Hormones are the endogenous ligands of most of the major classes of nuclear receptors, but EDCs may

91

also bind nuclear receptors. Nuclear receptors can act as repressors or activators of gene transcription

92

depending on ligand binding status, the identity of the ligand itself, and available coregulators.

93

Nuclear receptors are composed of several conserved structural domains, including a DNA

94

binding domain and a ligand binding domain (Weikum et al., 2018). The DNA binding domain is

95

responsible for interacting with the genome and typically interacts with specific sequences called DNA

96

response elements, whereas the ligand binding domain contains the binding pocket for small molecules.

97

Unliganded nuclear receptors can be found in the nucleus or cytoplasm and may bind to target genes,

98

resulting in recruitment of corepressors. Following endogenous ligand binding, conformational changes to

99

nuclear receptors disrupt corepressors leading to dimerization and interaction with DNA and co-

100

activators. Upon binding to DNA or interacting with tethering co-factors, nuclear receptors further

101

activate co-factors to facilitate transcription.

102

Endocrine disrupting chemicals can interact with nuclear receptors directly and elicit strong

103

biological consequences (Gore et al., 2015). Agricultural chemicals, including herbicides, insecticides,

104

rodenticides, and fungicides, are prime examples of synthetic endocrine disrupting molecules exogenous

105

to normal eukaryotic biology that interact with nuclear receptors (Beischlag et al., 2008). Table 1

106

provides examples of the biological effects of select agrochemicals that act through nuclear receptors.

107

Table 1: Agrochemical effects mediated through nuclear receptors Chemical atrazine

dibutyltin (DBT)

DDT

DDE dieldrin & aldrin

fenarimol

lindane (γ-HCH)

Mode of Action

Effect

Reference

AhR mediated

Nephrotoxicity

(Zhang et al., 2019)

PPARγ activation

Androgenic antagonist: inhibits dihydrotestosterone (DHT) binding to AR

ESR1 activation leading to altered expression of Pgr, Ccnd1, Cyp19a1 AR antagonist: inhibits DHT binding to AR AR antagonist AR antagonist leading to decrease in prostate binding protein, ornithin decarboxylase, and insulinlike-growth factor 1 mRNA levels Androgenic antagonist: inhibits DHT binding to AR

linuron

AR antagonist

methoxychlor

ESR1 and ESR2 agonist

methoxychlor procymidone

ESR1 and ESR2 agonist AR antagonist

propiconazole & tebuconazole 2,3,7,8tetrachlorodibenzodioxin (TCDD)

Pregnane X receptor (PXR) activation

2,3,7,8tetrachlorodibenzodioxin (TCDD) tributyltin (TBT)

AhR activation

AhR activation during in utero exposure

AhR activation

Increased expression of adipogenic genes, promoted adipogenic differentiation, increased lipid accumulation, decreased glucose tolerance, increased circulating leptin levels in males Decreased fertility and cryptorchidism. Reduction of testicular weight, decreased in number and motility of spermatozoa in epididymis, loss of gametes in lumen of seminiferous tubules, decreased testosterone production by testes, increased luteinizing hormone and follicle stimulating hormone Hormonal carcinogenesis in uterus and ovaries Decreased fertility and cryptorchidism Reproductive performance affected

(ChamorroGarcía et al., 2018)

(Kelce et al., 1995; Sakly, 2001)

(Kalinina et al., 2017) (Kelce et al., 1995) (Danzo, 1997)

Reduced weight of ventral prostate, seminal vesicles, and bulbourethral glands

(Vinggaard et al., 2005)

Altered testis histology

(Danzo, 1997)

Disruption of reproductive tract development, reduction of accessory gland weight, reduction of epidydimal weight, increased serum estradiol and luteinizing hormone Premature nuclear expression of ER gene in neonatal uterine epithelium Inhibition of folliculogenesis and stimulation of anti-Müllerian hormone production in the ovary In males: shortened anogenital distance, hypospadias, reduced weight and altered histology of prostate, permanent nipples, ectopic undescended testes Hepatocellular hypertrophy and hepatocellular steatosis Cleft palate Downregulation of uterine and hepatic ESR1 results in repressed estradiol function UGT1 induction resulting in decreased serum T4 concentrations and compensatory excretion of thyroid

(Lambright, 2000)

(Eroschenko et al., 1996) (Uzumcu et al., 2006) (Ostby et al., 1999) (Knebel et al., 2019) (Abbott et al., 1996) (Romkes et al., 1987) (Nishimura et al., 2002)

stimulating hormone PPARγ and retinoid X receptor (RXR) activation PPARγ induction and Esr1 repression tributyltin (TBT) vinclozolin AR antagonist

In utero exposure results in increase lipid accumulation in adipose tissue, liver, and testis Impaired metabolic functions on liver and pancreas via lipid accumulation in white adipose tissue and hepatic inflammation Hypospadias, undescended testes, delayed puberty, transgenerational prostate disease

(Grün et al., 2006) (Bertuloso et al., 2015) (Monosson et al., 1999; Christiansen et al., 2008; Anway, 2015)

108 109

The structural similarities between EDCs and endogenous hormones form the basis for nuclear

110

receptor-based mechanisms of endocrine disruption. Both classes of molecules are generally hydrophobic,

111

lipid-soluble small molecules. In addition, many of the most worrisome EDCs have structural elements

112

such as hydroxyl groups in the proper position to interact with amino acid residues in the ligand binding

113

pocket in the same manner as endogenous hormones. For example, the methoxychlor metabolite 2,2-bis-

114

(p-hydroxyphenyl)-1,1,1-trichloroethane (HPTE) contains both hydroxyl groups and hydrophobic

115

aromatic regions that allow it to bind to estrogen receptors, whereas the pyrethroid deltamethrin shares a

116

diphenyl ether structural moiety with thyroid hormones (Delfosse et al., 2015; Du et al., 2010) (Figure 2).

117

Once EDCs bind a receptor, they either activate the hormone receptor, amplifying physiological hormonal

118

activity, or antagonize endogenous hormone action to block activity. Due to the structural similarities

119

between EDCs and endogenous hormones as exemplified in Figure 2, EDCs are typically investigated as

120

competitive binders in the ligand binding domain of the receptor. In addition, EDCs which are not

121

structurally similar to hormones can still directly affect nuclear receptors by stimulation or inhibition of

122

receptor expression. This leads to imbalances in endocrine homeostasis through modification of hormone

123

receptor turnover/availability.

124 125

Figure 2: Structural similarities between endogenous hormones and agrochemicals. Top: Estradiol

126

and the methoxychlor metabolite HPTE share phenolic structure (blue) that facilitates the binding of

127

HPTE to human estrogen receptor 1 (ESR1) (Delfosse et al., 2015). The second hydroxyl group of HPTE

128

does not interact in the ESR1 binding pocket in the same way as the second hydroxyl on estradiol.

129

Bottom: The thyroid hormone triiodothyronine (T3) and pyrethroid deltamethrin share diphenyl ether

130

functional groups (green) that facilitate binding to thyroid receptors (Du et al., 2010).

131

In vitro assays are valuable tools for investigating the ability of EDCs to directly interact with

132

receptors and alter transcription processes. One commonly used assay is a recombinant receptor and

133

reporter gene assay in which a luciferase reporter gene is associated with a nuclear receptor response

134

element such that binding of a ligand to the receptor triggers transcription and luciferase activity (Legler

135

et al., 1999). Although these assays are specific, responsive, and quick, they do not measure binding

136

affinity nor identify which step of the nuclear receptor activation process is disrupted (Xiang et al., 2017;

137

Judson et al., 2018). High throughput screening of nuclear receptor hormone agonists and antagonists is a

138

top priority of regulatory agencies. In the US, the Tox21 program high throughput screening program

139

utilizes multiple in vitro and in silico assays to assess ligand binding, dimerization, DNA binding, RNA

140

transcription, and other steps in the activation process (Judson et al., 2018). However useful in vitro and

141

silico models are for predicting EDC interaction with nuclear receptors, these models do not integrate

142

multiple mechanisms of endocrine disruption that can occur in a whole animals. For this reason, in vivo

143

testing is also necessary to understand mechanisms that may occur simultaneously in multiple organ

144

systems to extrapolate to human and environmental health (Gore et al., 2015).

145

Estrogen Receptors (ESR)

146

Estrogen receptor alpha and estrogen receptor beta (ESR2) have unique and overlapping

147

physiological roles that are highly tissue and cell type dependent. Both receptors are expressed in the

148

brain, lungs, uterus, ovaries, breast, heart, and intestines. ESR1 is predominantly expressed in hepatocytes

149

of the liver and the hippocampus, whereas ESR2 is predominantly expressed in the prostate, vagina, and

150

cerebellum (Taylor and Al-Azzawi, 2000). ESR1 and ESR2 have quite similar binding pockets with

151

subtle differences in amino acids at the ligand binding domain that explain the ligand selectivity between

152

the two types (Kuiper et al., 1998). Both ESR1 and ESR2 regulate gene expression in response to

153

estrogen exposure via ligand dependent or ligand independent mechanisms, and each subtype can mediate

154

unique responses to ligands.

155

Inappropriate ESR signaling can lead to increased risk of hormone-dependent cancer, impaired

156

fertility, abnormal fetal growth and development, and altered metabolism in white adipose tissue (Zhao et

157

al., 2008). Many EDCs display estrogenic activity and interfere with normal estrogen signaling mediated

158

by ESR1 and ESR2. A notable characteristic of ESRs that makes them susceptible to direct interaction

159

with EDCs is their large binding pockets and broad specificity for ligands. The binding pockets are much

160

larger than the estrogen molecule itself (Pike et al., 1999; Brzozowski et al., 1997). Although ESR1 and

161

ESR2 have similar affinities for estrogen, EDCs and other exogenous ligands may display higher affinity

162

for one subtype over the other. For example, the o,p' isomer of DDT, which is the most estrogenic isomer

163

of DDT, has a higher relative binding affinity for ESR2 than ESR1 (Kuiper et al., 1998).

164

Organochlorine pesticides, including methoxychlor and DDT, are notorious estrogenic

165

agrochemicals. They bind both ESR1 and ESR2 (Kuiper et al., 1998) and elicit ER binding to DNA both

166

directly and through tethering as described above. DDT adversely affects the female reproductive tract by

167

stimulating uterine proliferation and impairing normal follicle development (Tiemann, 2008). Although

168

DDT was banned in the 1970s, it is persistent in the environment and accumulates in adipose tissue,

169

making it a relevant threat to public health today. DDT and its dechlorination metabolite

170

dichlorodiphenyldichloroethylene (DDE) have been detected in human adipose tissue around the world

171

many years after its use was terminated (Turusov et al., 2002).

172

Methoxychlor is another potent endocrine disruptor which stimulates uterotrophic activity and

173

impairs overall fertility in rat models (Cummings, 1997). Methoxychlor itself has low binding affinities

174

for ER. However, its major metabolite HPTE is a well-studied agonist for both ESR1 and ESR2 and is

175

likely the responsible agent for the endocrine disrupting effects of methoxychlor (Gaido et al., 2000).

176

HPTE acts as an agonist for ESR1 and antagonist for ESR2 (Gaido et al., 2000). ESR1 knockout mice do

177

not respond to HPTE treatment, indicating the effects of HPTE on gene regulation in the mouse uterus are

178

dependent on ESR1. HPTE may also mediate effects through non-classical ER signaling mechanisms

179

similar to estrogens. HPTE and estrogen treatment led to similar gene expression profiles in uteri from

180

mice expressing an ESR1 mutant deficient in DNA binding, which limits ESR1 mediated gene regulation

181

to the pathway in which ESR1 tethers to DNA through transcription factors like AP-1 and Sp1 (Hewitt

182

and Korach, 2011).

183

Pyrethroids also exhibit estrogenic and antiestrogenic activity, with variable results observed in

184

tests of different chemicals in vitro, suggesting that individual screening of members of a class of

185

pesticides is necessary to identify endocrine disrupting activity (Saillenfait et al., 2016). In addition to in

186

vitro screening, whole-organism bioassays have been used to detect the estrogenic activity of EDCs. In

187

transgenic medaka expressing green fluorescent protein, the herbicide linuron elicited anti-estrogenic

188

activity, whereas fenoxycarb, a less-studied insecticide, showed no effect on estrogen, androgen, or

189

thyroid signaling (Spirhanzlova et al., 2017). Recent studies have also investigated the estrogenic

190

properties of mixtures of pesticides. An in vitro analysis using multiple screening methods of individual

191

pesticides and mixtures on ESR1 and ESR2 found additive effects of the mixtures (Seeger et al., 2016),

192

suggesting the need for further experiments on mixtures, which are more representative of human

193

exposure.

194

Androgen Receptor

195

The androgen receptor (AR) is a ligand dependent nuclear transcription factor responsible for

196

male fetal development, secondary sex characteristics at puberty, and maintenance of spermatogenesis.

197

Beyond sex differentiation, the AR plays a critical role in development and maintenance of the

198

reproductive, musculoskeletal, cardiovascular, immune, neural, and haemopoietic systems (Rana et al.,

199

2014). Consequences of dysfunctional AR include infertility (Nenonen et al., 2011), delayed puberty

200

onset (Mouritsen et al., 2013), and cryptorchidism (Davis-Dao et al., 2012). AR signaling is also involved

201

in the development of tumors in the prostate, bladder, liver, kidney and lung.

202

The fungicides vinclozolin (Gray et al., 1994), procymidone (Hosokawa et al., 1993), and

203

prochloraz (Vinggaard et al., 2002) target the AR and their phenotypic consequences have been well

204

documented. Five triazole fungicides, tebuconazole, uniconazole, hexaconazole, peneconazole, and

205

bitertanol showed anti-androgenic activity toward human AR both before and after metabolism mediated

206

by human liver microsomes (Lv et al., 2017). Many insecticides and their metabolites have also been

207

shown to inhibit androgen receptor dependent transcriptional activity. These include multiple isomers of

208

DDT, DDE, and dichlorodiphenyldichloroethane (DDD) as well as methoxychlor and HPTE (Maness et

209

al., 1998). Urea based herbicides such as linuron have also been identified as endocrine disruptors via AR

210

antagonism (Lambright, 2000; Spirhanzlova et al., 2017). Agrochemicals have also been studied for their

211

effects against AR as mixtures. Various combinations of androgen antagonists have been shown to disrupt

212

male reproductive development in a cumulative and additive matter, regardless of their individual modes

213

of action, and differences in tissue selectivity (Wilson et al., 2008).

214 215

Peroxisome Proliferator Activated Receptor

The peroxisome proliferator activated receptor (PPAR) is a ligand activated nuclear receptor that

216

participates in energy combustion via stimulation of lipid catabolism and energy storage via stimulation

217

of adipogenesis (Neschen et al., 2007). Lipophilic hormones, monounsaturated fatty acids,

218

polyunsaturated fatty acids, and eicosanoids are all endogenous ligands of PPAR (Ayisi et al., 2018).

219

Three subtypes of PPAR, PPARα, PPARβ, and PPARγ exist, with differing tissue distribution, ligand

220

specificity, physiologic role, and mechanism of action. Once bound to their endogenous ligands, PPARs

221

can induce the expression of genes and enzymes involved in lipid metabolism through both genomic and

222

non-genomic mechanisms.

223

In a study of the PPARα and PPARγ activity of 200 pesticides using in vitro reporter gene assays,

224

only three pesticides, doclofop-methyl, pyrethrins, and imazalil, exhibited PPAR agonist activity, which

225

was further confirmed in vitro (Takeuchi et al., 2006). Due to its primary physiologic role as a regulator

226

of adipogenesis and lipolysis, PPARγ is a major target of agricultural chemicals that act as obesogens.

227

Tributyltin (TBT), an antifouling agent, alters PPAR-mediated differentiation of adipocytes and promotes

228

adipogenesis in liver and adipose tissue (Maradonna and Carnevali, 2018). Exposure to dibutyltin (DBT),

229

the major metabolite of TBT in the body, binds PPARγ to accelerate adipogenesis in both human and

230

mouse mesenchymal stem cells. Interestingly, human cells were shown to be significantly more

231

responsive to DBT than mouse cells (Chamorro-García et al., 2018). The fungicide triflumizole has also

232

been shown to activate PPARγ to promote adipogenesis, acting as an obesogen in vivo (Li et al., 2012).

233

Aryl Hydrocarbon Receptor (AhR) Complex

234

The aryl hydrocarbon receptor complex includes the aryl hydrocarbon receptor (AhR) and the

235

aryl hydrocarbon receptor nuclear translocator (ARNT). Although it is a basic region-helix/loop.helix

236

(bHLH) protein and not a nuclear receptor, the AhR is a ligand dependent transcription factor with similar

237

structure and function to nuclear receptors (Nebert, 2017). Following ligand binding, the AhR forms a

238

heterodimer with ARNT that binds to DNA at xenobiotic response elements (Figure 3). For many years

239

prior to the discovery of endogenous AhR ligands, the receptor’s most notable role was metabolism of

240

exogenous, synthetic chemicals. The Ahr is activated by exogenous chemicals, and relays environmental

241

signals to the cell (Elferinks et al., 1990). Thus, many mechanisms through which the AhR mediate toxic

242

effects in the body are well studied. Much of what is known today regarding mechanisms of AhR

243

mediated modulation of gene expression has been studied in the context of dioxin metabolism. Recently,

244

endogenous ligands of AhR have been identified, whereas the receptor’s physiological roles in cell

245

proliferation and differentiation, immune response, inflammation, and regulation of circadian rhythm are

246

less understood (Nebert, 2017).

247 248

Figure 3: Mechanism of AhR signaling. Synthetic chemicals such as dioxin (green triangles) activate

249

AhR by binding to the receptor and translocating to the nucleus, where the complex dimerizes with the

250

aryl hydrocarbon receptor nuclear translocator (ARNT) and controls transcription by interacting with

251

xenobiotic response elements (XRE) in DNA. Transcriptional regulation by dioxin and other EDCs leads

252

to an upregulation of xenobiotic metabolism to create toxic metabolites that impact the immune system,

253

neurological function, reproduction, and carcinogenesis.

254

2,3,7,8-Tetrachlorodibenzodioxin (TCDD, also known as dioxin) is one of the most widely

255

studied AhR ligands. The AhR has been shown to modulate the activity of other transcription factors

256

including ESRs and ARs (Ohtake et al., 2011). In the presence of estrogens and androgens, the AhR-

257

ARNT heterodimer downregulates transcriptional events that would otherwise be upregulated in the

258

presence of these hormones. Dioxin bound AhR acts as a substrate specific adaptor component that

259

targets estrogen and androgen bound receptors for degradation by the cullin 4B ubiquitin ligase complex

260

(Ohtake et al., 2007). The opposite effect on transcription has been observed when AhR is activated in the

261

absence of estrogen and androgen hormones. The AhR-ARNT complex physically associates with

262

unliganded estrogen and androgen receptors bringing transcriptional co-activators to the promotors of

263

each of these receptors (Ohtake et al., 2003). Hexachlorobenzene, a banned but persistent dioxin-like

264

fungicide, also activates AhR to trigger both genomic and non-genomic effects (van Birgelen, 1998; Miret

265

et al., 2019).

266

Reporter gene assays of 23 pesticides and insecticides in human and rat cell lines indicated AhR

267

agonistic effects by iprodione, chlorpyrifos, and prochlorax, whereas five additional pesticides exhibited

268

mixed activity in the two cell lines (Long et al., 2003). Another reporter gene assay study identified

269

eleven pesticides that agonize AhR function, while two inhibited AhR activity (Ghisari et al., 2015).

270

Pyrethroids have structural elements reminiscent of dioxins, suggesting potential AhR binding activity

271

(Brander et al., 2016); cypermethrin has been shown to antagonize TCDD-induced AhR transactivation

272

(Ghisari et al., 2015). The fungicide propiconazole also activates AhR (Knebel et al., 2018). This has

273

been confirmed in silico, in vitro, and in vivo, wherein AhR was activated in a luciferase reporter gene

274

assay in a human cell line and further increased mRNA and enzyme expression of genes controlled by

275

AhR in the livers of rats treated with propiconazole via diet (Knebel et al., 2018).

276

Thyroid Receptors

277

Thyroid receptors (TR) are another class of ligand-dependent transcription factors. Studies show

278

that four active isoforms of thyroid receptors (TRα1, TRβ1, TRβ2, and TRβ3) bind the endogenous

279

thyroid hormones triiodothyronine (T3) and thyroxine (T4) (Zoeller, 2012). TRα and TRβ isoforms are

280

differentially expressed in tissues throughout the body. Isoform distribution is particularly important

281

during fetal brain development when thyroid hormone signaling is vital for normal development. Thyroid

282

receptors are also important for physiological control of metabolism and the cardiovascular system

283

(Zoeller, 2012).

284

Extensive evidence in humans and animals indicates that pesticides including chlorpyrifos, DDT,

285

and methoxychlor can disrupt thyroid signaling and neurodevelopment (Boas et al., 2012). However,

286

most evidence on pesticides implicates thyroid hormone synthesis, bioavailability, and metabolism for

287

these effects (Ghassabian and Trasande, 2018). Pesticides generally do not have a high degree of

288

structural similarity to thyroid hormones and thus have been less investigated as TR binders compared to

289

environmental chemicals such as polychlorinated biphenyls (PCBs) and polybrominated diphenyl ethers

290

(PBDEs) that have been shown bind directly to TR (Ghassabian and Trasande, 2018; Zoeller, 2007).

291

However, in vitro reporter gene assays have indicated the ability of multiple pesticides to bind to TR. Ten

292

pyrethroids or pyrethroid metabolites have demonstrated antagonistic activity against TR (Du et al.,

293

2010). In an in vitro reporter assay analysis of the binding of 21 pesticides to human TR, 13 bound

294

directly to TR, with 5 exhibiting agonistic activity, including procymidone, imidacloprid, mancozeb, and

295

atrazine, whereas 11 were antagonistic and 4 exhibited both agonistic and antagonistic activity (Xiang et

296

al., 2017).

297

Non-genomic Signaling

298

While genomic mechanisms of endocrine disruption involve activation of nuclear receptors that

299

translocate to the nucleus to modulate gene expression, EDCs also exhibit non-genomic mechanisms of

300

action. These actions are characterized as rapid effects that do not directly or initially influence gene

301

expression (Figure 1C) (Lösel and Wehling, 2003). A non-genomic effect can be observed in a much

302

shorter time frame (seconds to minutes of exposure to experimental chemical) than genomic effects that

303

are constrained by transcription and translation. These effects are also typically unaffected by

304

actinomycin D or cycloheximide treatment, which inhibit transcription and translation, respectively.

305

Many non-genomic actions are initiated at the level of the cell membrane through interactions

306

with membrane-associated receptors. The fungicides prochloraz and vinclozolin have been shown to

307

directly target the membrane androgen receptor ZIP9, a member of the solute carrier protein family, to

308

block testosterone-driven zinc influx and apoptosis in PC3 prostate cancer cells (Thomas and Dong,

309

2019). Another study has demonstrated that the herbicide atrazine and pesticide trans-nonachlor target the

310

epidermal growth factor receptor (EGFR) and block receptor activation and autophosphorylation

311

(Hardesty et al., 2018). Further protein-ligand docking simulations suggest that trans-nonachlor acts as a

312

competitive antagonist to the EGFR, whereas atrazine blocks the tyrosine kinase activity of the receptor

313

(Hardesty et al., 2018). The insecticide cypermethrin can also inhibit EGFR activity and downstream

314

MAPK activation by interfering with non-classical testosterone signaling in sertoli cells, leading to

315

reduced cell viability and proliferation (Wang et al., 2019).

316

Activation of membrane receptors triggers signal transduction cascades to regulate cellular

317

responses like changes in gene expression, proliferation, and metabolism. The mitogen activated protein

318

kinase pathway (MAPK) is a kinase cascasde activated downstream of membrane receptors for growth

319

factors and cytokines that regulate cell growth, survival, and differentiation (Morrison, 2012). Several

320

studies have shown MAPK pathway activation to be critical in mediating the effects of agricultural

321

chemicals like nonylphenol, atrazine, and cypermethrin on Sertoli cell viability, testosterone production in

322

Leydig cells, and gonadotropin production in cultured gonadotropes, respectively (Choi et al., 2014;

323

Pogrmic-Majkic et al., 2016; F. Li et al., 2018). One study observed reduced membrane associated

324

connexin43 expression and impaired inter- and intracellular signaling in liver progenitor cells treated with

325

either methyoxylchlor or vinclonzin (Babica et al., 2016). This effect was dependent on p38 MAPK

326

activity as well as activation of protein kinase C.

327

The phosphoinositide-3-kinase–protein kinase B/AKT (PI3K/AKT) pathway is another target of

328

agricultural chemicals. The PI3K/AKT pathway is often triggered upstream by activation of membrane

329

receptors targeted by growth factors and cytokines, and plays a critical role in cell proliferation, survival,

330

and metabolism (Hemmings and Restuccia, 2012). Current studies have demonstrated the suppression of

331

this signaling pathway by nonylphenol and ziram in the testes of exposed rats, resulting in the generation

332

of reactive oxygen species and increased apoptosis (Huang et al., 2016; Xie et al., 2018).

333

Another common target of agricultural chemicals is the cyclic adenosine 3′,5′-monophosphate

334

(cAMP) pathway. The second messenger cAMP accumulates in the cell through activation of adenylyl

335

cyclase by membrane associated G-protein coupled receptors (Sassone-Corsi, 2012). In turn, cAMP

336

activates protein kinase A (PKA) to regulate cell metabolism, ion transport, and gene expression

337

(Sassone-Corsi, 2012). Atrazine has been shown to increase cAMP levels in Leydig cells by inhibiting the

338

cAMP specific phosphodiesterase, an enzyme that catalyzes the hydrolysis of cAMP to the inactive 5-

339

AMP (Karmaus and Zacharewski, 2015). Excessive accumulation of cAMP by atrazine increases PKA

340

activity to modulate testosterone production in Leydig cells (Samardzija et al., 2016; Pogrmic-Majkic et

341

al., 2016; Karmaus and Zacharewski, 2015). Other chemicals, like methyoxychlor and its metabolite

342

HTPE, have been shown to inhibit follicle stimulating hormone (FSH) stimulated cAMP production,

343

resulting in reduced estrogen production (Harvey et al., 2015). Another study demonstrated that exposure

344

to a mixture containing organochlorine insecticides impaired cAMP signaling, leading to decreased

345

cAMP mediated post-translational processing of the steroidogenic acute regulatory protein to its active

346

form and reduced steroidogenesis in cultured Leydig cells (Enangue Njembele et al., 2014).

347

EDCs can also influence cell function by targeting membrane associated ion-channels to mediate

348

rapid non-genomic responses. These channels open to form a pore, allowing the passive diffusion of

349

specific ions into the cell that will mediate numerous cellular functions, including the control of electrical

350

excitability and the exocytosis of secreted proteins (Jentsch et al., 2004). Insecticides cypermethrin and

351

DDE as well as glyphosate-based herbicides have been shown to target membrane bound voltage gated

352

calcium channels and/or intracellular ryanodine receptors and increase intracellular calcium

353

concentrations (Tavares et al., 2013; De Liz Oliveira Cavalli et al., 2013; Ye et al., 2017; F. Li et al.,

354

2018). Perturbation in calcium homeostasis by agricultural chemicals has been linked to several

355

physiological effects, including reduced acrosomal integrity and mobility in sperm, increased oxidative

356

damage and decreased viability in Leydig cells, and increased glucose-stimulated insulin secretion from

357

pancreatic β-cells through generation of reactive oxygen species (Tavares et al., 2013, 2015; De Liz

358

Oliveira Cavalli et al., 2013; Chen et al., 2017). Another study demonstrated that cypermethrin induced

359

calcium influx triggered rapid activation of MAPK signaling, leading to increased gonadotropin secretion

360

from cultured pituitary cells (F. Li et al., 2018). Cypermethrin has also been shown to target voltage gated

361

sodium channels in the hypothalamus to increase gonadotropin-releasing hormone pulse frequency (Ye et

362

al., 2017).

363

The non-genomic effects of endocrine disrupting chemicals are not fully understood, and even

364

less is known about those driven by agricultural EDCs. This is partly due to the duration of treatment.

365

Many studies using agrochemicals involve long term treatment periods spanning days to weeks, which

366

make it difficult to discern the rapid non-genomic actions from the slower genomic actions. Use of shorter

367

exposure periods in in vitro culture systems and the inclusion of actinomycin D and cycloheximide will

368

greatly increase our understanding of the non-genomic mechanisms of agricultural EDCs.

369

Hormone Synthesis and Degradation

370

Hormones in the body can be characterized into three main groups depending on their major

371

components: peptide/protein hormones, steroid hormones, and amino acid analogue hormones.

372

Peptide/protein hormones are the most abundant in the body and include gonadotropin-releasing

373

hormone, growth hormone, FSH, luteinizing hormone (LH), and thyroid stimulating hormone (TSH).

374

Important steroid hormones include estrogens, progesterone, and testosterone. Lastly, amino acid

375

analogues include iodothyronines such as thyroxine (T4) and 3,5,3’-triiodothyronine (T3) and amines

376

such as dopamine and serotonin. The synthesis and degradation of peptide/protein, steroid, and thyroid

377

hormones can be negatively affected by EDCs, including agrochemicals. Interfering with hormone action

378

may cause negative downstream effects on reproductive and non-reproductive health.

379

Peptide/protein hormones

380 381

Peptide and protein hormones are composed of short and long amino acid chains, respectively. Synthesis occurs in the nucleus and cytoplasm of secretory cells via gene transcription, translation into the

382

peptide chain, and finally post-translational modifications. After hormones are synthesized and packaged

383

into secretory granules, neural and hormonal signals cause their secretion into extracellular space

384

(Malandrino and Smith, 2018). Some peptide and protein hormones are secreted in pulsatile patterns that

385

may have rhythmic changes that contribute to feedback mechanisms, such as the hypothalamic-pituitary-

386

gonadotropin (HPG) axis, to control other hormone production in the body. Most peptide/protein

387

hormones are soluble in aqueous solvents and do not need carrier proteins for transport throughout the

388

blood stream. Therefore, they are susceptible to rapid protease degradation, leading to a short half-life and

389

duration of action (Malandrino and Smith, 2018). TSH, a glycoprotein hormone that stimulates the

390

production of T4, is susceptible to EDC influence. Perinatal exposure in rats to a glyphosate-based

391

herbicide resulted in decreased TSH levels, decreased gene expression of deiodinases and transporters,

392

and altered other genes regulated by thyroid hormones or involved in thyroid hormone metabolism in

393

male offspring (de Souza et al., 2017). Epidemiological studies of agricultural workers have shown

394

associations between pesticide exposure and levels of FSH and LH (Recio et al., 2005; Cremonese et al.,

395

2017). Extensive evidence in animal studies also indicates that chlorpyrifos treatment alters levels of FSH

396

and LH, in addition to steroid hormones (Li et al., 2019).

397

Steroid Hormones

398

Most steroid hormone synthesis takes place in the adrenal glands, ovary, testes, placenta, and

399

adipose tissue. The synthesis of steroid hormones is known as steroidogenesis. Cholesterol is the pre-

400

cursor for all steroid hormones, and it is transferred across the mitochondrial membrane by steroidogenic

401

acute regulatory protein (StAR). Cholesterol is then converted to pregnenolone in a process that is

402

common to all steroidogenic pathways (Miller and Auchus, 2011). After the production of pregnenolone,

403

the pathway varies depending on the hormone being produced; pregnenolone may undergo various

404

biotransformations to form aldosterone and cortisol in the adrenal gland or progesterone, estradiol, and

405

testosterone in the gonads (Acconcia and Marino, 2018; Auchus, 2014) (Figure 4). Most steroids are

406

hydrophobic in nature so hormones and their precursors can leave steroidogenic cells easily and are not

407

stored (Acconcia and Marino, 2018). After a biological response occurs, hormone secretion ceases which

408

contributes to feedback loops in the body, including the HPG axis and the hypothalamic-pituitary-adrenal

409

(HPA) axis (Acconcia and Marino, 2018). Carrier proteins transport steroid hormones throughout the

410

body and control the proportions of bound and unbound hormones (Leung and Farwell, 2018). Steroid

411

hormones are eliminated through enzymatic reactions followed by transport across the cell membrane.

412

Many of these reactions occur in the liver and include hydroxylation, conjugation, and reduction-

413

oxidation (You, 2004). After metabolites reach circulation, they are readily excreted in both urine and bile

414

as unconjugated or conjugated metabolites, but some may remain in circulation by binding to serum

415

proteins (You, 2004).

416 417

Figure 4: Steroidogenesis. Cholesterol is imported by steroidogenic acute regulatory protein (StAR) and

418

transformed into pregnenolone, which is further converted to adrenal and gonadal hormones.

419

Agrochemical exposure causes detrimental effects on the process of steroidogenesis that occurs in

420

the testis, ovary, and adrenal gland. These effects have been observed in both in vivo and in vitro

421

experimental designs. Alteration of steroidogenesis is a widely studied mode of action of EDCs; thus,

422

Table 2 provides examples of recent studies on the impacts of agrochemicals on steroid hormones.

423

Considering the literature, some common targets of agrochemical EDCs emerge. Multiple chemicals have

424

been shown to affect the synthesis of cholesterol, which is the pre-cursor for all steroid hormones. In the

425

adrenal glands, agrochemicals negatively affect the hormones, genes, and proteins involved in the

426

synthesis of steroid hormones. In the ovary, agrochemicals disrupt the production of estradiol by altering

427

the levels of enzymes and pre-cursor hormones involved in steroidogenesis in both in vivo and in vitro

428

studies. In the testes, agrochemicals impair the synthesis of testosterone, leading to detrimental effects on

429

reproduction. Table 2: Agrochemical effects on steroid hormone synthesis, metabolism, and concentrations Chemical

Mode of Action

Effect

Reference

2,4-D

Decreased serum testosterone levels, testis testosterone levels, Cyp17a1 levels, and total cholesterol levels in Leydig cells

Decreased pregnancy rate and number of pups

(Harada et al., 2016)

Decreased luminal spermatozoa

(Zhang et al., 2017)

Decreased testis, seminal vesicle, and prostate weight

(Pogrmic et al., 2009)

Altered dilation of seminiferous tubules and decreased size of Leydig cells

(Victor-Costa et al., 2010)

Altered steroidogenesis and aromatase activity

(Tinfo et al., 2011)

Decreased percent cell viability in Leydig cells

(Abarikwu et al., 2011)

Decreased levels of estradiol, but increased levels of progesterone in granulosa cells

Impaired reproductive efficiency

(Basini et al., 2012)

Altered levels of steroidogenic enzymes and increased levels of progesterone and testosterone in Leydig cells

Altered steroidogenesis

(Forgacs et al., 2013)

Increased levels of estrogen and androgens in adrenal cells

Interference with androgen synthesis

(Háhn et al., 2016)

Reduced levels of 3β-HSD expression

Decreased testis weight and heterogeneous testis morphology Decreased female fertility

(MartinsSantos et al., 2017) (PogrmicMajkic et al., 2018) (Ji et al., 2019)

Decreased serum testosterone levels atrazine

Decreased steroidogenic enzymes in Leydig cells, decreased serum levels of dihydrotestosterone and testosterone, decreased transport of cholesterol to mitochondria Decreased testosterone levels and increased estradiol levels, decreased protein levels of 3β-hydroxysteroid dehydrogenase (3β-HSD) expression Increased estradiol and progesterone levels in granulosa cells, increased estradiol, estrone, and progesterone levels in adrenal cells Altered levels of steroidogenic enzymes

Reduced levels of steroidogenic enzymes

cypermethrin

Altered levels of steroidogenic enzymes, increased levels of estrogen, cortisol, and aldosterone in adrenal cells

Altered the steroidogenic pathway

Increased levels of steroidogenic enzymes, increased levels of luteinizing hormone, follicle-stimulating hormone, and testosterone

Accelerated onset of puberty in males

Altered levels of steroidogenic enzymes, decreased levels of testosterone, increased levels of estradiol in vivo and in vitro

Altered testis weight over time, decreased germ cells, impaired spermatogenesis and steroidogenesis Decreased testis weight, sperm count, sperm motility, sperm viability, sperm production, and inhibited spermatogenesis Impaired spermatogenesis and decreased sperm count

endosulfan

Decreased steroidogenic enzyme and testosterone levels

fenvalerate

Decreased expression of steroidogenic enzymes and decreased serum and testicular testosterone Decreased levels of corticosterone, testosterone, cholesterol, but increased weight of adrenal glands Altered levels of steroidogenic enzymes, decreased progesterone levels, but increased estradiol levels Increased levels of cholesterol but decreased levels of steroidogenic enzymes in the ovary, decreased levels of cholesterol, but increased levels of steroidogenic enzymes in the adrenal glands, and deceased serum levels of LH, FSH, estradiol, and progesterone Decreased levels of steroidogenic enzymes and levels of testosterone, FSH, and LH

glyphosate and glyphosatebased herbicides lambdacyhalothrin

mancozeb

Decreased levels of steroidogenic enzymes and levels of testosterone

Decreased testosterone and FSH levels

methoxychlor

ziram

Altered levels of steroidogenic enzymes, decreased testosterone levels, increased estradiol levels Decreased levels of steroidogenic enzymes and testosterone Decreased levels of most steroidogenic enzymes and decreased levels of estradiol, testosterone, androstenedione, and progesterone Decreased expression of steroidogenic enzymes, levels of testosterone, and levels of FSH Decreased steroidogenic enzyme, testosterone, and FSH levels

430 431

Amino Acid Hormones

Disrupted function of HPA axis

Decreased ovary weight, altered ovarian histology, and sex ratio of pups Altered ovarian structural degenerations and follicular maturation

Delayed Leydig cell development Decreased testis, epididymis, seminal vesicle, vas deferens, and prostate weight, decreased sperm parameters Increased abnormal sperm morphology and sperm viability Decreased testis weight and disrupted spermatogenesis

(Ye et al., 2017) (Huang and Li, 2014)

(Aly and Khafagy, 2014)

(Zhang et al., 2010) (Pandey and Rudraiah, 2015) (Ren et al., 2018) (R. Ghosh et al., 2018)

(H. Li et al., 2018) (Girish and Reddy, 2018)

(Elsharkawy et al., 2019) (Du et al., 2014)

Decreased testis weight, cauda sperm count, and sperm motility Decreased antral follicle growth

(Aly and Azhar, 2013)

Decreased epididymis and testis weight

(Guo et al., 2017)

Disrupted Leydig cell development

(Xie et al., 2018)

(Basavarajappa et al., 2012)

432

Amino acid analogue hormones develop from amino acids. Specifically, the amines are derived

433

from tyrosine and are secreted from both the thyroid and adrenal medulla (Koibuchi, 2018). The thyroid

434

gland synthesizes the endogenous thyroid hormones T4 and T3, which require dietary iodine. Thyroid

435

hormones are produced extracellularly in the follicular lumen (Koibuchi, 2018). The lumen is filled with

436

the glycoprotein thyroglobulin, which is synthesized within the epithelial cells and secreted into the

437

lumen (Koibuchi, 2018). Iodine is oxidized by the enzyme thyroperoxidase and then binds to tyrosine

438

residues in the thyroglobulin, resulting in the formation of iodotyrosines (Koibuchi, 2018; Kopp et al.,

439

2008). The iodotyrosines are coupled by thyroperoxidase to form T4, which is further converted into T3

440

in target tissues by deiodinases. Thyroid hormones are transported via carrier proteins including thyroid

441

binding globulin and transthyretin. The levels of T4 and T3 are regulated by deiodinases in peripheral

442

tissues and metabolism in the liver (Boas et al., 2012).

443

Although pesticides are generally poor structural mimics for thyroid hormones, they have been

444

shown to exert significant effects on thyroid hormone synthesis, concentrations, and carrier proteins.

445

Nonylphenol, a now-banned surfactant previously used in pesticide formulations, inhibits thioperoxides

446

necessary for the synthesis of T4. (Schmutzler et al., 2004) The triazole herbicide amitrole has been

447

shown to increase the transcription of thyroglobulin gene, while decreasing the uptake of iodide (Pan

448

Hongmei et al., 2011). Further, DDT has been shown to down-regulate the iodine-accumulation function

449

of follicular thyrocytes by suppressing sodium/iodide symporter synthesis and disrupting regular thyroid

450

function in male rats (Yaglova and Yaglov, 2015). Multiple human epidemiological studies have

451

indicated an association between disrupted thyroid hormone levels and pesticide use (Cremonese et al.,

452

2017; Recio et al., 2005; Piccoli et al., 2016; Blanco-Muñoz et al., 2016; Lacasaña et al., 2010).

453

Malathion, nonylphenol, ioxynil, and pentachlorophenol exposure decreased T3 binding to Japanese quail

454

transthyretin (TTR), one of the carrier proteins, with no observed interaction with the ligand binding

455

domain of thyroid receptors (Ishihara, Nishiyama, et al., 2003). Dicofol was shown to alter T3 binding to

456

bullfrog TTR in a biphasic manner, with low doses of dicofol increasing binding and high doses

457

inhibiting binding (Ishihara, Sawatsubashi, et al., 2003). Recently, enhanced binding of T4 to human TTR

458

has been demonstrated in the presence of organophosphate triesters, which are typically used as flame

459

retardants, but have similar structures to organophosphate pesticides (Hill et al., 2018).

460

Epigenetics

461

Epigenetics encompasses changes in gene expression without changing DNA sequences. The

462

epigenome, which consists of all chemical and structural marks that control the accessibly of the genome,

463

is highly sensitive to environmental conditions including nutrition and stress as well as chemical

464

exposures. Changes to the epigenome can be heritable and can manifest in health impacts and disease

465

long after the exposure has ended, even multiple generations after the exposure occurred (Rattan and

466

Flaws, 2019). Observed transgenerational effects of environmental chemicals were one of the first

467

indications that EDCs could alter the epigenome. For example, vinclozolin and methoxylchlor exposure

468

during pregnancy have been shown to decrease fertility in male descendants for up to four generations

469

(Anway et al., 2005). The effects of these chemicals on germ cell genomes make these outcomes

470

heritable. Development is also a particularly sensitive window of exposure due to the extensive genetic

471

programming that occurs in this time period. Exposures during periods of development can alter the

472

epigenome to increase susceptibility to disease later in life, after the exposure has ended. This forms the

473

mechanistic basis for the developmental origins of health and disease (DOHaD) (Heindel and

474

Vandenberg, 2015).

475

In the past 15 years, mechanistic understanding of how EDCs can alter the epigenome has

476

significantly improved and three mechanisms of epigenetic modification have been identified in recent

477

EDC literature. DNA methylation and histone modification involve structural modifications to chromatin

478

that control the accessibility of genes to transcription factors. Non-coding RNAs, particularly micro-

479

RNAs (miRNAs), from the non-translated genome also regulate the epigenome and can be altered by

480

EDCs. These mechanisms are illustrated in Figure 5.

481 482

Figure 5: Mechanisms of epigenetic modifications. Chromosomes are composed of chromatin that is

483

wrapped around proteins called histone. Modifications to histone tails and direct DNA methylation can

484

control transcriptional access to DNA without modifying DNA sequence. Non-coding RNAs from the

485

untranslated genome also regulate transcription.

486

DNA Methylation

487

Modification of DNA with methyl groups is the most well-studied epigenetic modification.

488

Methylation of chromatin occurs at the 5-carbon position of cytosine to form 5-methylcytosine (5mC).

489

The cytosine is typically adjacent to guanine in the 5' → 3' direction (CpG). CpG sites are less represented

490

in the genome than expected by chance and appear in clusters known as CpG island in promoter regions

491

of genes. Methylation is generally a silencing mark, which acts by blocking access of transcription factors

492

to genes. DNA methyltransferases (DNMTs) are responsible for methylation and demethylation of CpG

493

sites. DNMT1 performs maintenance of methyl marks during replication to preserve epigenetic marks

494

through mitosis, whereas DNMT3a and DNMT3b perform de novo methylation.

495

Extensive reprogramming of methylation marks occurs during germ cell development and early

496

embryonic development (Reik et al., 2001). In both cases, methylation marks are removed and

497

remethylated, providing opportunities for disruption by environmental chemicals. Germ cell

498

modifications are heritable, which can lead to the preservation of altered methylation marks in offspring

499

and later generations, leading to transgenerational effects. For example, methoxychlor increased the

500

incidence of kidney disease, ovarian disease, and obesity as well as decreased sperm concentrations in F3

501

mice following prenatal exposure by altering methylation patterns (Manikkam et al., 2014; Stouder and

502

Paoloni-Giacobino, 2011). Gestational exposure of the F1 generation to the herbicide atrazine led to

503

increased testis disease, lean phenotype, early puberty in females, and behavioral changes in the F3

504

generation (McBirney et al., 2017). Differently methylated regions in sperm were identified for each

505

differently-exposed generation (F1 as embryo; F2 as germ cells; F3 no direct exposure), with consistently

506

differently methylated regions in the F3 generation corresponding to observation of lean phenotype

507

(McBirney et al., 2017). In sperm from F3 rats descended from F0 rats exposed gestationally to

508

vinclozolin, differentially methylated regions were associated with testis disease, prostate disease, and

509

kidney disease (Nilsson et al., 2018). In another transgenerational study of gestationally exposed rats,

510

vinclozolin induced differential methylation in sperm and brain in F1 and F3 generations (Gillette et al.,

511

2018). Adult exposure to methyl-parathion has been shown to decrease sperm quality in mice

512

immediately following exposure due to increased methylation at promoter regions of DNA repair genes as

513

well as global hypomethylation (Hernandez-Cortes et al., 2018). Although the heritability of these

514

observed changes in male germ cells was not investigated, effects due to adult exposures can be inherited

515

through the germ line. One study has examined methylation in sperm from humans exposed to TCDD,

516

finding differentially methylated regions associated with peripubertal serum TCDD measurements

517

(Pilsner et al., 2018).

518

Alternations in reprogramming during embryonic development can cause immediate toxicity or

519

increase adult diseases. The insecticide fipronil is composed of an enantiomeric mixture, one of which is

520

significantly more developmentally toxic to zebrafish. The toxic S-enantiomer hyper-methylated CpG

521

regions during development, leading to disruption of signaling pathways including MAPK, Wnt, and

522

hedgehog (Qian et al., 2017). Methoxychlor induced adult ovarian dysfunction in neonatally exposed rats.

523

Methylation analysis showed hypermethylation in ERβ promoter regions and increased Dnmt3b

524

expression (Zama and Uzumcu, 2009).

525

Histone Modification

526

DNA is organized by winding around proteins called histones to form chromatin that is compact

527

enough to fit in the nucleus of a cell (Strahl and Allis, 2000). DNA must unwind from the histones to be

528

accessible to transcription factors and thus gene expression is regulated by the structure of the histone.

529

Like methylation of DNA, histones can be modified with chemical tags that limit the ability of DNA to be

530

transcribed. Methylation and acetylation are two of the most common forms of post-transcriptional

531

modifications on histone tails that can limit the accessibility of genes.

532

Atrazine treatment of mice leads to heritable transgenerational effects on sperm mediated through

533

histone modifications. In male offspring following developmental exposure, trimethylation of the fourth

534

lysine on histone 3 (H3K4me3) was altered in promoters of key pluripotency-associated genes in sperm

535

(Hao et al., 2016). Furthermore, these marks were preserved in the F3 generation (Hao et al., 2016).

536

These marks were compared to a previous study on vinclozolin exposed mice, showing overlap in

537

differently methylated regions indicative of regions of the genome sensitive to epigenetic modifications

538

by environmental chemicals (Guerrero-Bosagna et al., 2012). Another study on atrazine exposure in adult

539

male mice found disruption of meiosis in sperm with alterations in H3K4me3 marks as well (Gely-Pernot

540

et al., 2015).

541

Recent studies have examined changes in histone modification and other epigenetic modification

542

as first steps in elucidating the mechanisms of action of less widely studied agrochemicals. The fungicide

543

carbendazim has been shown to alter spermatogenesis and steroidogenesis in mouse testes. Epigenetic

544

analysis has shown alterations in trimethylation at lysine 27 on H3 (H3K27me3) as well as changes in

545

DNA methylation (Liu et al., 2019). The organophosphate pesticide chlorpyrifos has been shown to

546

increase expression of histone deacetylase 1 (HDAC1) in mammary tissue of rats (Ventura et al., 2019).

547

The estrogenic pesticide endosulfan has also been shown to increase histone deacetylase (HDAC) and

548

DNMT expression in MCF-7 cells (K. Ghosh et al., 2018). Although each of these studies shows that

549

EDCs can act through epigenetic pathways, the detailed mechanisms through which these chemicals alter

550

expression of epigenetic regulatory proteins are largely unknown.

551

Non-Coding RNA Expression Non-coding RNAs are nucleic acids transcribed from sections of DNA that do not code for

552 553

proteins, once known as “junk” DNA. Non-coding RNAs, including small microRNAs (miRNAs), are

554

important regulators of post-transcriptional gene expression by binding to untranslated regions of mRNAs

555

to block protein translation. MiRNAs can target enzymes responsible for epigenetic marks such as

556

DNMTs and HDACs. In addition, epigenetic marks control the expression of miRNAs (Yao et al., 2019).

557

MiRNAs have been implicated in various human diseases, especially cancers (Tüfekci et al., 2014), but

558

the role of miRNAs and other non-coding RNAs is less widely studied in relation to the endocrine

559

system. However, several miRNAs that directly target ESR1 have been identified (Bhat-Nakshatri et al.,

560

2009).

561 562

Developmental exposure to atrazine in zebrafish significantly altered both human and zebrafish miRNA expression. Further analysis revealed that the altered miRNAs are associated with epigenetic

563

regulation of carcinogenesis, the cell cycle, and cell signaling (Wirbisky et al., 2016). In MCF-7 breast

564

cancer cells, DDT induced a distinct pattern of miRNA expression compared to vehicle control, estradiol,

565

or bisphenol A (BPA) (Tilghman et al., 2012). These preliminary studies show that EDCs can alter

566

miRNA expression to epigenetically regulate biological processes, but further investigations are necessary

567

for deeper understanding of the mechanism of action of EDCs on these miRNAs and identify miRNA

568

targets.

569

Conclusions

570

As this review illustrates, environmental chemicals can act as EDCs through a variety of

571

mechanisms. The diversity of pathways and precision of biological hormone actions in the endocrine

572

system makes it particularly susceptible to disruption by exogenous agents. In addition, the wide range of

573

possible phenotypes and endpoints makes integration of studies on EDCs to understand mechanisms a

574

difficult task. However, the requirement in the European Union of evidence of a plausible mode of action

575

for EDC regulation underscores the importance of mechanistic studies and analyses of these compounds

576

(Solecki et al., 2017).

577

Agrochemicals represent a significant class of EDCs that humans and animals around the world

578

are exposed to constantly. With such diversity of structures and uses, examples of agrochemicals that

579

exhibit each of the major mechanisms of endocrine disruption have been identified. Much like other

580

environmental contaminants, legacy pesticides continue to present a threat to human health, while newer

581

replacement chemicals are less well understood in terms of their endocrine disrupting potential.

582

Furthermore, newer chemicals with shorter half-lives and different routes of exposure pose challenges to

583

understanding their mechanisms of action, much like other classes of replacement EDCs.

584

Given the diversity of mechanisms through which natural hormones and EDCs act, it is vital that

585

21st century toxicology incorporates the principles of endocrinology into assessment of the safety of

586

chemicals in our environment (Schug et al., 2016; Vandenberg et al., 2019, 2013, 2012). Future studies

587

should recognize the prevalence of non-monotonic dose response curves and the importance of low dose

588

studies. In addition, mechanistic studies are needed on newer chemicals on the market and suspected

589

EDCs; legacy chemicals and controversial EDCs with lots of public interest have received most of the

590

scientific attention to date. Improvements in assays and techniques to elucidate EDC mechanisms of

591

action for computational, in vitro, and whole animal studies will facilitate interdisciplinary cooperation to

592

identify additional unstudied mechanisms of action and help regulators and the public understand the

593

various modes of action through which EDCs operate.

594

Acknowledgements

595

Thank you to Katie Chiang for Figure 5. This work was supported by Endocrine, Developmental, and

596

Reproductive Toxicology Training Grant NIH T32 ES 007326 and NIH R01 ES028661.

597

Competing interests

598

The authors declare no competing interests.

599 600

References

601 602

Abarikwu, S.O. et al. (2011) Atrazine induces transcriptional changes in marker genes associated with steroidogenesis in primary cultures of rat Leydig cells. Toxicol. Vitr., 25, 1588–1595.

603 604 605

Abbott, B.D. et al. (1996) Rapid distribution of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) to embryonic tissues in C57BL/6N mice and correlation with palatal uptake in vitro. Toxicol. Appl. Pharmacol., 141, 256–263.

606 607 608

Acconcia, F. and Marino, M. (2018) Principles of Endocrinology and Hormone Action. In, Belfiore,A. and LeRoith,D. (eds), Principles of Endocrinology and Hormone Action, Endocrinology. Springer International Publishing, pp. 43–72.

609 610

Aly, H.A.A. and Azhar, A.S. (2013) Methoxychlor induced biochemical alterations and disruption of spermatogenesis in adult rats. Reprod. Toxicol., 40, 8–15.

611 612

Aly, H.A.A. and Khafagy, R.M. (2014) Taurine reverses endosulfan-induced oxidative stress and apoptosis in adult rat testis. Food Chem. Toxicol., 64, 1–9.

613 614

Anway, M.D. et al. (2005) Epigenetic transgenerational actions of endocrine disruptors and male fertility. Science, 308, 1466–9.

615 616

Anway, M.D. and S.K.M. (2015) Transgenerational Effects of the Endocrine Disruptor Vinclozolin on the Prostate Transcriptome and Adult Onset Disease. Prostate, 1, 517–529.

617 618

Auchus, R.J. (2014) Human Steroid Biosynthesis. Knobil Neill’s Physiol. Reprod. Two-Volume Set, 1, 295–312.

619 620

Ayisi, C.L. et al. (2018) Genes, transcription factors and enzymes involved in lipid metabolism in fin fish. Agri Gene, 7, 7–14.

621 622

Babica, P. et al. (2016) Methoxychlor and vinclozolin induce rapid changes in intercellular and intracellular signaling in liver progenitor cells. Toxicol. Sci., 153, 174–185.

623 624

Balaguer, P. et al. (2019) Mechanisms of endocrine disruption through nuclear receptors and related pathways. Curr. Opin. Endocr. Metab. Res., 7, 1–8.

625 626

Basavarajappa, M.S. et al. (2012) Methoxychlor induces atresia by altering Bcl2 factors and inducing caspase activity in mouse ovarian antral follicles in vitro. Reprod. Toxicol., 34, 545–551.

627 628

Basini, G. et al. (2012) Atrazine disrupts steroidogenesis, VEGF and NO production in swine granulosa cells. Ecotoxicol. Environ. Saf., 85, 59–63.

629 630

Beischlag, T. V. et al. (2008) The aryl hydrocarbon receptor complex and the control of gene expression. Crit. Rev. Eukaryot. Gene Expr., 18, 207–250.

631 632

Bertuloso, B.D. et al. (2015) Tributyltin chloride leads to adiposity and impairs metabolic functions in the rat liver and pancreas. Toxicol. Lett., 235, 45–59.

633 634

Bhat-Nakshatri, P. et al. (2009) Estradiol-regulated microRNAs control estradiol response in breast cancer cells. Nucleic Acids Res., 37, 4850–4861.

635 636

van Birgelen, A.P. (1998) Hexachlorobenzene as a possible major contributor to the dioxin activity of human milk. Environ. Health Perspect., 106, 683–688.

637 638

Blanco-Muñoz, J. et al. (2016) Association between organochlorine pesticide exposure and thyroid hormones in floriculture workers. Environ. Res., 150, 357–363.

639 640

Boas, M. et al. (2012) Thyroid effects of endocrine disrupting chemicals. Mol. Cell. Endocrinol., 355, 240–248.

641 642

Brander, S.M. et al. (2016) Pyrethroid pesticides as endocrine disruptors: Molecular mechanisms in vertebrates with a focus on fishes. Environ. Sci. Technol., 50, 8977–8992.

643 644

Brzozowski, A.M. et al. (1997) Molecular basis of agonism and antagonism in the oestrogen receptor. Nature, 389, 753–758.

645

Carson, R. (1962) Silent Spring Houghton Mifflin Company.

646 647 648

Chamorro-García, R. et al. (2018) Effects of Perinatal Exposure to Dibutyltin Chloride on Fat and Glucose Metabolism in Mice, and Molecular Mechanisms, in Vitro. Environ. Health Perspect., 126, 057006.

649 650

Chen, Y.W. et al. (2017) Tributyltin exposure at noncytotoxic doses dysregulates pancreatic β-cell function in vitro and in vivo. Arch. Toxicol., 91, 3135–3144.

651 652 653

Choi, M.-S. et al. (2014) Nonylphenol-induced apoptotic cell death in mouse TM4 Sertoli cells via the generation of reactive oxygen species and activation of the ERK signaling pathway. J. Appl. Toxicol., 34, 628–636.

654 655

Christiansen, S. et al. (2008) Combined exposure to anti-androgens causes markedly increased frequencies of hypospadias in the rat. Int. J. Androl., 31, 241–247.

656 657

Cremonese, C. et al. (2017) Occupational exposure to pesticides, reproductive hormone levels and sperm quality in young Brazilian men. Reprod. Toxicol., 67, 174–185.

658 659

Cummings, A.M. (1997) Methoxychlor as a Model for Environmental Estrogens. Crit. Rev. Toxicol., 27, 367–379.

660 661 662

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

663 664

Davis-Dao, C. et al. (2012) Shorter androgen receptor CAG repeat lengths associated with cryptorchidism risk among hispanic white boys. J. Clin. Endocrinol. Metab., 97, 393–399.

665 666

Delfosse, V. et al. (2015) A structural perspective on nuclear receptors as targets of environmental compounds. Acta Pharmacol. Sin., 36, 88–101.

667 668

Du, G. et al. (2010) Assessing hormone receptor activities of pyrethroid insecticides and their metabolites in reporter gene assays. Toxicol. Sci., 116, 58–66.

669 670

Du, X. et al. (2014) Perinatal exposure to low-dose methoxychlor impairs testicular development in C57BL/6 mice. PLoS One, 9, 1–10.

671 672

Elferinks, J. et al. (1990) Protein-DNA Interactions at a Dioxin-responsive Enhancer. J. Biol. Chem., 265, 6575–6580.

673 674

Elsharkawy, E.E. et al. (2019) Mancozeb impaired male fertility in rabbits with trials of glutathione detoxification. Regul. Toxicol. Pharmacol., 105, 86–98.

675 676

Enangue Njembele, A.N. et al. (2014) In Vitro Exposure of Leydig Cells to an Environmentally Relevant Mixture of Organochlorines Represses Early Steps of Steroidogenesis1. Biol. Reprod., 90, 1–10.

677 678

Eroschenko, V.P. et al. (1996) Estradiol or methoxychlor stimulates estrogen receptor (ER) expression in uteri. Reprod. Toxicol., 10, 265–271.

679 680

Forgacs, A.L. et al. (2013) Triazine Herbicides and Their Chlorometabolites Alter Steroidogenesis in BLTK1 Murine Leydig Cells. Toxicol. Sci., 134, 155–167.

681 682

Gaido, K.W. et al. (2000) Interaction of methoxychlor and related compounds with estrogen receptor alpha and beta, and androgen receptor: structure-activity studies. Mol. Pharmacol., 58, 852–858.

683 684

Gely-Pernot, A. et al. (2015) The epigenetic processes of meiosis in male mice are broadly affected by the widely used herbicide atrazine. BMC Genomics, 16, 1–22.

685 686 687

Ghassabian, A. and Trasande, L. (2018) Disruption in thyroid signaling pathway: A mechanism for the effect of endocrine-disrupting chemicals on child neurodevelopment. Front. Endocrinol. (Lausanne)., 9.

688 689

Ghisari, M. et al. (2015) Effects of currently used pesticides and their mixtures on the function of thyroid hormone and aryl hydrocarbon receptor in cell culture. Toxicol. Appl. Pharmacol., 284, 292–303.

690 691

Ghosh, K. et al. (2018) The persistent organochlorine pesticide endosulfan modulates multiple epigenetic regulators with oncogenic potential in MCF-7 cells. Sci. Total Environ., 624, 1612–1622.

692 693

Ghosh, R. et al. (2018) Antigonadal and endocrine-disrupting activities of lambda cyhalothrin in female rats and its attenuation by taurine. Toxicol. Ind. Health, 34, 146–157.

694

Gillette, R. et al. (2018) Passing experiences on to future generations: endocrine disruptors and

695

transgenerational inheritance of epimutations in brain and sperm. Epigenetics, 13, 1106–1126.

696 697 698

Girish, B.P. and Reddy, P.S. (2018) Forskolin ameliorates mancozeb-induced testicular and epididymal toxicity in Wistar rats by reducing oxidative toxicity and by stimulating steroidogenesis. J. Biochem. Mol. Toxicol., 32.

699 700

Gore, A.C. et al. (2015) Executive Summary to EDC-2: The Endocrine Society’s Second Scientific Statement on Endocrine-Disrupting Chemicals. Endocr. Rev., 36, E1–E150.

701 702

Gray, L.E. et al. (1994) Developmental Effects of an Environmental Antiandrogen: The Fungicide Vinclozolin Alters Sex Differentiation of the Male Rat. Toxicol. Appl. Pharmacol., 129, 46–52.

703 704

Grün, F. et al. (2006) Endocrine-disrupting organotin compounds are potent inducers of adipogenesis in vertebrates. Mol. Endocrinol., 20, 2141–2155.

705 706

Guerrero-Bosagna, C. et al. (2012) Epigenetic transgenerational inheritance of vinclozolin induced mouse adult onset disease and associated sperm epigenome biomarkers. Reprod. Toxicol., 34, 694–707.

707

Guo, X. et al. (2017) Ziram delays pubertal development of rat Leydig cells. Toxicol. Sci., 160, 329–340.

708 709

Háhn, J. et al. (2016) Development of a combined method to assess the complex effect of atrazine on sex steroid synthesis in H295R cells. Chemosphere, 154, 507–514.

710 711 712

Hao, C. et al. (2016) Exposure to the widely used herbicide atrazine results in deregulation of global tissue-specific RNA transcription in the third generation and is associated with a global decrease of histone trimethylation in mice. Nucleic Acids Res., 44, 9784–9802.

713 714

Harada, Y. et al. (2016) PPARα-dependent cholesterol/testosterone disruption in Leydig cells mediates 2,4-dichlorophenoxyacetic acid-induced testicular toxicity in mice. Arch. Toxicol., 90, 3061–3071.

715 716

Hardesty, J.E. et al. (2018) Epidermal growth factor receptor signaling disruption by endocrine and metabolic disrupting chemicals. Toxicol. Sci., 162, 622–634.

717 718 719

Harvey, C.N. et al. (2015) Methoxychlor and its metabolite HPTE inhibit cAMP production and expression of estrogen receptors α and β in the rat granulosa cell in vitro. Reprod. Toxicol., 51, 72– 78.

720 721

Heindel, J.J. and Vandenberg, L.N. (2015) Developmental Origins of Health and Disease: A Paradigm for Understanding Disease Etiology and Prevention. Curr Opin Pediatr, 27, 248–253.

722 723

Heldring, N. and Pike, A. (2007) Estrogen receptors: how do they signal and what are their targets. Physiol. Rev., 905–931.

724 725

Hemmings, B.A. and Restuccia, D.F. (2012) PI3K-PKB / Akt Pathway. Cold Spring Harb Perspect Biol, 1–4.

726 727

Hernandez-Cortes, D. et al. (2018) Epigenetic modulation of Nrf2 and Ogg1 gene expression in testicular germ cells by methyl parathion exposure. Toxicol. Appl. Pharmacol., 346, 19–27.

728 729 730

Hewitt, S.C. and Korach, K.S. (2011) Estrogenic activity of bisphenol A and 2,2-bis(p-Hydroxyphenyl)1,1,1-trichloroethane (HPTE) demonstrated in mouse uterine gene profiles. Environ. Health Perspect., 119, 63–70.

731 732

Hill, K.L. et al. (2018) Organophosphate triesters and selected metabolites enhance binding of thyroxine to human transthyretin in vitro. Toxicol. Lett., 285, 87–93.

733

Hosokawa, S. et al. (1993) The affinity of procymidone to androgen receptor in rats and mice. J. Toxicol.

734

Sci., 18, 83–93.

735 736

Huang, C. and Li, X. (2014) Maternal cypermethrin exposure during the perinatal period impairs testicular development in C57BL male offspring. PLoS One, 9.

737 738

Huang, W. et al. (2016) Nonylphenol induced apoptosis and autophagy involving the Akt/mTOR pathway in prepubertal Sprague-Dawley male rats in vivo and in vitro. Toxicology, 373, 41–53.

739 740 741

Ishihara, A., Sawatsubashi, S., et al. (2003) Endocrine disrupting chemicals: Interference of thyroid hormone binding to transthyretins and to thyroid hormone receptors. Mol. Cell. Endocrinol., 199, 105–117.

742 743 744

Ishihara, A., Nishiyama, N., et al. (2003) The effect of endocrine disrupting chemicals on thyroid hormone binding to Japanese quail transthyretin and thyroid hormone receptor. Gen. Comp. Endocrinol., 134, 36–43.

745 746

Jentsch, T.J. et al. (2004) Ion channels: Function unravelled by dysfunction. Nat. Cell Biol., 6, 1039– 1047.

747 748

Ji, C. et al. (2019) Enantioselectivity in endocrine disrupting effects of four cypermethrin enantiomers based on in vitro models. Chemosphere, 220, 766–773.

749 750

Judson, R.S. et al. (2018) New approach methods for testing chemicals for endocrine disruption potential. Curr. Opin. Toxicol., 9, 40–47.

751 752

Kalinina, T.S. et al. (2017) Expression of hormonal carcinogenesis genes and related regulatory microRNAs in uterus and ovaries of DDT-treated female rats. Biochem., 82, 1118–1128.

753 754

Karmaus, A.L. and Zacharewski, T.R. (2015) Atrazine-mediated disruption of steroidogenesis in BLTK1 murine leydig cells. Toxicol. Sci., 148, 544–554.

755

Kelce, W.R. et al. (1995) Androgen Receptor Antagonist. 4, 1054–1057.

756 757

Knebel, C. et al. (2019) Pregnane X receptor mediates steatotic effects of propiconazole and tebuconazole in human liver cell lines. Arch. Toxicol., 93, 1311–1322.

758 759

Knebel, C. et al. (2018) Propiconazole is an activator of AHR and causes concentration additive effects with an established AHR ligand. Arch. Toxicol., 92, 3471–3486.

760 761 762

Koibuchi, N. (2018) Principles of Endocrinology and Hormone Action. In, Belfiore,A. and LeRoith,D. (eds), Principles of Endocrinology and Hormone Action, Endocrinology. Springer International Publishing, pp. 73–81.

763 764

Kopp, P. et al. (2008) Pendred syndrome and iodide transport in the thyroid. Trends Endocrinol. Metab., 19, 260–268.

765 766

Kuiper, G.G.J.M. et al. (1998) Interaction of estrogenic chemicals and phytoestrogens with estrogen receptor β. Endocrinology, 139, 4252–4263.

767 768

Lacasaña, M. et al. (2010) Association between organophosphate pesticides exposure and thyroid hormones in floriculture workers. Toxicol. Appl. Pharmacol., 243, 19–26.

769 770

Lambright, C. (2000) Cellular and Molecular Mechanisms of Action of Linuron: An Antiandrogenic Herbicide that Produces Reproductive Malformations in Male Rats. Toxicol. Sci., 56, 389–399.

771 772

Legler, J. et al. (1999) Development of a stably transfected estrogen receptor-mediated luciferase reporter gene assay in the human T47D breast cancer cell line. Toxicol. Sci., 48, 55–66.

773 774

Leung, A.M. and Farwell, A.P. (2018) The Endocrine System. In, Wylie and Churchill-Davidsons: A Practice of Anesthesia, Seventh Edition., pp. 3–25.

775 776

Li, F. et al. (2018) Pyrethroid Insecticide Cypermethrin Modulates Gonadotropin Synthesis via Calcium Homeostasis and ERK1/2 Signaling in LβT2 Mouse Pituitary Cells. Toxicol. Sci., 162, 43–52.

777 778

Li, H. et al. (2018) Lambda-cyhalothrin delays pubertal Leydig cell development in rats. Environ. Pollut., 242, 709–717.

779 780

Li, J. et al. (2019) Chlorpyrifos induces metabolic disruption by altering levels of reproductive hormones. J. Agric. Food Chem., 67, 10553–10562.

781 782

Li, X. et al. (2012) Triflumizole is an obesogen in mice that acts through peroxisome proliferator activated receptor gamma (PPARγ). Environ. Health Perspect., 120, 1720–1726.

783 784

Liu, J. et al. (2019) Low dose carbendazim disrupts mouse spermatogenesis might Be through estrogen receptor related histone and DNA methylation. Ecotoxicol. Environ. Saf., 176, 242–249.

785 786

De Liz Oliveira Cavalli, V.L. et al. (2013) Roundup disrupts male reproductive functions by triggering calcium-mediated cell death in rat testis and Sertoli cells. Free Radic. Biol. Med., 65, 335–346.

787 788

Long, M. et al. (2003) Effects of currently used pesticides in the AhR-CALUX assay: Comparison between the human TV101L and the rat H4IIE cell line. Toxicology, 194, 77–93.

789 790

Lösel, R. and Wehling, M. (2003) Nongenomic actions of steroid hormones. Nat. Rev. Mol. Cell Biol., 4, 46–56.

791 792

Lv, X. et al. (2017) Effects of triazole fungicides on androgenic disruption and CYP3A4 enzyme activity. Environ. Pollut., 222, 504–512.

793 794 795

Malandrino, N. and Smith, R.J. (2018) Synthesis, Secretion, and Transport of Peptide Hormones. In, Belfiore,A. and LeRoith,D. (eds), Principles of Endocrinology and Hormone Action. Springer International Publishing, pp. 29–42.

796 797 798

Maness, S.C. et al. (1998) Inhibition of androgen receptor-dependent transcriptional activity by DDT isomers and methoxychlor in HepG2 human hepatoma cells. Toxicol. Appl. Pharmacol., 151, 135– 142.

799 800

Manikkam, M. et al. (2014) Pesticide methoxychlor promotes the epigenetic transgenerational inheritance of adult-onset disease through the female germline. PLoS One, 9.

801 802

Maradonna, F. and Carnevali, O. (2018) Lipid metabolism alteration by endocrine disruptors in animal models: An overview. Front. Endocrinol. (Lausanne)., 9, 1–14.

803 804

Martins-Santos, E. et al. (2017) Persistent testicular structural and functional alterations after exposure of adult rats to atrazine. Reprod. Toxicol., 73, 201–213.

805 806

McBirney, M. et al. (2017) Atrazine induced epigenetic transgenerational inheritance of disease, lean phenotype and sperm epimutation pathology biomarkers.

807 808

Miller, W.L. and Auchus, R.J. (2011) The molecular biology, biochemistry, and physiology of human steroidogenesis and its disorders. Endocr. Rev., 32, 81–151.

809 810

Miret, N. V. et al. (2019) Impact of endocrine disruptor hexachlorobenzene on the mammary gland and breast cancer: The story thus far. Environ. Res., 173, 330–341.

811

Monosson, E. et al. (1999) Peripubertal exposure to the antiandrogenic fungicide, vinclozolin, delays

812 813

puberty, inhibits the development of androgen-dependent tissues, and alters androgen receptor function in the male rat. Toxicol. Ind. Health, 15, 65–79.

814

Morrison, D.K. (2012) MAP Kinase Pathways. Cold Spring Harb. Perspect. Biol., 4, a011254–a011254.

815 816

Mouritsen, A. et al. (2013) Androgen receptor CAG repeat length is associated with body fat and serum SHBG in boys: A prospective cohort study. J. Clin. Endocrinol. Metab., 98, 605–609.

817 818 819

Nebert, D.W. (2017) Aryl hydrocarbon receptor (AHR): “pioneer member” of the basic-helix/loop/helix per-Arnt-sim (bHLH/PAS) family of “sensors” of foreign and endogenous signals. Prog. Lipid Res., 67, 38–57.

820 821

Nenonen, H.A. et al. (2011) Non-linear association between androgen receptor cag repeat length and risk of male subfertility - a meta-analysis. Int. J. Androl., 34, 327–332.

822 823

Neschen, S. et al. (2007) N-3 Fatty Acids Preserve Insulin Sensitivity in Vivo in a Peroxisome Proliferator-Activated Receptor-Α-Dependent Manner. Diabetes, 56, 1034–1041.

824 825

Nilsson, E. et al. (2018) Vinclozolin induced epigenetic transgenerational inheritance of pathologies and sperm epimutation biomarkers for specific diseases. PLoS One, 13, 1–29.

826 827 828

Nishimura, N. et al. (2002) Immunohistochemical localization of thyroid stimulating hormone induced by a low oral dose of 2,3,7,8-tetrachlorodibenzo-p-dioxin in female Sprague-Dawley rats. Toxicology, 171, 73–82.

829 830

Ohtake, F. et al. (2011) Cross-talk of dioxin and estrogen receptor signals through the ubiquitin system. J. Steroid Biochem. Mol. Biol., 127, 102–107.

831

Ohtake, F. et al. (2007) Dioxin receptor is a ligand-dependent E3 ubiquitin ligase. Nature, 446, 562–566.

832 833

Ohtake, F. et al. (2003) Modulation of oestrogen receptor signalling by association with the activated dioxin receptor. Nature, 423, 545–550.

834 835

Ostby, J. et al. (1999) The fungicide procymidone alters sexual differentiation in the male rat by acting as an androgen-receptor antagonist in vivo and in vitro. Toxicol. Ind. Health, 15, 80–93.

836 837

Pan Hongmei et al. (2011) The effects of amitrole on thyroglobulin and iodide uptake in FRTL-5 cells. Toxicol. Ind. Health, 27, 187–192.

838 839

Pandey, A. and Rudraiah, M. (2015) Analysis of endocrine disruption effect of Roundup® in adrenal gland of male rats. Toxicol. Reports, 2, 1075–1085.

840 841

Piccoli, C. et al. (2016) Pesticide exposure and thyroid function in an agricultural population in Brazil. Environ. Res., 151, 389–398.

842 843

Pike, A.C.W. et al. (1999) Structure of the ligand-binding domain of oestrogen receptor beta in the presence of a partial agonist and a full antagonist. EMBO J., 18, 4608–4618.

844 845

Pilsner, J.R. et al. (2018) Peripubertal serum dioxin concentrations and subsequent sperm methylome profiles of young Russian adults. Reprod. Toxicol., 78, 40–49.

846 847

Pogrmic-Majkic, K. et al. (2016) Atrazine activates multiple signaling pathways enhancing the rapid hCG-induced androgenesis in rat Leydig cells. Toxicology, 368–369, 37–45.

848 849 850

Pogrmic-Majkic, K. et al. (2018) Atrazine suppresses FSH-induced steroidogenesis and LH-dependent expression of ovulatory genes through PDE-cAMP signaling pathway in human cumulus granulosa cells. Mol. Cell. Endocrinol., 461, 79–88.

851 852

Pogrmic, K. et al. (2009) Atrazine oral exposure of peripubertal male rats downregulates steroidogenesis gene expression in leydig cells. Toxicol. Sci., 111, 189–197.

853 854

Qian, Y. et al. (2017) Fipronil-induced enantioselective developmental toxicity to zebrafish embryolarvae involves changes in DNA methylation. Sci. Rep., 7, 1–11.

855 856

Rana, K. et al. (2014) Human androgen deficiency: Insights gained from androgen receptor knockout mouse models. Asian J. Androl., 16, 169–177.

857 858

Rattan, S. and Flaws, J.A. (2019) The epigenetic impacts of endocrine disruptors on female reproduction across generations†. Biol. Reprod., 0, 1–10.

859 860

Recio, R. et al. (2005) Pesticide exposure alters follicle-stimulating hormone levels in Mexican agricultural workers. Environ. Health Perspect., 113, 1160–1163.

861 862

Reik, W. et al. (2001) Epigenetic reprogramming in mammalian development. Science (80-. )., 293, 1089–1093.

863 864

Ren, X. et al. (2018) Effects of glyphosate on the ovarian function of pregnant mice, the secretion of hormones and the sex ratio of their fetuses. Environ. Pollut., 243, 833–841.

865 866

Romkes, M. et al. (1987) Effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin on hepatic and uterine estrogen receptor levels in rats. Toxicol. Appl. Pharmacol., 87, 306–314.

867 868

Saillenfait, A.M. et al. (2016) The estrogenic and androgenic potential of pyrethroids in vitro. Review. Toxicol. Vitr., 34, 321–332.

869

Sakly, M. (2001) Reproductive toxicity of DDT in adult male rats. Hum. Exp. Toxicol., 20, 393–397.

870 871

Samardzija, D. et al. (2016) Atrazine blocks ovulation via suppression of Lhr and Cyp19a1 mRNA and estradiol secretion in immature gonadotropin-treated rats. Reprod. Toxicol., 61, 10–18.

872

Sassone-Corsi, P. (2012) The Cyclic AMP pathway. Cold Spring Harb. Perspect. Biol., 4, 3–5.

873 874 875

Schmutzler, C. et al. (2004) Endocrine active compounds affect thyrotropin and thyroid hormone levels in serum as well as endpoints of thyroid hormone action in liver, heart and kidney. Toxicology, 205, 95–102.

876 877

Schug, T.T. et al. (2016) Minireview: Endocrine Disruptors: Past Lessons and Future Directions. Mol. Endocrinol., 30, 833–847.

878 879

Seeger, B. et al. (2016) Mixture effects of estrogenic pesticides at the human estrogen receptor α and β. PLoS One, 11, 1–15.

880 881

Solecki, R. et al. (2017) Scientific principles for the identification of endocrine-disrupting chemicals: a consensus statement. Arch. Toxicol., 91, 1001–1006.

882 883

de Souza, J.S. et al. (2017) Perinatal exposure to glyphosate-based herbicide alters the thyrotrophic axis and causes thyroid hormone homeostasis imbalance in male rats. Toxicology, 377, 25–37.

884 885 886

Spirhanzlova, P. et al. (2017) Using short-term bioassays to evaluate the endocrine disrupting capacity of the pesticides linuron and fenoxycarb. Comp. Biochem. Physiol. Part - C Toxicol. Pharmacol., 200, 52–58.

887 888

Stouder, C. and Paoloni-Giacobino, A. (2011) Specific transgenerational imprinting effects of the endocrine disruptor methoxychlor on male gametes. Reproduction, 141, 207–216.

889

Strahl, B.D. and Allis, C.D. (2000) The language of covalent histone modifications. Nature, 403, 41–45.

890 891 892

Takeuchi, S. et al. (2006) In vitro screening of 200 pesticides for agonistic activity via mouse peroxisome proliferator-activated receptor (PPAR)α and PPARγ and quantitative analysis of in vivo induction pathway. Toxicol. Appl. Pharmacol., 217, 235–244.

893 894

Tavares, R.S. et al. (2015) In vitro exposure to the organochlorine p,p′-DDE affects functional human sperm parameters. Chemosphere, 120, 443–446.

895 896

Tavares, R.S. et al. (2013) p,p’-DDE activates CatSper and compromises human sperm function at environmentally relevant concentrations. Hum. Reprod., 28, 3167–3177.

897 898

Taylor, A.H. and Al-Azzawi, F. (2000) Immunolocalisation of oestrogen receptor beta in human tissues. J. Mol. Endocrinol., 24, 145–155.

899 900 901

Thomas, P. and Dong, J. (2019) Novel mechanism of endocrine disruption by fungicides through binding to the membrane androgen receptor, ZIP9 (SLC39A9), and antagonizing rapid testosterone induction of the intrinsic apoptotic pathway. Steroids, 149, 108415.

902 903 904

Tiemann, U. (2008) In vivo and in vitro effects of the organochlorine pesticides DDT, TCPM, methoxychlor, and lindane on the female reproductive tract of mammals: A review. Reprod. Toxicol., 25, 316–326.

905 906

Tilghman, S.L. et al. (2012) Endocrine disruptor regulation of microRNA expression in breast carcinoma cells. PLoS One, 7.

907 908

Tinfo, N.S. et al. (2011) Understanding the effects of atrazine on steroidogenesis in rat granulosa and H295R adrenal cortical carcinoma cells. Reprod. Toxicol., 31, 184–193.

909

Tüfekci, K.U. et al. (2014) The role of microRNAs in human diseases. Methods Mol. Biol., 1107, 33–50.

910

Turusov, V. et al. (2002) DDT Ubiquity persistence and risks. Environ. Heal. Perspect., 110, 125–128.

911 912

Uzumcu, M. et al. (2006) Early postnatal methoxychlor exposure inhibits folliculogenesis and stimulates anti-Mullerian hormone production in the rat ovary. J. Endocrinol., 191, 549–558.

913 914

Vandenberg, L.N. et al. (2019) Endocrine disruptors and the future of toxicology testing — lessons from CLARITY–BPA. Nat. Rev. Endocrinol., 1.

915 916

Vandenberg, L.N. et al. (2012) Hormones and Endocrine-Disrupting Chemicals: Low-Dose Effects and Nonmonotonic Dose Responses. Endocr. Rev., 33, 1–78.

917 918

Vandenberg, L.N. et al. (2013) Regulatory decisions on endocrine disrupting chemicals should be based on the principles of endocrinology. Reprod. Toxicol., 38, 1–15.

919 920

Ventura, C. et al. (2019) Effects of the pesticide chlorpyrifos on breast cancer disease. Implication of epigenetic mechanisms. J. Steroid Biochem. Mol. Biol., 186, 96–104.

921 922

Victor-Costa, A.B. et al. (2010) Changes in testicular morphology and steroidogenesis in adult rats exposed to Atrazine. Reprod. Toxicol., 29, 323–331.

923 924

Vinggaard, A.M. et al. (2005) Antiandrogenic effects in short-term in vivo studies of the fungicide fenarimol. Toxicology, 207, 21–34.

925 926

Vinggaard, A.M. et al. (2002) Antiandrogenic effects in vitro and in vivo of the fungicide prochloraz. Toxicol. Sci., 69, 344–353.

927 928

Wang, Q. et al. (2019) The anti-androgenic effects of cypermethrin mediated by non-classical testosterone pathway activation of mitogen-activated protein kinase cascade in mouse Sertoli cells.

929

Ecotoxicol. Environ. Saf., 177, 58–65.

930 931

Weikum, E.R. et al. (2018) The nuclear receptor superfamily: A structural perspective. Protein Sci., 27, 1876–1892.

932 933

Welshons, W. V. et al. (2003) Large Effects from Small Exposures. I. Mechanisms for EndocrineDisrupting Chemicals with Estrogenic Activity. Environ. Health Perspect., 111, 994–1006.

934 935

Wilson, V.S. et al. (2008) Diverse mechanisms of anti-androgen action: Impact on male rat reproductive tract development. Int. J. Androl., 31, 178–185.

936 937

Wirbisky, S.E. et al. (2016) Embryonic atrazine exposure alters zebrafish and human miRNAs associated with angiogenesis, cancer, and neurodevelopment. Food Chem. Toxicol., 98, 25–33.

938 939

Xiang, D. et al. (2017) Structure-based investigation on the binding and activation of typical pesticides with thyroid receptor. Toxicol. Sci., 160, 205–216.

940 941

Xie, L. et al. (2018) Delayed Puberty by Ziram Is Associated with Down Regulation of Testicular Phosphorylated AKT1 and SIRT1/PGC-1α Signaling. Chem. Res. Toxicol., 31, 1315–1322.

942 943 944

Yaglova, N. V. and Yaglov, V. V. (2015) Mechanisms of Disruptive Action of Dichlorodiphenyltrichloroethane (DDT) on the Function of Thyroid Follicular Epitheliocytes. Bull. Exp. Biol. Med., 160, 231–233.

945 946

Yao, Q. et al. (2019) The roles of microRNAs in epigenetic regulation. Curr. Opin. Chem. Biol., 51, 11– 17.

947 948

Ye, X. et al. (2017) Pyrethroid Insecticide Cypermethrin Accelerates Pubertal Onset in Male Mice via Disrupting Hypothalamic-Pituitary-Gonadal Axis. Environ. Sci. Technol., 51, 10212–10221.

949 950

You, L. (2004) Steroid hormone biotransformation and xenobiotic induction of hepatic steroid metabolizing enzymes. Chem. Biol. Interact., 147, 233–246.

951 952

Zama, A.M. and Uzumcu, M. (2009) Fetal and neonatal exposure to the endocrine disruptor methoxychlor causes epigenetic alterations in adult ovarian genes. Endocrinology, 150, 4681–4691.

953 954

Zhang, C. et al. (2019) Nuclear receptor AHR-mediated xenobiotic detoxification pathway involves in atrazine-induced nephrotoxicity in quail (Coturnix C. coturnix). Environ. Pollut., 253, 889–898.

955 956

Zhang, D. et al. (2017) Exposure to 2,4-dichlorophenoxyacetic acid induces oxidative stress and apoptosis in mouse testis. Pestic. Biochem. Physiol., 141, 18–22.

957 958

Zhang, H. et al. (2010) Pubertal and early adult exposure to fenvalerate disrupts steroidogenesis and spermatogenesis in mice at adulthood. J. Appl. Toxicol., 30, 369–377.

959

Zhao, C. et al. (2008) Estrogen receptor beta: an overview and update. Nucl. Recept. Signal., 6, 1–10.

960 961

Zoeller, R.T. (2007) Environmental chemicals impacting the thyroid: Targets and consequences. Thyroid, 17, 811–817.

962 963

Zoeller, R.T. (2012) Neuroendocrine regulation of development, growth and metabolism - thyroid Elsevier Inc.

964 965 966